MXPA06008516A - Susceptibility gene for myocardial infraction, stroke, and paod; methods of treatment. - Google Patents

Susceptibility gene for myocardial infraction, stroke, and paod; methods of treatment.

Info

Publication number
MXPA06008516A
MXPA06008516A MXPA06008516A MXPA06008516A MXPA06008516A MX PA06008516 A MXPA06008516 A MX PA06008516A MX PA06008516 A MXPA06008516 A MX PA06008516A MX PA06008516 A MXPA06008516 A MX PA06008516A MX PA06008516 A MXPA06008516 A MX PA06008516A
Authority
MX
Mexico
Prior art keywords
allele
seq
leukotriene
flap
haplotype
Prior art date
Application number
MXPA06008516A
Other languages
Spanish (es)
Inventor
Anna Helgadottir
Hakon Hakonarson
Jeffrey R Gulcher
Mark E Gurney
Original Assignee
Decode Genetics Ehf
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
Priority claimed from US10/769,744 external-priority patent/US20050282855A1/en
Priority claimed from US10/830,477 external-priority patent/US7851486B2/en
Priority claimed from US10/829,674 external-priority patent/US7507531B2/en
Application filed by Decode Genetics Ehf filed Critical Decode Genetics Ehf
Priority claimed from PCT/US2005/003312 external-priority patent/WO2005075022A2/en
Publication of MXPA06008516A publication Critical patent/MXPA06008516A/en

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Linkage of myocardial infarction (MI) and a locus on chromosome 13q12 is disclosed. In particular, the FLAP gene within this locus is shown by genetic association analysis to be a susceptibility gene for MI and ACS, as well as stroke and PAOD. Pathway targeting for treatment and diagnostic applications in identifying those who are at risk of developing MI, ACS, stroke or PAOD, in particular are described. The invention also provides for compositions comprising a leukotriene synthesis inhibitor and a stating and methods of using these compositions to reduce C-reactive protein in a human subject at risk of MI, ACS, stroke and/or PAOD.

Description

GENETIC SUSCEPTIBILITY FOR INFARCTION TO MYOCARDIUM, APOPLEJÍA AND PAOD; METHODS OF TREATMENT BACKGROUND OF THE INVENTION Myocardial Infarction (MI) and the Syndrome Acute coronary (ACS), for example, unstable angina, myocardial infarction without ST elevation (NSTEMI) or myocardial infarction with ST elevation (STEMI), are the main causes of hospital admissions in industrialized countries. Cardiovascular disease continues to be the leading cause of death in the United States, Europe and Japan. The costs of the disease are high both in terms of morbidity and mortality, as well as in terms of the economic burden on health care systems. Myocardial infarction usually occurs when there is an abrupt decrease in coronary blood flow followed by a thrombotic occlusion of a coronary artery previously damaged by atherosclerosis. In most cases, the infarction occurs when an atherosclerotic plaque clears, fractures or ulcerates, and when conditions favor thrombogenesis. In rare cases, the infarction may be due to coronary artery occlusion caused by coronary embolism, congenital abnormalities, coronary spasm, and a wide variety of systemic diseases, particularly inflammatory ones. Medical risk factors for MI include smoking, diabetes, hypertension, and total serum cholesterol levels of > 200 mg / dL, high serum LDL cholesterol, and low serum HDL cholesterol. Percentages of cases in individuals without a prior history of cardiovascular disease are approximately 1%. In individuals who have had a first MI or ACS, the risk of repeat MI within the following year is 10-14%, despite maximum medical management including angioplasty and stenting. Atherosclerosis can affect the vascular beds in many large and medium arteries. Myocardial infarction and unstable angina (acute coronary syndrome) (ACS)) originates from atherosclerosis of the coronary artery, whereas ischemic stroke is more frequently a consequence of atherosclerosis of the carotid or cerebral artery. Member ischemia caused by peripheral arterial occlusive disease (PAOD) can occur as a consequence of atherosclerosis of the iliac, femoral, and popliteal arteries. Atherosclerotic diseases are common despite the widespread use of drugs that inhibit thrombosis (aspirin) or treat medical risk factors such as high blood cholesterol levels (statins), diabetes or hypertension (diuretics and anti-hypertensives). Atherosclerotic disease is initiated by the accumulation of lipids within the arterial wall, and in particular, the accumulation of low density lipoprotein (LDL) cholesterol. The trapped LDL becomes oxidized and internalized by macrophages. This causes the formation of atherosclerotic lesions that contain accumulations of macrophages flooded with cholesterol, referred to as "foam cells". When the disease progresses, the smooth muscle cells proliferate and grow inside the arterial wall forming a "fibrous cap" of extracellular matrix that contains a necrotic nucleus, rich in lipids. Present in the arterial walls of most people throughout their lives, fibrous atherosclerotic plaques are relatively stable. Such fibrous lesions cause extensive remodeling of the arterial wall, displacing the outer, elastic membrane without the reduction in luminal diameter or the serious impact on the oxygen supply to the heart. Consequently, patients may develop large fibrous atherosclerotic lesions without luminal shrinkage until the last hour in the disease process. However, the coronary artery lumen may eventually shrink over time and in some cases affect blood flow to the heart, especially under high states of demand such as exercise. This can result in reversible ischemia causing chest pain mitigated by rest called stable angina. In contrast to the relative stability of fibrous atherosclerotic lesions, the culprit lesions associated with myocardial infarction and unstable angina (each of which are part of the acute coronary syndrome) are characterized by a thin fibrous cap, a large lipid nucleus, and the infiltration of inflammatory cells such as T- lymphocytes and monocytes / macrophages. Noninvasive imaging techniques have shown that most MIs occur in sites with low or intermediate grade stenosis, which indicates that coronary artery occlusion is most often due to the rupture of culprit lesions with consequent formation of a thrombus or blood clot and is not due solely to luminal shrinkage due to stenosis. The rupture of the plaque may be due to erosion or unequal thinning of the fibrous cap, usually in the margins of the lesion where the macrophages enter, accumulate and become activated by a local inflammatory process. The thinning of the fibrous cap can result from the degradation of the extracellular matrix by proteases released from activated macrophages. These changes that produce plaque instability and MI risk can be increased by the production of tissue factor pro-coagulant and other factors that increase the likelihood of thrombosis. In acute coronary syndrome, the culprit lesion showing rupture or erosion with local thrombosis is usually treated by angioplasty or by dilation and balloon placement of a stent to maintain luminal evidence. Patients who experience ACS are at high risk for a second coronary episode due to the nature of multiple blood vessels of coronary artery disease with percentages of cases approaching 10-14% within 12 months after the first incident . The incipient view of MI is as an anti-inflammatory disease of the arterial vascular wall in pre-existing chronic atherosclerotic lesions, which sometimes trigger rupture of culprit lesions and lead to local thrombosis and infarction to the subsequent myocardium. The process that activates and sustains arterial wall inflammation leading to plaque instability is unknown, however, it results in the release within the circulation of the tumor necrosis factor of alpha and interleukin-6. These and other cytokines or biological mediators released from the damaged vascular wall stimulate an inflammatory response in the liver causing elevation in several non-specific general inflammatory markers including C-reactive protein. Although not specific to atherosclerosis, C-reactive protein (CRP) and serum amyloid A seem to predict the risk of MI, perhaps as substitutes for vascular wall inflammation. Although classical risk factors such as smoking, hyperlipidemia, hypertension and diabetes are associated with many cases of coronary heart disease (CHD) and MI, many patients have no implication of these risk factors. In fact, many patients who exhibit one or more of these risk factors do not develop MI. Family history has long been recognized as one of the greatest risk factors. Although some of the MI family groups reflect the genetic contribution to the other conventional risk factors, a large number of studies have suggested that there are significant genetic susceptibility factors, beyond the known risk factors (Friedlander Y, et al. , Br. Herat J. 1985; 53: 382-7, Shea S. et al. , J. Am. Coll. Cardiol. 1984, 4: 793-801, and Hopkins P.N., et al. , Am. J. Cardiol. 1988; 62: 703-7). The main genetic susceptibility factors have only been identified for rare Mendelian forms of hyperlipidemia such as familial hypercholesterolemia. Genetic risk is conferred by subtle differences in genes between individuals in a population. Genes differ among individuals more frequently due to single nucleotide polymorphisms (SNPs), although other variations are important. The SNPs are located on average every 1000 base pairs in the human genome. Therefore, a typical human gene containing 250,000 base pairs can contain 250 different SNPs. Only a small number of SNPs are located in exons and alter the amino acid sequence of the protein encoded by the gene. The majority of SNPs have no effect on genetic function, while others can alter the transcription, splicing, translation or stability of the mRNA encoded by the gene. The additional genetic polymorphism in the human genome is caused by the insertion, deletion, displacement or reversal of any short or long broadening of the DNA. Genetic polymorphisms that confer disease risk can therefore directly alter the amino acid sequence of the proteins, increase the amount of protein produced from the gene, or decrease the amount of protein produced by the gene. When genetic polymorphisms that confer risk of disease are not covered, genetic testing for such risk factors becomes important for clinical medicine. Examples are the apolipoprotein E test that identifies the genetic carriers of the apoE4 polymorphism in patients with dementia for the differential diagnosis of Alzheimer's disease and the Leiden test of Factor V for predisposition to deep vein thrombosis. Most notably, in the treatment of cancer, the diagnosis of genetic variants in tumor cells is used for the selection of the most appropriate treatment regimen for the individual patient. In breast cancer, the genetic variation in estrogen receptor expression or tyrosine kinase expression of hergulin receptor type 2 (Her2) determines whether the anti-estrogenic drugs (tamoxifen) or the anti-Her2 antibody (Herceptin) will be incorporated into the treatment plan. In the diagnosis of chronic chromosome genetic shift (CML) Philadelphia chromosomes that mix the genes encoding the tyrosine kinases of Bcr and Abl receptors indicate that Gleevec (STI571), a specific inhibitor of the Bcr-Abl kinase should be used to the treatment of cancer. For patients with CML with such genetic alteration, inhibition of Bcr-Abl kinase leads to rapid elimination of tumor cells and remission from leukemia. Many general inflammatory markers predict the risk of coronary heart disease, although these markers are not specific to atherosclerosis. For example, Stein (Stein, S., Am. J. Cardiol, 87 (suppl): 21A-26A (2001)) discusses the use of any of the following inflammatory markers in serum as substitutes to predict the risk of coronary heart disease. including reactive protein C (CRP), amyloid serum A, fibrinogen, interleukin-6, tissue factor necrosis alpha, soluble vascular cell adhesion molecules (sVCAM), soluble intervascular adhesion molecules (sICAM), E-selectin, metalloprotease type 1 matrix, type 2 matrix metalloprotease, type 3 matrix metalloprotease and type 9 matrix metalloprotease. Elevation in one or more of these inflammatory markers in serum is not specific for coronary heart disease, but it also occurs with age or in association with cerebrovascular disease, peripheral vascular disease, non-insulin-dependent diabetes, osteoarthritis, bacterial infection and sepsis. Serum reactive protein C (CRP) is examined as a convenient and sensitive marker of systemic inflammation. Generally, CRP is measured in serum samples using enzyme-linked immunosorbent assay (EIA). Consistent across multiple published studies is the finding of a correlation between the increased risk of coronary artery disease with CRP in increased serum. For example, in the omen's Health Study, CRP was measured in 27,939 apparently healthy American women. The cut-off points for serum CRP quintiles in women were: less than or equal to 0.49, more than 0.49 to 1.08, more than 1.08 to 2.09, more than 2.09 to 4.19, and more than 4.19 mg of CRP per liter, see Ridker, PM et al. , New England, J. Med., 347: 1557-1565 (2001). Compared to the lowest quintiles, and even when adjusted for age, each quintile that has more than 0.49 mg of CRP per liter was associated with the increased risk for coronary heart disease with the highest relative risk of 4.5 seen for those women in the highest quintile of CRP in serum (more than 4.19 mg of CRP per liter). A similar correlation has been reported between increased serum CRP and increased risk for coronary heart disease in women (Ridker, PM, et al., New England, J. Med., 342: 836-843 (2000) and Bermudez, EA, et al., Arterioscler, Thromb. Vasc. Biol., 22: 1668-1613 (2002)). Men also show a correlation between increased inflammatory markers in serum such as CR and the increased risk for coronary heart disease has been reported (Doggen, CJM et al., J. Internal! Med., 248: 406-414 (2000 ) and Ridker, PM et al., New England, J. Med., 336: 913-979 (1997). The quintiles for serum CRP as reported by Doggen et al., were less than 0.65, greater than 0.65 a. 1.18, greater than 1.18 to 2.07, greater than 2.07 to 4.23, and greater than 4.23 mg of CRP per liter.Unlike in women, elevated CRP in serum correlates with the increased relative risk for coronary heart disease only in the 4th and 5th fifth of CRP (relative risk of 1.7x and 1.9x, respectively) Serum CRP in women has also been measured along with lipid markers such as serum low density lipoprotein cholesterol (LDL-C) levels In the study by Ridker, PM et al. (2002), the CRP and serum LDL-C correlate minimally, they are classified for both serum markers that provide better prognostic indication than either alone. Thus, women with CRP in serum over medium values (more than 1.52 mg of CRP per liter) and also serum LDL-C over medium values (more than 123.7 mg of LDL-C per deciliter) were at risk more elevated for coronary heart disease. Elevated CRP or other inflammatory markers in serum is also a high prognosis of increased risk of a second myocardial infarction in patients with previous myocardial infarction (Retterstol, L. • et al., Atheroscler., 150: 433-440 ( 2002)). Since CRP occurs in the liver, there is no a priori mechanistic explanation of why elevation in CRP and other inflammatory markers in serum should be the prognosis for coronary artery disease. As discussed by Doggen, C. J. M., et al. , one or more of the following factors were speculated to determine the observed correlation; (1) intrinsic inflammation and tissue damage within the arterial lesions, (2) previous infection by Helicobacter pylori or by Chlamydia pneumoniae, (3) release of peptide cytokines including interleukin-6, or (4) activation of the complementary system. The end products of the leukotriene pathway are potent mediators of inflammatory lipids derived from arachidonic acid. These can potentially contribute to the development of atherosclerosis and the destabilization of atherosclerotic plaques through the oxidation of lipids and / or the pro-inflammatory effects. It is known that LTC4, LTD4 and LTE4 induce vasoconstriction. Alien et al. , Circulation, 57: 2406-2413 (1998) describes a novel mechanism in which atherosclerosis is associated with the appearance of leukotriene receptors capable of inducing hyperactivity of the human epicardial coronary arteries in response to LTC4 and LTD4. LTB4, on the other hand, is a strong pro-inflammatory agent. The increased production of these end products, from the leukotriene pathway, could therefore serve as a risk factor for MI and atherosclerosis, while both inflammation and vasoconstriction / vasospasm have a well-established role in the pathogenesis of MI and Atherosclerosis It has also been shown that a heterozygous deficiency of the 5-LO enzyme in a knockout mouse model decreases the size of the atherosclerotic lesion in LDLR - / - mice by approximately 95% (Mehrabian et al., Circulation Research, 91: 120 (2002 )). However, no such genetic evidence has been reported for involvement of leukotriene in MI or atherosclerosis in humans. Mehrabian et al. , did not report a very small genetic association study when examining the correlation between promoter polymorphisms of 5-LO and intimate thickening of the carotid in normal individuals. However, their data suggest paradoxically that a lower amount of leukotriene production correlates with atherosclerosis in the carotid.
COMPENDIUM OF THE INVENTION As dibed herein, a gene on chromosome 13ql2-13 has been identified as playing a major role in myocardial infarction (MI). This gene, hereinafter referred to as the MI gene, comprises nucleic acid encoding the 5-lipoxygenase activation protein (AL0X5AP or FLAP) herein after referred to as FLAP. The gene has also been shown to play a role in stroke and PAOD. The invention relates to methods of treatment (prophylactic and / or therapeutic) for certain diseases and conditions (e.g., MI, ACS, atherosclerosis, stroke, PAOD) associated with FLAP or with other members of the leukotriene pathway (e.g., biosynthetic enzymes or proteins such as FLAP, arachidonate 4-liposigenase (5-LO), leukotriene C4 synthase (LTC4S), leukotriene A4 hydrolase (LTA4H), leukotriene B4 12-hydroxideshydrogenase (LTB4DH)); receptors and / or binding agents of the enzymes; and receptors for leukotrienes LTA4, LTB4, LTC4, LTD4, LTE4, Cys LT1, Cys LT2, including leukotriene receptor B4 (BLT1), leukotriene receptor B4 (BLT2), cysteinyl leukotriene receptor 1 (CysLTRl), cysteinyl leukotriene receptor 2 (CysLTR2). The methods include the following: methods of treatment for myocardial infarction or susceptibility to myocardial infarction; prophylaxis therapy methods for myocardial infarction; treatment methods for transient ischemic attack, transient monocular blindness or stroke, or sus- ceptibility to stroke; treatment methods for claudication, PAOD or susceptibility to PAOD; treatment methods for acute coronary syndrome (eg, unstable angina, myocardial infarction without ST elevation (NSTEMI) or myocardial infarction with ST elevation), methods to reduce the risk of MI, stroke or PAOD in people with ankle index / asymptomatic brachialis less than 0.9, methods to reduce the risk of a second myocardial infarction or stroke, methods of treatment for atherosclerosis, such as for patients requiring treatment (eg, angioplasty, stents, revascularization procedure) restore blood flow in arteries (eg, coronary, carotid and / or femoral arteries), treatment methods for asymptomatic ankle / brachial index of less than 0.9, and / or methods to decrease leukotriene synthesis (eg, for the treatment of myocardial infarction, stroke or PAOD).
The invention provides methods for prophylaxis therapy for myocardial infarction (MI). These methods comprise selecting a human subject susceptible to MI, by administering to the subject a composition comprising a therapeutically effective amount of a therapeutic agent MI that inhibits the synthesis of leukotriene in vivo, wherein the therapeutic agent MI inhibits the synthesis of leukotriene by inhibiting the activity of at least one protein selected from the protein that activates 5-lipoxygenase (FLAP) and 5-lipoxygenase (5-LO). The methods also comprise monitoring the level of myeloperoxidase before and during prophylaxis treatment, wherein the therapeutic agent MI is administered in an effective amount to reduce MPO levels in a subject. These methods may further comprise monitoring at least one additional inflammatory marker, such as a C reactive protein, in the human subject before and during prophylaxis therapy. In the methods of the invention, a leukotriene synthesis inhibitor is administered to an individual in a therapeutically effective amount. The leukotriene synthesis inhibitor can be an agent that inhibits or antagonizes a member of the leukotriene synthesis pathway (e.g., FLAP, 5-LO, LTC4S, LTA4H and LTB4DH). For example, the leukotriene synthesis inhibitor can be an agent that inhibits or antagonizes the polypeptide activity of FLAP (eg, a FLAP inhibitor) and / or the expression of FLAP nucleic acid as dibed herein (e.g., a FLAP nucleic acid antagonist). In another embodiment, the leukotriene synthesis inhibitor is an agent that inhibits or antagonizes polypeptide activity and / or nucleic acid expression of another member of the biosynthetic pathway of leukotriene (e.g., LTC4S, LTA4H) or that increases disruption of leukotrienes (for example, LTB4DH). In preferred embodiments, the agent alters the activity and / or expression of the nucleic acid of FLAP or 5-LO. Preferred agents include those set forth in Table I of the Agent herein. In another embodiment, preferred agents may be: 1- ((4-chlorophenyl) methyl) -3- ((1,1-dimethylethyl) thio) -alpha, alpha-dimethyl-5- (2-quinolinylmethoxy) -lH- acid Indole-2-propanoic or otherwise known as MK-0591, (R) - (+) - alpha-cyclopentyl-4- (2-quinolinemethoxy) -Benzenacetic acid otherwise known as BAY-x-1005, acid 3 - (3- (1,1-dimethylethylthio-5- (quinolin-2-ylmethoxy) -1- (4-chloromethylphenyl) indol-2-yl) -2,2-dimethylpropionaldehyde oxime-0-2-acetic acid otherwise known as A-81834, enantiomerics, salts, chemical derivatives and optically pure analogues; or they can be zileuton, atreleuton, 6 - ((3-fluoro-5- (tetrahydro-4-methoxy-2H-pyran-4-yl) phenoxy) methyl) -1-methyl-2 (1H) -quinolinone otherwise known as ZD-2138, 1- ((4-chlorophenyl) methyl) -3- ((1,1-dimethylethyl) thio) -alpha, alpha-dimethyl-5- (2-quinolinomethoxy) -lH-Indol-2 -propanoic otherwise known as MK-886, 4- (3- (4- (2-Methyl-imidazol-1-yl) -phenylsulfanyl) -phenyl) -tetrahydro-pyran-4-carboxylic acid amide another mode known as CJ-13610, its enantiomers, salts, chemical derivatives and optically pure analogues. In another embodiment, the agent alters the metabolism or activity of a leukotriene (eg, LTA4, LTB4, LTC4, LTD4, LTE4, Cys LT1, Cys LT2) such as leukotriene antagonists or antibodies to leukotrienes, as well as agents which alter the activity of a leukotriene receptor (eg, BLT1, BLT2, CysLTR1 and CysLTR2). The results in Example 10 demonstrate that in patients with the FLAP and LTA4 haplotypes at risk, a FLAP inhibitor (DG-031 also known as Bay-X-1005) had an extremely significant and dose-dependent effect on the total cellular blood and the level of urinary metabolite that includes a reduction of 26% in the production of leukotriene B4 by activated neutrophils, a reduction of 13% of myeloperoxidase in whole blood, and a 27% increase in urinary leukotriene E4. In addition, there was evidence of a persistent effect, followed by discontinuation of the FLAP inhibitor, in sensitive, high reactive C protein and amyloid serum A. This reduction in CRP and amyloid serum A was observed in the upper part of the beneficial effects that may have been achieved by statins taken by 85% of the subjects under study. The invention provides compositions comprising a leukotriene synthesis inhibitor and a statin. The invention also provides the use of a leukotriene synthesis inhibitor and a statin for the manufacture of a medicament for reducing CRP levels in a human subject. Such compositions are intended for human administration, and preferably further comprise (at least one) a pharmaceutically acceptable diluent, adjuvant, excipient or carrier. The materials and methods for formulation and co-formulation are well known, and many are described herein in greater detail. In one variation, formulation of the composition in convenient unit dose formulations, such as pills or capsules for oral administration, including sustained release formulations, is specifically contemplated. In another variation, transdermal co-administration, for example, through a skin patch is contemplated. In yet another variation, the administration of one or both agents through a drug-eluting stent is specifically contemplated. In particular, the compositions may comprise a leukotriene synthesis inhibitor that inhibits the activity of a member of the leukotriene synthesis pathway such as 5-lipoxygenase, 5-lipoxygenase activation protein (FLAP), leukotriene C4 synthase, leukotriene A4 hidolase, arachidonate 4-lipoxingenase, leukotriene B4 12-hydroxideshydrogenase, leukotriene A4 receptor, leukotriene B4 receptor, leukotriene C4 receptor, leukotriene D4 receptor, leukotriene E4 receptor, leukotriene B4 receptor 1, leukotriene receptor B4, cysteinyl leukotriene receptor 1 and cysteinyl leukotriene receptor 2. Any LT inhibitor is suitable for the practice of the invention, and several LT inhibitors are described herein. To help minimize side effects, an LT inhibitor that is specific for a member of the LT synthesis path is preferred. Exemplary inhibitors include both small molecules and biological inhibitors of proteins (e.g., antibody substances, peptides), and biological inhibitors that operate at the nucleic acid level (e.g., non-coding nucleic acids and nucleic acids from interfering RNA and proteins). of zinc finger). Preferred agents that inhibit the activity of a member of the leukotriene path are listed in Table I of the Agent herein, including the following agents: 1- ((4-chlorophenyl) methyl) -3- ((1, 1- 'dimethylethyl) thio) -alpha, alpha-dimethyl-5- (2- (2- quinolinylmethoxy) -1H- indol-2-propanoic acid (R) - (+) - alphal-cyclopentyl-4- (2-quinolinylmethoxy) -Benzenacetic acid, 3- (3- (1,1-dimethylethylthio-5- ( quinolin-2-ylmethoxy) -1- (4-chloromethylphenyl) indol-2-yl) -2,2-dimethylpropionaldehyde oxime-0-2-acetic acid, zileuton, atreleuton, 6- ((3-fluoro-5- (tetrahydro -4-methoxy-2H-pyran-4-yl) phenoxy) methyl) -1-methyl-2 (1H) -quinolinone, 1- ((4-chlorophenyl) methyl) -3- ((1, l-dimethylethyl) thio) - alpha, alf -dimethyl-5- (2-quinolinylmethoxy) -lH-indol-2-propanoic acid and 4- (3- (4- (2-Methyl-imidazol-l-yl) -phenylsulfanyl) -phenyl ester) -tetrahydro-pyran-4-carboxylic acid In one variation the inhibitor LT is a FLAP inhibitor.A preferred group of compounds is described herein as BAY X105 (also known as DG-031) as well as related compounds described s in Mohrs, et al., U.S. Patent No. 4,970,215, incorporated herein by reference in its entirety. In another variation, the LT inhibitor is an LTA4H inhibitor. Other preferred agents include those set forth in Table II of the Agent and the list of the LTA4H Agent set forth herein. Additional preferred agents include those described in Penning et al. , Med Chem. 2002 45 (16): 3482-90, Penning, Curr Pharm Des. 2001, 7 (3): 163-79 and Penning et al. , J Med Chem. 2000 43 (4): 721-35.
TABLE II AGENT In addition, the following LTA4H inhibitors are described in USP2003 / 0004101A1, the teachings of which are incorporated herein by reference in their entirety: ADDITIONAL LIST OF AGENT LTA4H 1. 1- [2- [4- (phenylmethyl) phenoxy] ethyl] -2-methyl-4-tetrazolylpiperidine 2. 1- [2- [4- (4-oxazolyl) phenoxy) phenoxy] ethyl] irrolidine 3. 3- [methyl] [3- [4- (2-thienylmethyl)] phenoxy] propyl] amino] propionic 4. 3- [methyl [3- [4- (2-thienylmethyl) phenoxy] propyl] amino] propionate methyl 3- [methyl] [3- [4- (3-thienylmethyl]] ) phenoxy] pro-pil] amino] propionic 6. 3- [methyl] [3-4- (3-thienylmethyl) phenoxy] propyl] amino] propionate 7. acid 3 [methyl] 3-4- (4-fluorophenoxy) ) phenoxy] propyl] amino] propionic 8. 3- [methyl [3- [4- (4-biphenyloxy) phenoxy] propyl] amino] propionic acid 9. N- [3- [[3- [4- (phenylmethyl) phenoxy] propyl] methylamino] propionyl] benzenesulfonamide 10. 1- [2- [4-phenylmethyl) phenoxy] ethyl] -2-methyl-4- (1H- tetrazol-5-yl) piperidine 11. 1- [2- [4- (phenylmethyl) phenoxy] ethyl] -4- (1H-tetrazol-5-yl) piperidine In some embodiments, the compositions of the invention comprise a statin, and the methods of the invention comprise the administration of a statin. In this context, the term "statin" should be understood to refer to any of the class of inhibitors of 3-hydroxy-3-methylglutarylcoenzyme A (HMG-CoA) reductase, the enzyme that converts HGM-CoA to the mevalonic acid cholesterol precursor. Numerous compounds with high specificity for this enzyme have been developed and approved for human therapy. The compositions of the invention may comprise a statin that is listed in Table III of the present Agent, such as rovuvastatin (also known as visastatin), fluvastatin, atorvastatin, lovastatin (also known as mevolin), simvastatin, pravastatin, pitavastatin, mevastatin, crevastatin, ML-236a, ML-236B, MBV-530A and MB-530B. References to agents should be understood to include salts, acids, bases, esters, pro-drugs, metabolites and other pharmaceutically acceptable common formulation variants of the agents. A growing body of new evidence identifies serum CRP as a marker for cardiovascular morbidity / mortality, and correlates with reductions in serum CRP for better clinical outcomes (See for ele, Ridker et al., N. Engl. J. Med. 352 (1): 20-28 (2005); Nissen et al., N. Engl. J. Med. 352 (1): 29-38 (2005); and Pearson et al., Circulation 107: 499-511 (2003)). Serum CRP in excess of 3.0 mg / l is considered high risk; 1.0 to 3.0 risk on average; and below 1 mg / l of low risk. (Pearson et al.) The compositions and methods of the invention provide tools for reducing CRP in serum. Reductions in CRP can be measured on a concentration basis, where compositions and methods that achieve CRP below 3.0 mg / l are preferred; with even more preferred targets of 2.75 mg / L, 2.5 mg / L, 2.25 mg / L, 2.0 mg / L, 1.75 mg / L, 1.5 mg / L, 1.25 mg / L, 1.0 mg / L, 0.75 mg / L 0.5 mg / l. Reductions in CRP can also be measured on a percentage basis, where clinical effectiveness is evaluated as a percentage reduction in CRP in a patient compared to no drug therapy or compared to single drug therapy. Depending on the initial CRP measurement, compositions and methods that reduce CRP anywhere from 10% -90% or more are contemplated, for example, reductions of 10%, 20%, 25%, 30%, 40% , 50%, 60%, 65%, 70%, 75%, 80% or any objective between these values. The invention also contemplates methods for reducing MPO and a method for monitoring MPO levels. Reductions in MPO can be measured in a concentration basis, where the compositions and methods that reduce the MPO level relative to the quartile distribution of MPO in the normal population (ie, from the fourth quartile to the 3rd quartile or from the 3a to 2a) are preferred. Reductions in MPO can also be measured on a percentage basis, where clinical effectiveness is evaluated as a percentage reduction in MPO in a patient compared to no drug therapy or compared to single drug therapy. Depending on the initial MPO measurement, compositions and methods that reduce MPO anywhere from 10% -90% or more are contemplated, for example, reductions of 10%, 20%, 25%, 30%, 40%, 50 %, 60%, 65%, 70%, 75%, 80% or any objective between these values. In some variations of the invention, the composition of the invention includes the leukotriene synthesis inhibitor in an amount effective to reduce serum reactive protein C (CRP) in a human subject. In some variations, the composition of the invention includes the statin in an effective amount that lowers low density lipoprotein cholesterol (LDL) and CRP reduces serum CRP in a human subject. In at least one preliminary and short term study described herein, human subjects who already enjoyed the benefits that decrease CRP of statin therapy were administered the BAY-X1005 LT inhibitor, and significant additional reductions in CRP were detected. Combination therapy of a longer duration may result in an additional CRP reduction than the 20-30% effect observed in the short term study. In one embodiment of the invention, the compositions comprise a leukotriene synthesis inhibitor in an amount effective to reduce serum CRP in a human subject and a statin. In another embodiment, the compositions comprise a statin in an amount effective to reduce serum LDL-C in a human subject and a leukotriene synthesis inhibitor. The invention also encompasses compositions comprising a leukotriene synthesis inhibitor and a statin in amounts effective to synergistically reduce CRP in a human subject. In one variation, the leukotriene inhibitor and the statin are included in the composition of the invention in effective amounts that synergistically reduce the reactive protein C in serum in a human subject. For the practice of the invention with BAY-X1005, doses of 50-750 mg per day for human adult patients are contemplated. The dose of 100-500 mg is contemplated, from one to five times per day. The dose of 250-375 mg, from one to three times per day, is preferred. The dosage for clinically approved statins has been developed and published by the manufacturers.
In a preferred embodiment, the statin is co-formulated with the inhibitor LT in a pill or a capsule for administrations of 1-4 times per day. The invention provides methods for using these compositions that reduce risk factors for cardiovascular diseases such as for MI, ACS, stroke or PAOD. In one method, a composition comprising a leukotriene synthesis inhibitor and a statin is administered to a human subject exhibiting one or more risk factors for MI, ACS, stroke or PAOD, wherein the composition is administered in an effective amount that reduces at least one risk factor for MI, ACS, stroke or PAOD. Preferably, the risk factor is elevated serum LDL-C or an elevated inflammatory marker such as CRP or amyloid serum A. In a highly preferred embodiment, LDL-C and CRP both are clinically significant quantities reduced, wherein a quantity clinically significant is an amount that correlates with a statistically significant measurable reduction in risk of an adverse cardiovascular event, when analyzed in a population, for example, in a clinical study. The invention also provides a method for using these compounds that reduce CRP in a human subject. In one variation, the invention is a method for reducing reactive protein C (CRP) in a human subject, which comprises administering to a human being in need of treatment to reduce CRP a composition of the invention containing the LT inhibitor and the statin as described above, in an amount effective to reduce reactive protein C in serum in the human subject. The identification of a human being in need of treatment for CRP reduction can be based on a variety of factors described herein, including genetic factors, CRP measurements, measurements of other inflammatory markers, and measurements of non-genetic and non-inflammatory markers. MI risk. In one variation, the method includes selecting for the administration step a human subject at risk of a disease or condition selected from the group consisting of myocardial infarction, acute coronary syndrome, stroke or peripheral arterial occlusive disease. Thus, the invention provides a method comprising selecting a human subject at risk of MI, ACS, stroke or PAOD and administering to the subject a composition comprising a leukotriene synthesis inhibitor and a statin wherein the composition is in an amount effective that reduces CRP in serum in a human subject. The method may further comprise the step of measuring CRP in serum in the human subject to verify the therapeutic efficacy of the composition, wherein a decrease in serum CRP followed by administration of the composition indicates therapeutic efficacy. In yet another variation, the verification of risk factors and / or toxicity is used to adjust dosage or dosage. For example, the dose or dosage of a statin or a leukotriene synthesis inhibitor is increased if the serum CRP and / or serum LDL and / or serum or urinary leukotriene measurements does not decrease to a target level, such as a level equivalent to the 50th lowest percentile, 40th percentile, 30th percentile, 20th percentile, 10th percentile, 1th percentile of a population, or other objective percentile among these exemplary objectives. As described above, the verification can also be used to adjust the dosage that achieves an objective level of CRP in serum, or to achieve an objective percentage reduction in CRP for a particular human subject. The verification may involve parameters in addition to CRP. A benefit of the statin for many human subjects will be the reduction in serum LDL, and the methods of the invention include administering the composition of the invention in an amount effective to reduce serum LDL and serum leukotrienes in the human subject. In this modality, serum LDL can be monitored. Other markers described herein, including amyloid serum A and myeloperoxidase, can be monitored. In certain embodiments of the invention, the individual or human subject selected for treatment is an individual who has at least one risk factor, such as a haplotype at risk of myocardial infarction, stroke or PAOD; in a haplotype at risk in the FLAP gene; a polymorphism in a FLAP nucleic acid; a polymorphism at risk in the 5-LO genetic promoter. The invention provides methods for selecting a human subject susceptible to MI comprising determining a FLAP genotype or haplotype of a human subject, and selecting to treat a human subject with a FLAP genotype or haplotype that correlates with an increased risk of MI. Methods of the invention include selecting a human subject with the presence of at least one haplotype at risk in or near the FLAP gene such as a haplotype shown in Table 14; a haplotype shown in Table 15; a haplotype shown in Table 19; haplotype B4; haplotype B5; haplotype B6; haplotype A4; haplotype A5; HapB haplotype, HapCl haplotype, HapC2 haplotype, HapC3 haplotype, HapC4-A haplotype and HapC4-B haplotype. The methods of the invention also include selecting a human subject for treatment, wherein the presence in such a subject of a haplotype comprising the marker SG13S106 (SNP DG00AAHII) (SEQ ID NO: 1, position 176579), allele G , identifies the subject as having a susceptibility to MI; the presence of a haplotype comprised of markers SG13S99 (DGOOAAFIU), allele T (SEQ.
FROM IDENT. NO .: 1, position 138551), SG13S377 (DGOOAAJFF) (SEQ ID NO: 1, position 169965), allele G; SG13S106 [SNP DGOOAAHII] (SEQ ID NO: 1, position 176579), allele G; SG13S32 (SEQ ID NO: 1, position 198547), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G identifies the subject as having a susceptibility to MI; the presence in such a subject of a haplotype comprised of markers: SG13S375 (SEQ ID NO: 1, position 164874), T allele; SG13S25 (SEQ ID NO: 1, position 165553), allele G; SG13S32 (SEQ ID NO: 1, position 176579), allele A; and SG13S106 (SEQ ID NO: 1, item 198547), allele G or A identifies the subject as having a susceptibility to MI, the presence of such a subject of a haplotype comprised of the marker SG13S375 (SNP DG00AAJFC) (SEQ. DE IDENTITY NO .: 1, position 164874), allele T and SG13S25 (SEQ ID NO: 1, position 165553), allele G identified the subject as having a susceptibility to MI the presence of such subject to a haplotype comprised of marker SG13S375 (SNP DG00AAJFC) (SEQ ID NO: 1, position 164874), allele T; and SG13S25 (SEQ ID NO: 1, position 165553), allele G and SG13S32 (SEQ ID NO: 1, item 198547) identified the subject as having a susceptibility to MI, the presence of such a subject of a haplotype comprised of marker SG13S106 (SNP DG00AAHII) (SEQ ID NO: 1, position 176579), allele G, SG13S30 (SEQ ID NO: 1, position 193840), allele G; and SG13S42 (SEQ ID NO: 1, position 203877), allele A, identifies the subject as having a susceptibility to MI, the presence of such a subject of a haplotype comprised of markers: SG13S377 (SEQ ID NO. 1, position 169965), allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G, identifies the subject as having a susceptibility to MI. In another embodiment, the invention provides a method for selecting a human subject susceptible to MI that comprises analyzing the nucleic acid of a human subject for the presence or absence of at least one FLAP polymorphism that correlates with an MI susceptibility. FLAP polymorphisms that correlate to MI susceptibility include SG13S377 (SEQ ID NO: 1, position 169965); allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), G allele. Additional FLAP polymorphisms that correlate to a susceptibility to MI include SG13S375 (SEQ ID NO: 1, position 164874), allele T, SG13S25 (SEQ ID NO: 1, position 165553), allele G; SG13S32 (SEQ ID NO: 1, position 176579), allele A; and SG13S106 (SEQ ID NO: 1, item 198547), allele G or A. The methods may further comprise selecting a subject in the presence of at least one FLAP polymorphism and in the presence of elevated CRP or MPO. In another embodiment, the invention provides methods for prophylaxis therapy for myocardial infarction (MI) which comprises analyzing nucleic acid from a human subject for the presence and absence of a FLAP haplotype, wherein the haplotype is comprised of markers: SG13S377 (SEC IDENTIFICATION NO .: 1, position 169965), allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G, and selection for the treatment of a human subject having nucleic acid in the presence of the FLAP haplotype. This method further comprises administering to the subject a composition comprising a therapeutically effective amount of an MI therapeutic agent that inhibits leukotriene synthesis in vivo, wherein the MI therapeutic agent inhibits leukotriene synthesis by inhibiting the activity of at least one selected protein. from the activation protein of 5-lipoxygenase (FLAP) y.5-lipoxygenase (5-LO). In a modality, the invention provides methods for decreasing the risk of a subsequent myocardial infarction in an individual who has had at least one myocardial infarction, which comprises administering a therapeutically effective amount of a therapeutic agent MI to the individual, wherein the therapeutic agent MI inhibits synthesis of leukotriene by inhibiting the activity of at least one protein selected from the protein that activates 5-lipoxygenase (FLAP) and 5-lipoxygenase (5-LO) and monitoring myeloperoxidase (MPO) in the individual before and during administration of the agent Therapeutic, wherein the therapeutic agent is administered in an amount effective to reduce the level of leukotriene in a subject. In another embodiment, the invention provides methods for selecting a human subject for susceptibility to MI that comprises analyzing nucleic acid from a human subject for the presence and absence of the FLAP haplotype comprising markers.; SG13S377 (SEQ ID NO: 1, position 169965), allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G, and identifying the subject that has a susceptibility to MI, wherein the presence of the FLAP haplotype correlates with an increased risk of MI. Individuals or human subjects selected for treatment may have at least one risk factor for a family or medical history such as diabetes; hypertension; hypercholesterolemia; elevated triglycerides; Ip (a). high; obesity; ankle / branchial index (ABI) less than 0.9; smoker in the past or currently; Transient ischemic attack; transient monocular blindness; endarterectomy of the carotid artery; asymptomatic carotid stenosis; claudication; limb ischaemia leading to gangrene, ulceration or amputation; a graft of vascular or peripheral artery revascularization; increased cholesterol in serum LDL and / or decreased HDL cholesterol; total serum cholesterol > 200 mg / dL, increased leukotriene synthesis; and / or at least one previous myocardial infarction, ACS, stable angina, previous transient ischemic attack, transient monocular blindness, or stroke, asymptomatic carotid stenosis or carotid endarterectomy, atherosclerosis, requires treatment for restoration -coronary artery blood flow (for example, angioplasty, stent, revascularization procedure). In addition, individuals or human subjects selected for treatment may have a high inflammatory marker, for example, a marker such as a reactive protein C (CRP) amyloid serum A, fibrinogen, a leukotriene, a metabolite of leukotriene, interleukin-6, tissue necrosis factor-alpha, a soluble vascular cell adhesion molecule (sVCAM), a soluble intervascular adhesion molecule (sICAM), E-selectin, type 1 matrix metalloprotease, type 2 matrix metalloprotease, type 3 matrix metalloprotease, type 9 matrix metalloprotease, myeloperoxidase (MPO), and N-tyrosine). The invention provides methods for prophylaxis therapy for MI which comprises administering a therapeutic agent MI in an amount effective to reduce the elevated serum level of at least one of the elevated inflammatory markers. In a particular embodiment, the invention provides methods for myocardial infarction (MI) prophylaxis comprising administering to a subject in need of prophylaxis for myocardial infarction a composition comprising a therapeutically effective amount of a MI therapeutic agent that inhibits leukotriene synthesis. in vivo, and verifying the level of myeloperoxidase (MPO) in the human subject before and during the prophylaxis treatment, wherein the therapeutic agent MI is administered in an effective amount to reduce the MPO level in a subject. The invention also provides methods for selecting a human subject at risk of developing myocardial infarction, comprising contacting a blood sample from the human subject with a calcium ionophore that stimulates the production of a leukotriene; and measuring the production of a leukotriene in the blood sample after the contact stage, where the production of leukotriene-high compared to a control correlates with the increased risk of developing myocardial infarction (MI). Control in these methods can be a human being of the same sex as the subject selected for treatment or a human being of equal age to the subject selected for treatment. Human subjects who are already treated with statins can enjoy the benefit of the present invention if the therapy of subjects that includes an LT antagonist is modified. Thus, in yet another embodiment, the invention is a method for reducing reactive protein C (CRP) in a human subject, comprising: selecting a human subject receiving statin therapy to reduce serum LDL, wherein the therapy with statin optionally reduces CRP in serum in the human subject; and administering to the human subject a leukotriene synthesis antagonist, in an amount effective to further reduce CRP in the human subject. In still another embodiment, the invention is a method for reducing reactive protein C (CRP) in a human subject, comprising: identifying a human subject in need of treatment to reduce CRP in serum; administer to the human subject a composition comprising a statin; and administering to the human subject a composition comprising a leukotriene synthesis inhibitor, wherein the statin and the leukotriene synthesis inhibitor are administered in effective amounts that reduce CRP in serum in the human subject. The statin and LT inhibitor can be administered simultaneously as a single composition, as described above; they can be administered simultaneously as separate compositions; or they can be administered sequentially. Depending on the dosing schedule, the daily administration regimen may include simultaneous administration at some times and separate administration at other times, for example if one agent is administered twice a day and the other three times a day. In certain embodiments of the invention, the individual or human subject selected for treatment is an individual who has at least one risk factor, such as a haplotype at risk of myocardial infarction, stroke or PAOD; a haplotype at risk in the FLAP gene; a polymorphism in a FLAP nucleic acid; a polymorphism at risk in the 5-LO genetic promoter; diabetes; hypertension hypercholesterolemia; elevated triglycerides; lp (a) 'elevated obesity; ankle / gill index (ABI) less than 0.9 smoker in the past or currently; transient attack of ischemia; transient monocular blindness; endarterectomy of the carotid artery; asymptomatic carotid stenosis; claudication; limb ischaemia leading to gangrene, ulceration or amputation; a graft of vascular or peripheral artery revascularization; an elevated inflammatory marker (e.g., a marker such as C reactive protein (CRP), amyloid serum A, fibrinogen, a leukotriene, a metabolite of leukotriene, interleukin-6, tissue necrosis factor-alpha, a vascular cell adhesion molecule soluble (sVCAM), a soluble intervascular adhesion molecule (sICAM), E-selectin, type 1 matrix metalloprotease, type 2 matrix metalloprotease, type 3 matrix metalloprotease, type 9 matrix metalloprotease, myeloperoxidase (MPO), and N -tyrosine); increased LDL cholesterol and / or decreased HDL cholesterol; increased leukotriene synthesis; and / or at least one previous myocardial infarction, ACS, stable angina, previous transient ischemic attack, transient monocular blindness, or stroke, asymptomatic carotid stenosis or carotid endarterectomy, atherosclerosis, requires treatment for restoration of coronary artery blood flow ( for example, angioplasty, stent, revascularization procedure). The invention also relates to methods for evaluating an individual for an increased risk of MI, ACS, atherosclerosis, stroke, or PAOD, by evaluating or monitoring a level of a leukotriene metabolite (e.g., LTE4, LTD4, LTB4) in the individual (for example, in a sample of blood, serum, plasma or urine). An individual or human subject selected for treatment may have a high measurement of a leukotriene or leukotriene metabolite, such as LTC4, LTD4, LTB4 and LTE4. The level of leukotrienes and leukotriene metabolites can be measured in serum, plasma, blood or urine in the individual. An increased level of leukotriene metabolite is indicative of an increased risk. The invention also encompasses methods for evaluating an individual for an increased risk of MI, ACS, atherosclerosis, stroke, transient ischemic attack, transient monocular blindness, asymptomatic carotid stenosis, PAOD, claudication, or limb ischemia by stimulating the production of a leukotriene or a metabolite of leukotriene in a test sample from the individual (eg, a sample comprising neutrophils), using a calcium ionophore, and comparing the level of the leukotriene or leukotriene metabolite with a control level. A level of leukotriene or leukotriene metabolite production that is significantly higher than the control level is indicative of increased risk. The invention also relates to methods for evaluating the response to treatment with a leueotriene synthesis inhibitor., evaluating or monitoring a level of a leukotriene or a leukotriene metabolite in the individual prior to treatment, and comparing the level to a level of the leukotriene or leukotriene metabolite evaluated during or after treatment. A level that is significantly lower during or after treatment, than before treatment, is indicative of efficacy of the leukotriene synthesis inhibitor treatment. The level of leukotriene can be monitored in serum, plasma, blood or urine collected from the subject before, during or after treatment. The invention further relates to methods for evaluating the response to treatment with a leukotriene synthesis inhibitor, by stimulating the production of a leukotriene or a leukotriene metabolite in a first test sample from the individual (eg, a sample comprising neutrophils) before treatment, using a calcium ionophore, and comparing the level of leukotriene or leukotriene metabolite with a level of production of leukotriene or leukotriene in a second sample test from the individual, during or after treatment. A production level of the leukotriene or leukotriene metabolite in the second test sample that is significantly lower than the level in the first test sample is indicative of efficacy of the treatment, for example, the treatment or therapeutic agent reduces the level of leukotriene in the subject at the average level of leukotrienes in human subjects in the general population or lower than the average level. Similarly, the invention encompasses methods for evaluating response to treatment with a leukotriene synthesis inhibitor, evaluating or monitoring a level of an inflammatory marker in the individual prior to treatment, and during or after treatment. A level of inflammatory marker during or after treatment, which is significantly lower than the level of inflammatory marker before treatment, is indicative of treatment efficacy. To determine the effectiveness of the compositions of the present invention comprising a statin, total cholesterol, LDL-C and / or triglycerides can be evaluated from measurements of risk factor markers in the serum of a human subject administered to the composition. A level of total cholesterol in serum, LDL-C and / or triglycerides during or after treatment, which is significantly lower than the level of total cholesterol, LDL-C and / or triglycerides before treatment is indicative of the efficacy of the treatment . The invention also relates to the use of leukotriene synthesis inhibitors for the manufacture of a medicament for the treatment of MI, ACS, apoplexy, PAOD, and / or atherosclerosis, as described herein, as well as for the manufacture of a medication for the reduction of leukotriene synthesis.
BRIEF DESCRIPTION OF THE DRAWINGS The foregoing and other objects, features and advantages of the invention will be apparent from the following more particular description of the preferred embodiments of the invention. FIGURE 1 shows the results from a haplotype case-control analysis of 437 patients with MI women versus 721 controls using combinations of microsatellite markers 4 and 5 to define the test haplotypes. The p-value of the association is plotted on the y-axis, and the position of the markers on the x-axis. Only haplotypes showing association with a p < 10-5 are shown in the figure. The most significant haplotype association of the microsatellite marker is found using markers DG13S1103, DG13S166, DG13S1287, DG13S1061 and DG13S301, with alleles 4, 0, 2, 14 and 3, respectively (p-value of 1.02x 10"7). The carrier of the haplotype is 7.3% in patients with MI women and 0.3% in controls.The segment that is common to all the haplotypes shown in the figure includes only one gene, FLAP.FIGURE 2 shows the alleles of the markers that define the haplotypes Most significant microsatellite markers The segment defined with a black square is common to all the most significantly associated haplotypes The FLAP nucleic acid is located between markers DG13S166 and D13S1238.The haplotype of two markers involving the 0 and -2 alleles for markers DG13S166 and D13S1238, respectively, are found in excess in patients.The carrier frequency of this haplotype is 27% in patients and 15.4% in controls (p value 1 x 10"3). Therefore, the association analysis confirms that the gene associated with narrower MI within the linkage peak is FLAP. FIGURE 3 shows the relative location of the Key SNP and exons of the AL0X5AP / FLAP gene (exons shown in vertical rectangles). The haplotype length varies between 33 and 68 kb. FIGURE 4 shows a significant positive correlation between serum LTE4 levels and serum CRP levels. FIGURE 5 describes the production of LTB4 of neutrophils stimulated by ionomycin from MI patients (n = 41) and controls (n = 35). The transformed logarithm values (average + SD) measured in 15 and 30 minutes of stimulated cells are shown. (7.1) The production of LTB4 in MI patients and controls. The difference in mean values between patients and controls is tested using a t-test of two samples of the transformed logarithm values. (7.2) The production of LTB4 in MI male carriers and non-carriers of A4 haplotype. The average values of controls are included for comparison. It is important that men with the A4 haplotype produce higher amounts of LTB4 (p <0.005 compared to controls). (7.3). The schematic representation of the 5-LO pathway with bioactive leukotriene products. FIGURE 6 shows a schematic view of the linkage region of chromosome 13 showing the gene FLAP. (9.1) Linkage scan for patients with MI women and the LOD fall region that includes the FLAP gene; (9.2) The microsatellite association for all MI patients; the association of a single marker and the haplotype association of two, three, four and five markers. The arrows indicate the location of the most significant haplotype association through the FLAP gene in men and women. (9.3) The genetic structure FLAP, with exons shown as cylinders, and the location of all the SNPs classified in the region (vertical lines). The vertical lines indicate the position of the microsatellites (shown in 9.2) and the SNPs (shown in 9.3) used in the analysis. FIGURE 7 shows a linkage scan using structure microsatellite markers on chromosome 13 for male patients with ischemic stroke or TIA (n = 342 in 164 families at 6 miosis). The degree of LOD is expressed on the y axis, and the distance from the ether on Kosambi cM on the x axis. FIGURE 8 shows a linkage disequilibrium in pairs (LD) between the SNPs in a 60 kb region encompassing FLAP. The markers are drawn equidistantly. Two LD measurements are shown; D 'in the upper left triangle and the P values in the lower right triangle. The shaded lines indicate the positions of the FLAP exons and the stars indicate the location of the A4 haplotype markers at risk. The scales for the LD resistance are provided for both measurements on the right. FIGURE 9 provides a diagram of the clinical trial schedule. This figure shows that in Visit 2 (on day 1 of the study) the subjects were chosen at random within each of the three branches and to any placebo or active drug within each branch. A washout period of 2 weeks separated the treatment periods of 4 weeks. The transition was made in week 6. FIGURE 10 shows the analysis of the transfer effect for CRP and SAA (on a logarithm scale).
DETAILED DESCRIPTION OF THE INVENTION Extensive genealogical information has been combined with powerful genetic participation methods to map a gene on chromosome 13ql2-13 that is associated with myocardial infarction. A broad genome investigation for gene susceptibility to MI, which uses a structure map of 1000 microsatellite markers, revealed a suggestive ligament site at 13ql2-13. Sixty families with 159 patients with MI women who were grouped within and including 6 meiotic episodes were used in linkage analysis. First, only patients with MI women were used in the linkage analysis in an effort to enrich patients with stronger genetic factors contributing to their risk for MI. The epidemiological study of a population-based sample of patients with Icelandic MI has previously suggested that genetic factors for MI may be stronger for women than for men, since the relative risk for siblings of MI women is significantly higher than the relative risk for male probands (1.59 (Cl 1.47-1.73) vs. 1.35 (Cl 1.28-1.42)) (unpublished data). The highest LOD score (2.5) was found in marker D13S289. The resulting LOD score for families remained the same after adding 14 microsatellite markers to the candidate region. The inclusion of additional markers increased the information on the share by descent from 0.7 to 0.8, around the markers that gave the highest LOD scores. This linkage analysis mapped a gene that contributes MI to chromosome 13ql2-13. The MY candidate site on chromosome 13ql2-13 was then finally mapped with microsatellite markers. Patients with myocardial infarction and controls were initially genotyped with microsatellite markers with an average space between markers of less than 100 kb over the 12 Mb candidate region. The initial haplotype association analysis that included all classified microsatellite markers in genotype through candidate site MI, resulted in several extended haplotypes composed of 4 and 5 microsatellite markers that were significantly associated with MI women (see, for example, Tables 14 and 15 below). A region common to all these extended haplotypes is defined by markers DG13S166 and D13S1238. This region includes only one gene, the FLAP nucleic acid sequence. The haplotype of two markers involving 0 and -2 alleles for the markers DG13S166 and D13S1238, respectively, was found in excess in patients. Specific variants of the gene were then discovered that were associated with MI. In order to classify the SNPs in the FLAP gene, the complete gene was sequenced, both in exons and in introns. Initially, 9 of the SNPs, identified within the gene, were classified as genotypes in patients and controls. Additional microsatellite markers near or within the FLAP gene were also genotyped in all patients and controls. Five of the publicly known SNPs that are located within a 5 'distance of 200 kb to the FLAP gene were genotyped also in patients and control. The haplotype association analysis in this case-control study that includes these additional markers showed several different variants of the same haplotype that were significantly associated with MI women (see for example, Table 8). Table 9 shows two haplotypes that are representative of these MI risk haplotypes which are referred to herein as "at risk" haplotypes of MI women. The relative risk for patients with MI women who had the haplotype "at risk" of MI women increased (see for example, Table 9), which indicates that the "at risk" haplotype of MI women also increased the risk of having an MI in men. These results further strengthened the hypothesis that FLAP was a gene with MI susceptibility.Association of SNP Haplotype to MI, and Subsequently to Stroke and PAOD In an effort to identify haplotypes involving only SNP markers that are associated with MI, additional SNPs were identified by sequencing the FLAP gene and the region flanking the gene. Currently, a total of 45 SNPs in 1343 patients and 624 unrelated controls have been classified by genotype. Two correlated SNP haplotype series have been observed in excess in patients, denoted as A and B in Table 7. The length of the haplotypes varies between 33 and 69 kb, and the haplotypes cover one or two blocks of linkage disequilibrium. Both series of haplotypes (HapA and HapB) contain the common G allele of SN13S25 of the SNPs. HapC2, identified in the analysis of the North American group (see Example 13), also contains the G allele of SNP SG13S25. All haplotypes in series A contain SNP SG13S114, while all haplotypes in series B contain SNP SG13S106. In the B series, the B4, B5 and B6 haplotypes have a relative risk (RR) greater than 2 and with allelic frequencies over 10%. Haplotypes in series A have significantly lower RR and lower p-value, but higher frequency (15-16%). The haplotypes in the B and A series are strongly correlated, that is, the haplotypes in B define a subset of haplotypes in A. Therefore, the haplotypes in the B series are more specific than in A. However, the Haplotypes in the A series are more sensitive, that is, they capture more individuals with the putative mutation, as observed in the risk attributable to the population which is lower for B than for A. In addition, these haplotypes show similar risk relationships and allele frequencies for early-onset patients (defined as the beginning of the first MI before 55 years) and for both genders. In addition, when analyzing several groups of patients with known risk factors, such as hypertension, high cholesterol, smoking and diabetes, no significant correlation was found with these haplotypes, suggesting that the haplotypes in the FLAP gene represent an independent genetic susceptibility factor. for me . The analysis of the North American group (described in Example 12) identified another haplotype C which is associated with MI as demonstrated in Table 33 (Example 13). HapC is defined by the T allele of marker SG13S375. ' There are 4 additional variations of the HapC haplotype which comprises the SNP in addition to the T allele of SG13S375. HapC2 is defined by the T allele of SG13S375 of the SNP • and the G allele of SNP SG13S25. HapC3 is defined by the T allele of SG13S375 of the SNPs and the G allele of SNP SG13S25 and the A allele of SG13S32 of the SNP. HapC4-A is defined by allele A of SNP SG13S106 in addition to the -del allele of SN13S375 of SNPs, allele G of SG13S25 of SNP and allele A of SG13S32 of SNP. HapC4-B is defined by the G allele of the SN13 SG13S106 in addition to the T allele of the SN13S375 of the SNPs, the G allele of the SNP SG13S25 and the A allele of SG13S32 of the SNP. HapC4-A and HapC-4B correlates with HapA and HapB. Because stroke and PAOD are diseases that are closely related to MI (everything occurs at the base of atherosclerosis), the SNP haplotype in the FLAP gene that confers risk to MI was evaluated to determine whether it also confers risk of stroke and / or PAOD. Table 20 shows that the A4 haplotype increases the risk of having a stroke to a similar degree as when the risk of having an MI increases. Table 34 demonstrates that HapA is associated with the risk of stroke in a Scottish group (Example 14). Although not as significantly, the 4A haplotype also confers the risk of developing PAOD. The FLAP nucleic acid encodes a protein that activates 5-lipoxygenase which, in combination with 5-lipoxygenase (5-LO), is required for leukotriene synthesis. FLAP acts in coordination with 5-LO to catalyze the first stage in the synthesis of leukotrienes from arachidonic acid. This catalyzes the conversion of ar'aquidónico acid to the acid 5 (S) -hidroxiperoxi-6-trans-8, 11, 14-cis-eicosatetranoic (5-HPETE), and in addition to the epoxide acid 5 (S) -trans7,9 trans 11, 14-cis-eicosatetranoic allylic (leukotriene A4, LTA4). Leukotrienes are a family of highly potent biological mediators of inflammatory processes produced mainly by leukocytes derived from spinal cord such as monocytes, macrophages and neutrophils. Both FLAP and 5-LO are detected within atherosclerotic lesions (Proc Nati Acad Sci U S A. 2003 Feb 4; 100 (3): 1238-43. ), indicating that the vessel itself can be a source of leukotrienes. It was found at the beginning that the FLAP haplotype of risk to MI was associated with higher levels of leukotriene. The increased production of leukotriene in individuals with pre-existing atherosclerosis can lead to instability or friability of the fibrous cap plate leading to local thrombotic events. If this occurs in arteries of the coronary artery, this leads to MI or unstable angina. If this occurs in the cerebrovasculature, this leads to apoplexy or transient ischemic attack. If this occurs in large arteries to the limbs, this causes or exacerbates limb ischemia in people with peripheral arterial occlusive disease (PAOD). Therefore, those with a genetically influenced predisposition to produce higher leukotriene levels have a higher risk for episodes due to pre-existing atherosclerosis such as MI. Inhibitors of FLAP function prevent the displacement of 5-LO from the cytoplasm to the cell membrane and inhibit the activation of 5-LO and therefore decrease the synthesis of leukotriene. As a result of these discoveries, the methods are now available, for the treatment of myocardial infarction (MI) and acute coronary syndrome (ACS), as well as stroke. and PAOD, through the use of leukotriene inhibitors, such as agents that inhibit leukotriene biosynthesis or antagonize signaling through leukotriene receptors. The term "treatment" as used herein, refers not only to improving the symptoms associated with the disease or condition, but also to preventing or delaying the onset of the disease or condition; avoiding or diminishing the appearance of a second episode of the disease or condition; and / or also reducing the severity or frequency of symptoms of the disease or condition. In the case of atherosclerosis, "treatment" also refers to a minimization or reversal of plaque development. The methods are also available to assess an individual's risk for MI, ACS, stroke or PAOD. In a preferred embodiment, the individual being treated is an individual who is susceptible (at increased risk) to MI, ACS, stroke or PAOD, such as an individual who is in one of the representative target populations described herein.
Target Representative Populations In one embodiment of the invention, an individual who is at risk for MI, ACS, stroke or PAOD is an individual who has a haplotype at risk in FLAP, as described herein. In one embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, stroke or PAOD comprises markers SG13S99, SG13S25, SG13S377, SG13S106, SG13S32 and SG13S35 at site 13ql2-13. In another embodiment, the haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S99, SG13S25, SG13S106, SG13S30 and SG13S42 at site 13ql2. In a third modality, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S106, SG13S30 and SG13S42 at site 13ql2-13. In a fourth embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S99, SG13S25, SG13S114, SG13S89 and SG13S32 at site 13ql2-13. In a fifth modality, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S114, SG13S89 and SG13S32 at site 13ql2-13. In another modality, a haplotype is associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises the marker SG13S375 at the 13ql2-13 site. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25 and SG13S375 at the 13ql2-13 site. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S375, and SG13S32 at the 13ql2-13 site. In a further embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S375, SG13S32 and SG13S106 at the 13ql2-13 site. Additional haplotypes associated with a susceptibility to myocardial infarction, ACS, stroke or PAOD include the haplotypes shown in Tables 4, 8, 9, 14, 15, 17 and 19, as well as haplotypes comprising markers shown in Table 13. The increased risk for MI, ACS, stroke or PAOD in individuals with a haplotype at risk for FLAP is logically conferred by increased production of leukotrienes in the arterial vascular wall or in inflammatory cells derived from the spinal cord within blood and / or the arterial vascular wall. It was shown in the present that the haplotypes at risk of FLAP are associated with the highest production of LTB4 ex vivo. It was further shown herein that serum leukotriene levels (specifically, leukotriene E4) correlate with serum CRP levels in patients with myocardial infarction. The genetic variation FLAP can lead to high levels of leukotriene (within the blood vessel and / or systematically), which in turn can lead to higher CRP levels which have been shown to be a risk factor for MI. Therefore, individuals with a haplotype at risk for FLAP are prone to have elevated CRP in serum as well as other inflammatory markers in serum. The level of CRP in serum or other inflammatory markers in serum can be used as a substitute for the level of arterial wall inflammation initiated by lipid deposition and atherogenesis conferred by the presence of the FLAP haplotype at risk. In another embodiment of the invention, an individual who is at risk of MI, ACS, stroke or PAOD is an individual who has a polymorphism in a FLAP gene, where the presence of the polymorphism is indicative of a susceptibility to MI, ACS, stroke. or PAOD. The term "gene" as used herein, refers not only to the sequence of nucleic acids encoding a polypeptide, but also to promoter regions, elements that enhance transcription, splice donors / acceptor sites, and other non-transcribed nucleic acid elements. Representative polymorphisms include those presented in Table 13, below. In a further embodiment of the invention, an individual who is at risk for MI, ACS, stroke or PAOD is an individual who has a polymorphism at risk in the 5-LO gene in the promoter region, as described herein. In a fourth modality, an individual who is at risk for MI, ACS, stroke or PAOD is an individual who has an elevated inflammatory marker. An "elevated inflammatory marker" as used herein, is the presence of an amount of an inflammatory marker that is greater, by an amount that is statistically significant, than the amount that is typically found in control individuals or in comparison to the risk of disease in a population associated with the lowest measuring band (for example, below average or average, the lowest quartile or the lowest quintile) compared to higher measurement bands (for example, above the average or term) medium, the second, third or fourth quartile; the second, third, fourth or fifth quintile). An "inflammatory marker" refers to a molecule that is indicative of the presence of inflammation in an individual, e.g., C-reactive protein (CRP), amyloid serum A, fibrinogen, leukotriene levels (e.g., leukotriene B4, leukotriene C4). ), leukotriene metabolites (eg, leukotriene E4), interleukin-6, tissue necrosis factor-alpha, soluble vascular cell adhesion molecules (sVCAM), soluble intervascular adhesion molecules (sICAM), E-selectin, matrix metalloprotease type 1, matrix metalloprotease type 2, matrix metalloprotease type 3, matrix metalloprotease type 9, myeloperoxidase (MPO), N-tyrosine) or other markers (see for example, Doggen, et al., J. Intemal Med., 248: 406-414 (2000); Ridker, PM et al., New England, J. Med. 1997: 335: 973-979, Rettersol, L et al., 2002: 16O.-433-440; Ridker, PM, et al., New England, J. Med., 2002: 347: 1557-1565, Bermudez, EA et al., Arterioscler, Thromb. Biol., 2002: 22: 1668-1673). In certain embodiments, the presence of such inflammatory markers can be measured in serum or urine. In a fifth modality, an individual who is at risk for MI, ACS, stroke or PAOD is an individual who has increased LDL cholesterol levels and / or decreased HDL cholesterol. For example, the American Heart Association states that an LDL cholesterol level of less than 100 mg / dL is optimal; from 100-129 mg / dL is near / above the optimum; from 130-159 mg / dL is at the elevated edge; from 160-189 it is elevated; and from 190 upwards it is very high. Therefore, an individual who is at risk of MI, ACS, stroke or PAOD due to an increased LDL cholesterol level, is for example, an individual who has more than 100 mg / dL of cholesterol, such as an individual who it has an almost / above optimal level, a high edge level, a high level or a very high level. Similarly, the American Heart Association states that an HDL cholesterol level of less than 40 mg / dL 'is a major risk factor for heart disease; and an HDL cholesterol level of 60 mg / dL or more is protective against heart disease. Thus, an individual who is at risk of MI, ACS, stroke or PAOD due to a decreased HDL cholesterol level, is for example, an individual who has less than 60 mg / dL of HDL cabbages, such as an individual who has less than 40 mg / dL of HDL cholesterol.
In a sixth modality, an individual who is at risk of MI, ACS, stroke or PAOD is an individual who has increased leukotriene synthesis. The "increased leukotriene synthesis" as used herein, indicates a quantity of leukotriene production that is greater, by an amount that is statistically significant, than the amount of leukotriene production that is normally found in control or comparison individuals. to the production of leukotriene in a population associated with the lowest measurement band (for example, below the mean or average, the lowest quartile or the lowest quintile) compared to the highest measurement bands (for example, over the average or average term, the second, third or fourth quartile, the second, third, fourth or fifth quintile). For example, haplotypes at risk of FLAP correlate with increased serum leukotriene synthesis levels, and with increased ex vivo production of leukotrienes. An individual can be evaluated for the presence of increased leukotriene synthesis by a variety of methods. For example, an individual may be evaluated for an increased risk of MI, ACS, stroke, PAOD or atherosclerosis by evaluating the level of leukotriene metabolite (e.g., LTE4) in a sample (e.g., serum, plasma or urine) to from the individual. Samples containing blood, cells or tissue can also be obtained from an individual and used to evaluate the production of leukotriene or leukotriene metabolite ex vivo under appropriate assay conditions. An increased level of leukotriene metabolites and / or an increased level of leukotriene production ex vivo is indicative of the increased production of leukotrienes in the individual, and of an increased risk of MI, ACS, stroke, PAOD or atherosclerosis. In a further embodiment, an individual who is at risk for MI, ACS or stroke is an individual who has already experienced at least one MI, episode of ACS or stroke, or who has stable angina, and is therefore at risk of a second MI, episode of ACS or stroke. In another modality, an individual who is at risk for MI, ACS, stroke or PAOD is an individual who has atherosclerosis or who requires treatment (eg, angioplasty, stents, revascularization procedure) to restore blood flow in arteries. In additional modalities, an individual who is at risk of MI, stroke or PAOD is an individual who has an asymptomatic ankle / branchial index of less than 0.9.; an individual who is at risk of stroke is an individual who has had one or more transient attacks; who has had transient monocular blindness; has had a carotid endarterectomy; or has asymptomatic carotid stenosis; an individual who is at risk of PAOD, is an individual who has (or had) claudication, limb ischemia leading to gangrene, ulceration or amputation, or has had a revascularization procedure. In additional modalities, an individual who is at risk for MI, ACS, stroke or PAOD is an individual who has diabetes; hypertension; hypercholesterolemia; high triglycerides (e.g.,> 200 mg / dL); lp (a) elevated; obesity; ankle / branchial index (ABI) less than 0.9; and / or smoker in the past or currently. Individuals at risk for MI, ACS, stroke or PAOD may be within one of these representative target populations. For example, an individual may have experienced at least one MI, episode of ACS, transient ischemic attack, transient monocular blindness or stroke, and may also have an increased level of an inflammatory marker. As used herein, the term "individual in a target population" refers to an individual who is at risk of MI, ACS, stroke or PAOD, who falls into at least one of the representative target populations described above.
Evaluation for Haplotypes at risk A "haplotype" as described herein, refers to a combination of genetic markers ("alleles") such as those set forth in Table 13. In a certain embodiment, the haplotype may comprise one or more alleles (for example, a haplotype that contains a single SNP), two or more alleles, three or more alleles, four or more alleles, or five or more alleles. Genetic markers are particular "alleles" in "polymorphic sites" associated with FLAP. A nucleotide position in which more than one sequence is possible in a population (either a natural population or a synthetic population, eg, a library of synthetic molecules), is referred to herein as a "polymorphic site". Where a polymorphic site is a single nucleotide in length, the site is referred to as a single nucleotide polymorphism ("SNP"). For example, if at a particular chromosomal location, a member of a population has an adenine and another member of the population has a thymine at the same position, then this position is a polymorphic site, and more specifically, the polymorphic site is a. SNP. Polymorphic sites may allow differences in sequences based on substitutions, insertions or deletions. Each version of the sequence with respect to the polymorphic site is referred to herein as an "allele" of the polymorphic site. Thus, in the previous example, the SNP allows both an adenine allele and a thymine allele. Normally, a reference sequence refers to a particular sequence. Alleles that differ from the reference are referred to as "variant" alleles. For example, the reference FLAP sequence is described herein by the SEC. FROM IDENT. NO .: 1. The term "variant FLAP" as 5 is used herein, refers to a sequence that differs from the SEC. FROM IDENT. DO NOT. : 1, but is otherwise substantially similar. The genetic markers that constitute the haplotypes described herein are FLAP variants. Additional variants may include changes that affect a polypeptide, for example, the FLAP polypeptide. These sequence differences, when compared to a reference nucleotide sequence, can include the insertion or deletion of a single nucleotide, or more 15 of a nucleotide, resulting in a frame change; the change of at least one nucleotide, resulting in a change in the encoded amino acid; the change of at least one nucleotide, resulting in the generation of a premature stop codon; the elimination of several nucleotides, resulting 20 in a deletion of one or more amino acids encoded by the nucleotides; the insertion of one or several nucleotides, such as by unequal recombination or genetic conversion, resulting in an interruption of the coding sequence of a reading frame; the duplication of all or part of a 25. sequence; the transposition; or a re-arrangement of a nucleotide sequence, as described in detail above. Such sequence changes alter the polypeptide encoded by a FLAP nucleic acid. For example, if the change in the nucleic acid sequence causes a frame change, the frame change may result in a change in the encoded amino acids, and / or may result in the generation of a premature stop codon, causing generation of a truncated polypeptide. Alternatively, a polymorphism associated with a susceptibility to MI, ACS, stroke or PAOD may be a synonym change in one or more nucleotides (i.e., a change that does not result in a change in the amino acid sequence). Such a polymorphism may, for example, alter splice sites, affect the stability or transport of mRNA, or otherwise affect the transcription or translation of the polypeptide. The polypeptide encoded by the reference nucleotide sequence is the "reference" polypeptide with a particular reference amino acid sequence, and polypeptides encoded by variant alleles are referred to as "variant" polypeptides with variant amino acid sequences. Haplotypes are a combination of genetic markers, for example, particular alleles at polymorphic sites. The haplotypes described herein, for example, which have markers such as those shown in Table 13, are found more frequently in individuals with MI, ACS, stroke or PAOD than in individuals without MI, ACS, stroke or PAOD. Therefore, these haplotypes have predictive value to detect a susceptibility to MI, ACS, stroke or PAOD in an individual. The haplotypes described herein are in some cases a combination of several genetic markers, for example, SNPs and microsatellites. Therefore, haplotype detection can be achieved by methods known in the art for detecting sequences at polymorphic sites, such as the methods described above. In certain methods described herein, an individual who is at risk for MI, ACS, stroke or PAOD is an individual in whom a haplotype at risk is identified. In a modality, the haplotype at risk is one that confers a significant risk of MI, ACS, apoplexy or PAOD. In one embodiment, the importance associated with a haplotype is measured by a probability ratio. In an additional modality, importance is measured by a percentage. In one embodiment, a significant risk is measured as a probability ratio of at least about 1.2, including, but not limited to, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, and 1.9. In an additional mode, a probability ratio of at least 1.2 is significant. In a further embodiment, a probability ratio of at least about 1.5 is significant. In a further mode, a significant increase in risk of at least about 1.7 is significant. In a further embodiment, a significant increase in risk of at least 20%, including, but not limited to, approximately 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% and 98%. In a further embodiment, a significant increase in risk is at least about 50%. In yet another modality, a haplotype at risk has a p <value; 0.05. It is understood, however, that identifying that any risk is medically significant may also depend on a variety of factors, including the specific disease, the haplotype, and often environmental factors. A haplotype at risk in, or comprising portions of the FLAP gene, in one where the haplotype occurs most frequently in an individual at risk of MI, ACS, stroke or PAOD (affected), compared to the frequency of its presence in an individual healthy (control), and where the presence of the haplotype is indicative of susceptibility to MI, ACS, apoplexy or PAOD. As an example of a simple test for correlation, it would be an exact Fisher test on a two-by-two table. Given a group of chromosomes, the two-by-two table is constructed from the number of chromosomes that include both haplotypes, one from the haplotype, but not the other and none from the haplotypes. In certain embodiments, a haplotype at risk is a haplotype at risk in or near FLAP that correlates significantly with a haplotype such as a haplotype shown in Table 14; a haplotype shown in Table 15; a haplotype shown in Table 19; haplotype B4; haplotype B5; haplotype B6; haplotype A4; haplotype A5; or HapB haplotype. In other modalities, a haplotype at risk comprises a haplotype at risk in or near FLAP that significantly correlates with susceptibility to myocardial infarction or stroke. In a particular embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S99, SG13S25, SG13S377, SG13S106, SG13S32 and SG13S35 at site 13ql2-13. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S99, SG13S25, SG13S106, SG13S30 and SG13S42 at site 13ql2-13. In a third modality, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S106, SG13S30 and SG13S42 at site 13ql2-13. In a fourth embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S99, SG13S25, SG13S114, SG13S89 and SG13S32 at site 13ql2-13. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises the marker SG13S375 at the 13ql2-13 site. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25 and SG13S375 at the 13ql2-13 site. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S375 and SG13S32 at the 13ql2-13 site. In a further embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S375, SG13S32 and SG13S106. In other modalities, the haplotype at risk is selected from the group consisting of: haplotype B4, B5, B6, A4, A5, Cl, C2, C3, C4-A and C4-B. The haplotype at risk may also comprise a combination of markers in the B4, B5, B6, A4, A5, Cl, C2, C3, C4-A and / or C4-B haplotypes. In the additional modalities, the haplotype at risk may be a HapB haplotype. In other embodiments, the at-risk haplotype comprises a polymorphism shown in Table 13. Standard techniques can be used to genotype the presence of SNPs and / or microsatellite markers, such as fluorescent-based techniques (Chen, et al., Genome. Res. 9, 492 (1999)), PCR, LCR; Nested PCR and other techniques for nucleic acid amplification. In a preferred embodiment, the method comprises evaluating in an individual the presence or frequency of the SNPs and / or microsatellites in, comprising portions of the FLAP gene, wherein an excess or higher frequency of the SNPs and / or microsatellites compared to a healthy control individual is indicative that the individual is susceptible to MI, ACS, apoplexy or PAOD. See for example Table 13 (below) for SNPs and markers that can form haplotypes that can be used to select tools. These markers and SNPs can be identified in haplotypes at risk. For example, in a haplotype at risk, microsatellite markers and / or SNPs such as those set forth in Table 13 may be included. The presence of the haplotype is indicative of a susceptibility to MI, ACS, stroke or PAOD, and therefore is indicative of an individual who is within a target population for the treatment methods described herein. Haplotype analysis involves defining a candidate susceptibility site that uses LOD scores. The defined regions are then ultra-fine mapped with microsatellite markers with an average spacing between markers of less than 100 kb. All usable microsatellite markers found in public databases and mapped within this region can be used. In addition, microsatellite markers identified within the sequence assembly of the COCO genetics of the human genome can be used. The frequencies of the haplotypes in the patient and the control groups can be calculated using an expectation-maximization algorithm (Dempster A. et al., 1977, J. R. Stat. Soc. B, 39: 1-389). An implementation of this algorithm that can handle missing genotypes and uncertainty with the phase can be used. Under the null hypothesis, patients and controls are assumed to have identical frequencies. When using a probability method, an alternative hypothesis is tested, in which a haplotype at candidate risk, which may include the markers described herein, is allowed to have a higher frequency in patients than controls, while the frequency relationships of other haplotypes are assumed to be the same in both groups. The probabilities are maximized separately under both hypotheses and a corresponding 1-df probability ratio statistic is used to evaluate the statistical significance. To search for haplotypes at risk in the 1-lod fall, for example, the association of all possible combinations of markers classified in genotype is studied, provided that those markers encompass a practical region. The combined patient and the control groups can be randomly divided into two sets, equal in size to the original group of patients and controls. The haplotype analysis is then repeated and the most significant p-value recorded is determined. This random scheme can be repeated, for example, for 100 times to construct an empirical distribution of p-values. In a preferred embodiment, a p-value of <0.05 is indicative of a haplotype at risk. A detailed discussion of the haplotype analysis is described in International Application No. PCT / US03 / 32556, filed on October 16, 2003, which is incorporated herein by reference in its entirety.
Methods of Treatment The present invention encompasses treatment methods (prophylactic and / or therapeutic, as described above) for MI, ACS, stroke or PAOD in individuals, such as individuals in the target populations described above, as well as for other diseases and conditions associated with FLAP or with other members of the leukotriene pathway (eg, for atherosclerosis). Members of the "leukotriene pathway" as used herein, include polypeptides (e.g., enzymes, receptors) and other molecules that are associated with the production of leukotrienes: e.g., proteins or enzymes such as FLAP, 5- LO, other leukotriene biosynthetic enzymes (for example, leukotriene C4 synthase, leukotriene A4 hydrolase); receptors or binding agents of the enzymes; leukotrienes such as LTA4, LTB4, LTC4, LTD4, LTE4; and leukotriene receptors (e.g., leukotriene B4 receptor 1 (BLT1), leukotriene B4 receptor 2 (BLT2), cysteinyl leukotriene receptor 1 (CysLTRl), cysteinyl leukotriene receptor 2 (CysLTR2)). In particular, the invention relates to methods of treatment for myocardial infarction or susceptibility to myocardial infarction (eg, for individuals in a population at risk such as that described above); as well as treatment methods for acute coronary syndrome (for example, unstable angina, myocardial infarction without ST elevation (NSTEMI) or myocardial infarction with ST elevation (STEMI)); methods to reduce the risk of MI, stroke or PAOD in people with asymptomatic ankle / brachial index less than 0. 9; to decrease the risk of a second myocardial infarction; for apoplexy or susceptibility to stroke; for transient ischemic attack; for transient monocular blindness; to decrease the risk of a second stroke; for PAOD or susceptibility to PAOD; for ABI less than 0.9; for claudication or member ischemia; for atherosclerosis, such as for patients requiring treatment (eg, angioplasty, stents, vascularization procedure) to restore blood flow in arteries (eg, coronary, carotid and / or femoral arteries); for asymptomatic ankle / brachial index treatment of less than 0.9; and / or to decrease leukotriene synthesis (for example, for the treatment of MI, ACS, stroke or PAOD). The invention further relates to the use of one or more inhibitors of leukotriene synthesis, as described herein, for the manufacture of a medicament for the treatment of MI, ACS, stroke, PAOD and / or atherosclerosis, for example, using the methods described herein. The invention also provides the use of one or more inhibitors of leukotriene synthesis, as described herein, for the manufacture of a medicament for reducing the risk of MI, ACS, PAOD, stroke and / or atherosclerosis using the methods described in the present . These medicaments may comprise a leukotriene synthesis inhibitor alone or in combination with a statin, as described herein. In the methods of the invention, a "leukotriene synthesis inhibitor" is used. In one embodiment, a "leukotriene synthesis inhibitor" is an agent that inhibits FLAP polypeptide activity and / or expression of the FLAP nucleic acid, as described herein (e.g., a nucleic acid antagonist). In another embodiment, a leukotriene synthesis inhibitor is an agent that inhibits polypeptide activity and / or nucleic acid expression of another member of the leukotriene biosynthetic pathway (e.g., 5-LO, LTC4S, LTA4H, LTB4DH) . In yet another embodiment, a leukotriene synthesis inhibitor is an agent that alters the activity or metabolism of a leukotriene (eg, an antagonist of a leukotriene, an antagonist of a leukotriene receptor). In the preferred modalities, the inhibitor of leukotriene synthesis alters the activity and / or the expression of the nucleic acid of FLAP or of 5-LO, or alters the interaction between FLAP and 5-LO. Leukotriene synthesis inhibitors can alter the activity of the polypeptide or the expression of the nucleic acid of a member of the leukotriene pathway by a variety of means, such as, for example, by degrading, deregulating or catalytically interfering with expression, transcription or translation of a nucleic acid encoding the member of the leukotriene path; altering the post-translational processing of the polypeptide; altering the transcription of the splice variants; or by interfering with the activity of the polypeptide (e.g., by binding to the polypeptide, or by binding to another polypeptide that interacts with that member of the leukotriene pathway, such as a FLAP binding agent as described herein or some other binding agent of a member of the leukotriene pathway, by altering the interaction between two or more members of the leukotriene pathway (e.g., the interaction between FLAP and 5-LO), or by antagonizing the activity of a member of the leukotriene pathway. Representative leukotriene synthesis inhibitors include the following: agents that inhibit the activity of a member of the leukotriene biosynthetic pathway (e.g., FLAP, 5-LO), LTC4S, LTA4H, such as the agents presented in Table I of the Subsequent agent: agents that inhibit the activity of receptors of leukotriene pathway members, such as FLAP receptors, LTA4 receptors, LTB4 receptors, r LTC4 receptors, LTD4 receptors, LTE4 receptors, Cys LTl receptors, Cys LT2 receptors, 5-LO receptors; BLT1; BLT2; CysLTRl; CysLTR2; agents that bind to members of the leukotriene pathway, such as FLAP binding agents (e.g., 5-LO) or agents that bind to receptors of leukotriene pathway members (e.g., leukotriene receptor antagonists) ); agents that bind to a leukotriene (e.g., LTA4, LTB4, LTC4-, LTD4, LTE4, Cys LT1, Cys LT2); agents that increase the interruption of leukotrienes (e.g., LTB4DH); or other agents that otherwise affect (e.g., increase or decrease) leukotriene activity; antibodies to leukotrienes; non-coding nucleic acids or double-stranded interference RNAs, small nucleic acids encoding FLAP, 5-LO, or a leukotriene synthetase or other member of the leukotriene pathway, or fragments or derivatives thereof, including non-nucleic acids. encoding nucleic acids encoding FLAP, 5-LO or leukotriene synthetase polypeptides, and vectors comprising such non-coding nucleic acids (e.g., nucleic acid, cDNA and / or mRNA, double-stranded RNA interference, or an acid nucleic acid encoding an active fragment or derivative thereof, or an oligonucleotide, for example, the complement of one of SEQ ID NOS: 1 or 3, or a nucleic acid complementary to the nucleic acid encoding SEQ. IDENT. NO .: 2, or fragments or derivatives thereof); peptidomimetics; fusion proteins or prodrugs thereof; ribozymes; other small molecules; and other agents that alter (e.g., inhibit or antagonize) the expression of a member of the leukotriene pathway, such as the expression of the FLAP or 5-LO nucleic acid or the activity of the polypeptide, or that regulate the transcription of variants splicing or splice variants of 5-LO (for example, agents that affect the splice variants, or that affect the amount of each splice variant are expressed) are expressed. - More than one leukotriene synthesis inhibitor can be used at the same time, if desired. The therapy is designed to alter the activity of a FLAP polypeptide, a 5-LO polypeptide, or another member of the leukotriene pathway in an individual, such as by inhibiting or antagonizing activity. For example, a leukotriene synthesis inhibitor may be administered in order to decrease the synthesis of the leukotrienes within the individual, or to deregulate or decrease the expression or availability of the FLAP nucleic acid or the splice variants specific to the FLAP nucleic acid. The deregulation or decreasing expression or availability of a native FLAP nucleic acid or a particular splice variant could minimize the expression or activity of a defective nucleic acid or the particular splice variant and therefore minimize the impact of the defective nucleic acid or variant of particular splice. Similarly, for example, a leukotriene synthesis inhibitor can be administered in order to deregulate or decrease the expression or availability of the nucleic acid encoding 5-LO or the splice variants specific for the nucleic acid encoding 5-LO. The leukotriene synthesis inhibitor (s) is administered in a therapeutically effective amount (ie, an amount that is sufficient to treat the disease or condition, such as by improving symptoms associated with the disease or condition, by preventing or delaying the onset of disease or condition, and / or also reducing the severity or frequency of symptoms of the disease or condition). The amount which will be therapeutically effective in the treatment of a particular disease or condition of the individual will depend on the symptoms and severity of the disease, and can be determined by standard clinical techniques. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dose ranges. The precise dose that is used in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of a practitioner and each circumstance of the patient. The effective dose can be extrapolated from dose response curves derived from in vitro or animal model test systems. In the preferred embodiments of the invention, the leukotriene synthesis inhibiting agent is an agent that inhibits the activity of FLAP and / or 5-LO. Preferred agents include the following, as set forth in Table I of the Agent Subsequently: TABLE AGENT I co H t tSi H L? Or L? Or L? TABLE AGENT I In the preferred methods of the invention, the agents set forth in Table III of the Agent can be used for prophylactic and / or therapeutic treatment for diseases and conditions associated with FLAP or with other members of the leukotriene pathway, or with the increased synthesis of leukotriene. . In particular, they can be used for the treatment of myocardial infarction or susceptibility to myocardial infarction, such as for individuals in a population at risk, as described above (for example, based on the identified risk factors such as high cholesterol). , high C reactive protein, and / or genotype); for individuals suffering from acute coronary syndrome, such as unstable angina, myocardial infarction without ST elevation (NSTEMI) or myocardial infarction with ST elevation (STEMI); methods to reduce the risk of MI, stroke, PAOD in people with asymptomatic ankle / brachial index less than 0.9; to decrease the risk of a subsequent myocardial infarction, such as in individuals who have already had one or more myocardial infarctions; for apoplexy or susceptibility to stroke; to decrease the risk of a second stroke; for PAOD or susceptibility to PAOD; for the treatment of atherosclerosis, such as in patients requiring treatment (eg, angioplasty, stents, revascularization procedure) to restore blood flow in arteries (eg, coronary, carotid, and / or femoral arteries); for the treatment of an asymptomatic ankle / brachial index of less than 0.9; and / or to decrease the synthesis of leukotriene (eg, for the treatment of myocardial infarction, ACS, stroke or PAOD.) In a preferred embodiment of the invention, the leukotriene synthesis inhibitor is a FLAP inhibitor such as acid. - ((4-chlorophenyl) methyl) -3- ((1,1-dimethylethyl) thio) -alpha, alpha-dimethyl-5- (2-quinolinylmethoxy) -1H-indole-2-propanoic otherwise known as MK -0591, acid (R) - (+) - alpha-cyclopentyl-4- (2-quinolinylmethoxy) -Benzenacetic otherwise known as BAY-x-1005, 3- (3- (1,1-dimethylethylthio-5) - (quinolin-2-ylmethoxy) -1- (4-chloromethylphenyl) indol-2-yl) -2,2-dimethylpropionaldehyde oxime-0-2-acetic acid otherwise known as A-81834, its enantiomers, salts, derivatives chemicals, analogs or other optically pure compounds that inhibit FLAP that effectively decrease the biosynthesis of leukotriene when administered to humans.In another preferred embodiment of the invention, the synthesis inhibitor of the leukotriene is a 5LO inhibitor such as zileuton, atreleuton, 6- ((3-fluoro-5- (tetrahydro-4-methoxy-2H-pyran-4-yl) phenoxy) methyl) -l-methyl-2 (lH) -quinolinone otherwise known as ZD-2138, l - ((4-chlorophenyl) methyl) -3- ((1,1-dimethylethyl) thio) -alpha, alpha-dimethyl-5- (2-quinolinylmethoxy) -1H- indole-2-propanoic otherwise known as MK-886, 4- (3- (4- (2-Methyl-imidazol-1-yl) phenylsulfanyl) -phenyl) -tetrahydro-pyran-4-carboxylic acid amide another mode known as CJ-13610, its enantiomers, salts, chemical derivatives, analogs or other optically pure compounds that inhibit 5-LO that effectively decreases leukotriene biosynthesis when administered to humans. The compound can be represented by the following formula: in M is selected from the group consisting of hydrogen, a pharmaceutically acceptable cation, and a pharmaceutically acceptable metabolically cleavable group; B is a linear or branched divalent alkylene group of from one to twelve carbon atoms; Z is thiazolyl, optionally substituted with alkyl of one to six carbon atoms or haloalkyl of one to six carbon atoms; L is selected from the group consisting of (a) alkylene from 1-6 carbon atoms, (b) alkenylene from 2-6 carbon atoms, (c) alkynylene from 2-6 carbon atoms, (d) hydroxyalkyl of 1-6 carbon atoms, (e) >; C = 0, (f) > C = N-OR ?, wherein Rx is hydrogen or Ci-C6 alkyl, (g) (CHR1) n (CO) (CHR2) m, when n and m are independently selected from an integer from one to six and Rx and R2 are independently selected from hydrogen and C -C6 alkyl, (h) - (CHR?) N C = NOR2, where Ri, R2 and n are as defined above; (i) - (CHR?) n ON = CR2, where Rx, R2 and n are as defined above; (j) - (CHRxJn-O- (CHR2) m-, where Rx, R2, n and m are as defined above, (k) - (CHRi) n-NR2 (CHR3) ra-, where i, R2, n and m are as defined above and R 3 is selected from hydrogen and C 6 -alkyl; (1) - (CHR 1) n S -CHR 2) m-, wherein Ri, R 2, n and m are as defined above; and (m) - (CHR!) n- (S02) - (CHR2) m-, wherein R1 R2, n and m are as defined above; A is carbocyclic aryl optionally substituted with alkyl of one to six carbon atoms, haloalkyl of one to six carbon atoms, hydroxyalkyl of one to six carbon atoms, alkoxy of one to twelve carbon atoms, alkoxyalkoxy in which the two portions Alkoxy can each independently contain from one to six carbon atoms, alkylthio from one to six carbon atoms, hydroxy, halogen, cyano, amino, alkylamino of one to six carbon atoms, dialkylamino in which the two alkyl groups can contain independently from one to six carbon atoms, alkanoylamino from two to eight carbon atoms, N-alkanoyl-N-alkylamino wherein the alkanoyl is from two to eight carbon atoms and the alkyl group is from one to six carbon atoms, alkylaminocarbonyl from two to eight carbon atoms, dialkylaminocarbonyl in which the two alkyl groups are independently from one to six carbon atoms, boxyl, alkoxycarbonyl of from two to eight carbon atoms, phenyl, optionally substituted with alkyl of one to six carbon atoms, haloalkyl of one to six carbon atoms, alkoxy of one to six carbon atoms, hydroxy or halogen, phenoxy, optionally substituted with alkyl of one to six carbon atoms, haloalkyl of one to six carbon atoms, alkoxy of one to six carbon atoms, hydroxy or halogen, and phenylthio, optionally substituted with alkyl of one to six carbon atoms, haloalkyl of one to six carbon atoms, alkoxy from one to six carbon atoms, hydroxy or halogen. Preferably, the compound is a compound or a pharmaceutically acceptable salt thereof having the name (R) -N-. { 3- [5- (4-fluorophenylmethyl) thiazo-2-yl] -lmethyl-2-propynyl} -N-hydroxyurea. See U.S. Patent No. 4,615,596, incorporated herein by reference. The compound is replaced by the following formula: or a pharmaceutically acceptable salt thereof, wherein A is selected from the group consisting of linear or branched divalent alkylene or from one to twelve carbon atoms and divalent cycloalkylene from three to eight carbon atoms; Ra is selected from the group consisting of hydrogen, alkylthio or from one to six carbon atoms, phenylthio, optionally substituted by alkyl of one to six carbon atoms, alkoxy or from one to six carbon atoms, or halogen, phenylalkylthio wherein the alkyl portion contains from one to six carbon atoms, and the phenyl group is optionally substituted with alkyl of one to six carbon atoms, alkoxy of one to six carbon atoms, or halogen, R2 is selected from the group which consists of COOB wherein B is selected from hydrogen, a pharmaceutically acceptable cation or a metabolically unlinkable group, -COOalkyl wherein the alkyl portion contains from one to six carbon atoms, -COOalkylcarbocyclylaryl wherein the alkyl portion contains from one to six carbon atoms and the aryl portion is optionally substituted with alkyl from one to six carbon atoms, alkoxy or from one to six atom carbon, or halogen, -CONR5R6 wherein R5 is selected from the group consisting of hydrogen, hydroxyl, alkyl or from one to six carbon atoms, and alkoxy from one to six carbon atoms, and R6 is selected from the group consisting of hydrogen and alkyl from one to six carbon atoms, -COR6 and -OH; R3 is selected from the group consisting of phenylalkyl wherein the alkyl portion contains from one to six carbon atoms, and the phenyl group is optionally substituted with alkyl or from one to six carbon atoms, alkoxy or from one to six atoms of carbon, or halogen, R 4 is selected from the group consisting of thiazolylalkoxy wherein the alkyl portion contains from one to six carbon atoms, and the heteroaryl portion is optionally substituted with alkyl from one to six carbon atoms, alkoxy from one to six carbon atoms, or halogen, and thiazolyloxy optionally substituted with alkyl of one to six carbon atoms, alkoxy of one to six carbon atoms, or halogen. See U.S. Patent No. 5,288,743, incorporated herein by reference. The compound can be represented by the formula: or a pharmaceutically acceptable salt thereof, wherein M is selected from the group consisting of hydrogen, and a pharmaceutically acceptable cation; B is a linear or branched divalent alkylene group of from one to twelve carbon atoms; Z is selected from the group consisting of: (a) furyl, optionally substituted with alkyl of one to six carbon atoms, or haloalkyl of one to six carbon atoms, and (b) thienyl, optionally substituted with alkyl from one to six carbon atoms, or haloalkyl from one to six carbon atoms; and L is alkylene from 1-6 carbon atoms; A is phenyl optionally substituted with alkyl of one to six carbon atoms, haloalkyl of one to six carbon atoms, hydroxyalkyl of one to six carbon atoms, alkoxy of one to twelve carbon atoms, alkoxyalkoxy in which the two alkoxy portions they can each independently contain from one to six carbon atoms, alkylthio from one to six carbon atoms, hydroxy, halogen, cyano, amino, alkylamino from one to six carbon atoms, dialkylamino in which the two alkyl groups can independently contain from one to six carbon atoms, alkanoylamino of from two to eight carbon atoms, N-alkanoyl-N-alkylamino in which the ancanoyl is from two to eight carbon atoms and the alkyl group is from one to six carbon atoms, alkylaminocarbonyl from two to eight carbon atoms, dialkylaminocarbonyl in which the two alkyl groups are independently from one to six carbon atoms, carboxyl, alkoxycarbonyl of from two to eight carbon atoms, phenyl, optionally substituted with alkyl of one to six carbon atoms, haloalkyl of one to six carbon atoms, alkoxy of one to six carbon atoms, hydroxy or halogen, phenoxy, optionally substituted with alkyl of one to six carbon atoms, haloalkyl of one to six carbon atoms, alkoxy of one to six carbon atoms, hydroxy or halogen, or phenylthio, optionally substituted by alkyl or from one to six carbon atoms, haloalkyl from one to six carbon atoms, alkoxy from one to six carbon atoms, hydroxy or halogen. Preferably, the compound is a compound or a pharmaceutically acceptable salt thereof selected from the group consisting of: N-. { 3- (5- (4-fluorophenylmethyl) fur-2-yl) -3-butin-2-yl} -N-hydroxyurea; N-. { 3- (5- (4-fluorophenylmethyl) -2-thienyl) -l-methyl-2-propynyl} -N-hydroxyurea; (R) -N-. { 3- (5- (4-fluorophenylmethyl) -2-thienyl) -1-methyl-2-propynyl} -N-hydroxyurea; and (R) -N-. { 3- (5- (4-chlorophenylmethyl) -2-thienyl) -l-methyl-2-propynyl} -N-hydroxyurea; (S) -N-. { 3- [5- (4-fluorophenylmethyl) -2-thienyl] -l-methyl-2-propynyl} -N-hydroxyurea. See U.S. Patent No. 5,288,751, incorporated herein by reference. The compound can be represented by the formula: or a pharmaceutically acceptable salt thereof, wherein A is selected from the group consisting of linear or branched divalent alkylene of one to twelve carbon atoms, linear or branched divalent alkenylene of two to twelve carbon atoms, and divalent cycloalkylene of three to eight carbon atoms; R1 is alkylthio of one to six carbon atoms; R6 is selected from the group consisting of hydrogen and alkyl of one to six carbon atoms; R7 is selected from the group consisting of (carboxyl) alkyl in which the alkyl portion is from one to six carbon atoms, (alkoxycarbonyl) alkyl in which the alkoxycarbonyl portion is from two to six carbon atoms and the alkyl portion is from one to six carbon atoms , (aminocarbonyl) alkyl in which the alkyl portion is from one to six carbon atoms, ((alkylamino) carbonyl) alkyl in which each alkyl portion is independently from one to six carbon atoms, and ((dialkylamino) carbonyl) alkyl in which each alkyl portion is independently from one to six carbon atoms; R3 is phenylalkyl in which the alkyl portion is from one to six carbon atoms; R 4 is 2-, 3- or 6-quinolylmethoxy, optionally substituted with alkyl of one to six carbon atoms, haloalkyl of one to six carbon atoms, alkoxy of one to twelve carbon atoms, halogen, or hydroxy. Preferably, the compound is selected from the group consisting of: 3- (3-1, 1-dimethylethylthio) -5- (quinolin-2-ylmethoxy-1- (4-chlorophenylmethyl) -indol-2-yl} acid ) -2, 2-dimethylpropionaldehyde oxime-0-2 acetic; 3- (3- (1,1-dimethylethylthio) -5- (quinolin-2-ylmethoxy) -1- (4-chloro-phenylmethyl) indol-2-yl) -2,2-dimethylpropionaldehyde oxime-0-2 (3-methyl) butyric; 3- (3- (1,1-dimethylethylthio) -5- (6,7-dichloroquinolin-2-ylmethoxy) -1- (4-chlorophenylmethyl) -indol-2-yl) -2,2-dimethylpropionaldehyde oxime- 0-2-acetic; and 3- (3- (1,1-dimethylethylthio) -5- (6-fluoroquinolin-2-ylmethoxy) -1- (4-chlorophenylmethyl) indol-2-yl) -2,2-dimethylpropionaldehyde oxime-0- 2-propionic; or a pharmaceutically acceptable salt or ester thereof. See U.S. Patent No. 5,459,150, incorporated herein by reference. The compound can be represented by the formula: -Ar Q2 or pharmaceutically acceptable salts thereof, wherein Q is a 9, 10 or 11-membered heterocyclic-bicyclic moiety containing one to two nitrogen heteroatoms and optionally containing an additional heteroatom selected from nitrogen, oxygen and sulfur, and Q can optionally support up to four substituents selected from halogen, hydroxy, cyano, formyl, oxo, thioxo, alkyl of (Cl-4), alkenillo of. (C3-4), alkynyl (C3-4), alkoxy of (Cl-4), fluoroalkyl of (Cl-4), hydroxyalkyl of (Cl-4), alkanoyl of (C2-5), phenyl, benzoyl and benzyl, and wherein the phenyl substituents , benzoyl and benzyl optionally can support one or two substituents selected from halogen, alkyl of (Cl-4) and alkoxy of (Cl-4); X is oxy, thio, sulfinyl or sulfonyl;. Ar is phenylene, pyridindiyl, pyrimidindiyl, thiophendiyl, furandiyl, thiazoldiyl, oxazoldiyl, thiadiazoldiyl or oxadiazoldiyl which may optionally support one or two substituents selected from halogen, cyano, trifluoromethyl, hydroxy, amino, alkyl (Cl-4), alkoxy of (Cl-4), alkylamino of (Cl-4) and dialkylamino of (Cl-4); and Q is selected from groups of formulas II and III: p m where R is hydrogen, alkanoyl or benzyl of (C2-5), and where the benzoyl group can withstand Optionally one or two substituents selected from halogen, alkyl of (Cl-4) and alkoxy of (Cl-4); R is alkyl of (Cl-4); and R is hydrogen or (Cl-4) alkyl; or R and R are joined to form a methylene, vinylene, ethylene or trimethylene group. Preferably, the compound is selected from the group consisting of: (2S, 4R) -4- [5-fluoro-3- (1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-ylthio) phenyl] -4-hydroxy-2-ethyltetrahydropyran, (2S, R) -4- [5-fluoro-3- (1-methyl-2-oxo-l, 2,3,4-tetrahydroquinolin-6-ylsulfonyl) phenyl] -4-hydroxy-2-methyltetrahydropyran, (2S, 4R ) -4-hydroxy-2-methyl-4- [2- (1-methyl-2-oxo-l, 2, 3, 4-tetrahydroquinolin-6-ylthio) thiazol-5-yl] tetrahydropyran, (2S, 4R ) -4-hydroxy-2-methyl-4- [2- (1-methyl-2-oxo-l, 2,3,4-tetrahydroquinolin-6-ylsulfonyl) thiazol-5-yl] tetrahydropyran, (2S, 4R ) -4- [2- (7-Fluoro-l-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-ylthio) thiazol-5-yl] -4-hydroxy-2-methyltetrahydropyran, ( 2S, 4R) -4-hydroxy-2-methyl-4- [2- (1-methyl-2-oxoindolin-5-ylthio) thiazol-5-yl] tetrahydropyran, (2S, 4R) -4-hydroxy-2 -methyl-4- [2 (1-methyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-ylthio) thien-4-yl) tetrahydropyran, (2S, 4R) -4-hydroxy-2- methyl-4- [2- (1-methyl-2? xo-l, 2, 3, 4-tetrahydroquinolin-6-ylsulfonyl) thien-4-yl] tetrahydropyran, (2S, 4R) -4-hydroxy-2 -methyl-4- [2- (l-methyl -2-oxo-l, 2,3,4-tetrahydroquinolin-6-ylthio) thien-5-yl] tetrahydropyran, (2S, 4R) -4-hydroxy-2-methyl-4- [2- (l-methyl -2-oxo-l, 2-dihydroquinolin-6-ylthio) thien-4-yl] tetrahydropyran, (2S, 4R) -4-hydroxy-2-methyl-4- [2- (1, 8-dimethyl-2 -oxo-1, 2,3,4-tetrahydroquinolin-6-ylthio) thien-4-yl] tetrahydropyran, 4- [2- (8-fluoro-1-methyl-2-oxo-1, 2,3,4 -tetrahydroquinolin-6-ylthio) thien-4-yl] -4-hydroxy-2-methyltetrahydropyran, 4- [2- (7-fluoro-l-methyl-2-oxo-l, 2, 3, 4-tetrahydroquinolin- 6-ylthio) thien-4-yl] -4-hydroxy-2-methyltetrahydropyran, (2S, 4R) -4-hydroxy-2-methyl-4- [2- (l-methyl-2-oxoindolin-5-ylthio ) thien-4-yl] tetrahydropyran, (2S, 4R) -4-hydroxy-2-methyl-4- [3- (1-methyl-2-oxo-l, 2, 3,4-tetrahydroquinolin-6-ylthio ) phenyl] tetrahydropyran, (2S, 4R) -4-hydroxy-2-methyl-4- [3- (l-methyl-2-oxo-l, 2, 3, 4-tetrahydroquinolin-6-ylsulfonyl) phenyl] tetrahydropyran , (2S, 4R) -4 - [3- (1-ethyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-ylthio) phenyl] -4-hydroxy-2-methyltetrahydropyran, (2S, 4R ) -4- [3- (7-fluoro-l-me) til-2-oxo-1,2,3,4-tetrahydroquinolin-6-ylthio) phenyl] -4-hydroxy-2-methyltetrahydropyran, (2S, 4R) -4-hydroxy-2-methyl-4- [3- (1-methyl-2-oxo-l, 2-dihydroquinolin-6-ylthio) phenyl) tetrahydropyran, (2S, 4R) -4- [3- (8-chloro-l-methyl-2-oxo-1, 2 , 3,4-tetrahydroquinolin-6-ylthio) phenyl] -4-hydroxy-2-methyltetrahydropyran and (2S, 4R) -4-hydroxy-2-methyl-4- [3- (1-methyl-2-oxoindoline- 5-ylthio) phenyl] tetrahydropyran. EP 623614 Bl, incorporated herein by reference. The compound can be represented by the formula: wherein Q is a 10-membered heterocyclic-bicyclic portion containing one or two nitrogen heteroatoms which support one or two thioxo substituents, and whose heterocyclic portion may optionally support one, two or three additional substituents selected from halogen, hydroxy , cyano, amino, alkyl of (Cl-4), alkoxy of (Cl-4), fluoroalkyl of (Cl-4), alkylamino of (Cl-4), di- [(Cl-4) alkyl] amino, -aminoalkyl of (Cl-4), alkylamino of (Cl-4) -alkyl of (Cl-4), di- [(Cl-4) alkyl] amino-alkyl of (Cl-4), phenyl and phenylalkyl of (Cl-4), and wherein the phenyl or phenylalkyl substituent of (Cl-4) may optionally support a substituent selected from halogen, (Cl-4) alkyl and (Cl-4) alkoxy; wherein A is a direct linkage to X or is alkylene of (Cl-3); wherein X is oxy, thio, sulfinyl, sulfonyl or imino; wherein Ar is phenylene which can optionally support one or two substituents selected from halogen, hydroxy, amino, nitro, cyano, carbamoyl, ureido, alkyl of (Cl-4), alkoxy of (Cl-4), alkylamino of (Cl-4), di- [(Cl-4) alkyl] amino, fluoroalkyl of (Cl-4) and (C 2-4) alkanoylamino; or Ar is pyridylene; wherein R is (Cl-4) alkyl, (C3-4) alkenyl or (C3-4) alkynyl; and wherein R and R together form a group of the formula -AXA-, which together with the carbon atom to which A and A come together, defines a ring having 5 to 7 atoms in the ring, wherein A and A, which may be the same or different, each is alkylene of (Cl-3) and X is oxy, thio, sulfinyl or sulfonyl, and whose ring can support one, two or three substituents, which may be the same or different, selected from hydroxy, alkyl of (Cl-4) and alkoxy of (Cl-4); or wherein R and R together form a group of the formula -AXA-, which together with the oxygen atom to which A is attached and with the carbon atom to which A is attached, defines a ring having 5 to 7 atoms in the ring, where A and A, which may be the same or different, each is alkylene of (Cl-3) and X is oxy, thio, sulfinyl or sulfonyl, and whose ring can support one, two or three alkyl substituents of (Cl-4), and wherein R is (Cl-4) alkyl, (C 2-4) alkenyl or (C 2-4) alkynyl; or a pharmaceutically acceptable salt thereof. Preferably, the compound is selected from the group consisting of: 4- (5-fluoro-3- (l-methyl-2-thioxo-1,2-dihydroquinolin-6-ylmethoxy) phenyl] -4-ethoxytetrahydropyran and 4- (5-fluoro-3- (l-methyl-2-thioxo-1,2,4,4-tetrahydroquinolin-6-ylmethoxy) phenyl] -4-methoxytetrahydropyran, 4- (5-fluoro-3- (l -methyl-2-thioxo-1,2,3,4-tetrahydroquinolin-6-ylthio) phenyl] -4-methoxytetrahydropyran and a pharmaceutically acceptable salt thereof See EP 466452 Bl, incorporated herein by reference. to be a substituted 4- (quinolin-2-61-methoxy) phenylacetic acid derivative represented by the following formula: or a pharmaceutically acceptable salt thereof, wherein R 1 represents a group of the formula: -OR2 -N \ R and R3 are identical or different and represent hydrogen, lower alkyl, phenyl, benzyl or a group of the formula: R * S4 -CH- -C02R5 C 1 H -CH 2 O • CH- R 4 represents hydrogen, lower alkyl, phenyl or benzyl, which may be optionally substituted by hydroxyl, carboxyl, lower alkoxycarbonyl, lower alkylthio, heteroaryl or carbamoyl, R 5 represents hydrogen, lower alkyl, phenyl or benzyl, R 6 represents a group of the formula - COR5 or -C0R2R5, R7 represents hydrogen, lower alkyl or phenyl, Y represents a group of the formula: wherein R8 represents hydrogen, lower alkyl or phenyl and n denotes a number from 0 to 5, Z represents norbornyl or represents a group of the formula: wherein R9 and R10 are identical or different and denote hydrogen, lower alkyl or phenyl, or R9 and R10 can together form a saturated carbocyclic ring having up to 6 carbon atoms and m denotes a number from 1 to 6, and A and B are identical or different and denote hydrogen, lower alkyl or halogen, or a pharmaceutically acceptable salt thereof. Preferably, the compounds are selected from the group consisting of: 2- [4- (quinolin-2-yl-methoxy) phenyl] -2-cyclopentyl acetic acid, 2- [4- (quinolin-2-yl- methoxy) phenyl] -2-cyclohexylacetic acid and 2- [4- (quinolin-2-yl-methoxy) phenyl] -2-cycloheptylacetic acid, (+) -enantiomer of 2- [4- (quinolin-2-yl- methoxy) phenyl] -2-cyclopentylacetic, (-) -enantiomer of 2- [4- (quinolin-2-yl-methoxy) phenyl] -2-cyclopentyl acetic acid and pharmaceutically acceptable salts thereof. See U.S. Patent No. 4,970,215, incorporated herein by reference. The compound can be represented by the formula: wherein R, R, R, R and R are independently hydrogen, halogen, lower alkyl, lower alkenyl, lower alkynyl, -CF3, -CN, -N02, -N3, -C (OH) RR, -C02R, -SR , -S (0) R, -S (0) 2 R, -S (0) 2 NRR, -OR, -NRR, -C (0) RO - (CH 2) t R; R is hydrogen, -CH 3, -CF 3, -C (0) H, X-R O -X-R; R and R are independently: alkyl, - (CH2) uPh (R) 2 or - (CH2) uTh (R) 2; R is -CF3 or R; R is hydrogen or X-r; each R is independently hydrogen or lower alkyl, or two R on the same carbon atom join to form a cycloalkyl ring of 3 to 6 carbon atoms; R is hydrogen, lower alkyl or -CH2R; R is lower alkyl or - (CH2) rR; R is -CF3 or R; R is hydrogen, -C (0) R, R, or two R in the same nitrogen can be joined to form a monocyclic heterocyclic ring of 4 to 6 atoms containing up to 2 heteroatoms chosen from O, S or N; R is hydrogen, -CF3, lower alkyl, lower alkenyl, lower alkynyl or - (CH2) rR; R is - (CH2) s-C (RR) - (CH2) s-R or -CH2C (0) NRR; R is hydrogen or lower alkyl; R is a) a heterocyclic monocyclic or bicyclic ring containing from 3 to 9 nuclear carbon atoms and 1 or 2 nuclear heteroatoms selected from N, S or O with each ring in the heterocyclic radical which is formed of 5 or 6 atoms, or b) the radical -R; R is alkyl or C (0) R; R is phenyl substituted with 1 or 2 R groups; R is hydrogen, halogen, lower alkyl, lower alkoxy, lower alkylthio, lower alkylsulfonyl, lower alkylcarbonyl, -CF3, -CN, -N02 or -N3; R is alkyl, cycloalkyl, monoheterocyclic monocyclic ring; R is the residual structure of a standard amino acid, or R and R attached to the same N can be cyclized to form a proline residue; m is 0 to 1; n is 0 to 3; p is 1 to 3, when m is 1; p is 0 to 3 when m is 0; r is 0 to 2; s is 0 to 3; t is 0 to 2; u is 0 to 3; v is 0 or 1; is 0, S or NR; X is 0 or NR; X is C (O), CRR, S, S (O) or S (0) 2, X is C (O), CRR, S (0) 2 or a bond; And it's X or X; Q is -C02R, -C (O) NHS (O) 2R, -NHS (0) 2R, -S (0) 2NHR -C (0) NRR, -C02R, -C (0) NRR, -CH20H or 1H - or 2H-tetrazol-5-yl; and pharmaceutically acceptable salts thereof. Preferred embodiments of the compounds are selected from the following and pharmaceutically acceptable salts thereof: 3- [N- (p-chlorobenzyl) -3- (t-butylthio) -5- (quinolin-2-ylmethoxy) ) indol-2-yl] -2, 2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3-methyl-5- (quinolin-2-ylmethoxy) indol-2-yl) -2,2-dimethylpropanoic acid; 3- [N- (p-t-butylthiobenzyl) -3- (t-butylthio) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3- (phenylthio) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3- (phenylsulfonyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, N-oxide; 3- [N- (p-chlorobenzyl) -3- (phenylsulfonyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3- (phenylsulfinyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3-benzoyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3-benzyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3- (3,3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2- acid dimethylpropanoic; 2- [N- (p-chlorobenzyl) -3- (t-butylthio) -5- (quinolin-2-ylmethoxy) indol-2-yl] ethoxyethoic acid; 3- [N- (p-chlorobenzyl) -3- (3, 3-dimethyl-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3- (t-butylthio) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2-methylpropanoic acid; 3- [N- (p-chlorobenzyl) -3-methyl-5- (6,7-dichloroquinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -3-methyl-5- (7-chloroquinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -4-allyl-5- (quinolin-2-ylmethoxy) -3- (t-butylthio) indol-2-yl] -2,2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -4-allyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -6- (quinolin-2-ylmethoxy) -3- (t-butylthio) indol-2-yl] -2,2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -4- (quinolin-2-ylmethoxy) -3- (t-butylthio) indol-2-yl] -2,2-dimethylpropanoic acid; 3- [N- (p-chlorobenzyl) -7- (quinolin-2-ylmethoxy) -3- (t-butylthio) indol-2-yl] -2,2-dimethylpropanoic acid; 2- [2- [N- (p-chlorobenzyl) -3- (t-butylthio) -5- (quinolin-2-ylmethoxy) indol-2-yl] ethoxy] propanoic acid; 3- [N- (p-chlorobenzyl) -4- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid; 3- [N-methyl-3- (p-chlorobenzoyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N-methyl-3- (p. -chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3-y-propoxy-5- (quinolin-2) -ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (t-butylthio) -5- (quinolin-2-yl-methoxy) indole-2] -yl] -2-ethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3-trifluoroacetyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [3- N- (4-chlorobenzyl) -3- (3, 3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2-methylpropanoic acid, 3- [ 3- (3, 3-dimethyl-l-oxo-l-butyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N- (4-trifluoromethylbenzyl) -3- (3, 3-dimethyl-1-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N-benzyl-3] - (3, 3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropa noic acid, 3- [N- (3-methoxybenzyl) -3- (3, 3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N-allyl-3- (3, 3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2- dimethylpropanoic acid, 3- [N- (4-methoxybenzyl) -3- (3, 3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2- dimethylpropanoic, 3- [N-methyl-3- (3, 3-dimethyl-l-oxo-3-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, acid 3- [3- (4-chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid. 3- [N- (phenylsulfonyl) -3- (4-chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N-benzyl-3-] acid (4-chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (t-butylsulfonyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (t-butylsulfinyl) -5- (quinolin-2-ylmethoxy) indole] -2-yl] -2, 2-dimethylpropanoic acid, 3- [N-allyl-3- (4-chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (n-propyl) -3- (4-chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N-ethyl- 3- (4-Chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (4-t-butylbenzoyl) ) -5- (quinolin-2-yl-methoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (4-chlorobenzoyl) -5- (quinolin -2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid 3- [N- (4-chlorobenzyl) -3- (1, 1-dimethylethyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic, 3- [N- ( 4-chlorobenzyl) -3-acetyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3-cyclopropanecarbonyl-5- ( quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (3-cyclopentylpropanoyl) -5- (quinolin-2-ylmethoxy) indole- 2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (3-methylbutanoyl) -5- (quinolin-2-yl-methoxy) indol-2-yl] -2 , 2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3-propanoyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- ( 4-chlorobenzyl) -3- (2-methylpropanoyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] 2, 2-dimethylpropanoic, 3- [N- (4-chlorobenzyl) -3-trimethylacetyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- ( 4-chlorobenzyl) -3-phenylacetyl-5- (q'uinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-fluorobenzyl) -3- (3, 3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N- (4-bromobenzyl) -3- ( 3,3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2 -dimethylpropanoic acid, 3- [N- (4-iodobenzyl) -3- - (3,3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- acid. { N- (4-chlorobenzyl) -3- (1,1-dimethylbutyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) ) -3- (1, 1-dimethylpropyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (3-fluorobenzyl) -3- (1, 1-dimethylethyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (3-methylethyl) -5- ( quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3-cyclopropyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (1-methyl-1-cyclopropyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2 -dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3-cyclopentyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4- chlorobenzyl) -3-cyclohexyl-5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (alpha, alpha-dimethylbenzyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (2-. { 4-chloro-alpha, alpha-dimethylbenzyl} -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (1-adamantyl) -5- (quinolin-2-) ilmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- ((1-adamantyl) methyl) -5- (quinolin-2-ylmethoxy) indole-2 -yl] -2,2-dimethylpropanoic acid, 3- [N- (1,1-dimethylethyl) -3- (4-chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2, 2 -dimethylpropanoic acid, 3- [N- (1,1-dimethylpropyl) -3- (4-chlorobenzyl) -6- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoic acid, 3- [N- (4-chlorobenzyl) -3- (3, 3-dimethyl-l-oxo-l-butyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-diethylpropanoic acid, 3 - [N- (4-chlorobenzyl) -3,6-bis (acetyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoate or 3- [N- (4- chlorobenzyl) -3,6-bis (cyclopropancarbonyl) -5- (quinolin-2-ylmethoxy) indol-2-yl] -2,2-dimethylpropanoate methyl. See EP 419049 Bl, incorporated herein by reference. The term "alkyl" refers to a monovalent group derived from a straight or branched chain saturated hydrocarbon by the removal of a single hydrogen atom. Alkyl groups are exemplified by methyl, ethyl, n- and isopropyl, n-, sec-, iso- and tert-butyl and the like. The term "hydroxyalkyl" represents an alkyl group, as defined above, substituted by one to three hydroxyl groups with the proviso that no more than one hydroxy group can be attached to a single carbon atom of the alkyl group. The term "alkylamino" refers to a group having the structure -NHR 'wherein R' is alkyl, as previously defined, examples of alkylamino include methylamino, ethylamine, isopropylamino and the like. The term "alkylaminocarbonyl" refers to an alkylamino group as previously defined, attached to the parent molecular moiety through a carbonyl group. Examples of alkylaminocarbonyl include methylaminocarbonyl, ethylaminocarbonyl, iso-propylaminocarbonyl and the like. The term "alkylthio" refers to an alkyl group, as defined above, attached to the parent molecular moiety through a sulfur atom and includes such examples as methylthio, ethylthio, propylthio, n-, sec- and tert-butylthio. and similar. The term "alkanoyl" represents an alkyl group, as defined above, linked to the parent molecular moiety through a carbonyl group. The alkanoyl groups are exemplified by formyl, acetyl, propionyl, butanoyl and the like. The term "alkanoylamino" refers to an alkanoyl group as previously defined, attached to the parent molecular moiety through a nitrogen atom. Examples of alkanoylamino include formamido, acetamido and the like. The term "N-alkanoyl-N-alkylamino" refers to an alkanoyl group, as previously defined, attached to the parent molecular moiety through an aminoalkyl group. Examples of N-alkanoyl-N-alkylamino include N-methylformamido, N-methyl acetamido and the like. The terms "alkoxy" or "alkoxy" denote an alkyl group, as defined above, attached to the parent molecular moiety through an oxygen atom. Representative alkoxy groups include methoxy, ethoxy, propoxy, butoxyl and the like. The term "alkoxyalkoxy" refers to an alkyl group, as defined above, linked through an oxygen to an alkyl group, as defined above, in turn linked through an oxygen to the parent molecular moiety. Examples of alkoxyalkoxy include methoxymethoxy, methoxyethoxy, ethoxyethoxy and the like. The term "alkoxyalkyl" refers to an alkoxy group, as defined above, linked through an alkylene group to the parent molecular moiety. The term "alkoxycarbonyl" represents an ester group, that is, an alkoxy group, attached to the parent molecular moiety through a carbonyl group such as methoxycarbonyl, ethoxycarbonyl, and the like. The term "alkenyl" denotes a monovalent group derived from a hydrocarbon containing at least one carbon-carbon double bond by the removal of a single hydrogen atom. Alkenyl groups include, for example, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl and the like. The term "alkylene" denotes a divalent group derived from a saturated straight-chain or branched hydrocarbon by the removal of two hydrogen atoms, for example, methylene, 1,2-ethylene, 1,1-ethylene, 1, 3-propylene, 2,2-dimethylpropylene and the like. The term "alkenylene" denotes a divalent group derived from a straight or branched chain hydrocarbon containing at least one carbon-carbon double bond. Examples of alkenylene include -CH = CH, -CH2 CH = CH-, C (CH3) = CH-, -CH2 CH = CHCH2- and the like. The term "cycloalkylene" refers to a divalent group derived from a carbocyclic hydrocarbon saturated by the removal of two hydrogen atoms, for example, cyclopentylene, cyclohexylene and the like. The term "cycloalkyl" denotes a monovalent group derived from a saturated, monocyclic or bicyclic carbocyclic ring compound by the removal of a single hydrogen atom. Examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, bicyclo [2.2.1] heptanil and bicyclo [2.2.2] octanyl. The term "alkynylene" refers to a divalent group derived by the removal of two hydrogen atoms from a straight or branched chain acyclic hydrocarbon group containing a carbon-carbon triple bond. Examples of alkynylene include -CH = CH-, -CH = CH-CH2, -CH = CH-CH (CH3) -, and the like. The term "carbocyclic aryl" denotes a group in the monovalent carbocyclic ring derived by the removal of a single hydrogen atom from a combined or unbranched monocyclic or bicyclic ring system that obeys the "4n + 2p electron" or Huckel aromaticity. Examples of carbocyclic aryl groups include, phenyl, 1 and 2-naphthyl, biphenylyl, fluorenyl and the like. The term "(carbocyclic aryl) alkyl" refers to a group on the carbocyclic aryl ring as defined above, attached to the parent molecular moiety through an alkylene group. Representative (carbocyclic aryl) alkyl groups include phenylmethyl, phenylethyl, phenylpropyl, 1-naphthylmethyl and the like. The term "carbocycliclaryl coxy" refers to an alkyl carbocyclic alkyl group, as defined above, attached to the parent molecular moiety through an oxygen atom. The term "carbocyclic aryloxyalkyl" refers to a carbocyclic aryl group, as defined above, attached to the progenitor molecular moiety through an oxygen atom and therefore through an alkylene group. Such groups are exemplified by phenoxymethyl, 1 and 2-naphthyloxymethyl, phenoxyethyl and the like. The term "(carbocyclic aryl) alkoxyalkyl" denotes a carbocyclic aryl group as defined above, attached to the progenitor molecular moiety through an alkoxyalkyl group. Representative (carbocyclic aryl) alkoxyalkyl groups include phenylmethoxymethyl, phenylethoxymethyl, 1- and 2-naphthylmethoxyethyl and the like. "carbocyclic arylthioalkyl" represents a carbocyclic aryl group as defined above, attached to the parent molecular moiety through a sulfur atom and therefore through an alkylene group and typified by phenylthiomethyl, 1- and 2-naphthylthioethyl and Similar. The term "dialkylamino" refers to a group having the structure -NR'R "wherein R 'and R" are independently selected from alkyl, as previously defined. Additionally, R 'and R "taken together can optionally be - (CH2) - e where kk is an integer from 2 to 6. Examples of dialkylamino include, dimethylamino, diethylaminocarbonyl, methylethylamino, piperidino and the like The term" halo or halogen " "denotes fluorine, chlorine, bromine or iodine The term" haloalkyl "denotes an alkyl group, as defined above, having one, two or three halogen atoms attached thereto and exemplified by such groups as chloromethyl, bromoethyl, trifluoromethyl and the like The term "hydroxyalkyl" represents an alkyl group, as defined above, substituted by one to three hydroxyl groups with the proviso that no more than one hydroxy group can be attached to a single carbon atom of the alkyl group. "Phenoxy" refers to a phenyl group attached to the parent molecular moiety through an oxygen atom.The term "phenylthio" refers to a phenyl group attached to the parent molecular moiety. through a sulfur atom. The term "pyridyloxy" refers to a pyridyl group attached to the parent molecular moiety through an oxygen atom. The terms "heteroaryl" or "heterocyclic aryl" as used herein refer to aromatic groups on the 5 or 6 membered ring containing one oxygen atom, one, two, three or four nitrogen atoms, one nitrogen and one nitrogen atom. a sulfur atom, or a nitrogen and an oxygen atom. The term "heteroaryl" also includes bi-or tricyclic groups wherein the aromatic heterocyclic ring is combined with one or two benzene rings. Representative heteroaryl groups are pyridyl, thienyl, indolyl, pyrazinyl, isoquinolyl, pyrrolyl, pyrimidyl, benzothienyl, furyl, benzo [b] furyl, imidazolyl, thiazolyl, carbazolyl and the like. The term "heteroarylalkyl" denotes a heteroaryl group, as defined above, attached to the parent molecular moiety through an alkylene group. The term "heteroaryloxy" denotes a heteroaryl group, as defined above, attached to the progenitor molecular moiety through an oxygen atom. The term "heteroarylalkoxy" denotes a heteroarylalkyl group, as defined above, attached to the parent molecular moiety through an oxygen atom.
Method for Reducing Risk Factors for Cardiovascular Disease The present invention encompasses compositions and methods for reducing risk factors for MI, ACS, stroke and / or PAOD. The method for reducing risk factors comprises administering a composition comprising a leukotriene synthesis inhibitor, described in detail herein alone, or in combination with a statin, to an individual at risk for any of these conditions. Individuals at risk include the target population described herein, especially individuals with elevated CRP, and those at risk for other diseases and conditions associated with FLAP and / or other members of the leukotriene pathway. In particular, the invention encompasses methods for reducing levels of plasma CRP or plasma amyloid A serum levels comprising administering an effective amount of the leukotriene inhibitor alone or in combination with a statin. Statins are competitive inhibitors of 3-hydroxy-3-methylglutarlcoenzyme A (HMG-CoA) reductase, the enzyme that converts HMG-CoA to mevalonic acid, the precursor of cholesterol. In the active site binding of HMG-CoA reductase, statins alter the enzyme's confirmation, so it is avoided from obtaining a functional structure. The conformational change of the active site HMG-CoA reductase makes very effective and specific statin drugs. The inhibition of HMG-CoA reductase reduces the synthesis of intracellular cholesterol in hepatocytes. The reduction of intracellular cholesterol results in an increase in hepatic LDL receptors on the cell surface, which in turn reduces the level of circulating LDL and its precursors, intermediate density lipoproteins (IDL) and very low density lipoproteins (VLDL). In addition, statins inhibit hepatic synthesis of apolipoprotein B-100, which results in a decrease in the synthesis and secretion of triglyceride-rich lipoproteins. Additional beneficial effects of statins on lipid biosynthesis include the inhibition of LDL oxidation, and the inhibition of the expression of scavenger receptors. Statins also reduce the accumulation of esterified cholesterol in macrophages, increase the synthesis of nitric oxide of the endothelial cell, reduce inflammatory processes, increase the stability of atherosclerotic plaques, and restore platelet activity and the coagulation process. Due to its beneficial effects and high specificity, statins have become some of the most prescribed medicines in the industrialized world. In the preferred embodiments of the invention, the statin is one of the following agents: rovuvastatin, fluvastatin, atorvastatin, lovastatin, simvastatin, pravastatin or pitavastatin. These agents are described in detail in H H 3 to o Mevastatin and related compounds are described in U.S. Patent No. 3,983,140. Lovastatin (mevinolin) and related compounds are described in U.S. Patent No. 4,231,938. Keto analogues of mevinolin (lovastatin) are described in European Patent Application No. 0,142,146 A2, and quinoline and pyridine derivatives are described in US Patent No. 5,506,219 and 5,691,322. Pravastatin and related compounds are described in U.S. Patent No. 4,346,227. Simvastatin and related compounds are described in U.S. Patent Nos. 4,448,784 and 4,450,171. Fluvastatin and related compounds are described in U.S. Patent No. 5,354,772. Cerivastatin and related compounds are described in U.S. Patent Nos. 5,006,530 and 5,177,080. Atorvastatin and related compounds are described in U.S. Patent Nos. 4,681,893; 5,273,995; 5,385,929 and 5,686,104. Pitavastatin (nisvastatin (NK-104) or itavastatin) and related compounds are described in U.S. Patent No. 5,011,930. Rosuvastatin (visastatin (ZD-4522)) and related compounds are described in U.S. Patent No. 5,260,440. Other possible molecules of HMG CoA reductase are described in US Patent Nos. 5,753,675; 4,613,610; 4,686,237; 4,647,576; and 4,499,289; and British Patent No. GB 2205837. The patents cited in relation to statins or other agents identified herein are now described for making and using statins / agents, as well as biochemically active homologs thereof, salts, prodrugs, metabolites and the like. . Such patents are incorporated herein by reference in their entirety. Dosages for statins have also been described in the patent and commercial literature (for example, Physician's Desk Reference 2004, incorporated herein by reference) and by the manufacturers and clinical practitioners who prescribe them. Combination therapy that uses statin dosages similar to what is used when statins are prescribed alone, or less, is specifically contemplated. Compositions comprising a leukotriene synthesis inhibitor alone or in combination with a statin can comprise a leukotriene synthesis inhibitor in an amount effective to reduce a risk factor such as CRP or amyloid serum A. Effective daily doses of the inhibitors of leukotriene synthesis are between .01 mg and 100 g, more preferably 0.1 mg to 1 g, and all individual doses within these ranges are specifically contemplated. Doses in single exemplary adults include 10 mg, 25 mg, 50 mg, 75 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 500 mg, and 750 mg, from one to four times a day. The compositions may comprise a statin in an amount effective to reduce total serum cholesterol, serum LDL, and / or serum CRP. Effective daily doses are between 0.01 mg and 100 g, more preferably 0.1 mg to 1 g, and all individual doses within these ranges are specifically contemplated. Exemplary individual doses include 5 mg, -10 mg, 15 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, and 80 mg, 100 mg, 150 mg, 200 mg, 250 mg, and 500 mg from one to four times a day. The incipient evidence suggests that elevated CRP is an independent risk factor for adverse clinical outcomes. See, for example, Ridker et al. , N. Engl. J. Med. 352: 1 (January 6, 2005). In another variation, the invention provides compositions, unit doses, and methods of treatment wherein a leukotriene synthesis inhibitor and a statin are included or administered in amounts that act synergistically to reduce serum CRP levels. Synergistically effective amounts are • amounts that either (a) achieve a greater percent reduction in CRP than is achieved in an average patient using any type of agent alone, in a safe and effective amount, or (b) reduce CRP by a comparable amount to single-agent therapy, with few side effects; or (c) reduces CRP by an amount comparable to single agent therapy, and also reduces at least one other cardiovascular risk factor more effectively than just single agent therapy. In one variation, the invention provides a composition comprising a leukotriene synthesis inhibitor and a statin for simultaneous administration, for example, in a dose. A composition in the form of a tablet, pill or capsule, including sustained release formulations, is specifically contemplated. In another variation, a unit dose comprising a single dose of the leukotriene synthesis inhibitor and a single dose of the statin, packaged together, but not in admixture, is contemplated. In another variation, the methods of the invention involve administering a composition comprising a leukotriene inhibitor and a composition comprising a statin at the same or different times, for example, by administering the leukotriene synthesis inhibitor before or after the administration of a composition comprising a statin. The compositions for and methods for administering the agents to an individual continuously (for example, through a patch or iv), once or twice a day, once a day, every other day, twice a week, weekly, or monthly for one or more weeks, months or years, or for the complete life of a patient, depending on the level of risk for the individual, is specifically contemplated, to manage CRP in serum and other levels of factor in cardiovascular risk. It is contemplated that these compositions will be used for the treatment and lifestyle management plans for prevention of primary or secondary MI, ACS, stroke or PAOD.
Therapc Agents of Nucleic Acid In another embodiment, a nucleic acid of the invention; a nucleic acid complementary to a nucleic acid of the invention; or a portion of such nucleic acid (for example, an oligonucleotide as described below); or a nucleic acid encoding a member of the leukotriene pathway (eg, 5-LO), can be used in "non-coding" therapy wherein a nucleic acid (eg, an oligonucleotide) which specifically hybridizes to the mRNA and / or genomic DNA of a nucleic acid is generally administered in situ. The non-coding nucleic acid that specifically hybridizes to the mRNA and / or DNA inhibits the expression of the polypeptide encoded by that mRNA and / or DNA, for example, by inhibiting translation and / or transcription. The binding of the non-coding nucleic acid can be by complementarity of conventional base pairs, or for example, in the case of DNA duplex binding, through the specific interaction in the largest groove of the double helix. A non-coding construct can be delivered, for example, as an expression plasmid as described above. When the plasmid is transcribed in the cell, it produces RNA that is complementary to a portion of the mRNA and / or the DNA encoding the polypeptide for the leukotriene pathway member (e.g., FLAP or 5-LO). Alternatively, the non-coding construct can be an oligonucleotide probe that is generated ex vivo and is introduced into the cells; then it inhibits expression by hybridizing with the mRNA and / or the genomic DNA of the polypeptide. In one embodiment, oligonucleotide probes are modified oligonucleotides that are resistant to endogenous nucleases, for example, exonucleases and / or endonucleases, so they are stable in vivo. Exemplary nucleic acid molecules for use as non-coding oligonucleotides are DNA phosphoramidate, phosphothioate and methylphosphonate analogues (see also US Pat. Nos. 5,176,996, 5,264,564 and 5,256,775). Additionally, general methods for constructing oligomers useful in non-coding therapy are also described, for example, by Van der Krol et al. , (Biotechniques 6: 958-976 (1988)); and Stein et al. (Cancer Res. 48: 2659-2668 (1988)). With respect to the non-coding DNA, oligodeoxyribonucleotides derived from the translation initiation site are preferred. To perform non-coding therapy, oligonucleotides (mRNA, cDNA or DNA) are designated as being complementary to mRNA encoding the polypeptide. The non-coding oligonucleotides bind to mRNA transcripts and prevent translation. Absolute complementarity, although preferred, is not required. A sequence "complementary" to a portion of an RNA, as referred to herein, indicates that a sequence has sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; in the case of double-stranded non-coding nucleic acids, a single strand of the duplex DNA can thus be tested, or a triple formation can be evaluated. The ability to hybridize will depend both on the degree of complementarity and on the length of the non-coding nucleic acid, as described in detail above. Generally, the longer the hybridizing nucleic acid, the more base incompatibility with an RNA can contain and still form a stable duplex (or triplex), according to the case) . One skilled in the art can determine a tolerable degree of incompatibility by the use of standard procedures. The oligonucleotides used in non-coding therapy can be DNA, RNA or mixtures or chemical derivatives or modified versions thereof, single-stranded or double-stranded. The oligonucleotides can be modified in the base portion, the sugar portion, or the phosphate structure, for example, to improve the stability of the molecule, hybridization, etc. Oligonucleotides may include other adjoining groups such as peptides (e.g., to direct host cell receptors in vivo), or agents that facilitate transport across the cell membrane (see for example, Letsinger et al., Proc. Nati. Acad Sci. USA 86: 6553-6556 (1989); Lemaitre et al. , Proc. Nati Acad. Sci. USA 84: 648-652 (1987); PCT International Application No. WO 88/09810) or the blood-brain barrier (see, for example, PCT International Publication No. WO 89/10134), or hybridization-activated cleavage agents (see for example, Krol et al. ., BioTechniques 6: 958-976 (1988)) or intercalation agents. (See, for example, Zon, Pharm, Res. 5: 539-549 (1988)). For this purpose, the oligonucleotide can be conjugated to another molecule (for example, a peptide, crosslinking agent activated by hybridization, transport agent, splitting agent activated by hybridization). The non-coding molecules are delivered to cells expressing the leukotriene pathway member in vivo. A number of methods can be used to deliver non-coding DNA or RNA to the cells; for example, the non-coding molecules can be injected directly into the tissue site, or the modified non-coding molecules, designed to target the desired cells (eg, non-coding bound to peptides or antibodies that specifically bind receptors or antigens expressed on the surface). target cell) can be administered systematically. Alternatively, in a preferred embodiment, a recombinant DNA construct is used, wherein the non-coding oligonucleotide is placed under the control of a strong promoter (eg, pol III or pol II). The use of such a construct to transfect target cells in the patient results in the transcription of sufficient amounts of single-stranded RNAs that will form base pairs complementary to the endogenous transcriptionists and consequently prevent translation of the mRNA. For example, a vector can be introduced in vivo so that it is absorbed by a cell and the transcription of a non-coding RNA is directed. Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired non-coding RNA. Such vectors can be constructed by recombinant DNA technology methods standard in the art and described above. For example, a plasmid, cosmid, YAC or viral vector can be used to prepare the recombinant DNA construct that can be introduced directly into the tissue site. Alternatively, viral vectors may be used, which selectively infect the desired tissue, in which case administration may be achieved by another route (eg, systematically). In another embodiment of the invention, small double-stranded interference RNA (RNA interference (RNAi)) can be used. RNAi is a post-transcription process, where double-stranded RNA is introduced, and specific genetic silencing results, through catalytic degradation of the target mRNA. See, for example, Elbashir, S.M., et al. , Nature 411: 494-498 (2001); Lee, N.S., Nature Biotech. 15: 500-505 (2002); Lee, S-K. et al. , Nature Medicine 8 (7): 681-686 (200'2); the complete teachings of these references are incorporated herein by reference. RNAi is used routinely to investigate genetic function in a high throughput form or to modulate gene expression in human diseases (Chi et al., PNAS, 100 (11): 6343-6346 (2003)). The introduction of long double-stranded RNA leads to the sequence-specific degradation of the homologous genetic transcriptionists. The long double-stranded RNA is metabolized to siRNA of 21-23 small nucleotides (interfering RNA, small). The siRNA is then linked to the protein RISC complex (RNA-induced silencing complex) with dual-function helicase. The helicase has RNAase activity and is able to separate the RNA. Unlinked siRNA allows a non-coding strand to bind to a target. This results in the sequence dependent degradation of the cognate mRNA. Apart from the endogenous RNAi, exogenous RNAi, chemically synthesized or recombina produced can also be used. When using non-intronic portions of the FLAP gene, such corresponding mRNA portions of SEC. FROM IDENT. DO NOT. : 1, or portions of the SEC. FROM IDENT. NO .: 3, target regions of the FLAP gene that are accessible to RNAi are targeted and silenced. With this technique, it is possible to conduct a genetic run of RNAi of the nucleic acids of the FLAP gene and determine the amount of inhibition of the protein product. In this way, it is possible to design specific genetic therapies directing the mRNA of the gene. The endogenous expression of a member of the leukotriene pathway (e.g., FLAP, 5-LO) can also be reduced by inactivating or "knocking out" the gene or its promoter using target homologous recombination (e.g. see Smithies et al., Nature 317 : 230-234 (1985); Thomas & Capecchi, Cell 51: 503-512 (1987); Thompson et al. , Cell 5: 313-321 (1989)). For example, a non-functional gene, altered from a member of the leukotriene pathway (or an unrelated DNA sequence) flanked by the DNA homologous to the endogenous gene (any of the coding regions or regulatory regions of the gene) can used, with or without a selectable marker and / or a negative selectable marker, to transfect cells expressing the gene in vivo. The insertion of the DNA construct, through objective homologous recombination, results in the inactivation of the gene. Recombinant DNA constructs can be administered or directed directly to the required in vivo site using appropriate vectors, as described above. Alternatively, the expression of unaltered genes can be increased using a similar method; The objective homologous recombination can be used to insert a DNA construct comprising an unaltered functional gene, or the complement thereof, or a portion thereof, in place of a gene in the cell, as described above. In another embodiment, the target homologous recombination can be used to insert a DNA construct comprising a nucleic acid encoding a polypeptide variant that differs from that present in the cell. Alternatively, the endogenous expression of a member of the leukotriene pathway can be reduced by directing deoxyrubonucleotide sequences complementary to the regulatory region of the leukotriene pathway member (i.e., the promoter and / or enhancers) to form the triple helical structures that they avoid transcription of the gene in target cells in the body. (See generally, Helene, C, Anticancer Drug Des., 6 (6): 569-84 (1991); Helene, C. et al., Ann., NY Acad. Sci. 660: 27-36 (1992); and Maher, LJ Bioassays 14 (12): 807-12 (1992)). Likewise, the non-coding constructs described herein, by antagonizing the normal biological activity of one of the members of the leukotriene pathway, can be used in tissue manipulation, eg, tissue differentiation, both in vivo and - for ex vivo tissue cultures. In addition, non-coding techniques (eg, microinjection of non-coding molecules, or transfection with plasmids whose transcriptionists are non-coding with respect to a nucleic acid RNA or nucleic acid sequence) can be used to investigate the role of one or more members of the leukotriene trajectory in the development of conditions related to diseases. Such techniques can be used in cell culture, but can also be used in the creation of transgenic animals. The therapeutic agents as described herein may be delivered in a composition, as described above, or by themselves. These can be administered systematically, or they can be directed to a particular tissue. The therapeutic agents can be produced by a variety of means, including chemical synthesis; recombinant production; in vivo production (e.g., a transgenic animal, such as U.S. Patent No. 4,873,316 to Meade et al.), for example, and can be isolated using standard means such as those described herein. In addition, a combination of any of the above methods of treatment (eg, administration of a non-altered polypeptide together with the non-coding therapy that directs the altered mRNA to a member of the leukotriene pathway; administration of a first variant of splicing together with the non-coding therapy that directs a second splice variant) can also be used. The invention further relates to the use of such therapeutic agents, as described herein, for the manufacture of a medicament for the treatment of MI, ACS, stroke, PAOD and / or atherosclerosis, for example, using the methods described in I presented.
Monitoring Progress of Treatment The present invention also relates to methods for monitoring the response of an individual, such as an individual in one of the target populations described above, to treatment with a leukotriene synthesis inhibitor. Because the level of inflammatory markers can rise in individuals who are in the target populations described above, an assessment of the level of inflammatory markers of individuals both before and during treatment with the leukotriene synthesis inhibitor will indicate whether the treatment has The production of leukotrienes in the arterial vascular wall or in the inflammatory cells derived from the spinal cord has been successfully reduced. For example, in one embodiment of the invention, an individual who is a member of a target population as described above (e.g., an individual at risk of MI, ACS, stroke or PAOD, such as an individual who is at risk due to a FLAP haplotype) can be evaluated for response to treatment with a leukotriene synthesis inhibitor, by examining levels of leukotriene or levels of leukotriene metabolite in the individual. Leukotrienes in blood, serum, plasma or urine (for example, leukotriene? 4, cysteinyl leukotriene 1), or the ex vivo production of leukotrienes (for example, in blood samples stimulated with a calcium ionophore to produce leukotrienes), or metabolites of leukotriene, can be measured before, and during or after treatment with the leukotriene synthesis inhibitor. The level of leukotriene or leukotriene metabolite before treatment is compared to the level of leukotriene or leukotriene metabolite during or after treatment. The efficacy of the treatment is indicated by a decrease in leukotriene production: a level of leukotriene or leukotriene metabolite during or after treatment that is significantly lower than the level of leukotriene or leukotriene metabolite before treatment is indicative of effectiveness. A level that is lower during or after treatment may be shown, for example, by reduced serum or urine leukotrienes, or decreased ex vivo production of leukotrienes, or decreased leukotriene metabolites. A level that is "significantly lower" as used herein, is a level that is lower than the amount that is normally found in the control (s), or is lower in a comparison of the risk of disease in a population associated with the other measurement bands (for example, the median or median, the highest quartile or the highest quintile) compared to the lower measurement bands (for example, the median or median, the other quartiles, the other quintiles). For example, in one embodiment of the invention, the level of a leukotriene or leukotriene metabolite is evaluated in an individual prior to treatment with a leukotriene synthesis inhibitor; and during or after treatment with the leukotriene synthesis inhibitor, and the levels are compared. A level of leukotriene or the leukotriene metabolite during or after treatment that is significantly lower than the level of leukotriene or leukotriene metabolite prior to treatment is indicative of efficacy of the inhibitor * fe_: 73X leukotriene synthesis. In another embodiment, the production of a leukotriene or leukotriene metabolite is stimulated in a first test sample from the individual, using a calcium ionophore, prior to treatment with an inhibitor of l.eukotriene synthesis, and is also stimulated in a second test sample from the individual, using a calcium ionophore, during or after treatment with the leukotriene synthesis inhibitor, and the level of production in the first test sample is compared to the level of leukotriene production or the goal of leukotriene in the second test sample. A level of leukotriene or leukotriene metabolite in the second test sample that is significantly lower than the level of the leukotriene or leukotriene metabolite in the first test sample, is indicative of efficacy of treatment with the leukotriene synthesis inhibitor. In another embodiment of the invention, an individual who is a member of a target population of individuals at risk of MI, ACS, stroke or PAOD (eg, an individual in a target population described above, such as an individual at risk due to high C reactive protein) can be evaluated for response to treatment with a leukotriene synthesis inhibitor, by examining levels of inflammatory markers in the individual. For example, levels of the inflammatory marker in an appropriate test sample (eg, serum, plasma or urine) can be measured before, and during or after treatment with the leukotriene synthesis inhibitor. The level of the inflammatory marker before treatment is compared to the level of the inflammatory marker during or after treatment. The efficacy of the treatment is indicated by a decrease in the level of the inflammatory marker, that is, a level of the inflammatory marker during or after the treatment that is significantly lower (eg, significantly lower) than the level of the inflammatory marker before the treatment, is indicative of efficacy. Representative inflammatory markers include: C-reactive protein (CRP), serum. Amyloid A, fibrinogen, a leukotriene (for example, LTB4, LTC4, LTD4, LTE4), a metabolite of leukotriene, interleukin-6, tissue factor alpha necrosis, soluble vascular cell adhesion molecules (sVCAM), soluble intervascular adhesion molecules (sICAM), E-selectin, matrix metalloprotease type 1, matrix metalloprotease type 2, matrix metalloprotease type 3, matrix metalloprotease type 9, myeloperoxidase (MPO), and N-tyrosine. In a preferred embodiment, the marker is CRP or MPO. The efficacy of the treatment of a leukotriene synthesis inhibitor can be monitored by measuring biomarkers at risk in plasma, serum or urine. Clinical trials are available by the following biomarkers: CRP, amyloid serum A, IL-lß, IL-6, IL-8, IL-10, TNF-a, E-selectin, P-selectin and the intracellular adhesion molecule 1 , the vascular cell adhesion molecule 1. The relative risk of a cardiovascular event predicted by CRP levels that is low risk is less than 1 mg / L, the average is 1.0-3.0 mg / L and high risk patients have more than 3.0 mg / L. Thus, the optimal therapeutic effect of a leukotriene synthesis inhibitor alone or in combination with a statin is to reduce the CRP level to 2.0 mg / L or lower. The effectiveness of the treatment of a statin is monitored by measuring the level of cholesterol in total serum, serum LDL and / or serum triglycerides. A level of total cholesterol in serum, LDL-C and / or triglycerides during or after treatment, which is significantly lower than the level of total cholesterol, LDL-C and / or triglycerides before treatment is indicative of the efficacy of the treatment. For cholesterol management purposes, "high-risk patients" have an LDL level of 130 mg / dL or higher and optimally the statin treatment will reduce the LDL level to less than 100 mg / dL. "Moderately high risk patients" are those individuals with two or more factors. of risk for coronary heart disease with a 10-20% risk of heart attack in the span of ten years. Optimally, the statin treatment will maintain the LDL level below 129 mg / dL. More recent studies show an additional benefit in morbidity and mortality when statin therapy decreased serum LDL-C less than 70 mg / dL. (Ridker et al., N. Engl. J. Med. 352 (1): 20-28, 2005; Nissen et al. , N. Engl. J. Med. 352 (1): 29-38, 2005). This optimal therapeutic effect of a statin would be at lower LDL-C levels below 70 mg / dL, as delineated by Ridker et al., N. Engl. J. Med. 352 (1): 20-28, 2005 and? Issen et al. , N. Engl. J. Med. 352 (1): 29-38, 2005, statin therapy can reduce CRP. He . CRP is an additional parameter that can be monitored along with statin therapy.
Increased Risk Assessment The present invention is further related to methods for evaluating an individual (e.g., an individual who is in a target population as described herein, such as an individual who is at risk for MI, ACS, stroke or PAOD), for an increased risk of MI, ACS, atherosclerosis, stroke, transient ischemic attack, transient monocular blindness, asymptomatic carotid stenosis, PAOD, claudication, or limb ischemia. The methods comprise evaluating the level of a leukotriene metabolite (e.g., LTE4, LTD4, LTB4) in the individual, wherein an increased level of leukotriene metabolite is indicative of an increased risk. The level can be measured in any appropriate tissue or fluid sample, such as blood, serum, plasma or urine. In a particular embodiment, the sample comprises neutrophils. The level of the leukotriene metabolite can be measured by standard methods, such as the methods described herein. For example, in one embodiment, the production of the leukotriene metabolite is stimulated in a first test sample from the individual, using a calcium ionophore. The production level is compared to a control level. The control level is a level that is normally found in the control individual (s), such as an individual who is not at risk of MI, ACS, stroke or PAOD; alternatively, a control level is the level that is found in comparison of the risk of disease in a population associated with the lowest measurement band (for example, below the median or median, the lowest quartile or the lowest quintile) compared to the highest measurement bands (for example, on the average or median, the second, third or fourth quartile, the second, third, fourth or fifth quintile). A production level of the leukotriene metabolite that is significantly higher than the control level is indicative of an increased risk. Individuals at increased risk are candidates for treatments described herein.
Pharmaceutical Compositions The present invention also relates to pharmaceutical compositions comprising agents described herein, for example, an agent that is a leukotriene synthesis inhibitor as described herein. For example, a leukotriene synthesis inhibitor can be formulated with a physiologically acceptable carrier or excipient to prepare a pharmaceutical composition. The carrier and the composition can be sterile. The formulation must adjust the mode of administration. The invention also provides compositions comprising a leukotriene synthesis inhibitor, as set forth in Table I of the Agent, and a statin, as set forth in Table III of the Agent. The leukotriene synthesis inhibitor and the statin can be co-formulated with a physiologically acceptable carrier or excipient to prepare a pharmaceutical composition. This composition can be a formulation for delivering the leukotriene synthesis inhibitor and a statin in a single dose. Processes for the isolation and purification of statins and other HMG-CoA reductase inhibitors include different combinations of extraction methods, chromatography, lactonization and crystallization. Examples of formulations for statins, statin derivatives and statin salts are found in. the following, all incorporated by reference in its entirety, the US patents Nos. 6,316,460, 6,589,959, RE37,314, 5,354,772, 5,356,896, 5,686,104, 5,969,156, 6,126,971, 5,030,447, 5,180,589, 5,622,985, 6,825,015, 6,838,566, -5,403,860, 5,763,653 and 5,763,646 , International Patent Publications WO 86/03488, WO 86/07054, French Patent No. 2596393, European Patent Application No. 0221025, British Patent Nos. 2055100A and 2073199A and European Patent No. 65,835. Suitable pharmaceutically acceptable carriers include, but are not limited to water, saline solutions (eg, NaCl), saline, regulated saline solution, alcohols, glycerol, ethanol, gum arabic, vegetable oils, benzylic alcohols, polyethylene glycols, gelatin, carbohydrates such as lactose, amylose or starch, dextrose, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid esters, hydroxymethyl cellulose, polyvinyl pyrrolidone, etc., as well as combinations thereof. The pharmaceutical preparations can, if desired, be mixed with auxiliary agents, for example, lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, dyes, flavorings and / or aromatics and the like, which do not they react noxiously with the active agents. The composition, if desired, may also contain minor amounts of wetting agents or emulsifiers, or pH regulating agents. The composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation or powder. The composition can be formulated as a suppository, with binders and traditional carriers such as triglycerides. The oral formulation may include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, polyvinylpyrrolidone, sodium saccharin, cellulose, magnesium carbonate, etc. Methods of introducing these compositions include, but are not limited to, intradermal, intramuscular, intraperitoneal, intraocular, intravenous, subcutaneous, topical, oral and intranasal. Other suitable methods of introduction may also include gene therapy (as described below), rechargeable or biodegradable devices, particle acceleration devices ("genetic cannons") and slow-release polymeric devices. The pharmaceutical compositions of this invention can also be administered as part of a combination therapy with other agents. The composition can be formulated according to routine procedures as a pharmaceutical composition adapted for administration to humans. For example, compositions for intravenous administration are usually solutions in a sterile isotonic aqueous buffer. When necessary, the composition may also include a solubilizing agent and a local anesthetic to alleviate pain at the site of injection. Generally, the ingredients are either separately supplied or mixed together in unit dosage form, for example, as a dry lyophilized powder or a water-free concentrate in a hermetically sealed container such as a vial or sachet indicating a quantity of the agent active. When the composition is to be administered by infusion, it can be distributed with an infusion bottle containing pharmaceutical grade water, sterile, saline or dextrose / water. When the composition is administered by injection, a vial of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
For topical application, non-sprayable, viscous to semi-solid forms or solid forms comprising a carrier compatible with topical application and having a dynamic viscosity preferably greater than water, may be employed. Suitable formulations include, but are not limited to solutions, suspensions, emulsions, creams, ointments, powders, enemas, lotions, suns-chemical colloids, liniments, balms, aerosols, etc., which are, if desired, sterilized or mixed with auxiliary agents, for example, preservatives, stabilizers, wetting agents, buffers or salts for influencing osmotic pressure, etc. The agent can be incorporated into a cosmetic formulation. For topical application, sprayable aerosol preparations are also suitable wherein the active ingredient, preferably in combination with a solid or liquid inert carrier material, is packaged in a squeeze bottle or in a mixture with a pressurized, volatile, normally gaseous propellant , for example, pressurized air. The agents described herein can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium , ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylaminoethanol, histidine, procaine, etc. The agents are administered in a therapeutically effective amount. The amount of agents that will be therapeutically effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dose ranges. The precise dose to be used in the formulation will also depend on the route of administration, and the seriousness of the symptoms, and should be decided according to the judgment of one practitioner and each of the patient's circumstances. Effective doses can be extrapolated from dose response curves derived from in vitro or animal model test systems. The invention also provides a pharmaceutical package or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such or such containers may be a notice in the form prescribed by a governmental agent that regulates the manufacture, the use or sale of pharmaceutical or biological products, whose notice reflects the approval by the manufacturing agency, the use of sale for human administration. The package or equipment can be labeled with information regarding the mode of administration, the sequence of drug administration (eg, separate, sequential or jointly) or the like. The package or equipment may also include means to remind the patient of the therapy. The package or equipment can be a single unit dose of the combination therapy or it can be a plurality of unit doses. In particular, the agents can be separated, mixed together in any combination, present in a single bottle or a tablet. For example, a package or equipment of the invention may contain a single dose for the delivery of both the leukotriene synthesis inhibitor and a statin at the same time, or contain two or more doses wherein a dose will supply a synthesis inhibitor. of leukotriene and one dose will supply a statin either in parallel or one after the other. Agents integrated in a blister pack or other distribution means are preferred. For the purpose of the invention, the unit dose is intended to mean a dose that is dependent on the individual pharmacodynamics of each agent and is administered in doses approved by the FDA in standard time courses.
Nucleic Acids of the Invention FLAP Nucleic Acids, Portions and Variants In addition, the invention relates to isolated nucleic acid molecules comprising a human FLAP nucleic acid. The term "FLAP nucleic acid" as used herein, refers to a FLAP polypeptide that encodes an isolated nucleic acid molecule. The FLAP nucleic acid molecules of the present invention can be RNA, for example, mRNA or DNA such as cDNA and genomic DNA. DNA molecules can be double-stranded or single-stranded; Single-stranded DNA RNA can be either the coding strand or uncoded strand, or non-coding strand. The nucleic acid molecule can include all or a portion of the coding sequence of the gene or nucleic acid and can further comprise non-coding sequences such as introns and 3 'and 5' sequences without coding (including regulatory sequences, for example, as well as promoters, transcription enhancing elements, splice donors / acceptor sites, etc.). For example, a FLAP nucleic acid may consist of SECs. FROM IDENT. NOS. : 1 or 3 or the complement thereof, or a portion of the fragment of such an isolated nucleic acid molecule (e.g., cDNA or nucleic acid) encoding the FLAP polypeptide (e.g., a polypeptide such as SEQ. IDENT. NO .: 2). In a preferred embodiment, the isolated nucleic acid molecule comprises a nucleic acid molecule selected from the group consisting of SEQ. FROM IDENT. NOS .: 1 or 3, or its complement of them. Additionally, the nucleic acid molecules of the invention can be combined to a marker sequence, for example, a sequence encoding a polypeptide that aids in the isolation or purification of the polypeptide. Such sequences include, but are not limited to, those that encode a glutathione-S-transferase (GST) fusion protein and those that encode a polypeptide marker of hemagglutinin A (HA) from influenza. An "isolated" nucleic acid molecule as used herein, is one that is separated from nucleic acids that normally flank the gene or nucleic acid sequence (as in genomic sequences) and / or has been completely purified. or partially from other transcribed sequences (for example, as in an RNA library). For example, an isolated nucleic acid of the invention can be substantially isolated with respect to the complex cellular medium where it occurs naturally, or the culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized. In some cases, the isolated material will form part of a composition (for example, an unpurified extract containing other substances), a buffer system or a reactive mixture. In other circumstances, the material can be purified to essential homogeneity, for example, as determined by PAGE or column chromatography such as HPLC. In certain embodiments, an isolated nucleic acid molecule comprises at least about 50, 80 or 90% (on a molar basis) of all macromolecular species present. With respect to genomic DNA, the term "isolated" may also refer to nucleic acid molecules that are separated from the chromosome with which the genomic DNA is naturally associated. For example, the isolated nucleic acid molecule can contain less than about 5 kb, including, but not limited to 4 kb, 3 kb, 2 kb, Ikb, 0.5 kb or 0.1 kb nucleotides which flank the nucleic acid molecule in the genomic DNA of the cell from which the nucleic acid molecule is derived. The nucleic acid molecule can be combined with other coding or regulatory sequences and still be considered isolated. Thus, the recombinant DNA contained in a vector is included in the definition of "isolated" as used herein. As well, isolated nucleic acid molecules include recombinant DNA molecules in heterologous host cells, as well as partial DNA molecules or substantially purified in solution. "Isolated" nucleic acid molecules also include RNA transcriptionists in vivo and in vi tro of the DNA molecules of the present invention. An isolated nucleic acid molecule or nucleic acid sequence can include a nucleic acid molecule or nucleic acid sequence that is synthesized chemically or by recombinant means. Therefore, the recombinant DNA contained in a vector is included in the definition of "isolated" as used herein. Also, the isolated nucleotide sequences include recombinant DNA molecules in heterologous organisms, as well as partial DNA molecules or substantially purified in solution. RNA transcriptionists in vivo and in vitro of the DNA molecules of the present invention are also encompassed by "isolated" nucleotide sequences. Such isolated nucleotide sequences are useful in the manufacture of the encoded polypeptide, as probes for isolating homologous sequences (e.g., from other mammalian species), for gene mapping (e.g., by in situ hybridization with chromosomes), or for detecting the expression of the nucleic acid in tissue (e.g., human tissue) such as by Northern blot analysis. The present invention also relates to nucleic acid molecules that are not necessarily found in nature, but which encode a FLAP polypeptide (e.g., a polypeptide having an amino acid sequence comprising an amino acid sequence of SEQ. IDENT NO: 2), or another variant of splicing of a FLAP polypeptide or polymorphic variant thereof. Thus, for example, DNA molecules comprising a sequence that is different from the nucleic acid sequence of natural origin but which, due to the degeneracy of the genetic code, encode a FLAP polypeptide of the present invention are also the objects of this invention. The invention also encompasses nucleotide sequences that encode portions (fragments), or that encode variant polypeptides such as analogs or derivatives of a FLAP polypeptide. Such variants may be of natural origin, such as in the case of allelic variation or single nucleotide polymorphisms, or non-natural, such as those induced by various mutagenic and mutagenic processes. The intended variants include, but are not limited to the addition, deletion and substitution of one or more nucleotides that may result in conservative or non-conservative amino acid changes, including additions and deletions. Preferably, the nucleotide changes (and / or the resulting amino acid) are silent and preserved; that is, they do not alter the characteristics or activity of a FLAP polypeptide. In a preferred embodiment, the nucleotide sequences are fragments comprising one or more polymorphic microsatellite markers. In another preferred embodiment, the nucleotide sequences are fragments comprising one or more single nucleotide polymorphisms in a FLAP nucleic acid (eg, the simple nucleotide polymorphisms set forth in Table 13, below). Other alterations of the nucleic acid molecules of the invention may include, for example, labeling, methylation, internucleotide modifications such as uncharged linkages (eg, methyl phosphonates, phosphotriesters, phosphoamidates, carbamates), charged linkages (eg example, phosphorothioates, phosphorodithioates), pendant portions (e.g., polypeptides), intercalators (e.g., acridine, psoralen), chelators, alkylators, and modified linkages (e.g., anomeric alpha nucleic acids). Also included are synthetic molecules that mimic nucleic acid molecules in the ability to bind to a designated sequence through a hydrogen bond and other chemical interactions. Such molecules include, for example, those in which the peptide ligaments replace the phosphate ligaments in the structure of the molecule. The invention also relates to nucleic acid molecules that hybridize under high stringency hybridization conditions, such as for selective hybridization, to a nucleic acid sequence described herein (e.g., nucleic acid molecules which specifically hybridize to a nucleic acid sequence encoding polypeptides described herein, and optionally, have a polypeptide activity). In one embodiment, the invention includes variants described herein which hybridize under high stringency hybridization conditions (e.g., for selective hybridization) to a nucleic acid sequence which comprises a nucleic acid sequence selected from the group that consists of the SEC. FROM IDENT. NOS .: 1 or 3 or the complement of them. In another embodiment, the invention includes variants described herein which hybridize under high stringency hybridization conditions (eg, for selective hybridization) to a nucleic acid sequence encoding an amino acid sequence of SEQ. FROM IDENT. DO NOT. : 2 or a polymorphic variant thereof. In a preferred embodiment, the variant that hybridizes under high stringency hybridizations has an activity of a FLAP. Such nucleic acid molecules can be detected and / or isolated by specific hybridization (eg, under conditions of high stringency). "Specific hybridization" as used herein, refers to the ability, of a first nucleic acid to hybridize to a second nucleic acid in a manner such that the first nucleic acid does not hybridize to any nucleic acid different from the second nucleic acid (e.g., when the first nucleic acid has a higher similarity to the second nucleic acid than to any other nucleic acid in a sample where the hybridization is to be performed). The "stringency conditions" for hybridization is a term of the art which refers to incubation and washing conditions, for example, temperature conditions and buffer concentration, which allows the hybridization of a particular nucleic acid to a second nucleic acid; the first nucleic acid may be perfectly (ie, 100%) complementary to the second, or the first and second may share some degree of complementarity that is less than the perfect (eg, 70%, 75%, 85%, 95% ). For example, certain conditions of high severity can be used, which perfectly distinguish complementary nucleic acids from those of less complementarity. "Conditions of high severity", "conditions of moderate severity" and "conditions of low severity" for nucleic acid hybridization are explained on pages 2.10.1-2.10.16 and pages 6.3.1-6.3.6 on Curren Protocols in Molecular Biology (Ausubel, FM et al. , "Current Protocols in Molecular Biology", John Wiley & Sons, (1998), the complete teachings of which are incorporated in the present for preference). The exact conditions which determine the severity of hybridization depend not only on the ionic strength (eg, 0.2X SSC, 0.1 X SSC), temperature (eg, room temperature, 42 ° C, 68 ° C) and the concentration of destabilizing agents such as formamide or denaturing agents such as SDS, but also factors such as the length of the nucleic acid sequence, base composition, percent of incompatibility between the hybridization sequences and the frequency of appearance of subsets of that sequence within other non-identical sequences. Thus, equivalent conditions can be determined by varying one or more of these parameters while maintaining a similar degree of identity or similarity between the two nucleic acid molecules. Typically, the conditions are used so that sequences of at least about 60%, at least about 70%, at least about 80%, at least about 90% or at least about 95% or more identical to each other remain hybridized one to the other. other. By varying the hybridization conditions from a level of severity at which hybridization does not occur to a level at which the hybridization is first observed, the conditions which will allow a given sequence to hybridize (eg, selectively) with the most similar in the sample can be determined. Exemplary conditions are described in Krause, M. H. and S. A. Aaronson, Methods in Enzymology 200: 546-556 (1991) and in Ausubel, et al. , "Current Protocols in Molecular Biology", John Wiley & Sons, (1998), which describes the determination of washing conditions to moderate or reduce severity conditions. Washing is the stage where the conditions are usually established to determine a minimum level of complementarity of the hybrids. Generally, at the beginning of the lowest temperature at which only homologous hybridization occurs, each ° C by which the final wash temperature is reduced (maintains the concentration SSC constant) allows a 1% increase in the maximum extent of incompatibility between the sequences that hybridize. Generally, doubling the concentration of SSC results in an increase in Tm of -17 ° C. When using these guidelines, the wash temperature can be determined empirically for high, moderate or low severity, depending on the level of incompatibility sought. For example, a low severity wash may comprise a wash in a solution containing 0.2X SSC / 0.1% SDS for 10 minutes at room temperature; a wash of moderate severity may include washing in a pre-heated solution (42 ° C) solution containing 0.2X SSC / 0.1% SDS for 15 minutes at 42 ° C; and a high severity wash may comprise washing in a pre-heated solution (68 ° C) containing 0. IX SSC / 0.1% SDS for 15 minutes at 68 ° C. In addition, the washings may be repeated or sequentially to obtain a desired result as is known in the art. Equivalent conditions can be determined by varying one or more of the given parameters as an example, as is known in the art, although a similar degree of identity or similarity is maintained between the target nucleic acid molecule and the primer or probe used. The percentage of homology or identity of two nucleotide or amino acid sequences can be determined by aligning the sequences for optimal comparison purposes (for example, the openings can be introduced in the sequence of a first sequence for optimal alignment).
The nucleotides or amino acids in the corresponding positions are then compared, and the percentage of identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e.,% identity = # of identical positions / total # of positions x 100). When a position in a sequence is occupied by the same nucleotide or amino acid residue as the corresponding position in the other sequence, then the molecules are homologous in position. As used herein, the "homology" of nucleic acid or amino acid is equivalent to the "identity" of nucleic acid or amino acid. In certain embodiments, the length of an aligned sequence for comparison purposes is at least 30%, for example, at least 40%, in certain modalities at least 60%, and in other modalities at least 70%, 80%, 90% or 95% of the length of the reference sequence. The current comparison of the two sequences can be achieved by well-known methods, for example, using a mathematical algorithm. A preferred non-limiting example of such a mathematical algorithm is described in Karlin et al. , Proc. Nati Acad. Sci. USA 90-5873-5877 (1993). Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) as described in Altschul et al. , Nucleic Acids Res. 25: 389-3402 (1997). When using BLAST and Gapped programs BLAST, the missing parameters of the respective programs (for example, NBLAST) can be used. In one embodiment, the parameters for sequence comparison can be set to a score = 100, word length = 12, or can be varied (for example, W = 5 or W = 20). Another non-limiting, preferred example of a mathematical algorithm used for the comparison of sequences is the algorithm of Myers and Miller, CABIOS 4 (1): 11-17 (1988). Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package (Accelrys, Cambridge, UK). When using the ALIGN program for comparison of amino acid sequences, a PAM120 weight residue table, an opening length penalty of 12, and an opening penalty of 4 may be used. Additional algorithms for sequence analysis are known in the art and include ADVANCE and ADAM as described in Torellis and Robotti, Comput. Appl. Biosci. 10: 3-5 (1994), and FASTA described in Pearson and Lipman, Proc. Nati Acad. Sci. USA 85: 2444-8 (1998). In another embodiment, the percent identity between two amino acid sequences can be achieved using the GAP program in the GCG software package using either a BLOSUM63 matrix or a PAM250 matrix, and an aperture weight of 12, 10, 8, 6 or 4 and a weight length of 2, 3 or 4. In yet another embodiment, the percent identity between two nucleic acid sequences can be achieved using the GAP program in the GCG software package using an aperture weight of 50 and a weight 3. The present invention also provides isolated nucleic acid molecules that contain a fragment or portion that hybridizes under high stringency conditions to the nucleic acid sequence comprising SEQ. FROM IDENT. NO .: 1 or 3 or the complement of the SEC. FROM IDENT. NO .: 1 or 3, and also provides isolated nucleic acid molecules containing a fragment or moiety that hybridizes under high stringency conditions to a nucleic acid sequence encoding an amino acid sequence of the invention or a polymorphic variant thereof. The nucleic acid fragments of the invention are at least about 15, for example, at least about 18, 20, 23 or 25 nucleotides, and may be 30, 40, 50, 100, 200 or more nucleotides in length. Longer fragments, eg, 30 or more nucleotides in length, encoding antigenic polypeptides described herein are particularly useful, such as for the generation of antibodies as described below.
Probes and Primers In a related aspect, the nucleic acid fragments of the invention are used as probes or primers in assays such as those described herein. "Probes" or "primers" are oligonucleotides that hybridize in a base-specific manner to a complementary strand of nucleic acid molecules. Such probes and primers include polypeptide nucleic acids, as described in Nielsen et al., (Science 254: 1497-1500 (1991)). A probe or primer comprises a nucleic acid region that hybridizes to at least about 15, for example, about 20-25, and in certain 40, 50 or 75 consecutive nucleotides of a nucleic acid of the invention, such as a nucleic acid comprising a continuous nucleic acid sequence of SEQ. FROM IDENT. NOS .: 1 or 3 or the complement of the SEC. FROM IDENT. US: 1 or 3, or a nucleic acid sequence encoding an amino acid sequence of SEQ. FROM IDENT. DO NOT. : 2 or polymorphic variant thereof. In preferred embodiments, a probe or primer comprises 100 or fewer nucleotides, in certain embodiments, from 6 to 50 nucleotides, for example, from 12 to 30 nucleotides. In other embodiments, the probe or primer is at least 70% identical to the contiguous nucleic acid sequence or the complement of the contiguous nucleotide sequence., for example, at least 80% identical, in certain embodiments at least 90% identical, and in other embodiments at least 95% identical, or even capable of selectively hybridizing to the contiguous nucleic acid sequence or complement of the nucleotide sequence contiguous Frequently, the probe or primer further comprises a tag, for example, a radioisotope, a fluorescent compound, an enzyme or an enzyme co-factor. Particularly useful probes and primers of the invention are those that hybridize marker-locations (eg, in the FLAP gene) and those that allow amplification (eg, using PCR) or a small DNA fragment that includes a marker. interest, especially markers forming haplotypes of the invention, the equipment containing one or two or three or more such probes and primers are contemplated as aspects of the invention. The nucleic acid molecules of the invention are those described above and can be identified and isolated using standard molecule biology techniques and the sequence information provided herein. For example, nucleic acid molecules can be amplified and isolated using the polymerase chain reaction and synthetic oligonucleotide primers based on one or more of SEQ. FROM IDENT. NOS. : 1 or 3, or the complement thereof, or designed based on nucleotides based on sequences encoding one or more of the amino acid sequences provided herein. See generally PCR Technology: Principles and Applications for DNA Amplification (ed. H. A. Erlich, Freeman Press, NY, NY, 1992); PCR Protocols: A Guide to Methods and Applications (Eds. Innis et al., Academic Press, San Diego, CA, 1990); Mattila et al. , Nucí. Acids Res. 19: 4967 (1991); Eckert et al., PCR Methods and Applications 1:17 (1991); PCR (eds. McPherson et al., IRL Press, Oxford); and U.S. Patent 4,683,202. The nucleic acid molecules can be amplified using cDNA, mRNA or genomic DNA as a template, cloned into an appropriate vector and characterized by a DNA sequence analysis. Other suitable amplification methods include the ligase chain reaction (LCR) (see Wu and Wallace, Genomics 4: 560 (1989), Landegren et al., Science 241: 1077 (1988), transcription amplification (Kwoh et al. , Proc. Nati, Acad. Sci. USA 86: 1173 (1989)) and self-sustained sequence replication (Guatelli et al., Proc. Nat. Acad. Sci. USA 87: 1874 (1990)) and sequence amplification. based on nucleic acid (NASBA) The last two methods of amplification involve isothermal reactions based on isothermal transcription, which produces both single-stranded RNA (ssRNA) and double-stranded DNA (dsDNA) as the amplification products in a ratio of about 30 or 100 to 1, respectively.The amplified DNA can be labeled, for example, radiolabelled and used as a probe to select a cDNA library derived from human cells, mRNA in Express zap, ZIPLOX or other suitable vector. clones corresp Intents can be isolated, DNA can be obtained followed by excision in vivo, and the cloned insert can be sequenced in either or both orientations by methods recognized in the art to identify the correct reading frame encoding a polypeptide of the appropriate molecular weight. For example, direct analysis of the nucleic acid molecules of the present invention can be achieved using well-known methods that are commercially available. See, for example, Sambrook et al. , Molecular Cloning, A Laboratory Manual (2nd Ed., CSHP, New York 1989); Zyskind et al. , Recombinant DNA Laboratory Manual, (Acad. Press, 1988)). By using these similar methods, the polypeptide and the DNA encoding the polypeptide can be isolated, sequenced and further characterized. The non-coding nucleic acid molecules of the invention can be designed using the nucleotide sequences of SEQ. FROM IDENT. NOS .: 1 or 3 and / or the complement of one or more of the SEC. FROM IDENT. NOS. : 1 or 3 and / or a portion of one or more of the SECs. FROM IDENT. NOS .: 1 or 3, or the complement of one or more of the SEC. FROM IDENT. US: 1 or 3 and / or a sequence encoding the amino acid sequence of SEC. FROM IDENT. DO NOT. : 2 or coding a portion of one or more of the SECs. FROM IDENT. NOS. : 1 or 3 or its complement. These can be constructed using chemical synthesis and enzymatic ligation reactions using methods known in the art. For example, a non-coding nucleic acid molecule (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally-occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the non-coding and coding acids, for example, phosphorothioate derivatives and nucleotides substituted with acridine can be used. Alternatively, the non-coding nucleic acid molecule can be produced biologically using an expression vector wherein a nucleic acid molecule has been subcloned in an antisense orientation (i.e., the RNA transcribed from the inserted nucleic acid molecule will be a non-coding orientation to a target nucleic acid of interest). The nucleic acid sequences can also be used to compare with the endogenous DNA sequences in patients to identify one or more of the FLAP-related disorders., and as probes, such as to hybridize and discover the related DNA sequences or to subtract known sequences from a sample. The nucleic acid sequences may also be used to derive primers for genetic fingerprinting, to raise anti-polypeptide antibodies using DNA immunization techniques, and as an antigen to raise anti-DNA antibodies or produce immune responses. The portions or fragments of the nucleotide sequences identified herein (and the corresponding complete genetic sequences) can be used in numerous ways as polynucleotide reagents. For example, these sequences can be used to: (i) map their respective genes on a chromosome; and, thereby locating genetic regions or nucleic acid regions associated with a genetic disease; (ii) identify an individual from a tiny biological sample (tissue classification); and (iii) assist in the forensic identification of a biological sample. Additionally, the nucleotide sequences of the invention can be used to identify and express recombinant polypeptides for analysis, characterization or therapeutic use, or as markers for tissues wherein the corresponding polypeptide is expressed, either constitutively, during tissue differentiation, or disease states. The nucleic acid sequences may additionally be used as reagents in the selection and / or diagnostic assays described herein, and may also be included as a component of equipment (eg, reagent kits) for use in screening and / or diagnostic assays. described in the present.
Vectors Another aspect of the invention relates to nucleic acid constructs containing a nucleic acid molecule of SEQ. FROM IDENT. NOS .: 1 or 3 or the complement of the same (or a portion thereof). Yet another aspect of the invention relates to nucleic acid constructs containing a nucleic acid molecule encoding an amino acid of SEQ. FROM IDENT. DO NOT. : 2 or a polymorphic variant thereof. The constructs comprise a vector (e.g., an expression vector) wherein a sequence of the invention has been inserted in a coding or non-coding orientation. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid" which refers to a loop of double-stranded DNA in which additional DNA segments can be bundled. Another type of vector is a viral vector, where additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (eg, bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell at the introduction of the host cell, and therefore replicate together with the host genome. In addition certain vectors, such as expression vectors, are capable of directing the expression of genes or nucleic acids to which they are operably linked. In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and associated adenoviruses) that perform equivalent functions. Preferred recombinant expression vectors of the invention comprise a nucleic acid molecule of the invention in a form suitable for expression of the nucleic acid molecule in a host cell. This means that the recombinant expression vectors include one or more regulatory sequences, selected at the base of the host cells that are used for expression, which is operably linked to the nucleic acid sequence that is expressed. Within a recombinant expression vector, "operably linked" or "operably linked" is intended to mean that the nucleic acid sequence of interest binds to the regulatory sequence (s) in a manner which allows the expression of the acid sequence nucleic acid (eg, in a transcription / translation system in vi tro or in a host cell when the vector is introduced into the host cell). The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, "Gene Expression Technology", Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those that direct the constitutive expression of a nucleic acid sequence in many types of host cell and those that direct the expression of the nucleic acid sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector may depend on such factors as the choice of the host cell to be transformed and the level of expression of the desired polypeptide. The expression vectors of the invention can be introduced into host cells which therefore produce polypeptides, including fusion polypeptides, encoded by nucleic acid molecules as described herein. The recombinant expression vectors of the invention can be designed for expression of a polypeptide of the invention in prokaryotic or eukaryotic cells, for example, bacterial cells such as E. coli, insect cells (using baculovirus expression vectors), yeast cells or mammalian cells. Suitable host cells are further discussed in Goeddel, supra.
Alternatively, the recombinant expression vector can be transcribed and translated in vi tro, for example using T7 promoter regulatory sequences and T7 polymerase. Another aspect of the invention relates to host cells in which a recombinant expression vector of the invention has been introduced. The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular target cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in subsequent generations due to any mutation or environmental influences, such progeny can not in fact be identical to the progenitor cell, but are still included within the scope of the terms as used herein. . A host cell can be any prokaryotic or eukaryotic cell. For example, a nucleic acid molecule of the invention can be expressed in bacterial cells (e.g., E. coli), insect cells, yeast or mammalian cells (such as Chinese hamster ovary (CHO) cells or cells). COS). Other suitable host cells are known to those skilled in the art. The vector DNA can be introduced into prokaryotic or eukaryotic cells through conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of techniques recognized by the art to introduce a foreign nucleic acid molecule (eg, DNA) into a host cell, including -precipitation of calcium phosphate or calcium chloride, transfection mediated by DEAE-dextran, lipofection or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (supra) and other laboratory manuals. For stable transfection in mammalian cells, it is known that, depending on the expression vector and the transfection technique used, only a small fraction of cells can integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene or nucleic acid encoding a selectable marker (eg, for resistance to antibiotics) is generally introduced into the host cells together with the gene or nucleic acid of interest. Preferred selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. The nucleic acid molecules encoding a selectable marker can be introduced into a host cell in the same vector as the nucleic acid molecule of the invention or can be introduced into a separate vector. Cells stably transfected with the introduced nucleic acid molecule can be identified by drug selection (for example, cells that have incorporated the selectable marker gene or nucleic acid will survive, while the other cells will die). A host cell of the invention, such as a prokaryotic host cell or eukaryotic host cell in culture, can be used to produce (ie, express) a polypeptide of the invention. Accordingly, the invention further provides methods for producing a polypeptide using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of the invention (into which a recombinant expression vector encoding a polypeptide of the invention has been introduced) in a suitable medium such that the polypeptide is produced. In another embodiment, the method further comprises isolating the polypeptide from the medium or host cell. The host cells of the invention can also be used to produce non-human transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell within which a nucleic acid molecule of the invention has been introduced (e.g., an exogenous FLAP nucleic acid, or an exogenous nucleic acid encoding a FLAP polypeptide). Such host cells can then be used to create transgenic non-human animals in which exogenous nucleotide sequences have been introduced into the genome or homologous recombinant animals in which the endogenous nucleotide sequences have been altered. Such animals are useful for studying the function and / or activity of the nucleic acid sequence and the polypeptide encoded by the sequence and for identifying and / or evaluating modulators of its activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or a mouse, wherein one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens and amphibians. A transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, so that the expression of a genetic product encoded in one is directed or more cell types or tissues of the transgenic animal. As used herein, a "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous gene has been altered by homologous recombination between the endogenous gene and a DNA molecule. exogenously introduced into an animal cell, for example, an embryonic cell of the animal, prior to the development of the animal. Methods for generating transgenic animals through embryonic manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866 and 4,870,009, U.S. Patent No. 4,873,191 and in Hogan, Manipulating the Mouse Embryo (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1986). Methods for constructing homologous recombination vectors and homologous recombinant animals are described in detail in Bradley, Current Opinion in BioTechnology 2: 823-829 (1991) and in PCT Publications Nos: WO 90/11354, WO 91/01140, WO 92/0968 and WO 93/04169. The clones of the non-human transgenic animals described herein may also be produced according to the methods described in Wilmut et al. , Nature 385: 810-813 (1997) and PCT Publications Nos. WO 97/07668 and WO 97/07669.
Polypeptides of the Invention The present invention also relates to isolated polypeptides encoded by FLAP nucleic acids ("FLAP polypeptides") and fragments and variants thereof, as well as polypeptides encoded by nucleotide sequences described herein (eg, other variants of splicing). The term "polypeptide" refers to a polymer of amino acids, and not to a specific length; thus, peptides, oligopeptides and proteins are included within the definition of a polypeptide. As used herein, a polypeptide will be "isolated" or "purified" when it is substantially free of cellular material, when it is isolated from recombinant and non-recombinant cells, or is free of chemical precursors or other chemicals when chemically synthesized. . A polypeptide, however, can bind to another polypeptide with which it is normally associated in a cell (e.g., in a "fusion protein") and still "isolate" or "purify". A detailed discussion of methods for making a polypeptide of the invention is provided in International Application No. PCT / US03 / 32556, filed October 16, 2003, which is incorporated herein by reference in its entirety.
Antibodies of the Invention Polyclonal and / or monoclonal antibodies that specifically bind a form of the polypeptide or nucleic acid product (e.g., a polypeptide encoded by a nucleic acid having a SNP as set forth in Table 13), but not a another form of the polypeptide or the nucleic acid product, are also provided. Antibodies are also provided which bind to a portion of any polypeptide encoded by nucleic acids of the invention (eg, SEQ ID NO: 1 or SEQ ID NO: 3, or complement of SEQ ID NO: SEQ ID NO: 3) or a polypeptide encoded by nucleic acids of the invention containing a polymorphic site or sites. The invention also provides antibodies to polypeptides and polypeptide fragments of the invention, or a portion thereof, or having an amino acid sequence encoded by a nucleic acid molecule comprising all or a portion of SEQ. FROM IDENT. NOS .: 1 or 3, or the complement thereof, or another variant or portion thereof. A detailed discussion of the methods for making the antibodies of the invention is provided in International Application No. PCT / US03 / 32556, filed on October 16, 2003, which is incorporated by reference in its entirety e? the present .
Diagnostic Assays The nucleic acids, probes, primers, polypeptides and antibodies described herein may be used in methods of diagnosing a susceptibility to MI, ACS, stroke or PAOD, or to another disease or condition associated with an MI gene, such as FLAP. , as well as in equipment useful for the diagnosis of a susceptibility to MI, ACS, stroke or PAOD, or to another disease or condition associated with FLAP. In one embodiment, the kit useful for diagnosis of susceptibility to MI, ACS stroke or PAOD, or to another disease or condition associated with FLAP comprises primers as described herein, wherein the primers contain one or more of the SNPs identified in Table 13. In one embodiment of the invention, the diagnosis of susceptibility to MI, ACS, apoplexy or PAOD (or diagnosis of susceptibility to another disease or condition associated with FLAP), is made by detecting a polymorphism in a FLAP nucleic acid as describes in the present. The polymorphism may be an alteration in a FLAP nucleic acid, such as the insertion or deletion of a single nucleotide, or of more than one nucleotide, resulting in an alteration of structure change; the change of at least one nucleotide, which results in a change in the encoded amino acid; the change of at least one nucleotide, which results in the generation of a premature stop codon; the elimination of several nucleotides; which results in a deletion of one or more amino acids encoded by the nucleotides; the insertion of one or more nucleotides, such as by unequal recombination or genetic conversion, resulting in an interruption of the coding sequence of the gene or nucleic acid; the duplication of all or a part of the gene or nucleic acid; the transposition of all or a part of the gene or nucleic acid; or the rearrangement of all or a part of the gene or nucleic acid. More than one alteration can occur in a single gene or nucleic acid. Such sequence changes cause an alteration in the polypeptide encoded by a FLAP nucleic acid. For example, if the alteration is an alteration of structure change, the structure change may result in a change in the encoded amino acids, and / or may result in the generation of a premature stop codon, causing the generation of a truncated polypeptide . Alternatively, a polymorphism associated with a disease or condition associated with a FLAP nucleic acid or a susceptibility to a disease or condition associated with a FLAP nucleic acid may be a synonymous alteration in one or more nucleotides (i.e., an alteration that does not result in a change in the polypeptide encoded by a FLAP nucleic acid). Such a polymorphism can alter splice sites, affect the stability or transport of mRNA, or otherwise affect the transcription or translation of the nucleic acid. A FLAP nucleic acid having any alteration described above is referred to herein as an "altered nucleic acid". In a first method for diagnosing a susceptibility to MI, ACS, apoplexy or PAOD, hybridization methods, such as Southern analysis, Northern analysis, or in situ hybridization can be used (see Current Protocols in Molecular Biology, Ausubel, F. et al. , eds., John Wileyn &Sons, including all supplements until 1999). For example, a biological sample from a test subject (a "test sample") of the genomic DNA, RNA or cDNA, is obtained from an individual suspected of having been susceptible to, predisposed to carry a defect for a susceptibility to a disease or condition associated with a FLAP nucleic acid (the "test subject"). The individual can be an adult, a child, or a fetus. The test sample can be from any source containing genomic DNA, such as a blood sample, the amniotic fluid sample, the cerebrospinal fluid sample, or the tissue sample from the skin, muscle or buccal mucosa or conjunctival, placenta, gastrointestinal tract or other organs. A DNA test sample from fetal or tissue cells can be obtained by appropriate methods, such as by amniocentesis or chorionic villus sampling. The DNA, RNA or cDNA sample is then examined to determine if a polymorphism in a MI nucleic acid is present, and / or to determine which splice variants or encoded by the FLAP are presented. The presence of the polymorphism or the splice variant (s) may be indicated by the hybridization of the nucleic acid in the DNA, RNA or genomic cDNA to a nucleic acid probe. A "nucleic acid probe" as used herein, may be a DNA probe or an RNA probe; the nucleic acid probe may contain at least one polymorphism in a FLAP nucleic acid or contains a nucleic acid encoding a particular splice variant of a FLAP nucleic acid. The probe can be any of the nucleic acid molecules described above (eg, the nucleic acid, fragment, a vector comprising the nucleic acid, a probe or a primer, etc.). To diagnose a susceptibility to MI, ACS, apoplexy or PAOD (or another disease or condition associated with FLAP), the test sample containing a FLAP nucleic acid is contacted with at least one nucleic acid probe to form a hybridization sample. A preferred probe for detecting mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing mRNA or genomic DNA sequences described herein. The nucleic acid probe can be for example, a full-length nucleic acid molecule, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to appropriate mRNA or DNA genomic For example, the nucleic acid probe can be all or a portion of one of the SECs. FROM IDENT. US. : 1 and 3, or the complement of it or a portion of it; or it can be a nucleic acid encoding all or a portion of one of SEQ. FROM IDENT. DO NOT . : 2. Other probes suitable for use in the diagnostic assays of the invention are described above (see, for example, probes and primers discussed under the title "Nucleic Acids of the Invention"). The hybridization sample is maintained under conditions that are sufficient to allow specific hybridization of the nucleic acid probe to a FLAP nucleic acid. "Specific hybridization" as shown herein, indicates the exact hybridization (eg, without incompatibilities). The specific hybridization can be carried out under severe stringent conditions or moderate stringent conditions, for example, as described above. In a particularly preferred embodiment, the hybridization conditions for specific hybridization are of high severity. The specific hybridization, if present, is then detected using standard methods. If the specific hybridization occurs between the nucleic acid probe and the FLAP nucleic acid in the test sample, then the FLAP has the polymorphism, or is the splice variant, that occurs in the nucleic acid probe. More than one nucleic acid probe can also be used at the same time in this method. The specific hybridization of any of the nucleic acid probes is indicative of a polymorphism in the FLAP nucleic acid, or the presence of a particular splice variant encoding the FLAP nucleic acid, and is therefore diagnostic for a susceptibility to disease or a condition associated with FLAP (for example, MI, ACS, stroke, or PAOD). In Northern analysis (see Current Protocols in Molecular Biology, Ausubel, F. et al., Eds., John Wiley &Sons, supra) the hybridization methods described above are used to identify the presence of a polymorphism or a variant of particular splice, associated with a susceptibility to a disease or condition associated with FLAP (eg, MI, ACS, stroke, or PAOD). For Northern analysis, an RNA test sample is obtained from the individual by appropriate means. The specific hybridization of a nucleic acid probe, as described above, to RNA from the individual is indicative of a polymorphism in a FLAP nucleic acid, or the presence of a particular splice variant, encoded by a FLAP nucleic acid, and is therefore diagnostic for susceptibility to a disease or condition associated with FLAP (e.g., MI, ACS, stroke or PAOD). For representative examples of the use of nucleic acid probes, see for example, U.S. Patent Nos. 5,288,611 and 4,851,330. Alternatively, a peptide nucleic acid (PNA) probe can be used in place of a nucleic acid probe in the hybridization methods described above. PNA is an imitation of DNA having an inorganic structure similar to peptide, such as units of N- (2-aminoethyl) glycine with an organic base (A, G, C, T or U) attached to the glycine nitrogen through a methylenecarbonyl linker (see for example, Nielsen, PE et al., Bioconjugate Chemistry 5, American Chemical Society, p. 1 (1994). The PNA probe can be designed to hybridize specifically to a nucleic acid having a polymorphism associated with a susceptibility to a disease or condition associated with FLAP (e.g., MI). Hybridization of the PNA probe to a FLAP nucleic acid as described herein is diagnostic of the susceptibility to the disease or condition. In another method of the invention, mutation analysis by restriction digestion can be used to detect an altered nucleic acid, or nucleic acids containing the polymorphism (s), if the mutation or polymorphism in the nucleic acid results in the creation or elimination of a restriction site. A test sample containing genomic DNA is obtained from the individual.
The polymerase chain reaction (PCR) can be used to amplify a FLAP nucleic acid (and, if necessary, flanking sequences) in the genomic DNA test sample from the test individual. The RFLP analysis is conducted as described (see Current Protocols in Molecular Biology, supra). The digestion pattern of the relevant DNA fragment indicates the presence or absence of the alteration or polymorphism in the FLAP nucleic acid, and therefore indicates the presence or absence of susceptibility to a disease or condition associated with FLAP (e.g. MI, ACS, apoplexy or PAOD) Sequence analysis can also be used to detect specific polymorphisms in the FLAP nucleic acid. A test sample of DNA or RNA is obtained from the test individual. PCR or other appropriate methods can be used to amplify the nucleic acid, and / or its flanking sequences, if desired. The sequence of a FLAP nucleic acid, or a fragment of the nucleic acid, or cDNA, or fragment of the cDNA, or mRNA, or fragment of the mRNA, is determined using standard methods. The nucleic acid sequence, nucleic acid fragment, cDNA, cDNA fragment, mRNA, or mRNA fragment is compared to the known nucleic acid sequence of the nucleic acid, cDNA (e.g., one or more of SEQ ID. NOS .: 1 or 3, and / or the complement of SEQ ID NO: 1 or 3), or the nucleic acid sequence encoding SEC. FROM IDENT. DO NOT. : 2 or a fragment thereof) or mRNA, as appropriate. The presence of a polymorphism in FLAP indicates that the individual has a susceptibility to a disease associated with FLAP (eg, MI, ACS, stroke, or PAOD). Allele-specific oligonucleotides can be used to detect the presence of polymorphisms or polymorphisms in the FLAP nucleic acid, through the use of spot blot hybridization of oligonucleotides amplified with allele-specific oligonucleotide (ASO) probes (see for example, Saiki , R. et al., Nature 324: 163-166 (1986)). An "allele-specific oligonucleotide" (also referred to herein as an "allele-specific oligonucleotide probe") is an oligonucleotide of about 10-50 base pairs, eg, about 15-30 base pairs, which specifically hybridize to a FLAP nucleic acid, and containing a polymorphism associated with a susceptibility to a disease or condition associated with FLAP (e.g., MI, ACS, stroke, or PAOD). An allele-specific oligonucleotide probe that is specific for particular polymorphisms in a FLAP nucleic acid can be prepared, using standard methods (see Current Protocols in Molecular Biology, supra). To identify polymorphisms in the nucleic acid associated with susceptibility to the disease, a DNA test sample is obtained from the individual. The PCR can be used to amplify all or a fragment of a FLAP nucleic acid, and its flanking sequences. The DNA containing the amplified FLAP nucleic acid (or fragment 'of the nucleic acid) is spotted with spot, using standard methods (see Current Protocols in Molecular Biology, supra) and the point is contacted with the oligonucleotide probe. The presence of specific hybridization of the probe to the amplified FLAP is then detected. The specific hybridization of an allele-specific oligonucleotide probe to DNA from the individual is indicative of a polymorphism in the FLAP, and is therefore indicative of a susceptibility to a disease or condition associated with FLAP (e.g., MI, ACS , apoplexy or PAOD). A specific allele primer hybridizes to a target site or DNA that overlaps a polymorphism and only primes the amplification in an allelic manner to which the primer exhibits perfect complementarity. See Gibbs, Nucleic Acids Res. 17, 2427-2448 (1989). This priming is used together with a second primer which hybridizes at a distal site. The amplification proceeds from two primers, resulting in a detectable product which indicates that the particular allelic form is presented. A control is usually performed with a second pair of primers, one of which shows a single base incompatibility at the polymorphic site and the other of which exhibits perfect complementarity to a distal site. The base-single incompatibility prevents amplification and the detectable product is not formed. The working method improves when the incompatibility is included in the majority of the 3 'position of the oligonucleotide aligned with the polymorphism because this position is more destabilizing for elongation from the primer (see for example, WO 93/22456). With the addition of such analogs as blocked nucleic acids (the LNAs), the size of primers and probes can be reduced as little as 8 bases. LNAs are a novel class of bicyclic DNA analogs in which the 2 'and 4' positions in the furanose ring are bound through an O-methylene (oxy-LNA), S-methylene (thio-LNA), or a portion of amino methylene (amino-LNA). Common to all these variants LAN is an affinity towards the complementarity of nucleic acids, which is by far the most highly reported by an analogue DNA. For example, all the particular oxy-LNA nonamers have been shown to have melting temperatures of 64 ° C and 74 ° C when in complex the complementarity of DNA or RNA, respectively, as opposed to 28 ° C for DNA and RNA for the nonamer of Corresponding DNA Substantial increases in Tm are also obtained when the LNA monomers are used in combination with DNA or RNA monomers. For primers and probes, depending on where the LNA monomers are included (for example, the 3 'end, the 5' end or in the center), the Tm could be increased considerably. In another embodiment, the arrangements of the oligonucleotide probes that are complementary to the target nucleic acid sequence segments from an individual can be used to identify polymorphisms in a FLAP nucleic acid. For example, in one embodiment, an oligonucleotide array can be used. Oligonucleotide arrays typically comprise a plurality of different oligonucleotide probes that attach to a surface of a substrate at different known locations. These oligonucleotide arrays also described as "Genechips ™" have been generally described in the art, for example, U.S. Patent No. 5,143,854 and PCT Patent Publication Nos. WO 90/15070 and WO 92/10092. These arrangements can generally be produced using mechanical synthesis methods or light-directed synthesis methods incorporating a combination of photolithographic methods and solid-phase oligonucleotide synthesis methods. See Fodor et al. , Science 251: 767-777 (1991); Pirrung, et al. , U.S. Patent 5,143,854; (see also PCT Application WO 90/15070); Fodor et al. , PCT Application WO 92/10092; and U.S. Patent No. 5,424,186, the complete teachings for each of which is incorporated herein by reference. Techniques for the synthesis of these arrangements using mechanical synthesis methods are described in, for example, US Pat. No. 5,384,261, the complete teachings which are incorporated herein by reference. In another example, the linear arrangements can be used. Once an oligonucleotide array is prepared, a nucleic acid of interest is hybridized to the array and scavenged for polymorphisms. Hybridization and scanning are generally carried out by methods described herein and also in, for example, PCT Applications Nos. WO 92/10092 and WO 95/11995, and US Patent No. 5,424,186, the complete teachings which are incorporated for reference herein. In brief, a nucleic acid sequence that includes one or more previously identified polymorphic markers is amplified using well-known amplification techniques, eg, PCR. Typically, this involves the use of primer sequences that are complementary to the two strands of the target sequence both upstream and downstream from the polymorphism. Asymmetric PCR techniques can also be used. The amplified target, which generally incorporates a tag, is then hybridized to the arrangement under appropriate conditions. At the termination of hybridization and washing of the arrangement, the array is scanned to determine the position at which the target sequence is hybridized. The hybridization data obtained from the sweep are usually in the form of fluorescence intensities as a function of location in the array. In an inverse method, a probe, which contains a polymorphism, can be applied to a solid surface and PCR amplicons are then added to hybridize to these probes. Although described primarily in terms of a single detection block, for example, the detection of single polymorphism arrangements may include multiple detection blocks, and thus is capable of analyzing multiple specific polymorphisms. It will be generally understood that the detection blocks can be grouped within a single arrangement or in multiple separate arrangements so that variable optimal conditions can be used during the hybridization of the target to the arrangement. For example, it may often be desired to provide detection of those polymorphisms that fall within G-C-rich stretches of a genomic sequence, separately from those that fall into segments rich in A-T. This allows the separate optimization of hybridization conditions for each situation.
Additional uses of oligonucleotide arrays for the detection of polymorphisms can be found, for example, in U.S. Patent Nos. 5,858,659 and 5,837,832, the entire teachings which are incorporated herein by reference. Other methods of nucleic acid analysis can be used to detect polymorphisms in a nucleic acid described herein, or variants encoded by a nucleic acid described herein. Representative methods include direct manual sequencing (Church and Gilber, Proc. Nati, Acad. Sci. USA 81: 1991-1995 (1988); Sanger, F. et al., Proc. Nati. Acad. Sci. USA 74: 5463 -5467 (1977); Beavis et al., US Patent No. 5,288,644); automated fluorescent sequencing; Single-strand conformation polymorphism assays (SSCP); denaturing gel electrophoresis (CDGE); denaturing gradient gel electrophoresis (DGGE) (Sheffield, VC et al., Proc. Nati, Acad. Sci. USA 86: 232-236 (1989)), mobility change analysis (Orita, M. et al., Proc. Nati, Acad. Sci. USA 86: 2766-2770 (1989)), restriction enzyme analysis (Flavell et al., Cell 15:25 (1978)); Geever et al. , Proc.
Nati Acad. Sci. USA 78: 5081 (1981)); heteroduplex analysis; splitting of chemical incompatibility (CMC) (Cotton et al., Proc. Nati, Acad. Sci. USA 85: 4397-4401 (1985)); RNase protection assays (Myers, R.M. et al., Science 230: 1242 (1985)); use of polypeptides which recognize nucleotide incompatibilities, such as mutS protein E. coli; Allele-specific PCR, for example. In one embodiment of the invention, the diagnosis of a susceptibility to a disease or condition associated with FLAP (for example, MI, ACS, stroke, or PAOD) can also be done by quantitative PCR expression analysis (kinetic thermal cyclization). This technique that uses TaqMan® can be used to allow the identification of polymorphisms and if a patient is homozygous or heterozygous. The technique can evaluate the presence of an alteration in the expression or composition of the polypeptide encoded by a FLAP nucleic acid or splice variants encoded by a FLAP nucleic acid. In addition, the expression of the variants can be quantified as physically or functionally different. In another embodiment of the invention, the diagnosis of a susceptibility to MI, ACS, apoplexy or PAOD (or of another disease or condition associated with FLAP) can also be used by examining the expression and / or composition of a FLAP polypeptide by a variety of methods, including enzyme-linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. A test sample from an individual is evaluated for the presence of an alteration in the expression and / or an alteration in composition of the polypeptide encoded by a FLAP nucleic acid, or for the presence of a particular variant encoded by a FLAP nucleic acid . An alteration in expression of a polypeptide encoded by a FLAP nucleic acid can be, for example, an alteration in the quantitative polypeptide expression (i.e., the amount of polypeptide produced); an alteration in the composition of a polypeptide encoded by a FLAP nucleic acid is an alteration in the qualitative polypeptide expression (for example, the expression of an altered FLAP polypeptide or a different splicing variant). In a preferred embodiment, the diagnosis of a susceptibility to a disease or condition associated with FLAP is made by detecting a particular splice variant encoded by that FLAP variant, or a particular pattern of splice variants. Both alterations (quantitative and qualitative) can also be presented. An "alteration" in the expression or polypeptide composition, refers to an alteration in the expression or composition in a test sample, as compared to the expression or composition of polypeptide by FLAP nucleic acid in a control sample. A control sample is a sample that corresponds to a test sample (for example, it is from the same cell type) and it is from an individual who is not affected by the disease or a susceptibility to a disease or condition associated with an acid nucleic FLAP. An alteration in the expression or composition of the polypeptide in the test sample, as compared to the control sample, is indicative of a susceptibility to a disease or a condition associated with FLAP (e.g., MI, ACS-, stroke or PAOD) ). Similarly, the presence of one or more different splice variants in the test sample, or the presence of significantly different amounts of different splice variants in the test sample, as compared to the control sample, is indicative of a susceptibility to a disease or condition associated with a FLAP nucleic acid. Various means for examining the expression or composition of the polypeptide encoded by a FLAP nucleic acid can be used, including: spectroscopy, colorimetry, electrophoresis, isoelectric concentration and immunoassays (e.g., David et al., U.S. Patent 4,376,110) such as immunoblotting (see also Current Protpcols in Molecular Biology, particularly chapter 10). For example, in one embodiment, an antibody capable of binding to the polypeptide (e.g., as described above), preferably an antibody with a detectable label, can be used. The antibodies can be polyclonal, or more preferably monoclonal. An intact antibody, or a fragment thereof (eg, Fab or F (ab ') 2 may be used.The term "labeling", with respect to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically joining) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and a final labeling of a biotin DNA probe so that it can be detected with fluorescently labeled streptavidin.The Western blot analysis, using an antibody as described above, which specifically binds to a polypeptide encoded by an altered FLAP (for example, a FLAP that has an SNP as shown in Table 13), or an antibody Because it binds specifically to a polypeptide encoded by a non-altered nucleic acid, or an antibody that specifically binds to a particular splice variant encoded by a nucleic acid, it can be used to identify the presence of a test sample of a variant of particular splice or of a polypeptide encoded by a polymorphic or altered FLAP, or the absence of a test sample of a particular splice variant or of a polypeptide encoded by a non-polymorphic or non-altered nucleic acid. The presence of a polypeptide encoded by a polymorphic or altered nucleic acid, or the absence of a polypeptide encoded by a non-polymorphic or non-altered nucleic acid, is diagnostic for a susceptibility to a disease or condition associated with FLAP, such as the presence ( or absence) of particular splice variants encoded by the FLAP nucleic acid. In one embodiment of this method, the level or amount of polypeptide encoded by a FLAP nucleic acid in a test sample is compared to the level or amount of the polypeptide encoded by FLAP in a control sample. A level or amount of the polypeptide in the test sample that is higher or lower than the level or amount of the polypeptide in the control sample, so that the difference is statistically significant, is indicative of an alteration in the expression of the polypeptide encoded by the FLAP, and is diagnostic for a susceptibility to a disease or a condition associated with that FLAP. Alternatively, the composition of the polypeptide encoded by a FLAP nucleic acid in a test sample is compared to the composition of the polypeptide encoded by FLAP in a control sample. (for example, the presence of different splice variants). A difference in the composition of the polypeptide in the test sample, as compared to the composition of the polypeptide in the control sample, is diagnostic for a susceptibility to a disease or condition associated with that FLAP. In another embodiment, both the level or the amount and the composition of the polypeptide can be evaluated in the test sample and in the control sample. A difference in the amount or level of the polypeptide in the test sample, compared to the control sample; a difference in composition in the test sample, compared to the control sample; or both a difference in the amount or a level, and a difference in composition, is indicative of a susceptibility to a disease or condition associated with FLAP (e.g., MI). The invention also relates to a method for the diagnosis and identification of susceptibility to myocardial infarction, ACS, apoplexy or PAOD in an individual, by identifying a haplotype at risk in FLAP. In one modality, the haplotype at risk is one which confers a significant risk of MI, ACS, apoplexy or PAOD. In one embodiment, the importance associated with a haplotype is measured by a probability ratio. In an additional modality, importance is measured by a percentage. In one modality, a significant risk is measured as a probability ratio of at least 1.2, including but not limited to: 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, and 1.9. In an additional mode, a probability ratio of at least 1.2 is significant. In a further embodiment, a probability ratio of at least about 1.5 is significant. In an additional mode, a significant increase in risk is at least approximately 1.7 is significant. In a further embodiment, a significant increase in risk is at least about 20%, including but not limited to about 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% and 98%. In a further embodiment, a significant increase in risk of at least about 50%. In yet another modality, a haplotype at risk has a value p < 0.05. It is understood, however, that identifying whether a risk is medically significant may also depend on a variety of factors, including the specific disease, the haplotype, and often environmental factors. The invention also relates to methods for diagnosing a susceptibility to myocardial infarction, ACS, stroke or PAOD in an individual, comprising selecting an at-risk haplotype in the FLAP nucleic acid that occurs most frequently in an individual susceptible to myocardial infarction. (affected), compared to the frequency of its presence in a healthy individual (control), where the presence of the haplotype is indicative of susceptibility to myocardial infarction. Standard techniques for classifying the genotype for the presence of SNP and / or microsatellite markers that are associated with myocardial infarction, ACS, stroke or PAOD can be used, such as fluorescent-based techniques (Chen, et al., Genome Res. , 492 (1999), PCR, LCR, Nested PCR and other techniques for nucleic acid amplification In a preferred embodiment, the method comprises evaluating in an individual the presence or frequency of SNPs and / or microsatellites in the FLAP nucleic acid. which are associated with myocardial infarction, ACS, apoplexy or PAOD, where a higher excess or frequency of SNPs and / or microsatellites compared to a healthy control individual is indicative that the individual is susceptible to myocardial infarction, ACS, apoplexy or PAOD See Table 7 for SNPs, which includes haplotypes that can be used as selection tools See also Table 13 that establishes SNPs and markers for use as tools In one embodiment, the at-risk haplotype is characterized by the presence of polymorphism (s) represented in Table 13. For example, SG13S99, wherein the SNP may be a "C" or a "T"; SG13S25, where the SNP can be a "G" or an "A"; SG13S377 wherein the SNP can be a "G" or an "A" / SG13S106, wherein the SNP can be a "G" or an "A"; SG13S114, wherein the SNP may be a "T" or an "A"; SG13S89, wherein the SNP can be a "G" or an "A"; SG13S30, wherein the SNP can be a "G" or a "T"; SG13S32, wherein the SNP may be a "C" or an "A"; SG13S42, wherein the SNP may be a "G" or an "A"; and SG13S35, where the SNP can be a "G" or an "A". In addition, SG13A375, wherein the SNP can be a "T", SG13S32, wherein the SNP can be an "A", and SG13S106, wherein the SNP can be a "G" or an "A". Equipment (e.g., reactive kits) useful in the diagnostic methods comprises useful components in any of the methods described herein, including for example, the waves and the hybridization primers as described herein (e.g., probes or tagged primers), reagents for detection of tagged molecules, restriction enzymes (e.g., for RFLP analysis), allele-specific oligonucleotides, antibodies which bind to altered or unaltered FLAP polypeptide (native), media for nucleic acid amplification comprising a FLAP, or means for analyzing the nucleic acid sequence of a nucleic acid described herein, or for analyzing the amino acid sequence of a polypeptide as described herein, etc. In one embodiment, a kit for diagnosing susceptibility to MI, ACS, stroke or PAOD may comprise primers for nucleic acid amplification of a region in the FLAP nucleic acid comprising an at-risk haplotype that is more frequently present in an individual who has MI. , ACS, apoplexy or PAOD or susceptible to MI, ACS, stroke or PAOD. The primers can be designed using portions of the nucleic acids flanking the SNPs that are indicative of MI. In a particularly preferred embodiment, the primers are designed to amplify regions of the FLAP nucleic acid associated with a haplotype at risk for MI, ACS, apoplexy or PAOD, as shown in Table 7, or more particularly the haplotype defined by the following markers. of SNP: In one modality, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S99, SG13S25, SG13S377, SG13S106, SG13S32 and SG13S35, at the 13ql2-13 site. In a particular modality, the presence of the T, G, G, G, A and G alleles in SG13S99, SG13S25, SG13S377, SG13S106, SG13S32 and SG13S35, respectively (the B6 haplotype), is diagnostic of susceptibility to myocardial infarction, ACS, apoplexy or PAOD. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S99, SG13S25, SG13S106, SG13S30 and SG13S42 at site 13ql2-13. In a particular modality, the presence of the T, G, G, G and A alleles in SG13S99, SG13S25, SG13S106, SG13S30 and SG13S42, respectively (the B5 haplotype), is diagnosis of susceptibility to myocardial infarction, ACS, apoplexy or PAOD. In a third modality, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S106, SG13S30 and SG13S42 at site 13ql2-13. In a particular embodiment, the presence of alleles G, G, G and A in SG13S25, SG13S106, SG13S30 and SG13S42, respectively (the B4 haplotype), is diagnostic of susceptibility to myocardial infarction, ACS, stroke or PAOD. In a fourth embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S99, SG13S25, SG13S114, SG13S89 and SG13S32 at site 13ql2-13. In a particular modality, the presence of the T, G, T, G and A alleles in SG13S99, SG13S25, SG13S114, SG13S89 and SG13S32, respectively (the A5 haplotype), is diagnostic of susceptibility to myocardial infarction, ACS, apoplexy or PAOD. In a fifth modality, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S114, SG13S89 and SG13S32 at site 13ql2-12. In a particular embodiment, the presence of alleles G, T, G and A in SG13S25, SG13S114, SG13S89 and SG13S32, respectively (the A4 haplotype), is diagnosis of susceptibility to myocardial infarction, ACS, stroke or PAOD. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises the marker SG13S375 at the 13ql2-13 site. In a particular modality, the presence of T in SG13S375 (the HapCl haplotype) is diagnostic of susceptibility to myocardial infarction, ACS, apoplexy or PAOD. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25 and SG13S375 at the 13ql2-13 site. In a particular embodiment, the presence of T and G in SG13S375 and SG13S25, respectively (the HapC2 haplotype) is diagnostic of susceptibility to myocardial infarction, ACS, apoplexy or PAOD. In another embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS, apoplexy or PAOD comprises markers SG13S25, SG13S375 and SG13S32 at the 13ql2-13 site. In a particular modality, the presence of T, G and A in Sgl3S375, SG13S25 and SG13S32, respectively (HapC3 haplotype), is diagnostic of susceptibility to myocardial infarction, ACS, apoplexy or PAOD. In a further embodiment, a haplotype associated with a susceptibility to myocardial infarction, ACS-, apoplexy or PAOD comprises markers SG13S25, SG13S375, SG13S32 and SG13S106 at the 13ql2-13 site. In a particular embodiment, the presence of T, G, A and G in SG13S375, SG13S25, SG13S32 and SG13S106, respectively (HapC4-A haplotype) is diagnostic of susceptibility to myocardial infarction, ACS, apoplexy or PAOD. In a particular embodiment, the presence of T, G, A and A in SG13S375, SG13S25, SG13S32 and SG13S106, respectively (HapC4-B haplotype) is diagnostic of susceptibility to myocardial infarction, ACS, apoplexy or PAOD.
Selection Assays and Agents Identified Thereby The invention provides methods (also referred to herein as "screening assays") to identify the presence of a nucleotide that hybridizes to a nucleic acid of the invention, as well as to identify the presence of a polypeptide encoded by a nucleic acid of the invention. In one embodiment, the presence (or absence) of a nucleic acid molecule of interest (eg, a nucleic acid having significant homology to a nucleic acid of the invention) in a sample can be evaluated by contacting the sample with an acid nucleic acid comprising a nucleic acid of the invention (eg, a nucleic acid having the sequence of one of SEQ ID NOS: 1 or 3 or the complement thereof, or a nucleic acid encoding an amino acid) having the sequence of SEQ ID NO: 2, or a fragment or a variant of such nucleic acids), under stringent conditions as described above, and then evaluating the sample for the presence (or absence) of hybridization . In a preferred embodiment, high severity conditions are suitable conditions for selective hybridization. In another embodiment, a sample containing a nucleic acid molecule of interest is contacted with a nucleic acid containing a contiguous nucleic acid sequence (e.g., a primer or a probe as described above) that is at least partially complementary to a part of the nucleic acid molecule of interest (eg, a FLAP nucleic acid), and the contacted sample is evaluated for the presence or absence of hybridization. In a preferred embodiment, the nucleic acid containing a contiguous nucleic acid sequence is completely complementary from the nucleic acid molecule of interest. In any of these embodiments, all or a portion of the nucleic acid of interest may be subjected to prior amplification to perform hybridization. In another embodiment, the presence (or absence) of a polypeptide of interest, such as a polypeptide of the invention or a fragment or variant thereof, in a sample can be evaluated by contacting the sample with an antibody that specifically hybridizes to the polypeptide of interest (eg, an antibody such as those described above), and then evaluating the sample for the presence (or absence) of binding of the antibody to the polypeptide of interest. In another embodiment, the invention provides methods for identifying agents (e.g., fusion proteins, polypeptides, peptidomimetics, prodrugs, receptors, binding agents, antibodies, small molecules or other drugs or ribozymes that alter (e.g., increase or decrease) the activity of the polypeptides described herein, or which otherwise interact with the polypeptides herein, For example, such agents may be agents that bind to polypeptides described herein (e.g., binding agent for members of the leukotriene pathway, such as FLAP binding agents), which have a stimulatory or inhibitory effect on, for example, the activity of polypeptides of the invention, or which change (e.g., improve or inhibit) the ability of the polypeptides of the invention to interact with members of the leukotriene path binding agents (e.g. other binding agents); or which alter the post-translational processing of the leukotriene pathway member polypeptide, such as a FLAP polypeptide (e.g., agents that alter proteolytic processing to direct the polypeptide from where it is normally synthesized to another location in the cell, such as the cell surface, agents that alter proteolytic processing so that more polypeptide is released from the cell, etc.). In one embodiment, the invention provides assays for screening candidate or test agents that bind to or modulate the activity of polypeptides described herein (one or biologically active portions thereof) as well as agents identifiable by the assays. The test agents can be obtained using any of the numerous approaches in the combinatorial library methods known in the art, including: biological libraries; spatially steerable parallel phase or solid phase solution phase libraries; synthetic library methods that require descircunvolution; the "one pearl one compound" library method; and synthetic library methods that utilize the affinity chromatography selection. The biological library approach is limited to polypeptide libraries, while the other four approaches are applicable to polypeptide, oligomer without peptide or libraries of small molecules of compounds (Lam, K.S., Anticancer Drug Des. 12: 145 (1997)). In one embodiment, to identify agents which alter the activity of a FLAP polypeptide, a cell, a cell lysate, or solution containing or expressing a FLAP polypeptide (e.g., SEQ ID NO: 2 or other variant) of splicing encoded by a nucleic acid FLAP, such as a nucleic acid comprising a SNP as shown in Table 13), or a fragment or derivative thereof (As described above), you can contact an agent to test yourself; alternatively, the polypeptide can be contacted directly with the agent being tested. The level (amount) or activity of FLAP is evaluated (for example, the level (amount) of the FLAP activity is measured, either directly or indirectly), and compared to the level of activity in a control (ie, the level of activity of the FLAP polypeptide or active fragment or derivative thereof in the absence of the agent being tested). If the level of activity in the presence of the agent differs, by an amount that is statistically significant, from the level of the activity in the absence of the agent, then the agent is an agent that alters the activity of a FLAP polypeptide. An increase in the level of FLAP activity in the presence of the agent relative to activity in the absence of the agent indicates that the agent is an agent that improves FLAP activity. Similarly, a decrease in the level of FLAP activity in the presence of the agent, relative to activity in the absence of the agent, indicates that the agent is an agent that inhibits FLAP activity. In another embodiment, the level of activity of a FLAP polypeptide or derivative or fragment thereof in the presence of the agent being tested, is compared to a control level that has been previously established. A statistically significant difference in the level of activity in the presence of the agent from the control level indicates that the agent alters FLAP activity. The present invention also relates to an assay for identifying agents which alter the expression of a FLAP nucleic acid (eg, non-coding nucleic acids, fusion proteins, polypeptides, peptide mimetics, pro-drugs, receptors, binding agents, antibodies, small molecules or other drugs or ribozymes, which alter (e.g., increase or decrease) the expression (e.g., transcription or translation) of the nucleic acid or which otherwise interact with the nucleic acids described herein, as well as agents identifiable by the assays For example, a solution containing a nucleic acid encoding a FLAP polypeptide (eg, a FLAP nucleic acid) may be contacted with an agent being tested.The solution may comprise, for example , cells that contain the nucleic acid or a cellular lysate that contains the nucleic acid, alternatively, the solution can be another solution comprising elements necessary for the transcription / translation of the nucleic acid. Cells not suspended in solution can also be used, if desired. The level and / or pattern of the FLAP expression (e.g., the level and / or mRNA pattern or protein expressed, such as the level and / or pattern of different splice variants) is evaluated, and compared with the level and / or expression pattern in a control (ie, the level and / or pattern of the FLAP expression in the absence of the agent being tested).
If the level and / or pattern in the presence of the agent differ, by an amount or in a manner that is statistically significant, from the level and / or the pattern in the absence of the agent, then the agent is an agent that alters the expression of the FLAP nucleic acid. The improvement of FLAP expression indicates that the agent is an activator of FLAP activity. Similarly, the inhibition of FLAP expression indicates that the agent is a repressor of FLAP activity. In another embodiment, the level and / or pattern of the FLAP polypeptide (s) (eg, different splice variants) in the presence of the agent being tested is compared to a control level and / or the pattern that has been previously established . A level and / or pattern in the presence of the agent that differs from the level and / or the control pattern by an amount or in a manner that statistically indicates that the agent alters the FLAP expression. In another embodiment of the invention, agents that alter the expression of a FLAP nucleic acid or which otherwise interact with the nucleic acids described herein, can be identified using a cell, a cellular lysate, or a solution containing a nucleic acid encoding the region promoter of FLAP nucleic acid operably linked to a reporter gene. After contacting an agent to be tested, the level of expression of the reporter gene (e.g., the level of mRNA or protein expressed) is evaluated, and compared to the level of expression in a control (i.e. level of expression of the reporter gene in the absence of the agent being tested). If the level in the presence of the agent differs, by an amount or in a manner that is statistically significant, from the level in the absence of the agent, then the agent is an agent that alters the expression of the FLAP nucleic acid, as indicated for its ability to alter the expression of a nucleic acid that is operably linked to the FLAP nucleic acid promoter. The improvement of the reporter's expression indicates that the agent is an activator of the FLAP expression. Similarly, inhibition of reporter expression indicates that the agent is a repressor of FLAP expression. In another embodiment, the level of expression of the reporter in the presence of the test agent is compared to the control level that has been previously established. A level in the presence of the agent that differs from the control level by an amount or in a manner that is statistically significant indicates that the agent alters the expression. The agents which alter the amounts of different splice variants encoded by a FLAP nucleic acid (for example, an agent which improves the expression of a first splicing variant, and which inhibits the expression of a second splice variant), as well as agents which stimulate the activity of a first splicing variant and inhibit the activity of a second splice variant, can be easily identified using these methods described above. In other embodiments of the invention, the assays can be used to evaluate the impact of a test agent on the activity of a polypeptide relative to a FLAP binding agent. For example, a cell that expresses a compound that interacts with a FLAP nucleic acid (herein referred to as a "FLAP binding agent", which may be a polypeptide or another molecule that interacts with a FLAP nucleic acid, such as a receptor, or another molecule, such as 5-LO) is contacted with a FLAP in the presence of a test agent, and the ability of the test agent to alter the interaction between the FLAP and the FLAP binding agent is determined . Alternatively, a cell lysate or a solution containing the FLAP binding agent can be used. An agent which binds to the FLAP or the FLAP binding agent may alter the interaction by interfering with, or improving the ability of the FLAP to bind to, associate with, or otherwise interact with the FLAP binding agent. Determining the ability of the test agent to bind to a FLAP nucleic acid or a FLAP nucleic acid binding agent can be achieved, for example, by coupling the test agent with a radioisotope or enzyme tag so that the binding of the test agent the polypeptide can be determined by detecting the labeling with 125I, 3SS, 14C or 3H, either directly or indirectly, and the radioisotope detected by the direct counting of radioemission or by scintillation counting. Alternatively, the test agents can be labeled enzymatically with, for example, horseradish peroxidase, alkaline phosphatase or luciferase, and the enzymatic label detected by the determination of conversion of an appropriate substrate to the product. It is also within the scope of the invention of this invention to determine the ability of a test agent to interact with the polypeptide without the labeling of any of the interactive agents. For example, a microphysiometer can be used to detect the interaction of a test agent with a FLAP or a FLAP binding agent without the labeling of any test agent, FLAP or FLAP binding agent, McConnell, H.M et al. , Science 257: 1906-1912 (1992). As used herein, a "microphysiometer" (e.g., Cytosensor ™) is an analytical instrument that measures the rate at which a cell acidifies its environment using a light-sensitive potentiometric sensor (LAPS). Changes in this rate of acidification can be used as an indicator of the interaction between the ligand and the polypeptide. Thus, these receptors can be used to select compounds that are agonists for use in the treatment of a disease or condition associated with FLAP or a susceptibility to a disease or condition associated with FLAP, or antagonists to study a susceptibility to a disease or condition. a condition associated with FLAP (for example, MI, ACS, stroke, or PAOD). The drugs can be designed to regulate FLAP activation, which in turn can be used to regulate the signaling pathways and transcription events of downstream genes or of proteins or polypeptides that interact with FLAP (eg, 5-LO). In another embodiment of the invention, assays can be used to identify polypeptides that interact with one or more FLAP polypeptides, as described herein. For example, a yeast two-hybrid system such as that described by Fields and Song (Fields, S. and Song, 0., Nature 340: 245-246 (1989)) can be used to identify polypeptides that interact with one or more FLAP polypeptides. In such a two-hybrid yeast system, vectors are constructed based on the flexibility of a transcription factor having two functional domains (a DNA binding domain and a transcription activation domain). If the two domains separate, but combine to two different proteins that interact with each other, transcriptional activation can be achieved, and transcription of specific markers (eg, nutritional markers such as His and Ade, or color markers such as lacZ) can be used to identify the presence of interaction and transcriptional activation. For example, in the methods of the invention, a first vector is used which includes a nucleic acid encoding a DNA binding domain and also a FLAP polypeptide, a splice variant or fragment or derivative thereof, and a second vector which includes a nucleic acid encoding a transcription activation domain and also a nucleic acid encoding a polypeptide which can potentially interact with the FLAP polypeptide, splice variant or fragment or derivative thereof (eg, a FLAP polypeptide binding agent or receptor). Incubation of the yeast containing the first vector and the second vector under appropriate conditions (for example, coupling conditions such as that used in the Matchmaker ™ system from Clontech (Palo Alto, California, USA)) allows the identification of colonies that express the markers of interest. These colonies can be examined to identify the polypeptide (s) that interact with the FLAP polypeptide or fragment or derivative thereof. Such polypeptides may be useful as agents that alter the expression activity of a FLAP polypeptide as described above. In more than one embodiment of the above test methods of the present invention, it may be desirable to immobilize either the FLAP, the FLAP binding agent or other test components on a solid support, in order to facilitate the separation of complex shapes from non-complexes of one or both of the polypeptides, as well as to accommodate the automation of the assay. The binding of a test agent to the polypeptide, or the interaction of the polypeptide with a binding agent in the presence and / or absence of a test agent, can be achieved in any suitable container that contains the reagents. Examples of such containers include microtitre plates, test tubes and micro-centrifuge tubes. In one embodiment, a fusion protein (eg, a glutathione-S-transferase fusion protein) can be provided which adds a domain that allows a FLAP nucleic acid or a FLAP binding agent that binds to one matrix or another solid support. In another embodiment, the modulators of expression of nucleic acid molecules of the invention are identified in a method wherein a cell, cell lysate or solution containing a nucleic acid encoding a FLAP nucleic acid is contacted with a test agent and the expression of the appropriate mRNA or polypeptide (e.g., the splice variant (s)) in the cell, cell lysate or solution, is determined. The level of expression of the appropriate mRNA or polypeptide (s) in the presence of the test agent is compared to the level of expression of mRNA or polypeptide (s) in the absence of the test agent. The test agent can then be identified as an expression modulator based on this comparison. For example, when mRNA or polypeptide expression is higher (statistically significantly greater) in the presence of the test agent than in its absence, the test agent is identified as a stimulator or enhancer of mRNA or polypeptide expression. Alternatively, when the expression of the mRNA or polypeptide is lower (statistically, significantly less) in the presence of the test agent than in its absence, the test agent is identified as an inhibitor of mRNA or polypeptide expression. The level of mRNA or polypeptide expression in the cells can be determined by methods described herein to detect mRNA or polypeptide. In yet another embodiment, the invention provides methods for identifying agents (e.g., fusion proteins, polypeptides, peptide mimetics, pro-drugs, binding receptors, antibodies, small molecules or other drugs or ribozymes) which they alter (e.g. increase or decrease) the activity of a member of the leukotriene pathway binding agent, such as a FLAP binding agent (eg, 5-LO), as described herein. For example, such agents may be agents which have a stimulatory or inhibitory effect on, for example, the activity of a member of the leukotriene pathway binding agent, such as a FLAP binding agent.; which changes (e.g., improves or inhibits) the ability of a member of the leukotriene pathway binding agents (e.g., receptors or other binding agents) to interact with the polypeptides of the invention; or which alter the post-translational processing of the leukotriene path binding agent member, (eg, agents that alter proteolytic processing to direct the leukotriene path binding agent member from where it is normally synthesized to another location in the cell, such as the cell surface, agents that alter the proteolytic processing so that more than one active binding agent is released from the cell, etc.). For example, the invention provides assays for screening candidate or test agents that bind to, a or modulate the activity of a member of the leukotriene pathway (or enzymatically active portion or portions thereof), as well as agents identifiable by the assays. .As described above, the test agents can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; solid phase libraries or parallel spatially steerable solution phase; synthetic library methods that require descircunvolution; the compound of "a pearl a compound"; and methods of synthetic library using affinity chromatography selection. The biological library approach is limited to polypeptide libraries, while the other four approaches are applicable to polypeptide, non-polypeptide oligomer or small molecule libraries of compounds (Lam, K.S. Anticancer Drug Des., 12: 145 (1997)). In one embodiment, to identify agents which alter the activity of a member of the leukotriene pathway (such as a FLAP binding agent, or an agent which binds to a member of the leukotriene pathway (a "link agent"). ")), a cell, a cell lysate, or a solution that contains or expresses a binding agent (e.g., 5-LO or a leukotriene pathway member receptor, or other binding agent), or a fragment ( for example, an enzymatically active fragment) or derivative thereof, can be contacted with an agent that is tested; alternatively, the linking agent (or fragment or derivative thereof) can be contacted directly with the agent to be tested. The level (amount) of the activity of the binding agent is evaluated (either directly or indirectly), and compared with the level of activity in a control (ie, the level of activity in the absence of the agent being tested) . If the level of activity in the presence of the agent differs, by an amount that is statistically significant, from the level of activity in the absence of the agent, then the agent is an agent that alters the activity of the leukotriene path member. An increase in the level of activity relative to control indicates that the agent is an agent that improves (is an agonist) activity. Similarly, a decrease in the level of activity relative to a control indicates that the agent is an agent that inhibits (is an antagonist of) the activity. In another embodiment, the level of activity in the presence of the agent that is used is compared to a control level that has been previously established. A level of activity in the presence of the agent that is deferred from the control level by an amount that is statistically significant indicates that the agent alters the activity. This invention also relates to novel agents identified by the screening assays described above. Accordingly, it is within the scope of this invention to further utilize an identified agent. as described herein in an appropriate animal model. For example, an agent identified as described herein (for example, a test agent that is a modulating agent, a non-coding nucleic acid molecule, a specific antibody, or a polypeptide binding agent) can be used in an animal model to determine the efficacy, toxicity or side effects of treatment with such an agent. Alternatively, an agent identified as described herein may be used in an animal model to determine the mechanism of action of such an agent. In addition, this invention relates to the use of novel agents identified by the screening assays described above for treatments as described herein: In addition, an agent identified as described herein may be used to alter the activity of a coded polypeptide. by a FLAP nucleic acid, or to alter the expression of a FLAP nucleic acid, by contacting the polypeptide or the nucleic acid (or by contacting a cell comprising the polypeptide or the nucleic acid) with the identified agent as described in the present . The present invention is now illustrated by the following Examples, which are not intended to be limited in any way. The teachings of all cited references are incorporated herein in their entirety.
Example 1; Identification of the Gene and Haplotypes Associated with MI A comprehensive genome scan of 296 multiple Icelandic families with 713 patients with MI was performed. Through the ligament suggestive of a site on chromosome 13ql2-13 for patients with MI women and patients with early MI onset, and analysis of haplotype association, the gene encoding the 5-lipoxygenase activation protein (FLAP) was identified and a 4 -SNP haplotype within the gene was determined to confer an almost twice-MI risk. Male patients showed stronger association in haplotype at risk. Independent confirmation of association of FLAP to MI was obtained in a group of British patients with sporadic MI. These findings maintain FLAP as the first isolated specific gene that confers substantial risk of the complex MI trait.
Methods Study population Patients participating in the study were recruited from one. record that includes all MIs that occurred before 75 years (more than 8,000 patients) in Iceland from 1981 to 2000. This record is a part of the MONICA Project of the World Health Organization (The World Health Organization MONICA Project, WHO MONICA Project Principal Investigators, J. Clin Epidemiol 41 105-14 (1988)). The diagnosis of all patients in the registry followed strict diagnostic rules based on signs, symptoms, electrocardiograms, cardiac enzymes and necropsy findings. Genotypes from 713 patients with MI and 1741 from their first-degree relatives were used in the linkage analysis. For the microsatellite association study of the MI site, 802 patients with unrelated MI (n = 233 women, n = 624 men and n = 302 in early onset) and 837 controls based on the population were used. For the SNP association study in and around the FLAP gene 779 unrelated MI patients were classified by genotype (n = 293 women, n = 486 men and n = 358 in early onset). The control group for the SNP association study It was a population based and comprised of 628 unrelated men and women in the age range of 30-85 years, whose medical history is unknown. The study was approved by the Icelandic Data Protection Commission and the National Bioethics Committee of Iceland. Informed consent was obtained from all study participants. Personal identifiers associated with medical information and blood samples were coded with a third party coding system as previously described (Gulcher, JR, Kristjansson, K., Gudbjartsson, H. &Stefansson, K., Eur J Hum Genet 8, 739.42 (2000)). Statistical analysis A broad genome scan was performed as previously described (Gretarsdottir, S. et al., Am J Hum Genet 70, 593-603 (2002)), using a set of approximately 1000 microsatellite markers. Exchange methods of affected allele only by multipoint (Kong, A. &Cox, N.J., Am J Hum Genet 61, 1179-88 (1997)) were used to evaluate binding evidence. All results were obtained using the Allegro program (Gudbjartsson, D.F., Jonasson, K., Frigge, M.L. &Kong, A.
Allegro, Nat Genet 25, 12-3 (2000)) and the genetic map of CODE (Kong, A. et al., Nat Genes 31, 241-7 (2002)). The Spares classification function (Whittemore, A.S. &Halpern, J., Biometrics 50, 118-27 (1994); Kruglyak, L., Daly, M.J., Reeve-Daly, M.P. & Lander, E.S., Am J Hum Genet 58, 1347-63 (1996)) was used, as did the exponential allele exchange model (Kong, A. &Cox,? .J. Am J Hum Genet 61, 1179-88 (1997)) to generate relevant 1-df statistics (degree of freedom). When family grades are combined to obtain a total degree, a specific value scheme was used, which is partially a logarithm to scale between the specific value of each affected pair equally and the specific value of each family as well. In the analysis, all individuals, classified by genotype who were not affected, were treated as "unknown". Due to the concern with the small sample behavior, the corresponding P values were usually computed in two different comparison modes, and one less significant was reported. The first P value is computed based on the large sample theory; Zir = (2 logc (10) LOD) and is distributed approximately as a standard normal distribution under the null hypothesis of no linkage (Kong, A. &Cox, N.J. Am J Hum Genet 61, 1179-88 (1997)). A second P value was calculated by comparing the observed grade of LOD to its full data sampling distribution under the null hypothesis (Gudbjartsson, DF, Jonasson, K., Frigge, ML &Kong, A. Allegro, Nat Genet 25, 12-3 (2000)). When a data set consists of more than a handful of families, these two P values tend to be very similar. The measure of information that was used (? Icolae, D. University of Chicago (1999)), and implemented in Allegro, is closely related to a classical measure of information (Dempster, A., Laird,? M, Rubin, DB ., JR Stat Soc B 39, 1-38 (1977) and has a property that is between 0, if the marker genotypes are completely non-informative, and 1, if the genotypes determine the exact amount of interchange of alleles by descent between the Affected relatives For single label association studies, Fisher's exact test was used to calculate two-sided P values for each allele.All P values were not adjusted for multiple comparisons unless specifically indicated. allele instead of microsatellite carriers, SNPs and haplotypes, to minimize any deviation due to the relevance of patients recruited as families for linkage analysis. and second grade were removed from the patient list. For the haplotype analysis, the NEMO program was used (Gretarsdottir, S. et al., Nat Genet n35, 131-8 (2003)), which manages the absent genotypes and the uncertainty with the phase through a probability procedure, using the expectation-maximization algorithm as a calculation tool to calculate the haplotype frequencies. Under the null hypothesis, affected individuals and controls are assumed to have identical haplotype frequencies. Under the alternative hypothesis, the candidate haplotype at risk is allowed to have a higher frequency in affected individuals than in controls, while the frequency relationships of all other haplotypes are assumed to be the same in both groups. The probabilities are maximized separately under both hypotheses, and a corresponding 1-df probability relation statistic is used to evaluate the statistical significance (id). Even though the investigations were carried out only for haplotypes that increase the risk, all the values reported P are double, unless otherwise stated. To assess the importance of the correct haplotype association for multiple testing, a randomization test was carried out using the same genotype data. The groups of affected individuals and controls were taken at random and the analysis repeated. This procedure was repeated up to 1,000 times and the P value present is the fraction of replications that produce a P value for a tested haplotype that is less than or equal to the P value observed using the original patient and the control groups. For both single marker and haplotype analyzes, the relative risk (RR) and the risk attributable to the population were calculated assuming a multiplicative model (Terwilliger, JD &Ott, JA, Hum Hered 42, 337-46 (1992); Falk, CT &Rubinstein, P., Ann Hum Genet 51 (Pt 3), 227-33 (1987)) in which the risk of the two haplotype alleles to a person is multiplied. The LD was calculated between pairs of SNPs using the standard definition of D '(Lewontin, RC, Genetics 50, 757-82 (1964)) and R2 (Hill, WG &Robertson, A., Genetics 60, 615-28 (1968)). When using NEMO, the frequencies of the two allele marker combinations are calculated by a maximum probability, and the deviation from the linkage equilibrium is evaluated by a probability relation test. When all the SNP combinations are plotted to produce the LD structure in a particular region, D 'was plotted in the upper left corner and the P value in the lower right corner. In the presented LD diagrams, the markers are drawn equidistantly more than according to their physical positions.
Identification of DNA polymorphism New polymorphic repeats (eg, dinucleotide or trinucleotide repeats) were identified with the Sputnik program. For microsatellite alleles: sample CEPH 1347-02 (Center d'Etudes du Polymorphisme Humain, genomics repository) is used as a reference. The lower allele of each microsatellite in this sample is set to 0 and all other alleles in other samples are listed according to this reference. Thus, the allele is 1 bp longer than the lower allele in the CEPH sample, allele 2 is 2 bp longer than the lower allele in the CEPH sample, allele 3 is 3 bp longer than the allele lower in the CEPH sample, allele 4 is 4 bp longer than the lower allele in the CEPH sample, allele 1 is 1 bp shorter than the lower allele in the CEPH sample, allele 2 is 2 bp more short than the lower allele in the CEPH sample, etc. Simple nucleotide polymorphisms in the gene were detected by exonic and intronic regions of PCR sequencing from patients and controls. The simple public de-nucleotide polymorphisms were obtained from the NCBI SNP database. SNPs were genotyped using a method to detect SNPs with the addition of dye terminator targeted by fluorescent polarization template (SNP-FP-TDI assay) (Chen, X., Zehnbauer, B., Gnirke, A. & Kwok, PY, Proc Nati Acad Sci USA 94, 10756-61. (1997)) and TaqMan assays (Applied Biosystems).
RESULTS Linkage analysis A genome wide scan was performed in the search for MI susceptibility genes using a structure set of about 1000 microsatellite markers. The initial linkage analysis included 713 patients with MI who met the search criteria WHO MONICA (The World Health Organization MONICA Project, WHO MONICA Project Principal Investigators, J Clin Epidemiol 41, 105-14 (1988)) and grouped into 296 families extended. The linkage analysis was also repeated for early onset, separately in male and female patients. The description of the number of patients and families in each analysis was given in Table 1. TABLE 1: Number of patients grouped into families and the corresponding number of families used in linkage analysis Phenotype Number Number Number of Relatives patients, even families classified as genotype51 All patients with MI 713 296 863 1741 men 575 248 724 1385 and women 140 56 108 366 Home 194 93 156 739 early a Patients classified by genotype were used to increase information in IBD exchange among patients in the linkage analysis None of these analysis produced a site of broad genome importance. However, the most prominent LOD grade (LOD = 2.86) was observed on chromosome 13ql2-13 for patients with MI women at the D13S289 peak marker (data not shown). This site also had the most prominent LOD grade (LOD = 2.03) for patients with MI in early onset. After increasing the information in identity exchange by descent over 90% by classifying 14 additional microsatellite markers in a region of 30 centiMorgan (cM) around D13S289, the LOD grade from the women's analysis fell to .2.48 (P value = 0.00036), while the highest grade LOD remained at D13S289 (FIGURE 6.1).
Microsatellite association study The 7.6 Mb region corresponding to a one-degree LOD drop in the linkage analysis of women who have MI, contains 40 known genes (Table 2). Table 2: Genes residing within a fall region LOD of the ligation peak of chromosome 13ql2-13 LL symbol [Nop? Bre_genético_LL USP12L1 protease 12 specific for ubiquitin similar to 1 RP 21 ribosomal protein L21 GTF3A general transcription factor IIIA MTIF3 factor 3 of transnational mitochondrial initiation PDZRN1 domain PDZ containing ring finger 1 MGC9850 hypothetical protein MGC9850 POLR1D polypeptide D polymerase I (RNA) 16kDa GSH1 homeobox 1 GS GS1 factor 1 insulin promoter, homeodomain transcription factor CDX2 transcription factor 2 baking box type caudal FLT3 tyrosine kinase 3 related to fms L0C255967 protein hypothetical LOC255967 Tyrosine kinase 1 related to fms (vascular endothelial growth factor / receptor of the FLT1 factor vascular permeability) C13orfl2 reading frame 12 open on chromosome 13 LOC283537 protein hypothetical LOC283537 KIAA0774 protein KIAA0774 Family 7 carrier of solutes (cationic amino acid transporter, and + system), SLC7A1 member 1 UBL3 ubiquitin similar to 3 MGC2599 hypothetical MGC2599 protein similar to subunit A 2599 of katanin p60 HMGB1 group box 1 high mobility D13S106E highly loaded protein A OX5AP araqiaidonat activation protein or 5-lipoxygenase FLJ14834 hypothetical protein FLJ14834 MGC 0178 hypothetical protein MGC40178 HSPH1 104kDa / 110kDa heat shock protein B3GT similar to beta 3-glycosyltransferase similar to the G protein-coupled receptor that affects the testicular offspring GR? AT (H. sapiens) LOC196549 similar to hypothetical protein FLJ20897 13CDNA73 hypothetical protein CG003 BRCA2 breast cancer 2, early start CG018 gene CG018 hypothetical PRO0297 protein PRO0297 LOC88523 CG016 CG012 gene CG012 hypothetical gene CG030 hypothetical gene CG005 hypothetical protein from region CRA2 APRIN inhibitor proliferation induced by androgen KL Cloto STARD13 domain START containing 13 RFC3 replication factor C (activator 1) 3, 38kDa To determine which gene in this region most likely contributes to MI, 120 microsatellite markers placed within this region were classified, and an association study of cases and controls was performed using 802 unrelated MI patients and 837 population-based controls. The association study was also repeated for each of the three phenotypes that were used in the linkage study, that is, early onset, in patients with MI men and women. The initial association analysis was performed when the average spacing between the microsatellite markers was approximately 100 kb. This analysis revealed several extended haplotypes composed of 4 and 5 microsatellite markers that were significantly associated with MI in women (see FIGURES 1 and 2, and Tables 13 and 14). A region common to all these extended haplotypes is identified by markers DG13S166 and D13S1238. This region included only one gene, the FLAP nucleic acid sequence. The haplotype of two markers involving 0 and -2 alleles for markers DG13S166 and D13S1238, respectively, was found in excess in patients. This was the first evidence that the FLAP gene could be involved in the pathogenesis of myocardial infarction. Subsequent haplotype analysis that includes more microsatellite markers (n = 120) in the candidate region on chromosome 13ql2-13, now with a marker density of 1 microsatellite marker per 60 kb, showed reduced significance of the original haplotype association. However, the haplotype association analysis using increased density of markers again was directed towards the FLAP gene. This analysis strongly suggests that a region of 300 kb was involved in myocardial infarction susceptibility. As shown in FIGURE 5.2, the haplotype that showed association with the entire MI with the lowest P value (0.00009) covered a region containing 2 known genes, including the gene that encodes the protein that activates arachidonate 5-lipoxygenase (FLAP) and a gene with an unknown function called a highly charged protein. However, the haplotype association to MI in women in this region was less significant (P value = 0.005) than for all MI patients and surprisingly, the most significant haplotype association was observed for patients with MI men (P value = 0.000002 ). This MI haplotype in men was the only haplotype that remained important after adjusting all tested haplotypes. In view of the association results described above, FLAP was an attractive candidate and therefore focused efforts on this gene. Selection for FLAP polymorphisms and linkage disequilibrium mapping To determine if variations within the FLAP gene are significantly associated with MI and for the search for causal variations, the FLAP gene was sequenced in 93 patients and 93 controls. The sequenced region covers 60 kb containing the FLAP gene, including 5 known exons and introns and the 5 'region of 26 kb to the first exon and the 3' region of 7 kb to the fifth exon. In total, 144 SNPs were identified, of which 96 were excluded from additional analyzes either due to low frequency of minor allele or that were completely correlated with other SNPs and thus were repetitive. FIGURE 6 shows the distribution of 48 SNPs, used to classify by genotype, in relation to exons, introns and the flanking regions 5 'and 3' of the FLAP gene. Only one SNP was identified within a coding sequence (exon 2). This SNP was not driven to amino acid substitution. The locations of these SNPs in the NCBI human genome system, construction 34, are listed in Table 3. Table 3: Locations of all SNPs classified by genotype in NCBI construction 34 of the human genome system. name SNP Construcción34 start SG13S381 29083350 SG13S366 29083518 SG13S1 29086224 SG13S2 29087473 SG13S367 29088090 SG13S10 29088473 SG13S3 29089044 SG13S368 29089886 SG13S4 29090997 SG13S5 29091307 SG13S90 29091780 SG13S6 29092536 SG13S371 29093964 SG13S372 29094259 SG13S373 29096688 SG13S375 29096874 SG13S376 29096962 SG13S25 29097553 SG13S377 29101965 SG13S100 29104271 SG13S95 29106329 SG13S191 29107830 SG13S106 29108579 SG13S114 29110096 SG13S121 29112174 SG13S122 29112264 SG13S43 29112455 SG13S192 29116308 SG13S88 29116401 SG13S137 29118118 SG13S86 29118815 SG13S87 291 18873 SG13S39 29119740 SG13S26 29122253 SG13S27 29122283 SG13S29 29123643 SG13S89 29124441 SG13S96 29124906 SG13S30 29125840 SG13S97 29129139 SG13S32 29130547 SG13S41 29134045 SG13S42 29135877 SG13S34 29137100 SG13S35 29138117 SG13S181 29138633 SG13S184 29139435 SG13S188 29140805 In addition to the SNPs, a polymorphism consisting of a repeating monopolymer A that has been described in the FLAP promoter region was classified (Koshino, T. et al., Mol Cell Biol Res Commun 2, 32-5 (1999)) . The linkage disequilibrium (LD) block structure defined by 48 SNPs that were selected for classification by additional genotype is shown in FIGURE 8. A strong LD was detected across the FLAP region, although it appears that at least one recombination may have occurred by dividing the region into two strongly correlated LD blocks.
Haplotype Association to MI To perform a case-control association study, the 48 selected SNPs and the monopolymer A repeat marker were classified by genotype in a set of 779 unrelated MI patients and 628 population-based controls. Each of the 49 markers was individually tested for association to the disease. Three SNPs, one located 3 kb upstream of the first exon and the other two 1 and 3 kb downstream of the first exon, showed nominally significant association to MI (Table 4). Table 4: Allelic association of SNP in groups with MI Phenotype Marker Allele value P RR # Pat. % Pat: # Clt% Ctl All patients SG13S106 G 0.0044 1.29 681 72.0 530 66.6 SG13S100 A 0.020 1.29 388 69.6 377 63.9 SG13S114 T 0.021 1.21, 764 70.0 602 65.8 Men SG13S106 G 0.0037 1.35 422 7 722..99 530 66.6 SG13S100 A 0.0099 1.36 292 70.7 377 63.9 SG13S .114 T 0.026 1.24 477 70.4 602 65.8 early start SG13S100 A 0.0440 1.43 99 71.7 377 63.9 The nominally significant SNP association with the corresponding number of patients (# Pat.) And controls (# Ctrl.). RR refers to relative risk However, after adjusting the number of markers tested, these results were not significant. A search was then conducted for haplotypes showing association to the disease using the same groups. For calculation reasons, the search was limited to haplotype combinations constructed from two, three or four SNPs, and only haplotypes that were in excess in the patients were tested. P values were adjusted for all tested haplotypes by randomly taking patients and controls (see Methods). Several haplotypes were found to be significantly associated with the disease with an adjusted P value of less than 0.05 (Table 5). T7ABLA 5: SNP haplotypes that are significantly associated with patients with MI Icelandic. a Simple test P values, adjusted P values for all tested SNP haplotypes. c Correlation measure with Haplotype A4. The most significant association was observed for a four SNP haplotype spanning 33 kb, including the first four exons of the gene (Figure 6.3) with a nominal P value of 0.0000023 and an adjusted P value of 0.005. This haplotype, labeled A4, has a haplotype frequency of 15.8% (30.3% carrier frequency) in patients against 9.5% (17.9% carrier frequency) in controls (Table 6). Table 6: Association of haplotype A4 to MI, Stroke and PAOD Adjusted P value for the number of haplotypes tested. bNo to do. c Excluding known cases of MI. The FLAP A4 haplotype and the corresponding number of patients (n), haplotype frequency in patients (Frq Pat.), Relative risk (RR), risk attributed to population (PAR) and P values are shown. The haplotype A4 is defined by the following SNPs: SG13S25, SG13S114, SG13S89 and SG13S32 (Table 5). The same controls (n = 628) are used for the analysis of association in MI, stroke and PAOD as well as for men, women and the analysis of early onset. The haplotype frequency A4 in the control groups is 0.095. The relative risk conferred for the A4 haplotype compared to other haplotypes constructed in the same SNP, assuming a multiplicative model, was 1.8 and the risk attributable to the corresponding population (PAR) was 13.5%. As shown in Table 6, the A4 haplotype was observed in higher frequency in male patients (30.9% of carrier frequency) than in female patients (25.7% of carrier frequency). All other haplotypes that were significantly associated with an adjusted P value less than 0.05, were highly correlated with the A4 haplotype and variants of that haplotype should be considered (Table 5). Table 6 shows the results of the A4 haplotype association study using 770 patients with MI, 702 stroke patients, 577 patients with PAOD and 628 controls. First- and second-degree relatives were excluded from groups of patients. All known cases of MI were removed from groups with stroke and PAOD before the test by association. A significant association of the A4 haplotype to stroke was observed, with a relative risk of 1.67 (P value = 0.000095). In addition, it was determined if the A4 haplotype was associated mainly with a particular apoplexy sub-phenotype, and it was found that both ischemic and hemorrhagic strokes were significantly associated with the A4 haplotype (Table 22, later).
More variants of the A4 haplotype Two correlated SNP haplotype series were observed in excess in patients, denoted as A and B in Table 7. The length of the haplotypes varies between 33 and 69-kb, and the haplotypes cover one or two blocks of linkage disequilibrium. Both series of haplotypes contain the common G allele of SNP SG13S25. All haplotypes in series A contain SNP SG13S114, while all haplotypes in series B contain SNP SG13S106. In the B series, haplotypes B4, B5 and B6 have a relative risk (RR) greater than -2 and with allelic frequencies above 10%. Haplotypes in series A have slightly lower RR and lower p values, but higher frequency (15-16%). The haplotypes in the A and B series correlate strongly, that is, the haplotypes in B define a subset of the haplotypes in A. Therefore, the B haplotypes are more specific than A. The haplotypes A, however, are more sensitive , that is, they capture more individuals with the putative mutation, as observed in the risk attributable to the population, which is lower for B than for A. In addition, these haplotypes show similar risk relationships and allelic frequency for patients in the early stage ( defined as the beginning of the first MI before 55) and for both genders. In addition, when analyzing several groups of patients with known risk factors, such as hypertension, high cholesterol, smoking and diabetes, no significant correlation was found with these haplotypes, indicating that the haplotypes in the FLAP gene represent an independent genetic susceptibility factor. for me . Table 7: Selected SNP haplotypes and corresponding p-values Relative risk (RR), number of patients (#aff), allelic frequency in patients. (aff.frq.), carrier frequency in patients (carr.frq.), number of controls (#con), allelic frequency in controls (con.frq.), population attributable risk (PAR). The patients used for this analysis were not related within 4 miosis.
Haplotype association for MY women Before all the SNPs had been classified, which eventually lead to the identification of the A4 haplotype, a haplotype association analysis was performed that included 437 patients with MI women, 1049 patients with MI men, and 811 controls that had been classified by genotype with several SNPs and 3 microsatellite markers. These markers were all located within the 300 kb region encompassed by the FLAP gene. In a case-control study of MI patients using these data, several haplotypes were found, which were significantly over-represented in "patients with MI women compared to controls (see Table 8)." All these haplotypes were highly correlated yes Table 8: haplotypes in the FLAP region (FLAP and flanking nucleotide sequences) that were associated with MI women Table 9 shows two haplotypes that are representative of these haplotypes at risk women, with MI.The relative risk of these haplotypes were 2.4 and 4, and were carried out by 23% and 13% of patients with MI women, respectively Table 9: Two representative variants of the haplotypes "at risk" with MI women P-val: p-value for the association. N_aff: Number of patients used in the analysis Aff. Frq: haplotype frequency in patients N_ctrl: number of controls used in the analysis Ctrl, frq: frequency of haplotype in cont roles Rel_risk: Relative risk of the haplotype. PAR: risk attributable to population. Info: information content. Table 10 shows that these same haplotypes were over-represented also in patients with MI men compared to controls, although with a lower relative risk. It should be noted that after classifying and analyzing more SNPs in the FLAP region, these "at risk" haplotypes with MI women could no longer be considered significant after adjusting for multiple tests. Table 10: Haplotype frequencies "at risk" with MI women in male patients against controls.
P-val: p-value for the association. N_aff: Number of patients in the analysis. Aff. frq: haplotype frequency in patients. N_crtl: number of controls used in the analysis. Ctrl, frq: haplotype frequency in controls. Rel_ris: Relative risk of the haplotype. PAR: risk attributable to population. Info: information content. Table 11: The marker map for chromosome 13 is used in the linkage analysis.
- Table 12 Map Marker for the. Second stage of the Linkage Analysis Table 13 shows the exons with positions encoding the FLAP protein, markers, polymorphisms and the SNPs identified within the genomic sequence by the methods described herein.
M NJ H H L? Or L? Or L? Table 13 Construction Construction (%) of final position in NCBI 34; 34 of NCBI; frequency start > from the position of start in chr. stop at chr.SWP / marker / SNP allele allele SEC. FROM iaSEC. ID 13 (pb) 13 (pb) name of exon aliasl alias2 public minor variation minor IDEIM T. NO. IDEN T. NO .: 1 28932432 28932432 rs1556428 SG13S421 DG00AAFQR AGG 10.32 432432 28960356 28960356 SG13S417 SNP13B R1028729 rs1028729 C / TG 28 356 30.46 28 356 28965803 28965803 SG13S418 SNP13B Y1323898 rs1323898 A / GT 33803 37.38 33803 28974627 28974627 SG13S44 A / GG 0545 42627 42627 28975101 28975101 SG13S45 C / GG 1,111 43,101 43,101 28,975,315 28,975,315 SG13S46 A / GG 0328 43315 43315 28975353 28975353 SG13S50 C / TC 0495 43,353 43,353 28,975,774 28,975,774 SG13S52 A / GA 6993 43,774 43,774 28,985,244 28,985,244 SG13S53 rs1408167 A / CC 30876 53244 53244 28985303 28985303 SG13S55 rs1408169 A / 6 G 6,731 53,303 53,303 28,985,423 28,985,423 SG13S56 GTT 0353 53,423 53,423 28,985,734 28,985,734 SG13S57 rs6490471 C / TC 31,356 53,734 53,734 28,985,902 28,985,902 SG13S58 rs6490472 A / GA 30935 53902 53902 29003869 29003869 SG13S59 C / GG 5492 71,869 71,869 29,004,696 29,004,696 SG13S60 A / GA 1.812 72696 72696 29007670 29007670 SG13S419 SNP13B K912392 rs912392 C / TG 35.00 75670 75670 to L? 29015410 29015410 SG13S61 cn C 1,314 83,410 83,410 29,025,792 29,025,792 SG13S62 cp T 3521 93,792 93,792 29,026,202 29,026,202 SG13S63 ÍS7997114 A / GA 30,031 94,202 94,202 29,026,668 29,026,668 SG13S64 A / GA 1724 94,668 94,668 29,038,707 29,038,707 SG13S65 A / GA 0369 106707 106707 29042180 29042180 SG13S420 DG00AAFIV rs2248564 A / TA 13.66 110180 110132 29049355 29049355 SG13S66 A / GA 20.779 117355 117355 29049446 29049446 SG13S67 CT13966 29049355 29049447 SG13S67 A / CA 16.923 118416 118416 29059348 29059448 SG13S70 A / CA 34,364 127348 127348 A / GA 8.537 127383 127383 29059383 29059383 SG13S71 127402 127402 29059402 29059402 SG13S72 G / TT 25,536 131,702 131,949 29,063,702 29,063,949 D13S289 132359 132753 29064359 29064753 DG13S166 A / GA 37302 134272 134272 29066272 29066272 SG13S73 138551 138551 29070551 29070551 SG13S99 SNP 13 Y1323892 DG00AAFIU rs1323892 C / TC 6.25 0.49 149983 149983 29081983 29081983 SG13S382 FLA267479 A / GAA / GA 14.08 150200 150200 29082200 290822 00 SG13S383 FLA267696 267853 A G G 0.62 150357 150357 29082357 29082357 SG13S384 FLA AJER C G G 14.01 151350 151350 29083350 29083350 SG13S381 FLA268846 DG00A t NJ H H? Or L? Or L? 29083518 29083518 SG13S366 FLA269014 DG00AAJES rs4312166 A / GT 10.8 153190 153190 29086224 29086224 SG13S1 FLA271864 G / TG 30.00 154224 154224 29087473 29087473 SG13S2 FLA273371 A / GA 27.95 155473 155473 29088090 29088090 SG13S367 FLA273988 DG00AAJEU rs4474551 A / GG 2.41 156 090 156 090 29088186 29088186 SG13S388 FLA274084 A / GA 0.39 156 186 156 186 29088473 29088473 SG13S10 FLA274371 A / TT 10.23 156 473 156 473 29089044 29089044 SG13S3 FLA274942 cp T 15.17 157044 157044 29089886 29089886 SG13S368 FLA275784 DG00AAJEV cp T 13.60 157 886 157 886 29090025 29090025 SG13S369 F A275923 DGOOAAJEW GGG G 12.44 158 025 158 025 29090054 29090054 SG13S370 FLA275952 DGOOAA EX A / GA 1345 158054 158054 29090997 29090997 SG13S4 FLA276895 G / CG 14.59 158 997 158 997 29091307 29091307 SG13S5 FLA277205 rs4238133 G / TT 26.84 159 307 159307 29091580 29091580 SG13S389 FLA277478 A / G A 12.73 159580 159580 29091780 29091780 SG13S90 FLA277678 A / C c 43.67 159780 159780 29092287 29092287 SG13S390 FLA278185 rs5004913 A / GA 12.18 160287 160287 29092536 29092536 SG13S6 FLA278434 A / GA 8.38 160536 160536 29092594 29092594 SG13S391 FLA278492 A / GG 0.62 160,594 160,594 29,092,947 29,092,947 SG13S392 FLA278845 GTT 12.34 160947 160947 NJ 29093964 29093964 SG13S371 FLA279888 DGOOAAJEY rs4409939 A G G 25.34 161964 161964 L? L? 29094259 29094259 SG13S372 FLA280183 DGOOAAJEZ A / G c 0.24 162259 162259 29094999 29094999 SG13S393 FLA280923 A / TT 25.66 162,999 162,999 29,096,688 29,096,688 SG13S373 FLA282612 OG00AAJFA A / GA 14.84 164688 164688 29096813 29096813 SG13S374 FLA282737 DGOOAAJFB A / GG 12.37 164,813 164,813 29,096,874 29,096,874 SG13S375 FLA282798 DGOOAA FC OT c 14.55 164874 164874 29096962 29096962 SG13S376 FLA282886 DGOOAAJFD A / GG 11.99 164962 164962 29097476 29097476 SG13S394 FLA283400 C / GC 14.66 165476 165476 29097553 29097553 SG13S25 FLA283477 A / GA 12.21 165553 165553 29098486 29098486 SG13S395 FLA284410 A / GA 0.79 166486 166486 29098891 29098891 SG13S396 FLA284815 A / CC 10.15 166891 166891 29098979 29098979 SG13S397 FLA284903 cp C 3.53 166979 166979 29101965 29101965 SG13S377 FLA287889 DGOOAAJFF A / GA 1245 169965 169965 29103909 29103909 SG13S189 FLA289833 C / GC 0.62 171909 171909 29104271 29104271 SG13S100 FLA290195 DGOOAAHIK rs4073259 A / GG 31.55 172 271 172 271 29104629 C / GG 4.94 172629 1726 29 29104629 29104646 29104646 SG13S398 FLA290553 SG13S94 FLA290570 Cp rs4073261 C 15.51 172646 29105099 172646 29105099 SG13S101 FLA291023 rs4075474 C T T 173 099 27.91 173 099 29106329 29106329 SG13S95 FLA292253 G / T G 14.74 174 329 174 329 29106652 29106652 SG13S102 FLA292576 AGG T 1.17 174 652 174 652 ISJ to Lp or L? or L? 29107138 29107138 SG13S103 FLA293062 cp T 1.28 175,138 175,138 29,107,404 29,107,404 SG13S104 FLA293328 A / GA 2.17 175 404 175 404 29107668 29107812 EX0N1 175668 175812 29107830 29107830 SG13S191 FLA293754 DG00AAFJT IS4769055A / CA 30.11 175830 175830 29108398 29108398 SG13S105 FLA294322 A / GG 0.66 176,398 176,398 29,108,579 29,108,579 SG13S106 F A294503 DG00AAHII A / GA 28.31 176 579 176 579 29108919 29108919 SG13S107 FLA294843 rs4075131 A / GG 14.85 176919 176919 29108972 29108972 SG13S108 FA294896 rs4075132 C / TC 1.21 176972 176972 29109112 29109112 SG13S109 FLA295036 A / GA 1.04 177112 177112 29109182 29109182 SG13S110 FLA295106 A / GG 0.88 177,182 177,182 29,109,344 29,109,344 SG13S111 FLA295268 rs4597169 CT C 1.14 177344 177344 29109557 29109557 SG13S112 FLA295481 cp T 7.10 177,557 177,557 29,109,773 29,109,773 SG13S113 FLA295697 rs4293222 C / GC 22.52 177773 177773 29110096 29110096 SG13S 4 FLA296020 DG00AAHID A / TA 20.86 178096 178096 29110178 29110178 SG13S115 FLA296102 A / TT 13.83 178178 178178 29110508 29110508 SG13S116 FLA296432 rs4769871 CT 178508 29110630 178508 T 29110630 4.05 SG13S117 FLA296554 rs4769872A / G A 178630 29110689 178630 29110689 4.07 SG13S118 FLA296613 rs4769873 CT 178689 29110862 178689 T 29110862 4.07 SG13S119 FLA296786 A / G A 1.06 178 862 178 862 29111889 29111889 SG13S120 FLA297813 cp 179 889 179 889 t C 16.00 29112174 29112174 SG13S121 FLA298098 DGQOAAHIJ rs4503649 A / G G 49.36 180174 180174 Lp 29112264 29112264 SG13S122 FLA298188 DGOOAAHIH A / GA 29.75 180264 180264 29112306 29112306 SG13S123 FLA298230 CT T 5.06 180,306 180,306 29,112,455 29,112,455 SG13S43 FLA298379 rs3885907 A / CC 46.23 180455 180455 29112583 29112583 SG13S399 FLA298507 A / CC 1.59 180583 180583 29112680 29112680 SG13S124 FLA298604 rs3922435 C / TT 1.45 180680 180680 29113139 29113139 SG13S125 FLA299063 A / GG 1.32 181139 181139 29114056 29114056 SG13S400 FLA299980 A / GA 3.25 182,056 182,056 29,114,738 2911473B SG13S126 FLA300662 A / GA 34.12 182738 182738 29114940 29114940 SG13S127 FLA300864 A / GG 29.63 182940 182940 29115876 29115878 SG13S128 FLA302094 rs4254165A / GA 45.68 183878 163878 29116020 29116020 SG13S129 FLA302236 rs4360791 A / GG 36.65 184020 184020 29116068 29116068 SG13S130 FLA302284 G / TG 8.07 184068 184068 184196 184296 29116196 29116296 exon2 29116249 29116249 SG13S190 FLA302465 cp T 1.02 184,249 184,249 29,116,308 29,116,308 SG13S192 FLA302524 B_SNP_302524 rs3803277 A / CA 49.57 184 308 184 308 29 116344 29116344 SG13S193 FLA302560 A / G A 0.58 184344 184344 29116401 29116401 SG13S88 FLA302617 OA_SNP_302617 rs3803278 CT C 24.71 184401 184401 29116688 29116688 SG13S131 FLA302904 cp T 7.19 184688 .184688 29117133 29117133 SG13S132 FLA303349 A / C A 1.10 185133 185133 t lo H H L? or L? Or L? 29117546 29117546 SG13S133 FLA303762"4356336 CTT 37.65 185546 185546 29117553 29117553 SG13S38 FA303769 CS4584668 A / TA 45.50 185553 185553 29117580 29117580 SG13S134 FLA303796 CTT 1.22 185,580 185,580 29,117,741 29,117,741 SG13S135 FLA303957 rs4238137 CTT 0.89 185 741 185 741 29117954 29117954 SG13S136 FLA304170 rs4147063 C / TT 36.69 185 954 185 954 29118118 29118118 SG13S137 FLA304334 DG00AAHIG rs4147064 CTT 29.11 186118 186118 29118815 29118815 SG13S86 FLA305031 A / GA 30.19 186815 186815 29118873 29118873 SG13S87 FLA305089 DG00AAHOJ A / GG 29.3 186,873 186,873 29,119,069 29,119,069 SG13S138 FLA305285 cp T 36.96 187069 187069 29119138 29119138 SG13S139 FLA305354 C / GG 36.63 187138 187138 2919289 29119289 SG13S140 FLA305505 A / GTT 37.34 187289 29119462 187289 29119462 SG13S141 FLA305678 C / T c 1.15 187 462 187 462 29119740 29119740 SG13S39 FLA305956 G / TT 9.91 187,740 29,120,939 187,740 29,120,939 SG13S142 FLA307155 rs4387455 CT 188939 188939 29120949 c 3.36 SG13S143 29120949 FLA30716 5 rs4254166 CTT 36.24 188949 188949 29121342 29121342 SG13S144 FLA307558 rs4075692 A / GA 31.58 189342 189342 29121572 29121572 SG13S145 FLA307788 C / GG 0.45 189572 189572 29121988 29121988 SG13S146 FLA308204 cp T 1.14 189 988 189 988 29122253 29122253 SG13S26 FLA308469 cp T 46.57 190253 190253 29122283 29122283 SG13S27 FLA308499 A / GA 10.34 190283 190283 to L? 29122294 29122294 SG13S147 FLA308510 CTT 8.00 190294 190294 29122298 29122298 SG13S28 FLA308514 op T 33.71 190298 190298 29122311 29122311 SG13S148 FLA308527 G / TT 2.29 190311 190311 29123370 29123370 SG13S98 FLA309586 G / TG 1.19 191370 191370 29123635 29123635 SG13S149 FLA309851 A / GA 1.01 191635 191635 29123643 29123643 SG13S29 FA309859 A / CA 47.88 191 643 191 643 29124188 29124259 exon3 192188 192259 29124441 29124441 SG13S89 FLA310657 B_SNP_310657 rs4769874 A / GA 4.68 192 441 192 441 29124906 29124906 SG13S96 FLA311122 rs4072653 A / GG 29.72 192906 192906 29125032 29125032 SG13S150 FLA31248 CG c 8.22 193 032 193 032 29125521 29125521 SG13S401 FLA311737 cp C 21.10 193521 29125822 193521 29125822 SG13S151 FLA312038 cp 193 822 193 822 t 8.57 29,125,840 29,125,840 193,840 23.23 SG13S30 FLA312056 GTT 29127301 193840 29127301 SG13S31 FA313550 T cp 24.20 195301 195301 196080 196162 29128080 29128162 29128284 29128284 EXON4 SG13S152 FLA314500 C / G 23.89 196284 196284 29128316 c 2912 8316 SG13S402 FLA314532 rs4468448 C T T 19.363 196316 196316 29128798 29128798 SG13S403 FLA315014 SS4399410 A / G G 11.50 196798 196798 29129016 29129016 SG13S153 FLA315232 A / T t 3.08 197016 197016 29129139 29129139 SG13S97 FLA315355 A / G A 9.72 197139 197139 to t L? or L? L? 29129154 29129154 SG13S154 FLA315370 CT T 0.98 197,154 197,154 29,129,395 29,129,395 SG13S40 FLA315611 G / TT 2.24 197395 197395 29129915 29129915 SG13S155 FLA316131 rs4769875 A / GA 1.43 197915 197915 29130192 29130192 SG13S156 FLA316408 A / CA 1.80 198,192 198,192 29,130,256 29,130,256 SG13S157 FLA316472 A / GG 2.38 198,256 198,256 29130299 29130299 SG13S158 FLA316515 A / CA 0.61 198,299 198,299 29,130,353 29,130,353 SG13S159 FLA316569 G / TG 2.55 198,353 198,353 29,130,391 29,130,391 SG13S160 FLA316607 cp T 0.83 198,391 198,391 29,130,547 29,130,547 SG13S32 FLA316763 A / CC 48.50 198547 198547 29131280 29131280 SG13S161 FLA317496 A / GG 2.44 199,280 199,280 29,131,403 29131403 SG13S162 FLA317619 A / GG 2.45 199403 199403 29131404 29131404 SG13S163 FLA317620 C / TC 2.45 199404 199404 29131431 29131431 SG13S164 FLA317647 rs4769058 C / TC 2.55 199431 199431 29131517 29131517 SG13S165 FLA317733 m T 20.00 199517 199517 29131528 29131528 SG13S166 FLA317744 rs4769059 C / TT 2.46 199,528 199,528 29131599 29131599 SG13S167 FL A317815 rs4769876 A / G A 3.50 199599 199599 29132003 29132003 29132003 SG13S168 FA318219 A / C C 8.39 200003 200003 29133753 29133753 SG13S33 FLA319969 G / T t 8.99 201753 201753 29134045 29134045 SG13S41 FLA320261 A / G G 5.41 202045 202045 29134177 29134177 SG13S169 FLA320393 A / G G 4.12 202177 202177 t 29134379 29134379 SG13S404 FLA320595 rs4427651 G / T G 38.33 202379 202379 L? 29135558 29135558 SG13S170 FLA321774 rs3935645 C / T C 32.77 203558 203558 8 29135640 29135640 SG13S171 FLA321856 rs3935644A / GG 4803 203640 203640 29135750 29135750 SG13S172 FLA321966 A / GG 1.67 203,750 203,750 29,135,809 29,135,809 SG13S173 FLA322025 A / TA 0.68 203,809 203,809 29,135,877 29,135,877 SG13S42 FLA322093 rs4769060 A / GG 42.44 203877 203877 29136080 29136556 EX0N5 204080 204556 29136290 29136290 SG13S194 FLA322506 cp T 0.30 204,290 204,290 29,136,462 29,136,462 SG13S195 FLA322678 rsl132340 AG G 2.46 204 462 204 462 29136797 29136797 SG13S174 FLA323013 A / GG 0.56 204 797 204 797 29137100 29137100 SG13S34 FLA323316 G / TG 30.23 205100 205100 29137150 29137150 SG13S175 FLA323366 A / GA 2.40 205 150 205 150 29137607 29137607 SG13S176 FA323823 A / GA 24.2 205 607 205 607 29137651 29137651 SG13S177 FLA323867 cp T 1.64 205,651 205,651 29,137,905 29,137,905 SG13S178 FLA324121 C / GC 1.40 205,905 205,905 29,138,117 29,138,117 SG13S35 FLA324333 A / GA 9.52 206 117 206 117 29138375 29138375 SG13S179 FLA324591 A / GA 48.14 206375 206375 29138385 29138385 SG13S180 FLA324601 C / TT 2.50 20638 5 206385 181 FLA324849 DG00AAHIF rs4420371 C / G c 49.41 206633 206633 29138633 29138633 SG13S 29139153 29139153 SG13S182 FLA325369 cp T 2.36 207153 207153 to IO M H L? or Lp O L? 29139277 29139277 SG13S183 FLA325493 rs4466940 C / TT 12.07 207 277 207 277 29139435 29139435 SG13S184 FLA325651 DG00AArs4445746 A / GA 16.67 207435 207435 29139971 29139971 SG13S185 FLA326187 A / GG 7.66 207,971 207,971 29,140,441 29,140,441 SG13S405 FLA326657 A / GA 9.66 208441 208441 29140649 29140649 SG13S91 FLA326865 A / GA 7.78 208,649 208,649 29,140,695 29,140,695 SG13S186 FLA326911 rs4769877 A / TA 25.71 208695 208695 29140703 29140703 SG13S187 FLA326919 A / GA 1.43 208 703 208 703 29140805 29140805 SG13S188 FLA327021 DG00AAJFE A / GG 4.71 208,805 208,805 29,141,049 29,141,049 SG13S406 FLA327265 err T 0.56 209 049 209 049 29142392 29142392 SG13S92 FLA328644 rs4429158 CT T 210392 29142397 210392 29142397 8.33 SG13S93 FLA328649 A / GA 29142712 210397 29142712 210397 7.23 SG13S36 FLA328964 cr C 15.88 210712 210712 29144013 29144013 SG13S407 FLA33G265 CT T 3.29 212013 212013 29144203 29144203 T cp SG13S408 FLA330455 0.30 212203 212203 29144234 29144589 212589 29144255 212234 29144255 D13S1238 SG 13S7 FLA330507 cp T 16.28 212255 212255 29144877 29144877 SG13S37 FLA331129 AG G 16.70 212877 212877 29144982 29144982 SG13S409 FLA331234 A / G A 1.93 212982 212982 29144983 29144983 SG13S8 FLA331235 rs4491352A / C C 30.64 212983 212983 t T 20.57 213122 213122 Lp 29145122 29145122 29145143 SG13S410 FLA331374 rs4319601 CT 29145143 SG13S411 FLA331395 A / G A 1.54 213 143 213 143 29145171 29145171 SG13S9 FLA331423 213,171 213,171 cp C 16.37 29145221 29145221 SG13S412 FLA331473 rs4769062 A / G A 7.42 213 221 213 221 29145265 29145265 SG13S413 FLA331517 rs4238138 CT T 1.91 213,265 213,265 Table 14: 4 Haplotypes extended microsatellite markers Alleles #s': For alleles A of SNP = 0, C = 1, G = 2, T = 3; for microsatellite alleles: the CEPH sample (Center d'Etudes du Polymorphisme Humain, genomics repository) is used as a reference, as described above. Length = haplotype length in Mb. P-val = value p. RR = relative risk. N af = number of patients. P al = allelic haplotype frequency. P ca = frequency of haplotype carrier. N ct = number of controls. Alleles = alleles in the haplotype. Markers = markers in the haplotype. Table 15: 5 haplotypes extended microsatellite markers Length = haplotype length in Mb. P-val = value p. RR = relative risk. N af = Number of patients. P al = allelic frequency of haplotype. P ca = carrier frequency of haplotype. N ct = number of controls. Alleles = alleles in the haplotype. Markers = markers in the haplotype.
EXAMPLE 2: RELATIONSHIP BETWEEN THE POLYMORPHISM IN THE PROMOTER OF 5-LIPOXYGENASE AND MY A family of mutations in the binding region of the G-C rich transcription factor of the 5-LO gene has been previously identified. These mutations consist of the elimination of one, elimination of two, or addition of a zinc finger binding site (Spl / Egr-1) in the upstream region of 176 a. 147 bp from the start site of the ATG translation where there are normally 5 reasons for link Spl in the tandem. These mutations of natural origin in the 5-LO genetic promoter have been shown to modify the transcriptional binding and reporter genetic transcription. The ability of mutant forms of DNA to promote transcription of reporter CAT constructs have been shown to be significantly lower than those of wild type DNA (JL Clin, Invest, Volume 99, Number 5, March 1997, 1130-1137). . To test whether 5-LO is associated with atherosclerotic diseases, particularly myocardial infarction (MI) in the human population, this promoter polymorphism, which consists of a variable number of binding sites Spl / Egr-1 tandem, was classified by genotype in 1112 patients with MI, 748 patients with PAOD and 541 patients with apoplexy. The results, shown in Table 16, demonstrate that the wild-type allele (which represents the allele with the most active promoter and thus with the highest expression of 5-LO mRNA; "7. Clin. Invest. 99, Number 5, March 1997, 1130-1137) is significantly associated with MI (RR = 1.2, p <0.05). The results are consistent with a disease hypothesis in which the increased expression of 5-LO plays a role in the pathogenesis of MI. Table 16 Single-sided p values; Fisher's exact test. N_aff = number of affected individuals; Frg_aff = frequency of the wild-type allele of the promoter polymorphism in the affected group; N_ctrl = number of controls; Frg_ctrl = frequency of the wild-type allele of the promoter polymorphism in the control group; EXAMPLE 3: LTE4 BIOSYNTHESIS ELEVATED IN INDIVIDUALS WITH FLAP MY HAPLOTIPE AT HIS RISK Based on the known function of the end products of the leukotriene pathway and based on the 5-LO association data, the association of the FLAP haplotype with MI it is better explained by the increased expression and / or the increased function of the FLAP gene. In other words, individuals who have a "at risk" FLAP haplotype have either an increased amount of FLAP or a more active FLAP. This would lead to increased production of leukotrienes in these individuals. To demonstrate this theory, LTE4, a metabolite of leukotriene downstream, was measured in serum samples in patients. A quantitative determination of LTE4 in human serum was performed by ligation chromatography coupled with tandem mass spectrometry. The protocol was made as follows: ANALYTICAL METHOD Table Pl (Protocol 1): List of Abbreviations Type and conditions Table P2: Type and Analytical Conditions Other instruments Table P3: The apparatus used for the treatment and sample measurements Materials Table P4: Reagents for treatment and sample measurements Table P5: Reference substances Stock solutions A stock solution of LTE4 was prepared by the supplier at a concentration of 100 μg / ml in methanol. The stock solution was diluted to a concentration of 20 μg / ml in methanol and this usable solution was kept refrigerated. 2-8 ° C. An internal standard stock solution (LTE4-d3) was prepared by the supplier at a concentration of 49.5 μg / ml.
The stock solution was diluted in a concentration of 1 μg / ml in methanol and this usable solution was kept refrigerated at 2-8 ° C. The preparation of deoxidizing solutions, calibration standards and quality control samples. The deoxidizing solutions (SS) in the concentration range of 1 ng / ml to 10000 ng / ml were prepared by dilution of the usable Solution. The following deoxidizing solutions were prepared: Table P6: Deoxidant solutions for calibration standards Table P7: Deoxidant solutions for quality controls After the preparation, deoxidizing solutions were maintained at 2-8 ° C for calibration standards and quality controls.
Preparation of calibration standards and quality controls Recent calibration standards and quality controls (QCs) were prepared each day by deoxidizing white plasma as described in Tables P8 and P9, respectively. Table P8: Preparation of calibration standards Table P9: Preparation of quality controls Sample preparation The aliquots of 400 μl of each study sample, calibration standards, QC samples and the control blank were pipetted into an eppendorf vial. All samples other than blank were added with 20 μl of internal standard usable solution and the samples were vortexed for a few seconds. The pH of the plasma samples is adjusted to pH 4.5 using 60 μl of 10% acetic acid and centrifuged for 10 minutes at 4100 rpm immediately before extraction. The 96-well solid phase extraction plate was conditioned with 1 ml of methanol and 1 ml of water. Then, 400 μl of the plasma samples are loaded onto the plate. Vacuum is applied, followed by drying the disc for 1 minute. After washing with 2 ml of water and 1 ml of 30% methanol in 2% acetic acid. Then, the plate was eluted with 0.6 ml of methanol. The plate was then dried for a few minutes. The methanol eluate was evaporated almost until it was stable under a stream of nitrogen at 45 ° C. The residue was reconstituted in 30 μl mobile phase and mixed by vortex for a few minutes. Subsequently, the solutions are centrifuged for 10 minutes at 10,000 rpm, and 10 μl is injected by the autosampler into the LC-MS / MS system for quantification.
Measurement Performance Samples were prepared and measured in batches and a typical batch will consist of: A set of calibration standards, a lower, extra calibration standard and a blank. Samples are collected from 16 individuals and a set of control samples (CL, CM, CH). Samples are collected from 17 individuals and a set of control samples (C ^, CM, CH).
Quantitative Determination of Analyte in Plasma Samples The standard curve is calculated from the ANALYTE / INTERNAL STANDARD peak area ratios of the calibration standards and their nominal ANALYTE concentrations. The unknown samples of LTE4 were calculated from a quadratic regression equation where a weighting curve, 1 / X2, is used. The measured peak area of the samples was converted into a pictogram of the ANALYTE per milliliter (pg / ml) of plasma according to the equation calculated for the standard curve. The calculation of the regression for the standard curve and the calculations of the concentration of the unknown samples and the control samples are carried out with MassLynx software.
Acceptance Criteria Calibration Standards The determination coefficient (R2) for the calibration curve must exceed 0.98. The calibration curve includes the concentration range from 50 pg / ml - 1500 pg / ml. The concentration of the calibration standards must be within ± 25% of their nominal value when they are recalculated from the regression equation. The calibration standards that leave those criteria (at most 3 in each run) are rejected and the calibration is performed again with the remaining standards. If the standard curve is not accepted, the samples must be reanalyzed.
Control samples At least two thirds of the quality controls analyzed must be within ± 25% of their nominal value when calculated from the regression equation. If more than one third of the controls fall, the samples must be reanalyzed.
Results Table 17 (below) shows that the haplotype at risk "with MI women was associated more with patients with MI women who have high LTE4 levels (50th percentile higher) than with patients with MI women who have low LTE4 levels (50th percentile). In addition, the haplotype analysis, which compares patients with MI women with high LTE4 levels with low-level women, showed that those with high levels had an increased prevalence of the "at risk" haplotype of 1.6 times (see Table 18) Although the association did not rise to the level of statistical significance, the results clearly showed that the haplotypes "at risk" are found in the group of patients with MI who have high levels of LTE.4 The carrier frequency of the haplotypes "in risk "is 12% and 20% respectively, in the group with MY full women, but they increase to 15% and 24%, respectively, in the group with MY women who have high levels of LTE4. Respectively, the carrier frequency of the "at risk" haplotypes decreases to 8% and 18%, respectively, in the 'MI Group' women who have low levels of LTE4 (Note that the carrier frequencies are twice the frequency of disease allele 1 minus the disease allele frequency plus the square of the allele frequency of the disease). Note that LTE4 may simply reflect the leukotriene synthesis ratio of the synthetic pathway of leukotriene upstream of the key leukotriene metabolite involved in MI risk. For example, leukotriene B4 is probably more susceptible than leukotriene E4 to be involved in the inflammatory aspects of MI plates, but since B4 has a short half-life, it is difficult to measure reliability in serum samples, whereas E4 has long-term stability. Table 17: Association of haplotypes at risk for MI in women, with MI in women who also have high levels of LTE4 (> 50 pg / ml (just the 50th upper percentile).
P-val: p-value for the association. N_aff: Number of patients used in the analysis. Aff. frq: haplotype frequency in patients. N_ctrl: number of controls used in the analysis, ctrl.frq: haplotype frequency in controls. Rel_risk: Relative risk of the haplotype. PAR: risk attributable to population. Info: information content. There was less association between the haplotype at risk for MI in women, with MI in women who have low levels of LTE4 (< 50 pg / ml). Table 18: Association between haplotypes that were more significantly associated with MI in women, and serum LTE4 levels.
P-val: p-value for the association. N_aff: Number of patients used in the analysis. Aff. frq: frequency of. haplotype in patients. N_ctrl: number of controls used in the analysis. Ctrl, frq: haplotype frequency in controls. Rel_risk: Relative risk of the haplotype. PAR: risk attributable to population. Info: information content. Here, the group of affected individuals were defined as patients with MI women with elevated LTE4 in serum (greater than 50 pg / ml) and the control group defined as patients with MI women with LTE4 in low serum (posterior 50 pg / ml) .
Example 4: Elevated LTE4 Correlated with High Reactive Protein C (CRP) The relationship between the increased production of leukotrienes and the inflammatory CRP marker, as well as the risk factor established for MI, was then explored.
As shown in FIGURE 4, a significant positive correlation was found between serum LTE4 levels and serum CRP levels.
Example 5: Evaluation of the CRP Level in Patients with Haplotype at Risk The CRP level was evaluated in women patients with at-risk haplotypes with MI women, in order to evaluate whether there was a presence of an elevated level of the inflammatory marker in the presence of the haplotype at risk with MI women. The results are shown in Table 19. Although the association did not rise to the level of statistical significance, it was shown that the average CRP was elevated in those patients with the haplotype at risk against those without it. Table 19: All patients Women No. Average CRP SE CRP Affected: With haplotype 155 4.91 8.7 Without haplotype 218 4.35 6.13 Example 6: Elevated Levels of LTE-4 in Serum in Patients with MI Against Controls The final products of the leukotriene pathway are lipid mediators, potent inflammatory agents that can authoritatively contribute to the development of atherosclerosis and destabilization of atherosclerotic plaques through oxidation lipid and / or pro-inflammatory effects. Examples from one to five show that: 1) MI correlates with the genetic variation in FLAP; 2) MI correlates with the promoter polymorphism of elevated expression in 5-LO; 3) the levels of reactive protein C correlate with E4 of leukotriene in serum; and 4) Patients with FLAP haplotypes at risk of MI have higher levels of leukotriene E4 in serum and CRP. Based on these data, it was hypothesized that serum leukotriene E4 levels correlate with MI risk. To test this hypothesis, LTE4, a metabolite of leukotriene downstream, was measured in 488 patients with MI women and 164 control serum samples. LTE4 levels for patients were higher than controls for controls using a unilateral ilcoxon rank sum test. The p-value of the difference was 0.0092, thus confirming the hypothesis. Therefore, elevated E4 leukotriene represents a risk factor for MI. LTE4 levels in serum or plasma can be used to profile the risk of MI for individuals to help decide treatment and the lifestyle management plan is better for the prevention of primary or secondary MI. In the same way other metabolites of leukotriene can be used to profile the risk for MI.
Example 7: Production of Increased LTB4 in Activated Neutrophils from Patients with MI A major bioactive product from one of the two branches of the 5-LO pathway is LTB4. To determine whether patients with a past history of MI have increased 5-LO pathway activity compared to controls, the production of LTB4 in isolated blood neutrophils was measured before and after stimulation in vitro with the calcium ionophore, ionomycin. No difference was detected between the production of LTB4 in neutrophils at rest from patients with MI or controls (results not shown). In contrast, LTB4 generation by neutrophils from patients with MI stimulated with the ionophore was significantly greater than by neutrophils from controls in 15 and 30 minutes, respectively (FIGURE 5.1). In addition, as shown in FIGURE 5.2, the observed increase in LTB4 release was largely accounted for by male carriers of A4 haplotype, whose cells produced significantly more LTB4 than cells from controls (P value = 0.0042) (Table twenty). As shown in Table 20, there was also an intense TB4 response in guiding males that do not carry HapA, but of doubtful importance. This could be explained by additional variants in the FLAP gene that have not been revealed., or alternatively in the LTB4 response in some of the patients without the haplotype at FLAP risk. As shown in Table 20, differences in LTB4 response were not detected in women. However, due to a sample size from Pegueño, this can not be considered conclusive. Taken together, elevated levels of LTB4 production of neutrophils stimulated from male carriers of at-risk haplotype suggest that the variants associated with the disease in the FLAP gene increase the FLAP response to factors that stimulate inflammatory cells, resulting in increased leukotriene and increased risk for MI.
Methods Isolation and activation of neutrophils in peripheral blood 50 ml of blood were extracted in EDTA containing vacuum tubes from patients with MI of 43 and 45 years and compatible controls. All blood was drawn at the same time in the early hours of the morning after 12 hours of fasting. Neutrophils were isolated using Ficoll-Pague PLUS (Amersham Biosciences). In short, the red blood cell plates from the Ficoll gradient were harvested and the red blood cells were subsequently used in 0.165 M NH 4 CL for 10 minutes on ice. After washing with PBS, the neutrophils were counted and plated in cultures of 2 × 10 6 cells / ml in 4 ml of 15% fetal calf serum (FCS) (GIBCO BRL) in RPMI-1640 (GIBCO BRL). The cells were then stimulated with maximal effective ionomycin concentration (1 μM). 0, 15, 30, 60 minutes after 600 μl of ionomycin addition of the culture medium was aspirated and stored at -80 ° C for the measurement of LTB 4 release as described below. Cells were maintained at 37 ° C in a humid atmosphere of 5% C02 / 95% air. All samples were treated with indometasin (1 μM) to blog the enzyme cyclooxygenase.
Ionomycin-induced release of LTB4 in neutrophils It was used in LTB4 immunoassay (R & D systems) to quantify the concentration of LTB4 in the supernatant from neutrophils stimulated in cultured ionomycin. The assay used is based on the competitive binding technique in which the LTB4 present in the test samples (200 1) competes with a fixed amount of LTB4 etched with alkaline phosphatase for sites in a polyclonal rabbit antibody. During the incubation, the polyclonal Ab becomes bound to a coated goat anti-rabbit Ab in the microplates. After a wash to remove the excess conjugate and an unbound sample, a substrate solution is added to the wells to determine the activity of the bound enzyme. The color development stops and the absorbance is read at 405 nm. The intensity of the color is inversely proportional to the concentration of LTB4 in the sample. Each measurement of LTB4 using the LTB4 immunoassay was done in duplicate. Table 20: Levels of LTB4 after stimulation with ionomycin of isolated neutrophils3 After 15 minutes After 30 minutes Phenotype (n) Average (SD) value P Average (SD) value P Controls (35) 4.53 (1.00) 4.67 (0.88) Men (18) 4.61 (1.10) 4.68 (1.07) Women (17) 4.51 (0.88) 4.67 (0.62) MI (41) 5.18 (1.09) 0.011 5.24 (1.06) 0.016 Carriers (16) 5.26 (1.09) 0.027 5.27 (1.09) 0.051 Non-carriers (24) 5.12 (1.08) 0.040 5.22 (1.03) 0.035 Men with Ml (28) 5.37 (1.10) 0.0033 5.38 (1.09) 0.0076 Carriers (10) 5.66 (1.04) 0.0042 5.58 (1.12) 0.013 NO carriers (18) 5.20 (1.09) 0.039 5.26 (1.05) 0.041 Women with Ml (13) 4.78 (0.95) 0.46 4.95 (0.92) 0.36 Carriers (6) 4.59 (0.80) 0.90 4.75 (0.82) 0.85 Non-carriers (7) 4.94 (1.04) 0.34 5.12 (0.96) 0.25 at 1t Average + DE of logarithm values transformed from LTB4 levels of neutrophils stimulated with ionomycin from patients with MI and controls. The results are shown for two time points: 15 and 30 minutes. The results for men and women and for carriers of men and women with MI and non-carriers of HapA at risk are shown separately. The • double p values that correspond to a standard two-sample test of the difference in the average values between the patients with MI, their various sub-groups and the controls are shown.
Example 8: Haplotypes Associated with MI Also Confer Stroke Risk and PAOD. Because apoplexy and PAOD are diseases that are closely related to MI (all occur at the base of atherosclerosis), we examined whether the SNP haplotype in the FLAP gene that confers risk to MI also confers stroke risk and / or PAOD. The "at risk" haplotype (A4 haplotype) can be defined by the following 4 SNPs: SG13S25 with the G allele, SG13S114 with the T allele, SG13S89 with the G allele and SG13S32 with the A allele. Table 21 shows that the A4 haplotype increases the risk of having a stroke to a similar degree when the risk of having an MI increases. The haplotype "at risk" is transported by 28% of patients with stroke and 17% of controls, meaning that the relative risk of having a stroke for carriers of this haplotype is 1.7 (p '= 5.8 10 + 06). Aungue is not so significant, the "at risk" haplotype also confers risk of having PAOD. Table 21: The group of patients used in the association analysis shown in Table 21 may include first and second degree relatives. Table 21, discussed above, shows the results of the A4 haplotype association study using 779 patients with MI, 702 stroke patients, 577 patients with PAOD and 628 controls. First- and second-degree relatives were excluded from the group of patients. All known cases of MI were removed from the apoplexy and PAOD groups before trial 5 per association. A significant association of the A4 haplotype to stroke was observed, with a relative risk of 1.67 (P value = 0.000095). In addition, it was determined whether the A4 haplotype was associated mainly with a particular apoplexy sub-phenotype, and it was found that both ischemic and hemorrhagic strokes were significantly associated with the A4 haplotype (Table 22). Table 22: Association of the A4 haplotype to apoplexy subgroups Phenotype (n) Pat .Frq. RR - PAR Value Pb Apoplexy (702) 0.149 1.67 0.116 0.000095 Ischemic (484) 0.148 1.65 0.113 0.00053 AER (148) 0.137 1.51 0.090 0.058 Hemorrhagic (68) 0.167 1.91 0.153 0.024 20 aExcludes known cases of MI. Finally, the A4 haplotype was less significantly associated with PAOD (Table 21). It should be noted that similar to the strongest association of haplotype A4 to MI in men compared to MI in women, it also shows association 25 stronger in stroke and PAOD in men (Table 21).
Study Population The groups with apoplexy and PAOD used in this study have been previously described (Gretarsdottir, S. et al, Nat Genet 35, 131-8 (2003), Gretarsdottir, S. et al., Am J Hum Genet 70 , 593-603 (2002), Gudmundsson, G. et al., Am J Hum Genet 70, 586-92 (2002)). For apoplexy linkage analysis, genotypes were used from 342 male patients with ischemic stroke or AIA who joined at least one other male patient within and including 6 miosis in 164 families. For association studies of 702 patients with all forms of stroke (n = 329 women and n = 373 men) and 577 patients with PAOD (n = 221 women and n = 356 men) were analyzed. Patients with stroke or PAOD who also had MI were excluded. The controls used for studies of stroke association and PAOD were the same as those used in the MI S? P association study (n = 628). The study was approved by the Icelandic Data Protection Commission and the National Bioethics Committee of Iceland. Informed consent was obtained from all participants in the study. The personnel identifiers associated with medical information and blood samples were encrypted with a third-party encryption system as described (Gulcher, JR, Kristjansson, K., Gudbjartsson, H. &Stefansson, K., Eur J Hum Genet 8, 739-42 (2000)). In addition, in an independent linkage study of male patients with ischemic stroke or transient ischemic attack, linkage to the same sites was observed as a LOD grade of 1.51 at the same peak marker (FIGURE 7), further suggesting that a susceptibility factor cardiovascular could reside on this site.
Example 9; Haplotype Association to FLAP in a British Group In an independent study, it was determined if variants in the FLAP gene also have an impact on MI risk in a population outside of Iceland. The four SNPs, which define the A4 haplotype, were classified into a group of 750 patients from the United Kingdom who had sporadic MI, and 728 controls from the British population. Patients and controls come from 3 separate study groups recruited in Leicester and Sheffield. There were no significant differences in haplotype frequency between patients and controls (16.9% versus 15.3% respectively). However, when 9 additional SNPs, distributed through the FLAP gene, were classified in the British group and were investigated for other haplotypes that could be associated with MI, two SNPs showed association to MI with a nominally significant P value (data not shown) . In addition, three and four SNP haplotype combinations increased MI risk in the British group as well, and the most significant association was observed for a fourth SNP haplotype with a nominal P value = 0.00037 (Table 23).
Table 23 Association of the HapB haplotype to British patients with MI Phenotype (n) Frq. Pat. RR PAR P-value P-value MI. (750) 0.075 1.95 0.072 0.00037 0.046 Men (546) 0.075 1.97 0.072 0.00093 ND Women (204) 0.073 1.90 0.068 0.021 ND aValue p adjusted for the number of haplotypes tested using 1,000 randomization tests. The results for HapB showing the strongest association are shown. in the British group with MI. HapB is defined by the following SNPs: SG13S377, SG13S114, SG13S41 and SG13S35 (which have the following alleles A, A, A and G, respectively.) In the three phenotypes shown, the same set of British controls n = 728 is used and the The frequency of HapB in the control group is 0.040, the number of patients (n), frequency of haplotype in patients (Frq.pat.), relative risk (RR) and risk attributed to the population (PAR).
This was called HapB haplotype. The haplotype frequency of HapB is 7.5% in the group of the patient with MI (carrier frequency 14.4%) compared to 4.0% (carrier frequency 7.8%) in controls, conferring a relative risk of 1.95 (Table 23). This haplotype remained significant after adjusting all tested haplotypes, using 1000 stages of randomization, with a value P adjusted = 0. 046. No other SNP haplotype had an adjusted P value less than 0.05. The two A4 and HapB haplotypes at risk appear to be mutually exclusive without an example where the chromosome carries both haplotypes.
British population under study The recruitment method of 3 separate groups of British subjects has been described previously (Steeds, R., Adams, M., Smith, P., Channer, K. &Samani, NJ Thromb Haemost 79, 980- 4 (1998); Brouilette, S., Singh, RK, Thompson, JR, Goodall, AH &Samani, NJ, Arterioscler Thromb Vasc Biol 23, 842-6 (2003)). In brief, in the first two groups a total of 547 patients including those who were admitted to the coronary care units (CCU) of Leicester Royal Infirmary, Leicester (July 1993-April 1994) and the Royal Hallamshire Hospital, Sheffield (November 1995) -marzo 1997) and according to the World Health Organization criteria for acute MI in terms of symptoms, elevations in cardiac enzymes or electrocardiographic changes (Nomenclature and criteria for diagnosis of ischemic heart disease.) The report of the Join International Society and Federation of Cardiology / orld Health Organization forcing on standarization of clinical nomenclature, Circulation 59, 607-9 (1979)). A total of 530 control subjects were recruited in each hospital from adult visitors to patients without cardiovascular disease in pavilions of general medicine, surgery, orthopedics and obstetrics to provide subjects likely to be representative of the source population from whom the subjects they originated. Subjects who reported a history of coronary heart disease were excluded. In a third group, 203 subjects were recruited retrospectively from the records of 3 coronary care units in Leicester. All had suffered from MI according to WHO criteria before age 50. At the time of participation, at least 3 months had passed from the acute episode in the patients. The control group comprises 180 subjects without personnel or family history of premature coronary heart disease, coupled by age, sex and current smoking status with the cases. The control subjects were recruited from 3 primary care practices located within the same geographical area. All the subjects of the group were white people of Northern European origin.
Discussion These results show that variants of the gene encoding FLAP are associated with the increased risk of MI and stroke. In the Icelandic group, a haplotype that covers the FLAP gene carries 30% of all patients with MI and barely doubles the MI risk. These findings were subsequently replicated in an independent group of stroke patients. further, another haplotype that covers the FLAP gene is associated with MI in a British group. The linkage suggestive of chromosome 13ql2-13 was observed with several different phenotypes, including MI in women, early-stage MI of both sexes, and ischemic stroke or AIA in men. However, surprisingly, the strongest haplotype association was observed in men with MI or stroke. Therefore, there may be other variants or haplotypes within the FLAP gene, or in other genes within the linkage region, which may also confer risk to these cardiovascular phenotypes. These data also show that the at-risk haplotype of the FLAP gene has increased frequency in all stroke subgroups, including ischemic, TIA, and hemorrhagic strokes. Of interest is that the A4 haplotype confers significantly higher MI risk and stroke than does PAOD. This could be explained by differences in the pathogenesis of these diseases. Unlike patients with PAOD who have ischemic legs due to atherosclerotic lesions that are responsible for gradually decreasing blood flow in the legs, patients with MI and stroke have suffered from acute episodes, with interruption in the vascular wall suddenly decreasing blood flow to regions of the heart and brain. No association was found between the A4 haplotype and MI in a British group. However, the significant association to MI was found with a variant variance encompassing the FLAP gene. The fact that different haplotypes of the gene are conferring risk to MI in a second population is not surprising. A common disease similar to MI is associated with many different mutations or sequence variations, and the frequencies of these variants associated with the disease may differ between populations. In addition, the same mutations can be observed developing in different haplotypic antecedents. In summary, it has been found that: MI correlates with genetic variation in FLAP; MI correlates with the promoter polymorphism of high expression in 5-LO; Patients with FLAP haplotypes at risk for MI women have higher serum LTE4 levels; LTE4 levels correlate with serum CRP levels; and patients with FLAP haplotypes at risk for MI have elevated CRP. In addition, neutrophils isolated from patients with MI have been shown to produce more LTB4 when stimulated with ionomycin compared to controls. Taken together, these results show that increased leukotriene synthesis is a risk factor for MI, and that this risk is driven in part by the FLAP and 5-LO gene variants and is captured in part by the measurement of LTE4 levels and CRP in serum. In addition, the SNP haplotype in the FLAP gene that confers risk to MI also confers the risk of apoplexy and / or PAOD.
Markers Used in the Present Table 24: Position of base pairs of microsatellite markers (start and stop of construct 34 of the NCBI sequence set of amplimersin and primer sequences (forward and reverse).
All references cited herein are incorporated by reference in their entirety. Although this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and detail may be made herein without departing from the spirit of the invention encompassed by the claims. annexes.
Example 10; Clinical Trial Crossed, Controlled with Placebo / Randomized, Shows the Inhibition of Reduced Biomarkers by FLAP of Risk of Infarction to the Myocardium The trajectory of 5-lipoxygenase, through FLAP, leads to the production of leukotriene B4, one of the most potent chemokine mediators of arterial inflammation. The experiments described in Example 7 showed that patients with MI make more LTB4 than controls. Therefore, it seems that the variant at risk over-regulates the trajectory of leukotriene. A clinical trial was carried out to demonstrate that patients with genetic variation in FLAP who predispose to MI could benefit from inhibiting FLAP, with the FLAP inhibitor DG-031. In the short-term study, changes in the level of biomarkers that are associated with MI risk were classified as evidence of changes in MI risk.
Population of Patients All the patients in the study had a history of MI and were carriers of specific MI-associated haplotypes in the FLAP and / or LTA4 hydrolase genes.
(See US Patent Application No. 10 / 944,272 and PCT Application No. PCT / 2004/030582, incorporated for reference in its entirety.) The recruitment process included individuals who have previously participated in an MI genetics study (Helgadottir et al., Nat. Genet. 2004; 36 (3): 233-9. 2004). Apart from FLAP, the hydrolase gene LTA4 also shows significant association to MI in Iceland and the expression of baseline mRNA of the LTA hydrolase gene is higher in patients with MI than in control subjects; that is, subjects with variants at risk in both FLAP and hydrolase LTA4 genes are at increased risk of MI in support. In this way, carriers of both haplotypes at risk of FLAP or hydrolase LTA4 were recruited as their haplotypes were confirmed by DNA analysis from the blood sample collected in the study. Nine Simple Nucleotide Polymorphism (SNP) markers were genotyped to define haplotypes at risk. These SP markers are set forth in Table 25 below and are described in detail in Example 1. SNPs that are genotyped within FLAP and LTA4 hydrolase genes were made using Taqman platform based on SNP (ABI) as described in Helgadottir et al. , 2004 Mar; 36 (3): 233-9. The haplotypes carried by each individual were calculated using the NEMO program (version 1.01) and 902 in internal population controls, as described in Gretarsdottir et al., Nat Genet 35: 131-8, 2003. Table 25 Genotypes used to derive FLAP and haplotypes at risk of hydrolase LTA4 Haplotype Allele SNP Allele SNP Allele SNP 1 A3 (FLAP gene) G SG13S25 T SG13S114 A SG13S32 2 AF (FLAP gene) G SG13S25 T SG13S114 3 NA3 (FLAP gene) A SG13S122 C SG13S32 C SG13S8 4 HF (LTA4-0H gene) A SG12S25 C SG12S223 5 GF (LTA4-0H gene) A SG12S225 T SG12S233 Conscripts were asked for permission to use their medical and genetic information already collected in the genetics of CODE (Reykjavik, Iceland) for the clinical trial. Of more than 900 patients identified as eligible by clinical and genotypic criteria, 640 returned their signed consent providing permission to use their genetic and medical data. Genotypes for FLAP and hydrolase LTA4 genes were subsequently confirmed again, and those who were carriers of variants in FLAP and / or hydrolase LTA4 genes were judged eligible for the study if they also met the other inclusion criteria and none of the exclusion criteria established in Table 26. The baseline characteristics of the patients participating in the study are set forth in Table 27. All patients who participated gave informed consent and the protocol was approved by the National Bioethics Committee of Iceland. Table 26 Study eligibility criteria Table 27. Baseline characteristics of the study group * a common low-risk haplotype (RR 1.3) carried by 85-90% of the study subjects Study Conduct All study participants live in the metropolitan area of Reykjavik or its neighboring towns. All participants in the study were followed by the designated cardiologists of the University Hospital of Iceland, in their dispensaries or private clinics, and all the subjects had participated in a study in the genetics of MI. After the subject had given informed consent, the medical and medication history, including co-morbidities, concurrent medications, and specific details, about the subject's cardiovascular history, including current status, was completed. All study participants fasted and did not take their medications before the study visit. Cardiologists examined patients in all 8 visits and completed case report forms. All blood was collected and processed immediately after the sample. All blood specimens used for biomarker studies were processed within two hours of the blood sample.
Study Drug Patients (191 subjects) who met the eligibility criteria of the study were enrolled and randomized into 3 different dose level groups: (1) 64 patients on 250 mg / day therapy with DG-031 (250 mg qd) against placebo; (2) 64 patients on therapy of 500 mg / day with DG-031 (250 mg b.i.d.) against placebo; and (3) 63 patients on therapy of 750 mg / day with DG-031 (250 mg t.i.d.) against placebo. The dose of 750 mg / day was well tolerated in human studies of previous phase I-III (Dahlen et al., Thorax 1997; 52: 342-7; Hamilton et al., Thorax 1997; 52: 348-54), conducted by healthy volunteers and asthma patients as part of the drug development program for Bayer x 1005 (now DG-031). All patients received 3 tablets per day. The treatment periods, 4 weeks each, were separated by a washout period of 2 weeks. The placebo tablets were identical in configuration, color, shape and flavor to the active tablets except that they did not contain active drug ingredients. Treatment with DG-031 or placebo was in addition to the subject's standard care, including all medications and the treatment plan as prescribed by the subject's cardiologist prior to enrollment. The cross-study design is summarized in Figure 9. Due to the early termination of 19 subjects (mainly related to unavailability due to travel) 11 were replaced before the closure of the enlistment. In this way, a total of 191 subjects were enrolled, with 172 completing all 8 visits or 8 patients (4.4%) short of target. Three subjects did not return for their early termination visit.
Data Analysis, Randomization and Statistical Considerations All data were analyzed according to a pre-established analysis plan and by intention to treatment. The hypotheses were tested at a double nominal importance level of 0.05. Each branch of the study, as well as clusters (combining dose levels), was considered for the primary analysis. Each set is a standard AB / BA cross design and in the primary efficacy analysis, the levels of biomarkers of risk to MI at the end of the treatment periods (visits 4 and 7) were used as primary response variables. The differences between DG-031 and placebo treatment was the primary outcome, evaluated separately for each of the biomarkers. The treatment effect was tested using a t-test of two samples in period differences for appropriately transformed response variables, under an assumption of normality of the transformed data. The effect of the treatment is reported as half of the average differences observed in the t test of two samples, with 95% Cl. No subsequent cross-effect tests were performed as part of the primary analysis. The tests for transfer were made and reported separately from the results of the primary analysis. As pre-specified for the primary analysis, a simple Bonferroni adjustment based on 10 biomarkers for the primary objective for the pooled set of the two highest doses was used to report the outcome of the primary objective. All reported p values are nominal. To cancel potential temporal effects, cross-effects were also studied with t-tests of two samples comparing group AB measurements (drug / placebo) with the measurements of group BA (placebo / drug). To calculate the effect of the drug on visit 3 for group AB, (v3 - v2) with v3 and v2 denoting, respectively, measurements at visit 3 and visit 2, was used. Similarly (v4 - v2) and (v5 - v2) were used to calculate the effects on visits 4 and 5. To calculate the effect on visit 6 [(v6-v2) + (v3 - v2)] was used . Note that v6 from group BA includes the effect of drug after two weeks which cancels the effect of the drug on visit 3 from group AB. Similarly, [(v7 - v2) + (v4 - v2)] was used to calculate the effect on visit 7. The two AB groups of higher doses were used for all visits. The 3 BA groups are used for visits 3, 4 and 5 since they had received the same treatment until visit 5, but only the two high dose BA groups are used for visits 6 and 7. The sample size for this study was chosen so that each of the three branches is provided, after 5% of defections, at least 80% power (with a = 0.05, double) to detect a relative decrease of 15% for a variable of logarithm response normal, since one trial for that variable has a coefficient of variation of 20% and the intra-person coefficient of variation is as high as 25%. Based on these assumptions, the target recruitment included 180 subjects with randomization in 3 different dose level groups as described above. At the enlistment visit, an independent study nurse who did not observe the content of the drug, provided medication equipment according to a computer generated randomization list. The randomization of patients under study was stratified according to sex. For both strata, a block design combined with block size 12 was used to assign patients in each of the six study sequences. All biomarkers were transformed using a logarithmic transformation change (transformed value is the natural logarithm of the original value plus a change constant for each assay). The missing data were filled using an advance scheme carried by the last simple observation (LOCF), in cases where there is no previous measurement, the following observation was carried out again. Statistical margins for established data are entered based on the IQR distance from the middle term.
Biomarker measurements ELISA and mass spectrometry assays were used to measure MI levels in at-risk biomarkers and are summarized in Table 28. Apart from plasma measurements, LTB4 and MPO were also measured in whole blood preparations. live after ionomycin activation of leukocytes, using ELISA and mass spectrometry. Both dose and time dependent stimulations were performed to determine the result of LTB4 and maximum MPO of the cells. The correction was made for conted of whole blood, when the amount of these mediators produced is related to the number of cells in a fixed volume. On the logarithm scale, the adjustment was based on a linear model, with coefficients determined empirically at the time of blind review. Several tertiary markers were also measured including: IL-6, IL-12p40, TNFa, MMP-9, sICAM, sVCAM, P-selectin, E-selectin, MCP-1 and oxidized LDL.
Table 28: Methods and tests used to quantify study biomarkers Clinical Outcome The baseline values for the biomarker variables before treatment are shown in Table 29. For the primary efficacy endpoint, as specified in the statistical analysis plan, 10 variables were considered in the pooled set of subjects in 500 mg and 750 mg branches and the data are set forth in Table 30. The end point of the study's primary efficacy was confirmed by showing that DG-031 reduces the levels of LTB4 produced by ex vivo ionomycin-activated neutrophils for the pooled set of branches of 500 mg and 750 mg (p nominal = 0.0042), and this is statistically important after the correction for multiple testing. As shown in Table 30, the maximum reduction, in the production of LTB4 and MPO added to 26% for LBT4 (p nominal = 0.0026) and 13% for MPO (p nominal = 0.023) in the dose of 750 mg / day of DG-031. DG-031 also significantly reduced serum sICAM-1 (p = 0.02), but no effects were observed in other tertiary markers. Lp-PLA2 increased 9% (nominal p = 0.0056) in response to the higher dose of DG-031 and there was comparable increase observed in LDL cholesterol (8%) that correlated with Lp-PLA2. In contrast, the effects of the two lower doses (250 mg / day and 500 mg / day) on Lp-PLA2 were not significant. Urine levels of LTE4 increased by 27% in response to the higher dose of DG-031 (nominal p = 0.00002). The significant correlation between the inhibition of LTB4 and MPO production in response to DG-031 was observed (r = 0.65, p <; 0.00001). Table 29. Summary statistics of baseline biomarker values Assay 250 mg / day 500 mg / day 750 mg / day active placebo active placebo active placebo * w.b. = Complete blood base line is not available for LTB4 measured using mass spectrometry tcorr for wbc = corrected for white blood cell count Table 30. Treatment effect based on the t test of two samples for the treatment groups, the groups grouped for the two higher doses and all doses (natural logarithm scale) 250 mg / day 500 mg / day 750 mg / day 500 & 750 mg / day 250, 600 & 760 mg / day Test '"= M> ÍB? IW ÍBs fp = f27) mn / rfwfa = 191) Primary objectives * w.b. = Complete blood aunt measurement is not part of the primary analysis wrt fit for multiple test tcorr. for wbc = corrected for white blood cell count §m.s. = mass spec. = mass spectrometry Tests for cross-effects A cross-effect trial from the treatment phase to the placebo phase was performed as a t-test of two samples in the differences between visit 2 and 5 for patients with drug and placebo, respectively. The group taking the drug consists of patients with treatment of 500 mg / day and 750 mg / day and the placebo group includes patients with placebo from 3 routes. The resulting p-values and confidence intervals for the effect are given in Table 31 (the data for Lp-PLA2 and N-tyrosine were not available). No cross-effects were observed with LTB4 and MPO. In contrast, marked cross-effects were observed for CRP and SAA, with the reduction in CRP that was significant at the 5% level (p = 0.017). SAA showed similar cross-effects that were slightly below this level of importance (p = 0.051). Table 31 Cross-effect test for each study period p-value test 95% Cl effect CRP 0.017 -0.28 [-0, .52, -0.05] Amyloid A 0.051 -0.14 [-0, .29.0.00] LTE4 in urine 0.48 -0.06 [-0, .22,0.10] MCP-1 0.084 0.07 [-0. .01,0.15] MMP 9 0.56 -0.04 [-0 .16,0.09] Plasma MPO 0.28 -0.11 [-0 .31,0.09] Blood cell count 0.57 -0.01 [-0.06,0.03] LTB4 targets in whole blood, 0.45 -0.10 [-0 .36.0.16] corr. for wbct MPO in whole blood, 0.93 0.01 [-0 .13,0.15] corr. for wbct LTB4 in whole blood, 0.45 0.19 [-0 .33,0.71] mass spectrometry LTB4 in whole blood, 0.64 0.12 [-0. .40,0.64] corr. for bc, ms.§ tcorr. for w.b. = corrected for white blood cell count §m.s. = mass spect. = mass spectrometry Figure 10 shows the average effects calculated in CRP and SAA for the subjects receiving the highest drug doses in the first period. Note that measurements from subjects receiving the placebo contribute first to these calculations to nullify potential temporal effects. For visits 3 (after 2 weeks of therapy) and 4 (after 4 weeks of therapy), this constitutes the treatment effect, while cross-effects appear between visits 5 to 7. The level of CRP decreased in the visits 3 and 4, but not significantly. The reduction became more pronounced, approximately 25%, and the importance at visit 5 (p = 0.017), and seems to be persistent until visit 7, during the time the subjects took placebo. This prolonged effect is part of the reason that the effect of the drug was not detected in the primary analysis which did not take this scenario into account. The design of this test does not have maximum power to study such effects which are reflected by the large standard errors in the calculations, particularly for visits 6 and 7. Even though the measurements at visits 3 and 6 are not available for SAA, the observed changes of CRP and Amyloid A between visits 2 and 5 are highly correlated (r = 0.68, p <0.00001). Therefore, it seems that the drug has similar effects in both biomarkers. No difference was detected in the effects of DG-031 on biomarkers of MI risk among patients with FLAP or LTA4 hydrolase haplotypes when the data were analyzed separately. There was no difference in serious episodes between the treatment groups or dose branches in the study group. In particular, no difference was detected in liver transaminases between the groups in active drug or placebo. The only symptom that had significantly more frequency reported for the active drug was dizziness, experienced by 6 patients in active drug (any dose) and none in placebo (p = 0.032). This did not interfere with the daily activities of the subjects. When taken together, the data generated through the genetic isolation of MI (Example 1) and the clinical trial reported here, shows that DG031 is a safe and well-tolerated drug that can, at least in part, correct a biochemical defect that confers a relative risk of acute cardiovascular events that is similar to or greater than the risk conferred by the higher quintile of LDL cholesterol. Rather, the data suggest that DG-031 reduces serum levels of CRP and SAA by approximately 25%, suggesting that this will reduce the risk of acute cardiovascular events.
Example 11: Clinical Trial Investigating the Effect of Compositions Comprising a Leukotriene Synthesis Inhibitor and a Statin in Biomarkers of Risk of Myocardial Infarction A randomized, placebo-controlled, crossover clinical trial, as described in Example 10, is carried out to investigate the effect of the compositions comprising a leukotriene synthesis inhibitor and a statin at the levels of MI risk biomarkers. The participants for the study are optionally carriers of variants in the FLAP and / or LTA4 hydrolase genes set forth in Table 25. A group of participants receives a leukotriene synthesis inhibitor alone, such as DG031. Another group of participants receives a statin alone. A third group of participants receive a composition comprising both a leukotriene synthesis inhibitor and a statin. The fourth group of participants receives a placebo. Each participant receives treatment for at least two months and the levels of the biomarkers established in Table 28 are monitored in each participant for at least three months. It is expected that the group receiving a leukotriene synthesis inhibitor will only have a 25% decrease in CRP levels and the group receiving a statin alone will also have a 25% decrease in CRP levels. More substantial decrease in CRP from combination therapy is evidence that combination therapy is beneficial. In view of the data from the clinical trial described in Example 10; where almost all (approximately 85%) of the participants were on statin therapy, it is expected that the group receiving the combination therapy will exhibit a 50% decrease in CRP levels.
Example 12: Association of variants in the gene encoding ALOX5AP / FLAP to MI in a North American Population As described in Example 1, a variant in the gene encoding the 5-lipoxygenase activation protein (AL0X5AP / FLAP) confers risk for both MI and stroke in Iceland. Another SNP-based haplotype within AL0X5AP, HapB, showed significant association to MI in British groups as described in Example 9, • using similar techniques, the association between HapA and HapB and MI in an American group ("Cleveland") was analyzed. ). The AL0X5AP haplotype HapA is also associated with MI in a North American population. The SNPs that define HapA (SG13S25, SG13S114, SG13S89 and SG13S32) and HapB (SG13S377, SG13S114, SG13S41 and SG13S35) were classified by genotype in 696 patients with MI (553 men and 143 women) and 698 controls (314 men and 384 women ). The majority of the study subjects were Caucasian and approximately 10% were African-American. Ethnicity information on an individual level is not available. The SNP haplotype analysis was done using the NEMO program (Gretarsdottir et al., Nat Genet 35: 131-8, 2003). NEMO handles missing genotypes and phase uncertainty through a probability procedure, using the expectation-maximization algorithm as a calculation tool to calculate haplotype frequencies. For haplotypes at risk we calculated the relative risk (RR) assumed by a multiplicative model (Falk &Rubinstein P Ann Hum Genet 51 (Pt 3): 227-33, 1987: illiger &Ott Hum Hered. 42: 337-46 , 1992) where the risk of the two haplotypes to a person multiplies. The results of the haplotype association analysis for HapA and HapB are shown in Table 32. As demonstrated in the Icelandic population (see Example 1), the calculated frequency of HapA was significantly higher in the patient group than in the control group. In the total group, the allelic / haplotype frequency of HapA was 16.9% and 13.6% in patients and controls respectively (P = 0.014), which corresponds to a 29% increase in risk of MI for each copy of HapA carried. The relative risk of MI in the total group was 1.29 and a P value for the association was 0.014. In addition, HapA was over-represented in patients who had experienced MI relatively early in life (men before 55 and women before 65 years). As shown in Table 32, the relative risk of MI in the initial stage (men before 55 and women before 65 years) was 1.61 and the P value for the association was 0.0034.
Table 32. Association of HapA and HapB with MI HapA HapB Phenotype (n) Frequency RR P Frequency RR P Group Total Controls (698) 0.136 0.074 MI (696) 0.169 1.29 0.014 0.081 1.1 HL Initial stage (170) 0.205 1.61 0.0034 The association of HapB to MI in the study group was also studied. HApB has previously been shown to confer risk to MI in an English group (see Example 9). A slight excess of HapB was observed in the total patient group (8.1%) compared to all controls (7.4%), but was not significant (Table 32). This analysis demonstrated that a haplotype AL0X5AP, HapA, previously reported to confer risk of MI and stroke in an Icelandic group (Example 1), and stroke in a Scottish group (described later in Example 14), is associated with MI in a population North American. HapB that confers MI risk in a British group (Example 9) was not associated with MI in this North American group.
Example 13. Additional AL0X5AP / FLAP Haplotype Associated with MI in a North American Population From the analysis of the Cleveland group described in Example 12, another haplotype was identified, which is significantly associated with MI. This haplotype is denoted as HapC, and 5 variants of this haplotype were identified (HapCl, HapC2, HapC3, HapC4-A, HapC4-B). These haplotypes show the most significant association to MI in the Cleveland group. These haplotypes are defined in Table 33. HapCl is the T allele of the marker SG13S375. HapC2 has the T allele of the SG13S375 marker and the G allele of the SG13S25. HapC3 adds allele A of SG13S32 plus allele T of marker SG13S375 and allele G of SG13S25. The addition of the fourth SNP to SG13S106 divides HapC3 into two parts, or HapC4-A and HapC4-B. The G allele of SNP SG13S25, which is in HapC2, HapC3, HapC4-Á and HapC4-B is also a characteristic of HapA. The frequency of HapCl, HapC2, HapC3 and HapC4-A and B in different populations are shown in Table 33. HapCl, HapC2 and HapC3 are over-represented in patient groups in all tested populations. In groups from Iceland and the United Kingdom studied, the HapC4-A part of HapC3 seems to be the one that captures all the risk conferred by HapC3. All HapC variants except HapC4-B are correlated with HapA, denoting that chromosomes that carry HapC also tend to carry HapA. The correlation between HapC4-A and HapA is defined by a correlation coefficient (R2) of 0.52; the linkage disequilibrium (D ') of 0.77 and the P value (measure of importance) of 6.4 x 10-312. HapC is also correlated with HapB, although HapA and HapB correlate negatively. The correlation between HapC4-A and HapB haplotypes is defined by a correlation coefficient (R2) of 0.08; the linkage disequilibrium (D ') of 0.05 and the P value of 2.2 x 10-39. Table 33. t Lp H or H L? or L? w LO O to what H Lp or L? or L? I HAVE IT Example 14; Association of variants in the gene encoding AL0X5AP / FLAP 'apoplexy in a Scottish Population HapA and HapB haplotype analysis was carried out in a Scottish group as described in Example 1 and 9. The SNPs that define HapA (SG13S25, SG13S114, SG13S89 and SG13S32) and HapB (SG13S377, SG13S114, SG13S41 and SG13S35) were classified by genotype into 450 Scottish stroke patients and 710 controls. The patient and control groups have been previously described (MacLeod et al., Neurology 53: 418-20, 1999; Mei lejohn et al., Stroke 32: 57-62, 2001; Duthie et al., Am J Clin. 75: 908-13, 2002; Whalley et al., Am J Clin. Nutr., 2004). In short, 450 patients from northeastern Scotland with confirmed CT of ischemic stroke (including 26 patients with transient ischemic attacks (TIA)) were recruited between 1997 and 1999, within a week of admission of the acute stroke unit in Aberdeen Royan Infirmary. The patients were also subclassified according to the TOAST research criteria (Adams et al., Stroke 24: 35-41, 1993). One hundred fifty-five patients (34%) had a large vascular apoplexy, 96 (21.3%) had cardiogenic stroke and 109 (24.2%) had small vascular apoplexy. In 5 cases (1.1%), apoplexy was determined with another determined etiology, 7 (1.5%) had more than one etiology, and 78 (17.3%) had an unknown cause of stroke despite extensive evaluation. Seven hundred and ten controls without a history of stroke or AIA were recruited as a part of 1921 (n = 227) and 1936 (n = 371) Aberdeen Birth Cohort Studies (Duthie et al., Am. J. Clin. Nutr. 75: 908- 13, 2002; halley et al. , Am J Clin. Nutr., 2004) and primary care (n = 112) (Meiklejohn et al., Stroke 32: 57-62, 2001). The SNP haplotype analysis was done using the NEMO program (Gretardottir et al., Nat Genet 35: 131-8, 2003). NEMO handles missing genotypes and phase uncertainty through the probability procedure, using the expectation-maximization algorithm as a calculation tool to calculate haplotype frequencies. As the two tested haplotypes had previously been shown to confer risk of MI and stroke in an Icelandic group, and MI in an English group, the P values reported were unilateral. For haplotypes at risk, the relative risk (RR) that adds a multiplicative model was calculated (Falk &Rubinstein P Ann Hum Genet 51 (Pt 3): 227-33, 1987: Terwilliger &Ott Hum Hered. 42: 337-46 , 1992) in which the risk of the two haplotype alleles to a person multiplies. The results of the haplotype association analysis for HapA and HapB are shown in Table 34. The haplotype frequencies of HapA in Scottish stroke and control populations were higher than in the corresponding Icelandic populations. As demonstrated in the Icelandic population, the calculated frequency of HapA was significantly higher in Scottish patients with stroke than in Scottish controls. The carrier frequency of HapA in Scottish patients and controls was 33.4% and 26.4%, respectively, resulting in a high risk of 1.36 (P = 0.007) and a corresponding PAR of 9.6%. In the Icelandic population, a higher frequency of HapA was observed in male patients when compared to female patients with stroke or MI. This gender difference in the frequency of HapA was not observed in the Scottish population.
Table 34. Association of HapA and HapB with ischemic stroke HapA HapB Phenotype (n) Frequency RR P-value Frequency RR value-i? Scotland Controls (710) 0.142 0.058 Ischemic stroke (450) to 0.184 1.36 0.007 0.0S8 1.20 NS Men (253) 0.183 1.35 0.023 0.092 1.65 0.016 Women (181) 0.179 1.34 0.044 0.035 0.58 DK Iceland Controls (624) 0.095 0.07 Ischemic Apoplexy (632) 0.147 1.63 0.00013 0.073 1.09 NS Men (335) 0.155 1.75 0.0002 0.086 1.31 HK Women (297) 0.138 1.51 0.0079 0.058 0.86. NS The association of HapB to stroke in the Scottish group was also investigated. HapB has previously shown that it confers MI irrigation in an English group (Example 9). A slight excess of HapB was observed in the patient group (6.8%) compared to controls (5.8%), but it was not significant (Table 34). However, the gender-specific analysis showed that the frequency of HapB was higher in men with ischemic stroke (9.2%) than in controls, resulting in a RR of 1.65 (P = 0.016). The frequency of HapB in women with ischemic stroke was 3.5% which was lower, but not significantly different from the controls. The frequencies of HapB in men and women with ischemic stroke differed significantly (P = 0.0021). As shown in Table 34, similar trends were observed in the Icelandic group; the frequency of HapB is higher in men with ischemic stroke (8.6%) than in women with ischemic stroke (5.8%) although this was not significant (P = 0.055). In this way, HapA, the haplotype at risk of ALOX5AP, is associated with ischemic stroke in a Scottish group. HapB was not associated with ischemic stroke in the group, Scottish. However, HapB was over-represented in male patients.

Claims (1)

  1. CLAIMS 1. A method for evaluating a human subject, for susceptibility to myocardial infarction (MI), the method comprises analyzing the nucleic acid of a human subject for the presence or absence of a haplotype of 5-lipoxygenase activation protein (FLAP) ), wherein the FLAP haplotype is selected from the group consisting of: (a) a FLAP haplotype comprising markers SG13S377 (SEQ ID NO: 1, position 169965), allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G; (b) a FLAP haplotype comprising marker SG13S375 (SEQ ID NO: 1, position 164874), T allele; Y (c) a FLAP haplotype comprising markers SG13S25 (SEQ ID NO: 1, position 165553), G allele; SG13S99 (DG00AAFIU), allele T (SEQ ID NO: 1, position 138551); SG13S377 (DG00AAJFF) (SEQ ID NO: 1, position 169965), allele G; SG13S106 [SNP DG00AAHII] (SEQ ID NO: 1, position 176579), allele G; SG13S32 (SEQ ID NO: 1, position 198547), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G; and to evaluate the susceptibility to MI from the presence or absence of the haplotype, where the presence of the FLAP haplotype correlates with an increased susceptibility for myocardial infarction. The method of claim 1, further comprising isolating the nucleic acid from a biological sample of the human subject for analysis in the examination step. 3. The use of a therapeutic agent for the preparation of a medicament for prophylaxis therapy for myocardial infarction (MI) in a human subject identified as having a FLAP haplotype selected from the group consisting of: (a) a FLAP haplotype comprising markers SG13S377 (SEQ ID NO: 1, position 169965), allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G; (b) a FLAP haplotype comprising a marker SG13S375 (SEQ ID NO: 1, position 164874), T allele; Y (c) a FLAP haplotype comprising markers SG13S25 (SEQ ID NO: 1, position 165553), G allele; SG13S99 (DG00AAFIU), allele T (SEQ ID NO: 1, position 138551); SG13S377 (DG00AAJFF) (SEQ ID NO: 1, position 169965), allele G; SG13S106 [SNP DG00AAHII] (SEQ ID NO: 1, position 176579), allele G; SG13S32 (SEQ ID NO: 1, position 198547), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G; wherein the therapeutic agent is an inhibitor of leukotriene synthesis. . The method or use according to any of claims 1-3, wherein the FLAP haplotype comprises markers: SG13S377 (SEQ ID NO: 1, position 169965), allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and - SG13S35 (SEQ ID NO: 1, position 206117), allele G. The method or use according to claim 4, wherein the haplotype further comprises the marker SG13S375 (SNP DG00AAJFC) (SEQ ID NO: 1, position 165874), T allele; and SG13S25 (SEQ ID NO: 1, position 165553), allele G. 6. The method or use according to claim 4, wherein the haplotype further comprises the marker. SG13S375 (SNP DG00AAJFC) (SEQ ID NO: 1, position 164874), T allele; and SG13S25 (SEQ ID NO: 1, position 165553), allele G and SG13S32 (SEQ ID NO: 1, position 198547) allele A. 7. The method or use according to any of the claims 1-3, wherein the haplotype comprises marker SG13S375 (SEQ ID NO: 1, position 164874), allele T. 8. The method or use of claim 7, wherein the haplotype further comprises the marker SG13S25 (SEC. IDENT. DO NOT. : 1, position 165553), allele G. 9. The method or use of claim 8, wherein the haplotype further comprises SG13S32 (SEQ ID NO: 1, position 176579), allele A. 10. The method or use of claim 9, wherein the haplotype further comprises the marker SG13S106 (SEQ ID NO: 1, position 198547), allele G or A. 11. The method or use according to any of claims 1-3. , wherein the haplotype comprises markers: SG13S25 (SEQ ID NO: 1, position 165553), allele G; SG13S99 (DG00AAFIU), allele T (SEQ ID NO: 1, position 138551); SG13S377 (DG00AAJFF) (SEQ ID NO: 1, position 169965), allele G; SG13S106 [SNP DG00AAHI] (SEQ ID NO: 1, position 176579), allele G; SG13S32 (SEQ ID NO: 1, position 198547), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G. 12. The use according to any of claims 3-11, wherein the medicament comprises an amount of the effective therapeutic agent to reduce the level of at least one inflammatory marker selected from reactive protein C and myeloperoxidase (MPO) in the human subject during prophylaxis therapy. The use according to any of claims 3-12, wherein the medicament comprises an amount of the therapeutic agent effective to reduce the level of leukotriene in a human subject during prophylaxis therapy, wherein the reduction in the leukotriene level it is measured in a serum, plasma or urine sample from the human subject during the prophylaxis treatment. 1 . The use of claim 13, wherein the reduction in the level of leukotriene is measured ex vivo in a blood sample from the human subject, wherein the blood sample is stimulated with a calcium ionophore before measuring the production of leukotriene. 15. The use according to any of claims 3-14, wherein the medicament comprises the therapeutic agent in an effective condition to reduce the level of leukotriene in the subject below an average level of leukotrienes in human subjects. 16. A composition comprising a leukotriene synthesis inhibitor and a statin. 17. A composition according to claim 16, further comprising a pharmaceutically acceptable carrier. 18. The use according to any of claims 3-15, or the composition according to claim 16 or 17, wherein the leukotriene synthesis inhibitor is an agent that inhibits the activity of a synthetic pathway protein. leukotriene selected from the group consisting of 5-lipoxygenase, 5-lipoxygenase activation protein (FLAP), leukotriene C4 synthase, leukotriene A4 hydrolase, arachidonate 4-lipoxygenase, leukotriene B4 12-hydroxideshydrogenase; leukotriene A4 receptor, leukotriene B4 receptor, leukotriene C4 receptor, leukotriene D4 receptor, leukotriene E4 receptor, leukotriene B4 receptor 1, leukotriene B4 receptor 2, cysteinyl leukotriene receptor 1, and cysteinyl leukotriene receptor 2. 19. The use according to any of claims 3-15, or the composition according to any of claims 16-18, wherein the leukotriene synthesis inhibitor is selected from the group consisting of acid 1- ( (4-chlorophenyl) methyl) -3- ((1,1-dimethylethyl) thio) -alpha, alpha-dimethyl-5- (2-quinolinylmethoxy) -lH-indol-2-propanoic acid (R) - (+ ) -alpha-cyclopentyl-4- (2-quinolinylmethoxy) -benzeneacetic acid, 3- (3- (1, l-dimethylethylthio-5- (quinolin-2-ylmethoxy) -1- (4-chloromethylphenyl) indole-2-} il) -2, 2-dimethylpropionaldehyde oxime-0-2-acetic, zileuton, atreleuton, 6- ((3-fluoro-5- (tetrahydro-4-methoxy-2H-pyran-4-yl) phenoxy) methyl) - 1-methyl-2 (1H) -quinolinone, 1- ((4-chlorophenyl) methyl) -3- ((1,1-dimethylethyl) thio) -alpha, alpha-dimethyl-5- (2-quinolinylmethoxy) -1H -indol-2-propanoic acid 4- (3- (4- (2-methyl-imidazol-1-yl) -phenylsulfañyl) -phenyl) -tetrahydro-pyran-4-carboxylic acid amide 20. Use according to to any of the claims 3-15, or the composition according to any of claims 16-18, wherein the leukotriene synthesis inhibitor is a FLAP inhibitor. The use or composition according to claim 20, wherein the FLAP inhibitor comprises a compound represented by the formula: or a pharmaceutically acceptable salt thereof, wherein R1 represents a group of the formula: R2 and R3 are identical or different and represent hydrogen, lower alkyl, phenyl, benzyl or a group of the formula: -CH- -C02Rs -CH- -CH, -OR ° R 4 represents hydrogen, lower alkyl, phenyl or benzyl, which may be optionally substituted by hydroxyl, carboxyl, lower alkoxycarbonyl, lower alkylthio, heteroaryl or carbamoyl, R 5 represents hydrogen, lower alkyl, phenyl or benzyl, R 6 represents a group of the formula - COR5 or -C02R5, R7 represents hydrogen, lower alkyl or phenyl, Y represents a group of the formula: wherein R represents hydrogen, lower alkyl or phenyl and n denotes a number from 0 to 5, Z represents norbornyl, or represents a group of the formula: wherein R9 and R10 are identical or different and denote hydrogen, lower alkyl or phenyl, or R9 and R10 can together form a saturated carbocyclic ring having up to 6 carbon atoms and m denotes a number from 1 to 6, and A and B are identical or different and denote hydrogen, lower alkyl or halogen, or a pharmaceutically acceptable salt thereof. The use or composition according to claim 20, wherein the FLAP inhibitor comprises a compound selected from the group consisting of: 2- [4- (quinolin-2-yl-methoxy) phenyl] -2- cyclopentylacetic, 2- [4- (quinolin-2-yl-methoxy) phenyl] -2-cyclohexylacetic acid, and 2- [4- (quinolin-2-yl-methoxy) phenyl] -2-cycloheptylacetic acid, acid (+ ) - 2- [4- (quinolin-2-yl-methoxy) phenyl] -2-cyclopentyl acetic acid enantiomer, 2- (4- (quinolin-2-yl-methoxy) phenyl] - (-) - enantiomer 2-cyclopentylacetic and pharmaceutically acceptable salts thereof. 23. The use or composition according to claim 20, wherein the FLAP inhibitor comprises BAY-X-1005 or a pharmaceutically acceptable salt, formulation or prodrug thereof. 2 . The use or composition according to claim 20, wherein the leukotriene synthesis inhibitor is (R) - (+) - alpha-cyclopentyl-4- (2-quinolinylmethoxy) -Benzenacetic acid. 25. The composition according to any of claims 16-24, wherein the statin is selected from the group consisting of rovuvastatin, fluvastatin, atorvastatin, lovastatin, simvastatin, pravastatin or pitavastatin. 26. The composition according to any of claims 16-25, wherein the leukotriene synthesis inhibitor is included in the composition in an amount effective to reduce the serum reactive protein C (CRP) in a human subject. The composition according to any of claims 16-26, wherein the statin is included in the composition in an amount effective to reduce low density lipoprotein cholesterol (LDL) and reduce serum CRP in a human subject. . The composition according to any of claims 16-27, wherein the leukotriene inhibitor and the statin are included in the composition in effective amounts to synergistically reduce the reactive protein C in serum in a human subject. 29. A unit dose of the composition of any of claims 16-28 for administration to a human subject. 30. The composition or unit dose of any of claims 16-29, which is a pill or a capsule. 31. The composition or unit dose according to any of claims 16-30, which contains 50 to 750 milligrams of the FLAP inhibitor. 32. The composition or unit dose according to claim 31, which contains 250 to 375 milligrams of the FLAP inhibitor. 33. The composition or unit dose according to any of claims 16-32, which contains 1 to 200 milligrams of the statin. 34. The composition or unit dose according to claim 33, which contains 5 to 80 milligrams of the statin. 35. The use of the composition of any of claims 16-34 in the manufacture of a medicament for reducing serum LDL, leukotrienes in serum, and reactive protein C (CRP) in a human subject. 36. The use of claim 35, wherein the human subject is at risk for a disease or condition selected from the group consisting of myocardial infarction, acute coronary syndrome, stroke, or peripheral arterial occlusive disease. 37. A method for monitoring the efficiency of prophylaxis therapy for myocardial infarction (MI) in a human subject, wherein the prophylaxis therapy comprises administration of a leukotriene synthesis inhibitor to the subject, the method comprising monitoring the efficacy of therapy by measuring myeloperoxidase (MPO) in samples from the human subject before and during prophylaxis therapy, wherein an effective prophylaxis therapy reduces the level of MPO during prophylaxis therapy below the MPO level prior to therapy. prophylaxis. 38. The method of claim 37, further comprising the step of monitoring at least one additional inflammatory marker in samples from the human subject during prophylaxis therapy. 39. The method according to claim 38, wherein the additional inflammatory marker is a reactive protein C (CRP). 40. The method according to any of claims 37-39, wherein the monitoring further comprises monitoring a level of leukotriene in serum, plasma or urine from the human subject before and during the prophylaxis treatment, wherein a therapy of Effective prophylaxis reduces leukotriene level. 41. The method according to claim 40, wherein the monitoring comprises measuring leukotriene production ex vivo in a blood sample from the human subject, wherein the blood sample is stimulated with a calcium ionophore before measuring the production of leukotriene. 42. The method according to any of claims 37-41, wherein the prophylaxis therapy comprises the administration of an agent that inhibits FLAP activity. 43. The method according to any of claims 37-42, wherein the human subject has a FLAP haplotype selected from the group consisting of: (a) a FLAP haplotype comprising markers SG13S377 (SEQ ID NO: 1, position 169965), allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G; (b) a FLAP haplotype comprising marker SG13S375 (SEQ ID NO: 1, position 164874), T allele; Y (c) a FLAP haplotype comprising markers SG13S25 (SEQ ID NO: 1, position 165553), G allele; SG13S99 (DG00AAFIU), allele T (SEQ ID NO: 1, position 138551); SG13S377 (DG00AAJFF) (SEQ ID NO: 1, position 169965), allele G; SG13S106 [SNP DG00AAHII] (SEQ ID NO: 1, position 176579), allele G; SG13S32 (SEQ ID NO: 1, position 198547), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G; 44. The use of a leukotriene synthesis inhibitor in the preparation of a medicament for prophylaxis for myocardial infarction (MI) in a human subject, wherein the medicament comprises an amount of leukotriene synthesis inhibitor effective to inhibit the synthesis of leukotriene in vivo and reduces the level of myeloperoxidase (MPO) level in the human subject during prophylaxis treatment. 45. A kit comprising reagents for carrying out the method of any one of claims 11 or 37-43. 46. A reagent for evaluating a sample for the presence of a FLAP haplotype comprising the markers: SG13S377 (SEQ ID NO: 1, position 169965), allele A; SG13S114 (SEQ ID NO: 1, position 178096), allele A; SG13S41 (SEQ ID NO: 1, position 202045), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G. 47. A reagent of claim 46, wherein the haplotype further comprises the marker SG13S375 (SNP DG00AAJFC) (SEQ ID NO. : 1, position 164874), allele T; and SG13S25 (SEQ ID NO: 1, position 165553), allele G. 48. A reagent of claim 47, wherein the haplotype further comprises the marker SG13S375 (SNP DG00AAJFC) (SEQ ID NO. : 1, position 164874), allele T; and SG13S25 (SEQ ID NO: 1, position 165553), allele G and SG13S32 (SEQ ID NO: 1, position 198547), allele A. 49. A reagent for evaluating a sample for the presence of a FLAP haplotype comprising a marker SG13S375 (SEQ ID NO: 1, position 164874), allele T. 50. A reagent of claim 49, wherein the haplotype further comprises a marker SG13S25 (SEQ ID. NO .: 1, position 165553), allele G. 51. A reagent of claim 50, wherein the haplotype further comprises SG13S32 (SEQ ID NO: 1, position 176579), allele A. 52. A reagent of claim 51, wherein the haplotype further comprises a marker SG13S106 (SEQ ID NO: 1, position 198547), allele G or A. 53. A reagent for evaluating a sample for the presence of a haplotype FLAP comprising the markers: SG13S25 (SEQ ID NO: 1, position 165553), allele G; SG13S99 (DGOOAAFIU), allele T (SEQ ID NO: 1, position 138551); SG13S377 (DG00AAJFF) (SEQ ID NO: 1, position 169965), allele G; SG13S106 [SNP DG00AAHII] (SEQ ID NO: 1, position 176579), allele G; SG13S32 (SEQ ID NO: 1, position 198547), allele A; and SG13S35 (SEQ ID NO: 1, position 206117), allele G. 54. A reagent kit for evaluating a sample for the presence of a FLAP haplotype, comprising in separate containers, (a) one or more reagents according to any of claims 46-53; wherein the reagent is a probe that hybridizes to such FLAP haplotype markers and wherein the probe comprises a detectable label; and (b) reagents for the detection of such a label. 55. A reagent kit for evaluating a sample for the presence of a FLAP haplotype, comprising in separate containers, (a) one or more reagents according to any of claims 46-53; wherein the reagent is a primer for the amplification of a region in the FLAP nucleic acid comprising at least one of the FLAP haplotype markers, and (b) reagents for the amplification of the region in the FLAP nucleic acid.
MXPA06008516A 2004-01-30 2005-01-31 Susceptibility gene for myocardial infraction, stroke, and paod; methods of treatment. MXPA06008516A (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US76954204A 2004-01-30 2004-01-30
US10/769,744 US20050282855A1 (en) 2002-10-17 2004-01-30 Susceptibility gene for myocardial infarction, stroke, and PAOD; methods of treatment
US10/830,477 US7851486B2 (en) 2002-10-17 2004-04-22 Susceptibility gene for myocardial infarction, stroke, and PAOD; methods of treatment
US10/829,674 US7507531B2 (en) 2002-10-17 2004-04-22 Use of 5-lipoxygenase activating protein (FLAP) gene to assess susceptibility for myocardial infarction
US64290905P 2005-01-10 2005-01-10
PCT/US2005/003312 WO2005075022A2 (en) 2004-01-30 2005-01-31 Susceptibility gene for myocardial infraction, stroke, and paod; methods of treatment

Publications (1)

Publication Number Publication Date
MXPA06008516A true MXPA06008516A (en) 2007-03-21

Family

ID=40262935

Family Applications (1)

Application Number Title Priority Date Filing Date
MXPA06008516A MXPA06008516A (en) 2004-01-30 2005-01-31 Susceptibility gene for myocardial infraction, stroke, and paod; methods of treatment.

Country Status (1)

Country Link
MX (1) MXPA06008516A (en)

Similar Documents

Publication Publication Date Title
US8158362B2 (en) Methods of diagnosing susceptibility to myocardial infarction and screening for an LTA4H haplotype
US20060019269A1 (en) Susceptibility gene for myocardial infarction, stroke, and PAOD, methods of treatment
JP5301987B2 (en) Genetic variants in the TCF7L2 gene as a diagnostic marker for risk of type 2 diabetes
US20080146540A1 (en) Methods of diagnosis and treatment for asthma, allergic rhinitis and other respiratory diseases based on haplotype association
US20050287551A1 (en) Susceptibility gene for human stroke; methods of treatment
US20030054531A1 (en) Human stroke gene
US20050272051A1 (en) Methods of preventing or treating recurrence of myocardial infarction
US7851486B2 (en) Susceptibility gene for myocardial infarction, stroke, and PAOD; methods of treatment
US20050164220A1 (en) Susceptibility gene for human stroke: method of treatment
AU2004257748A1 (en) Method of diagnosis and treatment for asthma based on haplotype association
WO2005075022A2 (en) Susceptibility gene for myocardial infraction, stroke, and paod; methods of treatment
US20050282855A1 (en) Susceptibility gene for myocardial infarction, stroke, and PAOD; methods of treatment
AU2003290527A1 (en) Susceptibility gene for myocardial infarction
US20080293750A1 (en) Susceptibility Gene for Myocardial Infarction, Stroke, Paod and Methods of Treatment
US20100216863A1 (en) Susceptibility Gene for Myocardial Infarction, Stroke, and PAOD; Methods of Treatment
MXPA06008516A (en) Susceptibility gene for myocardial infraction, stroke, and paod; methods of treatment.
US20040014099A1 (en) Susceptibility gene for human stroke; methods of treatment
JP2006500068A (en) Human stroke susceptibility gene; therapy
WO2005123964A2 (en) Susceptibility gene for human stroke: methods of treatment

Legal Events

Date Code Title Description
FA Abandonment or withdrawal