WO2000068266A1 - Protease de proteine precurseur amyloide et composes d'acides nucleiques apparentes - Google Patents

Protease de proteine precurseur amyloide et composes d'acides nucleiques apparentes Download PDF

Info

Publication number
WO2000068266A1
WO2000068266A1 PCT/US2000/006707 US0006707W WO0068266A1 WO 2000068266 A1 WO2000068266 A1 WO 2000068266A1 US 0006707 W US0006707 W US 0006707W WO 0068266 A1 WO0068266 A1 WO 0068266A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
leu
val
ala
gly
Prior art date
Application number
PCT/US2000/006707
Other languages
English (en)
Inventor
Gerald Wayne Becker
John Edward Hale
William Francis Heath, Jr.
Edward Marion Johnstone
Sheila Parks Little
Yuan Tu
Wu-Kuang Yeh
Tinggui Yin
Original Assignee
Eli Lilly And Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eli Lilly And Company filed Critical Eli Lilly And Company
Priority to AU46406/00A priority Critical patent/AU4640600A/en
Publication of WO2000068266A1 publication Critical patent/WO2000068266A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6478Aspartic endopeptidases (3.4.23)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to proteases which cleave amyloid precursor protein and facilitate ⁇ -amyloid peptide release, and accordingly, have utility in identifying compounds which treat or prevent Alzheimer's Disease.
  • Alzheimer's disease is a degenerative brain disorder characterized clinically by progressive loss of memory, cognition, reasoning, judgment, and emotional stability that gradually leads to profound mental deterioration and ultimately death.
  • Alzheimer's disease is a very common cause of progressive mental failure (dementia) in aged humans and is believed to represent the fourth most common medical cause of death in the United States.
  • Alzheimer's disease has been observed in races and ethnic groups worldwide and presents a major present and future public health problem. The disease is currently estimated to affect about two to three million individuals in the United States alone. -Alzheimer's disease is at present incurable. No treatment that effectively prevents Alzheimer's disease or reverses its symptoms and course is currently known.
  • the brains of individuals with Alzheimer's disease exhibit characteristic lesions termed senile (or amyloid) plaques, amyloid angiopathy (amyloid deposits in blood vessels) and neurofibrillary tangles.
  • senile or amyloid
  • amyloid angiopathy amyloid deposits in blood vessels
  • neurofibrillary tangles Large numbers of these lesions, particularly amyloid plaques and neurofibrillary tangles, are generally found in several areas of the human brain important for memory and cognitive function in patients with Alzheimer's disease. Smaller numbers of these lesions in a more restrictive anatomical distribution are also found in the brains of most aged humans who do not have clinical Alzheimer's disease.
  • Amyloid plaques and amyloid angiopathy also characterize the brains of individuals with Trisomy 21 (Down's syndrome) and Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-Type (HCHWA-D).
  • amyloid angiopathy The principal chemical constituent of the amyloid plaques and vascular amyloid deposits (amyloid angiopathy), characteristic of Alzheimer's disease and the other disorders mentioned above, is an approximately 4.2 kilodalton (kD) protein of about 39-43 amino acids designated the ⁇ -amyloid peptide (this protein is also referred to in the literature as ⁇ AP, A ⁇ , A ⁇ P, A- beta, or ⁇ /A4).
  • ⁇ -Amyloid peptide was first purified and a partial amino acid sequence was provided by Glenner, et al.. Biochemical and Biophysical Research Communications, 120:885-890 (1984). The isolation procedure and the sequence data for the first 28 amino acids are described in United States Patent 4,666,829, the entire contents of which are herein incorporated by reference.
  • ⁇ -amyloid peptide is a small fragment of a much larger precursor protein, the amyloid precursor protein (APP), that is normally produced by cells in many tissues of various animals, including humans.
  • APP amyloid precursor protein
  • Knowledge of the structure of the gene encoding the amyloid precursor protein has demonstrated that the ⁇ -amyloid peptide arises as a peptide fragment that is cleaved from the amyloid precursor protein by proteases.
  • a normal (i.e., non-pathogenic) processing of the amyloid precursor protein occurs via cleavage by a putative " -secretase” which cleaves between amino acids 16 and 17 of the ⁇ -amyloid peptide region within the protein. Kang, et al., Nature (London), 325:773-776 (1987). It is further believed that pathogenic processing occurs in part via an enzyme designated " ⁇ -secretase" which cleaves at the amino-terminus of the ⁇ -amyloid peptide region within the precursor protein.
  • the identification, isolation, and purification of the proteases involved in the processing of amyloid precursor protein would permit chemical modeling of a critical event in the pathology of Alzheimer's disease and would allow the screening of compounds to determine their ability to inhibit formation of ⁇ -amyloid peptide.
  • the treatment methods would advantageously be based on drugs which are capable of inhibiting ⁇ -amyloid peptide release and/or its synthesis in vivo.
  • the present invention provides an isolated protein useful as an amyloid precursor protein protease, said compound comprising the amino acid sequence
  • SEQ ID NO:2 525 530 535 hereinafter referred to as SEQ ID NO:2.
  • amyloid precursor protein protease comprising the amino acid sequence Met Leu Arg Arg Arg Gly Ser Pro Gly 1 5
  • SEQ ID NO:4 525 530 535 hereinafter referred to as SEQ ID NO:4.
  • the invention also provides isolated nucleic acid compounds that comprise nucleic acid sequences which encode the amino acid compounds provided.
  • the isolated nucleic acid compounds that are provided are preferably DNA, however, nucleic acid compounds which are sense or antisense mRNA are also provided.
  • a particularly preferred nucleic acid compound is the DNA compound comprising the sequence
  • cag ctg gtg cac age ttt get gag ggc cag gac cag ggc age gcc tat 291 Gin Leu Val His Ser Phe Ala Glu Gly Gin Asp Gin Gly Ser Ala Tyr 75 80 85
  • gtg gcc get ccc tac teg aag ccc age atg ace ctg gag ccc aac aag 483 Val Ala Ala Pro Tyr Ser Lys Pro Ser Met Thr Leu Glu Pro Asn Lys 140 145 150
  • ctg act ggc aac gtg ace aeg teg cag atg gcc aac gag cag ggc ttg 627
  • aac etc ate tgg cag ctg aca gac ace aaa cag ctg gtg cac agt ttc 915 Asn Leu He Trp Gin Leu Thr Asp Thr Lys Gin Leu Val His Ser Phe 285 290 295
  • an even more preferred isolated nucleic acid compound is the DNA compound having the sequence agctgtcagc cgcctcacag gaag atg ctg cgt egg egg ggc age cct ggc 51
  • cag ctg gtg cac age ttt get gag ggc cag gac cag ggc age gcc tat 291
  • ggc tac cct gag get gag gtg ttc tgg cag gat ggg cag ggt gtg ccc 579 Gly Tyr Pro Glu Ala Glu Val Phe Trp Gin Asp Gly Gin Gly Val Pro 170 175 180 185
  • ctg act ggc aac gtg ace aeg teg cag atg gcc aac gag cag ggc ttg 627 Leu Thr Gly Asn Val Thr Thr Ser Gin Met Ala Asn Glu Gin Gly Leu
  • SEQ ID NO:3 SEQ ID NO:3.
  • nucleic acid vectors comprising nucleic acids which encode SEQ ID NO:2 or SEQ ID NO:4 or functional equivalents thereof.
  • the preferred nucleic acid vectors are those which are DNA.
  • DNA vectors of the present invention preferably comprise a promoter positioned to drive expression of said DNA sequence. Those vectors wherein said promoter functions in human embryonic kidney cells (293 cells), AV12 cells, yeast cells or E.coli cells are preferred.
  • the present invention also provides probes and primers useful for molecular biology techniques.
  • a compound which encodes all or part of SEQ ID NO:2 or SEQ ID NO:4 and which is at least 18 consecutive base pairs in length is provided.
  • the 18 base pair or more compound is DNA.
  • Most preferred for this use are the DNA compounds which comprise at least 18 consecutive base pairs of SEQ ID NO:l or SEQ ID NO:3.
  • a preferred host cell is an oocyte.
  • a preferred oocyte is one which has been injected with sense mRNA or DNA compounds of the present invention.
  • a more preferred oocyte is one which has been injected with sense mRNA or DNA compounds of the present invention in conjunction with DNA or sense mRNA which encodes APP.
  • this invention provides cells into which the nucleic acid compounds of the present invention may be transfected.
  • Host cells include those which are transfected with a nucleic acid compound which encodes SEQ ID NO:2 or SEQ ID NO:4.
  • Preferred cells include host cells transfected with a DNA vector comprising SEQ ID NO:l or SEQ ID NO:3.
  • the preferred transfected host cells which encode SEQ ID NO:2 or SEQ ID NO:4 are 293 cells, AV12 cells, yeast cells and E. coli cells.
  • the invention provides a method for identifying DNA homologous to a probe of the present invention, which comprises combining test nucleic acid with the probe under hybridizing conditions and identifying those test nucleic acids which hybridize.
  • Assays utilizing the compounds provided by the present invention are also provided.
  • the assays provided determine whether a substance is a ligand for ⁇ -secretase, said method comprising contacting ⁇ -secretase with said substance, monitoring ⁇ -secretase activity by physically detectable means, and identifying those substances which interact with or affect ⁇ -secretase.
  • Preferred assays of the present invention include a cell culture assay, a high-performance liquid chromotography (HPLC) assay or a synthetic competition assay.
  • Pre- ferred cell culture assays utilize oocytes, AN12, E. coli, yeast or 293 cells which co-express nucleic acids which encode ⁇ -secretase and APP.
  • the invention also provides methods for constructing a host cell capable of expressing a nucleic acid compound which encodes an amino acid compound comprising SEQ ID ⁇ O:2 or SEQ ID NO:4, said methods comprising transfecting a host cell with a DNA vector comprising a nucleic acid compound encoding SEQ ID NO:2 or SEQ ID NO:4.
  • a preferred method utilizes 293, AV12, yeast or E. coli cells as the host cells.
  • a more preferred method includes a DNA vector which comprises SEQ ID NO:l or SEQ ID NO:3.
  • a most preferred method includes a DNA vector which comprises SEQ ID NO:3.
  • Another preferred method comprises (a) a DNA vector which comprises SEQ ID NO: 3 and (b) a DNA expression vector which encodes the APP coding sequence.
  • methods for expressing a nucleic acid sequence which encodes SEQ ID NO:2 or SEQ ID NO:4, or functional equivalents thereof , in a transfected host cell comprise culturing a transfected host cell of the present invention under conditions suitable for gene expression.
  • a preferred method utilizes 293, AV12, yeast or E. coli cells as the transfected host cell.
  • a more preferred method utilizes a DNA vector to transfect the host cell.
  • a most preferred method utilizes a DNA vector comprising all or part of SEQ ID NO:3.
  • the present invention also provides processes for isolating and purifying an amyloid precursor protein protease, said process comprising: (a) establishing in a suitable medium, a culture of the host cells transformed with a polynucleotide encoding an amyloid precursor protein protease; and (b) isolating said protease from said culture.
  • compositions comprising a peptide isolated according to such processes are also provided.
  • polyclonal and monoclonal antibodies to the peptides of the present invention are also provided.
  • Base pair refers to DNA or RNA.
  • the abbreviations A, C, G, and T correspond to the 5'-monophosphate forms of the deoxyribonucleosides (deoxy)adenosine, (deoxy)cytidine, (deoxy)guanosine, and (deoxy)thymidine, respectively, when they occur in DNA molecules.
  • the abbreviations U, C, G, and A correspond to the 5'-monophosphate forms of the ribonucleosides uridine, cytidine, guanosine, and adenosine, respectively when they occur in RNA molecules.
  • base pair may refer to a partnership of A with T or C with G.
  • heteroduplex base pair may refer to a partnership of A with U or C with G. (See the definition of "complementary”, infra.)
  • restriction refers to the catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA ("sequence-specific endonucleases”).
  • restriction enzymes used herein are commercially available and their reaction conditions, cofactors, and other requirements were used as would be known to one of ordinary skill in the art. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer or can be readily found in the literature.
  • Ligation refers to the process of forming phosphodiester bonds between two double stranded nucleic acid fragments. Unless otherwise provided, ligation may be accomplished using known buffers and conditions with a DNA ligase, such as T4 DNA ligase.
  • plasmid refers to an extrachromosomal (usually) self- replicating genetic element. Plasmids are generally designated by a lower case “p” preceded and/or followed by letters and/or numbers.
  • the starting plasmids herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids in accordance with published procedures. In addition, equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.
  • reading frame means the nucleotide sequence from which translation occurs “read” in triplets by the translational apparatus of transfer RNA (tRNA) and ribosomes and associated factors, each triplet corresponding to a particular amino acid. To insure against improper translation, the triplet codons corresponding to the desired polypeptide must be aligned in multiples of three from the initiation codon, i.e. the correct "reading frame” being maintained.
  • Recombinant DNA cloning vector refers to any autonomously replicating agent, including, but not limited to, plasmids and phages, comprising a DNA molecule to which one or more additional DNA segments can or have been added.
  • recombinant DNA expression vector refers to any recombinant DNA cloning vector in which a promoter to control transcription of the inserted DNA has been incorporated.
  • expression vector system refers to a recombinant DNA expression vector in combination with one or more trans- acting factors that specifically influence transcription, stability, or replication of the recombinant DNA expression vector.
  • the trans-acting factor may be expressed from a co-transfected plasmid, virus, or other extrachromosomal element, or may be expressed from a gene integrated within the chromosome.
  • Transcription refers to the process whereby information contained in a nucleotide sequence of DNA is transferred to a complementary RNA sequence.
  • transfection refers to the taking up of an expression vector by a host cell whether or not any coding sequences are in fact expressed. Numerous methods of transfection are known to the ordinarily skilled artisan, for example, calcium phosphate co-precipitation, and electroporation. Successful transfection is generally recognized when any indication of the operation of this vector occurs within the host cell.
  • transformation means the introduction of DNA into an organism so that the DNA is replicable, either as an extrachromosomal element or by chromosomal integration.
  • Methods of transforming bacterial and eukaryotic hosts are well known in the art, many of which methods, such as nuclear injection, protoplast fusion or by calcium treatment using calcium chloride are summarized in J. Sambrook, et al.. MOLECULAR CLONING: A LABORATORY MANUAL, (1989).
  • transformation refers to the process whereby the genetic information of messenger RNA is used to specify and direct the synthesis of a polypeptide chain.
  • vector refers to a nucleic acid compound used for the transformation of cells in gene manipulation bearing polynucleotide sequences corresponding to appropriate protein molecules which when combined with appropriate control sequences confer specific properties on the host cell to be transformed. Plasmids, viruses, and bacteriophage are suitable vectors. Artificial vectors are constructed by cutting and joining DNA molecules from different sources using restriction enzymes and ligases.
  • vector as used herein includes Recombinant DNA cloning vectors and Recombinant DNA expression vectors.
  • complementary refers to the capacity of p urine and pyrimidine nucleotides to associate through hydrogen bonding to form double stranded nucleic acid molecules.
  • the following base pairs are related by complementarity: guanine and cytosine; adenine and thymine; and adenine and uracil.
  • complementary means that the aforementioned relationship applies to substantially all base pairs comprising two single-stranded nucleic acid molecules over the entire length of said molecules.
  • Partially complementary refers to the aforementioned relationship in which one of two single-stranded nucleic acid molecules is shorter in length than the other such that a portion of one of the molecules remains single-stranded.
  • hybridization refers to a process in which a strand of nucleic acid joins with a complementary strand through base pairing.
  • the conditions employed in the hybridization of two completely or nearly completely complementary nucleic acid strands varies with the degree of complementarity of the two strands and the length of the strands. Such techniques and conditions are well known to practitioners in this field.
  • isolated amino acid sequence refers to any amino acid sequence, however constructed or synthesized, which is locationally distinct from the naturally occurring sequence.
  • isolated DNA compound refers to any DNA sequence, however constructed or synthesized, which is locationally distinct from its natural location in genomic DNA.
  • isolated nucleic acid compound refers to any RNA or DNA sequence, however constructed or synthesized, which is locationally distinct from its natural location.
  • a "primer” is a nucleic acid fragment which functions as an initiating substrate for enzymatic or synthetic elongation.
  • promoter refers to a DNA sequence which directs transcription of DNA to RNA.
  • a “probe” as used herein is a nucleic acid compound or a fragment thereof which hybridizes with a nucleic acid compound which encodes either the entire sequence of SEQ ID NO:2 or SEQ ID NO:4, a sequence complementary to SEQ ID NO:2 or SEQ ID NO:4, or a part thereof.
  • EST or “expressed sequence tag” refers to a fragment or sampling of cDNA which encodes a polypeptide of unknown function.
  • stringency refers to a set of hybridization conditions which may be varied in order to vary the degree of nucleic acid affinity for other nucleic acid. (See the definition of "hybridization”, supra.)
  • telomere sequence refers to a sequence in which antibodies raised against an epitope of the proteins of the present invention, or a fragment thereof, may be used to differentiate between the proteins of the present invention and other ⁇ -secretase variants. This term may also be employed in the sense that such antibodies may be used to differentiate between the human ⁇ -secretase protein and analogous proteins derived from other species.
  • PCR refers to the widely-known polymerase chain reaction employing a thermally-stable polymerase. This technique, as appreciated by those skilled in the art, is employed to amplify a particular nucleic acid fragment.
  • BLAST refers to the widely known basic local alignment search tool.
  • This tool consists of a set of computer- based programs designed to permit examination of amino acid and nucleic acid sequence databases for similarity with an isolated sequence of interest.
  • RACE refers to the widely known rapid amplification of complimentary ends technique, to amplify and obtain the 5' and 3' ends of isolated cDNA.
  • proteins of the present invention can be synthesized by a number of different methods. All of the amino acid compounds of the invention can be made by chemical methods well known in the art, including solid phase peptide synthesis, or recombinant methods. Both methods are described in U.S. Patent 4,617,149, herein incorporated by reference.
  • peptides may be synthesized by solid-phase methodology utilizing an Applied Biosystems 430A peptide synthesizer (commercially available from Applied Biosystems, Foster City California) and synthesis cycles supplied by Applied Biosystems.
  • Protected amino acids such as t-butoxycarbonyl-protected amino acids, and other reagents are commercially available from many chemical supply houses. Sequential t-butoxycarbonyl chemistry using double couple protocols are applied to the starting /-.-methyl benzhydryl amine resins for the production of C-terminal carboxamides.
  • pyridine-2-aldoxime methiodide resin For the production of C-terminal acids , the corresponding pyridine-2-aldoxime methiodide resin is used. Asparagine, glutamine, and arginine are coupled using preformed hydroxy benzotriazole esters. The following side chain protection may be used:
  • the peptides may be deprotected and cleaved from the resin with anhydrous hydrogen fluoride containing 10% meta-cresol. Cleavage of the side chain protecting group(s) and of the peptide from the resin is carried out at zero degrees centigrade or below, preferably - 20°C for thirty minutes followed by thirty minutes at 0°C.
  • the proteins of the present invention may also be produced by recombinant methods. Recombinant methods are preferred if a high yield is desired.
  • a general method for the construction of any desired DNA sequence is provided in J. Brown, et al., Methods in Enzymology, 68:109 (1979). See also, J. Sambrook, et al., supra. The basic steps in the recombinant production of desired proteins are:
  • prokaryotes are used for cloning of DNA sequences in constructing the vectors of this invention.
  • Prokaryotes may also be employed in the production of the protein of interest.
  • the Escherichia coli K12 strain 294 ATCC No. 31446
  • Other strains of E. coli which may be used (and their relevant genotypes) include the following.
  • DH5a F- (j80dlacZDMl5), D(lacZYA-argF)U169 supE44, l-> hsdR17(r K -, m ⁇ + ), recAl, endAl, gyrA96, thi-1, relAl
  • strains are all commercially available from suppliers such as: Bethesda Research Laboratories, Gaithersburg, Maryland 20877 and Stratagene Cloning Systems, La Jolla, California 92037; or are readily available to the public from sources such as the American Type Culture
  • genotypes listed are illustrative of many of the desired characteristics for choosing a bacterial host and are not meant to limit the invention in any way.
  • the genotype designations are in accordance with standard nomenclature. See, for example, J. Sambrook, et al., supra.
  • bacilli such as Bacillus subtilis
  • enterobacteriaceae such as Salmonella tvphimurium or Serratia marcescans
  • various Pseudomonas species may be used.
  • other bacteria especially Streptomvces, spp., may be employed in the prokaryotic cloning and expression of the proteins of this invention.
  • Promoters suitable for use with prokaryotic hosts include the b- lactamase [vector pGX2907 (ATCC 39344) contains the replicon and b- lactamase gene] and lactose promoter systems [Chang et al.. Nature (London), 275:615 (1978); and Goeddel et al.. Nature (London), 281:544
  • alkaline phosphatase alkaline phosphatase
  • the tryptophan (trp) promoter system vector pATHl (ATCC 37695) is designed to facilitate expression of an open reading frame as a trpE fusion protein under control of the trp promoter] and hybrid promoters such as the tac promoter (isolatable from plasmid pDR540 ATCC- 37282).
  • trp tryptophan
  • vector pATHl ATCC 37695
  • hybrid promoters such as the tac promoter (isolatable from plasmid pDR540 ATCC- 37282).
  • other functional bacterial promoters whose nucleotide sequences are generally known, enable one of skill in the art to ligate them to DNA encoding the proteins of the instant invention using linkers or adapters to supply any required restriction sites. Promoters for use in bacterial systems also will contain a Shine -Dalgarno sequence operably linked to the DNA encoding the desired polypeptide
  • the proteins of this invention may be synthesized either by direct expression or as a fusion protein comprising the protein of interest as a translational fusion with another protein or peptide which may be removable by enzymatic or chemical cleavage. It is often observed in the production of certain peptides in recombinant systems that expression as a fusion protein prolongs the lifespan, increases the yield of the desired peptide, or provides a convenient means of purifying the protein of interest.
  • a variety of peptidases e.g. trypsin which cleave a polypeptide at specific sites or digest the peptides from the amino or carboxy termini (e.g. diaminopeptidase) of the peptide chain are known.
  • particular chemicals e.g.
  • cyanogen bromide will cleave a polypeptide chain at specific sites.
  • the skilled artisan will appreciate the modifications necessary to the amino acid sequence (and synthetic or semi-synthetic coding sequence if recombinant means are employed) to incorporate site-specific internal cleavage sites. See e.g., P. Carter, "Site Specific Proteolysis of Fusion Proteins", Chapter 13 in PROTEIN PURIFICATION: FROM MOLECULAR MECHANISMS TO LARGE SCALE PROCESSES, American Chemical Society, Washington, DC (1990).
  • the proteins of the present invention may also be produced in eukaryotic systems.
  • the present invention is not limited to use in a particular eukaryotic host cell.
  • a variety of eukaryotic host cells are available from depositories such as the American Type Culture Collection (ATCC) and are suitable for use with the vectors of the present invention.
  • ATCC American Type Culture Collection
  • the choice of a particular host cell depends to some extent on the particular expression vector used to drive expression of the amyloid precursor protein protease-encoding nucleic acids of the present invention.
  • Exemplary host cells suitable for use in the present invention are listed in Table I. Theses exemplary host cells are merely illustrative of the many eukaryotic cells available for use with the present invention and are not meant in any way to limit the scope of the present invention.
  • a preferred cell line employed in the expression of the protein of the present invention is the widely available 293 cell line. As noted, this cell line was constructed from human embryonal kidney tissue and is available from American Type Culture Collection under the accession number ATCC CCL 1573.
  • Some illustrative vectors include the pSV2-type vectors which comprise segments of the simian virus 40 (SN40) genome that constitute a defined eukaryotic transcription unit-promoter, intervening sequence, and polyadenylation site.
  • SN40 simian virus 40
  • the plasmid pSN2-type vectors transform mammalian and other eukaryotic host cells by integrating into the host cell chromosomal D ⁇ A.
  • Plasmid pSV2-type vectors such as plasmid pSN2-gpt, pSN2- neo, pSN2-dhfr, pSN2-hyg, and pSV2-b-globin, in which the SN40 promoter drives transcription of an inserted gene.
  • These vectors are suitable for use with the coding sequences of the present invention and are widely available from sources such as the ATCC or the Northern Regional Research Laboratory (NRRL), 1815 N. University Street, Peoria, Illinois, 61604.
  • the plasmid pSV2-dhfr (ATCC 37146) comprises a murine dihydrofolate reductase (dhfr) gene under the control of the SV40 early promoter. Under the appropriate conditions, the dhfr gene is known to be amplified, or copied, in the host chromosome. This amplification can result in the amplification of closely-associated DNA sequences and can, therefore, be used to increase production of a protein of interest. See, e.g., J. Schimke, Cell, 35:705-713 (1984). Plasmids constructed for expression of the proteins of the present invention in mammalian and other eukaryotic host cells can utilize a wide variety of promoters.
  • the present invention is in no way limited to the use of the particular promoters exemplified herein.
  • Promoters such as the SV40 late promoter, promoters from eukaryotic genes, such as, for example, the estrogen-inducible chicken ovalbumin gene, the interferon genes, the gluco-corticoid-inducible tyrosine aminotransferase gene, and the thymidine kinase gene, and the major early and late adenovirus genes can be readily isolated and modified to express the genes of the present invention.
  • Eukaryotic promoters can also be used in tandem to drive expression of a coding sequence of this invention.
  • Plasmid pRSVcat (ATCC 37152) comprises portions of a long terminal repeat of the Rous Sarcoma virus, a virus known to infect chickens and other host cells. This long terminal repeat contains a promoter which is suitable for use in the vectors of this invention. H. Gorman, et al., Proceedings of the National Academy of Sciences (USA). 79:6777 (1982).
  • the plasmid pMSNi (NRRL B- 15929) comprises the long terminal repeats of the Murine Sarcoma virus, a virus known to infect mouse and other host cells.
  • the mouse metallothionein promoter has also been well characterized for use in eukaryotic host cells and is suitable for use in the expression of the nucleic acids of the present invention.
  • the mouse metallothionein promoter is present in the plasmid pdBPN-MMTneo (ATCC 37224) which can serve as the starting material of other plasmids of the present invention.
  • Another usefull expression vector system employs one of a series of vectors containing the BK enhancer, an enhancer derived from the BK virus, a human papovavirus.
  • the most preferred such vector systems are those which employ not only the BK enhancer but also the adenovirus-2-early region IA (El A) gene product.
  • the E1A gene product (actually, the E1A gene produces two products, which are collectively referred to herein as "the E1A gene product”) is an immediate-early gene product of adenovirus, a large D ⁇ A virus.
  • An additional useful eukaryotic expression vector is the phd series of vectors which comprise a BK enhancer in tandem with the adenovirus late promoter to drive expression of useful products in eukaryotic host cells.
  • the construction and method of using the phd plasmid, as well as related plasmids, are described in U.S. Patents 5,242,688, issued September 7, 1993, and 4,992,373, issued February 12, 1991, all of which are herein incorporated by reference.
  • Escherichia coli K12 GM48 cells harboring the plasmid phd are available as part of the permanent stock collection of the Northern Regional Research Laboratory under accession number NRRL B- 18525. The plasmid may be isolated from this culture using standard techniques.
  • the plasmid phd contains a unique Bell site which may be utilized for the insertion of the gene encoding the protein of interest.
  • the skilled artisan understands that linkers or adapters may be employed in cloning the gene of interest into this Bell site.
  • the phd series of plasmids functions most efficiently when introduced into a host cell which produces the E1A gene product, cell lines such as AN12-664, 293 cells, and others, described supra.
  • Transformation of the mammalian cells can be performed by any of the known processes including, but not limited to, the protoplast fusion method, the calcium phosphate co-precipitation method, electroporation and the like. See, e.g., J. Sambrook, et al., supra, at 3:16.30-3:16.66.
  • viruses are also appropriate vectors.
  • the adenovirus, the adeno- associated virus, the vaccinia virus, the herpes virus, the baculovirus, and the rous sarcoma virus are useful.
  • Such a method is described in U.S. Patent 4,775,624, herein incorporated by reference.
  • Several alternate methods of expression are described in J. Sambrook, et al., supra, at 16.3-17.44.
  • eukaryotic microbes such as yeast cultures may also be used.
  • Saccharomyces cerevisiae or common baker's yeast, is the most commonly used eukaryotic microorganism, although a number of other strains are commonly available.
  • the plasmid YRp7 ATCC-40053
  • yeast cultures may also be used. See, e.g., L. Stinchcomb, et al., Nature (London), 282:39 (1979); J. Kingsman et al.. Gene, 7:141 (1979); S.
  • This plasmid already contains the trp_ gene which provides a selectable marker for a mutant strain of yeast lacking the ability to grow in tryptophan.
  • Suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase [found on plasmid pAP12BD (ATCC 53231) and described in U.S. Patent No. 4,935,350, issued June 19, 1990, herein incorporated by reference] or other glycolytic enzymes such as enolase [found on plasmid p AC 1 (ATCC 39532)], glyceraldehyde-3-phosphate dehydrogenase [derived from plasmid pHcGAPCl (ATCC 57090, 57091)], hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6- phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, p ho sp ho glucose isomerase, and glucokinase, as well as the alcohol dehydrogenase and pyru
  • yeast promoters which are inducible promoters, having the additional advantage of their transcription being controllable by varying growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein [contained on plasmid vector
  • P CL28XhoLHBPV (ATCC 39475) and described in United States Patent No. 4,840,896, herein incorporated by reference], glyceraldehyde 3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose [e.g. GAL1 found on plasmid pRY121 (ATCC 37658)] utilization.
  • Suitable vectors and promoters for use in yeast expression are further described in R.
  • Yeast enhancers such as the UAS Gal from Saccharomyces cerevisiae (found in conjunction with the CYC1 promoter on plasmid YEpsec-hllbeta ATCC 67024), also are advantageously used with yeast promoters.
  • Practitioners of this invention realize that, in addition to the above-mentioned expression systems, cloned cDNA may also be employed in the production of transgenic animals in which a test mammal, usually a mouse, in which expression or overexpression of the proteins of the present invention can be assessed.
  • the nucleic acids of the present invention may also be employed in the construction of "knockout" animals in which the expression of the native cognate of the gene is suppressed.
  • protease of the present invention can be isolated and purified from cultured host cells transiently expressing ⁇ - secretase activity.
  • the following examples further illustrate such isolation and purification processes, however, these examples are not in any way to be interpreted as limiting the scope of the present invention.
  • the enzymatic activity of beta-secretase was determined by an ELISA assay, modified in form over an assay appearing in application No. PCT/US96/09985, the entire content of which is herein incorporated by reference.
  • MBP-APPcl25sw as the substrate, the activity was determined by measuring the MBP-APPc26sw cleavage product with a polyclonal antibody, 192sw, highly specific to the product.
  • MBP-APPcl25sw is a fusion substrate in which maltose binding protein is fused to the 125- residue C-terminal portion of the Swedish amyloid precursor protein.
  • one picounit is defined as the amount of the enzyme that catalyzes the conversion of MBP-APP125sw to one picomole of MBP-APPc26sw per ml in 2-hr under the optimized reaction conditions of the Elisa assay (see Table II, infra).
  • the protein concentration was first determined using the well known Bio-Rad microassay method. (Rio-Rad Laboratories, Hercules, CA, 94547) Example 3 ⁇ -Secretase Purification
  • beta-secretase was carried out by a protocol modified over the purification methods appearing in application No. PCT/US96/09985, the entire content of which is herein incorporated by reference.
  • the enzyme purification protocol was undertaken at 4 C.
  • ⁇ - Secretase activity and protein recovery data are provided in Table III, infra.
  • Membrane Extract About 400-g of 293 cells were resuspended in 2,000-ml of 20 mM HEPES (Sigma Chemical Co., St Louis, MO), pH 7.5, 2 mM EDTA (Sigma Chemical Co., St Louis, MO) and 0.25% sucrose (Buffer IA). The cell suspension was homogenized in an 1000-ml aliquot and the cells were broken with an Brinkmann Homogenizer (POLYTRON, Switzerland) according to the following conditions: at setting 4 for 2- and 1-min; and at setting 5 for 1- and 0.5-min (with 3-min cooling time after each homogenization).
  • Brinkmann Homogenizer POLYTRON, Switzerland
  • the broken cell suspension was centrifuged at l,000xg for 20-min; the resulting supernatant (post-nuclear supernatant; PNSl) was saved.
  • the pellet was resuspended in 1,200-ml of Buffer IA, homogenized , broken and centrifuged as described above; the supernatant (PNS2) was saved and the pellet was discarded.
  • PNSl and PNS2 were combined (2,700-ml) and centrifuged at 45,000xg for 1-hr; the resulting pellet (P2; containing membranes) was saved and the supernatant was discarded.
  • This pellet was resuspended in 3,200-ml of 20 mM MES (Sigma Chemical Co., St Louis, MO), pH 6.0, 2 mM EDTA, 0.5% Triton X-100, 150 mM NaCl, 0.2 mM AEBSF, 5 mg/ml leupeptin, 2 mg/ml E64 and 1 mg/ml pepstatin A (Buffer IB); all four protease inhibitors were from Calbiochem; San Diego, CA).
  • the suspension was stirred continuously for 1-hr for extraction of beta-secretase and centrifuged at 16,000xg for 1-hr; the pellet was discarded.
  • the resulting supernatant (3,120- ml) was adjusted to pH 7.5 with 1 M Tris base and filtered through 0.45-mm Zapcap-S (Schleicher & Schuell; Keene, NH); the filtrate was the P2 membrane extract (P2ME, 3,170-ml).
  • a second P2ME was prepared similarly from another 400g of 293 cells.
  • the combined beta-secretase activity and protein data for the P2ME fractions are shown in Table III.
  • beta-secretase activity from each P2ME was purified separately in the next step.
  • WGA Wheat Germ Agglutinin
  • the first P2ME was loaded to a WGA-Agarose column (Vector Lab.; Burlingame, CA) (1000-ml), pre-equilibrated with 1500-ml of 20 mM Tris, pH 7.5, 2 mM EDTA, 0.5% Triton X-100 and 150 mM NaCl (Buffer 2A).
  • the WGA column which binds to N-acetyl-D-glucosamine glycopeptides, was washed with 1000-ml of Buffer 2A and bound proteins were eluted with a linear gradient of 0-10% chitin hydrolysate (Vector Lab.) in a total volume of 900-ml constructed from Buffer 2A and Buffer 2B (20 mM Tris, pH 7.5, 2 mM EDTA, 0.5% Triton X-100 and 10% chitin hydrolysate). The fractions containing higher specific activities of beta-secretase were pooled as the WGA-eluate (180-ml).
  • the WGA-Agarose column was regenerated and equilibrated.
  • the second WGA-eluate (170-ml) was obtained similarly from the second P2ME.
  • the two WGA-eluates were combined. (see Table III).
  • the two WGA- Agarose washings were combined. (see Table III).
  • the WGA-eluate and the WGA-wash were purified together in the next step.
  • HiTrap Q (HiQ) Eluate The WGA-eluate and the WGA-wash were diluted 6x and 4x, respectively, with 20 mM Tris, pH 8.0, 2 mM EDTA and 0.2% Triton X-100 (Buffer 3A) to reduce Triton X-100 and NaCl concentrations. Both diluted enzyme preparations were loaded consecutively to four connected, fast flow anion exchanger HiTrap Q columns (4x5-ml; Amersham Pharmacia Biotech, Piscataway, NJ), pre-equilibrated with 100-ml of Buffer 3A, and the column was washed with 200-ml of Buffer 3A.
  • Bound proteins were eluted first with a linear gradient of 0-500 mM NaCl in buffer 3A in a total volume of 420-ml and then eluted with 100-ml of Buffer 3B (Buffer 3A plus 500 mM NaCl). The fractions containing higher specific activities were pooled as the HiQ-eluate. (Table III) The HiQ-eluate was used directly in the next step.
  • HA Hydroxylapatite Wash: The HiQ-eluate was loaded to a Hydroxylapatite column (10-ml; Bio-Rad, Hercules, CA), pre-equilibrated with 100-ml of Buffer 4A (same as Buffer 3A), the column flow-through was collected. HA separation of proteins is based on charge absorption and desorption. The column was washed with 50-ml of Buffer 4A, and the bound proteins were eluted from the column with a linear gradient of 0-500 mM potassium phosphate in Buffer 4A in a total volume of 100-ml. Among the three fractions (the flow-through, the wash, and the eluate), the HA-wash (50-ml) showed the highest total , and specific activity of beta-secretase (Table III) and, thus, was used in the next step.
  • the HA-wash was de-salted to about 15 mM NaCl with Buffer 5A (same as Buffer 4A) using Centriprep 50 (Millipore, Bedford, MA ).
  • the de-salted HA-wash was loaded to a high resolving strong anion exchanger Mono Q column (1-ml; Amersham Pharmacia Biotech), pre- equilibrated with 30-ml of Buffer 5A.
  • the column was washed with 30-ml of buffer 5A, and bound proteins were eluted first with a linear gradient of 0- 500 mM in Buffer 5A in a total volume of 20-ml and then eluted with 10-ml of Buffer 5B (Buffer 5A plus 500 mM NaCl). Two activity peaks were observed and the active fractions from each activity peak were pooled. (Table III) The two Mono Q-eluates (2-ml & 4-ml)were purified separately in the next step.
  • Mono Q-eluate 1 was loaded to a high resolution gel-filtration Superdex 200 column (300-ml; Amersham Pharmacia Biotech), pre-equilibrated with 900-ml of 20 mM Tris, pH 8.0, 2 mM EDTA, 0.2% Triton X-100, 1 mM MgCl 2 , 1 mM CaCl 2 and 150 mM NaCl (Buffer 6).
  • the proteins were eluted with 300-ml of Buffer 6. Two activity peaks were observed and the active fractions from both peaks were pooled together (S- 200-eluate 1, 19.2-ml)(Table III) and used in the next step.
  • Mono Q-eluate 2 was similarly purified by the same Superdex 200 column; three activity peaks were observed.
  • the two Superdex 200-eluates that showed the higher specific activities (S-200 eluates 2a & 2b; Table III) were used separately in the next step.
  • the bound proteins were eluted from DEAE-Sepharose beads of the three samples with, respectively, 2x200-, 2x400- and 2x100 ⁇ l of Buffer 7B (Buffer 7A plus 1 M NaCl).
  • Buffer 7B Buffer 7A plus 1 M NaCl.
  • the three DEAE-eluates 400-, 800-, and 200 ⁇ l; Table III) were treated separately in the next step.
  • Buffer 9 A (same as Buffer 7A).
  • the bound proteins were washed and eluted with a step-wise NaCl gradient in Buffer 7A in a total volume of 35.6-ml as shown below: 7.6-ml at 0 mM NaCl, 16-ml from 0 to 250 mM, 8-ml from 250 to 500 mM and 4-ml at 500 mM. Two activity peaks were observed. Desalted De-Gly 2a were purified in a similar way; two activity peaks were also observed.
  • Desalted De-Gly 2b sample was purified with a Mini Q PC column (0.24-ml from Amersham Pharmacia Biotech) using a step-wise NaCl gradient in Buffer 7A in a total volume of 13.2-ml (4.8-ml at 0 mM NaCl, 4.8- ml from 0 to 250 mM, 2.4-ml from 250 to 500 mM and 1.2-ml at 500 mM). Only one major activity peak was observed. The resulting beta-secretase activity and protein data are shown in Table III.
  • Beta-secretase activity was purified up to 3, 000-fold to apparent homogeneity by the 9-step procedure. Note, however, that the actual activity enrichment was likely much greater excluding Triton X-100 interference to the low level protein determination in Step 9 above.
  • Mini Q-Eluate 2a represents the first peak from fraction Q-E la combined with the first peak from fraction
  • Mini Q-Eluate 2b represents the second peak from fraction Q-E la combined with the second peak from fraction Q-E lb.
  • SEQ ID NO: 2 or SEQ ID NO: 4 will fail to change the function of the amino acid compound.
  • some hydrophobic amino acids may be exchanged for other hydrophobic amino acids.
  • Those altered amino acid compounds which confer substantially the same function in substantially the same manner as the exemplified amino acid compounds are also encompassed within the present invention.
  • Typical such conservative substitutions attempt to preserve the: (a) secondary or tertiary structure of the polypeptide backbone; (b) the charge or hydrophobicity of the residue; or (c) the bulk of the side chain.
  • Some examples of such conservative substitutions of amino acids, resulting in the production of proteins which are functional equivalents of the protein of SEQ ID NO:2 or SEQ ID NO:4 are shown in Table II, infra.
  • substitutions may be introduced into the protein in a variety of ways, such as during the chemical synthesis or by chemical modification of an amino acid side chain after the protein has been prepared.
  • Alterations of the protein having a sequence which corresponds to the sequence of SEQ ID NO:2 or SEQ ID NO:4 may also be induced by alterations of the nucleic acid compounds which encodes these proteins. These mutations of the nucleic acid compound may be generated by either random mutagenesis techniques, such as those techniques employing chemical mutagens, or by site-specific mutagenesis employing oligonucleotides. In addition, allelic variants of the gene encoding the protein of the present invention may also be purified by the processes of the present invention. Those nucleic acid compounds which confer substantially the same function in substantially the same manner as the exemplified nucleic acid compounds are also encompassed within the present invention.
  • nucleic acid compounds which comprise isolated nucleic acid sequences which encode SEQ ID NO:2 or SEQ ID NO:4.
  • amino acid compounds of the invention can be encoded by a multitude of different nucleic acid sequences because most of the amino acids are encoded by more than one nucleic acid triplet due to the degeneracy of the amino acid code. Because these alternative nucleic acid sequences would encode the same amino acid sequences, the present invention further comprises these alternate nucleic acid sequences.
  • N- terminal fragments of the determined protein sequence were then searched against protein sequence databases using commercially available BLAST methodology.
  • a BLAST analysis of the proprietary INCYTE database revealed a series of ESTs showing homology to the determined experimental sequence. The ESTs were subsequently assembled into a contiguous cDNA sequence.
  • RACE-ready cDNA (CLONTECH Laboratories, Palo Alto, CA, 94303) was used as a template to elongate and amplify the 5' end until a starting methionine was detected. PCR was then employed to generate the full length cDNA clone. E. coli were subsequently transformed with the full length cDNAs comprising SEQ ID NO:l and SEQ ID NO:3 in order to generate sufficient copies of the nucleic acids for sequence determination.
  • the gene encoding the amyloid precursor protein protease of the present invention may also be produced using synthetic methodology, which synthesis is well known in the art. See, e.g., E.L. Brown, R. Belagaje, M.J. Ryan, and H.G. Khorana, Methods in Enzvmology. 68:109-151 (1979).
  • nucleic acid sequences corresponding to the gene encoding the amyloid precursor protein protease of the present invention can also be generated using conventional DNA synthesizing apparatuses such as the Applied Biosystems Model 380A or 380B DNA synthesizers, (commercially available from Applied Biosystems, Inc., 850 Lincoln Center Drive, Foster City, CA 94404) These synthesizers employ phosphoramidite chemistry. In the alternative, the more traditional phosphotriester chemistry may be employed to synthesize the nucleic acids of this invention. See, e.g., M.J. Gait, ed., OLIGONUCLEOTIDE SYNTHESIS, A PRACTICAL APPROACH, (1984).
  • the synthetic amyloid precursor protein protease gene may be designed to possess restriction endonuclease cleavage sites at either end of the transcript to facilitate isolation from and integration into expression and amplification plasmids.
  • the restriction sites are chosen so as to properly orient the coding sequence of the target enzyme with control sequences to achieve proper in-frame reading and expression of the amyloid precursor protein protease molecule.
  • a variety of other such cleavage sites may be incorporated depending on the particular plasmid constructs employed and may be generated by techniques well known in the art.
  • the desired DNA sequences can be generated using the polymerase chain reaction as described in U.S. Patent No. 4,889,818, which is herein incorporated by reference.
  • the present invention also encompasses ribonucleic acid compounds which have a sequence which encodes SEQ ID NO:2 or SEQ ID NO:4.
  • the present invention provides the RNA sequence represented by
  • SEQ ID NO:5 which is herein referred to as SEQ ID NO:5.
  • ribonucleic acids of the present invention may be prepared using the polynucleotide synthetic methods discussed supra or they may be prepared enzymatically using any one of various RNA polymerases to transcribe a DNA template.
  • This invention also provides nucleic acids, RNA or DNA, which are complementary to SEQ ID NO:l, SEQ ID NO:3, or SEQ ID NO:5
  • the present invention also provides probes and primers useful for molecular biology techniques.
  • the 18 or more base pair compound is DNA.
  • Primers and probes may be obtained by means well known in the art. For example, once a protein of interest is isolated, restriction enzymes and subsequent gel separation may be used to isolate the fragment of choice.
  • the term "selectively hybridize" as used herein may refer to either of two situations. In the first such embodiment of this invention, the nucleic acid compounds described supra hybridize to DNA or RNA encoding a human ⁇ -secretase protease under more stringent hybridization conditions than these same nucleic acid compounds would hybridize to nucleic acids encoding analogous ⁇ -secretases of another species. In the second such embodiment of this invention, these probes hybridize to the DNA or RNA encoding the ⁇ -secretase under more stringent hybridization conditions than other related compounds, including nucleic acid sequences encoding other amyloid precursor protein proteases.
  • probes and primers can be prepared enzymatically as described supra. In a most preferred embodiment, however, these probes and primers are synthesized using chemical means as described herein.
  • probes and primers are well known. For example, all or part of the probes or primers may be used to hybridize to the coding sequence. Then, through PCR amplification, the full length sequence may be generated. The full length sequence can be subsequently subcloned into any vector of choice.
  • the primers or probes may be radioactively labeled at the 5' end in order to screen cDNA libraries by conventional means.
  • a primer or probe can be labeled with a radioactive element which provides for an adequate signal as a means for detection and has sufficient half-life to be useful for detection, such as 32p 3j ⁇ , 14c or the like.
  • recombinant DNA cloning vectors and expression vectors comprising the nucleic acids of the present invention can be prepared. Many such vectors are illustrated and described above.
  • the preferred nucleic acid vectors are those which are DNA.
  • the preferred recombinant DNA vectors comprise the isolated DNA sequences as given by SEQ ID NO:l or SEQ ID NO:3.
  • the most preferred recombinant DNA vectors comprise the DNA sequence given by SEQ ID NO:3.
  • the type of cloning vector employed depends upon the availability of appropriate restriction sites, the type of host cell in which the vector is to be transfected or transformed, the purpose of the transfection or transformation (e.g., transient expression in an oocyte system, stable transformation as an extrachromosomal element, or integration into the host chromosome), the presence or absence of readily assayable markers (e.g., antibiotic resistance markers, metabolic markers, or the like), and the number of copies of the gene to be present in the cell.
  • readily assayable markers e.g., antibiotic resistance markers, metabolic markers, or the like
  • the type of vector employed to carry the nucleic acids of the present invention may be RNA viruses, DNA viruses, lytic bacteriophages, lysogenic bacteriophages, stable bacteriophages, plasmids, viroids, and the like.
  • a constitutive promoter i.e. a promoter which is functional at all times, instead of a regulatable promoter which may be activated or inactivated by the artisan using heat, addition or removal of a nutrient, addition of an antibiotic, and the like.
  • the practitioner also understands that the amount of the nucleic acid or protein to be produced dictates, in part, the selection of the expression system.
  • Plasmids can be readily modified to construct expression vectors in a variety of organisms, including, but not limited to, E. coli, Sf9 (as host for baculovirus), Spodoptera and Saccharomyces. See: Serook et al., Molecular Cloning: A Laboratory Manual (1989)
  • oligonucleotide-directed site-specific mutagenesis One of the most widely employed techniques for altering a nucleic acid sequence is by way of oligonucleotide-directed site-specific mutagenesis.
  • B Comack "Current Protocols in Molecular Biology", 8.01-8.5.9, (F. Ausubel, et al., eds. 1991).
  • an oligonucleotide whose sequence contains the mutation of interest, is synthesized as described supra.
  • This oligonucleotide is then hybridized to a template containing the wild-type sequence.
  • the template is a single-stranded template.
  • Particularly useful are plasmids which contain regions such as the fl intergenic region. This region allows the generation of single-stranded templates when a helper phage is added to the culture harboring the "phagemid".
  • a DNA-dependent DNA polymerase is then used to synthesize the second strand from the oligonucleotide, complementary to the template DNA.
  • the resulting product is a heteroduplex molecule containing a mismatch due to the mutation in the oligonucleotide.
  • Host cells which harbor the nucleic acids provided by the present invention can also be prepared.
  • One suitable host cell is an Xenopus sp. oocyte which has been injected with RNA or DNA compounds of the present invention.
  • Preferred oocytes of the present invention are those which harbor a sense mRNA of the present invention.
  • Other preferred host cells include HeLa and 293 cells which have been transfected and/or transformed with a vector which comprises a nucleic acid of the present invention.
  • a method for constructing a recombinant host cell capable of expressing SEQ ID NO:2 or SEQ ID NO:4 is also possible with regard to the present invention, said method comprising transforming a host cell with a recombinant DNA vector that comprises an isolated DNA sequence which encodes SEQ ID NO:2 or SEQ ID NO:4.
  • the preferred host cell is 293 cells.
  • a preferred vector for expression is one which comprises SEQ ID NO:l or SEQ ID NO:3, most preferably SEQ ID NO:l.
  • Transformed host cells may be cultured under conditions well known to skilled artisans such that SEQ ID NO: 2 or SEQ ID NO:4 are expressed, thereby producing the sequence of ⁇ - secretase in the recombinant host cell.
  • a further embodiment of the invention consists of a method of isolating and purifying an amyloid precursor protein protease from a host cell expressing said protein.
  • a host cell either prokaryotic or eukaryotic, expressing amyloid precursor protein protease
  • the second step of this embodiment is the isolation of amyloid precursor protein protease from the cultured cells.
  • the cultured cells are lysed by nitrogen cavitation in the presence of protease inhibitors.
  • a soluble fraction is prepared from the lysate by ultracentrifugation.
  • the resulting solution of cytosolic proteins contains ⁇ -secretase and is subjected to a series of purification procedures.
  • the soluble fraction of the cell lysate is run through a series of column chromatography procedures. Anion exchange chromatography is followed by hydrophobic interaction, molecular sizing, and finally another hydrophobic interaction technique where the conditions are such that the ⁇ - secretase binds the resin weakly. Each column is run individually, and the eluate is collected in fractions while monitoring for absorbance at 280 nm. Fractions are assayed for ⁇ -secretase activity, and those fractions with the desired activity are then run over the next column until a homogeneous solution of ⁇ -secretase is obtained.
  • Immunoaffinity purification using anti- ⁇ -secretase antibodies is an alternative to the series of chromatographic procedures already mentioned. Making antiserum or monoclonal antibodies directed against a purified protein is well known in the art, and skilled artisans readily will be able to prepare anti- ⁇ -secretase antibodies. Preparing an immunoaffinity matrix using such antibodies and isolating ⁇ -secretase using the immunoaffinity matrix is also well within the skill of the art. See, AFFINITY CHROMATOGRAPHY PRINCIPLES & METHODS, Pharmacia Fine Chemicals, 1983. The ability of an agent to inhibit the protein of the present invention is essential in the development of a multitude of indications.
  • such an assay includes a method for determining whether a substance is a functional ligand of ⁇ -secretase, said method comprising contacting a functional compound of the ⁇ -secretase with said substance, monitoring enzymatic activity by physically detectable means, and identifying those substances which effect a chosen response.
  • the instant invention provides such a screening system useful for discovering agents which inhibit the ⁇ -secretase, said screening system comprising the steps of:
  • Utilization of the screening system described above provides a sensitive and rapid means to determine compounds which inhibit ⁇ -secretase.
  • This screening system may also be adapted to automated procedures such as a
  • PANDEX® Boxter-Dade Diagnostics
  • a protein protease is prepared as elsewhere described herein, preferably using recombinant DNA technology.
  • a sample of a test compound is then introduced to the reaction vessel containing the protein protease followed by the addition of an appropriate substrate.
  • the substrate may be added simultaneously with the test compound.
  • the desirability of a bioactivity assay system which determines the response of ⁇ -secretase to a compound of interest is clear.
  • the instant invention provides such a bioactivity assay, said assay comprising the steps of: a) transfecting a mammalian host cell with an expression vector comprising DNA encoding ⁇ -secretase;
  • the present invention comprises a method of using said inhibitors of ⁇ -secretase in the treatment of patients with acquired disease states of the brain including Alzheimer's disease.
  • patient refers to a mammal such a rat, mouse, guinea pig, dog, or human. It is understood, however, that the preferred patient for use with the methods herein is a human patient.
  • treatment includes its generally acepted meaning which encompasses prohibiting, preventing, restraining, and slowing, stopping, or reversing the progression, severity, and resulting symptoms of acquired disease states of the brain.
  • themethods of the present invention encompass both therapeutic and prophylactic uses.
  • Compounds which are inhibitors of ⁇ -secretase are usually administered in the form of pharmaceutical compositions. These compounds can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. These compounds are effective as both injectable and oral compositions. Such compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.
  • the present invention also includes methods employing pharmaceutical compositions which contain, as the active ingredient, inhibitors of ⁇ -secretase associated with pharmaceutically acceptable carriers.
  • the active ingredient is usually mixed with an excipient, diluted by an excipient or enclosed within such a carrier which can be in the form of a capsule, sachet, paper or other container.
  • the excipient serves as a diluent, it can be a solid, semi- solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient.
  • compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing for example up to 10% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.
  • the active compound In preparing a formulation, it may be necessary to mill the active compound to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it ordinarily is milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size is normally adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.
  • excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose.
  • the formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxybenzoates; sweetening agents; and flavoring agents.
  • compositions of the invention can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.
  • the compositions are preferably formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • the active compound is effective over a wide dosage range.
  • dosages per day normally fall within the range of about 0.5 to about 30 mg/kg of body weight. In the treatment of adult humans, the range of about 1 to about 15 mg/kg/day, in single or divided dose, is especially preferred.
  • the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms, and therefore the above dosage ranges are not intended to limit the scope of the invention in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, provided that such larger doses are first divided into several smaller doses for administration throughout the day.
  • the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • a solid preformulation composition containing a homogeneous mixture of a compound of the present invention.
  • the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.
  • This solid preformulation is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of the active ingredient of the present invention.
  • the tablets or pills of the present invention may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action.
  • the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former.
  • the two components can be separated by enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release.
  • enteric layers or coatings such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.
  • liquid forms in which the novel compositions of the present invention may be incorporated for administration orally or by injection include aqueous solutions, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles.
  • compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders.
  • the liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra.
  • the compositions are administered by the oral or nasal respiratory route for local or systemic effect.
  • Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device may be attached to a face mask, tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices which deliver the formulation in an appropriate manner.
  • Hard gelatin capsules containing the following ingredients are prepared:
  • Quantity Ingredient (mg/capsule)
  • the above ingredients are mixed and filled into hard gelatin capsules in 340 mg quantities.
  • a tablet formula is prepared using the ingredients below:
  • Quantity Ingredient (mg/tablet)
  • Stearic acid 5.0 The components are blended and compressed to form tablets, each weighing 240 mg.
  • a dry powder inhaler formulation is prepared containing the following components:
  • the active mixture is mixed with the lactose and the mixture is added to a dry powder inhaling appliance.
  • Tablets each containing 30 mg of active ingredient, are prepared as follows:
  • Quantity Ingredient (mg/tablet)
  • the active ingredient, starch and cellulose are passed through a
  • Capsules each containing 40 mg of medicament are made as follows:
  • the active ingredient, cellulose, starch, and magnesium stearate are blended, passed, through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 150 mg quantities.
  • Suppositories each containing 25 mg of active ingredient are made as follows:
  • the active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of nominal 2.0 g capacity and allowed to cool.
  • Suspensions each containing 50 mg of medicament per 5.0 ml dose are made as follows:
  • Ingredient Amount Active Ingredient 50.0 mg Xanthan gum 4.0 mg
  • the medicament, sucrose and xanthan gum are blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of the microcrystalline cellulose and sodium carboxymethyl cellulose in water.
  • the sodium benzoate, flavor, and color are diluted with some of the water and added with stirring. Sufficient water is then added to produce the required volume.
  • Capsules each containing 15 mg of medicament, are made as follows:
  • the active ingredient, cellulose, starch, and magnesium stearate are blended, passed through a No. 20 mesh U.S. sieve, and filled into hard gelatin capsules in 425 mg quantities.
  • An intravenous formulation may be prepared as follows:
  • a topical formulation may be prepared as follows:
  • the white soft paraffin is heated until molten.
  • the liquid paraffin and emulsifying wax are incorporated and stirred until dissolved.
  • the active ingredient is added and stirring is continued until dispersed.
  • the mixture is then cooled until solid.
  • Sublingual or buccal tablets each containing 10 mg of active ingredient, may be prepared as follows:
  • the glycerol, water, sodium citrate, polyvinyl alcohol, and polyvinylpyrrolidone are admixed together by continuous stirring and maintaining the temperature at about 90°C.
  • the solution is cooled to about 50-55°C and the medicament is slowly admixed.
  • the homogenous mixture is poured into forms made of an inert material to produce a drug-containing diffusion matrix having a thickness of about 2-4 mm. This diffusion matrix is then cut to form individual tablets having the appropriate size.
  • transdermal delivery devices Such transdermal patches may be used to provide continuous or discontinuous infusion of the compounds of the present invention in controlled amounts.
  • transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Patent 5,023,252, issued June 11, 1991, herein incorporated by reference.
  • patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • Indirect techniques which are generally preferred, usually involve formulating the compositions to provide for drug latentiation by the conversion of hydrophilic drugs into lipid-soluble drugs or prodrugs.
  • Latentiation is generally achieved through blocking of the hydroxy, carbonyl, sulfate, and primary amine groups present on the drug to render the drug more lipid soluble and amenable to transportation across the blood-brain barrier.
  • the delivery of hydrophilic drugs may be enhanced by intra-arterial infusion of hypertonic solutions which can transiently open the blood-brain barrier.
  • the proteins of this invention as well as fragments of these proteins may be used as antigens for the synthesis of antibodies.
  • antibody as used herein describes antibodies, fragments of antibodies (such as, but not limited, to Fab, Fab', Fab2', and Fv fragments), and chimeric, humanized, veneered, resurfaced, or CDR-grafted antibodies capable of binding antigens of a similar nature as the parent antibody molecule from which they are derived.
  • the instant invention also encompasses single chain polypeptide binding molecules.
  • antibody as used herein is not limited by the manner in which the antibodies are produced, whether such production is in situ or not.
  • antibody as used in this specification encompasses those antibodies produced by recombinant DNA technology means including, but not limited, to expression in bacteria, yeast, insect cell lines, or mammalian cell lines.
  • mice The production of antibodies, both monoclonal and polyclonal, in animals, especially mice, is well known in the art. See, e.g., C. Milstein, HANDBOOK OF EXPERIMENTAL IMMUNOLOGY, (Blackwell Scientific Pub., 1986); J. Goding, MONOCLONAL ANTIBODIES: PRINCIPLES AND PRACTICE, (Academic Press, 1983).
  • monoclonal antibodies the basic process begins with injecting a mouse, or other suitable animal, with an immunogen. The mouse is subsequently sacrificed and cells taken from its spleen are fused with myeloma cells, resulting in a hybridoma that reproduces in vitro.
  • the population of hybridomas is screened to isolate individual clones, each of which secretes a single antibody species, specific for the immunogen.
  • the individual antibody species obtained in this way is each the product of a single B cell from the immune animal generated in response to a specific antigenic site, or epitope, recognized on the immunogenic substance.
  • Chimeric antibodies are described in U.S. Patent No. 4,816,567, which issued March 28, 1989 to S. Cabilly, et al. This reference discloses methods and vectors for the preparation of chimeric antibodies. The entire contents of U.S. Patent No. 4,816,567 are herein incorporated by reference.
  • An alternative approach to production of genetically engineered antibodies is provided in U.S. Patent No. 4,816,397, which also issued March 28, 1989 to M.
  • Boss, et al. the entire contents of which are herein incorporated by reference.
  • the Boss patent teaches the simultaneous co-expression of the heavy and light chains of the antibody in the same host cell.
  • the approach of U.S. Patent 4,816,397 has been further refined as taught in European Patent Publication No. 0 239 400, which published September 30, 1987.
  • the teachings of this European patent publication (Winter) are a preferred format for the genetic engineering of the reactive monoclonal antibodies of this invention.
  • the Winter technology involves the replacement of complementarity determining regions (CDRs) of a human antibody with the CDRs of a murine monoclonal antibody thereby converting the specificity of the human antibody to the specificity of the murine antibody which was the source of the CDR regions.
  • This "CDR grafting" technology affords a molecule containing minimal murine sequence and thus is less immunogenic.
  • Single chain antibody technology is yet another variety of genetically engineered antibody which is now well known in the art. See, e.g. R.E. Bird, et al., Science 242:423-426 (1988); PCT Publication No. WO 88/01649, which was published 10 March 1988; United States Patent 5,260,203, issued November 9, 1993, the entire contents of which are herein incorporated by reference.
  • the single chain antibody technology involves joining the binding regions of heavy and light chains with a polypeptide sequence to generate a single polypeptide having the binding specificity of the antibody from which it was derived.
  • the aforementioned genetic engineering approaches provide the skilled artisan with numerous means to generate molecules which retain the binding characteristics of the parental antibody while affording a less immunogenic format.
  • antibodies are used in diagnostics, therapeutics or in diagnostic/therapeutic combinations.
  • diagnosis as used herein is meant testing that is related to either the in vitro or in vivo diagnosis of disease states or biological status in mammals, preferably in humans.
  • therapeutics and “therapeutic/diagnostic combinations” as used herein is respectively meant the treatment or the diagnosis and treatment of disease states or biological status by the in vivo administration to mammals, preferably humans, of the antibodies of the present invention.
  • the antibodies of the present invention are especially preferred in the diagnosis and/or treatment of conditions associated with an excess or deficiency of ⁇ -secretase.

Abstract

La présente invention concerne une protéase de protéine précurseur amyloïde et des composés d'acides nucléiques apparentés. Cette invention concerne aussi des compositions, des vecteurs d'expression, des cellules hôtes transfectées, de même que des dosages et des techniques d'utilisation. Les composés de cette invention permettront de mieux cerner la maladie d'Alzheimer et d'autres états liés à une maladie neurodégénérative.
PCT/US2000/006707 1999-05-11 2000-05-09 Protease de proteine precurseur amyloide et composes d'acides nucleiques apparentes WO2000068266A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU46406/00A AU4640600A (en) 1999-05-11 2000-05-09 Amyloid precursor protein protease and related nucleic acid compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13342399P 1999-05-11 1999-05-11
US60/133,423 1999-05-11

Publications (1)

Publication Number Publication Date
WO2000068266A1 true WO2000068266A1 (fr) 2000-11-16

Family

ID=22458563

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/006707 WO2000068266A1 (fr) 1999-05-11 2000-05-09 Protease de proteine precurseur amyloide et composes d'acides nucleiques apparentes

Country Status (2)

Country Link
AU (1) AU4640600A (fr)
WO (1) WO2000068266A1 (fr)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001018204A1 (fr) * 1999-09-03 2001-03-15 Curagen Corporation Polynucleotides codant des membres de la famille humaine d'antigenes b7 d'activation des lymphocytes b et polypeptides ainsi codes
WO2001049098A2 (fr) * 1999-10-13 2001-07-12 Michael Jerome Bienkowski Secretase associee a la maladie d'alzheimer, substrats app et utilisations correspondants
WO2002022683A1 (fr) * 2000-09-12 2002-03-21 Kirin Beer Kabushiki Kaisha Nouvelle membrane a paroi cellulaire dendritique et son utilisation
EP1212344A1 (fr) * 1999-09-03 2002-06-12 Human Genome Sciences, Inc. Polynucleotides, polypeptides et anticorps de type b7
WO2003014293A2 (fr) * 2001-08-02 2003-02-20 Eli Lilly And Company Nouveaux analogues de polypeptides et proteines hybrides et leurs procedes d'utilisation
US6627739B1 (en) 1999-02-10 2003-09-30 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US6835565B1 (en) 1998-09-24 2004-12-28 Pharmacia & Upjohn Company Alzheimer's disease secretase
US6844148B1 (en) 1998-09-24 2005-01-18 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6965018B2 (en) 2000-06-06 2005-11-15 Bristol-Myers Squibb Company Antibodies directed to B7-related polypeptide, BSL-2
WO2005119262A2 (fr) * 2004-05-27 2005-12-15 Galapagos N.V. Procedes, compositions et essais sur composes pour l'inhibition de la production de la proteine beta-amyloide
US7115410B1 (en) 1999-02-10 2006-10-03 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7368554B2 (en) 2000-06-06 2008-05-06 Bristol-Myers Squibb Company Polynucleotides encoding BSL2v1c2-Ig
US7456007B1 (en) 1998-12-31 2008-11-25 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7514408B1 (en) 1999-12-02 2009-04-07 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7527969B2 (en) 2002-06-19 2009-05-05 Raven Biotechnologies, Inc. RAAG10 cell surface target and a family of antibodies recognizing that target
US7718774B2 (en) 2006-11-08 2010-05-18 Macrogenics, Inc. TES7 and antibodies that bind thereto
US7732131B2 (en) 2004-08-03 2010-06-08 Innate Pharma S.A. Therapeutic and diagnostic methods and compositions targeting 4Ig-B7-H3 and its counterpart NK cell receptor
US8394815B2 (en) 2002-08-19 2013-03-12 Lorus Therapeutics Inc. 2,4,5-trisubstituted imidazoles and their use as anti-microbial agents
US8840889B2 (en) 2009-08-13 2014-09-23 The Johns Hopkins University Methods of modulating immune function
US8969372B2 (en) 2003-11-14 2015-03-03 Aptose Boisciences Inc. Aryl imidazoles and their use as anti-cancer agents
US9150656B2 (en) 2010-03-04 2015-10-06 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9309247B2 (en) 2013-03-20 2016-04-12 Lorus Therapeutics Inc. 2-substituted imidazo[4,5-D]phenanthroline derivatives and their use in the treatment of cancer
US9441049B2 (en) 2010-03-04 2016-09-13 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US11104957B2 (en) 2013-10-04 2021-08-31 Aptose Biosciences, Inc. Compositions and methods for treating cancers
US11149047B2 (en) 2017-10-30 2021-10-19 Aptose Biosciences, Inc. Aryl imidazoles for treatment of cancer

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0653154A2 (fr) * 1993-11-12 1995-05-17 Hoechst Japan Limited Animal transgènique pour la maladie d'Alzheimer
WO1996040885A2 (fr) * 1995-06-07 1996-12-19 Athena Neurosciences, Inc. β-SECRETASE, ANTICORPS DIRIGES CONTRE LA β-SECRETASE ET DOSAGES PERMETTANT DE DETECTER L'INHIBITION DE LA β-SECRETASE
WO1999046281A2 (fr) * 1998-03-10 1999-09-16 Genentech, Inc. Nouveaux polypeptides et acides nucleiques les codant

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0653154A2 (fr) * 1993-11-12 1995-05-17 Hoechst Japan Limited Animal transgènique pour la maladie d'Alzheimer
WO1996040885A2 (fr) * 1995-06-07 1996-12-19 Athena Neurosciences, Inc. β-SECRETASE, ANTICORPS DIRIGES CONTRE LA β-SECRETASE ET DOSAGES PERMETTANT DE DETECTER L'INHIBITION DE LA β-SECRETASE
WO1999046281A2 (fr) * 1998-03-10 1999-09-16 Genentech, Inc. Nouveaux polypeptides et acides nucleiques les codant

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE GENEMBL 6 June 1996 (1996-06-06), HILLIER ET AL.: "yx480g8.r1 soares melanocyte 2NbHM Homo sapiens cDNA clone IMAGE:265022 5' similar to contains Alu repetitive element", XP002148339 *
DATABASE SWISSPROT 1 June 1998 (1998-06-01), HUSAINI ET AL.: "Bos taurus butyrophilin fragment", XP002148340 *

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7375187B2 (en) 1998-09-24 2008-05-20 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US7378511B2 (en) 1998-09-24 2008-05-27 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US7812123B2 (en) 1998-09-24 2010-10-12 Elan Pharmaceuticals, Inc. Alzheimer's disease secretase
US7456269B2 (en) 1998-09-24 2008-11-25 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates thereof, and uses thereof
US6790610B2 (en) 1998-09-24 2004-09-14 Pharmacia & Upjohn Company Alzheimer's disease, secretase, APP substrates therefor, and uses therefor
US6844148B1 (en) 1998-09-24 2005-01-18 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6420534B1 (en) 1998-09-24 2002-07-16 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses thereof
US7368536B2 (en) 1998-09-24 2008-05-06 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6440698B1 (en) 1998-09-24 2002-08-27 Mark E. Gurney Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6500667B1 (en) 1998-09-24 2002-12-31 Pharmacia & Upjohn Company Aspartyl protease 2 (Asp2) antisense oligonucleotides
US7041473B1 (en) 1998-09-24 2006-05-09 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses thereof
US6797487B2 (en) 1998-09-24 2004-09-28 Pharamcia & Upjohn Company Polynucleotides encoding alzheimer's disease secretase
US6913918B2 (en) 1998-09-24 2005-07-05 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6867018B1 (en) 1998-09-24 2005-03-15 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses thereof
US6835565B1 (en) 1998-09-24 2004-12-28 Pharmacia & Upjohn Company Alzheimer's disease secretase
US6828117B2 (en) 1998-09-24 2004-12-07 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6699671B1 (en) 1998-09-24 2004-03-02 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6706485B1 (en) 1998-09-24 2004-03-16 Pharmacia & Upjohn Company Method of identifying agents that inhibit APP processing activity
US6727074B2 (en) 1998-09-24 2004-04-27 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6737510B1 (en) 1998-09-24 2004-05-18 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses thereof
US6753163B2 (en) 1998-09-24 2004-06-22 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US6825023B1 (en) 1998-09-24 2004-11-30 Pharmacia & Upjohn Company Alzheimer's disease secretase, APP substrates therefor, and uses therefor
US7456007B1 (en) 1998-12-31 2008-11-25 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US6627739B1 (en) 1999-02-10 2003-09-30 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7067271B1 (en) 1999-02-10 2006-06-27 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7276349B2 (en) 1999-02-10 2007-10-02 Elan Pharmaceuticals, Inc. Beta-secretase enzyme compositions and methods
US7252963B2 (en) 1999-02-10 2007-08-07 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7115410B1 (en) 1999-02-10 2006-10-03 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7109017B1 (en) 1999-02-10 2006-09-19 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7427478B2 (en) 1999-02-10 2008-09-23 Elan Pharmaceuticals, Inc. Beta-secretase enzyme compositions and methods
EP1212344A1 (fr) * 1999-09-03 2002-06-12 Human Genome Sciences, Inc. Polynucleotides, polypeptides et anticorps de type b7
WO2001018204A1 (fr) * 1999-09-03 2001-03-15 Curagen Corporation Polynucleotides codant des membres de la famille humaine d'antigenes b7 d'activation des lymphocytes b et polypeptides ainsi codes
US6429303B1 (en) 1999-09-03 2002-08-06 Curagen Corporation Nucleic acids encoding members of the human B lymphocyte activation antigen B7 family and methods of using the same
EP1212344A4 (fr) * 1999-09-03 2004-08-04 Human Genome Sciences Inc Polynucleotides, polypeptides et anticorps de type b7
WO2001049098A3 (fr) * 1999-10-13 2003-11-20 Michael Jerome Bienkowski Secretase associee a la maladie d'alzheimer, substrats app et utilisations correspondants
WO2001050829A2 (fr) * 1999-10-13 2001-07-19 Michael Jerome Bienkowski Secretase de la maladie d'alzheimer, substrats app destines a celle-ci et utilisation de cette derniere
WO2001049097A3 (fr) * 1999-10-13 2003-11-20 Michael Jerome Bienkowski Secretase liee a la maladie d'alzheimer, substrats app et leurs utilisations
WO2001050829A3 (fr) * 1999-10-13 2003-12-04 Michael Jerome Bienkowski Secretase de la maladie d'alzheimer, substrats app destines a celle-ci et utilisation de cette derniere
WO2001049098A2 (fr) * 1999-10-13 2001-07-12 Michael Jerome Bienkowski Secretase associee a la maladie d'alzheimer, substrats app et utilisations correspondants
WO2001049097A2 (fr) * 1999-10-13 2001-07-12 Michael Jerome Bienkowski Secretase liee a la maladie d'alzheimer, substrats app et leurs utilisations
US7514408B1 (en) 1999-12-02 2009-04-07 Elan Pharmaceuticals, Inc. β-secretase enzyme compositions and methods
US7807786B2 (en) 2000-06-06 2010-10-05 Bristol-Myers Squibb Company BSL2v1c2 polypeptides
US9399052B2 (en) 2000-06-06 2016-07-26 Bristol-Myers Squibb Company Polynucleotides encoding BSL2v2c2-Ig
US7358354B2 (en) 2000-06-06 2008-04-15 Bristol-Myers Squibb Company Polynucleotides encoding BSL3
US7279567B2 (en) 2000-06-06 2007-10-09 Bristol-Myers Squibb Company Polynucleotides-encoding B7-related polypeptide, BSL2-L165-35B
US9409970B2 (en) 2000-06-06 2016-08-09 Bristol-Myers Squibb Company Polynucleotides encoding BSL2v2c2
US8664360B2 (en) 2000-06-06 2014-03-04 Bristols-Myers Squibb Company BSL3 polypeptides
US8354513B2 (en) 2000-06-06 2013-01-15 Bristol-Myers Squibb Company Polynucleotides encoding BSL2vcvc-lg
US8080636B2 (en) 2000-06-06 2011-12-20 Bristol-Myers Squibb Company BSL3 antibodies
US10308702B2 (en) 2000-06-06 2019-06-04 Bristol-Myers Squibb Comapny BSL2v2c2-Ig polypeptides
US8674076B2 (en) 2000-06-06 2014-03-18 Bristol-Myers Squibb Company BSL2vcvc-lg polypeptides
US10400023B2 (en) 2000-06-06 2019-09-03 Bristol-Myers Squibb Company B7-related nucleic acids and polypeptides useful for immunomodulation
US7723479B2 (en) 2000-06-06 2010-05-25 Bristol-Myers Squibb Company BSL3 polypeptides
US7368554B2 (en) 2000-06-06 2008-05-06 Bristol-Myers Squibb Company Polynucleotides encoding BSL2v1c2-Ig
US6965018B2 (en) 2000-06-06 2005-11-15 Bristol-Myers Squibb Company Antibodies directed to B7-related polypeptide, BSL-2
WO2002022683A1 (fr) * 2000-09-12 2002-03-21 Kirin Beer Kabushiki Kaisha Nouvelle membrane a paroi cellulaire dendritique et son utilisation
EP2080766A1 (fr) 2001-06-06 2009-07-22 Bristol-Myers Squibb Company Acides nucléiques et polypeptides apparentés à B7 utiles pour l'immunomodulation
WO2003014293A3 (fr) * 2001-08-02 2003-12-24 Lilly Co Eli Nouveaux analogues de polypeptides et proteines hybrides et leurs procedes d'utilisation
WO2003014293A2 (fr) * 2001-08-02 2003-02-20 Eli Lilly And Company Nouveaux analogues de polypeptides et proteines hybrides et leurs procedes d'utilisation
US7527969B2 (en) 2002-06-19 2009-05-05 Raven Biotechnologies, Inc. RAAG10 cell surface target and a family of antibodies recognizing that target
US8779098B2 (en) 2002-06-19 2014-07-15 Macrogenics West, Inc. B7-H3L cell surface antigen and antibodies that bind thereto
US9574007B2 (en) 2002-06-19 2017-02-21 Macrogenics, Inc. B7-H3L cell surface antigen and antibodies that bind thereto
US8394815B2 (en) 2002-08-19 2013-03-12 Lorus Therapeutics Inc. 2,4,5-trisubstituted imidazoles and their use as anti-microbial agents
US8969372B2 (en) 2003-11-14 2015-03-03 Aptose Boisciences Inc. Aryl imidazoles and their use as anti-cancer agents
US10080739B2 (en) 2003-11-14 2018-09-25 Aptose Biosciences Inc. Aryl imidazoles and their use as anti-cancer agents
WO2005119262A2 (fr) * 2004-05-27 2005-12-15 Galapagos N.V. Procedes, compositions et essais sur composes pour l'inhibition de la production de la proteine beta-amyloide
WO2005119262A3 (fr) * 2004-05-27 2006-06-15 Galapagos Nv Procedes, compositions et essais sur composes pour l'inhibition de la production de la proteine beta-amyloide
US8465931B2 (en) 2004-08-03 2013-06-18 Innate Pharma S.A. Therapeutic and diagnostic methods and compositions targeting 4Ig-B7-H3 and its counterpart NK cell receptor
US7732131B2 (en) 2004-08-03 2010-06-08 Innate Pharma S.A. Therapeutic and diagnostic methods and compositions targeting 4Ig-B7-H3 and its counterpart NK cell receptor
US8216570B2 (en) 2006-11-08 2012-07-10 Macrogenics, Inc. TES7 and antibodies that bind thereto
US7718774B2 (en) 2006-11-08 2010-05-18 Macrogenics, Inc. TES7 and antibodies that bind thereto
US8840889B2 (en) 2009-08-13 2014-09-23 The Johns Hopkins University Methods of modulating immune function
US9676856B2 (en) 2009-08-13 2017-06-13 The Johns Hopkins University Methods of modulating immune function
US10730945B2 (en) 2010-03-04 2020-08-04 Macrogenics, Inc. Antibodies reactive with B7-H3 and users thereof
US9714296B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9714295B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9896508B2 (en) 2010-03-04 2018-02-20 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US9441049B2 (en) 2010-03-04 2016-09-13 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US9150656B2 (en) 2010-03-04 2015-10-06 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US10683364B2 (en) 2010-03-04 2020-06-16 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
US9309247B2 (en) 2013-03-20 2016-04-12 Lorus Therapeutics Inc. 2-substituted imidazo[4,5-D]phenanthroline derivatives and their use in the treatment of cancer
US11104957B2 (en) 2013-10-04 2021-08-31 Aptose Biosciences, Inc. Compositions and methods for treating cancers
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US11591400B2 (en) 2016-04-15 2023-02-28 Macrogenics, Inc. B7-H3 directed antibody drug conjugates
US11149047B2 (en) 2017-10-30 2021-10-19 Aptose Biosciences, Inc. Aryl imidazoles for treatment of cancer

Also Published As

Publication number Publication date
AU4640600A (en) 2000-11-21

Similar Documents

Publication Publication Date Title
WO2000068266A1 (fr) Protease de proteine precurseur amyloide et composes d'acides nucleiques apparentes
DE69534402T2 (de) Apoptose-proteasen 3 und 4, ähnlich dem umwandlungsenzym für interleukin-1beta
AU744875B2 (en) Prostate tumor polynucleotide and antigen compositions
US6355478B1 (en) Rhesus monkey neuropeptide Y Y2 receptor
JPH11507205A (ja) ヒト腫瘍壊死因子受容体
EP0711832A2 (fr) Récepteur métabotropique humain, m Glu R2
SK287085B6 (sk) Izolovaný polypeptid, ktorý sa viaže na TRAF2 a modeluje aktivitu NF-kB, DNA sekvencia, ktorá ho kóduje, farmaceutický prostriedok s ich obsahom a ich použitie
JP2002512521A (ja) 32個のヒト分泌タンパク質
US6093397A (en) Amyloid precursor protein protease
US5618918A (en) Human brain sodium dependent inorganic phosphate cotransporter
JP2001523943A (ja) 2つのヒト・カテプシンタンパク質
US5840509A (en) Protease and related nucleic acid compounds
JP2002532054A (ja) 29個のヒト分泌タンパク質
US5698444A (en) Serotonin receptor protein and related nucleic acid compounds
JPH10201487A (ja) 哺乳動物のfin−1核酸および蛋白質配列およびその使用
EP0780472A2 (fr) Protéines de stress
JPH11501802A (ja) ユビキチン結合酵素7、8および9
US6653446B2 (en) Human hypoxanthine- (guanine) phosphoribosyl transferase-2
US6030791A (en) Antibody for a homolog of rat elastase IV derived from human pancreas
US5945321A (en) Ubiquitin conjugating enzymes 7, 8 and 9
JP2002204697A (ja) ヒトdnaリガーゼiv
JPH10508484A (ja) ヒトdnaリガーゼiii
JP2002233366A (ja) ヒトデオキシシチジンキナーゼ2
JP2002511771A (ja) 新規なカルボキシペプチダーゼインヒビター
JP2002095486A (ja) ヒトカリウムチャンネル1および2タンパク質

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 10018383

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP