WO2012131680A2 - Compositions and methods for treating inflammation - Google Patents

Compositions and methods for treating inflammation Download PDF

Info

Publication number
WO2012131680A2
WO2012131680A2 PCT/IL2012/050111 IL2012050111W WO2012131680A2 WO 2012131680 A2 WO2012131680 A2 WO 2012131680A2 IL 2012050111 W IL2012050111 W IL 2012050111W WO 2012131680 A2 WO2012131680 A2 WO 2012131680A2
Authority
WO
WIPO (PCT)
Prior art keywords
peptide
diseases
molecule
cell
autoimmune
Prior art date
Application number
PCT/IL2012/050111
Other languages
French (fr)
Other versions
WO2012131680A9 (en
WO2012131680A3 (en
Inventor
Einat TOISTER
Idit Shachar
Original Assignee
Yeda Research And Development Co. Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Yeda Research And Development Co. Ltd. filed Critical Yeda Research And Development Co. Ltd.
Publication of WO2012131680A2 publication Critical patent/WO2012131680A2/en
Publication of WO2012131680A9 publication Critical patent/WO2012131680A9/en
Publication of WO2012131680A3 publication Critical patent/WO2012131680A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention in some embodiments thereof, relates to compositions and methods for treating inflammation.
  • B and T lymphocytes develop in the primary lymphoid organs while their capabilities are employed throughout the body. Accordingly, B and T lymphocytes continually patrol the body for foreign antigens by circulating from the blood, through tissues, into lymph nodes (LNs), and back to the blood. Lymphocyte migration out of the vasculature is therefore a key regulatory point for controlling immune responses. It is controlled by multi-step processes that include chemoattraction, cell-cell adhesion and transmigration through cell layers. The continuous recirculation of lymphocytes and their movement from the blood into peripheral tissues is a central feature of immune surveillance, but also contributes to the pathogenesis of inflammatory diseases, autoimmune diseases, as well as the development and progression of allergic responses.
  • VLA4 vascular cell adhesion molecule- 1
  • VCAM-1 vascular cell adhesion molecule- 1
  • the VLA-4-VCAM-1 interaction is responsible, at least in part, for recruitment of T cells to areas of vascular inflammation and plays an essential role during various types of inflammation [Lobb and Hemler, J Clin Invest (1994) 94(5): 1722-8; Postigo et al, Res Immunol (1993) 144(9): 723-35; discussion 754-62].
  • VLA-4 also binds to fibronectin on the extracellular matrix (ECM) and this interaction is important for migration of T lymphocytes to antigenic sites [Mannion et al, J Immunol (1996) 157(5): 2039-47; Yuan et al, Biochem J (1996) 318 (Pt 2): 591-6], thus VLA-4 expression is critical for the migration of T lymphocytes into inflamed tissue.
  • ECM extracellular matrix
  • CD151 a member of the tetraspanins family (also known as the transmembrane 4 superfamily), is expressed in various cell types including epidermal basal cells, epithelial cells, skeletal, smooth and cardiac muscle, endothelial cells, T cells, neutrophils, platelets, and Schwann cells and has a strong molecular association with the ⁇ family of integrins.
  • CD151 possesses four highly conserved transmembrane domains, cytoplasmic amino and carboxyl termini, and two extracellular loops, the larger of which contains the distinctive pattern of cysteine residues that help to define the family.
  • CD151 modulates integrin activity and function, such as those of ⁇ integrins [Nishiuchi et al., Proc Natl Acad Sci U S A (2005) 102(6): 1939-44].
  • hasegawa et al. have reported that the expression level of CD151 on adult T cell leukemia (ATL) cells from the lymph nodes of lymphoma-type ATL patients were significantly higher than those on circulating ATL cells [Hasegawa et al., J of immunology (1998) 161 : 3087-3095]. Thus, they suggest that increased expression of CD 151 may act to retain ATL cells in the lymph nodes.
  • ATL adult T cell leukemia
  • WO 2007/146968 discloses multivalent binding peptides, including bi-specific binding peptides, having an immunoglobulin effector function.
  • the cell target of the peptide may be an angiogenesis target (e.g. CD 151) and the cell may be a T cell.
  • Methods for using such peptides to treat, prevent or ameliorate symptoms of a variety of diseases, disorders or conditions are also provided.
  • WO 2007/146968 contemplates targeted recruitment of effector cells (e.g. cytotoxic T lymphocytes) or to localizing therapeutic compounds (e.g. radiolabeled proteins) to cells, tissues, agents and foreign objects to be destroyed or sequestered (e.g. cancer cells).
  • effector cells e.g. cytotoxic T lymphocytes
  • therapeutic compounds e.g. radiolabeled proteins
  • Additional background art includes U.S. 20090324600 and U.S. 201000150902 teaching anti CD151 antibodies and uses of same for the treatment of cancer.
  • WO2010/137020 teaches the use of CD151 inhibitors for the treatment of cancer and inflammation.
  • an isolated peptide comprising at least 20 consecutive amino acids of the amino acid sequence set forth in SEQ ID NO: 2, the peptide being shorter than 100 amino acids and having a CD151 inhibitory activity.
  • the CD 151 inhibitory activity comprises inhibition of lymphocyte migration or homing.
  • the migration or homing is to a lymphoid organ.
  • the lymphocyte comprises a T cell.
  • the CD 151 inhibitory activity comprises inhibition of Vav phosphorylation.
  • the isolated peptide is a soluble peptide.
  • a molecule comprising the isolated peptide attached to a heterologous moiety.
  • the heterologous moiety is a proteinaceous moiety.
  • the heterologous moiety comprises an immunoglobulin domain.
  • the heterologous moiety is a non-proteinaceous moiety.
  • the non-proteinaceous moiety comprises a synthetic polymer.
  • the peptide is as set forth in SEQ ID NO: 2.
  • an isolated polynucleotide encoding the peptide there is provided an isolated polynucleotide encoding the peptide.
  • nucleic acid expression construct comprising the isolated polynucleotide under the transcriptional control of a cis-acting regulatory element.
  • a pharmaceutical composition comprising as an active ingredient, the isolated peptide or molecule, and a pharmaceutically acceptable carrier.
  • a method of treating inflammation in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the isolated peptide or molecule, thereby treating the inflammation in the subject.
  • the isolated peptide or molecule is used in the treatment of inflammation.
  • the inflammation is associated with a medical condition selected from the group consisting of a cancer, an autoimmune disease, a hypersensitivity, a diabetes, an infectious disease, a transplantation associated disease and an allergy.
  • the subject is a human being.
  • the inflammation is associated with an inflammatory bowel disease.
  • said inflammatory bowel disease comprises colitis.
  • FIGs. 1A-B are bar graphs showing that CD151ECD peptide inhibits the actin polymerization and migration of T cells.
  • Figure 1A - T cells were incubated with CD151ECD (SEQ ID NO: 2) or a control peptide (SEQ ID NO: 2, 20 or 40 ⁇ g/ml) for lh. The cells were then stimulated with CCL21 (0.1 mg/ml). 15 sec later, the cells were immediately fixed, permeabilized, stained with FITC-phalloidin. Actin polymerization was analyzed by flow cytometry. The results shown represent the average of three independent experiments.
  • Figure IB - T cells were suspended with CD151ECD (SEQ ID NO: 2) or a control peptide ((SEQ ID NO: 2) 40 ⁇ g/ml) for 1 hr. The cells were then placed in the upper chamber of a transwell. The migration towards the chemokine CCL21 (0.4 mg/ml), residing in the lower part of the apparatus, was analyzed after 3 hours by FACS. The graph shows the average of three independent experiments.
  • FIG. 2 is a micrograph showing that CD151ECD peptide downregulates Vavl phosphorylation.
  • T cells were incubated with CD151ECD (SEQ ID NO: 2) or a control peptide ((SEQ ID NO: 2) 40 ⁇ g/ml) for 1 min. Immediately after treatment, cells were washed and fast frozen in liquid nitrogen. Cells were then lysed and an aliquot reserved for total Vavl analysis. Phosphorylated proteins (pVav) from the remaining lysate were immunoprecipitated (IP) with an anti-Tyr(P) antibody. Immunoprecipitates and total lysate proteins were separated on 10% (w/v) SDS-PAGE and blotted with an anti-Vavl antibody. The results shown represent three separate experiments.
  • the present invention in some embodiments thereof, relates to compositions and methods for treating inflammation.
  • the progress in understanding the mechanisms of T cell activation, T-cell trafficking, migration and homing, as well as progress towards development of mechanisms for the inactivation and modulation of T-cells has been translated into strategies for treatment of autoimmune diseases.
  • the targets for selective immuno- intervention include modulation of antigen recognition, costimulation blockade, induction of regulatory cells, deviation to non-pathogenic or protective responses, neutralization of proinflammatory cytokines, induction or administration of anti-inflammatory cytokines, and modulation of leukocyte trafficking.
  • the surveillance of the body for foreign antigens is a critical function of the immune system. Lymphocytes migrate from the blood into tissues and secondary lymphoid organs, and return to the blood via lymph vessels and the thoractic duct. The majority of lymphocytes are capable of tissue selective trafficking (homing), recognizing organ-specific adhesion molecules on specialized endothelial cells. Previous studies focused on the specific recruitment of leukocytes to the lymph nodes (LN) or to sites of inflammation.
  • CD151 has been previously suggested as a target for exerting global suppressory effects on T cell trafficking and differentiation within the peripheral lymph.
  • WO2010/137020 teaches CD151 derived peptides for inhibition of CD151 homophilic interactions.
  • T cell CD151 is a key orchestrator of T cell motility, and its targeting by inhibitory peptides results in beneficial suppression of inflammation.
  • an isolated CD 151 peptide (having a CD151 amino acid sequence, also termed herein "peptide") comprising at least 20 consecutive amino acids of the amino acid sequence set forth in SEQ ID NO: 2, the peptide being shorter than 100 amino acids and having a CD151 inhibitory activity.
  • CD151 refers to a CD151 gene product (i.e., protein or mRNA) such as set forth in GenBank Accession Nos. NM_004357.4, NM_139030.3, NMJ39029.1, NM 001039490.1, NP_004348.2, NP_620599.1, NP_620598.1 and NP 001034579.1.
  • the CD151 is human CD151 such as set forth in SEQ ID NO: 1.
  • isolated refers to isolated or purified from the natural environment, e.g., biological samples e.g., serum, cells or tissues.
  • CD151 activity refers to a cell signaling activity (e.g., vav phosphorylation, ⁇ -catenin signaling), self-ligation activity (i.e., cdl51 homophilic interaction), actin cytoskeleton polymerization, cell adhesion, cell spreading, cell migration (e.g. towards a chemokine such as CCL21 or CXCL12) or homing (e.g. to a lymphoid organ).
  • a cell signaling activity e.g., vav phosphorylation, ⁇ -catenin signaling
  • self-ligation activity i.e., cdl51 homophilic interaction
  • actin cytoskeleton polymerization e.g., cell adhesion, cell spreading, cell migration (e.g. towards a chemokine such as CCL21 or CXCL12) or homing (e.g. to a lymphoid organ).
  • the CD151 inhibitory activity comprises inhibition of lymphocyte migration or homing.
  • the migration or homing is to a lymphoid organ.
  • lymphocyte refers to a white blood cell of the immune system.
  • a lymphocyte may include a T cell, a B cell and natural killer (NK) cell.
  • the lymphocyte is a T cell including CD4+ T cells, CD8+ T cells and any subsets therefrom (including naive, mature and memory T cells).
  • the CD151 inhibitory activity comprises inhibition of Vav phosphorylation.
  • the present invention provides ample teachings for assessing CD151 signaling, migration and homing, see Example 2 of the Examples section which follows.
  • the CD151 peptide can be at least 15 amino acids but shorter than 1 10 amino acids.
  • the Cdl51 amino acid sequence is 15- 110, 20-110, 20-100, 30-100, 50-100, 20-70, 30-70, 40-70, 50-70, 60-70 or 62-70, 63-70 amino acids long. It will be appreciated that any groups of ranges or examples provided herein is meant to read over the whole group or specific individual ranges/examples comprised therein.
  • the peptide is as set forth in SEQ ID NO: 2.
  • the peptide is soluble.
  • soluble refers to the absence of a cell-membrane anchoring moiety.
  • a molecule which comprises the CD151 peptide attached to a heterologous moiety forming.
  • the heterologous moiety may be a proteinaceous moiety i.e., a heterologous amino acid sequence.
  • heterologous amino acid sequence refers to an amino acid sequence which does not form a consecutive part of the CD151 polypeptide in nature. According to a specific embodiment the heterologous amino acid sequence is non-immunogenic. This sequence can confer solubility to the peptide of the present invention, thereby increasing the half-life of such a fusion polypeptide molecule in the serum.
  • heterologous amino acid sequence is generally localized at the amino- or carboxyl- terminus of the CD 151 of the present invention.
  • At least one heterologous amino acid sequence can be contiguously (N-terminally or C-terminally) attached to the CD151 peptide of the present invention.
  • the heterologous amino acid sequence may be attached to the CD151 amino acid sequence by any of peptide or non-peptide bond. Attachment of the CD 151 amino acid sequence to the heterologous amino acid sequence may be effected by direct covalent bonding (peptide bond or a substituted peptide bond) or indirect binding such as by the use of a linker having functional groups.
  • heterologous amino acid sequence which may be used in accordance with this aspect of the present invention is an immunoglobulin amino acid sequence, such as the hinge and Fc regions of an immunoglobulin heavy domain (see U.S. Pat. No. 6,777,196).
  • the immunoglobulin moiety in the chimeras of this aspect of the present invention may be obtained from IgGl, IgG2, IgG3 or IgG4 subtypes, IgA, IgE, IgD or IgM, as further discussed hereinbelow.
  • Immunoadhesins Chimeras constructed from a receptor sequence linked to an appropriate immunoglobulin constant domain sequence (immunoadhesins) are known in the art. Immunoadhesins reported in the literature include fusions of the T cell receptor; CD4; L- selectin (homing receptor); CD44; CD28 and B7; CTLA-4; CD22; TNF receptor; NP receptors; and IgE receptor a.
  • the chimeric molecule will retain at least functionally active hinge and CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain. Fusions can also be generated to the C-terminus of the Fc portion of a constant domain, or immediately N-terminal to the CHI of the heavy chain or the corresponding region of the light chain.
  • the exact site at which fusion (conjugation) between the heterologous sequence and the CD151 amino acid sequence is not critical. Particular sites are well known in the art and may be selected in order to optimize the biological activity, secretion or binding characteristics of such chimeric molecules of the present invention.
  • the CD151 amino acid sequence of the present invention may be conjugated to the CD151 amino acid sequence of the present invention, it is preferable to fuse shorter sequences.
  • a sequence beginning in the hinge region just upstream of the papain cleavage site, which defines IgG Fc chemically; residue 216, taking the first residue of heavy chain constant region to be 114, or analogous sites of other immunoglobulins is used in the fusion.
  • the CD151 amino acid sequence is fused to the hinge region and CH2 and CH3, or to the CHI, hinge, CH2 and CH3 domains of an IgGl, IgG2, or IgG3 heavy chain (see U.S. Pat. No. 6,777,196).
  • the precise site at which the fusion is made is not critical, and the optimal site can be determined by routine experimentation.
  • the immunoglobulin sequences used in the construction of the chimeric molecules of this aspect of the present invention may be from an IgG immunoglobulin heavy chain constant domain.
  • the use of human IgGl immunoglobulin sequences is exemplary.
  • a major advantage of using IgGl is that IgGl can be purified efficiently on immobilized protein A.
  • other structural and functional properties of immunoglobulins should be considered when choosing the Ig fusion partner for a particular chimera construction.
  • the IgG3 hinge is longer and more flexible, so it can accommodate larger CD151 amino acid sequences that may not fold or function properly when fused to IgGl .
  • IgG are bivalent homodimers, whereas Ig subtypes like IgA and IgM may give rise to dimeric or pentameric structures, respectively, of the basic Ig homodimer unit.
  • IgA and IgM may give rise to dimeric or pentameric structures, respectively, of the basic Ig homodimer unit.
  • Other considerations in selecting the immunoglobulin portion of the chimeric molecules of this aspect of the present invention are described in U.S. Pat. No. 6,777,196.
  • heterologous amino acid sequences i.e., moieties
  • GST glutathione-S-transferase
  • CTP carboxy terminal peptide from chorionic gonadotrophin
  • CAT chloramphenicol acetyltransferase
  • molecules i.e., peptide fusions
  • molecules may comprise heterologous amino acid sequences, as described above.
  • the peptide i.e., CD151 amino acid sequence
  • a non-proteinaceous moiety such molecules are preferably selected non-immunogenic in a subject (e.g., human subject).
  • Such a molecule is highly stable (resistant to in-vivo proteolytic activity probably due to steric hindrance conferred by the non-proteinaceous moiety) and may be produced using common solid phase synthesis methods which are inexpensive and highly efficient, as further described hereinbelow.
  • recombinant techniques may still be used, whereby the recombinant peptide product is subjected to in-vitro modification (e.g., PEGylation as further described hereinbelow).
  • non-proteinaceous moiety refers to a molecule not including peptide bonded amino acids that is attached to the above-described CD151 amino acid sequence.
  • non-proteinaceous moieties may be also attached to the above mentioned fusion molecules (i.e., which comprise a heterologous amino acid sequence) to promote stability and possibly solubility of the molecules.
  • Bioconjugation of such a non-proteinaceous moiety can confer the CD151 amino acid sequence with stability (e.g., against protease activities) and/or solubility (e.g., within a biological fluid such as blood, digestive fluid) while preserving its biological activity and prolonging its half-life.
  • stability e.g., against protease activities
  • solubility e.g., within a biological fluid such as blood, digestive fluid
  • peptide encompasses native peptides (either degradation products, synthetically synthesized peptides or recombinant peptides) and peptidomimetics (typically, synthetically synthesized peptides), as well as peptoids and semipeptoids which are peptide analogs, which may have, for example, modifications rendering the peptides more stable while in a body or more capable of penetrating into cells.
  • Methods for preparing peptidomimetic compounds are well known in the art and are specified, for example, in Quantitative Drug Design, C.A. Ramsden Gd., Chapter 17.2, F. Choplin Pergamon Press (1992), which is incorporated by reference as if fully set forth herein. Further details in this respect are provided hereinunder.
  • Natural aromatic amino acids, Trp, Tyr and Phe may be substituted for synthetic non-natural acid such as TIC, naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe or o-methyl-Tyr.
  • synthetic non-natural acid such as TIC, naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe or o-methyl-Tyr.
  • the peptides of the present invention may also include one or more modified amino acids or one or more non-amino acid monomers (e.g. fatty acids, complex carbohydrates etc).
  • amino acid or “amino acids” is understood to include the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine and phosphothreonine; and other unusual amino acids including, but not limited to, 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine and ornithine.
  • amino acid includes both D- and L-amino acids.
  • the peptides of the present invention are preferably utilized in a linear form, although it will be appreciated that in cases where cyclicization does not severely interfere with peptide characteristics, cyclic forms of the peptide can also be utilized.
  • the peptides of the present invention may be synthesized by any techniques that are known to those skilled in the art of peptide synthesis.
  • solid phase peptide synthesis a summary of the many techniques may be found in J. M. Stewart and J. D. Young, Solid Phase Peptide Synthesis, W. H. Freeman Co. (San Francisco), 1963 and J. Meienhofer, Hormonal Proteins and Peptides, vol. 2, p. 46, Academic Press (New York), 1973.
  • For classical solution synthesis see G. Schroder and K. Lupke, The Peptides, vol. 1, Academic Press (New York), 1965.
  • these methods comprise the sequential addition of one or more amino acids or suitably protected amino acids to a growing peptide chain.
  • amino acids or suitably protected amino acids Normally, either the amino or carboxyl group of the first amino acid is protected by a suitable protecting group.
  • the protected or derivatized amino acid can then either be attached to an inert solid support or utilized in solution by adding the next amino acid in the sequence having the complimentary (amino or carboxyl) group suitably protected, under conditions suitable for forming the amide linkage.
  • the protecting group is then removed from this newly added amino acid residue and the next amino acid (suitably protected) is then added, and so forth. After all the desired amino acids have been linked in the proper sequence, any remaining protecting groups (and any solid support) are removed sequentially or concurrently, to afford the final peptide compound.
  • a method of preparing the peptide compounds of the present invention involves solid phase peptide synthesis.
  • a method of producing the peptide of the invention involves recombinant techniques such as described by Bitter et al. (1987) Methods in Enzymol. 153:516-544; Studier et al. (1990) Methods in Enzymol. 185:60-89; Brisson et al. (1984) Nature 310:511-514; Takamatsu et al. (1987) EMBO J. 6:307-311; Coruzzi et al. (1984) EMBO J. 3: 1671-1680; Brogli et al. (1984) Science 224:838-843; Gurley et al. (1986) Mol. Cell. Biol. 6:559-565 and Weissbach & Weissbach, 1988, Methods for Plant Molecular Biology, Academic Press, NY, Section VIII, pp 421-463.
  • an expression construct i.e., expression vector
  • an isolated polynucleotide encoding the CD151 peptide of the invention e.g., SEQ ID NO: 3 encoding SEQ ID NO: 2
  • a nucleic acid sequence encoding the heterologous amino acid is positioned under the transcriptional control of a regulatory element, such as a promoter and is introduced into host cells.
  • Methods of introducing the expression construct into a host cell include, electroporation, lipofection and chemical transformation (e.g., calcium phosphate).
  • the "transformed” cells are cultured under suitable conditions, which allow the expression of the chimeric molecule encoded by the nucleic acid sequence.
  • the expressed peptide or chimeric peptide (including the heterologous amino acid sequence described above) is recovered from the cell or cell culture, and purification is effected according to the end use of the recombinant polypeptide.
  • any of a number of suitable transcription and translation elements including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, and the like, can be used in the expression vector [see, e.g., Bitter et al, (1987) Methods in Enzymol. 153:516-544].
  • the expression construct of the present invention can also include sequences engineered to optimize stability, production, purification, yield or toxicity of the expressed fusion protein.
  • prokaryotic or eukaryotic cells can be used as host-expression systems to express the fusion protein coding sequence.
  • These include, but are not limited to, microorganisms, such as bacteria transformed with a recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vector containing the peptide coding sequence; yeast transformed with recombinant yeast expression vectors containing the peptide coding sequence; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors, such as Ti plasmid, containing the chimera coding sequence.
  • Mammalian expression systems are preferably used to express the peptide of the present invention.
  • the choice of host cell line for the expression of the molecules depends mainly on the expression vector. Eukaryotic expression systems are preferred (e.g., mammalian and insects) since they allow post translational modifications (e.g., glyccosylation). Another consideration is the amount of protein that is required. Milligram quantities often can be produced by transient transfections.
  • the adenovirus EIA- transformed 293 human embryonic kidney cell line can be transfected transiently with pR 5 -based vectors by a modification of the calcium phosphate method to allow efficient expression.
  • CDM8-based vectors can be used to transfect COS cells by the DEAE-dextran method.
  • the molecules can be expressed after stable transfection of a host cell line. It will be appreciated that the presence of a hydrophobic leader sequence at the N-terminus of the molecule will ensure processing and secretion of the molecule by the transfected cells.
  • bacterial or yeast host systems may be preferable to reduce cost of production.
  • bacterial host systems are devoid of protein glycosylation mechanisms, a post production glycosylation may be needed.
  • transformed cells are cultured under effective conditions, which allow for the expression of high amounts of recombinant polypeptide.
  • Effective culture conditions include, but are not limited to, effective media, bioreactor, temperature, pH and oxygen conditions that permit protein production.
  • An effective medium refers to any medium in which a cell is cultured to produce the recombinant peptide molecule of the present invention.
  • Such a medium typically includes an aqueous solution having assimilable carbon, nitrogen and phosphate sources, and appropriate salts, minerals, metals and other nutrients, such as vitamins.
  • Cells of the present invention can be cultured in conventional fermentation bioreactors, shake flasks, test tubes, microtiter dishes, and petri plates. Culturing can be carried out at a temperature, pH and oxygen content appropriate for a recombinant cell. Such culturing conditions are within the expertise of one of ordinary skill in the art.
  • resultant proteins of the present invention may either remain within the recombinant cell, secreted into the fermentation medium, secreted into a space between two cellular membranes, such as the periplasmic space in E. coli; or retained on the outer surface of a cell or viral membrane.
  • recovery of the recombinant protein refers to collecting the whole fermentation medium containing the protein and need not imply additional steps of separation or purification.
  • Peptides of the present invention can be purified using a variety of standard protein purification techniques, such as, but not limited to, affinity chromatography, ion exchange chromatography, filtration, electrophoresis, hydrophobic interaction chromatography, gel filtration chromatography, reverse phase chromatography, concanavalin A chromatography, chromato focusing and differential solubilization.
  • Molecules of the present invention are preferably retrieved in "substantially pure” form.
  • substantially pure refers to a purity that allows for the effective use of the protein in the applications, described hereinbelow.
  • inhibitory activity of the peptides of the invention as manifested by their ability to inhibit cell migration and Vav signaling supports their use as agents in the treatment of inflammation.
  • a method of treating inflammation in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the isolated peptide or molecule described above, thereby treating the inflammation in the subject.
  • the term "subject in need thereof refers to a mammal, preferably a human subject at any age which may benefit from the treatment modality of the present invention. According to specific embodiments, the subject does not suffer from a medical condition which is not associated with inflammation.
  • the subject is a human being.
  • inflammation refers to any medical condition which comprises an inflammatory response in which migration of cells (e.g. to the lymph nodes) contributes to inflammation onset or progression.
  • Inflammatory diseases include chronic inflammatory diseases and acute inflammatory diseases.
  • hypersensitivity examples include, but are not limited to, Type I hypersensitivity, Type II hypersensitivity, Type III hypersensitivity, Type IV hypersensitivity, immediate hypersensitivity, antibody mediated hypersensitivity, immune complex mediated hypersensitivity, T lymphocyte mediated hypersensitivity and DTH.
  • Type I or immediate hypersensitivity such as asthma.
  • Type II hypersensitivity include, but are not limited to, rheumatoid diseases, rheumatoid autoimmune diseases, rheumatoid arthritis (Krenn V. et ah, Histol Histopathol 2000 Jul;15 (3):791), spondylitis, ankylosing spondylitis (Jan Voswinkel et al, Arthritis Res 2001; 3 (3): 189), systemic diseases, systemic autoimmune diseases, systemic lupus erythematosus (Erikson J. et al, Immunol Res 1998;17 (l-2):49), sclerosis, systemic sclerosis (Renaudineau Y. et al, Clin Diagn Lab Immunol.
  • myasthenic diseases myasthenic diseases, Lambert-Eaton myasthenic syndrome (Takamori M. Am J Med Sci. 2000 Apr;319 (4):204), paraneoplastic neurological diseases, cerebellar atrophy, paraneoplastic cerebellar atrophy, non-paraneoplastic stiff man syndrome, cerebellar atrophies, progressive cerebellar atrophies, encephalitis, Rasmussen's encephalitis, amyotrophic lateral sclerosis, Sydeham chorea, Gilles de la Tourette syndrome, polyendocrinopathies, autoimmune polyendocrinopathies (Antoine JC. and Honnorat J.
  • vasculitises necrotizing small vessel vasculitises, microscopic polyangiitis, Churg and Strauss syndrome, glomerulonephritis, pauci-immune focal necrotizing glomerulonephritis, crescentic glomerulonephritis (Noel LH. Ann Med Interne (Paris). 2000 May;151 (3): 178); antiphospholipid syndrome (Flamholz R. et al, J Clin Apheresis 1999; 14 (4): 171); heart failure, agonist- like beta-adrenoceptor antibodies in heart failure (Wallukat G. et al, Am J Cardiol.
  • Type IV or T cell mediated hypersensitivity include, but are not limited to, rheumatoid diseases, rheumatoid arthritis (Tisch R, McDevitt HO. Proc Natl Acad Sci U S A 1994 Jan 18; 91 (2):437), systemic diseases, systemic autoimmune diseases, systemic lupus erythematosus (Datta SK., Lupus 1998; 7 (9):591), glandular diseases, glandular autoimmune diseases, pancreatic diseases, pancreatic autoimmune diseases, Type 1 diabetes (Castano L. and Eisenbarth GS. Ann. Rev. Immunol. 8:647); thyroid diseases, autoimmune thyroid diseases, Graves' disease (Sakata S.
  • delayed type hypersensitivity examples include, but are not limited to, contact dermatitis and drug eruption.
  • T lymphocyte mediating hypersensitivity examples include, but are not limited to, helper T lymphocytes and cytotoxic T lymphocytes.
  • helper T lymphocyte-mediated hypersensitivity examples include, but are not limited to, T l lymphocyte mediated hypersensitivity and T 2 lymphocyte mediated hypersensitivity.
  • cardiovascular diseases include, but are not limited to, cardiovascular diseases, rheumatoid diseases, glandular diseases, gastrointestinal diseases, cutaneous diseases, hepatic diseases, neurological diseases, muscular diseases, nephric diseases, diseases related to reproduction, connective tissue diseases and systemic diseases.
  • autoimmune cardiovascular diseases include, but are not limited to atherosclerosis (Matsuura E. et al, Lupus. 1998;7 Suppl 2:S135), myocardial infarction (Vaarala O. Lupus. 1998;7 Suppl 2:S132), thrombosis (Tincani A. et al, Lupus 1998;7 Suppl 2:S 107-9), Wegener's granulomatosis, Takayasu's arteritis, Kawasaki syndrome (Praprotnik S. et al, Wien Klin Klin Klin Klinschr 2000 Aug 25;112 (15-16):660), anti-factor VIII autoimmune disease (Lacroix-Desmazes S.
  • autoimmune rheumatoid diseases include, but are not limited to rheumatoid arthritis (Krenn V. et al, Histol Histopathol 2000 Jul;15 (3):791; Tisch R, McDevitt HO. Proc Natl Acad Sci units S A 1994 Jan 18;91 (2):437) and ankylosing spondylitis (Jan Voswinkel et al, Arthritis Res 2001; 3 (3): 189).
  • autoimmune glandular diseases include, but are not limited to, pancreatic disease, Type I diabetes, thyroid disease, Graves' disease, thyroiditis, spontaneous autoimmune thyroiditis, Hashimoto's thyroiditis, idiopathic myxedema, ovarian autoimmunity, autoimmune anti-sperm infertility, autoimmune prostatitis and Type I autoimmune polyglandular syndrome.
  • Diseases include, but are not limited to autoimmune diseases of the pancreas, Type 1 diabetes (Castano L. and Eisenbarth GS. Ann. Rev. Immunol. 8:647; Zimmet P. Diabetes Res Clin Pract 1996 Oct;34 Suppl:S125), autoimmune thyroid diseases, Graves' disease (Orgiazzi J.
  • autoimmune gastrointestinal diseases include, but are not limited to, chronic inflammatory intestinal diseases (Garcia Herola A. et ah, Gastroenterol Hepatol. 2000 Jan; 23 (1): 16), celiac disease (Landau YE. and Shoenfeld Y. Harefuah 2000 Jan 16; 138 (2): 122), colitis, ileitis and Crohn's disease (see Example 3).
  • autoimmune cutaneous diseases include, but are not limited to, autoimmune bullous skin diseases, such as, but are not limited to, pemphigus vulgaris, bullous pemphigoid and pemphigus foliaceus.
  • autoimmune hepatic diseases include, but are not limited to, hepatitis, autoimmune chronic active hepatitis (Franco A. et al, Clin Immunol Immunopathol 1990 Mar; 54 (3):382), primary biliary cirrhosis (Jones DE. Clin Sci (Colch) 1996 Nov; 91 (5):551 ; Strassburg CP. et al, Eur J Gastroenterol Hepatol. 1999 Jun; 1 1 (6):595) and autoimmune hepatitis (Manns MP. J Hepatol 2000 Aug; 33 (2):326).
  • autoimmune neurological diseases include, but are not limited to, multiple sclerosis (Cross AH. et al, J Neuroimmunol 2001 Jan 1 ; 1 12 (1-2): 1), Alzheimer's disease (Oron L. et al, J Neural Transm Suppl. 1997;49:77), myasthenia gravis (Infante AJ. And Kraig E, Int Rev Immunol 1999; 18 (l-2):83; Oshima M. et al, Eur J Immunol 1990 Dec;20 (12):2563), neuropathies, motor neuropathies (Kornberg AJ. J Clin Neurosci.
  • autoimmune muscular diseases include, but are not limited to, myositis, autoimmune myositis and primary Sjogren's syndrome (Feist E. et al, Int Arch Allergy Immunol 2000 Sep; 123 (1):92) and smooth muscle autoimmune disease (Zauli D. et al, Biomed Pharmacother 1999 Jun;53 (5-6):234).
  • autoimmune nephric diseases include, but are not limited to, nephritis and autoimmune interstitial nephritis (Kelly CJ. J Am Soc Nephrol 1990 Aug; 1 (2): 140).
  • autoimmune diseases related to reproduction include, but are not limited to, repeated fetal loss (Tincani A. et ah, Lupus 1998; 7 Suppl 2:S107-9).
  • autoimmune connective tissue diseases include, but are not limited to, ear diseases, autoimmune ear diseases (Yoo TJ. et ah, Cell Immunol 1994 Aug; 157 (1):249) and autoimmune diseases of the inner ear (Gloddek B. et ah, Ann N Y Acad Sci 1997 Dec 29; 830:266).
  • autoimmune systemic diseases include, but are not limited to, systemic lupus erythematosus (Erikson J. et ah, Immunol Res 1998;17 (l-2):49) and systemic sclerosis (Renaudineau Y. et ah, Clin Diagn Lab Immunol. 1999 Mar;6 (2): 156); Chan OT. et al, Immunol Rev 1999 Jun; 169: 107).
  • infectious diseases include, but are not limited to, chronic infectious diseases, subacute infectious diseases, acute infectious diseases, viral diseases, bacterial diseases, protozoan diseases, parasitic diseases, fungal diseases, mycoplasma diseases and prion diseases.
  • diseases associated with transplantation of a graft include, but are not limited to, graft rejection, chronic graft rejection, subacute graft rejection, hyperacute graft rejection, acute graft rejection and graft versus host disease.
  • allergic diseases include, but are not limited to, asthma, hives, urticaria, pollen allergy, dust mite allergy, venom allergy, cosmetics allergy, latex allergy, chemical allergy, drug allergy, insect bite allergy, animal dander allergy, stinging plant allergy, poison ivy allergy and food allergy.
  • cancer examples include but are not limited to carcinoma, lymphoma, blastoma, sarcoma, and leukemia.
  • cancerous diseases include but are not limited to: Myeloid leukemia such as Chronic myelogenous leukemia. Acute myelogenous leukemia with maturation. Acute promyelocytic leukemia, Acute nonlymphocytic leukemia with increased basophils, Acute monocytic leukemia. Acute myelomonocytic leukemia with eosinophilia; Malignant lymphoma, such as Birkitt's Non-Hodgkin's; Lymphocytic leukemia, such as Acute lumphoblastic leukemia.
  • Myeloid leukemia such as Chronic myelogenous leukemia. Acute myelogenous leukemia with maturation. Acute promyelocytic leukemia, Acute nonlymphocytic leukemia with increased basophils, Acute monocytic leuk
  • Chronic lymphocytic leukemia Myeloproliferative diseases, such as Solid tumors Benign Meningioma, Mixed tumors of salivary gland, Colonic adenomas; Adenocarcinomas, such as Small cell lung cancer, Kidney, Uterus, Prostate, Bladder, Ovary, Colon, Sarcomas, Liposarcoma, myxoid, Synovial sarcoma, Rhabdomyosarcoma (alveolar), Extraskeletel myxoid chonodrosarcoma, Ewing's tumor; other include Testicular and ovarian dysgerminoma, Retinoblastoma, Wilms' tumor, Neuroblastoma, Malignant melanoma, Mesothelioma, breast, skin, prostate, and ovarian.
  • Adenocarcinomas such as Small cell lung cancer, Kidney, Uterus, Prostate, Bladder, Ovary, Colon, Sarcomas, Lipos
  • the inflammatory condition is a leukemia or lymphoma.
  • the inflammatory condition is rheumatoid arthritis, inflammatory bowel disease (e.g. ulcerative colitis and Crohn's disease) and asthma.
  • each of the peptides or molecules described hereinabove can be administered to the subject per se or as part of a pharmaceutical composition which also includes a physiologically acceptable carrier.
  • a pharmaceutical composition which also includes a physiologically acceptable carrier.
  • the purpose of a pharmaceutical composition is to facilitate administration of the active ingredient to an organism.
  • a "pharmaceutical composition” refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • active ingredient refers to the peptide having the CD151 inhibitory activity accountable for the biological effect.
  • pharmaceutically acceptable carrier refers to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • An adjuvant is included under these phrases.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intracardiac, e.g., into the right or left ventricular cavity, into the common coronary artery, intravenous, inrtaperitoneal, intranasal, or intraocular injections.
  • compositions of the present invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer.
  • physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the pharmaceutical composition can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient.
  • Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the active ingredients for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • compositions described herein may be formulated for parenteral administration, e.g., by bolus injection or continuos infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative.
  • the compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen- free water based solution, before use.
  • a suitable vehicle e.g., sterile, pyrogen- free water based solution
  • compositions of the present invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
  • compositions suitable for use in context of the present invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of active ingredients (the peptide) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., inflammation) or prolong the survival of the subject being treated.
  • a therapeutically effective amount means an amount of active ingredients (the peptide) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., inflammation) or prolong the survival of the subject being treated.
  • the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays.
  • a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
  • Animal models for inflammatory colon diseases include animal models of ulcerative colitis such as, but are not limited to, trinitrobenzene sulfonic acid (TNBS)- induced colitis in rats and mice [ Komori et al., J Gastroenterol (2005) 40: 591-599].
  • An animal model for adjuvant arthritis (AA, a model of rheumatoid arthritis) includes the rat heat-killed Mt strain H37Ra-induced AA [Kannan, Theor Biol Med Model. (2005) 2: 17].
  • An animal model for asthma includes the Ovalbumin (OVA) sensitization mouse model [Flaishon, L., et al, J. Immunol: Cutting edge 168: 3707 (2002)].
  • OVA Ovalbumin
  • Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals.
  • the data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage may vary depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p.l).
  • Dosage amount and interval may be adjusted individually to provide sufficient levels of the active ingredient to induce or suppress the biological effect (minimal effective concentration, MEC).
  • MEC minimum effective concentration
  • the MEC will vary for each preparation, but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
  • dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
  • the amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • the dosage and timing of administration will be responsive to a careful and continuous monitoring of the individual changing condition.
  • compositions of the present invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert.
  • Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as is further detailed above.
  • the subject may be administered with additional chemical drugs (e.g., anti-inflammatory).
  • additional chemical drugs e.g., anti-inflammatory
  • Anti-inflammatory agents which may be used according to the present teachings include, but are not limited to, Alclofenac; Alclometasone Dipropionate; Algestone Acetonide; Alpha Amylase; Amcinafal; Amcinafide; Amfenac Sodium; Amiprilose Hydrochloride; Anakinra; Anirolac; Anitrazafen; Apazone; Balsalazide Disodium; Bendazac; Benoxaprofen; Benzydamine Hydrochloride; Bromelains; Broperamole; Budesonide; Carprofen; Cicloprofen; Cintazone; Cliprofen; Clobetasol Propionate; Clobetasone Butyrate; Clopirac; Cloticasone Propionate; Cormethasone Acetate; Cortodoxone; Deflazacort; Desonide; Desoximetasone; Dexamethasone Dipropionate; Diclofenac Potassium; Diclofenac Sodium
  • Flurbiprofen Fluretofen; Fluticasone Propionate; Furaprofen; Furobufen; Halcinonide;
  • Halobetasol Propionate Halopredone Acetate; Ibufenac; Ibuprofen; Ibuprofen
  • Ibuprofen Piconol Inol
  • Ibuprofen Piconol Inonidap
  • Indomethacin Indomethacin Sodium
  • Indoprofen Indoxole
  • Intrazole Isoflupredone Acetate
  • Isoxepac Isoxicam
  • Ketoprofen Lofemizole Hydrochloride; Lomoxicam; Loteprednol Etabonate;
  • Nabumetone Naproxen; Naproxen Sodium; Naproxol; Nimazone; Olsalazine Sodium; Orgotein; Orpanoxin; Oxaprozin; Oxyphenbutazone; Paranyline Hydrochloride;
  • Piroxicam Piroxicam Cinnamate; Piroxicam Olamine; Pirprofen; Prednazate; Prifelone;
  • Prodolic Acid Proquazone; Proxazole; Proxazole Citrate; Rimexolone; Romazarit;
  • Salcolex Salcolex; Salnacedin; Salsalate; Sanguinarium Chloride; Seclazone; Sermetacin; Sudoxicam; Sulindac; Suprofen; Talmetacin; Talniflumate; Talosalate; Tebufelone;
  • Tixocortol Pivalate Tolmetin; Tolmetin Sodium; Triclonide; Triflumidate;
  • compositions, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
  • At least one compound may include a plurality of compounds, including mixtures thereof.
  • range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
  • method refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
  • treating includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition.
  • CD151 has a significant role in T cell migration, its activation induces T cell migration in vitro and in vivo.
  • blocking CD151 results in inhibition of T cell migration in vitro and their entry to the lymph nodes (LN) in vivo.
  • LN lymph nodes
  • CD151-ECD SEQ ID NO: 2 (Entelechon GmbH).
  • CD151ECD regulates T cell migration
  • T cells were stimulated with 100 ng/ml CCL21 chemokine (SLC) (PeproTech). Cytoskeleton rearrangement was analyzed by flow cytometry following staining with FITC-phalloidin, as previously described (Flaishon, L., et al.,. J. Biol. Chem., 2001 276(50):46701-6). Transwell migration
  • chemotaxis was assayed by using transwell chambers, as previously described (Flaishon, L., et al.,. J. Biol. Chem., 2001). Briefly, about 4 x 10 6 T cells were pretreated with either stimulating or blocking anti-CD151 antibody for 1 hour as described in Methods. The migration towards the chemokine CCL21 (SLC) (0.4 mg/ml) (PeproTech) residing in the lower part of the apparatus, was analyzed after 3 hours by F AC Sort.
  • SLC chemokine CCL21
  • the assays were double controlled using SEQ ID NO: 4 as an additional independent control peptide (results not shown).
  • Protein G-Sepharose beads (GE Healthcare) were conjugated to Tyr (P) monoclonal antibody (Santa Cruz) for 2 hours on 4 °C, followed by three washes in phosphate-buffered saline. Beads were added to the cell lysates and proteins were immunoprecipitated overnight. The protein G-bound material was washed three times with phosphate-buffered saline containing 0.1% SDS and 0.5% Nonidet P-40. Immunoprecipitates were separated by SDS-PAGE.
  • the protein bands were transferred onto a nitrocellulose membrane and probed with anti-Vavl (Cell Signaling) followed by horseradish peroxidase-conjugated anti-mouse (Jackson ImmunoResearch Labs,West Grove, PA).
  • CD151ECD regulates T cell migration
  • the present inventors have first analyzed the ability of T cells to polymerize their actin in the presence of CD151ECD or a control peptide.
  • T cells were incubated with various concentrations of CD151ECD (10-80 ⁇ g/ml) or a control peptide for 1 hour, and then stimulated with CCL21 (0.1 mg/ml) (PeproTech) for 15 sec.
  • CCL21 0.1 mg/ml
  • the cells were immediately fixed with paraformaldehyde, permeabilized, stained with FITC-phalloidin, and then analyzed by flow cytometry to determine the state of their cytoskeleton.
  • CD151ECD specifically inhibited the ability of T cells to polymerize their actin at the concentrations of 20 ⁇ g/ml and more significantly at 40 ⁇ g/ml.
  • CD151ECD regulates in vitro migration of T cells using a Transwell migration assay.
  • Naive T cells were suspended with CD151ECD or a control peptide (40 ⁇ / ⁇ 1) for 1 hr, and then placed in the upper chamber of a transwell.
  • CD151ECD significantly inhibited the migration of control naive T cells toward CCL21 by about 60%.
  • CD151ECD down-regulated the Vav 1 -phosphorylation.
  • CD151-ECD peptide inhibits the inflammatory response in DSS Colitis Induction of DSS Colitis
  • mice were divided into three groups with 10 mice in each group. Each group was intraperitoneally (i.p.) injected with PBS, 80 ⁇ g CD151-ECD (SEQ ID NO: 2), or control scrambled peptide (SEQ ID NO: 5 Entelechon GmbH) in 200 of PBS from day 0 (immediately after DSS induction) to day 10, daily.
  • PBS 80 ⁇ g CD151-ECD
  • SEQ ID NO: 5 Entelechon GmbH control scrambled peptide
  • Murine Colonoscopy Mice underwent murine colonoscopy on day 7 after disease induction.
  • a high-resolution murine video endoscopic system consisting of a miniature probe (1.9 mm outer diameter), a xenon light source, a triple chip HD camera, and an air pump ("Colo view,” Karl Storz) to achieve regulated inflation of the mouse colon.
  • Digitally recorded video files were processed with Windows Movie Maker software (Microsoft).
  • Endoscopic quantification of colitis was graded using the MEICS (murine endoscopic index of colitis severity) system which consisted of five parameters: thickening of the colon wall, changes in the normal vascular pattern, presence of fibrin, mucosal granularity and stool consistency as described (Becker et al, 2005 GUT 54:950-954).
  • Histological assessment of DSS Colitis - Colons were fixed in 4 % paraformaldehyde for histology with hematoxylin and eosin (H&E) staining. The degree of histological damage and inflammation was graded in a blinded fashion. The amount of inflammation (0-4) was evaluated by estimation of the number of inflammatory cells.
  • CD151-ECD dextran sulfate sodium
  • mice were administered 1% DSS in their drinking water for 5 days, followed by a 5 day rest period. During those 10 days, each group was intraperitoneally (i.p.) injected with CD151-ECD or control peptides. Body weight was monitored daily. On day 11 of the experiment, mice were sacrificed and the colon length was measured and was taken to histological evaluation. As shown in Figure 3 A, CD151-ECD had a favorable effect on weight maintenance, as the percentage of weight loss was significantly lower in the CD151-ECD group in comparison to the control peptide injected group.
  • This CD 151-ECD peptide acts as a competitive inhibitor.
  • the truncated CD151- ECD fragment was shown to inhibit the CD 151 induced- signaling cascade resulting in inhibition of Vav-1 phosphorylation, downregulation of actin polymerization and in vitro T cell migration.
  • Analysis of the effect of CD 151-ECD in the DSS-induced colitis model revealed that daily administration of CD 151-ECD significantly decreased the development of DSS-induced colitis, as reflected by its favorable effect on weight loss, shortening of the colon, endoscopic colitis score and histological inflammation score.

Abstract

An isolated peptide comprising at least 20 consecutive amino acids of the amino acid sequence set forth in SEQ ID NO: 2, the peptide being shorter than 100 amino acids and having a CD151 inhibitory activity. Also provided are uses of this peptide.

Description

COMPOSITIONS AND METHODS FOR TREATING INFLAMMATION
FIELD AND BACKGROUND OF THE INVENTION
The present invention, in some embodiments thereof, relates to compositions and methods for treating inflammation.
The surveillance of the body for foreign antigens is a critical function of the immune system. B and T lymphocytes develop in the primary lymphoid organs while their capabilities are employed throughout the body. Accordingly, B and T lymphocytes continually patrol the body for foreign antigens by circulating from the blood, through tissues, into lymph nodes (LNs), and back to the blood. Lymphocyte migration out of the vasculature is therefore a key regulatory point for controlling immune responses. It is controlled by multi-step processes that include chemoattraction, cell-cell adhesion and transmigration through cell layers. The continuous recirculation of lymphocytes and their movement from the blood into peripheral tissues is a central feature of immune surveillance, but also contributes to the pathogenesis of inflammatory diseases, autoimmune diseases, as well as the development and progression of allergic responses.
VLA4, a cell surface heterodimer in the integrin superfamily of adhesion receptors, is expressed on several different circulating blood cell types, including T cells. VLA4 is involved in cell-cell interactions and mediates cell attachment to vascular cell adhesion molecule- 1 (VCAM-1), which is expressed on the surface of endothelial cells as a result of their exposure to cytokines. The VLA-4-VCAM-1 interaction is responsible, at least in part, for recruitment of T cells to areas of vascular inflammation and plays an essential role during various types of inflammation [Lobb and Hemler, J Clin Invest (1994) 94(5): 1722-8; Postigo et al, Res Immunol (1993) 144(9): 723-35; discussion 754-62]. VLA-4 also binds to fibronectin on the extracellular matrix (ECM) and this interaction is important for migration of T lymphocytes to antigenic sites [Mannion et al, J Immunol (1996) 157(5): 2039-47; Yuan et al, Biochem J (1996) 318 (Pt 2): 591-6], thus VLA-4 expression is critical for the migration of T lymphocytes into inflamed tissue.
The protein CD151 (PETA-3/SFA-1), a member of the tetraspanins family (also known as the transmembrane 4 superfamily), is expressed in various cell types including epidermal basal cells, epithelial cells, skeletal, smooth and cardiac muscle, endothelial cells, T cells, neutrophils, platelets, and Schwann cells and has a strong molecular association with the βΐ family of integrins. CD151 possesses four highly conserved transmembrane domains, cytoplasmic amino and carboxyl termini, and two extracellular loops, the larger of which contains the distinctive pattern of cysteine residues that help to define the family. It was previously shown in human melanoma cells that homophilic interactions between CD151 proteins on the surface of neighboring cells increase c-Jun activity through the activation of FAK, Src, p38 MAPK, and INK, leading to enhanced cell motility and MMP-9 expression [Hong et al, J Biol Chem (2006) 281(34): 24279- 92]. Although the physiological function of CD151 is largely unknown, in vitro functional studies showed that CD151 is involved in cell adhesion, motility, and polarity [Yauch et al, Mol Biol Cell (1998) 9(10): 2751-65]. It was shown that antibodies to either CD151 or α3β1 lead to a 88-92 % reduction in neutrophil motility in response to f-Met-Leu-Phe on fibronectin, suggesting a functionally important role of these complexes in cell migration [Yauch et al, Mol Biol Cell (1998) 9(10): 2751-65]. Since cellular processes regulated by CD151 (such as cell adhesion, migration, and spreading) are integrin-mediated adhesive events, it has been proposed that CD151 modulates integrin activity and function, such as those of βΐ integrins [Nishiuchi et al., Proc Natl Acad Sci U S A (2005) 102(6): 1939-44].
Hasegawa et al. have reported that the expression level of CD151 on adult T cell leukemia (ATL) cells from the lymph nodes of lymphoma-type ATL patients were significantly higher than those on circulating ATL cells [Hasegawa et al., J of immunology (1998) 161 : 3087-3095]. Thus, they suggest that increased expression of CD 151 may act to retain ATL cells in the lymph nodes.
As was previously reported [Flaishon et al, Blood (2008) 112: 5016-5025; PCT Publication No. WO 2008/012796], picomolar [pM] levels of circulating CCL2 (also called Macrophage Chemotactic Protein- 1 or MCP-1) can exert global suppressive effects on T-cell trafficking and differentiation within peripheral lymph nodes. The impaired homing of T lymphocytes to the peripheral lymph nodes by CCL2 results in attenuated progression of both asthma and adjuvant arthritis. Thus, low levels of circulating CCL2 can exert global suppressive effects on T-cell trafficking and differentiation within peripheral lymph nodes, and may be clinically beneficial as an anti-inflammatory agent. PCT Publication No. WO 2007/146968 discloses multivalent binding peptides, including bi-specific binding peptides, having an immunoglobulin effector function. The cell target of the peptide may be an angiogenesis target (e.g. CD 151) and the cell may be a T cell. Methods for using such peptides to treat, prevent or ameliorate symptoms of a variety of diseases, disorders or conditions (e.g. inflammatory and autoimmune conditions) are also provided. For example, WO 2007/146968 contemplates targeted recruitment of effector cells (e.g. cytotoxic T lymphocytes) or to localizing therapeutic compounds (e.g. radiolabeled proteins) to cells, tissues, agents and foreign objects to be destroyed or sequestered (e.g. cancer cells).
Additional background art includes U.S. 20090324600 and U.S. 201000150902 teaching anti CD151 antibodies and uses of same for the treatment of cancer.
WO2010/137020 teaches the use of CD151 inhibitors for the treatment of cancer and inflammation. SUMMARY OF THE INVENTION
According to an aspect of some embodiments of the present invention there is provided an isolated peptide comprising at least 20 consecutive amino acids of the amino acid sequence set forth in SEQ ID NO: 2, the peptide being shorter than 100 amino acids and having a CD151 inhibitory activity.
According to some embodiments of the invention, the CD 151 inhibitory activity comprises inhibition of lymphocyte migration or homing.
According to some embodiments of the invention, the migration or homing is to a lymphoid organ.
According to some embodiments of the invention, the lymphocyte comprises a T cell.
According to some embodiments of the invention, the CD 151 inhibitory activity comprises inhibition of Vav phosphorylation.
According to some embodiments of the invention, the isolated peptide is a soluble peptide.
According to an aspect of some embodiments of the present invention there is provided a molecule comprising the isolated peptide attached to a heterologous moiety. According to some embodiments of the invention, the heterologous moiety is a proteinaceous moiety.
According to some embodiments of the invention, the heterologous moiety comprises an immunoglobulin domain.
According to some embodiments of the invention, the heterologous moiety is a non-proteinaceous moiety.
According to some embodiments of the invention, the non-proteinaceous moiety comprises a synthetic polymer.
According to some embodiments of the invention, the peptide is as set forth in SEQ ID NO: 2.
According to an aspect of some embodiments of the present invention there is provided an isolated polynucleotide encoding the peptide.
According to an aspect of some embodiments of the present invention there is provided a nucleic acid expression construct comprising the isolated polynucleotide under the transcriptional control of a cis-acting regulatory element.
According to an aspect of some embodiments of the present invention there is provided a pharmaceutical composition comprising as an active ingredient, the isolated peptide or molecule, and a pharmaceutically acceptable carrier.
According to an aspect of some embodiments of the present invention there is provided a method of treating inflammation in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the isolated peptide or molecule, thereby treating the inflammation in the subject.
According to some embodiments of the invention, the isolated peptide or molecule is used in the treatment of inflammation.
According to some embodiments of the invention, the inflammation is associated with a medical condition selected from the group consisting of a cancer, an autoimmune disease, a hypersensitivity, a diabetes, an infectious disease, a transplantation associated disease and an allergy.
According to some embodiments of the invention, the subject is a human being. According to some embodiments of the invention, the inflammation is associated with an inflammatory bowel disease. According to some embodiments of the invention, said inflammatory bowel disease comprises colitis.
Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.
BRIEF DESCRIPTION OF THE DRAWINGS
Some embodiments of the invention are herein described, by way of example only, with reference to the accompanying drawings. [IF IMAGES, REPHRASE] With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of embodiments of the invention. In this regard, the description taken with the drawings makes apparent to those skilled in the art how embodiments of the invention may be practiced.
In the drawings:
FIGs. 1A-B are bar graphs showing that CD151ECD peptide inhibits the actin polymerization and migration of T cells. Figure 1A - T cells were incubated with CD151ECD (SEQ ID NO: 2) or a control peptide (SEQ ID NO: 2, 20 or 40 μg/ml) for lh. The cells were then stimulated with CCL21 (0.1 mg/ml). 15 sec later, the cells were immediately fixed, permeabilized, stained with FITC-phalloidin. Actin polymerization was analyzed by flow cytometry. The results shown represent the average of three independent experiments. Figure IB - T cells were suspended with CD151ECD (SEQ ID NO: 2) or a control peptide ((SEQ ID NO: 2) 40 μg/ml) for 1 hr. The cells were then placed in the upper chamber of a transwell. The migration towards the chemokine CCL21 (0.4 mg/ml), residing in the lower part of the apparatus, was analyzed after 3 hours by FACS. The graph shows the average of three independent experiments.
FIG. 2 is a micrograph showing that CD151ECD peptide downregulates Vavl phosphorylation. T cells were incubated with CD151ECD (SEQ ID NO: 2) or a control peptide ((SEQ ID NO: 2) 40 μg/ml) for 1 min. Immediately after treatment, cells were washed and fast frozen in liquid nitrogen. Cells were then lysed and an aliquot reserved for total Vavl analysis. Phosphorylated proteins (pVav) from the remaining lysate were immunoprecipitated (IP) with an anti-Tyr(P) antibody. Immunoprecipitates and total lysate proteins were separated on 10% (w/v) SDS-PAGE and blotted with an anti-Vavl antibody. The results shown represent three separate experiments.
FIGs. 3A-F show that the CD151-ECD peptide inhibits the inflammatory response in DSS Colitis. Specifically shown are comparison of intraperitoneal (i.p.) injections of control scrambled peptide (SEQ ID NO: 5) or CD151-ECD treatment (80 μg, SEQ ID NO: 2), depending on the efficacy of the peptide in the in vitro assays). n=10-15 animals in each group. Shown are percent of weight loss over 10 days (Figure 3A), length of colon after sacrifice of the mice on day 11 (Figure 3B), representative pictures of colon length in each group (Figure 3C), endoscopic colitis score (Figure 3D), representative photographs of distal colons of healthy mouse, control peptide-treated mouse and CD151-ECD-treated mouse (Figure 3E), and inflammation score (Figure 3F).
DESCRIPTION OF SPECIFIC EMBODIMENTS OF THE INVENTION
The present invention, in some embodiments thereof, relates to compositions and methods for treating inflammation.
Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not necessarily limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways.
The progress in understanding the mechanisms of T cell activation, T-cell trafficking, migration and homing, as well as progress towards development of mechanisms for the inactivation and modulation of T-cells has been translated into strategies for treatment of autoimmune diseases. The targets for selective immuno- intervention include modulation of antigen recognition, costimulation blockade, induction of regulatory cells, deviation to non-pathogenic or protective responses, neutralization of proinflammatory cytokines, induction or administration of anti-inflammatory cytokines, and modulation of leukocyte trafficking. The surveillance of the body for foreign antigens is a critical function of the immune system. Lymphocytes migrate from the blood into tissues and secondary lymphoid organs, and return to the blood via lymph vessels and the thoractic duct. The majority of lymphocytes are capable of tissue selective trafficking (homing), recognizing organ-specific adhesion molecules on specialized endothelial cells. Previous studies focused on the specific recruitment of leukocytes to the lymph nodes (LN) or to sites of inflammation.
CD151 has been previously suggested as a target for exerting global suppressory effects on T cell trafficking and differentiation within the peripheral lymph. WO2010/137020 teaches CD151 derived peptides for inhibition of CD151 homophilic interactions.
Whilst reducing the present invention to practice, the present inventors have uncovered a novel region within CD 151 large extra cellular loop that contributes to T cell migration. This region corresponds to amino acids 118-180 of human CD151. The present inventors have synthesized a peptide corresponding to this region and found unprecedented efficacy in the inhibition of T cell migration and CD151 dependent Vav phosphorylation. . The CD151 fragment inhibited both T cell migration in vitro and in vivo and attenuated the development of DSS-induced colitis (Figures 3A-F). Thus, T cell CD151 is a key orchestrator of T cell motility, and its targeting by inhibitory peptides results in beneficial suppression of inflammation. These results suggest that such a peptide can be used as a potent anti inflammatory agent.
Thus according to an aspect of the invention there is provided an isolated CD 151 peptide (having a CD151 amino acid sequence, also termed herein "peptide") comprising at least 20 consecutive amino acids of the amino acid sequence set forth in SEQ ID NO: 2, the peptide being shorter than 100 amino acids and having a CD151 inhibitory activity.
As used herein the term "CD151" refers to a CD151 gene product (i.e., protein or mRNA) such as set forth in GenBank Accession Nos. NM_004357.4, NM_139030.3, NMJ39029.1, NM 001039490.1, NP_004348.2, NP_620599.1, NP_620598.1 and NP 001034579.1. According to a specific embodiment, the CD151 is human CD151 such as set forth in SEQ ID NO: 1. As used herein the term "isolated" refers to isolated or purified from the natural environment, e.g., biological samples e.g., serum, cells or tissues.
As used herein "CD151 activity" refers to a cell signaling activity (e.g., vav phosphorylation, β-catenin signaling), self-ligation activity (i.e., cdl51 homophilic interaction), actin cytoskeleton polymerization, cell adhesion, cell spreading, cell migration (e.g. towards a chemokine such as CCL21 or CXCL12) or homing (e.g. to a lymphoid organ).
According to a specific embodiment, the CD151 inhibitory activity comprises inhibition of lymphocyte migration or homing.
According to a specific embodiment the migration or homing is to a lymphoid organ.
As used herein the term "lymphocyte" refers to a white blood cell of the immune system. A lymphocyte may include a T cell, a B cell and natural killer (NK) cell. According to a specific embodiment of the present invention, the lymphocyte is a T cell including CD4+ T cells, CD8+ T cells and any subsets therefrom (including naive, mature and memory T cells).
According to a specific embodiment, the CD151 inhibitory activity comprises inhibition of Vav phosphorylation.
The present invention provides ample teachings for assessing CD151 signaling, migration and homing, see Example 2 of the Examples section which follows.
The CD151 peptide can be at least 15 amino acids but shorter than 1 10 amino acids. Thus according to specific embodiments the Cdl51 amino acid sequence is 15- 110, 20-110, 20-100, 30-100, 50-100, 20-70, 30-70, 40-70, 50-70, 60-70 or 62-70, 63-70 amino acids long. It will be appreciated that any groups of ranges or examples provided herein is meant to read over the whole group or specific individual ranges/examples comprised therein.
According to a specific embodiment, the peptide is as set forth in SEQ ID NO: 2.
According to an important embodiment of the invention the peptide is soluble. According to a specific embodiment, soluble refers to the absence of a cell-membrane anchoring moiety.
According to an aspect of the invention there is provided a molecule which comprises the CD151 peptide attached to a heterologous moiety forming. The heterologous moiety may be a proteinaceous moiety i.e., a heterologous amino acid sequence.
As used herein the phrase "heterologous amino acid sequence" refers to an amino acid sequence which does not form a consecutive part of the CD151 polypeptide in nature. According to a specific embodiment the heterologous amino acid sequence is non-immunogenic. This sequence can confer solubility to the peptide of the present invention, thereby increasing the half-life of such a fusion polypeptide molecule in the serum.
The heterologous amino acid sequence is generally localized at the amino- or carboxyl- terminus of the CD 151 of the present invention.
As mentioned, at least one heterologous amino acid sequence can be contiguously (N-terminally or C-terminally) attached to the CD151 peptide of the present invention. The heterologous amino acid sequence may be attached to the CD151 amino acid sequence by any of peptide or non-peptide bond. Attachment of the CD 151 amino acid sequence to the heterologous amino acid sequence may be effected by direct covalent bonding (peptide bond or a substituted peptide bond) or indirect binding such as by the use of a linker having functional groups. Functional groups include, without limitation, a free carboxylic acid (C(=0)OH), a free amino group (NH2), an ester group (C(=0)OR, where R is alkyl, cycloalkyl or aryl), an acyl halide group (C(=0)A, where A is fluoride, chloride, bromide or iodide), a halide (fluoride, chloride, bromide or iodide), a hydroxyl group (OH), a thiol group (SH), a nitrile group (C≡N), a free C- carbamic group (NR"-C(=0)-OR', where each of R' and R" is independently hydrogen, alkyl, cycloalkyl or aryl).
An example of a heterologous amino acid sequence which may be used in accordance with this aspect of the present invention is an immunoglobulin amino acid sequence, such as the hinge and Fc regions of an immunoglobulin heavy domain (see U.S. Pat. No. 6,777,196). The immunoglobulin moiety in the chimeras of this aspect of the present invention may be obtained from IgGl, IgG2, IgG3 or IgG4 subtypes, IgA, IgE, IgD or IgM, as further discussed hereinbelow.
Chimeras constructed from a receptor sequence linked to an appropriate immunoglobulin constant domain sequence (immunoadhesins) are known in the art. Immunoadhesins reported in the literature include fusions of the T cell receptor; CD4; L- selectin (homing receptor); CD44; CD28 and B7; CTLA-4; CD22; TNF receptor; NP receptors; and IgE receptor a.
Typically, in such fusions the chimeric molecule will retain at least functionally active hinge and CH2 and CH3 domains of the constant region of an immunoglobulin heavy chain. Fusions can also be generated to the C-terminus of the Fc portion of a constant domain, or immediately N-terminal to the CHI of the heavy chain or the corresponding region of the light chain.
The exact site at which fusion (conjugation) between the heterologous sequence and the CD151 amino acid sequence is not critical. Particular sites are well known in the art and may be selected in order to optimize the biological activity, secretion or binding characteristics of such chimeric molecules of the present invention.
Though it may be possible to conjugate the entire heavy chain constant region to the CD151 amino acid sequence of the present invention, it is preferable to fuse shorter sequences. For example, a sequence beginning in the hinge region just upstream of the papain cleavage site, which defines IgG Fc chemically; residue 216, taking the first residue of heavy chain constant region to be 114, or analogous sites of other immunoglobulins, is used in the fusion. In a particularly preferred embodiment, the CD151 amino acid sequence is fused to the hinge region and CH2 and CH3, or to the CHI, hinge, CH2 and CH3 domains of an IgGl, IgG2, or IgG3 heavy chain (see U.S. Pat. No. 6,777,196). The precise site at which the fusion is made is not critical, and the optimal site can be determined by routine experimentation.
As mentioned, the immunoglobulin sequences used in the construction of the chimeric molecules of this aspect of the present invention may be from an IgG immunoglobulin heavy chain constant domain. The use of human IgGl immunoglobulin sequences is exemplary. A major advantage of using IgGl is that IgGl can be purified efficiently on immobilized protein A. However, other structural and functional properties of immunoglobulins should be considered when choosing the Ig fusion partner for a particular chimera construction. For example, the IgG3 hinge is longer and more flexible, so it can accommodate larger CD151 amino acid sequences that may not fold or function properly when fused to IgGl . Another consideration may be valency; IgG are bivalent homodimers, whereas Ig subtypes like IgA and IgM may give rise to dimeric or pentameric structures, respectively, of the basic Ig homodimer unit. Other considerations in selecting the immunoglobulin portion of the chimeric molecules of this aspect of the present invention are described in U.S. Pat. No. 6,777,196.
Further examples of heterologous amino acid sequences (i.e., moieties) commonly used in fusion protein construction include, but are not limited to galactosidase, glucuronidase, glutathione-S-transferase (GST), carboxy terminal peptide (CTP) from chorionic gonadotrophin (CG ) and chloramphenicol acetyltransferase (CAT).
Thus, molecules (i.e., peptide fusions) of this aspect of the present invention may comprise heterologous amino acid sequences, as described above.
Additionally or alternatively as mentioned hereinabove the peptide (i.e., CD151 amino acid sequence) may be attached to a non-proteinaceous moiety, such molecules are preferably selected non-immunogenic in a subject (e.g., human subject).
Such a molecule is highly stable (resistant to in-vivo proteolytic activity probably due to steric hindrance conferred by the non-proteinaceous moiety) and may be produced using common solid phase synthesis methods which are inexpensive and highly efficient, as further described hereinbelow. However, it will be appreciated that recombinant techniques may still be used, whereby the recombinant peptide product is subjected to in-vitro modification (e.g., PEGylation as further described hereinbelow).
The phrase "non-proteinaceous moiety" as used herein refers to a molecule not including peptide bonded amino acids that is attached to the above-described CD151 amino acid sequence.
It will be appreciated that such non-proteinaceous moieties may be also attached to the above mentioned fusion molecules (i.e., which comprise a heterologous amino acid sequence) to promote stability and possibly solubility of the molecules.
Bioconjugation of such a non-proteinaceous moiety (such as PEGylation) can confer the CD151 amino acid sequence with stability (e.g., against protease activities) and/or solubility (e.g., within a biological fluid such as blood, digestive fluid) while preserving its biological activity and prolonging its half-life. Methods of PEGylation, for example, are well known in the art.
The term "peptide" as used herein encompasses native peptides (either degradation products, synthetically synthesized peptides or recombinant peptides) and peptidomimetics (typically, synthetically synthesized peptides), as well as peptoids and semipeptoids which are peptide analogs, which may have, for example, modifications rendering the peptides more stable while in a body or more capable of penetrating into cells. Such modifications include, but are not limited to N terminus modification, C terminus modification, peptide bond modification, including, but not limited to, CH2- NH, CH2-S, CH2-S=0, 0=C-NH, CH2-0, CH2-CH2, S=C-NH, CH=CH or CF=CH, backbone modifications, and residue modification. Methods for preparing peptidomimetic compounds are well known in the art and are specified, for example, in Quantitative Drug Design, C.A. Ramsden Gd., Chapter 17.2, F. Choplin Pergamon Press (1992), which is incorporated by reference as if fully set forth herein. Further details in this respect are provided hereinunder.
Peptide bonds (-CO-NH-) within the peptide may be substituted, for example, by N-methylated bonds (-N(CFB)-CO-), ester bonds (-C(R)H-C-0-0-C(R)-N-), ketomethylen bonds (-CO-CH2-), a-aza bonds (-NH-N(R)-CO-), wherein R is any alkyl, e.g., methyl, carba bonds (-CH2-NH-), hydroxyethylene bonds (-CH(OH)-CH2-), thioamide bonds (-CS-NH-), olefinic double bonds (-CH=CH-), retro amide bonds (- NH-CO-), peptide derivatives (-N(R)-CH2-CO-), wherein R is the "normal" side chain, naturally presented on the carbon atom.
These modifications can occur at any of the bonds along the peptide chain and even at several (2-3) at the same time.
Natural aromatic amino acids, Trp, Tyr and Phe, may be substituted for synthetic non-natural acid such as TIC, naphthylelanine (Nol), ring-methylated derivatives of Phe, halogenated derivatives of Phe or o-methyl-Tyr.
In addition to the above, the peptides of the present invention may also include one or more modified amino acids or one or more non-amino acid monomers (e.g. fatty acids, complex carbohydrates etc).
The term "amino acid" or "amino acids" is understood to include the 20 naturally occurring amino acids; those amino acids often modified post-translationally in vivo, including, for example, hydroxyproline, phosphoserine and phosphothreonine; and other unusual amino acids including, but not limited to, 2-aminoadipic acid, hydroxylysine, isodesmosine, nor-valine, nor-leucine and ornithine. Furthermore, the term "amino acid" includes both D- and L-amino acids. The peptides of the present invention are preferably utilized in a linear form, although it will be appreciated that in cases where cyclicization does not severely interfere with peptide characteristics, cyclic forms of the peptide can also be utilized.
The peptides of the present invention may be synthesized by any techniques that are known to those skilled in the art of peptide synthesis. For solid phase peptide synthesis, a summary of the many techniques may be found in J. M. Stewart and J. D. Young, Solid Phase Peptide Synthesis, W. H. Freeman Co. (San Francisco), 1963 and J. Meienhofer, Hormonal Proteins and Peptides, vol. 2, p. 46, Academic Press (New York), 1973. For classical solution synthesis see G. Schroder and K. Lupke, The Peptides, vol. 1, Academic Press (New York), 1965.
In general, these methods comprise the sequential addition of one or more amino acids or suitably protected amino acids to a growing peptide chain. Normally, either the amino or carboxyl group of the first amino acid is protected by a suitable protecting group. The protected or derivatized amino acid can then either be attached to an inert solid support or utilized in solution by adding the next amino acid in the sequence having the complimentary (amino or carboxyl) group suitably protected, under conditions suitable for forming the amide linkage. The protecting group is then removed from this newly added amino acid residue and the next amino acid (suitably protected) is then added, and so forth. After all the desired amino acids have been linked in the proper sequence, any remaining protecting groups (and any solid support) are removed sequentially or concurrently, to afford the final peptide compound. By simple modification of this general procedure, it is possible to add more than one amino acid at a time to a growing chain, for example, by coupling (under conditions which do not racemize chiral centers) a protected tripeptide with a properly protected dipeptide to form, after deprotection, a pentapeptide and so forth. Further description of peptide synthesis is disclosed in U.S. Pat. No. 6,472,505.
According to a specific embodiment a method of preparing the peptide compounds of the present invention involves solid phase peptide synthesis.
According to another specific embodiment, a method of producing the peptide of the invention involves recombinant techniques such as described by Bitter et al. (1987) Methods in Enzymol. 153:516-544; Studier et al. (1990) Methods in Enzymol. 185:60-89; Brisson et al. (1984) Nature 310:511-514; Takamatsu et al. (1987) EMBO J. 6:307-311; Coruzzi et al. (1984) EMBO J. 3: 1671-1680; Brogli et al. (1984) Science 224:838-843; Gurley et al. (1986) Mol. Cell. Biol. 6:559-565 and Weissbach & Weissbach, 1988, Methods for Plant Molecular Biology, Academic Press, NY, Section VIII, pp 421-463.
Briefly, an expression construct (i.e., expression vector), which includes an isolated polynucleotide encoding the CD151 peptide of the invention (e.g., SEQ ID NO: 3 encoding SEQ ID NO: 2) optionally in frame fused to a nucleic acid sequence encoding the heterologous amino acid (described above) is positioned under the transcriptional control of a regulatory element, such as a promoter and is introduced into host cells.
Methods of introducing the expression construct into a host cell are well known in the art and include, electroporation, lipofection and chemical transformation (e.g., calcium phosphate).
The "transformed" cells are cultured under suitable conditions, which allow the expression of the chimeric molecule encoded by the nucleic acid sequence.
Following a predetermined time period, the expressed peptide or chimeric peptide (including the heterologous amino acid sequence described above) is recovered from the cell or cell culture, and purification is effected according to the end use of the recombinant polypeptide.
Depending on the host/vector system utilized, any of a number of suitable transcription and translation elements including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, and the like, can be used in the expression vector [see, e.g., Bitter et al, (1987) Methods in Enzymol. 153:516-544].
Other than containing the necessary elements for the transcription and translation of the inserted coding sequence (encoding the CD151 peptide), the expression construct of the present invention can also include sequences engineered to optimize stability, production, purification, yield or toxicity of the expressed fusion protein.
A variety of prokaryotic or eukaryotic cells can be used as host-expression systems to express the fusion protein coding sequence. These include, but are not limited to, microorganisms, such as bacteria transformed with a recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vector containing the peptide coding sequence; yeast transformed with recombinant yeast expression vectors containing the peptide coding sequence; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors, such as Ti plasmid, containing the chimera coding sequence. Mammalian expression systems are preferably used to express the peptide of the present invention.
The choice of host cell line for the expression of the molecules depends mainly on the expression vector. Eukaryotic expression systems are preferred (e.g., mammalian and insects) since they allow post translational modifications (e.g., glyccosylation). Another consideration is the amount of protein that is required. Milligram quantities often can be produced by transient transfections. For example, the adenovirus EIA- transformed 293 human embryonic kidney cell line can be transfected transiently with pR 5 -based vectors by a modification of the calcium phosphate method to allow efficient expression. CDM8-based vectors can be used to transfect COS cells by the DEAE-dextran method. If larger amounts of protein are desired, the molecules can be expressed after stable transfection of a host cell line. It will be appreciated that the presence of a hydrophobic leader sequence at the N-terminus of the molecule will ensure processing and secretion of the molecule by the transfected cells.
It will be appreciated that the use of bacterial or yeast host systems may be preferable to reduce cost of production. However since bacterial host systems are devoid of protein glycosylation mechanisms, a post production glycosylation may be needed.
In any case, transformed cells are cultured under effective conditions, which allow for the expression of high amounts of recombinant polypeptide. Effective culture conditions include, but are not limited to, effective media, bioreactor, temperature, pH and oxygen conditions that permit protein production. An effective medium refers to any medium in which a cell is cultured to produce the recombinant peptide molecule of the present invention. Such a medium typically includes an aqueous solution having assimilable carbon, nitrogen and phosphate sources, and appropriate salts, minerals, metals and other nutrients, such as vitamins. Cells of the present invention can be cultured in conventional fermentation bioreactors, shake flasks, test tubes, microtiter dishes, and petri plates. Culturing can be carried out at a temperature, pH and oxygen content appropriate for a recombinant cell. Such culturing conditions are within the expertise of one of ordinary skill in the art.
Depending on the vector and host system used for production, resultant proteins of the present invention may either remain within the recombinant cell, secreted into the fermentation medium, secreted into a space between two cellular membranes, such as the periplasmic space in E. coli; or retained on the outer surface of a cell or viral membrane.
Following a predetermined time in culture, recovery of the recombinant protein is effected. The phrase "recovering the recombinant protein" refers to collecting the whole fermentation medium containing the protein and need not imply additional steps of separation or purification. Peptides of the present invention can be purified using a variety of standard protein purification techniques, such as, but not limited to, affinity chromatography, ion exchange chromatography, filtration, electrophoresis, hydrophobic interaction chromatography, gel filtration chromatography, reverse phase chromatography, concanavalin A chromatography, chromato focusing and differential solubilization.
Molecules of the present invention are preferably retrieved in "substantially pure" form. As used herein, "substantially pure" refers to a purity that allows for the effective use of the protein in the applications, described hereinbelow.
The inhibitory activity of the peptides of the invention as manifested by their ability to inhibit cell migration and Vav signaling supports their use as agents in the treatment of inflammation.
Thus, according to an aspect of the invention there is provided a method of treating inflammation in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the isolated peptide or molecule described above, thereby treating the inflammation in the subject.
As used herein the term "subject in need thereof refers to a mammal, preferably a human subject at any age which may benefit from the treatment modality of the present invention. According to specific embodiments, the subject does not suffer from a medical condition which is not associated with inflammation.
According to a specific embodiment the subject is a human being. As used herein the term "inflammation" refers to any medical condition which comprises an inflammatory response in which migration of cells (e.g. to the lymph nodes) contributes to inflammation onset or progression.
A number of diseases and conditions, which involve an inflammatory response, can be treated using the methodology described hereinabove. Examples of such diseases and conditions are summarized infra.
Inflammatory diseases - Include, but are not limited to, chronic inflammatory diseases and acute inflammatory diseases.
Inflammatory diseases associated with hypersensitivity
Examples of hypersensitivity include, but are not limited to, Type I hypersensitivity, Type II hypersensitivity, Type III hypersensitivity, Type IV hypersensitivity, immediate hypersensitivity, antibody mediated hypersensitivity, immune complex mediated hypersensitivity, T lymphocyte mediated hypersensitivity and DTH.
Type I or immediate hypersensitivity, such as asthma.
Type II hypersensitivity include, but are not limited to, rheumatoid diseases, rheumatoid autoimmune diseases, rheumatoid arthritis (Krenn V. et ah, Histol Histopathol 2000 Jul;15 (3):791), spondylitis, ankylosing spondylitis (Jan Voswinkel et al, Arthritis Res 2001; 3 (3): 189), systemic diseases, systemic autoimmune diseases, systemic lupus erythematosus (Erikson J. et al, Immunol Res 1998;17 (l-2):49), sclerosis, systemic sclerosis (Renaudineau Y. et al, Clin Diagn Lab Immunol. 1999 Mar;6 (2):156); Chan OT. et al, Immunol Rev 1999 Jun;169: 107), glandular diseases, glandular autoimmune diseases, pancreatic autoimmune diseases, diabetes, Type I diabetes (Zimmet P. Diabetes Res Clin Pract 1996 Oct;34 Suppl:S125), thyroid diseases, autoimmune thyroid diseases, Graves' disease (Orgiazzi J. Endocrinol Metab Clin North Am 2000 Jun;29 (2):339), thyroiditis, spontaneous autoimmune thyroiditis (Braley-Mullen H. and Yu S, J Immunol 2000 Dec 15;165 (12):7262), Hashimoto's thyroiditis (Toyoda N. et al, Nippon Rinsho 1999 Aug;57 (8): 1810), myxedema, idiopathic myxedema (Mitsuma T. Nippon Rinsho. 1999 Aug;57 (8): 1759); autoimmune reproductive diseases, ovarian diseases, ovarian autoimmunity (Garza KM. et ah, J Reprod Immunol 1998 Feb;37 (2):87), autoimmune anti-sperm infertility (Diekman AB. et ah, Am J Reprod Immunol. 2000 Mar;43 (3): 134), repeated fetal loss (Tincani A. et al, Lupus 1998;7 Suppl 2:S107-9), neurodegenerative diseases, neurological diseases, neurological autoimmune diseases, multiple sclerosis (Cross AH. et al, J Neuroimmunol 2001 Jan 1;112 (1-2): 1), Alzheimer's disease (Oron L. et al, J Neural Transm Suppl. 1997;49:77), myasthenia gravis (Infante AJ. And Kraig E, Int Rev Immunol 1999; 18 (1-2): 83), motor neuropathies (Kornberg AJ. J Clin Neurosci. 2000 May;7 (3): 191), Guillain-Barre syndrome, neuropathies and autoimmune neuropathies (Kusunoki S. Am J Med Sci. 2000 Apr;319 (4):234), myasthenic diseases, Lambert-Eaton myasthenic syndrome (Takamori M. Am J Med Sci. 2000 Apr;319 (4):204), paraneoplastic neurological diseases, cerebellar atrophy, paraneoplastic cerebellar atrophy, non-paraneoplastic stiff man syndrome, cerebellar atrophies, progressive cerebellar atrophies, encephalitis, Rasmussen's encephalitis, amyotrophic lateral sclerosis, Sydeham chorea, Gilles de la Tourette syndrome, polyendocrinopathies, autoimmune polyendocrinopathies (Antoine JC. and Honnorat J. Rev Neurol (Paris) 2000 Jan;156 (1):23); neuropathies, dysimmune neuropathies (Nobile-Orazio E. et al, Electroencephalogr Clin Neurophysiol Suppl 1999;50:419); neuromyotonia, acquired neuromyotonia, arthrogryposis multiplex congenita (Vincent A. et al, Ann N Y Acad Sci. 1998 May 13;841 :482), cardiovascular diseases, cardiovascular autoimmune diseases, atherosclerosis (Matsuura E. et al, Lupus. 1998;7 Suppl 2:S135), myocardial infarction (Vaarala O. Lupus. 1998;7 Suppl 2:S132), thrombosis (Tincani A. et al, Lupus 1998;7 Suppl 2:S107-9), granulomatosis, Wegener's granulomatosis, arteritis, Takayasu's arteritis and Kawasaki syndrome (Praprotnik S. et al, Wien Klin Wochenschr 2000 Aug 25;112 (15-16):660); anti-factor VIII autoimmune disease (Lacroix-Desmazes S. et al, Semin Thromb Hemost.2000;26 (2): 157); vasculitises, necrotizing small vessel vasculitises, microscopic polyangiitis, Churg and Strauss syndrome, glomerulonephritis, pauci-immune focal necrotizing glomerulonephritis, crescentic glomerulonephritis (Noel LH. Ann Med Interne (Paris). 2000 May;151 (3): 178); antiphospholipid syndrome (Flamholz R. et al, J Clin Apheresis 1999; 14 (4): 171); heart failure, agonist- like beta-adrenoceptor antibodies in heart failure (Wallukat G. et al, Am J Cardiol. 1999 Jun 17;83 (12A):75H), thrombocytopenic purpura (Moccia F. Ann Ital Med Int. 1999 Apr-Jun;14 (2): 114); hemolytic anemia, autoimmune hemolytic anemia (Efremov DG. et al, Leuk Lymphoma 1998 Jan;28 (3-4):285), gastrointestinal diseases, autoimmune diseases of the gastrointestinal tract, intestinal diseases, chronic inflammatory intestinal disease (Garcia Herola A. et al, Gastroenterol Hepatol. 2000 Jan;23 (1): 16), celiac disease (Landau YE. and Shoenfeld Y. Harefuah 2000 Jan 16; 138 (2): 122), autoimmune diseases of the musculature, myositis, autoimmune myositis, Sjogren's syndrome (Feist E. et al, Int Arch Allergy Immunol 2000 Sep; 123 (1):92); smooth muscle autoimmune disease (Zauli D. et al, Biomed Pharmacother 1999 Jun;53 (5-6):234), hepatic diseases, hepatic autoimmune diseases, autoimmune hepatitis (Manns MP. J Hepatol 2000 Aug; 33 (2):326) and primary biliary cirrhosis (Strassburg CP. et al, Eur J Gastroenterol Hepatol. 1999 Jun; 11 (6):595).
Type IV or T cell mediated hypersensitivity, include, but are not limited to, rheumatoid diseases, rheumatoid arthritis (Tisch R, McDevitt HO. Proc Natl Acad Sci U S A 1994 Jan 18; 91 (2):437), systemic diseases, systemic autoimmune diseases, systemic lupus erythematosus (Datta SK., Lupus 1998; 7 (9):591), glandular diseases, glandular autoimmune diseases, pancreatic diseases, pancreatic autoimmune diseases, Type 1 diabetes (Castano L. and Eisenbarth GS. Ann. Rev. Immunol. 8:647); thyroid diseases, autoimmune thyroid diseases, Graves' disease (Sakata S. et al, Mol Cell Endocrinol 1993 Mar;92 (1):77); ovarian diseases (Garza KM. et al, J Reprod Immunol 1998 Feb;37 (2):87), prostatitis, autoimmune prostatitis (Alexander RB. et al, Urology 1997 Dec;50 (6):893), polyglandular syndrome, autoimmune polyglandular syndrome, Type I autoimmune polyglandular syndrome (Hara T. et al, Blood. 1991 Mar 1;77 (5): 1127), neurological diseases, autoimmune neurological diseases, multiple sclerosis, neuritis, optic neuritis (Soderstrom M. et al, J Neurol Neurosurg Psychiatry 1994 May;57 (5):544), myasthenia gravis (Oshima M. et al, Eur J Immunol 1990 Dec;20 (12):2563), stiff-man syndrome (Hiemstra HS. et al, Proc Natl Acad Sci U S A 2001 Mar 27;98 (7):3988), cardiovascular diseases, cardiac autoimmunity in Chagas' disease (Cunha-Neto E. et al, J Clin Invest 1996 Oct 15;98 (8): 1709), autoimmune thrombocytopenic purpura (Semple JW. et al, Blood 1996 May 15;87 (10):4245), anti- helper T lymphocyte autoimmunity (Caporossi AP. et al, Viral Immunol 1998; 11 (1):9), hemolytic anemia (Sallah S. et al, Ann Hematol 1997 Mar;74 (3):139), hepatic diseases, hepatic autoimmune diseases, hepatitis, chronic active hepatitis (Franco A. et al, Clin Immunol Immunopathol 1990 Mar;54 (3):382), biliary cirrhosis, primary biliary cirrhosis (Jones DE. Clin Sci (Colch) 1996 Nov; 91 (5):551), nephric diseases, nephric autoimmune diseases, nephritis, interstitial nephritis (Kelly CJ. J Am Soc Nephrol 1990 Aug; 1 (2): 140), connective tissue diseases, ear diseases, autoimmune connective tissue diseases, autoimmune ear disease (Yoo TJ. et al, Cell Immunol 1994 Aug; 157 (1):249), disease of the inner ear (Gloddek B. et al, Ann N Y Acad Sci 1997 Dec 29; 830:266), skin diseases, cutaneous diseases, dermal diseases, bullous skin diseases, pemphigus vulgaris, bullous pemphigoid and pemphigus foliaceus.
Examples of delayed type hypersensitivity include, but are not limited to, contact dermatitis and drug eruption.
Examples of types of T lymphocyte mediating hypersensitivity include, but are not limited to, helper T lymphocytes and cytotoxic T lymphocytes.
Examples of helper T lymphocyte-mediated hypersensitivity include, but are not limited to, T l lymphocyte mediated hypersensitivity and T 2 lymphocyte mediated hypersensitivity.
Autoimmune diseases
Include, but are not limited to, cardiovascular diseases, rheumatoid diseases, glandular diseases, gastrointestinal diseases, cutaneous diseases, hepatic diseases, neurological diseases, muscular diseases, nephric diseases, diseases related to reproduction, connective tissue diseases and systemic diseases.
Examples of autoimmune cardiovascular diseases include, but are not limited to atherosclerosis (Matsuura E. et al, Lupus. 1998;7 Suppl 2:S135), myocardial infarction (Vaarala O. Lupus. 1998;7 Suppl 2:S132), thrombosis (Tincani A. et al, Lupus 1998;7 Suppl 2:S 107-9), Wegener's granulomatosis, Takayasu's arteritis, Kawasaki syndrome (Praprotnik S. et al, Wien Klin Wochenschr 2000 Aug 25;112 (15-16):660), anti-factor VIII autoimmune disease (Lacroix-Desmazes S. et al, Semin Thromb Hemost.2000;26 (2): 157), necrotizing small vessel vasculitis, microscopic polyangiitis, Churg and Strauss syndrome, pauci-immune focal necrotizing and crescentic glomerulonephritis (Noel LH. Ann Med Interne (Paris). 2000 May;151 (3): 178), antiphospholipid syndrome (Flamholz R. et al, J Clin Apheresis 1999;14 (4): 171), antibody-induced heart failure (Wallukat G. et al, Am J Cardiol. 1999 Jun 17;83 (12A):75H), thrombocytopenic purpura (Moccia F. Ann Ital Med Int. 1999 Apr-Jun;14 (2): 114; Semple JW. et al, Blood 1996 May 15;87 (10):4245), autoimmune hemolytic anemia (Efremov DG. et al, Leuk Lymphoma 1998 Jan;28 (3-4):285; Sallah S. et al, Ann Hematol 1997 Mar;74 (3): 139), cardiac autoimmunity in Chagas' disease (Cunha-Neto E. et al, J Clin Invest 1996 Oct 15;98 (8): 1709) and anti-helper T lymphocyte autoimmunity (Caporossi AP. et al, Viral Immunol 1998; 11 (1):9).
Examples of autoimmune rheumatoid diseases include, but are not limited to rheumatoid arthritis (Krenn V. et al, Histol Histopathol 2000 Jul;15 (3):791; Tisch R, McDevitt HO. Proc Natl Acad Sci units S A 1994 Jan 18;91 (2):437) and ankylosing spondylitis (Jan Voswinkel et al, Arthritis Res 2001; 3 (3): 189).
Examples of autoimmune glandular diseases include, but are not limited to, pancreatic disease, Type I diabetes, thyroid disease, Graves' disease, thyroiditis, spontaneous autoimmune thyroiditis, Hashimoto's thyroiditis, idiopathic myxedema, ovarian autoimmunity, autoimmune anti-sperm infertility, autoimmune prostatitis and Type I autoimmune polyglandular syndrome. Diseases include, but are not limited to autoimmune diseases of the pancreas, Type 1 diabetes (Castano L. and Eisenbarth GS. Ann. Rev. Immunol. 8:647; Zimmet P. Diabetes Res Clin Pract 1996 Oct;34 Suppl:S125), autoimmune thyroid diseases, Graves' disease (Orgiazzi J. Endocrinol Metab Clin North Am 2000 Jun;29 (2):339; Sakata S. et al, Mol Cell Endocrinol 1993 Mar;92 (1):77), spontaneous autoimmune thyroiditis (Braley-Mullen H. and Yu S, J Immunol 2000 Dec 15;165 (12):7262), Hashimoto's thyroiditis (Toyoda N. et al, Nippon Rinsho 1999 Aug;57 (8):1810), idiopathic myxedema (Mitsuma T. Nippon Rinsho. 1999 Aug;57 (8): 1759), ovarian autoimmunity (Garza KM. et al, J Reprod Immunol 1998 Feb;37 (2):87), autoimmune anti-sperm infertility (Diekman AB. et ah, Am J Reprod Immunol. 2000 Mar;43 (3): 134), autoimmune prostatitis (Alexander RB. et al, Urology 1997 Dec;50 (6):893) and Type I autoimmune polyglandular syndrome (Hara T. et al, Blood. 1991 Mar 1;77 (5): 1127).
Examples of autoimmune gastrointestinal diseases include, but are not limited to, chronic inflammatory intestinal diseases (Garcia Herola A. et ah, Gastroenterol Hepatol. 2000 Jan; 23 (1): 16), celiac disease (Landau YE. and Shoenfeld Y. Harefuah 2000 Jan 16; 138 (2): 122), colitis, ileitis and Crohn's disease (see Example 3).
Examples of autoimmune cutaneous diseases include, but are not limited to, autoimmune bullous skin diseases, such as, but are not limited to, pemphigus vulgaris, bullous pemphigoid and pemphigus foliaceus. Examples of autoimmune hepatic diseases include, but are not limited to, hepatitis, autoimmune chronic active hepatitis (Franco A. et al, Clin Immunol Immunopathol 1990 Mar; 54 (3):382), primary biliary cirrhosis (Jones DE. Clin Sci (Colch) 1996 Nov; 91 (5):551 ; Strassburg CP. et al, Eur J Gastroenterol Hepatol. 1999 Jun; 1 1 (6):595) and autoimmune hepatitis (Manns MP. J Hepatol 2000 Aug; 33 (2):326).
Examples of autoimmune neurological diseases include, but are not limited to, multiple sclerosis (Cross AH. et al, J Neuroimmunol 2001 Jan 1 ; 1 12 (1-2): 1), Alzheimer's disease (Oron L. et al, J Neural Transm Suppl. 1997;49:77), myasthenia gravis (Infante AJ. And Kraig E, Int Rev Immunol 1999; 18 (l-2):83; Oshima M. et al, Eur J Immunol 1990 Dec;20 (12):2563), neuropathies, motor neuropathies (Kornberg AJ. J Clin Neurosci. 2000 May;7 (3): 191); Guillain-Barre syndrome and autoimmune neuropathies (Kusunoki S. Am J Med Sci. 2000 Apr; 319 (4):234), myasthenia, Lambert-Eaton myasthenic syndrome (Takamori M. Am J Med Sci. 2000 Apr;319 (4):204); paraneoplastic neurological diseases, cerebellar atrophy, paraneoplastic cerebellar atrophy and stiff-man syndrome (Hiemstra HS. et al, Proc Natl Acad Sci units S A 2001 Mar 27;98 (7):3988); non-paraneoplastic stiff man syndrome, progressive cerebellar atrophies, encephalitis, Rasmussen's encephalitis, amyotrophic lateral sclerosis, Sydeham chorea, Gilles de la Tourette syndrome and autoimmune polyendocrinopathies (Antoine JC. and Honnorat J. Rev Neurol (Paris) 2000 Jan; 156 (1):23); dysimmune neuropathies (Nobile-Orazio E. et al, Electroencephalogr Clin Neurophysiol Suppl 1999;50:419); acquired neuromyotonia, arthrogryposis multiplex congenita (Vincent A. et al, Ann N Y Acad Sci. 1998 May 13;841 :482), neuritis, optic neuritis (Soderstrom M. et al, J Neurol Neurosurg Psychiatry 1994 May;57 (5):544) and neurodegenerative diseases.
Examples of autoimmune muscular diseases include, but are not limited to, myositis, autoimmune myositis and primary Sjogren's syndrome (Feist E. et al, Int Arch Allergy Immunol 2000 Sep; 123 (1):92) and smooth muscle autoimmune disease (Zauli D. et al, Biomed Pharmacother 1999 Jun;53 (5-6):234).
Examples of autoimmune nephric diseases include, but are not limited to, nephritis and autoimmune interstitial nephritis (Kelly CJ. J Am Soc Nephrol 1990 Aug; 1 (2): 140). Examples of autoimmune diseases related to reproduction include, but are not limited to, repeated fetal loss (Tincani A. et ah, Lupus 1998; 7 Suppl 2:S107-9).
Examples of autoimmune connective tissue diseases include, but are not limited to, ear diseases, autoimmune ear diseases (Yoo TJ. et ah, Cell Immunol 1994 Aug; 157 (1):249) and autoimmune diseases of the inner ear (Gloddek B. et ah, Ann N Y Acad Sci 1997 Dec 29; 830:266).
Examples of autoimmune systemic diseases include, but are not limited to, systemic lupus erythematosus (Erikson J. et ah, Immunol Res 1998;17 (l-2):49) and systemic sclerosis (Renaudineau Y. et ah, Clin Diagn Lab Immunol. 1999 Mar;6 (2): 156); Chan OT. et al, Immunol Rev 1999 Jun; 169: 107).
Infectious diseases
Examples of infectious diseases include, but are not limited to, chronic infectious diseases, subacute infectious diseases, acute infectious diseases, viral diseases, bacterial diseases, protozoan diseases, parasitic diseases, fungal diseases, mycoplasma diseases and prion diseases.
Graft rejection diseases
Examples of diseases associated with transplantation of a graft include, but are not limited to, graft rejection, chronic graft rejection, subacute graft rejection, hyperacute graft rejection, acute graft rejection and graft versus host disease.
Allergic diseases
Examples of allergic diseases include, but are not limited to, asthma, hives, urticaria, pollen allergy, dust mite allergy, venom allergy, cosmetics allergy, latex allergy, chemical allergy, drug allergy, insect bite allergy, animal dander allergy, stinging plant allergy, poison ivy allergy and food allergy.
Cancerous diseases
Examples of cancer include but are not limited to carcinoma, lymphoma, blastoma, sarcoma, and leukemia. Particular examples of cancerous diseases but are not limited to: Myeloid leukemia such as Chronic myelogenous leukemia. Acute myelogenous leukemia with maturation. Acute promyelocytic leukemia, Acute nonlymphocytic leukemia with increased basophils, Acute monocytic leukemia. Acute myelomonocytic leukemia with eosinophilia; Malignant lymphoma, such as Birkitt's Non-Hodgkin's; Lymphocytic leukemia, such as Acute lumphoblastic leukemia. Chronic lymphocytic leukemia; Myeloproliferative diseases, such as Solid tumors Benign Meningioma, Mixed tumors of salivary gland, Colonic adenomas; Adenocarcinomas, such as Small cell lung cancer, Kidney, Uterus, Prostate, Bladder, Ovary, Colon, Sarcomas, Liposarcoma, myxoid, Synovial sarcoma, Rhabdomyosarcoma (alveolar), Extraskeletel myxoid chonodrosarcoma, Ewing's tumor; other include Testicular and ovarian dysgerminoma, Retinoblastoma, Wilms' tumor, Neuroblastoma, Malignant melanoma, Mesothelioma, breast, skin, prostate, and ovarian.
In a specific embodiment of the present invention, the inflammatory condition is a leukemia or lymphoma.
In another specific embodiment, the inflammatory condition is rheumatoid arthritis, inflammatory bowel disease (e.g. ulcerative colitis and Crohn's disease) and asthma.
It will be appreciated that each of the peptides or molecules described hereinabove can be administered to the subject per se or as part of a pharmaceutical composition which also includes a physiologically acceptable carrier. The purpose of a pharmaceutical composition is to facilitate administration of the active ingredient to an organism.
As used herein a "pharmaceutical composition" refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
Herein the term "active ingredient" refers to the peptide having the CD151 inhibitory activity accountable for the biological effect.
Hereinafter, the phrases "physiologically acceptable carrier" and
"pharmaceutically acceptable carrier" which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound. An adjuvant is included under these phrases.
Herein the term "excipient" refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.
Techniques for formulation and administration of drugs may be found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference.
Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intracardiac, e.g., into the right or left ventricular cavity, into the common coronary artery, intravenous, inrtaperitoneal, intranasal, or intraocular injections.
Alternately, one may administer the pharmaceutical composition in a local rather than systemic manner, for example, via injection of the pharmaceutical composition directly into a tissue region of a patient.
Pharmaceutical compositions of the present invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
For injection, the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
For oral administration, the pharmaceutical composition can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient. Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
Pharmaceutical compositions which can be used orally, include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.
For administration by nasal inhalation, the active ingredients for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
The pharmaceutical composition described herein may be formulated for parenteral administration, e.g., by bolus injection or continuos infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative. The compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions.
Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen- free water based solution, before use.
The pharmaceutical composition of the present invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.
Pharmaceutical compositions suitable for use in context of the present invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of active ingredients (the peptide) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., inflammation) or prolong the survival of the subject being treated.
Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein. For any preparation used in the methods of the invention, the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.
Animal models for inflammatory colon diseases include animal models of ulcerative colitis such as, but are not limited to, trinitrobenzene sulfonic acid (TNBS)- induced colitis in rats and mice [Komori et al., J Gastroenterol (2005) 40: 591-599]. An animal model for adjuvant arthritis (AA, a model of rheumatoid arthritis) includes the rat heat-killed Mt strain H37Ra-induced AA [Kannan, Theor Biol Med Model. (2005) 2: 17]. An animal model for asthma includes the Ovalbumin (OVA) sensitization mouse model [Flaishon, L., et al, J. Immunol: Cutting edge 168: 3707 (2002)].
Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p.l).
Dosage amount and interval may be adjusted individually to provide sufficient levels of the active ingredient to induce or suppress the biological effect (minimal effective concentration, MEC). The MEC will vary for each preparation, but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.
Depending on the severity and responsiveness of the condition to be treated, dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.
The amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc. The dosage and timing of administration will be responsive to a careful and continuous monitoring of the individual changing condition.
Compositions of the present invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert. Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as is further detailed above.
Depending on the medical condition, the subject may be administered with additional chemical drugs (e.g., anti-inflammatory).
Anti-inflammatory agents which may be used according to the present teachings include, but are not limited to, Alclofenac; Alclometasone Dipropionate; Algestone Acetonide; Alpha Amylase; Amcinafal; Amcinafide; Amfenac Sodium; Amiprilose Hydrochloride; Anakinra; Anirolac; Anitrazafen; Apazone; Balsalazide Disodium; Bendazac; Benoxaprofen; Benzydamine Hydrochloride; Bromelains; Broperamole; Budesonide; Carprofen; Cicloprofen; Cintazone; Cliprofen; Clobetasol Propionate; Clobetasone Butyrate; Clopirac; Cloticasone Propionate; Cormethasone Acetate; Cortodoxone; Deflazacort; Desonide; Desoximetasone; Dexamethasone Dipropionate; Diclofenac Potassium; Diclofenac Sodium; Diflorasone Diacetate; Diflumidone Sodium; Diflunisal; Difluprednate; Diftalone; Dimethyl Sulfoxide; Drocinonide; Endrysone; Enlimomab; Enolicam Sodium; Epirizole; Etodolac; Etofenamate; Felbinac; Fenamole; Fenbufen; Fenclofenac; Fenclorac; Fendosal; Fenpipalone; Fentiazac; Flazalone; Fluazacort; Flufenamic Acid; Flumizole; Flunisolide Acetate; Flunixin; Flunixin Meglumine; Fluocortin Butyl; Fluorometholone Acetate; Fluquazone;
Flurbiprofen; Fluretofen; Fluticasone Propionate; Furaprofen; Furobufen; Halcinonide;
Halobetasol Propionate; Halopredone Acetate; Ibufenac; Ibuprofen; Ibuprofen
Aluminum; Ibuprofen Piconol; Ilonidap; Indomethacin; Indomethacin Sodium; Indoprofen; Indoxole; Intrazole; Isoflupredone Acetate; Isoxepac; Isoxicam;
Ketoprofen; Lofemizole Hydrochloride; Lomoxicam; Loteprednol Etabonate;
Meclofenamate Sodium; Meclofenamic Acid; Meclorisone Dibutyrate; Mefenamic
Acid; Mesalamine; Meseclazone; Methylprednisolone Suleptanate; Momiflumate;
Nabumetone; Naproxen; Naproxen Sodium; Naproxol; Nimazone; Olsalazine Sodium; Orgotein; Orpanoxin; Oxaprozin; Oxyphenbutazone; Paranyline Hydrochloride;
Pentosan Polysulfate Sodium; Phenbutazone Sodium Glycerate; Pirfenidone;
Piroxicam; Piroxicam Cinnamate; Piroxicam Olamine; Pirprofen; Prednazate; Prifelone;
Prodolic Acid; Proquazone; Proxazole; Proxazole Citrate; Rimexolone; Romazarit;
Salcolex; Salnacedin; Salsalate; Sanguinarium Chloride; Seclazone; Sermetacin; Sudoxicam; Sulindac; Suprofen; Talmetacin; Talniflumate; Talosalate; Tebufelone;
Tenidap; Tenidap Sodium; Tenoxicam; Tesicam; Tesimide; Tetrydamine; Tiopinac;
Tixocortol Pivalate; Tolmetin; Tolmetin Sodium; Triclonide; Triflumidate;
Zidometacin; Zomepirac Sodium.
As used herein the term "about" refers to ± 10 %.
The terms "comprises", "comprising", "includes", "including", "having" and their conjugates mean "including but not limited to".
The term "consisting of means "including and limited to".
The term "consisting essentially of means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
As used herein, the singular form "a", "an" and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a compound" or
"at least one compound" may include a plurality of compounds, including mixtures thereof.
Throughout this application, various embodiments of this invention may be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
Whenever a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases "ranging/ranges between" a first indicate number and a second indicate number and "ranging/ranges from" a first indicate number "to" a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals therebetween.
As used herein the term "method" refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
As used herein, the term "treating" includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition.
It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements. Various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below find experimental support in the following examples. EXAMPLES
Reference is now made to the following examples, which together with the above descriptions illustrate some embodiments of the invention in a non limiting fashion.
Generally, the nomenclature used herein and the laboratory procedures utilized in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are thoroughly explained in the literature. See, for example, "Molecular Cloning: A laboratory Manual" Sambrook et al, (1989); "Current Protocols in Molecular Biology" Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al, "Current Protocols in Molecular Biology", John Wiley and Sons, Baltimore, Maryland (1989); Perbal, "A Practical Guide to Molecular Cloning", John Wiley & Sons, New York (1988); Watson et al, "Recombinant DNA", Scientific American Books, New York; Birren et al. (eds) "Genome Analysis: A Laboratory Manual Series", Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057; "Cell Biology: A Laboratory Handbook", Volumes I-III Cellis, J. E., ed. (1994); "Current Protocols in Immunology" Volumes I-III Coligan J. E., ed. (1994); Stites et al. (eds), "Basic and Clinical Immunology" (8th Edition), Appleton & Lange, Norwalk, CT (1994); Mishell and Shiigi (eds), "Selected Methods in Cellular Immunology", W. H. Freeman and Co., New York (1980); available immunoassays are extensively described in the patent and scientific literature, see, for example, U.S. Pat. Nos. 3,791,932; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; 4,098,876; 4,879,219; 5,011,771 and 5,281,521; "Oligonucleotide Synthesis" Gait, M. J., ed. (1984); "Nucleic Acid Hybridization" Hames, B. D., and Higgins S. J., eds. (1985); "Transcription and Translation" Hames, B. D., and Higgins S. J., Eds. (1984); "Animal Cell Culture" Freshney, R. I., ed. (1986); "Immobilized Cells and Enzymes" IRL Press, (1986); "A Practical Guide to Molecular Cloning" Perbal, B., (1984) and "Methods in Enzymology" Vol. 1-317, Academic Press; "PCR Protocols: A Guide To Methods And Applications", Academic Press, San Diego, CA (1990); Marshak et al, "Strategies for Protein Purification and Characterization - A Laboratory Course Manual" CSHL Press (1996); all of which are incorporated by reference as if fully set forth herein. Other general references are provided throughout this document. The procedures therein are believed to be well known in the art and are provided for the convenience of the reader. All the information contained therein is incorporated herein by reference.
EXAMPLE 1
Construction and expression of soluble CD151 extracellular domain
(CD151ECD)
The present inventors have previously shown that CD151 has a significant role in T cell migration, its activation induces T cell migration in vitro and in vivo. In addition, blocking CD151, using a blocking antibody, results in inhibition of T cell migration in vitro and their entry to the lymph nodes (LN) in vivo. Thus, CD151 was established as a candidate for exerting global inhibitory effects on T cell trafficking and differentiation within peripheral lymph nodes and clinically beneficial as an anti-inflammatory agent.
Since homophilic interactions regulate CD151 activation, the present inventors have hypothesized that these interactions could be inhibited using a truncated CD151 fragment that can bind CD151, by acting as a competitive binder, thereby blocking the ability of the receptor to engage in further interactions. The present inventors have therefore designed, through laborious structural analysis and screening, a peptide consisting of amino acid coordinates 118-180 of SEQ ID NO: 1. termed, CD151-ECD, SEQ ID NO: 2 (Entelechon GmbH).
EXAMPLE 2
CD151ECD regulates T cell migration
Cytoskeleton rearrangement
Following incubation of T cells with CD151ECD (SEQ ID NO: 2) or a control peptide (20 or 40 μg/ml) (SEQ ID NO: 5, scrambled), T cells were stimulated with 100 ng/ml CCL21 chemokine (SLC) (PeproTech). Cytoskeleton rearrangement was analyzed by flow cytometry following staining with FITC-phalloidin, as previously described (Flaishon, L., et al.,. J. Biol. Chem., 2001 276(50):46701-6). Transwell migration
Following incubation of T cells with CD151ECD (SEQ ID NO: 2) or a control peptide (SEQ ID NO: 5, 40 μg/ml), chemotaxis was assayed by using transwell chambers, as previously described (Flaishon, L., et al.,. J. Biol. Chem., 2001). Briefly, about 4 x 106 T cells were pretreated with either stimulating or blocking anti-CD151 antibody for 1 hour as described in Methods. The migration towards the chemokine CCL21 (SLC) (0.4 mg/ml) (PeproTech) residing in the lower part of the apparatus, was analyzed after 3 hours by F AC Sort.
The assays were double controlled using SEQ ID NO: 4 as an additional independent control peptide (results not shown).
Vavl Immunoprecipitation
Protein G-Sepharose beads (GE Healthcare) were conjugated to Tyr (P) monoclonal antibody (Santa Cruz) for 2 hours on 4 °C, followed by three washes in phosphate-buffered saline. Beads were added to the cell lysates and proteins were immunoprecipitated overnight. The protein G-bound material was washed three times with phosphate-buffered saline containing 0.1% SDS and 0.5% Nonidet P-40. Immunoprecipitates were separated by SDS-PAGE. The protein bands were transferred onto a nitrocellulose membrane and probed with anti-Vavl (Cell Signaling) followed by horseradish peroxidase-conjugated anti-mouse (Jackson ImmunoResearch Labs,West Grove, PA).
Results
To determine whether CD151ECD regulates T cell migration, the present inventors have first analyzed the ability of T cells to polymerize their actin in the presence of CD151ECD or a control peptide. T cells were incubated with various concentrations of CD151ECD (10-80 μg/ml) or a control peptide for 1 hour, and then stimulated with CCL21 (0.1 mg/ml) (PeproTech) for 15 sec. The cells were immediately fixed with paraformaldehyde, permeabilized, stained with FITC-phalloidin, and then analyzed by flow cytometry to determine the state of their cytoskeleton. As shown in Figure 1A, CD151ECD specifically inhibited the ability of T cells to polymerize their actin at the concentrations of 20 μg/ml and more significantly at 40 μg/ml.
Thereafter, the present inventors determined whether CD151ECD regulates in vitro migration of T cells using a Transwell migration assay. Naive T cells were suspended with CD151ECD or a control peptide (40 μ§/ηι1) for 1 hr, and then placed in the upper chamber of a transwell. The migration towards the chemokine CCL21 (0.4 mg/ml) (PeproTech) residing in the lower part of the apparatus, was analyzed after 3 hours by FACS. As demonstrated in Figure IB, CD151ECD significantly inhibited the migration of control naive T cells toward CCL21 by about 60%.
Next, the effect of CD151ECD on the downstream signaling cascade was analyzed. The present inventors have previously shown that CD 151 activation induces Vav phosphorylation, therefore Vav phosphorylation was followed. T cells were incubated with CD151ECD or a control peptide (40 μg/ml) for 1 min. Cells were lysed, proteins were separated on SDS-PAGE and Vav phosohorylation was analyzed by probing the membrane with anti-Vavl antibody (Cell Signaling). As shown in Figure 2, CD151ECD down-regulated the Vav 1 -phosphorylation. Thus, blocking CD 151 with CD151ECD inhibited the CD151 induced-signaling cascade resulting in downregulation of actin polymerization and migration of T cells.
Example 3
CD151-ECD peptide inhibits the inflammatory response in DSS Colitis Induction of DSS Colitis
Materials and Methods
Animals -_DSS colitis was induced using 1% DSS (MP Biomedicals, Solon,
OH) in C57BL/6 mice, age 8 weeks, weighing 18-20 gr; as previously described (Ohkawara et al, 2002 Gastroenterology 123:256-270).
In vivo treatment with CD151-ECD - The mice were divided into three groups with 10 mice in each group. Each group was intraperitoneally (i.p.) injected with PBS, 80 μg CD151-ECD (SEQ ID NO: 2), or control scrambled peptide (SEQ ID NO: 5 Entelechon GmbH) in 200
Figure imgf000036_0001
of PBS from day 0 (immediately after DSS induction) to day 10, daily.
Murine Colonoscopy - Mice underwent murine colonoscopy on day 7 after disease induction. To monitor colitis, we used a high-resolution murine video endoscopic system, consisting of a miniature probe (1.9 mm outer diameter), a xenon light source, a triple chip HD camera, and an air pump ("Colo view," Karl Storz) to achieve regulated inflation of the mouse colon. Digitally recorded video files were processed with Windows Movie Maker software (Microsoft). Endoscopic quantification of colitis was graded using the MEICS (murine endoscopic index of colitis severity) system which consisted of five parameters: thickening of the colon wall, changes in the normal vascular pattern, presence of fibrin, mucosal granularity and stool consistency as described (Becker et al, 2005 GUT 54:950-954).
Histological assessment of DSS Colitis - Colons were fixed in 4 % paraformaldehyde for histology with hematoxylin and eosin (H&E) staining. The degree of histological damage and inflammation was graded in a blinded fashion. The amount of inflammation (0-4) was evaluated by estimation of the number of inflammatory cells.
Statistical Analysis - For statistical comparison of paired samples, a two-tailed
Student's t test was used.
Results
The powerful inhibitory effect of CD151-ECD on chemokine-triggered migration of naive T-lymphocytes in vitro suggested that this peptide might have anti- inflammatory activity in vivo . As a proof of this principle, the effect of CD151-ECD was examined in dextran sulfate sodium (DSS) colitis, a T cell dependent inflammatory disease induced by DSS administration through drinking water; which serves as an experimental model for human IBD (Velde et al, 2006 Inflammatory Bowel Diseases 12:995-999). It is generally believed that DSS is directly toxic to gut epithelial cells of the basal crypts, and affects the integrity of the mucosal barrier (Velde et al., 2006, supra).
Mice were administered 1% DSS in their drinking water for 5 days, followed by a 5 day rest period. During those 10 days, each group was intraperitoneally (i.p.) injected with CD151-ECD or control peptides. Body weight was monitored daily. On day 11 of the experiment, mice were sacrificed and the colon length was measured and was taken to histological evaluation. As shown in Figure 3 A, CD151-ECD had a favorable effect on weight maintenance, as the percentage of weight loss was significantly lower in the CD151-ECD group in comparison to the control peptide injected group. In addition, the shortening of colons in the CD151-ECD group was less pronounced than in the control group (Figures 3B-C) further suggesting that this CD 151 derived peptide exerts beneficial anti-inflammatory effects in this model. Therefore the present inventors have further assessed the inflammatory state of the control and the CD151 peptide treated mice by high-resolution live colonoscopy. CD151-ECD treated mice developed only mild intestinal inflammation, and a significant reduction in colonoscopy severity index was observed in this group compared to the control-peptide treated group (Figure 3D-E).
Next, the inflammation score of the differently treated mice by histology was assessed. As can be seen in Figure 3F, CD151-ECD significantly inhibited the development of inflammation, as was evaluated based on the histological examination that allowed us to estimate the number of inflammatory cells. These results further demonstrate that blocking the CD151 activity by CD151-ECD reduces the inflammatory response and further demonstrate the beneficial effect of CD151-ECD treatment. Thus, the samples of CD151-ECD group were less affected and showed milder colitis; thus, blocking CD151 activity by CD 151-ECD reduced the inflammatory response.
This CD 151-ECD peptide acts as a competitive inhibitor. The truncated CD151- ECD fragment was shown to inhibit the CD 151 induced- signaling cascade resulting in inhibition of Vav-1 phosphorylation, downregulation of actin polymerization and in vitro T cell migration. Analysis of the effect of CD 151-ECD in the DSS-induced colitis model revealed that daily administration of CD 151-ECD significantly decreased the development of DSS-induced colitis, as reflected by its favorable effect on weight loss, shortening of the colon, endoscopic colitis score and histological inflammation score.
Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims.
All publications, patents and patent applications mentioned in this specification are herein incorporated in their entirety by reference into the specification, to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention. To the extent that section headings are used, they should not be construed as necessarily limiting.

Claims

WHAT IS CLAIMED IS:
1. An isolated peptide comprising at least 20 consecutive amino acids of the amino acid sequence set forth in SEQ ID NO: 2, the peptide being shorter than 100 amino acids and having a CD151 inhibitory activity.
2. The isolated peptide of claim 1, wherein said CD151 inhibitory activity comprises inhibition of lymphocyte migration or homing.
3. The isolated peptide of claim 2, wherein said migration or homing is to a lymphoid organ.
4. The isolated peptide of claim 2, wherein said lymphocyte comprises a T cell.
5. The isolated peptide of claim 1, wherein said CD151 inhibitory activity comprises inhibition of Vav phosphorylation.
6. The isolated peptide of claim 1, being a soluble peptide.
7. A molecule comprising the isolated peptide of claim 1-6 attached to a heterologous moiety.
8. The molecule of claim 7, wherein said heterologous moiety is a proteinaceous moiety.
9. The molecule of claim 8, wherein said heterologous moiety comprises an immunoglobulin domain.
10. The molecule of claim 7, wherein said heterologous moiety is a non- proteinaceous moiety.
1 1. The molecule of claim 10, wherein said non-proteinaceous moiety comprises a synthetic polymer.
12. The isolated peptide of claim 1 or the molecule of claim 7, wherein said peptide is as set forth in SEQ ID NO: 2.
13. An isolated polynucleotide encoding the peptide of claim 1-9 or 12.
14. A nucleic acid expression construct comprising the isolated polynucleotide of claim 13 under the transcriptional control of a cis-acting regulatory element.
15. A pharmaceutical composition comprising as an active ingredient, the isolated peptide or molecule of any of claims 1-12, and a pharmaceutically acceptable carrier.
16. A method of treating inflammation in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the isolated peptide or molecule of any of claims 1-12, thereby treating the inflammation in the subject.
17. The isolated peptide or molecule of claim 1-12, for use in the treatment of inflammation.
18. The method of claim 16 or the use of claim 17, wherein said inflammation is associated with a medical condition selected from the group consisting of a cancer, an autoimmune disease, a hypersensitivity, a diabetes, an infectious disease, a transplantation associated disease and an allergy.
19. The method of claim 16, wherein said subject is a human being.
20. The method of claim 16, wherein said inflammation is associated with an inflammatory bowel disease.
21. The method of claim 16, wherein said inflammatory bowel disease comprises colitis.
PCT/IL2012/050111 2011-03-28 2012-03-28 Compositions and methods for treating inflammation WO2012131680A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161468109P 2011-03-28 2011-03-28
US61/468,109 2011-03-28

Publications (3)

Publication Number Publication Date
WO2012131680A2 true WO2012131680A2 (en) 2012-10-04
WO2012131680A9 WO2012131680A9 (en) 2012-12-13
WO2012131680A3 WO2012131680A3 (en) 2013-01-24

Family

ID=46051707

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2012/050111 WO2012131680A2 (en) 2011-03-28 2012-03-28 Compositions and methods for treating inflammation

Country Status (1)

Country Link
WO (1) WO2012131680A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules

Citations (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3791932A (en) 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3839153A (en) 1970-12-28 1974-10-01 Akzona Inc Process for the detection and determination of specific binding proteins and their corresponding bindable substances
US3850578A (en) 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3853987A (en) 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
US3879262A (en) 1972-05-11 1975-04-22 Akzona Inc Detection and determination of haptens
US3901654A (en) 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3935074A (en) 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3984533A (en) 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
US5011771A (en) 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US6472505B1 (en) 1997-05-14 2002-10-29 Aventis Pharmaceuticals Inc. Peptide parathyroid hormone analogs
US6777196B2 (en) 1997-02-18 2004-08-17 Genentech, Inc. Neurturin receptor
WO2007146968A2 (en) 2006-06-12 2007-12-21 Trubion Pharmaceuticals, Inc. Single-chain multivalent binding proteins with effector function
WO2008012796A2 (en) 2006-07-24 2008-01-31 Yeda Research And Development Co. Ltd. Pharmaceutical compositions comprising ccl2 and use of same for the treatment of inflammation
US20090324600A1 (en) 2006-10-18 2009-12-31 Pierre Fabre Medicament Use of an anti-cd151 antibody in the treatment of cancer
US20100150902A1 (en) 2008-04-11 2010-06-17 Jean-Francois Haeuw Anti-cd151 antibodies and their use in the treatment of cancer
WO2010137020A1 (en) 2009-05-28 2010-12-02 Yeda Research And Development Co. Ltd. Methods of treating inflammation

Patent Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3839153A (en) 1970-12-28 1974-10-01 Akzona Inc Process for the detection and determination of specific binding proteins and their corresponding bindable substances
US3791932A (en) 1971-02-10 1974-02-12 Akzona Inc Process for the demonstration and determination of reaction components having specific binding affinity for each other
US3901654A (en) 1971-06-21 1975-08-26 Biological Developments Receptor assays of biologically active compounds employing biologically specific receptors
US3853987A (en) 1971-09-01 1974-12-10 W Dreyer Immunological reagent and radioimmuno assay
US3867517A (en) 1971-12-21 1975-02-18 Abbott Lab Direct radioimmunoassay for antigens and their antibodies
US3879262A (en) 1972-05-11 1975-04-22 Akzona Inc Detection and determination of haptens
US3850578A (en) 1973-03-12 1974-11-26 H Mcconnell Process for assaying for biologically active molecules
US3935074A (en) 1973-12-17 1976-01-27 Syva Company Antibody steric hindrance immunoassay with two antibodies
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4034074A (en) 1974-09-19 1977-07-05 The Board Of Trustees Of Leland Stanford Junior University Universal reagent 2-site immunoradiometric assay using labelled anti (IgG)
US3984533A (en) 1975-11-13 1976-10-05 General Electric Company Electrophoretic method of detecting antigen-antibody reaction
US4098876A (en) 1976-10-26 1978-07-04 Corning Glass Works Reverse sandwich immunoassay
US4879219A (en) 1980-09-19 1989-11-07 General Hospital Corporation Immunoassay utilizing monoclonal high affinity IgM antibodies
US5011771A (en) 1984-04-12 1991-04-30 The General Hospital Corporation Multiepitopic immunometric assay
US4666828A (en) 1984-08-15 1987-05-19 The General Hospital Corporation Test for Huntington's disease
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683202B1 (en) 1985-03-28 1990-11-27 Cetus Corp
US4801531A (en) 1985-04-17 1989-01-31 Biotechnology Research Partners, Ltd. Apo AI/CIII genomic polymorphisms predictive of atherosclerosis
US5272057A (en) 1988-10-14 1993-12-21 Georgetown University Method of detecting a predisposition to cancer by the use of restriction fragment length polymorphism of the gene for human poly (ADP-ribose) polymerase
US5192659A (en) 1989-08-25 1993-03-09 Genetype Ag Intron sequence analysis method for detection of adjacent and remote locus alleles as haplotypes
US5281521A (en) 1992-07-20 1994-01-25 The Trustees Of The University Of Pennsylvania Modified avidin-biotin technique
US6777196B2 (en) 1997-02-18 2004-08-17 Genentech, Inc. Neurturin receptor
US6472505B1 (en) 1997-05-14 2002-10-29 Aventis Pharmaceuticals Inc. Peptide parathyroid hormone analogs
WO2007146968A2 (en) 2006-06-12 2007-12-21 Trubion Pharmaceuticals, Inc. Single-chain multivalent binding proteins with effector function
WO2008012796A2 (en) 2006-07-24 2008-01-31 Yeda Research And Development Co. Ltd. Pharmaceutical compositions comprising ccl2 and use of same for the treatment of inflammation
US20090324600A1 (en) 2006-10-18 2009-12-31 Pierre Fabre Medicament Use of an anti-cd151 antibody in the treatment of cancer
US20100150902A1 (en) 2008-04-11 2010-06-17 Jean-Francois Haeuw Anti-cd151 antibodies and their use in the treatment of cancer
WO2010137020A1 (en) 2009-05-28 2010-12-02 Yeda Research And Development Co. Ltd. Methods of treating inflammation

Non-Patent Citations (114)

* Cited by examiner, † Cited by third party
Title
"A Practical Guide to Molecular Cloning", 1984, PERBAL, B.
"Animal Cell Culture", 1986
"Basic and Clinical Immunology", 1994, APPLETON & LANGE
"Cell Biology: A Laboratory Handbook", vol. I-III, 1994
"Current Protocols in Immunology", vol. I-III, 1994
"Current Protocols in Molecular Biology", vol. I-III, 1994
"Genome Analysis: A Laboratory Manual Series", vol. 1-4, 1998, COLD SPRING HARBOR LABORATORY PRESS
"Immobilized Cells and Enzymes", 1986, IRL PRESS
"Methods in Enzymology", vol. 1-317, ACADEMIC PRESS
"Nucleic Acid Hybridization", 1985
"Oligonucleotide Synthesis", 1984
"PCR Protocols: A Guide To Methods And Applications", 1990, ACADEMIC PRESS
"Remington's Pharmaceutical .Sciences", MACK PUBLISHING CO.
"Selected Methods in Cellular Immunology", 1980, W. H. FREEMAN AND CO.
"Transcription and Translation", 1984
ALEXANDER RB. ET AL., UROLOGY, vol. 50, no. 6, December 1997 (1997-12-01), pages 893
ANTOINE JC.; HONNORAT J., REV NEUROL (PARIS, vol. 156, no. 1, January 2000 (2000-01-01), pages 23
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1989, JOHN WILEY AND SONS
BECKER ET AL., GUT, vol. 54, 2005, pages 950 - 954
BITTER ET AL., METHODS IN ENZYMOL., vol. 153, 1987, pages 516 - 544
BRALEY-MULLEN H.; YU S, J IMMUNOL, vol. 165, no. 12, 15 December 2000 (2000-12-15), pages 7262
BRISSON ET AL., NATURE, vol. 310, 1984, pages 511 - 514
BROGLI ET AL., SCIENCE, vol. 224, 1984, pages 838 - 843
CAPOROSSI AP ET AL., VIRAL IMMUNOL, vol. 11, no. 1, 1998, pages 9
CASTANO L.; EISENBARTH GS., ANN. REV. IMMUNOL., vol. 8, pages 647
CHAN OT. ET AL., IMMUNOL REV, vol. 169, June 1999 (1999-06-01), pages 107
CORUZZI ET AL., EMBO J., vol. 3, 1984, pages 1671 - 1680
CROSS AH. ET AL., J NEUROIMMUNOL, vol. 112, no. 1-2, 1 January 2001 (2001-01-01), pages 1
CUNHA-NETO E. ET AL., J CLIN INVEST, vol. 98, no. 8, 15 October 1996 (1996-10-15), pages 1709
DATTA SK, LUPUS, vol. 7, no. 9, 1998, pages 591
DIEKMAN AB. ET AL., AM J REPROD IMMUNOL., vol. 43, no. 3, March 2000 (2000-03-01), pages 134
EFREMOV DG. ET AL., LEUK LYMPHOMA, vol. 28, no. 3-4, January 1998 (1998-01-01), pages 285
ERIKSON J. ET AL., IMMUNOL RES, vol. 17, no. 1-2, 1998, pages 49
FEIST E. ET AL., INT ARCH ALLERGY IMMUNOL, vol. 123, no. 1, September 2000 (2000-09-01), pages 92
FINGL ET AL.: "The Pharmacological Basis of Therapeutics", 1975, pages: L
FLAISHON ET AL., BLOOD, vol. 112, 2008, pages 5016 - 5025
FLAISHON, L. ET AL., J. BIOL. CHEM., 2001
FLAISHON, L. ET AL., J. BIOL. CHEM., vol. 276, no. 50, 2001, pages 46701 - 6
FLAISHON, L. ET AL., J. IMMUNOL: CUTTING EDGE, vol. 168, 2002, pages 3707
FLAMHOLZ R. ET AL., J CLIN APHERESIS, vol. 14, no. 4, 1999, pages 171
FRANCO A. ET AL., CLIN IMMUNOL IMMUNOPATHOL, vol. 54, no. 3, March 1990 (1990-03-01), pages 382
G. SCHRODER; K. LUPKE: "The Peptides", vol. 1, 1965, ACADEMIC PRESS
GARCIA HEROLA A. ET AL., GASTROENTEROL HEPATOL., vol. 23, no. 1, January 2000 (2000-01-01), pages 16
GARZA KM. ET AL., J REPROD IMMUNOL, vol. 37, no. 2, February 1998 (1998-02-01), pages 87
GLODDEK B. ET AL., ANN N Y ACAD SCI, vol. 830, 29 December 1997 (1997-12-29), pages 266
GURLEY ET AL., MOL. CELL. BIOL., vol. 6, 1986, pages 559 - 565
HARA T. ET AL., BLOOD, vol. 77, no. 5, 1 March 1991 (1991-03-01), pages 1127
HASEGAWA ET AL., J OF IMMUNOLOGY, vol. 161, 1998, pages 3087 - 3095
HIEMSTRA HS. ET AL., PROC NATL ACAD SCI U S A, vol. 98, no. 7, 27 March 2001 (2001-03-27), pages 3988
HIEMSTRA HS. ET AL., PROC NATL ACAD SCI UNITS S A, vol. 98, no. 7, 27 March 2001 (2001-03-27), pages 3988
HONG ET AL., J BIOL CHEM, vol. 281, no. 34, 2006, pages 24279 - 92
INFANTE AJ.; AND KRAIG E, INT REV IMMUNOL, vol. 18, no. 1-2, 1999, pages 83
INFANTE AJ.; KRAIG E, INT REV IMMUNOL, vol. 18, no. 1-2, 1999, pages 83
J. M. STEWART; J. D. YOUNG: "Solid Phase Peptide Synthesis", 1963, W. H. FREEMAN CO.
J. MEIENHOFER: "Hormonal Proteins and Peptides", vol. 2, 1973, ACADEMIC PRESS, pages: 46
JAN VOSWINKEL ET AL., ARTHRITIS RES, vol. 3, no. 3, 2001, pages 189
JONES DE, CLIN SCI (COLCH, vol. 91, no. 5, November 1996 (1996-11-01), pages 551
KANNAN, THEOR BIOL MED MODEL., vol. 2, 2005, pages 17
KELLY CJ., J AM SOC NEPHROL, vol. 1, no. 2, August 1990 (1990-08-01), pages 140
KOMORI ET AL., J GASTROENTEROL, vol. 40, 2005, pages 591 - 599
KORNBERG AJ., J CLIN NEUROSCI., vol. 7, no. 3, May 2000 (2000-05-01), pages 191
KRENN V. ET AL., HISTOL HISTOPATHOL, no. 3, 15 July 2000 (2000-07-15), pages 791
KRENN V. ET AL., HISTOL HISTOPATHOL, vol. 15, no. 3, July 2000 (2000-07-01), pages 791
KUSUNOKI S., AM J MED SCI., vol. 319, no. 4, April 2000 (2000-04-01), pages 234
LACROIX-DESMAZES S. ET AL., SEMIN THROMB HEMOST., vol. 26, no. 2, 2000, pages 157
LANDAU YE.; SHOENFELD Y., HAREFUAH, vol. 138, no. 2, 16 January 2000 (2000-01-16), pages 122
LOBB; HEMLER, J CLIN INVEST, vol. 94, no. 5, 1994, pages 1722 - 8
MANNION ET AL., J IMMUNOL, vol. 157, no. 5, 1996, pages 2039 - 47
MANNS MP., J HEPATOL, vol. 33, no. 2, August 2000 (2000-08-01), pages 326
MARSHAK ET AL.: "Strategies for Protein Purification and Characterization - A Laboratory Course Manual", 1996, CSHL PRESS
MATSUURA E. ET AL., LUPUS, vol. 7, no. 2, 1998, pages 135
MATSUURA E. ET AL., LUPUS, vol. 7, no. 2, 1998, pages S135
MITSUMA T., NIPPON RINSHO., vol. 57, no. 8, August 1999 (1999-08-01), pages 1759
MOCCIA F., ANN ITAL MED INT., vol. 14, no. 2, April 1999 (1999-04-01), pages 114
NISHIUCHI ET AL., PROC NATL ACAD SCI U S A, vol. 102, no. 6, 2005, pages 1939 - 44
NOBILE-ORAZIO E. ET AL., ELECTROENCEPHALOGR CLIN NEUROPHYSIOL SUPPL, vol. 50, 1999, pages 419
NOEL LH., ANN MED INTERNE (PARIS)., vol. 151, no. 3, May 2000 (2000-05-01), pages 178
NOEL LH., ANN MED INTERNE (PARIS, vol. 151, no. 3, May 2000 (2000-05-01), pages 178
OHKAWARA ET AL., GASTROENTEROLOGY, vol. 123, 2002, pages 256 - 270
ORGIAZZI J. ENDOCRINOL METAB CLIN NORTH AM, vol. 29, no. 2, June 2000 (2000-06-01), pages 339
ORGIAZZI, J. ENDOCRINOL METAB CLIN NORTH AM, vol. 29, no. 2, June 2000 (2000-06-01), pages 339
ORON L. E, J NEURAL TRANSM SUPPL., vol. 49, 1997, pages 77
ORON L. ET AL., J NEURAL TRANSM SUPPL., vol. 49, 1997, pages 77
OSHIMA M. ET AL., EUR J IMMUNOL, vol. 20, no. 12, December 1990 (1990-12-01), pages 2563
PERBAL: "A Practical Guide to Molecular Cloning", 1988, JOHN WILEY & SONS
POSTIGO ET AL., RES IMMUNOL, vol. 144, no. 9, 1993, pages 723 - 35
PRAPROTNIK S. ET AL., WIEN KLIN WOCHENSCHR, vol. 112, no. 15-16, 25 August 2000 (2000-08-25), pages 660
RENAUDINEAU Y. ET AL., CLIN DIAGN LAB IMMUNOL., vol. 6, no. 2, March 1999 (1999-03-01), pages 156
SAKATA S. ET AL., MOL CELL ENDOCRINOL, vol. 92, no. 1, March 1993 (1993-03-01), pages 77
SALLAH S. ET AL., ANN HEMATOL, vol. 74, no. 3, March 1997 (1997-03-01), pages 139
SAMBROOK ET AL.: "Molecular Cloning: A laboratory Manual", 1989
SEMPLE JW. ET AL., BLOOD, vol. 87, no. 10, 15 May 1996 (1996-05-15), pages 4245
SODERSTROM M. ET AL., J NEUROL NEUROSURG PSYCHIATRY, vol. 57, no. 5, May 1994 (1994-05-01), pages 544
STRASSBURG CP. ET AL., EUR J GASTROENTEROL HEPATOL., vol. 11, no. 6, June 1999 (1999-06-01), pages 595
STUDIER ET AL., METHODS IN ENZYMOL., vol. 185, 1990, pages 60 - 89
TAKAMATSU ET AL., EMBO J., vol. 6, 1987, pages 307 - 311
TAKAMORI M., AM J MED SCI., vol. 319, no. 4, April 2000 (2000-04-01), pages 204
TINCANI A. ET AL., LUPUS, vol. 7, no. 2, 1998, pages 107 - 9
TISCH R; MCDEVITT HO, PROC NATL ACAD SCI U S A, vol. 91, no. 2, 18 January 1994 (1994-01-18), pages 437
TISCH R; MCDEVITT HO, PROC NATL ACAD SCI UNITS S A, vol. 91, no. 2, 18 January 1994 (1994-01-18), pages 437
TOYODA N. ET AL., NIPPON RINSHO, vol. 57, no. 8, August 1999 (1999-08-01), pages 1810
VAARALA O., LUPUS., vol. 7, no. 2, 1998, pages 132
VAARALA O., LUPUS., vol. 7, no. 2, 1998, pages S132
VELDE ET AL., INFLAMMATORY BOWEL DISEASES, vol. 12, 2006, pages 995 - 999
VINCENT A. ET AL., ANN N Y ACAD SCI., vol. 841, 13 May 1998 (1998-05-13), pages 482
WALLUKAT G. ET AL., AM J CARDIOL., vol. 83, no. 12A, 17 June 1999 (1999-06-17), pages 75H
WATSON ET AL.: "Recombinant DNA", SCIENTIFIC AMERICAN BOOKS
WEISSBACH; WEISSBACH: "Methods for Plant Molecular Biology", 1988, ACADEMIC PRESS, pages: 421 - 463
YAUCH ET AL., MOL BIOL CELL, vol. 9, no. 10, 1998, pages 2751 - 65
YOO TJ. ET AL., CELL IMMUNOL, vol. 157, no. 1, August 1994 (1994-08-01), pages 249
YUAN ET AL., BIOCHEM J, vol. 318, 1996, pages 591 - 6
ZAULI D. ET AL., BIOMED PHARMACOTHER, vol. 53, no. 5-6, June 1999 (1999-06-01), pages 234
ZIMMET P., DIABETES RES CLIN PRACT, vol. 34, October 1996 (1996-10-01), pages 125
ZIMMET P., DIABETES RES CLIN PRACT, vol. 34, October 1996 (1996-10-01), pages S125

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014159813A1 (en) 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules

Also Published As

Publication number Publication date
WO2012131680A9 (en) 2012-12-13
WO2012131680A3 (en) 2013-01-24

Similar Documents

Publication Publication Date Title
Bach et al. The IFNγ receptor: a paradigm for cytokine receptor signaling
ES2281072T3 (en) SURFACE RECEIVER OF ACTIVATED T CELLS: ACTS-4.
AU2001261585C1 (en) Compositions and methods for achieving immune suppression
JP2011050389A (en) Novel composition and method for treatment of immune related disease
JP2010099073A (en) Composition and method for treatment of natural killer cell-related disease
JP4901747B2 (en) Novel compositions and methods for the treatment of immune related diseases
KR100620334B1 (en) Ligand for herpes simplex virus entry mediator and methods of use
JP2002515246A (en) IL-17 homologous polypeptides and their therapeutic uses
WO2007094005A2 (en) Molecules and methods of using same for treating ccr5/ccr5 ligands associated diseases
CN102482324A (en) Novel chemokine binding polypeptides capable of inhibiting the course of autoimmunity, inflammation and cancer
JP2013240329A (en) Composition and method for treatment of systemic lupus erythematosus
KR102569494B1 (en) Isolated polypeptides of cd44 and uses therof
JP4602954B2 (en) Use of interleukin-22 polypeptide and antibody binding thereto
JP2007525972A (en) Anti-angiogenic peptides from the N-terminus of endostatin
WO2010103517A1 (en) Soluble compositions for the treatment of cxcr3-ligand associated diseases
EP2566500A2 (en) Use of ccl1 in therapy
WO2012131680A2 (en) Compositions and methods for treating inflammation
JP2013048621A (en) Composition and method for treatment of immune related diseases
JP4280444B2 (en) Compositions and methods for inhibiting neoplastic cell growth
WO2006126209A1 (en) Polypeptides and compositions comprising same and methods of using same for treating cxcr4 associated medical conditions
JP2006515748A (en) Compositions and methods for the treatment of rheumatoid arthritis
ES2321122T3 (en) CHEMIOCINE ANTAGONISTS CC.
US20120070449A1 (en) Methods of treating inflammation
JP2004520811A (en) Interleukin-22 polypeptide, nucleic acid encoding the same, and method for treating pancreatic disease
JP4166254B2 (en) Borkine polypeptide and nucleic acid

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12720011

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12720011

Country of ref document: EP

Kind code of ref document: A2