US20090075884A1 - Human il-17f and other il-17f-related proteins and methods related thereto - Google Patents

Human il-17f and other il-17f-related proteins and methods related thereto Download PDF

Info

Publication number
US20090075884A1
US20090075884A1 US12/175,336 US17533608A US2009075884A1 US 20090075884 A1 US20090075884 A1 US 20090075884A1 US 17533608 A US17533608 A US 17533608A US 2009075884 A1 US2009075884 A1 US 2009075884A1
Authority
US
United States
Prior art keywords
ctla
cells
protein
seq
proteins
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/175,336
Inventor
Kenneth Jacobs
Kerry Kelleher
McKeough Carlin
Samuel Goldman
Debra Pittman
Sha Mi
Steven Neben
Joanne Giannotti
Margaret M. Golden-Fleet
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Genetics Institute LLC
Original Assignee
Genetics Institute LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US08/514,014 external-priority patent/US5707829A/en
Priority claimed from US08/685,239 external-priority patent/US6074849A/en
Application filed by Genetics Institute LLC filed Critical Genetics Institute LLC
Priority to US12/175,336 priority Critical patent/US20090075884A1/en
Publication of US20090075884A1 publication Critical patent/US20090075884A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/521Chemokines
    • C07K14/523Beta-chemokines, e.g. RANTES, I-309/TCA-3, MIP-1alpha, MIP-1beta/ACT-2/LD78/SCIF, MCP-1/MCAF, MCP-2, MCP-3, LDCF-1, LDCF-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to human CTLA-8 proteins, nucleic acids encoding such proteins, methods of treatment using such proteins.
  • the invention also relates to the use of rat CTLA-8 proteins and herpesvirus Saimiri ORF 13 proteins in methods of treatment.
  • Cytokines are secreted proteins which act on specific hematopoietic target cells to cause a differentiation event or on other target cells to induce a particular physiological response, such as secretion of proteins characteristic of inflammation.
  • Cytokines also variously known as lymphokines, hematopoietins, interleukins, colony stimulating factors, and the like, can be important therapeutic agents, especially for diseases or conditions in which a specific cell population is depleted. For example, erythropoietin, G-CSF, and GM-CSF, have all become important for treatment of anemia and leukopenia, respectively.
  • Other cytokines such as interleukin-3, interleukin-6, interleukin-11 and interleukin-12 show promise in treatment of conditions such as thrombocytopenia and modulation of immune response.
  • novel cytokines were identified by assaying a particular cell such as a bone marrow cell, for a measurable response, such as proliferation.
  • the search for novel cytokines has thus been limited by the assays available, and if a novel cytokine has an activity which is unmeasurable by a known assay, the cytokine remains undetectable.
  • cDNAs encoding cytokines have been detected using the polymerase chain reaction (PCR) and oligonucleotide primers having homology to shared motifs of known cytokines or their receptors.
  • PCR approach is also limited by the necessity for knowledge of previously cloned cytokines in the same protein family.
  • Cytokines have also been cloned using subtractive hybridization to construct and screen cDNA libraries, or they can potentially be cloned using PCR followed by gel electrophoresis to detect differentially expressed genes.
  • the subtractive hybridization methods are based on the assumption that cytokine mRNAs are those that are differentially expressed, and these methods do not require any prior knowledge of the sequence of interest.
  • many cytokines may be encoded by mRNAs which are not differentially expressed, and thus are undetectable using these methods.
  • CTLA-8 human CTLA-8
  • CTLA-8 (now known as IL-17F) and active fragments thereof are disclosed.
  • CTLA-8 (now known as IL-17F) is used throughout the present specification to refer to both proteins and polynucleotides encoding those proteins and to refer to proteins and polynucleotides from all mammalian species.
  • the present invention provides an isolated polynucleotide comprising a nucleotide sequence selected from the group consisting of:
  • the polynucleotide of the invention encodes a protein having CTLA-8 activity.
  • the polynucleotide is operably linked to an expression control sequence.
  • the polynucleotide is contained in a vector suitable for in vivo expression in a mammalian subject.
  • Polynucleotides comprising the nucleotide sequence of SEQ ID NO:1 from nucleotide 55 to nucleotide 544, the nucleotide sequence of SEQ ID NO:1 from nucleotide 139 to nucleotide 544 or the nucleotide sequence of SEQ ID NO:1 from nucleotide 86 to nucleotide 544 are particularly preferred.
  • Host cells transformed with the polynucleotides of the invention are also provided, including mammalian cells.
  • Processes are also provided for producing a human CTLA-8 protein, said processes comprising:
  • Isolated human CTLA-8 protein is also provided which comprises an amino acid sequence selected from the group consisting of:
  • Proteins comprising the amino acid sequence of SEQ ID NO:2 and comprising the sequence from amino acids 29 to 163, from amino acid 31 to 163, or from amino acids 11 to 163 of SEQ ID NO:2 are particularly preferred.
  • the protein has CTLA-8 activity.
  • Pharmaceuticals composition comprising a human CTLA-8 protein of the invention and a pharmaceutically acceptable carrier are also provided.
  • compositions which comprise an antibody which specifically reacts with a human CTLA-8 protein of the invention.
  • Methods of treating a mammalian subject comprise administering a therapeutically effective amount of a pharmaceutical composition comprising a human CTLA-8 protein.
  • Rat CTLA-8 and active (i.e., having CTLA-8 activity) fragments thereof may also be used in such methods of treatment,
  • the rat protein is administered as a composition comprising a pharmaceutically acceptable carrier and a protein comprising an amino acid sequence selected from the group consisting of:
  • herpesvirus Saimiri ORF13 referred to herein as “herpes CTLA-8”, and active (i.e., having CTLA-8 activity) fragments thereof and active fragments thereof may also be used in such methods of treatment.
  • the herpes CTLA-8 protein is administered as a composition comprising a pharmaceutically acceptable carrier and a protein comprising an amino acid sequence selected from the group consisting of:
  • the invention also provides a method of treating a mammalian subject comprising administering a therapeutically effective amount of a composition comprising a pharmaceutically acceptable carrier and IL-17 or an active fragment thereof.
  • the subject is treated to produce an effect selected from the group consisting of inhibition of angiogenesis, inhibition of growth or proliferation of vascular endothelial cells, inhibition of tumor growth, inhibition of angiogenesis-dependent tissue growth, proliferation of myeloid cells or progenitors, proliferation of erythroid cells or progenitors, proliferation of lymphoid cells or progenitors, induction of IFN ⁇ production, induction of IL-3 production and induction of GM-CSF production.
  • FIG. 1 is a comparison of homologous regions of the amino acid sequences of human CTLA-8 (indicated as “B18_F1”), rat CTLA-8 (indicated as “Musctla8”) and herpes CTLA-8 (indicated as “Hsvie — 2”).
  • FIG. 2 depicts autoradiographs demonstrating expression of human CTLA-8 in COS cells.
  • FIG. 3 presents data relating to the ability of human CTLA-8 to inhibit angiogenesis.
  • FIGS. 4 and 5 present data relating to the ability of human CTLA-8 to produce or induce hematopoietic activity.
  • FIGS. 6 and 7 present data demonstrating the ability of human CTLA-8 to induce production of IL-6 and IL-8.
  • SEQ ID NO:1 provides the nucleotide sequence of a cDNA encoding the human CTLA-8 protein.
  • SEQ ID NO:2 provides the amino acid sequence of the human CTLA-8 protein.
  • the initiating methionine may be at amino acid 11 of SEQ ID NO:2.
  • the mature protein sequence is believed to begin at amino acid 31 of SEQ ID NO:2 (encoded by the sequence beginning with nucleotide 146 of SEQ ID NO:1).
  • the region from amino acid 29 to amino acid 163 of human CTLA-8 shows marked homology to portions of rat CTLA-8 (amino acids 18 to 150 of SEQ ID NO:4) and herpesvirus Saimiri ORF13 (“herpes CTLA-8”) (amino acids 19 to 151 of SEQ ID NO:5).
  • a cDNA sequence encoding rat CTLA-8 is listed at SEQ ID NO:3 and its corresponding amino acid sequence is reported at SEQ ID NO:4.
  • a cDNA sequence encoding herpes CTLA-8 is listed at SEQ ID NO:5 and its corresponding amino acid sequence is reported at SEQ ID NO:6. Homology between rat CTLA-8 and herpes CTLA-8 was reported by Rouvier et al., J. Immunol. 1993, 150, 5445-5456.
  • Golstein et al. (WO95/18826; Fossiez et al., Microbial Evasion and Subversion of Immunity 544:3222 (Abstract)) have also reported a species they initially identified as “human CTLA-8.”
  • examination of the sequence of the Golstein et al. species and the human CTLA-8 (B18) sequence of the present invention readily reveals that they are two different proteins, although they are homologous with each other and with the rat CTLA-8 and herpes CTLA-8 identified herein.
  • the Golstein et al. species has now been renamed as interleukin-17 (IL-17). Because of the homology between applicants' human CTLA-8 (B18) and IL-17, these proteins are expected to share some activities.
  • IL-17 interleukin-17
  • human CTLA-8 (B 18) forms homodimers when expressed.
  • human CTLA-8 proteins may possess activity in either monomeric or dimeric forms.
  • Human CTLA-8 proteins can also be produced as heterodimers with rat and herpes CTLA-8 proteins and with human IL-17. These heterodimers are also expected to have activities of the proteins of which they are comprised.
  • Forms of human CTLA-8 protein of less than full length are encompassed within the present invention and may be produced by expressing a corresponding fragment of the polynucleotide encoding the human CTLA-8 protein (SEQ ID NO:1). These corresponding polynucleotide fragments are also part of the present invention.
  • Modified polynucleotides as described above may be made by standard molecular biology techniques, including site-directed mutagenesis methods which are known in the art or by the polymerase chain reaction using appropriate oligonucleotide primers.
  • a protein has “CTLA-8 activity” if it either (1) displays biological activity in a factor-dependent cell proliferation assay (preferably an assay in which the corresponding species' full-length CTLA-8 is active) (including without limitation those assays described below), or (2) induces expression or secretion of ⁇ -IFN, or (3) displays chemoattractant or chemotactic activity in a chemoattraction or chemotaxis assay (preferably an assay in which the corresponding species' full-length CTLA-8 is active) or (4) induces expression or secretion of IL-3 or GM-CSF.
  • a factor-dependent cell proliferation assay preferably an assay in which the corresponding species' full-length CTLA-8 is active
  • (4) induces expression or secretion of IL-3 or GM-CSF
  • Human CTLA-8 protein or fragments thereof having CTLA-8 activity may be fused to carrier molecules such as immunoglobulins.
  • human CTLA-8 protein may be fused through “linker” sequences to the Fc portion of an immunoglobulin.
  • the invention also encompasses allelic variations of the nucleotide sequence as set forth in SEQ ID NO:1, that is, naturally-occurring alternative forms of the isolated polynucleotide of SEQ ID NO:1 which also encode human CTLA-8 or CTLA-8 proteins having CTLA-8 activity. Also included in the invention are isolated polynucleotides which hybridize to the nucleotide sequence set forth in SEQ ID NO:1 under highly stringent (0.2 ⁇ SSC at 65° C.), stringent (e.g. 4 ⁇ SSC at 65° C. or 50% formamide and 4 ⁇ SSC at 42° C.), or relaxed (4 ⁇ SSC at 50° C. or 30-40% formamide and 4 ⁇ SSC at 42° C.) conditions.
  • highly stringent 0.2 ⁇ SSC at 65° C.
  • stringent e.g. 4 ⁇ SSC at 65° C. or 50% formamide and 4 ⁇ SSC at 42° C.
  • Isolated polynucleotides which encode human CTLA-8 protein but which differ from the nucleotide sequence set forth in SEQ ID NO:1 by virtue of the degeneracy of the genetic code are also encompassed by the present invention. Variations in the nucleotide sequence as set forth in SEQ ID NO:1 which are caused by point mutations or by induced modifications which enhance CTLA-8 activity, half-life or production level are also included in the invention.
  • the isolated polynucleotides of the invention may be operably linked to an expression control sequence such as the pMT2 or pED expression vectors disclosed in Kaufman et al., Nucleic Acids Res. 19, 4485-4490 (1991), in order to produce the CTLA-8 protein recombinantly.
  • an expression control sequence such as the pMT2 or pED expression vectors disclosed in Kaufman et al., Nucleic Acids Res. 19, 4485-4490 (1991
  • Many suitable expression control sequences are known in the art. General methods of expressing recombinant proteins are also known and are exemplified in R. Kaufman, Methods in Enzymology 185, 537-566 (1990).
  • operably linked means enzymatically or chemically ligated to form a covalent bond between the isolated polynucleotide of the invention and the expression control sequence, in such a way that the CTLA-8 protein is expressed by a host cell which has been transformed (transfected) with the ligated polynucleotide/expression control sequence.
  • a number of types of cells may act as suitable host cells for expression of the human CTLA-8 protein. Any cell type capable of expressing functional human CTLA-8 protein may be used.
  • Suitable mammalian host cells include, for example, monkey COS cells, Chinese Hamster Ovary (CHO) cells, human kidney 293 cells, human epidermal A431 cells, human Colo205 cells, 3T3 cells, CV-1 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HeLa cells, mouse L cells, BHK, HL-60, U937, HaK, Rat2, BaF3, 32D, FDCP-1, PC12 or C2C12 cells.
  • monkey COS cells Chinese Hamster Ovary (CHO) cells
  • human kidney 293 cells human epidermal A431 cells
  • human Colo205 cells human Colo205 cells
  • CV-1 cells other transformed primate cell lines
  • normal diploid cells cell strains derived from in vitro culture of primary tissue, primary explants,
  • the human CTLA-8 protein may also be produced by operably linking the isolated polynucleotide of the invention to suitable control sequences in one or more insect expression vectors, and employing an insect expression system.
  • suitable control sequences in one or more insect expression vectors, and employing an insect expression system.
  • Materials and Methods for baculovirus/insect cell expression systems are commercially available in kit form from, e.g., Invitrogen, San Diego, Calif., U.S.A. (the MaxBac® kit), and such methods are well known in the art, as described in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987), incorporated herein by reference.
  • Soluble forms of the human CTLA-8 protein may also be produced in insect cells using appropriate isolated polynucleotides as described above.
  • the human CTLA-8 protein may be produced in lower eukaryotes such as yeast or in prokaryotes such as bacteria.
  • suitable yeast strains include Saccharomyces cerevisiae, Schizosaccharomyces pombe, Kluyveromyces strains, Candida , or any yeast strain capable of expressing heterologous proteins.
  • Suitable bacterial strains include Escherichia coli, Bacillus subtilis, Salmonella typhimurium , or any bacterial strain capable of expressing heterologous proteins.
  • the human CTLA-8 protein of the invention may also be expressed as a product of transgenic animals, e.g., as a component of the milk of transgenic cows, goats, pigs, or sheep which are characterized by somatic or germ cells containing a polynucleotide sequence encoding the human CTLA-8 protein.
  • the human CTLA-8 protein of the invention may be prepared by growing a culture of transformed host cells under culture conditions necessary to express the desired protein. The resulting expressed protein may then be purified from the culture medium or cell extracts. Soluble forms of the human CTLA-8 protein of the invention can be purified from conditioned media. Membrane-bound forms of human CTLA-8 protein of the invention can be purified by preparing a total membrane fraction from the expressing cell and extracting the membranes with a non-ionic detergent such as Triton X-100.
  • a non-ionic detergent such as Triton X-100.
  • the human CTLA-8 protein can be purified using methods known to those skilled in the art.
  • the human CTLA-8 protein of the invention can be concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • the concentrate can be applied to a purification matrix such as a gel filtration medium.
  • an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups.
  • the matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification.
  • a cation exchange step can be employed.
  • Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred (e.g., S-Sepharose® columns).
  • the purification of the human CTLA-8 protein from culture supernatant may also include one or more column steps over such affinity resins as concanavalin A-agarose, heparin-toyopearl® or Cibacrom blue 3GA Sepharose®; or by hydrophobic interaction chromatography using such resins as phenyl ether, butyl ether, or propyl ether; or by immunoaffinity chromatography.
  • RP-HPLC reverse-phase high performance liquid chromatography
  • hydrophobic RP-HPLC media e.g., silica gel having pendant methyl or other aliphatic groups
  • the human CTLA-8 protein is purified so that it is substantially free of other mammalian proteins.
  • human CTLA-8, active fragments and variants thereof, and CTLA-8 related proteins possess or induce cytokine activities.
  • Human CTLA-8 expression correlated with ⁇ -IFN expression in induced primary cells and can induce the expression of IL-3 and/or GM-CSF, which expression can in turn produce effects associated with the induced cytokine. Therefore, human CTLA-8 and CTLA-8 related proteins may have an effect on proliferation or function of myeloid cells, erythroid cells, lymphoid cells and their progenitors.
  • Human CTLA-8 proteins may also play a role in formation of platelets or their progenitors.
  • a protein of the present invention may exhibit cytokine, cell proliferation (either inducing or inhibiting) or cell differentiation (either inducing or inhibiting) activity or may induce production of other cytokines in certain cell populations.
  • cytokine cytokine
  • cell proliferation either inducing or inhibiting
  • cell differentiation either inducing or inhibiting
  • the activity of a protein of the present invention is evidenced by any one of a number of routine factor dependent cell proliferation assays for cell lines including, without limitation, 32D, DA2, DA1G, T10, B9, B9/11, BaF3, MC9/G, M+(preB M+), 2E8, RB5, DA1, 123, T1165, HT2, CTLL2, TF-1, Mo7e and CMK.
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • Assays for T-cell or thymocyte proliferation include without limitation those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W. Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Takai et al., J. Immunol. 137:3494-3500, 1986; Bertagnolli et al., J. Immunol.
  • Assays for cytokine production and/or proliferation of spleen cells, lymph node cells or thymocytes include, without limitation, those described in: Polyclonal T cell stimulation, Kruisbeek, A. M. and Shevach, E. M. In Current Protocols in Immunology . J. E Coligan et al. eds. Vol 1 pp. 3.12.1-3.12.14, John Wiley and Sons, Toronto. 1994; and Measurement of mouse and human Interferon ⁇ , Schreiber, R. D. In Current Protocols in Immunology . J. E Coligan et al. eds. Vol 1 pp. 6.8.1-6.8.8, John Wiley and Sons, Toronto. 1994.
  • Assays for proliferation and differentiation of hematopoietic and lymphopoietic cells include, without limitation, those described in: Measurement of Human and Murine Interleukin 2 and Interleukin 4, Bottomly, K., Davis, L. S, and Lipsky, P. E. In Current Protocols in Immunology . J. E Coligan et al. eds. Vol 1 pp. 6.3.1-6.3.12, John Wiley and Sons, Toronto. 1991; deVries et al., J. Exp. Med. 173:1205-1211, 1991; Moreau et al., Nature 336:690-692, 1988; Greenberger et al., Proc. Natl. Acad. Sci. U.S.A.
  • Assays for T-cell clone responses to antigens include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W. Strober Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function; Chapter 6, Cytokines and their cellular receptors; Chapter 7, Immunologic studies in Humans); Weinberger et al., Proc. Natl. Acad. Sci.
  • a protein of the present invention may also exhibit immune stimulating or immune suppressing activity, including without limitation the activities for which assays are described herein.
  • a protein may be useful in the treatment of various immune deficiencies and disorders (including severe combined immunodeficiency (SCID)), e.g., in regulating (up or down) growth and proliferation of T and/or B lymphocytes, as well as effecting the cytolytic activity of NK cells and other cell populations.
  • SCID severe combined immunodeficiency
  • These immune deficiencies may be genetic or be caused by viral (e.g., HIV) as well as bacterial or fungal infections, or may result from autoimmune disorders.
  • infectious diseases caused by viral, bacterial, fungal or other infection may be treatable using a protein of the present invention, including infections by HIV, hepatitis viruses, herpes viruses, mycobacteria, leshmania, malaria and various fungal infections such as candida.
  • a protein of the present invention may also be useful where a boost to the immune system generally would be indicated, i.e., in the treatment of cancer.
  • Autoimmune disorders which may be treated using a protein of the present invention include, for example, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes mellitis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease.
  • a protein of the present invention may also to be useful in the treatment of allergic reactions and conditions, such as asthma or other respiratory problems.
  • Other conditions, in which immune suppression is desired may also be treatable using a protein of the present invention.
  • a protein of the present invention may also suppress chronic or acute inflammation, such as, for example, that associated with infection (such as septic shock or systemic inflammatory response syndrome (SIRS)), inflammatory bowel disease, Crohn's disease or resulting from over production of cytokines such as TNF or IL-1 (such as the effect demonstrated by IL-11).
  • infection such as septic shock or systemic inflammatory response syndrome (SIRS)
  • SIRRS systemic inflammatory response syndrome
  • inflammatory bowel disease such as septic shock or systemic inflammatory response syndrome (SIRS)
  • Crohn's disease or resulting from over production of cytokines
  • TNF or IL-1 such as the effect demonstrated by IL-11.
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • Suitable assays for thymocyte or splenocyte cytotoxicity include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Herrmann et al., Proc. Natl. Acad. Sci. USA 78:2488-2492, 1981; Herrmann et al., J. Immunol. 128:1968-1974, 1982; Handa et al., J. Immunol.
  • T-cell-dependent immunoglobulin responses and isotype switching (which will identify, among others, proteins that modulate T-cell dependent antibody responses and that affect Th1/Th2 profiles) include, without limitation, those described in: Maliszewski, J. Immunol. 144:3028-3033, 1990; and Assays for B cell function: In vitro antibody production, Mond, J. J. and Brunswick, M. In Current Protocols in Immunology . J. E Coligan et al. eds. Vol 1 pp. 3.8.1-3.8.16, John Wiley and Sons, Toronto. 1994.
  • MLR Mixed lymphocyte reaction
  • Dendritic cell-dependent assays (which will identify, among others, proteins expressed by dendritic cells that activate naive T-cells) include, without limitation, those described in: Guery et al., J. Immunol. 134:536-544, 1995; Inaba et al., Journal of Experimenal Medicine 173:549-559, 1991; Macatonia et al., Journal of Immunology 154:5071-5079, 1995; Porgador et al., Journal of Experimental Medicine 182:255-260, 1995; Nair et al., Journal of Virology 67:4062-4069, 1993; Huang et al., Science 264:961-965, 1994; Macatonia et al., Journal of Experimental Medicine 169:1255-1264, 1989; Bhardwaj et al., Journal of Clinical Investigation 94:791-807, 1994; and Inaba et al., Journal of Experimental Medicine 172:631-640, 1990.
  • lymphocyte survival/apoptosis (which will identify, among others, proteins that prevent apoptosis after superantigen induction and proteins that regulate lymphocyte homeostasis) include, without limitation, those described in: Darzynkiewicz et al., Cytometry 13:795-808, 1992; Gorczyca et al., Leukemia 7:659-670, 1993; Gorczyca et al., Cancer Research 53:1945-1951, 1993; Itoh et al., Cell 66:233-243, 1991; Zacharchuk, Journal of Immunology 145:4037-4045, 1990; Zamai et al., Cytometry 14:891-897, 1993; Gorczyca et al., International Journal of Oncology 1:639-648, 1992.
  • Assays for proteins that influence early steps of T-cell commitment and development include, without limitation, those described in: Antica et al., Blood 84:111-117, 1994; Fine et al., Cellular Immunology 155:111-122, 1994; Galy et al., Blood 85:2770-2778, 1995; Toki et al., Proc. Nat. Acad. Sci. USA 88:7548-7551, 1991.
  • a protein of the present invention may be useful in regulation of hematopoiesis and, consequently, in the treatment of myeloid or lymphoid cell deficiencies. Even marginal biological activity in support of colony forming cells or of factor-dependent cell lines indicates involvement in regulating hematopoiesis, e.g.
  • erythroid progenitor cells alone or in combination with other cytokines, thereby indicating utility, for example, in treating various anemias or for use in conjunction with irradiation/chemotherapy to stimulate the production of erythroid precursors and/or erythroid cells; in supporting the growth and proliferation of myeloid cells such as granulocytes and monocytes/macrophages (i.e., traditional CSF activity) useful, for example, in conjunction with chemotherapy to prevent or treat consequent myelo-suppression; in supporting the growth and proliferation of megakaryocytes and consequently of platelets thereby allowing prevention or treatment of various platelet disorders such as thrombocytopenia, and generally for use in place of or complementary to platelet transfusions; and/or in supporting the growth and proliferation of hematopoietic stem cells which are capable of maturing to any and all of the above-mentioned hematopoietic cells and therefore find therapeutic utility in various stem cell disorders (such as those usually treated with
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • Assays for embryonic stem cell differentiation include, without limitation, those described in: Johansson et al. Cellular Biology 15:141-151, 1995; Keller et al., Molecular and Cellular Biology 13:473-486, 1993; McClanahan et al., Blood 81:2903-2915, 1993.
  • Assays for stem cell survival and differentiation include, without limitation, those described in: Methylcellulose colony forming assays, Freshney, M. G. In Culture of Hematopoietic Cells . R. I. Freshney, et al. eds. pp. 265-268, Wiley-Liss, Inc., New York, N.Y. 1994; Hirayama et al., Proc. Natl. Acad. Sci. USA 89:5907-5911, 1992; Primitive hematopoietic colony forming cells with high proliferative potential, McNiece, I. K. and Briddell, R. A.
  • CTLA-8 proteins are useful in the treatment of various immune deficiencies and disorders (including SCID), e.g., in regulating (up or down) growth, proliferation and/or activity of T and/or B lymphocytes, as well as the cytolytic activity of NK cells.
  • immune deficiencies may be caused by viral (e.g., HIV) as well as bacterial infections, or may result from autoimmune disorders.
  • infectious diseases caused by viral, bacterial, fungal or other infection may be treatable using CTLA-8 proteins, including infections by HIV, hepatitis, influenza, CMV, herpes, mycobacterium, leishmaniasis, malaria and various fungal infections (such as candida).
  • CTLA-8 proteins may also be useful where a boost to the immune system generally would be indicated, i.e., in the treatment of cancer or as an adjuvant to vaccines.
  • Autoimmune disorders which may be treated using factors of the present invention include, for example, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes mellitis and autoimmune inflammatory eye disease.
  • the CTLA-8 proteins are also expected to be useful in the treatment of allergic reactions and conditions.
  • CTLA-8 proteins are also expected to have chemotactic activity.
  • a protein or peptide has “chemotactic activity,” as used herein, if it can stimulate, directly or indirectly, the directed orientation or movement of cells, including myeloid and lymphoid cells.
  • the protein or peptide has the ability to directly stimulate directed movement of cells (particularly T-cells). Whether a particular protein or peptide has chemotactic activity for cells can be readily determined by employing such protein or peptide in any known assay for cell chemotaxis.
  • CTLA-8 proteins also inhibit growth and proliferation of vascular endothelial cells.
  • human CTLA-8 proteins are effective in inhibiting angiogenesis (i.e., vascular formation). This activity will also be useful in the treatment of tumors and other conditions in which angiogenesis is involved. Inhibition of angiogenesis by human CTLA-8 proteins will also result in inhibition or prevention of the condition to which normal angiogenesis would contribute.
  • Isolated CTLA-8 proteins purified from cells or recombinantly produced, may be used as a pharmaceutical composition when combined with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier may contain, in addition to CTLA-8 protein and carrier, diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art.
  • pharmaceutically acceptable means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier will depend on the route of administration.
  • the pharmaceutical composition of the invention may also contain cytokines, lymphokines, or other hematopoietic factors such as M-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-15, G-CSF, ⁇ -IFN, stem cell factor, and erythropoietin.
  • the pharmaceutical composition may contain thrombolytic or anti-thrombotic factors such as plasminogen activator and Factor VIII.
  • the pharmaceutical composition may further contain other anti-inflammatory agents.
  • CTLA-8 protein may be included in formulations of the particular cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent to minimize side effects of the cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent.
  • the pharmaceutical composition of the invention may be in the form of a liposome.
  • CTLA-8 protein and pharmaceutically acceptable carriers are combined with amphipathic agents such as lipids, which exist in aggregated forms, such as micelles, insoluble monolayers, liquid crystals, or lamellar layers, in aqueous solution.
  • Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. Preparation of such liposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Pat. No. 4,235,871; U.S. Pat. No. 4,501,728; U.S. Pat. No. 4,837,028; and U.S. Pat. No. 4,737,323, all of which are incorporated herein by reference.
  • the term “therapeutically effective amount” means the total amount of each active component of the pharmaceutical composition or method that is sufficient to show a meaningful patient benefit, e.g., amelioration of symptoms of, healing of, or increase in rate of healing of such conditions.
  • a meaningful patient benefit e.g., amelioration of symptoms of, healing of, or increase in rate of healing of such conditions.
  • the term refers to that ingredient alone.
  • the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
  • CTLA-8 protein is administered to a mammal.
  • CTLA-8 protein may be administered in accordance with the method of the invention either alone or in combination with other therapies such as treatments employing cytokines, lymphokines or other hematopoietic factors.
  • CTLA-8 protein may be administered either simultaneously with the cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering CTLA-8 protein in combination with cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors.
  • CTLA-8 protein used in the pharmaceutical composition or to practice the method of the present invention can be carried out in a variety of conventional ways, such as oral ingestion, inhalation, or cutaneous, subcutaneous, or intravenous injection. Intravenous administration to the patient is preferred.
  • CTLA-8 protein When a therapeutically effective amount of CTLA-8 protein is administered orally, CTLA-8 protein will be in the form of a tablet, capsule, powder, solution or elixir.
  • the pharmaceutical composition of the invention may additionally contain a solid carrier such as a gelatin or an adjuvant.
  • the tablet, capsule, and powder contain from about 5 to 95% CTLA-8 protein, and preferably from about 25 to 90% CTLA-8 protein.
  • a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils may be added.
  • the liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol.
  • the pharmaceutical composition When administered in liquid form, contains from about 0.5 to 90% by weight of CTLA-8 protein, and preferably from about 1 to 50% CTLA-8 protein.
  • CTLA-8 protein When a therapeutically effective amount of CTLA-8 protein is administered by intravenous, cutaneous or subcutaneous injection, CTLA-8 protein will be in the form of a pyrogen-free, parenterally acceptable aqueous solution.
  • a preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to CTLA-8 protein an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or other vehicle as known in the art.
  • the pharmaceutical composition of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additive known to those of skill in the art.
  • CTLA-8 protein in the pharmaceutical composition of the present invention will depend upon the nature and severity of the condition being treated, and on the nature of prior treatments which the patient has undergone. Ultimately, the attending physician will decide the amount of CTLA-8 protein with which to treat each individual patient. Initially, the attending physician will administer low doses of CTLA-8 protein and observe the patient's response. Larger doses of CTLA-8 protein may be administered until the optimal therapeutic effect is obtained for the patient, and at that point the dosage is not generally increased further.
  • the various pharmaceutical compositions used to practice the method of the present invention should contain about 0.1 ⁇ g to about 100 mg of CTLA-8 protein per kg body weight, preferably about 0.1 ⁇ g to about 10 mg of CTLA-8 protein per kg body weight, more preferably about 0.1 ⁇ g to about 100 ⁇ g of CTLA-8 protein per kg body weight, most preferably about 0.1 ⁇ g to about 10 ⁇ g of CTLA-8 protein per kg body weight.
  • the duration of intravenous therapy using the pharmaceutical composition of the present invention will vary, depending on the severity of the disease being treated and the condition and potential idiosyncratic response of each individual patient. It is contemplated that the duration of each application of the CTLA-8 protein will be in the range of 12 to 24 hours of continuous intravenous administration. Ultimately the attending physician will decide on the appropriate duration of intravenous therapy using the pharmaceutical composition of the present invention.
  • CTLA-8 protein of the invention may also be used to immunize animals to obtain polyclonal and monoclonal antibodies which specifically react with the CTLA-8 protein and which may inhibit CTLA-8 binding to its receptor. Such antibodies are also useful for performing diagnostics assays for CTLA-8 in accordance with known methods. Such antibodies may be obtained using the entire CTLA-8 protein as an immunogen, or by using fragments of human CTLA-8 protein.
  • the peptide immunogens additionally may contain a cysteine residue at the carboxyl terminus, and are conjugated to a hapten such as keyhole limpet hemocyanin (KLH). Additional peptide immunogens may be generated by replacing tyrosine residues with sulfated tyrosine residues.
  • KLH keyhole limpet hemocyanin
  • Neutralizing or non-neutralizing antibodies binding to human CTLA-8 protein may also be useful therapeutics for certain tumors and also in the treatment of conditions described above. These neutralizing monoclonal antibodies are capable of blocking the ligand binding to the human CTLA-8 protein or may promote clearance of protein from the patient.
  • rat CTLA-8 proteins, herpes CTLA-8 proteins and IL-17 proteins will also possess CTLA-8 activity as described above.
  • rat and herpes CTLA-8 proteins and IL-17 proteins, as well as active fragments and variants thereof can be used in the preparation of pharmaceutical compositions and in methods of treatment as described for human CTLA-8.
  • Rat and herpes CTLA-8 proteins, and active fragments and variants thereof can be produced as described above using the polynucleotides (or fragments or variants thereof) described in SEQ ID NO:3 and SEQ ID NO:5, respectively.
  • Rat and herpes CTLA-8 may also be produced as described in Rouvier et al., J. Immunol. 1993, 150, 5445-5456.
  • CTLA-8 proteins of other species can also be used as described herein.
  • cDNAs encoding rat CTLA-8 and herpes CTLA-8 were deposited with the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209, on Jul. 6, 1995 and assigned accession numbers ATCC 69867 and ATCC 69866, respectively.
  • IL-17 proteins may also be produced as described in Golstein et al., supra.
  • the human CTLA-8 (B18) proteins of the present invention may also share some activities with IL-17.
  • any of the proteins discussed or disclosed herein may be administered by in vivo expression of the protein in a mammalian subject.
  • a polynucleotide encoding the desired protein is administered to the subject in manner allowing expression in accordance with known methods, including without limitation the adenovirus methods disclosed herein.
  • a partial clone for human CTLA-8 was isolated from a cDNA library made from RNA isolated from stimulated human peripheral blood mononuclear cells. This partial clone was identified as “B18.” B18 is sometimes used herein to refer to the human CTLA-8 of the present invention. Homology searches identified this partial clone as being related to the herpes and rat CTLA-8 genes. DNA sequence of this partial clone was used to isolate the full-length clone.
  • a directional, full-length cDNA library was constructed by standard means in the COS expression vector pMV2.
  • the cDNA library was transformed into E. coli by electroporation.
  • the bulk of the original transformed cDNA library was frozen in glycerol at ⁇ 80° C.
  • An aliquot was titered to measure the concentration of transformed E. coli .
  • the E. coli were thawed, diluted to 76,000/0.1 ml in media containing ampicillin, and 0.1 ml was distributed into the wells of a microtiter dish in an 8 ⁇ 8 array.
  • the microtiter dish was placed at 37° C. overnight to grow the E. coli.
  • PCR was performed first on the DNA preps corresponding to the eight columns.
  • the PCR consisted of two sequential reactions with nested oligonucleotides using standard conditions.
  • the oligonucleotides used for the PCR reaction were derived from the sequence of the partial B18 clone. They were:
  • B185 CACAGGCATACACAGGAAGATACATTCA (SEQ ID NO:7)
  • B183 TCTTGCTGGATGGGAACGGAATTCA (SEQ ID NO:8)
  • B18N ATACATTCACAGAAGAGCTTCCTGCACA (SEQ ID NO:9)
  • the PCR conditions were 2.5 mM MgCl 2 and 95° C. ⁇ 2 min for one cycle, 95° C. ⁇ 1 min plus 68° C. ⁇ 1 min for 30 cycles, and 68° C. ⁇ 10 for one cycle.
  • Each reaction was 20 ⁇ l.
  • the first reaction contained oligonucleotides B185 and B183 and 1 ⁇ l of the DNA preparations.
  • the second reaction contained oligonucleotides B183 and B18N and 1 ⁇ l of the first reaction.
  • DNA preps that potentially contained a full-length B18 cDNA clone were identified by agarose gel electrophoresis on an aliquot of the second PCR reaction. A DNA band of the correct mobility was assumed to be derived from a B18 cDNA.
  • E. coli containing the putative full-length B18 cDNA clone concentration of E. coli in well A2 was measured by titering and plating dilutions of the well. Then 7600 E. coli were seeded into the wells of a second microtiter plate in an 8 ⁇ 8 array. The E. coli were grown overnight; wells were pooled, and DNA was prepared as described above. To identify which of these wells contained E. coli transformed with B18, sequential PCR reactions were performed essentially as described above. Agarose gel electrophoresis identified well B2 as potentially containing a B18 cDNA.
  • the E. coli containing this cDNA was further purified by seeding wells of a microtiter plate with 253 E. coli per well and proceeding as for the purification of the E. coli in well A2.
  • Well C3 was identified as containing a putative full-length B18 cDNA clone.
  • the exact E. coli was identified by plating the contents of the well onto bacterial culture media and then screening the E. coli colonies following established protocols.
  • the probe for these hybridizations was a PCR fragment generated by doing a PCR reaction on the B18 clone using as primers the oligonucleotides described above (SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9). When a single colony was identified, DNA was prepared and sequenced by standard methods. Comparison of this sequence to the sequence of the original partial clone confirmed identity and that the isolated cDNA was full-length.
  • the full-length B18 clone for human CTLA-8 was transfected into COS cells which were then labelled with 35 S-methionine. An aliquot of conditioned medium from the transfected cell culture was reduced, denatured and electrophoresed on polyacrylamide gels. Autoradiographs of those gels are reproduced in FIG. 2 . The band indicated by the arrow demonstrates expression of human CTLA-8.
  • angiostatic activity assay endothelial cell proliferation assay.
  • the assay was done in a 96 well plate.
  • Primary human umbilical cells (HUVECs) were seeded to 2 ⁇ 10 3 cells per well in EGM medium (Clonetics)/20% FCS and incubated at 37° C. for 24 hr.
  • the cells were then starved in M199 medium (GIBCO BRL) containing 10% charcoal treated serum (M199-CS) for 48 hr at 37° C.
  • Conditioned media containing B18 (human CTLA-8) was obtained from transfected COS or stably expressing CHO cells and 1:10, 1:50, 1:250, and 1:1250 dilutions prepared in MI 99-CS medium containing 100 ng/ml FGF. The dilutions of B18 were added to the starved cells and incubated for 72 hr at 37° C. The cells were then radiolabeled by [ 3 H]-thymidine for 6 hr. Radiolabeled cells were washed with PBS and trypsinized for liquid scintillation counting. Results were plotted using Kaleidograph software. The results are shown in FIG. 3 .
  • the B18 cDNA in the expression plasmid Adori 2-12 B18 was driven by the cytomegalovirus (CMV) immediate early promoter and enhancer.
  • CMV cytomegalovirus
  • the Adori 2-12 vector was created by addition of an SV40 origin and enhancer to a known adenovirus vector (Barr et al., Gene Therapy 1:51 (1994); Davidson et al., Nature Genetics 3:219 (1993)).
  • the HindIII/BamHI fragment encoding the SV40 origin and enhancer was isolated from the pMT2 mammalian expression vector, blunted with Klenow and cloned into the NatI site (blunted with Klenow) of the Ad5 expression vector.
  • the vector was derived by digesting pNOT-B18 cDNA with SalI, filling in the 5′ overhang with Klenow to generate a blunt end and digesting with EcoRI to isolate the B18 cDNA.
  • the blunted-EcoRI B18 fragment was inserted into the restriction sites EcoRV-EcoRI of the adenovirus vector Adori 2-12.
  • the CMV-B18 expression cassette was located downstream of the SV40 origin and enhancer, and 0-1 map units of the left hand end of the adenovirus type 5(Ad5).
  • the SV40 splice donor and acceptor were located between the CMV promoter and B18 cDNA. Following the insert was SV40 poly A site, 9-16 map units of Ad5 and the puc 19 origin.
  • a recombinant adenovirus was generated by homologous recombination in 293 cells. AscI linearized Adori 2-12 B18 and ClaI digested AdCMVlacZ were introduced into the 293 cells using lipofectamine. Recombinant adenovirus virus was isolated and amplified on 293 cells. The virus was released from infected 293 cells by three cycles of freeze-thawing. The virus was further purified by two cesium chloride centrifugation gradients and dialyzed against PBS at 4° C. Following dialysis of the virus glycerol was added to a concentration of 10% and the virus was stored at ⁇ 70° C. until use. The virus was characterized by expression of the transgene, plaque forming units on 293 cells, particles/ml and Southern analysis of the virus.
  • a single dose of 5 ⁇ 10 10 particles of recombinant adenovirus encoding B18 was injected into the tail vein of male C57/b16 mice, age 7-8 weeks. Control mice received an adenovirus encoding B-galactosidase.
  • mice from each experimental group were killed on day 7 and 14.
  • Blood was collected and automated hematologic analysis was performed using a Baker 9000. Differential counts were performed on blood smears. Tissue was harvested, fixed in formalin, and stained with hematoxylin and eosin for histopathology. In the first set of experiments, serum and tissues were analyzed 7 and 14 days post injection. A slight increase in peripheral platelet counts was observed.
  • mice from each group were killed on days 7, 14, and 21.
  • serum was collected via cardiac puncture for quantitation of systemic II-6 using a commercial kit (Endogen).
  • Spleen and liver were collected for histopathology, spleen and bone marrow hematopoietic progenitors were quantitated, and bone marrow smears were prepared and stained with Wright-Geimsa for cell counts.
  • Bone marrow cellularity was calculated from pooled femurs in each group (Table III). No significant differences were observed in either group. No significant changes were observed in bone marrow hematopoietic progenitors from day 7, 14, and 21.
  • the CFU-GM, BFU-E and CFU-MEG in the B18 mice were similar to the B-gal control (Table II).
  • the bone marrow myeloid: erythroid ratios (Table IV) suggest a granulocytic hyperplasia with a possible erythroid hypoplasia in mice that received adenovirus B18 on day 7. By day 21 the ratio in the B-gal group was higher. No changes were observed in the IL6 serum levels.
  • Hematopoietic precursors were determined from pooled spleen and bone marrow samples from four animals in each group.
  • CFU-GM and BFU-E either 1 ⁇ 10 4 bone marrow cells or 1 ⁇ 10 5 spleen cells were added to complete alpha methylcelluose medium (0.9% methylcellulose in alpha medium, 30% fetal bovine serum, 1% bovine serum albumin, 10 ⁇ 4 M 2-mercaptoethanol, 2 mM L-glutamine, 2% murine spleen cell conditioned medium, and 3 U/mL erythropoietin) and aliquoted into 35 mm tissue culture dishes in a final volume of 1.0 mL.
  • B18 (human CTLA-8) was tested for the ability to induce production of factors having hematopoietic activity in a factor-dependent cell proliferation assay using the human erythroleukemic cell line, TF-1 (Kitamura et al., J. Cell Physiol. 140:323 (1989)).
  • the cells were initially grown in the presence of rhGMCSF (100 U/ml).
  • the cells were fed three days prior to setting up the assay.
  • the assay conditions were as follows:
  • MRC5 cells were incubated in the presence of human CTLA-8 (B18) and production of IL-6 and IL-8 were measured.
  • Herpes CTLA-8 (IL-17) was used as a positive control.
  • Applicants' human CTLA-8 (B18) demonstrated titratable production of both IL-6 and IL-8 (see FIGS. 6 and 7 ).

Abstract

Polynucleotides encoding human CTLA-8 and related proteins are disclosed. Human CTLA-8 proteins and methods for their production are also disclosed. Methods of treatment using human CTLA-8 proteins, rat CTLA-8 proteins and herpesvirus herpes CTLA-8 proteins are also provided.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional application of U.S. application Ser. No. 11/034,843, filed Jan. 14, 2005, which is a divisional application of U.S. application Ser. No. 10/102,080, filed Mar. 19, 2002 (U.S. Pat. No. 6,902,735), which is a continuation application of U.S. application Ser. No. 09/507,150, filed on Feb. 18, 2000 (abandoned), which in turn is a divisional application of U.S. application Ser. No. 09/034,810, filed on Mar. 4, 1998 (U.S. Pat. No. 6,043,344), which in turn is a divisional application of U.S. application Ser. No. 08/685,239, filed Jul. 18, 1996 (U.S. Pat. No. 6,074,849), which in turn is a continuation-in-part application of U.S. application Ser. No. 08/504,032, filed Jul. 19, 1995 (abandoned), and a continuation-in-part of U.S. application Ser. No. 08/514,014, filed Aug. 11, 1995 (U.S. Pat. No. 5,707,829), which in turn claims the benefit of U.S. provisional application Ser. No. 60/035,347, filed Jul. 19, 1995 (abandoned). The contents of all of the aforementioned applications are hereby incorporated by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to human CTLA-8 proteins, nucleic acids encoding such proteins, methods of treatment using such proteins. The invention also relates to the use of rat CTLA-8 proteins and herpesvirus Saimiri ORF 13 proteins in methods of treatment.
  • BACKGROUND OF THE INVENTION
  • Cytokines are secreted proteins which act on specific hematopoietic target cells to cause a differentiation event or on other target cells to induce a particular physiological response, such as secretion of proteins characteristic of inflammation. Cytokines, also variously known as lymphokines, hematopoietins, interleukins, colony stimulating factors, and the like, can be important therapeutic agents, especially for diseases or conditions in which a specific cell population is depleted. For example, erythropoietin, G-CSF, and GM-CSF, have all become important for treatment of anemia and leukopenia, respectively. Other cytokines such as interleukin-3, interleukin-6, interleukin-11 and interleukin-12 show promise in treatment of conditions such as thrombocytopenia and modulation of immune response.
  • For these reasons a significant research effort has been expended in searching for novel cytokines and cloning the DNAs which encode them. In the past, novel cytokines were identified by assaying a particular cell such as a bone marrow cell, for a measurable response, such as proliferation. The search for novel cytokines has thus been limited by the assays available, and if a novel cytokine has an activity which is unmeasurable by a known assay, the cytokine remains undetectable. In a newer approach, cDNAs encoding cytokines have been detected using the polymerase chain reaction (PCR) and oligonucleotide primers having homology to shared motifs of known cytokines or their receptors. The PCR approach is also limited by the necessity for knowledge of previously cloned cytokines in the same protein family. Cytokines have also been cloned using subtractive hybridization to construct and screen cDNA libraries, or they can potentially be cloned using PCR followed by gel electrophoresis to detect differentially expressed genes. The subtractive hybridization methods are based on the assumption that cytokine mRNAs are those that are differentially expressed, and these methods do not require any prior knowledge of the sequence of interest. However, many cytokines may be encoded by mRNAs which are not differentially expressed, and thus are undetectable using these methods.
  • It would be desirable to develop new methods for identifying novel cytokines and other secreted factors and to isolate polynucleotides encoding them.
  • SUMMARY OF THE INVENTION
  • In developing the present invention, methods were employed which selectively identify polynucleotides which encode secreted proteins. One such polynucleotide was isolated which encodes “human CTLA-8.” In accordance with the present invention, polynucleotides encoding human CTLA-8 (now known as IL-17F) and active fragments thereof are disclosed. “CTLA-8” (now known as IL-17F) is used throughout the present specification to refer to both proteins and polynucleotides encoding those proteins and to refer to proteins and polynucleotides from all mammalian species.
  • In certain embodiments, the present invention provides an isolated polynucleotide comprising a nucleotide sequence selected from the group consisting of:
  • (a) the nucleotide sequence of SEQ ID NO:1 from nucleotide 146 to nucleotide 544;
  • (b) a nucleotide sequence capable of hybridizing to a nucleic acid sequence specified in (a);
  • (c) a nucleotide sequence varying from the sequence of the nucleotide sequence specified in (a) as a result of degeneracy of the genetic code; and
  • (d) an allelic variant of the nucleotide sequence specified in (a).
  • Preferably, the polynucleotide of the invention encodes a protein having CTLA-8 activity. In other embodiments the polynucleotide is operably linked to an expression control sequence. In other preferred embodiments, the polynucleotide is contained in a vector suitable for in vivo expression in a mammalian subject. Polynucleotides comprising the nucleotide sequence of SEQ ID NO:1 from nucleotide 55 to nucleotide 544, the nucleotide sequence of SEQ ID NO:1 from nucleotide 139 to nucleotide 544 or the nucleotide sequence of SEQ ID NO:1 from nucleotide 86 to nucleotide 544 are particularly preferred.
  • Host cells transformed with the polynucleotides of the invention are also provided, including mammalian cells.
  • Processes are also provided for producing a human CTLA-8 protein, said processes comprising:
  • (a) growing a culture of the host cell of the invention in a suitable culture medium; and
  • (b) purifying the human CTLA-8 protein from the culture.
  • Isolated human CTLA-8 protein is also provided which comprises an amino acid sequence selected from the group consisting of:
  • (a) the amino acid sequence of SEQ ID NO:2;
  • (b) the amino acid sequence of SEQ ID NO:2 from amino acids 11 to 163;
  • (c) the amino acid sequence of SEQ ID NO:2 from amino acids 29 to 163;
  • (d) the amino acid sequence of SEQ ID NO:2 from amino acids 31 to 163; and
  • (e) fragments of (a), (b), (c) or (d) having CTLA-8 activity.
  • Proteins comprising the amino acid sequence of SEQ ID NO:2 and comprising the sequence from amino acids 29 to 163, from amino acid 31 to 163, or from amino acids 11 to 163 of SEQ ID NO:2 are particularly preferred. Preferably, the protein has CTLA-8 activity. Pharmaceuticals composition comprising a human CTLA-8 protein of the invention and a pharmaceutically acceptable carrier are also provided.
  • Compositions are also disclosed which comprise an antibody which specifically reacts with a human CTLA-8 protein of the invention.
  • Methods of treating a mammalian subject are also provided which comprise administering a therapeutically effective amount of a pharmaceutical composition comprising a human CTLA-8 protein.
  • Rat CTLA-8 and active (i.e., having CTLA-8 activity) fragments thereof may also be used in such methods of treatment, Preferably the rat protein is administered as a composition comprising a pharmaceutically acceptable carrier and a protein comprising an amino acid sequence selected from the group consisting of:
  • (a) the amino acid sequence of SEQ ID NO:4;
  • (b) the amino acid sequence of SEQ ID NO:4 from amino acids 18 to 150; and
  • (c) fragments of (a) or (b) having CTLA-8 activity.
  • Herpesvirus Saimiri ORF13, referred to herein as “herpes CTLA-8”, and active (i.e., having CTLA-8 activity) fragments thereof and active fragments thereof may also be used in such methods of treatment. Preferably the herpes CTLA-8 protein is administered as a composition comprising a pharmaceutically acceptable carrier and a protein comprising an amino acid sequence selected from the group consisting of:
  • (a) the amino acid sequence of SEQ ID NO:6;
  • (b) the amino acid sequence of SEQ ID NO:6 from amino acids 19 to 151; and
  • (c) fragments of (a) or (b) having CTLA-8 activity.
  • The invention also provides a method of treating a mammalian subject comprising administering a therapeutically effective amount of a composition comprising a pharmaceutically acceptable carrier and IL-17 or an active fragment thereof.
  • In methods of treatment provided by the present invention, preferably the subject is treated to produce an effect selected from the group consisting of inhibition of angiogenesis, inhibition of growth or proliferation of vascular endothelial cells, inhibition of tumor growth, inhibition of angiogenesis-dependent tissue growth, proliferation of myeloid cells or progenitors, proliferation of erythroid cells or progenitors, proliferation of lymphoid cells or progenitors, induction of IFNγ production, induction of IL-3 production and induction of GM-CSF production.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a comparison of homologous regions of the amino acid sequences of human CTLA-8 (indicated as “B18_F1”), rat CTLA-8 (indicated as “Musctla8”) and herpes CTLA-8 (indicated as “Hsvie 2”).
  • FIG. 2 depicts autoradiographs demonstrating expression of human CTLA-8 in COS cells.
  • FIG. 3 presents data relating to the ability of human CTLA-8 to inhibit angiogenesis.
  • FIGS. 4 and 5 present data relating to the ability of human CTLA-8 to produce or induce hematopoietic activity.
  • FIGS. 6 and 7 present data demonstrating the ability of human CTLA-8 to induce production of IL-6 and IL-8.
  • DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
  • The inventors of the present application have identified and provided a polynucleotide encoding a human CTLA-8 protein. SEQ ID NO:1 provides the nucleotide sequence of a cDNA encoding the human CTLA-8 protein. SEQ ID NO:2 provides the amino acid sequence of the human CTLA-8 protein. Alternatively, the initiating methionine may be at amino acid 11 of SEQ ID NO:2. On the basis of amino terminal sequencing, the mature protein sequence is believed to begin at amino acid 31 of SEQ ID NO:2 (encoded by the sequence beginning with nucleotide 146 of SEQ ID NO:1).
  • The region from amino acid 29 to amino acid 163 of human CTLA-8 (SEQ ID NO:2) shows marked homology to portions of rat CTLA-8 (amino acids 18 to 150 of SEQ ID NO:4) and herpesvirus Saimiri ORF13 (“herpes CTLA-8”) (amino acids 19 to 151 of SEQ ID NO:5). A cDNA sequence encoding rat CTLA-8 is listed at SEQ ID NO:3 and its corresponding amino acid sequence is reported at SEQ ID NO:4. A cDNA sequence encoding herpes CTLA-8 is listed at SEQ ID NO:5 and its corresponding amino acid sequence is reported at SEQ ID NO:6. Homology between rat CTLA-8 and herpes CTLA-8 was reported by Rouvier et al., J. Immunol. 1993, 150, 5445-5456.
  • Applicants had previously incorrectly identified the rat sequences of SEQ ID NO:3 and SEQ ID NO:4 as applying to murine CTLA-8. Applicants' human CTLA-8 (B 18) does also show homology to the true murine CTLA-8 sequence.
  • Golstein et al. (WO95/18826; Fossiez et al., Microbial Evasion and Subversion of Immunity 544:3222 (Abstract)) have also reported a species they initially identified as “human CTLA-8.” However, examination of the sequence of the Golstein et al. species and the human CTLA-8 (B18) sequence of the present invention readily reveals that they are two different proteins, although they are homologous with each other and with the rat CTLA-8 and herpes CTLA-8 identified herein. The Golstein et al. species has now been renamed as interleukin-17 (IL-17). Because of the homology between applicants' human CTLA-8 (B18) and IL-17, these proteins are expected to share some activities.
  • It has also been preliminarily determined that human CTLA-8 (B 18) forms homodimers when expressed. As a result, human CTLA-8 proteins may possess activity in either monomeric or dimeric forms. Human CTLA-8 proteins can also be produced as heterodimers with rat and herpes CTLA-8 proteins and with human IL-17. These heterodimers are also expected to have activities of the proteins of which they are comprised.
  • Forms of human CTLA-8 protein of less than full length are encompassed within the present invention and may be produced by expressing a corresponding fragment of the polynucleotide encoding the human CTLA-8 protein (SEQ ID NO:1). These corresponding polynucleotide fragments are also part of the present invention. Modified polynucleotides as described above may be made by standard molecular biology techniques, including site-directed mutagenesis methods which are known in the art or by the polymerase chain reaction using appropriate oligonucleotide primers.
  • For the purposes of the present invention, a protein has “CTLA-8 activity” if it either (1) displays biological activity in a factor-dependent cell proliferation assay (preferably an assay in which the corresponding species' full-length CTLA-8 is active) (including without limitation those assays described below), or (2) induces expression or secretion of γ-IFN, or (3) displays chemoattractant or chemotactic activity in a chemoattraction or chemotaxis assay (preferably an assay in which the corresponding species' full-length CTLA-8 is active) or (4) induces expression or secretion of IL-3 or GM-CSF.
  • Human CTLA-8 protein or fragments thereof having CTLA-8 activity may be fused to carrier molecules such as immunoglobulins. For example, human CTLA-8 protein may be fused through “linker” sequences to the Fc portion of an immunoglobulin.
  • The invention also encompasses allelic variations of the nucleotide sequence as set forth in SEQ ID NO:1, that is, naturally-occurring alternative forms of the isolated polynucleotide of SEQ ID NO:1 which also encode human CTLA-8 or CTLA-8 proteins having CTLA-8 activity. Also included in the invention are isolated polynucleotides which hybridize to the nucleotide sequence set forth in SEQ ID NO:1 under highly stringent (0.2×SSC at 65° C.), stringent (e.g. 4×SSC at 65° C. or 50% formamide and 4×SSC at 42° C.), or relaxed (4×SSC at 50° C. or 30-40% formamide and 4×SSC at 42° C.) conditions. Isolated polynucleotides which encode human CTLA-8 protein but which differ from the nucleotide sequence set forth in SEQ ID NO:1 by virtue of the degeneracy of the genetic code are also encompassed by the present invention. Variations in the nucleotide sequence as set forth in SEQ ID NO:1 which are caused by point mutations or by induced modifications which enhance CTLA-8 activity, half-life or production level are also included in the invention.
  • The isolated polynucleotides of the invention may be operably linked to an expression control sequence such as the pMT2 or pED expression vectors disclosed in Kaufman et al., Nucleic Acids Res. 19, 4485-4490 (1991), in order to produce the CTLA-8 protein recombinantly. Many suitable expression control sequences are known in the art. General methods of expressing recombinant proteins are also known and are exemplified in R. Kaufman, Methods in Enzymology 185, 537-566 (1990). As defined herein “operably linked” means enzymatically or chemically ligated to form a covalent bond between the isolated polynucleotide of the invention and the expression control sequence, in such a way that the CTLA-8 protein is expressed by a host cell which has been transformed (transfected) with the ligated polynucleotide/expression control sequence.
  • A number of types of cells may act as suitable host cells for expression of the human CTLA-8 protein. Any cell type capable of expressing functional human CTLA-8 protein may be used. Suitable mammalian host cells include, for example, monkey COS cells, Chinese Hamster Ovary (CHO) cells, human kidney 293 cells, human epidermal A431 cells, human Colo205 cells, 3T3 cells, CV-1 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HeLa cells, mouse L cells, BHK, HL-60, U937, HaK, Rat2, BaF3, 32D, FDCP-1, PC12 or C2C12 cells.
  • The human CTLA-8 protein may also be produced by operably linking the isolated polynucleotide of the invention to suitable control sequences in one or more insect expression vectors, and employing an insect expression system. Materials and Methods for baculovirus/insect cell expression systems are commercially available in kit form from, e.g., Invitrogen, San Diego, Calif., U.S.A. (the MaxBac® kit), and such methods are well known in the art, as described in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987), incorporated herein by reference. Soluble forms of the human CTLA-8 protein may also be produced in insect cells using appropriate isolated polynucleotides as described above.
  • Alternatively, the human CTLA-8 protein may be produced in lower eukaryotes such as yeast or in prokaryotes such as bacteria. Suitable yeast strains include Saccharomyces cerevisiae, Schizosaccharomyces pombe, Kluyveromyces strains, Candida, or any yeast strain capable of expressing heterologous proteins. Suitable bacterial strains include Escherichia coli, Bacillus subtilis, Salmonella typhimurium, or any bacterial strain capable of expressing heterologous proteins.
  • The human CTLA-8 protein of the invention may also be expressed as a product of transgenic animals, e.g., as a component of the milk of transgenic cows, goats, pigs, or sheep which are characterized by somatic or germ cells containing a polynucleotide sequence encoding the human CTLA-8 protein.
  • The human CTLA-8 protein of the invention may be prepared by growing a culture of transformed host cells under culture conditions necessary to express the desired protein. The resulting expressed protein may then be purified from the culture medium or cell extracts. Soluble forms of the human CTLA-8 protein of the invention can be purified from conditioned media. Membrane-bound forms of human CTLA-8 protein of the invention can be purified by preparing a total membrane fraction from the expressing cell and extracting the membranes with a non-ionic detergent such as Triton X-100.
  • The human CTLA-8 protein can be purified using methods known to those skilled in the art. For example, the human CTLA-8 protein of the invention can be concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit. Following the concentration step, the concentrate can be applied to a purification matrix such as a gel filtration medium. Alternatively, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification. Alternatively, a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred (e.g., S-Sepharose® columns). The purification of the human CTLA-8 protein from culture supernatant may also include one or more column steps over such affinity resins as concanavalin A-agarose, heparin-toyopearl® or Cibacrom blue 3GA Sepharose®; or by hydrophobic interaction chromatography using such resins as phenyl ether, butyl ether, or propyl ether; or by immunoaffinity chromatography. Finally, one or more reverse-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups, can be employed to further purify the human CTLA-8 protein. Some or all of the foregoing purification steps, in various combinations or with other known methods, can also be employed to provide a substantially purified isolated recombinant protein.
  • Preferably, the human CTLA-8 protein is purified so that it is substantially free of other mammalian proteins.
  • It is believed that human CTLA-8, active fragments and variants thereof, and CTLA-8 related proteins (such as, for example, rat CTLA-8 and herpes CTLA-8) (collectively “CTLA-8 proteins”) possess or induce cytokine activities. Human CTLA-8 expression correlated with γ-IFN expression in induced primary cells and can induce the expression of IL-3 and/or GM-CSF, which expression can in turn produce effects associated with the induced cytokine. Therefore, human CTLA-8 and CTLA-8 related proteins may have an effect on proliferation or function of myeloid cells, erythroid cells, lymphoid cells and their progenitors. Human CTLA-8 proteins may also play a role in formation of platelets or their progenitors.
  • A protein of the present invention may exhibit cytokine, cell proliferation (either inducing or inhibiting) or cell differentiation (either inducing or inhibiting) activity or may induce production of other cytokines in certain cell populations. Many protein factors discovered to date, including all known cytokines, have exhibited activity in one or more factor dependent cell proliferation assays, and hence the assays serve as a convenient confirmation of cytokine activity. The activity of a protein of the present invention is evidenced by any one of a number of routine factor dependent cell proliferation assays for cell lines including, without limitation, 32D, DA2, DA1G, T10, B9, B9/11, BaF3, MC9/G, M+(preB M+), 2E8, RB5, DA1, 123, T1165, HT2, CTLL2, TF-1, Mo7e and CMK.
  • The activity of a protein of the invention may, among other means, be measured by the following methods:
  • Assays for T-cell or thymocyte proliferation include without limitation those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W. Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Takai et al., J. Immunol. 137:3494-3500, 1986; Bertagnolli et al., J. Immunol. 145:1706-1712, 1990; Bertagnolli et al., Cellular Immunology 133:327-341, 1991; Bertagnolli, et al., J. Immunol. 149:3778-3783, 1992; Bowman et al., J. Immunol. 152: 1756-1761, 1994.
  • Assays for cytokine production and/or proliferation of spleen cells, lymph node cells or thymocytes include, without limitation, those described in: Polyclonal T cell stimulation, Kruisbeek, A. M. and Shevach, E. M. In Current Protocols in Immunology. J. E Coligan et al. eds. Vol 1 pp. 3.12.1-3.12.14, John Wiley and Sons, Toronto. 1994; and Measurement of mouse and human Interferon γ, Schreiber, R. D. In Current Protocols in Immunology. J. E Coligan et al. eds. Vol 1 pp. 6.8.1-6.8.8, John Wiley and Sons, Toronto. 1994.
  • Assays for proliferation and differentiation of hematopoietic and lymphopoietic cells include, without limitation, those described in: Measurement of Human and Murine Interleukin 2 and Interleukin 4, Bottomly, K., Davis, L. S, and Lipsky, P. E. In Current Protocols in Immunology. J. E Coligan et al. eds. Vol 1 pp. 6.3.1-6.3.12, John Wiley and Sons, Toronto. 1991; deVries et al., J. Exp. Med. 173:1205-1211, 1991; Moreau et al., Nature 336:690-692, 1988; Greenberger et al., Proc. Natl. Acad. Sci. U.S.A. 80:2931-2938, 1983; Measurement of mouse and human interleukin 6-Nordan, R. In Current Protocols in Immunology. J. E Coligan et al. eds. Vol 1 pp. 6.6.1-6.6.5, John Wiley and Sons, Toronto. 1991; Smith et al., Proc. Natl. Acad. Sci. U.S.A. 83:1857-1861, 1986; Measurement of human Interleukin 11-Bennett, F., Giannotti, J., Clark, S. C. and Turner, K. J. In Current Protocols in Immunology. J. E Coligan et al. eds. Vol 1 pp. 6.15.1 John Wiley and Sons, Toronto. 1991; Measurement of mouse and human Interleukin 9-Ciarletta, A., Giannotti, J., Clark, S. C. and Turner, K. J. In Current Protocols in Immunology. J. E Coligan et al. eds. Vol 1 pp. 6.13.1, John Wiley and Sons, Toronto. 1991.
  • Assays for T-cell clone responses to antigens (which will identify, among others, proteins that affect APC-T cell interactions as well as direct T-cell effects by measuring proliferation and cytokine production) include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W. Strober Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function; Chapter 6, Cytokines and their cellular receptors; Chapter 7, Immunologic studies in Humans); Weinberger et al., Proc. Natl. Acad. Sci. USA 77:6091-6095, 1980; Weinberger et al., Eur. J. Immun. 11:405-411, 1981; Takai et al., J. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988.
  • A protein of the present invention may also exhibit immune stimulating or immune suppressing activity, including without limitation the activities for which assays are described herein. A protein may be useful in the treatment of various immune deficiencies and disorders (including severe combined immunodeficiency (SCID)), e.g., in regulating (up or down) growth and proliferation of T and/or B lymphocytes, as well as effecting the cytolytic activity of NK cells and other cell populations. These immune deficiencies may be genetic or be caused by viral (e.g., HIV) as well as bacterial or fungal infections, or may result from autoimmune disorders. More specifically, infectious diseases caused by viral, bacterial, fungal or other infection may be treatable using a protein of the present invention, including infections by HIV, hepatitis viruses, herpes viruses, mycobacteria, leshmania, malaria and various fungal infections such as candida. Of course, in this regard, a protein of the present invention may also be useful where a boost to the immune system generally would be indicated, i.e., in the treatment of cancer.
  • Autoimmune disorders which may be treated using a protein of the present invention include, for example, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes mellitis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease. Such a protein of the present invention may also to be useful in the treatment of allergic reactions and conditions, such as asthma or other respiratory problems. Other conditions, in which immune suppression is desired (including, for example, asthma and related respiratory conditions), may also be treatable using a protein of the present invention.
  • A protein of the present invention may also suppress chronic or acute inflammation, such as, for example, that associated with infection (such as septic shock or systemic inflammatory response syndrome (SIRS)), inflammatory bowel disease, Crohn's disease or resulting from over production of cytokines such as TNF or IL-1 (such as the effect demonstrated by IL-11).
  • The activity of a protein of the invention may, among other means, be measured by the following methods:
  • Suitable assays for thymocyte or splenocyte cytotoxicity include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Herrmann et al., Proc. Natl. Acad. Sci. USA 78:2488-2492, 1981; Herrmann et al., J. Immunol. 128:1968-1974, 1982; Handa et al., J. Immunol. 135:1564-1572, 1985; Takai et al., J. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988; Herrmann et al., Proc. Natl. Acad. Sci. USA 78:2488-2492, 1981; Herrmann et al., J. Immunol. 128:1968-1974, 1982; Ianda et al., J. Immunol. 135:1564-1572, 1985; Takai et al., J. Immunol. 137:3494-3500, 1986; Bowman et al., J. Virology 61:1992-1998; Takai et al., J. Immunol. 140:508-512, 1988; Bertagnolli et al., Cellular Immunology 133:327-341, 1991; Brown et al., J. Immunol. 153:3079-3092, 1994.
  • Assays for T-cell-dependent immunoglobulin responses and isotype switching (which will identify, among others, proteins that modulate T-cell dependent antibody responses and that affect Th1/Th2 profiles) include, without limitation, those described in: Maliszewski, J. Immunol. 144:3028-3033, 1990; and Assays for B cell function: In vitro antibody production, Mond, J. J. and Brunswick, M. In Current Protocols in Immunology. J. E Coligan et al. eds. Vol 1 pp. 3.8.1-3.8.16, John Wiley and Sons, Toronto. 1994.
  • Mixed lymphocyte reaction (MLR) assays (which will identify, among others, proteins that generate predominantly Th1 and CTL responses) include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, W Strober, Pub. Greene Publishing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Takai et al., J. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988; Bertagnolli et al., J. Immunol. 149:3778-3783, 1992.
  • Dendritic cell-dependent assays (which will identify, among others, proteins expressed by dendritic cells that activate naive T-cells) include, without limitation, those described in: Guery et al., J. Immunol. 134:536-544, 1995; Inaba et al., Journal of Experimenal Medicine 173:549-559, 1991; Macatonia et al., Journal of Immunology 154:5071-5079, 1995; Porgador et al., Journal of Experimental Medicine 182:255-260, 1995; Nair et al., Journal of Virology 67:4062-4069, 1993; Huang et al., Science 264:961-965, 1994; Macatonia et al., Journal of Experimental Medicine 169:1255-1264, 1989; Bhardwaj et al., Journal of Clinical Investigation 94:791-807, 1994; and Inaba et al., Journal of Experimental Medicine 172:631-640, 1990.
  • Assays for lymphocyte survival/apoptosis (which will identify, among others, proteins that prevent apoptosis after superantigen induction and proteins that regulate lymphocyte homeostasis) include, without limitation, those described in: Darzynkiewicz et al., Cytometry 13:795-808, 1992; Gorczyca et al., Leukemia 7:659-670, 1993; Gorczyca et al., Cancer Research 53:1945-1951, 1993; Itoh et al., Cell 66:233-243, 1991; Zacharchuk, Journal of Immunology 145:4037-4045, 1990; Zamai et al., Cytometry 14:891-897, 1993; Gorczyca et al., International Journal of Oncology 1:639-648, 1992.
  • Assays for proteins that influence early steps of T-cell commitment and development include, without limitation, those described in: Antica et al., Blood 84:111-117, 1994; Fine et al., Cellular Immunology 155:111-122, 1994; Galy et al., Blood 85:2770-2778, 1995; Toki et al., Proc. Nat. Acad. Sci. USA 88:7548-7551, 1991.
  • A protein of the present invention may be useful in regulation of hematopoiesis and, consequently, in the treatment of myeloid or lymphoid cell deficiencies. Even marginal biological activity in support of colony forming cells or of factor-dependent cell lines indicates involvement in regulating hematopoiesis, e.g. in supporting the growth and proliferation of erythroid progenitor cells alone or in combination with other cytokines, thereby indicating utility, for example, in treating various anemias or for use in conjunction with irradiation/chemotherapy to stimulate the production of erythroid precursors and/or erythroid cells; in supporting the growth and proliferation of myeloid cells such as granulocytes and monocytes/macrophages (i.e., traditional CSF activity) useful, for example, in conjunction with chemotherapy to prevent or treat consequent myelo-suppression; in supporting the growth and proliferation of megakaryocytes and consequently of platelets thereby allowing prevention or treatment of various platelet disorders such as thrombocytopenia, and generally for use in place of or complementary to platelet transfusions; and/or in supporting the growth and proliferation of hematopoietic stem cells which are capable of maturing to any and all of the above-mentioned hematopoietic cells and therefore find therapeutic utility in various stem cell disorders (such as those usually treated with transplantation, including, without limitation, aplastic anemia and paroxysmal nocturnal hemoglobinuria), as well as in repopulating the stem cell compartment post irradiation/chemotherapy, either in-vivo or ex-vivo (i.e. in conjunction with bone marrow transplantation) as normal cells or genetically manipulated for gene therapy.
  • The activity of a protein of the invention may, among other means, be measured by the following methods:
  • Suitable assays for proliferation and differentiation of various hematopoietic lines are cited above.
  • Assays for embryonic stem cell differentiation (which will identify, among others, proteins that influence embyronic differentation hematopoiesis) include, without limitation, those described in: Johansson et al. Cellular Biology 15:141-151, 1995; Keller et al., Molecular and Cellular Biology 13:473-486, 1993; McClanahan et al., Blood 81:2903-2915, 1993.
  • Assays for stem cell survival and differentiation (which will identify, among others, proteins that regulate lympho-hematopoiesis) include, without limitation, those described in: Methylcellulose colony forming assays, Freshney, M. G. In Culture of Hematopoietic Cells. R. I. Freshney, et al. eds. pp. 265-268, Wiley-Liss, Inc., New York, N.Y. 1994; Hirayama et al., Proc. Natl. Acad. Sci. USA 89:5907-5911, 1992; Primitive hematopoietic colony forming cells with high proliferative potential, McNiece, I. K. and Briddell, R. A. In Culture of Hematopoietic Cells. R. I. Freshney, et al. eds. pp. 23-39, Wiley-Liss, Inc., New York, N.Y. 1994; Neben et al., Experimental Hematology 22:353-359, 1994; Cobblestone area forming cell assay, Ploemacher, R. E. In Culture of Hematopoietic Cells. R. I. Freshney, et al. eds. pp. 1-21, Wiley-Liss, Inc., New York, N.Y. 1994; Long term bone marrow cultures in the presence of stromal cells, Spooncer, E., Dexter, M. and Allen, T. In Culture of Hematopoietic Cells. R. I. Freshney, et al. eds. pp. 163-179, Wiley-Liss, Inc., New York, N.Y. 1994; Long term culture initiating cell assay, Sutherland, H. J. In Culture of Hematopoietic Cells. R. I. Freshney, et al. eds. pp. 139-162, Wiley-Liss, Inc., New York, N.Y. 1994.
  • CTLA-8 proteins are useful in the treatment of various immune deficiencies and disorders (including SCID), e.g., in regulating (up or down) growth, proliferation and/or activity of T and/or B lymphocytes, as well as the cytolytic activity of NK cells. These immune deficiencies may be caused by viral (e.g., HIV) as well as bacterial infections, or may result from autoimmune disorders. More specifically, infectious diseases caused by viral, bacterial, fungal or other infection may be treatable using CTLA-8 proteins, including infections by HIV, hepatitis, influenza, CMV, herpes, mycobacterium, leishmaniasis, malaria and various fungal infections (such as candida). Of course, in this regard, the CTLA-8 proteins may also be useful where a boost to the immune system generally would be indicated, i.e., in the treatment of cancer or as an adjuvant to vaccines. Autoimmune disorders which may be treated using factors of the present invention include, for example, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, Guillain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes mellitis and autoimmune inflammatory eye disease. The CTLA-8 proteins are also expected to be useful in the treatment of allergic reactions and conditions.
  • CTLA-8 proteins are also expected to have chemotactic activity. A protein or peptide has “chemotactic activity,” as used herein, if it can stimulate, directly or indirectly, the directed orientation or movement of cells, including myeloid and lymphoid cells. Preferably, the protein or peptide has the ability to directly stimulate directed movement of cells (particularly T-cells). Whether a particular protein or peptide has chemotactic activity for cells can be readily determined by employing such protein or peptide in any known assay for cell chemotaxis.
  • CTLA-8 proteins also inhibit growth and proliferation of vascular endothelial cells. As a result, human CTLA-8 proteins are effective in inhibiting angiogenesis (i.e., vascular formation). This activity will also be useful in the treatment of tumors and other conditions in which angiogenesis is involved. Inhibition of angiogenesis by human CTLA-8 proteins will also result in inhibition or prevention of the condition to which normal angiogenesis would contribute.
  • Isolated CTLA-8 proteins, purified from cells or recombinantly produced, may be used as a pharmaceutical composition when combined with a pharmaceutically acceptable carrier. Such a composition may contain, in addition to CTLA-8 protein and carrier, diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art. The term “pharmaceutically acceptable” means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier will depend on the route of administration. The pharmaceutical composition of the invention may also contain cytokines, lymphokines, or other hematopoietic factors such as M-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-15, G-CSF, γ-IFN, stem cell factor, and erythropoietin. The pharmaceutical composition may contain thrombolytic or anti-thrombotic factors such as plasminogen activator and Factor VIII. The pharmaceutical composition may further contain other anti-inflammatory agents. Such additional factors and/or agents may be included in the pharmaceutical composition to produce a synergistic effect with CTLA-8 protein, or to minimize side effects caused by the CTLA-8 protein. Conversely, CTLA-8 protein may be included in formulations of the particular cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent to minimize side effects of the cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent.
  • The pharmaceutical composition of the invention may be in the form of a liposome. In the liposome, CTLA-8 protein and pharmaceutically acceptable carriers are combined with amphipathic agents such as lipids, which exist in aggregated forms, such as micelles, insoluble monolayers, liquid crystals, or lamellar layers, in aqueous solution. Suitable lipids for liposomal formulation include, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. Preparation of such liposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Pat. No. 4,235,871; U.S. Pat. No. 4,501,728; U.S. Pat. No. 4,837,028; and U.S. Pat. No. 4,737,323, all of which are incorporated herein by reference.
  • As used herein, the term “therapeutically effective amount” means the total amount of each active component of the pharmaceutical composition or method that is sufficient to show a meaningful patient benefit, e.g., amelioration of symptoms of, healing of, or increase in rate of healing of such conditions. When applied to an individual active ingredient, administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
  • In practicing the method of treatment or use of the present invention, a therapeutically effective amount of CTLA-8 protein is administered to a mammal. CTLA-8 protein may be administered in accordance with the method of the invention either alone or in combination with other therapies such as treatments employing cytokines, lymphokines or other hematopoietic factors. When co-administered with one or more cytokines, lymphokines, other hematopoietic factors or vaccine components (such as antigens or other adjuvants), CTLA-8 protein may be administered either simultaneously with the cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors, or sequentially. If administered sequentially, the attending physician will decide on the appropriate sequence of administering CTLA-8 protein in combination with cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors.
  • Administration of CTLA-8 protein used in the pharmaceutical composition or to practice the method of the present invention can be carried out in a variety of conventional ways, such as oral ingestion, inhalation, or cutaneous, subcutaneous, or intravenous injection. Intravenous administration to the patient is preferred.
  • When a therapeutically effective amount of CTLA-8 protein is administered orally, CTLA-8 protein will be in the form of a tablet, capsule, powder, solution or elixir. When administered in tablet form, the pharmaceutical composition of the invention may additionally contain a solid carrier such as a gelatin or an adjuvant. The tablet, capsule, and powder contain from about 5 to 95% CTLA-8 protein, and preferably from about 25 to 90% CTLA-8 protein. When administered in liquid form, a liquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oil, soybean oil, or sesame oil, or synthetic oils may be added. The liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol. When administered in liquid form, the pharmaceutical composition contains from about 0.5 to 90% by weight of CTLA-8 protein, and preferably from about 1 to 50% CTLA-8 protein.
  • When a therapeutically effective amount of CTLA-8 protein is administered by intravenous, cutaneous or subcutaneous injection, CTLA-8 protein will be in the form of a pyrogen-free, parenterally acceptable aqueous solution. The preparation of such parenterally acceptable protein solutions, having due regard to pH, isotonicity, stability, and the like, is within the skill in the art. A preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to CTLA-8 protein an isotonic vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or other vehicle as known in the art. The pharmaceutical composition of the present invention may also contain stabilizers, preservatives, buffers, antioxidants, or other additive known to those of skill in the art.
  • The amount of CTLA-8 protein in the pharmaceutical composition of the present invention will depend upon the nature and severity of the condition being treated, and on the nature of prior treatments which the patient has undergone. Ultimately, the attending physician will decide the amount of CTLA-8 protein with which to treat each individual patient. Initially, the attending physician will administer low doses of CTLA-8 protein and observe the patient's response. Larger doses of CTLA-8 protein may be administered until the optimal therapeutic effect is obtained for the patient, and at that point the dosage is not generally increased further. It is contemplated that the various pharmaceutical compositions used to practice the method of the present invention should contain about 0.1 μg to about 100 mg of CTLA-8 protein per kg body weight, preferably about 0.1 μg to about 10 mg of CTLA-8 protein per kg body weight, more preferably about 0.1 μg to about 100 μg of CTLA-8 protein per kg body weight, most preferably preferably about 0.1 μg to about 10 μg of CTLA-8 protein per kg body weight.
  • The duration of intravenous therapy using the pharmaceutical composition of the present invention will vary, depending on the severity of the disease being treated and the condition and potential idiosyncratic response of each individual patient. It is contemplated that the duration of each application of the CTLA-8 protein will be in the range of 12 to 24 hours of continuous intravenous administration. Ultimately the attending physician will decide on the appropriate duration of intravenous therapy using the pharmaceutical composition of the present invention.
  • CTLA-8 protein of the invention may also be used to immunize animals to obtain polyclonal and monoclonal antibodies which specifically react with the CTLA-8 protein and which may inhibit CTLA-8 binding to its receptor. Such antibodies are also useful for performing diagnostics assays for CTLA-8 in accordance with known methods. Such antibodies may be obtained using the entire CTLA-8 protein as an immunogen, or by using fragments of human CTLA-8 protein. The peptide immunogens additionally may contain a cysteine residue at the carboxyl terminus, and are conjugated to a hapten such as keyhole limpet hemocyanin (KLH). Additional peptide immunogens may be generated by replacing tyrosine residues with sulfated tyrosine residues. Methods for synthesizing such peptides are known in the art, for example, as in R. P. Merrifield, J. Amer. Chem. Soc. 85, 2149-2154 (1963); J. L. Krstenansky, et al., FEBS Lett. 211, 10 (1987).
  • Neutralizing or non-neutralizing antibodies (preferably monoclonal antibodies) binding to human CTLA-8 protein may also be useful therapeutics for certain tumors and also in the treatment of conditions described above. These neutralizing monoclonal antibodies are capable of blocking the ligand binding to the human CTLA-8 protein or may promote clearance of protein from the patient.
  • Because of their homology to human CTLA-8, rat CTLA-8 proteins, herpes CTLA-8 proteins and IL-17 proteins (the “human CTLA-8” of Golstein et al., supra) will also possess CTLA-8 activity as described above. As a result, rat and herpes CTLA-8 proteins and IL-17 proteins, as well as active fragments and variants thereof, can be used in the preparation of pharmaceutical compositions and in methods of treatment as described for human CTLA-8. Rat and herpes CTLA-8 proteins, and active fragments and variants thereof, can be produced as described above using the polynucleotides (or fragments or variants thereof) described in SEQ ID NO:3 and SEQ ID NO:5, respectively. Rat and herpes CTLA-8 may also be produced as described in Rouvier et al., J. Immunol. 1993, 150, 5445-5456. CTLA-8 proteins of other species can also be used as described herein. cDNAs encoding rat CTLA-8 and herpes CTLA-8 were deposited with the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209, on Jul. 6, 1995 and assigned accession numbers ATCC 69867 and ATCC 69866, respectively. IL-17 proteins may also be produced as described in Golstein et al., supra.
  • Because of its homology to IL-17, the human CTLA-8 (B18) proteins of the present invention may also share some activities with IL-17.
  • For the purposes of treatment or therapy, any of the proteins discussed or disclosed herein may be administered by in vivo expression of the protein in a mammalian subject. In such instances, a polynucleotide encoding the desired protein is administered to the subject in manner allowing expression in accordance with known methods, including without limitation the adenovirus methods disclosed herein.
  • Example 1 Isolation of Human CTLA-8 cDNA
  • A partial clone for human CTLA-8 was isolated from a cDNA library made from RNA isolated from stimulated human peripheral blood mononuclear cells. This partial clone was identified as “B18.” B18 is sometimes used herein to refer to the human CTLA-8 of the present invention. Homology searches identified this partial clone as being related to the herpes and rat CTLA-8 genes. DNA sequence of this partial clone was used to isolate the full-length clone.
  • In order to isolate a full-length cDNA for B18, a directional, full-length cDNA library was constructed by standard means in the COS expression vector pMV2. The cDNA library was transformed into E. coli by electroporation. The bulk of the original transformed cDNA library was frozen in glycerol at −80° C. An aliquot was titered to measure the concentration of transformed E. coli. The E. coli were thawed, diluted to 76,000/0.1 ml in media containing ampicillin, and 0.1 ml was distributed into the wells of a microtiter dish in an 8×8 array. The microtiter dish was placed at 37° C. overnight to grow the E. coli.
  • To prepare DNA for PCR, 20 μl aliquots of culture from each well were withdrawn and pooled separately for each row and column of eight wells, giving 16 pools of 160 μl each. The E. coli were pelleted, resuspended in 160 μl of standard lysis buffer consisting of 10 mM Tris HCl pH8, 1 mM EDTA, 0.01% Triton X-100, and lysed by heating to 95° C. for 10 minutes.
  • To identify which of the wells contained E. coli transformed with B18, PCR was performed first on the DNA preps corresponding to the eight columns. The PCR consisted of two sequential reactions with nested oligonucleotides using standard conditions. The oligonucleotides used for the PCR reaction were derived from the sequence of the partial B18 clone. They were:
  • B185: CACAGGCATACACAGGAAGATACATTCA (SEQ ID NO:7)
    B183: TCTTGCTGGATGGGAACGGAATTCA (SEQ ID NO:8)
    B18N: ATACATTCACAGAAGAGCTTCCTGCACA (SEQ ID NO:9)
  • The PCR conditions were 2.5 mM MgCl2 and 95° C.×2 min for one cycle, 95° C.×1 min plus 68° C.×1 min for 30 cycles, and 68° C.×10 for one cycle. Each reaction was 20 μl. The first reaction contained oligonucleotides B185 and B183 and 1 μl of the DNA preparations. The second reaction contained oligonucleotides B183 and B18N and 1 μl of the first reaction.
  • DNA preps that potentially contained a full-length B18 cDNA clone were identified by agarose gel electrophoresis on an aliquot of the second PCR reaction. A DNA band of the correct mobility was assumed to be derived from a B18 cDNA. Next the same sequence of PCR reactions and gel analysis was done on the DNA preps corresponding to the eight rows. The intersection of a row and a column identified well A2 as potentially containing B18, narrowing it down to the 76,000 E. coli originally seeded into that well.
  • To further purify the individual E. coli containing the putative full-length B18 cDNA clone, the concentration of E. coli in well A2 was measured by titering and plating dilutions of the well. Then 7600 E. coli were seeded into the wells of a second microtiter plate in an 8×8 array. The E. coli were grown overnight; wells were pooled, and DNA was prepared as described above. To identify which of these wells contained E. coli transformed with B18, sequential PCR reactions were performed essentially as described above. Agarose gel electrophoresis identified well B2 as potentially containing a B18 cDNA.
  • The E. coli containing this cDNA was further purified by seeding wells of a microtiter plate with 253 E. coli per well and proceeding as for the purification of the E. coli in well A2. Well C3 was identified as containing a putative full-length B18 cDNA clone. The exact E. coli was identified by plating the contents of the well onto bacterial culture media and then screening the E. coli colonies following established protocols. The probe for these hybridizations was a PCR fragment generated by doing a PCR reaction on the B18 clone using as primers the oligonucleotides described above (SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9). When a single colony was identified, DNA was prepared and sequenced by standard methods. Comparison of this sequence to the sequence of the original partial clone confirmed identity and that the isolated cDNA was full-length.
  • The full-length clone was deposited with the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209, on Jul. 6, 1995 and assigned accession number ATCC 69868.
  • Example 2 Expression of Human CTLA-8
  • The full-length B18 clone for human CTLA-8 was transfected into COS cells which were then labelled with 35S-methionine. An aliquot of conditioned medium from the transfected cell culture was reduced, denatured and electrophoresed on polyacrylamide gels. Autoradiographs of those gels are reproduced in FIG. 2. The band indicated by the arrow demonstrates expression of human CTLA-8.
  • Example 3 Inhibition of Angiogenesis by Human CTLA-8
  • The ability of human CTLA-8 to inhibit angiogenesis was examined in an angiostatic activity assay (endothelial cell proliferation assay). The assay was done in a 96 well plate. Primary human umbilical cells (HUVECs) were seeded to 2×103 cells per well in EGM medium (Clonetics)/20% FCS and incubated at 37° C. for 24 hr. The cells were then starved in M199 medium (GIBCO BRL) containing 10% charcoal treated serum (M199-CS) for 48 hr at 37° C. Conditioned media containing B18 (human CTLA-8) was obtained from transfected COS or stably expressing CHO cells and 1:10, 1:50, 1:250, and 1:1250 dilutions prepared in MI 99-CS medium containing 100 ng/ml FGF. The dilutions of B18 were added to the starved cells and incubated for 72 hr at 37° C. The cells were then radiolabeled by [3H]-thymidine for 6 hr. Radiolabeled cells were washed with PBS and trypsinized for liquid scintillation counting. Results were plotted using Kaleidograph software. The results are shown in FIG. 3. In the figure, “Med” is the mock control, “B18” and “B18-1” were conditioned medium from two independent transfections of COS with DNA encoding human CTLA-8 (B18). IFNγ was used as a positive control angiostatic (i.e., angiogenesis inhibition) activity. These data demonstrate that human CTLA-8 (B18) inhibits angiogenesis.
  • Example 4 Hematopoietic Activity of Human CTLA-8
  • The hematopoietic activity of human CTLA-8 (B18) expressed in vivo was examined by construction of a recombinant adenovirus vector.
  • The B18 cDNA in the expression plasmid Adori 2-12 B18 was driven by the cytomegalovirus (CMV) immediate early promoter and enhancer.
  • The Adori 2-12 vector was created by addition of an SV40 origin and enhancer to a known adenovirus vector (Barr et al., Gene Therapy 1:51 (1994); Davidson et al., Nature Genetics 3:219 (1993)). The HindIII/BamHI fragment encoding the SV40 origin and enhancer was isolated from the pMT2 mammalian expression vector, blunted with Klenow and cloned into the NatI site (blunted with Klenow) of the Ad5 expression vector.
  • The vector was derived by digesting pNOT-B18 cDNA with SalI, filling in the 5′ overhang with Klenow to generate a blunt end and digesting with EcoRI to isolate the B18 cDNA. The blunted-EcoRI B18 fragment was inserted into the restriction sites EcoRV-EcoRI of the adenovirus vector Adori 2-12. The CMV-B18 expression cassette was located downstream of the SV40 origin and enhancer, and 0-1 map units of the left hand end of the adenovirus type 5(Ad5). The SV40 splice donor and acceptor were located between the CMV promoter and B18 cDNA. Following the insert was SV40 poly A site, 9-16 map units of Ad5 and the puc 19 origin.
  • A recombinant adenovirus was generated by homologous recombination in 293 cells. AscI linearized Adori 2-12 B18 and ClaI digested AdCMVlacZ were introduced into the 293 cells using lipofectamine. Recombinant adenovirus virus was isolated and amplified on 293 cells. The virus was released from infected 293 cells by three cycles of freeze-thawing. The virus was further purified by two cesium chloride centrifugation gradients and dialyzed against PBS at 4° C. Following dialysis of the virus glycerol was added to a concentration of 10% and the virus was stored at −70° C. until use. The virus was characterized by expression of the transgene, plaque forming units on 293 cells, particles/ml and Southern analysis of the virus.
  • A single dose of 5×1010 particles of recombinant adenovirus encoding B18 was injected into the tail vein of male C57/b16 mice, age 7-8 weeks. Control mice received an adenovirus encoding B-galactosidase. Four mice from each experimental group were killed on day 7 and 14. Blood was collected and automated hematologic analysis was performed using a Baker 9000. Differential counts were performed on blood smears. Tissue was harvested, fixed in formalin, and stained with hematoxylin and eosin for histopathology. In the first set of experiments, serum and tissues were analyzed 7 and 14 days post injection. A slight increase in peripheral platelet counts was observed. The animals that received B18 exhibited a slight increase in spleen size. Macroscopic analysis of the spleen showed an increase in splenic extramedullary hematopoiesis on day 7 compared to the control. These results showed a hematopoietic growth activity associated with B18.
  • In a second set of experiments 5×1010 particles of recombinant adenovirus encoding B18 were injected into the tail vein of male C57/b16 mice, age 17-18 weeks. Control mice received an adenovirus encoding B-galactosidase. Blood samples were collected via retro-orbital sinus on days 2, 5, 7, 10, 14, and 21. The hematologic analyses were performed on the Baker 9000 automated cell counter with murine-specific settings. Analyses included WBC, RBC, HCT and PLT counts. Blood smears were prepared and stained with Wright-Geimsa for WBC differentials based on a 100 cell count. Reticulocytes and reticulated platelets were quantitated using flow cytometry. Four mice from each group were killed on days 7, 14, and 21. In addition to peripheral blood analysis, serum was collected via cardiac puncture for quantitation of systemic II-6 using a commercial kit (Endogen). Spleen and liver were collected for histopathology, spleen and bone marrow hematopoietic progenitors were quantitated, and bone marrow smears were prepared and stained with Wright-Geimsa for cell counts.
  • Administration of adenovirus encoding B18 resulted in a marked increase in peripheral blood neutrophils and WBC (FIG. 4). Maximum increases in neutrophils were observed at day 5 and day 7. The control mice showed little difference at day 5 and day 7. Peripheral blood neutrophils were similar in the control mice and mice that received B18 at day 21. In both the B18 and control groups an increase in white blood cells was also observed. The mice that received B18 had a greater increase in WBC between day 2 and day 7. By Day 21 a more pronounced increase was observed in the B-gal group. No other changes in cellular chemistries were observed (Table I).
  • Bone marrow cellularity was calculated from pooled femurs in each group (Table III). No significant differences were observed in either group. No significant changes were observed in bone marrow hematopoietic progenitors from day 7, 14, and 21. The CFU-GM, BFU-E and CFU-MEG in the B18 mice were similar to the B-gal control (Table II).
  • Administration of the adenovirus encoding B18 resulted in an increase in CFU-GM (myeloid) and BFU-E (erythroid) progenitors in the spleen compared to animals that received the B-gal virus on day 7. The increase in progenitors in the B18 mice was 11-fold in CFU-GM and a 52-fold in BFU-E (Table II). There was a 2-fold increase in CFU-MEG at day 7 for the B18 mice. By day 21 no significant differences were observed in splenic CFU-MEG or BFU-E between the groups (Table II). A 3-fold decrease in CFU-GM was observed in mice that received adenovirus encoding B18. A slight increase in spleen size at day 7 was observed in the B18 group. This is consistent with an increase in splenic cellularity. By day 14 and day 21 spleen weights were similar to the control group (Table III). Macroscopic analysis of the spleen showed an increase in splenic extramedullary hematopoiesis of the B18 mice on day 7 compared to the control.
  • The bone marrow myeloid: erythroid ratios (Table IV) suggest a granulocytic hyperplasia with a possible erythroid hypoplasia in mice that received adenovirus B18 on day 7. By day 21 the ratio in the B-gal group was higher. No changes were observed in the IL6 serum levels.
  • These results show a hematopoietic activity associated with the administration of adenovirus encoding B18 (human CTLA-8). Increases in neutrophils and white blood cells were observed at day 7 in animals that received B18 adenovirus. The data showed that B18 resulted in an increase in splenic CFU-GM and BFU-E 7 days post administration compared to the control animals. Splenic extramedullary hematopoiesis on day 7 demonstrates that B18 exhibits a hematopoietic growth activity. These data suggest that B18 may mobilize early hematopoietic precursors.
  • TABLE I
    Peripheral hematology for day 2, 5, 7, 10, 14 and 21
    WBC
    ×10{circumflex over ( )}3/ Neuts ANC Lymphs ALC Eos Monos RBC Retics Abs Retics HCT PLT RPlt Abs RPlt
    uL % ×10{circumflex over ( )}3/uL % ×10{circumflex over ( )}3/uL % % ×10{circumflex over ( )}6/uL % ×10{circumflex over ( )}6/uL % ×10{circumflex over ( )}3/uL % ×10{circumflex over ( )}3/uL
    Study A54-4B . . . B18 (Platelets) Day 2 . . . 4-25-96.
    Group A
    B-Gal #1 5.4 40 2.16 54 2.92 0 6 10.88 3.65 0.40 48.0 836 11.94 99.82
    B-Gal #2 7.4 25 1.85 65 4.81 3 7 12.34 2.04 0.25 56.6 900 10.10 90.90
    B-Gal #3 6.8 40 2.72 52 3.54 2 6 11.26 3.26 0.37 51.6 894 9.77 87.34
    B-Gal #4 8.8 23 2.02 64 5.63 1 12 12.00 2.55 0.31 54.8 840 10.63 89.29
    AVG 7.1 32.0 2.19 58.8 4.22 1.5 7.8 11.62 2.88 0.33 52.8 868 10.61 91.84
    SEM 0.7 4.6 0.19 3.4 0.61 0.6 1.4 0.33 0.36 0.03 1.9 17 0.48 2.76
    B18 #1 11.4 59 6.73 31 3.53 1 9 11.16 4.88 0.54 52.4 1242 14.92 185.31
    B18 #2 9.2 30 2.76 62 5.70 3 5 10.14 3.97 0.40 48.0 632 10.90 68.89
    B18 #3 5.0 51 2.55 40 2.00 0 9 11.16 3.23 0.36 51.2 832 11.18 93.02
    B18 #4 6.4 41 2.62 55 3.52 0 4 10.80 3.09 0.33 49.2 904 17.31 156.48
    AVG 8.0 45.3 3.67 47.0 3.69 1.0 6.8 10.82 3.79 0.41 50.2 903 13.58 125.92
    SEM 1.4 6.3 1.02 7.0 0.76 0.7 1.3 0.24 0.41 0.05 1.0 127 1.55 27.07
    Study A54-4B . . . B18 (Platelets) Day 5 . . . 4-28-96.
    Group B
    B-Gal #1 7.6 14 1.06 78 5.93 3 5 11.26 5.25 0.59 52.4 1082 15.51 167.82
    B-Gal #2 10.6 20 2.12 78 8.27 1 1 10.72 4.71 0.50 49.4 994 17.37 172.66
    B-Gal #3 8.8 18 1.51 69 6.07 2 11 11.12 3.40 0.38 51.2 916 9.55 87.48
    B-Gal #4 10.8 38 4.10 58 6.26 0 4 10.22 6.21 0.63 47.0 1092 13.93 152.12
    AVG 9.5 22.5 2.20 70.8 6.63 1.5 5.3 10.83 4.89 0.53 50.0 1021 14.09 145.02
    SEM 0.8 5.3 0.67 4.8 0.55 0.6 2.1 0.23 0.59 0.06 1.2 41 1.67 19.67
    B18 #1 14.8 18 2.66 71 10.51 1 10 12.66 2.31 0.29 57.0 1204 7.57 91.14
    B18 #2 14.2 37 5.25 53 7.53 2 8 9.80 3.32 0.33 44.6 888 14.33 127.25
    B18 #3 12.8 30 3.84 59 7.55 1 10 12.12 4.12 0.50 55.6 1134 10.15 115.10
    B18 #4 16.0 58 9.28 37 5.92 0 5 11.04 3.93 0.43 50.8 1166 15.75 183.65
    AVG 14.5 35.8 5.26 55.0 7.88 1.0 8.3 11.41 3.42 0.39 52.0 1098 11.95 129.28
    SEM 0.7 8.4 1.44 7.1 0.96 0.4 1.2 0.63 0.41 0.05 2.8 71 1.88 19.61
    Study A54-4B . . . B18 (Platelets) Day 7 . . . 4-30-96.
    Group C
    B-Gal #1 15.2 14 2.13 69 10.49 1 16 11.04 3.54 0.39 50.8 862 12.46 107.41
    B-Gal #2 14.0 12 1.68 81 11.34 0 7 11.38 5.05 0.57 52.6 1104 14.91 164.61
    B-Gal #3 14.8 14 2.07 73 10.80 1 12 10.92 5.42 0.59 49.6 952 11.49 109.38
    AVG 14.7 13.3 1.96 74.3 10.88 0.7 11.7 11.11 4.67 0.52 51.0 973 12.95 127.13
    SEM 0.4 0.7 0.14 3.5 0.25 0.3 2.6 0.14 0.58 0.06 0.9 71 1.02 18.75
    B18 #1 19.4 33 6.40 62 12.03 0 5 10.14 2.93 0.30 45.2 864 12.80 110.59
    B18 #2 25.4 39 9.91 53 13.48 0 8 9.46 6.05 0.57 43.6 1288 12.49 160.87
    B18 #3 23.6 44 10.38 50 11.80 0 6 9.74 5.17 0.50 44.4 1076 15.41 165.81
    B18 #4 12.8 15 1.92 75 9.60 0 10 9.54 6.26 0.60 43.4 1136 15.88 180.40
    AVG 20.3 32.8 7.15 60.0 11.72 0.0 7.3 9.72 5.10 0.49 44.2 1091 14.15 154.42
    SEM 2.8 6.3 1.96 5.6 0.80 0.0 1.1 0.15 0.76 0.07 0.4 88 0.87 15.19
    Study A54-4B . . . B18 (Platelets) Day 10 . . . 5-3-96.
    Group A
    B-Gal #1 18.6 17 3.16 69 12.83 3 11 10.22 12.41 1.27 46.8 1460 16.20 236.52
    B-Gal #2 13.2 16 2.11 79 10.43 1 4 10.48 6.00 0.63 48.8 1128 14.48 163.33
    B-Gal #3 19.6 16 3.14 74 14.50 0 10 10.72 6.25 0.67 49.4 1338 16.58 221.84
    B-Gal #4 18.6 21 3.91 72 13.39 3 4 10.44 7.59 0.79 48.4 1068 14.35 153.26
    AVG 17.5 17.5 3.08 73.5 12.79 1.8 7.3 10.47 8.06 0.84 48.4 1249 15.40 193.74
    SEM 1.5 1.2 0.37 2.1 0.86 0.8 1.9 0.10 1.49 0.15 0.6 91 0.58 20.78
    B18 #1 14.2 33 4.69 56 7.95 5 6 8.70 11.97 1.04 39.2 1760 14.49 255.02
    B18 #2 17.6 35 6.16 57 10.03 1 7 9.04 9.48 0.86 42.0 1104 18.88 208.44
    B18 #3 16.2 39 6.32 57 9.23 1 3 4.74 16.77 0.79 22.4 894 29.19 260.96
    B18 #4 14.2 25 3.55 66 9.37 1 8 9.30 9.93 0.92 42.0 1416 16.81 238.03
    AVG 15.6 33.0 5.18 59.0 9.15 2.0 6.0 7.95 12.04 0.90 36.4 1294 19.84 240.61
    SEM 0.8 2.9 0.66 2.3 0.43 1.0 1.1 1.08 1.67 0.05 4.7 189 3.24 11.77
    Study A54-4B . . . B18 (Platelets) Day 14 . . . 5-7-96.
    B-Gal #1 17.8 18 3.20 74 13.17 0 8 10.86 5.97 0.65 50.8 1360 11.03 150.01
    B-Gal #2 20.4 26 5.30 66 13.46 1 7 10.92 7.07 0.77 50.8 1616 8.18 132.19
    B-Gal #3 16.0 7 1.12 90 14.40 1 3 11.36 6.41 0.73 52.8 1298 7.36 95.53
    B-Gal #4 18.0 36 6.48 57 10.26 1 6 9.30 7.62 0.71 43.0 1672 10.05 168.04
    AVG 18.1 21.8 4.03 71.8 12.82 0.8 6.0 10.61 6.77 0.71 49.4 1487 9.16 136.44
    SEM 0.9 6.1 1.18 7.0 0.89 0.3 1.1 0.45 0.36 0.03 2.2 93 0.84 15.48
    B18 #1 15.4 9 1.39 81 12.47 1 9 10.62 5.74 0.61 48.2 1262 9.51 120.02
    B18 #2 15.4 31 4.77 58 8.93 2 9 9.76 10.33 1.01 44.6 1092 14.29 156.05
    B18 #3 13.4 42 5.63 39 5.23 0 19 10.34 4.99 0.52 46.6 1376 15.79 217.27
    B18 #4 11.6 57 6.61 34 3.94 2 7 9.38 5.57 0.52 43.0 1092 16.66 181.93
    AVG 14.0 34.8 4.60 53.0 7.64 1.3 11.0 10.03 6.66 0.66 45.6 1206 14.06 168.82
    SEM 0.9 10.0 1.14 10.7 1.93 0.5 2.7 0.28 1.23 0.12 1.1 70 1.59 20.54
    Study A54-4B . . . B18 (Platelets) Day 21 . . . 5-14-96.
    B-Gal #1 25.4 23 5.84 67 17.02 0 10 9.22 8.15 0.75 42.8 1776 9.61 170.67
    B-Gal #2 19.6 19 3.72 69 13.52 0 12 9.50 9.95 0.95 44.4 1662 9.44 156.89
    B-Gal #3 27.6 11 3.04 82 22.63 3 4 9.74 8.84 0.86 45.8 1684 11.45 192.82
    B-Gal #4 28.0 13 3.64 83 23.24 0 4 9.04 7.54 0.68 41.6 1346 10.48 141.06
    AVG 25.2 16.5 4.06 75.3 19.10 0.8 7.5 9.38 8.62 0.81 43.7 1617 10.25 165.36
    SEM 1.9 2.8 0.61 4.2 2.33 0.8 2.1 0.15 0.52 0.06 0.9 94 0.46 10.97
    B18 #1 18.6 9 1.67 83 15.44 1 7 9.84 7.40 0.73 43.8 1642 8.37 137.44
    B18 #2 16.2 48 7.78 45 7.29 1 6 11.10 7.97 0.88 51.4 1970 10.78 212.37
    B18 #3 14.8 53 7.84 42 6.22 0 5 7.52 20.26 1.52 38.0 1568 10.50 164.64
    B18 #4 15.2 18 2.74 74 11.25 2 6 9.64 7.02 0.68 43.0 1422 7.60 108.07
    AVG 16.2 32.0 5.01 61.0 10.05 1.0 6.0 9.53 10.66 0.95 44.1 1651 9.31 155.63
    SEM 0.9 10.9 1.63 10.3 2.10 0.4 0.4 0.74 3.21 0.20 2.8 116 0.78 22.16
  • TABLE II
    Bone marrow and Splenic Hematopoietic Progenitors
    CFU-MEG CFU-GM BFU-E
    B-Gal B18 B-Gal B18 B-Gal B18
    Bone Marrow*
    Day 7 16.0 ± 3.5  15.7 ± 3.1  307 ± 117 241 ± 78 51 ± 19 25 ± 11
    Day 14 10.7 ± 2.3  15.3 ± 1.2 233 ± 15 373 ± 35 30 ± 10 60 ± 30
    Day 21 5.7 ± 0.6  6.7 ± 3.1 170 ± 17 160 ± 27 40 ± 10 27 ± 6 
    Spleen**
    Day 7 9.3 ± 1.6 19.5 ± 1.5 27 ± 3 298 ± 6  1.3 ± 1.2 68 ± 10
    Day 14 9.7 ± 0.6 12.7 ± 0.6 267 ± 32 197 ± 21 33 ± 6  10 ± 10
    Day 21 17.0 ± 1.0  19.3 ± 2.5 187 ± 6   73 ± 15 23 ± 6  23 ± 6 
    *Bone marrow progenitors are represented as mean ± sd number of colonies per 105 cells.
    **Spleen progenitors are represented as mean ± sd number of colonies per 106 cells.
  • Hematopoietic precursors were determined from pooled spleen and bone marrow samples from four animals in each group. For quantitation of CFU-GM and BFU-E, either 1×104 bone marrow cells or 1×105 spleen cells were added to complete alpha methylcelluose medium (0.9% methylcellulose in alpha medium, 30% fetal bovine serum, 1% bovine serum albumin, 10−4M 2-mercaptoethanol, 2 mM L-glutamine, 2% murine spleen cell conditioned medium, and 3 U/mL erythropoietin) and aliquoted into 35 mm tissue culture dishes in a final volume of 1.0 mL. Cultures were incubated for 7 days at 37° C., and 5% CO2. Microscopic colonies were defined as clusters of 50 or more cells. For quantitation of CFU-MEG, either 1×105 bone marrow cells or 1×106 spleen cells were added to complete alpha methylcellulose medium and incubated as described above. Megakaryocyte colonies were defined as a group of 3 or more cells.
  • TABLE III
    Spleen Weights and Femur Cellularity
    Femur
    Spleen Wt. Cellularity
    (Mg) B-Gal B18 (×106) B-Gal B18
    Day
    7 187 ± 19 224 ± 29 Day 7 28 23
    Day 14 175 ± 13 170 ± 10 Day 14 28 27
    Day 21 174 ± 21 151 ± 27 Day 21 28 26
    Spleen weights were determined at time of sacrifice are represented as means ± sd from four animals.
  • TABLE IV
    Bone Marrow Myeloid:Erythroid Ratios
    Group Mouse # Day 7 Day 14 Day 21
    B-gal 1 1.43 2.12 5.78
    2 0.91 2.46 5.83
    3 1.62 1.03 3.66
    4 5.44 4.82
    AVG 1.32 2.76 5.02
    SD 0.37 0.37 1.89
    B18 1 5.59 2.01 2.02
    2 6.51 1.25 2.13
    3 5.49 1.58 1.81
    4 0.50 2.51 2.92
    AVG 4.52 1.86 2.22
    SD 1.29 2.72 0.56
    All entries represent the number of myeloid cells per 1 erythroid cell. Normal mouse ratios are approximately 1:1 to 2:1.
  • Example 5 Additional Experiments Relating to Hematopoietic Activity of Human CTLA-8
  • B18 (human CTLA-8) was tested for the ability to induce production of factors having hematopoietic activity in a factor-dependent cell proliferation assay using the human erythroleukemic cell line, TF-1 (Kitamura et al., J. Cell Physiol. 140:323 (1989)). The cells were initially grown in the presence of rhGMCSF (100 U/ml). The cells were fed three days prior to setting up the assay. The assay conditions were as follows:
  • cells/well 5000/200 μl
    incubation time
    3 days
    pulse time 4 hours
    amount of tritiated thymidine 0.5 μCi/well
    counting time 1 minute
    replicates 2
  • B18 alone, conditioned medium (CM) from B18 induced HS-5 cells were assayed. Buffer alone, CM from HS-5 cells induced with buffer and CM from uninduced HS-5 cells were assayed as controls. Results are shown in FIG. 5. B18 (human CTLA-8) demonstrated an ability to induce production of factors which induced TF-1 proliferation. This activity was substantially eliminated by the addition of anti-GMCSF antibodies. These data demonstrate that human CTLA-8 (B18) is able to induce hematopoiesis. Particularly, without being bound by any theory, it appears that human CTLA-8 (B18) induces production of GM-CSF and/or IL-3.
  • Example 6 Ability of Human CTLA-8 to Induce Production of IL-6 and IL-8
  • MRC5 cells were incubated in the presence of human CTLA-8 (B18) and production of IL-6 and IL-8 were measured. Herpes CTLA-8 (IL-17) was used as a positive control. Applicants' human CTLA-8 (B18) demonstrated titratable production of both IL-6 and IL-8 (see FIGS. 6 and 7).
  • All patent and literature references cited herein are incorporated by reference as if fully set forth.

Claims (7)

1-27. (canceled)
28. A method of stimulating hematopoiesis in a subject, the method comprising administering to the subject a therapeutically effective amount of a composition comprising an amino acid sequence selected from the group consisting of:
(a) the amino acid sequence of SEQ ID NO:2;
(b) the amino acid sequence of SEQ ID NO:2 from amino acids 11 to 163;
(c) the amino acid sequence of SEQ ID NO:2 from amino acids 29 to 163;
(d) the amino acid sequence of SEQ ID NO:2 from amino acids 31 to 163; and
(e) fragments of (a), (b), (c) or (d) having CTLA-8 activity.
29. The method of claim 28, wherein said subject is treated to produce an effect selected from the group consisting of proliferation of myeloid cells or progenitors, proliferation of erythroid cells or progenitors, proliferation of lymphoid cells or progenitors, induction of IL-3 production and induction of GM-CSF production.
30. The method of claim 33, wherein the myeloid cells are neutrophils.
31. A method of inhibiting angiogenesis in a subject, the method comprising administering to the subject a therapeutically effective amount of a composition comprising an amino acid sequence selected from the group consisting of:
(a) the amino acid sequence of SEQ ID NO:2;
(b) the amino acid sequence of SEQ ID NO:2 from amino acids 11 to 163;
(c) the amino acid sequence of SEQ ID NO:2 from amino acids 29 to 163;
(d) the amino acid sequence of SEQ ID NO:2 from amino acids 31 to 163; and
(e) fragments of (a), (b), (c) or (d) having CTLA-8 activity.
32. The method of claim 31, wherein the subject is treated to prevent growth or proliferation of vascular endothelial cells, tumor growth, or angiogenesis-dependent tissue growth.
33. The method of claim 29, wherein the effect is proliferation of myeloid cells.
US12/175,336 1995-07-19 2008-07-17 Human il-17f and other il-17f-related proteins and methods related thereto Abandoned US20090075884A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/175,336 US20090075884A1 (en) 1995-07-19 2008-07-17 Human il-17f and other il-17f-related proteins and methods related thereto

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US50403295A 1995-07-19 1995-07-19
US3534795P 1995-07-19 1995-07-19
US08/514,014 US5707829A (en) 1995-08-11 1995-08-11 DNA sequences and secreted proteins encoded thereby
US08/685,239 US6074849A (en) 1995-07-19 1996-07-18 Polynucleotides encoding human CTLA-8 related proteins
US09/034,810 US6043344A (en) 1995-07-19 1998-03-04 Human CTLA-8 and uses of CTLA-8-related proteins
US50715000A 2000-02-18 2000-02-18
US10/102,080 US6902735B1 (en) 1995-07-19 2002-03-19 Antibodies to human IL-17F and other CTLA-8-related proteins
US11/034,843 US20050214315A1 (en) 1995-07-19 2005-01-14 Antibodies to human IL-17F and other CTLA-8-related proteins and methods related thereto
US12/175,336 US20090075884A1 (en) 1995-07-19 2008-07-17 Human il-17f and other il-17f-related proteins and methods related thereto

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/034,843 Division US20050214315A1 (en) 1995-07-19 2005-01-14 Antibodies to human IL-17F and other CTLA-8-related proteins and methods related thereto

Publications (1)

Publication Number Publication Date
US20090075884A1 true US20090075884A1 (en) 2009-03-19

Family

ID=34624107

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/102,080 Expired - Fee Related US6902735B1 (en) 1995-07-19 2002-03-19 Antibodies to human IL-17F and other CTLA-8-related proteins
US11/034,843 Abandoned US20050214315A1 (en) 1995-07-19 2005-01-14 Antibodies to human IL-17F and other CTLA-8-related proteins and methods related thereto
US12/175,336 Abandoned US20090075884A1 (en) 1995-07-19 2008-07-17 Human il-17f and other il-17f-related proteins and methods related thereto

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/102,080 Expired - Fee Related US6902735B1 (en) 1995-07-19 2002-03-19 Antibodies to human IL-17F and other CTLA-8-related proteins
US11/034,843 Abandoned US20050214315A1 (en) 1995-07-19 2005-01-14 Antibodies to human IL-17F and other CTLA-8-related proteins and methods related thereto

Country Status (1)

Country Link
US (3) US6902735B1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110033451A1 (en) * 2005-02-14 2011-02-10 Wyeth Llc Interleukin-17f antibodies and other il-17f signaling antagonists and uses therefor
US11633503B2 (en) 2009-01-08 2023-04-25 Northwestern University Delivery of oligonucleotide-functionalized nanoparticles
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties
US11866700B2 (en) 2016-05-06 2024-01-09 Exicure Operating Company Liposomal spherical nucleic acid (SNA) constructs presenting antisense oligonucleotides (ASO) for specific knockdown of interleukin 17 receptor mRNA

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI1641822T1 (en) 2003-07-08 2013-08-30 Genentech, Inc. Il-17 a/f heterologous polypeptides and therapeutic uses thereof
WO2006088925A2 (en) * 2005-02-14 2006-08-24 Wyeth Use of il17-f in diagnosis and therapy of airway inflammation
AU2007226627B2 (en) 2006-03-10 2012-09-20 Zymogenetics, Inc. Antibodies that bind both IL-17A and IL-17F and methods of using the same
EP2029171A1 (en) * 2006-06-19 2009-03-04 Wyeth a Corporation of the State of Delaware Methods of modulating il-22 and il-17
TW200902064A (en) * 2007-03-28 2009-01-16 Wyeth Corp Methods and compositions for modulating IL-17F/IL-17A biological activity
KR20110025649A (en) 2008-05-05 2011-03-10 노비뮨 에스 에이 Anti-il-17a/il-17f cross-reactive antibodies and methods of use thereof
PE20160652A1 (en) 2009-05-05 2016-07-09 Novimmune Sa ANTIBODIES THAT JOIN IL-17F
UA117218C2 (en) 2011-05-05 2018-07-10 Мерк Патент Гмбх Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
WO2013177101A2 (en) 2012-05-22 2013-11-28 Bristol-Myers Squibb Company Bispecific antibodies and methods of using the same
US9622523B2 (en) 2014-01-06 2017-04-18 Exsomed International IP, LLC Ergonomic work gloves
KR20160114723A (en) * 2014-02-11 2016-10-05 빈 왕 Immunotherapeutic composition, therapeutic method and diagnostic method
EP3322716A2 (en) * 2015-07-15 2018-05-23 California Institute of Technology Il-17f-specific capture agents, compositions, and methods of using and making

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5536637A (en) * 1993-04-07 1996-07-16 Genetics Institute, Inc. Method of screening for cDNA encoding novel secreted mammalian proteins in yeast
US5707829A (en) * 1995-08-11 1998-01-13 Genetics Institute, Inc. DNA sequences and secreted proteins encoded thereby
US6043344A (en) * 1995-07-19 2000-03-28 Genetics Institute, Inc. Human CTLA-8 and uses of CTLA-8-related proteins
US6274711B1 (en) * 1993-06-14 2001-08-14 Inserm, Institut National De La Sante Et De La Recherche Medicale Purified mammalian CTLA-8 antigens and related reagents

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6562333B1 (en) 1993-06-14 2003-05-13 Schering Corporation Purified mammalian CTLA-8 antigens and related reagents
WO1996029408A1 (en) 1995-03-23 1996-09-26 Immunex Corporation Il-17 receptor

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5536637A (en) * 1993-04-07 1996-07-16 Genetics Institute, Inc. Method of screening for cDNA encoding novel secreted mammalian proteins in yeast
US6274711B1 (en) * 1993-06-14 2001-08-14 Inserm, Institut National De La Sante Et De La Recherche Medicale Purified mammalian CTLA-8 antigens and related reagents
US6043344A (en) * 1995-07-19 2000-03-28 Genetics Institute, Inc. Human CTLA-8 and uses of CTLA-8-related proteins
US6074849A (en) * 1995-07-19 2000-06-13 Genetics Institute, Inc. Polynucleotides encoding human CTLA-8 related proteins
US5707829A (en) * 1995-08-11 1998-01-13 Genetics Institute, Inc. DNA sequences and secreted proteins encoded thereby

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110033451A1 (en) * 2005-02-14 2011-02-10 Wyeth Llc Interleukin-17f antibodies and other il-17f signaling antagonists and uses therefor
US11633503B2 (en) 2009-01-08 2023-04-25 Northwestern University Delivery of oligonucleotide-functionalized nanoparticles
US11866700B2 (en) 2016-05-06 2024-01-09 Exicure Operating Company Liposomal spherical nucleic acid (SNA) constructs presenting antisense oligonucleotides (ASO) for specific knockdown of interleukin 17 receptor mRNA
US11696954B2 (en) 2017-04-28 2023-07-11 Exicure Operating Company Synthesis of spherical nucleic acids using lipophilic moieties

Also Published As

Publication number Publication date
US6902735B1 (en) 2005-06-07
US20050214315A1 (en) 2005-09-29

Similar Documents

Publication Publication Date Title
EP0839196B1 (en) Human ctla-8 and uses of ctla-8-related proteins
US6043344A (en) Human CTLA-8 and uses of CTLA-8-related proteins
US20090075884A1 (en) Human il-17f and other il-17f-related proteins and methods related thereto
WO1997004097A9 (en) Human ctla-8 and uses of ctla-8-related proteins
EP0994947B1 (en) Mu-1, member of the cytokine receptor family
EP1005552A1 (en) Member of the hematopoietin receptor superfamily
AU2004231176B2 (en) Human CTLA-8 and uses of CTLA-8-related proteins
AU776563B2 (en) Human CTLA-8 and uses of CTLA-8-related proteins
AU2008203282A1 (en) Human CTLA-8 and uses of CTLA-8-related proteins
MXPA98000507A (en) Ctla-8 human and uses of proteins related to the ctl
AU2003208117C1 (en) MU-1, member of the cytokine receptor family
WO2000029439A9 (en) The cxc chemokine h174 and methods for preventing damage to the nervous system
WO1999053066A1 (en) U4, a member of the hematopoietin receptor superfamily
WO2000066148A1 (en) Novel proteins

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION