AU2016204271B2 - Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration - Google Patents

Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration Download PDF

Info

Publication number
AU2016204271B2
AU2016204271B2 AU2016204271A AU2016204271A AU2016204271B2 AU 2016204271 B2 AU2016204271 B2 AU 2016204271B2 AU 2016204271 A AU2016204271 A AU 2016204271A AU 2016204271 A AU2016204271 A AU 2016204271A AU 2016204271 B2 AU2016204271 B2 AU 2016204271B2
Authority
AU
Australia
Prior art keywords
mrna
cfhr3
cfhr1
factor
seq
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
AU2016204271A
Other versions
AU2016204271A1 (en
Inventor
Gregory S. Hageman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Iowa Research Foundation UIRF
Original Assignee
University of Iowa Research Foundation UIRF
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2007272335A external-priority patent/AU2007272335B2/en
Priority claimed from AU2013202401A external-priority patent/AU2013202401A1/en
Application filed by University of Iowa Research Foundation UIRF filed Critical University of Iowa Research Foundation UIRF
Priority to AU2016204271A priority Critical patent/AU2016204271B2/en
Publication of AU2016204271A1 publication Critical patent/AU2016204271A1/en
Application granted granted Critical
Publication of AU2016204271B2 publication Critical patent/AU2016204271B2/en
Priority to AU2018206776A priority patent/AU2018206776A1/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

Disclosed are screening methods for determining a human subject's propensity to develop a vascular disorder and/or age-related macular degeneration (AMD), therapeutic or prophylactic compounds for treating disease or inhibiting its development, and methods of treating patients to alleviate symptoms of the disease, prevent or delay its onset, or inhibit its progression. The inventions are based on the discovery that persons with a genome having a deletion of the CFHR-1 and/or CFHR-3 gene, which normally lie on human chromosome 1 between DNA encoding CFH and CFHR-4, are at reduced risk of developing AMD, and elevated risk of developing vascular disease such as aneurysm.

Description

The inventions are based on the discovery that persons with a genome having a deletion of the CFHR-1 and/or CFHR-3 gene, which normally lie on human chromosome 1 between DNA encoding CFH and CFHR-4, are at reduced risk of developing AMD, and elevated risk of developing vascular disease such as aneurysm.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
METHODS AND REAGENTS FOR TREATMENT AND DIAGNOSIS OF VASCULAR DISORDERS AND AGE-RELATED MACULAR DEGENERATION
CROSS-REFERENCES TO RELATED APPLICATIONS [0001] This application claims priority to U.S. Provisional Patent Application No. 60/840,073, filed August 23, 2006. This application also claims priority to U.S. Provisional Patent Application No. 60/831,018, filed July 13, 2006. Both these applications are hereby incorporated by reference.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT [0002] This invention was made with government support under NIH R01 EY11515 and R24
ΕΥ017404, awarded by the National Institutes of Health. The government has certain rights in the invention.
FIELD OF THE INVENTION [0003] This invention relates to screening and therapeutic methods for complement-mediated diseases such as age-related macular degeneration and vascular diseases. The invention finds application in the fields of biology and medicine.
BACKGROUND OF THE INVENTION [0004] Complement Factor H (CFH) is a multifunctional protein that acts as a key regulator of the complement system. See Zipfel, 2001, Factor H and disease: a complement regulator affects vital body functions Semin Thromb Hemost. 27:191-9. The Factor H protein activities include: (1) binding to C-reactive protein (CRP), (2) binding to C3b, (3) binding to heparin, (4) binding to sialic acid; (5) binding to endothelial cell surfaces, (6) binding to cellular integrin receptors (7) binding to pathogens, including microbes (see Figure 3 of U.S. patent publication No. 20070020647), and (8) C3b co-factor activity. The Factor H gene, known as HF1, CFH and HF, is located on human chromosome 1, at position lq32. The lq32 locus contains a number of complement pathway-associated genes. One group of these genes, referred to as the regulators of complement activation (RCA) gene cluster, contains the genes that encode Factor H, five Factor H-related proteins (FHR-1, FHR-2, FHR-3, FHR-4 and FHR-5 or CFHR1, CFHR2,
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
CFHR3, CFHR4 and CFHR5, respectively), and the gene encoding the beta subunit of coagulation factor XIII. The Factor H and Factor H related proteins are composed almost entirely of short consensus repeats (SCRs). Factor H and FHL1 are composed of SCRs 1-20 and 1-7, respectively. FHR-1, FHR-2, FHR-3, FHR-4 and FHR-5 are composed of 5, 4, 5, 5 and 8 SCRs, respectively. The order of genes, from centromere to telomere is FH/FHL1, FHR-3, FHR-1, FHR-4, FHR-2 and FHR-5.
Factor H Gene [0005] The Factor H cDNA encodes a polypeptide 1231 amino acids in length having an apparent molecular weight of 155 kDa (see Ripoche et al., 1988, Biochem J 249:593-602). There is an alternatively spliced form of Factor H known as FHL-1 (and also has been referred to as HFL1 or CFHT). FHL-1 corresponds essentially to exons 1 through 9 of Factor H (see Ripoche et al., 1988, Biochem J 249:593-602). The FHL1 cDNA encodes a polypeptide 449 amino acids in length having an apparent molecular weight of 45-50 kDa. The first 445 amino acids of FH1 and FHL1 are identical, with FHL1 having four unique C-terminal amino acids (encoded by alternative exon 10A, which is located in the intron between exon 9 and exon 10. cDNA and amino acid sequence data for human Factor H and FHL1 are found in the EMBL/GenBank Data Libraries under accession numbers Y00716 and X07523, respectively. The 3926 base nucleotide sequence of the reference form of human Factor H cDNA has GenBank accession number Y00716 and the polypeptide has GenBank accession number Y00716. The 1658 base nucleotide sequence of the reference form of HFL1, the truncated form of the human Factor H, has GenBank accession number X07523, and the polypeptide sequence has GenBank accession number X07523. The Factor H gene sequence (150626 bases in length) has GenBank accession number AL049744. The Factor H promoter is located 5' to the coding region of the Factor H gene.
FHR-1 Gene [0006] The FHR-1 gene is also known as CFHR1, CFHL1, CFHL, FHR1 and HFL1. The FHR-1 cDNA encodes a polypeptide 330 amino acids in length having an predicted molecular weight of 39 kDa (see Estaller et al., 1991,7. Immunol. 146:3190-3196). cDNA and amino acid sequence data for human FHR-1 are found in the EMBL/GenBank Data Libraries under accession number M65292. The FHR-1 gene sequence is found under GenBank accession number AL049741.
FHR-2 Gene [0007] The FHR-2 gene is also known as CFHR2, CFHL2, FHR2 and HFL3. The FHR-2 cDNA encodes a polypeptide 270 amino acids in length having a predicted molecular weight of 31 kDa (see Strausberg et al., Proc. Natl. Acad. Sci USA 99:16899-16903). cDNA and amino acid sequence data for
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun2016 human FHR-2 are found in the EMBL/GenBank Data Libraries under accession number BC022283. The FHR-2 gene sequence is found under GenBank accession number AL139418.
FHR-3 Gene [0008] The FHR-3 gene is also known as CFHR3, CFHL3, FHR3 and HLF4. The FHR-3 cDNA encodes a polypeptide 330 amino acids in length having a predicted molecular weight of 38 kDa (see Strausberg et al., Proc. Natl. Acad. Sci USA 99:16899-16903). cDNA and amino acid sequence data for human FHR-3 are found in the EMBL/GenBank Data Libraries under accession number BC058009. The FHR-3 gene sequence is found under GenBank accession number AL049741.
FHR-4 Gene [0009] The FHR-4 gene is also known as CFHR4, CFHL4 and FHR4. The FHR-4 cDNA encodes a polypeptide 331 amino acids in length having a predicted molecular weight of 38 kDa (see Skerka et al., 1991, J. Biol. Chem. 272:5627-5634). cDNA and amino acid sequence data for human FHR-4 are found in the EMBL/GenBank Data Libraries under accession number X98337. The FHR-4 gene sequence is found under GenBank accession numbers AF190816 (5' end), AL139418 (3' end) and BX248415.
FHR-5 Gene [0010] The FHR-5 gene is also known as CFHR5, CFHL5 and FHR5. The CFHR5 cDNA encodes a polypeptide 569 amino acids in length having an apparent molecular weight of 65 kDa (see McRae et al., 2001, J. Biol.Chem. 276:6747-6754). cDNA and amino acid sequence data for human CFHR5 are found in the EMBL/GenBank Data Libraries under accession number AF295327. The 2821 base nucleotide sequence of the reference form of human CFHR5 has GenBank accession number AF295327, and the polypeptide sequence has GenBank accession number AAK15619. The CFHR5 genomic sequence is found under GenBank accession numbers AL139418 (5' end) and AL353809 (3' end). The FHR-5 promoter is located 5' to the coding region of the CFHR5 gene.
BRIEF SUMMARY OF THE INVENTION [0011] In one aspect, the invention provides a screening method for determining a human subject's propensity to develop a vascular disorder and/or age-related macular degeneration (AMD), involving analysis of a biological sample from the subject to detect the presence or absence of a deletion in chromosome 1 between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H-related 4 (CFHR4) gene, wherein the presence of a deletion is evidence
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 that the subject is at an increased risk of developing a vascular disorder and a decreased risk of developing AMD.
[0012] Examples of vascular disorders include aneurysms, such as abdominal aortic aneurysm (AAA) and brain intracranial aneurysm.
[0013] In one embodiment, the method comprises detecting the presence or absence of at least a portion of the complement Factor H-related 3 (CFHR3) gene. In a related embodiment the entire protein coding region of the CFHR3 gene is deleted. In a related embodiment the entire CFHR3 gene is deleted. In a related embodiment the entire CFHR3 gene and the region between the CFHR3 gene and complement Factor H-related 1 (CFHRl) gene are deleted.
[0014] In one embodiment, the method comprises detecting the presence or absence of at least a portion of the complement Factor H-related 1 (CFHRl) gene. In a related embodiment the entire protein coding region of the CFHRl gene is deleted. In a related embodiment the entire CFHR1 gene is deleted. In a related embodiment the entire CFHRl gene and the region between the CFHRl gene and complement factor H-related 4 (CFHR4) gene are deleted. In a related embodiment the entire CFHRl gene and the region between the CFHRl gene and CFHR3 gene are deleted.
[0015] In one embodiment, the method comprises detecting the presence or absence of at least a portion of the CFHR3 gene and at least a portion of the CFHRl gene. In a related embodiment both the entire protein coding regions of the CFHR3 and CFHRl genes are deleted. In a related embodiment the entire CFHR3 and CFHRl genes are deleted.
[0016] In one embodiment, a deletion or a partial deletion of an intergenic sequence selected from: a) a sequence between the CFH gene and the CFHR3 gene; b) a sequence between the CFHR3 gene and the CFHRl gene; c) a sequence between the CFHRl gene and the CFHR4 gene. In yet another embodiment, at least a portion of the CFH gene is deleted (e.g., at least a portion of exon 22 is deleted). [0017] In one embodiment, the presence or absence of the deletion is detected by assaying for a gene product encoded in chromosome 1 between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H-related 4 (CFHR4) gene, where the absence of the gene product, or a reduced level of expression of the gene product, indicates the presence of deletion. In another embodiment, the presence or absence of a CFHRl gene product and/or a CFHR3 gene product is detected, where the absence of a gene product is indicative of a deletion. In one instance, the gene product is a protein. In another embodiment, detecting the presence or absence of a deletion is performed by analyzing a chromosome or nucleic acid (e.g., DNA or RNA) from the subject.
[0018] In one embodiment the presence or absence of the deletion is detected by assaying for a truncated CFHRl or CRHR3 gene product, where detection of a truncated gene product is indicative of a
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 deletion. In a preferred embodiment, the CFHR1 gene is partially deleted and expresses a truncated polypeptide gene product.
[0019] In one embodiment the subject has a genotype of T at position 1277 of the coding region of the CFH gene of the chromosome comprising the deletion.
[0020] The subject may be homozygous or heterozygous for deletions. Thus, in one embodiment, deletions are present in both chromosomes 1 of the subject.
[0021] The presence or absence of the deletion may be detected in a biological sample from a patient by, for example, analyzing a chromosome or nucleic acid (e.g., DNA or RNA) sample from the subject. The presence or absence of the deletion also may be detected by, for example, determining the presence or absence of protein encoded by the (deleted) DNA in a biological sample from the subject, e.g., a body fluid or tissue sample of the subject, by detecting a variant or truncated form of the CFHR1 or CFHR3 polypeptides in a body fluid or tissue sample of the subject, or by measuring the level of CFHR1 or CFHR3 polypeptides in a body fluid or tissue sample of the subject.
[0022] The biological sample is any sample taken from a patient that is suitable for use in the invention. Examples of biological samples that include body fluids include blood, serum, urine, cerebral spinal fluid (CSF) and saliva. In one embodiment, the body fluid is blood, serum or urine. Examples of biological samples that comprise tissue samples include a skin biopsy and a cheek scraping. In one embodiment, the tissue sample is a skin biopsy.
[0023] Proteins (amount or presence) may be detected, for example, using an immunoassay such as a sandwich immunoassay, a competitive immunoassay, a radioimmunoassay, fluorophore-labelled immunoassay, an ELISA or a Western blot. Mass spectroscopy also may be used. Variant proteins (amount or presence) may be detected, for example, using variant-specific antibodies. Truncated proteins (amount or presence) may be detected, for example, by a difference in the size of the protein by Western blot analysis or mass spectroscopy.
[0024] In certain embodiments, the method comprises in the detecting step determining the presence of a deletion, for example, a deletion in a CFHR1 or CFHR3 gene, or the absence or a reduction of corresponding gene product (e.g., the amount or activity of the gene product) indicating a higher risk of the subject developing a vascular disorder.
[0025] In other embodiments, the method comprises in the detecting step determining the absence of a deletion, for example, the presence of a CFHR1 or CFHR3 gene, or the presence or an increase of the corresponding gene product (e.g., the amount or activity of the gene product) indicating a lower risk of the subject developing a vascular disorder.
[0026] In another embodiment, the method comprises in the detecting step determining the presence of a deletion, for example, a deletion in a CFHR1 or CFHR3 gene, or the absence or a reduction
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 of the corresponding gene product (e.g., the amount or activity of the gene product) indicating a lower risk of the subject developing AMD.
[00271 In yet another embodiment, the method comprises in the detecting step determining the absence of a deletion, for example, the presence of a CFHRl or CFHR3 gene, or the presence or an increase of the corresponding gene product (e.g., the amount or activity of the gene product) indicating a higher risk of the subject developing AMD. The increase in gene product, for example, can be at least 10%, at least 20%, at least 50%, or more.
[0028] In certain embodiments, the method further comprises detecting at least one other genetic variant or biomarker indicative of AMD and/or vascular disease. Genetic variants that may be detected in the invention include genetic variants of complement factor H (CFH) gene, HTRA1 gene, complement factor B (BF) gene and/or the complement component 2 (C2) gene. In an embodiment, the genetic variants include one or a plurality of polymorphic sites, such as those described herein.
[0029] In another aspect, the invention provides a method for treating a subject having (i.e., exhibiting symptoms of), or is at risk for developing, a vascular disorder, by administering a CFHRl polypeptide and/or a CFHR3 polypeptide to the subject. The polypeptide may be a full-length CFHRl polypeptide or a fragment or portion thereof. The polypeptide may be a full-length CFHR3 polypeptide or a fragment or portion thereof.
[0030] In another aspect the invention provides a pharmaceutical composition comprising a
CFHR3 protein or fragment thereof and at least one pharmaceutically effective excipient. In another aspect the invention provides a pharmaceutical composition comprising a CFHRl protein or fragment thereof and at least one pharmaceutically effective excipient.
[0031] In another aspect the invention provides the use of a protein comprising the gene product of at least a portion of the CFHR3 and/or CFHRl gene for the preparation of a medicament for the treatment of a vascular disorder.
[0032] In another aspect the invention provides gene therapy vectors comprising nucleic acid encoding a CFHR3 or CFHRl protein, or fragment thereof. The vector may include a promoter that drives expression of the CFHR3 or CFHRl gene in multiple cell types. Alternatively, the vector may include a promoter that drives expression of the CFHR3 or CFHRl gene only in specific cell types, for example, in cells of the retina or in cells of the kidney. In a related aspect pharmaceutical compositions are provided containing a gene therapy vector encoding a CFHR3 or CFHRl protein or fragment thereof and a pharmaceutically acceptable excipient.
[0033] In another aspect the invention provides a method of treating a subject having (i.e., exhibiting symptoms of), or susceptible to developing, age-related macular degeneration (AMD), by administering an agent that reduces the expression of the CFHRl and/or CFHR3 genes or reduces the
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 activity or amount of a gene product of the CFHR1 and/or CFHR3 genes. Agents include antisense RNA, siRNA or ribozyme that reduces expression of the CFHR1 and/or CFHR3 genes. In a related aspect the level of protein is reduced, for example by using plasmaphoresis or antibody-based inhibition, for example, using an anti-CFHRl antibody and/or an anti-CFHR.3 antibody.
[0034] In another aspect the invention provides a pharmaceutical composition comprising an anti-CFHRl antibody and a pharmaceutically acceptable carrier. In one embodiment, an anti-CFHRl antibody specifically binds the amino-terminus of a CFHR1 polypeptide. In another aspect the invention provides a pharmaceutical composition comprising an anti-CFHR3 antibody and a pharmaceutically acceptable carrier. In one embodiment, an anti-CFHR3 antibody specifically binds the carboxyl-terminus of a CFHR3 antibody.
[0035] In another aspect the invention provides a diagnostic kit for diagnosing susceptibility to a vascular disorder and/or AMD in a subject, comprising nucleic acid primers or probes that detect the presence or absence of a deletion in the DNA sequence between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H-related 4 (CFHR4) gene on human chromosome 1.
[0036] In another aspect the invention provides a diagnostic device comprising nucleic acid primers or probes that detect the presence or absence of a deletion in the DNA sequence between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor Hrelated 4 (CFHR4) gene on human chromosome 1 immobilized on a substrate, such as a microarray.
[0037] In another aspect the invention provides a diagnostic kit for diagnosing susceptibility to a vascular disorder and/or AMD in a subject, comprising antibodies that detect the presence or absence of the complement Factor H-related 3 (CFHR3) protein, or variant or truncated forms thereof, and/or complement Factor H related 1 (CFHR1) protein, or variant or truncated forms thereof, in a body fluid or tissue sample of the subject.
[0038] In another aspect the invention provides a drug screening method for screening for agents for use in treating a vascular disorder. The method involves a) combining (i) a cell that expresses CFHR3 and/or CFHR1 polypeptides; and (ii) a test agent; b) measuring the level of CFHR3 and/or CFHR1 polypeptides secreted into the medium; and c) comparing the level of CFHR3 and/or CFHR1 polypeptides secreted into the medium in the presence of the test agent with a reference value, said reference value being the level of CFHR3 and/or CFHR1 polypeptides secreted into the medium in the absence of the test agent, where a higher level of CFHR3 and/or CFHR1 polypeptides secreted into the medium in the presence of the test agent indicates the test agent may be useful for treating the vascular disorder.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 [0039] In another aspect the invention provides a method for identifying a CFH protein likely to protect against AMD development, by identifying a subject with a deletion in the DNA sequence between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H-related 4 (CFHR4) gene on human chromosome 1; determining the sequence of the CFH gene encoded by the gene contained in the chromosome containing the deletion; and determining the sequence of the protein encoded by the CFH gene, wherein said protein is different from wild-type CFH, said protein being a CFH protein likely to protect against AMD development. The invention also provides a protective CFH protein obtained using the method.
BRIEF DESCRIPTION OF THE FIGURES [0040] . Figure 1 is a diagram showing the organization of the regulators-of-complementactivation (RCA) gene cluster on chromosome lq32 and the arrangement of approximately 60-amino acid domains known as short consensus repeats (SCRs) in complement Factor H (CFH), Factor H-Like 1 (CFHL1) and Factor H-Related 1, 2, 3, 4 and 5 (CFHR1, CFHR2, CFHR3, CFHR4 and CFHR5). CFH has 20 SCRs. The interacting partners with some of these SCRs has been determined and is shown on the top right (CRP, C reactive protein; Hep, heparin). Complement factor H-like 1 (CFHL1) is a splice isoform of CFH, while complement factor H-related proteins 1-5 (CFHR1-5) are each encoded by a unique gene (CFHR1-5). The SCRs of CFHR1-5 are similar to some of the SCRs in CFH, as denoted by the numbers in the ovals. For example, CFHR5 has 9 SCRs, with the first two being similar to SCRs 6 and 7 of Factor H and therefore having CRP and heparin binding properties. SCRs 5-7 of CFHR5 have the numbers 12-14 within the corresponding ovals because these SCRs are similar to SCRs 12-14 of Factor H and have C3b and heparin binding properties.
[0041] Figure 2 shows regions of homology (genomic duplications) in the genes encoding CFH and the Factor H-related proteins. Exons are indicated as vertical lines. Regions labeled with the same letter (e.g., A, A', and A) have substantially identical sequences.
[0042] Figure 3 shows a Western blot of serum proteins from seven patients using an anti-human
CFH antibody. FHL-1, CFHR1 and CFHR2 indicate the positions of the truncated form of CFH, CFHR1 and CFHR2, respectively. The anti-human CFH antibody employed also cross-reacts with CFHR1 and CFHR2. No CFHR1 is detected in the serum of two patients (197-02 and 325-02) that have a homozygous deletion of the CFHR3 and CFHR1 genes, as determined by SSCP analysis and direct DNA sequencing.
[0043] Figure 4 shows a SSCP analysis of the CFH, CFHR3 and CFHR1 genes. 1, 2, 3, and 4 indicate four different SSCP patterns observed using primers from exon 22 of the CFH gene to PCR amplify DNA. SSCP patterns 1, 2 and 3 correspond to homozygous non-deletion or heterozygous
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 deletion of CFHR3 and CFHR1, and pattern 4 corresponds to homozygous deletion of CFHR3 and CFHRl.
[0044] Figure 5 shows a PCR analysis of the CFH and CFYi-related genes 1 to 5 in leukocytes from 20 patients that are separated into four groups according to the SSCP patterns using the CFH exon 22 primers (patterns 1-4 are as described in Figure 4). From left to right, in each panel (gel), 5 leukocytederived DNA samples each from patients displaying SSCP patterns 1, 2, 3 and 4 were subjected to PCR using primers specific for CFH, CFHRl, CFHR2, CFHR3, CFHR4 and CFHR5, as indicated. When SSCP analysis and direct DNA sequencing show a homozygous deletion of the CFHR3 and CFHRl genes, no PCR amplifiable CFHR3 and CFHRl DNA are detected.
[0045] Figure 6 shows an amino acid alignment of the CFH (SEQ ED NO: 2), CFHRl (SEQ ID
NO: 4), and CFHR3 (SEQ ID NO: 6) proteins.
[0046] Figure 7 shows a nucleotide alignment of the CFH (SEQ ID NO: 1), CFHRl (SEQ ID
NO: 3), and CFHR3 genes (SEQ ID NO: 5).
DETAILED DESCRIPTION
1. Definitions [0047] The following definitions are provided to aid in understanding the invention. Unless otherwise defined, all terms of art, notations and other scientific or medical terms or terminology used herein are intended to have the meanings commonly understood by those of skill in the arts of medicine and molecular biology. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not be assumed to represent a substantial difference over what is generally understood in the art.
[0048] A vascular disorder is a disease or condition of the vascular system. One type of vascular disorder is an aneurysm such as abdominal aortic aneurysm or brain intracranial aneurysm. Other types of vascular disorder include hypertension, cerebral vascular accidents, trans-ischemic accidents (e.g., stroke). Still other types of vascular disorders include coronary artery disease, peripheral artery disease, varicose veins, and peripheral vascular disease.
[0049] A nucleic acid, polynucleotide or oligonucleotide is a polymeric form of nucleotides of any length, may be DNA or RNA, and may be single- or double-stranded. Nucleic acids may include promoters or other regulatory sequences. Oligonucleotides are usually prepared by synthetic means. A reference to the sequence of one strand of a double-stranded nucleic acid defines the complementary sequence and except where otherwise clear from context, a reference to one strand of a nucleic acid also refers to its complement. For certain applications, nucleic acid (e.g., RNA) molecules may be modified to increase intracellular stability and half-life. Possible modifications include, but are not limited to, the use
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 of phosphorothioate or 2'-O-methyl rather than phosphodiesterase linkages within the backbone of the molecule. Modified nucleic acids include peptide nucleic acids (PNAs) and nucleic acids with nontraditional bases such as inosine, queosine and wybutosine and acetyl-, methyl-, thio-, and similarly modified forms of adenine, cytidine, guanine, thymine, and uridine which are not as easily recognized by endogenous endonucleases.
[0050] Hybridization probes are nucleic acids capable of binding in a base-specific manner to a complementary strand of nucleic acid. Such probes include nucleic acids and peptide nucleic acids (Nielsen et al., 1991). Hybridization may be performed under stringent conditions which are known in the art. For example, see, e.g., Berger and Kimmel (1987) METHODS IN ENZYMOLOGY, VOL. 152: GUIDE TO Molecular Cloning Techniques, San Diego: Academic Press, Inc.; Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual, 2nd Ed., Vols. 1-3, Cold Spring Harbor Laboratory; Sambook (2001) 3rd Edition; Rychlik, W. and Rhoads, R.E., 1989, Nucl. Acids Res. 17, 8543; Mueller, P.R. et al. (1993) In: CURRENT PROTOCOLS IN MOLECULAR BIOLOGY 15.5, Greene Publishing Associates, Inc. and John Wiley and Sons, New York; and Anderson and Young, QUANTITATIVE FILTER Hybridization in Nucleic Acid Hybridization (1985)). As used herein, the term probe includes primers. Probes and primers are sometimes referred to as oligonucleotides.
[0051] The term primer refers to a single-stranded oligonucleotide capable of acting as a point of initiation of template-directed DNA synthesis under appropriate conditions, in an appropriate buffer and at a suitable temperature. The appropriate length of a primer depends on the intended use of the primer but typically ranges from 15 to 30 nucleotides. A primer sequence need not be exactly complementary to a template but must be sufficiently complementary to hybridize with a template. The term primer site refers to the area of the target DNA to which a primer hybridizes. The term primer pair means a set of primers including a 5' upstream primer, which hybridizes to the 5' end of the DNA sequence to be amplified and a 3' downstream primer, which hybridizes to the complement of the 3' end of the sequence to be amplified.
[0052] Exemplary hybridization conditions for short probes and primers is about 5 to 12 degrees C below the calculated Tm. Formulas for calculating Tm are known and include: Tm = 4°C x (number of G’s and C’s in the primer) + 2°C x (number of A’s and T’s in the primer) for oligos <14 bases and assumes a reaction is carried out in the presence of 50mM monovalent cations. For longer oligos, the following formula can be used: Tm = 64.9°C + 41°C x (number of G’s and C’s in the primer - 16.4)/N, where N is the length of the primer. Another commonly used formula takes into account the salt concentration of the reaction (Rychlik, supra, Sambrook, supra, Mueller, supra.): Tm = 81.5°C + 16.6°C x (logl0[Na+] + [K+]) + 0.41°C x (%GC) - 675/N, where N is the number of nucleotides in the oligo. The aforementioned formulae provide a starting point for certain applications; however, the
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 design of particular probes and primers may take into account additional or different factors. Methods for design of probes and primers for use in the methods of the invention are well known in the art.
[0053] The term polymorphism refers to the occurrence of two or more genetically determined alternative sequences or alleles in a population. A polymorphic site is the locus at which sequence divergence occurs. Polymorphic sites have at least two alleles. A diallelic polymorphism has two alleles. A triallelic polymorphism has three alleles. Diploid organisms may be homozygous or heterozygous for allelic forms. A polymorphic site may be as small as one base pair. Examples of polymorphic sites include: restriction fragment length polymorphisms (RFLPs); variable number of tandem repeats (VNTRs); hypervariable regions; minisatellites; dinucleotide repeats; trinucleotide repeats; tetranucleotide repeats; and simple sequence repeats. As used herein, reference to a polymorphism can encompass a set of polymorphisms (i.e., a haplotype).
[0054] A single nucleotide polymorphism (SNP) occurs at a polymorphic site occupied by a single nucleotide, which is the site of variation between allelic sequences. The site is usually preceded by and followed by highly conserved sequences of the allele. A SNP usually arises due to substitution of one nucleotide for another at the polymorphic site. Replacement of one purine by another purine or one pyrimidine by another pyrimidine is called a transition. Replacement of a purine by a pyrimidine or vice versa is called a transversion. A synonymous SNP refers to a substitution of one nucleotide for another in the coding region that does not change the amino acid sequence of the encoded polypeptide. A nonsynonymous SNP refers to a substitution of one nucleotide for another in the coding region that changes the amino acid sequence of the encoded polypeptide. A SNP may also arise from a deletion or an insertion of a nucleotide or nucleotides relative to a reference allele.
[0055] The term deletion, when referring to a nucleic acid sequence, has the usual meaning in genetics of an allele in which one or more bases are missing compared to a reference or wild-type sequence. Deletions may be as short as one base-pair. Deletions detected in the present invention may be longer, such as a deletion of at least 100 bp, at least 200 bp, at least 300 bp, at least 400 bp, at least 500 bp, at least 600 bp, at least 700 bp, at least 800 bp, at least 900 bp, at least 1000 bp, at least 1100 bp, at least 1200 bp, at least 1300 bp, at least 1400 bp, at least 1500 bp, at least 1600 bp, at least 1700 bp, at least 1800 bp, at least 1900 bp, at least 2000 bp, at least 2500 bp, at least 3000 bp, at least 3500 bp, at least 4000 bp, at least 4500 bp, at least 5000 bp, at least 6000 bp, at least 7000 bp, at least 8000 bp, at least 9000 bp, at least 10,000 bp, at least 15,000 bp, at least 20,000 bp, at least 30,000 bp, at least 40,000 bp, at least 50,000 bp, at least 75,000 bp, at least 100,000 bp, at least 125,000 bp, at least 150,000 bp, at least 200,000 bp or at least 250,000 bp.
[0056] The term haplotype refers to the designation of a set of polymorphisms or alleles of polymorphic sites within a gene of an individual. For example, a 112 Factor H haplotype refers to the
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
Factor H gene comprising allele 1 at each of the first two polymorphic sites and allele 2 at the third polymorphic site. A diplotype is a haplotype pair.
[0057] An isolated nucleic acid means a nucleic acid species that is the predominant species present in a composition. Isolated means the nucleic acid is separated from at least one compound with which it is associated in nature. A purified nucleic acid comprises (on a molar basis) at least about 50, 80 or 90 percent of all macromolecular species present.
[0058] Two amino acid sequences are considered to have substantial identity when they are at least about 80% identical, preferably at least about 90% identical, more preferably at least about 95%, at least about 98% identical or at least about 99% identical. Percentage sequence identity is typically calculated by determining the optimal alignment between two sequences and comparing the two sequences. Optimal alignment of sequences may be conducted by inspection, or using the local homology algorithm of Smith and Waterman, 1981, Adv. Appl. Math. 2: 482, using the homology alignment algorithm of Needleman and Wunsch, 1970, J. Mol. Biol. 48: 443, using the search for similarity method of Pearson and Lipman, 1988, Proc. Natl. Acad. Sci. U.S.A. 85: 2444, by computerized implementations of these algorithms (e.g., in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.) using default parameters for amino acid comparisons (e.g., for gap-scoring, etc.). It is sometimes desirable to describe sequence identity between two sequences in reference to a particular length or region (e.g., two sequences may be described as having at least 95% identity over a length of at least 500 basepairs). Usually the length will be at least about 50, 100, 200, 300, 400, 500, 600, 700, 800, 900 or 1000 amino acids, or the full length of the reference protein. Two amino acid sequences can also be considered to have substantial identity if they differ by 1, 2, or 3 residues, or by from 2-20 residues, 2-10 residues, 3-20 residues, or 3-10 residues.
[0059] Linkage describes the tendency of genes, alleles, loci or genetic markers to be inherited together as a result of their location on the same chromosome. Linkage can be measured by percent recombination between the two genes, alleles, loci or genetic markers. Typically, loci occurring within a 50 centimorgan (cM) distance of each other are linked. Linked markers may occur within the same gene or gene cluster. Linkage disequilibrium or allelic association means the preferential association of a particular allele or genetic marker with a specific allele or genetic marker at a nearby chromosomal location more frequently than expected by chance for any particular allele frequency in the population. A marker in linkage disequilibrium can be particularly useful in detecting susceptibility to disease, even if the marker itself does not cause the disease.
[0060] The terms susceptibility, propensity, and risk refer to either an increased or decreased likelihood of an individual developing a disorder (e.g., a condition, illness, disorder or disease) relative to a control population. In one example, the control population may be individuals in the
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 population (e.g., matched by age, gender, race and/or ethnicity) without the disorder, or without the genotype or phenotype assayed for. In some contexts, the terms diagnosing and screening are used interchangeably (e.g., a person skilled in the art can diagnose a propensity to develop the disease).
[0061] The term diagnose and diagnosis refer to the ability to determine or identify whether an individual has a particular disorder (e.g., a condition, illness, disorder or disease).
[0062] The term “screen” or “screening” as used herein has a broad meaning. It includes processes intended for the diagnosis or for determining the susceptibility, propensity, risk, or risk assessment of an asymptomatic subject for developing a disorder later in life. Screening also includes the prognosis of a subject, i.e., when a subject has been diagnosed with a disorder, determinating in advance the progress of the disorder as well as the assessment of efficacy of therapy options to treat a disorder. [0063] The terms portion, fragment and/or truncated form when used in reference to a
Factor H-related gene product (e.g., CFHR.3 or CFHR1 gene product), refers to a nucleic acid or polypeptide sequence that is less than the full-length sequence (i.e., a portion of the full-length gene or polypeptide). A portion or fragment or truncated form of CFHR3 or CFHR1 gene or polypeptide can be at least 25, at least 50, at least 75, at least 100, at least 150, at least 200, at least 250, or at least 300 nucleotides or amino acids in length. Typically the portion includes at least 1, often at least two, and sometimes at least 3 or 4 complete SCRs.
[0064] As used herein, the term “gene product” means an RNA (e.g., mRNA) or protein that is encoded by the gene. A protein coding region is a region of DNA/RNA sequence within a gene that encodes a polypeptide or protein.
[0065] An assay is a procedure wherein the presence or amount or a property of a test substance, e.g., a nucleic acid or gene product, is detected or measured.
[0066] The terms “inhibit” and “reduce” refer to any inhibition, reduction, or decrease in expression or activity including partial or complete inhibition of gene expression or gene product activity.
2. Association of Polymorphisms in the CFHR1 And CFHR3 Genes and Risk Of Developing AMD and Vascular Disorders [0067] A correlation between polymorphic sites and haplotypes in the CFH gene and the likelihood of developing AMD has been discovered. See Hageman et al., 2005, Proc. Natl. Acad. Sci. U.S.A. 102:7227-32; Haines et al., 2005, Science 308:419-21; Klein et al., 2005, Science 308:385-9; Edwards et al., 2005, Science 308:421-4 and U.S. patent publication No. 20070020647, each incorporated by reference in its entirety for all purposes. Both CFH risk haplotypes and CFH protective haplotypes are known. Polymorphisms particularly associated with increased risk include a variant allele at: rsl061170 (402H; exon 9); rs203674 (intron 10) and the polymorphism at residue 1210 (1210C; exon 22).
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
Polymorphisms particularly associated with decreased risk include the protective H2 haplotype, which includes a variant allele in IVS6 (intron 6, rs3766404) and the H4 haplotype, which includes a variant allele in IVS1 (intron 1, rs529825) and a variant allele (162) (exon 2, rs800292).
[0068] It has now been discovered that an AMD protective haplotype is genetically linked to deletions in the DNA sequence between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H-related 4 (CFHR4) gene on human chromosome 1 (i.e., the DNA sequence encoding the CFHR1 and CFHR3 proteins). See Example 1, infra. The discovery that deletions at the CFHR1 and CFHR3 loci are associated with decreased risk of developing AMD has a number of specific applications, including screening individuals to ascertain risk of developing AMD and identification of new and optimal therapeutic approaches for individuals afflicted with, or at increased risk of developing, AMD. As discussued in Example 1, below, the deletion genotype is predominantly associated with the CE7/H4 haplotype. See Hageman et al., 2005, Proc. Natl. Acad. Sci. U.S.A. 102:7227-32. Thus, this deletion acts as a marker for decreased risk of conditions for which the H4 haplotype is protective.
[0069] Moreover, it has now been discovered that deletions in the DNA sequence between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H related 4 (CFHR4) gene on human chromosome 1 (i.e., the DNA sequence encoding the CFHR1 and CFHR3 proteins) are associated with increased risk of developing a vascular disease such as aortic aneurysm. See Example 1, infra. The discovery that deletions at the CFHR1 and CFHR3 loci are associated with increased risk of developing a vascular disorder has a number of specific applications, including screening individuals to ascertain risk of developing a vascular disorder and identification of new and optimal therapeutic approaches for individuals afflicted with, or at increased risk of developing, vascular disorders.
3. Screening Methods [0070] Based on the discoveries described herein, a subject's risk for AMD or vascular disease can be assessed by determining whether or not a the subject has a deletion within the region of chromosome 1 lying between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H-related 4 (CFHR4). The extent of the deletion may vary in different individuals or populations. For example, in one embodiment the all of most of the region between CFH exon 22 and CFHR4 exon 1 is deleted. Alternatively, a portion of the region may be deleted, such as, for example, a deletion of less than the entire region between CFH exon 22 and CFHR4 exon 1 but including the CFHR1 encoding sequence, or including the CFHR3 encoding sequence,
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 including both, or including a non-coding (e.g., intragenic) sequence. An individual may be homozygous for deletion (both chromosomes 1 have a deletion in the region) or may be heterozygous for deletion. [0071] For example and not limitation, the homozygous deletion of CFHR1 and/or CFHR3 can be detected from the absence of CFHR1 and/or CFHL3 protein in a body fluid or tissue sample (see Figure 3), by the absence of RNA encoded in the region between the 3' end of CFH exon 22 and the 5' end of CFHR4 exon 1 (e.g., absense of absence of CFHR1 and/or CFHL3 mRNAs), or by absense of genomic DNA in the region in the region between the 3' end of CFH exon 22 and the 5' end of CFHR4 exon 1. The present or absense of DNA or RNA sequences can be determined using art known methods, such as PCR. The absense of a nucleic acid sequence is deduced from the absense of an amplified PCR product in an assay of a tissue sample (see Figure 5). It will be understood that, although PCR is frequently cited herein as a method for genetic analysis, many other analytical methods are known and are suitable for detection of a deletion. For example and not limitation several are described below in the section captioned Analysis of Nucleic Acid Samples.
[0072] The heterozygous deletion of CFHR1 and/or CFHR3 can be determined, for illustration and not limitation, (1) from a reduction in the amount of protein in a body fluid or tissue sample as compared to the amount from a control having both alleles of CFHR1 and/or CFHR3 genes, (2) from a reduction in the amount of RNA, DNA, or amplified PCR product in a tissue sample as compared to the amount from similar sample of a homozygote without the deletion, or (3) by an assay using direct DNA sequencing, quantitative PCR or other methods known in the art. For example, the amount of a gene product may be reduced in a heterozygote by at least 10%, at least 20%, at least 30%, about 50% or or more compared to a homozygote without the deletion. Quantitative PCR and methods are available that would be able to detect a two-fold difference in mRNA or DNA in a sample.
[0073] As noted, a deletion lies in the region between CFH exon 22 and CFHR4 exon 1 but need not span the entire region. Deletions of a portion of the CFHR1 and/or CFHR3 genes (partial deletions) may result in truncated forms of CFHR1 and/or CFHR3 RNAs and polypeptides. Such partial deletions can be identified by a difference in size of a protein in a body fluid or tissue sample compared to the full-length protein, by detecting a size difference in the RNA, and by various methods well known in the art, including PCR amplification of DNA or RNA in a biological sample using primers selected to distinguish between a nucleic acid comprising a deletion and a nucleic acid not containg a deletion. Methods known in the art can be used to distinguish homozygotes from heterozygotes (see, e.g., Example 1).
[0074] The selection, design and manufacture of suitable primers or probes for analysis of nucleic acid is well known in the art. A person of ordinary skill in the art can use suitable combinations of primers to detect deletions. In an embodiment, the primers or probes are designed to hybridize at any
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun2016 position in the DNA sequence between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H-related 4 (CFHR4) gene. For instance, both primers may be located in the CHFR3 gene to detect its presence or absence. In another example. In other examples, one or more primers are located within intergenic (non-coding) sequence, e.g., intergenic sequence between between CFHR3 and CFHR1 or between CFHR1 and CFHR4.
[0075] In another embodiment, the invention includes a method of detecting a nonreciprocal transfer of genetic information, such as gene conversion. In one instance, the gene conversion results in replacement of a 3’ portion of the CFH gene with a portion of the 3’ CFHR1 gene, such that a chimeric protein with sequence derived from both the CFH gene and the CFHR1 gene is produced.
3.1 Analysis of Nucleic Acid Samples [0076] Methods for detection of polymorphisms and deletions in genetic sequences are well known in the art and can be adapted for use in the present invention.
[0077] In one embodiment, genomic DNA is analyzed. For assay of genomic DNA, virtually any biological sample containing genomic DNA or RNA, e.g., nucleated cells, is suitable. For example, genomic DNA can be obtained from peripheral blood leukocytes collected from case and control subjects (QIAamp DNA Blood Maxi kit, Qiagen, Valencia, CA). Other suitable samples include saliva, cheek scrapings, biopsies of retina, kidney, skin, or liver or other organs or tissues; amniotic fluid, cerebral spinal fluid (CSF) samples; and the like. Alternatively RNA or cDNA can be assayed. Methods for purification or partial purification of nucleic acids from patient samples for use in diagnostic or other assays are well known [0078] Methods for detecting polymorphisms and deletions in nucleic acids include, without limitation, Southern blot analysis (see Kees et al., Homozygous Deletion of the pl6/MTSl Gene in Pediatric Acute Lymphoblastic Leukemia Is Associated With Unfavorable Clinical Outcome, Blood 89:4161-4166, Fizzotti et al., Detection of homozygous deletions of the cyclin-dependent kinase 4 inhibitor (pi6) gene in acute lymphoblastic leukemia and association with adverse prognostic features, Blood 85(10):2685-2690, Kitada et al., Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism, Nature 392 (9):605-608); Northern Blot Analysis (see Fieschi et al., A novel form of complete IL-12/IL-23 receptor bl deficiency with cell surface-expressed nonfunctional receptors, Immunobiology 104(7):2095-2101) and amplification based method such as PCR-based methods are used to detect deletions in samples. PCR primers may be designed to target DNA sequences flanking a known mutation, in which a change in PCR product size in comparison to amplification reactions using WT DNA identifies a mutant template. Primers may also be targeted to deleted sequences, wherein an absence of a PCR product identifies a mutant template (Kitada et al., Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism, Nature 392:605-608) including multiplex PCR (Chong et al.,
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
Single-tube multiplex-PCR screen for common deletional determinants of α-thalassemia, Blood 95 (1):360-362).
[0079] Polymorphisms (e.g., deletions) can also be detected using allele-specific probes; use of allele-specific primers; direct sequence analysis; denaturing gradient gel electropohoresis (DGGE) analysis; single-strand conformation polymorphism (SSCP) analysis; and denaturing high performance liquid chromatography (DHPLC) analysis. Other well known methods to detect polymorphisms in DNA include use of: Molecular Beacons technology (see, e.g., Piatek et al., 1998; Nat. Biotechnol. 16:359-63; Tyagi, and Kramer, 1996, Nat. Biotechnology 14:303-308; and Tyagi, et al., 1998, Nat. Biotechnol. 16:49-53), Invader technology (see, e.g., Neri et al., 2000, Advances in Nucleic Acid and Protein Analysis 3826:117-125 and U.S. Patent No. 6,706,471), nucleic acid sequence based amplification (Nasba) (Compton, 1991), Scorpion technology (Thelwell et al., 2000, Nuc. Acids Res, 28:3752-3761 and Solinas et al., 2001, Duplex Scorpion primers in SNP analysis and FRET applications Nuc. Acids Res, 29:20), restriction fragment length polymorphism (RFLP) analysis, and the like.
[0080] The design and use of allele-specific probes for analyzing polymorphisms are described by e.g., Saiki et al., 1986; Dattagupta, EP 235,726; and Saiki, WO 89/11548. Briefly, allele-specific probes are designed to hybridize to a segment of target DNA from one individual but not to the corresponding segment from another individual, if the two segments represent different polymorphic forms. Hybridization conditions are chosen that are sufficiently stringent so that a given probe essentially hybridizes to only one of two alleles. Typically, allele-specific probes are designed to hybridize to a segment of target DNA such that the polymorphic site aligns with a central position of the probe.
[0081] Exemplary probes for analyzing deletions and polymorphisms are shown in Table 1 of
Example 1, but many others may be designed by one of skill.
[0082] Allele-specific probes are often used in pairs, one member of a pair designed to hybridize to the reference allele of a target sequence and the other member designed to hybridize to the variant allele. Several pairs of probes can be immobilized on the same support for simultaneous analysis of multiple polymorphisms within the same target gene sequence.
[0083] The design and use of allele-specific primers for analyzing polymorphisms are described by, e.g., WO 93/22456 and Gibbs, 1989. Briefly, allele-specific primers are designed to hybridize to a site on target DNA overlapping a polymorphism and to prime DNA amplification according to standard PCR protocols only when the primer exhibits perfect complementarity to the particular allelic form. A single-base mismatch prevents DNA amplification and no detectable PCR product is formed. The method works best when the polymorphic site is at the extreme 3'-end of the primer, because this position is most destabilizing to elongation from the primer.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun2016 [0084J Amplification products generated using PCR can be analyzed by the use of denaturing gradient gel electrophoresis (DGGE). Different alleles can be identified based on sequence-dependent melting properties and electrophoretic migration in solution. See Erlich, ed., PCR Technology, Principles and Applications for DNA Amplification, Chapter 7 (W.H. Freeman and Co, New York, 1992).
[0085] Alleles of target sequences can be differentiated using single-strand conformation polymorphism (SSCP) analysis. Different alleles can be identified based on sequence- and structuredependent electrophoretic migration of single stranded PCR products (Orita et al., 1989). Amplified PCR products can be generated according to standard protocols, and heated or otherwise denatured to form single stranded products, which may refold or form secondary structures that are partially dependent on base sequence.
[0086] Alleles of target sequences can be differentiated using denaturing high performance liquid chromatography (DHPLC) analysis. Different alleles can be identified based on base differences by alteration in chromatographic migration of single stranded PCR products (Frueh and Noyer-Weidner, 2003, Clin Chem Lab Med. 41(4):452-61). Amplified PCR products can be generated according to standard protocols, and heated or otherwise denatured to form single stranded products, which may refold or form secondary structures that are partially dependent on the base sequence.
[0087] Direct sequence analysis of polymorphisms can be accomplished using DNA sequencing procedures that are well-known in the art. See Sambrook et al., MOLECULAR CLONING, A LABORATORY Manual (2nd Ed., CSHP, New York 1989) and Zyskind et al., RECOMBINANT DNA LABORATORY MANUAL (Acad. Press, 1988).
[0088] Homozygote deletions can be identified by a variety of methods known in the art. For example, in one approach DNA samples are amplified for further analysis. In an embodiment, two CFHR1-specific primer pairs are used, for instance, (“CFHLlex6.F” [5’- AGTCGGTTTGGACAGTG -3’ (SEQ ID NO: 7)] & “CFHLlex6R” [5’- GCACAAGTTGGATACTCC -3’ (SEQ ID NO: 8)]; and/or “CHFLlex6.F2” [5’- CATAGTCGGTTTGGACAGTG -3’ (SEQ ID NO: 9)] & “CFHLlex6.R” [5’GCACAAGTTGGATACTCC -3’ (SEQ ID NO: 8)]). In another embodiment, CFHR3-specific primer pairs are used, for instance, (“CFHL3ex3.F” [5’- TCATTGCTATGTCCTTAGG -4’ (SEQ ID NO: 10)] & “CFHL3ex3.R” [5’- TCTGAGACTGTCGTCCG -3’ (SEQ ID NO: 11)]; and/or “CFHL3ex3seq.F” [5’TTTTGGATGTTTATGCG -3’ (SEQ ID NO: 12)] & “CFHL3ex3seq.R” [5’- AAATAGGTCCGTTGGC -3’ (SEQ ID NO: 13)]). Absence of the correct-sized PCR product indicates that the CFHL1 and/or CFHL3 gene(s) are deleted.
[0089] Similarly, heterozygote deletions can be identified by a variety of methods known in the art. For example, in one approach DNA samples are amplified for further analysis, for example with the same primers listed above, followed by direct sequencing. Heterozygotes are characterized, for instance,
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 by chromatograms in which one peak is approximately half the height of the second peak (in contrast to equal sized peaks) at the SNP positions (rs460897, rsl6840561, rs4230, rs414628 for CFHR1; rsl061170 for CFHR3). In another embodiment, a protocol employing ParAllele genotyping data, a copy number analysis is performed, in which samples that fail to genotype key markers (MRD 3855, MRD 3856, MRD_3857, rs385390, rs389897) in the region of these two genes are identified. All samples assigned a copy number of 0 (designated CN0) allow the haplotypes that contain the deletion to be defined. Having defined a deletion haplotype, linkage disequilibrium is used to infer whether samples could not carry a deletion. Specifically, if a sample is homozygous for a different allele than one that defines the haplotype, then it does not carry a deletion.
[0090] A wide variety of other methods are known in the art for detecting polymorphisms in a biological sample. For example and not limitation, see, e.g., Ullman et al. Methods for single nucleotide polymorphism detection U.S. Pat. No. 6,632,606; Shi, 2002, Technologies for individual genotyping: detection of genetic polymorphisms in drug targets and disease genes Am JPharmacogenomics 2:197205; and Kwok et al., 2003, Detection of single nucleotide polymorphisms Curr Issues Biol. 5:43-60).
3.2 Analysis of Protein Samples [0091] Methods for protein analysis that can be adapted for detection of proteins such as the
CFHR1 and CFHR3 gene products and variants or fragments thereof are well known. These methods include analytical biochemical methods such as electrophoresis (including capillary electrophoresis and one- and two-dimensional electrophoresis), chromatographic methods such as high performance liquid chromatography (HPLC), thin layer chromatography (TLC), hyperdiffusion chromatography, mass spectrometry, and various immunological methods such as fluid or gel precipitin reactions, immunodiffusion (single or double), Immunoelectrophoresis, radioimmnunoassay (RIA), enzyme-linked immunosorbent assays (ELISAs), immunofluorescent assays, Western blotting and others.
[0092] For example, a number of well established immunological binding assay formats suitable for the practice of the invention are known (see, e.g., Harlow, E.; Lane, D. ANTIBODIES: A LABORATORY MANUAL. Cold Spring Harbor, N.Y: Cold Spring Harbor Laboratory; 1988; and Ausubel et al., (2004) Current Protocols in Molecular Biology, John Wiley & Sons, New York NY. The assay may be, for example, competitive or non-competitive. Typically, immunological binding assays (or immunoassays) utilize a capture agent to specifically bind to and, often, immobilize the analyte. In one embodiment, the capture agent is a moiety that specifically binds to a variant or wild-type CFHR1 or CFHR3 polypeptide or subsequence (e.g., a fragment or truncated form of CFHR1 or CFHR3). The
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 bound protein may be detected using, for example, a detectably labeled anti-CFHRl or anti-CFHR3 antibody.
3.3 Screening Using Multiple Polymorphisms and Markers [0093] In diagnostic methods, analysis of CFHR1 and/or CFHR3 polymorphisms can be combined with analysis of polymorphisms in other genes associated with AMD or vascular disease (e.g., AAA), detection of protein markers of AMD (see, e.g., Hageman et al., patent publications US 20030017501; US 20020102581; WOO 184149; and WOO 106262; and US patent applications 11 /706,154 (entitled “Protective Complement Proteins and Age-Related Macular Degeneration”) and 11/706,074 (entitled “Variants in Complement Regulatory Genes Predict Age-Related Macular Degeneration”); Gorin et al., US20060281120; and Hoh, W02007/044897, each of which are incorporated herein by reference in their entirety for all purposes), assessment of other risk factors of AMD or vascular disease (such as family history).
[0094] For example, analysis of CFHR1 and/or CFHR3 polymorphisms (e.g., deletions) can be combined with the analysis of polymorphisms in the Complement Factor H gene (CFH). Genetic variants of the CFH gene that may be detected include, but are not limited to, a genotype of a T at position 1277 of the coding region of human CFH, any one or more of rs529825; rs800292; rs3766404; rsl061147; rsl061170; and rs203674; any one of more of intron 2 (IVS2 or insTT); rs2274700; exon 10A; and rs375046; one or both of rs529825 and rs800292; one or more of rsl061147, rsl061170 and rs203674; at least one of rs529825 and rs800292; and rs3766404; and at least one of rsl061147, rsl061170 and rs203674; at least rs529825, rs800292, rs3766404, rsl061170, and rs203674; and/or exon 22 (R1210C). See. e.g., Hartman et al., 2006, HTRAl promoter polymorphism in wet age-related macular degeneration Science 314:989-92, incorporated herein by reference.
[0095] In certain embodiments, the analysis of CFHR1 and/or CFHR3 polymorphisms can be combined with analysis of polymorphisms in the HTRAl gene (also known as the PRSS11 gene), the complement factor B (BF) gene, and/or the complement component 2 (C2) gene. Genetic variants of the HTRAl gene that may be detected include, but are not limited to, at least one of rsl0490924, rsl 1200638, rs760336, and rs763720. Each of the single nucleotide polymorphisms (SNPs) within the HTRAl gene are associated with increased risk of developing AMD. The genetic variants of the BF gene that may be detected include the presence of an A or G at rs641153 of the BF gene, or an R or Q at position 32 of the BF protein; and/or an A or T at rs4151667 of the BF gene, or L or H at position 9 of the BF protein. The genetic variants of the C2 protein that may be detected include a G or T at rs547154 of the C2 gene; and/or a C or G at rs9332379 of the C2 gene, or E of D at position 318 of the C2 protein. See, e.g., Gold
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 et al., 2006 Variation in factor B (BF) and complement component 2 (C2) genes is associated with agerelated macular degeneration Nat Genet. 38:458-62.
[0096] In addition, the analysis of CFHR1 and/or CFHR3 polymorphisms can be combined with an analysis of protein markers associated with AMD. The protein markers may include, but are not limited to, fibulin-3, vitronectin, β-crystallin A2, β-crystallin A3, β-crystallin A4, β-crystallin S, glucoseregulated protein 78 kD (GRP-78), calreticulin, 14-3-3 protein epsilon, serotransferrin, albumin, keratin, pyruvate carboxylase, villin 2, complement 1 q binding protein/hyaluronic acid binding protein (complement 1 q component), amyloid A (al amyloid A), amyloid P component, C5 and CSb-9 terminal complexes, HLA-DR, fibrinogen, Factor X, prothrombin, complements 3,5 and 9, complement reactive protein (CRP), HLA-DR, apolipoprotein A, apolipoprotein E, antichymotrypsin, p2 microglobulin, thrombospondin, elastin, collagen, ICAM-1, LFA1, LFA3, B7, IL-1, IL-6, IL-12, TNF-alpha, GM-CSF, heat shock proteins, colony stimulating factors (GM-CSF, M-CSFs), and IL-10.
4. Therapeutic Methods [0097] In an embodiment, the invention provides methods of treatment and/or prophylaxis of diseases associated with a deletion within a CFHR1 and/or CFHR3 gene, or with reduced amount or activity of a CFHR1 and/or CFHR3 gene product, though the administration of a CFHR1 or CFHR3 polypeptide, or at least one portion of a CFHR1 and/or a CFHR3 polypeptide, or mixtures thereof, to a subject. In one instance, the disease is vascular disease.
[0098] In an embodiment, the invention provides methods of treatment and/or prophylaxis of diseases associated with an absence of a deletion within a CFHR1 and/or CFHR3 gene, or with unchanged or increased amount or activity of a CFHR1 and/or CFHR3 gene product, though the administration of at least one agent that reduces or inhibits CFHR1 or CFHR3 polypeptide to a subject. In one instance, the disease is AMD.
4.1 Prevention and Treatment of Vascular Disorders [0099] A subject identified as having an elevated likelihood of developing a vascular disorder (e.g., aneurysm) can be treated by administering CFHR1 and/or CFHR3 polypeptides or biologically active fragments or variants thereof. The therapeutic polypeptide can be administered systemically (e.g., by intravenous infusion) or locally (e.g., directly to an organ or tissue, such as the eye or the liver). The polypeptides may have the sequence of wild-type (naturally occurring) polypeptides or may have an amino acid sequence substantially identical to the naturally occurring form.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 [0100] CFHRl and CFHR3 polypeptides or biologically active fragments or variants thereof may be isolated from blood (serum or plasma) or produced using conventional recombinant technology (see Ausubel et al., 2004, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, Greene Publishing and Wiley-Interscience, New York). Recombinant expression generally involved introducing the CFHRl or CFHR3 gene into an expression vector that include a promoter to drive transcription of the DNA which is adapted for expression in prokaryotic (e.g., E. coli) and eukaryotic (e.g., yeast, insect or mammalian cells) hosts. Suitable host cells include bacteria such as E. coli, yeast, filamentous fungi, insect cells, and mammalian cells, which are typically immortalized, including mouse, hamster, human, and monkey cell line. Usually, the promoter is a eukaryotic promoter for expression in a mammalian cell. Usually, transcription regulatory sequences comprise a heterologous promoter and optionally an enhancer, which is recognized by the host cell. Commercially available expression vectors can be used. Expression vectors can include host-recognized replication systems, amplifiable genes, selectable markers, host sequences useful for insertion into the host genome, and the like.
[0101] In another embodiment the recombinant CFHRl or CFHR3 is a full-length polypeptide, a variant thereof, or fragment thereof. In one embodiment the fragment is a biologically active fragment.
In this context, a biologically active CFHRl or CFHR3 polypeptide has an activity associated with wildtype CFHRl or CFHR3. For example, in some embodiments the fragment has heparin and/or CRP and/or C3b-protein binding activity. Preferably the fragment has substantial sequence identity to at least a portion of the wild-type proteins. Biologically active fragments may comprise varying lengths of sequence substantially identical to wild-type proteins, such as, for example, at least 100, 200, 500, 700, 900 or 1100 residues. Alternatively, biologically active fragments may comprise at least one SCR substantially identical to CFHRl or CFHR3, preferably at least 2, 3, 4, or 5 SCRs.
[0102] In specific embodiments, the biologically active fragment includes at least SCR 6-7. In another embodiment, the biologically active fragment includes at least SCR 19-20. In another embodiment, the biologically active CFH includes at least SCR1.
[0103] In certain embodiments the therapeutic Factor H polypeptides are chimeric or fusion proteins, and comprise sequence from other proteins. For example, a therapeutic Factor H polypeptide may contain portions of human CFHRl or CFHR3 as well as portions comprised, at least in part, of SCR (or CCP) consensus domains from other proteins (e.g., CR1, MCP, DAF, C4BP, CR2, CFH) and/or artificial SCR (CCP) consensus sequences. See U.S. Pat. No. 5,545,619, incorporated herein by reference.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
4.1.1 Therapeutic Compositions Containing CFHR1 or CFHR3 Polypeptides [0104] The invention provides therapeutic preparations of CFHR1 or CFHR3 polypeptides, which may be wild-type or variants (e.g., neutral or protective variants), and may be full length forms, truncated forms, or biologically active fragments, including splice variants and recombinant fusion proteins. Therapeutic CFHR1 or CFHR3 polypeptides can be made recombinantly. Therapeutic proteins can be recombinantly produced (e.g., in cultured bacterial or eukaryotic cells) and purified using methods well known in the art and described herein. Alternatively, CFHR1 or CFHR3 polypeptides can be isolated from cultured RPE cells (e.g., primary cultures) or other cells that express CFHR1 or CFHR3 endogenously. Recombinant or purified polypeptides subject to FDA approval must be tested for potency and identity, be sterile, be free of extraneous material, and all ingredients in a product (i.e., preservatives, diluents, adjuvants, and the like) must meet standards of purity, quality, and not be deleterious to the patient.
[0105] The invention provides a composition comprising a CFHR1 polypeptide or CFHR.3 polypeptide, and a pharmaceutically acceptable excipient or carrier. The term “pharmaceutically acceptable excipient or carrier” refers to a medium that is used to prepare a desired dosage form of a compound. A pharmaceutically acceptable excipient or carrier can include one or more solvents, diluents, or other liquid vehicles; dispersion or suspension aids; surface active agents; isotonic agents; thickening or emulsifying agents; preservatives; solid binders; lubricants; and the like. Remington’s Pharmaceutical Sciences, Fifteenth Edition, E.W. Martin (Mack Publishing Co., Easton, PA, 1975) and Handbook of Pharmaceutical Excipients, Third Edition, A.H. Kibbe ed. (American Pharmaceutical Assoc. 2000), disclose various carriers used in formulating pharmaceutical compositions and known techniques for the preparation thereof. The pharmaceutical compositions may be formulated using slow release agents or biodegradeable agents following techniques known in the art. In one embodiment, the pharmaceutically acceptable excipient is not deleterious to a mammal (e.g., human patient) if administered to the eye (e.g., by intraocular injection). For intraocular administration, for example and not limitation, the therapeutic agent can be administered in a Balanced Salt Solution (BSS) or Balanced Salt Solution Plus (BSS Plus) (Alcon Laboratories, Fort Worth, Texas, USA). In a related aspect, the invention provides a sterile container, e.g. vial, containing a therapeutically acceptable CFHR.1 or CFHR.3 polypeptides, optionally as a lyophilized preparation.
[0106] The amount of CFHR1 or CFHR.3 polypeptide, or biologically active fragment thereof, to be administered to an individual can be determined. In one embodiment, exogenous CFHR.1 or CFHR.3 can be administered to an individual in an amount sufficient to achieve a level similar to the plasma concentration of CFHR1 or CFHR.3 in a healthy individual, i.e., an amount sufficient to achieve a plasma level of from about 50 to 600 micrograms/ml, such as from about 100 to 560 micrograms/ml. The
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 amount of CFHRl or CFHR3 to be administered to an individual (e.g., a 160 pound subject) can be, for example and not for limitation, from about 10 milligrams to about 5000 milligrams per dose, from about 50 milligrams to about 2000 milligrams per dose, from about 100 milligrams to about 1500 milligrams per dose, from about 200 milligrams to about 1000 milligrams per dose, or from about 250 milligrams to about 750 milligrams per dose. The frequency with which CFHRl or CFHR3 can be administered to an individual can be, for example and not for limitation, twice per day, once per day, twice per week, once per week, once every two weeks, once per month, once every two months, once every six months, or once per year. The amount and frequency of administration of CFHRl or CFHR3 to an individual can be readily determined by a physician by monitoring the course of treatment.
[0107] Alternatively, the CFHRl or CFHR3 polypeptide, or biologically active fragment thereof, can be administered to an individual using gene therapy or cell therapy methods as described further below.
4.1.2 Gene Therapy Methods [0108] In another approach, CFHRl or CFHR3 polypeptide is administered by in vivo expression of protein encoded by exogenous polynucleotide (i.e., via gene therapy). In one example, gene therapy involves introducing into a cell a vector that expresses CFHRl or CFHR3 polypeptides or biologically active fragments of CFHRl or CFHR3. The cell may be an endogenous cell (i.e., a cell from the patient) or engineered exogenous cell.
[0109] Vectors can be viral or nonviral. A number of vectors derived from animal viruses are available, including those derived from adenovirus, adeno-associated virus, retroviruses, pox viruses, alpha viruses, rhadboviruses, and papillomaviruses. Usually the viruses have been attenuated to no longer replicate (see, e.g., Kay et al. 2001, Nature Medicine 7:33-40).
[0110] The nucleic acid encoding the polypeptide is typically linked to regulatory elements, such as a promoters and an enhancers, which drive transcription of the DNA in the target cells of an individual. The promoter may drive expression of the gene in all cell types. Alternatively, the promoter may drive expression of the CFHRl or CFHR3 gene only in specific cell types, for example, in cells of the retina, the liver or the kidney. The regulatory elements, operably linked to the nucleic acid encoding the polypeptide, are often cloned into a vector.
[0111] As will be understood by those of skill in the art, gene therapy vectors contain the necessary elements for the transcription and translation of the inserted coding sequence (and may include, for example, a promoter, an enhancer, other regulatory elements). Promoters can be constitutive or inducible. Promoters can be selected to target preferential gene expression in a target tissue, such as the RPE (for recent reviews see Sutanto et al., 2005, Development and evaluation of the specificity of a
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 cathepsin D proximal promoter in the eye Curr Eye Res. 30:53-61; Zhang et al., 2004, Concurrent enhancement of transcriptional activity and specificity of a retinal pigment epithelial cell-preferential promoter Mol Vis. 10:208-14; Esumi et al., 2004, Analysis of the VMD2 promoter and implication of E-box binding factors in its regulation J Biol Chem 279:19064-73; Camacho-Hubner et al., 2000, The Fugu rubripes tyrosinase gene promoter targets transgene expression to pigment cells in the mouse Genesis. 28:99-105; and references therein).
10112] Suitable viral vectors include DNA virus vectors (such as adenoviral vectors, adenoassociated virus vectors, lentivirus vectors, and vaccinia virus vectors), and RNA virus vectors (such as retroviral vectors). In one embodiment, an adeno-associated viral (AAV) vector is used. For recent reviews see Auricchio et al., 2005, Adeno-associated viral vectors for retinal gene transfer and treatment of retinal diseases Curr Gene Ther. 5:339-48; Martin et al., 2004, Gene therapy for optic nerve disease, Eye 18:1049-55; Ali, 2004, Prospects for gene therapy Novartis Found Symp. 255:165-72; Hennig et al., 2004, AAV-mediated intravitreal gene therapy reduces lysosomal storage in the retinal pigmented epithelium and improves retinal function in adult MPS VII mice Mol Ther. 10:106-16; Smith et al.,
2003, AAV-Mediated gene transfer slows photoreceptor loss in the RCS rat model of retinitis pigmentosa Mol Ther. 8:188-95; Broderick et al., 2005, Local administration of an adeno-associated viral vector expressing IL-10 reduces monocyte infiltration and subsequent photoreceptor damage during experimental autoimmune uveitis Mol Ther. 12:369-73; Cheng et al., 2005, Efficient gene transfer to retinal pigment epithelium cells with long-term expression. Retina 25:193-201; Rex et al., Adenovirusmediated delivery of catalase to retinal pigment epithelial cells protects neighboring photoreceptors from photo-oxidative stress. Hum Gene Ther. 15:960-7; and references cited therein).
[0113] Gene therapy vectors must be produced in compliance with the Good Manufacturing
Practice. (GMP) requirements rendering the product suitable for administration to patients. The present invention provides gene therapy vectors suitable for administration to patients including gene therapy vectors that are produced and tested in compliance with the GMP requirements. Gene therapy vectors subject to FDA approval must be tested for potency and identity, be sterile, be free of extraneous material, and all ingredients in a product (z.e., preservatives, diluents, adjuvants, and the like) must meet standards of purity, quality, and not be deleterious to the patient. For example, the nucleic acid preparation is demonstrated to be mycoplasma-free. See, e.g, Islam et al., 1997, An academic centre for gene therapy research-and clinical grade manufacturing capability, Ann Med 29, 579-583.
[0114] Methods for administering gene therapy vectors are known. In one embodiment, CFHR1 or CFHR3 expression vectors are introduced systemically (e.g., intravenously or by infusion). In one embodiment, expression vectors are introduced locally {i.e., directly to a particular tissue or organ, e.g., liver). In one embodiment, expression vectors are introduced directly into the eye (e.g., by intraocular
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 injection). As will be understood by those of skill in the art, the promoter chosen for the expression vectors will be dependent upon the target cells expressing the CFHR1 or CFHR3 polypeptides. In some embodiments, a cell type-specific promoter is used and in other embodiments, a constitutive or general promoter is used. For recent reviews see, e.g., Dinculescu et al., 2005, Adeno-associated virus-vectored gene therapy for retinal disease Hum Gene Ther. 16:649-63; Rex et al., 2004, Adenovirus-mediated delivery of catalase to retinal pigment epithelial cells protects neighboring photoreceptors from photooxidative stress Hum Gene Ther. 15:960-7; Bennett, 2004, Gene therapy for Leber congenital amaurosis Novartis Found Symp. 255:195-202; Hauswirth et al., Range of retinal diseases potentially treatable by AAV-vectored gene therapy Novartis Found Symp. 255:179-188, and references cited therein).
[0115] Thus in one aspect, the invention provides a preparation comprising a gene therapy vector encoding a CFHR1 or CFHR.3 polypeptide, optionally a viral vector, where the gene therapy vector is suitable for administration to a human subject and in an excipient suitable for administration to a human subject (e.g., produced using GLP techniques). Optionally the gene therapy vector comprises a promoter that is expressed preferentially or specifically in retinal pigmented epithelium cells.
[0116] Nonviral methods for introduction of CFHR1 or CFHR3 gene sequences, such as encapsulation in biodegradable polymers (e.g., polylactic acid (PLA); polyglycolic acid (PGA); and copolymers (PLGA) can also be used (for recent reviews see, e.g., Bejjani et al., 2005, Nanoparticles for gene delivery to retinal pigment epithelial cells Mol Vis. 11:124-32; Mannermaa et al., 2005, Longlasting secretion of transgene product from differentiated and filter-grown retinal pigment epithelial cells after nonviral gene transfer Curr Eye Res. 2005 30:345-53; and references cited therein). Alternatively, the nucleic acid encoding a CFHR1 or CFHR3 polypeptide may be packaged into liposomes, or the nucleic acid can be delivered to an individual without packaging without using a vector.
4.1.3. Cell Therapy Methods [0117] In another approach, CFHR1 or CFHR3 polypeptide is administered by in vivo expression of protein encoded by endogenous or exogenous CFHR1 or CFHR3 polynucleotide (i.e., via cell therapy). For example, hepatocyte transplantation has been used as an alternative to whole-organ transplantation to support many forms of hepatic insufficiency (see, e.g., Ohashi et al., Hepatocyte transplantation: clinical and experimental application, J Mol Med. 2001 79:617-30). According to this method, hepatocytes or other CFHR1- or CFHR3-expressing cells are administered (e.g., infused) to a patient in need of treatment. These cells migrate to the liver or other organ, and produce the therapeutic protein. Also see, e.g., Alexandrova et al., 2005, Large-scale isolation of human hepatocytes for therapeutic application Cell Transplant. 14(10):845-53; Cheong et al., 2004,Attempted treatment of
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 factor H deficiency by liver transplantation Pediatr Nephrol. 19:454-8; Ohashi et al., 2001, Hepatocyte transplantation: clinical and experimental application J Mol Med. 79:617-30; Serralta et al., 2005, Influence of preservation solution on the isolation and culture of human hepatocytes from liver grafts Cell Transplant. 14( 10):837-43; Yokoyama et al., 2006, In vivo engineering of metabolically active hepatic tissues in a neovascularized subcutaneous cavity Am. J. Transplant. 6(l):50-9; Dhawan et al., 2005, Hepatocyte transplantation for metabolic disorders, experience at King's College hospital and review of literature. Acta Grastroenterol. Belg. 68(4):457-60; Bruns et al., 2005, Injectable liver: a novel approach using fibrin gel as a matrix for culture and intrahepatic transplantation of hepatocytes Tissue Eng. 11(11-12):1718-26. Other cell types that may be used include, for illustration and not limitation, kidney and pancreatic cells. In one embodiment, the administered cells are engineered to express a recombinant form of the CFHRl or CFHR3 protein.
[0118] In another, related approach, therapeutic organ transplantation is used. Most of the body's systemic CFHRl and CFHR3 is produced by the liver, making transplantation of liver tissue the preferred method. See, Gerber et al., 2003, Successful (?) therapy of hemolytic-uremic syndrome with factor H abnormality Pediatr Nephrol. 18:952-5.
[0119] In another approach, a CFHRl or CFHR3 protein is delivered to the back of the eye by injection into the eye (e.g., intravitreal) or via encapsulated cells. Neurotech’s Encapsulated Cell Technology (ECT), as an example, is a unique technology that allows for the sustained, longterm delivery of therapeutic factors to the back of the eye. See (http://www.neurotech.fr). ECT implants consist of cells that have been genetically modified to produce a specific therapeutic protein that are encapsulated in a semi-permeable hollow fiber membrane. The cells continuously produce the therapeutic protein that diffuses out of the implant and into the eye (Bush et al., 2004, Encapsulated cell-based intraocular delivery of ciliary neurotrophic factor in normal rabbit: dose-dependent effects on ERG and retinal histology Invest Ophthalmol Vis Sci. 45:2420-30.). CNTF delivered to the human eye by ECT devices was recently shown to be completely successful and associated with minimal complications in 10 patients enrolled in a Phase I clinical trial (Sieving et al., 2006, Ciliary neurotrophic factor (CNTF) for human retinal degeneration: phase I trial of CNTF delivered by encapsulated cell intraocular implants Proc Natl Acad Sci USA 103(10):3896-901). Also see Song et al., 2003, Photoreceptor protection by cardiotrophin-1 in transgenic rats with the rhodopsin mutation s334ter IOVS, 44(9):4069-75; Tao et al., 2002, Encapsulated Cell-Based Delivery of CNTF Reduces Photoreceptor Degeneration in Animal Models of Retinitis Pigmentosa IOVS, 43 10:3292-3298; and Hammang et al., U.S. Pat. No. 6,649,184. In one embodiment of the present invention, a form of CFHRl or CFHR3 is expressed in cells and administered in an encapsulated form. In one embodiment, the cells used are the NTC-201 human RPE
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 line (ATCC # CRL-2302) available from the American Type Culture Collection P.O. Box 1549, Manassas, VA 20108.
4.2 Prevention and Treatment of AMD [0120] A subject identified as having an elevated likelihood of developing AMD, exhibiting symptoms of AMD, or susceptible to AMD, can be treated by reducing the expression, activity or amount of a gene product of the CFHR1 and/or CFHR3 genes. Any method of reducing levels of CFHR1 or CFHR3 in the eye or systemically may be used for treatment including, for example, inhibiting transcription of a CFHR1 or CFHR3 gene, inhibiting translation of CFHR1 or CFHR3 RNA, decreasing the amount or activity of CFHR1 or CFHR3 proteins (e.g., by plasmaphoresis, antibody-directed plasmaphoresis, or complexing with a CFHR1 or CFHR3 binding moiety (e.g., heparin or antibody), or by administration of inhibitory nucleic acids. In some embodiments levels of CFHR1 or CFHR3 are preferentially reduced in the eye (e.g., RPE) relative to other tissues. For illustration and not limitation, several methods are briefly described below.
4.2.1 Inhibitory Nucleic Acids [0121] Inhibitory nucleic acids are known and include antisense nucleic acids, interfering RNAs, ribozymes and others (see, e.g., Gomes et al., 2005, Intraocular delivery of oligonucleotides Curr Pharm Biotechnol. 6:7-15; and Henry et al., 2004, Setting sights on the treatment of ocular angiogenesis using antisense oligonucleotides Trends Pharmacol Sci 25:523-7; PCT Publications WO 98/53083; WO 99/32619; WO 99/53050; WO 00/44914; WO 01/36646; WO 01/75164; WO 02/44321; and U.S. Patent No. 6,107,094; Sui et al., 2002, A DNA vector-based RNAi technology to suppress gene expression in mammalian cells Proc Natl Acad Sci USA 99:5515-20; and Kasahara and Aoki, 2005, Gene silencing using adenoviral RNA vector in vascular smooth muscle cells and cardiomyocytes Methods Mol Λ/erf.l 12:155-72; US Patent Nos. US6180399; US5869254; US6025167; US5854038; US5591610; US5667969; US5354855;US5093246; US5180818; US5116742; US5037746; and US4987071; Dawson et al., 2000, Hammerhead ribozymes selectively suppress mutant type I collagen mRNA in osteogenesis imperfecta fibroblasts Nucleic Acids Res. 28:4013-20; Blalock et al., 2004 Hammerhead ribozyme targeting connective tissue growth factor mRNA blocks transforming growth factor-beta mediated cell proliferation Exp Eye Res. 78:1127-36; Kuan et al., 2004, Targeted gene modification using triplexforming oligonucleotides Methods Mol Biol. 262:173-94.
[0122] It will be understood that inhibitory nucleic acids can be administered as a pharmaceutical composition or using gene therapy or cell therapy methods.
WO 2008/008986
PCT/US2007/073514
2016204271 23Jun2016
4.2.2 Antibodies and Antibody Therapy [0123J In one aspect, an anti-CFHRl or anti-CFHR3 binding agents (e.g., antibodies) that reduce the activity or amount of the proteins is administered to an individual with or at risk for AMD. The antibody can be administered systemically or locally (see, e.g., Gaudreault et al., 2005, Preclinical pharmacokinetics of Ranibizumab (rhuFabV2) after a single intravitreal administration Invest Ophthalmol Vis Sci. 46:726-33).
[0124] In one embodiment, an anti-CFHRl antibody specifically binds an epitope of CFHR1, in particular human CFHR1. In certain embodiments, an anti-CFHRl antibody specifically binds an epitope located within the amino-terminus of a CFHR1 polypeptide. In particular, an anti-CFHRl antibody specifically binds an epitope located between amino acids 1-143 of SEQ ED NO: 4 as shown in Figure 6. In other embodiments, an anti-CFHRl antibody specifically binds an epitope within the CFHR1 short consensus repeats (SCRs) 6 and/or 7 as shown in Figure 1. The amino acid sequence of CFHR1 SCR6 is 35% homologous to the corresponding CFH SCR, and the amino acid sequence of CFHR SCR7 is 45% homologous to the corresponding CFH SCR. Anti-CFHRl antibodies of the invention specifically bind CFHR1 and do not cross-react with CFH or other factor H related proteins including CFHT, CFHR2, CFHR3, CFHR4, or CFHR5. A variety of immunoassay formats may be used to select antibodies that are specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with an antigen. See Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York.
Epitope mapping of the CFHR1 protein is within the skill of the art to determine epitopes that are most immunogenic for the generation of anti-CFHRl antibodies.
[0125] In another embodiment, an anti-CFHR3 antibody specifically binds an epitope of
CFHR3, in particular human CFHR3. In certain embodiments, an anti-CFHR3 antibody specifically binds an epitope located within the carboxyl-terminus of a CFHR3 polypeptide. For example, an antiCFHR3 antibody may specifically bind to an epitope between amino acids 144-330 of SEQ ID NO: 6 as shown in Figure 6. In other embodiments, an anti-CFHR3 antibody specifically binds an epitope within the CHFR3 SCRs 8, 19 and/or 20 as shown in Figure 1. The amino acid sequence of CFHR3 SCR8 is 63% homologous to the corresponding CRH SCR, the amino acid sequence of CFHR3 SCR19 is 62% homologous to the corresponding CFH SCR, and the amino acid sequence of CFHR3 SCR20 is 36% homologous to the corresponding CFH SCR. Anti-CFHR3 antibodies of the invention specifically bind CFHR3 and do not cross-react with CFH or other factor H related proteins including CFHT, CFHR1, CFHR2, CFHR4, or CFHR5. Epitope mapping of the CFHR3 protein is within the skill of the art to determine epitopes that may be immunogenic for the generation of anti-CFHR3 antibodies.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 [01261 It is understood that each of the antibodies discussed above can be an intact antibody, for example, a monoclonal antibody. Alternatively, the binding protein can be an antigen binding fragment of an antibody, or can be a biosynthetic antibody binding site. Antibody fragments include Fab, Fab’, (Fab’)2 or Fv fragments. Techniques for making such antibody fragments are known to those skilled in the art. A number of biosynthetic antibody binding sites are known in the art and include, for example, single Fv or sFv molecules, described, for example, in U.S. Patent Nos. 5,476,786. Other biosynthetic antibody binding sites include bispecific or bifunctional binding proteins, for example, bispecific or bifunctional antibodies, which are antibodies or antibody fragments that bind at least two different antigens. For example, bispecific binding proteins can bind CFHR1, CFHR3, and/or another antigen. Methods for making bispecific antibodies are known in art and, include, for example, by fusing hybridomas or by linking Fab’ fragments. See, e.g., Songsivilai et al. (1990) CLIN. EXP. IMMUNOL. 79: 315-325; Kostelny et al. (1992) J. IMMUNOL. 148: 1547-1553.
[0127] Anti-CFHRl and anti-CFHR3 antibodies can be produced using techniques well known in the art. Monoclonal antibodies can be produced using standard fusion techniques for forming hybridoma cells. See G. Kohler, et al., Nature, 256:456 (1975). Alternatively, monoclonal antibodies can be produced from cells by the method of Huse, et al., Science, 256:1275 (1989).
[0128] It is understood that the CDRs of the antibodies described herein confer the binding specificity to CFHR1 or CFHR3. The antibodies described herein can be used as diagnostic and/or therapeutic agents. It is understood that the antibodies of the invention can be modified to optimize performance depending upon the intended use of the antibodies. For example, when the antibody is being used as a therapeutic agent, the antibody can be modified to reduce its immunogenicity in the intended recipient. Alternatively or in addition, the antibody can be fused or coupled to another protein or peptide, for example, a growth factor, cytokine, or cytotoxin. Such modifications can be achieved by using routine gene manipulation techniques known in the art.
[0129] Various techniques for reducing the antigenicity of antibodies and antibody fragments are known in the art. These techniques can be used to reduce or eliminate the antigenicity of the antibodies of the invention. For example, when the antibodies are to be administered to a human, the antibodies preferably are engineered to reduce their antigenicity in humans. This process often is referred to as humanization. Preferably, the humanized binding proteins have the same or substantially the same affinity for the antigen as the original non-humanized binding protein it was derived from.
[0130] In one well known humanization approach, chimeric proteins are created in which immunoglobulin constant regions of antibodies from one species, e.g., mouse, are replaced with immunoglobulin constant regions from a second, different species, e.g., a human. In this example, the
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 resulting antibody is a mouse-human chimera, where the human constant region sequences, in principle, are less immunogenic than the counterpart murine sequences. This type of antibody engineering is described, for example, Morrison, et al. (1984) Proc. Nat. Acad. Sci. 81: 6851-6855, Neuberger et al., 1984, Nature 312: 604-608; U.S. Patent Nos. 6,893,625 (Robinson); 5,500,362 (Robinson); and 4,816,567 (Cabilly).
[0131] In another approach, known as CDR grafting, the CDRs of the light and heavy chain variable regions of an antibody of interest are grafted into frameworks (FRs) from another species. For example, murine CDRs can be grafted into human FR sequences. In some embodiments, the CDRs of the light and heavy chain variable regions of an anti-CFHRl antibody or an anti-CFHR3 antibody are grafted into human FRs or consensus human FRs. In order to create consensus human FRs, FRs from several human heavy chain or light chain amino acid sequences are aligned to identify a consensus amino acid sequence. CDR grafting is described, for example, in U.S. Patent Nos. 7,022,500 (Queen); 6,982,321 (Winter); 6,180,370 (Queen); 6,054,297 (Carter); 5,693,762 (Queen); 5,859,205 (Adair); 5,693,761 (Queen); 5,565,332 (Hoogenboom); 5,585,089 (Queen); 5,530,101 (Queen); Jones et al. (1986) NATURE 321: 522-525; Riechmann et al. (1988) NATURE 332: 323-327; Verhoeyen et al. (1988) SCIENCE 239: 1534-1536; and Winter (1998) FEBS LETT 430: 92-94.
[0132] In addition, it is possible to create fully human antibodies in mice. In this approach, human antibodies are prepared using a transgenic mouse in which the mouse’s antibody-producing genes have been replaced by a substantial portion of the human antibody producing genes. Such mice produce human immunoglobulin instead of murine immunoglobulin molecules. See, e.g., WO 98/24893 (Jacobovitz et al.) and Mendez et al., 1997, Nature Genetics 15: 146-156. Fully human anti-CFHRl and/or anti-CFHR3 monoclonal antibodies can be produced using the following approach. Transgenic mice containing human immunoglobulin genes are immunized with the antigen of interest, e.g., CFHR1 or CFHR3. Lymphatic cells from the mice then are obtained from the mice, which are then fused with a myeloid-type cell line to prepare immortal hybridoma cell lines. The hybridoma cell lines are screened and selected to identify hybridoma cell lines that produce antibodies specific to CLHR1 or CFHR3.
5. Drug Screening/Antagonists of Risk Variant Factor H or Variant CFHR5 [0133] The invention provides a drug screening method for screening for agents for use in treating vascular disorders. The method involves combining (i) a cell that expresses CFHR3 and/or CFHR1 polypeptides; and (ii) a test agent; b) measuring the level of CFHR3 and/or CFHR1 gene expression in the cell and c) comparing the level of CFHR3 and/or CFHR1 gene expression in the cell with a reference value, where the reference value is the level of CFHR3 and/or CFHR1 gene expression in
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 the absence of the test agent, where a higher level of CFHR3 and/or CFHR1 gene expression in the presence of the test agent indicates the test agent may be useful for treating the vascular disorders. Compounds from natural product libraries or synthetic combinatorial libraries may be screened. The level of CFHR3 and/or CFHR1 gene expression using a variety of approaches including measuring protein levels, measuring mRNA levels or other methods.
[0134] In one embodiment the method involves combining (i) a cell that expresses CFHR3 and/or CFHR1 polypeptides; and (ii) a test agent; b) measuring the level of CFHR3 and/or CFHR1 polypeptides produced by the cell {e.g., secreted into the medium); and c) comparing the level of CFHR3 and/or CFHR1 polypeptides secreted into the medium in the presence of the test agent with a reference value, said reference value being the level of CFHR3 and/or CFHR1 polypeptides produced (or secreted into the medium) in the absence of the test agent, where a higher level of CFHR3 and/or CFHR1 polypeptides secreted into the medium in the presence of the test agent indicates the test agent may be useful for treating the vascular disorders. Compounds from natural product libraries or synthetic combinatorial libraries may be screened.
6. Identifying Protective Forms of Complement Factor H Proteins [0135] As described above, deletions at the CFHR1 and CFHR3 loci are linked to the presence of a protective haplotype. Protective haplotypes and protective forms of CFH proteins are described in Hageman et al., 2005, Proc. Natl. Acad. Sci. U.S.A. 102:7227-32 and U.S. patent publication No. 20070020647. In one aspect, the invention provides a method for identifying a CFH protein likely to protect against development of AMD when administered to a subject having, or at risk of developing, AMD. The method involves identifying a subject with a deletion in the DNA sequence between the 3' end of exon 22 of the complement factor H {CFH) gene and the 5' end of exon 1 of complement Factor Hrelated 4 {CFHR4) gene on human chromosome 1; determining the sequence of the CFH gene encoded by the gene contained in the chromosome containing the deletion; and determining the sequence of the protein encoded by the CFH gene, wherein said protein is different from wild-type CFH, said protein being a CFH protein likely to protect against AMD development. The invention also provides a protective CFH protein obtained using the method. U.S. patent publication No. 20070020647 discloses the use of protective forms of CFH protein to protect against AMD development and to treat AMD.
7. Kits and Diagnostic Devices [0136] The invention provides reagents, devices and kits for detecting CFHR1 or CFHR3 deletions. A number of assay systems are known in the art, and it is within the skill of the art to arrive at means to determine the presence of variations associated with vascular disorders or AMD. The kit
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 reagents, such as multiple primers, multiple probes, combinations of primers, or combinations of probes, may be contained in separate containers prior to their use for diagnosis or screening. In an embodiment, the kit contains a first container containing a probe, primer, or primer pair for a first CFHR1 or CFHR3 allele described herein, and a second container containing a probe, primer, or primer pair for a second CFHR1 or CFHR3 allele described herein.
[0137] The kits may contain one or more pairs of CFHR1 and/or CFHR3 allele-specific oligonucleotides hybridizing to different forms of a polymorphism. The allele-specific oligonucleotides may be provided immobilized on a substrate.
[0138] The invention also provides devices and reagents useful for diagnostic, prognostic, drug screening, and other methods are provided. In one aspect, a device comprising immobilized primer(s) or probe(s) specific for detecting deletions in the CFHR1 and/or CFHR3 genes and optionally also including immobilized primeifs) or probe(s) specific for detecting polymorphic sites in CFH that are associated with AMD. Exemplary probes and polymorphic sites are described in U.S. patent publication No. 20070020647.
[0139] In one aspect, a device comprising immobilized primer(s) or probe(s) specific for one or more Factor H and/or CFHR5 and/or CFHR1 and/or CFHR3 gene products (polynucleotides or proteins) is provided. The primers or probes can bind polynucleotides (e.g., based on hybridization to specific polymorphic sites) or polypeptides (e.g., based on specific binding to a variant polypeptide).
[01401 In one embodiment, an array format is used in which a plurality (at least 2, usually at least 3 or more) of different primers or probes are immobilized. The term array is used in its usual sense and means that each of a plurality of primers or probes, usually immobilized on a substrate, has a defined location (address) e.g., on the substrate. The number of primers or probes on the array can vary depending on the nature and use of the device. For example, a dipstick format array can have as few as 2 distinct primers or probes, although usually more than 2 primers or probes, and often many more, will be present. One method for attaching the nucleic acids to a surface is by making high-density oligonucleotide arrays (see, Fodor et al., 1991, Science 251:767-73; Lockhart et al., 1996, Nature Biotech 14:1675; and U.S. Pat. Nos. 5,578,832; 5,556,752; and 5,510,270). It is also contemplated that, in some embodiments, a device comprising a single immobilized probe can be used.
[0141] In one embodiment, an array format is used in which a plurality (at least 2, usually at least 3 or more) of different primers or probes are immobilized. The term array is used in its usual sense and means that each of a plurality of primers or probes, usually immobilized on a substrate, has a defined location (address) e.g., on the substrate. The number of primers or probes on the array can vary depending on the nature and use of the device.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 [0142] In one embodiment, the immobilized probe is an antibody or other CFHR1 or CFHR3 binding moiety.
[0143] It will be apparent to the skilled practitioner guided by this disclosure than various polymorphisms and haplotypes can be detected, and used in combination with a deletion in the DNA sequence between the 3' end of exon 22 of the complement factor H (CFH) gene and the 5' end of exon 1 of complement Factor H-related 4 (CFHR4) gene on human chromosome 1, to assess the propensity of an individual to develop a Factor H related condition. Examples of CFH polymorphisms that may be assayed for include the following SNPs and combinations of SNPs: rs529825; rs800292; rs3766404; rsl061147; rsl061170; rs203674; and optionally including exon 22 (R1210C). In one embodiment the array includes primers or probes to determine the allele at at least one of the following polymorphic sites: rs529825; rs800292; intron 2 (IVS2 or insTT); rs3766404; rsl061147; rsl061170; exon 10A; rs203674; rs375046; and optionally including exon 22 (R1210C). In an embodiment the array includes primers or probes to determine the allele at at least one of the following polymorphic sites: (a) rs3753394; (b) rs529825; (c) rs800292; (d) intron 2 (IVS2 or insTT); (e) rs3766404; (f) rsl061147; (g) rs 1061170; (h) rs2274700; (i) rs203674; (]) rs3753396; (j) rsl065489; and optionally including exon 22 (R1210C). In one embodiment, the array includes primers or probes to determine the allele at at least one of the following polymorphic sites: rs800292 (I62V); IVS 2 (-18insTT); rsl061170 (Y402H); andrs2274700 (A473A). In one embodiment, the array includes primers or probes to determine the allele at at least one of the following polymorphic sites: rs9427661 (-249T>C); rs9427662 (-20T>C); and rsl2097550 (P46S). [0144] The array can include primers or probes to determine the allele at two of the above sites, at least three, at least four, at least five or at least six. In one embodiment the primers or probes distinguish alleles at rs529825. In one embodiment the primers or probes distinguish alleles at rs800292. In one embodiment the primers or probes distinguish alleles at rs3766404. In one embodiment the primers or probes distinguish alleles at rs 1061147. In one embodiment the primers or probes distinguish alleles at rsl061170. In one embodiment the primers or probes distinguish alleles at rs203674. In one embodiment the primers or probes distinguish alleles at exon 22 (R1210C). In one embodiment the primers or probes distinguish alleles at rs529825 and rs800292. In one embodiment the primers or probes distinguish alleles at two or three of rsl061147, rsl061170 and rs203674. In one embodiment the primers or probes distinguish alleles at rs529825 and rs800292, at rs3766404, two or three of rsl061147, rsl061170 and rs203674. In one embodiment the primers or probes distinguish alleles at rs529825, rs800292, rs3766404, rsl061170 and rs203674. In one embodiment, the primers or probes distinguish alleles at exon 22 (R1210C) and at rs529825; at rs800292; at rs3766404; at rsl061147; at rsl061170; at rs203674; at rs529825 and rs800292; at two or three of rsl061147, rsl061170 and rs203674; at rs529825 and rs800292, rs3766404, and two or three of rsl061147, rsl061170 and rs203674; or at rs529825,
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 rs800292, rs3766404, rsl 061170 and rs203674. In one embodiment, the primers or probes distinguish alleles at (a) any one or more of rs529825; rs800292; rs3766404; rs 1061147; rs 1061170; and rs203674; (b)any one of more of intron 2 (IVS2 or insTT); rs2274700; exon 10A; and rs375046; (c) one or both of rs529825 and rs800292; (d) one or more of rs 1061147, rsl 061170 and rs203674; (e) at least one of rs529825 and rs800292; and rs3766404; and at least one of rsl061147, rsl061170 and rs203674; (f) at least rs529825, rs800292, rs3766404, rs 1061170, and rs203674; (g) exon 22 (R1210C); (h) exon 22 (R1210C) and any of (a)-(g); or (i) any one or more of rs529825; rs800292; rs3766404; rs 1061147; rsl061170; rs203674; intron 2 (IVS2 or insTT); rs2274700; exon 10A; rs375046; and exon 22 (R1210C) and any one or more of rs9427661, rs9427662 and rsl2097550.
[0145] The array can include primers or probes to determine the allele at two of the above sites, at least three, at least four, at least five or at least six. In one embodiment the primers or probes distinguish alleles at rs529825. In one embodiment the primers or probes distinguish alleles at rs800292. In one embodiment the primers or probes distinguish alleles at intron 2 (IVS2 or insTT). In one embodiment the primers or probes distinguish alleles at rs3766404. In one embodiment the primers or probes distinguish alleles at rsl061147. In one embodiment the primers or probes distinguish alleles at rsl061170. In one embodiment the primers or probes distinguish alleles at exon 10A. In one embodiment the primers or probes distinguish alleles at rs2274700. In one embodiment the primers or probes distinguish alleles at rs203674. In one embodiment the primers or probes distinguish alleles at rs375046. In one embodiment the primers or probes distinguish alleles at exon 22 (R1210C). In one embodiment the primers or probes distinguish alleles at rs529825 and rs800292. In one embodiment the primers or probes distinguish alleles at two or three of rsl061147, rsl061170 and rs203674. In one embodiment the primers or probes distinguish alleles at of rs529825 and rs800292, at intron 2, at rs3766404, at two or three of rsl061147, rsl061170 and rs203674, at exon 10A, at rs2274700, and at rs375046. In one embodiment the primers or probes distinguish alleles at rs529825, rs800292, intron 2 (IVS2 or insTT), rs3766404, rsl061170, exon 10A, rs2274700, rs203674, and rs375046. In one embodiment, the primers or probes distinguish alleles at exon 22 (R1210C) and at either at rs529825; at rs800292; at intron 2 (IVS2 or insTT); at rs3766404; at rsl061147; at rsl061170; at rs2274700, at exon 10A; at rs203674; at rs375046; at rs529825 and rs800292; at two or three of rsl061147, rsl061170 and rs203674; at rs529825 and rs800292, intron 2 (IVS2 or insTT), rs3766404, two or three of rsl061147, rsl061170 and rs203674, rs2274700, exon 10A, and rs375046; or at rs529825, rs800292, intron 2 (IVS2 or insTT), rs3766404, rsl061170, rs2274700, exon 10A, rs203674, and rs375046. In one embodiment, the device distinguishes any combination of allelles at the sites listed above in the context of kits.
[0146] In one embodiment, the substrate comprises fewer than about 1000 distinct primers or probes, often fewer than about 100 distinct primers or probes, fewer than about 50 distinct primers or
WO 2008/008986
PCT/US2007/073514
2016204271 23Jun2016 probes, or fewer than about 10 distinct primers or probes. As used in this context, a primer is distinct from a second primer if the two primers do not specifically bind the same polynucleotide (i.e., such as cDNA primers for different genes). As used in this context, a probe is distinct from a second probe if the two probes do not specifically bind the same polypeptide or polynucleotide (i.e., such as cDNA probes for different genes). Primers or probes may also be described as distinct if they recognize different alleles of the same gene (i.e., CFH or CFHR5). Thus, in one embodiment diagnostic devices of the invention detect alleles of CFH only, CFHR5 only, CFH and CFHR5 only, or CFH, CFHR5 and up to 20, preferably up to 10, or preferably up to 5 genes other than CFH and/or CFHR5. That is, the device is particularly suited to screening for AMD and related complement-associated diseases. In one embodiment, the device comprises primers or probes that recognize CFH and/or one or more of CFHR1-5 only. In a related embodiment, the device contains primers and probes for up to 20, preferably up to 10, or preferably up to 5 other genes than CFH or CFHR1-5.
[0147] In one embodiment, the immobilized primer(s) is/are an allele-specific primerfs) that can distinguish between alleles at a polymorphic site in the Factor H or CHRF5 gene. Exemplary allelespecific primers to identify alleles at polymorphic sites in the Factor H gene are shown in TABLE 16A of U.S. patent publication No. 20070020647, incorporated by reference in its entirety for all purposes. The immobilized allele-specific primers hybridize preferentially to nucleic acids, either RNA or DNA, that have sequences complementary to the primers. The hybridization may be detected by various methods, including single-base extension with fluorescence detection, the oligonucleotide ligation assay, and the like (see Shi, M.M., 2001, Enabling large-scale pharmacogenetic studies by high-throughput mutation detection and genotyping technologies Clin. Chem. 47(2):164-172). Microarray-based devices to detect polymorphic sites are commercially available, including Affymetrix (Santa Clara, CA), Protogene (Menlo Park, CA), Genometrix (The Woodland, TX), Motorola BioChip Systems (Northbrook, IL), and Perlegen Sciences (Mountain View, CA).
[0148] The invention provides reagents and kits for detecting CFHR1 and/or CFHR3 proteins.
A number of assay systems are known in the art, and it is within the skill of the art to arrive at means to determine the presence or absence of CFHR1 and/or CFHR3, or variant or truncated forms thereof, associated with vascular disorders or AMD. The kit reagents, such as anti-CFHR3 or CFHR1 antibodies or other CFHR3 or CFHR1 binding moieties, may be contained in separate containers prior to their use for diagnosis or screening. In an embodiment, the kit contains a first container containing an antibody or binding moiety that specifically binds to CFHR1 protein, or a variant or truncated form thereof, and a second container containing an antibody or binding moiety that specifically binds to CFHR3 protein, or a variant or truncated form thereof. In some embodiments the binding moieties is an aptamer, such as a nucleic acid aptamer. Aptamers are RNA or DNA molecules selected in vitro from vast populations of
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 random sequence that recognize specific ligands by forming binding pockets. Aptamers are nucleic acids that are capable of three dimensional recognition that bind specific proteins or other molecules. See, e.g., US20050176940 Aptamers and Antiaptamers.
[0149] Thus, the invention provides reagents for conducting the screening methods of the invention, comprising a binding moiety capable of specifically binding CFHRl and/or CFHR3 protein or a portion thereof (e.g., a labeled binder that reacts preferentially with CFHRl and/or CFHR3 protein or a portion thereof or a labeled binder that reacts preferentially with CFHRl mRNA and/or CFHR3 mRNA or a portion thereof, or a labeled binder that reacts preferentially with CFHRl DNA and/or CFHR3 DNA). The binding moiety may comprise, for example, a member of a ligand-receptor pair, i.e., a pair of molecules capable of having a specific binding interaction (such as antibody-antigen, protein-protein, nucleic acid-nucleic acid, protein-nucleic acid, or other specific binding pair known in the art).
Optionally the binding moiety is labled (e.g., directly labeled) or is accompanied by a labeled molecule that reacts with the binding moiety (indirectly labeled). Detectable labels can be directly attached to or incorporated into the detection reagent by chemical or recombinant methods. Examples of detectable labels include, but are not limited to, radioisotopes, fluorophores, chromophores (e.g., colored particles), mass labels, electron dense particles, magnetic particles, spin labels, and molecules that emit chemiluminescence. Methods for labeling are well known in the art.
[0150] The kits may contain an instruction manual with instructions how to use the anti-CFHR3 or CFHRl antibodies or other CFHR3 or CFHRl binding moieties to detect CFHR3 or CFHRl proteins in body fluids or in tissue samples.
[0151] The kits may contain a control antibody or binding moiety. An example of a control antibody or binding moiety is an antibody that specifically binds to CFH protein.
[0152] The kits may contain one or more pairs of antibodies or binding moieties that specifically bind to different (i.e., not wild-type or full-length) forms (e.g., variant or truncated) of CFHRl or CFHR3 proteins.
[0153] In one embodiment, the antibodies or binding moieties are immobilized to a solid support such as an ordered array.
[0154] In one embodiment, the antibodies or binding moieties are used in Western blots.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
EXAMPLES
Example 1:
[0155] Polymerase chain reaction (PCR) amplification, single-strand conformation polymorphism (SSCP) analysis and direct DNA sequencing were used to characterize a deletion in the CFHR3 and CFHR1 genes located between the CFH and CFHR4 genes on chromosome 1. Examples of primers that can be used for PCR amplification of the CFT/gene and CF/7-related genes 1 to 5 are shown in Table 1 A. Examples of primers that can be used for SSCP analysis of the CFH and CFHR3 genes are shown in Table IB. Examples of primers that can be used for direct DNA sequencing of the CFH, CFHR1 and CFHR3 genes are shown in Table 1C and ID.
WO 2008/008986
PCT/US2007/073514
Ό
Ο
CM α
ζ co
CM
CCM o
CM
Ό
O
CM
C/0 <L>
a <D o
!
a £
.g *+-» o
<υ aS
Q i-4 £
Ό ω
o z
Θ ol w
o o
o
H
O
O
H
O
H
O <
O <
o
H
O
H
O'
W
OO o
z
Θ d
w o
to §
Θ o
W uo ω
a •c (X o
-s
H x
&
o
X o
z o£ ' O' ω 00
O H O < O < o o rl a
c x
to •S §
£ <D d
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 co (Z) ·< cn kg < S3 m
ό z
σ w
(Z) o
<
u u
H o
<
u k
o £
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 [0156] In a study directed toward further characterization of CFH and its associated haplotypes on chromosome lq, a complete deletion of the entire CFHL1 and CFHL3 genes was identified. In examining SSCP gels generated using CFH exon 22 primers (Table 1), several additional patterns of variation were observed due to the amplification of CFHR1 in addition to CFH. By designing another set of CFH-specific primers, it was determined that there were no variations in exon 22 of CFH. CFHR1specific primers were generated and used to identify a deletion of CFHR1. Further analysis of the CFHR1, CFHR2, CFHR3, CFHR4 and CFHR5 genes and intervening sequence 5' to CFHR3 (Table ID) using specific primers revealed a deletion that extends across the entire length of the CFHR1 and CFHR3 genes. The precise boundaries of the complete deletion have not be determined, but the mapping of the boundaries is within the skill of the art.
[0157] SSCP analysis and direct DNA sequencing was used to determine the frequency of the homozygous deletion of the CFHR3 and CFHR1 genes in a set of 1074 patients with and without a clinical history of AMD. The cohort included patients who had other systemic diseases, including vascular diseases, irrespective of their AMD status. As shown in Table 2, homozygous deletion of the CFHR1 and CFHR3 genes was found in -2.7% of the persons tested.
Table 2. Frequency of homozygous deletion of CFHR1 and CFHR3 genes
Genotype* Count Percent
+/+, +/Δ 1046 97.3%
Δ/Δ 28 2.7%
+/+, +/Δ, Δ/Δ 1074 100%
*Genotype refers to the deletion (Δ) or non-deletion (+) of the CFHR1 and CFHR3 genes by SSCP analysis and direct sequencing.
[0158] Initial analysis suggested that the deletion homozygotes were more common in control individuals than in AMD cases. To determine whether there was an association of the homozygous deletion of the CFHR3 and CFHR1 genes with AMD, a subset of the above patient population was analyzed by SSCP analysis and direct DNA sequencing. As shown in Table 3, in a study of 576 AMD patients and 352 age-matched non-AMD control patients, deletion homozygotes make up 5.1% of controls and 1.2% of cases. The homozygous deletion of CFHR1 and CFHR3 is strongly associated with controls, with χ2 = 10.2 and P value = 0.0014, demonstrating a highly significant protective effect of the homozygous CFHR1/CFHR3 deletion for AMD.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
Table 3. Association of homozygous deletion of CFHRl and CFHR3 genes with non-AMD
Genotype Non-AMD patients AMD patients
Count +/+, +/Δ 352 576
Count Δ/Δ 18 7
Frequency +/+, +/Δ 0.951 0.988
Frequency Δ/Δ 0.049 0.012
*Genotype refers to the deletion (Δ) or non-deletion (+) of the CFHRl and CFHR3 genes by SSCP analysis and direct sequencing.
[0159] To determine whether there was an association of the homozygous deletion of the
CFHR3 and CFHRl genes with vascular disorders, two subsets of the above patient population were analyzed by SSCP analysis and direct DNA sequencing. As shown in Table 4A, a study of 26 patients with abdominal aortic aneurysm (AAA) and 133 non-AAA patients revealed that the homozygous deletion of CFHRl and CFHR3 was strongly associated with AAA, with χ2 = 6.982329 and P = 0.0082. As shown in Table 4B, a second study of 86 patients with abdominal aortic aneurysm (AAA) and 221 non-AAA patients revealed that the homozygous deletion of CFHRl and CFHR3 was associated with AAA, with χ2 = 4.05 and P = 0.0442.
Table 4. Association of homozygous deletion of CFHRl and CFHR3 genes with AAA A. Study 1
Genotype Controls AAA
Count +/+, +/Δ 126 19
Count Δ/Δ 7 7
Total +/+, +/Δ, Δ/Δ 133 26
*Genotype refers to the deletion (Δ) or non-deletion (+) of the CFHRl and CFHR3 genes by SSCP analysis and direct sequencing.
B. Study 2
Genotype Controls AAA
Count +/+, +/Δ 221 86
Count Δ/Δ 12 11
Total +/+, +/Δ, Δ/Δ 233 97
*Genotype refers to the deletion (Δ) or non-deletion (+) of the CFHRl and CFHR3 genes by SSCP analysis and direct sequencing.
[0160] To determine whether previously identified protective haplotypes in the CFH gene were associated with the del (Δ) CFHRl allele, haplotype analysis was performed. As shown in Tables 5A-5E, the relationship between the del (Δ) CFHRl allele and SNPs in the CFH gene revealed strong linkage
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016 disequilibrium. The SNPs used in this haplotype analysis are described in U.S. patent publication No. 20070020647. In the table, letters refer to genotypes and numbers refer to SSCP shift patterns.
Table 5. CFH gene haplotype analysis in subjects with the del/del (Δ/Δ) CFHR1 allele A. Promoter 1 to Exon 3
Promoter 1 Promoter 4 Exon 2 Exon 3 a Exon 3
rs3753394 rs800292 same SNP as 3a
I62V
C-257T G184A IVS2-18insTT
1 AA TT GG SS SS
2 AA CC GG SS SS
3 AA CT GG SS SS
4 AA CC GG SS + GlOORhet SS + GlOORhet
5 AA CT GG SS SS
6 AA CT GG SS SS
7 AA CC GG SS SS
8 AA TT GG SS SS
9 AA CT GG SS SS
10 AA CC GG SS SS
11 AA CC GG SS SS
12 AA CC GG SS SS
13 AA CT GG SS SS
14 GG SS
15 GG SS
16 GG SS
17 GG SS
18 GG SS
19 GG SS
20 GG SS
21 GA SS
22 GG SS
23 SS
24 SS
25 SS
26 SS
B. IVS 6 to Exon 7b
IVS 6 IVS 6 IVS6 IVS6 Exon 7b
shift N or Del rs 16840419 rs3766404 rsl061147
A307A
A921C
1 3 NN GA CT CC
2 5 NDel 5 (GG) 5 (CC?) CC
3 2 NN GG CC CC
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
IVS6 IVS6 IVS6 IVS6 Exon 7b
shift N or Del rsl 6840419 rs3766404 rsl061147
4 2 NN GG CC CC
5 3 NN GA CT CC
6 1 NN AA TT AC
7 5 NDel 5 (GG) 5 (CC?) CC
8 1 NN AA TT CC
9 3 NN GA CT CC
10 2 NN GG CC CC
11 No DNA (3) NN No DNA (GA) No DNA (CT) AC
12 2 NN GG CC CC
13 1 NN AA TT CC
14
15
16
17
18 AA
19
20
21
22
23
24
25
26
C. Exon 9 to Exon 16b
Exon 9 Exon 10A Exon 10a Exon 13b Exon 16b
rsl061170 rs2274700 rs3753396 rs375046
Y402H A473A Q672Q IVS15
C1204T CFHtrunc G2016A A2089G
1 TT 1 AA AA
2 TT AA AA
3 TT 1 AA AA AA
4 TT 1 AA AA AA
5 TT 1 AA AA 4
6 CT 1 GA AA
7 TT AA
8 TT 1 AA AA AA
9 TT 1 AA AA AA
10 TT 1 AA AA AC?
11 CT 1 GA AA
12 TT AA
13 TT
14 TT 1 AA
15 TT 1 AA
16 CT 1 GA
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
Exon 9 Exon 10A Exon 10a Exon 13b Exon 16b
rslO61170 rs2274700 rs3753396 rs3 75046
Y402H A473A Q672Q I VS 15
C1204T CFHtrunc G2016A A2089G
17 TT 1 AA AA CC
18 CC 1 GG
19 TT 1 AA
20 TT 1 AA
21 TT 1 AA
22 TT 1 AA
23 TT
24 TT
25 TT
26 TT
D. Exon 17a to Exon 19a
Exon 17a Exon 17b Exon 18a Exon 18b Exon 19a
rslO65489 rs 1065489 rs534399
A892V E936D E936D V1007L
C2748T G2881T G2881T G3092T
1 1 CC GG GG GG
2 1 CC
3 1 CC GG GG GG
4 1 CC GG GG GG
5 1 CC GG GG GG
6 3 CC GG GG TT
7 1
8 1 CC GG GG GG
9 1 CC GG GG GG
10 1 CC GG GG GG
11 1 CC GG GG GG
12 1 CC
13 1 CC
14 GG
15 GG
16 GG
17 GG
18 1 CC GG GG GG
19 GG
20 GG
21 GG
22 GG
23 GG
24 GG
25 GG
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
Exon 17a Exon 17b Exon 18a Exon 18b Exon 19a
rsl065489 rsl065489 rs534399
A892V E936D E936D V1007L
C2748T G2881T G2881T G3092T
26 GG
E. Exon 20b to Exon22 split (detects both CFH and CFHR1)
Exon 20b Exon 22b Exon 22split
1191/1197/1210 1197
1 4 4 4
2 4
3 4 4 4
4 2 4 4
5 4 4 4
6 4 4 4
7 4
8 4 4 4
9 4 4 4
10 6 4 4
11 4 4 4
12 4
13 4
14 4
15 4 4
16 4 4
17 4
18 4 4
19 4 4
20 6 4
21 4 4
22 4 4
23 4 4
24 4 4
25 4 4
26 4 4
[0161] As shown in Table 6, in two studies it was found that the deletion of the CFHR1 and
CFHR3 genes was associated with 402T-containing haplotypes. This deletion is almost never found on the same 402C-containing haplotype as the major CFH risk allele, Y402H. The del (Δ) CFHR1 mutation is predominantly associated with the CT77H4 haplotype, a haplotype with T at position 1277 of the coding region of CFH (codon 402) shown previously shown to be protective for AMD. However, not every del (Δ) CFHR1 chromosome is on H4, and the protection of del/del (Δ/Δ) CFHR1 homozygotes for
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
AMD is even stronger than H4 homozygotes. Heterozygous deletion of the CFHR3 and CFHR1 genes was detected by direct DNA sequencing of the CFH, CFHR1 and CFHR3 genes using a CFH exon 22 primer.
Table 6. Association of homozygous deletion of CFHRl and CFHR3 genes with the TT genotype at position 1277 of the coding region of CFH (codon 402)
A. Study 1
CFH402 Genotype
Genotype TT TC CC
Count +/+, +/Δ 102 209 150
Count Δ/Δ 11 2 0
Count +/+, +/Δ, Δ/Δ 113 211 150
*Genotype refers to the deletion (Δ) or non-deletion (+) of the CFHRl and CFHR3 genes by SSCP analysis and direct sequencing.
**CFH402 Genotype refers to the nucleotide on both alleles at position 1277 of the coding region of human CFH. A T results in a tyrosine at codon 402, whereas a C results in a histidine at codon 402. ***Of the 474 patients, approximately 22+/-4% are heterozygous (+/Δ) for the deletion of the CFHRl and CFHR3 genes, as determined by direct DNA sequencing.
B. Study 2
CFH402 Genotype
Genotype TT TC CC
Count +/+, +/Δ 192 393 283
Count Δ/Δ 23 3 0
Count +/+, +/Δ, Δ/Δ 215 396 283
*Genotype refers to the deletion (Δ) or non-deletion (+) of the CFHRl and CFHR3 genes by SSCP analysis and direct sequencing.
**CFH402 Genotype refers to the nucleotide on both alleles at position 1277 of the coding region of human CFH. A T results in a tyrosine at codon 402, whereas a C results in a histidine at codon 402.
[0162] By Western blotting, it was determined that CFHRl protein, normally an abundant serum protein, is absent in sera derived from individuals homozygous for the CFHRlICFHR3 deletion. Figure 3 shows a representative Western blot of serum proteins from seven (out of a sample set of 52) patients using an anti-human CFH antibody. Serum proteins were separated by one-dimensional SDSpolyacrylamide gel electrophoresis and transferred to a nitrocellulose membrane. After transfer, the membrane was blocked with 5% non-fat dry milk, washed, and then incubated with a goat anti-human CFH (Calbiochem, 1:1000 dilution). After incubation, the membrane was washed, and then incubated
WO 2008/008986
PCT/US2007/073514
2016204271 23Jun2016 with horse radish peroxidase-conjugated rabbit anti-goat Ig antibody (Abeam, 1:4000 dilution). After incubation, the membrane was washed, and then incubated with extravidin (1:1500 dilution). Samples 197-02 and 325-02 were from patients with a TT 402 genotype (protective CFH H4 haplotype) and have homozygous deletion of CFHR1 and CFHR3 genes, as determined by SSCP analysis and direct sequencing. Figure 3 shows that no CFHR1 is detected in the serum from patients having a homozygous deletion of the CFHR1 and CFHR3 genes.
[0163] Western blotting using the same anti-human CFH antibody was used to detect CFH and
CFHR1 in serum from an additional 40 patients, separated according to SSCP patterns using the CFH exon 22 primers. Patterns 1-3 correspond to homozygous, or heterozygous for, non-deletion of CFHR1 and CFHR3 (+/+, +/Δ), and pattern 4 corresponds to homozygous deletion of CFHRl and CFHR3 (Δ/Δ) (see Figure 4). All 10 of the serum samples from patients displaying SSCP pattern 4 show no CFHR1, whereas all 30 of the serum samples from patients displaying SSCP patterns 1-3 show at least some CFHR1 (data not shown). Thus, analysis of serum from individuals with a CFHR1 del/del (Δ/Δ) genotype shows that they lack any detectable CFHR1 protein. This protein analysis confirms that these individuals lack both the CFHR1 gene and encoded protein. Individuals who are heterozygous for deletion of CFHR1 and CFHR3 can be recognized by protein analysis of serum samples by virtue of the intensity of the band corresponding to CFHR1 being roughly half the intensity in heterozygous (+/Δ) patients as compared to homozygous non-deletion (+/+) patients.
[0164] PCR experiments using leukocyte-derived DNA were performed to confirm that patients having a homozygous deletion of CFHR1 and CFHR3 do not have CFHR1 and CFHR3 DNA. Figure 5 shows a PCR analysis of CFH and CFHR1-5 from DNA samples from 20 patients, separated into four groups according to SSCP patterns using the CFH exon 22 primers mentioned above. Patterns 1-3 correspond to homozygous non-deletion or heterozygous deletion of CFHR1 and CFHR3 (+/+, +/Δ), and pattern 4 corresponds to homozygous deletion of CFHRl and CFHR3 (Δ/Δ). From left to right, 5 samples each from patients displaying SSCP patterns 1, 2, 3 and 4 were subjected to PCR using primers specific for CFH, CFHRl, CFHR2, CFHR3, CFHR4 and CFHR5, as indicated. This figure shows that CFH, CFHR4 and CFHR5 DNA are amplified in all of the samples, whereas CFHRl and CFHR3 DNA are amplified in samples from patients displaying SSCP patterns 1-3, but not from patients displaying SSCP pattern 4. The CFHR2 DNA was amplified in some, but not all, of the samples. Thus, when SSCP and direct sequencing show a homozygous deletion of the CFHRl and CFHR3 genes, no PCR amplifiable CFHRl and CFHR3 DNA are detected in samples.
WO 2008/008986
PCT/US2007/073514
2016204271 23 Jun 2016
Example 2: Production of Anti-CFHRl and Anti-CFHR3 Monoclonal Antibodies [0165] Mice will be immunized with recombinant human CFHR1 or CFHR3. Two mice with sera displaying the highest anti-CFHRl and anti-CFHR3 activity by Enzyme Linked Immunosorbent Assay (ELISA) will be chosen for subsequent fusion and spleens and lymph nodes from the appropriate mice will be harvested. B-cells will be harvested and fused with an myeloma line. Fusion products will be serially diluted on one or more plates to near clonality. Supernatants from the resulting fusions will be screened for their binding to hCFHRl or hCFHR3 by ELISA. Supernatants identified as containing antibodies to CFHR1 or CFHR3 will be further characterized by in vitro functional testing as discussed below. A panel of hybridomas will be selected and the hybridomas will be subcloned and expanded. The monoclonal antibodies will then be purified by affinity chromatography on Protein A/G resin under standard conditions.
[0166] Anti-CFHRl and anti-CFHR3 antibodies may be further characterized by in vitro functional testing using complement activation assays well known in the art. For example, complement activation assays may be conducted in solution (e.g., fluid phase in blood) or on immobilized surfaces. Exemplary assays may measure the ability of the anti-CFHRl and/or anti-CFHR3 antibodies to block or reduce CFH, C3b, heparin and/or C-reactive protein (CRP) binding to a substrate.
*** [0167] Although the present invention has been described in detail with reference to specific embodiments, those of skill in the art will recognize that modifications and improvements are within the scope and spirit of the invention, as set forth in the claims which follow. All publications and patent documents cited herein are incorporated herein by reference as if each such publication or document was specifically and individually indicated to be incorporated herein by reference. Citation of publications and patent documents (patents, published patent applications, and unpublished patent applications) is not intended as an admission that any such document is pertinent prior art, nor does it constitute any admission as to the contents or date of the same. The invention having now been described by way of written description, those of skill in the art will recognize that the invention can be practiced in a variety of embodiments and that the foregoing description is for purposes of illustration and not limitation of the following claims.
2016204271 07 Mar 2018

Claims (15)

  1. WHAT IS CLAIMED IS:
    1. An antibody that reduces the activity or amount of a gene product of the CFHR1 and/or CFHR3 genes, when used in the treatment of a subject having or at risk of developing AMD.
  2. 2. Use of an antibody that reduces the activity or amount of a gene product of the CFHR1 and/or CFHR3 genes in the manufacture of a medicament for the treatment of a subject having or at risk of developing AMD.
  3. 3. The antibody of Claim 1 or the use of Claim 2, wherein the antibody is an antibody to a CFHR1 protein.
  4. 4. The antibody or use of Claim 3, wherein said antibody binds the aminoterminus of a CFHR1 polypeptide.
  5. 5. The antibody or use of Claim 3, wherein said treatment further comprises administering an antibody to a CFHR3 protein.
  6. 6. The antibody of Claim 1 or the use of Claim 2, wherein the antibody is an antibody to a CFHR3 protein.
  7. 7. The antibody or use of Claim 6, wherein said antibody binds the carboxylterminus of a CFHR3 polypeptide.
  8. 8. The antibody or use of Claim 6, wherein said treatment further comprises administering an antibody to a CFHR1 protein.
  9. 9. A pharmaceutical composition comprising an anti-CFHRl antibody that specifically binds to CFHR1; and a pharmaceutically acceptable carrier, wherein the antibody is monoclonal.
    2016204271 07 Mar 2018
  10. 10. The pharmaceutical composition of claim 9 wherein the antibody specifically binds an epitope between amino acids 1-143 of SEQ ID NO: 4 within the amino-terminus of a CFHRl polypeptide.
  11. 11. A pharmaceutical composition comprising an anti-CFHR3 antibody that specifically binds to CFHR3; and a pharmaceutically acceptable carrier wherein the antibody is monoclonal.
  12. 12. The pharmaceutical composition of claim 11 wherein the antibody specifically binds an epitope between amino acids 144-330 of SEQ ID NO: 6 within the carboxyl-terminus of a CFHR3 polypeptide.
  13. 13. A method of treating a subject having or at risk of developing AMD, wherein the method comprises administering to the subject the antibody of any one of claims 1 and 3-8, or the pharmaceutical composition of any one of claims 9-12.
    WO 2008/008986
    PCT/US2007/073514
    2016204271 23 Jun 2016 g
    'q <
    o ώ
    r {Ms{ ω
    —J co x X X c\ X X Ϊ X X X X X ft X X X X O y. O o o o
    1 /15 c
    φ o
    ία.
    x! a. o x o 5 0 5
    Φ c
    Φ u Ο φ <f w
    3 =
    X O
    WO 2008/008986
    PCT/US2007/073514
    2016204271 23 Jun 2016
    2/15
    CFH
    CFHR3 CFHR1 CFHR4 CFHR2 CFHR5 o
    ILLII_Hl ΠΙ Ι_LlilU_II_lllLI_U1 11 I IL III I II I
    I_J Li I_l l_I (_I l_l I U I_I
    B A* C D B' A C’ D’ B D*‘
    50kb
    A
    FIG. 2
    WO 2008/008986
    PCT/US2007/073514
    2016204271 23 Jun 2016
    3/15
    CT) 00 OO cn oo 00 CD ro CD 00 00 1 O 1 cn 1 1 o 1 o 1 O 1 o cn O O cn cn cn N) ho
    FIG. 3
    WO 2008/008986
    PCT/US2007/073514
    4/15
    2016204271 23 Jun 2016
    2 4 13
    FIG. 4
    WO 2008/008986
    PCT/US2007/073514
    5/15
    2016204271 23 Jun 2016
    FIG. 5
    WO 2008/008986
    PCT/US2007/073514
    6/15
    2016204271 23 Jun 2016
    H Factor AA seq 1 CFHR1 AA seq. 1 CHFR3 AA seq. 1
    H Factor AA seq 51 CFHR1 AA seq. 1 CHFR3 AA seq. 1
    H Factor AA seq 101 CFHR1 AA seq. 1 CHFR3 AA seq. 1
    H Factor AA seq 151 CFHR1 AA seq. 1 CHFR3 AA seq. 1
    H Factor AA seq 201 CFHR1 AA seq. 5 CHFR3 AA seq. 5
    H Factor AA seq 251 CFHR1 AA seq. 16 CHFR3 AA seq. 16
    H Factor AA seq 301 CFHR1 AA seq. 23 CHFR3 AA seq. 23
    H Factor AA seq 351 CFHR1 AA seq. 49 CHFR3 AA seq. 49
    H Factor AA seq 401 CFHR1 AA seq. 99 CHFR3 AA seq. 99
    H Factor AA seq 450 CFHR1 AA seq. 139 CHFR3 AA seq. 140
    H Factor AA seq 500 CFHR1 AA seq. 139 CHFR3 AA seq. 140
    H Factor AA seq 550 CFHR1 AA seq. 139 CHFR3 AA seq. 140
    MRLLAKIICLMLWAICVAEDCNELPPRRNTEILTGSWSDQTYPEGTQAIY
    KCRPGYRSLGNVIMVCRKGEWVALNPLRKCQKRPCGHPGDTPFGTFTLTG
    GNVFEYGVKAVYTCNEGYQLLGEINYRECDTDGWTNDIPICEWKCLPVT
    APENGKIVSSAMEPDREYHFGQAVRFVCNSGYKIEGDEEMHCSDDGFWSK
    MWLL
    MLLL
    EKPKCVEISCKSPDVINGSPISQKIIYKENERFQYKCNMGYEYSERGDAV
    VS------------VILISRISS
    IN------------VILTLWVSC ★ * *
    CTESGWRPLPSCEEKSCDNPYIPNGDYSPLRIKHRTGDEITYQCRNGFYP
    -----------------------------------VGGEATF------------------------------------------ANGQVKP-------ATRGNTAKCTSTGWIPAPRCTLKPCDYPDIKHGGLYHENMRRPYFPVAVG
    ------------------------CDFPKINHGILYDEEKYKPFSQVPTG
    ------------------------CDFPDIKHGGLFHENMRRPYFPVAVG ★★*★*★* * ★ * *
    KYYSYYCDEHFETPSGSYWDHIHCTQDGWSPAVPCLRKCYFPYLENGYNQ
    EVFYYSCEYNFVSPSKSFWTRITCTEEGWSPTPKCLRLCFFPFVENGHSE
    KYYSYYCDEHFETPSGSYWDYIHCTQNGWSPAVPCLRKCYFPYLENGYNQ ** ★ * * * * * ★ ★ ★ ★ Jr i
    NHGRKFVQGKSIDVACHPGYALPKA-QTTVTCMENGWSPTPRCIRVKTCS
    S SGQTHLEGDTVQIICNTGYRLQNN-ENNISCVERGWSTPP--------NYGRKFVQGNSTEVACHPGYGLPKVRQTTVTCTENGWS PTP--------k k k k k k .....k k k k k k
    KSSIDIENGFISESQYTYALKEKAKYQCKLGYVTADGETSGSIRCGKDGW
    SAQPTCIKSCDIPVFMNARTKNDFTWFKLNDTLDYECHDGYESNTGSTTG
    SIVCGYNGWSDLPICYERECELPKIDVHLVPDRKKDQYKVGEVLKFSCKP
    100
    150
    200
    250
    300
    350
    400
    449
    138
    139
    499
    138
    139
    549
    138
    139
    599
    138
    139
    FIG. 6
    WO 2008/008986
    PCT/US2007/073514
    7/15
    2016204271 23 Jun 2016
    H Factor AA seq CFHR1 AA seq. CHFR3 AA seq. 600 139 140 H Factor AA seq 650 CFHR1 AA seq. 139 CHFR3 AA seq. 140 H Factor AA seq 700 CFHR1 AA seq. 139 CHFR3 AA seq. 140 H Factor AA seq 750 CFHR1 AA seq. 139 CHFR3 AA seq. 140 H Factor AA seq 800 CFHR1 AA seq. 144 CHFR3 AA seq. 144 H Factor AA seq 850 CFHR1 AA seq. 144 CHFR3 AA seq. 144 H Factor AA seq 900 CFHR1 AA seq. 144 CHFR3 AA seq. 144 H Factor AA seq 950 CFHR1 AA seq. 144 CHFR3 AA seq. 144 H Factor AA seq 1000 CFHR1 AA seq. 144 CHFR3 AA seq. 144 H Factor AA seq 1050 CFHR1 AA seq. 149 CHFR3 AA seq. 149
    GFTIVGPNSVQCYHFGLSPDLPICKEQVQSCGPPPELLNGNVKEKTKEEY 649
    -------------------------------------------------- 138
    -------------------------------------------------- 139
    GHSEWEYYCNPRFLMKGPNKIQCVDGEWTTLPVCIVEESTCGDIPELEH 699
    -------------------------------------------------- 138
    -------------------------------------------------- 139
    GWAQLSSPPYYYGDSVEFNCSESFTMIGHRSITCIHGVWTQLPQCVAIDK 749
    -------------------------------------------------- 138
    -------------------------------------------------- 139
    LKKCKSSNLIILEEHLKNKKEFDHNSNIRYRCRGKEGWIHTVCINGRWDP 799
    --KCRST------------------------------------------- 143
    --RCIR-------------------------------------------- 143 *
    EVNCSMAQIQLCPPPPQIPNSHNMTTTLNYRDGEKVSVLCQENYLIQEGE 849
    -------------------------------------------------- 143
    -------------------------------------------------- 143
    EITCKDGRWQSIPLCVEKIPCSQPPQIEHGTINSSRSSQESYAHGTKLSY 899
    -------------------------------------------------- 143
    -------------------------------------------------- 143
    TCEGGFRISEENETTCYMGKWSSPPQCEGLPCKSPPEISHGWAHMSDSY 949
    -------------------------------------------------- 143
    -------------------------------------------------- 143
    QYGEEVTYKCFEGFGIDGPAIAKCLGEKWSHPPSCIKTDCLSLPSFENAI 999
    -------------------------------------------------- 143
    -------------------------------------------------- 143
    PMGEKKDVYKAGEQVTYTCATYYKMDGASNVTCINSRWTGRPTCRDTSCV 1049
    ---------------------------------------------DTSCV 148
    ---------------------------------------------DRTCS 148 * . ★
    NPPTVQNAYIVSRQMSKYPSGERVRYQCRSPYEMFGDEE---VMCLNGNW 1096
    NPPTVQNAYIVSRQMSKYPSGERVRYQCRSPYEMFGDEE---VMCLNGNW 195
    KS DIEIENGFISESSSIYILNKEIQYKCKPGYATADGNS SGSITCLRNGW 198 * * * * * * * * *
    H Factor AA seq 1097 CFHR1 AA seq. 196 CHFR3 AA seq. 199
    H Factor AA seq 1147 CFHR1 AA seq. 246 CHFR3 AA seq. 249
    H Factor AA seq 1197 CFHR1 AA seq. 296 CHFR3 AA seq. 299
    TEPPQCKDSTGKCGPPPPIDNGDITSFPLSVYAPASSVEYQCQNLYQLEG 1146 TEPPQCKDSTGKCGPPPPIDNGDITSFPLSVYAPASSVEYQCQNLYQLEG 245 SAQPICINSSEKCGPPPPISNGDTTSFLLKVYVPQSRVEYQCQSYYELQG 248 , 4c 4c 4c , 4;4e4c4c4c4e4c4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4e 4c 4c 4c 4c 4c 4c 4c 4c 4e 4c
    NKRITCRNGQWSEPPKCLHPCVISREIMENYNIALRWTAKQKLYSRTGES 1196 NKRITCRNGQWS EP P KCLHP CVISREIMENYNIALRWTAKQKLYLRTGES 295 SNYVTCSNGEWSAPPRCIHPCIITEENMNKNNIKLKGRSDRKYYAKTGDT 298 . 4c 4c 4c 4c . 4c 4c 4c4c^4e 4e4e4c^4e^ 4c 4c 4c 4c 4c 4e 4e 4c 4c
    VEFVCKRGYRLSSRSHTLRTTCWDGKLEYPTCAKR 1231
    AEFVCKRGYRLSSRSHTLRTTCWDGKLEYPTCAKR 330
    IEFMCKLGYNANTSILSFQAVCREGIVEYPRCE 331
    4c 4c 4c 4c 4c 4e 4c 4c 4c 4c 4c 4c
    WO 2008/008986
    PCT/US2007/073514
    8/15
    2016204271 23 Jun 2016
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    1 ATGAGACTTCTAGCAAAGATTATTTGCCTTATGTTATGGGCTATTTGTGT 50 1 0 1 0
    51 AGCAGAAGATTGCAATGAACTTCCTCCAAGAAGAAATACAGAAATTCTGA 100 1 0
    1 0
    101 CAGGTTCCTGGTCTGACCAAACATATCCAGAAGGCACCCAGGCTATCTAT 150 1 0 1 ' 0
    151 AAATGCCGCCCTGGATATAGATCTCTTGGAAATGTAATAATGGTATGCAG 200 1 0
    1 0
    201 GAAGGGAGAATGGGTTGCTCTTAATCCATTAAGGAAATGTCAGAAAAGGC 250 1 0
    1 0
    251 CCTGTGGACATCCTGGAGATACTCCTTTTGGTACTTTTACCCTTACAGGA 300 1 0
    1 0
    301 GGAAATGTGTTTGAATATGGTGTAAAAGCTGTGTATACATGTAATGAGGG 350 1 0
    1 0
    351 GTATCAATTGCTAGGTGAGATTAATTACCGTGAATGTGACACAGATGGAT 400 1 0
    1 0
    401 GGACCAATGATATTCCTATATGTGAAGTTGTGAAGTGTTTACCAGTGACA 450 1 0
    1 0
    451 GCACCAGAGAATGGAAAAATTGTCAGTAGTGCAATGGAACCAGATCGGGA 500 1 0
    1 0
    501 ATACCATTTTGGACAAGCAGTACGGTTTGTATGTAACTCAGGCTACAAGA 550 1 0
    1 0
    H Factor mRNA
    CFHR1 mRNA
    CFHR3 mRNA
    H Factor mRNA
    CFHR1 mRNA
    CFHR3 mRNA
    FIG. 7
    WO 2008/008986
    PCT/US2007/073514
    9/15
    2016204271 23 Jun 2016
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    H Factor mRNA CFHRl mRNA CFHR3 mRNA
    551 TTGAAGGAGATGAAGAAATGCATTGTTCAGACGATGGTTTTTGGAGTAAA 600
    601 GAGAAACCAAAGTGTGTGGAAATTTCATGCAAATCCCCAGATGTTATAAA 650
    651 TGGATCTCCTATATCTCAGAAGATTATTTATAAGGAGAATGAACGATTTC 700
    701 AATATAAATGTAACATGGGTTATGAATACAGTGAAAGAGGAGATGCTGTA 750
    751 TGCACTGAATCTGGATGGCGTCCGTTGCCTTCATGTGAAGAAAAATCATG 800
    801 TGATAATCCTTATATTCCAAATGGTGACTACTCACCTTTAAGGATTAAAC 850
    851 ACAGAACTGGAGATGAAATCACGTACCAGTGTAGAAATGGTTTTTATCCT 900
    901 GCAACCCGGGGAAATACAGCCAAATGCACAAGTACTGGCTGGATACCTGC 950
    951 TCCGAGATGTACCTTGAAACCTTGTGATTATCCAGACATTAAACATGGAG 1000 1 0
    1 0
    1001 GTCTATATCATGAGAATATGCGTAGACCATACTTTCCAGTAGCTGTAGGA 1050 1 0
    1 0
    1051 AAATATTACTCCTATTACTGTGATGAACATTTTGAGACTCCGTCAGGAAG 1100 1 0
    1 0
    H Factor mRNA
    CFHRl mRNA
    CFHR3 mRNA
    H Factor mRNA
    CFHRl mRNA
    CFHR3 mRNA
    WO 2008/008986
    PCT/US2007/073514
    10/15
    2016204271 23 Jun 2016
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    1101 TTACTGGGATCACATTCATTGCACACAAGATGGATGGTCGCCAGCAGTAC 1150 1 0
    1 0
    1151 CATGCCTCAGAAAATGTTATTTTCCTTATTTGGAAAATGGATATAATCAA 1200 1 0
    1 0
    1201 AATCATGGAAGAAAGTTTGTACAGGGTAAATCTATAGACGTTGCCTGCCA 1250 1 0
    1 0
    1251 TCCTGGCTACGCTCTTCCAAAAGCGCAGACCACAGTTACATGTATGGAGA 1300 1 0
    1 0
    1301 ATGGCTGGTCTCCTACTCCCAGATGCATCCGTGTCAAAACATGTTCCAAA 1350 1 0
    1 0
    1351 TCAAGTATAGATATTGAGAATGGGTTTATTTCTGAATCTCAGTATACATA 1400 1 0
    1 0
    1401 TGCCTTAAAAGAAAAAGCGAAATATCAATGCAAACTAGGATATGTAACAG 1450 1 0
    1 0
    1451 CAGATGGTGAAACATCAGGATCAATTAGATGTGGGAAAGATGGATGGTCA 1500 1 0
    1 0
    1501 GCTCAACCCACGTGCATTAAATCTTGTGATATCCCAGTATTTATGAATGC 1550 1 0
    1 0
    1551 CAGAACTAAAAATGACTTCACATGGTTTAAGCTGAATGACACATTGGACT 1600 1 0
    1 0
    1601 ATGAATGCCATGATGGTTATGAAAGCAATACTGGAAGCACCACTGGTTCC 1650 1 0
    1 0
    H Factor mRNA
    CFHR1 mRNA
    CFHR3 mRNA
    H Factor mRNA
    CFHR1 mRNA
    CFHR3 mRNA
    WO 2008/008986
    PCT/US2007/073514
    11 /15
    2016204271 23 Jun 2016
    H Factor mRNA 1651 ATAGTGTGTGGTTACAATGGTTGGTCTGATTTACCCATATGTTATGAAAG 1700 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 1701 AGAATGCGAACTTCCTAAAATAGATGTACACTTAGTTCCTGATCGCAAGA 1750 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 1751 AAGACCAGTATAAAGTTGGAGAGGTGTTGAAATTCTCCTGCAAACCAGGA 1800 CFHRl mRNA , . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 1801 TTTACAATAGTTGGACCTAATTCCGTTCAGTGCTACCACTTTGGATTGTC 1850 CFHRl mRNA , , 1 0 CFHR3 mRNA 1 0 H Factor mRNA 1851 TCCTGACCTCCCAATATGTAAAGAGCAAGTACAATCATGTGGTCCACCTC 1900 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 1901 CTGAACTCCTCAATGGGAATGTTAAGGAAAAAACGAAAGAAGAATATGGA 1950 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 1951 CACAGTGAAGTGGTGGAATATTATTGCAATCCTAGATTTCTAATGAAGGG 2000 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2001 ACCTAATAAAATTCAATGTGTTGATGGAGAGTGGACAACTTTACCAGTGT 2050 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2051 GTATTGTGGAGGAGAGTACCTGTGGAGATATACCTGAACTTGAACATGGC 2100 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2101 TGGGCCCAGCTTTCTTCCCCTCCTTATTACTATGGAGATTCAGTGGAATT 2150 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2151 CAATTGCTCAGAATCATTTACAATGATTGGACACAGATCAATTACGTGTA 2200 CFHRl mRNA . 1 0 CFHR3 mRNA 1 0
    WO 2008/008986
    PCT/US2007/073514
    12/15
    2016204271 23 Jun 2016
    H Factor mRNA 2201 TTCATGGAGTATGGACCCAACTTCCCCAGTGTGTGGCAATAGATAAACTT 2250 CFHR1 mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2251 AAGAAGTGCAAATCATCAAATTTAATTATACTTGAGGAACATTTAAAAAA 2300 CFHR1 mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2301 CAAGAAGGAATTCGATCATAATTCTAACATAAGGTACAGATGTAGAGGAA 2350 CFHR1 mRNA . . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2351 AAGAAGGATGGATACACACAGTCTGCATAAATGGAAGATGGGATCCAGAA 2400 CFHR1 mRNA . , 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2401 GTGAACTGCTCAATGGCACAAATACAATTATGCCCACCTCCACCTCAGAT 2450 CFHR1 mRNA . . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2451 TCCCAATTCTCACAATATGACAACCACACTGAATTATCGGGATGGAGAAA 2500 CFHR1 mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2501 AAGTATCTGTTCTTTGCCAAGAAAATTATCTAATTCAGGAAGGAGAAGAA 2550 CFHR1 mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2551 ATTACATGCAAAGATGGAAGATGGCAGTCAATACCACTCTGTGTTGAAAA 2600 CFHR1 mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2601 AATTCCATGTTCACAACCACCTCAGATAGAACACGGAACCATTAATTCAT 2650 CFHR1 mRNA . 1 0 CFHR3 mRNA 1 0 H Factor mRNA 2651 CCAGGTCTTCACAAGAAAGTTATGCACATGGGACTAAATTGAGTTATACT 2700 CFHR1 mRNA . 1 ATGTGGCTCCTGGTCAGTGTAATTCTAAT 29 CFHR3 mRNA 1 ATGTTGTTACTAATCAATGTCATTCTGAC k k k k k k k k k k 29 H Factor mRNA 2701 TGTGAGGGTGGTTTCAGGATATCTGAAGAAAATGAAACAACATGCTACAT 2750 CFHR1 mRNA . 30 CTCACGGATATCCTCTG-----TTGGGGGAGAAGCAACATTTTGTGATTT 74 CFHR3 mRNA 30 CTTGTGGGTTTCCTGTG-----CTAATGGACAAGTGAAACCTTGTGATTT 74
    ★ Jr * * * * * * k k
    WO 2008/008986
    PCT/US2007/073514
    13/15
    2016204271 23 Jun 2016
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    2751 GGGAAAATGGAGTTCTCCACCTCAGTGTGAAGGCCTTCCTTGTAAATCTC 2800 75 TCCAAAAATAAACCATGGAATTCTATATGATGAAGAAAAATATAAGCCAT 124 75 TCCAGACATTAAACATGGAGGTCTATTTCATGAGAATATGCGTAGACCAT 124 i it * * * * * *
    2801 CACCTGAGATTTCTCATGGTGTTGTAGCTCACATGTCAGACAGTTATCAG 2850
    12 5 TTTCCCAGGTTCCTACAGGGGAAGTTTTCTA------------TTACTCC 162
    12 5 ACTTTCCAGTAGCTGTAGGAAAATATTACTC------------CTATTAC 162 * * * * * it ie
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    2851 TATGGAGAAGAAGTTACGTACAAATGTTTTGAAGGTTTTGGAATTGATGG 2900
    163 TGTGAATATAATTTTGTGT----CTCCTTCAAAATCATT-------TTGG 2 01
    163 TGTGATGAACATTTTGAGA----CTCCGTCAGGAAGTTA-------CTGG 201 * * * * * * * * * * * ***
    2901 GCCTGCAATTGCAAAATGCTTAGGAGAAAAATGGTCTCACCCTCCATCAT 2950
    202 ACTCGCA----TAACATGCACAGAAGAAGGATGGTCACCAACACCAAAGT 247
    202 GATTACA----TTCATTGCACACAAAATGGGTGGTCACCAGCAGTACCAT 247 ** ****** ****** * * *
    2951 GCATAAAAACAGATTGTCTCAGTTTACCTAGCTTTGAAAATGCCATACCC 3000
    248 GTCT-----CAGACTGTGTTTCTTTCCTTTTGTGGAAAATGGTCATTCTG 292
    248 GTCT-----CAGAAAATGTTATTTTCCTTATTTGGAAAATGGATATAATC 292 * * ** * * * * ** * * * * *** * **
    3001 ATGGGAGAGAAGAAGGATGTGTATAAGGCGGG--TGAGCAAGTGACTTAC 3048 293 AATCTTCAGGACAAACACATCTGGAAGGTGATACTGTGCAAATTATTTGC 342 293 AAAATTATGGAAGAAAGTTTGTACAGGGTAACTCTACAGAAGTTGCCTGC 342 * * * * * * * ** * ** * * *
    3049 A-CTTGTGCAACATATTACAAAATGGATGGAGCCAGTAATGTAACATGCA 3097
    3 43 AACACAGGATACAGACTTCAAAACAATGAGAAC----AACATTTCATGTG 3 88
    343 CATCCTGGCTACGGTCTTCCAAAAGTCCGTCAG-ACCACAGTTACATGTA 391 *** ***** * *****
    3098 TTAATAGCAGATGGACAGGAAGGCCAACATGCAGA------GACACCTCC 3141
    389 TAGAACGGGGCTGGTCCACCCCTCCCAAATGCAGGTCCACTGACACTTCC 438 392 CGGAGAATGGCTGGTCTCCTACTCCCAGATGCATCC---GAGACAGAACA 438 * * * * * * ** * ***** **** *
    3142 TGTGTGAATCCGCCCACAGTACAAAATGCTTATATAGTGTCGAGACAGAT 3191 439 TGTGTGAATCCGCCCACAGTACAAAATGCTTATATAGTGTCGAGACAGAT 488 439 TGCTCAAAATCAGATATAGAAATTGAAAATGGATTCATTTCTGAATCTTC 488 ** *** **** * * **** *
    3192 GAGTAAATATCCATCTGGTGAGAGAGTACGTTATCAATGTAGGAGCCCTT 3241 489 GAGTAAATATCCATCTGGTGAGAGAGTACGTTATCAATGTAGGAGCCCTT 538 489 CTCTATTTATATTTTAAATAAAGAAATACAATA.TAAATGTAAACCAGGAT 538 ** *** * ** **** ********* *
    3 242 ATGAAATGTT---TGGGGAT------GAAGAAGTGATGTGTTTAAATGGA 3 2 82
    53 9 ATGAAATGTT---TGGGGAT------GAAGAAGTGATGTGTTTAAATGGA 579
    53 9 ATGCAACAGCAGATGGAAATTCTTCAGGATCAATTACATGTTTGCGAAAT 58 8 ***** *** ** ** *** *****
    WO 2008/008986
    PCT/US2007/073514
  14. 14/15
    2016204271 23 Jun 2016
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA
    3283 AACTGGACGGAACCACCTCAATGCAAAGATTCTACAGGAAAATGTGGGCC 3332 580 AACTGGACGGAACCACCTCAATGCAAAGATTCTACGGGAAAATGTGGGCC 629 589 GGATGGTCAGCACAACCAATTTGCATTAATTCTTCAGAAAAGTGTGGACC 638 ★ *★ ★ ★ ★★ ★★★ ★ ★ ii ★ ★ ★ *★*
    3333 CCCTCCACCTATTGACAATGGGGACATTACTTCATTCCCGTTGTCAGTAT 3382 630 CCCTCCACCTATTGACAATGGGGACATTACTTCATTCCCGTTGTCAGTAT 679 639 TCCTCCACCTATTAGCAATGGTGATACCACCTCCTTTCTACTAAAAGTGT 688 ★ *★ ★ ★* ★★ ★i *
    3383 ATGCTCCAGCTTCATCAGTTGAGTACCAATGCCAGAACTTGTATCAACTT 3432 680 ATGCTCCAGCTTCATCAGTTGAGTACCAATGCCAGAACTTGTATCAACTT 729 689 ATGTGCCACAGTCAAGAGTCGAGTACCAATGCCAGTCCTACTATGAACTT 738
    4e ★ ★ •it 4c ★ 4c 4c 4c ★★★ ★ + 4c 4c 4c 4e 4c 4c 4c 4c 4e 4c
    3433 GAGGGTAACAAGCGAATAACATGTAGAAATGGACAATGGTCAGAACCACC 3482 73 0 GAGGGTAACAAGCGAATAACATGTAGAAATGGACAATGGTCAGAACCACC 77 9 739 CAGGGTTCTAATTATGTAACATGTAGTAATGGAGAGTGGTCGGCACCACC 788
    4c 4e 4e 4c 4c 4e 4e 4c4c4e4c4e4e4c4e4e4e 4c 4e 4c 4c 4c 4c 4c 4e 4e 4e 4c 4c 4e 4c 4c 4e 4c 4c 4c
    3483 AAAATGCTTACATCCGTGTGTAATATCCCGAGAAATTATGGAAAATTATA 3532 780 AAAATGCTTACATCCGTGTGTAATATCCCGAGAAATTATGGAAAATTATA 829 789 TAGATGCATACATCCATGTATAATAACTGAAGAAAACATGAATAAAAATA 838
    4c 4e 4c 4c 4c 4e 4e 4e 4c 4e 4e 4e 4e 4e 4c 4c 4c 4c 4c 4c 4e 4c 4e 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c
    3533 ACATAGCATTAAGGTGGACAGCCAAACAGAAGCTTTATTCGAGAACAGGT 3582 830 ACATAGCATTAAGGTGGACAGCCAAACAGAAGCTTTATTTGAGAACAGGT 879 839 ACATAAAGTTAAAAGGAAGAAGTGACAGAAAATATTATGCAAAAACAGGG 888
    4e 4c 4c 4c 4c 4c 4c 4c 4e 4e 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c
    3 583 GAATCAGTTGAATTTGTGTGTAAACGGGGATATCGTCTTTCATCACGTTC 3 632 880 GAATCAGCTGAATTTGTGTGTAAACGGGGATATCGTCTTTCATCACGTTC 929 889 GATACCATTGAATTTATGTGTAAATTGGGATATAATGCAAATACATCAAT 938
    4c 4c 4c ★★*★*** 4c4c4c4c4c4c4c4c 4c 4c 4e 4c 4c 4c 4c 4c 4c 4c
    3633 TCACACATTGCGAACAACATGTTGGGATGGGAAACTGGAGTATCCAACTT 3682 930 TCACACATTGCGAACAACATGTTGGGATGGGAAACTGGAGTATCCAACTT 979 939 TCTATCATTTCAAGCAGTGTGTCGGGAAGGGATAGTGGAATACCCCAGAT 988
    4c 4c 4c 4e 4c 4c 4c 4c 4e 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4e 4c 4c 4c
    3683 GTGCAAAAAGATAGAATCAAT-CATAAAGTGCACACCTTTATTCAGAACT 3731 980 GTGCAAAAAGATAGAATCAAT-CATAAAATGCACACCTTTATTCAGAACT 1028 989 GCG-AATAAGGCAGCATTGTTACCCTAAATGTATGTCCAACTTCCACTTT 1037 ★ 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c
    3 73 2 TTAGTATTAAATCAGTTCTCAATTTCATTTTTTATGTATTGTTTTACTCC 3 781 1029 TTAGTATTAAATCAGTTCTTAATTTAATTTTTAA-GTATTGTTTTACTCC 1077 1038 TCCACTTCTCACTCTTATGGTCTCAAAGCTTGCAAAGATAGCTTCTGATA 1087
    4e 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c
    3782 TTTTTATTCATACGTAAAATTTTGGATTAATTTGTGAAAATGTAATTATA 3831 1078 TTTTTATTCATACGTAAAATTTTGGATTAATTTGTGAAAATGTAATTATA 112 7 1088 TTGTTGTA-ATTTCTACTTTATTTCAAAGAAAATTAATATAATAGTTTCA 1136
    4c 4c 4c 4c 4c 4c 4c 4e 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c 4c
    3832 AGCTGAGACCGGTGGCTCTCTT 3853
    1128 AGCTGAGACCGGTGGCTCTCTTCTTAAAAGCACCATATTAAAACTTGGAA 1177
    WO 2008/008986
    PCT/US2007/073514
    2016204271 23 Jun 2016
  15. 15/15
    CFHR3 mRNA
    H Factor mRNA CFHR1 mRNA CFHR3 mRNA
    H Factor mRNA CFHL1 mRNA CFHL3 mRNA
    1137 ATTTGCAAC---TTAATATATTCTCAAAAATATATTAAAACAAACTAAAT 1183 * Mt Mr ★ Mr Mr ★ ★ Mr Me
    3854 3853
    1178 AACTGGAAAACT 1189
    1184 TATTGCTTATGCTTGTACTAAAATAATAAAAACTACTCTTATAAAAAAAA 1233
    3854 3853 1190 1189 1234 AAAAAAAAAAAAA 1246
AU2016204271A 2006-07-13 2016-06-23 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration Ceased AU2016204271B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2016204271A AU2016204271B2 (en) 2006-07-13 2016-06-23 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration
AU2018206776A AU2018206776A1 (en) 2006-07-13 2018-07-19 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US60/831,018 2006-07-13
US60/840,073 2006-08-23
AU2007272335A AU2007272335B2 (en) 2006-07-13 2007-07-13 Methods and reagents for treatment and diagnosis of vascular disorders and age-related macular degeneration
AU2013202401A AU2013202401A1 (en) 2006-07-13 2013-04-04 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration
AU2016204271A AU2016204271B2 (en) 2006-07-13 2016-06-23 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2013202401A Division AU2013202401A1 (en) 2006-07-13 2013-04-04 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2018206776A Division AU2018206776A1 (en) 2006-07-13 2018-07-19 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration

Publications (2)

Publication Number Publication Date
AU2016204271A1 AU2016204271A1 (en) 2016-07-07
AU2016204271B2 true AU2016204271B2 (en) 2018-04-19

Family

ID=56551053

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2016204271A Ceased AU2016204271B2 (en) 2006-07-13 2016-06-23 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration
AU2018206776A Abandoned AU2018206776A1 (en) 2006-07-13 2018-07-19 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU2018206776A Abandoned AU2018206776A1 (en) 2006-07-13 2018-07-19 Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration

Country Status (1)

Country Link
AU (2) AU2016204271B2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006062716A2 (en) * 2004-11-18 2006-06-15 Yale University Methods and compositions for treating ocular disorders
WO2007144621A2 (en) * 2006-06-13 2007-12-21 The Queen's University Of Belfast Protection against and treatment of age related macular degeneration

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006062716A2 (en) * 2004-11-18 2006-06-15 Yale University Methods and compositions for treating ocular disorders
WO2007144621A2 (en) * 2006-06-13 2007-12-21 The Queen's University Of Belfast Protection against and treatment of age related macular degeneration

Also Published As

Publication number Publication date
AU2018206776A1 (en) 2018-08-09
AU2016204271A1 (en) 2016-07-07

Similar Documents

Publication Publication Date Title
US20180371547A1 (en) Methods and Reagents for Treatment and Diagnosis of Vascular Disorders and Age-Related Macular Degeneration
US11168120B2 (en) Methods and reagents for treatment of age-related macular degeneration
EP2851432B1 (en) RCA locus analysis to assess susceptibility to AMD
WO2008118969A2 (en) Methods and agents for evaluating inflammatory bowel disease, and targets for treatment
AU2016204271B2 (en) Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration
AU2013202401A1 (en) Methods and reagents for the treatment and diagnosis of vascular disorders and age-related macular degeneration
AU2013203231A1 (en) RCA locus analysis to assess susceptibility to AMD and MPGNII

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired