WO2003009813A2 - Methodes de traitement de maladies induites par neuropiline - Google Patents

Methodes de traitement de maladies induites par neuropiline Download PDF

Info

Publication number
WO2003009813A2
WO2003009813A2 PCT/US2002/023868 US0223868W WO03009813A2 WO 2003009813 A2 WO2003009813 A2 WO 2003009813A2 US 0223868 W US0223868 W US 0223868W WO 03009813 A2 WO03009813 A2 WO 03009813A2
Authority
WO
WIPO (PCT)
Prior art keywords
tφrs
neuropilin
cells
binding
cell
Prior art date
Application number
PCT/US2002/023868
Other languages
English (en)
Other versions
WO2003009813A3 (fr
Inventor
Gene Liau
David Ho
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to AU2002332430A priority Critical patent/AU2002332430A1/en
Publication of WO2003009813A2 publication Critical patent/WO2003009813A2/fr
Publication of WO2003009813A3 publication Critical patent/WO2003009813A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/53Ligases (6)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/71Assays involving receptors, cell surface antigens or cell surface determinants for growth factors; for growth regulators
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/9015Ligases (6)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • This invention relates to the identification of the vascular endothelial cell growth factor receptor, neuropilin, as a selective binding partner for the anti-angiogenic molecule T2-TrpRS.
  • the invention particularly relates to methods of using T2-TrpRS that exploit this discovery.
  • Angiogenesis or the proliferation of new capillaries from preexisting blood vessels, is a fundamental process necessary for embryonic development, subsequent growth, and tissue repair.
  • Angiogenesis is a prerequisite for the development and differentiation of the vascular tree, as well as for a wide variety of fundamental physiological processes including embryogenesis, somatic growth, tissue and organ repair and regeneration, cyclical growth of the corpus luteum and endometrium, and development and differentiation of the nervous system.
  • Angiogenesis additionally occurs as part of the body's repair processes, e.g. in the healing of wounds and fractures.
  • Angiogenesis is also a factor in tumor growth, because a tumor just continuously stimulate growth of new capillary blood vessels in order to grow.
  • Angiogenesis is an essential part of the growth of human solid cancer, and abnormal angiogenesis is associated with other diseases such as rheumatoid arthritis, psoriasis, and diabetic retinopathy (Folkman et al, Science 235: 442- 447 (1987)).
  • VEGF vascular endothelial growth factor
  • the neuropilin (NRP) family is composed of two homologous members, neuropilin- 1 (NRP1) and neuropilin-2 (NRP2).
  • NRP1 was first identified in certain classes of neurons as a 140-kDa transmembrane glycoprotein expressed on the tips of actively growing axons in the developing nervous system.
  • NRP2 was subsequently identified by expression cloning.
  • NRP's are high-affinity receptors for several classes of the class 3 semaphorins, secreted proteins with potent axon repulsive activity that prevent improper innervation during embryogenesis.
  • NRP's have also been demonstrated to be receptors for NEGF (Rossignol et al, Genomics 70: 211-222 (2000)).
  • ⁇ RP1 is expressed in the nervous, cardiovascular, and skeletal systems. Capillaries and blood vessel endothelial cells (EC) and the mesenchymal cells surrounding blood vessels express ⁇ RP1. After birth, ⁇ RP1 expression in neural tissues is dimished, but ⁇ RP1 expression persists in many adult tissues and notably in the placenta and heart. ⁇ P1 and ⁇ RP2 have been reported to be the only VEGF receptors expressed at the surface of tumor cells, suggesting that NRP could directly mediate as yet unidentified signals upon VEGF binding to tumor cells (Soker et al, Cell 92: 735-745 (1998)).
  • NRP1 might be a mediator of angiogenesis.
  • NRP1 is expressed in EC in vitro and is associated with blood vessels in vivo (Kitasukawa et al, Development 121: 4309-4318 (1995); Soker et al, Cell 92: 735-745 (1998)). It binds VEGF 165 , which is a potent mitogenic and chemotactic factor for EC in vitro and an angiogenic factor in vivo (Soker et al, 1998).
  • VEGF 165 a potent mitogenic and chemotactic factor for EC in vitro and an angiogenic factor in vivo (Soker et al, 1998).
  • Overexpression of NRPl in transgenic mice results in embryonic death due to excess blood vessels and hemorrhaging (Kitasukawa et al, 1995). Homozygous NRPl knockouts are embryonic lethal due to defects in the cardiovascular system, impaired neural vascularization, and disruption of
  • Aminoacyl-tRNA synthetases which catalyze the aminoacylation of tRNA molecules, are ancient proteins that are essential for decoding genetic information during the process of translation. In higher eukaryotes, nine aminoacyl- tRNA synthetases associate with at least three other polypeptides to form a supramolecular multienzyme complex (Mirande et al, Eur. J. Biochem. 147: 281-89 (1985)).
  • Each of the eukaryotic tRNA synthetases consists of a core enzyme, which is closely related to the prokaryotic counterpart of the tRNA synthetase, and an additional domain that is appended to the amino-tenmnal or carboxyl-terminal end of the core enzyme (Mirande, Prog. Nucleic Acid Res. Mol. Biol. 40: 95-142 (1991)).
  • TrpRS Trytophanyl- tRNA synthetase catalyzes a reaction that involves aminoacylation of tRNA to incorporate tryptophan into protein.
  • T ⁇ RS has been found to be secreted under apoptotic conditions in cell culture. Structural analysis has revealed that T ⁇ RS contains structures similar to the CXC-chemokine family. However, T ⁇ RS lacks the ELR motif which is essential for PMN receptor binding and cytokine activities (Wakasugi et al, Science 284: 147-151 (1999)).
  • T ⁇ RS exists as two forms, the full length T ⁇ RS and the truncated T ⁇ RS (mini-T ⁇ RS), wherein the NH 2 -terminal 48 amino acids are deleted due to alternate mRNA splicing (Fig. 1) (Wakasugi et al, Proc. Natl. Acad. Sci. 99: 173-177 (2002)).
  • T ⁇ RS and mini-T ⁇ RS are further described in International Application Nos. PCT/US01/08966 and PCT/US01/08975, both filed March 21, 2001, the disclosures of which are inco ⁇ orated herein by reference in their entireties.
  • T ⁇ RS Treatment of T ⁇ RS with PMN elastase results into two additional products, the 47 (super mini-T ⁇ RS or Tl-T ⁇ RS) and 44 (T2-T ⁇ RS) kDa fragments. Terminal amino acid analysis has revealed Ser-71 and Ser-94, respectively, as the NH 2 -terminal residues for these fragments. These molecules, Tl-T ⁇ RS and T2-T ⁇ RS, have been shown to be potent antagonists of in vivo angiogenesis (Otani et al, Proc. Natl. Acad. Sci. 99: 178-183, 2002). Tl- T ⁇ RS and T2-T ⁇ RS are further described in U.S. Provisional Application No.
  • T ⁇ RS contains a small stretch of residues in which 13 of 39 residues are identical to semaphorin-E, a ligand for the NRP receptors (Fig. 2A). While interesting, this homology does not necessarily imply functional significance. Furthermore, the Schimmel lab has previously noted that the related tyrosyl-Trna synthetase (TyrRS) may function in a manner similar to the CXC chemokines (Wakasugi et al, Science 284: 147-151 (1999)), and they hypothesized that the angiogogenic and angiostatic activities of TyrRS and T ⁇ RS maybe via chemokine receptors.
  • the present disclosure supports for the first time a specific and su ⁇ rising interaction between T2-T ⁇ RS and neuropilin and that this binding inhibits VEGF-mediated activity. Furthermore, it is demonstrated herein that this interaction effectively blocks VEGF binding to neuropilin. Thus, it is hypothesized that T2-T ⁇ RS acts as a competitive antagonist of VEGF- neuropilin interaction with consequent disruption of angiogenesis. This hypothesis is supported by the finding described herein that T2-T ⁇ RS down-regulates VEGF-induced receptor tyrosine kinase activity and HUVEC protection against apoptosis but has no effect on fibroblast growth- factor mediated biological activity on HUVEC.
  • the present invention provides a method of treating a neuropilin-mediated disease in a mammal, comprising administering to the mammal an amount of T2-T ⁇ RS or a derivative thereof effective to treat the disease.
  • the T2-T ⁇ RS or a derivative thereof comprises an amino acid sequence having at least 90% sequence identity to SEQ ID NO:3.
  • the T2-T ⁇ RS or a derivative thereof comprises an amino acid sequence having at least 95% sequence identity to SEQ ID NO:3.
  • the T2-T ⁇ RS or a derivative thereof comprises an amino acid sequence having at least 99% sequence identity to SEQ ID NO:3.
  • T2-T ⁇ RS is administered to said mammal.
  • the mammal is a primate.
  • the primate is a human.
  • the disease is cancer.
  • the cancer is selected from the group consisting of lung, colon, breast, and prostate cancer.
  • the disease is injury to the nervous system.
  • disease is rheumatoid arthritis. i another preferred embodiment, disease is a development anomaly.
  • the T2-T ⁇ RS or derivative is administered as a recombinant protein.
  • the protein is administered orally, intravenously, intramuscularly, intraperitoneally, or intrathecally.
  • the T2-T ⁇ RS or derivative is administered by administering a vector comprising a nucleotide sequence encoding the T2-T ⁇ RS or derivative.
  • the vector is a plasmid, a lipid formulation, or a viral vector.
  • the vector is administered orally, intravenously, intramuscularly, intraperitoneally, or intrathecally.
  • the present invention also provides a method for identifying an antagonist to neuropilin, comprising: (a) designing molecules that are expected to bind to a neuropilin based on the chemistry and three-dimensional structure of neuropilin and the chemistry and three- dimensional structure of T2 -T ⁇ RS; (b) contacting the molecules with neuropilin in an in vitro or cellular assay; and (c) identifying which molecules bind to neuropilin.
  • Figure 1 is a schematic representation of full-length T ⁇ RS and mini-T ⁇ RS. Numbers on the left and right represent the NH 2 and COOH terminal residues.
  • Figure 2A shows a partial primary amino acid sequence alignment of T2-T ⁇ RS (SEQ ID NO.T) and SEM (SEQ ID NO:2). Identical residues are depicted in bold.
  • FIG. 2B shows the complete amino acid sequence of T2-T ⁇ RS (SEQ ID NO:3).
  • FIG. 3 A shows that T2-T ⁇ RS specifically binds to NRP-1 and NRP-2 but not Flt-1 and KDR.
  • FIG. 3B shows that the binding of T2-T ⁇ RS is reversible by VEGF but not bFGF.
  • FIG. 4A shows that T2-T ⁇ RS does not inhibit RTK activity.
  • FIG. 4B shows that T2-T ⁇ RS inhibits VEGF-induced, but not bFGF-induced, activity on HUVEC.
  • FIG. 4C shows that T2-T ⁇ RS, but not full-length T ⁇ RS, inhibits VEGF-induced RTK activity on HUVEC.
  • FIG. 5 shows that T2-T ⁇ RS blocks VEGF-mediated HUVEC survival but has no effect on bFGF protection of HUVEC apoptosis.
  • FIG. 6 Specific and saturable binding of biotinylated T2-T ⁇ RS to HUVECs is shown by subtracting the total binding from non-specific binding. Binding constants were determined using the equation in Example 4. Number of T2-T ⁇ RS binding sites were calculated using a standard curve as outlined in Example 4. Data points represent the means (+ SEM) of three observations.
  • Figure 7A Luciferase dsRNA, NRPl/al mismatch control and NRPl/al were electrophorated into 10 HUVEC. After 24 hrs, cells were lysed and lysates electrophoresed on a SDS PAGE. Gels were transferred to nitrocellulose membranes and silencing of NRP-1 expression by the dsRNA oligonucleotides was detected by Western blot using C19 antibody that recognized the cytoplasmic domain of NRP-1.
  • Figure 7B Experimental conditions were identical to Figure 7 A except that 40 ⁇ g of the dsRNA oligonucleotides were used. Cells were plated on tissue culture dishes and at the indicated time intervals, cells were lysed and the amount of NRP-1 expression was detected as described in Example 5.
  • FIG. 8 Binding of T2-T ⁇ RS to HUVEC with RNAi was conducted as indicated in Example 5.
  • EC endothelial cell
  • cells were electroporated in the presence of PBS or with the indicated concentrations of dsRNAi oligonucleotides.
  • Cells were incubated with either 300 nM biotin T2-T ⁇ RS for total binding determination and 300 nM biotin T2-T ⁇ RS plus 15 ⁇ M of T2-T ⁇ RS for non specific binding dete ⁇ nination. Data showed here is the specific binding of T2-T ⁇ RS where total binding was subtracted from the non specific binding. Data points represent the means (+ SEM) of three observations.
  • Figure 9A shows that the chromogenic substrate S-2765 is specific to Fxa and non- reactive to HUVEC membrane, the phospholipids PC/PS nor T2-T ⁇ RS.
  • FIG. 9B shows that T2-T ⁇ RS stimulates Fxa activities in the presence of HUVEC membrane fragments but not phospholipids or buffer alone (graph labelled no membrane).
  • FIG. 10 shows that T2-T ⁇ RS recombinant protein inhibits in vivo angiogenesis in a dose-dependent manner (average matrigel plug weight was 10 mg and the amount of VEGF used in these experiments was 0.5 nM).
  • Figure 11 A Northern analysis of RNA extracted from cells transduced with 500 PPC of Av3T2T ⁇ RS shows presence of expected 1.4 Kb band.
  • FIG. 1 IB Western analysis of conditioned media (CM) and cell lysate (CL) shows presence of T2T ⁇ RS protein in both the CM and CL fractions.
  • Figure 12 is a map of pAvCsT2T ⁇ RS.
  • Figure 13 is a map of pNDSQ3.1.
  • Neuropilin means neuropilin 1 (NRPl), neuropilin 2 (NRP2) and their isoforms, which are referred to collectively as neuropilin (Rossignol et al, Genomics 70: 211-222 (2000)).
  • T ⁇ RS means tryptophanyl-tRNA synthetase.
  • Truncated tRNA synthetase polypeptides means polypeptides that are shorter than the corresponding full-length tRNA synthetase.
  • Cell culture encompasses both the culture medium and the cultured cells.
  • isolated a polypeptide from the cell culture encompasses isolating a soluble or secreted polypeptide from the culture medium as well as isolating an integral membrane protein from the cultured cells.
  • Cell extract includes culture media, especially spent culture media from which the cells have been removed.
  • a cell extract that contains the DNA or protein of interest should be understood to mean a homogenate preparation or cell-free preparation obtained from cells that express the protein or contain the DNA of interest.
  • Plasmid is an autonomous, self-replicating extrachromosomal DNA molecule and is designated by a lower case “p” preceded and/or followed by capital 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 accord with published procedures.
  • equivalent plasmids to those described are known in the art and will be apparent to the ordinarily skilled artisan.
  • “Digestion” of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA.
  • the various restriction enzymes used herein are commercially available and their reaction conditions, cofactors and other requirements were used as would be known to the ordinarily skilled artisan.
  • For analytical pu ⁇ oses typically 1 ⁇ g of plasmid or DNA fragment is used with about 2 units of enzyme in about 20 ⁇ l of buffer solution.
  • For the pu ⁇ ose of isolating DNA fragments for plasmid construction typically 5 to 50 ⁇ g of DNA are digested with 20 to 250 units of enzyme in a larger volume. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer.
  • Polynucleotide embodying the present invention can be in the form of RNA or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA.
  • the DNA can be double-stranded or single-stranded, and if single stranded can be the coding strand or non- coding (anti-sense) strand.
  • Polynucleotide encoding a polypeptide encompasses a polynucleotide that includes only coding sequence for the polypeptide as well as a polynucleotide that includes additional coding and/or non-coding sequence.
  • Oligonucleotides refers to either a single stranded polynucleotide or two complementary polynucleotide strands that can be chemically synthesized. Such synthetic oligonucleotides have no 5' phosphate and thus will not ligate to another oligonucleotide without adding a phosphate with an A TP in the presence of a kinase. A synthetic oligonucleotide will ligate to a fragment that has not been dephosphorylated.
  • amino acid residue refers to an amino acid that is part of a polypeptide.
  • the amino acid residues described herein are preferably in the L" isomeric form. However, residues in the D" isomeric form can be substituted for any L-amino acid residue, as long as the desired functional property is retained by the polypeptide.
  • NH2 refers to the free amino group present at the amino terminus of a polypeptide.
  • COOH refers to the free carboxy group present at the carboxyl terminus of a polypeptide. All amino acid residue sequences represented herein by formulae have a left to right orientation in the conventional direction of amino-terminus to carboxyl-terminus.
  • a dash at the beginning or end of an amino acid residue sequence indicates a peptide bond to a further sequence of one or more amino acid residues or to an amino- terminal group such as NH 2 or to a carboxyl-terminal group such as COOH.
  • suitable conservative substitutions of amino acids are known to those of skill in this art and can be made generally without altering the biological activity of the resulting molecule.
  • Those of skill in this art recognize that, in general, single amino acid substitutions in non-essential regions of a polypeptide do not substantially alter biological activity (see, e.g., Watson et al. Molecular Biology of the Gene, 4th Edition, 1987, The Benjamin/Cummings Pub. Co., p.224). Such substitutions are preferably made in accordance with those set forth as follows:
  • a polypeptide “variant” or “derivative” refers to a polypeptide that is a mutagenized form of a polypeptide or one produced through recombination but that still retains a desired activity, such as the ability to bind to a ligand or a nucleic acid molecule or to modulate transcription
  • a derivative of T2-T ⁇ RS refers to an alteration of the native T2-T ⁇ RS to one produced through amino acid substitution, addition, or deletion.
  • a derivative of T2-T ⁇ RS includes a polypeptide in which one or more wild-type amino acids are substituted with alternate amino acids, and includes primary sequence changes.
  • T2-T ⁇ RS that retain T2-T ⁇ RS's ability to specifically bind to neuropilin and thereby inhibit VEGF-mediated activity.
  • Such truncations may include N- terminal deletions, C-terminal deletions, and internal deletions of amino acids.
  • nucleic acid cassette refers to the genetic material of interest which can express a protein, or a peptide, or RNA after it is inco ⁇ orated transiently, permanently or episomally into a cell.
  • the nucleic acid cassette is positionally and sequentially oriented in a vector with other necessary elements such that the nucleic acid in the cassette can be transcribed and, when necessary, translated in the cell.
  • Coding plasmid describes plasmid vectors that deliver nucleic acids into a packaging cell line for stable integration into a chromosome in the cellular genome.
  • Delivery plasmid is a plasmid vector that carries or delivers nucleic acids encoding a therapeutic gene or gene that encodes a therapeutic product or a precursor thereof or a regulatory gene or other factor that results in a therapeutic effect when delivered in vivo in or into a cell line, such as, but not limited to a packaging cell line, to propagate therapeutic viral vectors.
  • vectors are described herein. For example, one vector is used to deliver particular nucleic acid molecules into a packaging cell line for stable integration into a chromosome. These types of vectors are generally identified herein as complementing plasmids.
  • a further type of vector described herein carries or delivers nucleic acid molecules in or into a cell line (e.g., a packaging cell line) for the pu ⁇ ose of propagating therapeutic viral vectors; hence, these vectors are generally referred to herein as delivery plasmids.
  • a third "type" of vector described herein is used to carry nucleic acid molecules encoding therapeutic proteins or polypeptides or regulatory proteins or are regulatory sequences to specific cells or cell types in a subject in need of treatment; these vectors are generally identified herein as therapeutic viral vectors or recombinant adenoviral vectors or viral Ad-derived vectors and are in the form of a virus particle encapsulating a viral nucleic acid containing an expression cassette for expressing the therapeutic gene.
  • the terms “homology” and “identity” are often used interchangeably.
  • degree of homology or identity can be determined, for example, by comparing sequence information using a GAP computer program.
  • the GAP program utilizes the alignment method of Needleman and Wunsch, J. Mol. Biol 48:443 (1970), as revised by Smith and Waterman, Adv. Appl Math. 2:482 (1981). Briefly, the GAP program defines similarity as the number of aligned symbols (i.e., nucleotides or amino acids) that are similar, divided by the total number of symbols in the shorter of the two sequences.
  • the preferred default parameters for the GAP program can include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745 (1986), as described by Schwartz and Dayhoff, eds., Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, pp. 353-358 (1979); (2) a penalty of 3.0 for each gap and an additional 0.10 penalty for each symbol in each gap; and (3) no penalty for end gaps.
  • nucleic acid molecules have nucleotide sequences that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% "identical” can be determined using known computer algorithms such as the "FAST A” program, using for example, the default parameters as in Pearson and Lipman, Proc. Natl. Acad. Sci. USA 85:2444 (1988).
  • the BLAST function of the National Center for Biotechnology Information database can be used to determine identity. In general, sequences are aligned so that the highest order match is obtained. "Identity" er se has an art-recognized meaning and can be calculated using published techniques.
  • identity is well known to skilled artisans (Carillo, H. & Lipton, D., SIAMJ. Applied Math. 48:1073 (1988)). Methods commonly employed to determine identity or similarity between two sequences include, but are not limited to, those disclosed in Martin J. Bishop, ed., Guide to Huge Computers, Academic Press, San Diego, (1994), and Carillo, H. & Lipton, D., SIAMJ. Applied ⁇ ath. 48:1073 (1988). Methods to determine identity and similarity are codified in computer programs.
  • Preferred computer program methods to determine identity and similarity between two sequences include, but are not limited to, GCG program package (Devereux, J., et al, Nucleic Acids Research 12(1):387 (1984), BLASTP, BLASTN, FASTA (Atschul, S.F., et al, J. Molec. Biol 215:403 (1990)).
  • test polypeptide can be defined as any polypeptide that is 90% or more identical to a reference polypeptide.
  • at least “90% identical to” refers to percent identities from 90 to 99.99 relative to the reference polypeptides. Identity at a level of 90% or more is indicative of the fact that, assuming for exemplification pu ⁇ oses a test and reference polynucleotide length of 100 amino acids are compared. No more than 10% (i.e., 10 out of 100) amino acids in the test polypeptide differs from that of the reference polypeptides.
  • Comparisons can be made between a test and reference polynucleotides. Such differences can be represented as point mutations randomly distributed over the entire length of an amino acid sequence or they' can be clustered in one or more locations of varying length up to the maximum allowable, e.g. 10/100 amino acid difference (approximately 90% identity). Differences are defined as nucleic acid or amino acid substitutions, or deletions.
  • the terms “gene therapy” and “genetic therapy” refer to the transfer of heterologous DNA to the certain cells, target cells, of a mammal, particularly a human, with a disorder or conditions for which such therapy is sought.
  • the DNA is introduced into the selected target cells in a manner such that the heterologous DNA is expressed and a therapeutic product encoded thereby is produced.
  • the heterologous DNA can in some manner mediate expression of DNA that encodes the therapeutic product, it can encode a product, such as a peptide or RNA that in some manner mediates, directly or indirectly, expression of a therapeutic product.
  • Genetic therapy can also be used to nucleic acid encoding a gene product replace a defective gene or supplement a gene product produced by the mammal or the cell in which it is introduced.
  • the introduced nucleic acid can encode a therapeutic compound, such as a growth factor inhibitor thereof, or a tumor necrosis factor or inhibitor thereof, such as a receptor therefor, that is not normally produced in the mammalian host or that is not produced in therapeutically effective amounts or at a therapeutically useful time.
  • the heterologous DNA encoding the therapeutic product can be modified prior to introduction into the cells of the afflicted host in order to enhance or otherwise alter the product or expression thereof.
  • Heterologous DNA is DNA that encodes RNA and proteins that are not normally produced in vivo by the cell in which it is expressed or that mediates or encodes mediators that alter expression of endogenous DNA by affecting transcription, translation, or other regulatable biochemical processes. Heterologous DNA can also be referred to as foreign DNA. Any DNA that one of skill in the art would recognize or consider as heterologous or foreign to the cell in which is expressed is herein encompassed by heterologous DNA. Examples of heterologous DNA include, but are not limited to, DNA that encodes traceable marker proteins, such as a protein that confers drug resistance, DNA that encodes therapeutically effective substances, such as anti-cancer agents, enzymes and hormones, and DNA that encodes other types of proteins, such as antibodies.
  • heterologous DNA refers to a DNA molecule not present in the exact orientation and position as the counte ⁇ art DNA molecule found in the corresponding wild-type adenovirus. It can also refer to a DNA molecule from another organism or species (i.e., exogenous) or from another Ad serotype.
  • “Therapeutically effective DNA product” is a product that is encoded by heterologous DNA so that, upon introduction of the DNA into a host, a product is expressed that effectively ameliorates or eliminates the symptoms, manifestations of an inherited or acquired disease or that cures said disease.
  • DNA encoding the desired heterologous DNA is cloned into a plasmid vector and introduced by routine methods, such as calcium-phosphate mediated DNA uptake or microinjection, into producer cells, such as packaging cells. After amplification in producer cells, the vectors that contain the heterologous DNA are introduced into selected target cells.
  • “Expression or delivery vector” refers to any plasmid or virus into which a foreign or heterologous DNA can be inserted for expression in a suitable host cell- i.e., the protein or polypeptide encoded by the DNA is synthesized in the host cell's system.
  • Vectors capable of directing the expression of DNA segments (genes) encoding one or more proteins are referred to herein as "expression vectors.” Also included are vectors that allow cloning of cDNA (complementary DNA) from mRNAs produced using reverse transcriptase.
  • Gene is a nucleic acid molecule whose nucleotide sequence encodes RNA or polypeptide.
  • a gene can be either RNA or DNA. Genes can include regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).
  • isolated with reference to a nucleic acid molecule, polypeptide, or other biomolecule, means that the nucleic acid or polypeptide has separated from the genetic environment from which the polypeptide or nucleic acid was obtained. It can also mean altered from the natural state. For example, a polynucleotide or a polypeptide naturally present in a living animal is not “isolated,” but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is "isolated", as the term is employed herein. Thus, a polypeptide or polynucleotide produced and/or contained within a recombinant host cell is considered isolated.
  • isolated polypeptide or an “isolated polynucleotide” are polypeptides or polynucleotides that have been purified, partially or substantially, from a recombinant host cell or from a native source.
  • a recombinantly produced version of a compounds can be substantially purified by the one-step method described in Smith and Johnson, Gene 67:31-40 (1988).
  • isolated and “purified” are sometimes used interchangeably.
  • Such polynucleotide could be part of a vector and/or such polynucleotide or polypeptide could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
  • isolated polynucleotide is meant that the nucleic acid is free of the coding sequences of those genes that, in the naturally occurring genome of the organism (if any) immediately flank the gene encoding the nucleic acid of interest.
  • Isolated DNA can be single- stranded or double-stranded, and can be genomic DNA, cDNA, recombinant hybrid DNA, or synthetic DNA. It can be identical to a native DNA sequence, or can differ from such sequence by the deletion, addition, or substitution of one or more nucleotides.
  • isolated or purified as it refers to preparations made from biological cells or hosts means any cell extract containing the indicated DNA or protein including a crude extract of ' the DNA or protein of interest.
  • a purified preparation in the case of a protein, can be obtained following an individual technique or a series o preparative or biochemical techniques and the DNA or protein of ' interest can be present at various degrees o/purity in these preparations.
  • the procedures can include for example, but are not limited to, ammonium sulfate fractionation, gel filtration, ion exchange change chromatography, affinity chromatography, density gradient centrifugation and electrophoresis.
  • a preparation of DNA or protein that is "substantially pure” or “isolated” means a preparation free from naturally occurring materials with which such DNA or protein is normally associated in nature. "Essentially pure” should be understood to mean a “highly” purified preparation that contains at least 95 % of the DNA or protein of interest.
  • Packaging cell line is a cell line that provides a missing gene product or its equivalent.
  • Adenovirus viral particle is the minimal structural or functional unit of a virus.
  • a virus can refer to a single particle, a stock of particles or a viral genome.
  • the adenovirus (Ad) particle is relatively complex and can be resolved into various substructures.
  • PRE Post-transcription regulatory element
  • viral or cellular messenger RNA that is not spliced, i.e. intronless messages. Examples include, but are not limited to, human hepatitis virus, woodchuck hepatitis virus, the TK gene and mouse histone gene.
  • the PRE can be placed before a polyA sequence and after a heterologous DNA sequence
  • “Pseudotyping” describes the production of adenoviral vectors having modified capsid protein or capsid proteins from a different serotype than the serotype of the vector itself.
  • One example is the production of an adenovirus 5 vector particle containing an Ad37 fiber protein. This can be accomplished by producing the adenoviral vector in packaging cell lines expressing different fiber proteins.
  • Promoters of interest herein can be inducible or constitutive. Inducible promoters will initiate transcription only in the presence of an additional molecule; constitutive promoters do not require the presence of any additional molecule to regulate gene expression, a regulatable or inducible promoter can also be described as a promoter where the rate or extent of RNA polymerase binding and initiation is modulated by external stimuli. Such stimuli include, but are not limited to various compounds or compositions, light, heat, stress and chemical energy sources. Inducible, suppressible and repressible promoters are considered regulatable promoters. Preferred promoters herein, are promoters that are selectively expressed in ocular cells, particularly photoreceptor cells.
  • Receptor refers to a biologically active molecule that specifically binds to (or with) other molecules.
  • receptor protein can be used to more specifically indicate the proteinaceous nature of a specific receptor.
  • Recombinant refers to any progeny formed as the result of genetic engineering. This can also be used to describe a virus formed by recombination of plasmids in a packaging cell.
  • Transgene or “therapeutic nucleic acid molecule” includes DNA and RNA molecules encoding an RNA or polypeptide. Such molecules can be “native” or naturally derived sequences; they call also be “non-native” or “foreign” that are naturally or recombinantly derived.
  • the term “transgene,” which can be used interchangeably herein with the term “therapeutic nucleic acid molecule,” is often used to describe a heterologous or foreign (exogenous) gene that is carried by a viral vector and transduced into a host cell.
  • Therapeutic nucleotide nucleic acid molecules include antisense sequences or nucleotide sequences that can be transcribed into antisense sequences.
  • Therapeutic nucleotide sequences all include nucleic acid molecules that function to produce a desired effect in the cell or cell nucleus into which said therapeutic sequences are delivered.
  • a therapeutic nucleic acid molecule can include a sequence of nucleotides that encodes a functional protein intended for delivery into a cell which is unable to produce that functional protein.
  • Promoter region refers to the portion of DNA of a gene that controls transcription of the DNA to which it is operatively linked.
  • the promoter region includes specific sequences of DNA that are sufficient for RNA polymerase recognition, binding and transcription initiation. This portion of the promoter region is referred to as the promoter.
  • the promoter region includes sequences that modulate this recognition, binding and transcription initiation activity of the RNA polymerase. These sequences can be cis acting or can be responsive to trans acting factors. Promoters, depending upon the nature of the regulation, can be constitutive or regulated.
  • “Operatively linked” means that the sequences or segments have been covalently joined into one piece of DNA, whether in single or double stranded form, whereby control sequences on one segment control expression or replication or other such control of other segments.
  • the two segments are not necessarily contiguous, however.
  • “Complex” as used herein refers to the product of a specific binding reaction such as an antibody-antigen or receptor-ligand reaction. Exemplary complexes are immunoreaction products.
  • Label and" Indicating means in their various grammatical forms refer to single atoms and molecules that are either directly or indirectly involved in the production of a detectable signal to indicate the presence of a complex. Any label or indicating means can be linked to or inco ⁇ orated in an expressed protein, polypeptide, or antibody molecule that is part of an antibody or monoclonal antibody composition of the present invention or used separately, and those atoms or molecules can be used alone or in conjunction with additional reagents. Such labels are themselves well known in clinical diagnostic chemistry and constitute a part of this invention only insofar as they are utilized with otherwise novel proteins methods and/or systems.
  • the present invention includes methods for the treatment of all diseases impacted by T2-T ⁇ RS or T2-T ⁇ RS derivatives via their interaction with neuropilins.
  • the T2- T ⁇ RS or derivatives can be delivered as a recombinant protein or by gene therapy vectors including but not limited to adenoviral, AAV, HS V vector, retroviral, lentiviral, and plasmid vectors.
  • the invention further includes small molecules or other drugs based on this interaction.
  • the present invention further provides for the use of surrogate markers such as ove ⁇ roduction of VEGF to target specific cancers for treatment and evaluation of T2-T ⁇ RS- based treatment.
  • one aspect of the present invention is directed to the use of T2-T ⁇ RS, its derivatives, modifications, or small molecules or drugs based on this interaction for the treatment of cancer because neuropilin- 1 is known to be expressed by newly formed blood vessels and by tumor cells, including but not limited to metastatic prostate tumor cells, breast carcinomas, and melanoma (Soker et al, Cell 92:735-745 (1998); Latil et al, Int. J. Cancer 89: 167-171 (2000)).
  • Neuropilin expression by tumor cells promotes tumor angiogenesis and progression (Miao et al, FASEB J. 14: 2532-2539 (2000)).
  • increased expression of endothelial neuropilin is associates with neuroblastoma stages I-IV (Fakhari et al, Cancer 94(1): 258-263 (2001)).
  • T2 -T ⁇ RS T2 -T ⁇ RS
  • its derivatives, modifications, or small molecules or drugs based on this interaction for the treatment of nervous system injury
  • semaphorin 3 A sem3 A
  • neuropilin 1 prolonged binding of semaphorin 3 A (sem3 A) to neuropilin 1 induces cell death (Bagnard et al, J. ofNeurosci. 21(10): 3332-41 (2001)).
  • T2-T ⁇ RS can act as an antagonist of sem3A-nuropilin-l interaction to prevent apoptosis and promote cell survial, and possibly proliferation of neuronal stem cells.
  • a second example is that the interaction between semaphorin IV (semlV) and neurophilin-2 defines specific repulsive axon guidance events in vivo (Kolodkin et al, Neuron 21: 1079-92 (1998)); thus, T2-T ⁇ RS can bind to neuropilin-2 and therapeutically modify this process.
  • Another aspect of the present invention is directed to the use of T2-T ⁇ RS, its derivatives, modifications, or small molecules or drugs based on this interaction for the treatment of rheumatoid arthritis.
  • Neuropilin up-regulation has been detected in the synovial tissues of rheumatoid arthritis patients and this correlated with increased vascular density (Ikeda et al, J. of Path. 191: 426-433 (2000)).
  • T2-T ⁇ RS T2-T ⁇ RS
  • its derivatives, modifications, or small molecules or drugs based on this interaction for the treatment of organogenesis and development anomalies.
  • semaphorin III nerves, bones, and heart
  • neuropilin-1 blood vessel
  • a double knockout of NRP-1 and NRP -2 revealed more severe vascular defects, suggesting that both are required for appropriate blood vessel development (Takashima et al, Proc. Natl. Acad. Sci. 99: 3657-3662 (2002)).
  • T2-T ⁇ RS T2-T ⁇ RS
  • KDR/Flk-1 ocular-related diseases
  • ocular-related diseases such as Proliferate Diabetic Retinopathy and Age Related Macular Degeneration
  • neuropilin-1 and KDR/Flk-1 have been found to co-localized in the area of neovascularized vessels of the retina (Maramatsu et al, Inves Ophthalmol Vis. Sci. 42(6): 1172-8 (2001); Ikeda et al, Inves Ophthalmol Vis. Sci. 41(7): 1649-56 (2000)).
  • Another aspect of the present invention is directed to the use use of T2-T ⁇ RS, its derivatives, modifications, or small molecules or drugs based on this interaction for the treatment of angiogenesis diseases associated with inappropriate arterial- venous junctions or conversion such as in vein graft stenosis, because neuropilin-1 is preferentially express by arterial and neuropilin-2 is preferentially expressed by venous blood vessels (Herzog et al, Mech. Dev. 109 (1): 115-9 (2001)).
  • T2 -T ⁇ RS discovery of the mechanism of action of T2 -T ⁇ RS creates the opportunity to model the molecule and design even more potent antagonists of angiogenesis to treat angiogenesis-related diseases and potentially other diseases mediated by neuropilin. It is also envisioned that this discovery enables identification of small molecule antagonists by further defining the interaction. For example, using peptides and site-directed mutagenesis to identify the sequences or residue(s) that are important for T2-T ⁇ RS binding to neuropilin would allow the creation of novel molecules with a better binding interaction. By understanding the charge- charge interactions and hydrophobicity/hydrophilicity required for binding, it would be possible to design a small molecule agonist or antagonist of the interaction.
  • T2-T ⁇ RS contains a region of high sequence of homology to semaphorin E (SemE), a binding partner for neuropilin (see Figure 2A).
  • SemE semaphorin E
  • FIG. 2A The N-terminal of T2-T ⁇ RS contains a region of high sequence of homology to semaphorin E (SemE), a binding partner for neuropilin (see Figure 2A).
  • SemE semaphorin E
  • FIG. 2A The N-terminal of T2-T ⁇ RS contains a region of high sequence of homology to semaphorin E (SemE), a binding partner for neuropilin (see Figure 2A).
  • T2-T ⁇ RS Tyrioo -> Lys or T2-T ⁇ RS Phe 107 ->Ile could alter the charge-to-charge interaction between the binding surfaces and enhance T2-T ⁇ RS binding to neuropilin.
  • the findings described herein indicate a common neuropilin binding element between the semaphor
  • neuropilin belongs to a large class of receptors that contain CUB and blood coagulation factor V/VIII (F V/NIII) domains.
  • CUB domains are widespread in other proteins and are important in developmental regulation.
  • CUB domains have been found in several classes of proteases, such as spermadhesins, procollagen C-proteinase, and the complement system proteins.
  • Database mining indicates that there are other proteins that share sequence homology to the CUB domain of neuropilin. We have identified these as endothelial and smooth muscle cell-derived neuropilin-like molecule (ESDN), neuropilin and tolloid-like 1 and 2 (NETO1 andNETO2) (Kobuke et al, J. Biol. Chem.
  • NEGF neuropilin binding to neuropilin has been found to reside within the bl/b2 domain and the binding is enhanced by the al/a2 domain (J. Biol. Chem. 277(27): 24818-25 (July 5, 2002)). Because NEGF binds to neuropilin and is able to compete with T2-T ⁇ RS for neuropilin binding, it is likely that the al/a2 and/or bl/b2 domains of neuropilin are involve in the binding to T2-T ⁇ RS. Therefore, factor N, factor NIII, ⁇ ETO1. ⁇ ETO2 and ESDN may represent additional binding partners for T2-T ⁇ RS and represent novel activities of T2-T ⁇ RS.
  • the blood protein factor Xa (Fxa) binds to the membrane bound EPR-1 protein that is similar to the light chain of factor Na (Altieri, FASEB J. 9: 860-865 (1995)); this protein can act as a cofactor for Fxa to catalyze prothrombin activation in the absence of added factor Na (Ambrosini et al, J. Biol. Chem. 271: 1243-1248 (1996).
  • T2-T ⁇ RS may be involved in the regulation of blood coagulation through their interactions with factor Va or factor Va-like protein such as EPR-1.
  • Suitable D ⁇ A viral vectors for use herein includes, but are not limited to an adenovirus (Ad), adeno-associated virus (AAV), he ⁇ es virus, vaccinia virus or a polio virus.
  • a suitable R ⁇ A virus for use herein includes but is not limited to a retrovirus or Sindbis virus. It is to be understood by those skilled in the art that several such D ⁇ A and R ⁇ A viruses exist that may be suitable for use herein.
  • Adenoviral vectors have proven especially useful for gene transfer into eukaryotic cells and are widely available to one skilled in the art and is suitable for use herein.
  • Adeno-associated virus has been used as a gene transfer system with applications in gene therapy. See U.S. Patents Nos. 5,139,941; 5,436,146; and 5,622,856. He ⁇ es simplex virus type-1 (HSV-1) vectors are available and are especially useful in the nervous system because of its neurotropic property. See U.S. Patent No. 5,288,641. Vaccinia viruses, of the poxvirus family, have also been developed as expression vectors. Each of the above-described vectors is widely available and is suitable for use herein. Retroviral vectors are capable of infecting a large percentage of the target cells and integrating into the cell genome.
  • Preferred retroviruses include lentiviruses, and also include, but are not limited to, HIV, BLV and SIN. See U.S. Patents ⁇ os.5,665,577; 5,994,136; 6,013,516; 5,672,510; 5,707,865 and 5,817,491.
  • viral vectors that can be used for gene therapy as taught herein include adenovirus (See U.S. Patent No.5,935,935), he ⁇ es virus, vaccinia, adeno-associated virus (AAV), or, preferably, an RNA virus such as a retrovirus, and also include a modified viral vector, such as an adenovirus, known as a "gutless" vector.
  • the retroviral vector is a derivative of a murine or avian retrovirus, or is a lentiviral vector.
  • the preferred retroviral vector is a lentiviral vector.
  • retroviral vectors in which a single foreign gene can be inserted include, but are not limited to: Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTN), SIN, BIN, HIN and Rous Sarcoma Virus (RSV).
  • MoMuLV Moloney murine leukemia virus
  • HaMuSV Harvey murine sarcoma virus
  • MuMTN murine mammary tumor virus
  • SIN BIN
  • HIN Rous Sarcoma Virus
  • RSV Rous Sarcoma Virus
  • Retroviral vectors can be made target specific by inserting, for example, a polynucleotide encoding a protein. Preferred targeting is accomplished by using an antibody to target the retroviral vector.
  • Those of skill in the art know of, or can readily ascertain without undue experimentation, specific polynucleotide sequences which can be inserted into the retroviral genome to allow target specific delivery of the retroviral vector containing the zinc fmger-nucleotide binding protein polynucleotide. Because recombinant retroviruses are defective, they require assistance in order to produce infectious vector particles.
  • helper cell lines that contain plasmids encoding all of the structural genes of the retrovirus under the control of regulatory sequences within the LTR. These plasmids are missing a nucleotide sequence which enables the packaging mechanism to recognize an RNA transcript for encapsidation.
  • Helper cell lines which have deletions of the packaging signal include but are not limited to 2, PA317 and PA12, for example. These cell lines produce empty virions, since no genome is packaged. If a retroviral vector is introduced into such cells in which the packaging signal is intact, but the structural genes are replaced by other genes of interest, the vector can be packaged and vector virion produced. The vector virions produced by this method can then be used to infect a tissue cell line, such as NIH 3T3 cells, to produce large quantities of chimeric retroviral virions.
  • the viral vector used is a "gutless" adenoviral vector.
  • Such vectors are devoid of all viral coding regions and contain only the essential adenovirus packaging signals and the transgene expression cassette. (See Example 6). They may be prepared by techniques known to those of skill in the art. Sandig et al. PNAS 97:1002-1007 (2000)
  • Non- viral delivery techniques for gene therapy include DNA-ligand complexes, adenovirus-ligand-DNA complexes, direct injection of DNA, CaPO 4 precipitation, gene gun techniques, electroporation, liposomes and lipofection. Any of these methods are available to one skilled in the art and would be suitable for use herein. Other suitable methods are available to one skilled in the art, and it is to be understood that the herein may be accomplished using any of the available methods of transfection.
  • colloidal dispersion systems include macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes, which are preferred.
  • Liposomes are artificial membrane vesicles which are useful as delivery vehicles in vitro and in vivo. It has been shown that large unilamellar vesicles (LUN), which range in size from 0.2-4.0 ⁇ m can encapsulate a substantial percentage of an aqueous buffer containing large macromolecules. RNA, DNA and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley et al, Trends Biochem. Sci., 6:77, 1981).
  • LUN large unilamellar vesicles
  • Lipofection may be accomplished by encapsulating an isolated nucleic acid molecule within a liposomal particle and contacting the liposomal particle with the cell membrane of the target cell.
  • Liposomes are self-assembling, colloidal particles in which a lipid bilayer, composed of amphiphilic molecules such as phosphatidyl serine or phosphatidyl choline, encapsulates a portion of the surrounding media such that the lipid bilayer surrounds a hydrophilic interior.
  • Unilammellar or multilammellar liposomes can be constructed such that the interior contains a desired chemical, drug, or, as provide herein, an isolated nucleic acid molecule.
  • Liposomes have been used for delivery of polynucleotides in plant, yeast and bacterial cells as well as mammalian cells.
  • characteristics among the following should be present: (1) encapsulation of the genes of interest at high efficiency while not compromising their biological activity; (2) preferential and substantial binding to a target cell in comparison to non-target cells; (3) delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic information (Mannino et al, Biotechniques, 6:682, 1988).
  • the composition of the liposome is usually a combination of phospholipids, particularly high-phase-transition-temperature phospholipids, usually in combination with steroids, especially cholesterol. Other phospholipids or other lipids may also be used.
  • the physical characteristics of liposomes depend on pH, ionic strength, and the presence of divalent cations.
  • lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and gangliosides. Particularly useful are diacylphosphatidylglycerols, where the lipid moiety contains from 14-18 carbon atoms, particularly from 16-18 carbon atoms, and is saturated.
  • Illustrative phospholipids include egg phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphosphatidylcholine.
  • the targeting of liposomes has been classified based on anatomical and mechanistic factors.
  • Anatomical classification is based on the level of selectivity, for example, organ- specific, cell-specific, and organelle-specific.
  • Mechanistic targeting can be distinguished based upon whether it is passive or active. Passive targeting uses the natural tendency of liposomes to distribute to cells of the reticulo-endothelial system (RES) in organs which contain sinusoidal capillaries.
  • RES reticulo-endothelial system
  • Active targeting involves alteration of the liposome by coupling the liposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein, or by changing the composition or size of the liposome in order to achieve targeting to organs and cell types other than the naturally occurring sites of localization.
  • a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein
  • the surface of the targeted delivery system may be modified in a variety of ways.
  • lipid groups can be inco ⁇ orated into the lipid bilayer of the liposome in order to maintain the targeting ligand in stable association with the liposomal bilayer.
  • Various linking groups can be used for joining the lipid chains to the targeting ligand.
  • the compounds bound to the surface of the targeted delivery system are ligands and receptors permitting the targeted delivery system to find and "home in" on the desired cells.
  • a ligand may be any compound of interest that interacts with another compound, such as a receptor.
  • Antibodies are preferred receptors. Antibodies can be used to target liposomes to specific cell-surface ligands. For example, certain antigens expressed specifically on tumor cells, referred to as tumor-associated antigens (TAAs), may be exploited for the pu ⁇ ose of targeting antibody-zinc finger-nucleotide binding protein-containing liposomes directly to the malignant tumor. Since the zinc finger-nucleotide binding protein gene product may be indiscriminate with respect to cell type in its action, a targeted delivery system offers a significant improvement over randomly injecting non-specific liposomes.
  • TAAs tumor-associated antigens
  • Antibody-targeted liposomes can include monoclonal or polyclonal antibodies or fragments thereof such as Fab, or F(ab') 2 , as long as they bind efficiently to an the antigenic epitope on the target cells. Liposomes may also be targeted to cells expressing receptors for hormones or other serum factors.
  • the cells may be transfected in vivo, ex vivo or in vitro.
  • the cells may be transfected as primary cells isolated from a patient or a cell line derived from primary cells, and are not necessarily autologous to the patient to whom the cells are ultimately administered. Following ex vivo or in vitro transfection, the cells may be implanted into a host. Genetic modification of the cells may be accomplished using one or more techniques well known in the gene therapy field (see, e.g., (1994) Human Gene Therapy 5:543-563).
  • nucleic acid molecules provided herein to a target cell in vivo may be accomplished using any of a variety of techniques well known to those skilled in the art.
  • the vectors of the herein may be administered orally, parentally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles.
  • Suppositories for rectal administration of the drag can be prepared by mixing the drug with a suitable non-irritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and therefore melt in the rectum and release the drug.
  • the dosage regimen for treating a disorder or a disease with the vectors and/or compositions provided is based on a variety of factors, including the type of disease, the age, weight, sex, medical condition of the patient, the severity of the condition, the route of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined empirically using standard methods.
  • the pharmaceutically active compounds can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals.
  • the pharmaceutical composition may be in the form of, for example, a capsule, a tablet, a suspension, or liquid.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a given amount of DNA or viral vector particles (collectively referred to as "vector").
  • vector may contain an amount of vector from about 10 3 -10 15 viral vector particles, preferably from about 10 6 -10 12 viral particles.
  • a suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods.
  • the vector may also be administered by injection as a composition with suitable carriers including saline, dextrose, or water.
  • nucleic acids and /or vectors herein can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more vectors or other agents.
  • the therapeutic agents can be formulated as separate compositions that are given at the same time or different times, or the therapeutic agents can be given as a single composition.
  • Ligands similarly may be delivered by any suitable mode of administration, including by oral, parenteral, intravenous, intramuscular and other known routes. Any known pharmaceutical formulations is contemplated.
  • EXAMPLE 1 Binding of T2-T ⁇ RS to NRP is Specific and Can Be Competed by VEGF
  • Binding assays were performed in 96 well PolySorb plates (Fisher Scientific, Newark, DE). Wells were absorbed with 0.5 ⁇ M of test protein for 2 hrs at room temperature, washed 3 times with PBS, and incubated with 200 ⁇ l of 0.2% Tween 20 and 0.5% BSA in PBS for one hr to block non-specific binding sites. The wells were then washed 3 times with washing buffer (0.05%) Tween 20, 0.5% BSA in PBS). After washing, recombinant Biotin T2-T ⁇ RS was added and binding was allowed to occur overnight at 4°C. Wells were washed 3 times with wash buffer to remove unbound T2-T ⁇ RS.
  • T2-T ⁇ RS 500 nM was mixed with VEGF at the indicated concentrations and incubated on soluble NRP-1 (sNRP-1) containing plate and treated as above. Avidin-HRP (Sigma, St. Louis, MO) was used to detect the bound T2-T ⁇ RS. The wells were washed 3 times with PBS and the amount of T2- T ⁇ RS bound was detected colormetrically by reading A 15 .
  • T2 -T ⁇ RS binding to sNRP-1 was examined utilising ELISA based binding assays.
  • the K for T2-T ⁇ RS binding to NRP-1 was determined to be 50- 160 nM in three separate binding experiments and the K ⁇ j for T2-T ⁇ RS binding to NRP-2 was determined to be 20-100 nM in two separate binding experiments; whereas, the binding of T2- T ⁇ RS to Flt-1 and KDR remained low and statistically indistinguishable from back ground binding (Fig. 3A).
  • VEGF can bind to NRP-1 with high affinity
  • VEGF as well as bFGF (used as a negative control) were used in a competition binding experiment to determine their capacities to inhibit T2-T ⁇ RS binding to NRP-1. It was found that VEGF, but not bFGF, inhibited T2-T ⁇ RS binding to sNRP-1 with an IC 50 ⁇ 100 nM (Fig. 3B) which is similar to the K ⁇ j of T2-T ⁇ RS binding to sNRP-1. Despite the fact that K and IC 50 values are incomparable, it is apparent that the saturable binding observed with T2-T ⁇ RS to sNRP-1 is readily reversible with VEGF. Thus, it is postulated that these molecules share at least one domain on the same binding surface on the sNRP-1 molecule.
  • VEGF competes with T2-T ⁇ RS for binding to NRP-1 : IC 50 ⁇ 100 nM.
  • EXAMPLE 2 T2-T ⁇ RS Inhibits VEGF-induced Receptor Tyrosine Kinase Activity
  • Receptor tyrosine kinase (RTK) assays were conducted according to method by Schraag et al, Anal. Biochem. 221: 233-239 (1993) with minor modifications. Briefly, HUVECs (passages 5-7) membrane fragments were prepared from endothelial cells grown to ⁇ 90% confluency. Cells were detached from the surface with lOmM EDTA in PBS and centrifuged at 1000 ⁇ m for 5 min. The cell pellet was resuspended in 5 ml of extraction buffer (PBS, Protease inhibitors, 0.2nM Na 3 VO 4 , 5mM mercaptoethanol, ImM EDTA) and lysed by homogenizing for 2 min.
  • PBS Protease inhibitors, 0.2nM Na 3 VO 4 , 5mM mercaptoethanol, ImM EDTA
  • the extract was transferred to microcentrifuge tubes and centrifuged for 5 minutes at maximum speed (14,000 ⁇ m). The supernatant was centrifuged twice at 100,000 g to pellet the cell membrane.
  • Cell membrane was resuspended in PBS and total protein concentration was determined using the BCA protein determination method (BioRad, Hercules, CA). Cell membrane was flash freeze and kept frozen until use.
  • the RTK activity of the extracts was determined using Oncogen's protein tyrosine kinase assay kit (Oncogene, Boston, MA). Essentially, the membrane preparations were incubated on plates containing polymeric tyrosine residues that serve as substrates for RTKs. After a 30-minute incubation, the reaction was terminated and the amount of phosphorylated tyrosine quantitated by binding to an HRP- conjugated phosphotyrosine specific antibody. Experiments were done in triplicate.
  • T2-T ⁇ RS was examined in the RTK activity assay and found to have no effect on basal RTK activity in the range of concentrations tested (Fig. 4A). However, T2-T ⁇ RS was a potent inhibitor of VEGF-induced RTK activity and suppressed VEGF activity with an IC 50 of -lOOnM (Fig. 4A). T2-T ⁇ RS blocked VEGF induced RTK stimulation (Fig. 4A and B) but had no effect on bFGF-induced RTK activity (Fig. 4B). The readings were converted into relative percent of RTK activity to allow comparison between VEGF and FGF. Full length T ⁇ RS was also tested and found to have no effect on VEGF-induced membrane RTK activity (Fig. 4C).
  • VEGF 0.5 nM
  • T2-T ⁇ RS 100 nM
  • Apoptosis assays were conducted according to methods described by Gerber et al, J. Biol. Chem. 273: 30336-30343 (1998); Kanellis et al, Am. J. Physiol. Renal Physiol 278: F905-915 (2000); Kwak et al, FEBSLett. 448: 249-253 (1999); and Mesri et al, Am. J. Pathol. 158: 1757-1765 (2001) with minor modifications. Briefly, Human umbilical vein endothelial cells (HUVECs) (Vec Technologies, Inc.
  • Rensselaer, NY at passage 7 were seeded at 10 4 per well in a 96-well flat bottom plate (CoStar, Corning, New York) in MCDB131 medium (Vec Technologies, Inc. Rensselaer, NY) and incubated in a 37°C, 5%> CO 2 humidified incubator overnight. Cells were incubated in MCDB131 and 10% FBS (BioWittaker, Walkersville, MD) for 24 hours. Wells were washed twice with 37°C HBSS prior to the addition of the treatments. All treatments were added in triplicate in a final volume of 200 ⁇ l of Ml 99 per well. The cells were incubated at 37°C for 24 hr and the degree of apoptosis was determined by ELISA.
  • the Cell Death Detection ELISA PLUS (Roche Molecular Biochemicals, Germany) is a photometric enzyme-immunoassay for the in vitro determination of cytoplasmic histone-associated-DNA fragments after cell death. Briefly, a 20 ⁇ l aliquot of cell lysate was placed into a streptavidin- coated MTP plate followed by the addition of 80 ⁇ l of an anti-histone-biotin and anti-DNA- POD mixture (Roche Molecular Biochemicals, Germany). After a 2 hour incubation the unbound antibodies were washed off.
  • the amount of nucleosomes retained by the POD was determined photometrically at 405 nm on a plate reader (Spectra Max, Molecular Devices) with ABTS as the substrate (reference wavelength at 490 nm).
  • the numerical values obtained from A4 05 minus the reference A 490 readings are used as the apoptosis index. Experiments were done in triplicates. The effect of T2-T ⁇ RS on HUVEC apoptosis alone or in combination with 0.5 nM VEGF or bFGF was examamined.
  • T2-T ⁇ RS When T2-T ⁇ RS was added at concentrations between 0.01- l,000nM, alone or in the presence of either VEGF or bFGF, T2-T ⁇ RS consistently enhanced the level of endothelial cell apoptosis in the presence of VEGF, but not bFGF (Fig. 5).
  • the apoptosis index was -1.0, as the concentration of T2-T ⁇ RS increased, the apoptosis index increased to a maximum of 2.2 (Fig. 5).
  • the concentration in which T2-T ⁇ RS induced 50%> of cell death (IC 5 o) was estimated to be ⁇ 100nM. The results are summarized in the following table.
  • VEGF 0.5 nM
  • T2-T ⁇ RS 100 nM
  • the nonradioactive cell binding assay was adapted from method by Vieira A. (Molecidar Biotechnology 247(10): 247-50 (1998)).
  • the following buffers were prepared before the experiment: ST Buffer (1 nM EDTA, 50 mM NaCl, 100 mM Tris-HCL, 0.1% SDS, 1% Triton X-100, 0.2% BSA, pH 7.4); Buffer A (200 mM HEPES, 100 mM potassium acetate, 85 mM sucrose, 1 mM magnesium acetate, pH 7.4); Buffer Bl (200 mM HEPES, 100 mM potassium acetate, 75 mM sucrose, 1 mM magnesium acetate, pH 7.4); Buffer B2 (200 mM HEPES, 100 mM potassium acetate, 1 mM magnesium acetate, pH 7.4); Binding Buffer (200 mM HEPES, 100 mM potassium acetate, 1 mM magnesium acetate,
  • the ligand-capture plates were prepared as follow: The ELISA plate was coated with 100 ⁇ l of 10 nM anti 6 His tag monoclonal Ab diluted in PBS for 2 hrs at room temperature wells were blocked with 200 ⁇ l PBS, 0.5% BSA, 0.2% Tween 20 for 2 hrs at room temperature.
  • Perforated HUVEC's were prepared as follows: Passage cells at 70% confluency in a 150 mm dish ( ⁇ 3.5 x 10 6 cells). Cells were washed three times with cold Buffer A, then scraped and collected in a siliconized tube containing 40 ml of ice-cold Buffer A. Cells were resuspended and centrifuged at 1000 g for 5 min. at 4°C, the supernatant was discarded and the pellet was resuspended in 2 volume of Buffer Bl. Cells were frozen and kept in liquid nitrogen.
  • Biotin T2-T ⁇ RS was prepared as follows: Biotinylation of T2-T ⁇ RS was conducted as outlined in the protocol provided by the manufacture (Pierce, Rockford, IL). Briefly, 100 ⁇ l of the stock T2-T ⁇ RS (2 mg/ml) was incubated with 50 fold molar excess of the Sulfo-NHS- biotin for 2 hrs on ice. The unbound biotin was removed by a desalting column and the amount of biotin bound to T2-T ⁇ RS was calculated to be 1:1 molar ratio using the HABA method for determining biotin inco ⁇ oration.
  • the binding assay was done as follows: Cells were thawed on ice followed by a 5 second centrifuge at 3500 g. The supernatant was removed and cells were resuspended in ice- cold Buffer B2 at the concentration of ⁇ 5 x 10 6 cells /ml. For total binding, the biotinylated T2- T ⁇ RS was added and for non-specific binding, biotin-T2-T ⁇ RS was added with 100 fold molar excess of unlabelled T2-T ⁇ RS. The final volume of 100 ⁇ l contained 500,000 cells reaction mixture tubes was gently mixed and incubate at 4°C for 3 hr.
  • T2-T ⁇ RS The binding of T2-T ⁇ RS to HUVECs was detected as follows: The reaction mixture tubes were centrifuged for 5 seconds at 14,000 ⁇ m to pellet cells. Pellet was washed three times in 1 ml of ice-cold Binding Buffer then centrifuged for 5 seconds to remove supernatant. Cells were lysed by adding 200 ⁇ l of ST Buffer. 50 ⁇ l of cell lysates was added to the wells of the ligand capture plate and incubated overnight with gentle rocking. To detect binding, wells were washed with 200 ⁇ l PBS, 0.5% BSA, 0.05% Tween 20 three times.
  • T2- T ⁇ RS bound was detected by adding Avidin-HRP followed by p-nitropheyl phosphate to develop a color reaction which was measured at 415 nm with an ELISA plate reader.
  • the amount of T2-T ⁇ RS bound was converted into number of moles using the standard curve and divided by the number of cells to determine to number of T2-T ⁇ RS binding site per cell.
  • T2-T ⁇ RS binding to HUNECs was examined utilising perforated cell based binding assays.
  • the binding assays indicated that T2-T ⁇ RS binding to HUNECs was saturable and specific. Total binding of T2-T ⁇ RS binding to HUNECs was dete ⁇ nined. In the presence of 50-100 fold molar excess of unlabeled T2-T ⁇ Rs, non-specific binding was demonstrated. Subtracting total from non-specific binding curves, the specific binding of T2-T ⁇ RS to HUNECs was determined to be biphasic.
  • the high affinity, low capacity site was calculated to have a binding constant of 3 nM and 18,000 sites/cell, whereas the low affinity, high capacity site was 200 nM and 60,000 sites/cell ( Figure 6).
  • siRNAs chosen had a G/C content of about 50%> and consisted of 21-nt sense and antisense strands that paired to produce a 19-nt duplex region with 2-nt, dTdT, overhangs at each 3' terminus.
  • BLAST searches NCBI database
  • annealed siRNA duplexes were purchased from Dharmacon Research, Inc. (Lafayette, CO).
  • the siRNAs used in this study are given in the table below. Specificity was controlled by inco ⁇ orating four mis-matches in the Nl duplex to create aNRP-lal control RNAi.
  • the luc control duplex targeted to the luciferase gene was used to show that an unrelated dsRNAi also has no effect on NRP-1 expression.
  • Samples were then combined with 0.5 ml of MCDB-131 complete media and transferred to a 60 mm dish containing complete media. Dishes were placed in a CO 2 incubator at 37°C overnight. On average there was approximately 50-70% cell survival the following morning.
  • T2-T ⁇ RS binding sites/cell decreased to 40,000 and further decreased to 20,000 binding sites/cell when treated with 40 ⁇ g dsRNA/al, compaired to cells untreated with dsRNA.
  • the number of T2-T ⁇ RS binding to cells remained unchanged in the negative control, the dsRNA/al control and dsRNA Luciferase (Fig. 8).
  • Fxa Factor Xa
  • the test conditions were set up where 10 nM of Fxa (Haematologic Tech, Essez Junction, VT) was incubated with either 20 mM of phospholipids (4 parts phosphatidylcholine and 1 part phosphatidylserine (Sigma, St. Louis, MO) or 20 ⁇ g of HUVEC membrane fragments (described in Example 3) in an assay buffer containing 0.05 M Tris-HCl, pH 7.3, 0.2% BSA and 25 mM CaCl 2 .
  • 10 mM of Fxa was incubated with assay buffer alone.
  • T2-T ⁇ RS was added into these test conditions at various concentrations and incubated at 37°C for 5 minutes.
  • 1 mM of the chromogenic substrate S-2765 (DiaPharma, West Chester, OH) was added into each sample. After 3 minutes, the reactions were stopped with 20%> of acetic acid and Fxa activities were measured by reading the A o 5 .
  • S2765 was demonstrated to be specific to Fxa and not reactive to membrane fragments nor T2-T ⁇ RS, even though this chromogenic substrate has been demonstrated to be reactive to other serine proteases.
  • T2-T ⁇ RS induced a 75% enhancement of Fxa activity when compared to Fxa in combination with buffer alone (Fig. 9B).
  • Fxa is vital in regulation of blood coagulation as well as an active chemoattractant for smooth muscle cells (Gasic et al, PNAS USA 89: 2317-2320 (1992)) and endothelial cells (Mackie, The Oxford Text Book of Medicine 3 rd Ed., 3613-3627 (1996)) migration
  • T2-T ⁇ RS to modulate Fxa activites may influence these process and contribute to the overall effectiveness of the molecule as an anti-angiogenesis agent.
  • T2-T ⁇ RS is a Potent Inhibitor of VEGF Induced Angiogenesis in vivo
  • mice Female nude (nu/nu) mice were purchased from Taconic (Model # NCRNU-M, Taconic, Germantown, NY) and grouped into 10 per group. The mice were kept for one week in an animal quarantine room before being released to holding rooms.
  • Matrigel Assays were performed. Various concentrations of the recombinant T2-T ⁇ RS protein were mixed with 0.45 ml matrigel (Fisher Scientific, Neward, DE) in the presence of VEGF (3mg/ml) (R&D, Minneapolis, Mn) and 20 U/ml heparin (Sigma, St. Louis, MO).
  • matrigel was mixed with test substances and preloaded into a 1.0 ml tuberculin syringes with G27 needles. Animals were anaesthetised and 0.45 ml of the mixed Matrigel were injected subcutaneously into the dorsal caudal portion of the midline (1 injection/animal) using a G27 needle. The Matrigel rapidly formed a solid gel that persisted for over 7 days. On Day 7, all animals were euthanized by CO 2 and matrigel plugs were harvested for haemoglobin measurement.
  • Haemoglobin measurement of the matrigel plug was a means to evaluate the angiogenic index.
  • T2-T ⁇ RS is a potent suppressor of VEGF-induced angiogenesis.
  • 10 nM of T2-T ⁇ RS brought about the onset of anti-angiogenesis activity and at 1000 nM, the matrigel plug was clear of hemoglobin.
  • T2-T ⁇ RS demonstrated that it is a potent anti-angiogenic molecule. Indeed, the concentration in which T2-T ⁇ RS induced 50%) of anti-angiogenic effect (IC 50 ) was estimated to be ⁇ 100nM (Fig. 10).
  • the 1.2 kb Nliel fragment, containing cDNA encoding human T2-T ⁇ RS was cloned into the adeno-shuttle plasmid pAvCsxlx digested with Spel to generate the adenoviral shuttle plasmid pAvT2T ⁇ RS ( Figure 12).
  • the recombinant adenovirus encoding human T2-T ⁇ RS was constructed by a rapid vector generation protocol using Cre recombinase-mediated recombination of two lox-site containing plasmids, pNDSQ3.1 (containing a lox site 5' to the right hand portion of the adenoviral vector genome; Figure 13), and the adenoviral shuttle plasmid pAvT2T ⁇ RS (containing a loxt site 3' to the left end of the viral genome and the T2T ⁇ RS expression cassette).
  • pNDSQ3.1 digested with CM
  • pAvT2T ⁇ RS digested with vwii
  • Cre-encoding plasmid pC-Cre3.1
  • the transfected cells were treated with dexamethasone (0.33 ⁇ M) and the plasmids were joined by Cre-mediated recombination, generating the adenovirus encoding T2-T ⁇ RS (Av3T2T ⁇ RS).
  • the Av3CsNull vector was generated in a similar manner, but lacks a transgene.
  • the cells were harvested a week after transfection and passaged until a cytopathic effect (CPE) was observed.
  • CPE cytopathic effect
  • cells were freeze (dry ice)/thawed (37°C) 5 times to obtain a CVL (crude viral lysate), which was centrifuged to remove the cell debris and then used to infect fresh S8 cells in the presence of dexamethasone (0.33 ⁇ M).
  • CPE cytopathic effect
  • the supernatant was loaded on a discontinuous Cesium Chloride gradient (1.25 g/ml CsCl and 1.4 g/ml CsCl) and centrifuged for 1 hr at 28,000 ⁇ m (in a SW28 swing bucket rotor).
  • the bottom viral band was pulled from the gradient and centrifuged on a CsCl continuous gradient (1.33 g/ml CsCl) overnight at 60,000 ⁇ m (in an NNT-65 rotor).
  • the purified viral band was pulled from the gradient, glycerol was added to a final concentration of 10% and the mixture was dialyzed in 200 mM Tris pH 8.0, 50 mM Hepes, 10% glycerol for >16 hr at 4°C.
  • concentration of vector was determined by spectrophotometric analysis (Mittereder et al, J. Virol. 70: 7498-7509 (1996)) and the vector was then aliquoted and stored at - 70°C.
  • the genomic organization and purity of Av3T2T ⁇ RS was verified by D ⁇ A analysis of isolated viral D ⁇ A by restriction endonuclease digestion.
  • D ⁇ A was isolated from Av3T2T ⁇ RS by first digesting the virus with Proteinase-K followed by Phenol/Chloroform extraction and ethanol precipitation of the viral D ⁇ A. The viral D ⁇ A was then digested with BamHI (Gibco BRL, Rockville, MD), EcoRN, Notl and Pvu ⁇ (NEB, Beverly, MA) and applied to a 0.4% agarose/TA ⁇ gel. The D ⁇ A restriction analysis revealed the expected sizes.
  • RNAzol B (Cat# CS-104, Tel-Test Inc., Friendswood, TX) according to the manufacturer provided protocol.
  • Ten micro grams of total RNA resuspended in DEPC dH 2 O (Cat# 9920, Arnbion, Austin, TX) was mixed with 12.5 volumes of RNA Sample Loading Buffer with Ethidium Bromide (Cat# 2-631636, 5Prime ⁇ 3Prime, Boulder, CO) and heated at 56°C for 15 minutes and quickly cooled on ice.
  • RNA was transferred to a Nytran membrane (0.45 ⁇ m) (Cat# 77412, Schleicher & Schuell, Keene, NH) according to the protocol in Molecular Cloning (Maniatis, pp 7.46-7.48, 1989) via capillary transfer overnight at room temperature.
  • the membrane was UV crosslinked and blocked in prehybridization solution (8 ml of 1M NaPO 4 , 32 ⁇ l 0.5M EDTA, 0.5% BSA, 11.2 ml 10% SDS) at 65°C for 2 hours.
  • prehybridization solution 8 ml of 1M NaPO 4 , 32 ⁇ l 0.5M EDTA, 0.5% BSA, 11.2 ml 10% SDS
  • the membrane was washed with 2X SSC (Cat# 15557-044 Gibco BRL, Rockville, MD) + 0.1% SDS (Cat# 15553-035 Gibco BRL, Rockville, MD) one time at room temperature for 30 minutes. After the initial wash, the membrane was washed two additional times with 0.2X SSC + 0.1% SDS for 30 minutes at 64°C with shaking. Following the washes, the membrane was exposed to Kodak X-OMAT AR film (Cat# 1651579 Kodak, Rochester, NY) at -80°C for 24 hours to detect the signal.
  • 2X SSC Cat# 15557-044 Gibco BRL, Rockville, MD
  • SDS Cat# 15553-035 Gibco BRL, Rockville, MD
  • S8 cells were infected with AV3T2T ⁇ RS at 500 ppc.
  • Conditioned media 25 ml
  • cells were harvested 72 hours post-infection.
  • the cell pellet was lysed using 300 ⁇ l lysis buffer containing protease inhibitor cocktail.
  • the cell lysate (1.35 ⁇ l) and the conditioned media (16.3 ⁇ l) were mixed with the corresponding amounts of NuPAGE LDS Sample Buffer 4X (6.3 ⁇ l) (Cat# NP0007 Invitrogen, Carlsbad, CA) and NuPAGE Sample Reducing Agent 10X (2.5 ⁇ l) (Cat# NP0004 Invitrogen, Carlsbad, CA) and heated at 70°C for 10 minutes.
  • the membrane was incubated with His-probe (H- 15) rabbit polyclonal IgG (200 ⁇ g/ml) diluted 1:200 (Cat# sc-803 Santa Cruz Biotechnology, Santa Cruz, CA) for one hour at room temperature with shaking.
  • His-probe H- 15
  • rabbit polyclonal IgG 200 ⁇ g/ml
  • 1:200 Cat# sc-803 Santa Cruz Biotechnology, Santa Cruz, CA
  • the membrane was incubated with a 1:5000 dilution of anti-rabbit IgG HRP-linked F(Ab') 2 secondary antibody (Cat# NA9340, Amersham Pharmacia, Piscataway, NJ) for 1 hour, followed by four additional washes and a five minute incubation in chemiluminescence detection reagent mix (Cat# 34075 Supersignal West Dura Extended Duration Substrate, Pierce, Rockford, IL). The blot was exposed to a Kodax Biomax MR X-ray film (Cat# 876-1520, Kodak, Rochester, NY).
  • the results of the Western analysis revealed the presence of T2T ⁇ RS protein in the conditioned media and the cell lysate of the cell transduced with the Av3T2T ⁇ RS vector.
  • No T2T ⁇ RS protein was detected in either conditioned media or cell lysate from the Av3CsNull vector transduced cells ( Figure 1 IB).
  • the T2T ⁇ RS protein was purified from the conditioned media of the cells transduced with Av3T2T ⁇ RS vector using Ni-NTA agarose beads. This semi-purified protein was found biologically active in a variety of assays including receptor tyrosine kinase (RTK) assay using HUVEC membrane and apoptosis assay explained in previous examples.
  • RTK receptor tyrosine kinase
  • EXAMPLE 10 Generation of Bovine Immunodeficiency Virus (BIN) Based Lentiviral vectors Encoding Human T2T ⁇ RS
  • the shuttle plasmid was generated by linearizing the parent shuttle plasmid, pBIVUniversalCMV, with Nhel and ligating it with the 1.2 Kb T2T ⁇ RS-N/zeI fragment.
  • the lentiviral supernatant was obtained by co-transfecting 293T cells with pBIVUniversalCMVT2T ⁇ RS, packaging construct and env construct by CaPO 4 transient transfection method per manufacturer protocol.
  • 293T cells were seeded in 10cm tissue culture plates in appropriate medium (DMEM+10% Heat Inactivated FBS+l%Penn/Strep). The following day, the cells were serum stimulated by changing the medium. 15 ⁇ g each of the packaging plasmid and the shuttle plasmid and 4.5 ⁇ g of the envelope plasmid were diluted in distilled water and mixed with calcium phosphate. D ⁇ A solution was added drop-wise to the 2X HBSS solution while vortexing. D ⁇ A precipitate was then added to the cells and the cells were incubated at 37°C, 5% CO 2 , overnight. Next day, the medium was aspirated and fresh medium was added to the plates.
  • appropriate medium DMEM+10% Heat Inactivated FBS+l%Penn/Strep

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Immunology (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Dermatology (AREA)
  • Emergency Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Le récepteur du facteur de croissance des cellules endothéliales, la neuropiline, est un partenaire de liaison sélectif pour la molécule anti-angiogénique T2-TrpRS. Cette liaison inhibe l'activité induite par VEGF en bloquant efficacement la liaison de VEGF à la neuropiline. L'hypothèse est que T2- TrpRS agit comme un antagoniste concurrentiel de l'interaction VEGF-neuropiline avec perturbation consécutive de l'angiogenèse. Cette découverte est exploitée aux fins de proposer de nouvelles méthodes pour le traitement de maladies ou pathologies induites par T2-TrpRS ou des dérivées de T2-TrpRS par leur interaction avec des neuropilines.
PCT/US2002/023868 2001-07-26 2002-07-25 Methodes de traitement de maladies induites par neuropiline WO2003009813A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002332430A AU2002332430A1 (en) 2001-07-26 2002-07-25 Methods of treating neuropilin-mediated diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US30787201P 2001-07-26 2001-07-26
US60/307,872 2001-07-26
US35555102P 2002-02-07 2002-02-07
US60/355,551 2002-02-07

Publications (2)

Publication Number Publication Date
WO2003009813A2 true WO2003009813A2 (fr) 2003-02-06
WO2003009813A3 WO2003009813A3 (fr) 2003-08-14

Family

ID=26975973

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/023868 WO2003009813A2 (fr) 2001-07-26 2002-07-25 Methodes de traitement de maladies induites par neuropiline

Country Status (2)

Country Link
AU (1) AU2002332430A1 (fr)
WO (1) WO2003009813A2 (fr)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006016217A1 (fr) * 2004-08-02 2006-02-16 Angiosyn, Inc. FRAGMENTS D'ARNt SYNTHÉTASE
WO2005117954A3 (fr) * 2004-06-04 2006-10-12 Scripps Research Inst Compositions et méthodes pour le traitement de maladies néovasculaires
EP1853113A2 (fr) * 2005-02-24 2007-11-14 The Scripps Research Institute Revascularisation de tissu retinien ischemique et procede de criblage associe
US8282921B2 (en) 2004-08-02 2012-10-09 Paul Glidden tRNA synthetase fragments
US8404471B2 (en) 2008-06-26 2013-03-26 Atyr Pharma, Inc. Compositions and methods comprising glycyl-tRNA synthetases having non-canonical biological activities
US8404242B2 (en) 2009-03-16 2013-03-26 Atyr Pharma, Inc. Compositions and methods comprising histidyl-tRNA synthetase splice variants having non-canonical biological activities
US8828395B2 (en) 2009-12-11 2014-09-09 Atyr Pharma, Inc. Antibodies that bind tyrosyl-tRNA synthetases
US9029506B2 (en) 2010-08-25 2015-05-12 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of tyrosyl-tRNA synthetases
US9062302B2 (en) 2010-05-04 2015-06-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex
US9062301B2 (en) 2010-05-04 2015-06-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutamyl-prolyl-tRNA synthetases
US9068177B2 (en) 2010-04-29 2015-06-30 Atyr Pharma, Inc Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
US9127268B2 (en) 2009-12-11 2015-09-08 Atyr Pharma, Inc. Aminoacyl tRNA synthetases for modulating inflammation
US9499810B2 (en) 2008-06-11 2016-11-22 Atyr Pharma, Inc. Thrombopoietic activity of tyrosyl-tRNA synthetase polypeptides
US9623093B2 (en) 2010-04-29 2017-04-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl tRNA synthetases
WO2017066482A1 (fr) * 2015-10-15 2017-04-20 The Scripps Research Institute Activité de liaison de l'aminoacyl-arnt synthétase dans la neuropathie de charcot marie tooth (cmt) et les maladies neurologiques afférentes à la maladie de cmt
US9637730B2 (en) 2010-07-12 2017-05-02 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of histidyl-tRNA synthetases
US9687533B2 (en) 2010-05-14 2017-06-27 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-tRNA synthetases
US9688978B2 (en) 2011-12-29 2017-06-27 Atyr Pharma, Inc. Aspartyl-tRNA synthetase-Fc conjugates
US9714419B2 (en) 2011-08-09 2017-07-25 Atyr Pharma, Inc. PEGylated tyrosyl-tRNA synthetase polypeptides
US9790482B2 (en) 2010-05-17 2017-10-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
US9796972B2 (en) 2010-07-12 2017-10-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
US9816084B2 (en) 2011-12-06 2017-11-14 Atyr Pharma, Inc. Aspartyl-tRNA synthetases
US9822353B2 (en) 2011-12-06 2017-11-21 Atyr Pharma, Inc. PEGylated aspartyl-tRNA synthetase polypeptides
US9896515B2 (en) 2010-04-27 2018-02-20 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases
US9896680B2 (en) 2009-03-31 2018-02-20 Atyr Pharma, Inc. Compositions and methods comprising aspartyl-tRNA synthetases having non-canonical biological activities
US10030077B2 (en) 2010-04-26 2018-07-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-tRNA synthetase
US10093915B2 (en) 2013-03-15 2018-10-09 Atyr Pharma Inc. Histidyl-tRNA synthetase-Fc conjugates
US10150958B2 (en) 2010-04-27 2018-12-11 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl-tRNA synthetases
US10179906B2 (en) 2010-05-03 2019-01-15 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-tRNA synthetases
US10189911B2 (en) 2010-04-29 2019-01-29 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Valyl-tRNA synthetases
US10563191B2 (en) 2010-10-06 2020-02-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related protein fragments of tryptophanyl tRNA synthetases
US11767520B2 (en) 2017-04-20 2023-09-26 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2403864B1 (fr) 2009-02-27 2015-08-12 Atyr Pharma, Inc. Motifs structuraux de polypeptides associés à une activité de signalisation cellulaire
EP2563911B1 (fr) 2010-04-28 2021-07-21 aTyr Pharma, Inc. Compositions thérapeutiques, diagnostiques et d'anticorps à base de fragments de protéines d'aminoacyl-arnt synthétases
EP2566496B1 (fr) 2010-05-03 2018-02-28 aTyr Pharma, Inc. Découverte innovante de compositions thérapeutiques, diagnostiques et à base d'anticorps liées des fragments protéiques de méthionyl-arnt-synthétases
CA2797277C (fr) 2010-05-03 2021-02-23 Atyr Pharma, Inc. Decouverte innovante de compositions therapeutiques, de diagnostic et d'anticorps liees a des fragments proteiques d'arginyle-arnt synthetases
US8946157B2 (en) 2010-05-03 2015-02-03 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of seryl-tRNA synthetases
CA2800281C (fr) 2010-06-01 2021-01-12 Atyr Pharma, Inc. Decouverte innovante de compositions therapeutiques, diagnostiques, et d'anticorps associes a des fragments de proteine de lysyl-tarn synthetases
US8999321B2 (en) 2010-07-12 2015-04-07 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
US8969301B2 (en) 2010-07-12 2015-03-03 Atyr Pharma Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of aspartyl-tRNA synthetases
CN104334196B (zh) 2012-02-16 2018-04-10 Atyr 医药公司 用于治疗自身免疫疾病和炎性疾病的组氨酰‑tRNA合成酶

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020044941A1 (en) * 1999-03-12 2002-04-18 Rosen Craig A. Nucleic acids, proteins and antibodies

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU3395900A (en) * 1999-03-12 2000-10-04 Human Genome Sciences, Inc. Human lung cancer associated gene sequences and polypeptides

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020044941A1 (en) * 1999-03-12 2002-04-18 Rosen Craig A. Nucleic acids, proteins and antibodies

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE GENBANK [Online] RUBEN: 'Lung cancer associated polypeptide', XP002961696 Database accession no. (AAB58220) & WO 00 55180 A2 *

Cited By (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2005249586B2 (en) * 2004-06-04 2010-12-23 The Scripps Research Institute Compositions and methods for treatment of neovascular diseases
WO2005117954A3 (fr) * 2004-06-04 2006-10-12 Scripps Research Inst Compositions et méthodes pour le traitement de maladies néovasculaires
JP4943324B2 (ja) * 2004-06-04 2012-05-30 ザ スクリプス リサーチ インスティチュート 新生血管病の治療のための組成物および方法
JP2008501718A (ja) * 2004-06-04 2008-01-24 ザ・スクリプス・リサーチ・インステイチユート 新生血管病の治療のための組成物および方法
JP2008508349A (ja) * 2004-08-02 2008-03-21 アンジオシン,インコーポレイティド tRNAシンセターゼフラグメント
US8282921B2 (en) 2004-08-02 2012-10-09 Paul Glidden tRNA synthetase fragments
WO2006016217A1 (fr) * 2004-08-02 2006-02-16 Angiosyn, Inc. FRAGMENTS D'ARNt SYNTHÉTASE
EP1853113A2 (fr) * 2005-02-24 2007-11-14 The Scripps Research Institute Revascularisation de tissu retinien ischemique et procede de criblage associe
EP1853113A4 (fr) * 2005-02-24 2011-03-30 Scripps Research Inst Revascularisation de tissu retinien ischemique et procede de criblage associe
AU2006216678B2 (en) * 2005-02-24 2011-11-17 The Scripps Research Institute Revascularization of ischemic retinal tissue and screening method therefor
EP1902725A2 (fr) * 2005-02-24 2008-03-26 The Scripps Research Institute Revascularisation de tissu rétinal ischémique
JP2008532941A (ja) * 2005-02-24 2008-08-21 ザ・スクリプス・リサーチ・インステイチユート 虚血性網膜組織の血行再建及びそのためのスクリーニング方法
EP1902725A3 (fr) * 2005-02-24 2008-04-02 The Scripps Research Institute Revascularisation de tissu rétinal ischémique
US9499810B2 (en) 2008-06-11 2016-11-22 Atyr Pharma, Inc. Thrombopoietic activity of tyrosyl-tRNA synthetase polypeptides
US9157076B2 (en) 2008-06-26 2015-10-13 Atyr Pharma, Inc. Compositions and methods comprising glycyl-tRNA synthetases having non-canonical biological activities
US8404471B2 (en) 2008-06-26 2013-03-26 Atyr Pharma, Inc. Compositions and methods comprising glycyl-tRNA synthetases having non-canonical biological activities
US8747840B2 (en) 2008-06-26 2014-06-10 Atyr Pharma, Inc. Compositions and methods comprising glycyl-tRNA synthetases having non-canonical biological activities
US9585946B2 (en) 2008-06-26 2017-03-07 Atyr Pharma, Inc. Compositions and methods comprising glycyl-tRNA synthetases having non-canonical biological activities
US11078299B2 (en) 2009-03-16 2021-08-03 Atyr Pharma, Inc. Compositions and methods comprising histidyl-tRNA synthetase splice variants having non-canonical biological activities
US8753638B2 (en) 2009-03-16 2014-06-17 Atyr Pharma, Inc. Compositions and methods comprising histidyl-TRNA synthetase splice variants having non-canonical biological activities
US10526419B2 (en) 2009-03-16 2020-01-07 Atyr Pharma, Inc. Compositions and methods comprising histidyl-tRNA synthetase splice variants having non-canonical biological activities
US10941214B2 (en) 2009-03-16 2021-03-09 Atyr Pharma, Inc. Compositions and methods comprising histidyl-tRNA synthetase splice variants having non-canonical biological activities
US8404242B2 (en) 2009-03-16 2013-03-26 Atyr Pharma, Inc. Compositions and methods comprising histidyl-tRNA synthetase splice variants having non-canonical biological activities
US10017582B2 (en) 2009-03-16 2018-07-10 Atyr Pharma, Inc. Compositions and methods comprising histidyl-trna synthetase splice variants having non-canonical biological activities
US9605265B2 (en) 2009-03-16 2017-03-28 Atyr Pharma, Inc. Compositions and methods comprising histidyl-tRNA synthetase splice variants having non-canonical biological activities
US9896680B2 (en) 2009-03-31 2018-02-20 Atyr Pharma, Inc. Compositions and methods comprising aspartyl-tRNA synthetases having non-canonical biological activities
US9127268B2 (en) 2009-12-11 2015-09-08 Atyr Pharma, Inc. Aminoacyl tRNA synthetases for modulating inflammation
US8828395B2 (en) 2009-12-11 2014-09-09 Atyr Pharma, Inc. Antibodies that bind tyrosyl-tRNA synthetases
US9328340B2 (en) 2009-12-11 2016-05-03 Atyr Pharma, Inc. Amino acyl tRNA synthetases for modulating inflammation
US9943577B2 (en) 2009-12-11 2018-04-17 Atyr Pharma, Inc. Aminoacyl tRNA synthetases for modulating inflammation
US10717786B2 (en) 2010-04-26 2020-07-21 aTye Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Cysteinyl-tRNA synthetase
US10030077B2 (en) 2010-04-26 2018-07-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of cysteinyl-tRNA synthetase
US10563192B2 (en) 2010-04-27 2020-02-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl-tRNA synthetases
US10150958B2 (en) 2010-04-27 2018-12-11 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of threonyl-tRNA synthetases
US9896515B2 (en) 2010-04-27 2018-02-20 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of isoleucyl tRNA synthetases
US10189911B2 (en) 2010-04-29 2019-01-29 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Valyl-tRNA synthetases
US9623093B2 (en) 2010-04-29 2017-04-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of asparaginyl tRNA synthetases
US9068177B2 (en) 2010-04-29 2015-06-30 Atyr Pharma, Inc Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutaminyl-tRNA synthetases
US10179906B2 (en) 2010-05-03 2019-01-15 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-alpha-tRNA synthetases
US10160814B2 (en) 2010-05-04 2018-12-25 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutamyl-prolyl-tRNA synthetases
US9062301B2 (en) 2010-05-04 2015-06-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glutamyl-prolyl-tRNA synthetases
US9062302B2 (en) 2010-05-04 2015-06-23 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of p38 multi-tRNA synthetase complex
US10220080B2 (en) 2010-05-14 2019-03-05 aTyr Pharam, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-tRNA synthetases
US9687533B2 (en) 2010-05-14 2017-06-27 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of phenylalanyl-beta-tRNA synthetases
US10179908B2 (en) 2010-05-17 2019-01-15 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
US9790482B2 (en) 2010-05-17 2017-10-17 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of leucyl-tRNA synthetases
US9637730B2 (en) 2010-07-12 2017-05-02 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of histidyl-tRNA synthetases
US10669533B2 (en) 2010-07-12 2020-06-02 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of Histidyl-tRNA synthetases
US9796972B2 (en) 2010-07-12 2017-10-24 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
US10196628B2 (en) 2010-07-12 2019-02-05 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of histidyl-tRNA synthetases
US10196629B2 (en) 2010-07-12 2019-02-05 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of glycyl-tRNA synthetases
US9029506B2 (en) 2010-08-25 2015-05-12 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related to protein fragments of tyrosyl-tRNA synthetases
US10563191B2 (en) 2010-10-06 2020-02-18 Atyr Pharma, Inc. Innovative discovery of therapeutic, diagnostic, and antibody compositions related protein fragments of tryptophanyl tRNA synthetases
US9714419B2 (en) 2011-08-09 2017-07-25 Atyr Pharma, Inc. PEGylated tyrosyl-tRNA synthetase polypeptides
US9822353B2 (en) 2011-12-06 2017-11-21 Atyr Pharma, Inc. PEGylated aspartyl-tRNA synthetase polypeptides
US9816084B2 (en) 2011-12-06 2017-11-14 Atyr Pharma, Inc. Aspartyl-tRNA synthetases
US9688978B2 (en) 2011-12-29 2017-06-27 Atyr Pharma, Inc. Aspartyl-tRNA synthetase-Fc conjugates
US10472618B2 (en) 2013-03-15 2019-11-12 Atyr Pharma, Inc. Histidyl-tRNA synthetase-Fc conjugates
US10711260B2 (en) 2013-03-15 2020-07-14 Atyr Pharma, Inc. Histidyl-tRNA synthetase-Fc conjugates
US10093915B2 (en) 2013-03-15 2018-10-09 Atyr Pharma Inc. Histidyl-tRNA synthetase-Fc conjugates
US11072787B2 (en) 2013-03-15 2021-07-27 Atyr Pharma Inc. Histidyl-tRNA synthetase-Fc conjugates
WO2017066482A1 (fr) * 2015-10-15 2017-04-20 The Scripps Research Institute Activité de liaison de l'aminoacyl-arnt synthétase dans la neuropathie de charcot marie tooth (cmt) et les maladies neurologiques afférentes à la maladie de cmt
US11767520B2 (en) 2017-04-20 2023-09-26 Atyr Pharma, Inc. Compositions and methods for treating lung inflammation

Also Published As

Publication number Publication date
AU2002332430A1 (en) 2003-02-17
WO2003009813A3 (fr) 2003-08-14

Similar Documents

Publication Publication Date Title
WO2003009813A2 (fr) Methodes de traitement de maladies induites par neuropiline
Blumer et al. Expression analysis and subcellular distribution of the two G-protein regulators AGS3 and LGN indicate distinct functionality: localization of LGN to the midbody during cytokinesis
Rettig et al. Isoform-specific interaction of the alpha1A subunits of brain Ca2+ channels with the presynaptic proteins syntaxin and SNAP-25.
JP7386210B2 (ja) Nmeバリアント種の発現および抑制
JPH10510718A (ja) 血管内皮増殖因子−b
US20150307589A1 (en) Peptidic antagonists of class iii semaphorins/neuropilins complexes
Mason et al. Cell adhesion molecules and adhesion abnormalities in prostate cancer
Duval et al. Molecular architecture of acetylcholinesterase collagen‐tailed forms; construction of a glycolipid‐tailed tetramer.
ES2331441T3 (es) Dna que codifica dp-75 y un proceso para su uso.
CN101812127A (zh) 微管结合蛋白及其编码基因与应用
CN107827970B (zh) 一种抑制foxm1的抗肿瘤蛋白肽
Gringel et al. Nuclear fibroblast growth factor-2 interacts specifically with splicing factor SF3a66
TW201200151A (en) Methods and compositions related to reduced MET phosphorylation by leukocyte cell-derived chemotaxin 2 in tumor cells
KR101123130B1 (ko) Ptk7 단백질의 기능 저해를 통한 세포의 이동, 침윤 또는 혈관신생 억제제
KR102236843B1 (ko) 새로운 재생 치료제로서의 camkk1
JP2018021026A (ja) 線維化抑制剤
CN102958532A (zh) 使用结缔组织生长因子的药物组合物
WO2000045771A2 (fr) Suppression de la transformation de cellules par le facteur de transcription egr
JPH11506325A (ja) Δp62、その変異体、核酸配列及びこれらの使用
BG63548B1 (bg) Полипептиди, съдържащи протеинови домени на gax, включени в транскрипцията и/или взаимнодействащи сдруги протеини, съответни нуклеинови киселини и тяхното използване
US20100111913A1 (en) Method of enhancing migration of neural precursor cells
CN107812192B (zh) 抑制znf498蛋白表达量的物质在制备预防和治疗癌症的产品中的应用
WO2007088882A1 (fr) Polypeptide ayant une affinité pour un constituant de virus à enveloppe et son utilisation dans le transfert d'une substance dans une cellule
CN110054678B (zh) 一种膜结合型mFLT3LG蛋白及其应用
WO2021020505A1 (fr) Composition pharmaceutique pour le traitement du cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC 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 MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ UG ZM ZW AM AZ BY KG KZ RU TJ TM AT BE BG CH CY CZ DK EE ES FI FR GB GR IE IT LU MC PT SE SK TR BF BJ CF CG CI GA GN GQ 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)
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 in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP