WO2007149798A2 - Biomarkers for the progression of alzheimer's disease - Google Patents

Biomarkers for the progression of alzheimer's disease Download PDF

Info

Publication number
WO2007149798A2
WO2007149798A2 PCT/US2007/071421 US2007071421W WO2007149798A2 WO 2007149798 A2 WO2007149798 A2 WO 2007149798A2 US 2007071421 W US2007071421 W US 2007071421W WO 2007149798 A2 WO2007149798 A2 WO 2007149798A2
Authority
WO
WIPO (PCT)
Prior art keywords
leu
disease
ser
alzheimer
giu
Prior art date
Application number
PCT/US2007/071421
Other languages
French (fr)
Other versions
WO2007149798A3 (en
Inventor
Yunsheng He
Baltazar Gomez-Mancilla
Joanne Meyer
Giorgio Rovelli
Rainer R. Kuhn
Graeme Bilbe
Original Assignee
Novartis Ag
Novartis Pharma Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag, Novartis Pharma Gmbh filed Critical Novartis Ag
Priority to US12/305,053 priority Critical patent/US20100035251A1/en
Priority to EP07798679A priority patent/EP2035582A2/en
Priority to BRPI0713738-9A2A priority patent/BRPI0713738A2/en
Priority to JP2009516649A priority patent/JP2009541336A/en
Priority to AU2007261095A priority patent/AU2007261095A1/en
Priority to MX2008016524A priority patent/MX2008016524A/en
Priority to CA002657980A priority patent/CA2657980A1/en
Publication of WO2007149798A2 publication Critical patent/WO2007149798A2/en
Publication of WO2007149798A3 publication Critical patent/WO2007149798A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/172Haplotypes

Definitions

  • This invention relates generally to the analytical testing of tissue samples in vitro, and more particularly to aspects of genetic polymorphisms of the leucine-rich repeat kinase 2 (LRRK2) gene.
  • LRRK2 leucine-rich repeat kinase 2
  • Therapy specific diagnostics (a.k.a., theranostics) is an emerging medical technology field, which provides tests useful to diagnose a disease, choose the correct treatment regime and monitor a subject's response. That is, theranostics are useful to predict and assess drug response in individual subjects, i.e., individualized medicine. Theranostic tests are also useful to select subjects for treatments that are particularly likely to benefit from the treatment or to provide an early and objective indication of treatment efficacy in individual subjects, so that the treatment can be altered with a minimum of delay.
  • SNPs single nucleotide polymorphisms
  • LRRK2 leucine-rich repeat kinase 2
  • AD Alzheimer's disease
  • mutations of the LRRK2 gene causing a change in the protein such as T1602S and T2352, in particular T2352M
  • T1602S and T2352, in particular T2352M may have an impact on Alzheimer's disease and can be used as a biomarker of Alzheimer's disease progression and age-at-onset of Alzheimer's disease.
  • the invention provides for the use of a LRRK2 modulating agent in the manufacture of a medicament for the treatment of Alzheimer's disease a selected patient population.
  • the patient population is selected on the basis of polymorphisms in theLRRK2 gene that are indicative of progression from mild cognitive impairment (MCI) to Alzheimer's disease.
  • the LRRK2 modulating agent is a heterocyclic compound that slows the progression by the patient from mild cognitive impairment to Alzheimer's disease.
  • the LRRK2 modulating agent is a heterocyclic compound that slows the progression by the patient from moderate Alzheimer's disease to severe Alzheimer's disease.
  • the polymorphism in the LRRK2 gene can be T 1602 S or T2352. The invention also provides methods for the predicting Alzheimer's disease progression or age-at-onset of Alzheimer's disease.
  • FIG 1 is a depiction of the LRRK2 protein structure and location of the two common
  • LRRK2 polymorphisms T1602S and T2352M.
  • LRRK2-T1602S was significantly associated with conversion from mild cognitive impairment to Alzheimer's disease.
  • the mild cognitive impairment patients with TT genotype were at greater risk to progress to Alzheimer's disease.
  • the LRRK2-T2352 also showed a trend for conversion to Alzheimer's disease.
  • the mild cognitive impairment patients with CC genotype tended to progress to Alzheimer's disease.
  • LRRK2-T1602S and LRRK2-T2352 showed a same trend of the association observed in the mild cognitive impairment study.
  • the Alzheimer's disease patients with TT genotype of LRRK2-T1602S or CC genotype of LRRK2-T2352 tended to decline faster on cognitive performance over 6 months, especially in the presence of a BuChE-K variant.
  • LRRK2 may play a role in Alzheimer's disease pathogenesis, especially disease progression and that polymorphisms of LRRK2 can be used as biomrkers of this progression.
  • the human LRKK2 gene (SEQ ID NO : 1 ) is located in the P ARK8 locus on chromosome 12ql2. The gene has multiple-domains.
  • LRKK2 protein (SEQ ID NO:2)is a receptor interacting protein (RIP) kinase.
  • the G2019S mutation is the most common pathogenic mutation of LRRK2 (5-6% of autosomal dominant and ⁇ 1% of sporadic late-onset cases).
  • the G2019S mutation is located in the kinase domain of the LRKK2 gene.
  • FIG. 1 shows LRRK2 protein structure and location of two other common polymorphisms.
  • T1602S and T2352M are common polymorphisms; and (2) they are missense polymorphisms.
  • Amino acid change caused by the polymorphisms are as follows: T1602S - Thr -> Ser (1602 A>T); T2352M - Thr -> Met (2352 C>T).
  • LD linkage disequilibrium
  • the various aspects of the present invention relate to diagnostic/theranostic methods and kits that use the LRRK2 mutations of the invention to identify individuals predisposed to disease or to classify individuals with regard to drug responsiveness, side effects, or optimal drug dose.
  • the invention provides methods for compound validation and a computer system for storing and analyzing data related to the LRRK2 mutations of the invention. Accordingly, various particular embodiments that illustrate these aspects follow.
  • allele means a particular form of a gene or DNA sequence at a specific chromosomal location (locus).
  • the term “antibody” includes, but is not limited to, polyclonal antibodies, monoclonal antibodies, humanized or chimeric antibodies and biologically functional antibody fragments sufficient for binding of the antibody fragment to the protein. Antibodies can be used in assays to determine the presence of variant proteins and peptides where the genetic polymorphisms of the invention are in the coding region of the gene..
  • the term “clinical response” means any or all of the following: a quantitative measure of the response, no response, and adverse response (i.e., side effects).
  • phase I phase II
  • phase III phase III clinical trials. Standard methods are used to define the patient population and to enrol subjects.
  • the term "effective amount" of a compound is a quantity sufficient to achieve a desired therapeutic and/or prophylactic effect, for example, an amount which results in the prevention of or a decrease in the symptoms associated with a disease that is being treated, e.g., the diseases associated with LRRK2 mutant polynucleotides and mutant polypeptides identified herein (particularly Alzheimer's disease and Parkinson's disease).
  • the amount of compound administered to the subject will depend on the type and severity of the disease and on the characteristics of the individual, such as general health, age, sex, body weight and tolerance to drugs. It will also depend on the degree, severity and type of disease. The skilled artisan will be able to determine appropriate dosages depending on these and other factors.
  • an effective amount of the compounds of the present invention sufficient for achieving a therapeutic or prophylactic effect, range from about 0.000001 mg per kilogram body weight per day to about 10,000 mg per kilogram body weight per day.
  • the dosage ranges are from about 0.0001 mg per kilogram body weight per day to about 100 mg per kilogram body weight per day.
  • the compounds of the present invention can also be administered in combination with each other, or with one or more additional therapeutic compounds.
  • expression includes but is not limited to one or more of the following: transcription of the gene into precursor mRNA; splicing and other processing of the precursor mRNA to produce mature mRNA; mRNA stability; translation of the mature mRNA into protein (including codon usage and tRNA availability); and glycosylation and/or other modifications of the translation product, if required for proper expression and function.
  • gene means a segment of DNA that contains all the information for the regulated biosynthesis of an RNA product, including promoters, exons, introns, and other untranslated regions that control expression.
  • genotype means an unphased 5' to 3' sequence of nucleotide pairs found at one or more polymorphic sites in a locus on a pair of homologous chromosomes in an individual.
  • genotype includes a full-genotype and/or a sub-genotype.
  • locus means a location on a chromosome or DNA molecule corresponding to a gene or a physical or phenotypic feature, in particular the LRRK2 gene.
  • LRRK2 modulating agent is any compound that alters (e.g., increases or decreases) the expression level or biological activity level of LRRK2 polypeptide compared to the expression level or biological activity level of LRRK2 polypeptide in the absence of the LRRK2 modulating agent.
  • LRRK2 modulating agent can be a small molecule, polypeptide, carbohydrate, lipid, nucleotide, or combination thereof.
  • the LRRK2 modulating agent may be an organic compound or an inorganic compound.
  • mutant means any heritable variation from the wild-type that is the result of a mutation, e.g., single nucleotide polymorphism.
  • mutant is used interchangeably with the terms “marker”, “biomarker”, and “target” throughout the specification.
  • the term "medical condition” includes, but is not limited to, any condition or disease manifested as one or more physical and/or psychological symptoms for which treatment is desirable, and includes previously and newly identified diseases and other disorders.
  • nucleotide pair means the nucleotides found at a polymorphic site on the two copies of a chromosome from an individual.
  • polymorphic site means a position within a locus at which at least two alternative sequences are found in a population, the most frequent of which has a frequency of no more than 99%.
  • phased means, when applied to a sequence of nucleotide pairs for two or more polymorphic sites in a locus, the combination of nucleotides present at those polymorphic sites on a single copy of the locus is known.
  • polymorphism means any sequence variant present at a frequency of >1% in a population.
  • the sequence variant may be present at a frequency significantly greater than 1% such as 5% or 10 % or more.
  • the term may be used to refer to the sequence variation observed in an individual at a polymorphic site.
  • Polymorphisms include nucleotide substitutions, insertions, deletions and microsatellites and may, but need not, result in detectable differences in gene expression or protein function.
  • polynucleotide means any RNA or DNA, which may be unmodified or modified RNA or DNA.
  • Polynucleotides include, without limitation, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, RNA that is mixture of single- and double-stranded regions, and hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions.
  • polynucleotide refers to triple-stranded regions comprising RNA or DNA or both
  • polypeptide means any polypeptide comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres. Polypeptide refers to both short chains, commonly referred to as peptides, glycopeptides or oligomers, and to longer chains, generally referred to as proteins. Polypeptides may contain amino acids other than the 20 gene-encoded amino acids.
  • Polypeptides include amino acid sequences modified either by natural processes, such as post- translational processing, or by chemical modification techniques that are well-known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature.
  • SNP nucleic acid means a nucleic acid sequence, which comprises a nucleotide that is variable within an otherwise identical nucleotide sequence between individuals or groups of individuals, thus existing as alleles. Such SNP nucleic acids are preferably from about 15 to about 500 nucleotides in length.
  • the SNP nucleic acids may be part of a chromosome, or they may be an exact copy of a part of a chromosome, e.g., by amplification of such a part of a chromosome through PCR or through cloning.
  • SNPs The SNP nucleic acids are referred to hereafter simply as "SNPs”.
  • a SNP is the occurrence of nucleotide variability at a single position in the genome, in which two alternative bases occur at appreciable frequency (i.e., >1%) in the human population.
  • a SNP may occur within a gene or within intergenic regions of the genome.
  • SNP probes according to the invention are oligonucleotides that are complementary to a SNP nucleic acid.
  • the term "subject” means that preferably the subject is a mammal, such as a human, but can also be an animal, e.g., domestic animals (e.g., dogs, cats and the like), farm animals (e.g., cows, sheep, pigs, horses and the like) and laboratory animals (e.g., monkey (e.g., cynmologous monkey, rats, mice, guinea pigs and the like).
  • the administration of an agent or drug to a subject or patient includes self-administration and the administration by another. It is also to be appreciated that the various modes of treatment or prevention of medical conditions as described are intended to mean “substantial", which includes total but also less than total treatment or prevention, and wherein some biologically or medically relevant result is achieved.
  • the LRRK2 modulating agent can be a hetrocyclic compound inhibitor of LRRK2 protein (SEQ ID NO:2).
  • the heterocyclic compound can be 5-[5-Methoxy-2-oxo-l,2- dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl- 1 H-pyrrole-3-carboxylic acid (3-amino- propyl)-amide; 5-[6-Methoxy-2-oxo- 1 ,2-dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl- 1 H- pyrrole-3-carboxylic acid (3-amino-propyl)-amide; 5-[7-Methoxy-2-oxo-l,2-dihydro-indol- (3Z)-ylidenemethyl]-2,4-dimethyl-l H-pyr
  • the heteroccyclic compound can be 3-[l-(3,5-Dimethyl-lH- pyrrol-2-yl)-methyl-(Z)-ylidene]-5-methoxy-l,3-dihydro-indol-2-one; 3-[l-(lH-Indol-2-yl)- meth-(Z)-ylidene]-5-methoxy-l,3-dihydro-indol-2-one; 5-Methoxy-3-[l-(4,5,6,7-tetrahydro- l H-indol-2-yl)-meth-(Z)-ylidene]-l,3-dihydro-indol-2-one; 3-[l-(3,5-Dimethyl-lH-pyrrol-2- yl)-meth-(Z)-ylidene]-5-methoxy-2-oxo-2,3-dihydro-indol-4-carbox
  • the pharmacological properties of the LRRK2 modulating agents can be evaluated, for example, in Drug Pull-Down experiments.
  • the above-mentioned heterocyclic compounds can show activity in Drug Pull-Down experiments at concentrations below 20 ⁇ M.
  • Compound 12 shows an IC 50 value of ⁇ 1 ⁇ M.
  • the LRRK2 gene (SEQ ID NO: l)may play a role in progression from mild cognitive impairment to Alzheimer's disease and progression from moderate Alzheimer's disease to more severe Alzheimer's disease. Therefore, the LRRK2 modulating agents may be able to be used to treat patients with MCI or Alzheimer's disease, to slow the progression from mild cognitive impairment to Alzheimer's disease or from moderate Alzheimer's disease to more severe Alzheimer's disease.
  • SNPs have the potential to be important tools for locating genes that are involved in human disease conditions. See e.g., Wang et al, Science 280: 1077-1082 (1998). It is increasingly clear that the risk of developing many common disorders and the metabolism of medications used to treat these conditions are substantially influenced by underlying genomic variations, although the effects of any one variant might be small.
  • a SNP is said to be "allelic" in that due to the existence of the polymorphism, some members of a species may have an unmutated sequence ⁇ i.e., the original allele) whereas other members may have a mutated sequence ⁇ i.e., the variant or mutant allele).
  • An association between a SNP and a particular phenotype does not necessarily indicate or require that the SNP is causative of the phenotype. Instead, the association may merely be due to genome proximity between a SNP and those genetic factors actually responsible for a given phenotype, such that the SNP and said genetic factors are closely linked. That is, a SNP may be in linkage disequilibrium ("LD") with the "true" functional variant. LD ⁇ a.k.a., allelic association) exists when alleles at two distinct locations of the genome are more highly associated than expected. Thus, a SNP may serve as a marker that has value by virtue of its proximity to a mutation that causes a particular phenotype.
  • nucleic acid molecules containing the gene may be complementary double stranded molecules and thus reference to a particular site on the sense strand refers as well to the corresponding site on the complementary antisense strand. That is, reference may be made to the same polymorphic site on either strand and an oligonucleotide may be designed to hybridize specifically to either strand at a target region containing the polymorphic site.
  • the invention also includes single-stranded polynucleotides that are complementary to the sense strand of the genomic variants described herein.
  • SNPs Single-strand conformation polymorphism (SSCP) analysis, heteroduplex analysis by denaturing high-performance liquid chromatography (DHPLC) and direct DNA sequencing and computational methods. Shi et al, Clin. Chem. 47:164-172 (2001). There is a wealth of sequence information in public databases.
  • SNP-typing methods currently include hybridization, primer extension, and cleavage methods. Each of these methods must be connected to an appropriate detection system. Detection technologies include fluorescent polarization (Chan et al., Genome Res.
  • Polymorphisms can also be detected using commercially available products, such as INVADERTM technology (available from Third Wave Technologies Inc. Madison, Wisconsin, USA).
  • INVADERTM technology available from Third Wave Technologies Inc. Madison, Wisconsin, USA.
  • a specific upstream "invader” oligonucleotide and a partially overlapping downstream probe together form a specific structure when bound to complementary DNA template. This structure is recognized and cut at a specific site by the Cleavase enzyme, resulting in the release of the 5' flap of the probe oligonucleotide. This fragment then serves as the "invader” oligonucleotide with respect to synthetic secondary targets and secondary fluorescently labelled signal probes contained in the reaction mixture.
  • polymorphisms may also be determined using a mismatch detection technique including, but not limited to, the RNase protection method using riboprobes (Winter et al, Proc. Natl. Acad. ScL USA 82:7575 (1985); Meyers et al, Science 230:1242 (1985)) and proteins which recognize nucleotide mismatches, such as the E.
  • variant alleles can be identified by single strand conformation polymorphism (SSCP) analysis (Orita et ah, Genomics 5:874-879 (1989); Humphries et al, in Molecular Diagnosis of Genetic Diseases, R. Elles, ed., pp. 321-340 (1996)) or denaturing gradient gel electrophoresis (DGGE) (Wartell et al, Nucl Acids. Res. 18:2699-2706 (1990); Sheffield et al, Proc. Natl. Acad. Sci. USA 86:232-236 (1989)).
  • SSCP single strand conformation polymorphism
  • DGGE denaturing gradient gel electrophoresis
  • a polymerase-mediated primer extension method may also be used to identify the polymorphisms.
  • Several such methods have been described in the patent and scientific literature and include the "Genetic Bit Analysis” method (WO 92/15712) and the ligase/polymerase mediated genetic bit analysis (U.S. Pat. No. 5,679,524). Related methods are disclosed in WO 91/02087, WO 90/09455, WO 95/17676, and U.S. Pat. Nos. 5,302,509 and 5,945,283. Extended primers containing a polymorphism may be detected by mass spectrometry as described in U.S. Pat. No. 5,605,798.
  • Another primer extension method is allele-specific PCR (Ruafio et al, Nucl Acids. Res. 17:8392 (1989); Ruafio et al, Nucl Acids. Res. 19: 6877-6882 (1991); WO 93/22456; Turki et al, J. Clin. Invest. 95:1635-1641 (1995)).
  • multiple polymorphic sites may be investigated by simultaneously amplifying multiple regions of the nucleic acid using sets of allele-specific primers as described in PCT patent application WO 89/10414.
  • blood samples from patients can be collected at the time of patient screening and DNA was extracted using, for example, the PUREGENETM DNA Isolation Kit (D-50K). Genotyping can be performed using the TaqMan® technology or using the Third Wave Technologies Invader Assay technique.
  • the invention provides methods and compositions for haplotyping and/or genotyping the gene in an individual.
  • the terms "genotype” and “haplotype” mean the genotype or haplotype containing the nucleotide pair or nucleotide, respectively, that is present at one or more of the polymorphic sites described herein and may optionally also include the nucleotide pair or nucleotide present at one or more additional polymorphic sites in the gene.
  • the additional polymorphic sites may be currently known polymorphic sites or sites that are subsequently discovered.
  • compositions of the invention contain oligonucleotide probes and primers designed to specifically hybridize to one or more target regions containing, or that are adjacent to, a polymorphic site.
  • Oligonucleotide compositions of the invention are useful in methods for genotyping and/or haplotyping a gene in an individual.
  • the methods and compositions for establishing the genotype or haplotype of an individual at the polymorphic sites described herein are useful for studying the effect of the polymorphisms in the aetiology of diseases affected by the expression and function of the protein, studying the efficacy of drugs targeting, predicting individual susceptibility to diseases affected by the expression and function of the protein and predicting individual responsiveness to drugs targeting the gene product.
  • Genotyping oligonucleotides of the invention may be immobilized on or synthesized on a solid surface such as a microchip, bead, or glass slide. See, e.g., WO 98/20020 and WO 98/20019.
  • Genotyping oligonucleotides may hybridize to a target region located one to several nucleotides downstream of one of the polymorphic sites identified herein. Such oligonucleotides are useful in polymerase-mediated primer extension methods for detecting one of the polymorphisms described herein and therefore such genotyping oligonucleotides are referred to herein as "primer-extension oligonucleotides”.
  • a genotyping method of the invention may involve isolating from an individual a nucleic acid mixture comprising the two copies of a gene of interest or fragment thereof, and determining the identity of the nucleotide pair at one or more of the polymorphic sites in the two copies.
  • the two "copies" of a gene in an individual may be the same allele or may be different alleles.
  • the genotyping method comprises determining the identity of the nucleotide pair at each polymorphic site.
  • the nucleic acid mixture is isolated from a biological sample taken from the individual, such as a blood sample or tissue sample.
  • a haplotyping method of the invention may include isolating from an individual a nucleic acid molecule containing only one of the two copies of a gene of interest, or a fragment thereof, and determining the identity of the nucleotide at one or more of the polymorphic sites in that copy.
  • Direct haplotyping methods include, for example, CLASPER SystemTM technology (U.S. Pat. No. 5,866,404) or allele- specific long-range PCR (Michalotos-Beloin et al, Nucl. Acids. Res.
  • the nucleic acid may be isolated using any method capable of separating the two copies of the gene or fragment. As will be readily appreciated by those skilled in the art, any individual clone will only provide haplotype information on one of the two gene copies present in an individual.
  • a haplotype pair is determined for an individual by identifying the phased sequence of nucleotides at one or more of the polymorphic sites in each copy of the gene that is present in the individual.
  • the haplotyping method comprises identifying the phased sequence of nucleotides at each polymorphic site in each copy of the gene.
  • the identity of a nucleotide (or nucleotide pair) at a polymorphic site may be determined by amplifying a target regions containing the polymorphic sites directly from one or both copies of the gene, or fragments thereof, and sequencing the amplified regions by conventional methods.
  • the genotype or haplotype for the gene of an individual may also be determined by hybridization of a nucleic sample containing one or both copies of the gene to nucleic acid arrays and subarrays such as described in published PCT patent application WO 95/11995.
  • polymorphic sites in linkage disequilibrium may be indirectly determined by genotyping other polymorphic sites in linkage disequilibrium with those sites of interest. As described above, two sites are said to be in linkage disequilibrium if the presence of a particular variant at one site is indicative of the presence of another variant at a second site. Stevens JC, MoI. Diag. 4: 309-317 (1999). Polymorphic sites in linkage disequilibrium with the polymorphic sites of the invention may be located in regions of the same gene or in other genomic regions.
  • the target regions may be amplified using any oligonucleotide-directed amplification method, including but not limited to polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • OLA oligonucleotide ligation assay
  • Oligonucleotides useful as primers or probes in such methods should specifically hybridize to a region of the nucleic acid that contains or is adjacent to the polymorphic site.
  • the oligonucleotides are between 10 and 35 nucleotides in length and preferably, between 15 and 30 nucleotides in length. Most preferably, the oligonucleotides are 20 to 25 nucleotides long. The exact length of the oligonucleotide will depend on many factors that are routinely considered and practiced by the skilled artisan.
  • nucleic acid amplification procedures may be used to amplify the target region including transcription-based amplification systems (U.S. Pat. No. 5,130,238; EP 329,822; U.S. Pat. No. 5,169,766, published PCT patent application WO 89/06700) and isothermal methods (Walker et al, Proc. Natl. Acad. ScL USA 89:392-396 (1992)).
  • Hybridizing Allele-Specific Oligonucleotide to a Target Gene A polymorphism in the target region may be assayed before or after amplification using one of several hybridization- based methods known in the art.
  • allele-specific oligonucleotides are utilized in performing such methods.
  • the allele-specific oligonucleotides may be used as differently labelled probe pairs, with one member of the pair showing a perfect match to one variant of a target sequence and the other member showing a perfect match to a different variant.
  • more than one polymorphic site may be detected at once using a set of allele- specific oligonucleotides or oligonucleotide pairs.
  • the members of the set have melting temperatures within 5°C, and more preferably within 2°C, of each other when hybridizing to each of the polymorphic sites being detected.
  • Hybridization of an allele-specific oligonucleotide to a target polynucleotide may be performed with both entities in solution, or such hybridization may be performed when either the oligonucleotide or the target polynucleotide is covalently or noncovalently affixed to a solid support. Attachment may be mediated, for example, by antibody-antigen interactions, poly-L-Lys, streptavidin or avidin-biotin, salt bridges, hydrophobic interactions, chemical linkages, UV cross-linking, baking, etc. Allele-specific oligonucleotide may be synthesized directly on the solid support or attached to the solid support subsequent to synthesis.
  • Solid- supports suitable for use in detection methods of the invention include substrates made of silicon, glass, plastic, paper and the like, which may be formed, for example, into wells ⁇ as in 96-well plates), slides, sheets, membranes, fibres, chips, dishes, and beads.
  • the solid support may be treated, coated or derivatised to facilitate the immobilization of the allele-specific oligonucleotide or target nucleic acid.
  • the invention provides a method for determining the frequency of a genotype or haplotype in a population.
  • the method comprises determining the genotype or the haplotype for a gene present in each member of the population, wherein the genotype or haplotype comprises the nucleotide pair or nucleotide detected at one or more of the polymorphic sites in the gene, and calculating the frequency at which the genotype or haplotype is found in the population.
  • the population may be a reference population, a family population, a same sex population, a population group, or a trait population (e.g., a group of individuals exhibiting a trait of interest such as a medical condition or response to a therapeutic treatment).
  • a trait population e.g., a group of individuals exhibiting a trait of interest such as a medical condition or response to a therapeutic treatment.
  • frequency data for genotypes and/or haplotypes found in a reference population are used in a method for identifying an association between a trait and a genotype or a haplotype.
  • the trait may be any detectable phenotype, including but not limited to susceptibility to a disease or response to a treatment.
  • the method involves obtaining data on the frequency of the genotypes or haplotypes of interest in a reference population and comparing the data to the frequency of the genotypes or haplotypes in a population exhibiting the trait.
  • Frequency data for one or both of the reference and trait populations may be obtained by genotyping or haplotyping each individual in the populations using one of the methods described above.
  • the haplotypes for the trait population may be determined directly or, alternatively, by the predictive genotype to haplotype approach described above.
  • the frequency data for the reference and/or trait populations are obtained by accessing previously determined frequency data, which may be in written or electronic form.
  • the frequency data may be present in a database that is accessible by a computer. Once the frequency data are obtained, the frequencies of the genotypes or haplotypes of interest in the reference and trait populations are compared.
  • haplotype frequency data for different groups are examined to determine whether they are consistent with Hardy- Weinberg equilibrium.
  • D.L. Hartl et ah Principles of Population Genomics, 3rd Ed. (Sinauer Associates, Sunderland, Massachusetts, 1997).
  • the analysis includes an assigning step, as follows: First, each of the possible haplotype pairs is compared to the haplotype pairs in the reference population.
  • haplotype pairs in the reference population matches a possible haplotype pair and that pair is assigned to the individual.
  • only one haplotype represented in the reference haplotype pairs is consistent with a possible haplotype pair for an individual, and in such cases the individual is assigned a haplotype pair containing this known haplotype and a new haplotype derived by subtracting the known haplotype from the possible haplotype pair.
  • a detectable genotype or haplotype that is in linkage disequilibrium with a genotype or haplotype of interest may be used as a surrogate marker.
  • a genotype that is in linkage disequilibrium with another genotype is indicated where a particular genotype or haplotype for a given gene is more frequent in the population that also demonstrates the potential surrogate marker genotype than in the reference population. If the frequency is statistically significant, then the marker genotype is predictive of that genotype or haplotype, and can be used as a surrogate marker.
  • the trait is susceptibility to a disease, severity of a disease, the staging of a disease or response to a drug. Such methods have applicability in developing diagnostic tests and therapeutic treatments for all pharmacogenetic applications where there is the potential for an association between a genotype and a treatment outcome, including efficacy measurements, pharmacokinetic measurements and side-effect measurements.
  • the trait of interest is a clinical response exhibited by a patient to some therapeutic treatment, for example, response to a drug targeting or to a therapeutic treatment for a medical condition.
  • genotype or haplotype data is obtained on the clinical responses exhibited by a population of individuals who received the treatment, hereinafter the "clinical population". This clinical data may be obtained by analyzing the results of a clinical trial that has already been run and/or by designing and carrying out one or more new clinical trials.
  • the individuals included in the clinical population are usually graded for the existence of the medical condition of interest. This grading of potential patients could employ a standard physical exam or one or more lab tests. Alternatively, grading of patients could use haplotyping for situations where there is a strong correlation between haplotype pair and disease susceptibility or severity.
  • the therapeutic treatment of interest is administered to each individual in the trial population, and each individual's response to the treatment is measured using one or more predetermined criteria. It is contemplated that in many cases, the trial population will exhibit a range of responses and that the investigator will choose the number of responder groups ⁇ e.g., low, medium, high) made up by the various responses. In addition, the gene for each individual in the trial population is genotyped and/or haplotyped, which may be done before or after administering the treatment. [78] These results are then analyzed to determine if any observed variation in clinical response between polymorphism groups is statistically significant. Statistical analysis methods, which may be used, are described in L.D. Fisher & G. vanBelle, Biostatistics: A Methodology for the Health Sciences (Wiley-lnterscience, New York, 1993). This analysis may also include a regression calculation of which polymorphic sites in the gene contribute most significantly to the differences in phenotype.
  • Fishers Exact tests are performed to evaluate response as a function of genotype.
  • correlations between individual response and genotype or haplotype content are created. Correlations may be produced in several ways. In one method, individuals are grouped by their genotype or haplotype (or haplotype pair) (also referred to as a polymorphism group), and then the averages and standard deviations of clinical responses exhibited by the members of each polymorphism group are calculated.
  • the identification of an association between a clinical response and a genotype or haplotype (or haplotype pair) for the gene may be the basis for designing a diagnostic method to determine those individuals who will or will not respond to the treatment, or alternatively, will respond at a lower level and thus may require more treatment, i.e., a greater dose of a drug.
  • the diagnostic method may take one of several forms: for example, a direct DNA test ⁇ i.e., genotyping or haplotyping one or more of the polymorphic sites in the gene), a serological test, or a physical exam measurement. The only requirement is that there be a good correlation between the diagnostic test results and the underlying genotype or haplotype. In a preferred embodiment, this diagnostic method uses the predictive haplotyping method described above.
  • analysis is performed using a logistic remodel to take into account gender and age in addition to treatment and "high responder" (to therapeutic treatment) genotype status.
  • an ANCOVA model can applied using the baseline value of patient response assessments as a quantitative co-variant.
  • the measured level of the gene expression product falls within 1.5 standard deviations of the mean of any of the control groups then that individual may be assigned to that genotype group. In yet another embodiment, if the measured level of the gene expression product is 1.0 or less standard deviations of the mean of any of the control groups levels then that individual may be assigned to that genotype group.
  • the standard control levels of the gene expression product would then be compared with the measured level of a gene expression product in a given patient.
  • This gene expression product could be the characteristic mRNA associated with that particular genotype group or the polypeptide gene expression product of that genotype group.
  • the patient could then be classified or assigned to a particular genotype group based on how similar the measured levels were compared to the control levels for a given group.
  • the invention also provides a computer system for storing and displaying polymorphism data determined for the gene.
  • the computer system comprises a computer processing unit, a display, and a database containing the polymorphism data.
  • the polymorphism data includes the polymorphisms, the genotypes and the haplotypes identified for a given gene in a reference population.
  • the computer system is capable of producing a display showing haplotypes organized according to their evolutionary relationships.
  • a computer may implement any or all analytical and mathematical operations involved in practicing the methods of the present invention.
  • the computer may execute a program that generates views (or screens) displayed on a display device and with which the user can interact to view and analyze large amounts of information relating to the gene and its genomic variation, including chromosome location, gene structure, and gene family, gene expression data, polymorphism data, genetic sequence data, and clinical population data (e.g., data on ethnogeographic origin, clinical responses, genotypes, and haplotypes for one or more populations).
  • the polymorphism data described herein may be stored as part of a relational database (e.g., an instance of an Oracle database or a set of ASCII flat files).
  • polymorphism data may be stored on the computer's hard drive or may, for example, be stored on a CD-ROM or on one or more other storage devices accessible by the computer.
  • the data may be stored on one or more databases in communication with the computer via a network.
  • the invention provides SNP probes, which are useful in classifying subjects according to their types of genetic variation.
  • the SNP probes according to the invention are oligonucleotides, which discriminate between SNPs in conventional allelic discrimination assays.
  • the oligonucleotides according to this aspect of the invention are complementary to one allele of the SNP nucleic acid, but not to any other allele of the SNP nucleic acid.
  • Oligonucleotides according to this embodiment of the invention can discriminate between SNPs in various ways. For example, under stringent hybridization conditions, an oligonucleotide of appropriate length will hybridize to one SNP, but not to any other.
  • the oligonucleotide may be labelled using a radiolabel or a fluorescent molecular tag.
  • an oligonucleotide of appropriate length can be used as a primer for PCR, wherein the 3' terminal nucleotide is complementary to one allele containing a SNP, but not to any other allele.
  • the presence or absence of amplification by PCR determines the haplotype of the SNP.
  • Genomic and cDNA fragments of the invention comprise at least one polymorphic site identified herein, have a length of at least 10 nucleotides, and may range up to the full length of the gene.
  • a fragment according to the present invention is between 100 and 3000 nucleotides in length, and more preferably between 200 and 2000 nucleotides in length, and most preferably between 500 and 1000 nucleotides in length.
  • kits of the Invention provides nucleic acid and polypeptide detection kits useful for haplotyping and/or genotyping the gene in an individual. Such kits are useful for classifying individuals for the purpose of classifying individuals. Specifically, the invention encompasses kits for detecting the presence of a polypeptide or nucleic acid corresponding to a marker of the invention in a biological sample, e.g., any bodily fluid including, but not limited to, serum, plasma, lymph, cystic fluid, urine, stool, cerebrospinal fluid, ascities fluid or blood, and including biopsy samples of body tissue.
  • a biological sample e.g., any bodily fluid including, but not limited to, serum, plasma, lymph, cystic fluid, urine, stool, cerebrospinal fluid, ascities fluid or blood, and including biopsy samples of body tissue.
  • the kit can comprise a labelled compound or agent capable of detecting a polypeptide or an mRNA encoding a polypeptide corresponding to a marker of the invention in a biological sample and means for determining the amount of the polypeptide or mRNA in the sample, e.g., an antibody which binds the polypeptide or an oligonucleotide probe which binds to DNA or mRNA encoding the polypeptide.
  • Kits can also include instructions for interpreting the results obtained using the kit.
  • the invention provides a kit comprising at least two genotyping oligonucleotides packaged in separate containers.
  • the kit may also contain other components such as hybridization buffer (where the oligonucleotides are to be used as a probe) packaged in a separate container.
  • the kit may contain, packaged in separate containers, a polymerase and a reaction buffer optimized for primer extension mediated by the polymerase, such as in the case of PCR.
  • such kit may further comprise a DNA sample collecting means.
  • the kit can comprise, e.g., (1) a first antibody, e.g., attached to a solid support, which binds to a polypeptide corresponding to a marker or the invention; and, optionally (2) a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable label.
  • a first antibody e.g., attached to a solid support
  • a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable label.
  • the kit can comprise, e.g., (1) an oligonucleotide, e.g., a detectably-labelled oligonucleotide, which hybridizes to a nucleic acid sequence encoding a polypeptide corresponding to a marker of the invention; or (2) a pair of primers useful for amplifying a nucleic acid molecule corresponding to a marker of the invention.
  • the kit can also comprise, e.g., a buffering agent, a preservative or a protein- stabilizing agent.
  • the kit can further comprise components necessary for detecting the detectable-label, e.g., an enzyme or a substrate.
  • the kit can also contain a control sample or a series of control samples, which can be assayed and compared to the test sample.
  • Each component of the kit can be enclosed within an individual container and all of the various containers can be within a single package, along with instructions for interpreting the results of the assays performed using the kit.
  • the invention comprises one or more isolated polynucleotides.
  • the invention also encompasses allelic variants of the same, that is, naturally occurring alternative forms of the isolated polynucleotides that encode mutant polypeptides that are identical, homologous or related to those encoded by the polynucleotides.
  • non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis techniques well-known in the art.
  • nucleic acid sequences capable of hybridizing at low stringency with any nucleic acid sequences encoding mutant polypeptide of the present invention are considered to be within the scope of the invention.
  • Standard stringency conditions are well characterized in standard molecular biology cloning texts. See, for example Molecular Cloning A Laboratory Manual, 2nd Ed., ed., Sambrook, Fritsch, & Maniatis (Cold Spring Harbor Laboratory Press, 1989); DNA Cloning, Volumes I and II, D.N. Glover, ed. (1985); Oligonucleotide Synthesis, M.J. Gait, ed. (1984); Nucleic Acid Hybridization, B.D. Hames & SJ. Higgins, eds (1984).
  • biological sample is intended to include tissues, cells, biological fluids and isolates thereof, isolated from a subject, as well as tissues, cells and fluids present within a subject.
  • Many expression detection methods use isolated RNA.
  • any RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of RNA from cells. See, e.g., Ausubel et al, Ed., Curr. Prot. MoI. Biol. (John Wiley & Sons, New York, 1987-1999).
  • the level of the mRNA expression product of the target gene is determined.
  • Methods to measure the level of a specific mRNA are well-known in the art and include Northern blot analysis, reverse transcription PCR and real time quantitative PCR or by hybridization to a oligonucleotide array or microarray.
  • the determination of the level of expression may be performed by determination of the level of the protein or polypeptide expression product of the gene in body fluids or tissue samples including but not limited to blood or serum. Large numbers of tissue samples can readily be processed using techniques well-known to those of skill in the art, such as, e.g., the single-step RNA isolation process of U.S. Pat. No. 4,843,155.
  • the isolated mRNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, PCR analyses and probe arrays.
  • One preferred diagnostic method for the detection of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to the mRNA encoded by the gene being detected.
  • the nucleic acid probe can be, e.g., a full-length cDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to an mRNA or genomic DNA encoding a marker of the present invention.
  • probes for use in the diagnostic assays of the invention are described herein. Hybridization of an mRNA with the probe indicates that the marker in question is being expressed.
  • the probes are immobilized on a solid surface and the mRNA is contacted with the probes, for example, in an Affymetrix gene chip array (Affymetrix, Calif. USA).
  • a skilled artisan can readily adapt known mRNA detection methods for use in detecting the level of mRNA encoded by the markers of the present invention.
  • An alternative method for determining the level of mRNA corresponding to a marker of the present invention in a sample involves the process of nucleic acid amplification, e.g., by RT-PCR (the experimental embodiment set forth in U.S. Pat. No. 4,683,202); ligase chain reaction (Barany et al, Proc. Natl. Acad. Sci. USA 88:189-193 (1991)) self-sustained sequence replication (Guatelli et al, Proc. Natl. Acad. Sci. USA 87: 1874-1878 (1990)); transcriptional amplification system (Kwoh et al, Proc. Natl. Acad. Sci.
  • amplification primers are defined as being a pair of nucleic acid molecules that can anneal to 5' or 3' regions of a gene (plus and minus strands, respectively, or vice- versa) and contain a short region in between.
  • amplification primers are from about 10-30 nucleotides in length and flank a region from about 50-200 nucleotides in length.
  • RT-PCR Real-time quantitative PCR
  • the RT-PCR assay utilizes an RNA reverse transcriptase to catalyze the synthesis of a DNA strand from an RNA strand, including an mRNA strand.
  • the resultant DNA may be specifically detected and quantified and this process may be used to determine the levels of specific species of mRNA.
  • TAQMAN® PE Applied Biosystems, Foster City, Calif, USA
  • AMPLITAQ GOLDTM DNA polymerase exploits the 5' nuclease activity of AMPLITAQ GOLDTM DNA polymerase to cleave a specific form of probe during a PCR reaction.
  • This is referred to as a TAQMANTM probe. See Luthra et al., Am. J. Pathol. 153: 63-68 (1998); Kuimelis et al., Nucl. Acids Symp. Ser. 37: 255-256 (1997); and Mullah et al, Nucl Acids Res. 26(4): 1026-1031 (1998)).
  • cleavage of the probe separates a reporter dye and a quencher dye, resulting in increased fluorescence of the reporter.
  • the accumulation of PCR products is detected directly by monitoring the increase in fluorescence of the reporter dye. Heid et al, Genome Res. 6(6): 986-994 (1996)). The higher the starting copy number of nucleic acid target, the sooner a significant increase in fluorescence is observed. See Gibson, Heid & Williams et al, Genome Res. 6: 995-1001 (1996).
  • Detection of Polypeptides can be detected by a probe which is detectably labelled, or which can be subsequently labelled.
  • the term "labelled", with regard to the probe or antibody is intended to encompass direct-labelling of the probe or antibody by coupling, i.e., physically linking, a detectable substance to the probe or antibody, as well as indirect- labelling of the probe or antibody by reactivity with another reagent that is directly-labelled. Examples of indirect labelling include detection of a primary antibody using a fluorescently- labelled secondary antibody and end-labelling of a DNA probe with biotin such that it can be detected with fluorescently-labelled streptavidin.
  • the probe is an antibody that recognizes the expressed protein.
  • a variety of formats can be employed to determine whether a sample contains a target protein that binds to a given antibody.
  • Immunoassay methods useful in the detection of target polypeptides of the present invention include, but are not limited to, e.g., dot blotting, western blotting, protein chips, competitive and noncompetitive protein binding assays, enzyme-linked immunosorbant assays (ELISA), immunohistochemistry, fluorescence activated cell sorting (FACS), and others commonly used and widely-described in scientific and patent literature, and many employed commercially.
  • a skilled artisan can readily adapt known protein/antibody detection methods for use in determining whether cells express a marker of the present invention and the relative concentration of that specific polypeptide expression product in blood or other body tissues.
  • Proteins from individuals can be isolated using techniques that are well-known to those of skill in the art. The protein isolation methods employed can, e.g., be such as those described in Harlow & Lane, Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1988)).
  • various host animals may be immunized by injection with the polypeptide, or a portion thereof. Such host animals may include, but are not limited to, rabbits, mice and rats.
  • adjuvants may be used to increase the immunological response, depending on the host species including, but not limited to, Freund's (complete and incomplete), mineral gels, such as aluminium hydroxide; surface active substances, such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin and dinitrophenol; and potentially useful human adjuvants, such as bacille Camette-Guerin (BCG) and Corynebacterium parvum.
  • BCG Bacille Camette-Guerin
  • Monoclonal antibodies which are homogeneous populations of antibodies to a particular antigen, may be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique of Kohler & Milstein, Nature 256: 495-497 (1975); and U.S. Pat. No. 4,376,110; the human B-cell hybridoma technique of Kosbor et ah, Immunol. Today 4: 72 (1983); Cole et ah, Proc. Natl. Acad. ScL USA 80: 2026-2030 (1983); and the EBV- hybridoma technique of Cole et ah, Monoclonal Antibodies and Cancer Therapy (Alan R. Liss, Inc., 1985) pp. 77-96.
  • chimeric antibodies are a molecule in which different portions are derived from different animal species, such as those having a variable or hypervariable region derived form a murine niAb and a human immunoglobulin constant region.
  • Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody.
  • Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros and magnetite.
  • a useful method for ease of detection, is the sandwich ELISA, of which a number of variations exist, all of which are intended to be used in the methods and assays of the present invention.
  • sandwich assay is intended to encompass all variations on the basic two-site technique. Immunofluorescence and EIA techniques are both very well- established in the art. However, other reporter molecules, such as radioisotopes, chemiluminescent or bioluminescent molecules may also be employed. It will be readily apparent to the skilled artisan how to vary the procedure to suit the required use.
  • Whole genome monitoring of protein i.e., the "proteome” can be carried out by constructing a microarray in which binding sites comprise immobilized, preferably monoclonal, antibodies specific to a plurality of protein species encoded by the cell genome.
  • binding sites comprise immobilized, preferably monoclonal, antibodies specific to a plurality of protein species encoded by the cell genome.
  • antibodies are present for a substantial fraction of the encoded proteins, or at least for those proteins relevant to testing or confirming a biological network model of interest.
  • methods for making monoclonal antibodies are well-known. See, e.g., Harlow & Lane, Antibodies: A Laboratory ManuaF (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1988)).
  • monoclonal antibodies are raised against synthetic peptide fragments designed based on genomic sequence of the cell. With such an antibody array, proteins from the cell are contacted to the array and their binding is measured with assays known in the art.
  • Two-Dimensional Gel Electrophoresis Two-dimensional gel electrophoresis is well-known in the art and typically involves isoelectric focusing along a first dimension followed by SDS-PAGE electrophoresis along a second dimension. See, e.g., Hames et al, Gel Electrophoresis of Proteins: A Practical Approach (IRL Press, New York, 1990); Shevchenko et al, Proc. Natl. Acad. Sci. USA 93: 14440-14445 (1996); Sagliocco et al, Yeast 12: 1519-1533 (1996); and Lander, Science 274: 536-539 (1996).
  • MS-based analysis methodology is useful for analysis of isolated target polypeptide as well as analysis of target polypeptide in a biological sample.
  • MS formats for use in analyzing a target polypeptide include ionization (I) techniques, such as, but not limited to, matrix assisted laser desorption (MALDI), continuous or pulsed electrospray ionization (ESI) and related methods, such as ionspray or thermospray, and massive cluster impact (MCI).
  • I ionization
  • MALDI matrix assisted laser desorption
  • ESI electrospray ionization
  • MCI massive cluster impact
  • Such ion sources can be matched with detection formats, including linear or non-linear reflectron time of flight (TOF), single or multiple quadrupole, single or multiple magnetic sector, Fourier transform ion cyclotron resonance (FTICR), ion trap and combinations thereof such as ion-trap/TOF.
  • TOF linear or non-linear reflectron time of flight
  • FTICR Fourier transform ion cyclotron resonance
  • ion trap and combinations thereof such as ion-trap/TOF.
  • numerous matrix/wavelength combinations ⁇ e.g., matrix assisted laser desorption (MALDI)) or solvent combinations (e.g., ESI) can be employed.
  • MALDI matrix assisted laser desorption
  • ESI solvent combinations
  • the target polypeptide can be solubilised in an appropriate solution or reagent system.
  • a solution or reagent system e.g., an organic or inorganic solvent
  • MS of peptides also is described, e.g., in International PCT Application No.
  • a solvent is selected that minimizes the risk that the target polypeptide will be decomposed by the energy introduced for the vaporization process.
  • a reduced risk of target polypeptide decomposition can be achieved, e.g., by embedding the sample in a matrix.
  • a suitable matrix can be an organic compound such as a sugar, e.g., a pentose or hexose, or a polysaccharide such as cellulose. Such compounds are decomposed thermolytically into CO 2 and H 2 O such that no residues are formed that can lead to chemical reactions.
  • the matrix also can be an inorganic compound, such as nitrate of ammonium, which is decomposed essentially without leaving any residue.
  • an inorganic compound such as nitrate of ammonium, which is decomposed essentially without leaving any residue.
  • Use of these and other solvents is known to those of skill in the art. See, e.g., U.S. Pat. No. 5,062,935.
  • Electrospray MS has been described by Fenn et al, J. Phys. Chem. 88: 4451-4459 (1984); and PCT Application No. WO 90/14148; and current applications are summarized in review articles. See Smith et al., Anal. Chem. 62: 882-89 (1990); and Ardrey, Spectroscopy 4: 10-18 (1992).
  • the mass of a target polypeptide determined by MS can be compared to the mass of a corresponding known polypeptide.
  • the corresponding known polypeptide can be the corresponding non-mutant protein, e.g., wild-type protein.
  • ESI the determination of molecular weights in femtomole amounts of sample is very accurate due to the presence of multiple ion peaks, all of which can be used for mass calculation.
  • Sub-attomole levels of protein have been detected, e.g., using ESI MS (Valaskovic et al, Science 273: 1199-1202 (1996)) and MALDI MS (Li et al, J. Am. Chem. Soc. 118: 1662-1663 (1996)).
  • Matrix Assisted Laser Desorption The level of the target protein in a biological sample, e.g., body fluid or tissue sample, may be measured by means of mass spectrometric (MS) methods including, but not limited to, those techniques known in the art as matrix-assisted laser desorption/ionization, time-of-flight mass spectrometry (MALDI- TOF-MS) and surfaces enhanced for laser desorption/ionization, time-of-flight mass spectrometry (SELDI-TOF-MS) as further detailed below.
  • MS mass spectrometric
  • Methods for performing MALDI are well-known to those of skill in the art. See, e.g., Juhasz et al, Analysis, Anal. Chem.
  • MALDI-TOF-MS has been described by Hillenkamp et al, Biological Mass Spectrometry, Burlingame & McCloskey, eds. (Elsevier Science Publ., Amsterdam, 1990) pp. 49-60. [120] A variety of techniques for marker detection using mass spectroscopy can be used. See Bordeaux Mass Spectrometry Conference Report, Hillenkamp, Ed., pp.
  • MS techniques allow the successful volatilization of high molecular weight biopolymers, without fragmentation, and have enabled a wide variety of biological macromolecules to be analyzed by mass spectrometry.
  • SMDl Surfaces Enhanced for Laser Desorption/lonization
  • Other techniques are used which employ new MS probe element compositions with surfaces that allow the probe element to actively participate in the capture and docking of specific analytes, described as Affinity Mass Spectrometry (AMS). See SELDI patents U.S. Pat. Nos. 5,719,060; 5,894,063; 6,020,208; 6,027,942; 6,124,137; and U.S. Patent application No. U.S. 2003/0003465.
  • SEAC probe elements have been designed with Surfaces Enhanced for Affinity Capture (SEAC). See Hutchens & Yip, Rapid Commun. Mass Spectrom. 7: 576-580 (1993).
  • SEAC probe elements have been used successfully to retrieve and tether different classes of biopolymers, particularly proteins, by exploiting what is known about protein surface structures and biospecific molecular recognition.
  • the immobilized affinity capture devices on the MS probe element surface, i.e., SEAC determines the location and affinity (specificity) of the analyte for the probe surface, therefore the subsequent analytical MS process is efficient.
  • SELDI Surfaces Enhanced for Neat Desorption
  • the probe element surfaces i.e., sample presenting means
  • EAM Energy Absorbing Molecules
  • SEAC SEAC
  • the probe element surfaces i.e., sample presenting means
  • affinity capture devices to facilitate either the specific or non-specific attachment or adsorption (so-called docking or tethering) of analytes to the probe surface, by a variety of mechanisms (mostly non-covalent).
  • SEPAR Photolabile Attachment and Release
  • the probe element surfaces i.e., sample presenting means
  • the analyte e.g., protein
  • the chemical specificities determining the type and number of the photolabile molecule attachment points between the SEPAR sample presenting means (i.e., probe element surface) and the analyte may involve any one or more of a number of different residues or chemical structures in the analyte (e.g. , His, Lys, Arg, Tyr, Phe and Cys residues in the case of proteins and peptides).
  • aspects of the biological activity state, or mixed aspects can be measured in order to obtain drug and pathway responses.
  • the activities of proteins relevant to the characterization of cell function can be measured, and embodiments of this invention can be based on such measurements.
  • Activity measurements can be performed by any functional, biochemical or physical means appropriate to the particular activity being characterized. Where the activity involves a chemical transformation, the cellular protein can be contacted with natural substrates, and the rate of transformation measured. Where the activity involves association in multimeric units, e.g., association of an activated DNA binding complex with DNA, the amount of associated protein or secondary consequences of the association, such as amounts of mRNA transcribed, can be measured.
  • response data may be formed of mixed aspects of the biological state of a cell.
  • Response data can be constructed from, e.g., changes in certain mRNA abundances, changes in certain protein abundances and changes in certain protein activities.
  • the objective of this EXAMPLE was to test whether variations in the LRRK2 gene are associated with the progression to Alzheimer's disease (AD) in subjects with mild cognitive impairment (MCI).
  • GLY2019SER mutation In a screen of patients, with results confirmed by re- sequencing, we found the following: For Parkinson's disease (PD): 6 out of 483 patients carry the G2019S mutation (1.24%). For Parkinson's disease with dementia (PDD): 1 out of 391 patients carry the G2019S mutation (0.26%). For Alzheimer's disease (AD): None of the 373 patients carry the G2019S mutation. For Mild cognitive impairment (MCI): None of the 448 patients carry the G2019S mutation. For Amyotrophic lateral sclerosis (ALS): None of the 483 patients carry the G2019S mutation.
  • PD Parkinson's disease
  • PDD Parkinson's disease with dementia
  • AD Alzheimer's disease
  • MCI Mild cognitive impairment
  • MCI Mild cognitive impairment
  • ALS Amyotrophic lateral sclerosis
  • LRRK2 leucine-rich repeat kinase 2
  • Lys Lys Leu lie VaI Arg Leu Asn Asn VaI GIn GIu GIy Lys GIn lie
  • GIu Thr Leu VaI GIn lie Leu GIu Asp Leu Leu VaI Phe Thr Tyr Ser
  • Arg Lys lie Leu Leu Ser Lys GIy lie His Leu Asn VaI Leu GIu Leu
  • Leu Leu Lys Arg Lys Arg Lys lie Leu Ser Ser Asp Asp Ser Leu Arg 945 950 955 960
  • GIy lie Cys Ser Pro Leu Arg Leu Lys GIu Leu Lys lie Leu Asn Leu
  • Ser Lys Asn His lie Ser Ser Leu Ser GIu Asn Phe Leu GIu Ala Cys
  • Cys Lys Ala Lys Asp lie lie Arg Phe Leu GIn GIn Arg Leu Lys Lys 1315 1320 1325
  • GIn Leu lie Pro Asp Cys Tyr VaI GIu Leu GIu Lys lie lie Leu Ser
  • GIu Arg Lys Asn VaI Pro lie GIu Phe Pro VaI lie Asp Arg Lys Arg
  • Lys Trp Leu Cys Lys lie Met Ala GIn lie Leu Thr VaI Lys VaI GIu
  • GIy Cys Pro Lys His Pro Lys GIy lie lie Ser Arg Arg Asp VaI GIu
  • Leu GIu lie Ser Pro Tyr Met Leu Ser GIy Arg GIu Arg Ala Leu Arg
  • 1810 1815 1820 lie Leu Leu Asp Asp Leu Met Lys Lys Ala GIu GIu GIy Asp Leu Leu 1825 1830 1835 1840
  • VaI Lys lie Phe Asn Lys His Thr Ser Leu Arg Leu Leu Arg GIn GIu 1905 1910 1915 1920
  • Leu His Ser Ala Met lie lie Tyr Arg Asp Leu Lys Pro His Asn VaI 1985 1990 1995 2000
  • Leu Leu Pro Lys Asn VaI lie VaI GIu Cys Met VaI Ala Thr His His 2145 2150 2155 2160
  • GIu VaI Ala Asp Ser Arg lie Leu Cys Leu Ala Leu VaI His Leu Pro
  • VaI GIu Lys GIu Ser Trp lie VaI Ser GIy Thr GIn Ser GIy Thr Leu
  • Leu VaI lie Asn Thr GIu Asp GIy Lys Lys Arg His Thr Leu GIu Lys 2225 2230 2235 2240
  • Lys lie Leu Asn lie GIy Asn VaI Ser Thr Pro Leu Met Cys Leu Ser

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Pathology (AREA)
  • General Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The genetic polymorphism LRRK2 (leucine-rich repeat kinase 2)-T1602S is significantly associated with conversion from mild cognitive impairment (MCI) to Alzheimer's disease (AD), with the patients with TT genotype being at greater risk to progress to Alzheimer's disease. The LRRK2-T2352 also showed a trend for conversion to Alzheimer's disease, with the patients with CC genotype tending to progress to Alzheimer's disease. Similar to the APOE-E4 allele, in the presence of a BuChE-K variant, LRRK2-T1602S and LRRK2-T2352 showed a greater association with the rate of conversion from mild cognitive impairment to Alzheimer's disease. In another study with placebo-treated Alzheimer's disease patients, LRRK2-T1602S and LRRK2-T2352 showed a same trend of association. The Alzheimer's disease patients with TT genotype of LRRK2-T1602S or CC genotype of LRRK2-T2352 tended to decline faster on cognitive performance over 6 months, especially in the presence of a BuChE-K variant. The association between the two common LRRK2 polymorphisms and Alzheimer's disease progression shows that LRRK2 may play a role in Alzheimer's disease pathogenesis, especially disease progression, and that polymorphisms of LRRK2 can be used as biomrkers of this progression.

Description

BIOMARKERS FOR THE PROGRESSION OF ALZHEIMER'S DISEASE
FIELD OF THE INVENTION
[01] This invention relates generally to the analytical testing of tissue samples in vitro, and more particularly to aspects of genetic polymorphisms of the leucine-rich repeat kinase 2 (LRRK2) gene.
BACKGROUND OF THE INVENTION
[02] Therapy specific diagnostics (a.k.a., theranostics) is an emerging medical technology field, which provides tests useful to diagnose a disease, choose the correct treatment regime and monitor a subject's response. That is, theranostics are useful to predict and assess drug response in individual subjects, i.e., individualized medicine. Theranostic tests are also useful to select subjects for treatments that are particularly likely to benefit from the treatment or to provide an early and objective indication of treatment efficacy in individual subjects, so that the treatment can be altered with a minimum of delay.
[03] Progress in pharmacogenetics, which establishes correlations between responses to specific drugs and the genetic profile of individual patients, is foundational to the development of new theranostic approaches. As such, there is a need in the art for the evaluation of patient-to-patient variations in gene sequence and gene expression. A common form of genetic profiling relies on the identification of DNA sequence variations called single nucleotide polymorphisms ("SNPs"), which are one type of genetic mutation leading to patient-to-patient variation in individual drug response. It follows that there is a need in the art to identify and characterize genetic mutations, such as SNPs, which are useful to identify the genotypes of subjects associated with drug responsiveness, side-effects, or optimal dose.
SUMMARY OF THE INVENTION
[04] Polymorphisms of the of the leucine-rich repeat kinase 2 (LRRK2) gene can be used as biomarkers of Alzheimer's disease(AD) progression. In particular, mutations of the LRRK2 gene causing a change in the protein (such as T1602S and T2352, in particular T2352M) may have an impact on Alzheimer's disease and can be used as a biomarker of Alzheimer's disease progression and age-at-onset of Alzheimer's disease. Accordingly, the invention provides for the use of a LRRK2 modulating agent in the manufacture of a medicament for the treatment of Alzheimer's disease a selected patient population. The patient population is selected on the basis of polymorphisms in theLRRK2 gene that are indicative of progression from mild cognitive impairment (MCI) to Alzheimer's disease. In one embodiment, the LRRK2 modulating agent is a heterocyclic compound that slows the progression by the patient from mild cognitive impairment to Alzheimer's disease. In another embodiment, the LRRK2 modulating agent is a heterocyclic compound that slows the progression by the patient from moderate Alzheimer's disease to severe Alzheimer's disease. In yet another embodiment, the polymorphism in the LRRK2 gene can be T 1602 S or T2352. The invention also provides methods for the predicting Alzheimer's disease progression or age-at-onset of Alzheimer's disease.
BRIEF DESCRIPTION OF THE DRAWINGS
[05] The drawing figure depicts preferred embodiments by way of example, not by way of limitations.
[06] FIG 1 is a depiction of the LRRK2 protein structure and location of the two common
LRRK2 polymorphisms (T1602S and T2352M).
DETAILED DESCRIPTION OF THE INVENTION
[07] Progression to Alzheimer's disease data of a 3-4 year study in mild cognitive impairment (MCI) patients was used to investigate the effect of the two common LRRK2 polymorphisms, T1602S and T2352, on rate of progression to Alzheimer's disease. To verify the findings , we further tested the correlation between the two common LRRK2 polymorphisms and cognitive performance over 6 months in placebo-treated Alzheimer's disease patients enrolled in another study.
[08] We found that LRRK2-T1602S was significantly associated with conversion from mild cognitive impairment to Alzheimer's disease. The mild cognitive impairment patients with TT genotype were at greater risk to progress to Alzheimer's disease. The LRRK2-T2352 also showed a trend for conversion to Alzheimer's disease. The mild cognitive impairment patients with CC genotype tended to progress to Alzheimer's disease. Similar to the APOE- E4 allele, in the presence of a BuChE-K variant, LRRK2-T1602S and LRRK2-T2352 showed a greater association with the rate of conversion from mild cognitive impairment to AD. [09] In the study with the placebo-treated Alzheimer's disease patients, LRRK2-T1602S and LRRK2-T2352 showed a same trend of the association observed in the mild cognitive impairment study. The Alzheimer's disease patients with TT genotype of LRRK2-T1602S or CC genotype of LRRK2-T2352 tended to decline faster on cognitive performance over 6 months, especially in the presence of a BuChE-K variant.
[10] The association between the two common LRRK2 polymorphisms and Alzheimer's disease progression shows that LRRK2 may play a role in Alzheimer's disease pathogenesis, especially disease progression and that polymorphisms of LRRK2 can be used as biomrkers of this progression.
[1 1] The human LRKK2 gene (SEQ ID NO : 1 ) is located in the P ARK8 locus on chromosome 12ql2. The gene has multiple-domains. LRKK2 protein (SEQ ID NO:2)is a receptor interacting protein (RIP) kinase. The G2019S mutation is the most common pathogenic mutation of LRRK2 (5-6% of autosomal dominant and ~1% of sporadic late-onset cases). The G2019S mutation is located in the kinase domain of the LRKK2 gene. [12] FIG. 1 shows LRRK2 protein structure and location of two other common polymorphisms. We focused on the polymorphisms T1602S and T2352M, because (1) they are common polymorphisms; and (2) they are missense polymorphisms. [13] The minor allele frequency is as follows: Tl 602S = 30%; T2352M = 34%. Amino acid change caused by the polymorphisms are as follows: T1602S - Thr -> Ser (1602 A>T); T2352M - Thr -> Met (2352 C>T). According to linkage disequilibrium (LD) analysis, T1602S and T2352M are in strong LD (D'=0.979).
[14] It is to be appreciated that certain aspects, modes, embodiments, variations and features of the invention are described below in various levels of detail in order to provide a substantial understanding of the present invention. The various aspects of the present invention relate to diagnostic/theranostic methods and kits that use the LRRK2 mutations of the invention to identify individuals predisposed to disease or to classify individuals with regard to drug responsiveness, side effects, or optimal drug dose. In other aspects, the invention provides methods for compound validation and a computer system for storing and analyzing data related to the LRRK2 mutations of the invention. Accordingly, various particular embodiments that illustrate these aspects follow.
[15] Definitions. The definitions of certain terms as used in this specification are provided below. Definitions of other terms may be found in the glossary provided by the U.S. Department of Energy, Office of Science, Human Genome Project
(http://www.ornl.gov/sci/techresources/Human Genome/glossary/)- In practicing the present invention, many conventional techniques in molecular biology, microbiology and recombinant DNA are used. These techniques are well-known and are explained in, e.g., Current Protocols in Molecular Biology, VoIs. I-III, Ausubel, ed. (1997); Sambrook et ah, Molecular Cloning: A Laboratory Manual, Second Edition (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989); DNA Cloning: A Practical Approach, VoIs. I and II, Glover D, ed. (1985); Oligonucleotide Synthesis, Gait, ed. (1984); Nucleic Acid Hybridization, Hames & Higgins, eds. (1985); Transcription and Translation, Hames & Higgins, eds. (1984); Animal Cell Culture, Freshney, ed. (1986); Immobilized Cells and Enzymes (IRL Press, 1986); Perbal, A Practical Guide to Molecular Cloning; the series, Methods in Enzymol. (Academic Press, Inc., 1984); Gene Transfer Vectors for Mammalian Cells, Miller & Calos, eds. (Cold Spring Harbor Laboratory, NY, 1987); and Methods in Enzymology, VoIs. 154 and 155, Wu & Grossman, and Wu, eds., respectively. [16] As used herein, the term "allele" means a particular form of a gene or DNA sequence at a specific chromosomal location (locus).
[17] As used herein, the term "antibody" includes, but is not limited to, polyclonal antibodies, monoclonal antibodies, humanized or chimeric antibodies and biologically functional antibody fragments sufficient for binding of the antibody fragment to the protein. Antibodies can be used in assays to determine the presence of variant proteins and peptides where the genetic polymorphisms of the invention are in the coding region of the gene.. [18] As used herein, the term "clinical response" means any or all of the following: a quantitative measure of the response, no response, and adverse response (i.e., side effects). [19] As used herein, the term "clinical trial" means any research study designed to collect clinical data on responses to a particular treatment, and includes but is not limited to phase I, phase II and phase III clinical trials. Standard methods are used to define the patient population and to enrol subjects.
[20] As used herein, the term "effective amount" of a compound is a quantity sufficient to achieve a desired therapeutic and/or prophylactic effect, for example, an amount which results in the prevention of or a decrease in the symptoms associated with a disease that is being treated, e.g., the diseases associated with LRRK2 mutant polynucleotides and mutant polypeptides identified herein (particularly Alzheimer's disease and Parkinson's disease). The amount of compound administered to the subject will depend on the type and severity of the disease and on the characteristics of the individual, such as general health, age, sex, body weight and tolerance to drugs. It will also depend on the degree, severity and type of disease. The skilled artisan will be able to determine appropriate dosages depending on these and other factors. Typically, an effective amount of the compounds of the present invention, sufficient for achieving a therapeutic or prophylactic effect, range from about 0.000001 mg per kilogram body weight per day to about 10,000 mg per kilogram body weight per day. Preferably, the dosage ranges are from about 0.0001 mg per kilogram body weight per day to about 100 mg per kilogram body weight per day. The compounds of the present invention can also be administered in combination with each other, or with one or more additional therapeutic compounds.
[21] As used herein, "expression" includes but is not limited to one or more of the following: transcription of the gene into precursor mRNA; splicing and other processing of the precursor mRNA to produce mature mRNA; mRNA stability; translation of the mature mRNA into protein (including codon usage and tRNA availability); and glycosylation and/or other modifications of the translation product, if required for proper expression and function. [22] As used herein, the term "gene" means a segment of DNA that contains all the information for the regulated biosynthesis of an RNA product, including promoters, exons, introns, and other untranslated regions that control expression. [23] As used herein, the term "genotype" means an unphased 5' to 3' sequence of nucleotide pairs found at one or more polymorphic sites in a locus on a pair of homologous chromosomes in an individual. As used herein, genotype includes a full-genotype and/or a sub-genotype.
[24] As used herein, the term "locus" means a location on a chromosome or DNA molecule corresponding to a gene or a physical or phenotypic feature, in particular the LRRK2 gene. [25] As used herein, the term "LRRK2 modulating agent" is any compound that alters (e.g., increases or decreases) the expression level or biological activity level of LRRK2 polypeptide compared to the expression level or biological activity level of LRRK2 polypeptide in the absence of the LRRK2 modulating agent. LRRK2 modulating agent can be a small molecule, polypeptide, carbohydrate, lipid, nucleotide, or combination thereof. The LRRK2 modulating agent may be an organic compound or an inorganic compound. [26] As used herein, the term "mutant" means any heritable variation from the wild-type that is the result of a mutation, e.g., single nucleotide polymorphism. The term "mutant" is used interchangeably with the terms "marker", "biomarker", and "target" throughout the specification.
[27] As used herein, the term "medical condition" includes, but is not limited to, any condition or disease manifested as one or more physical and/or psychological symptoms for which treatment is desirable, and includes previously and newly identified diseases and other disorders.
[28] As used herein, the term "nucleotide pair" means the nucleotides found at a polymorphic site on the two copies of a chromosome from an individual.
[29] As used herein, the term "polymorphic site" means a position within a locus at which at least two alternative sequences are found in a population, the most frequent of which has a frequency of no more than 99%.
[30] As used herein, the term "phased" means, when applied to a sequence of nucleotide pairs for two or more polymorphic sites in a locus, the combination of nucleotides present at those polymorphic sites on a single copy of the locus is known.
[31] As used herein, the term "polymorphism" means any sequence variant present at a frequency of >1% in a population. The sequence variant may be present at a frequency significantly greater than 1% such as 5% or 10 % or more. Also, the term may be used to refer to the sequence variation observed in an individual at a polymorphic site.
Polymorphisms include nucleotide substitutions, insertions, deletions and microsatellites and may, but need not, result in detectable differences in gene expression or protein function.
[32] As used herein, the term "polynucleotide" means any RNA or DNA, which may be unmodified or modified RNA or DNA. Polynucleotides include, without limitation, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions, single- and double-stranded RNA, RNA that is mixture of single- and double-stranded regions, and hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions. In addition, polynucleotide refers to triple-stranded regions comprising RNA or DNA or both
RNA and DNA. The term polynucleotide also includes DNAs or RNAs containing one or more modified bases and DNAs or RNAs with backbones modified for stability or for other reasons. [33] As used herein, the term "polypeptide" means any polypeptide comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres. Polypeptide refers to both short chains, commonly referred to as peptides, glycopeptides or oligomers, and to longer chains, generally referred to as proteins. Polypeptides may contain amino acids other than the 20 gene-encoded amino acids. Polypeptides include amino acid sequences modified either by natural processes, such as post- translational processing, or by chemical modification techniques that are well-known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature.
[34] As used herein, the term "SNP nucleic acid" means a nucleic acid sequence, which comprises a nucleotide that is variable within an otherwise identical nucleotide sequence between individuals or groups of individuals, thus existing as alleles. Such SNP nucleic acids are preferably from about 15 to about 500 nucleotides in length. The SNP nucleic acids may be part of a chromosome, or they may be an exact copy of a part of a chromosome, e.g., by amplification of such a part of a chromosome through PCR or through cloning. The SNP nucleic acids are referred to hereafter simply as "SNPs". A SNP is the occurrence of nucleotide variability at a single position in the genome, in which two alternative bases occur at appreciable frequency (i.e., >1%) in the human population. A SNP may occur within a gene or within intergenic regions of the genome. SNP probes according to the invention are oligonucleotides that are complementary to a SNP nucleic acid.
[35] As used herein, the term "subject" means that preferably the subject is a mammal, such as a human, but can also be an animal, e.g., domestic animals (e.g., dogs, cats and the like), farm animals (e.g., cows, sheep, pigs, horses and the like) and laboratory animals (e.g., monkey (e.g., cynmologous monkey, rats, mice, guinea pigs and the like). [36] As used herein, the administration of an agent or drug to a subject or patient includes self-administration and the administration by another. It is also to be appreciated that the various modes of treatment or prevention of medical conditions as described are intended to mean "substantial", which includes total but also less than total treatment or prevention, and wherein some biologically or medically relevant result is achieved.
[37] LRRK2 Modulating Agents. In one embodiment, the LRRK2 modulating agent can be a hetrocyclic compound inhibitor of LRRK2 protein (SEQ ID NO:2). [38] In several embodiments, the heterocyclic compound can be 5-[5-Methoxy-2-oxo-l,2- dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl- 1 H-pyrrole-3-carboxylic acid (3-amino- propyl)-amide; 5-[6-Methoxy-2-oxo- 1 ,2-dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl- 1 H- pyrrole-3-carboxylic acid (3-amino-propyl)-amide; 5-[7-Methoxy-2-oxo-l,2-dihydro-indol- (3Z)-ylidenemethyl]-2,4-dimethyl-l H-pyrrole-3-carboxylic acid (3-amino-propyl)-amide; 5- [5-Methoxy-2-oxo-l,2-dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl-lH-pyrrole-3- carboxylic acid (2-diethylamino-ethyl)-amide; or 5-[5-Dimethylsulfamoyl-2-oxo-l,2-dihydro- indol-(3Z)-ylidenemethyl]-2,4-dimethyl-l H-pyrrole-3-carboxylic acid (2-diethylamino-ethyl)- amide.
[39] In other embodiments, the heteroccyclic compound can be 3-[l-(3,5-Dimethyl-lH- pyrrol-2-yl)-methyl-(Z)-ylidene]-5-methoxy-l,3-dihydro-indol-2-one; 3-[l-(lH-Indol-2-yl)- meth-(Z)-ylidene]-5-methoxy-l,3-dihydro-indol-2-one; 5-Methoxy-3-[l-(4,5,6,7-tetrahydro- l H-indol-2-yl)-meth-(Z)-ylidene]-l,3-dihydro-indol-2-one; 3-[l-(3,5-Dimethyl-lH-pyrrol-2- yl)-meth-(Z)-ylidene]-5-methoxy-2-oxo-2,3-dihydro-indol-4-carboxylic acid methyl ester; ;3- [ 1 -(3,5-Dimethyl-l H-pyrrol-2-yl)-meth-(Z)-ylidene]-5-methoxy-2-oxo-2,3-dihydro-indol-4- carboxylic acid ethyl ester; 3-[l-(3,5-Dimethyl-lH-pyrrol-2-yl)-meth-(Z)-ylidene]-5- methoxy-2-oxo-2,3-dihydro-indol-4-carboxylic acid methyl ester; or 3-[l-(3,5-Dimethyl-lH- pyrrol-2-yl)-meth-(Z)-ylidene]-5-methoxy-2-oxo-2,3-dihydro-indol-4-carboxylic acid. [40] Alternatively, the heterocyclic compound may be selected from:
Figure imgf000009_0001
or
Figure imgf000010_0001
[Compound 14]
[41] The pharmacological properties of the LRRK2 modulating agents can be evaluated, for example, in Drug Pull-Down experiments. The above-mentioned heterocyclic compounds can show activity in Drug Pull-Down experiments at concentrations below 20 μM. Compound 12 shows an IC50 value of ~1 μM.
[42] The LRRK2 gene (SEQ ID NO: l)may play a role in progression from mild cognitive impairment to Alzheimer's disease and progression from moderate Alzheimer's disease to more severe Alzheimer's disease. Therefore, the LRRK2 modulating agents may be able to be used to treat patients with MCI or Alzheimer's disease, to slow the progression from mild cognitive impairment to Alzheimer's disease or from moderate Alzheimer's disease to more severe Alzheimer's disease.
[43] Identification and Characterization of Gene Sequence Variation. Due to their prevalence and widespread nature, SNPs have the potential to be important tools for locating genes that are involved in human disease conditions. See e.g., Wang et al, Science 280: 1077-1082 (1998). It is increasingly clear that the risk of developing many common disorders and the metabolism of medications used to treat these conditions are substantially influenced by underlying genomic variations, although the effects of any one variant might be small. [44] A SNP is said to be "allelic" in that due to the existence of the polymorphism, some members of a species may have an unmutated sequence {i.e., the original allele) whereas other members may have a mutated sequence {i.e., the variant or mutant allele). [45] An association between a SNP and a particular phenotype does not necessarily indicate or require that the SNP is causative of the phenotype. Instead, the association may merely be due to genome proximity between a SNP and those genetic factors actually responsible for a given phenotype, such that the SNP and said genetic factors are closely linked. That is, a SNP may be in linkage disequilibrium ("LD") with the "true" functional variant. LD {a.k.a., allelic association) exists when alleles at two distinct locations of the genome are more highly associated than expected. Thus, a SNP may serve as a marker that has value by virtue of its proximity to a mutation that causes a particular phenotype. [46] In describing the polymorphic sites of the invention, reference is made to the sense strand of the gene for convenience. As recognized by the skilled artisan, however, nucleic acid molecules containing the gene may be complementary double stranded molecules and thus reference to a particular site on the sense strand refers as well to the corresponding site on the complementary antisense strand. That is, reference may be made to the same polymorphic site on either strand and an oligonucleotide may be designed to hybridize specifically to either strand at a target region containing the polymorphic site. Thus, the invention also includes single-stranded polynucleotides that are complementary to the sense strand of the genomic variants described herein.
[47] Identification and Characterization of SNPs, Many different techniques can be used to identify and characterize SNPs, including single-strand conformation polymorphism (SSCP) analysis, heteroduplex analysis by denaturing high-performance liquid chromatography (DHPLC) and direct DNA sequencing and computational methods. Shi et al, Clin. Chem. 47:164-172 (2001). There is a wealth of sequence information in public databases. [48] The most common SNP-typing methods currently include hybridization, primer extension, and cleavage methods. Each of these methods must be connected to an appropriate detection system. Detection technologies include fluorescent polarization (Chan et al., Genome Res. 9:492-499 (1999)), luminometric detection of pyrophosphate release (pyrosequencing) (Ahmadiian et α/., Anal. Biochem. 280:103-10 (2000)), fluorescence resonance energy transfer (FRET)-based cleavage assays, DHPLC, and mass spectrometry (Shi, Clin. Chem. 47:164-172 (2001); U.S. Pat. No. 6,300,076 Bl). Other methods of detecting and characterizing SNPs are those disclosed in U.S. Pat. Nos. 6,297,018 and 6,300,063.
[49] Polymorphisms can also be detected using commercially available products, such as INVADER™ technology (available from Third Wave Technologies Inc. Madison, Wisconsin, USA). In this assay, a specific upstream "invader" oligonucleotide and a partially overlapping downstream probe together form a specific structure when bound to complementary DNA template. This structure is recognized and cut at a specific site by the Cleavase enzyme, resulting in the release of the 5' flap of the probe oligonucleotide. This fragment then serves as the "invader" oligonucleotide with respect to synthetic secondary targets and secondary fluorescently labelled signal probes contained in the reaction mixture. See also, Ryan D et al, Molecular Diagnosis 4(2): 135-144 (1999) and Lyamichev V et ah, Nature Biotechnology 17: 292-296 (1999), see also U.S. Pat. Nos. 5,846,717 and 6,001,567. [50] The identity of polymorphisms may also be determined using a mismatch detection technique including, but not limited to, the RNase protection method using riboprobes (Winter et al, Proc. Natl. Acad. ScL USA 82:7575 (1985); Meyers et al, Science 230:1242 (1985)) and proteins which recognize nucleotide mismatches, such as the E. coli mutS protein (Modrich P, Ann Rev Genet 25:229-253 (1991)). Alternatively, variant alleles can be identified by single strand conformation polymorphism (SSCP) analysis (Orita et ah, Genomics 5:874-879 (1989); Humphries et al, in Molecular Diagnosis of Genetic Diseases, R. Elles, ed., pp. 321-340 (1996)) or denaturing gradient gel electrophoresis (DGGE) (Wartell et al, Nucl Acids. Res. 18:2699-2706 (1990); Sheffield et al, Proc. Natl. Acad. Sci. USA 86:232-236 (1989)). A polymerase-mediated primer extension method may also be used to identify the polymorphisms. Several such methods have been described in the patent and scientific literature and include the "Genetic Bit Analysis" method (WO 92/15712) and the ligase/polymerase mediated genetic bit analysis (U.S. Pat. No. 5,679,524). Related methods are disclosed in WO 91/02087, WO 90/09455, WO 95/17676, and U.S. Pat. Nos. 5,302,509 and 5,945,283. Extended primers containing a polymorphism may be detected by mass spectrometry as described in U.S. Pat. No. 5,605,798. Another primer extension method is allele-specific PCR (Ruafio et al, Nucl Acids. Res. 17:8392 (1989); Ruafio et al, Nucl Acids. Res. 19: 6877-6882 (1991); WO 93/22456; Turki et al, J. Clin. Invest. 95:1635-1641 (1995)). In addition, multiple polymorphic sites may be investigated by simultaneously amplifying multiple regions of the nucleic acid using sets of allele-specific primers as described in PCT patent application WO 89/10414.
[51] In one embodiment, applicable to the results shown in the EXAMPLES below, blood samples from patients can be collected at the time of patient screening and DNA was extracted using, for example, the PUREGENE™ DNA Isolation Kit (D-50K). Genotyping can be performed using the TaqMan® technology or using the Third Wave Technologies Invader Assay technique.
[52] Haplotyping and Genotyping Oligonucleotides. The invention provides methods and compositions for haplotyping and/or genotyping the gene in an individual. As used herein, the terms "genotype" and "haplotype" mean the genotype or haplotype containing the nucleotide pair or nucleotide, respectively, that is present at one or more of the polymorphic sites described herein and may optionally also include the nucleotide pair or nucleotide present at one or more additional polymorphic sites in the gene. The additional polymorphic sites may be currently known polymorphic sites or sites that are subsequently discovered. [53] The compositions of the invention contain oligonucleotide probes and primers designed to specifically hybridize to one or more target regions containing, or that are adjacent to, a polymorphic site. Oligonucleotide compositions of the invention are useful in methods for genotyping and/or haplotyping a gene in an individual. The methods and compositions for establishing the genotype or haplotype of an individual at the polymorphic sites described herein are useful for studying the effect of the polymorphisms in the aetiology of diseases affected by the expression and function of the protein, studying the efficacy of drugs targeting, predicting individual susceptibility to diseases affected by the expression and function of the protein and predicting individual responsiveness to drugs targeting the gene product.
[54] Genotyping oligonucleotides of the invention may be immobilized on or synthesized on a solid surface such as a microchip, bead, or glass slide. See, e.g., WO 98/20020 and WO 98/20019.
[55] Genotyping oligonucleotides may hybridize to a target region located one to several nucleotides downstream of one of the polymorphic sites identified herein. Such oligonucleotides are useful in polymerase-mediated primer extension methods for detecting one of the polymorphisms described herein and therefore such genotyping oligonucleotides are referred to herein as "primer-extension oligonucleotides".
[56] Direct Genotyping Method of the Invention. A genotyping method of the invention may involve isolating from an individual a nucleic acid mixture comprising the two copies of a gene of interest or fragment thereof, and determining the identity of the nucleotide pair at one or more of the polymorphic sites in the two copies. As will be readily understood by the skilled artisan, the two "copies" of a gene in an individual may be the same allele or may be different alleles. In a particularly preferred embodiment, the genotyping method comprises determining the identity of the nucleotide pair at each polymorphic site. Typically, the nucleic acid mixture is isolated from a biological sample taken from the individual, such as a blood sample or tissue sample. Suitable tissue samples include whole blood, semen, saliva, tears, urine, faecal material, sweat, buccal smears, skin and hair. [57] Direct Haplotyping Method of the Invention. A haplotyping method of the invention may include isolating from an individual a nucleic acid molecule containing only one of the two copies of a gene of interest, or a fragment thereof, and determining the identity of the nucleotide at one or more of the polymorphic sites in that copy. Direct haplotyping methods include, for example, CLASPER System™ technology (U.S. Pat. No. 5,866,404) or allele- specific long-range PCR (Michalotos-Beloin et al, Nucl. Acids. Res. 24: 4841-4843 (1996)). The nucleic acid may be isolated using any method capable of separating the two copies of the gene or fragment. As will be readily appreciated by those skilled in the art, any individual clone will only provide haplotype information on one of the two gene copies present in an individual. In one embodiment, a haplotype pair is determined for an individual by identifying the phased sequence of nucleotides at one or more of the polymorphic sites in each copy of the gene that is present in the individual. In a preferred embodiment, the haplotyping method comprises identifying the phased sequence of nucleotides at each polymorphic site in each copy of the gene.
[58] In both the genotyping and haplotyping methods, the identity of a nucleotide (or nucleotide pair) at a polymorphic site may be determined by amplifying a target regions containing the polymorphic sites directly from one or both copies of the gene, or fragments thereof, and sequencing the amplified regions by conventional methods. The genotype or haplotype for the gene of an individual may also be determined by hybridization of a nucleic sample containing one or both copies of the gene to nucleic acid arrays and subarrays such as described in published PCT patent application WO 95/11995.
[59] Indirect Genotyping Method using Polymorphic Sites in Linkage Disequilibrium with a Target Polymorphism. In addition, the identity of the alleles present at any of the polymorphic sites of the invention may be indirectly determined by genotyping other polymorphic sites in linkage disequilibrium with those sites of interest. As described above, two sites are said to be in linkage disequilibrium if the presence of a particular variant at one site is indicative of the presence of another variant at a second site. Stevens JC, MoI. Diag. 4: 309-317 (1999). Polymorphic sites in linkage disequilibrium with the polymorphic sites of the invention may be located in regions of the same gene or in other genomic regions. [60] Amplifying a Target Gene Region. The target regions may be amplified using any oligonucleotide-directed amplification method, including but not limited to polymerase chain reaction (PCR). (U.S. Pat. No. 4,965,188), ligase chain reaction (LCR) (Barany et al, Proc. Natl. Acad. ScL USA 88:189-193 (1991); published PCT patent application WO 90/01069), and oligonucleotide ligation assay (OLA) (Landegren et al, Science 241 : 1077-1080 (1988)). Oligonucleotides useful as primers or probes in such methods should specifically hybridize to a region of the nucleic acid that contains or is adjacent to the polymorphic site. Typically, the oligonucleotides are between 10 and 35 nucleotides in length and preferably, between 15 and 30 nucleotides in length. Most preferably, the oligonucleotides are 20 to 25 nucleotides long. The exact length of the oligonucleotide will depend on many factors that are routinely considered and practiced by the skilled artisan.
[61] Other known nucleic acid amplification procedures may be used to amplify the target region including transcription-based amplification systems (U.S. Pat. No. 5,130,238; EP 329,822; U.S. Pat. No. 5,169,766, published PCT patent application WO 89/06700) and isothermal methods (Walker et al, Proc. Natl. Acad. ScL USA 89:392-396 (1992)). [62] Hybridizing Allele-Specific Oligonucleotide to a Target Gene. A polymorphism in the target region may be assayed before or after amplification using one of several hybridization- based methods known in the art. Typically, allele-specific oligonucleotides are utilized in performing such methods. The allele-specific oligonucleotides may be used as differently labelled probe pairs, with one member of the pair showing a perfect match to one variant of a target sequence and the other member showing a perfect match to a different variant. In some embodiments, more than one polymorphic site may be detected at once using a set of allele- specific oligonucleotides or oligonucleotide pairs. Preferably, the members of the set have melting temperatures within 5°C, and more preferably within 2°C, of each other when hybridizing to each of the polymorphic sites being detected.
[63] Hybridization of an allele-specific oligonucleotide to a target polynucleotide may be performed with both entities in solution, or such hybridization may be performed when either the oligonucleotide or the target polynucleotide is covalently or noncovalently affixed to a solid support. Attachment may be mediated, for example, by antibody-antigen interactions, poly-L-Lys, streptavidin or avidin-biotin, salt bridges, hydrophobic interactions, chemical linkages, UV cross-linking, baking, etc. Allele-specific oligonucleotide may be synthesized directly on the solid support or attached to the solid support subsequent to synthesis. Solid- supports suitable for use in detection methods of the invention include substrates made of silicon, glass, plastic, paper and the like, which may be formed, for example, into wells {as in 96-well plates), slides, sheets, membranes, fibres, chips, dishes, and beads. The solid support may be treated, coated or derivatised to facilitate the immobilization of the allele-specific oligonucleotide or target nucleic acid.
[64] Determining Population Genotypes and Haplotypes and Correlating Them with a Trait. The invention provides a method for determining the frequency of a genotype or haplotype in a population. The method comprises determining the genotype or the haplotype for a gene present in each member of the population, wherein the genotype or haplotype comprises the nucleotide pair or nucleotide detected at one or more of the polymorphic sites in the gene, and calculating the frequency at which the genotype or haplotype is found in the population. The population may be a reference population, a family population, a same sex population, a population group, or a trait population (e.g., a group of individuals exhibiting a trait of interest such as a medical condition or response to a therapeutic treatment). [65] In another aspect of the invention, frequency data for genotypes and/or haplotypes found in a reference population are used in a method for identifying an association between a trait and a genotype or a haplotype. The trait may be any detectable phenotype, including but not limited to susceptibility to a disease or response to a treatment. The method involves obtaining data on the frequency of the genotypes or haplotypes of interest in a reference population and comparing the data to the frequency of the genotypes or haplotypes in a population exhibiting the trait. Frequency data for one or both of the reference and trait populations may be obtained by genotyping or haplotyping each individual in the populations using one of the methods described above. The haplotypes for the trait population may be determined directly or, alternatively, by the predictive genotype to haplotype approach described above.
[66] The frequency data for the reference and/or trait populations are obtained by accessing previously determined frequency data, which may be in written or electronic form. For example, the frequency data may be present in a database that is accessible by a computer. Once the frequency data are obtained, the frequencies of the genotypes or haplotypes of interest in the reference and trait populations are compared.
[67] When polymorphisms are being analyzed, a calculation may be performed to correct for a significant association that might be found by chance. For statistical methods useful in the methods of the invention, see Statistical Methods in Biology, 3rd edition, Bailey NTJ, (Cambridge Univ. Press, 1997); Waterman MS, Introduction to Computational Biology (CRC Press, 2000) and Bioinformatics, Baxevanis AD & Ouellette BFF editors (John Wiley & Sons, Inc., 2001).
[68] In another embodiment, the haplotype frequency data for different groups are examined to determine whether they are consistent with Hardy- Weinberg equilibrium. D.L. Hartl et ah, Principles of Population Genomics, 3rd Ed. (Sinauer Associates, Sunderland, Massachusetts, 1997).
[69] In another embodiment, statistical analysis is performed by the use of standard ANOVA tests with a Bonferoni correction or a bootstrapping method that simulates the genotype phenotype correlation many times and calculates a significance value. ANOVA is used to test hypotheses about whether a response variable is caused by or correlates with one or more traits or variables that can be measured. LD Fisher & G vanBelle, Biostatistics: A Methodology for the Health Sciences, Ch. 10 (Wiley-lnterscience, New York, 1993). [70] In one embodiment for predicting a haplotype pair, the analysis includes an assigning step, as follows: First, each of the possible haplotype pairs is compared to the haplotype pairs in the reference population. Generally, only one of the haplotype pairs in the reference population matches a possible haplotype pair and that pair is assigned to the individual. Occasionally, only one haplotype represented in the reference haplotype pairs is consistent with a possible haplotype pair for an individual, and in such cases the individual is assigned a haplotype pair containing this known haplotype and a new haplotype derived by subtracting the known haplotype from the possible haplotype pair.
[71] In another embodiment, a detectable genotype or haplotype that is in linkage disequilibrium with a genotype or haplotype of interest may be used as a surrogate marker. A genotype that is in linkage disequilibrium with another genotype is indicated where a particular genotype or haplotype for a given gene is more frequent in the population that also demonstrates the potential surrogate marker genotype than in the reference population. If the frequency is statistically significant, then the marker genotype is predictive of that genotype or haplotype, and can be used as a surrogate marker.
[72] Another method for finding correlations between haplotype content and clinical responses uses predictive models based on error-minimizing optimization algorithms, one of which is a genetic algorithm. See, R Judson, "Genetic Algorithms and Their Uses in Chemistry" in Reviews in Computational Chemistry, Ch. 10, KB Lipkowitz & DB Boyd, eds. (VCH Publishers, New York, 1997) pp. 1-73. Simulated annealing (Press et ah, Numerical Recipes in C: The Art of Scientific Computing, Ch. 10 (Cambridge University Press, Cambridge, 1992), neural networks (E Rich & K Knight, Artificial Intelligence, 2nd Edition, Ch. 10 (McGraw-Hill, New York, 1991), standard gradient descent methods (Press et ah, supra Ch. 10), or other global or local optimization approaches (see discussion in Judson, supra) can also be used.
[73] Correlating Subject Genotype or Haplotype to Treatment Response. In preferred embodiments, the trait is susceptibility to a disease, severity of a disease, the staging of a disease or response to a drug. Such methods have applicability in developing diagnostic tests and therapeutic treatments for all pharmacogenetic applications where there is the potential for an association between a genotype and a treatment outcome, including efficacy measurements, pharmacokinetic measurements and side-effect measurements. [74] In another preferred embodiment, the trait of interest is a clinical response exhibited by a patient to some therapeutic treatment, for example, response to a drug targeting or to a therapeutic treatment for a medical condition.
[75] To deduce a correlation between a clinical response to a treatment and a genotype or haplotype, genotype or haplotype data is obtained on the clinical responses exhibited by a population of individuals who received the treatment, hereinafter the "clinical population". This clinical data may be obtained by analyzing the results of a clinical trial that has already been run and/or by designing and carrying out one or more new clinical trials. [76] The individuals included in the clinical population are usually graded for the existence of the medical condition of interest. This grading of potential patients could employ a standard physical exam or one or more lab tests. Alternatively, grading of patients could use haplotyping for situations where there is a strong correlation between haplotype pair and disease susceptibility or severity.
[77] The therapeutic treatment of interest is administered to each individual in the trial population, and each individual's response to the treatment is measured using one or more predetermined criteria. It is contemplated that in many cases, the trial population will exhibit a range of responses and that the investigator will choose the number of responder groups {e.g., low, medium, high) made up by the various responses. In addition, the gene for each individual in the trial population is genotyped and/or haplotyped, which may be done before or after administering the treatment. [78] These results are then analyzed to determine if any observed variation in clinical response between polymorphism groups is statistically significant. Statistical analysis methods, which may be used, are described in L.D. Fisher & G. vanBelle, Biostatistics: A Methodology for the Health Sciences (Wiley-lnterscience, New York, 1993). This analysis may also include a regression calculation of which polymorphic sites in the gene contribute most significantly to the differences in phenotype.
[79] In one embodiment, as a first pass analysis, Fishers Exact tests are performed to evaluate response as a function of genotype.
[80] After both the clinical and polymorphism data have been obtained, correlations between individual response and genotype or haplotype content are created. Correlations may be produced in several ways. In one method, individuals are grouped by their genotype or haplotype (or haplotype pair) (also referred to as a polymorphism group), and then the averages and standard deviations of clinical responses exhibited by the members of each polymorphism group are calculated.
[81] From the analyses described above, the skilled artisan that predicts clinical response as a function of genotype or haplotype content may readily construct a mathematical model. The identification of an association between a clinical response and a genotype or haplotype (or haplotype pair) for the gene may be the basis for designing a diagnostic method to determine those individuals who will or will not respond to the treatment, or alternatively, will respond at a lower level and thus may require more treatment, i.e., a greater dose of a drug. The diagnostic method may take one of several forms: for example, a direct DNA test {i.e., genotyping or haplotyping one or more of the polymorphic sites in the gene), a serological test, or a physical exam measurement. The only requirement is that there be a good correlation between the diagnostic test results and the underlying genotype or haplotype. In a preferred embodiment, this diagnostic method uses the predictive haplotyping method described above.
[82] In one embodiment, analysis is performed using a logistic remodel to take into account gender and age in addition to treatment and "high responder" (to therapeutic treatment) genotype status. In addition, an ANCOVA model can applied using the baseline value of patient response assessments as a quantitative co-variant.
[83] Assigning a Subject to a Genotype Group. As one of skill in the art will understand, there will be a certain degree of uncertainty involved in making this determination. Therefore, the standard deviations of the control group levels would be used to make a probabilistic determination and the methods of this invention would be applicable over a wide range of probability based genotype group determinations. Thus, for example and not by way of limitation, in one embodiment, if the measured level of the gene expression product falls within 2.5 standard deviations of the mean of any of the control groups, then that individual may be assigned to that genotype group. In another embodiment if the measured level of the gene expression product falls within 2.0 standard deviations of the mean of any of the control groups then that individual may be assigned to that genotype group. In still another embodiment, if the measured level of the gene expression product falls within 1.5 standard deviations of the mean of any of the control groups then that individual may be assigned to that genotype group. In yet another embodiment, if the measured level of the gene expression product is 1.0 or less standard deviations of the mean of any of the control groups levels then that individual may be assigned to that genotype group.
[84] Thus this process allows determination, with various degrees of probability, which group a specific subject should be placed in, and such assignment to a genotype group would then determine the risk category into which the individual should be placed. [85] Correlation between Clinical Response and Genotype or Haplotype. In order to deduce a correlation between clinical response to a treatment and a genotype or haplotype, it is necessary to obtain data on the clinical responses exhibited by a population of individuals who received the treatment, hereinafter the "clinical population." This clinical data may be obtained by analyzing the results of a clinical trial that has already been run and/or the clinical data may be obtained by designing and carrying out one or more new clinical trials. [86] The standard control levels of the gene expression product, thus determined in the different control groups, would then be compared with the measured level of a gene expression product in a given patient. This gene expression product could be the characteristic mRNA associated with that particular genotype group or the polypeptide gene expression product of that genotype group. The patient could then be classified or assigned to a particular genotype group based on how similar the measured levels were compared to the control levels for a given group.
[87] Computer System for Storing or Displaying Polymorphism Data. The invention also provides a computer system for storing and displaying polymorphism data determined for the gene. The computer system comprises a computer processing unit, a display, and a database containing the polymorphism data. The polymorphism data includes the polymorphisms, the genotypes and the haplotypes identified for a given gene in a reference population. In a preferred embodiment, the computer system is capable of producing a display showing haplotypes organized according to their evolutionary relationships. A computer may implement any or all analytical and mathematical operations involved in practicing the methods of the present invention. In addition, the computer may execute a program that generates views (or screens) displayed on a display device and with which the user can interact to view and analyze large amounts of information relating to the gene and its genomic variation, including chromosome location, gene structure, and gene family, gene expression data, polymorphism data, genetic sequence data, and clinical population data (e.g., data on ethnogeographic origin, clinical responses, genotypes, and haplotypes for one or more populations). The polymorphism data described herein may be stored as part of a relational database (e.g., an instance of an Oracle database or a set of ASCII flat files). These polymorphism data may be stored on the computer's hard drive or may, for example, be stored on a CD-ROM or on one or more other storage devices accessible by the computer. For example, the data may be stored on one or more databases in communication with the computer via a network.
[88] Nucleic Acid-based Diagnostics. In another aspect, the invention provides SNP probes, which are useful in classifying subjects according to their types of genetic variation. The SNP probes according to the invention are oligonucleotides, which discriminate between SNPs in conventional allelic discrimination assays. In certain preferred embodiments, the oligonucleotides according to this aspect of the invention are complementary to one allele of the SNP nucleic acid, but not to any other allele of the SNP nucleic acid. Oligonucleotides according to this embodiment of the invention can discriminate between SNPs in various ways. For example, under stringent hybridization conditions, an oligonucleotide of appropriate length will hybridize to one SNP, but not to any other. The oligonucleotide may be labelled using a radiolabel or a fluorescent molecular tag. Alternatively, an oligonucleotide of appropriate length can be used as a primer for PCR, wherein the 3' terminal nucleotide is complementary to one allele containing a SNP, but not to any other allele. In this embodiment, the presence or absence of amplification by PCR determines the haplotype of the SNP. [89] Genomic and cDNA fragments of the invention comprise at least one polymorphic site identified herein, have a length of at least 10 nucleotides, and may range up to the full length of the gene. Preferably, a fragment according to the present invention is between 100 and 3000 nucleotides in length, and more preferably between 200 and 2000 nucleotides in length, and most preferably between 500 and 1000 nucleotides in length.
[90] Kits of the Invention. The invention provides nucleic acid and polypeptide detection kits useful for haplotyping and/or genotyping the gene in an individual. Such kits are useful for classifying individuals for the purpose of classifying individuals. Specifically, the invention encompasses kits for detecting the presence of a polypeptide or nucleic acid corresponding to a marker of the invention in a biological sample, e.g., any bodily fluid including, but not limited to, serum, plasma, lymph, cystic fluid, urine, stool, cerebrospinal fluid, ascities fluid or blood, and including biopsy samples of body tissue. For example, the kit can comprise a labelled compound or agent capable of detecting a polypeptide or an mRNA encoding a polypeptide corresponding to a marker of the invention in a biological sample and means for determining the amount of the polypeptide or mRNA in the sample, e.g., an antibody which binds the polypeptide or an oligonucleotide probe which binds to DNA or mRNA encoding the polypeptide. Kits can also include instructions for interpreting the results obtained using the kit.
[91] In another embodiment, the invention provides a kit comprising at least two genotyping oligonucleotides packaged in separate containers. The kit may also contain other components such as hybridization buffer (where the oligonucleotides are to be used as a probe) packaged in a separate container. Alternatively, where the oligonucleotides are to be used to amplify a target region, the kit may contain, packaged in separate containers, a polymerase and a reaction buffer optimized for primer extension mediated by the polymerase, such as in the case of PCR. In a preferred embodiment, such kit may further comprise a DNA sample collecting means.
[92] For antibody-based kits, the kit can comprise, e.g., (1) a first antibody, e.g., attached to a solid support, which binds to a polypeptide corresponding to a marker or the invention; and, optionally (2) a second, different antibody which binds to either the polypeptide or the first antibody and is conjugated to a detectable label.
[93] For oligonucleotide-based kits, the kit can comprise, e.g., (1) an oligonucleotide, e.g., a detectably-labelled oligonucleotide, which hybridizes to a nucleic acid sequence encoding a polypeptide corresponding to a marker of the invention; or (2) a pair of primers useful for amplifying a nucleic acid molecule corresponding to a marker of the invention. [94] The kit can also comprise, e.g., a buffering agent, a preservative or a protein- stabilizing agent. The kit can further comprise components necessary for detecting the detectable-label, e.g., an enzyme or a substrate. The kit can also contain a control sample or a series of control samples, which can be assayed and compared to the test sample. Each component of the kit can be enclosed within an individual container and all of the various containers can be within a single package, along with instructions for interpreting the results of the assays performed using the kit.
[95] Nucleic Acid Sequences of the Invention. In one aspect, the invention comprises one or more isolated polynucleotides. The invention also encompasses allelic variants of the same, that is, naturally occurring alternative forms of the isolated polynucleotides that encode mutant polypeptides that are identical, homologous or related to those encoded by the polynucleotides. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis techniques well-known in the art. [96] Accordingly, nucleic acid sequences capable of hybridizing at low stringency with any nucleic acid sequences encoding mutant polypeptide of the present invention are considered to be within the scope of the invention. Standard stringency conditions are well characterized in standard molecular biology cloning texts. See, for example Molecular Cloning A Laboratory Manual, 2nd Ed., ed., Sambrook, Fritsch, & Maniatis (Cold Spring Harbor Laboratory Press, 1989); DNA Cloning, Volumes I and II, D.N. Glover, ed. (1985); Oligonucleotide Synthesis, M.J. Gait, ed. (1984); Nucleic Acid Hybridization, B.D. Hames & SJ. Higgins, eds (1984).
[97] Characterizing Gene Expression Level. Methods to detect and measure mRNA levels (i.e., gene transcription level) and levels of polypeptide gene expression products (i.e., gene translation level) are well-known in the art and include the use of nucleotide microarrays and polypeptide detection methods involving mass spectrometers and/or antibody detection and quantification techniques. See also, Tom Strachan & Andrew Read, Human Molecular Genetics, 2nd Edition. (John Wiley and Sons, Inc. Publication, New York, 1999)). [98] Determination of Target Gene Transcription. The determination of the level of the expression product of the gene in a biological sample, e.g., the tissue or body fluids of an individual, may be performed in a variety of ways. The term "biological sample" is intended to include tissues, cells, biological fluids and isolates thereof, isolated from a subject, as well as tissues, cells and fluids present within a subject. Many expression detection methods use isolated RNA. For in vitro methods, any RNA isolation technique that does not select against the isolation of mRNA can be utilized for the purification of RNA from cells. See, e.g., Ausubel et al, Ed., Curr. Prot. MoI. Biol. (John Wiley & Sons, New York, 1987-1999). [99] In one embodiment, the level of the mRNA expression product of the target gene is determined. Methods to measure the level of a specific mRNA are well-known in the art and include Northern blot analysis, reverse transcription PCR and real time quantitative PCR or by hybridization to a oligonucleotide array or microarray. In other more preferred embodiments, the determination of the level of expression may be performed by determination of the level of the protein or polypeptide expression product of the gene in body fluids or tissue samples including but not limited to blood or serum. Large numbers of tissue samples can readily be processed using techniques well-known to those of skill in the art, such as, e.g., the single-step RNA isolation process of U.S. Pat. No. 4,843,155. [100] The isolated mRNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, PCR analyses and probe arrays. One preferred diagnostic method for the detection of mRNA levels involves contacting the isolated mRNA with a nucleic acid molecule (probe) that can hybridize to the mRNA encoded by the gene being detected. The nucleic acid probe can be, e.g., a full-length cDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to an mRNA or genomic DNA encoding a marker of the present invention. Other suitable probes for use in the diagnostic assays of the invention are described herein. Hybridization of an mRNA with the probe indicates that the marker in question is being expressed. [101] In one format, the probes are immobilized on a solid surface and the mRNA is contacted with the probes, for example, in an Affymetrix gene chip array (Affymetrix, Calif. USA). A skilled artisan can readily adapt known mRNA detection methods for use in detecting the level of mRNA encoded by the markers of the present invention. [102] An alternative method for determining the level of mRNA corresponding to a marker of the present invention in a sample involves the process of nucleic acid amplification, e.g., by RT-PCR (the experimental embodiment set forth in U.S. Pat. No. 4,683,202); ligase chain reaction (Barany et al, Proc. Natl. Acad. Sci. USA 88:189-193 (1991)) self-sustained sequence replication (Guatelli et al, Proc. Natl. Acad. Sci. USA 87: 1874-1878 (1990)); transcriptional amplification system (Kwoh et al, Proc. Natl. Acad. Sci. USA 86: 1173-1 177 (1989)); Q-Beta Replicase (Lizardi et al, Biol. Technology 6: 1197 (1988)); rolling circle replication (U.S. Pat. No. 5,854,033); or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well-known to those of skill in the art. These detection schemes are especially useful for the detection of the nucleic acid molecules if such molecules are present in very low numbers. As used herein, "amplification primers" are defined as being a pair of nucleic acid molecules that can anneal to 5' or 3' regions of a gene (plus and minus strands, respectively, or vice- versa) and contain a short region in between. In general, amplification primers are from about 10-30 nucleotides in length and flank a region from about 50-200 nucleotides in length. [103] Real-time quantitative PCR (RT-PCR) is one way to assess gene expression levels, e.g., of genes of the invention, e.g., those containing SNPs and polymorphisms of interest. The RT-PCR assay utilizes an RNA reverse transcriptase to catalyze the synthesis of a DNA strand from an RNA strand, including an mRNA strand. The resultant DNA may be specifically detected and quantified and this process may be used to determine the levels of specific species of mRNA. One method for doing this is TAQMAN® (PE Applied Biosystems, Foster City, Calif, USA) and exploits the 5' nuclease activity of AMPLITAQ GOLD™ DNA polymerase to cleave a specific form of probe during a PCR reaction. This is referred to as a TAQMAN™ probe. See Luthra et al., Am. J. Pathol. 153: 63-68 (1998); Kuimelis et al., Nucl. Acids Symp. Ser. 37: 255-256 (1997); and Mullah et al, Nucl Acids Res. 26(4): 1026-1031 (1998)). During the reaction, cleavage of the probe separates a reporter dye and a quencher dye, resulting in increased fluorescence of the reporter. The accumulation of PCR products is detected directly by monitoring the increase in fluorescence of the reporter dye. Heid et al, Genome Res. 6(6): 986-994 (1996)). The higher the starting copy number of nucleic acid target, the sooner a significant increase in fluorescence is observed. See Gibson, Heid & Williams et al, Genome Res. 6: 995-1001 (1996). [104] Other technologies for measuring the transcriptional state of a cell produce pools of restriction fragments of limited complexity for electrophoretic analysis, such as methods combining double restriction enzyme digestion with phasing primers (see, e.g., EP 0 534858 Al), or methods selecting restriction fragments with sites closest to a defined mRNA end. (See, e.g., Prashar & Weissman, Proc. Natl. Acad. Sci. USA 93(2) 659-663 (19%)). [105] Other methods statistically sample cDNA pools, such as by sequencing sufficient bases, e.g., 20-50 bases, in each of multiple cDNAs to identify each cDNA, or by sequencing short tags, e.g., 9-10 bases, which are generated at known positions relative to a defined mRNA end pathway pattern. See, e.g., Velculescu, Science 270: 484-487 (1995). The cDNA levels in the samples are quantified and the mean, average and standard deviation of each cDNA is determined using by standard statistical means well-known to those of skill in the art. Norman TJ. Bailey, Statistical Methods In Biology, 3rd Edition (Cambridge University Press, 1995).
[106] Detection of Polypeptides. Immunological Detection Methods. Expression of the protein encoded by the genes of the invention can be detected by a probe which is detectably labelled, or which can be subsequently labelled. The term "labelled", with regard to the probe or antibody, is intended to encompass direct-labelling of the probe or antibody by coupling, i.e., physically linking, a detectable substance to the probe or antibody, as well as indirect- labelling of the probe or antibody by reactivity with another reagent that is directly-labelled. Examples of indirect labelling include detection of a primary antibody using a fluorescently- labelled secondary antibody and end-labelling of a DNA probe with biotin such that it can be detected with fluorescently-labelled streptavidin. Generally, the probe is an antibody that recognizes the expressed protein. A variety of formats can be employed to determine whether a sample contains a target protein that binds to a given antibody. Immunoassay methods useful in the detection of target polypeptides of the present invention include, but are not limited to, e.g., dot blotting, western blotting, protein chips, competitive and noncompetitive protein binding assays, enzyme-linked immunosorbant assays (ELISA), immunohistochemistry, fluorescence activated cell sorting (FACS), and others commonly used and widely-described in scientific and patent literature, and many employed commercially. A skilled artisan can readily adapt known protein/antibody detection methods for use in determining whether cells express a marker of the present invention and the relative concentration of that specific polypeptide expression product in blood or other body tissues. Proteins from individuals can be isolated using techniques that are well-known to those of skill in the art. The protein isolation methods employed can, e.g., be such as those described in Harlow & Lane, Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1988)). [107] For the production of antibodies to a protein encoded by one of the disclosed genes, various host animals may be immunized by injection with the polypeptide, or a portion thereof. Such host animals may include, but are not limited to, rabbits, mice and rats. Various adjuvants may be used to increase the immunological response, depending on the host species including, but not limited to, Freund's (complete and incomplete), mineral gels, such as aluminium hydroxide; surface active substances, such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin and dinitrophenol; and potentially useful human adjuvants, such as bacille Camette-Guerin (BCG) and Corynebacterium parvum.
[108] Monoclonal antibodies (mAbs), which are homogeneous populations of antibodies to a particular antigen, may be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique of Kohler & Milstein, Nature 256: 495-497 (1975); and U.S. Pat. No. 4,376,110; the human B-cell hybridoma technique of Kosbor et ah, Immunol. Today 4: 72 (1983); Cole et ah, Proc. Natl. Acad. ScL USA 80: 2026-2030 (1983); and the EBV- hybridoma technique of Cole et ah, Monoclonal Antibodies and Cancer Therapy (Alan R. Liss, Inc., 1985) pp. 77-96.
[109] In addition, techniques developed for the production of "chimeric antibodies" (see Morrison et ah, Proc. Natl. Acad. ScL USA 81 : 6851-6855 (1984); Neuberger et ah, Nature 312: 604-608 (1984); and Takeda et ah, Nature 314: 452-454 (1985)), by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable or hypervariable region derived form a murine niAb and a human immunoglobulin constant region.
[1 10] Alternatively, techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778; Bird, Science 242: 423-426 (1988); Huston et ah, Proc. Natl. Acad. ScL USA 85: 5879-5883 (1988); and Ward et ah, Nature 334: 544-546 (1989)) can be adapted to produce differentially expressed gene single-chain antibodies.
[I l l] Techniques useful for the production of "humanized antibodies" can be adapted to produce antibodies to the proteins, fragments or derivatives thereof. Such techniques are disclosed in U.S. Pat. Nos. 5,932,448; 5,693,762; 5,693,761; 5,585,089; 5,530,101; 5,569,825; 5,625,126; 5,633,425; 5,789,650; 5,661,016; and 5,770,429. [1 12] Antibodies or antibody fragments can be used in methods, such as Western blots or immunofluorescence techniques, to detect the expressed proteins. In such uses, it is generally preferable to immobilize either the antibody or proteins on a solid support. Suitable solid phase supports or carriers include any support capable of binding an antigen or an antibody. Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros and magnetite.
[1 13] A useful method, for ease of detection, is the sandwich ELISA, of which a number of variations exist, all of which are intended to be used in the methods and assays of the present invention. As used herein, "sandwich assay" is intended to encompass all variations on the basic two-site technique. Immunofluorescence and EIA techniques are both very well- established in the art. However, other reporter molecules, such as radioisotopes, chemiluminescent or bioluminescent molecules may also be employed. It will be readily apparent to the skilled artisan how to vary the procedure to suit the required use. [1 14] Whole genome monitoring of protein, i.e., the "proteome," can be carried out by constructing a microarray in which binding sites comprise immobilized, preferably monoclonal, antibodies specific to a plurality of protein species encoded by the cell genome. Preferably, antibodies are present for a substantial fraction of the encoded proteins, or at least for those proteins relevant to testing or confirming a biological network model of interest. As noted above, methods for making monoclonal antibodies are well-known. See, e.g., Harlow & Lane, Antibodies: A Laboratory ManuaF (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York, 1988)). In a preferred embodiment, monoclonal antibodies are raised against synthetic peptide fragments designed based on genomic sequence of the cell. With such an antibody array, proteins from the cell are contacted to the array and their binding is measured with assays known in the art.
[115] Detection of Polypeptides. Two-Dimensional Gel Electrophoresis. Two-dimensional gel electrophoresis is well-known in the art and typically involves isoelectric focusing along a first dimension followed by SDS-PAGE electrophoresis along a second dimension. See, e.g., Hames et al, Gel Electrophoresis of Proteins: A Practical Approach (IRL Press, New York, 1990); Shevchenko et al, Proc. Natl. Acad. Sci. USA 93: 14440-14445 (1996); Sagliocco et al, Yeast 12: 1519-1533 (1996); and Lander, Science 274: 536-539 (1996). [1 16] Detection of Polypeptides. Mass Spectroscopy. The identity as well as expression level of target polypeptide can be determined using mass spectrocopy technique (MS). MS-based analysis methodology is useful for analysis of isolated target polypeptide as well as analysis of target polypeptide in a biological sample. MS formats for use in analyzing a target polypeptide include ionization (I) techniques, such as, but not limited to, matrix assisted laser desorption (MALDI), continuous or pulsed electrospray ionization (ESI) and related methods, such as ionspray or thermospray, and massive cluster impact (MCI). Such ion sources can be matched with detection formats, including linear or non-linear reflectron time of flight (TOF), single or multiple quadrupole, single or multiple magnetic sector, Fourier transform ion cyclotron resonance (FTICR), ion trap and combinations thereof such as ion-trap/TOF. For ionization, numerous matrix/wavelength combinations {e.g., matrix assisted laser desorption (MALDI)) or solvent combinations (e.g., ESI) can be employed.
[117] For mass spectroscopy (MS) analysis, the target polypeptide can be solubilised in an appropriate solution or reagent system. The selection of a solution or reagent system, e.g., an organic or inorganic solvent, will depend on the properties of the target polypeptide and the type of MS performed, and is based on methods well-known in the art. See, e.g., Vorm et al., Anal. Chem. 61 : 3281 (1994) for MALDI; and Valaskovic et al, Anal. Chem. 67: 3802 (1995), for ESI. MS of peptides also is described, e.g., in International PCT Application No. WO 93/24834 and U.S. Pat. No. 5,792,664. A solvent is selected that minimizes the risk that the target polypeptide will be decomposed by the energy introduced for the vaporization process. A reduced risk of target polypeptide decomposition can be achieved, e.g., by embedding the sample in a matrix. A suitable matrix can be an organic compound such as a sugar, e.g., a pentose or hexose, or a polysaccharide such as cellulose. Such compounds are decomposed thermolytically into CO2 and H2O such that no residues are formed that can lead to chemical reactions. The matrix also can be an inorganic compound, such as nitrate of ammonium, which is decomposed essentially without leaving any residue. Use of these and other solvents is known to those of skill in the art. See, e.g., U.S. Pat. No. 5,062,935. Electrospray MS has been described by Fenn et al, J. Phys. Chem. 88: 4451-4459 (1984); and PCT Application No. WO 90/14148; and current applications are summarized in review articles. See Smith et al., Anal. Chem. 62: 882-89 (1990); and Ardrey, Spectroscopy 4: 10-18 (1992).
[118] The mass of a target polypeptide determined by MS can be compared to the mass of a corresponding known polypeptide. For example, where the target polypeptide is a mutant protein, the corresponding known polypeptide can be the corresponding non-mutant protein, e.g., wild-type protein. With ESI, the determination of molecular weights in femtomole amounts of sample is very accurate due to the presence of multiple ion peaks, all of which can be used for mass calculation. Sub-attomole levels of protein have been detected, e.g., using ESI MS (Valaskovic et al, Science 273: 1199-1202 (1996)) and MALDI MS (Li et al, J. Am. Chem. Soc. 118: 1662-1663 (1996)).
[119] Matrix Assisted Laser Desorption (MALDI). The level of the target protein in a biological sample, e.g., body fluid or tissue sample, may be measured by means of mass spectrometric (MS) methods including, but not limited to, those techniques known in the art as matrix-assisted laser desorption/ionization, time-of-flight mass spectrometry (MALDI- TOF-MS) and surfaces enhanced for laser desorption/ionization, time-of-flight mass spectrometry (SELDI-TOF-MS) as further detailed below. Methods for performing MALDI are well-known to those of skill in the art. See, e.g., Juhasz et al, Analysis, Anal. Chem. 68: 941-946 (1996), and see also, e.g., U.S. Pat. Nos. 5,777,325; 5,742,049; 5,654,545; 5,641,959; 5,654,545 and 5,760,393 for descriptions of MALDI and delayed extraction protocols. Numerous methods for improving resolution are also known. MALDI-TOF-MS has been described by Hillenkamp et al, Biological Mass Spectrometry, Burlingame & McCloskey, eds. (Elsevier Science Publ., Amsterdam, 1990) pp. 49-60. [120] A variety of techniques for marker detection using mass spectroscopy can be used. See Bordeaux Mass Spectrometry Conference Report, Hillenkamp, Ed., pp. 354-362 (1988); Bordeaux Mass Spectrometry Conference Report, Karas & Hillenkamp, eds., pp. 416-417 (1988); Karas & Hillenkamp, Anal. Chem. 60: 2299-2301 (1988); and Karas et al, Biomed Environ. Mass Spectrum 18: 841-843 (1989). The use of laser beams in TOF-MS is shown, e.g., in U.S. Patent Nos. 4,694,167; 4,686,366, 4,295,046 and 5,045,694, which are incorporated herein by reference in their entireties. Other MS techniques allow the successful volatilization of high molecular weight biopolymers, without fragmentation, and have enabled a wide variety of biological macromolecules to be analyzed by mass spectrometry. [121] Surfaces Enhanced for Laser Desorption/lonization (SELDl). Other techniques are used which employ new MS probe element compositions with surfaces that allow the probe element to actively participate in the capture and docking of specific analytes, described as Affinity Mass Spectrometry (AMS). See SELDI patents U.S. Pat. Nos. 5,719,060; 5,894,063; 6,020,208; 6,027,942; 6,124,137; and U.S. Patent application No. U.S. 2003/0003465. Several types of new MS probe elements have been designed with Surfaces Enhanced for Affinity Capture (SEAC). See Hutchens & Yip, Rapid Commun. Mass Spectrom. 7: 576-580 (1993). SEAC probe elements have been used successfully to retrieve and tether different classes of biopolymers, particularly proteins, by exploiting what is known about protein surface structures and biospecific molecular recognition. The immobilized affinity capture devices on the MS probe element surface, i.e., SEAC, determines the location and affinity (specificity) of the analyte for the probe surface, therefore the subsequent analytical MS process is efficient.
[122] Within the general category of SELDI are three separate subcategories: (1) Surfaces Enhanced for Neat Desorption (SEND), where the probe element surfaces, i.e., sample presenting means, are designed to contain Energy Absorbing Molecules (EAM) instead of "matrix" to facilitate desorption/ionizations of analytes added directly (neat) to the surface. (2) SEAC, where the probe element surfaces, i.e., sample presenting means, are designed to contain chemically defined and/or biologically defined affinity capture devices to facilitate either the specific or non-specific attachment or adsorption (so-called docking or tethering) of analytes to the probe surface, by a variety of mechanisms (mostly non-covalent). (3) Surfaces Enhanced for Photolabile Attachment and Release (SEPAR), where the probe element surfaces, i.e., sample presenting means, are designed or modified to contain one or more types of chemically defined cross-linking molecules to serve as covalent docking devices. The chemical specificities determining the type and number of the photolabile molecule attachment points between the SEPAR sample presenting means (i.e., probe element surface) and the analyte (e.g., protein) may involve any one or more of a number of different residues or chemical structures in the analyte (e.g. , His, Lys, Arg, Tyr, Phe and Cys residues in the case of proteins and peptides).
[123] Other Aspects of the Biological State. In various embodiments of the invention, aspects of the biological activity state, or mixed aspects can be measured in order to obtain drug and pathway responses. The activities of proteins relevant to the characterization of cell function can be measured, and embodiments of this invention can be based on such measurements. Activity measurements can be performed by any functional, biochemical or physical means appropriate to the particular activity being characterized. Where the activity involves a chemical transformation, the cellular protein can be contacted with natural substrates, and the rate of transformation measured. Where the activity involves association in multimeric units, e.g., association of an activated DNA binding complex with DNA, the amount of associated protein or secondary consequences of the association, such as amounts of mRNA transcribed, can be measured. Also, where only a functional activity is known, e.g., as in cell cycle control, performance of the function can be observed. However known and measured, the changes in protein activities form the response data analyzed by the methods of this invention. In alternative and non-limiting embodiments, response data may be formed of mixed aspects of the biological state of a cell. Response data can be constructed from, e.g., changes in certain mRNA abundances, changes in certain protein abundances and changes in certain protein activities.
[124] The following EXAMPLES are presented in order to more fully illustrate the preferred embodiments of the invention. These EXAMPLE should in no way be construed as limiting the scope of the invention, as defined by the appended claims.
EXAMPLE 1
ASSOCIATION OF THE COMMON POLYMORPHISMS IN THE LRRK2 GENE WITH
PROGRESSION OF ALZHEIMER'S DISEASE (AD)
[125] The objective of this EXAMPLE was to test whether variations in the LRRK2 gene are associated with the progression to Alzheimer's disease (AD) in subjects with mild cognitive impairment (MCI).
[126] We tested for 2 common polymorphisms of the LRRK2 gene: T1602S and T2352M.
For the LRRK2 gene (SEQ ID NO:1), pairwise Linkage Disequilibrium (LD) analysis showed that T1602S and T2352M are in strong LD (D'=0.979). For the T1602S mutation, the allele frequency (for the minor allele) was found to be as follows for the patient populations: PD =
27%, AD = 28%, MCI = 29%, ALS = 31%.
[127] THR1602SER mutation. Progression to Alzheimer's disease data of a 3-4 year study in
537 subjects was used to investigate the effect of the two LRRK2 common polymorphisms on the likelihood of progression to AD. The Investigation Into Delay to Diagnosis of Alzheimer's Disease With Exelon (InDDex) study was a placebo-controlled, 4-year longitudinal study to evaluate efficacy of Exelon® in the individuals with mild cognitive impairment (MCI). The clinical trial followed patients suffering from mild cognitive impairment and given either Exelon® (rivastigmine) at various doses or placebo and followed their conversion to Alzheimer's disease (AD). Feldman H et ah, Neurology 62: 1199-1201 (2004). The trial had an optional DNA collection component.
[ 128] In the InDDEx (MCI) study, we found that the TT genotype (or Thr/Thr) of the polymorphism T1602S was significantly associated with higher rate of progression to Alzheimer's disease (TABLE 1).
TABLE 1
Rate of conversion from MCI to AD bv T1602S genotype
LRRK2 genotype (T 1602S)
Conversion Thr/Thr Thr/Ser Ser/Ser Hazard 95% CI P to AD (n=38) (n=174) (n=217) ratio* value**
Yes % 34 21 14 94 17 05
No, % 6579 KM Zlfs 3 °°9 α63' 5-56> °;°021
*Cox proportional hazards. Age, gender and years of education were included in the model.
**Log-rank test for the time to conversion.
[ 129] Similar to the APOE-E4 allele, in the presence of a BuChE-K variant, LRRK2 polymorphism T1602S showed a greater association with the rate of conversion from mild cognitive impairment to Alzheimer's disease.
[130] To verify these findings, we further tested the correlation between this common LRRK2 polymorphism and cognitive performance over 6 months in the 178 placebo-treated AD patients enrolled in the IDEAL study. In the IDEAL (AD) study, LRRK2 polymorphism T1602S showed a same trend of the association observed in the MCI study. The Alzheimer's disease patients with TT genotype of T1602S tended to decline faster on cognitive performance over 6 months, especially in the presence of a BuChE-K variant. [131] THR2352MET mutation. In addition, in the InDDeX study, the CC genotype (or Thr/Thr) of T2352 showed a trend of associated with higher rate of conversion from mild cognitive impairment to Alzheimer's disease (TABLE 2). TABLE 2
Rate of conversion from MCI to AD bv T2352M genotype
LRRK2 Renotype (T2352M)
Conversion Met/Met Thr/Met Thr/Thr Hazard 95% CI P to AD (n=60) Tn= 196) (n=188) ratio* value**
Yes, % 16.67 15.31 21.81 (0 93 2 22) 0 0823
No, % 83.33 84.69 78.19 ^ ' }
*Cox proportional hazards. Age, gender and years of education were included in the model as co variants.
**Log-rank test for the time to conversion.
[132] To verify these findings, we further tested the correlation between the two common LRRK2 polymorphisms (see above) and cognitive performance over 6 months in the 178 placebo-treated AD patients enrolled in the IDEAL study. In the IDEAL (AD) study, LRRK2 polymorphisms T1602S and T2352 both showed a same trend of the association observed in the MCI study. The Alzheimer's disease patients with CC genotype of LRRK2-T2352 tended to decline faster on cognitive performance over 6 months, especially in the presence of a BuChE-K variant.
[133] Thus, common polymorphisms in the LRRK2 gene influence the rate of progression to Alzheimer's disease in subjects with mild cognitive impairment, suggesting that LRRK2 affects Alzheimer's disease pathogenesis.
EXAMPLE 2
ANALYSIS OF GENETIC VARIATIONS OF LRRK2
[134] GLY2019SER mutation. In a screen of patients, with results confirmed by re- sequencing, we found the following: For Parkinson's disease (PD): 6 out of 483 patients carry the G2019S mutation (1.24%). For Parkinson's disease with dementia (PDD): 1 out of 391 patients carry the G2019S mutation (0.26%). For Alzheimer's disease (AD): None of the 373 patients carry the G2019S mutation. For Mild cognitive impairment (MCI): None of the 448 patients carry the G2019S mutation. For Amyotrophic lateral sclerosis (ALS): None of the 483 patients carry the G2019S mutation.
[135] Only 4 of the 6 subjects carrying the G2019S mutation had clinical data. All 4 are male Caucasians. Progress was relatively faster (mutant vs wild-type, for 26 weeks), with the following results: UPDRSII: 1 vs 0.27. UPDRSIII: 4 vs -0.15. [136] We concluded that: Approximately 1.24% sporadic late-onset cases carry the mutation, which is similar to the frequency reported in the literature. Only about 0.26% PDD cases carry the G2019S mutation. This mutation is not common in AD, MCI and ALS. The mutation may be correlated with faster decline of motor function
EQUIVALENTS
[137] The details of one or more embodiments of the invention are set forth in the accompanying description above. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described. Other features, objects, and advantages of the invention will be apparent from the description and the claims. In the specification and the appended claims, the singular forms include plural referents unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All references cited herein are incorporated herein by reference in their entirety and for all purposes to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.
[138] The present invention is not to be limited in terms of the particular embodiments described in this application, which are intended as single illustrations of individual aspects of the invention. Many modifications and variations of this invention can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods and apparatuses within the scope of the invention, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. The present invention is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. SEQUENCE LISTING
<210> 1
<211> 9234
<212> DNA
<213> Homo sapiens
<220> <221> gene <222> ( 1 ) . . . ( 9234 )
<223> Homo sapiens leucine-rich repeat kinase 2 ( LRRK2 ) , mRNA ( NM 198578 )
<400> 1 cgctggctgc gggcggtgag ctgagctcgc ccccggggag ctgtggccgg cgcccctgcc 60 ggttccctga gcagcggacg ttcatgctgg gagggcggcg ggttggaagc aggtgccacc 120 atggctagtg gcagctgtca ggggtgcgaa gaggacgagg aaactctgaa gaagttgata 180 gtcaggctga acaatgtcca ggaaggaaaa cagatagaaa cgctggtcca aatcctggag 240 gatctgctgg tgttcacgta ctccgagcac gcctccaagt tatttcaagg caaaaatatc 300 catgtgcctc tgttgatcgt cttggactcc tatatgagag tcgcgagtgt gcagcaggtg 360 ggttggtcac ttctgtgcaa attaatagaa gtctgtccag gtacaatgca aagcttaatg 420 ggaccccagg atgttggaaa tgattgggaa gtccttggtg ttcaccaatt gattcttaaa 480 atgctaacag ttcataatgc cagtgtaaac ttgtcagtga ttggactgaa gaccttagat 540 ctcctcctaa cttcaggtaa aatcaccttg ctgatattgg atgaagaaag tgatattttc 600 atgttaattt ttgatgccat gcactcattt ccagccaatg atgaagtcca gaaacttgga 660 tgcaaagctt tacatgtgct gtttgagaga gtctcagagg agcaactgac tgaatttgtt 720 gagaacaaag attatatgat attgttaagt gcgtcaacaa attttaaaga tgaagaggaa 780 attgtgcttc atgtgctgca ttgtttacat tccctagcga ttccttgcaa taatgtggaa 840 gtcctcatga gtggcaatgt caggtgttat aatattgtgg tggaagctat gaaagcattc 900 cctatgagtg aaagaattca agaagtgagt tgctgtttgc tccataggct tacattaggt 960 aattttttca atatcctggt attaaacgaa gtccatgagt ttgtggtgaa agctgtgcag 1020 cagtacccag agaatgcagc attgcagatc tcagcgctca gctgtttggc cctcctcact 1080 gagactattt tcttaaatca agatttagag gaaaagaatg agaatcaaga gaatgatgat 1140 gagggggaag aagataaatt gttttggctg gaagcctgtt acaaagcatt aacgtggcat 1200 agaaagaaca agcacgtgca ggaggccgca tgctgggcac taaataatct ccttatgtac 1260 caaaacagtt tacatgagaa gattggagat gaagatggcc atttcccagc tcatagggaa 1320 gtgatgctct ccatgctgat gcattcttca tcaaaggaag ttttccaggc atctgcgaat 1380 gcattgtcaa ctctcttaga acaaaatgtt aatttcagaa aaatactgtt atcaaaagga 1440 atacacctga atgttttgga gttaatgcag aagcatatac attctcctga agtggctgaa 1500 agtggctgta aaatgctaaa tcatcttttt gaaggaagca acacttccct ggatataatg 1560 gcagcagtgg tccccaaaat actaacagtt atgaaacgtc atgagacatc attaccagtg 1620 cagctggagg cgcttcgagc tattttacat tttatagtgc ctggcatgcc agaagaatcc 1680 agggaggata cagaatttca tcataagcta aatatggtta aaaaacagtg tttcaagaat 1740 gatattcaca aactggtcct agcagctttg aacaggttca ttggaaatcc tgggattcag 1800 aaatgtggat taaaagtaat ttcttctatt gtacattttc ctgatgcatt agagatgtta 1860 tccctggaag gtgctatgga ttcagtgctt cacacactgc agatgtatcc agatgaccaa 1920 gaaattcagt gtctgggttt aagtcttata ggatacttga ttacaaagaa gaatgtgttc 1980 ataggaactg gacatctgct ggcaaaaatt ctggtttcca gcttataccg atttaaggat 2040 gttgctgaaa tacagactaa aggatttcag acaatcttag caatcctcaa attgtcagca 2100 tctttttcta agctgctggt gcatcattca tttgacttag taatattcca tcaaatgtct 2160 tccaatatca tggaacaaaa ggatcaacag tttctaaacc tctgttgcaa gtgttttgca 2220 aaagtagcta tggatgatta cttaaaaaat gtgatgctag agagagcgtg tgatcagaat 2280 aacagcatca tggttgaatg cttgcttcta ttgggagcag atgccaatca agcaaaggag 2340 ggatcttctt taatttgtca ggtatgtgag aaagagagca gtcccaaatt ggtggaactc 2400 ttactgaata gtggatctcg tgaacaagat gtacgaaaag cgttgacgat aagcattggg 2460 aaaggtgaca gccagatcat cagcttgctc ttaaggaggc tggccctgga tgtggccaac 2520 aatagcattt gccttggagg attttgtata ggaaaagttg aaccttcttg gcttggtcct 2580 ttatttccag ataagacttc taatttaagg aaacaaacaa atatagcatc tacactagca 2640 agaatggtga tcagatatca gatgaaaagt gctgtggaag aaggaacagc ctcaggcagc 2700 gatggaaatt tttctgaaga tgtgctgtct aaatttgatg aatggacctt tattcctgac 2760 tcttctatgg acagtgtgtt tgctcaaagt gatgacctgg atagtgaagg aagtgaaggc 2820 tcatttcttg tgaaaaagaa atctaattca attagtgtag gagaatttta ccgagatgcc 2880 gtattacagc gttgctcacc aaatttgcaa agacattcca attccttggg gcccattttt 2940 gatcatgaag atttactgaa gcgaaaaaga aaaatattat cttcagatga ttcactcagg 3000 tcatcaaaac ttcaatccca tatgaggcat tcagacagca tttcttctct ggcttctgag 3060 agagaatata ttacatcact agacctttca gcaaatgaac taagagatat tgatgcccta 3120 agccagaaat gctgtataag tgttcatttg gagcatcttg aaaagctgga gcttcaccag 3180 aatgcactca cgagctttcc acaacagcta tgtgaaactc tgaagagttt gacacatttg 3240 gacttgcaca gtaataaatt tacatcattt ccttcttatt tgttgaaaat gagttgtatt 3300 gctaatcttg atgtctctcg aaatgacatt ggaccctcag tggttttaga tcctacagtg 3360 aaatgtccaa ctctgaaaca gtttaacctg tcatataacc agctgtcttt tgtacctgag 3420 aacctcactg atgtggtaga gaaactggag cagctcattt tagaaggaaa taaaatatca 3480 gggatatgct cccccttgag actgaaggaa ctgaagattt taaaccttag taagaaccac 3540 atttcatccc tatcagagaa ctttcttgag gcttgtccta aagtggagag tttcagtgcc 3600 agaatgaatt ttcttgctgc tatgcctttc ttgcctcctt ctatgacaat cctaaaatta 3660 tctcagaaca aattttcctg tattccagaa gcaattttaa atcttccaca cttgcggtct 3720 ttagatatga gcagcaatga tattcagtac ctaccaggtc ccgcacactg gaaatctttg 3780 aacttaaggg aactcttatt tagccataat cagatcagca tcttggactt gagtgaaaaa 3840 gcatatttat ggtctagagt agagaaactg catctttctc acaataaact gaaagagatt 3900 cctcctgaga ttggctgtct tgaaaatctg acatctctgg atgtcagtta caacttggaa 3960 ctaagatcct ttcccaatga aatggggaaa ttaagcaaaa tatgggatct tcctttggat 4020 gaactgcatc ttaactttga ttttaaacat ataggatgta aagccaaaga catcataagg 4080 tttcttcaac agcgattaaa aaaggctgtg ccttataacc gaatgaaact tatgattgtg 4140 ggaaatactg ggagtggtaa aaccacctta ttgcagcaat taatgaaaac caagaaatca 4200 gatcttggaa tgcaaagtgc cacagttggc atagatgtga aagactggcc tatccaaata 4260 agagacaaaa gaaagagaga tctcgtccta aatgtgtggg attttgcagg tcgtgaggaa 4320 ttctatagta ctcatcccca ttttatgacg cagcgagcat tgtaccttgc tgtctatgac 4380 ctcagcaagg gacaggctga agttgatgcc atgaagcctt ggctcttcaa tataaaggct 4440 cgcgcttctt cttcccctgt gattctcgtt ggcacacatt tggatgtttc tgatgagaag 4500 caacgcaaag cctgcatgag taaaatcacc aaggaactcc tgaataagcg agggttccct 4560 gccatacgag attaccactt tgtgaatgcc accgaggaat ctgatgcttt ggcaaaactt 4620 cggaaaacca tcataaacga gagccttaat ttcaagatcc gagatcagct tgttgttgga 4680 cagctgattc cagactgcta tgtagaactt gaaaaaatca ttttatcgga gcgtaaaaat 4740 gtgccaattg aatttcccgt aattgaccgg aaacgattat tacaactagt gagagaaaat 4800 cagctgcagt tagatgaaaa tgagcttcct cacgcagttc actttctaaa tgaatcagga 4860 gtccttcttc attttcaaga cccagcactg cagttaagtg acttgtactt tgtggaaccc 4920 aagtggcttt gtaaaatcat ggcacagatt ttgacagtga aagtggaagg ttgtccaaaa 4980 caccctaagg gcattatttc gcgtagagat gtggaaaaat ttctttcaaa aaaaaggaaa 5040 tttccaaaga actacatgtc acagtatttt aagctcctag aaaaattcca gattgctttg 5100 ccaataggag aagaatattt gctggttcca agcagtttgt ctgaccacag gcctgtgata 5160 gagcttcccc attgtgagaa ctctgaaatt atcatccgac tatatgaaat gccttatttt 5220 ccaatgggat tttggtcaag attaatcaat cgattacttg agatttcacc ttacatgctt 5280 tcagggagag aacgagcact tcgcccaaac agaatgtatt ggcgacaagg catttactta 5340 aattggtctc ctgaagctta ttgtctggta ggatctgaag tcttagacaa tcatccagag 5400 agtttcttaa aaattacagt tccttcttgt agaaaaggct gtattctttt gggccaagtt 5460 gtggaccaca ttgattctct catggaagaa tggtttcctg ggttgctgga gattgatatt 5520 tgtggtgaag gagaaactct gttgaagaaa tgggcattat atagttttaa tgatggtgaa 5580 gaacatcaaa aaatcttact tgatgacttg atgaagaaag cagaggaagg agatctctta 5640 gtaaatccag atcaaccaag gctcaccatt ccaatatctc agattgcccc tgacttgatt 5700 ttggctgacc tgcctagaaa tattatgttg aataatgatg agttggaatt tgaacaagct 5760 ccagagtttc tcctaggtga tggcagtttt ggatcagttt accgagcagc ctatgaagga 5820 gaagaagtgg ctgtgaagat ttttaataaa catacatcac tcaggctgtt aagacaagag 5880 cttgtggtgc tttgccacct ccaccacccc agtttgatat ctttgctggc agctgggatt 5940 cgtccccgga tgttggtgat ggagttagcc tccaagggtt ccttggatcg cctgcttcag 6000 caggacaaag ccagcctcac tagaacccta cagcacagga ttgcactcca cgtagctgat 6060 ggtttgagat acctccactc agccatgatt atataccgag acctgaaacc ccacaatgtg 6120 ctgcttttca cactgtatcc caatgctgcc atcattgcaa agattgctga ctacggcatt 6180 gctcagtact gctgtagaat ggggataaaa acatcagagg gcacaccagg gtttcgtgca 6240 cctgaagttg ccagaggaaa tgtcatttat aaccaacagg ctgatgttta ttcatttggt 6300 ttactactct atgacatttt gacaactgga ggtagaatag tagagggttt gaagtttcca 6360 aatgagtttg atgaattaga aatacaagga aaattacctg atccagttaa agaatatggt 6420 tgtgccccat ggcctatggt tgagaaatta attaaacagt gtttgaaaga aaatcctcaa 6480 gaaaggccta cttctgccca ggtctttgac attttgaatt cagctgaatt agtctgtctg 6540 acgagacgca ttttattacc taaaaacgta attgttgaat gcatggttgc tacacatcac 6600 aacagcagga atgcaagcat ttggctgggc tgtgggcaca ccgacagagg acagctctca 6660 tttcttgact taaatactga aggatacact tctgaggaag ttgctgatag tagaatattg 6720 tgcttagcct tggtgcatct tcctgttgaa aaggaaagct ggattgtgtc tgggacacag 6780 tctggtactc tcctggtcat caataccgaa gatgggaaaa agagacatac cctagaaaag 6840 atgactgatt ctgtcacttg tttgtattgc aattcctttt ccaagcaaag caaacaaaaa 6900 aattttcttt tggttggaac cgctgatggc aagttagcaa tttttgaaga taagactgtt 6960 aagcttaaag gagctgctcc tttgaagata ctaaatatag gaaatgtcag tactccattg 7020 atgtgtttga gtgaatccac aaattcaacg gaaagaaatg taatgtgggg aggatgtggc 7080 acaaagattt tctccttttc taatgatttc accattcaga aactcattga gacaagaaca 7140 agccaactgt tttcttatgc agctttcagt gattccaaca tcataacagt ggtggtagac 7200 actgctctct atattgctaa gcaaaatagc cctgttgtgg aagtgtggga taagaaaact 7260 gaaaaactct gtggactaat agactgcgtg cactttttaa gggaggtaat ggtaaaagaa 7320 aacaaggaat caaaacacaa aatgtcttat tctgggagag tgaaaaccct ctgccttcag 7380 aagaacactg ctctttggat aggaactgga ggaggccata ttttactcct ggatctttca 7440 actcgtcgac ttatacgtgt aatttacaac ttttgtaatt cggtcagagt catgatgaca 7500 gcacagctag gaagccttaa aaatgtcatg ctggtattgg gctacaaccg gaaaaatact 7560 gaaggtacac aaaagcagaa agagatacaa tcttgcttga ccgtttggga catcaatctt 7620 ccacatgaag tgcaaaattt agaaaaacac attgaagtga gaaaagaatt agctgaaaaa 7680 atgagacgaa catctgttga gtaagagaga aataggaatt gtctttggat aggaaaatta 7740 ttctctcctc ttgtaaatat ttattttaaa aatgttcaca tggaaagggt actcacattt 7800 tttgaaatag ctcgtgtgta tgaaggaatg ttattatttt taatttaaat atatgtaaaa 7860 atacttacca gtaaatgtgt attttaaaga actatttaaa acacaatgtt atatttctta 7920 taaataccag ttactttcgt tcattaatta atgaaaataa atctgtgaag tacctaattt 7980 aagtactcat actaaaattt ataaggccga taattttttg ttttcttgtc tgtaatggag 8040 gtaaacttta ttttaaattc tgtgcttaag acaggactat tgcttgtcga tttttctaga 8100 aatctgcacg gtataatgaa aatattaaga cagtttccca tgtaatgtat tccttcttag 8160 attgcatcga aatgcactat catatatgct tgtaaatatt caaatgaatt tgcactaata 8220 aagtcctttg ttggtatgtg aattctcttt gttgctgttg caaacagtgc atcttacaca 8280 acttcactca attcaaaaga aaactccatt aaaagtacta atgaaaaaac atgacatact 8340 gtcaaagtcc tcatatctag gaaagacaca gaaactctct ttgtcacaga aactctctgt 8400 gtctttccta gacataatag agttgttttt caactctatg tttgaatgtg gataccctga 8460 attttgtata attagtgtaa atacagtgtt cagtccttca agtgatattt ttattttttt 8520 attcatacca ctagctactt gttttctaat ctgcttcatt ctaatgctta tattcatctt 8580 ttccctaaat ttgtgatgct gcagatccta catcattcag atagaaacct tttttttttt 8640 cagaattata gaattccaca gctcctacca agaccatgag gataaatatc taacactttt 8700 cagttgctga aggagaaagg agctttagtt atgatggata aaaatatctg ccaccctagg 8760 cttccaaatt atacttaaat tgtttacata gcttaccaca ataggagtat cagggccaaa 8820 tacctatgta ataatttgag gtcatttctg ctttaggaaa agtactttcg gtaaattctt tggccctgac cagtattcat tatttcagat aattccctgt gataggacaa ctagtacatt 8940 taatattctc agaacttatg gcattttact atgtgaaaac tttaaattta tttatattaa 9000 gggtaatcaa attcttaaag atgaaagatt ttctgtattt taaaggaagc tatgctttaa 9060 cttgttatgt aattaacaaa aaaatcatat ataatagagc tctttgttcc agtgttatct 9120 ctttcattgt tactttgtat ttgcaatttt ttttaccaaa gacaaattaa aaaaatgaat 9180 accatattta aatggaataa taaaggtttt ttaaaaactt taaaaaaaaa aaaa 9234
<210> 2 <211> 2527 <212> PRT <213> Homo sapiens
<220>
<221> PROPEP <222> (1) ... (2527)
<223> Homo sapiens leucine-rich repeat kinase 2 (LRRK2) preprotein (NM_198578)
<400> 2
Met Ala Ser GIy Ser Cys GIn GIy Cys GIu GIu Asp GIu GIu Thr Leu
1 5 10 15
Lys Lys Leu lie VaI Arg Leu Asn Asn VaI GIn GIu GIy Lys GIn lie
20 25 30
GIu Thr Leu VaI GIn lie Leu GIu Asp Leu Leu VaI Phe Thr Tyr Ser
35 40 45
GIu His Ala Ser Lys Leu Phe GIn GIy Lys Asn lie His VaI Pro Leu
50 55 60
Leu He VaI Leu Asp Ser Tyr Met Arg VaI Ala Ser VaI GIn GIn VaI 65 70 75 80
GIy Trp Ser Leu Leu Cys Lys Leu He GIu VaI Cys Pro GIy Thr Met
85 90 95
GIn Ser Leu Met GIy Pro GIn Asp VaI GIy Asn Asp Trp GIu VaI Leu
100 105 HO
GIy VaI His GIn Leu He Leu Lys Met Leu Thr VaI His Asn Ala Ser
115 120 125
VaI Asn Leu Ser VaI He GIy Leu Lys Thr Leu Asp Leu Leu Leu Thr
130 135 140
Ser GIy Lys He Thr Leu Leu He Leu Asp GIu GIu Ser Asp He Phe 145 150 155 160
Met Leu He Phe Asp Ala Met His Ser Phe Pro Ala Asn Asp GIu VaI
165 170 175
GIn Lys Leu GIy Cys Lys Ala Leu His VaI Leu Phe GIu Arg VaI Ser
180 185 190
GIu GIu GIn Leu Thr GIu Phe VaI GIu Asn Lys Asp Tyr Met He Leu
195 200 205
Leu Ser Ala Ser Thr Asn Phe Lys Asp GIu GIu GIu He VaI Leu His
210 215 220
VaI Leu His Cys Leu His Ser Leu Ala He Pro Cys Asn Asn VaI GIu 225 230 235 240
VaI Leu Met Ser GIy Asn VaI Arg Cys Tyr Asn He VaI VaI GIu Ala
245 250 255
Met Lys Ala Phe Pro Met Ser GIu Arg He GIn GIu VaI Ser Cys Cys
260 265 270
Leu Leu His Arg Leu Thr Leu GIy Asn Phe Phe Asn He Leu VaI Leu
275 280 285
Asn GIu VaI His GIu Phe VaI VaI Lys Ala VaI GIn GIn Tyr Pro GIu
290 295 300
Asn Ala Ala Leu GIn He Ser Ala Leu Ser Cys Leu Ala Leu Leu Thr 305 310 315 320
GIu Thr He Phe Leu Asn GIn Asp Leu GIu GIu Lys Asn GIu Asn GIn
325 330 335
GIu Asn Asp Asp GIu GIy GIu GIu Asp Lys Leu Phe Trp Leu GIu Ala
340 345 350
Cys Tyr Lys Ala Leu Thr Trp His Arg Lys Asn Lys His VaI GIn GIu
355 360 365
Ala Ala Cys Trp Ala Leu Asn Asn Leu Leu Met Tyr GIn Asn Ser Leu
370 375 380
His GIu Lys He GIy Asp GIu Asp GIy His Phe Pro Ala His Arg GIu 385 390 395 400
VaI Met Leu Ser Met Leu Met His Ser Ser Ser Lys GIu VaI Phe GIn
405 410 415
Ala Ser Ala Asn Ala Leu Ser Thr Leu Leu GIu GIn Asn VaI Asn Phe
420 425 430
Arg Lys lie Leu Leu Ser Lys GIy lie His Leu Asn VaI Leu GIu Leu
435 440 445
Met GIn Lys His lie His Ser Pro GIu VaI Ala GIu Ser GIy Cys Lys
450 455 460
Met Leu Asn His Leu Phe GIu GIy Ser Asn Thr Ser Leu Asp lie Met 465 470 475 480
Ala Ala VaI VaI Pro Lys He Leu Thr VaI Met Lys Arg His GIu Thr
485 490 495
Ser Leu Pro VaI GIn Leu GIu Ala Leu Arg Ala He Leu His Phe He
500 505 510
VaI Pro GIy Met Pro GIu GIu Ser Arg GIu Asp Thr GIu Phe His His
515 520 525
Lys Leu Asn Met VaI Lys Lys GIn Cys Phe Lys Asn Asp He His Lys
530 535 540
Leu VaI Leu Ala Ala Leu Asn Arg Phe He GIy Asn Pro GIy He GIn 545 550 555 560
Lys Cys GIy Leu Lys VaI He Ser Ser He VaI His Phe Pro Asp Ala
565 570 575
Leu GIu Met Leu Ser Leu GIu GIy Ala Met Asp Ser VaI Leu His Thr
580 585 590
Leu GIn Met Tyr Pro Asp Asp GIn GIu He GIn Cys Leu GIy Leu Ser
595 600 605
Leu He GIy Tyr Leu He Thr Lys Lys Asn VaI Phe He GIy Thr GIy
610 615 620
His Leu Leu Ala Lys He Leu VaI Ser Ser Leu Tyr Arg Phe Lys Asp 625 630 635 640
VaI Ala GIu He GIn Thr Lys GIy Phe GIn Thr He Leu Ala He Leu
645 650 655
Lys Leu Ser Ala Ser Phe Ser Lys Leu Leu VaI His His Ser Phe Asp
660 665 670
Leu VaI He Phe His GIn Met Ser Ser Asn He Met GIu GIn Lys Asp
675 680 685
GIn GIn Phe Leu Asn Leu Cys Cys Lys Cys Phe Ala Lys VaI Ala Met
690 695 700
Asp Asp Tyr Leu Lys Asn VaI Met Leu GIu Arg Ala Cys Asp GIn Asn 705 710 715 720
Asn Ser He Met VaI GIu Cys Leu Leu Leu Leu GIy Ala Asp Ala Asn
725 730 735
GIn Ala Lys GIu GIy Ser Ser Leu He Cys GIn VaI Cys GIu Lys GIu
740 745 750
Ser Ser Pro Lys Leu VaI GIu Leu Leu Leu Asn Ser GIy Ser Arg GIu
755 760 765
GIn Asp VaI Arg Lys Ala Leu Thr He Ser He GIy Lys GIy Asp Ser
770 775 780
GIn He He Ser Leu Leu Leu Arg Arg Leu Ala Leu Asp VaI Ala Asn 785 790 795 800
Asn Ser He Cys Leu GIy GIy Phe Cys He GIy Lys VaI GIu Pro Ser
805 810 815
Trp Leu GIy Pro Leu Phe Pro Asp Lys Thr Ser Asn Leu Arg Lys GIn
820 825 830
Thr Asn He Ala Ser Thr Leu Ala Arg Met VaI He Arg Tyr GIn Met
835 840 845
Lys Ser Ala VaI GIu GIu GIy Thr Ala Ser GIy Ser Asp GIy Asn Phe 850 855 860
Ser GIu Asp VaI Leu Ser Lys Phe Asp GIu Trp Thr Phe lie Pro Asp 865 870 875 880
Ser Ser Met Asp Ser VaI Phe Ala GIn Ser Asp Asp Leu Asp Ser GIu
885 890 895
GIy Ser GIu GIy Ser Phe Leu VaI Lys Lys Lys Ser Asn Ser lie Ser
900 905 910
VaI GIy GIu Phe Tyr Arg Asp Ala VaI Leu GIn Arg Cys Ser Pro Asn
915 920 925
Leu GIn Arg His Ser Asn Ser Leu GIy Pro lie Phe Asp His GIu Asp
930 935 940
Leu Leu Lys Arg Lys Arg Lys lie Leu Ser Ser Asp Asp Ser Leu Arg 945 950 955 960
Ser Ser Lys Leu GIn Ser His Met Arg His Ser Asp Ser lie Ser Ser
965 970 975
Leu Ala Ser GIu Arg GIu Tyr lie Thr Ser Leu Asp Leu Ser Ala Asn
980 985 990
GIu Leu Arg Asp lie Asp Ala Leu Ser GIn Lys Cys Cys lie Ser VaI
995 1000 1005
His Leu GIu His Leu GIu Lys Leu GIu Leu His GIn Asn Ala Leu Thr
1010 1015 1020
Ser Phe Pro GIn GIn Leu Cys GIu Thr Leu Lys Ser Leu Thr His Leu 1025 1030 1035 1040
Asp Leu His Ser Asn Lys Phe Thr Ser Phe Pro Ser Tyr Leu Leu Lys
1045 1050 1055
Met Ser Cys lie Ala Asn Leu Asp VaI Ser Arg Asn Asp lie GIy Pro
1060 1065 1070
Ser VaI VaI Leu Asp Pro Thr VaI Lys Cys Pro Thr Leu Lys GIn Phe
1075 1080 1085
Asn Leu Ser Tyr Asn GIn Leu Ser Phe VaI Pro GIu Asn Leu Thr Asp
1090 1095 1100
VaI VaI GIu Lys Leu GIu GIn Leu lie Leu GIu GIy Asn Lys lie Ser 1105 1110 1115 1120
GIy lie Cys Ser Pro Leu Arg Leu Lys GIu Leu Lys lie Leu Asn Leu
1125 1130 1135
Ser Lys Asn His lie Ser Ser Leu Ser GIu Asn Phe Leu GIu Ala Cys
1140 1145 1150
Pro Lys VaI GIu Ser Phe Ser Ala Arg Met Asn Phe Leu Ala Ala Met
1155 1160 1165
Pro Phe Leu Pro Pro Ser Met Thr lie Leu Lys Leu Ser GIn Asn Lys
1170 1175 1180
Phe Ser Cys lie Pro GIu Ala lie Leu Asn Leu Pro His Leu Arg Ser 1185 1190 1195 1200
Leu Asp Met Ser Ser Asn Asp lie GIn Tyr Leu Pro GIy Pro Ala His
1205 1210 1215
Trp Lys Ser Leu Asn Leu Arg GIu Leu Leu Phe Ser His Asn GIn lie
1220 1225 1230
Ser lie Leu Asp Leu Ser GIu Lys Ala Tyr Leu Trp Ser Arg VaI GIu
1235 1240 1245
Lys Leu His Leu Ser His Asn Lys Leu Lys GIu lie Pro Pro GIu lie
1250 1255 1260
GIy Cys Leu GIu Asn Leu Thr Ser Leu Asp VaI Ser Tyr Asn Leu GIu 1265 1270 1275 1280
Leu Arg Ser Phe Pro Asn GIu Met GIy Lys Leu Ser Lys lie Trp Asp
1285 1290 1295
Leu Pro Leu Asp GIu Leu His Leu Asn Phe Asp Phe Lys His lie GIy
1300 1305 1310
Cys Lys Ala Lys Asp lie lie Arg Phe Leu GIn GIn Arg Leu Lys Lys 1315 1320 1325
Ala VaI Pro Tyr Asn Arg Met Lys Leu Met lie VaI GIy Asn Thr GIy
1330 1335 1340
Ser GIy Lys Thr Thr Leu Leu GIn GIn Leu Met Lys Thr Lys Lys Ser 1345 1350 1355 1360
Asp Leu GIy Met GIn Ser Ala Thr VaI GIy lie Asp VaI Lys Asp Trp
1365 1370 1375
Pro lie GIn lie Arg Asp Lys Arg Lys Arg Asp Leu VaI Leu Asn VaI
1380 1385 1390
Trp Asp Phe Ala GIy Arg GIu GIu Phe Tyr Ser Thr His Pro His Phe
1395 1400 1405
Met Thr GIn Arg Ala Leu Tyr Leu Ala VaI Tyr Asp Leu Ser Lys GIy
1410 1415 1420
GIn Ala GIu VaI Asp Ala Met Lys Pro Trp Leu Phe Asn lie Lys Ala 1425 1430 1435 1440
Arg Ala Ser Ser Ser Pro VaI lie Leu VaI GIy Thr His Leu Asp VaI
1445 1450 1455
Ser Asp GIu Lys GIn Arg Lys Ala Cys Met Ser Lys lie Thr Lys GIu
1460 1465 1470
Leu Leu Asn Lys Arg GIy Phe Pro Ala lie Arg Asp Tyr His Phe VaI
1475 1480 1485
Asn Ala Thr GIu GIu Ser Asp Ala Leu Ala Lys Leu Arg Lys Thr lie
1490 1495 1500 lie Asn GIu Ser Leu Asn Phe Lys lie Arg Asp GIn Leu VaI VaI GIy 1505 1510 1515 1520
GIn Leu lie Pro Asp Cys Tyr VaI GIu Leu GIu Lys lie lie Leu Ser
1525 1530 1535
GIu Arg Lys Asn VaI Pro lie GIu Phe Pro VaI lie Asp Arg Lys Arg
1540 1545 1550
Leu Leu GIn Leu VaI Arg GIu Asn GIn Leu GIn Leu Asp GIu Asn GIu
1555 1560 1565
Leu Pro His Ala VaI His Phe Leu Asn GIu Ser GIy VaI Leu Leu His
1570 1575 1580
Phe GIn Asp Pro Ala Leu GIn Leu Ser Asp Leu Tyr Phe VaI GIu Pro 1585 1590 1595 1600
Lys Trp Leu Cys Lys lie Met Ala GIn lie Leu Thr VaI Lys VaI GIu
1605 1610 1615
GIy Cys Pro Lys His Pro Lys GIy lie lie Ser Arg Arg Asp VaI GIu
1620 1625 1630
Lys Phe Leu Ser Lys Lys Arg Lys Phe Pro Lys Asn Tyr Met Ser GIn
1635 1640 1645
Tyr Phe Lys Leu Leu GIu Lys Phe GIn lie Ala Leu Pro lie GIy GIu
1650 1655 1660
GIu Tyr Leu Leu VaI Pro Ser Ser Leu Ser Asp His Arg Pro VaI lie 1665 1670 1675 1680
GIu Leu Pro His Cys GIu Asn Ser GIu lie lie lie Arg Leu Tyr GIu
1685 1690 1695
Met Pro Tyr Phe Pro Met GIy Phe Trp Ser Arg Leu lie Asn Arg Leu
1700 1705 1710
Leu GIu lie Ser Pro Tyr Met Leu Ser GIy Arg GIu Arg Ala Leu Arg
1715 1720 1725
Pro Asn Arg Met Tyr Trp Arg GIn GIy lie Tyr Leu Asn Trp Ser Pro
1730 1735 1740
GIu Ala Tyr Cys Leu VaI GIy Ser GIu VaI Leu Asp Asn His Pro GIu 1745 1750 1755 1760
Ser Phe Leu Lys lie Thr VaI Pro Ser Cys Arg Lys GIy Cys lie Leu
1765 1770 1775
Leu GIy GIn VaI VaI Asp His lie Asp Ser Leu Met GIu GIu Trp Phe 1780 1785 1790
Pro GIy Leu Leu GIu lie Asp lie Cys GIy GIu GIy GIu Thr Leu Leu
1795 1800 1805
Lys Lys Trp Ala Leu Tyr Ser Phe Asn Asp GIy GIu GIu His GIn Lys
1810 1815 1820 lie Leu Leu Asp Asp Leu Met Lys Lys Ala GIu GIu GIy Asp Leu Leu 1825 1830 1835 1840
VaI Asn Pro Asp GIn Pro Arg Leu Thr lie Pro lie Ser GIn lie Ala
1845 1850 1855
Pro Asp Leu lie Leu Ala Asp Leu Pro Arg Asn lie Met Leu Asn Asn
1860 1865 1870
Asp GIu Leu GIu Phe GIu GIn Ala Pro GIu Phe Leu Leu GIy Asp GIy
1875 1880 1885
Ser Phe GIy Ser VaI Tyr Arg Ala Ala Tyr GIu GIy GIu GIu VaI Ala
1890 1895 1900
VaI Lys lie Phe Asn Lys His Thr Ser Leu Arg Leu Leu Arg GIn GIu 1905 1910 1915 1920
Leu VaI VaI Leu Cys His Leu His His Pro Ser Leu lie Ser Leu Leu
1925 1930 1935
Ala Ala GIy lie Arg Pro Arg Met Leu VaI Met GIu Leu Ala Ser Lys
1940 1945 1950
GIy Ser Leu Asp Arg Leu Leu GIn GIn Asp Lys Ala Ser Leu Thr Arg
1955 1960 1965
Thr Leu GIn His Arg lie Ala Leu His VaI Ala Asp GIy Leu Arg Tyr
1970 1975 1980
Leu His Ser Ala Met lie lie Tyr Arg Asp Leu Lys Pro His Asn VaI 1985 1990 1995 2000
Leu Leu Phe Thr Leu Tyr Pro Asn Ala Ala lie lie Ala Lys lie Ala
2005 2010 2015
Asp Tyr GIy lie Ala GIn Tyr Cys Cys Arg Met GIy lie Lys Thr Ser
2020 2025 2030
GIu GIy Thr Pro GIy Phe Arg Ala Pro GIu VaI Ala Arg GIy Asn VaI
2035 2040 2045 lie Tyr Asn GIn GIn Ala Asp VaI Tyr Ser Phe GIy Leu Leu Leu Tyr
2050 2055 2060
Asp lie Leu Thr Thr GIy GIy Arg lie VaI GIu GIy Leu Lys Phe Pro 2065 2070 2075 2080
Asn GIu Phe Asp GIu Leu GIu lie GIn GIy Lys Leu Pro Asp Pro VaI
2085 2090 2095
Lys GIu Tyr GIy Cys Ala Pro Trp Pro Met VaI GIu Lys Leu lie Lys
2100 2105 2110
GIn Cys Leu Lys GIu Asn Pro GIn GIu Arg Pro Thr Ser Ala GIn VaI
2115 2120 2125
Phe Asp lie Leu Asn Ser Ala GIu Leu VaI Cys Leu Thr Arg Arg lie
2130 2135 2140
Leu Leu Pro Lys Asn VaI lie VaI GIu Cys Met VaI Ala Thr His His 2145 2150 2155 2160
Asn Ser Arg Asn Ala Ser lie Trp Leu GIy Cys GIy His Thr Asp Arg
2165 2170 2175
GIy GIn Leu Ser Phe Leu Asp Leu Asn Thr GIu GIy Tyr Thr Ser GIu
2180 2185 2190
GIu VaI Ala Asp Ser Arg lie Leu Cys Leu Ala Leu VaI His Leu Pro
2195 2200 2205
VaI GIu Lys GIu Ser Trp lie VaI Ser GIy Thr GIn Ser GIy Thr Leu
2210 2215 2220
Leu VaI lie Asn Thr GIu Asp GIy Lys Lys Arg His Thr Leu GIu Lys 2225 2230 2235 2240
Met Thr Asp Ser VaI Thr Cys Leu Tyr Cys Asn Ser Phe Ser Lys GIn 2245 2250 2255
Ser Lys GIn Lys Asn Phe Leu Leu VaI GIy Thr Ala Asp GIy Lys Leu
2260 2265 2270
Ala lie Phe GIu Asp Lys Thr VaI Lys Leu Lys GIy Ala Ala Pro Leu
2275 2280 2285
Lys lie Leu Asn lie GIy Asn VaI Ser Thr Pro Leu Met Cys Leu Ser
2290 2295 2300
GIu Ser Thr Asn Ser Thr GIu Arg Asn VaI Met Trp GIy GIy Cys GIy 2305 2310 2315 2320
Thr Lys lie Phe Ser Phe Ser Asn Asp Phe Thr lie GIn Lys Leu lie
2325 2330 2335
GIu Thr Arg Thr Ser GIn Leu Phe Ser Tyr Ala Ala Phe Ser Asp Ser
2340 2345 2350
Asn He He Thr VaI VaI VaI Asp Thr Ala Leu Tyr He Ala Lys GIn
2355 2360 2365
Asn Ser Pro VaI VaI GIu VaI Trp Asp Lys Lys Thr GIu Lys Leu Cys
2370 2375 2380
GIy Leu He Asp Cys VaI His Phe Leu Arg GIu VaI Met VaI Lys GIu 2385 2390 2395 2400
Asn Lys GIu Ser Lys His Lys Met Ser Tyr Ser GIy Arg VaI Lys Thr
2405 2410 2415
Leu Cys Leu GIn Lys Asn Thr Ala Leu Trp He GIy Thr GIy GIy GIy
2420 2425 2430
His He Leu Leu Leu Asp Leu Ser Thr Arg Arg Leu He Arg VaI He
2435 2440 2445
Tyr Asn Phe Cys Asn Ser VaI Arg VaI Met Met Thr Ala GIn Leu GIy
2450 2455 2460
Ser Leu Lys Asn VaI Met Leu VaI Leu GIy Tyr Asn Arg Lys Asn Thr 2465 2470 2475 2480
GIu GIy Thr GIn Lys GIn Lys GIu He GIn Ser Cys Leu Thr VaI Trp
2485 2490 2495
Asp He Asn Leu Pro His GIu VaI GIn Asn Leu GIu Lys His He GIu
2500 2505 2510
VaI Arg Lys GIu Leu Ala GIu Lys Met Arg Arg Thr Ser VaI GIu 2515 2520 2525

Claims

CLAIMS We claim:
1. Use of a LRRK2 modulating agent in the manufacture of a medicament for the treatment of Alzheimer's disease a selected patient population, wherein the patient population is selected on the basis of polymorphisms in the leucine-rich repeat kinase 2 (LRRK2) gene that are indicative of progression from mild cognitive impairment (MCI) to Alzheimer's disease.
2. The use of claim 1 , wherein the LRRK2 modulating agent is a heterocyclic compound.
3. The use of claim 1, wherein the treatment of Alzheimer's disease slows the progression by the patient from mild cognitive impairment (MCI) to Alzheimer's disease.
4. The use of claim 1, wherein the treatment of Alzheimer's disease slows the progression by the patient from moderate Alzheimer's disease to severe Alzheimer's disease.
5. The use of claim 1, wherein the polymorphism in the LRRK2 gene is selected from the group consisting T1602S and T2352.
6. The use of claim 5, wherein theT1602S locus of the patient has a TT (Thr/Thr) genotype.
7. The use of claim 5, wherein T2352 locus of the patient has a CC (Thr/Thr) genotype.
8. A method for predicting the progression of Alzheimer's disease in a subject, comprising the steps of:
(a) obtaining a tissue sample from a subject;
(b) assaying the sample for the presence of a genetic polymorphism indicative of progression of the subject from mild cognitive impairment (MCI) to Alzheimer's disease; wherein the presence of a genetic polymorphism indicative of progression of the subject from mild cognitive impairment to Alzheimer's disease in the subject predicts that the subject is at increased risk for progression from mild cognitive impairment to Alzheimer's disease.
9. The method of claim 8, wherein the tissue sample is a blood sample.
10. The method of claim 8, wherein the genetic polymorphism is selected from the group consisting T1602S and T2352.
11. The method of claim 8, further comprising the step of.
(c) if the subject is predicted to have a genetic polymorphism indicative of progression of the subject from mild cognitive impairment to Alzheimer's disease, then administering to the subject a LRRK2 modulating agent to slow the progression from mild cognitive impairment to Alzheimer's disease or from moderate Alzheimer's disease to severe Alzheimer's disease.
PCT/US2007/071421 2006-06-20 2007-06-18 Biomarkers for the progression of alzheimer's disease WO2007149798A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US12/305,053 US20100035251A1 (en) 2006-06-20 2007-06-18 BioMarkers for the Progression of Alzheimer's Disease
EP07798679A EP2035582A2 (en) 2006-06-20 2007-06-18 Biomarkers for the progression of alzheimer's disease
BRPI0713738-9A2A BRPI0713738A2 (en) 2006-06-20 2007-06-18 BIOMARKERS FOR ALZHEIMER DISEASE PROGRESSION
JP2009516649A JP2009541336A (en) 2006-06-20 2007-06-18 Biomarkers for the progression of Alzheimer's disease
AU2007261095A AU2007261095A1 (en) 2006-06-20 2007-06-18 Biomarkers for the progression of Alzheimer's disease
MX2008016524A MX2008016524A (en) 2006-06-20 2007-06-18 Biomarkers for the progression of alzheimer's disease.
CA002657980A CA2657980A1 (en) 2006-06-20 2007-06-18 Biomarkers for the progression of alzheimer's disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US81561006P 2006-06-20 2006-06-20
US60/815,610 2006-06-20

Publications (2)

Publication Number Publication Date
WO2007149798A2 true WO2007149798A2 (en) 2007-12-27
WO2007149798A3 WO2007149798A3 (en) 2008-07-24

Family

ID=38834281

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/071421 WO2007149798A2 (en) 2006-06-20 2007-06-18 Biomarkers for the progression of alzheimer's disease

Country Status (11)

Country Link
US (1) US20100035251A1 (en)
EP (1) EP2035582A2 (en)
JP (1) JP2009541336A (en)
KR (1) KR20090019848A (en)
CN (1) CN101473044A (en)
AU (1) AU2007261095A1 (en)
BR (1) BRPI0713738A2 (en)
CA (1) CA2657980A1 (en)
MX (1) MX2008016524A (en)
RU (1) RU2009101384A (en)
WO (1) WO2007149798A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102009042160B3 (en) * 2009-09-11 2011-04-14 Eberhard-Karls-Universität Tübingen Universitätsklinikum Method for diagnosing and / or predicting the development of neurodegenerative diseases
WO2012028629A1 (en) 2010-09-02 2012-03-08 Glaxo Group Limited 2 - (benzyloxy) benzamides as lrrk2 kinase inhibitors
US9156845B2 (en) 2012-06-29 2015-10-13 Pfizer Inc. 4-(substituted amino)-7H-pyrrolo[2,3-d] pyrimidines as LRRK2 inhibitors
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
US9932325B2 (en) 2016-06-16 2018-04-03 Denali Therapeutics Inc. Compounds, compositions, and methods
RU2661022C2 (en) * 2012-07-06 2018-07-11 Рандокс Лабораторис Лтд Tropomyosin isoforms related to alzheimer's disease and moderate cognitive impairments
US10039753B2 (en) 2015-09-14 2018-08-07 Pfizer Inc. Imidazo[4,5-c]quinoline and imidazo[4,5-c][1,5]naphthyridine derivatives as LRRK2 inhibitors
WO2019012093A1 (en) 2017-07-14 2019-01-17 Glaxosmithkline Intellectual Property Development Limited Inhibitors of leucine rich repeat kinase 2
WO2020092136A1 (en) 2018-10-31 2020-05-07 Merck Sharp & Dohme Corp. N-heteroaryl indazole derivatives as lrrk2 inhibitors, pharmaceutical compositions, and uses thereof
WO2020247298A2 (en) 2019-06-06 2020-12-10 Merck Sharp & Dohme Corp. 1-pyrazolyl, 5-, 6- disubstituted indazole derivatives as lrrk2 inhibitors, pharmaceutical compositions, and uses thereof
US11028080B2 (en) 2016-03-11 2021-06-08 Denali Therapeutics Inc. Substituted pyrimidines as LRKK2 inhibitors
US11214565B2 (en) 2015-11-20 2022-01-04 Denali Therapeutics Inc. Compound, compositions, and methods
US12030872B2 (en) 2019-10-25 2024-07-09 Merck Sharp & Dohme Llc N-heteroaryl indazole derivatives as LRRK2 inhibitors, pharmaceutical compositions, and uses thereof

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012075046A1 (en) 2010-11-30 2012-06-07 Genentech, Inc. Assays and biomarkers for lrrk2
WO2012135631A1 (en) * 2011-03-30 2012-10-04 Arrien Pharmaeuticals Llc Substituted 5-(pyrazin-2-yl)-1h-pyrazolo [3, 4-b] pyridine and pyrazolo [3, 4-b] pyridine derivatives as protein kinase inhibitors
CN105331721A (en) * 2015-11-27 2016-02-17 首都医科大学宣武医院 Method, primers and kit for detecting PD (Parkinsons Disease) pathogenic gene mutation
CN114736959B (en) * 2022-05-09 2023-09-22 上海市精神卫生中心(上海市心理咨询培训中心) Mild cognitive impairment diagnostic markers, kit and application thereof
CN117230184B (en) * 2023-11-13 2024-03-19 深圳康美生物科技股份有限公司 Nucleic acid combination for detecting Alzheimer disease gene based on time-of-flight nucleic acid mass spectrometry technology and application

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006045392A2 (en) * 2004-10-21 2006-05-04 GSF-Forschungszentrum für Umwelt und Gesundheit GmbH Kaspp (lrrke) gene, its production and use for the detection and treatment of neurodegenerative disorders

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006045392A2 (en) * 2004-10-21 2006-05-04 GSF-Forschungszentrum für Umwelt und Gesundheit GmbH Kaspp (lrrke) gene, its production and use for the detection and treatment of neurodegenerative disorders

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
GIASSON BENOIT I ET AL: "Biochemical and pathological characterization of Lrrk2" ANNALS OF NEUROLOGY, vol. 59, no. 2, February 2006 (2006-02), pages 315-322, XP002482037 ISSN: 0364-5134 *
HERNANDEZ ET AL: "The dardarin G2019S mutation is a common cause of Parkinson's disease but not other neurodegenerative diseases" NEUROSCIENCE LETTERS, LIMERICK, IE, vol. 389, no. 3, 9 December 2005 (2005-12-09), pages 137-139, XP005072146 ISSN: 0304-3940 *
PARDO LUBA M ET AL: "In search of genes involved in neurodegenerative disorders" MUTATION RESEARCH, vol. 592, no. 1-2, December 2005 (2005-12), pages 89-101, XP005194014 ISSN: 0027-5107 *
ROSS OWEN A ET AL: "Lrrk2 and Lewy body disease" ANNALS OF NEUROLOGY, vol. 59, no. 2, February 2006 (2006-02), pages 388-393, XP002482038 ISSN: 0364-5134 *
TOFT ET AL: "LRRK2 mutations are not common in Alzheimer's disease" MECHANISMS OF AGEING AND DEVELOPMENT, ELSEVIER SEQUOIA, LAUSANNE, CH, vol. 126, no. 11, 1 November 2005 (2005-11-01), pages 1201-1205, XP005091276 ISSN: 0047-6374 *
ZIMPRICH A ET AL: "Mutations in LRRK2 cause autosomal-dominant Parkinsonism with pleomorphic pathology" NEURON, CELL PRESS, vol. 44, no. 4, 18 November 2004 (2004-11-18), pages 601-607, XP002371964 ISSN: 0896-6273 *

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102009042160B3 (en) * 2009-09-11 2011-04-14 Eberhard-Karls-Universität Tübingen Universitätsklinikum Method for diagnosing and / or predicting the development of neurodegenerative diseases
WO2012028629A1 (en) 2010-09-02 2012-03-08 Glaxo Group Limited 2 - (benzyloxy) benzamides as lrrk2 kinase inhibitors
US9156845B2 (en) 2012-06-29 2015-10-13 Pfizer Inc. 4-(substituted amino)-7H-pyrrolo[2,3-d] pyrimidines as LRRK2 inhibitors
US9642855B2 (en) 2012-06-29 2017-05-09 Pfizer Inc. Substituted pyrrolo[2,3-d]pyrimidines as LRRK2 inhibitors
RU2661022C2 (en) * 2012-07-06 2018-07-11 Рандокс Лабораторис Лтд Tropomyosin isoforms related to alzheimer's disease and moderate cognitive impairments
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
US10039753B2 (en) 2015-09-14 2018-08-07 Pfizer Inc. Imidazo[4,5-c]quinoline and imidazo[4,5-c][1,5]naphthyridine derivatives as LRRK2 inhibitors
US11214565B2 (en) 2015-11-20 2022-01-04 Denali Therapeutics Inc. Compound, compositions, and methods
US11028080B2 (en) 2016-03-11 2021-06-08 Denali Therapeutics Inc. Substituted pyrimidines as LRKK2 inhibitors
US11840529B2 (en) 2016-03-11 2023-12-12 Denali Therapeutics Inc. Substituted pyrimidines as LRKK2 inhibitors
US10590114B2 (en) 2016-06-16 2020-03-17 Denali Therapautics Inc. Compounds, compositions, and methods
US11111235B2 (en) 2016-06-16 2021-09-07 Denali Therapeutics Inc. Compounds, compositions, and methods
US9932325B2 (en) 2016-06-16 2018-04-03 Denali Therapeutics Inc. Compounds, compositions, and methods
US11591316B2 (en) 2016-06-16 2023-02-28 Denali Therapeutics Inc. Compounds, compositions, and methods
US11834439B2 (en) 2016-06-16 2023-12-05 Denali Therapeutics Inc. Compounds, compositions, and methods
WO2019012093A1 (en) 2017-07-14 2019-01-17 Glaxosmithkline Intellectual Property Development Limited Inhibitors of leucine rich repeat kinase 2
WO2020092136A1 (en) 2018-10-31 2020-05-07 Merck Sharp & Dohme Corp. N-heteroaryl indazole derivatives as lrrk2 inhibitors, pharmaceutical compositions, and uses thereof
WO2020247298A2 (en) 2019-06-06 2020-12-10 Merck Sharp & Dohme Corp. 1-pyrazolyl, 5-, 6- disubstituted indazole derivatives as lrrk2 inhibitors, pharmaceutical compositions, and uses thereof
US12030872B2 (en) 2019-10-25 2024-07-09 Merck Sharp & Dohme Llc N-heteroaryl indazole derivatives as LRRK2 inhibitors, pharmaceutical compositions, and uses thereof

Also Published As

Publication number Publication date
AU2007261095A1 (en) 2007-12-27
US20100035251A1 (en) 2010-02-11
KR20090019848A (en) 2009-02-25
CN101473044A (en) 2009-07-01
RU2009101384A (en) 2010-07-27
CA2657980A1 (en) 2007-12-27
EP2035582A2 (en) 2009-03-18
JP2009541336A (en) 2009-11-26
BRPI0713738A2 (en) 2014-06-24
MX2008016524A (en) 2009-03-09
WO2007149798A3 (en) 2008-07-24

Similar Documents

Publication Publication Date Title
US20100035251A1 (en) BioMarkers for the Progression of Alzheimer&#39;s Disease
WO2006104812A2 (en) Biomarkers for pharmacogenetic diagnosis of type 2 diabetes
US20100249107A1 (en) Biomarkers for Alzheimer&#39;s Disease Progression
US20090118350A1 (en) Biomarkers for Identifying Efficacy of Tegaserod in Patients with Chronic Constipation
WO2006060429A2 (en) Identification of variants in histone deacetylase 1 (hdac1) to predict drug response
US20070065821A1 (en) Methods for the prediction of suicidality during treatment
CN104937113B (en) Method for predicting the onset of extrapyramidal symptoms (EPS) induced by antipsychotic-based therapy
WO2006110478A2 (en) Mutations and polymorphisms of epidermal growth factor receptor
AU2006227283B2 (en) Biomarkers for efficacy of aliskiren as a hypertensive agent
US20090281090A1 (en) Biomarkers for the prediction of responsiveness to clozapine treatment
RU2408363C2 (en) Biomarkers for estimating efficacy of aliskiren as hypertensive agent
WO2007109183A2 (en) Mutations and polymorphisms of fms-related tyrosine kinase 1
WO2007002217A2 (en) Mutations and polymorphisms of bcl-2
WO2007121017A2 (en) Mutations and polymorphisms of fms-like tyrosine kinase 4
CN101146915A (en) Biomarkers for efficacy of aliskiren as a hypertensive agent

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780022902.3

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 9860/DELNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2007261095

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2007798679

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2657980

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 12305053

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2009516649

Country of ref document: JP

Ref document number: MX/A/2008/016524

Country of ref document: MX

Ref document number: 1020087030933

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2007261095

Country of ref document: AU

Date of ref document: 20070618

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2009101384

Country of ref document: RU

Kind code of ref document: A