WO1997033899A1 - Apoptosis inducing molecule i - Google Patents

Apoptosis inducing molecule i Download PDF

Info

Publication number
WO1997033899A1
WO1997033899A1 PCT/US1996/003773 US9603773W WO9733899A1 WO 1997033899 A1 WO1997033899 A1 WO 1997033899A1 US 9603773 W US9603773 W US 9603773W WO 9733899 A1 WO9733899 A1 WO 9733899A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
aim
polynucleotide
leu
ser
Prior art date
Application number
PCT/US1996/003773
Other languages
French (fr)
Inventor
Steven M. Ruben
Original Assignee
Human Genome Sciences, Inc.
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 Human Genome Sciences, Inc. filed Critical Human Genome Sciences, Inc.
Priority to PCT/US1996/003773 priority Critical patent/WO1997033899A1/en
Priority to AU57111/96A priority patent/AU5711196A/en
Publication of WO1997033899A1 publication Critical patent/WO1997033899A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates, in part, to newly identified polynucleotides and polypeptides; variants and derivatives of the polynucleotides and polypeptides; processes for making the polynucleotides and the polypeptides, and their variants and derivatives; agonists and antagonists of the polypeptides; and uses of the polynucleotides, polypeptides, variants, derivatives, agonists and antagonists.
  • the invention relates to polynucleotides and polypeptides of human Apoptosis Inducing Molecule I (AIM-I).
  • TNF- ⁇ and ⁇ are related members of a broad class of polypeptide mediators, which includes the interferons. interieukins and growth factors, collectively called cytokines (Beutler, B. and Cerami, A.. Annu. Rev. Immunol. 7:625-655, 1989).
  • Tumor necrosis factor (TNF- ⁇ and TNF-3) was originally discovered as a result of its anti-tumor activity, however, now it is recognized as a pleiotropic cytokine capable of numerous biological activities including apoptosis of some transformed cell lines, mediation of cell activation and proliferation and also as playing important roles in immune retaliation and inflammation.
  • TNF- ⁇ tumor necrosis factor
  • TNF-/3 lymphotoxin-c.
  • LT- 3. OX40L
  • Fas ligand CD30L
  • CD27L CD40L
  • 4- 1BBL 4- 1BBL.
  • the ligands of the TNF ligand superfamily are acidic, TNF-like molecules with approximately 20% sequence homology in the extracellular domains (range, 12%-36%) and exist mainly as membrane-bound forms with the biologically active form being a trimeric/multimeric complex. Soluble forms of the TNF ligand superfamily have only been identified so far for TNF. LT ⁇ , and Fas ligand (for a general review, see Gruss, H. and Dower, S.K. , Blood, 85 (12): 3378-3404, 1995), which is hereby incorporated by reference in its entirety.
  • Apoptosis plays a critical role in the destruction of immune thymocytes that recognize self antigens. Failure of this normal elimination process may play a role in autoimmune diseases (Gammon et al. Immunology Today. 12: 192. 1991 )
  • Fas antigen is a cell surface protein of relative MW of 45 Kd.
  • Both human and murine genes for Fas have been cloned by Watanabe-Fukunaga et al , (J. Immunolo. 148: 1274, 1992) and Itoh et al (Cell, 66:233, 1991).
  • the proteins encoded by these genes are both transmembrane proteins with structural homology to the nerve growth factor/tumor necrosis factor receptor superfamily, which includes two TNF receptors, the low affinity nerve growth factor receptor and CD40, CD27, CD30, and OX40.
  • Fas ligand has been described (Suda et al, Cell, 75: 1169, 1993). The amino acid sequence indicates that Fas ligand is a type 11 transmembrane protein belonging to the TNF family. Fas ligand is expressed in splenocytes and thymocytes, consistent with T-cell mediated cytotoxicity. The purified Fas ligand has a MW of 40 Kd.
  • Fas/Fas ligand interactions are required for apoptosis following the activation of T-cells (Ju et al Nature, 373:444, 1995; Brunner et al . Nature, 57J.441 , 1995).
  • Activation of T-cells induces expression of both proteins on the cell surface.
  • Subsequent interaction between the ligand and receptor results in apoptosis of the cells. This supports the possible regulatory role for apoptosis induced by Fas/ Fas ligand interaciion during normal immune responses.
  • polypeptide of the present invention has been identified as a novel member of the TNF ligand super- family based on structural and biological similarities.
  • AIM-I novel Apoptosis Inducing Molecule I
  • polynucleotides that encode AIM-I pa ⁇ icularly polynucleotides that encode the polypeptide herein designated AIM-I.
  • the polynucleotide comprises the region encoding human AIM-I in the sequence set out in Figure 1.
  • an isolated nucleic acid molecule encoding a mature polypeptide expressed by the human cDNA contained in ATCC Deposit No. 97448.
  • this aspect of the invention there are provided isolated nucleic acid molecules encoding human AIM-I. including mRNAs, cDNAs, genomic DNAs and, in further embodiments of this aspect of the invention, biologically, diagnostically, clinically or therapeuticallv useful variants, analogs, derivatives and/or fragments thereof, including fragments ot the ⁇ ar ⁇ ants. analogs and derivatives.
  • isolated nucleic acid molecules encoding human AIM-I. including mRNAs, cDNAs, genomic DNAs and, in further embodiments of this aspect of the invention, biologically, diagnostically, clinically or therapeuticallv useful variants, analogs, derivatives and/or fragments thereof, including fragments ot the ⁇ ar ⁇ ants. analogs and derivatives.
  • particularly preferred embodiments of this aspect of the invention are naturally occurring allelic variants of human AIM-I r
  • AIM-I polypeptides particularly human AIM-I polypeptides.
  • w hich may be employed to treat lymphadenopathy, autoimmune disease, graft versus host disease; which may be used to stimulate peripheral tolerance, destroy pathologic transformed cell lines, mediate cell activation and proliferation; and are functionally linked as primary mediators of immune regulation and inflammatory response.
  • This aspect of the invention provides novel polypeptides of human origin referred to herein as AIM-I as well as biologically, diagnostically or therapeutically useful fragments, variants and derivatives thereof, variants and derivatives of the fragments, and analogs of all of the foregoing.
  • AIM-I novel polypeptides of human origin referred to herein as AIM-I as well as biologically, diagnostically or therapeutically useful fragments, variants and derivatives thereof, variants and derivatives of the fragments, and analogs of all of the foregoing.
  • variants of human AIM-I encoded by naturally occurring alleles of the human AIM-I gene are particularly preferred embodiments of human AIM-I encoded by naturally occurring alleles of the human AIM-I gene.
  • methods for producing the aforementioned AIM-I polypeptides comprising culturing host cells having expressibly incorporated therein an exogenously-derived human AIM-I-encoding polynucleotide under conditions for expression of human AIM-I in the host and then recovering the expressed polypeptide.
  • products, compositions and methods for, among other things: assessing AIM-I expression in cells by determining AIM-I polypeptides or AIM-I-encoding mRN A; treaiinc disease or disorder caused by under-expression of the AIM-I in vitro, ex vivo or in vivo by exposing cells to AIM-I polypeptides or polynucleotides as disclosed herein; assaying genetic variation and aberrations, such as defects, in AIM-I genes; and administering a AIM-I polypeptide or polynucleotide to an organism to augment AIM-I function or remediate AIM-I dysfunction.
  • Ce ⁇ ain additional preferred aspects related to the above aspects of the invention provides antibodies against AIM-I polypeptides.
  • the antibodies are highly selective for human AIM-I, and may be employed, inter alia, to treat autoimmune diseases.
  • AIM-I agonists are provided.
  • preferred agonists are molecules that mimic AIM-I, that bind to AIM-I-binding molecules or receptor molecules, and that elicit or augment AIM-I- induced responses.
  • AIM-I antagonists are provided.
  • preferred antagonists are those which mimic AIM-I so as to bind to AIM-I receptor or binding molecules but not elicit an AIM-I-induced response or more than one AIM-I-induced response.
  • molecules that bind to or interact with AIM-I so as to inhibit an effect of AIM-I or more than one effect of AIM-I or which prevent expression of AIM-I.
  • the antagonists may be employed to prevent septic shock, inflammation, cerebral malaria, activation of the HIV virus, graft-host rejection, bone resorption, rheumatoid arthritis and cachexia.
  • compositions comprising an AIM-I polynucleotide or an AIM-I polypeptide for administration to cells in vitro. to cells ex vivo and to cells in vivo, or to a multicellular organism.
  • the compositions comprise an AIM-I polynucleotide for expression of an AIM-I polypeptide in a host organism for treatment of disease.
  • Pa ⁇ icularly preferred in this regard is expression in a human patient for treatment of a dysfunction associated with aberrant endogenous activity of AIM-I.
  • Figure 1 shows the nucleotide and deduced amino acid sequence of human AIM-I.
  • Figure 2 shows the regioas of similarity and identity between amino acid sequence of AIM-I of the present invention and human Fas ligand polypeptide (SEQ ID NO:3).
  • Figure 3 shows regions of similarity and identity between amino acid sequences of AIM-I of the present invention and human Fas ligand polypeptide, TNF ⁇ and TNF / 3.
  • Figure 4 shows structural and functional features of AIM-I deduced by the indicated techniques, as a function of amino acid sequence.
  • the term "digestion of DNA” refers to catalytic cleavage of DNA with a restriction enzyme that acts only upon ce ⁇ ain sequences in the DNA.
  • the various restriction enzymes referred to herein are all commercially available and their reaction conditions, cofactors and other requirements for use are known and routine to the skilled a ⁇ isan.
  • plasmid or DNA fragment is digested with about 2 units of enzyme in about 20 ⁇ l of reaction buffer.
  • isolating DNA fragments for plasmid construction typically 5 to 50 ⁇ g of DNA are digested with 20 to 250 units of enzyme in propo ⁇ ionately larger volumes.
  • Incubation times ol about 1 hour at 37 "C are ordinarily used, but conditions may vary in accordance with standard procedures, the supplier's instructions and the pa ⁇ iculars of the reaction.
  • reactions may be analyzed, and fragments may be purified by electrophoresis through an agarose or polyacrylamide gel, using well known methods that are routine for those skilled in the an.
  • genetic element generally means a polynucleotide comprising a region that encodes a polypeptide or a region that regulates transcription or translation or other processes impo ⁇ ant to expression of the polypeptide in a host cell, or a polynucleotide comprising both a region that encodes a polypeptide and a region operably linked thereto that regulates expression.
  • Genetic elements may be comprised within a vector that replicates as an episomal element; that is, as a molecule physically independent of the host cell genome. They may be comprised within mini-chromosomes, such as those that arise during amplification of transfected DNA by methotrexate selection in eukaryotic cells. Genetic elements also may be comprised within a host cell genome; not in their natural state but, rather, following manipulation such as isolation, cloning and introduction into a host cell in the form of purified DNA or in a vector, among others.
  • isolated means altered “by the hand of man” from its natural state; i.e. , that, if it occurs in nature, it has been changed or removed from its original environment, or both.
  • a naturally occurring polynucleotide or a polypeptide naturally present in a living animal in its natural state 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.
  • isolated means that it is separated from the chromosome and cell in which it naturally occurs
  • polynucleotides can be joined to other polynucleotides, such as DNAs. for mutagenesis, to form fusion proteins, and for propagation or expression in a host, for instance.
  • the isolated polynucleotides, alone or joined to other polynucleotides such as vectors, can be introduced into host cells, in culture or in whole organisms. Introduced into host cells in culture or in whole organisms, such DNAs still would be isolated, as the term is used herein, because they would not be in their naturally occurring form or environment.
  • polynucleotides and polypeptides may occur in a composition, such as a media formulations, solutions for introduction of polynucleotides or polypeptides, for example, into cells, compositions or solutions for chemical or enzymatic reactions, for instance, which are not naturally occurring compositions, and, therein remain isolated polynucleotides or polypeptides within the meaning of that term as it is employed herein.
  • a composition such as a media formulations, solutions for introduction of polynucleotides or polypeptides, for example, into cells, compositions or solutions for chemical or enzymatic reactions, for instance, which are not naturally occurring compositions, and, therein remain isolated polynucleotides or polypeptides within the meaning of that term as it is employed herein.
  • ligation refers to the process of forming phosphodiester bonds between two or more polynucleotides, which most often are double stranded DNAs. Techniques for ligation are well known to the an and protocols for ligation are described in standard laboratory manuals and references, such as, for instance, Sambrook et al. Molecular Cloning. A Laboratory Manual. 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989) and Maniatis et al, pg. 146, as cited below.
  • oligonucleotide(s) refers to relatively short polynucleotides. Often the term refers to single-stranded deoxyribonucleotides, but it can refer as well to single- or double-stranded ribonucleotides, RNA:DNA hybrids and double-stranded DNAs, among others.
  • Oligonucleotides such as single-stranded DNA probe oligonucleotides. often are synthesized by chemical makersxls. such as those implemented on automated oligonucleotide synthesizers. However, oligonucleotides can be made by a variety of other methods, including in vitro recombinant DNA-mediated techniques and by expression of DNAs in cells and organisms.
  • oligonucleotides typically are obtained without a 5' phosphate.
  • the 5 ' ends of such oligonucleotides are not substrates for phosphodiester bond formation by ligation reactions that employ DNA ligases typically used to form recombinant DNA molecules.
  • a phosphate can be added by standard techniques, such as those that employ a kinase and ATP.
  • the 3' end of a chemically synthesized oligonucleotide generally has a free hydroxyl group and, in the presence of a ligase, such as T4 DNA ligase, readily will form a phosphodiester bond with a 5' phosphate of another polynucleotide, such as another oligonucleotide. As is well known, this reaction can be prevented selectively, where desired, by removing the 5' phosphates of the other polynucleotide(s) prior to ligation.
  • a ligase such as T4 DNA ligase
  • Plasmids generally are designated herein by a lower case p preceded and/or followed by capital letters and/or numbers, in accordance with standard naming conventions that are familiar to those of skill in the art.
  • plasmids disclosed herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids by routine application of well known, published procedures. Many plasmids and other cloning and expression vectors that can be used in accordance with the present invention are well known and readily available to those of skill in the art. Moreover, those of skill readily may construct any number of other plasmids suitable for use in the invention. The properties, construction and use of such plasmids, as well as other vectors, in the present invention w ill be readily apparent to those of skill from the present disclosure.
  • polynucleoiidcisr generally refers to any polyribonucleotide or polydeoxribonucleotide. which mj> he unmodified RNA or DNA or modified RNA or DNA.
  • polynucleotides as used herein refers to, among others, single-and double-stranded DNA.
  • polynucleotide as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • the strands in such regions may be from the same molecule or from different molecules.
  • the regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules.
  • One of the molecules of a triple-helical region often is an oligonucleotide.
  • polynucleotide includes DNAs or RNAs as described above that contain one or more modified bases.
  • DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein.
  • DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples are polynucleotides as the term is used herein.
  • polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolicallv modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells, inter alia.
  • polypeptides as used herein, includes all polypeptides as described below.
  • the basic structure of polypeptides is well known and has been described in innumerable textbooks and other publications in the art.
  • the term is used herein to refer to any peptide or protein comprising two or more amino acids joined to each other in a linear chain by peptide bonds.
  • the term refers to both sho ⁇ chains, which also commonly are referred to in the an as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the an as proteins, of which there are many types.
  • polypeptides often contain amino acids other than the 20 amino acids commonly referred to as the 20 naturally occurring amino acids, and that many amino acids, including the terminal amino acids, may be modified in a given polypeptide, either by natural processes, such as processing and other post-translational modifications, but also by chemical modification techniques which are well known to the an. Even the common modifications that occur naturally in polypeptides are too numerous to list exhaustively here, but they are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature, and they are well known to those of skill in the an.
  • polypeptides of the present are, to name an illustrative few, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation. oxidation, proteolytic processing, phosphorylation. prenylation. racemization. selenoylation. sulfation. transfer-RNA mediated addition of amino acids to proteins such as
  • polypeptides are not always entirely linear.
  • polypeptides may be branched as a result of ubiquitination, and they may be circular, with or without branching, generally as a result of posttranslation events, including natural processing event and events brought about by human manipulation which do not occur naturally.
  • Circular, branched and branched circular polypeptides may be synthesized by non-translation natural process and by entirely synthetic methods, as well.
  • Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini.
  • blockage of the amino or carboxyl group in a polypeptide, or both, by a covalent modification is common in naturally occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention, as well.
  • the amino terminal residue of polypeptides made in E. coli, prior to proteolytic processing almost invariably w ill be N-formylmethionine.
  • modifications that t ⁇ cur in a polypeptide often will be a function of how it is made.
  • polypeptides made b ⁇ expressing a cloned gene in a host for instance, the nature and extent of the modifications in large part will be determined by the host cell posttranslational modification capacity and the modification signals present in the polypeptide amino acid sequence
  • glycosylation often does not occur in bacterial hosts such as E. coli. Accordingly, when glycosylation is desired, a polypeptide should be expressed in a glycosylating host, generally a eukaryotic cell.
  • Insect cell often carry out the same posttranslational glycosylations as mammalian cells and, for this reason, insect cell expression systems have been developed to express efficiently mammalian proteins having native patterns of glycosylation, inter alia. Similar considerations apply to other modifications.
  • polypeptide encompasses all such modifications, particularly those that are present in polypeptides synthesized by expressing a polynucleotide in a host cell.
  • variant(s) of polynucleotides or polypeptides, as the term is used herein, are polynucleotides or polypeptides that differ from a reference polynucleotide or polypeptide, respectively. Variants in this sense are described below and elsewhere in the present disclosure in greater detail.
  • a polynucleotide variant can be, for example, a polynucleotide that differs in nucleotide sequence from another, reference polynucleotide. Generally, differences are limited so that the nucleotide sequences of the reference and the variant are closely similar overall and. in many regions, identical.
  • changes m the nucleotide sequence of the variant may be silent. That is, they may not alter the ammo acids encoded by the polynucleotide. Where alterations are limited to silent changes of this type a variant will encode a polypeptide with the same amino acid sequence as the reference. Also as noted below, changes in t.' ⁇ nucleotide sequence of the variant may alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Such nucleotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below.
  • a polynucleotide variant can be, for example, a polypeptide that differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference and the variant are closely similar overall and, in many region, identical.
  • a variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions, fusions and truncations, which may be present in any combination.
  • receptor molecule refers to molecules which bind or interact specifically with AIM-I polypeptides of the present invention, including not only classic receptors, which are preferred, but also other molecules that specifically bind to or interact with polypeptides of the invention (which also may be referred to as “binding molecules” and “interaction molecules. " respectively and as "AIM-I binding molecules” and "AIM-I interaction molecules.
  • Binding between polypeptides of the invention and such molecules, including receptor or binding or interaction molecules may be exclusive to polypeptides of the invention, which is very highly preferred, or it may be highly specific for polypeptides of the invention, which is highly preferred, or it may be highly specific to a group of proteins that includes polypeptides of the invention, which is preferred, or it may be specific to several groups of proteins at least one of which includes polypeptides of the invention.
  • Receptors also may be non-naturallv occurring, such as antibodies and antibody- derived reagents that bind specifically to polypeptides of the invention.
  • the present invention relates to novel AIM-I polypeptides and polynucleotides, among other things, as described in greater detail below.
  • the invention relates to polypeptides and polv nucleotides of a novel human AIM-I, which is related by amino acid sequence homology to known human AIM-I.
  • the invention relates especially to AIM-I having the nucleotide and amino acid sequences set out in Figure 1 , and to the AIM-I nucleotide and amino acid sequences of the cDNA in ATCC Deposit No. 97448, which is herein referred to as "the deposited clone" or as the "cDNA of the deposited clone.
  • nucleotide and amino acid sequences set out in Figure 1 were obtained by sequencing the cDNA of the deposited clone.
  • sequence of the deposited clone is controlling as to any discrepancies between the two and any reference to the sequences of Figure 1 include reference to the sequence of the human cDNA of the deposited clone.
  • isolated polynucleotides which encode the AIM-I polypeptide having the deduced amino acid sequence of Figure 1 or the AIM-I polypeptide encoded by the cDNA in the deposited clone.
  • a polynucleotide of the present invention encoding human AIM-I polypeptide may be obtained using standard cloning and screening procedures, such as those for cloning cDNAs using human tissue as starting material.
  • standard cloning and screening procedures such as those for cloning cDNAs using human tissue as starting material.
  • the polynucleotide set out in Figure 1 was discovered in a cDNA library derived from cells of a human pancreas rumor.
  • Human AIM-I of the is structurally related to other proteins of the TNF family, as shown by the results ot sequencing the human cDNA encoding human AIM-I in the deposited clone
  • the cl)V ⁇ sequence thus obtained is set out in Figure 1 . It contains an open readinj. frame encodini! a protein of about 281 amino acid residues with a deduced molecular weight of about 31 kDa. The protein exhibits greatest homology to known human AIM I . among known proteins.
  • the AIM-I of Figure 1 has about 22.892 % identity and about 48.594 % similarity with the amino acid sequence of human Fas ligand.
  • Polynucleotides of the present invention may be in the form of RNA, such as mRNA, or in the form cDNA and genomic DNA obtained by cloning or produced by chemical synthetic techniques or by a combination thereof.
  • the DNA may be double- stranded or single-stranded.
  • Single-stranded DNA may be the coding strand, also known as the sense strand, or it may be the non-coding strand, also referred to as the anti-sense strand.
  • the coding sequence which encodes the polypeptide may be identical to the coding sequence of the polynucleotide shown in Figure 1. It also may be a polynucleotide with a different sequence, which, as a result of the redundancy (degeneracy) of the genetic code, encodes the polypeptide of the DNA of Figure 1.
  • Polynucleotides of the present invention which encode the polypeptide of Figure 1 may include, but are not limited to the coding sequence for the mature polypeptide, by itself; the coding sequence for the mature polypeptide and additional coding sequences, such as those encoding a leader or secretory sequence, such as a pre-, or pro- or prepro- protein sequence: the coding sequence of the mature polypeptide, with or without the aforementioned additional coding sequences, together with additional, non-coding sequences, including for example, but not limited to introns and non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription.
  • the polypeptide may be fused to a marker sequence, such as a peptide. which facilitates purification of the fused polypeptide.
  • the marker sequence is a hexa-histidine peptide, such as the tag provided in the pQE vector (Qiagen, Inc. , Chatsworth, CA), : -long others, many of which are commercially available. As described in Gentz et al , Proc. Natl. Acad. Sci..
  • hexa-histidine provides for convenient purification of the fusion protein.
  • the HA tag corresponds to an epitope derived of influenza hemagglutinin protein, which has been described by Wilson et al , Cell 37:161 (1984), for instance.
  • polynucleotide encoding a polypeptide encompasses polynucleotides which include a sequence encoding a polypeptide of the present invention, particularly the human AIM-I having the amino acid sequence set out in Figure 1.
  • the term encompasses polynucleotides that include a single continuous region or discontinuous regions encoding the polypeptide (for example, interrupted by introns) together with additional regions, that also may contain coding and/or non-coding sequences.
  • the present invention further relates to variants of the herein above described polynucleotides which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of Figure 1.
  • a variant of the polynucleotide may be a namrally occurring variant such as a naturally occurring allelic variant, or it may be a variant that is not known to occur naturally.
  • Such non-narurally occurring variants of the polynucleotide may be made by mutagenesis techniques, including those applied to polynucleotides, cells or organisms.
  • variants in this regard are variants that differ from the aforementioned polynucleotides by nucleotide substimtions. deletions or additions. The substitutions, deletions or additions may involve one or more nucleotides. The variants may be altered in coding or non-coding regions or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions.
  • polynucleotides encoding polypeptides having the amino acid sequence of AIM-I set out in Figure 1 ; variants, analogs, derivatives and fragments thereof, and fragments of the variants, analogs and derivatives.
  • polynucleotides encoding AIM-I variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments which have the amino acid sequence of the AIM-I polypeptide of Figure 1 in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination.
  • Fu ⁇ her preferred embodiments of the invention are polynucleotides that are at least 70% identical to a polynucleotide encoding the AIM-I polypeptide having the amino acid sequence set out in Figure 1 , and polynucleotides which are complementary to such polynucleotides.
  • most highly preferred are polynucleotides that comprise a region that is at least 80% identical to a polynucleotide encoding the AIM-I polypeptide of the cDNA of the deposited clone and polynucleotides complementary thereto.
  • polynucleotides at least 90% identical to the same are particularly preferred, and among these pa ⁇ icularly preferred polynucleotides, those with at least 95 % are especially preferred. Furthermore, those with at least 97% are highly preferred among those with at least 95%. and among these those with at least 98% and at least 99 c i are particularly highly preferred, with at least 99% being the more preferred.
  • polynucleotides which encode polypeptides which retain substantially the same biological function or activity as the mature pol ⁇ peptide encoded by the cDNA of Figure 1.
  • the present invention further relates to polynucleotides that hybridize to the herein above-described sequences
  • the present invention especially relates to polynucleotides which hybridize under stringent conditions to the herein above- described polynucleotides.
  • stringent conditions means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences.
  • polynucleotides of the invention may be used as a hybridization probe for cDNA and genomic DNA to isolate full-length cDNAs and genomic clones encoding human AIM-I and to isolate cDNA and genomic clones of other genes that have a high sequence similarity to the human AIM-I gene.
  • Such probes generally will comprise at least 15 bases.
  • such probes will have at least 30 bases and may have at least 50 bases.
  • Particularly preferred probes will have at least 30 bases and will have 50 bases or less.
  • the coding region of the AIM-I gene may be isolated by screening using the known DNA sequence to synthesize an oligonucleotide probe.
  • a labeled oligonucleotide having a sequence complementary to that of a gene of the present invention is then used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes to.
  • polynucleotides and polypeptides of the present invention may be employed as research reagents and materials for discovery of treatments and diagnostics to human disease, as further discussed herein relating to polynucleotide assays, inter alia.
  • the polynucleotides ma> encode a polypeptide which is the mature protein plus additional amino or carboxyl-terminal amino acids, or amino acids interior to the mature polypeptide (when the mature l rm has more than one polypeptide chain, for instance).
  • Such sequences may play a role in processing of a protein from precursor to a mature form, may facilitate protein trafficking, may prolong or shorten protein half-life or may facilitate manipulation of a protein for assay or production, among other things.
  • the additional amino acids may be processed away from the mamre protein by cellular enzymes.
  • a precursor protein, having the mamre form of the polypeptide fused to one or more prosequences may be an inactive form of the polypeptide.
  • inactive precursors When prosequences are removed such inactive precursors generally are activated. Some or all of the prosequences may be removed before activation. Generally, such precursors are called proproteins.
  • a polynucleotide of the present invention may encode a mamre protein, a mamre protein plus a leader sequence (which may be referred to as a preprotein), a precursor of a mature protein having one or more prosequences which are not the leader sequences of a preprotein. or a preproprotein. which is a precursor to a proprotein, having a leader sequence and one or more prosequences, which generally are removed during processing steps that produce active and mamre forms of the polypeptide.
  • a leader sequence which may be referred to as a preprotein
  • a precursor of a mature protein having one or more prosequences which are not the leader sequences of a preprotein.
  • a preproprotein which is a precursor to a proprotein, having a leader sequence and one or more prosequences, which generally are removed during processing steps that produce active and mamre forms of the polypeptide.
  • a deposit containing a human AIM-I cDNA has been deposited with the American Type Culture Collection, as noted above. Also as noted above, the human cDNA deposit is referred to herein as “the deposited clone” or as “the cDNA of the deposited clone. "
  • the deposited clone was deposited with the American Type Culture Collection, 12301 Park Lawn Dme. Rockv ille. Mar land 20852, USA. on February 20, 1996, and assigned ATCC Deposit No. 97448.
  • the deposited material is a pBluescript SK (-) plasmid (Stratagene, La Jolla, CA) that contains the full length AIM-I cDNA. referred to as "PF261 " upon deposit.
  • the deposit has been made under the terms of the Budapest Treaty on the international recognition of the deposit of micro-organisms for purposes of patent procedure. The strain will be irrevocably and without restriction or condition released to the public upon the issuance of a patent.
  • the deposit is provided merely as convenience to those of skill in the art and is not an admission that a deposit is required for enablement, such as that required under 35 U.S.C. ⁇ 112.
  • sequence of the polynucleotides contained in the deposited material, as well as the amino acid sequence of the polypeptide encoded thereby, are controlling in the event of any conflict with any description of sequences herein.
  • a license may be required to make, use or sell the deposited materials, and no such license is hereby granted.
  • the present invention further relates to a human AIM-I polypeptide which has the deduced amino acid sequence of Figure 1.
  • the invention also relates to fragments, analogs and derivatives of these polypeptides.
  • fragment when referring to the polypeptide of Figure 1 means a polypeptide which retains essentially the same biological function or activity as such polypeptide.
  • an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mamre polypeptide.
  • polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic In ce ⁇ ain preferred embodiments it is a recombinant polypeptide.
  • the fragmeni. derivative or analog of the polypeptide of Figure 1 may be (i) one in which one or more of the ammo acid residues are substituted with a conserved or non-conserved ammo acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mamre polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mamre polypeptide.
  • particularly preferred embodiments of the invention in this regard are polypeptides having the amino acid sequence of AIM-I set out in Figure 1 , variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments.
  • particularly preferred embodiments of the invention in this regard are polypeptides having the amino acid sequence of the AIM-I of the cDNA in the deposited clone, variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments.
  • Such substimtions are those that substitute a given amino acid in a polypeptide by another amino acid of like characteristics. Typically seen as conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala. Val. Leu and He; interchange of the hydroxyl residues Ser and Thr. exchange of the acidic residues Asp and Glu. substimtion between the amide residues Asn and Gin, exchange of the basic residues Lys and Arg and replacements among the aromatic residues Phe. Tyr.
  • variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments having the amino acid sequence of the AIM-I polypeptide of Figure 1 in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination.
  • silent substimtions, additions and deletions which do not alter the properties and activities of the AIM-I.
  • conservative substimtions are also especially preferred in this regard.
  • polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
  • polypeptides of the present invention include the polypeptide of SEQ ID NO: 2 (in particular the mamre polypeptide) as well as polypeptides which have at least 70% similarity (preferably at least 70% identity) to the polypeptide of SEQ ID NO:2 and more preferably at least 90% similarity (more preferably at least 90% identity) to the polypeptide of SEQ ID NO:2 and still more preferably at least 95% similarity (still more preferably at least 95 % identity) to the polypeptide of SEQ ID NO:2 and also include portions of such polypeptides with such portion of the polypeptide generally containing at least 30 amino acids and more preferably at least 50 amino acids.
  • Fragments or portions of the polypeptides of the present invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention.
  • polypeptides comprising fragments of AIM-I, most particularly fragments of the AIM-I having the amino acid set out in Figure 1 , and fragments of variants and derivatives of the AIM-I of Figure 1.
  • a fragment is a polypeptide having an amino acid sequence that entirely is the same as part but not all of the amino acid sequence of the aforementioned AIM-I polypeptides and variants or derivatives thereof.
  • fragments may be "free-standing, " i.e. , not part of or fused to other amino acids or polypeptides, or they may be comprised within a larger polypeptide of which they form a part or region. When comprised within a larger polypeptide. the presently discussed fragments most preferably form a single continuous region. However, several fragments may be comprised within a single larger polypeptide. For instance, certain preferred embodiments relate to a fragment of an AIM-I polypeptide of the present comprised within a precursor polypeptide designed for expression in a host and having heterologous pre and pro-polypeptide regions fused to the amino terminus of the AIM-I fragment and an additional region fused to the carboxyl terminus of the fragment. Therefore, fragments in one aspect of the meaning intended herein, refers to the portion or portions of a fusion polypeptide or fusion protein derived from AIM-I.
  • polypeptide fragments of the invention there may be mentioned those w hich have from about 100 to about 281 amino acids.
  • “about” includes the particularly recited range and ranges larger or smaller by several, a few. 5.
  • about 100-281 amino acids in this context means a polypeptide fragment of 100 plus or minus several, a few . 5, 4, 3, 2 or 1 amino acids to 281 plus or minus several a few . 5.
  • 4. 3. 2 or I amino acid residues, i.e. ranges as broad as 100 minus several amino acids 10 281 plus several amino acids to as narrow as 100 plus several amino acids to 281 minus several amino acids.
  • Truncation mutants include AIM-I polypeptides having the amino acid sequence of Figure 1, or of variants or derivatives thereof, except for deletion of a continuous series of residues (that is, a continuous region, pan or po ⁇ ion) that includes the amino terminus, or a continuous series of residues that includes the carboxyl terminus or, as in double truncation mutants, deletion of two continuous series of residues, one including the amino terminus and one including the carboxyl terminus. Fragments having the size ranges set out about also are preferred embodiments of truncation fragments, which are especially preferred among fragments generally.
  • fragments characterized by structural or functional attributes of AIM-I are also preferred in this aspect of the invention.
  • Preferred embodiments of the invention in this regard include fragments that comprise alpha-helix and alpha-helix forming regions ("alpha-regions"), beta-sheet and beta-sheet-forming regions ("beta-regions"), turn and turn-forming regions ("turn- regions”), coil and coil-forming regions ("coil-regions”), hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface-forming regions and high antigenic index regions of AIM-I.
  • Ce ⁇ ain preferred regions in these regards are set out in Figure 3, and include, but are not limited to. regions of the aforementioned types identified by analysis of the amino acid sequence set out in Figure 1 .
  • such preferred regions include Gamier- Robson alpha-regions, beta-regions, turn-regions and coil-regions, Chou-Fasman alpha-regions, beta-regions and turn-regions, Kyte-Doolittle hydrophilic n. _ions and hydrophilic regions. Eisenberg alpha and beta amphipathic regions, Karplus- Schulz flexible regions. Emini surface-forming regions and Jameson-Wolf high antigenic index regions.
  • highly preferred fragments in this regard are those that comprise regions of AIM-I that combine several structural features, such as several of the features set out above.
  • the regions defined by the residues about 1 to about 281 of Figure 1 which all are characterized by amino acid compositions highly characteristic of turn-regions, hydrophilic regions, flexible-regions, surface-forming regions, and high antigenic index-regions, are especially highly preferred regions.
  • Such regions may be comprised within a larger polypeptide or may be by themselves a preferred fragment of the present invention, as discussed above. It will be appreciated that the term "about” as used in this paragraph has the meaning set out above regarding fragments in general.
  • a highly preferred fragment comprises amino acids 39 through 281 which constitute a soluble po ⁇ ion of the overall AIM-I polypeptide sequence of Figure 1.
  • Fu ⁇ her preferred regions are those that mediate activities of AIM-I.
  • Most highly preferred in this regard are fragments that have a chemical, biological or other activity of AIM-I, including those with a similar activity or an improved activity, or with a decreased undesirable activity.
  • Highly preferred in this regard are fragments that contain regions that are homologs in sequence, or in position, or in both sequence and to active regions of related polypeptides. such as the related polypeptides set out in Figure 2. which include members of the TNF family.
  • particularly preferred fragments in these regards are truncation mutants, as discussed above.
  • the invention also relates to. among others, polynucleotides encoding the aforementioned fragments, polynucleotides that hybridize to polynucleotides encoding the tragments. particularly those that hybridize under stringent conditions, and po I nucleotides. such as PCR primers, for amplifying polynucleotides that encode the tragments
  • preferred polynucleotides are those that correspondent to the preferred fragments, as discussed above.
  • the present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.
  • Host cells can be genetically engineered to incorporate polynucleotides and express polypeptides of the present invention.
  • polynucleotides may be introduced into host cells using well known techniques of infection, transduction, transfection, transvection and transformation.
  • the polynucleotides may be introduced alone or with other polynucleotides.
  • Such other polynucleotides may be introduced independently, co-introduced or introduced joined to the polynucleotides of the invention.
  • polynucleotides of the invention may be transfected into host cells with another, separate, polynucleotide encoding a selectable marker, using standard techniques for co-transfection and selection in, for instance, mammalian cells.
  • polynucleotides generallv w ill be stably incorporated into the host cell genome.
  • the polynucleotides may be joined to a vector containing a selectable marker for propagation in a host.
  • the vector construct may be introduced into host cells by the aforementioned techniques.
  • a plasmid vector is introduced as DNA in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid tlectroporation also may be used to introduce polynucleotides into a host.
  • the vector is a virus, it may be packaged in vitro or introduced into a packaging cell and the packaged virus may be transduced into cells.
  • the vector may be, for example, a plasmid vector, a single or double-stranded phage vector, a single or double-stranded RNA or DNA viral vector.
  • Such vectors may be introduced into cells as polynucleotides, preferably DNA, by well known techniques for introducing DNA and RNA into cells.
  • the vectors in the case of phage and viral vectors also may be and preferably are introduced into cells as packaged or encapsidated virus by well known techniques for infection and transduction.
  • Viral vectors may be replication competent or replication defective. In the latter case viral propagation generally will occur only in complementing host cells.
  • vectors are those for expression of polynucleotides and polypeptides of the present invention.
  • such vectors comprise -acting control regions effective for expression in a host operatively linked to the polynucleotide to be expressed.
  • Appropriate trans-acting factors either are supplied by the host, supplied by a complementing vector or supplied by the vector itself upon introduction into the host.
  • the vectors provide for specific expression.
  • Such specific expression may be inducible expression or expression only in ce ⁇ ain types of cells or both inducible and cell-specific.
  • Pa ⁇ icularly preferred among inducible vectors are vectors that can be induced for expression by environmental factors that are easy to manipulate, such as temperamre and nutrient additives.
  • a variety of vectors suitable to this aspect of the invention, including constimtive and inducible expression vectors for use in prokaryotic and eukaryotic hosts, are well known and employed routinely bv those of skill in the an.
  • the engineered host cells can be cultured in conventional nutrient media, which may be modified as appropriate for. inter alia, activating promoters, selecting transformants or amplifying genes. Culture conditions, such as temperamre, pH and the 1 > '. previously used with the host cell selected for expression generally will be suitable for expression of polypeptides of the present invention as will be apparent to those of skill in the an.
  • a great variety of expression vectors can be used to express a polypeptide of the invention.
  • Such vectors include chromosomal, episomal and virus-derived vectors e.g.
  • any vector suitable to maintain, propagate or express polynucleotides to express a polypeptide in a host may be used for expression in this regard.
  • the appropriate DNA sequence may be inse ⁇ ed into the vector by any of a variety of well-known and routine techniques.
  • a DNA sequence for expression is joined to an expression vector by cleaving the DNA sequence and the expression vector with one or more restriction endonucleases and then joining the restriction fragments together using T4 DNA ligase.
  • Procedures for restriction and ligation that can be used to this end are well known and routine to those of skill. Suitable procedures in this regard, and for constmcting expression vectors using alternative techniques, which also are well known and routine to those skill, are set forth in great detail in Sambrook et al cited elsewhere herein.
  • the DNA sequence in the expression vector is operatively linked to appropriate expression control sequence! s».
  • appropriate expression control sequence! s» including, for instance, a promoter to direct mRNA transcription.
  • promoters include the phage lambda PL promoter, the E. coli lac. tip and tae promoters, the SV40 early and late promoters and promoters of retroviral LTRs. to name just a few of the well-known promoters. It will be understood that numerous promoters not mentioned are suitable for use in this aspect of the invention are well known and readily may be employed by those of skill in the manner illustrated by the discussion and the examples herein.
  • expression constructs will contain sites for transcription initiation and termination, and, in the transcribed region, a ribosome binding site for translation.
  • the coding po ⁇ ion of the mamre transcripts expressed by the constructs will include a translation initiating AUG at the beginning and a termination codon appropriately positioned at the end of the polypeptide to be translated.
  • constmcts may contain control regions that regulate as well as engender expression.
  • control regions that regulate as well as engender expression.
  • such regions will operate by controlling transcription, such as repressor binding sites and enhancers, among others.
  • Vectors for propagation and expression generally will include selectable markers. Such markers also may be suitable for amplification or the vectors may contain additional markers for this purpose.
  • the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells.
  • Preferred markers include dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, and tetracycline or ampicillin resistance genes for culruring £ coli and other bacteria.
  • the vector containing the appropriate DNA sequence as described elsewhere herein, as well as an appropriate promoter, and other appropriate control sequences, may be introduced into an appropriate host using a variety of well known techniques suitable to expression therein of a desired polypeptide.
  • appropriate hosts include bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as east cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells, animal cells such as CHO, COS and Bowes melanoma cells; and plant cells.
  • the present invention also includes recombinant constmcts, such as expression constmcts, comprising one or more of the sequences described above.
  • the constmcts comprise a vector, such as a plasmid or viral vector, into which such a sequence of the invention has been inse ⁇ ed.
  • the sequence may be inse ⁇ ed in a forward or reverse orientation.
  • the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence.
  • regulatory sequences including, for example, a promoter, operably linked to the sequence.
  • vectors preferred for use in bacteria are pQE70, pQE60 and pQE- 9, available from Qiagen: pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A. pNHl ⁇ a, pNH18A, pNH46A. available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540. pRIT5 available from Pharmacia.
  • preferred eukaryotic vectors are pWLNEO.
  • pMSG and pSVL available from Pharmacia. These vectors are listed solely by way of illustration of the many commercially available and well known vectors that are available to those of skill in the art for use in accordance with this aspect of the present invention. It will be appreciated that any other plasmid or vector suitable for. for example, introduction, maintenance, propagation or expression of a polynucleotide or polypeptide of the invention in a host may be used in this aspect of the invention.
  • Promoter regions can be selected from any desired gene using vectors that contain a reporter transcript ion unit lacking a promoter region, such as a chloramphenicol acetyl transferase ( " cat" ) transcription unit, downstream of restriction site or sites for introducing a candidate promoter fragment; i.e. , a fragment that may contain a promoter.
  • a reporter transcript ion unit lacking a promoter region such as a chloramphenicol acetyl transferase ( " cat” ) transcription unit, downstream of restriction site or sites for introducing a candidate promoter fragment; i.e. , a fragment that may contain a promoter.
  • introduction into the vector of a promoter- containing fragment at the restriction site upstream of the cat gene engenders production of CAT activity, which can be detected by standard CAT assays.
  • Vectors suitable to this end are well known and readily available. Two such vectors are pKK232-8 and pCM7.
  • bacterial promoters suitable for expression of polynucleotides and polypeptides in accordance with the present invention are the £. coli lad and lacZ and promoters, the T3 and T7 promoters, the T5 tae promoter, the lambda PR, PL promoters and the trp promoter.
  • known eukaryotic promoters suitable in this regard are the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous sarcoma vims ("RSV"). and metallothionein promoters, such as the mouse metallothionein-I promoter.
  • the present invention also relates to host cells containing the above-described constmcts discussed above.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukar otic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • constructs into the host cell can be effected by calcium phosphate transfection. DE ⁇ F.-dextran mediated transfection, cationic lipid-mediated transfection, electroporation. transduction. infection or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al. Basic Methods In Molecular Biolo ⁇ v. ( 1986). Constmcts in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
  • Mamre proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constmcts of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook et al. Molecular Cloning: A Laboratory Manual. 2nd Ed.. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989).
  • recombinant expression vectors will include origins of replication, a promoter derived from a highly -expressed gene to direct transcription of a downstream structural sequence, and a selectable marker to permit isolation of vector containing cells after exposure to the vector.
  • Enhancers are cis-acting elements of DNA. usually about from 10 to 300 bp that act to increase transcriptional activity of a promoter in a given host cell-type.
  • enhancers include the SV4O enhancer, w hich is located on the late side of the replication origin at bp 100 to 270. the cv iomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovims enhancers.
  • Polynucleotides of the invention encoding the heterologous structural sequence of a polypeptide of the invention generally w ill be inse ⁇ ed into the vector using standard techniques so that it is onerablv linked to the promoter for expression.
  • the polynucleotide will he positioned so that the transcription sta ⁇ site is located appropriately 5' to a ribosome binding site.
  • the ribosome binding site will be 5' to the AUG that initiates translation of the polypeptide to be expressed.
  • secretion signals may be incorporated into the expressed polypeptide.
  • the signals may be endogenous to the polypeptide or they may be heterologous signals.
  • the polypeptide may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals but also additional heterologous functional regions.
  • a region of additional amino acids, particularly charged amino acids may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification or during subsequent handling and storage.
  • region also may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide.
  • the addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the an.
  • the selected promoter is inducible it is induced by appropriate means (e.g.. temperature shift or exposure to chemical inducer) and cells are cultured for an additional period. Cells typically then are harvested by centrifugation. dismpted by physical or chemical means, and the resulting cmde extract retained for further purification.
  • Microbial cells employed in expression of proteins can be dismpted by any convenient method, including freeze-thaw cycling, sonication, mechanical dismption. or use of cell lysing agents, such methods are well know to those skilled in the art.
  • mammalian cell cul re systems can be employed for expression, as well.
  • mammalian expression systems include the COS-7 lines of monkey kidney fibroblast, described in Gluzman et al, Cell, 23: 175 (1981).
  • Other cell lines capable of expressing a compatible vector include for example, the C127, 3T3, CHO, HeLa, human kidney 293 and BHK cell lines.
  • the AIM-I polypeptide can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxy lapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification.
  • HPLC high performance liquid chromatography
  • Polypeptides of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosv lated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host- mediated processes.
  • AIM-I polynucleotides and polypeptides may be used in accordance with the present invention for a variety of applications, particularly those that make use of the chemical and biological properties AIM-I. Among these are applications in autoimmune disease and aberrant cellular proliferation. Additional applications relate to diagnosis and to treatment of disorders of cells, tissues and organisms. These aspects of the invention are illustrated further by the following discussion.
  • This invention is also related to the use of the AIM-I polynucleotides to detect complementary polynucleotides such as, for example, as a diagnostic reagent. Detection of a mutated form of AIM-I associated with a dysfunction will provide a diagnostic tool that can add or define a diagnosis of a disease or susceptibility to diseases which results from under-expression over-expression or altered expression of AIM-I, such as, for example, autoimmune diseases.
  • Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material.
  • the genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR prior to analysis.
  • PCR Saiki et al, Nature, 324: 163-166, ( 1986).
  • RNA or cDNA may also be used in the same ways.
  • PCR primers complementary to the nucleic acid encoding AIM-I can be used to identify and analyze AIM-1 expression and mutations.
  • deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype.
  • Point mutations can be identified by hybridizing amplified DNA to radiolabeled AIM-I RS ⁇ or alterrutivel .
  • radiolabeled AIM-I antisense DNA sequences Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melung lemperatures.
  • Sequence differences between a reference gene and genes having mutations also may be revealed by direct DNA sequencing.
  • cloned DNA segments may be employed as probes to detect specific DNA segments.
  • the sensitivity of such methods can be great Iv enhanced bv appropriate use of PCR or another amplification method.
  • a sequencing primer is used with double-stranded PCR product or a single-stranded template molecule generated by a modified PCR.
  • the sequence determination is performed by conventional procedures with radiolabeled nucleotide or by automatic sequencing procedures with fluorescent- tags.
  • DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels, with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g. , Myers et al, Science, 230: 1242, 1985).
  • Sequence changes at specific locations also may be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method (e.g. , Cotton er a/.. Proc. Natl. Acad. Sci., USA, 55:4397-4401 , 1985).
  • nuclease protection assays such as RNase and SI protection or the chemical cleavage method (e.g. , Cotton er a/.. Proc. Natl. Acad. Sci., USA, 55:4397-4401 , 1985).
  • the detection of a specific DNA sequence may be achieved by methods such as hybridization. RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g.. restriction fragment length polymorphisms ("RFLP”) and Southern blotting of genomic DNA.
  • restriction enzymes e.g.. restriction fragment length polymorphisms ("RFLP")
  • RFLP restriction fragment length polymorphisms
  • mutations also can be detected h ⁇ in situ analysis.
  • sequences of the present invention are also valuable for chromosome identification.
  • the sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome.
  • Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location.
  • the mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease.
  • the cDNA herein disclosed is used to clone genomic DNA of an AIM-I gene. This can be accomplished using a variety of well known techniques and libraries, which generally are available commercially.
  • the genomic DNA the is used for in situ chromosome mapping using well known techniques for this purpose. Typically, in accordance with routine procedures for chromosome mapping, some trial and error may be necessary to identify a genomic probe that gives a good in situ hybridization signal.
  • sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA. Computer analysis of the 3' untranslated region of the gene is used to rapidly select primers that do not span more than one exon in the genomic DNA. thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment.
  • PCR mapping of somatic cell hybrids is a rapid procedure for assigning a pa ⁇ icular DNA to a particular chromosome.
  • suhlocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner.
  • Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreemng w ith labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries.
  • Fluorescence in situ hybridization of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step.
  • This technique can be used with cDNA as short as 50 or 60.
  • Verma et al Human Chromosomes: A Manual Of Basic Techniques, Pergamon Press, New York (1988).
  • a cDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 jnu 500 potential causative genes. (This assumes 1 megabase mapping resolution and one gene per 20 kb).
  • the present invention alv> relates to a diagnostic assays such as quantitative and diagnostic assays for detecting levels of AIM-I protein in cells and tissues, including determination of normal and abnormal levels.
  • a diagnostic assay in accordance with the invention tor detecting over-expression of AIM-I protein compared to normal control tissue samples may be used to detect the presence of aberrant cellular proliferation such as cancer, for example.
  • Assay techniques that can be used to determine levels of a protein, such as an AIM-I protein of the present invention, in a sample derived from a host are well-known to those of skill in the an. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays.
  • An ELISA assay initially comprises preparing an antibody specific to AIM-I, preferably a monoclonal antibody.
  • a reponer antibody generally is prepared which binds to the monoclonal antibody.
  • the reponer antibody is attached a detectable reagent such as radioactive, fluorescent or enzymatic reagent, in this example horseradish peroxidase enzyme.
  • a sample is removed from a host and incubated on a solid suppo ⁇ . e.g. a polystyrene dish, that binds the proteins in the sample. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein such as bovine semm albumin.
  • the monoclonal antibody is incubated in the dish during which time the monoclonal antibodies attach to any AIM-I proteins attached to the polystyrene dish. Unbound monoclonal antibody is washed out with buffer.
  • the reponer antibody linked to horseradish peroxidase is placed in the dish resulting in binding of the reponer antibody to any monoclonal antibody bound to AIM-I.
  • Unattached reponer antibody is then washed out.
  • Reagents for peroxidase activity including a colorimetric substrate are then added to the dish.
  • Immobilized peroxidase, linked to AIM-I through the primary and secondary antibodies, produces a colored reaction product.
  • the amount of color developed in a given time period indicates the amount of AIM-I protein present in the sample.
  • Quantitative results typically are obtained by reference to a standard curve.
  • a competition assay may be employed wherein antibodies specific to AIM-I attached to a solid suppo ⁇ and labeled AIM-I and a sample derived from the host are passed over the solid suppo ⁇ and the amount of label detected attached to the solid suppo ⁇ can be correlated to a quantity of AIM-I in the sample.
  • the polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto. These antibodies can be, for example, polyclonal or monoclonal antibodies.
  • the present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the an may be used for the production of such antibodies and fragments.
  • Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
  • any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler. G. and Milstein, C., Nature, 256:495-497 (1975), the trioma technique, the human B-cell hybridoma technique (Kozbor et al. Immunology Today, 4:12 (1983) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al . pg. 77-96 in Monoclonal Antibodies And Cancer Therapy, Alan R. Liss. Inc. ( 1985 ).
  • the AIM-I of the present invention may be employed to treat lymphoproliferative disease which results in lymphadenopathy, the AIM-I mediates apoptosis by stimulating clonal deletion of T-cells and may therefore, by employed to treat autoimmune disease, to stimulate peripheral tolerance and cytotoxic T-cell mediated apoptosis.
  • the AIM-1 may also be employed as a research tool in elucidating the biology of autoimmune disorders including systemic lupus erythematosus, immunoproliferative disease lymphadenopathy (IPL), angioimmunoproliferative lymphadenopathy (AIL), rheumatoid a ⁇ hritis. diabetes, and multiple sclerosis, and to treat graft versus host disease.
  • the AIM-I may of the present invention may also be employed to inhibit neoplasia, such as tumor cell growth.
  • the AIM-I polypeptide may be responsible for tumor destmction through apoptosis and cytotoxicity to ce ⁇ ain cells.
  • AIM-I may also be employed to treat diseases which require growth promotion activity, for example, restenosis, since AIM-I has proliferation effects on cells of endothelial origin.
  • AIM-I may. therefore, also be employed to regulate hematopoiesis in endothelial cell development.
  • the polynucleotide encoding the AlM-1 may be employed as a diagnostic marker for determining expression of the polypeptide of the present invention since the gene is found in many tumor cell lines including pancreatic tumor, testes mmor, endometrial rumor and T-cell lymphoma.
  • This invention also provides a method for identification of molecules, such as receptor molecules, that bind AIM-I.
  • Genes encoding proteins that bind AIM-I, such as receptor proteins can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting. Such methods are described in many laboratory manuals such as, for instance, Coligan et al, Current Protocols in Immunology 1(2): Chapter 5 (1991).
  • expression cloning may be employed for this purpose.
  • polyadenylated RNA is prepared from a cell responsive to AIM-I
  • a cDNA library is created from this RNA.
  • the library is divided into pools and the pools are transfected individually into cells that are not responsive to AIM-I.
  • the transfected cells then are exposed to labeled AIM-I.
  • AIM-I can be labeled by a variety of well-known techniques including standard methods of radio-iodination or inclusion of a recognition site for a site-specific protein kinase.
  • the cells are fixed and binding of AIM-I is determined. These procedures conveniently are carried out on glass slides.
  • Pools are identified of cDNA that produced AIM-I-binding cells. Sub-pools are prepared from these positives, transfected into host cells and screened as described above. Using an iterative sub-pooling and re-screening process, one or more single clones that encode the putative binding molecule, such as a receptor molecule, can be isolated.
  • a labeled ligand can be photoaffinity linked to a cell extract, such as a membrane or a membrane extract, prepared from cells that express a molecule that it binds, such as a receptor molecule.
  • Cross-linked material is resolved by polyacrylamide gel electrophoresis ( "PAGE") and exposed to X-ray film.
  • the labeled complex containing the ligand-receptor can be excised, resolved into peptide fragments, and subjected to protein microsequencing.
  • the amino acid sequence obtained from microsequencing can be used to design unique or degenerate oligonucleotide probes to screen cDNA libraries to identify genes encoding the putative receptor molecule.
  • Polypeptides of the invention also can be used to assess AIM-I binding capacity of AIM-I binding molecules, such as receptor molecules, in cells or in cell-free preparations.
  • the invention also provides a method of screening compounds to identify those which enhance or block the action of AIM-I on cells, such as its interaction with AIM-I- binding molecules such as receptor molecules.
  • An agonist is a compound which increases the namral biological functions of AIM-I or which functions in a manner similar to AIM-I, while antagonists decrease or eliminate such functions.
  • a cellular compartment such as a membrane or a preparation thereof, such as a membrane-preparation, may be prepared from a cell that expresses a molecule that binds AIM-I.
  • AIM-I a molecule of a signaling or regulatory pathway modulated by AIM-I.
  • the preparation is incubated with labeled AIM-I in the absence or the presence of a candidate molecule which may be an AIM-I agonist or antagonist.
  • the ability of the candidate molecule to bind the binding molecule is reflected in decreased binding of the labeled ligand.
  • AlM-I-like effects of potential agonists and antagonists may by measured, for instance, by determining activ ity of a second messenger system following interaction of the candidate molecule with a cell or appropriate cell preparation, and comparing the effect with that of AIM-I or molecules that elicit the same effects as AIM-I.
  • Second messenger systems that may be useful in this regard include but are not limited to AMP guanylate cyclase. ion channel or phosphoinositide hydrolysis second messenger systems.
  • Another example of an assay for AIM-I antagonists is a competitive assay that combines AIM-I and a potential antagonist with membrane-bound AIM-I receptor molecules or recombinant AIM-I receptor molecules under appropriate conditions for a competitive inhibition assay.
  • AIM-I can be labeled, such as by radioactivity, such that the number of AIM-I molecules bound to a receptor molecule can be determined accurately to assess the effectiveness of the potential antagonist.
  • Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to a polypeptide of the invention, and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, a peptide, a polypeptide such as a closely related protein or antibody that binds the same sites on a binding molecule, such as a receptor molecule, without inducing AIM-I-induced activities, thereby preventing the action of AIM-I by excluding AIM-I from binding.
  • Antisense molecules can be used to control gene expression through antisense DNA or RNA or through triple-helix formation. Antisense techniques are discussed, for example, in Okano, J. Neurochem, 56:560. 1991 : Oligodeoxvnucleotides As Antisense Inhibitors Of Gene Expression, CRC Press. Boca Raton. FL ( 1988). Triple helix formation is discussed in, for instance Lee et al. Nucleic Acids Research, 3:173, 1979; Cooney et al , Science, 241:456 1988: and Dervan et al . Science, 257: 1360 1991. The methods are based on binding of a polynucleotide to a complementary DNA or RNA.
  • the 5' coding portion of a pol ucleotide that encodes the mamre polypeptide of the present invention may be used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length ⁇ DS ⁇ oligonucleotide is designed to be complementary to a region of the gene inv lved in transcription thereby preventing transcription and the production of AIM-l
  • the antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into AIM-I polypeptide.
  • the oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of AIM-I.
  • the antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g. , as hereinafter described.
  • the antagonists may be employed for instance to treat cachexia which is a lipid clearing defect resulting from a systemic deficiency of lipoprotein lipase. which is suppressed by AIM-I.
  • the AIM-I antagonists may also be employed to treat cerebral malaria in which AIM-I appears to play a pathogenic role.
  • the antagonists may also be employed to treat rheumatoid arthritis by inhibiting AIM-I induced production of inflammatory cytokines, such as IL1 in the synovial cells. When treating arthritis, AIM-I is preferably injected intra-articularly.
  • the AIM-I antagonists may also be employed to prevent graft-host rejection by preventing the stimulation of the immune system in the presence of a graft.
  • the AIM-I antagonists may also be employed to inhibit bone resorption and, therefore, to treat and/or prevent osteoporosis.
  • the antagonists may also be employed as anti-inflammatory agents, and to treat endotoxic shock. This critical condition results from an exaggerated response to bacterial and other types of infection.
  • compositions comprising the polynucleotide or the polypeptides discussed above or the agonists or antagonists.
  • the polypeptides of the present invention may be employed in combination with a non-sterile or sterile carrier or carriers for use with cells, tissues or organisms, such as a pharmaceutical carrier suitable for administration to a subject.
  • a pharmaceutical carrier suitable for administration to a subject such as a pharmaceutical carrier suitable for administration to a subject.
  • Such compositions comprise, for instance, a media additive or a therapeutically effective amount of a polypeptide of the ;.' ention and a pharmaceutically acceptable carrier or excipient.
  • Such carriers may include, but are not limited to. saline, buffered saline, dextrose, water, glycerol, ethanol and combinations thereof. The formulation should suit the mode of administration.
  • the invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention.
  • Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacmre, use or sale of pharmaceuticals or biological products, reflecting approval by the agency of the manufacmre, use or sale of the product for human administration.
  • Polypeptides and other compounds of the present invention may be employed alone or in conjunction with other compounds, such as therapeutic compounds.
  • compositions may be administered in any effective, convenient manner including, for instance, administration by topical, oral, anal, vaginal, intravenous, intraperitoneal. intramuscular, subcutaneous, intranasal or intradermal routes among others.
  • compositions generally are administered in an amount effective for treatment or prophylaxis of a specific indication or indications.
  • the compositions are administered in an amount of at least about 10 ⁇ g/kg body weight. In most cases they will be administered in an amount not in excess of about 8 mg/kg body weight per day. Preferably, in most cases, dose is from about 10 ⁇ g/kg to about 1 mg/kg body weight, daily. It will be appreciated that optimum dosage will be determined by standard methods for each treatment modality and indication, taking into account the indication, us severity . route of administration, complicating conditions and the like.
  • polypeptides, agonists and antagonists that are polypeptides may be employed in accordance with the present invention by expression of such polypeptides in vivo, in treatment modalities often referred to as "gene therapy.
  • a polynucleotide such as a DNA or RNA
  • the engineered cells then can be provided to a patient to be treated with the polypeptide.
  • cells may be engineered ex vivo by the use of a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention.
  • cells may be engineered in vivo for expression of a polypeptide in vivo by procedures known in the an.
  • a polynucleotide of the invention may be engineered for expression in a replication defective retroviral vector, as discussed above.
  • the retroviral expression construct then may be isolated and introduced into a packaging cell is transduced with a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention such that the packaging cell now produces infectious viral panicles containing the gene of interest.
  • These producer cells may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo.
  • Retrovimses from which the retroviral plasmid vectors herein above mentioned may be derived include, but are not limited to. Moloney Murine Leukemia Vims, spleen necrosis vims, retrov iruses such as Rous Sarcoma Vims, Harvey Sarcoma Vims, avian leukosis vims, gibbon ape leukemia v ims, human immunodeficiency vims, adenovims. Myeloproliferative Sarcoma Vims, and mammary tumor vims.
  • the retroviral plasmid vector is derived from Moloney Murine Leukemia Vims.
  • Such vectors well include one or more promoters for expressing the polypeptide.
  • Suitable promoters which may be employed include, but are not limited to, the retroviral LTR: the SV40 promoter: and the human cytomegalovirus (CMV) promoter described in Miller et al. Biotechntquei. 7 980-990 (1989), or any other promoter (e.g. , cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, RNA polymerase III, and ⁇ -actin promoters).
  • CMV human cytomegalovirus
  • viral promoters which may be employed include, but are not limited to, adenovims promoters, thymidine kinase (TK) promoters, and B19 parvovirus promoters. The selection of a suitable promoter will be apparent to those skilled in the an from the teachings contained herein.
  • Suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovims (CMV) promoter; the respiratory syncytial vims (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter: human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs herein above described); the ⁇ -actin promoter; and human growth hormone promoters.
  • the promoter also may be the native promoter which controls the gene encoding the
  • the retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines.
  • packaging cells which may be transfected include, but are not limited to. the PE501 . PA317, Y-2, Y-AM, PA12. T19-14X, VT- 19-17-H2. YCRE. YCRIP. GP + E-86. GP+envAml2. and DAN cell lines as described in Miller, A., Human Gene Therapv. / :5- 14 ( 1990).
  • the vector may be transduced into the packaging cells through any means known in the an. Such means include, but are not limited to. electroporation. the use of liposomes, and CaPO 4 precipitation.
  • the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid. and then administered to a host.
  • the producer cell line w ill generate infectious retroviral vector panicles, which include the nucleic acid sequence(s) encoding the polypeptides.
  • retroviral vector particles then may be employed to transduce eukaryotic cells, either in vitro or in vivo.
  • the transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide.
  • Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.
  • ligations were accomplished using standard buffers, incubation temperatures and times, approximately equimolar amounts of the DNA fragments to be ligated and approximately 10 units of T4 DNA ligase ("ligase") per 0.5 ⁇ g of DNA.
  • ligase T4 DNA ligase
  • the DNA sequence encoding human AIM-I in the deposited polynucleotide was amplified using PCR oligonucleotide primers specific to the amino acid carboxyl terminal sequence of the human AIM-I protein and to vector sequences 3' to the gene. Additional nucleotides containing restriction sites to facilitate cloning were added to the 5' and 3' sequences respectively.
  • the 5' oligonucleotide primer had the sequence 5' GCG GCG GGA TCC ATG GCT ATG ATG GAG GTC CAG 3' containing the underlined BamHI restriction site, which encodes a start AUG. followed by 18 nucleotides of the human AIM-I coding sequence set out in Figure 1.
  • the 3' primer had the sequence 5 CGC GCG TCT AGA GCT TAG GCA ACT AAA AAG GCC 3 " containing the underlined Xbal restriction site followed by 21 nucleotides complementary to the last 21 nucleotides of the AIM-I coding sequence set out in Figure 1. including the stop codon.
  • the restrictions sites were convenient to restriction enzyme sites in the bacterial expression vectors pQi ⁇ which were used tor bacterial expression in these examples.
  • C ⁇ i pQE9 encodes ampicillin antibiotic resistance ("Amp r ”) and contains a bacterial origin of replication ("ori"), an IPTG inducible promoter, a ribosome binding site ⁇ " RBS” ⁇ . a 6-His tag and restriction enzyme sites.
  • the ligation mixture was transformed into competent £. coli cells using standard procedures. Such procedures are described in Sambrook et al. Molecular Cloning: A Laboratory Manual, 2nd Ed. ; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989).
  • This strain which is only one of many that are suitable for expressing AIM-I, is available commercially from Qiagen.
  • Transformants were identified by their ability to grow on LB plates in the presence of ampicillin. Plasmid DNA was isolated from resistant colonies and the identity of the cloned DNA was confirmed by restriction analysis.
  • Clones containing the desired constmcts were grown overnight ("O/N") in liquid culture in LB media supplemented with both ampicillin (100 ⁇ g/ml) and kanamycin (25 ⁇ g/ml).
  • the O/N culture was used to inoculate a large culture, at a dilution of approximately 1 : 100 to 1 :250.
  • the cells were grown to an optical density at 600 nm ("ODioo") of between 0.4 and 0.6.
  • Isopropyl-B-D-thiogalactopyranoside (“IPTG”) was then added to a final concentration of 1 mM to induce transcription from lac repressor sensitive promoters, by inactivating the lad repressor. Cells subsequently were incubated further for 3 to 4 hours. Cells then were harvested by centrifugation and dismpted, by standard methods.
  • the cDNA sequence encoding the full length human AIM-I protein, in the deposited clone is amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the gene:
  • the 5' primer has the sequence 5' CCG CGC GGA TCC ATC ATG GCT ATG ATG GAG GTC C 3 ' containing the underlined restriction enzyme site followed by 22 bases of the sequence of AIM-I of Figure 1 .
  • An efficient signal for initiation of translation in eukaryotic cells as described by Kozak. M . J Mol. Biol , 196:941-950 (1987) is appropriately located in the vector portion of the construct.
  • the 3' primer has the sequence 5 ' CGC GCG TCT AGA GCT TAG CCA ACT AAA AAG GCC 3' containing the underlined Xbal restriction followed by nucleotides complementary to the last 21 nucleotides of the AIM-I coding sequence set out in Figure 1 , including the stop codon.
  • the amplified fragment is isolated from a 1 % agarose gel using a commercially available kit ("Geneclean, " BIO 101 Inc. , La Jolla, Ca.). The fragment then is digested with BamHI and Asp718 and again is purified on a 1 % agarose gel. This fragment is designated herein F2.
  • the vector pA2 is used to express the AIM-I protein in the baculovims expression system, using standard methods, such as those described in Summers et al, A Manual Of Methods For Baculovims Vectors And Insect Cell Culture Procedures, Texas Agricultural Experimental Station Bulletin No. 1555 (1987).
  • This expression vector contains the strong polyhedrin promoter of the Autographa califomica nuclear polyhedrosis vims (AcMNPV) followed by convenient restriction sites.
  • the signal peptide of AcMNPV gp67. including the N-terminal methionine, is located just upstream of a BamHI site.
  • the polyadenylation site of the simian vims 40 (“SV40") is used for efficient polyadenylation.
  • SV40 simian vims 40
  • the beta- galactosidase gene from £. coli is inserted in the same orientation as the polyhedrin promoter and is followed by the polyadenylation signal of the polyhedrin gene.
  • the polyhedrin sequences are flanked at both sides by viral sequences for cell-mediated homologous recombination with wild-type viral DNA to generate viable vims that express the cloned polynucleotide.
  • baculovims vectors could be used in place of pA2, such as pAc373, pVL941 and pAcIM l provided, as those of skill readily will appreciate, that constmction provides appropriately K»cated signals for transcription, translation, trafficking and the like, such as an in-frame Al G and a signal peptide, as required.
  • K K»cated signals for transcription, translation, trafficking and the like, such as an in-frame Al G and a signal peptide, as required.
  • Such vectors are described in Lucko et al . Vint ms . 170: 1 -39, among others.
  • the plasmid is digested w ith the restriction enzymes BamHI and Xbal and then is dephosphorylated using calf intestinal phosphatase, using routine procedures known in the art.
  • the DNA is then isolated from a 1 % agarose gel using a commercially available kit ("Geneclean" BIO 101 Inc., La Jolla, Ca.). This vector DNA is designated herein "V2" .
  • Fragment F2 and the dephosphorylated plasmid V2 are ligated together with T4 DNA ligase.
  • £. coli HB101 cells are transformed with ligation mix and spread on culture plates.
  • Bacteria are identified that contain the plasmid with the human AIM-I gene by digesting DNA from individual colonies using BamHI and Xbal and then analyzing the digestion product by gel electrophoresis. The sequence of the cloned fragment is confirmed by DNA sequencing. This plasmid is designated herein pBac AIM-I.
  • 5 ⁇ g of the plasmid pBacAIM-I is co-transfected with 1.0 ⁇ g of a commercially available linearized baculovims DNA ("BaculoGoldTM baculovims DNA”. Pharmingen, San Diego, CA.), using the lipofection method described by Feigner et al , Proc. Natl. Acad. Sci. USA, 54:7413-7417 ( 1987).
  • BaculoGoldTM vims DNA and 5 ⁇ g of the plasmid pBacAIM-I are mixed in a sterile well of a microtiter plate containing 50 ⁇ l of semm free Grace's medium (Life Technologies Inc. , Gaithersburg, MD).
  • plaque assay After four days the supernatant is collected and a plaque assay is performed, as described by Summers and Smith, cited above.
  • An agarose gel with "Blue Gal” (Life Technologies Inc. , Gaithersburg) is used to allow easy identification and isolation of gal-expressing clones, which p ⁇ xluce blue-stained plaques. (A detailed description of a "plaque assay” of this type can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10).
  • V- AIM-I A clone containing properly inserted AIM-I is identified by DNA analysis including restriction mapping and sequencing. This is designated herein as V- AIM-I.
  • Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS.
  • the cells are infected with the recombinant baculovims V- AIM-I at a multiplicity of infection ("MOD of about 2 (about 1 to about 3).
  • MOD multiplicity of infection
  • the medium is removed and is replaced with SF900 II medium minus methionine and cysteine (available from Life Technologies Inc. , Gaithersburg). 42 hours later, 5 ⁇ Ci of 35 S- methionine and 5 ⁇ Ci "S cysteine (available from Amersham) are added.
  • the cells are further incubated for 16 hours and then they are harvested by centrifugation, lysed and the labeled proteins are visualized by SDS-PAGE and autoradiography .
  • the expression plasmid. ⁇ IM-I HA. is made by cloning a cDNA encoding AIM-I into the expression vector pcDN Al/Amp (which can be obtained from Invitrogen, Inc.).
  • the expression vector pcDNAI/amp contains: (1) an E. coli origin of replication effective for propagation in £.
  • coli and other prokaryotic cell (2) an ampicillin resistance gene for selection of plasmid-containing prokaryotic cells; (3) an SV40 origin of replication for propagation in eukaryotic cells; (4) a CMV promoter, a polylinker, an SV40 intron, and a polyadenylation signal arranged so that a cDNA conveniently can be placed under expression control of the CMV promoter and operably linked to the SV40 intron and the polyadenylation signal by means of restriction sites in the polylinker.
  • a DNA fragment encoding the entire AIM-I precursor and a HA tag fused in frame to its 3' end is cloned into the polylinker region of the vector so that recombinant protein expression is directed by the CMV promoter.
  • the HA tag corresponds to an epitope derived from the influenza hemagglutinin protein described by Wilson et al, Cell 37: 767 (1984). The fusion of the HA tag to the target protein allows easy detection of the recombinant protein with an antibody that recognizes the HA epitope.
  • the plasmid construction strategy is as follows.
  • the AIM-I cDNA of the deposited clone is amplified using primers that contained convenient restriction sites, much as described above regarding the constmction of expression vectors for expression of AIM-I in £. coli and 5. fur ⁇ erda.
  • one of the primers contains a hemagglutinin tag ("HA tag") as described above.
  • Suitable primers include that following, which are used in this example.
  • the 5' primer containing the underlined restriction enzyme site, an AUG start codon and 22 codons thereafter, forming the hexapeptide haemagglutinin tag, has the sequence: 5' CCG CGC GGA ICC ATC ATG GCT ATG ATG GAG GTC C 3'
  • the 3' primer containing the underlined Xbal site and 21 nucleotides of 3' coding sequence (at the 3' end) has the sequence: 5' CGC GCG TCT AGA GCT TAG CCA ACT AAA AAG GCC 3'
  • the PCR amplified DNA fragment and the vector, pcDNAI/Amp, are digested with and then ligated.
  • the ligation mixmre is transformed into E. coli strain SURE (available from Stratagene Cloning Systems, 11099 North To ⁇ ey Pines Road, La Jolla, CA 92037) the transformed culture is plated on ampicillin media plates which then are incubated to allow growth of ampicillin resistant colonies. Plasmid DNA is isolated from resistant colonies and examined by restriction analysis and gel sizing for the presence of the AIM-I-encoding fragment.
  • COS cells are transfected with an expression vector, as described above, using DEAE-DEXTRAN, as described, for instance, in Sambrook et al . Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, Cold Spring Harbor, New York (1989). Cells are incubated under conditions for expression of AIM-l by the vector.
  • AIM-I HA fusion protein is detected by radiolabelling and immunoprecipitation. using methods described in, for example Harlow et al . Antibodies: A Laboratory Manual. 2nd Ed. ; Cold Spring Harbor Laboratory Press, Cold Spring Harbor. Ne York ( 1988). To this end, two days after transfection, the cells are labeled by incubation in media containing 35 S-cysteine for 8 hours. The cells and the media are collected, and the cells are washed and the lysed with detergent-containing RIPA buffer: 150 mM NaCl. I S N P-40. 0. 1 % SDS, 1 % NP-40, 0.5% DOC, 50 mM TRIS. pH 7.5, as described by Wilson et al. cited above.
  • Proteins are precipitated from the cell lysate and from the culture media using an HA-specific monoclonal antibody. The precipitated proteins then are analyzed by SDS-PAGE gels and autoradiography. .'., expression product of the expected size is seen in the cell lysate, which is not seen in negative controls.
  • Example 4 Tissue Distribution of AIM-I Expression
  • RNAzolTM B system Biotecx Laboratories, Inc. 6023 South Loop East, Houston, TX 77033.
  • RNA is isolated from tissue samples. The RNA is size resolved by electrophoresis through a 1 % agarose gel under strongly denaturing conditions. RNA is blotted from the gel onto a nylon filter, and the filter then is prepared for hybridization to a detectably labeled polynucleotide probe.
  • the antisense strand of the coding region of the cDNA inse ⁇ in the deposited clone is labeled to a high specific activity.
  • the cDNA is labeled by primer extension, using the Prime-It kit, available from Stratagene.
  • the reaction is carried out using 50 ng of the cDNA, following the standard reaction protocol as recommended by the supplier.
  • the labeled polynucleotide is purified away from other labeled reaction components by column chromatography using a Select-G-50 column, obtained from 5-Prime - 3-Prime, Inc. of 5603 Arapahoe Road. Boulder. CO 80303.
  • the labeled probe is hybridized to the filter, at a concentration of 1.000,000 cp /ml. in a small volume of 7 r ; SDS. 0.5 M NaPO 4 , pH 7.4 at 65 * C, overnight. Thereafter the probe solution is drained and the filter is washed twice at room temperamre and twice at 60 * C ith 0.5 x SSC, 0.1 % SDS. The filter then is dried and exposed to film at -70 overnight ith an intensifying screen. Autoradiography shows that mRNA for AIM-I is abundant in human hea ⁇ . bone marrow, CD4 + and CD19 * peripheral blood lymphocytes, and less so in lung and kidney tissue.
  • Example 5 Gene Therapeutic Expression of Human AIM-I
  • Fibroblasts are obtained from a subject by skin biopsy.
  • the resulting tissue is placed in tissue-culmre medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culmre flask, approximately ten pieces are placed in each flask.
  • the flask is turned upside down, closed tight and left at room temperamre overnight. After 24 hours at room temperamre, the flask is inve ⁇ ed - the chunks of tissue remain fixed to the bottom of the flask - and fresh media is added (e.g. , Ham's F12 media, with 10% FBS, penicillin and streptomycin).
  • the tissue is then incubated at 37 'C for approximately one week. At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culmre. a monolayer of fibroblasts emerges. The monolayer is trypsinized and scaled into larger flasks.
  • a vector for gene therap is digested with restriction enzymes for cloning a fragment to be expressed.
  • the digested vector is treated with calf intestinal phosphatase to prevent self-ligation.
  • the dephosphorylated, linear vector is fractionated on an agarose gel and purified.
  • AIM-I cDNA capable of expressing active AIM-I is isolated.
  • the ends of the fragment are modified, if necessary , for cloning into the vector. For instance, 5" overhanging may be treated w ith DNA polymerase to create blunt ends. 3 ' overhanging ends may be removed using S I nuclease. Linkers may be ligated to blunt ends with T4 DNA ligase
  • Equal quantities of the Moloney murine leukemia vims linear backbone and the AIM-I fragment are mixed together and joined using T4 DNA ligase.
  • the ligation mixmre is used to transform £. coli and the bacteria are then plated onto agar-containing kanamycin. Kanamycin phenotype and restriction analysis confirm that the vector has the properly inse ⁇ ed gene.
  • Packaging cells are grown in tissue culmre to confluent density in Dulbecco's Modified Eagle's Medium (DMEM) with 10% calf semm (CS), penicillin and streptomycin.
  • DMEM Dulbecco's Modified Eagle's Medium
  • CS calf semm
  • penicillin and streptomycin The vector containing the AIM-I gene is introduced into the packaging cells by standard techniques. Infectious viral panicles containing the AIM-I gene are collected from the packaging cells, which now are called producer cells.
  • Fresh media is added to the producer cells, and after an appropriate incubation period media is harvested from the plates of confluent producer cells.
  • the media containing the infectious viral particles, is filtered through a Millipore filter to remove detached producer cells.
  • the filtered media then is used to infect fibroblast cells.
  • Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the filtered media.
  • Polybrene Aldrich
  • the media is removed and replaced with fresh media. If the titer of vims is high, then virtually all fibroblasts will be infected and no selection is required. If the titer is low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his, to select out transduced cells for expansion.
  • Engineered fibroblasts then may be injected into rats, either alone or after having been grown to confluence on m ⁇ cn carr ⁇ er beads, such as cytodex 3 beads.
  • the injected fibroblasts produce AIM-I product, and the biological actions of the protein are conveyed to the host
  • ADDRESSEE Carella, Byrne, Bain, Gilfillan, Cecchi,
  • MOLECULE TYPE DNA (genomic)
  • CAAAACAAAC AAACAGAAAA CAGAAAACAA AAAAACCTCT ATGCAATCTG AGTAGAGCAG 1044
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)

Abstract

The invention relates to AIM-I polypeptides, polynucleotides encoding the polypeptides, methods for producing the polypeptides, in particular by expressing the polynucleotides, and agonists and antagonists of the polypeptides. The invention further relates to methods for utilizing such polynucleotides, polypeptides, agonists and antagonists for applications, which relate, in part, to research, diagnostic and clinical arts.

Description

Apoptosis Inducing Molecule I
This invention relates, in part, to newly identified polynucleotides and polypeptides; variants and derivatives of the polynucleotides and polypeptides; processes for making the polynucleotides and the polypeptides, and their variants and derivatives; agonists and antagonists of the polypeptides; and uses of the polynucleotides, polypeptides, variants, derivatives, agonists and antagonists. In particular, in these and in other regards, the invention relates to polynucleotides and polypeptides of human Apoptosis Inducing Molecule I (AIM-I).
BACKGROUND OF THE INVENTION
Human tumor necrosis factors (TNF-α) and β (TNF-0 or lymphotoxin) are related members of a broad class of polypeptide mediators, which includes the interferons. interieukins and growth factors, collectively called cytokines (Beutler, B. and Cerami, A.. Annu. Rev. Immunol. 7:625-655, 1989).
Tumor necrosis factor (TNF-α and TNF-3) was originally discovered as a result of its anti-tumor activity, however, now it is recognized as a pleiotropic cytokine capable of numerous biological activities including apoptosis of some transformed cell lines, mediation of cell activation and proliferation and also as playing important roles in immune retaliation and inflammation. To date, mere are nine known members of the TNF-ligand superfamily, TNF-α, TNF-/3 (lymphotoxin-c.), LT- 3. OX40L, Fas ligand, CD30L, CD27L, CD40L and 4- 1BBL. The ligands of the TNF ligand superfamily are acidic, TNF-like molecules with approximately 20% sequence homology in the extracellular domains (range, 12%-36%) and exist mainly as membrane-bound forms with the biologically active form being a trimeric/multimeric complex. Soluble forms of the TNF ligand superfamily have only been identified so far for TNF. LTα, and Fas ligand (for a general review, see Gruss, H. and Dower, S.K. , Blood, 85 (12): 3378-3404, 1995), which is hereby incorporated by reference in its entirety.
These proteins are involved in regulation of cell proliferation, activation, and differentiation, including control of cell survival or death by apoptosis or cytotoxicity (Armitage, R.J. , Curr. Opin. Immunol. 6:407, 1994 and Smith, C.A. , Cell, 75:959, 1994).
Mammalian development is dependent on both the proliferation and differentiation of cells as well as programmed cell death which occurs through apoptosis (Walker et al, Methods Achiev. Exp. Pathol 13: 18. 1988). Apoptosis plays a critical role in the destruction of immune thymocytes that recognize self antigens. Failure of this normal elimination process may play a role in autoimmune diseases (Gammon et al. Immunology Today. 12: 192. 1991 )
Itoh et al . Cell. (V>:233. I W| . described a cell surface antigen, Fas/CD23 that mediates apoptosis and is involved in cional deletion of T-cells. Fas is expressed in activated T-cells. B-cells. neutrophils and in thymus, liver, heart and lung and ovary in adult mice (Watanahe-Fukunaga et al . J. Immunol, 148: 1274, 1992) in addition to activated T-cells, B-cells. neutorυphils. In experiments where a monoclonal antibody is cross-linked to Fas. apoptosis is induced (Yonehara et al, J. Exp. Med, 169: 1141, 1989: Trauth et al. Science. 2« 5: 01 . 1989). In addition, there is an example where binding of a monoclonal antibody to Fas ligand is stimulatory to T-cells under certain conditions (Alderson et al, J. Exp. Med 178:2231, 1993).
Fas antigen is a cell surface protein of relative MW of 45 Kd. Both human and murine genes for Fas have been cloned by Watanabe-Fukunaga et al , (J. Immunolo. 148: 1274, 1992) and Itoh et al (Cell, 66:233, 1991). The proteins encoded by these genes are both transmembrane proteins with structural homology to the nerve growth factor/tumor necrosis factor receptor superfamily, which includes two TNF receptors, the low affinity nerve growth factor receptor and CD40, CD27, CD30, and OX40.
Recently the Fas ligand has been described (Suda et al, Cell, 75: 1169, 1993). The amino acid sequence indicates that Fas ligand is a type 11 transmembrane protein belonging to the TNF family. Fas ligand is expressed in splenocytes and thymocytes, consistent with T-cell mediated cytotoxicity. The purified Fas ligand has a MW of 40 Kd.
Recently it has been demonstrated that Fas/Fas ligand interactions are required for apoptosis following the activation of T-cells (Ju et al Nature, 373:444, 1995; Brunner et al . Nature, 57J.441 , 1995). Activation of T-cells induces expression of both proteins on the cell surface. Subsequent interaction between the ligand and receptor results in apoptosis of the cells. This supports the possible regulatory role for apoptosis induced by Fas/ Fas ligand interaciion during normal immune responses.
Clearly, there is a need tor factors that regulate activation, and differentiation of normal and abnormal cells There is a need, therefore, for identification and characterization of such factors that modulate activation and differentiation of cells, both normally and in disease states. In particular, there is a need to isolate and characterize additional molecules that mediaie apoptosis. and treat autoimmune disease, graft versus host disease and lymphadenopathy. and. among other things, can play a role in preventing, ameliorating or correcting dysfunctions or diseases. SUMMARY OF THE INVENTION
The polypeptide of the present invention has been identified as a novel member of the TNF ligand super- family based on structural and biological similarities.
Toward these ends, and others, it is an object of the present invention to provide polypeptides, inter alia, that have been identified as novel Apoptosis Inducing Molecule I (AIM-I) polypeptides by homology between the amino acid sequence set out in Figure 1 and known amino acid sequences of other proteins such as human Fas ligand (Suda et al, Cell 75: 1169, 1993).
It is a further object of the invention, moreover, to provide polynucleotides that encode AIM-I, paπicularly polynucleotides that encode the polypeptide herein designated AIM-I. In a particularly preferred embodiment of this aspect of the invention the polynucleotide comprises the region encoding human AIM-I in the sequence set out in Figure 1. In accordance with this aspect of the present invention there is provided an isolated nucleic acid molecule encoding a mature polypeptide expressed by the human cDNA contained in ATCC Deposit No. 97448.
In accordance w ith this aspect of the invention there are provided isolated nucleic acid molecules encoding human AIM-I. including mRNAs, cDNAs, genomic DNAs and, in further embodiments of this aspect of the invention, biologically, diagnostically, clinically or therapeuticallv useful variants, analogs, derivatives and/or fragments thereof, including fragments ot the \ arιants. analogs and derivatives. Among the particularly preferred embodiments of this aspect of the invention are naturally occurring allelic variants of human AIM-I r
It also is an object of the invention to provide AIM-I polypeptides, particularly human AIM-I polypeptides. w hich may be employed to treat lymphadenopathy, autoimmune disease, graft versus host disease; which may be used to stimulate peripheral tolerance, destroy pathologic transformed cell lines, mediate cell activation and proliferation; and are functionally linked as primary mediators of immune regulation and inflammatory response.
This aspect of the invention provides novel polypeptides of human origin referred to herein as AIM-I as well as biologically, diagnostically or therapeutically useful fragments, variants and derivatives thereof, variants and derivatives of the fragments, and analogs of all of the foregoing. Among the particularly preferred embodiments of this aspect of the invention are variants of human AIM-I encoded by naturally occurring alleles of the human AIM-I gene.
It is another object of the invention to provide a process for producing the aforementioned polypeptides. polypeptide fragments, variants and derivatives, fragments of the variants and derivatives, and analogs of the foregoing. In a preferred embodiment of this aspect of the invention there are provided methods for producing the aforementioned AIM-I polypeptides comprising culturing host cells having expressibly incorporated therein an exogenously-derived human AIM-I-encoding polynucleotide under conditions for expression of human AIM-I in the host and then recovering the expressed polypeptide.
In accordance w uh another object the invention there are provided products, compositions, processes and methods that utilize the aforementioned polypeptides and polynucleotides for research, biological, clinical and therapeutic purposes, inter alia.
In accordance w nh certain preferred embodiments of this aspect of the invention, there are provided products, compositions and methods, inter alia, for, among other things: assessing AIM-I expression in cells by determining AIM-I polypeptides or AIM-I-encoding mRN A; treaiinc disease or disorder caused by under-expression of the AIM-I in vitro, ex vivo or in vivo by exposing cells to AIM-I polypeptides or polynucleotides as disclosed herein; assaying genetic variation and aberrations, such as defects, in AIM-I genes; and administering a AIM-I polypeptide or polynucleotide to an organism to augment AIM-I function or remediate AIM-I dysfunction.
In accordance with ceπain preferred embodiments of these and other aspects of the invention there are provided probes that hybridize to human AIM-I sequences.
Ceπain additional preferred aspects related to the above aspects of the invention provides antibodies against AIM-I polypeptides. In paπicularly preferred embodiments in this regard, the antibodies are highly selective for human AIM-I, and may be employed, inter alia, to treat autoimmune diseases.
In accordance with another aspect of the present invention, there are provided AIM-I agonists. Among preferred agonists are molecules that mimic AIM-I, that bind to AIM-I-binding molecules or receptor molecules, and that elicit or augment AIM-I- induced responses. Also among preferred agonists are molecules that interact with AIM-I or AIM-I polypeptides. or with other modulators of AIM-I activities, and thereby potentiate or augment an effect of AIM-I or more than one effect of AIM-I.
In accordance with yet another aspect of the present invention, there are provided AIM-I antagonists. Among preferred antagonists are those which mimic AIM-I so as to bind to AIM-I receptor or binding molecules but not elicit an AIM-I-induced response or more than one AIM-I-induced response. Also among preferred antagonists are molecules that bind to or interact with AIM-I so as to inhibit an effect of AIM-I or more than one effect of AIM-I or which prevent expression of AIM-I. The antagonists may be employed to prevent septic shock, inflammation, cerebral malaria, activation of the HIV virus, graft-host rejection, bone resorption, rheumatoid arthritis and cachexia.
In a further aspect of the invention there are provided compositions comprising an AIM-I polynucleotide or an AIM-I polypeptide for administration to cells in vitro. to cells ex vivo and to cells in vivo, or to a multicellular organism. In certain paπicularly preferred embodiments of this aspect of the invention, the compositions comprise an AIM-I polynucleotide for expression of an AIM-I polypeptide in a host organism for treatment of disease. Paπicularly preferred in this regard is expression in a human patient for treatment of a dysfunction associated with aberrant endogenous activity of AIM-I.
Other objects, features, advantages and aspects of the present invention will become apparent to those of skill from the following description. It should be understood, however, that the following description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only. Various changes and modifications within the spirit and scope of the disclosed invention will become readily apparent to those skilled in the an from reading the following description and from reading the other pans of the present disclosure.
BRIEF DESCRIPTION OF THE DRAWINGS
The following drawings depict ceπain embodiments of the invention. They are illustrative only and do not limit the invention otherwise disclosed herein.
Figure 1 shows the nucleotide and deduced amino acid sequence of human AIM-I.
Figure 2 shows the regioas of similarity and identity between amino acid sequence of AIM-I of the present invention and human Fas ligand polypeptide (SEQ ID NO:3).
Figure 3 shows regions of similarity and identity between amino acid sequences of AIM-I of the present invention and human Fas ligand polypeptide, TNFα and TNF/3. Figure 4 shows structural and functional features of AIM-I deduced by the indicated techniques, as a function of amino acid sequence.
DETAILED DESCRIPTION OF THE INVENTION
The following illustrative elucidations are provided to facilitate understanding of certain terms used frequently herein, paπicularly in the examples. The elucidations are provided as a convenience and are not limitative of the invention.
The term "digestion of DNA" refers to catalytic cleavage of DNA with a restriction enzyme that acts only upon ceπain sequences in the DNA. The various restriction enzymes referred to herein are all commercially available and their reaction conditions, cofactors and other requirements for use are known and routine to the skilled aπisan.
For analytical purposes, typically, 1 μg of plasmid or DNA fragment is digested with about 2 units of enzyme in about 20 μl of reaction buffer. For the purpose of isolating DNA fragments for plasmid construction, typically 5 to 50 μg of DNA are digested with 20 to 250 units of enzyme in propoπionately larger volumes.
Appropriate buffers and substrate amounts for paπicular restriction enzymes are described in standard laboratory manuals, such as those referenced below, and they are specified by commercial suppliers
Incubation times ol about 1 hour at 37 "C are ordinarily used, but conditions may vary in accordance with standard procedures, the supplier's instructions and the paπiculars of the reaction. After digestion, reactions may be analyzed, and fragments may be purified by electrophoresis through an agarose or polyacrylamide gel, using well known methods that are routine for those skilled in the an. The term "genetic element" generally means a polynucleotide comprising a region that encodes a polypeptide or a region that regulates transcription or translation or other processes impoπant to expression of the polypeptide in a host cell, or a polynucleotide comprising both a region that encodes a polypeptide and a region operably linked thereto that regulates expression.
Genetic elements may be comprised within a vector that replicates as an episomal element; that is, as a molecule physically independent of the host cell genome. They may be comprised within mini-chromosomes, such as those that arise during amplification of transfected DNA by methotrexate selection in eukaryotic cells. Genetic elements also may be comprised within a host cell genome; not in their natural state but, rather, following manipulation such as isolation, cloning and introduction into a host cell in the form of purified DNA or in a vector, among others.
The term "isolated" means altered "by the hand of man" from its natural state; i.e. , that, if it occurs in nature, it has been changed or removed from its original environment, or both.
For example, a naturally occurring polynucleotide or a polypeptide naturally present in a living animal in its natural state 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. For example, with respect to polynucleotides. the term isolated means that it is separated from the chromosome and cell in which it naturally occurs
As pan of or following isolation, such polynucleotides can be joined to other polynucleotides, such as DNAs. for mutagenesis, to form fusion proteins, and for propagation or expression in a host, for instance. The isolated polynucleotides, alone or joined to other polynucleotides such as vectors, can be introduced into host cells, in culture or in whole organisms. Introduced into host cells in culture or in whole organisms, such DNAs still would be isolated, as the term is used herein, because they would not be in their naturally occurring form or environment. Similarly, the polynucleotides and polypeptides may occur in a composition, such as a media formulations, solutions for introduction of polynucleotides or polypeptides, for example, into cells, compositions or solutions for chemical or enzymatic reactions, for instance, which are not naturally occurring compositions, and, therein remain isolated polynucleotides or polypeptides within the meaning of that term as it is employed herein.
The term "ligation" refers to the process of forming phosphodiester bonds between two or more polynucleotides, which most often are double stranded DNAs. Techniques for ligation are well known to the an and protocols for ligation are described in standard laboratory manuals and references, such as, for instance, Sambrook et al. Molecular Cloning. A Laboratory Manual. 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989) and Maniatis et al, pg. 146, as cited below.
The tern "oligonucleotide(s)" refers to relatively short polynucleotides. Often the term refers to single-stranded deoxyribonucleotides, but it can refer as well to single- or double-stranded ribonucleotides, RNA:DNA hybrids and double-stranded DNAs, among others.
Oligonucleotides. such as single-stranded DNA probe oligonucleotides. often are synthesized by chemical metruxls. such as those implemented on automated oligonucleotide synthesizers. However, oligonucleotides can be made by a variety of other methods, including in vitro recombinant DNA-mediated techniques and by expression of DNAs in cells and organisms.
Initially, chemically sy nthesized DNAs typically are obtained without a 5' phosphate. The 5' ends of such oligonucleotides are not substrates for phosphodiester bond formation by ligation reactions that employ DNA ligases typically used to form recombinant DNA molecules. Where ligation of such oligonucleotides is desired, a phosphate can be added by standard techniques, such as those that employ a kinase and ATP.
The 3' end of a chemically synthesized oligonucleotide generally has a free hydroxyl group and, in the presence of a ligase, such as T4 DNA ligase, readily will form a phosphodiester bond with a 5' phosphate of another polynucleotide, such as another oligonucleotide. As is well known, this reaction can be prevented selectively, where desired, by removing the 5' phosphates of the other polynucleotide(s) prior to ligation.
The term "plasmids" generally are designated herein by a lower case p preceded and/or followed by capital letters and/or numbers, in accordance with standard naming conventions that are familiar to those of skill in the art.
Starting plasmids disclosed herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids by routine application of well known, published procedures. Many plasmids and other cloning and expression vectors that can be used in accordance with the present invention are well known and readily available to those of skill in the art. Moreover, those of skill readily may construct any number of other plasmids suitable for use in the invention. The properties, construction and use of such plasmids, as well as other vectors, in the present invention w ill be readily apparent to those of skill from the present disclosure.
The term "pols nucleoiidcisr generally refers to any polyribonucleotide or polydeoxribonucleotide. which mj> he unmodified RNA or DNA or modified RNA or DNA. Thus, for instance, polynucleotides as used herein refers to, among others, single-and double-stranded DNA. DNA that is a mixture of single-and double-stranded regions, single- and double-stranded RNA. and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double- stranded regions.
In addition, polynucleotide as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules. One of the molecules of a triple-helical region often is an oligonucleotide.
As used herein, the term polynucleotide includes DNAs or RNAs as described above that contain one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are "polynucleotides" as that term is intended herein. Moreover. DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein.
It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the an. The term polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolicallv modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells, inter alia.
The term "polypeptides" as used herein, includes all polypeptides as described below. The basic structure of polypeptides is well known and has been described in innumerable textbooks and other publications in the art. In this context, the term is used herein to refer to any peptide or protein comprising two or more amino acids joined to each other in a linear chain by peptide bonds. As used herein, the term refers to both shoπ chains, which also commonly are referred to in the an as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the an as proteins, of which there are many types.
It will be appreciated that polypeptides often contain amino acids other than the 20 amino acids commonly referred to as the 20 naturally occurring amino acids, and that many amino acids, including the terminal amino acids, may be modified in a given polypeptide, either by natural processes, such as processing and other post-translational modifications, but also by chemical modification techniques which are well known to the an. Even the common modifications that occur naturally in polypeptides are too numerous to list exhaustively here, but they are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature, and they are well known to those of skill in the an. Among the known modifications which may be present in polypeptides of the present are, to name an illustrative few, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation. oxidation, proteolytic processing, phosphorylation. prenylation. racemization. selenoylation. sulfation. transfer-RNA mediated addition of amino acids to proteins such as arginylation. and ubiquitination.
Such modifications are well known to those of skill and have been described in great detail in the scientific literature. Several particularly common modifications, glycosylation, lipid attachment, sulfation. gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation. for instance, are described in most basic texts, such as. for instance Proteins-Structure And Molecular Properties, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993). Many detailed reviews are available on this subject, such as. for example, those provided by Wold, F. , Posttranslational Protein Modifications: Perspectives and Prospects, pgs. 1-12 in Posttranslational Covalent Modification Of Proteins, B. C. Johnson, Ed., Academic Press, New York (1983); Seifter et al, Analysis for protein modifications and nonprotein cofactors, Meth. EnzymoL, 182:626-646 (1990) and Rattan et al . Protein Synthesis: Posttranslational Modifications and Aging, Ann. N. Y. Acad. Sci., 663:48-62 (1992).
It will be appreciated, as is well known and as noted above, that polypeptides are not always entirely linear. For instance, polypeptides may be branched as a result of ubiquitination, and they may be circular, with or without branching, generally as a result of posttranslation events, including natural processing event and events brought about by human manipulation which do not occur naturally. Circular, branched and branched circular polypeptides may be synthesized by non-translation natural process and by entirely synthetic methods, as well.
Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. In fact, blockage of the amino or carboxyl group in a polypeptide, or both, by a covalent modification, is common in naturally occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention, as well. For instance, the amino terminal residue of polypeptides made in E. coli, prior to proteolytic processing, almost invariably w ill be N-formylmethionine.
The modifications that tκcur in a polypeptide often will be a function of how it is made. For polypeptides made b\ expressing a cloned gene in a host, for instance, the nature and extent of the modifications in large part will be determined by the host cell posttranslational modification capacity and the modification signals present in the polypeptide amino acid sequence For instance, as is well known, glycosylation often does not occur in bacterial hosts such as E. coli. Accordingly, when glycosylation is desired, a polypeptide should be expressed in a glycosylating host, generally a eukaryotic cell. Insect cell often carry out the same posttranslational glycosylations as mammalian cells and, for this reason, insect cell expression systems have been developed to express efficiently mammalian proteins having native patterns of glycosylation, inter alia. Similar considerations apply to other modifications.
It will be appreciated that the same type of modification may be present in the same or varying degree at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications.
In general, as used herein, the term polypeptide encompasses all such modifications, particularly those that are present in polypeptides synthesized by expressing a polynucleotide in a host cell.
The term "variant(s)"of polynucleotides or polypeptides, as the term is used herein, are polynucleotides or polypeptides that differ from a reference polynucleotide or polypeptide, respectively. Variants in this sense are described below and elsewhere in the present disclosure in greater detail.
A polynucleotide variant can be, for example, a polynucleotide that differs in nucleotide sequence from another, reference polynucleotide. Generally, differences are limited so that the nucleotide sequences of the reference and the variant are closely similar overall and. in many regions, identical.
As noted below , changes m the nucleotide sequence of the variant may be silent. That is, they may not alter the ammo acids encoded by the polynucleotide. Where alterations are limited to silent changes of this type a variant will encode a polypeptide with the same amino acid sequence as the reference. Also as noted below, changes in t.' ^ nucleotide sequence of the variant may alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Such nucleotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below.
A polynucleotide variant can be, for example, a polypeptide that differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference and the variant are closely similar overall and, in many region, identical. A variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions, fusions and truncations, which may be present in any combination.
The term "receptor molecule" as used herein, refers to molecules which bind or interact specifically with AIM-I polypeptides of the present invention, including not only classic receptors, which are preferred, but also other molecules that specifically bind to or interact with polypeptides of the invention (which also may be referred to as "binding molecules" and "interaction molecules. " respectively and as "AIM-I binding molecules" and "AIM-I interaction molecules. " ) Binding between polypeptides of the invention and such molecules, including receptor or binding or interaction molecules may be exclusive to polypeptides of the invention, which is very highly preferred, or it may be highly specific for polypeptides of the invention, which is highly preferred, or it may be highly specific to a group of proteins that includes polypeptides of the invention, which is preferred, or it may be specific to several groups of proteins at least one of which includes polypeptides of the invention.
Receptors also may be non-naturallv occurring, such as antibodies and antibody- derived reagents that bind specifically to polypeptides of the invention.
The present invention relates to novel AIM-I polypeptides and polynucleotides, among other things, as described in greater detail below. In particular, the invention relates to polypeptides and polv nucleotides of a novel human AIM-I, which is related by amino acid sequence homology to known human AIM-I. The invention relates especially to AIM-I having the nucleotide and amino acid sequences set out in Figure 1 , and to the AIM-I nucleotide and amino acid sequences of the cDNA in ATCC Deposit No. 97448, which is herein referred to as "the deposited clone" or as the "cDNA of the deposited clone. " It will be appreciated that the nucleotide and amino acid sequences set out in Figure 1 were obtained by sequencing the cDNA of the deposited clone. Hence, the sequence of the deposited clone is controlling as to any discrepancies between the two and any reference to the sequences of Figure 1 include reference to the sequence of the human cDNA of the deposited clone.
In accordance with one aspect of the present invention, there are provided isolated polynucleotides which encode the AIM-I polypeptide having the deduced amino acid sequence of Figure 1 or the AIM-I polypeptide encoded by the cDNA in the deposited clone.
Using the information provided herein, such as the polynucleotide sequence set out in Figure 1 , a polynucleotide of the present invention encoding human AIM-I polypeptide may be obtained using standard cloning and screening procedures, such as those for cloning cDNAs using human tissue as starting material. Illustrative of the invention, the polynucleotide set out in Figure 1 was discovered in a cDNA library derived from cells of a human pancreas rumor.
Human AIM-I of the
Figure imgf000019_0001
is structurally related to other proteins of the TNF family, as shown by the results ot sequencing the human cDNA encoding human AIM-I in the deposited clone The cl)V\ sequence thus obtained is set out in Figure 1 . It contains an open readinj. frame encodini! a protein of about 281 amino acid residues with a deduced molecular weight of about 31 kDa. The protein exhibits greatest homology to known human AIM I . among known proteins. The AIM-I of Figure 1 has about 22.892 % identity and about 48.594 % similarity with the amino acid sequence of human Fas ligand. Polynucleotides of the present invention may be in the form of RNA, such as mRNA, or in the form cDNA and genomic DNA obtained by cloning or produced by chemical synthetic techniques or by a combination thereof. The DNA may be double- stranded or single-stranded. Single-stranded DNA may be the coding strand, also known as the sense strand, or it may be the non-coding strand, also referred to as the anti-sense strand.
The coding sequence which encodes the polypeptide may be identical to the coding sequence of the polynucleotide shown in Figure 1. It also may be a polynucleotide with a different sequence, which, as a result of the redundancy (degeneracy) of the genetic code, encodes the polypeptide of the DNA of Figure 1.
Polynucleotides of the present invention which encode the polypeptide of Figure 1 may include, but are not limited to the coding sequence for the mature polypeptide, by itself; the coding sequence for the mature polypeptide and additional coding sequences, such as those encoding a leader or secretory sequence, such as a pre-, or pro- or prepro- protein sequence: the coding sequence of the mature polypeptide, with or without the aforementioned additional coding sequences, together with additional, non-coding sequences, including for example, but not limited to introns and non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription. mRNA processing - including splicing and polyadenylation signals, for example - ribosome binding and stability of mRNA; additional coding sequence which codes for additional amino acids, such as those which provide additional functionalities. Thus, for instance, the polypeptide may be fused to a marker sequence, such as a peptide. which facilitates purification of the fused polypeptide. In certain preferred embodiments of this aspect of the invention, the marker sequence is a hexa-histidine peptide, such as the tag provided in the pQE vector (Qiagen, Inc. , Chatsworth, CA), : -long others, many of which are commercially available. As described in Gentz et al , Proc. Natl. Acad. Sci.. USA, 56:821 -824 ( 1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. The HA tag corresponds to an epitope derived of influenza hemagglutinin protein, which has been described by Wilson et al , Cell 37:161 (1984), for instance.
In accordance with the foregoing, the term "polynucleotide encoding a polypeptide" as used herein encompasses polynucleotides which include a sequence encoding a polypeptide of the present invention, particularly the human AIM-I having the amino acid sequence set out in Figure 1. The term encompasses polynucleotides that include a single continuous region or discontinuous regions encoding the polypeptide (for example, interrupted by introns) together with additional regions, that also may contain coding and/or non-coding sequences.
The present invention further relates to variants of the herein above described polynucleotides which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of Figure 1. A variant of the polynucleotide may be a namrally occurring variant such as a naturally occurring allelic variant, or it may be a variant that is not known to occur naturally. Such non-narurally occurring variants of the polynucleotide may be made by mutagenesis techniques, including those applied to polynucleotides, cells or organisms.
Among variants in this regard are variants that differ from the aforementioned polynucleotides by nucleotide substimtions. deletions or additions. The substitutions, deletions or additions may involve one or more nucleotides. The variants may be altered in coding or non-coding regions or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions.
Among the particularly preferred embodiments of the invention in this regard are polynucleotides encoding polypeptides having the amino acid sequence of AIM-I set out in Figure 1 ; variants, analogs, derivatives and fragments thereof, and fragments of the variants, analogs and derivatives. Further paπicuiarly preferred in this regard are polynucleotides encoding AIM-I variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments, which have the amino acid sequence of the AIM-I polypeptide of Figure 1 in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination. Especially preferred among these are silent substitutions, additions and deletions, which do not alter the propeπies and activities of the AIM-I. Also especially preferred in this regard are conservative substitutions. Most highly preferred are polynucleotides encoding polypeptides having the amino acid sequence of Figure 1 without substimtions.
Fuπher preferred embodiments of the invention are polynucleotides that are at least 70% identical to a polynucleotide encoding the AIM-I polypeptide having the amino acid sequence set out in Figure 1 , and polynucleotides which are complementary to such polynucleotides. Alternatively, most highly preferred are polynucleotides that comprise a region that is at least 80% identical to a polynucleotide encoding the AIM-I polypeptide of the cDNA of the deposited clone and polynucleotides complementary thereto. In this regard, polynucleotides at least 90% identical to the same are particularly preferred, and among these paπicularly preferred polynucleotides, those with at least 95 % are especially preferred. Furthermore, those with at least 97% are highly preferred among those with at least 95%. and among these those with at least 98% and at least 99ci are particularly highly preferred, with at least 99% being the more preferred.
Particularly preterred embodiments in this respect, moreover, are polynucleotides which encode polypeptides which retain substantially the same biological function or activity as the mature pol\ peptide encoded by the cDNA of Figure 1.
The present invention further relates to polynucleotides that hybridize to the herein above-described sequences In this regard, the present invention especially relates to polynucleotides which hybridize under stringent conditions to the herein above- described polynucleotides. As herein used, the term "stringent conditions" means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences.
As discussed additionally herein regarding polynucleotide assays of the invention, for instance, polynucleotides of the invention as discussed above, may be used as a hybridization probe for cDNA and genomic DNA to isolate full-length cDNAs and genomic clones encoding human AIM-I and to isolate cDNA and genomic clones of other genes that have a high sequence similarity to the human AIM-I gene. Such probes generally will comprise at least 15 bases. Preferably, such probes will have at least 30 bases and may have at least 50 bases. Particularly preferred probes will have at least 30 bases and will have 50 bases or less.
For example, the coding region of the AIM-I gene may be isolated by screening using the known DNA sequence to synthesize an oligonucleotide probe. A labeled oligonucleotide having a sequence complementary to that of a gene of the present invention is then used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes to.
The polynucleotides and polypeptides of the present invention may be employed as research reagents and materials for discovery of treatments and diagnostics to human disease, as further discussed herein relating to polynucleotide assays, inter alia.
The polynucleotides ma> encode a polypeptide which is the mature protein plus additional amino or carboxyl-terminal amino acids, or amino acids interior to the mature polypeptide (when the mature l rm has more than one polypeptide chain, for instance). Such sequences may play a role in processing of a protein from precursor to a mature form, may facilitate protein trafficking, may prolong or shorten protein half-life or may facilitate manipulation of a protein for assay or production, among other things. As generally is the case in situ, the additional amino acids may be processed away from the mamre protein by cellular enzymes.
A precursor protein, having the mamre form of the polypeptide fused to one or more prosequences may be an inactive form of the polypeptide. When prosequences are removed such inactive precursors generally are activated. Some or all of the prosequences may be removed before activation. Generally, such precursors are called proproteins.
In sum, a polynucleotide of the present invention may encode a mamre protein, a mamre protein plus a leader sequence (which may be referred to as a preprotein), a precursor of a mature protein having one or more prosequences which are not the leader sequences of a preprotein. or a preproprotein. which is a precursor to a proprotein, having a leader sequence and one or more prosequences, which generally are removed during processing steps that produce active and mamre forms of the polypeptide.
A deposit containing a human AIM-I cDNA has been deposited with the American Type Culture Collection, as noted above. Also as noted above, the human cDNA deposit is referred to herein as "the deposited clone" or as "the cDNA of the deposited clone. "
The deposited clone was deposited with the American Type Culture Collection, 12301 Park Lawn Dme. Rockv ille. Mar land 20852, USA. on February 20, 1996, and assigned ATCC Deposit No. 97448.
The deposited material is a pBluescript SK (-) plasmid (Stratagene, La Jolla, CA) that contains the full length AIM-I cDNA. referred to as "PF261 " upon deposit. The deposit has been made under the terms of the Budapest Treaty on the international recognition of the deposit of micro-organisms for purposes of patent procedure. The strain will be irrevocably and without restriction or condition released to the public upon the issuance of a patent. The deposit is provided merely as convenience to those of skill in the art and is not an admission that a deposit is required for enablement, such as that required under 35 U.S.C. §112. The sequence of the polynucleotides contained in the deposited material, as well as the amino acid sequence of the polypeptide encoded thereby, are controlling in the event of any conflict with any description of sequences herein. A license may be required to make, use or sell the deposited materials, and no such license is hereby granted.
The present invention further relates to a human AIM-I polypeptide which has the deduced amino acid sequence of Figure 1.
The invention also relates to fragments, analogs and derivatives of these polypeptides. The terms "fragment." "derivative" and "analog" when referring to the polypeptide of Figure 1 means a polypeptide which retains essentially the same biological function or activity as such polypeptide. Thus, an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mamre polypeptide.
The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic
Figure imgf000025_0001
In ceπain preferred embodiments it is a recombinant polypeptide.
The fragmeni. derivative or analog of the polypeptide of Figure 1 may be (i) one in which one or more of the ammo acid residues are substituted with a conserved or non-conserved ammo acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mamre polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mamre polypeptide. such as a leader or secretory sequence or a sequence which is employed for purification of the mamre polypeptide or a proprotein sequence. Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
Among the particularly preferred embodiments of the invention in this regard are polypeptides having the amino acid sequence of AIM-I set out in Figure 1 , variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments. Alternatively, particularly preferred embodiments of the invention in this regard are polypeptides having the amino acid sequence of the AIM-I of the cDNA in the deposited clone, variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments.
Among preferred variants are those that vary from a reference by conservative amino acid substimtions. Such substimtions are those that substitute a given amino acid in a polypeptide by another amino acid of like characteristics. Typically seen as conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala. Val. Leu and He; interchange of the hydroxyl residues Ser and Thr. exchange of the acidic residues Asp and Glu. substimtion between the amide residues Asn and Gin, exchange of the basic residues Lys and Arg and replacements among the aromatic residues Phe. Tyr.
Further particularly preferred in this regard are variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments, having the amino acid sequence of the AIM-I polypeptide of Figure 1 in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination. Especially preferred among these are silent substimtions, additions and deletions, which do not alter the properties and activities of the AIM-I. Also especially preferred in this regard are conservative substimtions. Most highly preferred are polypeptides having the amino acid sequence of Figure 1 without substimtions.
The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
The polypeptides of the present invention include the polypeptide of SEQ ID NO: 2 (in particular the mamre polypeptide) as well as polypeptides which have at least 70% similarity (preferably at least 70% identity) to the polypeptide of SEQ ID NO:2 and more preferably at least 90% similarity (more preferably at least 90% identity) to the polypeptide of SEQ ID NO:2 and still more preferably at least 95% similarity (still more preferably at least 95 % identity) to the polypeptide of SEQ ID NO:2 and also include portions of such polypeptides with such portion of the polypeptide generally containing at least 30 amino acids and more preferably at least 50 amino acids.
As known in the an "similarity" between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide.
Fragments or portions of the polypeptides of the present invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention.
Also among preferred embodiments of this aspect of the present invention are polypeptides comprising fragments of AIM-I, most particularly fragments of the AIM-I having the amino acid set out in Figure 1 , and fragments of variants and derivatives of the AIM-I of Figure 1. In this regard a fragment is a polypeptide having an amino acid sequence that entirely is the same as part but not all of the amino acid sequence of the aforementioned AIM-I polypeptides and variants or derivatives thereof.
Such fragments may be "free-standing, " i.e. , not part of or fused to other amino acids or polypeptides, or they may be comprised within a larger polypeptide of which they form a part or region. When comprised within a larger polypeptide. the presently discussed fragments most preferably form a single continuous region. However, several fragments may be comprised within a single larger polypeptide. For instance, certain preferred embodiments relate to a fragment of an AIM-I polypeptide of the present comprised within a precursor polypeptide designed for expression in a host and having heterologous pre and pro-polypeptide regions fused to the amino terminus of the AIM-I fragment and an additional region fused to the carboxyl terminus of the fragment. Therefore, fragments in one aspect of the meaning intended herein, refers to the portion or portions of a fusion polypeptide or fusion protein derived from AIM-I.
As representative examples of polypeptide fragments of the invention, there may be mentioned those w hich have from about 100 to about 281 amino acids.
In this context, "about" includes the particularly recited range and ranges larger or smaller by several, a few. 5. 4. 3. 2 or 1 amino acid at either extreme or at both extremes. For instance, about 100-281 amino acids in this context means a polypeptide fragment of 100 plus or minus several, a few . 5, 4, 3, 2 or 1 amino acids to 281 plus or minus several a few . 5. 4. 3. 2 or I amino acid residues, i.e. , ranges as broad as 100 minus several amino acids 10 281 plus several amino acids to as narrow as 100 plus several amino acids to 281 minus several amino acids.
Highly preferred in this regard are the recited ranges plus or minus as many as 5 amino acids at either or at both extremes. Particularly highly preferred are the recited ranges plus or minus as many as 3 ammo acids at either or at both the recited extremes. Especially particularly highly preferred are ranges plus or minus 1 amino acid at either or at both extremes or the recited ranges with no additions or deletions. Most highly preferred of all in this regard are fragments from about 100 to about 281 amino acids.
Among especially preferred fragments of the invention are truncation mutants of AIM-I. Truncation mutants include AIM-I polypeptides having the amino acid sequence of Figure 1, or of variants or derivatives thereof, except for deletion of a continuous series of residues (that is, a continuous region, pan or poπion) that includes the amino terminus, or a continuous series of residues that includes the carboxyl terminus or, as in double truncation mutants, deletion of two continuous series of residues, one including the amino terminus and one including the carboxyl terminus. Fragments having the size ranges set out about also are preferred embodiments of truncation fragments, which are especially preferred among fragments generally.
Also preferred in this aspect of the invention are fragments characterized by structural or functional attributes of AIM-I. Preferred embodiments of the invention in this regard include fragments that comprise alpha-helix and alpha-helix forming regions ("alpha-regions"), beta-sheet and beta-sheet-forming regions ("beta-regions"), turn and turn-forming regions ("turn- regions"), coil and coil-forming regions ("coil-regions"), hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface-forming regions and high antigenic index regions of AIM-I.
Ceπain preferred regions in these regards are set out in Figure 3, and include, but are not limited to. regions of the aforementioned types identified by analysis of the amino acid sequence set out in Figure 1 . As set out in Figure 3, such preferred regions include Gamier- Robson alpha-regions, beta-regions, turn-regions and coil-regions, Chou-Fasman alpha-regions, beta-regions and turn-regions, Kyte-Doolittle hydrophilic n. _ions and hydrophilic regions. Eisenberg alpha and beta amphipathic regions, Karplus- Schulz flexible regions. Emini surface-forming regions and Jameson-Wolf high antigenic index regions. Among highly preferred fragments in this regard are those that comprise regions of AIM-I that combine several structural features, such as several of the features set out above. In this regard, the regions defined by the residues about 1 to about 281 of Figure 1 , which all are characterized by amino acid compositions highly characteristic of turn-regions, hydrophilic regions, flexible-regions, surface-forming regions, and high antigenic index-regions, are especially highly preferred regions. Such regions may be comprised within a larger polypeptide or may be by themselves a preferred fragment of the present invention, as discussed above. It will be appreciated that the term "about" as used in this paragraph has the meaning set out above regarding fragments in general. Further, a highly preferred fragment comprises amino acids 39 through 281 which constitute a soluble poπion of the overall AIM-I polypeptide sequence of Figure 1.
Fuπher preferred regions are those that mediate activities of AIM-I. Most highly preferred in this regard are fragments that have a chemical, biological or other activity of AIM-I, including those with a similar activity or an improved activity, or with a decreased undesirable activity. Highly preferred in this regard are fragments that contain regions that are homologs in sequence, or in position, or in both sequence and to active regions of related polypeptides. such as the related polypeptides set out in Figure 2. which include members of the TNF family. Among particularly preferred fragments in these regards are truncation mutants, as discussed above.
It will be appreciated thai the invention also relates to. among others, polynucleotides encoding the aforementioned fragments, polynucleotides that hybridize to polynucleotides encoding the tragments. particularly those that hybridize under stringent conditions, and po I nucleotides. such as PCR primers, for amplifying polynucleotides that encode the tragments In these regards, preferred polynucleotides are those that correspondent to the preferred fragments, as discussed above. The present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.
Host cells can be genetically engineered to incorporate polynucleotides and express polypeptides of the present invention. For instance, polynucleotides may be introduced into host cells using well known techniques of infection, transduction, transfection, transvection and transformation. The polynucleotides may be introduced alone or with other polynucleotides. Such other polynucleotides may be introduced independently, co-introduced or introduced joined to the polynucleotides of the invention.
Thus, for instance, polynucleotides of the invention may be transfected into host cells with another, separate, polynucleotide encoding a selectable marker, using standard techniques for co-transfection and selection in, for instance, mammalian cells. In this case the polynucleotides generallv w ill be stably incorporated into the host cell genome.
Alternatively, the polynucleotides may be joined to a vector containing a selectable marker for propagation in a host. The vector construct may be introduced into host cells by the aforementioned techniques. Generally, a plasmid vector is introduced as DNA in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid tlectroporation also may be used to introduce polynucleotides into a host. 11 the vector is a virus, it may be packaged in vitro or introduced into a packaging cell and the packaged virus may be transduced into cells. A wide variety of techniques suitable for making polynucleotides and for introducing polynucleotides into cells in accordance w ith this aspect of the invention are well known and routine to those of skill in the an. Such techniques are reviewed at length in Sambrook et al cited above, which is illustrative of the many laboratory manuals that detail these techniques. In accordance with this aspect of the invention the vector may be, for example, a plasmid vector, a single or double-stranded phage vector, a single or double-stranded RNA or DNA viral vector. Such vectors may be introduced into cells as polynucleotides, preferably DNA, by well known techniques for introducing DNA and RNA into cells. The vectors, in the case of phage and viral vectors also may be and preferably are introduced into cells as packaged or encapsidated virus by well known techniques for infection and transduction. Viral vectors may be replication competent or replication defective. In the latter case viral propagation generally will occur only in complementing host cells.
Preferred among vectors, in certain respects, are those for expression of polynucleotides and polypeptides of the present invention. Generally, such vectors comprise -acting control regions effective for expression in a host operatively linked to the polynucleotide to be expressed. Appropriate trans-acting factors either are supplied by the host, supplied by a complementing vector or supplied by the vector itself upon introduction into the host.
In cenain preferred embodiments in this regard, the vectors provide for specific expression. Such specific expression may be inducible expression or expression only in ceπain types of cells or both inducible and cell-specific. Paπicularly preferred among inducible vectors are vectors that can be induced for expression by environmental factors that are easy to manipulate, such as temperamre and nutrient additives. A variety of vectors suitable to this aspect of the invention, including constimtive and inducible expression vectors for use in prokaryotic and eukaryotic hosts, are well known and employed routinely bv those of skill in the an.
The engineered host cells can be cultured in conventional nutrient media, which may be modified as appropriate for. inter alia, activating promoters, selecting transformants or amplifying genes. Culture conditions, such as temperamre, pH and the 1 > '. previously used with the host cell selected for expression generally will be suitable for expression of polypeptides of the present invention as will be apparent to those of skill in the an. A great variety of expression vectors can be used to express a polypeptide of the invention. Such vectors include chromosomal, episomal and virus-derived vectors e.g. , vectors derived from bacterial plasmids, from bacteriophage, from yeast episomes, from yeast chromosomal elements, from viruses such as baculovimses, papova vimses, such as SV40, vaccinia vimses, adenoviruses, fowl pox vimses, pseudorabies vimses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids, all may be used for expression in accordance with this aspect of the present invention. Generally, any vector suitable to maintain, propagate or express polynucleotides to express a polypeptide in a host may be used for expression in this regard.
The appropriate DNA sequence may be inseπed into the vector by any of a variety of well-known and routine techniques. In general, a DNA sequence for expression is joined to an expression vector by cleaving the DNA sequence and the expression vector with one or more restriction endonucleases and then joining the restriction fragments together using T4 DNA ligase. Procedures for restriction and ligation that can be used to this end are well known and routine to those of skill. Suitable procedures in this regard, and for constmcting expression vectors using alternative techniques, which also are well known and routine to those skill, are set forth in great detail in Sambrook et al cited elsewhere herein.
The DNA sequence in the expression vector is operatively linked to appropriate expression control sequence! s». including, for instance, a promoter to direct mRNA transcription. Representatives of such promoters include the phage lambda PL promoter, the E. coli lac. tip and tae promoters, the SV40 early and late promoters and promoters of retroviral LTRs. to name just a few of the well-known promoters. It will be understood that numerous promoters not mentioned are suitable for use in this aspect of the invention are well known and readily may be employed by those of skill in the manner illustrated by the discussion and the examples herein. In general, expression constructs will contain sites for transcription initiation and termination, and, in the transcribed region, a ribosome binding site for translation. The coding poπion of the mamre transcripts expressed by the constructs will include a translation initiating AUG at the beginning and a termination codon appropriately positioned at the end of the polypeptide to be translated.
In addition, the constmcts may contain control regions that regulate as well as engender expression. Generally, in accordance with many commonly practiced procedures, such regions will operate by controlling transcription, such as repressor binding sites and enhancers, among others.
Vectors for propagation and expression generally will include selectable markers. Such markers also may be suitable for amplification or the vectors may contain additional markers for this purpose. In this regard, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells. Preferred markers include dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, and tetracycline or ampicillin resistance genes for culruring £ coli and other bacteria.
The vector containing the appropriate DNA sequence as described elsewhere herein, as well as an appropriate promoter, and other appropriate control sequences, may be introduced into an appropriate host using a variety of well known techniques suitable to expression therein of a desired polypeptide. Representative examples of appropriate hosts include bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as east cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells, animal cells such as CHO, COS and Bowes melanoma cells; and plant cells. Hosts for of a great variety of expression constmcts are well known, and those of skill will be enabled bv the present disclosure readily to select a host for expressing a polypeptides in accordance with t is aspect of the present invention. More paπicularly, the present invention also includes recombinant constmcts, such as expression constmcts, comprising one or more of the sequences described above. The constmcts comprise a vector, such as a plasmid or viral vector, into which such a sequence of the invention has been inseπed. The sequence may be inseπed in a forward or reverse orientation. In certain preferred embodiments in this regard, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the an. and there are many commercially available vectors suitable for use in the present invention.
The following vectors, which are commercially available, are provided by way of example. Among vectors preferred for use in bacteria are pQE70, pQE60 and pQE- 9, available from Qiagen: pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A. pNHlόa, pNH18A, pNH46A. available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540. pRIT5 available from Pharmacia. Among preferred eukaryotic vectors are pWLNEO. pSV2CAT. pOG44, pXTl and pSG available from Stratagene; and pSVK3, pBPV. pMSG and pSVL available from Pharmacia. These vectors are listed solely by way of illustration of the many commercially available and well known vectors that are available to those of skill in the art for use in accordance with this aspect of the present invention. It will be appreciated that any other plasmid or vector suitable for. for example, introduction, maintenance, propagation or expression of a polynucleotide or polypeptide of the invention in a host may be used in this aspect of the invention.
Promoter regions can be selected from any desired gene using vectors that contain a reporter transcript ion unit lacking a promoter region, such as a chloramphenicol acetyl transferase ( "cat" ) transcription unit, downstream of restriction site or sites for introducing a candidate promoter fragment; i.e. , a fragment that may contain a promoter. As is well known, introduction into the vector of a promoter- containing fragment at the restriction site upstream of the cat gene engenders production of CAT activity, which can be detected by standard CAT assays. Vectors suitable to this end are well known and readily available. Two such vectors are pKK232-8 and pCM7. Thus, promoters for expression of polynucleotides of the present invention include not only well known and readily available promoters, but also promoters that readily may be obtained by the foregoing technique, using a reporter gene.
Among known bacterial promoters suitable for expression of polynucleotides and polypeptides in accordance with the present invention are the £. coli lad and lacZ and promoters, the T3 and T7 promoters, the T5 tae promoter, the lambda PR, PL promoters and the trp promoter. Among known eukaryotic promoters suitable in this regard are the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous sarcoma vims ("RSV"). and metallothionein promoters, such as the mouse metallothionein-I promoter.
Selection of appropriate vectors and promoters for expression in a host cell is a well known procedure and the requisite techniques for expression vector constmction, introduction of the vector into the host and expression in the host are routine skills in the art.
The present invention also relates to host cells containing the above-described constmcts discussed above. The host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukar otic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
Introduction of the construct into the host cell can be effected by calcium phosphate transfection. DEΛF.-dextran mediated transfection, cationic lipid-mediated transfection, electroporation. transduction. infection or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al. Basic Methods In Molecular Bioloεv. ( 1986). Constmcts in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
Mamre proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constmcts of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook et al. Molecular Cloning: A Laboratory Manual. 2nd Ed.. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989).
Generally, recombinant expression vectors will include origins of replication, a promoter derived from a highly -expressed gene to direct transcription of a downstream structural sequence, and a selectable marker to permit isolation of vector containing cells after exposure to the vector.
Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes may be increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA. usually about from 10 to 300 bp that act to increase transcriptional activity of a promoter in a given host cell-type. Examples of enhancers include the SV4O enhancer, w hich is located on the late side of the replication origin at bp 100 to 270. the cv iomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovims enhancers.
Polynucleotides of the invention, encoding the heterologous structural sequence of a polypeptide of the invention generally w ill be inseπed into the vector using standard techniques so that it is onerablv linked to the promoter for expression. The polynucleotide will he positioned so that the transcription staπ site is located appropriately 5' to a ribosome binding site. The ribosome binding site will be 5' to the AUG that initiates translation of the polypeptide to be expressed. Generally, there will be no other open reading frames that begin with an initiation codon, usually AUG, and lie between the ribosome binding site and the initiating AUG. Also, generally, there will be a translation stop codon at the end of the polypeptide and there will be a polyadenylation signal and a transcription termination signal appropriately disposed at the 3' end of the transcribed region.
For secretion of the translated protein into the lumen of the endoplasmic reticulum, into the periplasmic space or into the extracellular environment, appropriate secretion signals may be incorporated into the expressed polypeptide. The signals may be endogenous to the polypeptide or they may be heterologous signals.
The polypeptide may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals but also additional heterologous functional regions. Thus, for instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification or during subsequent handling and storage. Also, region also may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the an.
Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, here the selected promoter is inducible it is induced by appropriate means (e.g.. temperature shift or exposure to chemical inducer) and cells are cultured for an additional period. Cells typically then are harvested by centrifugation. dismpted by physical or chemical means, and the resulting cmde extract retained for further purification. Microbial cells employed in expression of proteins can be dismpted by any convenient method, including freeze-thaw cycling, sonication, mechanical dismption. or use of cell lysing agents, such methods are well know to those skilled in the art.
Various mammalian cell cul re systems can be employed for expression, as well. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblast, described in Gluzman et al, Cell, 23: 175 (1981). Other cell lines capable of expressing a compatible vector include for example, the C127, 3T3, CHO, HeLa, human kidney 293 and BHK cell lines.
The AIM-I polypeptide can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxy lapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification.
Polypeptides of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosv lated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host- mediated processes.
AIM-I polynucleotides and polypeptides may be used in accordance with the present invention for a variety of applications, particularly those that make use of the chemical and biological properties AIM-I. Among these are applications in autoimmune disease and aberrant cellular proliferation. Additional applications relate to diagnosis and to treatment of disorders of cells, tissues and organisms. These aspects of the invention are illustrated further by the following discussion.
This invention is also related to the use of the AIM-I polynucleotides to detect complementary polynucleotides such as, for example, as a diagnostic reagent. Detection of a mutated form of AIM-I associated with a dysfunction will provide a diagnostic tool that can add or define a diagnosis of a disease or susceptibility to diseases which results from under-expression over-expression or altered expression of AIM-I, such as, for example, autoimmune diseases.
Individuals carrying mutations in the human AIM-I gene may be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material. The genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR prior to analysis. PCR (Saiki et al, Nature, 324: 163-166, ( 1986). RNA or cDNA may also be used in the same ways. As an example, PCR primers complementary to the nucleic acid encoding AIM-I can be used to identify and analyze AIM-1 expression and mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled AIM-I RS \ or alterrutivel . radiolabeled AIM-I antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melung lemperatures.
Sequence differences between a reference gene and genes having mutations also may be revealed by direct DNA sequencing. In addition, cloned DNA segments may be employed as probes to detect specific DNA segments. The sensitivity of such methods can be great Iv enhanced bv appropriate use of PCR or another amplification method. For example, a sequencing primer is used with double-stranded PCR product or a single-stranded template molecule generated by a modified PCR. The sequence determination is performed by conventional procedures with radiolabeled nucleotide or by automatic sequencing procedures with fluorescent- tags.
Genetic testing based on DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels, with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g. , Myers et al, Science, 230: 1242, 1985).
Sequence changes at specific locations also may be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method (e.g. , Cotton er a/.. Proc. Natl. Acad. Sci., USA, 55:4397-4401 , 1985).
Thus, the detection of a specific DNA sequence may be achieved by methods such as hybridization. RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g.. restriction fragment length polymorphisms ("RFLP") and Southern blotting of genomic DNA.
In addition to more conventional gel-electrophoresis and DNA sequencing, mutations also can be detected h\ in situ analysis.
The sequences of the present invention are also valuable for chromosome identification. The sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome. Moreover, there is a current need for identifying particular sites on the chromosome. Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location. The mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease.
In ceπain preferred embodiments in this regard, the cDNA herein disclosed is used to clone genomic DNA of an AIM-I gene. This can be accomplished using a variety of well known techniques and libraries, which generally are available commercially. The genomic DNA the is used for in situ chromosome mapping using well known techniques for this purpose. Typically, in accordance with routine procedures for chromosome mapping, some trial and error may be necessary to identify a genomic probe that gives a good in situ hybridization signal.
In some cases, in addition, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA. Computer analysis of the 3' untranslated region of the gene is used to rapidly select primers that do not span more than one exon in the genomic DNA. thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment.
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a paπicular DNA to a particular chromosome. Using the present invention with the same oligonucleotide primers. suhlocalization can be achieved with panels of fragments from specific chromosomes or pools of large genomic clones in an analogous manner. Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreemng w ith labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific-cDNA libraries. Fluorescence in situ hybridization ("FISH") of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step. This technique can be used with cDNA as short as 50 or 60. For a review of this technique, see Verma et al, Human Chromosomes: A Manual Of Basic Techniques, Pergamon Press, New York (1988).
Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found, for example, in V. McKusick, Mendelian Inheritance In Man, available on line through Johns Hopkins University, Welch Medical Library. The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes).
Next, it is necessary to determine the differences in the cDNA or genomic sequence between affected and unaffected individuals. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease.
With current resolution of physical mapping and genetic mapping techniques, a cDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 jnu 500 potential causative genes. (This assumes 1 megabase mapping resolution and one gene per 20 kb).
The present invention alv> relates to a diagnostic assays such as quantitative and diagnostic assays for detecting levels of AIM-I protein in cells and tissues, including determination of normal and abnormal levels. Thus, for instance, a diagnostic assay in accordance with the invention tor detecting over-expression of AIM-I protein compared to normal control tissue samples may be used to detect the presence of aberrant cellular proliferation such as cancer, for example. Assay techniques that can be used to determine levels of a protein, such as an AIM-I protein of the present invention, in a sample derived from a host are well-known to those of skill in the an. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays. Among these ELISAs frequently are preferred. An ELISA assay initially comprises preparing an antibody specific to AIM-I, preferably a monoclonal antibody. In addition a reponer antibody generally is prepared which binds to the monoclonal antibody. The reponer antibody is attached a detectable reagent such as radioactive, fluorescent or enzymatic reagent, in this example horseradish peroxidase enzyme.
To carry out an ELISA a sample is removed from a host and incubated on a solid suppoπ. e.g. a polystyrene dish, that binds the proteins in the sample. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein such as bovine semm albumin. Next, the monoclonal antibody is incubated in the dish during which time the monoclonal antibodies attach to any AIM-I proteins attached to the polystyrene dish. Unbound monoclonal antibody is washed out with buffer. The reponer antibody linked to horseradish peroxidase is placed in the dish resulting in binding of the reponer antibody to any monoclonal antibody bound to AIM-I. Unattached reponer antibody is then washed out. Reagents for peroxidase activity, including a colorimetric substrate are then added to the dish. Immobilized peroxidase, linked to AIM-I through the primary and secondary antibodies, produces a colored reaction product. The amount of color developed in a given time period indicates the amount of AIM-I protein present in the sample. Quantitative results typically are obtained by reference to a standard curve.
A competition assay may be employed wherein antibodies specific to AIM-I attached to a solid suppoπ and labeled AIM-I and a sample derived from the host are passed over the solid suppoπ and the amount of label detected attached to the solid suppoπ can be correlated to a quantity of AIM-I in the sample. The polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto. These antibodies can be, for example, polyclonal or monoclonal antibodies. The present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the an may be used for the production of such antibodies and fragments.
Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler. G. and Milstein, C., Nature, 256:495-497 (1975), the trioma technique, the human B-cell hybridoma technique (Kozbor et al. Immunology Today, 4:12 (1983) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al . pg. 77-96 in Monoclonal Antibodies And Cancer Therapy, Alan R. Liss. Inc. ( 1985 ).
Techniques described for the production of single chain antibodies (U.S. Patent No. 4.946,778) can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention. Also, transgenic mice, or other organisms such as other mammals, may be used to express humanized antibodies to immunogenic polypeptide products of this invention. The above-described antibodies may be employed to isolate or to identify clones expressing the polypeptide or purify the polypeptide of the present invention by attachment of the antibody to a solid suppoπ for isolation and/or purification by affinity chromatography. These antibodies may be employed to treat auto-immune diseases by preventing the ligand from binding its receptor.
Thus, among others, the AIM-I of the present invention may be employed to treat lymphoproliferative disease which results in lymphadenopathy, the AIM-I mediates apoptosis by stimulating clonal deletion of T-cells and may therefore, by employed to treat autoimmune disease, to stimulate peripheral tolerance and cytotoxic T-cell mediated apoptosis. The AIM-1 may also be employed as a research tool in elucidating the biology of autoimmune disorders including systemic lupus erythematosus, immunoproliferative disease lymphadenopathy (IPL), angioimmunoproliferative lymphadenopathy (AIL), rheumatoid aπhritis. diabetes, and multiple sclerosis, and to treat graft versus host disease.
The AIM-I may of the present invention may also be employed to inhibit neoplasia, such as tumor cell growth. The AIM-I polypeptide may be responsible for tumor destmction through apoptosis and cytotoxicity to ceπain cells. AIM-I may also be employed to treat diseases which require growth promotion activity, for example, restenosis, since AIM-I has proliferation effects on cells of endothelial origin. AIM-I may. therefore, also be employed to regulate hematopoiesis in endothelial cell development.
The polynucleotide encoding the AlM-1 may be employed as a diagnostic marker for determining expression of the polypeptide of the present invention since the gene is found in many tumor cell lines including pancreatic tumor, testes mmor, endometrial rumor and T-cell lymphoma. This invention also provides a method for identification of molecules, such as receptor molecules, that bind AIM-I. Genes encoding proteins that bind AIM-I, such as receptor proteins, can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting. Such methods are described in many laboratory manuals such as, for instance, Coligan et al, Current Protocols in Immunology 1(2): Chapter 5 (1991).
For instance, expression cloning may be employed for this purpose. To this end polyadenylated RNA is prepared from a cell responsive to AIM-I, a cDNA library is created from this RNA. the library is divided into pools and the pools are transfected individually into cells that are not responsive to AIM-I. The transfected cells then are exposed to labeled AIM-I. (AIM-I can be labeled by a variety of well-known techniques including standard methods of radio-iodination or inclusion of a recognition site for a site-specific protein kinase.) Following exposure, the cells are fixed and binding of AIM-I is determined. These procedures conveniently are carried out on glass slides.
Pools are identified of cDNA that produced AIM-I-binding cells. Sub-pools are prepared from these positives, transfected into host cells and screened as described above. Using an iterative sub-pooling and re-screening process, one or more single clones that encode the putative binding molecule, such as a receptor molecule, can be isolated.
Alternatively a labeled ligand can be photoaffinity linked to a cell extract, such as a membrane or a membrane extract, prepared from cells that express a molecule that it binds, such as a receptor molecule. Cross-linked material is resolved by polyacrylamide gel electrophoresis ( "PAGE") and exposed to X-ray film. The labeled complex containing the ligand-receptor can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing can be used to design unique or degenerate oligonucleotide probes to screen cDNA libraries to identify genes encoding the putative receptor molecule. Polypeptides of the invention also can be used to assess AIM-I binding capacity of AIM-I binding molecules, such as receptor molecules, in cells or in cell-free preparations.
The invention also provides a method of screening compounds to identify those which enhance or block the action of AIM-I on cells, such as its interaction with AIM-I- binding molecules such as receptor molecules. An agonist is a compound which increases the namral biological functions of AIM-I or which functions in a manner similar to AIM-I, while antagonists decrease or eliminate such functions.
For example, a cellular compartment, such as a membrane or a preparation thereof, such as a membrane-preparation, may be prepared from a cell that expresses a molecule that binds AIM-I. such as a molecule of a signaling or regulatory pathway modulated by AIM-I. The preparation is incubated with labeled AIM-I in the absence or the presence of a candidate molecule which may be an AIM-I agonist or antagonist. The ability of the candidate molecule to bind the binding molecule is reflected in decreased binding of the labeled ligand. Molecules which bind gratuitously, i.e. , without inducing the effects of AIM-I on binding the AIM-I binding molecule, are most likely to be good antagonists. Molecules that bind well and elicit effects that are the same as or closely related to AIM-I. are good agonists.
AlM-I-like effects of potential agonists and antagonists may by measured, for instance, by determining activ ity of a second messenger system following interaction of the candidate molecule with a cell or appropriate cell preparation, and comparing the effect with that of AIM-I or molecules that elicit the same effects as AIM-I. Second messenger systems that may be useful in this regard include but are not limited to AMP guanylate cyclase. ion channel or phosphoinositide hydrolysis second messenger systems. Another example of an assay for AIM-I antagonists is a competitive assay that combines AIM-I and a potential antagonist with membrane-bound AIM-I receptor molecules or recombinant AIM-I receptor molecules under appropriate conditions for a competitive inhibition assay. AIM-I can be labeled, such as by radioactivity, such that the number of AIM-I molecules bound to a receptor molecule can be determined accurately to assess the effectiveness of the potential antagonist.
Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to a polypeptide of the invention, and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, a peptide, a polypeptide such as a closely related protein or antibody that binds the same sites on a binding molecule, such as a receptor molecule, without inducing AIM-I-induced activities, thereby preventing the action of AIM-I by excluding AIM-I from binding.
Other potential antagonists include antisense molecules. Antisense technology can be used to control gene expression through antisense DNA or RNA or through triple-helix formation. Antisense techniques are discussed, for example, in Okano, J. Neurochem, 56:560. 1991 : Oligodeoxvnucleotides As Antisense Inhibitors Of Gene Expression, CRC Press. Boca Raton. FL ( 1988). Triple helix formation is discussed in, for instance Lee et al. Nucleic Acids Research, 6:3073, 1979; Cooney et al , Science, 241:456 1988: and Dervan et al . Science, 257: 1360 1991. The methods are based on binding of a polynucleotide to a complementary DNA or RNA. For example, the 5' coding portion of a pol ucleotide that encodes the mamre polypeptide of the present invention may be used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length Λ DSΛ oligonucleotide is designed to be complementary to a region of the gene inv lved in transcription thereby preventing transcription and the production of AIM-l The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into AIM-I polypeptide. The oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of AIM-I. The antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g. , as hereinafter described.
The antagonists may be employed for instance to treat cachexia which is a lipid clearing defect resulting from a systemic deficiency of lipoprotein lipase. which is suppressed by AIM-I. The AIM-I antagonists may also be employed to treat cerebral malaria in which AIM-I appears to play a pathogenic role. The antagonists may also be employed to treat rheumatoid arthritis by inhibiting AIM-I induced production of inflammatory cytokines, such as IL1 in the synovial cells. When treating arthritis, AIM-I is preferably injected intra-articularly.
The AIM-I antagonists may also be employed to prevent graft-host rejection by preventing the stimulation of the immune system in the presence of a graft.
The AIM-I antagonists may also be employed to inhibit bone resorption and, therefore, to treat and/or prevent osteoporosis.
The antagonists may also be employed as anti-inflammatory agents, and to treat endotoxic shock. This critical condition results from an exaggerated response to bacterial and other types of infection.
The invention also relates 10 compositions comprising the polynucleotide or the polypeptides discussed above or the agonists or antagonists. Thus, the polypeptides of the present invention may be employed in combination with a non-sterile or sterile carrier or carriers for use with cells, tissues or organisms, such as a pharmaceutical carrier suitable for administration to a subject. Such compositions comprise, for instance, a media additive or a therapeutically effective amount of a polypeptide of the ;.' ention and a pharmaceutically acceptable carrier or excipient. Such carriers may include, but are not limited to. saline, buffered saline, dextrose, water, glycerol, ethanol and combinations thereof. The formulation should suit the mode of administration. The invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacmre, use or sale of pharmaceuticals or biological products, reflecting approval by the agency of the manufacmre, use or sale of the product for human administration.
Polypeptides and other compounds of the present invention may be employed alone or in conjunction with other compounds, such as therapeutic compounds.
The pharmaceutical compositions may be administered in any effective, convenient manner including, for instance, administration by topical, oral, anal, vaginal, intravenous, intraperitoneal. intramuscular, subcutaneous, intranasal or intradermal routes among others.
The pharmaceutical compositions generally are administered in an amount effective for treatment or prophylaxis of a specific indication or indications. In general, the compositions are administered in an amount of at least about 10 μg/kg body weight. In most cases they will be administered in an amount not in excess of about 8 mg/kg body weight per day. Preferably, in most cases, dose is from about 10 μg/kg to about 1 mg/kg body weight, daily. It will be appreciated that optimum dosage will be determined by standard methods for each treatment modality and indication, taking into account the indication, us severity . route of administration, complicating conditions and the like.
The AIM-1 polynucleotides. polypeptides, agonists and antagonists that are polypeptides may be employed in accordance with the present invention by expression of such polypeptides in vivo, in treatment modalities often referred to as "gene therapy. " Thus, for example, cells from a patient may be engineered with a polynucleotide, such as a DNA or RNA, encoding a polypeptide ex vivo, and the engineered cells then can be provided to a patient to be treated with the polypeptide. For example, cells may be engineered ex vivo by the use of a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention. Such methods are well-known in the an and their use in the present invention will be apparent from the teachings herein.
Similarly, cells may be engineered in vivo for expression of a polypeptide in vivo by procedures known in the an. For example, a polynucleotide of the invention may be engineered for expression in a replication defective retroviral vector, as discussed above. The retroviral expression construct then may be isolated and introduced into a packaging cell is transduced with a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention such that the packaging cell now produces infectious viral panicles containing the gene of interest. These producer cells may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo. These and other methods for administering a polypeptide of the present invention by such method should be apparent to those skilled in the an from the teachings of the present invention.
Retrovimses from which the retroviral plasmid vectors herein above mentioned may be derived include, but are not limited to. Moloney Murine Leukemia Vims, spleen necrosis vims, retrov iruses such as Rous Sarcoma Vims, Harvey Sarcoma Vims, avian leukosis vims, gibbon ape leukemia v ims, human immunodeficiency vims, adenovims. Myeloproliferative Sarcoma Vims, and mammary tumor vims. In one embodiment, the retroviral plasmid vector is derived from Moloney Murine Leukemia Vims.
Such vectors well include one or more promoters for expressing the polypeptide. Suitable promoters which may be employed include, but are not limited to, the retroviral LTR: the SV40 promoter: and the human cytomegalovirus (CMV) promoter described in Miller et al. Biotechntquei. 7 980-990 (1989), or any other promoter (e.g. , cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, RNA polymerase III, and β-actin promoters). Other viral promoters which may be employed include, but are not limited to, adenovims promoters, thymidine kinase (TK) promoters, and B19 parvovirus promoters. The selection of a suitable promoter will be apparent to those skilled in the an from the teachings contained herein.
The nucleic acid sequence encoding the polypeptide of the present invention will be placed under the control of a suitable promoter. Suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovims (CMV) promoter; the respiratory syncytial vims (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter: human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs herein above described); the β-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter which controls the gene encoding the polypeptide.
The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to. the PE501 . PA317, Y-2, Y-AM, PA12. T19-14X, VT- 19-17-H2. YCRE. YCRIP. GP + E-86. GP+envAml2. and DAN cell lines as described in Miller, A., Human Gene Therapv. / :5- 14 ( 1990). The vector may be transduced into the packaging cells through any means known in the an. Such means include, but are not limited to. electroporation. the use of liposomes, and CaPO4 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid. and then administered to a host.
The producer cell line w ill generate infectious retroviral vector panicles, which include the nucleic acid sequence(s) encoding the polypeptides. Such retroviral vector particles then may be employed to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide. Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.
The present invention is further described by the following examples. The examples are provided solely to illustrate the invention by reference to specific embodiments. These exemplification's, while illustrating certain specific aspects of the invention, do not portray the limitations or circumscribe the scope of the disclosed invention.
Certain terms used herein are explained in the foregoing glossary. All examples were carried out using standard techniques, which are well known and routine to those of skill in the an. except where otherwise described in detail. Routine molecular biology techniques of the following examples can be carried out as described in standard laboratory manuals, such as Sambrook et al.. Molecular Cloning: A Laboratory Manual, 2nd Ed. ; Cold Spring Harbor Laboratory Press, Cold Spring Harbor. N.Y. (1989), herein referred to as "Sambrook. " All parts or amounts set out in the following examples are by weight, unless otherwise specified.
Unless otherw i e stated si/e separation of fragments in the examples below was carried out using standard techniques of agarose and polyacrylamide gel electrophoresis ("PAGE") in Sambrtπik and numerous other references such as, for instance, by Goeddel et al . Nucleic Acids
Figure imgf000054_0001
. 5:4057 ( 1980).
Unless described otherw ise, ligations were accomplished using standard buffers, incubation temperatures and times, approximately equimolar amounts of the DNA fragments to be ligated and approximately 10 units of T4 DNA ligase ("ligase") per 0.5 μg of DNA.
Example 1 Expression and Purification of Human AIM-I Using Bacteria
The DNA sequence encoding human AIM-I in the deposited polynucleotide was amplified using PCR oligonucleotide primers specific to the amino acid carboxyl terminal sequence of the human AIM-I protein and to vector sequences 3' to the gene. Additional nucleotides containing restriction sites to facilitate cloning were added to the 5' and 3' sequences respectively.
The 5' oligonucleotide primer had the sequence 5' GCG GCG GGA TCC ATG GCT ATG ATG GAG GTC CAG 3' containing the underlined BamHI restriction site, which encodes a start AUG. followed by 18 nucleotides of the human AIM-I coding sequence set out in Figure 1.
The 3' primer had the sequence 5 CGC GCG TCT AGA GCT TAG GCA ACT AAA AAG GCC 3" containing the underlined Xbal restriction site followed by 21 nucleotides complementary to the last 21 nucleotides of the AIM-I coding sequence set out in Figure 1. including the stop codon.
The restrictions sites were convenient to restriction enzyme sites in the bacterial expression vectors pQi ^ which were used tor bacterial expression in these examples. (Qiagen, Inc.. Chatswoπh. C λ i pQE9 encodes ampicillin antibiotic resistance ("Ampr") and contains a bacterial origin of replication ("ori"), an IPTG inducible promoter, a ribosome binding site < "RBS" ι. a 6-His tag and restriction enzyme sites.
The amplified human AIM -I DNA and the vector pQE9 both were digested with BamHI and Xbal and the digested DNAs then were ligated together. Insertion of the AIM-I DNA into the restricted vector placed the AIM-I coding region downstream of and operably linked to the vector's IPTG-inducible promoter and in-frame with an initiating AUG appropriately positioned for translation of AIM-I.
The ligation mixture was transformed into competent £. coli cells using standard procedures. Such procedures are described in Sambrook et al. Molecular Cloning: A Laboratory Manual, 2nd Ed. ; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989). £. coli strain M15/rep4, containing multiple copies of the plasmid pREP4. which expresses lac repressor and confers kanamycin resistance ("Kanr"), was used in carrying out the illustrative example described here. This strain, which is only one of many that are suitable for expressing AIM-I, is available commercially from Qiagen.
Transformants were identified by their ability to grow on LB plates in the presence of ampicillin. Plasmid DNA was isolated from resistant colonies and the identity of the cloned DNA was confirmed by restriction analysis.
Clones containing the desired constmcts were grown overnight ("O/N") in liquid culture in LB media supplemented with both ampicillin (100 μg/ml) and kanamycin (25 μg/ml).
The O/N culture was used to inoculate a large culture, at a dilution of approximately 1 : 100 to 1 :250. The cells were grown to an optical density at 600 nm ("ODioo") of between 0.4 and 0.6. Isopropyl-B-D-thiogalactopyranoside ("IPTG") was then added to a final concentration of 1 mM to induce transcription from lac repressor sensitive promoters, by inactivating the lad repressor. Cells subsequently were incubated further for 3 to 4 hours. Cells then were harvested by centrifugation and dismpted, by standard methods. Inclusion bodies were purified from the dismpted cells using routine collection techniques, and protein was solubilized from the inclusion bodies into 8M urea. The 8M urea solution containing the solubilized protein was passed over a PD-10 column in 2X phosphate buffered saline ("PBS"), thereby removing the urea, exchanging the buffer and refolding the protein. The protein was purified by a further step of chromatography to remove endotoxin. Then, it was sterile filtered. The sterile filtered protein preparation was stored in 2X PBS at a concentration of 95 micrograms per mL.
Analysis of the preparation by standard methods of polyacrylamide gel electrophoresis revealed that the preparation contained above 90% monomer AIM-I having the expected molecular weight of, approximately, 31 kDa.
Example 2 Cloning and Expression of Human AIM-I in a Baculovirus Expression System
The cDNA sequence encoding the full length human AIM-I protein, in the deposited clone is amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the gene:
The 5' primer has the sequence 5' CCG CGC GGA TCC ATC ATG GCT ATG ATG GAG GTC C 3' containing the underlined restriction enzyme site followed by 22 bases of the sequence of AIM-I of Figure 1 . Inserted into an expression vector, as described below, the 5' end of the amplified fragment encoding human AIM-I provides an efficient signal peptide. An efficient signal for initiation of translation in eukaryotic cells, as described by Kozak. M . J Mol. Biol , 196:941-950 (1987) is appropriately located in the vector portion of the construct.
The 3' primer has the sequence 5' CGC GCG TCT AGA GCT TAG CCA ACT AAA AAG GCC 3' containing the underlined Xbal restriction followed by nucleotides complementary to the last 21 nucleotides of the AIM-I coding sequence set out in Figure 1 , including the stop codon. The amplified fragment is isolated from a 1 % agarose gel using a commercially available kit ("Geneclean, " BIO 101 Inc. , La Jolla, Ca.). The fragment then is digested with BamHI and Asp718 and again is purified on a 1 % agarose gel. This fragment is designated herein F2.
The vector pA2 is used to express the AIM-I protein in the baculovims expression system, using standard methods, such as those described in Summers et al, A Manual Of Methods For Baculovims Vectors And Insect Cell Culture Procedures, Texas Agricultural Experimental Station Bulletin No. 1555 (1987). This expression vector contains the strong polyhedrin promoter of the Autographa califomica nuclear polyhedrosis vims (AcMNPV) followed by convenient restriction sites. The signal peptide of AcMNPV gp67. including the N-terminal methionine, is located just upstream of a BamHI site. The polyadenylation site of the simian vims 40 ("SV40") is used for efficient polyadenylation. For an easy selection of recombinant virus the beta- galactosidase gene from £. coli is inserted in the same orientation as the polyhedrin promoter and is followed by the polyadenylation signal of the polyhedrin gene. The polyhedrin sequences are flanked at both sides by viral sequences for cell-mediated homologous recombination with wild-type viral DNA to generate viable vims that express the cloned polynucleotide.
Many other baculovims vectors could be used in place of pA2, such as pAc373, pVL941 and pAcIM l provided, as those of skill readily will appreciate, that constmction provides appropriately K»cated signals for transcription, translation, trafficking and the like, such as an in-frame Al G and a signal peptide, as required. Such vectors are described in Lucko et al . Vint ms . 170: 1 -39, among others.
The plasmid is digested w ith the restriction enzymes BamHI and Xbal and then is dephosphorylated using calf intestinal phosphatase, using routine procedures known in the art. The DNA is then isolated from a 1 % agarose gel using a commercially available kit ("Geneclean" BIO 101 Inc., La Jolla, Ca.). This vector DNA is designated herein "V2" .
Fragment F2 and the dephosphorylated plasmid V2 are ligated together with T4 DNA ligase. £. coli HB101 cells are transformed with ligation mix and spread on culture plates. Bacteria are identified that contain the plasmid with the human AIM-I gene by digesting DNA from individual colonies using BamHI and Xbal and then analyzing the digestion product by gel electrophoresis. The sequence of the cloned fragment is confirmed by DNA sequencing. This plasmid is designated herein pBac AIM-I.
5 μg of the plasmid pBacAIM-I is co-transfected with 1.0 μg of a commercially available linearized baculovims DNA ("BaculoGold™ baculovims DNA". Pharmingen, San Diego, CA.), using the lipofection method described by Feigner et al , Proc. Natl. Acad. Sci. USA, 54:7413-7417 ( 1987). lμg of BaculoGold™ vims DNA and 5 μg of the plasmid pBacAIM-I are mixed in a sterile well of a microtiter plate containing 50 μl of semm free Grace's medium (Life Technologies Inc. , Gaithersburg, MD). Afterwards 10 μl Lipofectin plus 90 μl Grace's medium are added, mixed and incubated for 15 minutes at room temperamre. Then the transfection mixture is added drop-wise to Sf9 insect cells (ATCC CRL 171 1 ) seeded in a 35 mm tissue culture plate with 1 ml Grace's medium without semm. The plate is rocked back and forth to mix the newly added solution. The plate is then incubated for 5 hours at 27 'C. After 5 hours the transfection solution i removed from the plate and 1 ml of Grace's insect medium supplemented with \()r<' fetal calf semm is added. The plate is put back into an incubator and cultivation is continued at 27 "C for four days.
After four days the supernatant is collected and a plaque assay is performed, as described by Summers and Smith, cited above. An agarose gel with "Blue Gal" (Life Technologies Inc. , Gaithersburg) is used to allow easy identification and isolation of gal-expressing clones, which pπxluce blue-stained plaques. (A detailed description of a "plaque assay" of this type can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10).
Four days after serial dilution, the vims is added to the cells. After appropriate incubation, blue stained plaques are picked with the tip of an Eppendorf pipette. The agar containing the recombinant vimses is then resuspended in an Eppendorf mbe containing 200 μl of Grace's medium. The agar is removed by a brief centrifugation and the supernatant containing the recombinant baculovims is used to infect Sf9 cells seeded in 35 mm dishes. Four days later the supernatants of these culture dishes are harvested and then they are stored at 4'C. A clone containing properly inserted AIM-I is identified by DNA analysis including restriction mapping and sequencing. This is designated herein as V- AIM-I.
Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS. The cells are infected with the recombinant baculovims V- AIM-I at a multiplicity of infection ("MOD of about 2 (about 1 to about 3). Six hours later the medium is removed and is replaced with SF900 II medium minus methionine and cysteine (available from Life Technologies Inc. , Gaithersburg). 42 hours later, 5 μCi of 35S- methionine and 5 μCi "S cysteine (available from Amersham) are added. The cells are further incubated for 16 hours and then they are harvested by centrifugation, lysed and the labeled proteins are visualized by SDS-PAGE and autoradiography .
Example 3 Expression of AIM-I in COS Cells
The expression plasmid. ΛIM-I HA. is made by cloning a cDNA encoding AIM-I into the expression vector pcDN Al/Amp (which can be obtained from Invitrogen, Inc.). The expression vector pcDNAI/amp contains: (1) an E. coli origin of replication effective for propagation in £. coli and other prokaryotic cell; (2) an ampicillin resistance gene for selection of plasmid-containing prokaryotic cells; (3) an SV40 origin of replication for propagation in eukaryotic cells; (4) a CMV promoter, a polylinker, an SV40 intron, and a polyadenylation signal arranged so that a cDNA conveniently can be placed under expression control of the CMV promoter and operably linked to the SV40 intron and the polyadenylation signal by means of restriction sites in the polylinker.
A DNA fragment encoding the entire AIM-I precursor and a HA tag fused in frame to its 3' end is cloned into the polylinker region of the vector so that recombinant protein expression is directed by the CMV promoter. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein described by Wilson et al, Cell 37: 767 (1984). The fusion of the HA tag to the target protein allows easy detection of the recombinant protein with an antibody that recognizes the HA epitope.
The plasmid construction strategy is as follows. The AIM-I cDNA of the deposited clone is amplified using primers that contained convenient restriction sites, much as described above regarding the constmction of expression vectors for expression of AIM-I in £. coli and 5. furςψerda.
To facilitate detection, purification and characterization of the expressed AIM-I, one of the primers contains a hemagglutinin tag ("HA tag") as described above.
Suitable primers include that following, which are used in this example.
The 5' primer, containing the underlined restriction enzyme site, an AUG start codon and 22 codons thereafter, forming the hexapeptide haemagglutinin tag, has the sequence: 5' CCG CGC GGA ICC ATC ATG GCT ATG ATG GAG GTC C 3' The 3' primer, containing the underlined Xbal site and 21 nucleotides of 3' coding sequence (at the 3' end) has the sequence: 5' CGC GCG TCT AGA GCT TAG CCA ACT AAA AAG GCC 3'
The PCR amplified DNA fragment and the vector, pcDNAI/Amp, are digested with and then ligated. The ligation mixmre is transformed into E. coli strain SURE (available from Stratagene Cloning Systems, 11099 North Toπey Pines Road, La Jolla, CA 92037) the transformed culture is plated on ampicillin media plates which then are incubated to allow growth of ampicillin resistant colonies. Plasmid DNA is isolated from resistant colonies and examined by restriction analysis and gel sizing for the presence of the AIM-I-encoding fragment.
For expression of recombinant AIM-I, COS cells are transfected with an expression vector, as described above, using DEAE-DEXTRAN, as described, for instance, in Sambrook et al . Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, Cold Spring Harbor, New York (1989). Cells are incubated under conditions for expression of AIM-l by the vector.
Expression of the AIM-I HA fusion protein is detected by radiolabelling and immunoprecipitation. using methods described in, for example Harlow et al . Antibodies: A Laboratory Manual. 2nd Ed. ; Cold Spring Harbor Laboratory Press, Cold Spring Harbor. Ne York ( 1988). To this end, two days after transfection, the cells are labeled by incubation in media containing 35S-cysteine for 8 hours. The cells and the media are collected, and the cells are washed and the lysed with detergent-containing RIPA buffer: 150 mM NaCl. I S N P-40. 0. 1 % SDS, 1 % NP-40, 0.5% DOC, 50 mM TRIS. pH 7.5, as described by Wilson et al. cited above. Proteins are precipitated from the cell lysate and from the culture media using an HA-specific monoclonal antibody. The precipitated proteins then are analyzed by SDS-PAGE gels and autoradiography. .'., expression product of the expected size is seen in the cell lysate, which is not seen in negative controls. Example 4 Tissue Distribution of AIM-I Expression
Northern blot analysis is carried out to examine the levels of expression of AIM-I in human tissues, using methods described by, among others, Sambrook et al, cited above. Total cellular RNA samples are isolated with RNAzol™ B system (Biotecx Laboratories, Inc. 6023 South Loop East, Houston, TX 77033).
About lOμg of total RNA is isolated from tissue samples. The RNA is size resolved by electrophoresis through a 1 % agarose gel under strongly denaturing conditions. RNA is blotted from the gel onto a nylon filter, and the filter then is prepared for hybridization to a detectably labeled polynucleotide probe.
As a probe to detect mRNA that encodes AIM-I, the antisense strand of the coding region of the cDNA inseπ in the deposited clone, is labeled to a high specific activity. The cDNA is labeled by primer extension, using the Prime-It kit, available from Stratagene. The reaction is carried out using 50 ng of the cDNA, following the standard reaction protocol as recommended by the supplier. The labeled polynucleotide is purified away from other labeled reaction components by column chromatography using a Select-G-50 column, obtained from 5-Prime - 3-Prime, Inc. of 5603 Arapahoe Road. Boulder. CO 80303.
The labeled probe is hybridized to the filter, at a concentration of 1.000,000 cp /ml. in a small volume of 7 r; SDS. 0.5 M NaPO4, pH 7.4 at 65 * C, overnight. Thereafter the probe solution is drained and the filter is washed twice at room temperamre and twice at 60* C ith 0.5 x SSC, 0.1 % SDS. The filter then is dried and exposed to film at -70 overnight ith an intensifying screen. Autoradiography shows that mRNA for AIM-I is abundant in human heaπ. bone marrow, CD4+ and CD19* peripheral blood lymphocytes, and less so in lung and kidney tissue. Example 5 Gene Therapeutic Expression of Human AIM-I
Fibroblasts are obtained from a subject by skin biopsy. The resulting tissue is placed in tissue-culmre medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culmre flask, approximately ten pieces are placed in each flask. The flask is turned upside down, closed tight and left at room temperamre overnight. After 24 hours at room temperamre, the flask is inveπed - the chunks of tissue remain fixed to the bottom of the flask - and fresh media is added (e.g. , Ham's F12 media, with 10% FBS, penicillin and streptomycin). The tissue is then incubated at 37 'C for approximately one week. At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culmre. a monolayer of fibroblasts emerges. The monolayer is trypsinized and scaled into larger flasks.
A vector for gene therap is digested with restriction enzymes for cloning a fragment to be expressed. The digested vector is treated with calf intestinal phosphatase to prevent self-ligation. The dephosphorylated, linear vector is fractionated on an agarose gel and purified.
AIM-I cDNA capable of expressing active AIM-I, is isolated. The ends of the fragment are modified, if necessary , for cloning into the vector. For instance, 5" overhanging may be treated w ith DNA polymerase to create blunt ends. 3' overhanging ends may be removed using S I nuclease. Linkers may be ligated to blunt ends with T4 DNA ligase
Equal quantities of the Moloney murine leukemia vims linear backbone and the AIM-I fragment are mixed together and joined using T4 DNA ligase. The ligation mixmre is used to transform £. coli and the bacteria are then plated onto agar-containing kanamycin. Kanamycin phenotype and restriction analysis confirm that the vector has the properly inseπed gene.
Packaging cells are grown in tissue culmre to confluent density in Dulbecco's Modified Eagle's Medium (DMEM) with 10% calf semm (CS), penicillin and streptomycin. The vector containing the AIM-I gene is introduced into the packaging cells by standard techniques. Infectious viral panicles containing the AIM-I gene are collected from the packaging cells, which now are called producer cells.
Fresh media is added to the producer cells, and after an appropriate incubation period media is harvested from the plates of confluent producer cells. The media, containing the infectious viral particles, is filtered through a Millipore filter to remove detached producer cells. The filtered media then is used to infect fibroblast cells. Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the filtered media. Polybrene (Aldrich) may be included in the media to facilitate transduction. After appropriate incubation, the media is removed and replaced with fresh media. If the titer of vims is high, then virtually all fibroblasts will be infected and no selection is required. If the titer is low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his, to select out transduced cells for expansion.
Engineered fibroblasts then may be injected into rats, either alone or after having been grown to confluence on mιcn carrιer beads, such as cytodex 3 beads. The injected fibroblasts produce AIM-I product, and the biological actions of the protein are conveyed to the host
It will be clear that the invention may be practiced otherwise than as paπicularly described in the foregoing description and examples. Numerous modifications and variations of the present invention are possible in light of the above teachings and. therefore, are within the scope of the appended claims. SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: Ruben, Steven M (ii) TITLE OF INVENTION: Apoptosis Inducing Molecule I (iii) NUMBER OF SEQUENCES: 10
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Carella, Byrne, Bain, Gilfillan, Cecchi,
Stewart & Olstein
(B) STREET: 6 Becker Farm Road
(C) CITY: Roseland
(D) STATE: NJ
(E) COUNTRY: US
(F) ZIP: 07068
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.30
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: PCT/US96/03773
(B) FILING DATE: 14-MAR-1996
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: MULLINS, J.G.
(B) REGISTRATION NUMBER: 33,073
(C) REFERENCE/DOCKET NUMBER: 325Θ00-549
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (201) 994-1700
(B) TELEPAX: (201) 994-1744
(2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1643 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 52..894
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:1 : GGCACGAGCG GCTGCCTGGC TGACTTACAG CAGTCAGACT CTGACAGGTT C ATG GCT 57
Met Ala
1
ATG ATG GAG GTC CAG GGG GGA CCC AGC CTG GGA CAG ACC TGC GTG CTG 105 Met Met Glu Val Gin Gly Gly Pro Ser Leu Gly Gin Thr Cys Val Leu
5 10 15
ATC GTG ATC TTC ACA GTG CTC CTG CAG TCT CTC TGT GTG GCT GTA ACT 153
He Val He Phe Thr Val Leu Leu Gin Ser Leu Cys Val Ala Val Thr 20 25 30
TAC GTG TAC TTT ACC AAC GAG CTG AAG CAG ATG CAG GAC AAG TAC TCC 201
Tyr Val Tyr Phe Thr Asn Glu Leu Lys Gin Met Gin Asp Lys Tyr Ser 35 40 45 50
AAA AGT GGC ATT GCT TGT TTC TTA AAA GAA GAT GAC AGT TAT TGG GAC 249
Lys Ser Gly He Ala Cys Phe Leu Lys Glu Asp Asp Ser Tyr Trp Asp 55 60 65
CCC AAT GAC GAA GAG AGT ATG AAC AGC CCC TGC TGG CAA GTC AAG TGG 297
Pro Asn Asp Glu Glu Ser Met Asn Ser Pro Cys Trp Gin Val Lys Trp 70 75 80
CAA CTC CGT CAG CTC GTT AGA AAG ATG ATT TTG AGA ACC TCT GAG GAA 345
Gin Leu Arg Gin Leu Val Arg Lys Met He Leu Arg Thr Ser Glu Glu 85 90 95
ACC ATT TCT ACA GTT CAA GAA AAG CAA CAA AAT ATT TCT CCC CTA GTG 393
Thr He Ser Thr Val Gin Glu Lys Gin Gin Asn He Ser Pro Leu Val 100 105 110
AGA GAA AGA GGT CCT CAG AGA GTA GCA GCT CAC ATA ACT GGG ACC AGA 441
Arg Glu Arg Gly Pro Gin Arg Val Ala Ala His He Thr Gly Thr Arg 115 120 125 130
GGA AGA AGC AAC ACA TTG TCT TCT CCA AAC TCC AAG AAT GAA AAG GCT 489
Gly Arg Ser Asn Thr Leu Ser Ser Pro Asn Ser Lys Asn Glu Lys Ala 135 140 145
CTG GGC CGC AAA ATA AAC TCC TGG GAA TCA TCA AGG AGT GGG CAT TCA 537
Leu Gly Arg Lys He Asn Ser Trp Glu Ser Ser Arg Ser Gly His Ser 150 155 160
TTC CTG AGC AAC TTG CAC TTG AGG AAT GGT GAA CTG GTC ATC CAT GAA 585
Phe Leu Ser Asn Leu His Leu Arg Asn Gly Glu Leu Val He His Glu 165 170 175
AAA GGG TTT TAC TAC ATC TAT TCC CAA ACA TAC TTT CGA TTT CAG GAG 633
Lys Gly Phe Tyr Tyr He Tyr Ser Gin Thr Tyr Phe Arg Phe Gin Glu 180 185 190
GAA ATA AAA GAA AAC ACA AAG AAC GAC AAA CAA ATG GTC CAA TAT ATT 681
Glu He Lys Glu Asn Thr Lys Asn Asp Lys Gin Met Val Gin Tyr He 195 200 205 210
TAC AAA TAC ACA AGT TAT CCT GAC CCT ATA TTG TTG ATG AAA AGT GCT 729
Tyr Lys Tyr Thr Ser Tyr Pro Asp Pro He Leu Leu Met Lys Ser Ala 215 220 225 AGA AAT AGT TGT TGG TCT AAA GAT GCA GAA TAT GGA CTC TAT TCC ATC 777
Arg Asn Ser Cys Trp Ser Lys Asp Ala Glu Tyr Gly Leu Tyr Ser He 230 235 240
TAT CAA GGG GGA ATA TTT GAG CTT AAG GAA AAT GAC AGA ATT TTT GTT 825
Tyr Gin Gly Gly He Phe Glu Leu Lys Glu Asn Asp Arg He Phe Val 245 250 255
TCT GTA ACA AAT GAG CAC TTG ATA GAC ATG GAC CAT GAA GCC AGT TTT 873
Ser Val Thr Asn Glu His Leu He Asp Met Asp His Glu Ala Ser Phe 260 265 270
TTC GGG GCC TTT TTA GTT GGC TAACTGACCT GGAAAGAAAA AGCAATAACC 924
Phe Gly Ala Phe Leu Val Gly 275 280
TCAAAGTGAC TATTCAGTTT TCAGGATGAT ACACTATGAA GATGTTTCAA AAAATCTGAC 984
CAAAACAAAC AAACAGAAAA CAGAAAACAA AAAAACCTCT ATGCAATCTG AGTAGAGCAG 1044
CCACAACCAA AAAATTCTAC AACACACACT GTTCTGAAAG TGACTCACTT ATCCCAAGAA 1104
AATGAAATTG CTGAAAGATC TTTCAGGACT CTACCTCATA TCAGTTTGCT AGCAGAAATC 1164
TAGAAGACTG TCAGCTTCCA AACATTAATG CAATGGTTAA CATCTTCTGT CTTTATAATC 1224
TACTCCTTGT AAAGACTGTA GAAGAAAGCG CAACAATCCA TCTCTCAAGT AGTGTATCAC 1284
AGTAGTAGCC TCCAGGTTTC CTTAAGGGAC AACATCCTTA AGTCAAAAGA GAGAAGAGGC 1344
ACCACTAAAA GATCGCAGTT TGCCTGGTGC AGTGGCTCAC ACCTGTAATC CCAACATTTT 1404
GGGAACCCAA GGTGGGTAGA TCACGAGATC AAGAGATCAA GACCATAGTG ACCAACATAG 1464
TGAAACCCCA TCTCTACTGA AAGTGCAAAA ATTAGCTGGG TGTGTTGGCA CATGCCTGTA 1524
GTCCCAGCTA CTTGAGAGGC TGAGGCAGGA GAATCGTTTG AACCCGGGAG GCAGAGGTTG 1584
CAGTGTGGTG AGATCATGCC ACTACACTCC AGCCTGGCGA CAGAGCGAGA CTTGGTTTC 1643
(2) INFORMATION FOR SEQ ID NO:2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 281 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Met Ala Met Met Glu Val Gin Gly Gly Pro Ser Leu Gly Gin Thr Cys 1 5 10 15
Val Leu He Val He Phe Thr Val Leu Leu Gin Ser Leu Cys Val Ala 20 25 30
Val Thr Tyr Val Tyr Phe Thr Asn Glu Leu Lys Gin Met Gin Asp Lys 35 40 45
Tyr Ser Lys Ser Gly He Ala Cys Phe Leu Lys Glu Asp Asp Ser Tyr 50 55 60 Trp Asp Pro Asn Asp Glu Glu Ser Met Asn Ser Pro Cys Trp Gin Val 65 70 75 80
Lys Trp Gin Leu Arg Gin Leu Val Arg Lys Met He Leu Arg Thr Ser 85 90 95
Glu Glu Thr He Ser Thr Val Gin Glu Lys Gin Gin Asn He Ser Pro
100 105 110
Leu Val Arg Glu Arg Gly Pro Gin Arg Val Ala Ala His He Thr Gly 115 120 125
Thr Arg Gly Arg Ser Asn Thr Leu Ser Ser Pro Asn Ser Lys Asn Glu 130 135 140
Lys Ala Leu Gly Arg Lys He Asn Ser Trp Glu Ser Ser Arg Ser Gly 145 150 155 160
His Ser Phe Leu Ser Asn Leu His Leu Arg Asn Gly Glu Leu Val He 165 170 175
His Glu Lys Gly Phe Tyr Tyr He Tyr Ser Gin Thr Tyr Phe Arg Phe 180 185 190
Gin Glu Glu He Lys Glu Asn Thr Lys Asn Asp Lys Gin Met Val Gin 195 200 205
Tyr He Tyr Lys Tyr Thr Ser Tyr Pro Asp Pro He Leu Leu Met Lys 210 215 220
Ser Ala Arg Asn Ser Cys Trp Ser Lys Asp Ala Glu Tyr Gly Leu Tyr 225 230 235 240
Ser He Tyr Gin Gly Gly He Phe Glu Leu Lys Glu Asn Asp Arg He 245 250 255
Phe Val Ser Val Thr Asn Glu His Leu He Asp Met Asp His Glu Ala 260 265 270
Ser Phe Phe Gly Ala Phe Leu Val Gly 275 280
(2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 267 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
Val Asp Ser Ser Ala Ser Ser Pro Trp Ala Pro Pro Gly Thr Val Leu
1 5 10 15
Pro Cys Pro Thr Ser Val Pro Arg Arg Pro Gly Gin Arg Arg Pro Pro 20 25 30
Pro Pro Pro Pro Pro Pro Pro Leu Pro Pro Pro Pro Pro Pro Pro Pro 35 40 45
Leu Pro Pro Leu Pro Leu Pro Pro Leu Lys Lys Arg Gly Asn His Ser 50 55 60 Thr Gly Leu Cys Leu Leu Val Met Phe Phe Met Val Leu Val Ala Leu 65 70 75 80
Val Gly Leu Gly Leu Gly Met Phe Gin Leu Phe His Leu Gin Lys Glu 85 90 95
Leu Ala Glu Leu Arg Glu Ser Thr Ser Gin Met His Thr Ala Ser Ser 100 105 110
Leu Glu Lys Gin He Gly His Pro Ser Pro Pro Pro Glu Lys Lys Glu 115 120 125
Leu Arg Lys Val Ala His Leu Thr Gly Lys Ser Asn Ser Arg Ser Met 130 135 140
Pro Leu Glu Trp Glu Asp Thr Tyr Gly He Val Leu Leu Ser Gly Val 145 150 155 160
Lys Tyr Lys Lys Gly Gly Leu Val He Asn Glu Thr Gly Leu Tyr Phe 165 170 175
Val Tyr Ser Lys Val Tyr Phe Arg Gly Gin Ser Cys Asn Asn Leu Pro 180 185 190
Leu Ser His Lys Val Tyr Met Arg Asn Ser Lys Tyr Pro Gin Asp Leu 195 200 205
Val Met Met Glu Gly Lys Met Met Ser Tyr Cys Thr Thr Gly Gin Met 210 215 220
Trp Ala Arg Ser Ser Tyr Leu Gly Ala Val Phe Asn Leu Thr Ser Ala 225 230 235 240
Asp His Leu Tyr Val Asn Val Ser Glu Leu Ser Leu Val Asn Phe Glu 245 250 255
Glu Ser Gin Thr Phe Phe Gly Leu Tyr Lys Leu 260 265
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 279 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
Met Gin Gin Pro Met Asn Tyr Pro Cys Pro Gin He Phe Trp Val Asp 1 5 10 15
Ser Ser Ala Thr Ser Ser Trp Ala Pro Pro Gly Ser Val Phe Pro Cys 20 25 30
Pro Ser Cys Gly Pro Arg Gly Pro Asp Gin Arg Arg Pro Pro Pro Pro 35 40 45
Pro Pro Pro Val Ser Pro Leu Pro Pro Pro Ser Gin Pro Leu Pro Leu 50 55 60
Pro Pro Leu Thr Pro Leu Lys Lys Lys Asp His Asn Thr Asn Leu Trp 65 70 75 80 Leu Pro Val Val Phe Phe Met Val Leu Val Ala Leu Val Gly Met Gly 85 90 95
Leu Gly Met Tyr Gin Leu Phe His Leu Gin Lys Glu Leu Ala Glu Leu 100 105 110
Arg Glu Phe Thr Asn Gin Ser Leu Lys Val Ser Ser Phe Glu Lys Gin 115 120 125
He Ala Asn Pro Ser Thr Pro Ser Glu Lys Lys Glu Pro Arg Ser Val 130 135 140
Ala His Leu Thr Gly Asn Pro His Ser Arg Ser He Pro Leu Glu Trp 145 150 155 160
Glu Asp Thr Tyr Gly Thr Ala Leu He Ser Gly Val Lys Tyr Lys Lys 165 170 175
Gly Gly Leu Val He His Glu Thr Gly Leu Tyr Phe Val Tyr Ser Lys 180 185 190
Val Tyr Phe Arg Gly Gin Ser Cys Asn Asn Gin Pro Leu Asn His Lys 195 200 205
Val Tyr Met Arg Asn Ser Lys Tyr Pro Glu Asp Leu Val Leu Met Glu 210 215 220
Glu Lys Arg Leu Asn Tyr Cys Thr Thr Gly Gin He Trp Ala His Ser 225 230 235 240
Ser Tyr Leu Gly Ala Val Phe Asn Leu Thr Ser Ala Asp His Leu Tyr 245 250 255
Val Asn He Ser Gin Leu Ser Leu He Asn Phe Glu Glu Ser Lys Thr 260 265 270
Phe Phe Gly Leu Tyr Lys Leu 275
(2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 233 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:
Met Ser Thr Glu Ser Met He Arg Asp Val Glu Leu Ala Glu Glu Ala 1 5 10 15
Leu Pro Lys Lys Thr Gly Gly Pro Gin Gly Ser Arg Arg Cys Leu Phe 20 25 30
Leu Ser Leu Phe Ser Phe Leu He Val Ala Gly Ala Thr Thr Leu Phe 35 40 45
Cys Leu Leu His Phe Gly Val He Gly Pro Gin Arg Glu Glu Ser Pro 50 55 60
Arg Asp Leu Ser Leu He Ser Pro Leu Ala Gin Ala Val Arg Ser Ser 65 70 75 80 Ser Arg Thr Pro Ser Asp Lys Pro Val Ala His Val Val Ala Asn Pro 85 90 95
Gin Ala Glu Gly Gin Leu Gin Trp Leu Asn Arg Arg Ala Asn Ala Leu 100 105 110
Leu Ala Asn Gly Val Glu Leu Arg Asp Asn Gin Leu Val Val Pro Ser 115 120 125
Glu Gly Leu Tyr Leu He Tyr Ser Gin Val Leu Phe Lys Gly Gin Gly 130 135 140
Cys Pro Ser Thr His Val Leu Leu Thr His Thr He Ser Arg He Ala 145 150 155 160
Val Ser Tyr Gin Thr Lys Val Asn Leu Leu Ser Ala He Lys Ser Pro 165 170 175
Cys Gin Arg Glu Thr Pro Glu Gly Ala Glu Ala Lys Pro Trp Tyr Glu 180 185 190
Pro He Tyr Leu Gly Cys Val Phe Gin Leu Glu Lys Gly Asp Arg Leu 195 200 205
Ser Ala Glu He Asn Arg Pro Asp Tyr Leu Asp Phe Ala Glu Ser Gly 210 215 220
Gin Val Tyr Phe Gly He He Ala Leu 225 230
(2) INFORMATION FOR SEQ ID N0:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 205 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:
Met Thr Pro Pro Glu Arg Leu Phe Leu Pro Arg Val Cys Gly Thr Thr 1 5 10 15
Leu His Leu Leu Leu Leu Gly Leu Leu Leu Val Leu Leu Pro Gly Ala 20 25 30
Gin Gly Leu Pro Gly Val Gly Leu Thr Pro Ser Ala Ala Gin Thr Ala 35 40 45
Arg Gin His Pro Lys Met His Leu Ala His Ser Thr Leu Lys Pro Ala 50 55 60
Ala His Leu He Gly Asp Pro Ser Lys Gin Asn Ser Leu Leu Trp Arg 65 70 75 80
Ala Asn Thr Asp Arg Ala Phe Leu Gin Asp Gly Phe Ser Leu Ser Asn 85 90 95
Asn Ser Leu Leu Val Pro Thr Ser Gly He Tyr Phe Val Tyr Ser Gin 100 105 110
Val Val Phe Ser Cys Lys Ala Tyr Ser Pro Lys Ala Pro Ser Ser Pro 115 120 125 Leu Tyr Leu Ala His Glu Val Cys Leu Phe Ser Ser Gin Tyr Pro Phe 130 135 140
His Val Pro Leu Leu Ser Ser Gin Lys Met Val Tyr Pro Gly Leu Gin 145 150 155 160
Glu Pro Trp Leu His Ser Met Tyr His Gly Ala Ala Phe Gin Leu Thr 165 170 175
Gin Gly Asp Gin Leu Ser Thr His Thr Asp Gly He Pro His Leu Val 180 185 190
Leu Ser Pro Ser Thr Val Phe Phe Gly Ala Phe Ala Leu 195 200 205
(2) INFORMATION FOR SEQ ID NO:7 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: GCGGCGGGAT CCATGGCTAT GATGGAGGTC CAG 33
(2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:8: CGCGCGTCTA GAGCTTAGGC AACTAAAAAG GCC 33
(2) INFORMATION FOR SEQ ID NO:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 34 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
, i) SEQUENCE DESCRIPTION: SEQ ID NO:9: CCGCGCGGAT CCATCATGGC TATGATGGAG GTCC 34
(2) INFORMATION FOR SEQ ID NO:10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic)
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10: CGCGCGTCTA GAGCTTAGCC AACTAAAAAG GCC 33

Claims

WHAT IS CLAIMED IS:
1. An isolated polynucleotide comprising a member selected from the group consisting of:
(a) a polynucleotide having at least a 70% identity to a polynucleotide encoding a polypeptide comprising an amino acid sequence of SEQ ID NO:2;
(b) a polynucleotide which is complementary to the polynucleotide of (a); and
(c) a polynucleotide comprising at least 15 bases of the polynucleotide of (a).
2. The polynucleotide of Claim 1 wherein the polynucleotide is DNA.
3. The polynucleotide of Claim 1 wherein the polynucleotide is RNA.
4. The polynucleotide of Claim 1 wherein the polynucleotide is genomic DNA.
5. The polynucleotide of Claim 2 which encodes the polypeptide comprising amino acids 39 to 281 of SEQ ID NO:2.
6. An isolated polynucleotide comprising a member selected from the group consisting of:
(a) a polynucleotide which encodes a mature polypeptide having the amino acid sequence expressed by the human cDNA contained in ATCC Deposit No. 97448;
(b) a polynucleotide which is complementary to the polynucleotide of (a); and
(c) a polynucleotide comprising at least 15 bases of the polynucleotide of (a) or (b).
7. The polynucleotide of claim 1 comprising the sequence as set forth in SEQ ID No. 1 from nucleotide 1 to nucleotide 1642.
8. The polynucleotide of claim 1 comprising the sequence as set foπh in SEQ ID No. 1 from nucleotide 52 to nucleotide 843.
9. A vector comprising the DNA of Claim 2.
10. A host cell comprising the vector of Claim 9.
11. A process for producing a polypeptide comprising: expressing from the host cell of Claim 10 the polypeptide encoded by said DNA.
12. A process for producing a cell which expresses a polypeptide comprising genetically engineering the cell with the vector of Claim 9.
13. A polypeptide comprising a member selected from the group consisting of:
(a) a polypeptide having an amino acid sequence set foπh in SEQ ID NO:2; and
(b) a polypeptide which is at least 70% identical to the polypeptide of (a).
14. The polypeptide of Claim 13 wherein the polypeptide comprises amino acid 1 to amino acid 281 of SEQ ID NO:2.
15. A compound which inhibits activation of the polypeptide of claim 13.
16. A compound which activates the polypeptide of claim 13.
17. A method for the treatment of a patient having need of AIM-I comprising: administering to the patient a therapeutically effective amount of the polypeptide of claim 13.
18. The method of Claim 17 wherein said therapeutically effective amount of the polypeptide is administered by providing to the patient DNA encoding said polypeptide and expressing said polypeptide in vivo.
19. A method for the treatment of a patient having need to inhibit AIM-I polypeptide comprising: administering to the patient a therapeutically effective amount of the compound of Claim 15.
20. A process for diagnosing a disease or a susceptibility to a disease related to an under-expression of the polypeptide of claim 13 comprising determining a mutation in a nucleic acid sequence encoding said polypeptide.
21. A diagnostic process comprising analyzing for the presence of the polypeptide of claim 13 in a sample derived from a host.
22. A method for identifying compounds which bind to and inhibit activation of the polypeptide of claim 13 comprising: contacting a cell expressing on the surface thereof a receptor for the polypeptide, said receptor being associated with a second component capable of providing a detectable signal in response to the binding of a compound to said receptor, with an analytically detectable AIM-I polypeptide and a compound under conditions to permit binding to the receptor; and
determining whether the compound binds to and inhibits the receptor by detecting the absence of a signal generated from the interaction of the AIM-I with the receptor.
PCT/US1996/003773 1996-03-14 1996-03-14 Apoptosis inducing molecule i WO1997033899A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
PCT/US1996/003773 WO1997033899A1 (en) 1996-03-14 1996-03-14 Apoptosis inducing molecule i
AU57111/96A AU5711196A (en) 1996-03-14 1996-03-14 Apoptosis inducing molecule i

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US1996/003773 WO1997033899A1 (en) 1996-03-14 1996-03-14 Apoptosis inducing molecule i

Publications (1)

Publication Number Publication Date
WO1997033899A1 true WO1997033899A1 (en) 1997-09-18

Family

ID=22254869

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/003773 WO1997033899A1 (en) 1996-03-14 1996-03-14 Apoptosis inducing molecule i

Country Status (2)

Country Link
AU (1) AU5711196A (en)
WO (1) WO1997033899A1 (en)

Cited By (153)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0835305A1 (en) * 1995-06-29 1998-04-15 Immunex Corporation Cytokine that induces apoptosis
WO1999007408A1 (en) * 1997-08-04 1999-02-18 Bio Merieux Protein factor associated with a neuro-degenerative or autoimmune and/or inflammatory disease
US6030945A (en) * 1996-01-09 2000-02-29 Genentech, Inc. Apo-2 ligand
US6046048A (en) * 1996-01-09 2000-04-04 Genetech, Inc. Apo-2 ligand
US6235878B1 (en) 1996-07-19 2001-05-22 Takeda Chemical Industries, Ltd. Fas ligand-like protein, its production and use
US6284236B1 (en) 1995-06-29 2001-09-04 Immunex Corporation Cytokine that induces apoptosis
WO2002002641A1 (en) 2000-06-16 2002-01-10 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
US6346388B1 (en) 1997-08-13 2002-02-12 Smithkline Beecham Corporation Method of identifying agonist and antagonists for tumor necrosis related receptors TR1 and TR2
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
WO2002083704A1 (en) 2001-04-13 2002-10-24 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
WO2002097033A2 (en) 2001-05-25 2002-12-05 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to trail receptors
WO2003060071A2 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
US6635743B1 (en) 1996-03-22 2003-10-21 Human Genome Sciences, Inc. Apoptosis inducing molecule II and methods of use
WO2003086458A1 (en) 2002-04-12 2003-10-23 Medimmune, Inc. Recombinant anti-interleukin-9 antibodies
WO2003100437A1 (en) * 2002-05-29 2003-12-04 Charite - Universitätsmedizin Berlin Method for identifying type-i interferon responsive ms patients by determining trail expression
US6713061B1 (en) 1996-03-12 2004-03-30 Human Genome Sciences, Inc. Death domain containing receptors
US6740739B1 (en) 1998-01-15 2004-05-25 Genentech, Inc. Substitutional variants of APO-2 ligand
US6746668B2 (en) 1996-01-09 2004-06-08 Genentech, Inc. Apo-2 ligand
US6759513B2 (en) 1996-03-12 2004-07-06 Human Genome Sciences, Inc. Death domain containing receptors
WO2005042743A2 (en) 2003-08-18 2005-05-12 Medimmune, Inc. Humanization of antibodies
WO2005077042A2 (en) 2004-02-09 2005-08-25 Human Genome Sciences, Inc. Albumin fusion proteins
WO2005117967A2 (en) 2004-04-12 2005-12-15 Medimmune, Inc. Anti-il-9 antibody formulations and uses thereof
WO2006102095A2 (en) 2005-03-18 2006-09-28 Medimmune, Inc. Framework-shuffling of antibodies
WO2007002543A2 (en) 2005-06-23 2007-01-04 Medimmune, Inc. Antibody formulations having optimized aggregation and fragmentation profiles
WO2007147090A2 (en) 2006-06-14 2007-12-21 Macrogenics, Inc. Methods for the treatment of autoimmune disorders using monoclonal antibodies with reduced toxicity
EP1870464A2 (en) * 1999-06-02 2007-12-26 Genentech, Inc. Methods and compositions for inhibiting neoplastic cell growth
WO2008019199A2 (en) 2006-06-26 2008-02-14 Macrogenics, Inc. FCγRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
US7357927B2 (en) 1996-03-12 2008-04-15 Human Genome Sciences, Inc. Death domain containing receptors
WO2008073312A2 (en) 2006-12-08 2008-06-19 Attenuon, Llc Urokinase-type plasminogen activator receptor epitope, monoclonal antibodies derived therefrom and methods of use thereof
WO2008136694A1 (en) 2007-05-04 2008-11-13 Technophage, Investigação E Desenvolvimento Em Biotecnologia, Sa Engineered rabbit antibody variable domains and uses thereof
WO2008157379A2 (en) 2007-06-21 2008-12-24 Macrogenics, Inc. Covalent diabodies and uses thereof
EP2027874A2 (en) 2000-11-28 2009-02-25 Medimmune, Inc. Methods of administering/dosing anti-rsv antibodies for prophylaxis and treatment
WO2009092011A1 (en) 2008-01-18 2009-07-23 Medimmune, Llc Cysteine engineered antibodies for site-specific conjugation
WO2009118300A1 (en) 2008-03-25 2009-10-01 Novartis Forschungsstiftung Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by down-regulating frizzled-4 and/or frizzled-1
WO2010027364A1 (en) 2008-09-07 2010-03-11 Glyconex Inc. Anti-extended type i glycosphingolipid antibody, derivatives thereof and use
WO2010052288A1 (en) 2008-11-07 2010-05-14 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Teneurin and cancer
EP2206516A1 (en) 2002-06-14 2010-07-14 Medimmune, LLC Stabilized liquid anti-RSV antibody formulations
EP2206720A1 (en) 2000-04-12 2010-07-14 Human Genome Sciences, Inc. Albumin fusion proteins
WO2010080538A1 (en) 2008-12-19 2010-07-15 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2010093993A2 (en) 2009-02-12 2010-08-19 Human Genome Sciences, Inc. Use of b lymphocyte stimulator protein antagonists to promote transplantation tolerance
WO2010100247A1 (en) 2009-03-06 2010-09-10 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Novel therapy for anxiety
EP2241323A1 (en) 2009-04-14 2010-10-20 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Tenascin-W and brain cancers
WO2010120524A2 (en) 2009-03-31 2010-10-21 Altair Therapeutics, Inc. Methods of modulating an immune response to a viral infection
US7855066B1 (en) 1999-06-28 2010-12-21 Genentech, Inc. Methods for making Apo-2 ligand using divalent metal ions
EP2266628A2 (en) 2003-05-13 2010-12-29 Novartis Vaccines and Diagnostics, Inc. Method of determining the susceptibility to bone meatastases by EPhA2 expression
EP2292266A1 (en) 2009-08-27 2011-03-09 Novartis Forschungsstiftung, Zweigniederlassung Treating cancer by modulating copine III
EP2292663A2 (en) 2006-08-28 2011-03-09 Kyowa Hakko Kirin Co., Ltd. Antagonistic human light-specific human monoclonal antibodies
EP2298806A1 (en) 2002-10-16 2011-03-23 Purdue Pharma L.P. Antibodies that bind cell-associated CA 125/0722P and methods of use thereof
WO2011036118A1 (en) 2009-09-22 2011-03-31 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mex-3
WO2011044368A1 (en) 2009-10-07 2011-04-14 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
WO2011045352A2 (en) 2009-10-15 2011-04-21 Novartis Forschungsstiftung Spleen tyrosine kinase and brain cancers
EP2316487A1 (en) 2003-04-11 2011-05-04 MedImmune, LLC Recombinant IL-9 antibodies & uses thereof
WO2011051392A1 (en) 2009-10-30 2011-05-05 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and cancer
EP2332575A1 (en) 2003-12-23 2011-06-15 Genentech, Inc. Treatment of cancer with novel anti-IL 13 monoclonal antibodies
US7964190B2 (en) 1996-03-22 2011-06-21 Human Genome Sciences, Inc. Methods and compositions for decreasing T-cell activity
EP2357192A1 (en) 1999-02-26 2011-08-17 Human Genome Sciences, Inc. Human endokine alpha and methods of use
WO2011107586A1 (en) 2010-03-05 2011-09-09 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research, Smoc1, tenascin-c and brain cancers
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2371389A2 (en) 2002-08-14 2011-10-05 MacroGenics, Inc. FcgammaRIIB-specific antibodies and methods of use thereof
WO2011131611A1 (en) 2010-04-19 2011-10-27 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Modulating xrn1
WO2011154485A1 (en) 2010-06-10 2011-12-15 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mammalian sterile 20-like kinase 3
EP2407548A1 (en) 2006-10-16 2012-01-18 MedImmune, LLC Molecules with reduced half-lives, compositions and uses thereof
WO2012019061A2 (en) 2010-08-05 2012-02-09 Stem Centrx, Inc. Novel effectors and methods of use
WO2012018687A1 (en) 2010-08-02 2012-02-09 Macrogenics, Inc. Covalent diabodies and uses thereof
EP2422811A2 (en) 2004-10-27 2012-02-29 MedImmune, LLC Modulation of antibody specificity by tailoring the affinity to cognate antigens
WO2012027723A1 (en) 2010-08-27 2012-03-01 Stem Centrx, Inc Notum protein modulators and methods of use
WO2012031273A2 (en) 2010-09-03 2012-03-08 Stem Centrx, Inc. Novel modulators and methods of use
WO2012032143A1 (en) 2010-09-10 2012-03-15 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and metastasis
EP2431054A2 (en) 2000-06-15 2012-03-21 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
WO2012058768A1 (en) 2010-11-05 2012-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
WO2012065937A1 (en) 2010-11-15 2012-05-24 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Anti-fungal agents
WO2012078813A2 (en) 2010-12-08 2012-06-14 Stem Centrx, Inc. Novel modulators and methods of use
US8211858B2 (en) 2007-04-27 2012-07-03 The University Of Toledo Modified plasminogen activator inhibitor type-1 molecule and methods based thereon
WO2012103165A2 (en) 2011-01-26 2012-08-02 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
WO2012112943A1 (en) 2011-02-18 2012-08-23 Stem Centrx, Inc. Novel modulators and methods of use
EP2500030A2 (en) 2005-11-04 2012-09-19 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
WO2012162068A2 (en) 2011-05-21 2012-11-29 Macrogenics, Inc. Deimmunized serum-binding domains and their use for extending serum half-life
WO2012168259A1 (en) 2011-06-06 2012-12-13 Novartis Forschungsstiftung, Zweigniederlassung Protein tyrosine phosphatase, non-receptor type 11 (ptpn11) and triple-negative breast cancer
US8388965B2 (en) 2007-10-15 2013-03-05 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2570432A1 (en) 2002-06-14 2013-03-20 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
EP2573114A1 (en) 2005-08-10 2013-03-27 MacroGenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2013043071A1 (en) 2011-09-23 2013-03-28 Technophage, Investigação E Desenvolvimento Em Biotecnologia, Sa Modified albumin-binding domains and uses thereof to improve pharmacokinetics
WO2013063702A1 (en) 2011-11-04 2013-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
WO2013068432A1 (en) 2011-11-08 2013-05-16 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Early diagnostic of neurodegenerative diseases
WO2013068431A1 (en) 2011-11-08 2013-05-16 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
WO2013093809A1 (en) 2011-12-23 2013-06-27 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
EP2610267A1 (en) 2006-12-18 2013-07-03 Genentech, Inc. Antagonist anti-Notch3 antibodies and their use in the prevention and treatment of Notch3-related diseases
WO2013102825A1 (en) 2012-01-02 2013-07-11 Novartis Ag Cdcp1 and breast cancer
EP2626371A1 (en) 2007-07-31 2013-08-14 MedImmune, LLC Multispecific epitope binding proteins and uses thereof
WO2013119960A2 (en) 2012-02-08 2013-08-15 Stem Centrx, Inc. Novel modulators and methods of use
EP2639301A2 (en) 2006-06-30 2013-09-18 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
WO2013144240A1 (en) 2012-03-29 2013-10-03 Friedrich Miescher Institute For Biomedical Research Inhibition of interleukin- 8 and/or its receptor cxcrl in the treatment her2/her3 -overexpressing breast cancer
WO2014001482A1 (en) 2012-06-29 2014-01-03 Novartis Forschungsstiftung, Zweigniererlassung, Friedrich Miescher Institute For Biomedical Research Treating diseases by modulating a specific isoform of mkl1
WO2014006114A1 (en) 2012-07-05 2014-01-09 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
WO2014006115A1 (en) 2012-07-06 2014-01-09 Novartis Ag Combination of a phosphoinositide 3-kinase inhibitor and an inhibitor of the il-8/cxcr interaction
US8637026B2 (en) 2007-12-26 2014-01-28 Vaccinex, Inc. Anti-C35 antibody combination therapies and methods
WO2014018625A1 (en) 2012-07-25 2014-01-30 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
US8647622B2 (en) 2007-08-29 2014-02-11 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
EP2703007A1 (en) 2007-03-30 2014-03-05 MedImmune, LLC Antibodies with decreased deamidation profiles
EP2711018A1 (en) 2009-06-22 2014-03-26 MedImmune, LLC Engineered Fc regions for site-specific conjugation
WO2014059028A1 (en) 2012-10-09 2014-04-17 Igenica, Inc. Anti-c16orf54 antibodies and methods of use thereof
WO2014082179A1 (en) 2012-11-28 2014-06-05 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
EP2769737A1 (en) 2009-07-20 2014-08-27 Bristol-Myers Squibb Company Combination of an anti-CTLA4 antibody with etoposide for the synergistic treatment of proliferative diseases
US8852608B2 (en) 2009-02-02 2014-10-07 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
WO2014197849A2 (en) 2013-06-06 2014-12-11 Igenica Biotherapeutics, Inc. Anti-c10orf54 antibodies and uses thereof
US8937169B2 (en) 1996-01-11 2015-01-20 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
WO2015019286A1 (en) 2013-08-07 2015-02-12 Friedrich Miescher Institute For Biomedical Research New screening method for the treatment friedreich's ataxia
US8986972B2 (en) 2012-02-24 2015-03-24 Stem Centrx, Inc. Nucleic acid encoding DLL3 antibodies
US9133273B2 (en) 2010-11-19 2015-09-15 Eisai R&D Management Co., Ltd. Nucleic acids encoding neutralizing anti-CCL20 antibodies
WO2015175375A1 (en) 2014-05-13 2015-11-19 Short Jay M Conditionally active biological proteins
WO2015181805A1 (en) 2014-05-28 2015-12-03 Zymeworks Inc. Modified antigen binding polypeptide constructs and uses thereof
WO2015189816A1 (en) 2014-06-13 2015-12-17 Friedrich Miescher Institute For Biomedical Research New treatment against influenza virus
WO2015198202A1 (en) 2014-06-23 2015-12-30 Friedrich Miescher Institute For Biomedical Research Methods for triggering de novo formation of heterochromatin and or epigenetic silencing with small rnas
WO2016001830A1 (en) 2014-07-01 2016-01-07 Friedrich Miescher Institute For Biomedical Research Combination of a brafv600e inhibitor and mertk inhibitor to treat melanoma
US9244074B2 (en) 2011-06-07 2016-01-26 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
WO2016046768A1 (en) 2014-09-24 2016-03-31 Friedrich Miescher Institute For Biomedical Research Lats and breast cancer
EP3037544A1 (en) 2005-10-13 2016-06-29 Human Genome Sciences, Inc. Methods and compositions for use in treatment of systemic lupus erythematosus (sle) patients with autoantibody positive diseases
WO2016139482A1 (en) 2015-03-03 2016-09-09 Kymab Limited Antibodies, uses & methods
EP3073267A1 (en) 2004-09-21 2016-09-28 Medimmune, Inc. Antibodies against and methods for producing vaccines for respiratory syncytial virus
WO2016166304A1 (en) 2015-04-15 2016-10-20 Van Berkel Patricius Hendrikus Cornelis Site-specific antibody-drug conjugates
US9505823B2 (en) 2006-08-07 2016-11-29 TEV A Biopharmaceuticals USA, Inc. Albumin-insulin fusion proteins
US9561274B2 (en) 2011-06-07 2017-02-07 University Of Hawaii Treatment and prevention of cancer with HMGB1 antagonists
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
WO2017059551A1 (en) 2015-10-08 2017-04-13 Zymeworks Inc. Antigen-binding polypeptide constructs comprising kappa and lambda light chains and uses thereof
WO2017072669A1 (en) 2015-10-28 2017-05-04 Friedrich Miescher Institute For Biomedical Research Tenascin-w and biliary tract cancers
WO2017096051A1 (en) 2015-12-02 2017-06-08 Stcube & Co., Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
WO2018083248A1 (en) 2016-11-03 2018-05-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
US9968687B2 (en) 2013-02-22 2018-05-15 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
EP3338793A1 (en) 2013-08-28 2018-06-27 AbbVie Stemcentrx LLC Novel sez6 modulators and methods of use
US10017580B2 (en) 2014-04-15 2018-07-10 ADC Therpeutics S.A. Humanized anti-Tn-MUC1 antibodies and their conjugates
US10035853B2 (en) 2013-08-28 2018-07-31 Abbvie Stemcentrx Llc Site-specific antibody conjugation methods and compositions
WO2018222685A1 (en) 2017-05-31 2018-12-06 Stcube & Co., Inc. Methods of treating cancer using antibodies and molecules that immunospecifically bind to btn1a1
WO2018222689A1 (en) 2017-05-31 2018-12-06 Stcube & Co., Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
WO2018226671A1 (en) 2017-06-06 2018-12-13 Stcube & Co., Inc. Methods of treating cancer using antibodies and molecules that bind to btn1a1 or btn1a1-ligands
EP3479844A1 (en) 2005-04-15 2019-05-08 MacroGenics, Inc. Covalent diabodies and uses thereof
WO2019102435A1 (en) 2017-11-27 2019-05-31 Euro-Celtique S.A. Humanized antibodies targeting human tissue factor
US10308721B2 (en) 2014-02-21 2019-06-04 Abbvie Stemcentrx Llc Anti-DLL3 antibodies and drug conjugates for use in melanoma
EP3502256A2 (en) 2008-09-26 2019-06-26 Tocagen Inc. Recombinant vectors
US10513699B2 (en) 2014-09-03 2019-12-24 Bioatla, Llc Discovering and producing conditionally active biologic proteins in the same eukaryotic cell production hosts
EP3590539A1 (en) 2014-03-04 2020-01-08 Kymab Limited Antibodies, uses & methods
WO2020016459A1 (en) 2018-07-20 2020-01-23 Pierre Fabre Medicament Receptor for vista
US10563194B2 (en) 2015-02-24 2020-02-18 BioAlta, LLC Conditionally active biological proteins
EP3623478A1 (en) 2012-10-25 2020-03-18 Tocagen Inc. Retroviral vector with mini-promoter cassette
US10766959B2 (en) 2014-12-11 2020-09-08 Pierre Fabre Medicament Anti-C10ORF54 antibodies and uses thereof
WO2020198731A2 (en) 2019-03-28 2020-10-01 Danisco Us Inc Engineered antibodies
EP3738981A1 (en) 2014-01-24 2020-11-18 NGM Biopharmaceuticals, Inc. Antibodies binding beta klotho domain 2 and methods of use thereof
WO2020242989A1 (en) 2019-05-24 2020-12-03 Sanofi Methods for treating systemic sclerosis
US11008389B2 (en) 2011-03-16 2021-05-18 Sanofi Uses of a dual V region antibody-like protein
EP3888690A2 (en) 2014-05-16 2021-10-06 MedImmune, LLC Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
WO2021202473A2 (en) 2020-03-30 2021-10-07 Danisco Us Inc Engineered antibodies
US11253590B2 (en) 2015-12-02 2022-02-22 Stsciences, Inc. Antibodies specific to glycosylated BTLA (B- and T- lymphocyte attenuator)
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
WO2024015953A1 (en) 2022-07-15 2024-01-18 Danisco Us Inc. Methods for producing monoclonal antibodies

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CELL, 17 December 1993, Vol. 75, SUDA et al., "Molecular Cloning and Expression of the Fas Ligand, a Novel Member of the Tumor Necrosis Factor Family", pages 1169-1178. *
THE JOURNAL OF IMMUNOLOGY, 1995, Vol. 154, CASCINO et al., "Three Functional Soluble Forms of the Human Apoptosis-Inducing Fas Molecule Are Produced by Alternative Splicing", pages 2706-2713. *
THE JOURNAL OF IMMUNOLOGY, 1996, Vol. 156, PAPOFF et al., "An N-Terminal Domain Shared by Fas/Apo-1 (CD95) Soluble Variants Prevents Cell Death in Vitro", pages 4622-4630. *

Cited By (284)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0835305A4 (en) * 1995-06-29 2002-02-06 Immunex Corp Cytokine that induces apoptosis
US7736637B2 (en) 1995-06-29 2010-06-15 Immunex Corporation Use of TRAIL polypeptides to induce apoptosis
US6521228B1 (en) 1995-06-29 2003-02-18 Immunex Corporation Antibodies directed against trail
US7972812B2 (en) 1995-06-29 2011-07-05 Immunex Corporation DNA encoding a soluble fragment of TRAIL polypeptide
EP0835305A1 (en) * 1995-06-29 1998-04-15 Immunex Corporation Cytokine that induces apoptosis
US6284236B1 (en) 1995-06-29 2001-09-04 Immunex Corporation Cytokine that induces apoptosis
EP1666591A1 (en) * 1995-06-29 2006-06-07 Immunex Corporation Cytokine that induces apoptosis
US7285533B2 (en) 1996-01-09 2007-10-23 Genentech, Inc. Apo-2 ligand
US6998116B1 (en) 1996-01-09 2006-02-14 Genentech, Inc. Apo-2 ligand
US6746668B2 (en) 1996-01-09 2004-06-08 Genentech, Inc. Apo-2 ligand
US6046048A (en) * 1996-01-09 2000-04-04 Genetech, Inc. Apo-2 ligand
US6030945A (en) * 1996-01-09 2000-02-29 Genentech, Inc. Apo-2 ligand
US8937169B2 (en) 1996-01-11 2015-01-20 Human Genome Sciences, Inc. Human G-protein chemokine receptor HSATU68
US8105589B2 (en) 1996-03-12 2012-01-31 Human Genome Sciences, Inc. Use of DR3 antibodies in the treatment of inflammatory disease
US6713061B1 (en) 1996-03-12 2004-03-30 Human Genome Sciences, Inc. Death domain containing receptors
US7357927B2 (en) 1996-03-12 2008-04-15 Human Genome Sciences, Inc. Death domain containing receptors
US7708996B2 (en) 1996-03-12 2010-05-04 Human Genome Sciences, Inc. DR3 antibodies
US6759513B2 (en) 1996-03-12 2004-07-06 Human Genome Sciences, Inc. Death domain containing receptors
US6635743B1 (en) 1996-03-22 2003-10-21 Human Genome Sciences, Inc. Apoptosis inducing molecule II and methods of use
US7964190B2 (en) 1996-03-22 2011-06-21 Human Genome Sciences, Inc. Methods and compositions for decreasing T-cell activity
US6235878B1 (en) 1996-07-19 2001-05-22 Takeda Chemical Industries, Ltd. Fas ligand-like protein, its production and use
WO1999007408A1 (en) * 1997-08-04 1999-02-18 Bio Merieux Protein factor associated with a neuro-degenerative or autoimmune and/or inflammatory disease
US6346388B1 (en) 1997-08-13 2002-02-12 Smithkline Beecham Corporation Method of identifying agonist and antagonists for tumor necrosis related receptors TR1 and TR2
US6740739B1 (en) 1998-01-15 2004-05-25 Genentech, Inc. Substitutional variants of APO-2 ligand
EP2357192A1 (en) 1999-02-26 2011-08-17 Human Genome Sciences, Inc. Human endokine alpha and methods of use
EP1870464A2 (en) * 1999-06-02 2007-12-26 Genentech, Inc. Methods and compositions for inhibiting neoplastic cell growth
EP1870464A3 (en) * 1999-06-02 2008-03-12 Genentech, Inc. Methods and compositions for inhibiting neoplastic cell growth
EP2339003A2 (en) 1999-06-28 2011-06-29 Genentech, Inc. APO-2 ligand substitutional variants
US7855066B1 (en) 1999-06-28 2010-12-21 Genentech, Inc. Methods for making Apo-2 ligand using divalent metal ions
EP2311956A1 (en) 1999-06-28 2011-04-20 Genentech, Inc. Methods for making APO-2 ligand using divalent metal ions
EP2295456A1 (en) 2000-04-12 2011-03-16 Human Genome Sciences, Inc. Albumin fusion proteins
EP2236152A1 (en) 2000-04-12 2010-10-06 Human Genome Sciences, Inc. Albumin fusion proteins
EP2298355A2 (en) 2000-04-12 2011-03-23 Human Genome Sciences, Inc. Albumin fusion proteins
EP2275557A1 (en) 2000-04-12 2011-01-19 Human Genome Sciences, Inc. Albumin fusion proteins
EP2357008A1 (en) 2000-04-12 2011-08-17 Human Genome Sciences, Inc. Albumin fusion proteins
EP2311872A1 (en) 2000-04-12 2011-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
EP2213743A1 (en) 2000-04-12 2010-08-04 Human Genome Sciences, Inc. Albumin fusion proteins
EP2267026A1 (en) 2000-04-12 2010-12-29 Human Genome Sciences, Inc. Albumin fusion proteins
EP2206720A1 (en) 2000-04-12 2010-07-14 Human Genome Sciences, Inc. Albumin fusion proteins
EP2216409A1 (en) 2000-04-12 2010-08-11 Human Genome Sciences, Inc. Albumin fusion proteins
EP2431054A2 (en) 2000-06-15 2012-03-21 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
EP2275449A1 (en) 2000-06-16 2011-01-19 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
EP2281843A1 (en) 2000-06-16 2011-02-09 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
EP2281842A1 (en) 2000-06-16 2011-02-09 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to BLyS
WO2002002641A1 (en) 2000-06-16 2002-01-10 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to blys
EP2412384A1 (en) 2000-11-28 2012-02-01 MedImmune, LLC Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
EP2027874A2 (en) 2000-11-28 2009-02-25 Medimmune, Inc. Methods of administering/dosing anti-rsv antibodies for prophylaxis and treatment
EP2338512A1 (en) 2000-11-28 2011-06-29 MedImmune, LLC Methods of administering/dosing anti-RSV antibodies for prophylaxis and treatment
EP3569610A2 (en) 2000-12-12 2019-11-20 Medlmmune, LLC Molecules with extended half lives, compositions and uses thereof
EP2354149A1 (en) 2000-12-12 2011-08-10 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
EP2357187A1 (en) 2000-12-12 2011-08-17 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
EP2341060A1 (en) 2000-12-12 2011-07-06 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
WO2002083704A1 (en) 2001-04-13 2002-10-24 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
EP2228389A2 (en) 2001-04-13 2010-09-15 Human Genome Sciences, Inc. Antibodies against vascular endothelial growth factor 2
WO2002097033A2 (en) 2001-05-25 2002-12-05 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to trail receptors
EP2277910A1 (en) 2001-12-21 2011-01-26 Human Genome Sciences, Inc. Albumin fusion proteins
EP2277888A2 (en) 2001-12-21 2011-01-26 Human Genome Sciences, Inc. Fusion proteins of albumin and erythropoietin
EP2261250A1 (en) 2001-12-21 2010-12-15 Human Genome Sciences, Inc. Albumin fusion proteins
WO2003060071A2 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
EP2990417A1 (en) 2001-12-21 2016-03-02 Human Genome Sciences, Inc. Albumin insulin fusion protein
EP1997829A1 (en) 2001-12-21 2008-12-03 Human Genome Sciences, Inc. Albumin fusion proteins
EP2277889A2 (en) 2001-12-21 2011-01-26 Human Genome Sciences, Inc. Fusion proteins of albumin and interferon beta
WO2003086458A1 (en) 2002-04-12 2003-10-23 Medimmune, Inc. Recombinant anti-interleukin-9 antibodies
EP2270049A2 (en) 2002-04-12 2011-01-05 Medimmune, Inc. Recombinant anti-interleukin-9-antibody
EP1369695A1 (en) * 2002-05-29 2003-12-10 Universitätsklinikum Charité der Humboldt- Universität zu Berlin Method for identifying type-I interferon responsive MS patients by determining TRAIL expression
WO2003100437A1 (en) * 2002-05-29 2003-12-04 Charite - Universitätsmedizin Berlin Method for identifying type-i interferon responsive ms patients by determining trail expression
EP2570432A1 (en) 2002-06-14 2013-03-20 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
EP2206516A1 (en) 2002-06-14 2010-07-14 Medimmune, LLC Stabilized liquid anti-RSV antibody formulations
EP2327421A1 (en) 2002-06-14 2011-06-01 MedImmune, LLC Stabilized liquid anti-RSV antibody formulations
EP2371389A2 (en) 2002-08-14 2011-10-05 MacroGenics, Inc. FcgammaRIIB-specific antibodies and methods of use thereof
EP2298806A1 (en) 2002-10-16 2011-03-23 Purdue Pharma L.P. Antibodies that bind cell-associated CA 125/0722P and methods of use thereof
EP2891666A1 (en) 2002-10-16 2015-07-08 Purdue Pharma L.P. Antibodies that bind cell-associated CA 125/O722P and methods of use thereof
EP3301114A1 (en) 2002-10-16 2018-04-04 Purdue Pharma LP Fusion polypeptides of antibodies that bind cell-associated ca 125/o722p
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2316487A1 (en) 2003-04-11 2011-05-04 MedImmune, LLC Recombinant IL-9 antibodies & uses thereof
EP2266628A2 (en) 2003-05-13 2010-12-29 Novartis Vaccines and Diagnostics, Inc. Method of determining the susceptibility to bone meatastases by EPhA2 expression
WO2005042743A2 (en) 2003-08-18 2005-05-12 Medimmune, Inc. Humanization of antibodies
EP2272566A2 (en) 2003-08-18 2011-01-12 MedImmune, LLC Humanisation of antibodies
EP2351584A1 (en) 2003-12-23 2011-08-03 Genentech, Inc. Novel anti-IL 13 antibodies and uses thereof
EP3718564A1 (en) 2003-12-23 2020-10-07 Genentech, Inc. Novel anti-il 13 antibodies and uses thereof
EP2805728A1 (en) 2003-12-23 2014-11-26 Genentech, Inc. Novel anti-IL 13 antibodies and uses thereof
EP2332575A1 (en) 2003-12-23 2011-06-15 Genentech, Inc. Treatment of cancer with novel anti-IL 13 monoclonal antibodies
WO2005077042A2 (en) 2004-02-09 2005-08-25 Human Genome Sciences, Inc. Albumin fusion proteins
WO2005117967A2 (en) 2004-04-12 2005-12-15 Medimmune, Inc. Anti-il-9 antibody formulations and uses thereof
EP3073267A1 (en) 2004-09-21 2016-09-28 Medimmune, Inc. Antibodies against and methods for producing vaccines for respiratory syncytial virus
EP2422811A2 (en) 2004-10-27 2012-02-29 MedImmune, LLC Modulation of antibody specificity by tailoring the affinity to cognate antigens
WO2006102095A2 (en) 2005-03-18 2006-09-28 Medimmune, Inc. Framework-shuffling of antibodies
EP3479844A1 (en) 2005-04-15 2019-05-08 MacroGenics, Inc. Covalent diabodies and uses thereof
WO2007002543A2 (en) 2005-06-23 2007-01-04 Medimmune, Inc. Antibody formulations having optimized aggregation and fragmentation profiles
EP2573114A1 (en) 2005-08-10 2013-03-27 MacroGenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP3037544A1 (en) 2005-10-13 2016-06-29 Human Genome Sciences, Inc. Methods and compositions for use in treatment of systemic lupus erythematosus (sle) patients with autoantibody positive diseases
EP2500030A2 (en) 2005-11-04 2012-09-19 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
EP2998318A1 (en) 2005-11-04 2016-03-23 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
EP3299027A1 (en) 2005-11-04 2018-03-28 Genentech, Inc. Use of complement pathway inhibitors to treat ocular diseases
EP2815764A1 (en) 2006-06-14 2014-12-24 Macrogenics, Inc. Methods for the treatment of autoimmune disorders using monoclonal antibodies with reduced toxicity
WO2007147090A2 (en) 2006-06-14 2007-12-21 Macrogenics, Inc. Methods for the treatment of autoimmune disorders using monoclonal antibodies with reduced toxicity
EP2505209A1 (en) 2006-06-26 2012-10-03 MacroGenics, Inc. Fcgamma-RIIB-specific antibodies and methods of the use thereof
WO2008019199A2 (en) 2006-06-26 2008-02-14 Macrogenics, Inc. FCγRIIB-SPECIFIC ANTIBODIES AND METHODS OF USE THEREOF
EP2671946A1 (en) 2006-06-30 2013-12-11 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
EP2639301A2 (en) 2006-06-30 2013-09-18 Bristol-Myers Squibb Company Polynucleotides encoding novel PCSK9 variants
US9505823B2 (en) 2006-08-07 2016-11-29 TEV A Biopharmaceuticals USA, Inc. Albumin-insulin fusion proteins
EP2484696A1 (en) 2006-08-28 2012-08-08 Kyowa Hakko Kirin Co., Ltd. Antagonistic human light-specific human monoclonal antibodies
EP2292663A2 (en) 2006-08-28 2011-03-09 Kyowa Hakko Kirin Co., Ltd. Antagonistic human light-specific human monoclonal antibodies
EP2407548A1 (en) 2006-10-16 2012-01-18 MedImmune, LLC Molecules with reduced half-lives, compositions and uses thereof
WO2008073312A2 (en) 2006-12-08 2008-06-19 Attenuon, Llc Urokinase-type plasminogen activator receptor epitope, monoclonal antibodies derived therefrom and methods of use thereof
EP2610267A1 (en) 2006-12-18 2013-07-03 Genentech, Inc. Antagonist anti-Notch3 antibodies and their use in the prevention and treatment of Notch3-related diseases
EP2703007A1 (en) 2007-03-30 2014-03-05 MedImmune, LLC Antibodies with decreased deamidation profiles
US8211858B2 (en) 2007-04-27 2012-07-03 The University Of Toledo Modified plasminogen activator inhibitor type-1 molecule and methods based thereon
WO2008136694A1 (en) 2007-05-04 2008-11-13 Technophage, Investigação E Desenvolvimento Em Biotecnologia, Sa Engineered rabbit antibody variable domains and uses thereof
WO2008157379A2 (en) 2007-06-21 2008-12-24 Macrogenics, Inc. Covalent diabodies and uses thereof
EP3424951A1 (en) 2007-06-21 2019-01-09 MacroGenics, Inc. Covalent diabodies and uses thereof
EP2626371A1 (en) 2007-07-31 2013-08-14 MedImmune, LLC Multispecific epitope binding proteins and uses thereof
US9815902B2 (en) 2007-08-29 2017-11-14 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
US9175087B2 (en) 2007-08-29 2015-11-03 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9228019B2 (en) 2007-08-29 2016-01-05 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9243067B2 (en) 2007-08-29 2016-01-26 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US8647622B2 (en) 2007-08-29 2014-02-11 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US8980262B2 (en) 2007-08-29 2015-03-17 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9732162B2 (en) 2007-10-15 2017-08-15 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2574630A1 (en) 2007-10-15 2013-04-03 Sanofi Antibodies that bind il-4 and/or il-13 and their uses
US11453727B2 (en) 2007-10-15 2022-09-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US8388965B2 (en) 2007-10-15 2013-03-05 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US10759871B2 (en) 2007-10-15 2020-09-01 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2573116A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2573118A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2573115A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2573117A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP3686220A1 (en) 2007-10-15 2020-07-29 Sanofi Antibodies that bind il-4 and/or il-13 and their uses
EP2573121A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind il-4 and/or il-13 and their uses
EP2573119A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US9738728B2 (en) 2007-10-15 2017-08-22 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2574629A1 (en) 2007-10-15 2013-04-03 Sanofi Antibodies that bind il-4 and/or il-13 and their uses
EP2574626A1 (en) 2007-10-15 2013-04-03 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US8637026B2 (en) 2007-12-26 2014-01-28 Vaccinex, Inc. Anti-C35 antibody combination therapies and methods
WO2009092011A1 (en) 2008-01-18 2009-07-23 Medimmune, Llc Cysteine engineered antibodies for site-specific conjugation
WO2009118300A1 (en) 2008-03-25 2009-10-01 Novartis Forschungsstiftung Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by down-regulating frizzled-4 and/or frizzled-1
WO2010027364A1 (en) 2008-09-07 2010-03-11 Glyconex Inc. Anti-extended type i glycosphingolipid antibody, derivatives thereof and use
EP3502256A2 (en) 2008-09-26 2019-06-26 Tocagen Inc. Recombinant vectors
WO2010052288A1 (en) 2008-11-07 2010-05-14 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Teneurin and cancer
EP3482769A1 (en) 2008-12-19 2019-05-15 MacroGenics, Inc. Covalent diabodies and uses thereof
WO2010080538A1 (en) 2008-12-19 2010-07-15 Macrogenics, Inc. Covalent diabodies and uses thereof
EP2786762A2 (en) 2008-12-19 2014-10-08 MacroGenics, Inc. Covalent diabodies and uses thereof
US9499590B2 (en) 2009-02-02 2016-11-22 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
US8852608B2 (en) 2009-02-02 2014-10-07 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
WO2010093993A2 (en) 2009-02-12 2010-08-19 Human Genome Sciences, Inc. Use of b lymphocyte stimulator protein antagonists to promote transplantation tolerance
WO2010100247A1 (en) 2009-03-06 2010-09-10 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Novel therapy for anxiety
WO2010120511A2 (en) 2009-03-31 2010-10-21 Altair Therapeutics, Inc. Method of treating respiratory disorders
WO2010120524A2 (en) 2009-03-31 2010-10-21 Altair Therapeutics, Inc. Methods of modulating an immune response to a viral infection
EP2241323A1 (en) 2009-04-14 2010-10-20 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Tenascin-W and brain cancers
EP2711018A1 (en) 2009-06-22 2014-03-26 MedImmune, LLC Engineered Fc regions for site-specific conjugation
EP2769737A1 (en) 2009-07-20 2014-08-27 Bristol-Myers Squibb Company Combination of an anti-CTLA4 antibody with etoposide for the synergistic treatment of proliferative diseases
EP3659596A1 (en) 2009-07-20 2020-06-03 Bristol-Myers Squibb Company Combination of anti-ctla4 antibody with paclitaxel for the synergistic treatment of proliferative diseases
EP2947098A1 (en) 2009-07-20 2015-11-25 Bristol-Myers Squibb Company Combination of anti-ctla4 antibody with gemcitabine for the synergistic treatment of proliferative diseases
EP2292266A1 (en) 2009-08-27 2011-03-09 Novartis Forschungsstiftung, Zweigniederlassung Treating cancer by modulating copine III
WO2011036118A1 (en) 2009-09-22 2011-03-31 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mex-3
US9096877B2 (en) 2009-10-07 2015-08-04 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
WO2011044368A1 (en) 2009-10-07 2011-04-14 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
WO2011045352A2 (en) 2009-10-15 2011-04-21 Novartis Forschungsstiftung Spleen tyrosine kinase and brain cancers
WO2011051392A1 (en) 2009-10-30 2011-05-05 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and cancer
WO2011107586A1 (en) 2010-03-05 2011-09-09 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research, Smoc1, tenascin-c and brain cancers
WO2011131611A1 (en) 2010-04-19 2011-10-27 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Modulating xrn1
WO2011154485A1 (en) 2010-06-10 2011-12-15 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Treating cancer by modulating mammalian sterile 20-like kinase 3
WO2012018687A1 (en) 2010-08-02 2012-02-09 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2012019061A2 (en) 2010-08-05 2012-02-09 Stem Centrx, Inc. Novel effectors and methods of use
WO2012027723A1 (en) 2010-08-27 2012-03-01 Stem Centrx, Inc Notum protein modulators and methods of use
WO2012031273A2 (en) 2010-09-03 2012-03-08 Stem Centrx, Inc. Novel modulators and methods of use
WO2012032143A1 (en) 2010-09-10 2012-03-15 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Phosphorylated twist1 and metastasis
WO2012058768A1 (en) 2010-11-05 2012-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
WO2012065937A1 (en) 2010-11-15 2012-05-24 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Anti-fungal agents
US9133273B2 (en) 2010-11-19 2015-09-15 Eisai R&D Management Co., Ltd. Nucleic acids encoding neutralizing anti-CCL20 antibodies
US9809647B2 (en) 2010-11-19 2017-11-07 Eisai R&D Management Co., Ltd. Neutralizing anti-CCL20 antibodies
WO2012078813A2 (en) 2010-12-08 2012-06-14 Stem Centrx, Inc. Novel modulators and methods of use
WO2012118547A1 (en) 2010-12-08 2012-09-07 Stem Centrx, Inc. Novel modulators and methods of use
WO2012103165A2 (en) 2011-01-26 2012-08-02 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
EP3763740A1 (en) 2011-01-26 2021-01-13 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
WO2012112943A1 (en) 2011-02-18 2012-08-23 Stem Centrx, Inc. Novel modulators and methods of use
EP3763388A1 (en) 2011-02-18 2021-01-13 AbbVie Stemcentrx LLC Novel modulators and methods of use
US11008389B2 (en) 2011-03-16 2021-05-18 Sanofi Uses of a dual V region antibody-like protein
WO2012162068A2 (en) 2011-05-21 2012-11-29 Macrogenics, Inc. Deimmunized serum-binding domains and their use for extending serum half-life
US9181553B2 (en) 2011-06-06 2015-11-10 Novartis Forschungsstiftung Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Method of treatment of breast cancers over-expressing the SHP2 signature genes
WO2012168259A1 (en) 2011-06-06 2012-12-13 Novartis Forschungsstiftung, Zweigniederlassung Protein tyrosine phosphatase, non-receptor type 11 (ptpn11) and triple-negative breast cancer
US9561274B2 (en) 2011-06-07 2017-02-07 University Of Hawaii Treatment and prevention of cancer with HMGB1 antagonists
US9244074B2 (en) 2011-06-07 2016-01-26 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
WO2013043071A1 (en) 2011-09-23 2013-03-28 Technophage, Investigação E Desenvolvimento Em Biotecnologia, Sa Modified albumin-binding domains and uses thereof to improve pharmacokinetics
WO2013063702A1 (en) 2011-11-04 2013-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
WO2013068431A1 (en) 2011-11-08 2013-05-16 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
WO2013068432A1 (en) 2011-11-08 2013-05-16 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Early diagnostic of neurodegenerative diseases
WO2013093809A1 (en) 2011-12-23 2013-06-27 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
WO2013102825A1 (en) 2012-01-02 2013-07-11 Novartis Ag Cdcp1 and breast cancer
WO2013119960A2 (en) 2012-02-08 2013-08-15 Stem Centrx, Inc. Novel modulators and methods of use
US9089617B2 (en) 2012-02-24 2015-07-28 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates
US9345784B1 (en) 2012-02-24 2016-05-24 Stemcentrx, Inc. Methods of delivering DLL3 antibody drug conjugates
US9352051B1 (en) 2012-02-24 2016-05-31 Stemcentrx, Inc. Kits containing DLL3 antibody drug conjugates
US9353182B2 (en) 2012-02-24 2016-05-31 Stemcentrx, Inc. Anti-DLL3 antibodies
US9358304B1 (en) 2012-02-24 2016-06-07 Stemcentrx, Inc. Methods of making DLL3 antibody drug conjugates
US9334318B1 (en) 2012-02-24 2016-05-10 Stemcentrx, Inc. Multivalent DLL3 antibodies
US10137204B2 (en) 2012-02-24 2018-11-27 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates for treating cancer
US9173959B1 (en) 2012-02-24 2015-11-03 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates
US9937268B2 (en) 2012-02-24 2018-04-10 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates and methods of use
US9481727B2 (en) 2012-02-24 2016-11-01 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US9480757B2 (en) 2012-02-24 2016-11-01 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US9486537B2 (en) 2012-02-24 2016-11-08 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US9155803B1 (en) 2012-02-24 2015-10-13 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates and methods of use
EP3095797A1 (en) 2012-02-24 2016-11-23 Stemcentrx, Inc. Anti dll3 antibodies and methods of use thereof
US9133271B1 (en) 2012-02-24 2015-09-15 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates and methods of use
US9107961B2 (en) 2012-02-24 2015-08-18 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates for treating cancer
US9089615B2 (en) 2012-02-24 2015-07-28 Stemcentrx, Inc. Anti-DLL3 antibodies
US9931420B2 (en) 2012-02-24 2018-04-03 Abbvie Stemcentrx Llc Methods of making DLL3 antibody drug conjugates
US9931421B2 (en) 2012-02-24 2018-04-03 Abbvie Stemcentrx Llc Methods of delivering DLL3 antibody drug conjugates
US9089616B2 (en) 2012-02-24 2015-07-28 Stemcentrx, Inc. Anti-DLL3 antibody drug conjugates and methods of use
US9090683B2 (en) 2012-02-24 2015-07-28 Stemcentrx, Inc. Methods of detection, diagnosis, and monitoring using anti-DLL3 antibodies
US9878053B2 (en) 2012-02-24 2018-01-30 Abbvie Stemcentrx Llc Methods of delivering DLL3 antibody drug conjugates
US9764042B1 (en) 2012-02-24 2017-09-19 Abbvie Stemcentrx Llc Methods of making DLL3 antibody drug conjugates
US9770518B1 (en) 2012-02-24 2017-09-26 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US9775916B1 (en) 2012-02-24 2017-10-03 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates for treating cancer
US8986972B2 (en) 2012-02-24 2015-03-24 Stem Centrx, Inc. Nucleic acid encoding DLL3 antibodies
US11033634B2 (en) 2012-02-24 2021-06-15 Abbvie Stemcentrx Llc Light chain variable regions
US9855343B2 (en) 2012-02-24 2018-01-02 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US9861708B2 (en) 2012-02-24 2018-01-09 Abbvie Stemcentrx Llc Kits containing DLL3 antibody drug conjugates
US9867887B1 (en) 2012-02-24 2018-01-16 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US10525130B2 (en) 2012-03-26 2020-01-07 Sanofi Stable IGG4 based binding agent formulations
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
WO2013144240A1 (en) 2012-03-29 2013-10-03 Friedrich Miescher Institute For Biomedical Research Inhibition of interleukin- 8 and/or its receptor cxcrl in the treatment her2/her3 -overexpressing breast cancer
WO2014001482A1 (en) 2012-06-29 2014-01-03 Novartis Forschungsstiftung, Zweigniererlassung, Friedrich Miescher Institute For Biomedical Research Treating diseases by modulating a specific isoform of mkl1
WO2014006114A1 (en) 2012-07-05 2014-01-09 Novartis Forschungsstiftung, Zweigniederlassung Friedrich Miescher Institute For Biomedical Research New treatment for neurodegenerative diseases
WO2014006115A1 (en) 2012-07-06 2014-01-09 Novartis Ag Combination of a phosphoinositide 3-kinase inhibitor and an inhibitor of the il-8/cxcr interaction
EP3381943A1 (en) 2012-07-25 2018-10-03 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
EP4063391A1 (en) 2012-07-25 2022-09-28 Celldex Therapeutics, Inc. Anti-kit antibodies and uses thereof
WO2014018625A1 (en) 2012-07-25 2014-01-30 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
WO2014059028A1 (en) 2012-10-09 2014-04-17 Igenica, Inc. Anti-c16orf54 antibodies and methods of use thereof
EP3623478A1 (en) 2012-10-25 2020-03-18 Tocagen Inc. Retroviral vector with mini-promoter cassette
WO2014082179A1 (en) 2012-11-28 2014-06-05 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US10478509B2 (en) 2013-02-22 2019-11-19 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates for treating cancer
US9968687B2 (en) 2013-02-22 2018-05-15 Abbvie Stemcentrx Llc Anti-DLL3 antibody drug conjugates
US10100123B2 (en) 2013-06-06 2018-10-16 Pierre Fabre Medicament Anti-C10orf54 antibodies and uses thereof
US10421818B2 (en) 2013-06-06 2019-09-24 Pierre Fabre Medicament Anti-C10orf54 antibodies and uses thereof
US10414823B2 (en) 2013-06-06 2019-09-17 Pierre Fabre Medicament Anti-C10orf54 antibodies and uses thereof
WO2014197849A2 (en) 2013-06-06 2014-12-11 Igenica Biotherapeutics, Inc. Anti-c10orf54 antibodies and uses thereof
WO2015019286A1 (en) 2013-08-07 2015-02-12 Friedrich Miescher Institute For Biomedical Research New screening method for the treatment friedreich's ataxia
EP3338793A1 (en) 2013-08-28 2018-06-27 AbbVie Stemcentrx LLC Novel sez6 modulators and methods of use
EP3892294A1 (en) 2013-08-28 2021-10-13 AbbVie Stemcentrx LLC Site-specific antibody conjugation methods and compositions
US10035853B2 (en) 2013-08-28 2018-07-31 Abbvie Stemcentrx Llc Site-specific antibody conjugation methods and compositions
EP3738981A1 (en) 2014-01-24 2020-11-18 NGM Biopharmaceuticals, Inc. Antibodies binding beta klotho domain 2 and methods of use thereof
US10308721B2 (en) 2014-02-21 2019-06-04 Abbvie Stemcentrx Llc Anti-DLL3 antibodies and drug conjugates for use in melanoma
EP3590539A1 (en) 2014-03-04 2020-01-08 Kymab Limited Antibodies, uses & methods
US10017580B2 (en) 2014-04-15 2018-07-10 ADC Therpeutics S.A. Humanized anti-Tn-MUC1 antibodies and their conjugates
WO2015175375A1 (en) 2014-05-13 2015-11-19 Short Jay M Conditionally active biological proteins
US10329556B2 (en) 2014-05-13 2019-06-25 Bioatla, Llc Conditionally active biological proteins
EP3888690A2 (en) 2014-05-16 2021-10-06 MedImmune, LLC Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
WO2015181805A1 (en) 2014-05-28 2015-12-03 Zymeworks Inc. Modified antigen binding polypeptide constructs and uses thereof
EP4026850A1 (en) 2014-05-28 2022-07-13 Zymeworks Inc. Modified antigen binding polypeptide constructs and uses thereof
WO2015189816A1 (en) 2014-06-13 2015-12-17 Friedrich Miescher Institute For Biomedical Research New treatment against influenza virus
WO2015198202A1 (en) 2014-06-23 2015-12-30 Friedrich Miescher Institute For Biomedical Research Methods for triggering de novo formation of heterochromatin and or epigenetic silencing with small rnas
WO2016001830A1 (en) 2014-07-01 2016-01-07 Friedrich Miescher Institute For Biomedical Research Combination of a brafv600e inhibitor and mertk inhibitor to treat melanoma
US10513699B2 (en) 2014-09-03 2019-12-24 Bioatla, Llc Discovering and producing conditionally active biologic proteins in the same eukaryotic cell production hosts
WO2016046768A1 (en) 2014-09-24 2016-03-31 Friedrich Miescher Institute For Biomedical Research Lats and breast cancer
EP3800202A1 (en) 2014-12-11 2021-04-07 Pierre Fabre Medicament Anti-c10orf54 antibodies and uses thereof
US10766959B2 (en) 2014-12-11 2020-09-08 Pierre Fabre Medicament Anti-C10ORF54 antibodies and uses thereof
US11873339B2 (en) 2014-12-11 2024-01-16 Pierre Fabre Medicament Anti-C10orf54 antibodies and uses thereof
US10563194B2 (en) 2015-02-24 2020-02-18 BioAlta, LLC Conditionally active biological proteins
US11254932B2 (en) 2015-02-24 2022-02-22 Bioatla, Inc. Conditionally active biological proteins
WO2016139482A1 (en) 2015-03-03 2016-09-09 Kymab Limited Antibodies, uses & methods
EP4137157A1 (en) 2015-03-03 2023-02-22 Kymab Limited Antibodies, uses and methods
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
WO2016166304A1 (en) 2015-04-15 2016-10-20 Van Berkel Patricius Hendrikus Cornelis Site-specific antibody-drug conjugates
WO2017059551A1 (en) 2015-10-08 2017-04-13 Zymeworks Inc. Antigen-binding polypeptide constructs comprising kappa and lambda light chains and uses thereof
WO2017072669A1 (en) 2015-10-28 2017-05-04 Friedrich Miescher Institute For Biomedical Research Tenascin-w and biliary tract cancers
EP3909983A1 (en) 2015-12-02 2021-11-17 STCube & Co. Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
WO2017096051A1 (en) 2015-12-02 2017-06-08 Stcube & Co., Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
US11253590B2 (en) 2015-12-02 2022-02-22 Stsciences, Inc. Antibodies specific to glycosylated BTLA (B- and T- lymphocyte attenuator)
WO2018083248A1 (en) 2016-11-03 2018-05-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
WO2018222685A1 (en) 2017-05-31 2018-12-06 Stcube & Co., Inc. Methods of treating cancer using antibodies and molecules that immunospecifically bind to btn1a1
WO2018222689A1 (en) 2017-05-31 2018-12-06 Stcube & Co., Inc. Antibodies and molecules that immunospecifically bind to btn1a1 and the therapeutic uses thereof
US11542331B2 (en) 2017-06-06 2023-01-03 Stcube & Co., Inc. Methods of treating cancer using antibodies and molecules that bind to BTN1A1 or BTN1A1-ligands
WO2018226671A1 (en) 2017-06-06 2018-12-13 Stcube & Co., Inc. Methods of treating cancer using antibodies and molecules that bind to btn1a1 or btn1a1-ligands
US11401345B2 (en) 2017-11-27 2022-08-02 Purdue Pharma L.P. Humanized antibodies targeting human tissue factor
WO2019102435A1 (en) 2017-11-27 2019-05-31 Euro-Celtique S.A. Humanized antibodies targeting human tissue factor
WO2020016459A1 (en) 2018-07-20 2020-01-23 Pierre Fabre Medicament Receptor for vista
WO2020198731A2 (en) 2019-03-28 2020-10-01 Danisco Us Inc Engineered antibodies
US11827671B2 (en) 2019-05-24 2023-11-28 Sanofi Methods for treating systemic sclerosis
WO2020242989A1 (en) 2019-05-24 2020-12-03 Sanofi Methods for treating systemic sclerosis
WO2021202473A2 (en) 2020-03-30 2021-10-07 Danisco Us Inc Engineered antibodies
WO2024015953A1 (en) 2022-07-15 2024-01-18 Danisco Us Inc. Methods for producing monoclonal antibodies

Also Published As

Publication number Publication date
AU5711196A (en) 1997-10-01

Similar Documents

Publication Publication Date Title
EP0897390B1 (en) Human tumor necrosis factor delta and epsilon
US5874240A (en) Human 4-1BB receptor splicing variant
WO1997033899A1 (en) Apoptosis inducing molecule i
AU733603B2 (en) Chemokine alpha 2
CA2249251A1 (en) Human cytokine polypeptide
US5945303A (en) Human hematopoietic-specific protein
US6509170B1 (en) Polynucleotides encoding human tumor necrosis factor delta
US20060171918A1 (en) Apoptosis inducing molecule I
US6448044B2 (en) DNA encoding human natural killer cell activating factor II
WO1997033898A1 (en) Human 4-1bb receptor splicing variant
AU2003236393B2 (en) Methods of Treating or Preventing Inflammation or Rheumatoid Arthritis
AU761216B2 (en) Human tumor necrosis factor delta and epsilon
US5952197A (en) DNA encoding the chemotactic cytokine III
EP0937147A1 (en) Human natural killer cell activating factor ii
AU767338B2 (en) Chemokine alpha 2
US20040197874A1 (en) Human cytokine polypeptide
WO1997032993A1 (en) Chemotactic cytokine iii
JP2008194038A (en) HUMAN TUMOR NECROSIS FACTOR delta and epsilon
KR19990087832A (en) Human tumor necrosis factor delta and epsilon

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AM AU BG BR BY CA CN CZ EE FI GE HU JP KG KP KR KZ LT LV MD MN MX NO NZ PL RO RU SG SI SK TJ TM UA US UZ VN

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

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)
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97532552

Format of ref document f/p: F

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

Ref country code: CA