WO2010070047A1 - Soluble polypeptides for use in treating autoimmune and inflammatory disorders - Google Patents

Soluble polypeptides for use in treating autoimmune and inflammatory disorders Download PDF

Info

Publication number
WO2010070047A1
WO2010070047A1 PCT/EP2009/067411 EP2009067411W WO2010070047A1 WO 2010070047 A1 WO2010070047 A1 WO 2010070047A1 EP 2009067411 W EP2009067411 W EP 2009067411W WO 2010070047 A1 WO2010070047 A1 WO 2010070047A1
Authority
WO
WIPO (PCT)
Prior art keywords
soluble
sirpα
polypeptide
seq
binding polypeptide
Prior art date
Application number
PCT/EP2009/067411
Other languages
French (fr)
Inventor
Marianne Raymond
Marie Sarfati
Karl Welzenbach
Maximilian Woisetschlaeger
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to CN2009801511374A priority Critical patent/CN102257001A/en
Priority to EP09796708A priority patent/EP2379584A1/en
Priority to AU2009327058A priority patent/AU2009327058A1/en
Priority to EA201100947A priority patent/EA201100947A1/en
Priority to CA2747678A priority patent/CA2747678A1/en
Priority to BRPI0923442A priority patent/BRPI0923442A2/en
Priority to MX2011006621A priority patent/MX2011006621A/en
Priority to US13/132,400 priority patent/US20110237498A1/en
Priority to JP2011541441A priority patent/JP2012512640A/en
Publication of WO2010070047A1 publication Critical patent/WO2010070047A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present Invention relates to soluble CD47 binding polypeptides, for use as a medicament, in particular for the prevention or treatment of autoimmune and inflammatory disorders, for example anergic asthma and inflammatory bowel diseases.
  • the invention more specifically relates to a soluble CD47 binding polypeptide for use as a medicament, comprising an extracellular domain of SIRP ⁇ (CD172a) or functional derivatives which bind to human CD47.
  • CD47 is a cell surface glycoprotein which binds to SIRP ⁇ (alias SHPS-1) and SIRP ⁇ on opposing cells. This interaction leads to negative regulation of immune celt function or can serve to mediating cellular adhesion and migration.
  • CD47 was suggested for use as a biologies in the treatment of autoimmune disorders (WO199 ⁇ /040940). In contrast, there are very few evidence of a potential use of CD47 tigands, such as SIRP ⁇ for similar therapeutic purposes.
  • One explanation is the ubiquitous expression of CD47 that would prevent from using CD47 binding polypeptides as potential drugs.
  • the present invention provides first in vivo evidence that SIRP ⁇ -Fc construct is suitable for preventing or stopping Th1/Th17- and Th2-driven diseases in animal models of disease. These data provide the basis for this invention and support the drugab ⁇ ity of StRP ⁇ -derived protein therapeutics.
  • the invention is based, in part, on the discovery that the manipulation of CD47/SIRP ⁇ pathway suppresses immunogenic CD103 * dendritic cells-driven pathogenesis of Th1r ⁇ i17- (arthritis and colitis) as well as Th2 driven diseases (allergic asthma).
  • the invention provides soluble CO47 binding polypeptides, for use as a medicament, comprising a SIRP ⁇ -d ⁇ rived polypeptide selected among the group consisting of a) an extracellular domain of SIRP ⁇ (SEQ ID NO:3); b) a fragment of SEQ ID NO 1 , and, c) a variant polypeptide of SEQ ID NO.1 having at least 75% identity to SEQ ID NO:3; wherein said SIRP ⁇ -derived polypeptide binds to human CD47 (SEQ ID NO:24)
  • the variant polypeptide of SEQ ID NO:3 is at least 80%, 85%, 90%, 9$% or 99% identical to SEQ ID NO:3.
  • soluble CD47 binding polypeptides according to the present invention are hereafter designated as the "Soluble Polypeptides of the Invention".
  • said SIRP ⁇ -derived polypeptide is selected among antagonist of CD47, i.e., a polypeptide that competitively inhibits the binding of a CD47 Kgand to CD47.
  • CD47 Kenya include, without limitation, SIRP ⁇ , SIRP ⁇ or TSP1.
  • said SIRP ⁇ -derived polypeptide is selected among agonist of CD47, i.e., a polypeptide that is capable of inducing CD47 signaling activity
  • said soluble CD47 binding polypeptide is selected among those that bind to human CD47 with a Ko of 2 ⁇ M or less and/or inhibits induced cytokine secretion as measured in an immune complex-stimulated dendritic cell cytokine release assay.
  • said SIRP ⁇ -derived polypeptide is an extracellular domain of SIRP ⁇ comprising at least the V-region of SIRP ⁇ (SEQ ID NO:2).
  • the Soluble Polypeptide of the Invention is a fusion polypeptide comprising a first component consisting of a SIRP ⁇ -derived polypeptide fused to a second heterologous polypeptide.
  • the Soluble Polypeptide further comprises a spacer between the second heterologous polypeptide and the SIRP ⁇ -derived polypeptide.
  • the SIRP ⁇ derived polypeptide is fused to an IgO Fc domain.
  • said Fc domain is a silent Fc fragment of human IgGI isotype.
  • said Fc domain is an aglycosylated mutant variant of human IgGI isotype.
  • the Soluble Polypeptides of the Invention are used a ⁇ drugs m the treatment of autoimmune and inflammatory disorders. Preferred indications are selected among the group consisting of Th2 ⁇ mediated airway inflammation, anergic disorders, asthma, inflammatory bowel diseases and ischemic disorders.
  • the Soluble Polypeptides of the Invention may be used as drugs in the treatment of leukemias or cancer.
  • CO47 refers to human CD47.
  • Human CD47 includes SEQ ID NO:24 but also any natural polymorphic, for example, comprising single nucleotide polymorphisms (SNPs), or splice variants of human CO47. Examples of splice variants or SNPs in CO47 nucleotide sequence found in human are described in TaWe 1.
  • SIRP ⁇ refers to the Signal Regulatory Protein Alpha (also designated CD172a or SHPS- 1) which shows adhesion to CD47 integrin associated protein.
  • SIRP ⁇ refers to human SIRP ⁇ as defined in SEQ ID NO.23.
  • Human SIRP ⁇ contains an amino acids extracellular domain (SEQ ID NO:3), with one V-type domain (SEQ ID NO.2), and two C 1 -type Ig domains and three potential N-glycosylation sites. It has a 110 amino acids cytoplasmic sequence with ITIM motifs that recruit tyrosine phosphatases SHP- 1 and SHP-2 when phosphorylated.
  • human SIRP ⁇ further includes, without limitation, any natural polymorphic, for example, comprising single nucleotide polymorphisms (SNPs), or splice variants of human SIRP ⁇ . Examples of splice variants or SNPs in SIRP ⁇ nucleotide sequence found in human are described in Table 2.
  • a polypeptide is "soluble" when K lacks any transmembrane domain or protein domain that anchors or integrates the polypeptide into the membrane of a cell expressing such polypeptide.
  • the Soluble Polypeptides of the Invention may likewise exclude transmembrane and Intracellular domains of SIRP ⁇ .
  • a polypeptide that "binds to CD47” is intended to refer to a polypeptide that binds to human CD47 with a K 0 of a 20 ⁇ M or less, 2 ⁇ M or less, 0.2 ⁇ M or less.
  • a polypeptide that binds to CD47 further binds to surfactant protein A (SP-A) and/or surfactant protein O (SP-O).
  • SP-A surfactant protein A
  • SP-O surfactant protein O
  • a polypeptide that inhibits induced cytokine secretion as measured in a immune complex-stimulated dendritic cell cytokine release assay is a polypeptide that inhibits cytokine (e.g. IL- ⁇ , IL-10, IL-12p70, IL-23, IL-8 and/or TNF- ⁇ ) release from peripheral blood monocytes, conventional dendritic ce ⁇ s (DCs) as weH as monocyte-derived DCs stimulated with Staphylococcus Aureus Cowan 1 (Pansorbin) or soluble CD40L and IFN- ⁇ .
  • cytokine e.g. IL- ⁇ , IL-10, IL-12p70, IL-23, IL-8 and/or TNF- ⁇
  • the Soluble Polypeptides of the invention inhibit cytokine secretion as measured in an immune complex- stimulated dendritic c ⁇ U cytokine release assay at an IC 90 of 1 ⁇ M or less, 10OnM or less, or 1OnM or less.
  • a polypeptide that inhibits T cefl proliferation may be measured in a mixed lymphocyte reaction assay as described in the Example.
  • K MMC or "K,” as used herein, is intended to refer to the association rate of a particular protein-protein interaction
  • K- or “K*” as used herein, is intended to refer to the dissociation rate of a particular protein-protein interaction
  • K 0 is intended to refer to the dissociation constant, which is obtained from the ratio of K 4 to K « (i.e. K 4 ZK,) and is expressed as a molar concentration (M).
  • K 0 values for protein-protein interaction can be determined using methods well established in the art. A method for determining the K 0 of an protein/protein interaction is by using surface plasmon resonance, or using a biosensor system such as a Biacore* system.
  • affinity refers to the strength of interaction between the polypeptide and its target at a single site. Within each site, the binding region of the polypeptide interacts through weak non-covalent forces with its target at numerous sites; the more interactions, the stronger the affinity.
  • high affinity for a binding polypeptide refers to a polypeptide having a K 0 of 1OnM or less, for example, 1nM or less, for its target.
  • the term "subject” includes any human or nonhuman animal.
  • Tbe term “nonhuman animal” includes ait vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc.
  • the term, "optimized" means that a nucleotide sequence has been altered to encode an amino acid sequence using codons that are preferred in the production cell or organism, either a eukaryotic ceil, for example, a cell of Pichia or Saocharomyces, a cell of Trichod ⁇ rma, a Chinese Hamster Ovary ceo (CHO) or a human ceB, or a prokaryotic ce ⁇ , for example, a strain of Escherichia coli.
  • a eukaryotic ceil for example, a cell of Pichia or Saocharomyces
  • a cell of Trichod ⁇ rma a Chinese Hamster Ovary ceo (CHO) or a human ceB
  • a prokaryotic ce ⁇ for example, a strain of Escherichia coli.
  • the optimized nucleotide sequence is engineered to retain completely or as much as possible the amino acid sequence originatty encoded by the starting nucleotide sequence, which is also known as the "parental" sequence.
  • the optimized sequences herein have been engineered to have codons that are preferred in the corresponding production cell or organism, for example a mammalian cell, however optimized expression of these sequences in other prokaryotic or eukaryotic c ⁇ fl ⁇ is also envisioned herein.
  • the amino acid sequences encoded by optimized nucleotide sequences are also referred to as optimized.
  • Soluble Polypeptides of the Invention comprise SIRP ⁇ -derived polypeptides that are selected among the group consisting of a) an extracellular domain of SIRP ⁇ (SEQ ID NO:3); b) a fragment of SEQ ID NO: 3, and, c) a variant polypeptide of SEQ ID NO:3; wherein said SIRP ⁇ -derived polypeptide binds to human CD47 (SEQ ID NO.24).
  • Soluble Polypeptides of the Invention and their SIRP ⁇ -derived fragments should retain the capacity to bind to CD47. Fragments of SEQ ID NO:3 can therefore be selected among those fragments comprising the CD47 binding domain of SIRP ⁇ . Those fragments generally do not comprise the transmembrane and intracellular domains of SIRP ⁇ .
  • SIRP ⁇ -derived polypeptide essentially consists of SEQ ID NO:3 or SEQ ID NO.2.
  • SIRP ⁇ -derived polypeptides further include, without limitation, variant polypeptides of SEQ ID NO.3 where amino acids residues have been mutated by amino acid d ⁇ tetton, insertion or substitution, yet have at least 60, 70, 80, 90 or 95 percent identity to SEQ ID NO: 3, so long as changes to the native sequence do not substantially affect the biological activity of the molecule, in particular its binding to CD47.
  • it include mutant amino acid sequences wherein no more than 1, 2, 3, 4 or 5 amino acids have been mutated by amino acid deletion, insertion or substitution in the SIRP ⁇ -derived polypeptide when compared with SEQ ID NO:2.
  • mutant ammo acid sequences are those sequences derived from single nucleotide polymorphisms (see Table 2).
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described below.
  • the percent identity between two amino acid sequences can be determined using the algorithm of E. Meyers and W. Miller (Comput. Appi. Biosci., 4:11-17, 1988) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. MoI, Biol. 48:444-453, 1970) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, ⁇ . or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
  • the SIRP ⁇ -derived polypeptide includes changes to SEQ ID NO.3 or SEQ ID NO.2 that contain conservative amino acid substitutions.
  • Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginme, histidine), acidic side chains (e.g., aspartJc acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, gkitamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isote ⁇ cine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g..).
  • basic side chains e.g., lysine, arginme, hist
  • one or more amino acid residues within the CD47 binding region of SIRP ⁇ - derjved polypeptide can be reptaoed with other amino add residues from the same side chain family, and the new polypeptide variant can be tested for retained function using the binding or functional assays described herein.
  • the SIRP ⁇ -derived polypeptides are selected among those that retain the capacity to inhft ⁇ t cytokine secretion as measured in a immune complex-stimulated dendritic DC cytokine release assay at least to the same extent as the polypeptide of SEQ IO NO: 3 comprising the extracellular domain of human SIRP ⁇ .
  • the SIRP ⁇ -derived polypeptides are selected among those that retain the capacity to inhibit T cell proliferation as measured in a mixed lymphocyte reaction assay.
  • the SIRP ⁇ -derived polypeptides are selected among those that cross-react with non-human primate CD47.
  • the Soluble Polypeptides of the Invention are fusion polypeptides comprising the SIRP ⁇ -derived polypeptides.
  • the Soluble Polypeptides of the Invention are fusion polypeptides comprising the SIRP ⁇ -derived polypeptides and a second heterologous amino acid sequence, e.g. a portion of one or more proteins other than SIRP ⁇ , covatentfy bound to the SIRP ⁇ -deriv ⁇ d polypeptide at the letter's N- and/or C-terminus, and optionally further comprising a linker.
  • the non SIRPu-derived protein can be preferably a soluble single chain polypeptide, which, when fused to another heterologous protein, is capable of increasing half life of the resulting fusion protein in blood.
  • the non-SIRP ⁇ -derived protein comprises a domain for multimerization of the fusion polypeptide.
  • the non SIRP ⁇ -derived protein can be, for example, an immunoglobulin, serum albumin and fragments thereof.
  • the non SIRP ⁇ -derived protein can also be a polypeptide capable of binding to serum albumin proteins to increase half life of the resulting molecule when administered in a subject Such approach is for example described in Nygren et a!., EP 0 488 525.
  • the non-SIRP ⁇ derived protein is an Fc domain.
  • Fc moiety for making soluble construct with increased in vivo half life in human is weJ! known in the art and for example described in Capon et al (US5.428.130).
  • Fc domain refers to the constant region of an immunoglobulin.
  • An Fc domain comprises at least the CH2 and CH3 domain, optionally, the hinge region which is located between the heavy chain CH1 domain and CH2.
  • Fc fragments could be obtained for example by papain digestion of an immunoglobulin.
  • Fc domain further include Fc variants into which a substitution, deletion or insertion of at least one amino acid has been introduced.
  • the hinge region of CW is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased.
  • the number of cysteine residues m the hinge region of CH1 is altered to, for example, facHitate assembly of the light and heavy chains or to increase or decrease the stab ⁇ ty of the fusion polypeptide.
  • the Fc region is modified to increase its biological half-life.
  • one or more of the following mutations can be introduced: T252L, T254S, T256F, as described in U.S. Patent No. 6,277,375 to Ward.
  • the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector functions of the Fc portion.
  • one or more amino adds can be replaced with a different amino acid residue such that the Fc portion has an altered affinity for an effector ligand.
  • the effector Rgand to which affinity » altered can be, for example, an Fc receptor or the C 1 component of complement.
  • one or more amino acids selected from amino acid residues can be replaced with a different amino acid residue such that the resulting Fc portion has altered C1q binding and/or reduced or abolished complement dependent cytotoxicity (CDC).
  • CDC complement dependent cytotoxicity
  • one or more amino acid residues are altered to thereby alter the ability of the Fc region to fix complement. This approach is described further in PCT Publication WO 94/29351 by Bodmer et al.
  • the Fc region is modified to increase the ability of the fusion polypeptide to mediate antibody dependent ceflular cytotoxicity (ADCC) and/or to increase or decrease the affinity of the Fc region for an Fey receptor by modifying one or more amino acids.
  • ADCC antibody dependent ceflular cytotoxicity
  • the Fc domain is of human origin and may be from any of the tmmunoglobu ⁇ n classes, such as IgG or IgA and from any subtype such as human IgGI , lgG2, igG3 and igG4.
  • the Fc domain is from a non-human animal, for example, but not limited to, a mouse, rat, rabbit, camel, shark, non-human primate or hamster.
  • the Fc domain of IgGI isotype is used.
  • a mutant variant of IgGI Fc fragment is used, e.g. a stent IgGI Fc which reduces or eliminates the ability of the fusion polypeptide to mediate antibody dependent ceflular cytotoxicity (AOCC) and/or to bind to an Fey receptor.
  • An example of an IgGI isotype silent mutant is a so-caS ⁇ d LALA mutant, wherein Leucine residue is replaced by Alanine residue at amino acid positions 234 and 235 as described in J. Virol 2001 D ⁇ c;75 ⁇ 24):i2i ⁇ i-8 by Hezareh et al.
  • the Fc domain is a mutant preventing glycosyfation at residue at position 297 of Fc domain.
  • an amino acid substitution of asparagtne residue at position 297 of the Fc domain is the replacement of N297 by a glycine or an alanine.
  • the Fc domain comprises a c ⁇ merization domain, preferably via cysteine capable of making covalent disulfide bridge between two fusion polypeptides comprising such Fc domain.
  • the SIRP ⁇ -derived polypeptide can be fused directly in frame with the non-SIRP ⁇ -derived protein or via a polypep ⁇ dic linker (spacer).
  • spacer may be a single amino acid (such as for example, a glycine residue) or between 5-100 amino acids, for example between 5-20 amino acids.
  • the linker should permit the SIRP ⁇ -derived domain to assume the proper spatial orientation to form a binding site with CD47.
  • the glycosylate pattern of the Soluble Polypeptide of the Invention can be altered compared to typical mammalian glycosylate pattern such as those obtained in CHO or human cefl lines.
  • an agiycosiated polypeptides can be made by using prokaryotlc cefl Hnes as host ce ⁇ s or mammalian cells that has been engineered to lack glycosylate.
  • Carbohydrate modifications can also be accomplished by; for example, altering one or more sites of glycosylate within the Soluble Polypeptide.
  • a glycosylated polypeptide can be made that has an altered type of glycosylate.
  • Such carbohydrate modifications can be accomplished by, for example, expressing the Soluble Polypeptides in a host cell with altered glycosylate machinery, Le the glycosylate pattern of the Soluble Polypeptide is altered compared to the glycosylate pattern observed in corresponding wild type ce ⁇ s.
  • Cells with altered glycosylate machinery have been described in the art and can be used as host cells in which to express recombinant Soluble Polypeptides of the Invention to thereby produce such Soluble Polypeptides with altered glycosylate.
  • the Soluble Polypeptides of the Invente can be produced in yeast, e.g., Pichi ⁇ pastoas, or filamentous fungi, e.g., Trichod ⁇ rma f&es ⁇ i engineered for mammalian-like glycosylate pattern (see for example EP129717261).
  • yeast e.g., Pichi ⁇ pastoas
  • filamentous fungi e.g., Trichod ⁇ rma f&es ⁇ i engineered for mammalian-like glycosylate pattern
  • Advantages of those giycoengineered host celts are, inter alia, the previse of polypeptide composites with homogeneous glycosylate pattern and/or higher yield.
  • Soluble Polypeptides herein that is contemplated by the invention is pegylation.
  • the Soluble Polypeptides of the invente for example, consisting essentially of SIRP ⁇ -derived polypeptides can be pegyiated.
  • Pegylatton is a well-known technology to increase the biological (e.g.. serum) half-life of the resulting biologies as compared to the same biologies without pegylation.
  • the polypeptide is typically reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the polypeptide*.
  • PEG polyethylene glycol
  • the p ⁇ gytation can be carried out by an acylatkm reaction or an afeyJatJon reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer).
  • a reactive PEG molecule or an analogous reactive water-soluble polymer.
  • polyethylene glycol is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (C1-C10) aikoxy- or arytoxy-polyethylene glycol or polyethylene glycol-maleimide.
  • Methods for pegyteting proteins are known in the art and can be applied to the Soluble Polypeptides of the invention. See for example, EP 0 154 316 by NterAnura et aJ. and EP 0 401 384 by IsNkawa et a!.
  • the polymeric carrier may comprise at least one natural or synthetic branched, finear or dendritic polymer.
  • the polymeric carrier is preferably soluble in water and body fluids and is preferably a pharmaceutically acceptable polymer. Water soluble polymer moieties include, but are not limited to, e.g.
  • poty ⁇ lkyiene glycol and derivatives thereof including PEG, PEG homopoJym ⁇ r ⁇ , mPEG, potypropytenegtycol homopoJyrner ⁇ , copolymers of ethylene glycol with propylene glycol, wherein said homopolymers and copolymers are unsubstituted or substituted at one end e.g. with an acykjroup; polygiycerines or polysia ⁇ c acid; carbohydrates, polysaccharides, cellulose and cellulose derivatives, including methylceNulose and carboxymethylceHulo ⁇ e; starches (e.g.
  • hydroxyalkyi starch especially hydroxyethyl starch (HES) and dextrin ⁇ s, and derivatives thereof
  • dextran and dextran derivatives including d ⁇ xtransutfat, cros ⁇ ttnked dextrin, and carboxymethyl dextrin
  • chitosan a linear polysaccharide
  • heparin and fragments of heparin polyvinyl alcohol and polyvinyl ethyl ethers
  • polyvinylpyrrollidon alpha, beta-poM ⁇ 2-hydroxyethyl)-OL-aspaftamide
  • polyoxy- ethyfated pofyote polyoxy- ethyfated pofyote.
  • the Soluble Polypeptides of the Invention have been shown to protect from inflammatory disorders such as allergic asthma or inflammatory bowel diseases in animal model and therefore may be used as a medicament, in particular to decrease or suppress (in a statistically or biologically significant manner) the Inflammatory and/or autoimmune response, in particular, a response mediated by SIRPa+ cells in a subject.
  • nucleic acid molecules that encode the Soluble Polypeptides of the Invention or at least SIRP ⁇ -derived polypeptides, including without limitation, the embodiments related to fusion polypeptides.
  • nucleotide sequences encoding Soluble Polypeptides of the Invention comprise SEQ ID NOs: 26 or 27.
  • the nucleic acids may be present in whole cells, in a ceU lysate, or may be nucleic acids in a partially purified or substantially pure form.
  • a nucleic add is Isolated" or "rendered substantially pure” when purified away from other cellular components or other contaminants, e.g., other ceH ⁇ lar nucleic adds or proteins, by standard techniques, induding aHcaline/SDS treatment, CsCI banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, F. Ausubel, et al., ed. 1087 Current Protocols in Molecular Biology, Greene Publishing and WHey Inter ⁇ cienc ⁇ , New York.
  • a nucleic acid of the invention can be, for example, DNA or RNA and may or may not contain kitronic sequences.
  • the nucleic acid is a cDNA molecule.
  • the nucleic acid may be present in a vector such as a phage display vector, or in a recombinant plasmid vector.
  • DNA fragments encoding the Soluble Polypeptides of the Invention are obtained, for example, fusion polypeptide comprising the SIRP ⁇ -derived polypeptides as described above, these DNA fragments can be further manipulated by standard recombinant DNA techniques, for example to include any signal sequence for appropriate secretion in expression system, any purification tag and cleavable tag for further purification steps. In these manipulations, a DNA fragment is operative ⁇ finked to another DNA molecule, or to a fragment encoding another protein, such as a purification/secretion tag or a flexible linker.
  • operatively linked is intended to mean that the two DNA fragments are joined in a functional manner, for example, such that the amino acid sequences encoded by the two DNA fragments remain in-frame, or such that the protein is expressed under control of a desired promoter.
  • the Soluble Polypeptides of the Invention can be produced in a host cell transfectoma using, for example, a combination of recombinant DNA techniques and gene transfection methods as is well known in the art.
  • a host cell transfectoma using, for example, a combination of recombinant DNA techniques and gene transfection methods as is well known in the art.
  • DNAs encoding the corresponding polypeptides can be obtained by standard molecular biology techniques (e.g., PCR amplification or cONA cloning using a hybridoma that expresses the polypeptides of interest) and the ONAs can be inserted into expression vectors such that the corresponding gene is operattv ⁇ ty linked to transcriptional and transiational control sequences.
  • the term "operatively linked” is intended to mean that a gene » Hgated into a vector such that transcriptional and transiational control sequences within the vector serve their intended function of regulating the transcription and translation of the gene.
  • the expression vector and expression control sequences are chosen to be compatible with the expression host c ⁇ H used.
  • the gene encoding Soluble Polypeptides or intermediates are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the gene fragment and vector, or blunt end ligation if no restriction sites are present).
  • AdcHtionaDy or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the polypeptide chain from a host cell.
  • the gene can be cloned into the vector such that the signal peptide is linked in frame to the amino terminus of the polypeptide chain.
  • the signal peptide can be a SIRP ⁇ signal peptide or a heterologous signal peptide (i.e., a signal peptide not naturally associated with SIRP ⁇ sequence).
  • the recombinant expression vectors of the invention carry regulatory sequences that control the expression of the gene in a host cell.
  • the term "regulatory sequence * is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the polypeptide chain genes.
  • Such regulatory sequences are described, for example, in Goedd ⁇ l (Gene Expression Technology. Methods in Enzymology 185, Academic Press. San Diego, CA 1990). It wt ⁇ be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences, may depend on such factors as the choice of the host ce ⁇ to be transformed, the level of expression of protein desired, etc.
  • Regulatory sequences for mammalian host ceil expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV), Simian Virus 40 (SV40), adenovirus (e.g., the adenovirus major late promoter (AdMLP)), and polyoma.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • polyoma e.g., the adenovirus major late promoter (AdMLP)
  • nonviral regulatory sequences may be used, such as the ubiquitin promoter or P-globin promoter.
  • regulatory elements composed of sequences from different sources such as the SRa promoter system, which contains sequences from the SV40 early promoter and the long terminal repeat of human T cefl leukemia virus type 1 (Takebe, Y.
  • the recombinant expression vectors of the invention may carry additional sequences, such as sequence* that regulate replication of the vector in host cefi ⁇ (e.g., origins of replication) and selectable marker genes.
  • sequence* that regulate replication of the vector in host cefi ⁇ (e.g., origins of replication)
  • selectable marker genes e.g., origins of replication
  • the selectable marker gene fac ⁇ itates selection of host cells into which the vector has been introduced (see, e.g., U.S. Pat Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et aJ ).
  • the selectable marker gene confers resistance to drugs, such as G418, hygromyctn or methotrexate, on a host cell into which the vector has been introduced.
  • Selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr- host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
  • DHFR dihydrofolate reductase
  • neo gene for G418 selection.
  • the expression vectors) encoding the Soluble Polypeptide or intermediates such as SIRP ⁇ -derived polypeptide is transfect ⁇ d into a host celt by standard techniques.
  • the various forms of the term "tran ⁇ fection" are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., etectroporation, cak ⁇ um-pho ⁇ phate precipitation, DEAE-dextran transection and the like. It is theoretically possible to express the Soluble Polypeptides of the Invention in either prokaryotic or eukaryotic host ce ⁇ .
  • glycoprotein in eukaryotic ceils in particular mammalian host cells, is discussed because such eukaryotic cefls, and in particular mammalian ceBs, are more likely than prokaryotic cells to assemble and secrete a properly folded and biologically active glycoprotein such as the Soluble Polypeptides of the Invention.
  • Mammalian host cells for expressing the Soluble Polypeptides and intermediates such as SIRP ⁇ -derived polypeptides of the invention include Chinese Hamster Ovary (CHO eels) (including dhfr- CHO c ⁇ Hs, described Urlaub and Chastn, 1980 Proc. Natl. Acad. Sd. USA 77:4216-4220 used with a DH FR selectable marker, e.g., as described in RJ. Kaufman and PA Sharp, 1982 MoI. Biol. 159:601-621. NSO myeloma cells, COS cells and SP2 cells) or human ceB lines (including PER-C ⁇ cell Hoes, Crucell).
  • Chinese Hamster Ovary CHO eels
  • dhfr- CHO c ⁇ Hs described Urlaub and Chastn, 1980 Proc. Natl. Acad. Sd. USA 77:4216-4220 used with a DH FR selectable marker, e.g., as
  • another expression system is the GS gene expression system shown in WO 87/04462, WO 89/01036 and EP 338,841.
  • the Soluble Polypeptides or intermediates such as SIRP ⁇ -derived polypeptides are produced by cuJturing the host cells for a period of time sufficient to aflow for expression of the recombinant polypeptides in the host cells or secretion of the recombinant polypeptides into the culture medium in which the host cett ⁇ are grown.
  • the polypeptides can then be recovered from the culture medium using standard protein purification methods.
  • the present invention provides multivalent proteins comprising at least two identical or different Soluble Polypeptides of the Invention binding to CD47.
  • the multivalent proteins comprise at least two, three or four Soluble Polypeptides of the Invention.
  • the Soluble CD47-binding Polypeptides can be linked together via protein fusion or covalent or non covalent linkage.
  • the multivalent proteins of the present invention can be prepared by conjugating the constituent binding specificities, using methods known in the art. For example, each binding specificity of the multivalent protein can be generated separately and then conjugated to one another.
  • cross-linking agents include protein A, carbodt ⁇ nide, N-su ⁇ midyl-S-ac ⁇ tyMhioac ⁇ tat ⁇ (SATA), 5.5'-dithiob ⁇ s(2-nrtrob ⁇ nzo»c acid) (OTNB).
  • o-phenylenedimaleimide oPDM
  • SPDP N- suo ⁇ nfmidyi-3 ⁇ 2-pyridyldithk>)propionate
  • suifosuccinlmidyl 4-(N- maleimidomethyl) cyctohaxan ⁇ -l-carboxyt ⁇ te suffo-SMCC
  • Karpovsky et al. 1984 J. Exp. Med. 160:1686; Uu, MA et al., 1985 Proc. Natl. Acad. Sd. USA 82:8648.
  • Other methods include those described in PauJus, 1985 Behring Ins. Mitt. No.
  • Covalent Hnkage can be obtained by disulfide bridge between two cysteines, for example disulfide bridge from cysteine of an Fc domain.
  • the hinge region of an Fc domain fused to SIRP ⁇ -derived polypeptide is modified to contain an odd number of sulfhydryl residues, for example one, prior to conjugation.
  • both binding specificities can be encoded in the same vector and expressed and assembled in the same host cell.
  • the present invention provides a composition, e.g., a pharmaceutical composition, containing one or a combination of the Soluble Polypeptides of the present Invention, formulated together with a pharmaceutically acceptable carrier.
  • Pharmaceutica) formulations comprising a Soluble Polypeptide of the invention may be prepared for storage by mixing the polypeptide having the desired degree of purity with optional physiotogicaHy acceptable carriers, ex ⁇ pients or stabilizers ⁇ Remington: The Science and Practice of Pharmacy 20th edition (2000)), in the form of aqueous solutions, lyophiMzed or other dried formulations. Therefore, the invention further relates to a lyophized composition comprising at least the Soluble Polypeptide of the invention.
  • compositions of the invention also can be administered in combination therapy, i.e., combined with other agents.
  • the combination therapy can include a Soluble Polypeptide of the present Invention combined with at least one other antiinflammatory or another chemotherapeutic agent. Examples of therapeutic agents that can be used in combination therapy are described in greater detail below in the section on uses of the Soluble Polypeptides of the Invention.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the Ice that are physiologically compatible.
  • the carrier should be suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion).
  • the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • the pharmaceutical compounds of the invention may include one or more pharmaceutically acceptable salts.
  • a "pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxfcotogicai effects (see e.g., Berge, S.M., et at , 1977 J. Pharm. Sd. 66.1-19). Examples of such salts include acid addition salts and base addition salts.
  • Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydrok ⁇ fic, phosphorous and the Wee, as well as from nontoxic organic acids such as alphatic mono* and dt-carboxytic acids, phenyl-substituted a&anoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like.
  • nontoxic inorganic acids such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydrok ⁇ fic, phosphorous and the Wee
  • nontoxic organic acids such as alphatic mono* and dt-carboxytic acids, phenyl-substituted a&anoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like.
  • Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as N 1 N * - dibenzylethyfenediamine, N-methyiglucamine, chloroprocaine, choline, diethanoiamine, ethyienediamine, procaine and the like.
  • a pharmaceutical composition of the Invention also may Include a pharmac ⁇ uticaly acceptable anti-oxktam
  • pharmaceutica ⁇ y acceptable antioxidants include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisutfate, sodium metabisuJfite, sodium sulfite and the Mke; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butyiated hydroxytoluene (BHT), lecithin, propyl gaHate, alpha-tocopherol, and the like; and metal chelating agents, such as citric add, ethytenediamine tetraacetic acid (EDTA), sorbitol, tartaric add, phosphoric acid, and the Bke.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisutfate, sodium metabisuJfite, sodium sulfite and the Mke
  • aqueous and nonaqueous carriers examples include water, ethanol, polyote (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as ofive ofl, and injectable organic esters, such as ethyl oieate.
  • polyote such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as ofive ofl
  • injectable organic esters such as ethyl oieate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures, supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, cntorobutanol, phenol sorbic acid, and the Bee. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as, aluminum monostearate and gelatin.
  • adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • compositions typically must be sterile and stable under the conditions of manufacture and storage.
  • the composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyoJ (for example, glycerol, propylene glycol, and Squid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of the injectable compositions can be brought about by including In the composition an agent that delays absorption for example, monostearate salts and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, fo ⁇ owed by sterilization microfUtration.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, and the particular mode of administration.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect. Generally, out of one hundred percent, this amount wHI range from about 0.01 per cent to about ninety-nine percent of active ingredient, from about 0.1 per cent to about 70 per cent, or from about 1 percent to about 30 percent of active ingredient in combination with a pharmaceutically acceptable carrier.
  • Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the requir ⁇ d pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuate.
  • the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight.
  • dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg.
  • An exemplary treatment regime entails administration once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months or once every three to ⁇ months.
  • Dosage regimens for a Soluble Polypeptide of the Invention include 1 mg/kg body weight or 3 mg/kg body weight by intravenous administration, with the polypeptide being given using one of the following doting schedules: every four weeks for six dosages, then every three months; every three weeks; 3 mg/kg body weight once followed by 1 mg/kg body weight every three weeks.
  • the Soluble Polypeptide is usually administered on multiple occasions. Intervals between single dosages can be, for example, weekly, monthly, every three months or yearly. Intervals can also be irregular as indicated by measuring blood levels of Soluble Polypeptide in the patient. In some methods, dosage is adjusted to achieve a plasma polypeptide concentration of about 1-1000 ⁇ g/ml and in some methods about 25-300 ug/ml.
  • the Soluble Polypeptide can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the Soluble Polypeptide in the patient. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated or until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level wi8 depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex. weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a "th ⁇ rapeuticaty effective dosage" of Soluble Polypeptide of the Invention can result in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction.
  • a composition of the present invention can be administered by one or more routes of administration using one or more of a variety of methods known hi the art.
  • routes and/or mode of administration wiH vary depending upon the desired results.
  • Routes of administration for Soluble Polypeptides of the Invention include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion.
  • parenteral administration' 1 means modes of administration other than enteral and topical administration, usua ⁇ y by injection, and includes, without limitation, intravenous, intramuscular, Intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrastemal injection and infusion.
  • a Soluble Polypeptide of the Invention can be administered by a nonparenteral route, such as a topical, epidermal or mucosal route of administration, for example, intranasalty, oratty, vaginally, recta ⁇ y, sublingualy ortopica ⁇ y.
  • a nonparenteral route such as a topical, epidermal or mucosal route of administration, for example, intranasalty, oratty, vaginally, recta ⁇ y, sublingualy ortopica ⁇ y.
  • the active compounds can be prepared with carriers that win protect the compound against rapid release, such as a controfl ⁇ d release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • a controfl ⁇ d release formulation including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, poryglycolic acid, collagen, poiyorthoesters, and poryiactic acid. Many methods for the preparation of such formulations are patented or generally known to those sktRed In the art. See. e.g., Sustained and Controlled Release Drug Delivery Systems, J R. Robinson, ed., Marcel Dekker, Inc., New York, 1 ⁇ 78.
  • compositions can be administered with medical devices known In the art.
  • a therapeutic composition of the invention can be administered with a needleless hypodermic Injection device, such as the devices shown in U.S. Patent No ⁇ . 5,399.163; 5.383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824 or 4,596,556.
  • a needleless hypodermic Injection device such as the devices shown in U.S. Patent No ⁇ . 5,399.163; 5.383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824 or 4,596,556.
  • wei known implants and modules useful in the present invention include: U.S. Patent No. 4,487,603, which shows an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Patent No. 4,486,194, which shows a therapeutic device for administering medicants through the skin; U.S. Patent No.
  • the Soluble Polypeptides of the Invention can be formulated to ensure proper distribution hi vivo.
  • the blood-brain barrier (BBB) excludes many highly hydrophiHc compounds.
  • BBB blood-brain barrier
  • the therapeutic compounds of the invention cross the BBB (if desired ⁇ , they can be formulated, for example, in liposomes.
  • liposomes For methods of manufacturing liposomes, see, e.g., U.S. Patents 4,522,811; 5,374,548; and 5,399,331.
  • the liposomes may comprise one or more moieties which are selectively transported into specific cells or organs, thus enhance targeted drug delivery (see, e.g., V. V. Ranade, 1989 J. CHne Pharmacol. 29:685).
  • Exemplary targeting moieties include folate or biotin (see, e.g., U.S. Patent 5,416,016 to Low et al.); mannosides (Umezawa et a!., 1988 Biochem. Biophys. Res. Commun. 153:1038); antibodies (P.G. Bioeman et al., 1995 FEBS Lett. 357:140; M. Owais et al., 1995 Antimicrob. Agents Chemother. 39:180); surfactant protein A receptor (Briscoe et al., 1995 Am. J. Physiol.1233: 134); pi20 (Schreier et a) , 1994 J. Biol. Chem. 269:9090); see also K. Keinanen; M.L Laukkanen, 1994 FEBSLett. 346:123; J J. Killton; IJ. Ftdler, 1994 lmmunomethods 4:273.
  • biotin see,
  • Soluble Polypeptides of the Invention have in vitro and in vivo diagnostic and therapeutic utilities.
  • these molecules can be administered to cells in culture, e.g. in vitm or in vivo, or in a subject, e.g., in vivo, to treat, prevent or diagnose a variety of disorders.
  • Non-human animals include a ⁇ vertebrates, e.g., mammals and non-mammals, such as non- human primates, sheep, dogs, cats, cows, horses, chickens, amphibians, and reptiles.
  • the methods are particularly suitable for treating, preventing or diagnosing autoimmune and inflammatory disorders mediated by SIRPa+ cells, e.g., a ⁇ ergic asthma or ulcerative colitis.
  • SIRPa+ cells e.g., a ⁇ ergic asthma or ulcerative colitis.
  • autoimmune and inflammatory disorders mediated by SIRPa+ cells
  • a ⁇ ergic asthma or ulcerative colitis include Inflammatory conditions, allergies and allergic conditions, autoimmune diseases, ischemic disorders, severe infections, and ceHs, organ or tissue transplant rejection, including xenotransplant (i.e. transplant from different species, for example from non-human species to human) of cells, tissues or organs.
  • autoimmune diseases includes, without limitation, arthritis (for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases, including inflammatory conditions and rheumatic diseases involving bone toss, inflammatory pain, spondyloarhropathies including ankylosing spondylitis, ReHer syndrome, reactive arthritis, psoriatic arthritis, and enterophath ⁇ arthritis, hypersensitivity (Including both airways hypersensitivity and dermal hypersensitivity) and aiergies.
  • Autoimmune diseases include autoimmune haematoiogicai disorders (including e.g.
  • hemolytic anaemia aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia
  • systemic lupus erythematosus inflammatory muscle disorders, polychondritis, scterodoma, Wegener granulomatosis, derrnatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis.
  • Steven-Johnson syndrome idiopathic sprue, endocrine ophthalmopathy, Graves disease, sarcoidosis, multiple sclerosis, primary biliary cirrhosis, juvenile diabetes (diabetes mettitus type I), uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g.
  • Th ⁇ Soluble Polypeptides of the Invention are also useful for the treatment, prevention, or amelioration of asthma, bronchitis, pneumoconiosis, pulmonary emphysema, and other obstructive or inflammatory diseases of the airways.
  • IgE mediated disorders include atopic disorders, which are characterized by an inherited propensity to respond immunologicafly to many common naturally occurring inhaled and ingested antigens and the continual production of IgE antibodies.
  • Specific atopic disorders includes aflergic asthma, allergic rhinitis, atopic dermatitis and allergic gastroenteropathy.
  • disorders associated with elevated IgE levels are not limited to those with an inherited (atopic) etiology.
  • Other disorders associated with elevated IgE levels, that appear to be IgE-mediated and are treatable with the formulations of this present invention include hypersensitivity (e. g., anaphylactic hypersensitivity), eczema, urticaria, allergic bronchopulmonary aspergillosis, parasitic diseases, hyper-lgE syndrome, ataxia- telangiectasia, Wiskott-Aldrich syndrome, thymic atymphoplasia, IgE myeloma and graft- versus-host reaction.
  • hypersensitivity e. g., anaphylactic hypersensitivity
  • eczema urticaria
  • allergic bronchopulmonary aspergillosis parasitic diseases
  • hyper-lgE syndrome ataxia- telangiectasia
  • Wiskott-Aldrich syndrome thymic atymphoplasia
  • the Soluble Polypeptides of the Invention may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to or in combination to, other drugs e.g. immunosuppressive or immunomodiiating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above.
  • other drugs e.g. immunosuppressive or immunomodiiating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above.
  • the Soluble Polypeptides of the invention may be used in combination with DMARD, e.g.
  • Gold salts sulphasalazine, antimalaria ⁇ , methotrexate, D-penidllamine, azathioprine, mycopheno ⁇ c add, cydosporine A, tacrolimus, siroiimus, minocycline, leflunomide, glucocorticoids; a calcmeurin inhibitor, e.g. cyclosporin A or FK 506; a modulator of lymphocyte recirculation, e.g. FTY720 and FTY720 analogs; a mTOR inhibitor, e.g.
  • rapamycin 40-O-(2-hydroxyethyl)- rapamycin, CCI779, ABT578, AP23573 or TAFA-93; an ascomydn having immunosuppressive properties, e.g. ABT-281 , ASM981, etc.; corticosteroids; cyclo-phos-phamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenotic add; myco-pheno-late mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC 1 CO2, CD3, CD4, CO7, CD8.
  • immunosuppressive monoclonal antibodies e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC 1 CO2, CD3, CD4, CO7, CD8.
  • LFA-1 antagonists ICAM- 1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g. pacHtaxe), gemcitabine, dspiatinum, doxorubicin or 5-fluorouracil; anti TNF agents, e.g. monoclonal antibodies to TNF, e.g. infliximab, ada&mimab, CDP870, or receptor constructs to TNF-RI or TNF-RII, e.g. Etanercept, PEG-TNF-RI; blockers of proinflammatory cytokines, . IL-1 blockers, e.g.
  • Anakinra or IL- 1 trap AAL160, ACZ 885, IL-6 blockers; chemokines blockers, e g inhibitors or activators of proteases, e.g. meta ⁇ oproteases, antt-IL-15 antibodies, antJ-IL-6 antibodies, anU-CD20 antibodies, anti-CD22 antibodies, antML17 antibodies, NSAIDs, such as aspirin or an artWnfectiou ⁇ agent (list not limited to the agent mentioned).
  • chemokines blockers e g inhibitors or activators of proteases, e.g. meta ⁇ oproteases, antt-IL-15 antibodies, antJ-IL-6 antibodies, anU-CD20 antibodies, anti-CD22 antibodies, antML17 antibodies, NSAIDs, such as aspirin or an artWnfectiou ⁇ agent (list not limited to the agent mentioned).
  • the Soluble Polypeptides of the Invention are also useful as co-therapeutic agents for use in conjunction with anti-inflammatory or bronchodilatory drug substances, particularly in the treatment of obstructive or inflammatory airways diseases such as those mentioned hereinbefore, for example as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs.
  • An agent of the invention may be mixed with the ariti-inflammatory or bronchodilatory drug in a fixed pharmaceutical composition or it may be administered separately, before, simultaneously with or after the antiinflammatory or bronchodilatory drug.
  • Such anti-inflammatory drugs include steroids, in particular giucocorticosteroids such as budesonide, be amethasone, fluticasone or mometasone, and dopamine receptor agonists such as cabergoiine, bromocriptine or ropinJrol ⁇ .
  • bronchodilatory drugs include anticholinergic or antimuscarMc agents, in particular ipratropium bromide, oxftropium bromide and tiotropium bromide.
  • the present invention also provides a method for the treatment of an obstructive or inflammatory airways disease which comprises administering to a subject, particularly a human subject, in need thereof a SoIuWe Polypeptide, as hereinbefore described.
  • the invention provides a Soluble Polypeptide, as hereinbefor ⁇ described for use in the preparation of a medicament for the treatment of an obstructive or Inflammatory airways disease.
  • the Soluble Polypeptides of the Invention are also particularly useful for the treatment, prevention, or amelioration of chronic gastrointestinal inflammation, such as inflammatory bowel diseases, including Crohn's disease and ulcerative ooHtis.
  • Chronic gastrointestinal inflammation refers to inflammation of the mucosal of the gastrointestinal tract that is characterized by a relatively longer period of onset, is long- lasting (e.g., from several days, weeks, months, or years and up to the We of the subject), and is associated with infiltration or influx of mononuclear cells and can be further associated with periods of spontaneous remission and spontaneous occurrence. Thus, subjects with chronic gastrointestinal inflammation may be expected to require a long period of supervision, observation, or care.
  • “Chronic gastrointestinal inflammatory conditions” also referred to as “chronic gastrointestinal inflammatory diseases” having such chronic inflammation include, but are not necessarily limited to, inflammatory bowel disease (IBD), co ⁇ tts induced by environmental insults (e.g..
  • gastrointestinal inflammation e.g., colitis
  • a therapeutic regimen such as administration of chemotherapy, radiation therapy, and the like
  • colitis in conditions such as chronic granulomatous disease (Schappi et al. Arch CKs Child.
  • celiac disease a heritable disease in which the intestinal lining is inflamed Ri response to the ingestion of a protein known as gluten
  • food allergies gastritis, infectious gastritis or enterocolitis (e.g., Helicobacter pylori-infected chronic active gastritis) and other forms of gastrointestinal inflammation caused by an infectious agent, and other like conditions.
  • inflammatory bowel disease or "IBO” refers to any of a variety of diseases characterized by inflammation of a ⁇ or part of the intestines.
  • examples of inflammatory bowel disease include, but are not limited to, Crohn's disease and ulcerative colitis.
  • Reference to IBD throughout the specification is often referred to in the specification as exemplary of gastrointestinal inflammatory conditions, and is not meant to be limiting.
  • the present invention also provides a method for the treatment of chronic gastrointestinal inflammation or inflammatory bowel diseases, such as ulcerative colitis, which comprises administering to a subject, particularly a human subject, in need thereof, a Soluble Polypeptide, as hereinbefore described.
  • the invention provides a Soluble Polypeptide, as hereinbefore described for use in the preparation of a medicament for the treatment of chronic gastrointestinal inflammation or inflammatory bowel diseases.
  • the present invention is also useful in the treatment, prevention or amelioration of leukemias or other cancer disorders.
  • Also encompassed within the scope of the present invention is a method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of a Soluble Polypeptide, and at least one second drug substance, said second drug substance being a immunosuppressive / immunomodulatory, antiinflammatory chemotherapeutic or anti-infectious drug, e.g. as indicated above.
  • a therapeutic combination e.g. a kit, comprising of a therapeutically effective amount of a) a Soluble Polypeptide of the Invention and b) at least one second substance selected from a immuno- ⁇ uppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti-infectious drug, e.g. as indicated above.
  • the kit may comprise instructions for its administration.
  • Soluble Polypeptides of the Invention are administered in conjunction with other immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti- infectious therapy
  • dosages of the co-administered combination compound will of course vary depending on the type of co-drug employed, on the condition being treated and so forth.
  • the Soluble Polypeptides of the invention can be used to detect levels of CD47+ dendritic cells, or levels of cells that contain CD47. This can be achieved, for example, by contacting a sample (such as an in vitro sample) and a control sample with a Soluble Polypeptide of the Invention under conditions that allow for the formation of a complex between the Soluble Polypeptides and CD47 expressing cells. Any complexes formed are detected and compared in the sample and the control. For example, standard detection methods, well known in the art, such as flow cytometric assays, can be performed using the compositions of the invention.
  • the invention further provides methods for detecting the presence of CD47 (e.g., human CD47) in a sample, or measuring the amount of CO47, comprising contacting the sample, and a control sample, with a Soluble Polypeptide of the Invention, under conditions that aftow for formation of a complex between the Soluble Pofyp ⁇ ptkJ ⁇ and CD47. The formation of a complex is then detected, wherein a difference in complex formation between the sample compared to the control sample is indicative of the presence of CD47 in the sample.
  • CD47 e.g., human CD47
  • kits consisting of the compositions of the invention and instructions for use.
  • the kit can further contain a least one additional reagent.
  • Kits typicaBy include a label indicating the intended use of the contents of the kit The term label includes any writing, or recorded material supplied on or with the kit, or which otherwise accompanies the kit.
  • the kit may further comprise tools for diagnosing whether a patient belongs to a group that wifl respond to a treatment, as defined above.
  • FIG. 1 Murine SIRP ⁇ -Fc binds CO47+/+ (WT) but not CD47-7- (KO) cede.
  • Murine SIRP ⁇ - Fc binding to CD47+/+ (WT) or CD47-/- (KO) murine sptenocytes was detected by flow cytometry as described.
  • the fluorescence resulting from SIRP ⁇ -Fc binding (SIRP) is plotted as dot piot versus FL3.
  • SIRP ⁇ -Fc binds to CD47+/+ expressing (Jin8CD47) but not CD47 deficient Jurkat T celts (J ⁇ i8). SIRP ⁇ -Fc binding was quantified by flow cytometry as described. The fluorescence resulting from SIRP ⁇ -Fc binding (SlRP) is plotted as histogram in bold Hne, Bold line indicates binding in presence of lOug/mL anti CD47 clone B6H12.
  • FIG. 3 Blocking CD47/SIRP ⁇ at priming prevents aHergJc disease development in BALB/c mice.
  • (3a) Mice were OVA-sensitized i.p. on day 0 and 5 in the presence or absence of SIRP ⁇ -Fc fusion molecules, and received OVA-aerosol challenges on day 12, 16 and 20 and euthanized on day 21 (n 4 to 7 mice per group).
  • FIG. 1 Mechanisms of disease protection in SIRP ⁇ -Fc treated BALB/c mice. Mice were OVA-sensitized on day 0 and 5 in the presence or absence of SIRP ⁇ -Fc fusion molecule, received OVA-aerosol challenge* on day 12, 16 and 20 and were euthanized on day 21.
  • OVA- aiumn immunization in the absence (P8S) or presence of SIRP ⁇ -Fc and CFSE eel dilution was examined in mLNs after 2 days.
  • Data is one representative experiments performed on 4 mice per group.
  • (4c) At day 21, absolute numbers of eosinophils (CCR3+) were assessed by flow cytometry in ex vivo isolated mLNs. Data are mean ⁇ SEM (n 3 to 4 mice per group).
  • Colitis was induced and assessed as described. 100 ug /animal murine SIRP ⁇ -Fc was applied at day 0 and 24h after i.p.. Alternatively PBS was injected i.p. ⁇ odyweight of animals was assessed until day 4 post TNBS injection.
  • the affinity of human SIRP ⁇ -Fc to monomeric CD47 or divalent CD47-FC protein can be assessed by Biacore.
  • Monovalent interaction of CO47 V-domain with SIRPu is reported around 1 ⁇ M (Heathertey et al MoI Cell. 2008).
  • an APP-tagged CD47 V domain protein is expressed in HEK293 cells and compared to a divalent-CD47-Fc protein as ligand.
  • the monovalent interaction with SIRP ⁇ - Fc was measured as K 0 of 0.8 ⁇ M while the binding strength (avidity) of a divalent CD47-Fc protein increased by a factor of 10 to K D ⁇ 6OnM. In contrast binding of a murine CD47-Fc fusion protein could not be observed indicating the specificity of the assessed interaction.
  • 5x10 5 CD47 * and CD4r J ⁇ rkat T cell lines are resuspended in 50 ⁇ l of FACS BUFFER (PBS 2% FCS 2mM EDTA) containing 200 ⁇ g/ml human IgG and 5 ⁇ g/ml SiRPa-Fc for 30 min at 4°C. After washing, cells are stained with FITC-labeled streptavidin (1/1000) for 30 min at 4°C.
  • inhibition/blocking studies of biotinylated SIRP ⁇ -Fc binding to CHO CD47 cell lines may be performed using anti-CD47 mAbs directed against different epitopes (i.e. B6H12. 2D3, BRIC 126, IF7 and 10G2 donee, different anti- human SIRP- ⁇ x(CD172a) mAbs, recombinant human Thrombospondin-1 (TSP-1) or TSP-1 c-terminal (4N1K) and control (4NG6) peptides.
  • anti-CD47 mAbs directed against different epitopes i.e. B6H12. 2D3, BRIC 126, IF7 and 10G2 donee
  • different anti- human SIRP- ⁇ x(CD172a) mAbs recombinant human Thrombospondin-1 (TSP-1) or TSP-1 c-terminal (4N1K) and control (4NG6) peptides.
  • Inhibition/blocking studies of directiy labeled anti-CD47 mAb biding to CHO CD47 oeK lines may be performed using non biotinyiated human SIRP- ⁇ -Fc.
  • Peripheral blood monocytes (CD14+ CD16-) as well as monocyte-derived dendritic cells (DCs) are prepared as described (Lato ⁇ r et ai, J of Immunol, 2001: 167:2547).
  • Conventional (DCs) are isolated as CDH(M-, lineage-, by a FACS Ana (BD Biosciences) by using aflophycocyanin (APC)-lab ⁇ l ⁇ d anti-CD11c (B-Jy ⁇ ), a mixture of FITOIab ⁇ t ⁇ d mAb ⁇ against lineage markers.
  • CD3 ,CD14, CD15, CD16, CD19 and CD56 and APC-Cy7-iabeled CD4 (RPA-T4) to reach >99% purity.
  • APCs are stimulated with Staphylococcus Aureus Cowan 1 at 1/40.000 (Pansorbin) or soluble CD40L (1 ⁇ g/ml) and IFN- ⁇ (500 U/ml) in the presence of various concentrations of human SIRP ⁇ -Fc (1 to 20 ⁇ g/mt) in HB101 serum-free medium.
  • Cytokine (IL-1, IL-6, IL- 10, IL-12p70, IL-23, IL-8 and TNF- ⁇ ) release is assessed by ELISA in the 24h or 48h culture supernatant ⁇ .
  • Mitomycin c-treated mature DCs will be cocuKured with allogeneic unfractionated, naTve (CD45RA * 0062L 1 *") or memory (CD45RO * CD62L taw ) adult CD4+ T cells ( 10 6 ZmI) at various stimulators (DCs)/ responder (T cede) ratios in the presence or absence of Soluble Polypeptides of the Invention (5 to 50 ⁇ g/ml). T cell proliferation ( 3 H thymidine uptake) and IFN- ⁇ release will be assessed in the culture supernatants of 5 to 6 day primary cultures. 4. In vivo date with murine animal models for use of 8IRPa-Fc in the treatment of asthma
  • mice were sensitized by i.p. injection of 10ug OVA (Sigma, Grade V) adsorbed to 1mg Imject Alum (Pierce) on days 0 and 5.
  • OVA aerosol Sigma, Grade V
  • mice were chaUenged for 30 minutes with a 0.5% OVA aerosol (Sigma, Grade V) delivered by a vibrating mesh nebulizer system (Omron).
  • Omron vibrating mesh nebulizer system
  • mice 24 hours after the last chafl ⁇ nge, mice were sacrificed with 75mg/kg sodium pentobarbital overdose and bled.
  • BAL was co ⁇ ected 3 times with 0.5ml physiologic saline and lung and mLN were isolated.
  • Lymphocytes were smafi, non-granular, non-aut ⁇ fluorescent, expressing CD3 or B220 and the other mononuclear ceils, including macrophages and DCs, were lacking CD3, B220 and CCR3.
  • mLNs and lungs were first treated with liberase. minced and cells were counted. Lung ce ⁇ suspensions were treated with NH4CI for red blood cell lysis and washed before staining.
  • anti-CD4 FITC or APC (BD Biosciences), anti- CD25 PE (Caltag) or FITC (BD Biosciences), anti-CD44 APC (clone IM7 8.1) were used.
  • FITC FITC
  • anti-CD44 APC clone IM7 8.1
  • mast ceils and basophils were first identified with extracellular antMgE-biotinylated and anti-CD117 (c-Kit, BioLegend) and CD4 T cells were stained with anti-CD4 APC.
  • Cells were fixed, permeabilized and stained with antML-13 PE (eBioscience).
  • IL-2, IL-4, IL-5, 1 L- 10, IFN-y (BO Btoscience ⁇ ), IL- 13 (R&D Systems) are measured in mLN ⁇ culture supernatants and king explants by ELISA.
  • CD47 and SIRP ⁇ appear as important molecules in the initiation and perpetuation of SIRP- ⁇ +CD103- DC-driven Th2 immunity. Thus they may therefore be harnessed therapeutically to reduce lung inflammation and ameliorate airway disease.
  • SIRP ⁇ -Fc Fc region of human IgGI
  • T ⁇ nitrobenzene sulfonic acid (2 or 3 mg) was dissolved in 50% ethanol and instffled into the colons of male Bato/c mice (WT and CD47 KO) via a 3.5F catheter. Control mice were given ethanol atone. Mice were weighed every 24 hours and sacrificed on day 2 (early time point) or day 4. In the chronic TNBS colitis model, 1.5 mg of TNBS was instHted intrarectal on day 0 and again on day 7, and mice were sacrificed on day 12. Serum, mesenteric lymph nodes and colons were harvested for further analysis.
  • Colons were scored macro ⁇ copteaJfy using the Wallace criteria which takes into account the presence of diarrhea, adhesions, thickening of the bowel wall and ulceration. They were also evaluated for microscopic markers of inflammation using the Ameho criteria, a scoring system based upon thickening of the subm ⁇ co ⁇ a, infiltration of the submucosa and lamina intestinal with mononuclear ceHs, mucous depletion, loss of crypt architecture, and edema (not shown).
  • a recombinant mouse SIRP ⁇ -Fc fusion protein was administered i ⁇ traperitoneatty (100 ug/mouse) just prior to TNBS colitis induction and 24 hours thereafter. Control mice received saline alone. Injection of murine SIRP ⁇ -Fc 100 ⁇ g/animal on day 0, 30 min before TNBS induction and on day 1 blocked statistically significantly the disease development as assessed by bodyweight toss.
  • Solution A and solution B were injected as emulsion into naTve male DBA/1 mice. The mice are anaesthetized by s.c. injection of a sterile filtrated mixture of k ⁇ tamine. Upon narcosis, the root of the tail of each mouse is shaved and subsequently. 0.1 ml of the collagen-emulsion per mouse (containing 100 ⁇ g of collagen) is injected i.d. into the base of the tail.

Abstract

The present invention relates to soluble CD47 binding polypeptides, for use as a medicament, in particular for the prevention or treatment of autoimmune and inflammatory disorders, for example allergic asthma and inflammatory bowel diseases. The Invention more specifically relates to a soluble CD47 binding polypeptide for use as a medicament, comprising an extracellular domain of SIRPα (CD172a) or functional derivatives which bind to human CD47.

Description

SOLUBLE POLYPEPTIDES FOR USE IN TREATING AUTOIMMUNE AND INFLAMMATORY DISORDERS
The present Invention relates to soluble CD47 binding polypeptides, for use as a medicament, in particular for the prevention or treatment of autoimmune and inflammatory disorders, for example anergic asthma and inflammatory bowel diseases. The invention more specifically relates to a soluble CD47 binding polypeptide for use as a medicament, comprising an extracellular domain of SIRPα (CD172a) or functional derivatives which bind to human CD47.
CD47 is a cell surface glycoprotein which binds to SIRPα (alias SHPS-1) and SIRPγ on opposing cells. This interaction leads to negative regulation of immune celt function or can serve to mediating cellular adhesion and migration. CD47 was suggested for use as a biologies in the treatment of autoimmune disorders (WO199Θ/040940). In contrast, there are very few evidence of a potential use of CD47 tigands, such as SIRPα for similar therapeutic purposes. One explanation is the ubiquitous expression of CD47 that would prevent from using CD47 binding polypeptides as potential drugs. Data shown by Yu βt at 2006 (J Invest Dermatol, 126, 797-807) suggest that a fusion protein made from the extracellular domains of SIRPα fused to an immunoglobulin Fc domain can prevent the migration from skin derived dendritic cβfls (DCs) to draining lymph nodes in mice and thereby attenuate (at least partially) contact hypersensitivity response in mice. Migration and function of DCs is essential for immune or inflammatory responses. Under disease condition these exacerbated responses of DCs can lead to the perpetuation of disease. Interfering with migration of pathogenic DCs from tissue to lymphoid organs would be an attractive opportunity to stop the vicious cycle driving autoimmune- or inflammatory diseases. The present invention provides first in vivo evidence that SIRPα-Fc construct is suitable for preventing or stopping Th1/Th17- and Th2-driven diseases in animal models of disease. These data provide the basis for this invention and support the drugabϋity of StRPα-derived protein therapeutics. The invention is based, in part, on the discovery that the manipulation of CD47/SIRPα pathway suppresses immunogenic CD103* dendritic cells-driven pathogenesis of Th1røi17- (arthritis and colitis) as well as Th2 driven diseases (allergic asthma). These new findings offer previously unknown common mechanism on underlying causes of disease and represent therapeutic perspectives for multiple autoimmune and inflammatory disorders. In addition, recent evidence in published reports indicate that Kgation of CD47 could be beneficial for treatment of several cancers (Majeti et al CeN 2009). WNIe the reports indicate the use of CD47 antibodies, the Invention here relates to the use of SIRPα-derived polypeptides for treatment of these diseases.
Therefore, in one aspect, the invention provides soluble CO47 binding polypeptides, for use as a medicament, comprising a SIRPα-dβrived polypeptide selected among the group consisting of a) an extracellular domain of SIRPα (SEQ ID NO:3); b) a fragment of SEQ ID NO 1 , and, c) a variant polypeptide of SEQ ID NO.1 having at least 75% identity to SEQ ID NO:3; wherein said SIRPα-derived polypeptide binds to human CD47 (SEQ ID NO:24) In certain embodiments, the variant polypeptide of SEQ ID NO:3 is at least 80%, 85%, 90%, 9$% or 99% identical to SEQ ID NO:3.
For ease of reading, the soluble CD47 binding polypeptides according to the present invention are hereafter designated as the "Soluble Polypeptides of the Invention".
In one embodiment, said SIRPα-derived polypeptide is selected among antagonist of CD47, i.e., a polypeptide that competitively inhibits the binding of a CD47 Kgand to CD47. CD47 Uganda include, without limitation, SIRPα, SIRPγ or TSP1.
In another embodiment, said SIRPα-derived polypeptide is selected among agonist of CD47, i.e., a polypeptide that is capable of inducing CD47 signaling activity In one embodiment, said soluble CD47 binding polypeptide is selected among those that bind to human CD47 with a Ko of 2μM or less and/or inhibits induced cytokine secretion as measured in an immune complex-stimulated dendritic cell cytokine release assay.
In another embodiment, said SIRPα-derived polypeptide is an extracellular domain of SIRPα comprising at least the V-region of SIRPα (SEQ ID NO:2).
In certain embodiments, the Soluble Polypeptide of the Invention is a fusion polypeptide comprising a first component consisting of a SIRPα-derived polypeptide fused to a second heterologous polypeptide. In one embodiment, the Soluble Polypeptide further comprises a spacer between the second heterologous polypeptide and the SIRPα-derived polypeptide. In one specific embodiment, the SIRPα derived polypeptide is fused to an IgO Fc domain. In a preferred embodiment, said Fc domain is a silent Fc fragment of human IgGI isotype. In one embodiment, said Fc domain is an aglycosylated mutant variant of human IgGI isotype. In another related embodiment, the Soluble Polypeptides of the Invention are used aβ drugs m the treatment of autoimmune and inflammatory disorders. Preferred indications are selected among the group consisting of Th2~mediated airway inflammation, anergic disorders, asthma, inflammatory bowel diseases and ischemic disorders. In addition, the Soluble Polypeptides of the Invention may be used as drugs in the treatment of leukemias or cancer.
In order that the present invention may be more readily understood, certain terms are first defined. Additional definitions are set forth throughout the detailed description.
The term CO47 refers to human CD47. Human CD47 includes SEQ ID NO:24 but also any natural polymorphic, for example, comprising single nucleotide polymorphisms (SNPs), or splice variants of human CO47. Examples of splice variants or SNPs in CO47 nucleotide sequence found in human are described in TaWe 1.
Figure imgf000004_0001
The term SIRPα refers to the Signal Regulatory Protein Alpha (also designated CD172a or SHPS- 1) which shows adhesion to CD47 integrin associated protein. In some embodiment, the term SIRPα refers to human SIRPα as defined in SEQ ID NO.23. Human SIRPα contains an amino acids extracellular domain (SEQ ID NO:3), with one V-type domain (SEQ ID NO.2), and two C 1 -type Ig domains and three potential N-glycosylation sites. It has a 110 amino acids cytoplasmic sequence with ITIM motifs that recruit tyrosine phosphatases SHP- 1 and SHP-2 when phosphorylated. The term human SIRPα further includes, without limitation, any natural polymorphic, for example, comprising single nucleotide polymorphisms (SNPs), or splice variants of human SIRPα. Examples of splice variants or SNPs in SIRPα nucleotide sequence found in human are described in Table 2.
Figure imgf000005_0001
As used herein, a polypeptide is "soluble" when K lacks any transmembrane domain or protein domain that anchors or integrates the polypeptide into the membrane of a cell expressing such polypeptide. In particular, the Soluble Polypeptides of the Invention may likewise exclude transmembrane and Intracellular domains of SIRPα. Ae used herein, a polypeptide that "binds to CD47" is intended to refer to a polypeptide that binds to human CD47 with a K0 of a 20μM or less, 2μM or less, 0.2μM or less. In some embodiment, a polypeptide that binds to CD47 further binds to surfactant protein A (SP-A) and/or surfactant protein O (SP-O).
As used herein, a polypeptide that inhibits induced cytokine secretion as measured in a immune complex-stimulated dendritic cell cytokine release assay is a polypeptide that inhibits cytokine (e.g. IL-β, IL-10, IL-12p70, IL-23, IL-8 and/or TNF-α) release from peripheral blood monocytes, conventional dendritic ceϋs (DCs) as weH as monocyte-derived DCs stimulated with Staphylococcus Aureus Cowan 1 (Pansorbin) or soluble CD40L and IFN- γ. One example of an immune complex-stimulated dendritic oeH cytokine release assay is described in more details in the examples below. In some embodiments, the Soluble Polypeptides of the invention inhibit cytokine secretion as measured in an immune complex- stimulated dendritic cβU cytokine release assay at an IC90 of 1μM or less, 10OnM or less, or 1OnM or less.
As used herein, a polypeptide that inhibits T cefl proliferation may be measured in a mixed lymphocyte reaction assay as described in the Example.
The term "KMMC" or "K,", as used herein, is intended to refer to the association rate of a particular protein-protein interaction, whereas the term "K-," or "K*" as used herein, is intended to refer to the dissociation rate of a particular protein-protein interaction. The term "K0", as used herein, is intended to refer to the dissociation constant, which is obtained from the ratio of K4 to K« (i.e. K4ZK,) and is expressed as a molar concentration (M). K0 values for protein-protein interaction can be determined using methods well established in the art. A method for determining the K0 of an protein/protein interaction is by using surface plasmon resonance, or using a biosensor system such as a Biacore* system.
As used herein, the term "Affinity" refers to the strength of interaction between the polypeptide and its target at a single site. Within each site, the binding region of the polypeptide interacts through weak non-covalent forces with its target at numerous sites; the more interactions, the stronger the affinity.
As used herein, the term "high affinity" for a binding polypeptide refers to a polypeptide having a K0 of 1OnM or less, for example, 1nM or less, for its target.
As used herein, the term "subject" includes any human or nonhuman animal. Tbe term "nonhuman animal" includes ait vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc.
As used herein, the term, "optimized" means that a nucleotide sequence has been altered to encode an amino acid sequence using codons that are preferred in the production cell or organism, either a eukaryotic ceil, for example, a cell of Pichia or Saocharomyces, a cell of Trichodβrma, a Chinese Hamster Ovary ceo (CHO) or a human ceB, or a prokaryotic ceβ, for example, a strain of Escherichia coli.
The optimized nucleotide sequence is engineered to retain completely or as much as possible the amino acid sequence originatty encoded by the starting nucleotide sequence, which is also known as the "parental" sequence. The optimized sequences herein have been engineered to have codons that are preferred in the corresponding production cell or organism, for example a mammalian cell, however optimized expression of these sequences in other prokaryotic or eukaryotic cβflβ is also envisioned herein. The amino acid sequences encoded by optimized nucleotide sequences are also referred to as optimized.
Various aspects of the invention are described in further detail in the following subsections.
Assays to evaluate the effects of the Soluble Polypeptides of the Invention on functional properties of CD47 are described in further detail in the Examples.
SIRPα-derived polypeptides
Soluble Polypeptides of the Invention comprise SIRPα-derived polypeptides that are selected among the group consisting of a) an extracellular domain of SIRPα (SEQ ID NO:3); b) a fragment of SEQ ID NO: 3, and, c) a variant polypeptide of SEQ ID NO:3; wherein said SIRPα-derived polypeptide binds to human CD47 (SEQ ID NO.24).
The Soluble Polypeptides of the Invention and their SIRPα-derived fragments should retain the capacity to bind to CD47. Fragments of SEQ ID NO:3 can therefore be selected among those fragments comprising the CD47 binding domain of SIRPα. Those fragments generally do not comprise the transmembrane and intracellular domains of SIRPα. In non-limiting illustrative embodiments, SIRPα-derived polypeptide essentially consists of SEQ ID NO:3 or SEQ ID NO.2. SIRPα-derived polypeptides further include, without limitation, variant polypeptides of SEQ ID NO.3 where amino acids residues have been mutated by amino acid dβtetton, insertion or substitution, yet have at least 60, 70, 80, 90 or 95 percent identity to SEQ ID NO: 3, so long as changes to the native sequence do not substantially affect the biological activity of the molecule, in particular its binding to CD47. In some embodiments, it include mutant amino acid sequences wherein no more than 1, 2, 3, 4 or 5 amino acids have been mutated by amino acid deletion, insertion or substitution in the SIRPα-derived polypeptide when compared with SEQ ID NO:2. Examples of mutant ammo acid sequences are those sequences derived from single nucleotide polymorphisms (see Table 2).
As used herein, the percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i. e., % identity = # of identical positions/total # of position* x 100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described below.
The percent identity between two amino acid sequences can be determined using the algorithm of E. Meyers and W. Miller (Comput. Appi. Biosci., 4:11-17, 1988) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. In addition, the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. MoI, Biol. 48:444-453, 1970) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, β. or 4 and a length weight of 1, 2, 3, 4, 5, or 6.
In a specific embodiment the SIRPα-derived polypeptide includes changes to SEQ ID NO.3 or SEQ ID NO.2 that contain conservative amino acid substitutions.
Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginme, histidine), acidic side chains (e.g., aspartJc acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, gkitamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoteυcine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g.. tyrosine, phenylalanine, tryptophan, hiβtktoiβ). Thus, one or more amino acid residues within the CD47 binding region of SIRPα- derjved polypeptide can be reptaoed with other amino add residues from the same side chain family, and the new polypeptide variant can be tested for retained function using the binding or functional assays described herein.
In some embodiments, the SIRPα-derived polypeptides Are selected among those that retain the capacity to inhftπt cytokine secretion as measured in a immune complex-stimulated dendritic cei cytokine release assay at least to the same extent as the polypeptide of SEQ IO NO: 3 comprising the extracellular domain of human SIRPα.
In some embodiments, the SIRPα-derived polypeptides are selected among those that retain the capacity to inhibit T cell proliferation as measured in a mixed lymphocyte reaction assay.
In another embodiment, the SIRPα-derived polypeptides are selected among those that cross-react with non-human primate CD47.
Fusion polypeptides
In one aspect, the Soluble Polypeptides of the Invention are fusion polypeptides comprising the SIRPα-derived polypeptides.
In a preferred embodiment, the Soluble Polypeptides of the Invention are fusion polypeptides comprising the SIRPα-derived polypeptides and a second heterologous amino acid sequence, e.g. a portion of one or more proteins other than SIRPα, covatentfy bound to the SIRPα-derivβd polypeptide at the letter's N- and/or C-terminus, and optionally further comprising a linker.
The non SIRPu-derived protein can be preferably a soluble single chain polypeptide, which, when fused to another heterologous protein, is capable of increasing half life of the resulting fusion protein in blood. Alternatively or in addition, the non-SIRPα-derived protein comprises a domain for multimerization of the fusion polypeptide.
The non SIRPα-derived protein can be, for example, an immunoglobulin, serum albumin and fragments thereof. The non SIRPα-derived protein can also be a polypeptide capable of binding to serum albumin proteins to increase half life of the resulting molecule when administered in a subject Such approach is for example described in Nygren et a!., EP 0 488 525. In one specific embodiment, the non-SIRPα derived protein is an Fc domain. The use of Fc moiety for making soluble construct with increased in vivo half life in human is weJ! known in the art and for example described in Capon et al (US5.428.130).
As used herein the term "Fc domain" refers to the constant region of an immunoglobulin. An Fc domain comprises at least the CH2 and CH3 domain, optionally, the hinge region which is located between the heavy chain CH1 domain and CH2. Fc fragments could be obtained for example by papain digestion of an immunoglobulin. As used herein, the term Fc domain further include Fc variants into which a substitution, deletion or insertion of at least one amino acid has been introduced.
In one embodiment, the hinge region of CW is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Patent No. 5,677,425 by Bodmer et al. The number of cysteine residues m the hinge region of CH1 is altered to, for example, facHitate assembly of the light and heavy chains or to increase or decrease the stabΦty of the fusion polypeptide. In another embodiment, the Fc region is modified to increase its biological half-life. Various approaches are possible. For example, one or more of the following mutations can be introduced: T252L, T254S, T256F, as described in U.S. Patent No. 6,277,375 to Ward. In yet other embodiments, the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector functions of the Fc portion. For example, one or more amino adds can be replaced with a different amino acid residue such that the Fc portion has an altered affinity for an effector ligand. The effector Rgand to which affinity » altered can be, for example, an Fc receptor or the C 1 component of complement. This approach is described in further detail in U.S. Patent Nos. 5,624,821 and 5,648,260, both by Winter et al. In another embodiment, one or more amino acids selected from amino acid residues can be replaced with a different amino acid residue such that the resulting Fc portion has altered C1q binding and/or reduced or abolished complement dependent cytotoxicity (CDC). This approach is described in further detail in U.S. Patent Nos. 6,194,551 by ldusogie et al.
In another embodiment, one or more amino acid residues are altered to thereby alter the ability of the Fc region to fix complement. This approach is described further in PCT Publication WO 94/29351 by Bodmer et al. In yet another embodiment, the Fc region is modified to increase the ability of the fusion polypeptide to mediate antibody dependent ceflular cytotoxicity (ADCC) and/or to increase or decrease the affinity of the Fc region for an Fey receptor by modifying one or more amino acids. This approach is described further in PCT Publication WO 00742072 by Presta Moreover, the binding sites on human IgGI for FcγRI, FcyRil, FcyRIII and FcRn have been mapped and variants with improved binding have been described (see Shields, R L. et al.. 2001 J. Biol. Chβm. 276:6591-6604).
In one embodiment, the Fc domain is of human origin and may be from any of the tmmunoglobuβn classes, such as IgG or IgA and from any subtype such as human IgGI , lgG2, igG3 and igG4. In other embodiments the Fc domain is from a non-human animal, for example, but not limited to, a mouse, rat, rabbit, camel, shark, non-human primate or hamster.
In certain embodiments, the Fc domain of IgGI isotype is used. In some specific embodiments, a mutant variant of IgGI Fc fragment is used, e.g. a stent IgGI Fc which reduces or eliminates the ability of the fusion polypeptide to mediate antibody dependent ceflular cytotoxicity (AOCC) and/or to bind to an Fey receptor. An example of an IgGI isotype silent mutant, is a so-caSβd LALA mutant, wherein Leucine residue is replaced by Alanine residue at amino acid positions 234 and 235 as described in J. Virol 2001 Dβc;75<24):i2iβi-8 by Hezareh et al. In certain embodiments, the Fc domain is a mutant preventing glycosyfation at residue at position 297 of Fc domain. For example, an amino acid substitution of asparagtne residue at position 297 of the Fc domain. Example of such amino acid substitution is the replacement of N297 by a glycine or an alanine.
In one embodiment, the Fc domain comprises a cβmerization domain, preferably via cysteine capable of making covalent disulfide bridge between two fusion polypeptides comprising such Fc domain.
The SIRPα-derived polypeptide can be fused directly in frame with the non-SIRPα-derived protein or via a polypepϋdic linker (spacer). Such spacer may be a single amino acid (such as for example, a glycine residue) or between 5-100 amino acids, for example between 5-20 amino acids. The linker should permit the SIRPα-derived domain to assume the proper spatial orientation to form a binding site with CD47. Suitable polypeptide tinkers may be selected among those that adopt a flexible conformation. Examples of such linkers are (without limitation) those linkers comprising Glycine and Serine residues, for example, (GIy4SeT)n wherein n=1-12
Glycosylate modifications
In still another embodiment, the glycosylate pattern of the Soluble Polypeptide of the Invention can be altered compared to typical mammalian glycosylate pattern such as those obtained in CHO or human cefl lines. For example, an agiycosiated polypeptides can be made by using prokaryotlc cefl Hnes as host ceβs or mammalian cells that has been engineered to lack glycosylate. Carbohydrate modifications can also be accomplished by; for example, altering one or more sites of glycosylate within the Soluble Polypeptide.
Additionally or alternatively, a glycosylated polypeptide can be made that has an altered type of glycosylate. Such carbohydrate modifications can be accomplished by, for example, expressing the Soluble Polypeptides in a host cell with altered glycosylate machinery, Le the glycosylate pattern of the Soluble Polypeptide is altered compared to the glycosylate pattern observed in corresponding wild type ceβs. Cells with altered glycosylate machinery have been described in the art and can be used as host cells in which to express recombinant Soluble Polypeptides of the Invention to thereby produce such Soluble Polypeptides with altered glycosylate. For example, EP 1 ,176,195 by Hang et al. describes a cell Nne with a functionary disrupted FUT8 gene, which encodes a fucosyt transferase, such that glycoproteins expressed in such a cell Bne exhtott hypofucosytation. PCT Publication WO 03/035835 by Presta describes a variant CHO cell Kne, Led3 cells, with reduced ability to attach fucose to Asn(297Htnked carbohydrates, also resulting in hypofucoβytation of glycoproteins expressed in that host cefl (see also Shields, R.I. et al., 2002 J. Biol. Chem. 277.26733-26740). Alternatively, the Soluble Polypeptides of the Invente can be produced in yeast, e.g., Pichiβ pastoas, or filamentous fungi, e.g., Trichodβrma f&esβi engineered for mammalian-like glycosylate pattern (see for example EP129717261). Advantages of those giycoengineered host celts are, inter alia, the previse of polypeptide composites with homogeneous glycosylate pattern and/or higher yield.
Peoylated Soluble Polypeptides and other conjugates
Another embodiment of the Soluble Polypeptides herein that is contemplated by the invention is pegylation. The Soluble Polypeptides of the invente, for example, consisting essentially of SIRPα-derived polypeptides can be pegyiated. Pegylatton is a well-known technology to increase the biological (e.g.. serum) half-life of the resulting biologies as compared to the same biologies without pegylation. To pegyiatβ a polypeptide, the polypeptide is typically reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the polypeptide*. The pβgytation can be carried out by an acylatkm reaction or an afeyJatJon reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer). As used herein, the term "polyethylene glycol" is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (C1-C10) aikoxy- or arytoxy-polyethylene glycol or polyethylene glycol-maleimide. Methods for pegyteting proteins are known in the art and can be applied to the Soluble Polypeptides of the invention. See for example, EP 0 154 316 by NterAnura et aJ. and EP 0 401 384 by IsNkawa et a!.
Alternative conjugates or polymeric carrier can be used, in particular to improve the pharmacokinetic properties of the resulting conjugates. The polymeric carrier may comprise at least one natural or synthetic branched, finear or dendritic polymer. The polymeric carrier is preferably soluble in water and body fluids and is preferably a pharmaceutically acceptable polymer. Water soluble polymer moieties include, but are not limited to, e.g. potyβlkyiene glycol and derivatives thereof, including PEG, PEG homopoJymβrβ, mPEG, potypropytenegtycol homopoJyrnerβ, copolymers of ethylene glycol with propylene glycol, wherein said homopolymers and copolymers are unsubstituted or substituted at one end e.g. with an acykjroup; polygiycerines or polysiaϋc acid; carbohydrates, polysaccharides, cellulose and cellulose derivatives, including methylceNulose and carboxymethylceHuloβe; starches (e.g. hydroxyalkyi starch (HAS), especially hydroxyethyl starch (HES) and dextrinβs, and derivatives thereof; dextran and dextran derivatives, including dβxtransutfat, crosβttnked dextrin, and carboxymethyl dextrin; chitosan (a linear polysaccharide), heparin and fragments of heparin; polyvinyl alcohol and polyvinyl ethyl ethers; polyvinylpyrrollidon; alpha, beta-poM{2-hydroxyethyl)-OL-aspaftamide; and polyoxy- ethyfated pofyote.
Mae of the Soluble Polypeptides as a medicament
The Soluble Polypeptides of the Invention have been shown to protect from inflammatory disorders such as allergic asthma or inflammatory bowel diseases in animal model and therefore may be used as a medicament, in particular to decrease or suppress (in a statistically or biologically significant manner) the Inflammatory and/or autoimmune response, in particular, a response mediated by SIRPa+ cells in a subject. Nucleic acid molecules encoding the Soluble Polypeptides of the Invention
Another aspect of the invention pertains to nucleic acid molecules that encode the Soluble Polypeptides of the Invention or at least SIRPα-derived polypeptides, including without limitation, the embodiments related to fusion polypeptides. Examples of nucleotide sequences encoding Soluble Polypeptides of the Invention comprise SEQ ID NOs: 26 or 27.
The nucleic acids may be present in whole cells, in a ceU lysate, or may be nucleic acids in a partially purified or substantially pure form. A nucleic add is Isolated" or "rendered substantially pure" when purified away from other cellular components or other contaminants, e.g., other ceHυlar nucleic adds or proteins, by standard techniques, induding aHcaline/SDS treatment, CsCI banding, column chromatography, agarose gel electrophoresis and others well known in the art. See, F. Ausubel, et al., ed. 1087 Current Protocols in Molecular Biology, Greene Publishing and WHey Interβciencβ, New York. A nucleic acid of the invention can be, for example, DNA or RNA and may or may not contain kitronic sequences. In an embodiment, the nucleic acid is a cDNA molecule. The nucleic acid may be present in a vector such as a phage display vector, or in a recombinant plasmid vector.
Once DNA fragments encoding the Soluble Polypeptides of the Invention are obtained, for example, fusion polypeptide comprising the SIRPα-derived polypeptides as described above, these DNA fragments can be further manipulated by standard recombinant DNA techniques, for example to include any signal sequence for appropriate secretion in expression system, any purification tag and cleavable tag for further purification steps. In these manipulations, a DNA fragment is operative^ finked to another DNA molecule, or to a fragment encoding another protein, such as a purification/secretion tag or a flexible linker. The term "operatively linked", as used in this context, is intended to mean that the two DNA fragments are joined in a functional manner, for example, such that the amino acid sequences encoded by the two DNA fragments remain in-frame, or such that the protein is expressed under control of a desired promoter.
Generation of transfectomas producing the SIRPα-derived polypeptide or Soluble Polypeptides
The Soluble Polypeptides of the Invention can be produced in a host cell transfectoma using, for example, a combination of recombinant DNA techniques and gene transfection methods as is well known in the art. For example, to express the Soluble Polypeptides of Invention or intermecKates thereof, such as SIRPα-dβrived polypeptides. DNAs encoding the corresponding polypeptides, can be obtained by standard molecular biology techniques (e.g., PCR amplification or cONA cloning using a hybridoma that expresses the polypeptides of interest) and the ONAs can be inserted into expression vectors such that the corresponding gene is operattvβty linked to transcriptional and transiational control sequences. In this context, the term "operatively linked" is intended to mean that a gene » Hgated into a vector such that transcriptional and transiational control sequences within the vector serve their intended function of regulating the transcription and translation of the gene. The expression vector and expression control sequences are chosen to be compatible with the expression host cβH used. The gene encoding Soluble Polypeptides or intermediates are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction sites on the gene fragment and vector, or blunt end ligation if no restriction sites are present). AdcHtionaDy or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the polypeptide chain from a host cell. The gene can be cloned into the vector such that the signal peptide is linked in frame to the amino terminus of the polypeptide chain. The signal peptide can be a SIRPα signal peptide or a heterologous signal peptide (i.e., a signal peptide not naturally associated with SIRPα sequence).
In addition to the polypeptide encoding sequence, the recombinant expression vectors of the invention carry regulatory sequences that control the expression of the gene in a host cell. The term "regulatory sequence* is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the polypeptide chain genes. Such regulatory sequences are described, for example, in Goeddβl (Gene Expression Technology. Methods in Enzymology 185, Academic Press. San Diego, CA 1990). It wtø be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences, may depend on such factors as the choice of the host ceβ to be transformed, the level of expression of protein desired, etc. Regulatory sequences for mammalian host ceil expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV), Simian Virus 40 (SV40), adenovirus (e.g., the adenovirus major late promoter (AdMLP)), and polyoma. Alternatively, nonviral regulatory sequences may be used, such as the ubiquitin promoter or P-globin promoter. Still further, regulatory elements composed of sequences from different sources, such as the SRa promoter system, which contains sequences from the SV40 early promoter and the long terminal repeat of human T cefl leukemia virus type 1 (Takebe, Y. βt al., 1988 MoI. CeN. Bid . 8:466-472) In addition to this, the recombinant expression vectors of the invention may carry additional sequences, such as sequence* that regulate replication of the vector in host cefiβ (e.g., origins of replication) and selectable marker genes. The selectable marker gene facβitates selection of host cells into which the vector has been introduced (see, e.g., U.S. Pat Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et aJ ). For example, typicaβy the selectable marker gene confers resistance to drugs, such as G418, hygromyctn or methotrexate, on a host cell into which the vector has been introduced. Selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr- host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
For expression of the polypeptide, the expression vectors) encoding the Soluble Polypeptide or intermediates such as SIRPα-derived polypeptide is transfectβd into a host celt by standard techniques. The various forms of the term "tranβfection" are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., etectroporation, cakϋum-phoβphate precipitation, DEAE-dextran transection and the like. It is theoretically possible to express the Soluble Polypeptides of the Invention in either prokaryotic or eukaryotic host ceββ. Expression of glycoprotein in eukaryotic ceils, in particular mammalian host cells, is discussed because such eukaryotic cefls, and in particular mammalian ceBs, are more likely than prokaryotic cells to assemble and secrete a properly folded and biologically active glycoprotein such as the Soluble Polypeptides of the Invention.
Mammalian host cells for expressing the Soluble Polypeptides and intermediates such as SIRPα-derived polypeptides of the invention include Chinese Hamster Ovary (CHO eels) (including dhfr- CHO cβHs, described Urlaub and Chastn, 1980 Proc. Natl. Acad. Sd. USA 77:4216-4220 used with a DH FR selectable marker, e.g., as described in RJ. Kaufman and PA Sharp, 1982 MoI. Biol. 159:601-621. NSO myeloma cells, COS cells and SP2 cells) or human ceB lines (including PER-Cβ cell Hoes, Crucell). In particular, for use with NSO myeloma cells, another expression system is the GS gene expression system shown in WO 87/04462, WO 89/01036 and EP 338,841. When recombinant expression vectors encoding polypeptides are introduced into mammalian host cells, the Soluble Polypeptides or intermediates such as SIRPα-derived polypeptides are produced by cuJturing the host cells for a period of time sufficient to aflow for expression of the recombinant polypeptides in the host cells or secretion of the recombinant polypeptides into the culture medium in which the host cettβ are grown. The polypeptides can then be recovered from the culture medium using standard protein purification methods.
Multivalent proteins
In another aspect, the present invention provides multivalent proteins comprising at least two identical or different Soluble Polypeptides of the Invention binding to CD47. In one embodiment the multivalent proteins comprise at least two, three or four Soluble Polypeptides of the Invention. The Soluble CD47-binding Polypeptides can be linked together via protein fusion or covalent or non covalent linkage. The multivalent proteins of the present invention can be prepared by conjugating the constituent binding specificities, using methods known in the art. For example, each binding specificity of the multivalent protein can be generated separately and then conjugated to one another.
A variety of coupling or cross-unking agents can be used for covalent conjugation. Examples of cross-linking agents include protein A, carbodtønide, N-suα^midyl-S-acβtyMhioacβtatβ (SATA), 5.5'-dithiob<s(2-nrtrobβnzo»c acid) (OTNB). o-phenylenedimaleimide (oPDM), N- suo^nfmidyi-3^2-pyridyldithk>)propionate (SPDP)1 and suifosuccinlmidyl 4-(N- maleimidomethyl) cyctohaxanβ-l-carboxytøte (suffo-SMCC) (see e.g., Karpovsky et al., 1984 J. Exp. Med. 160:1686; Uu, MA et al., 1985 Proc. Natl. Acad. Sd. USA 82:8648). Other methods include those described in PauJus, 1985 Behring Ins. Mitt. No. 78.118-132; Brennan et al., 1985 Science 229:81-83). and Glennie et al., 1987 J. Immunol. 139: 2367-2375). Covalent Hnkage can be obtained by disulfide bridge between two cysteines, for example disulfide bridge from cysteine of an Fc domain.
In a particular embodiment, the hinge region of an Fc domain fused to SIRPα-derived polypeptide is modified to contain an odd number of sulfhydryl residues, for example one, prior to conjugation. Alternatively, both binding specificities can be encoded in the same vector and expressed and assembled in the same host cell.
Pharmaceutical compositions
In another aspect, the present invention provides a composition, e.g., a pharmaceutical composition, containing one or a combination of the Soluble Polypeptides of the present Invention, formulated together with a pharmaceutically acceptable carrier. Pharmaceutica) formulations comprising a Soluble Polypeptide of the invention may be prepared for storage by mixing the polypeptide having the desired degree of purity with optional physiotogicaHy acceptable carriers, exάpients or stabilizers {Remington: The Science and Practice of Pharmacy 20th edition (2000)), in the form of aqueous solutions, lyophiMzed or other dried formulations. Therefore, the invention further relates to a lyophized composition comprising at least the Soluble Polypeptide of the invention.
Pharmaceutical compositions of the invention also can be administered in combination therapy, i.e., combined with other agents. For example, the combination therapy can include a Soluble Polypeptide of the present Invention combined with at least one other antiinflammatory or another chemotherapeutic agent. Examples of therapeutic agents that can be used in combination therapy are described in greater detail below in the section on uses of the Soluble Polypeptides of the Invention.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the Ice that are physiologically compatible. The carrier should be suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion). Depending on the route of administration, the active compound may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
The pharmaceutical compounds of the invention may include one or more pharmaceutically acceptable salts. A "pharmaceutically acceptable salt" refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxfcotogicai effects (see e.g., Berge, S.M., et at , 1977 J. Pharm. Sd. 66.1-19). Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydrokκfic, phosphorous and the Wee, as well as from nontoxic organic acids such as alphatic mono* and dt-carboxytic acids, phenyl-substituted a&anoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like. Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as N1N*- dibenzylethyfenediamine, N-methyiglucamine, chloroprocaine, choline, diethanoiamine, ethyienediamine, procaine and the like. A pharmaceutical composition of the Invention also may Include a pharmacβuticaly acceptable anti-oxktam Examples of pharmaceuticaβy acceptable antioxidants include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisutfate, sodium metabisuJfite, sodium sulfite and the Mke; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butyiated hydroxytoluene (BHT), lecithin, propyl gaHate, alpha-tocopherol, and the like; and metal chelating agents, such as citric add, ethytenediamine tetraacetic acid (EDTA), sorbitol, tartaric add, phosphoric acid, and the Bke.
Examples of suitable aqueous and nonaqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyote (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as ofive ofl, and injectable organic esters, such as ethyl oieate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures, supra, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, cntorobutanol, phenol sorbic acid, and the Bee. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as, aluminum monostearate and gelatin.
Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions.
Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyoJ (for example, glycerol, propylene glycol, and Squid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, one can include isotonic agents, for example, sugars, potyaJcohols such as mannitol. sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including In the composition an agent that delays absorption for example, monostearate salts and gelatin. Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, foβowed by sterilization microfUtration. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, and the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect. Generally, out of one hundred percent, this amount wHI range from about 0.01 per cent to about ninety-nine percent of active ingredient, from about 0.1 per cent to about 70 per cent, or from about 1 percent to about 30 percent of active ingredient in combination with a pharmaceutically acceptable carrier.
Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the requirβd pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuate.
For administration of the Soluble Polypeptides of the Invention, the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight. For example dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg. An exemplary treatment regime entails administration once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months or once every three to β months. Dosage regimens for a Soluble Polypeptide of the Invention include 1 mg/kg body weight or 3 mg/kg body weight by intravenous administration, with the polypeptide being given using one of the following doting schedules: every four weeks for six dosages, then every three months; every three weeks; 3 mg/kg body weight once followed by 1 mg/kg body weight every three weeks.
The Soluble Polypeptide is usually administered on multiple occasions. Intervals between single dosages can be, for example, weekly, monthly, every three months or yearly. Intervals can also be irregular as indicated by measuring blood levels of Soluble Polypeptide in the patient. In some methods, dosage is adjusted to achieve a plasma polypeptide concentration of about 1-1000 μg/ml and in some methods about 25-300 ug/ml.
Alternatively, the Soluble Polypeptide can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the Soluble Polypeptide in the patient. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated or until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level wi8 depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex. weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
A "thβrapeuticaty effective dosage" of Soluble Polypeptide of the Invention can result in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction.
A composition of the present invention can be administered by one or more routes of administration using one or more of a variety of methods known hi the art. As will be appreciated by the skilled artisan, the route and/or mode of administration wiH vary depending upon the desired results. Routes of administration for Soluble Polypeptides of the Invention include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase "parenteral administration'1 as used herein means modes of administration other than enteral and topical administration, usuaβy by injection, and includes, without limitation, intravenous, intramuscular, Intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrastemal injection and infusion.
Alternatively, a Soluble Polypeptide of the Invention can be administered by a nonparenteral route, such as a topical, epidermal or mucosal route of administration, for example, intranasalty, oratty, vaginally, rectaβy, sublingualy ortopicaβy.
The active compounds can be prepared with carriers that win protect the compound against rapid release, such as a controflβd release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, poryglycolic acid, collagen, poiyorthoesters, and poryiactic acid. Many methods for the preparation of such formulations are patented or generally known to those sktRed In the art. See. e.g., Sustained and Controlled Release Drug Delivery Systems, J R. Robinson, ed., Marcel Dekker, Inc., New York, 1Θ78.
Therapeutic compositions can be administered with medical devices known In the art. For example, Ni one embodiment, a therapeutic composition of the invention can be administered with a needleless hypodermic Injection device, such as the devices shown in U.S. Patent Noβ. 5,399.163; 5.383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824 or 4,596,556. Examples of wei known implants and modules useful in the present invention include: U.S. Patent No. 4,487,603, which shows an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Patent No. 4,486,194, which shows a therapeutic device for administering medicants through the skin; U.S. Patent No. 4,447,233, which shows a medication infusion pump for delivering medication at a precise infusion rate; U.S. Patent No. 4,447,224, which shows a variable flow implantable infusion apparatus for continuous drug delivery; U.S. Patent No. 4,439.196, which shows an osmotic drug delivery system raving multi-chamber compartments; and U.S. Patent Na 4,475,196, which shows an osmotic drug delivery system. Many other such implants, delivery systems, and modules are known to those skilled in the art.
In certain embodiments, the Soluble Polypeptides of the Invention can be formulated to ensure proper distribution hi vivo. For example, the blood-brain barrier (BBB) excludes many highly hydrophiHc compounds. To ensure that the therapeutic compounds of the invention cross the BBB (if desired}, they can be formulated, for example, in liposomes. For methods of manufacturing liposomes, see, e.g., U.S. Patents 4,522,811; 5,374,548; and 5,399,331. The liposomes may comprise one or more moieties which are selectively transported into specific cells or organs, thus enhance targeted drug delivery (see, e.g., V. V. Ranade, 1989 J. CHne Pharmacol. 29:685). Exemplary targeting moieties include folate or biotin (see, e.g., U.S. Patent 5,416,016 to Low et al.); mannosides (Umezawa et a!., 1988 Biochem. Biophys. Res. Commun. 153:1038); antibodies (P.G. Bioeman et al., 1995 FEBS Lett. 357:140; M. Owais et al., 1995 Antimicrob. Agents Chemother. 39:180); surfactant protein A receptor (Briscoe et al., 1995 Am. J. Physiol.1233: 134); pi20 (Schreier et a) , 1994 J. Biol. Chem. 269:9090); see also K. Keinanen; M.L Laukkanen, 1994 FEBSLett. 346:123; J J. Killton; IJ. Ftdler, 1994 lmmunomethods 4:273.
Uses and methods of the invention The Soluble Polypeptides of the Invention have in vitro and in vivo diagnostic and therapeutic utilities. For example, these molecules can be administered to cells in culture, e.g. in vitm or in vivo, or in a subject, e.g., in vivo, to treat, prevent or diagnose a variety of disorders.
The term "subject" as used herein is intended to include human and non-human animals. Non-human animals include aβ vertebrates, e.g., mammals and non-mammals, such as non- human primates, sheep, dogs, cats, cows, horses, chickens, amphibians, and reptiles.
The methods are particularly suitable for treating, preventing or diagnosing autoimmune and inflammatory disorders mediated by SIRPa+ cells, e.g., aβergic asthma or ulcerative colitis. These include Inflammatory conditions, allergies and allergic conditions, autoimmune diseases, ischemic disorders, severe infections, and ceHs, organ or tissue transplant rejection, including xenotransplant (i.e. transplant from different species, for example from non-human species to human) of cells, tissues or organs.
Examples of autoimmune diseases includes, without limitation, arthritis (for example rheumatoid arthritis, arthritis chronica progrediente and arthritis deformans) and rheumatic diseases, including inflammatory conditions and rheumatic diseases involving bone toss, inflammatory pain, spondyloarhropathies including ankylosing spondylitis, ReHer syndrome, reactive arthritis, psoriatic arthritis, and enterophathβ arthritis, hypersensitivity (Including both airways hypersensitivity and dermal hypersensitivity) and aiergies. Autoimmune diseases include autoimmune haematoiogicai disorders (including e.g. hemolytic anaemia, aplastic anaemia, pure red cell anaemia and idiopathic thrombocytopenia), systemic lupus erythematosus, inflammatory muscle disorders, polychondritis, scterodoma, Wegener granulomatosis, derrnatomyositis, chronic active hepatitis, myasthenia gravis, psoriasis. Steven-Johnson syndrome, idiopathic sprue, endocrine ophthalmopathy, Graves disease, sarcoidosis, multiple sclerosis, primary biliary cirrhosis, juvenile diabetes (diabetes mettitus type I), uveitis (anterior and posterior), keratoconjunctivitis sicca and vernal keratoconjunctivitis, interstitial lung fibrosis, psoriatic arthritis and glomerulonephritis (with and without nephrotic syndrome, e.g. including idiopathic nephrotic syndrome or minimal change nephropathy), tumors, multiple sclerosis, inflammatory disease of skin and cornea, myositis, loosening of bone implants, metabolic disorders, such as atherosclerosis, diabetes, and dislipidemia. Thβ Soluble Polypeptides of the Invention are also useful for the treatment, prevention, or amelioration of asthma, bronchitis, pneumoconiosis, pulmonary emphysema, and other obstructive or inflammatory diseases of the airways.
The Soluble Polypeptides of the Invention are also useful for the treatment of IgE-mediated disorders. IgE mediated disorders include atopic disorders, which are characterized by an inherited propensity to respond immunologicafly to many common naturally occurring inhaled and ingested antigens and the continual production of IgE antibodies. Specific atopic disorders includes aflergic asthma, allergic rhinitis, atopic dermatitis and allergic gastroenteropathy.
However, disorders associated with elevated IgE levels are not limited to those with an inherited (atopic) etiology. Other disorders associated with elevated IgE levels, that appear to be IgE-mediated and are treatable with the formulations of this present invention include hypersensitivity (e. g., anaphylactic hypersensitivity), eczema, urticaria, allergic bronchopulmonary aspergillosis, parasitic diseases, hyper-lgE syndrome, ataxia- telangiectasia, Wiskott-Aldrich syndrome, thymic atymphoplasia, IgE myeloma and graft- versus-host reaction.
The Soluble Polypeptides of the Invention may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to or in combination to, other drugs e.g. immunosuppressive or immunomodiiating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of diseases mentioned above. For example, the Soluble Polypeptides of the invention may be used in combination with DMARD, e.g. Gold salts, sulphasalazine, antimalariaβ, methotrexate, D-penidllamine, azathioprine, mycophenoϋc add, cydosporine A, tacrolimus, siroiimus, minocycline, leflunomide, glucocorticoids; a calcmeurin inhibitor, e.g. cyclosporin A or FK 506; a modulator of lymphocyte recirculation, e.g. FTY720 and FTY720 analogs; a mTOR inhibitor, e.g. rapamycin, 40-O-(2-hydroxyethyl)- rapamycin, CCI779, ABT578, AP23573 or TAFA-93; an ascomydn having immunosuppressive properties, e.g. ABT-281 , ASM981, etc.; corticosteroids; cyclo-phos-phamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenotic add; myco-pheno-late mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC1 CO2, CD3, CD4, CO7, CD8. CD25. CD28, CD40. CD45. CD58, CD80. CD86 or their ligands; other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTlMIg (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA-1 antagonists, ICAM- 1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g. pacHtaxe), gemcitabine, dspiatinum, doxorubicin or 5-fluorouracil; anti TNF agents, e.g. monoclonal antibodies to TNF, e.g. infliximab, ada&mimab, CDP870, or receptor constructs to TNF-RI or TNF-RII, e.g. Etanercept, PEG-TNF-RI; blockers of proinflammatory cytokines, . IL-1 blockers, e.g. Anakinra or IL- 1 trap, AAL160, ACZ 885, IL-6 blockers; chemokines blockers, e g inhibitors or activators of proteases, e.g. metaβoproteases, antt-IL-15 antibodies, antJ-IL-6 antibodies, anU-CD20 antibodies, anti-CD22 antibodies, antML17 antibodies, NSAIDs, such as aspirin or an artWnfectiouβ agent (list not limited to the agent mentioned).
The Soluble Polypeptides of the Invention are also useful as co-therapeutic agents for use in conjunction with anti-inflammatory or bronchodilatory drug substances, particularly in the treatment of obstructive or inflammatory airways diseases such as those mentioned hereinbefore, for example as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs. An agent of the invention may be mixed with the ariti-inflammatory or bronchodilatory drug in a fixed pharmaceutical composition or it may be administered separately, before, simultaneously with or after the antiinflammatory or bronchodilatory drug. Such anti-inflammatory drugs include steroids, in particular giucocorticosteroids such as budesonide, be amethasone, fluticasone or mometasone, and dopamine receptor agonists such as cabergoiine, bromocriptine or ropinJrolβ. Such bronchodilatory drugs include anticholinergic or antimuscarMc agents, in particular ipratropium bromide, oxftropium bromide and tiotropium bromide.
Combinations of agents of the invention and steroids may be used, for example, in the treatment of COPD or, particularty, asthma. Combinations of agents of the invention and anticholinergic or anttmuscarinic agents or dopamine receptor agonists may be used, for example, in the treatment of asthma or, particularty , COPD. In accordance with the foregoing, the present invention also provides a method for the treatment of an obstructive or inflammatory airways disease which comprises administering to a subject, particularly a human subject, in need thereof a SoIuWe Polypeptide, as hereinbefore described. In another aspect, the invention provides a Soluble Polypeptide, as hereinbeforβ described for use in the preparation of a medicament for the treatment of an obstructive or Inflammatory airways disease.
The Soluble Polypeptides of the Invention are also particularly useful for the treatment, prevention, or amelioration of chronic gastrointestinal inflammation, such as inflammatory bowel diseases, including Crohn's disease and ulcerative ooHtis.
"Chronic gastrointestinal inflammation" refers to inflammation of the mucosal of the gastrointestinal tract that is characterized by a relatively longer period of onset, is long- lasting (e.g., from several days, weeks, months, or years and up to the We of the subject), and is associated with infiltration or influx of mononuclear cells and can be further associated with periods of spontaneous remission and spontaneous occurrence. Thus, subjects with chronic gastrointestinal inflammation may be expected to require a long period of supervision, observation, or care. "Chronic gastrointestinal inflammatory conditions" (also referred to as "chronic gastrointestinal inflammatory diseases") having such chronic inflammation include, but are not necessarily limited to, inflammatory bowel disease (IBD), coβtts induced by environmental insults (e.g.. gastrointestinal inflammation (e.g., colitis) caused by or associated with (e.g., as a side effect) a therapeutic regimen, such as administration of chemotherapy, radiation therapy, and the like), colitis in conditions such as chronic granulomatous disease (Schappi et al. Arch CKs Child. 2001 February; 1984(2): 147« 151), celiac disease, celiac sprue (a heritable disease in which the intestinal lining is inflamed Ri response to the ingestion of a protein known as gluten), food allergies, gastritis, infectious gastritis or enterocolitis (e.g., Helicobacter pylori-infected chronic active gastritis) and other forms of gastrointestinal inflammation caused by an infectious agent, and other like conditions.
As used herein, "inflammatory bowel disease" or "IBO" refers to any of a variety of diseases characterized by inflammation of aβ or part of the intestines. Examples of inflammatory bowel disease include, but are not limited to, Crohn's disease and ulcerative colitis. Reference to IBD throughout the specification is often referred to in the specification as exemplary of gastrointestinal inflammatory conditions, and is not meant to be limiting. In accordance with the foregoing, the present invention also provides a method for the treatment of chronic gastrointestinal inflammation or inflammatory bowel diseases, such as ulcerative colitis, which comprises administering to a subject, particularly a human subject, in need thereof, a Soluble Polypeptide, as hereinbefore described. In another aspect, the invention provides a Soluble Polypeptide, as hereinbefore described for use in the preparation of a medicament for the treatment of chronic gastrointestinal inflammation or inflammatory bowel diseases.
The present invention is also useful in the treatment, prevention or amelioration of leukemias or other cancer disorders.
Also encompassed within the scope of the present invention is a method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of a Soluble Polypeptide, and at least one second drug substance, said second drug substance being a immunosuppressive / immunomodulatory, antiinflammatory chemotherapeutic or anti-infectious drug, e.g. as indicated above.
Or, a therapeutic combination, e.g. a kit, comprising of a therapeutically effective amount of a) a Soluble Polypeptide of the Invention and b) at least one second substance selected from a immuno-βuppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti-infectious drug, e.g. as indicated above. The kit may comprise instructions for its administration.
Where the Soluble Polypeptides of the Invention are administered in conjunction with other immuno-suppressive / immunomodulatory, anti-inflammatory chemotherapeutic or anti- infectious therapy, dosages of the co-administered combination compound will of course vary depending on the type of co-drug employed, on the condition being treated and so forth.
In one embodiment, the Soluble Polypeptides of the invention can be used to detect levels of CD47+ dendritic cells, or levels of cells that contain CD47. This can be achieved, for example, by contacting a sample (such as an in vitro sample) and a control sample with a Soluble Polypeptide of the Invention under conditions that allow for the formation of a complex between the Soluble Polypeptides and CD47 expressing cells. Any complexes formed are detected and compared in the sample and the control. For example, standard detection methods, well known in the art, such as flow cytometric assays, can be performed using the compositions of the invention.
Accordingly, in one aspect, the invention further provides methods for detecting the presence of CD47 (e.g., human CD47) in a sample, or measuring the amount of CO47, comprising contacting the sample, and a control sample, with a Soluble Polypeptide of the Invention, under conditions that aftow for formation of a complex between the Soluble PofypβptkJβ and CD47. The formation of a complex is then detected, wherein a difference in complex formation between the sample compared to the control sample is indicative of the presence of CD47 in the sample.
Also within the scope of the invention are kits consisting of the compositions of the invention and instructions for use. The kit can further contain a least one additional reagent. Kits typicaBy include a label indicating the intended use of the contents of the kit The term label includes any writing, or recorded material supplied on or with the kit, or which otherwise accompanies the kit. The kit may further comprise tools for diagnosing whether a patient belongs to a group that wifl respond to a treatment, as defined above.
The invention having been fully described, it is further illustrated by the foβowing examples and claims, which are iβustrative and are not meant to be further limiting.
BRiEF DESCRIPTION OF THE FIGURES
Figure 1. Murine SIRPα-Fc binds CO47+/+ (WT) but not CD47-7- (KO) cede. Murine SIRPα- Fc binding to CD47+/+ (WT) or CD47-/- (KO) murine sptenocytes was detected by flow cytometry as described. The fluorescence resulting from SIRPα-Fc binding (SIRP) is plotted as dot piot versus FL3.
Figure 2. Human SIRPα-Fc binds to CD47+/+ expressing (Jin8CD47) but not CD47 deficient Jurkat T celts (Jύi8). SIRPα-Fc binding was quantified by flow cytometry as described. The fluorescence resulting from SIRPα-Fc binding (SlRP) is plotted as histogram in bold Hne, Bold line indicates binding in presence of lOug/mL anti CD47 clone B6H12.
Figure 3. Blocking CD47/SIRPα at priming prevents aHergJc disease development in BALB/c mice. (3a) Mice were OVA-sensitized i.p. on day 0 and 5 in the presence or absence of SIRPα-Fc fusion molecules, and received OVA-aerosol challenges on day 12, 16 and 20 and euthanized on day 21 (n=4 to 7 mice per group). (3b) Lung sections of naϊve (PBS), OVA immunized, OVA plus SIRP-α-Fotreated mice stained with H&E and PAS. Data are representative of 3 individually analyzed lungs. (3c) Differential BALF cell numbers were analyzed by flow cytometry. (3d) Serum levels of OVA-βpβdfic IgE measured at day 21. (3e) IL-4, IL-5 and IL-13 levels in the supernatants of mLN cell cultures after 3 days of in vitro rβstimulafon with OVA. (3f) At day 21 , % of CD4+ T cells and IL-13 producing CD4+ T cells were assessed by flow cytometry in ex vivo isolated mLNs. Shown is one representative experiment out of 3. (3g) IL-4, IL-5, IL-13 and (h) eotaxin release in lung explants. Data are mean ± SEM. *P<0.05. "P<0.01, "*P<0.001.
Figure 4. Mechanisms of disease protection in SIRPα-Fc treated BALB/c mice. Mice were OVA-sensitized on day 0 and 5 in the presence or absence of SIRPα-Fc fusion molecule, received OVA-aerosol challenge* on day 12, 16 and 20 and were euthanized on day 21. (4a) Subpopulations of CD11blowCD103+ and CD11bhighCD103-DCs (gated on CD11c+) in mLNs. Data are presented as % DC subsets (n = 3 to 4 mice per group). (4b) BALB/c mice were passively transferred with CFSE-labeled CD47+/+CD4+Tg T cells a day prior i.p. OVA- aiumn immunization in the absence (P8S) or presence of SIRPα-Fc and CFSE eel dilution was examined in mLNs after 2 days. Data is one representative experiments performed on 4 mice per group. (4c) At day 21, absolute numbers of eosinophils (CCR3+) were assessed by flow cytometry in ex vivo isolated mLNs. Data are mean ± SEM (n= 3 to 4 mice per group).
Figure 6. Protection of TNBS-colitis by SIRPα-Fc administration
Colitis was induced and assessed as described. 100 ug /animal murine SIRPα-Fc was applied at day 0 and 24h after i.p.. Alternatively PBS was injected i.p. θodyweight of animals was assessed until day 4 post TNBS injection.
EXAMPLES 1. Examples of Soluble Polypeptides of the invention
The following table 3 provides examples of Soluble Polypeptides of the Invention that may be produced by recombinant methods using ONA encoding the disclosed amino acid sequences.
Figure imgf000032_0001
2. Affinity determination 2.1 Affinity to CD47
The affinity of human SIRPα-Fc to monomeric CD47 or divalent CD47-FC protein can be assessed by Biacore. Monovalent interaction of CO47 V-domain with SIRPu is reported around 1 μM (Heathertey et al MoI Cell. 2008). For example, an APP-tagged CD47 V domain protein is expressed in HEK293 cells and compared to a divalent-CD47-Fc protein as ligand. The monovalent interaction with SIRPα- Fc was measured as K0 of 0.8 μM while the binding strength (avidity) of a divalent CD47-Fc protein increased by a factor of 10 to KD < 6OnM. In contrast binding of a murine CD47-Fc fusion protein could not be observed indicating the specificity of the assessed interaction.
Figure imgf000033_0001
2.2 Cell binding to murine cellular CD47
5x105 CD47" and CD47~ murine splenocytes from wild type or CD47 knock out mice are resuspended in 50 μl of FACS BUFFER ( PBS 2% FCS 2mM EDTA ) containing 200μg/mi human IgG and 5 μg/ml murine SIRPα-Fc for 30 min at 4°C. After washing, cells are stained with FITC-labeled strβptavtdin (1/1000) for 30 min at 4°C. The results show that SIRPα-Fc binds to lymphocytes of CD474* but not to lymphocytes from CD47V knockout mice (Fig 1 ).
2.3 Cell binding to human cellular CD47
5x105 CD47* and CD4r Jυrkat T cell lines are resuspended in 50 μl of FACS BUFFER (PBS 2% FCS 2mM EDTA) containing 200μg/ml human IgG and 5 μg/ml SiRPa-Fc for 30 min at 4°C. After washing, cells are stained with FITC-labeled streptavidin (1/1000) for 30 min at 4°C.
The results show that SIRPα-Fc binds to lymphocytes of CD47*" Jυrkat cells (Jin8CD47) but not to Jυrkat cells which do not express CD47 (Jin8) (Fig 2). Binding to cells was specific as indicated by complete blockade with arrti CD47 antibody B6H12.
2.4 Inhibition/blocking studies of biotlnylated SIRP-α Fc binding to CHO CD47 cell lines
Alternatively, inhibition/blocking studies of biotinylated SIRPα-Fc binding to CHO CD47 cell lines may be performed using anti-CD47 mAbs directed against different epitopes (i.e. B6H12. 2D3, BRIC 126, IF7 and 10G2 donee, different anti- human SIRP-<x(CD172a) mAbs, recombinant human Thrombospondin-1 (TSP-1) or TSP-1 c-terminal (4N1K) and control (4NG6) peptides.
2.5 M∞ltmg/lnhibftion studies of directly labeled antJ-CD47 mAb binding to CHO CD47 cell lines
As a complementary approach, Inhibition/blocking studies of directiy labeled anti-CD47 mAb biding to CHO CD47 oeK lines may be performed using non biotinyiated human SIRP-α-Fc.
Inhibition/blocking studies of biotinyiatβd CD47-Fc bindino to L ceβs transfected with human SIRPα-Fc using SIRPα-Fc can ateo be assessed.
3. SIRPHX-FC functional assays
3.1 immune complex-stimulated dendritic cell cytokine release assay
Peripheral blood monocytes (CD14+ CD16-) as well as monocyte-derived dendritic cells (DCs) are prepared as described (Latoυr et ai, J of Immunol, 2001: 167:2547). Conventional (DCs) are isolated as CDH(M-, lineage-, by a FACS Ana (BD Biosciences) by using aflophycocyanin (APC)-labθlβd anti-CD11c (B-Jyβ), a mixture of FITOIabβtβd mAbβ against lineage markers. CD3 ,CD14, CD15, CD16, CD19 and CD56 and APC-Cy7-iabeled CD4 (RPA-T4) to reach >99% purity. APCs are stimulated with Staphylococcus Aureus Cowan 1 at 1/40.000 (Pansorbin) or soluble CD40L (1 μg/ml) and IFN-γ (500 U/ml) in the presence of various concentrations of human SIRPα-Fc (1 to 20 μg/mt) in HB101 serum-free medium. Cytokine (IL-1, IL-6, IL- 10, IL-12p70, IL-23, IL-8 and TNF-α) release is assessed by ELISA in the 24h or 48h culture supernatantβ.
3.2 Mixed lymphocyte reactions (MLR) assay
Mitomycin c-treated mature DCs (SAC or LPS stimulated) will be cocuKured with allogeneic unfractionated, naTve (CD45RA* 0062L1*") or memory (CD45RO* CD62Ltaw) adult CD4+ T cells ( 106ZmI) at various stimulators (DCs)/ responder (T cede) ratios in the presence or absence of Soluble Polypeptides of the Invention (5 to 50 μg/ml). T cell proliferation (3H thymidine uptake) and IFN-γ release will be assessed in the culture supernatants of 5 to 6 day primary cultures. 4. In vivo date with murine animal models for use of 8IRPa-Fc in the treatment of asthma
BALB/c mice were sensitized by i.p. injection of 10ug OVA (Sigma, Grade V) adsorbed to 1mg Imject Alum (Pierce) on days 0 and 5. On days 12, 16 and 20, mice were chaUenged for 30 minutes with a 0.5% OVA aerosol (Sigma, Grade V) delivered by a vibrating mesh nebulizer system (Omron). 24 hours after the last chaflβnge, mice were sacrificed with 75mg/kg sodium pentobarbital overdose and bled. BAL was coθected 3 times with 0.5ml physiologic saline and lung and mLN were isolated. One third of the Kings were rinsed in PBS supplemented with antibiotics, cut into small pieces and put in culture in flat-bottom 24 wβH plates for 24h in 1ml R PM 11640 (Wment Inc.) supplemented with 10% fetal bovine serum, 500U/ml Penicillin, 500ug/ml Streptomycin, 1OmM HEPES buffer and 1mM 2-ME. MLN cells (4x106 cβBs/ml) were cultured in flat-bottom 96 well plates and reβtimulated with OVA (100μ/ml) for 72h. Total BAL cβPs were washed, counted and stained for 30 minutes with anti-CCR3 PE (R&D systems), anti-CD3 FUC (clone 145-2C11) and anti-B220 FTTC (R&D systems). As described in Van Rijt LS. et al (Immunol Methods. 2004 May;288(1- 2): 111-21), granulocytes were found to be granular, rrøtvautoftuorescent, lacking the expression of C03 and B220. Eosinophils were distinguished from neutrophils by CCR3 expression. Lymphocytes were smafi, non-granular, non-autøfluorescent, expressing CD3 or B220 and the other mononuclear ceils, including macrophages and DCs, were lacking CD3, B220 and CCR3. To identify the OC subsets, mLNs and lungs were first treated with liberase. minced and cells were counted. Lung ceβ suspensions were treated with NH4CI for red blood cell lysis and washed before staining. Cells were stained with anti-CD11c FiTC (BD Biosciences) or anti-CD11c APC (clone N418), anti-CD11b PE (Caltβg), antW-Ad/MEd PE (BD Biosciences), anti-GR1, antf-8220 FITC (R&D systems), 120G8 FITC and antt- CD103-bk>tiny1atβd followed by SA-APC or CD103 PE (BD Biosciences) anti~CD47 and anti- SIRP-α mAbs. In lungs, autofluorescent alveolar macrophages were excluded from the analysis gates. To identify regulatory T cells, anti-CD4 FITC or APC (BD Biosciences), anti- CD25 PE (Caltag) or FITC (BD Biosciences), anti-CD44 APC (clone IM7 8.1) were used. To measure IL-13 production ex vivo, mast ceils and basophils were first identified with extracellular antMgE-biotinylated and anti-CD117 (c-Kit, BioLegend) and CD4 T cells were stained with anti-CD4 APC. Cells were fixed, permeabilized and stained with antML-13 PE (eBioscience). Cells were first stained for extracellular markers (anti-CO4 APC and anti- CD25 FITC), fixed, permeabilized and stained with anti-FoxP3 PE (kit from eBioscience). Mast ceHs and basophils + lntracytoplasmic IL-13 staining. All the data were acquired on a FACSCaRbur or Cantoil Ftow Cytometer (BD Btoscienceβ) and analysed with CeHquest or DIVA software (BO Bioβciences).
Cytokine measurements
IL-2, IL-4, IL-5, 1 L- 10, IFN-y (BO Btoscienceβ), IL- 13 (R&D Systems) are measured in mLNε culture supernatants and king explants by ELISA.
Mediastinal LN cefis from OVA-sβnsitizβd and challenged mice are restimulated in vitro for 3 days with OVA protein (1mg/ml) and IL-4, IL-5 and IL-13 production are quantified by ELISA in the culture supematante. Lungs expiants are cultured overnight in complete medium and culture supernatants are collected to measure cytokine release.
Results
lti vivo data with murine animal models for use of SIRPα-Fc in the treatment of allergic asthma
CD47 and SIRPα appear as important molecules in the initiation and perpetuation of SIRP- α+CD103- DC-driven Th2 immunity. Thus they may therefore be harnessed therapeutically to reduce lung inflammation and ameliorate airway disease. We here assessed the efficacy of SIRP-α plus the Fc region of human IgGI (SIRPα-Fc) on the development of allergic airway inflammation. BALB/c mice administered either SIRPα-Fc on days 0 and 5 of OVA immunization (Fig 3a) had very few or no inflammatory cell infiltration of the lung tissues after OVA aerosol challenge (Fig 3b). A strong reduction or an absence of eosinophils, neutrophils and lymphocytes in the BALF (Fig 3c) occurred together with a drop in serum OVA-speciftc IgE (Rg 3d), a 50% reduction in lymph node ceiutaήty and a drastic inhibition of IL-4, IL-5 and IL-13 production in the mLNs (Fig 3e and 0- The protection from airway disease development was not correlated with an increase in IL-10 nor IFN-γ release, which in fact was also suppressed in treated mice (data not shown). We next examined cytokine and chemokine release in the culture supernatants of lung explants of CD47- and SIRPα-Fc- treated mice and found that IL-5. IL-13 and eotaxin release were inhibited while IL-4 expression remained unchanged (Fig 3 g and h).
We next explored the potential mechanisms that governed this inhibition and resulted in protection from airway disease. We found a diminution in the accumulation of SIRP- (X+CD103- DCs in the mLNs of CD47-Fc-treated mice (Fig 4a). The administration of SIRPα- Fc ateo led to a reduction in the proportion of CFSE-labeled Tg T cβte in the mlNs of OVA- immunteed mice treated with CD47-FC (Rg 4b). Finally, we observed a decrease in the proportion and accumulation of eosinophils in the mLns of SIRPα-Fc-treated mice (Fig 4c).
These data demonstrate that CD47/SIRPα interruption at primary Ag sensitization drastically reduced type 2 responses in mLNs and lungs as well as IgE-dependent airway inflammation.
6. In vivo data with murine animal models for use of SIRPα-Fc in the treatment of colitis
Tήnitrobenzene sulfonic acid (TNBS) (2 or 3 mg) was dissolved in 50% ethanol and instffled into the colons of male Bato/c mice (WT and CD47 KO) via a 3.5F catheter. Control mice were given ethanol atone. Mice were weighed every 24 hours and sacrificed on day 2 (early time point) or day 4. In the chronic TNBS colitis model, 1.5 mg of TNBS was instHted intrarectal on day 0 and again on day 7, and mice were sacrificed on day 12. Serum, mesenteric lymph nodes and colons were harvested for further analysis. Colons were scored macroβcopteaJfy using the Wallace criteria which takes into account the presence of diarrhea, adhesions, thickening of the bowel wall and ulceration. They were also evaluated for microscopic markers of inflammation using the Ameho criteria, a scoring system based upon thickening of the submυcoβa, infiltration of the submucosa and lamina propria with mononuclear ceHs, mucous depletion, loss of crypt architecture, and edema (not shown). A recombinant mouse SIRPα-Fc fusion protein was administered iπtraperitoneatty (100 ug/mouse) just prior to TNBS colitis induction and 24 hours thereafter. Control mice received saline alone. Injection of murine SIRPα-Fc 100 μg/animal on day 0, 30 min before TNBS induction and on day 1 blocked statistically significantly the disease development as assessed by bodyweight toss.
β. in vivo murine animal models for use of SIRPα-Fc in the treatment of arthritis
Collagen induced arthritis model:
Mycobacterium tuberculosis are mixed with Freund's complete adjuvant and thoroughly shaken (= solution A). Bovine collagen solution aϋςuots are weH mixed on ice with sterile PBS (= solution B). Solution A and solution B were injected as emulsion into naTve male DBA/1 mice. The mice are anaesthetized by s.c. injection of a sterile filtrated mixture of kβtamine. Upon narcosis, the root of the tail of each mouse is shaved and subsequently. 0.1 ml of the collagen-emulsion per mouse (containing 100 μg of collagen) is injected i.d. into the base of the tail. A second injection of 100 μl of collagen/PBS (1:5 dilution) is given i.p. on day 22 after the first immunization (= booster). Swelling and scoring of disease is assessed as described in Nat Protoc. 2007;2(5): 1269-75 by Brand et at.
7. USEFUL AMINO ACID AND NUCLEOTIDE SEQUENCES FOR PRACTICING THE INVENTION
Figure imgf000038_0001
Figure imgf000039_0001

Claims

CLAlMS
1. A soluble CD47 binding polypeptide for use as a medicament, comprising a SIRPα- derived polypeptide selected among the group consisting of: a) an extracellular domain of SIRPα (SEQ IO NO.3); b) a fragment of SEQ ID NO:3, and, c) a variant polypeptide of SEQ ID NO:3 having at least 75% identity to SEQ IO NO.2; wherein said SJRPα-derived polypeptide binds to human CD47 (SEQ ID NO:24).
2. The soluble CD47 binding polypeptide accorcβng to claim 1, wherein said soluble CD47 binding polypeptide binds to human CD47 with a K0 of 2 μM or less, and inhibits induced cytokine secretion as measured in an immune complex-stimulated dendritic cβB cytokine release assay.
3. The soluble CD47 binding polypeptide according to claim 1 or 2, wherein said SIRPα derived polypeptide is fused to an IgG Fc fragment
4. The soluble CD47 binding polypeptide according to Claim 1, 2 or 3, wherein said IgG Fc fragment is a mutant aglycosylated Fc fragment.
5. The soluble CD47 binding polypeptide according to any of the preceding claims, wherein said extracellular domain of SIRPα comprises at least the V-region of SIRPα (SEQ ID NO:2).
6. The soluble CD47 binding polypeptide according to any of the preceding claims, for use as a drug in the treatment of autoimmune and inflammatory disorders.
7. The soluble CD47 binding polypeptide according to any of Claims 1-β. for use as a drug in the treatment of a) Th2-mediated airway inflammation; b) allergic disorders; c) asthma; d) inflammatory bowel diseases; e) arthritis; f) ischemic disorders; or, g) teukemias or cancer.
The soluble CD47 binding polypeptide according to any of Claims 1-7, wherein said polypeptide essentially consists of an extracellular domain of SIRPα fused to the Fc fragment of a human IgO.
A protein comprising at least two soluble CD47 binding polypeptides according to any of Claims 1-8.
A pharmaceutical composition comprising the soluble CD47 binding polypeptide according to any of Claims 1-8 or the protein according to Claim 9.
The pharmaceutical composition of Claim 10, in combination with one or more of a pharmaceutically acceptable excipient, diluent or carrier.
An isolated nucleic acid encoding the soluble CD47 binding polypeptide according to any of Claims 1-8.
A cloning or expression vector comprising one or more nucleic acids according to Claim 12.
A cloning or expression vector according to Claim 13, which comprises at least one nucleic acid of SEQ ID NO.25 or 26.
A host ceH comprising one or more cloning or expression vectors according to Claim 13 or 14.
The host cell according to Claim 15, wherein said host cell is a mammalian cell, for example, CHO ceil.
PCT/EP2009/067411 2008-12-19 2009-12-17 Soluble polypeptides for use in treating autoimmune and inflammatory disorders WO2010070047A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
CN2009801511374A CN102257001A (en) 2008-12-19 2009-12-17 soluble polypeptides for use in treating autoimmune and inflammatory disorders
EP09796708A EP2379584A1 (en) 2008-12-19 2009-12-17 Soluble polypeptides for use in treating autoimmune and inflammatory disorders
AU2009327058A AU2009327058A1 (en) 2008-12-19 2009-12-17 Soluble polypeptides for use in treating autoimmune and inflammatory disorders
EA201100947A EA201100947A1 (en) 2008-12-19 2009-12-17 SOLUBLE POLYPEPTIDES INTENDED FOR APPLICATION IN THE TREATMENT OF AUTOIMMUNE AND INFLAMMATORY DISORDERS
CA2747678A CA2747678A1 (en) 2008-12-19 2009-12-17 Soluble polypeptides for use in treating autoimmune and inflammatory disorders
BRPI0923442A BRPI0923442A2 (en) 2008-12-19 2009-12-17 soluble polypeptides for use in treating autoimmune and inflammatory disorders.
MX2011006621A MX2011006621A (en) 2008-12-19 2009-12-17 Soluble polypeptides for use in treating autoimmune and inflammatory disorders.
US13/132,400 US20110237498A1 (en) 2008-12-19 2009-12-17 Soluble polypeptides for use in treating autoimmune and inflammatory disorders
JP2011541441A JP2012512640A (en) 2008-12-19 2009-12-17 Soluble polypeptides for use in the treatment of autoimmune and inflammatory disorders

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP08172369 2008-12-19
EP08172369.4 2008-12-19
US20355808P 2008-12-22 2008-12-22
US61/203,558 2008-12-22
EP08172751.3 2008-12-23
EP08172751 2008-12-23

Publications (1)

Publication Number Publication Date
WO2010070047A1 true WO2010070047A1 (en) 2010-06-24

Family

ID=42268343

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2009/067411 WO2010070047A1 (en) 2008-12-19 2009-12-17 Soluble polypeptides for use in treating autoimmune and inflammatory disorders

Country Status (11)

Country Link
US (1) US20110237498A1 (en)
EP (1) EP2379584A1 (en)
JP (1) JP2012512640A (en)
KR (1) KR20110112299A (en)
CN (1) CN102257001A (en)
AU (1) AU2009327058A1 (en)
BR (1) BRPI0923442A2 (en)
CA (1) CA2747678A1 (en)
EA (1) EA201100947A1 (en)
MX (1) MX2011006621A (en)
WO (1) WO2010070047A1 (en)

Cited By (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011101791A1 (en) * 2010-02-18 2011-08-25 Tcl Pharma Anti-cd28 humanized antibodies
WO2013155109A1 (en) * 2012-04-09 2013-10-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods for generation of pluripotent and multipotent cells
CN103635490A (en) * 2011-06-16 2014-03-12 诺瓦提斯公司 Soluble proteins for use as therapeutics
WO2014094122A1 (en) 2012-12-17 2014-06-26 Trillium Therapeutics Inc. Treatment of cd47+ disease cells with sirp alpha-fc fusions
JP2015504899A (en) * 2012-01-17 2015-02-16 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー High affinity SIRP-alpha reagent
WO2016179399A1 (en) * 2015-05-06 2016-11-10 The Board Of Trustees Of The Leland Stanford Junior University High affinity cd47 analogs
EP3128005A1 (en) * 2015-08-07 2017-02-08 Alexo Therapeutics Inc. Sirp-alpha variant constructs and uses thereof
US9566347B2 (en) 2011-02-07 2017-02-14 The Trustees Of The University Of Pennsylvania Peptides and methods using same
WO2017027422A1 (en) 2015-08-07 2017-02-16 Alexo Therapeutics Inc. Constructs having a sirp-alpha domain or variant thereof
CN106535914A (en) * 2014-08-08 2017-03-22 阿列索治疗公司 Sirp-alpha variant constructs and uses thereof
US20170291945A1 (en) * 2014-09-15 2017-10-12 The Board Of Trustees Of The Leland Stanford Junior University Targeting aneurysm disease by modulating phagocytosis pathways
WO2017194586A1 (en) * 2016-05-10 2017-11-16 Université Pierre Et Marie Curie - Paris 6 (Upmc) Agents that activate cd47 and their use in the treatment of inflammation
WO2018081898A1 (en) 2016-11-03 2018-05-11 Trillium Therapeutics Inc. Improvements in cd47 blockade therapy by hdac inhibitors
EP3261671A4 (en) * 2015-02-27 2018-10-24 The Board of Trustees of The Leland Stanford Junior University Combination therapy for treatment of coronary artery disease
EP3287470A4 (en) * 2015-04-24 2018-10-31 Immuneonco Biopharmaceuticals (Shanghai) Co., Ltd Novel recombinant bi-functional fusion protein and preparation and application therefor
US10188701B2 (en) * 2015-10-01 2019-01-29 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US10329354B2 (en) 2013-09-18 2019-06-25 The Board Of Trustees Of The Leland Stanford Junior University Modulation of efferocytosis pathways for treatment of atherosclerotic disease
EP3180363B1 (en) 2014-08-15 2019-09-25 Merck Patent GmbH Sirp-alpha immunoglobulin fusion proteins
US10513548B2 (en) 2017-02-27 2019-12-24 Shattuck Labs, Inc. CSF1R-based chimeric proteins
WO2020047651A1 (en) 2018-09-04 2020-03-12 Trillium Therapeutics Inc. Cd47 blockade with parp inhibition for disease treatment
US10780121B2 (en) 2018-08-29 2020-09-22 Shattuck Labs, Inc. FLT3L-based chimeric proteins
EP3585418A4 (en) * 2017-02-27 2020-11-25 Shattuck Labs, Inc. Tigit- and light-based chimeric proteins
WO2021011544A1 (en) 2019-07-16 2021-01-21 Gilead Sciences, Inc. Hiv vaccines and methods of making and using
US10907209B2 (en) 2009-05-15 2021-02-02 University Health Network Compositions and methods for treating hematological cancers targeting the SIRPα CD47 interaction
US10918095B2 (en) 2017-03-31 2021-02-16 Beijing Biocytogen Co., Ltd Genetically modified mice expressing humanized CD47
US10961318B2 (en) 2017-07-26 2021-03-30 Forty Seven, Inc. Anti-SIRP-α antibodies and related methods
US10973212B2 (en) * 2017-03-31 2021-04-13 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Genetically modified non-human animal with human or chimeric SIRPa
WO2021076908A1 (en) 2019-10-18 2021-04-22 Forty Seven, Inc. Combination therapies for treating myelodysplastic syndromes and acute myeloid leukemia
WO2021087064A1 (en) 2019-10-31 2021-05-06 Forty Seven, Inc. Anti-cd47 and anti-cd20 based treatment of blood cancer
WO2021130638A1 (en) 2019-12-24 2021-07-01 Carna Biosciences, Inc. Diacylglycerol kinase modulating compounds
WO2021163064A2 (en) 2020-02-14 2021-08-19 Jounce Therapeutics, Inc. Antibodies and fusion proteins that bind to ccr8 and uses thereof
US11401329B2 (en) 2017-08-02 2022-08-02 Phanes Therapeutics, Inc. Anti-CD47 antibodies and uses thereof
WO2022190058A1 (en) 2021-03-12 2022-09-15 Dcprime B.V. Methods of vaccination and use of cd47 blockade
US11446315B2 (en) 2016-11-03 2022-09-20 Pf Argentum Ip Holdings Llc Enhancement of CD47 blockade therapy by proteasome inhibitors
WO2022221304A1 (en) 2021-04-14 2022-10-20 Gilead Sciences, Inc. CO-INHIBITION OF CD47/SIRPα BINDING AND NEDD8-ACTIVATING ENZYME E1 REGULATORY SUBUNIT FOR THE TREATMENT OF CANCER
WO2022229818A1 (en) 2021-04-27 2022-11-03 Pf Argentum Ip Holdings Llc Enhancement of cd47 blockade therapy with dhfr inhibitors
US11529425B2 (en) * 2017-09-07 2022-12-20 Dingfu Biotarget Co., Ltd. Immunoconjugates comprising signal regulatory protein alpha
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
US11613564B2 (en) 2019-05-31 2023-03-28 ALX Oncology Inc. Methods of treating cancer
WO2023076983A1 (en) 2021-10-28 2023-05-04 Gilead Sciences, Inc. Pyridizin-3(2h)-one derivatives
WO2023077030A1 (en) 2021-10-29 2023-05-04 Gilead Sciences, Inc. Cd73 compounds
WO2023073580A1 (en) 2021-10-29 2023-05-04 Pfizer Inc. Enhancement of cd47 blockade with taxanes for cd47+ cancer therapy
WO2023079438A1 (en) 2021-11-08 2023-05-11 Pfizer Inc. Enhancement of cd47 blockade therapy with anti-vegf agents
WO2023122615A1 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
US11702458B2 (en) 2017-01-05 2023-07-18 Kahr Medical Ltd. PD1-41BBL fusion protein and methods of use thereof
WO2023147418A1 (en) 2022-01-28 2023-08-03 Gilead Sciences, Inc. Parp7 inhibitors
EP4245756A1 (en) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023183817A1 (en) 2022-03-24 2023-09-28 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
US11771764B2 (en) 2017-11-06 2023-10-03 Pfizer Inc. CD47 blockade with radiation therapy
WO2023196784A1 (en) 2022-04-05 2023-10-12 Gilead Sciences, Inc. Combinations of antibody therapies for treating colorectal cancer
WO2023205719A1 (en) 2022-04-21 2023-10-26 Gilead Sciences, Inc. Kras g12d modulating compounds
WO2023228044A1 (en) 2022-05-25 2023-11-30 Pfizer Inc. Dosing regimens of sirp alpha fusion proteins for treatment of cancer
WO2024006929A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Cd73 compounds
WO2024015741A1 (en) 2022-07-12 2024-01-18 Gilead Sciences, Inc. Hiv immunogenic polypeptides and vaccines and uses thereof
US11891423B2 (en) 2018-04-17 2024-02-06 Hangzhou Sumgen Biotech Co., Ltd. Fusion protein binding to CD47 protein and application thereof
US11897937B2 (en) 2017-01-05 2024-02-13 Kahr Medical Ltd. SIRPalpha-41BBL fusion protein and methods of use thereof
WO2024040151A1 (en) 2022-08-18 2024-02-22 Pfizer Inc. Sirp alpha fusion protein and anti-cd38 antibody combination therapies
RU2815515C2 (en) * 2018-07-11 2024-03-18 КАР Медикал Лтд. Embodiments of sirp-alpha-4-1bbl fusion protein and methods of use thereof
WO2024064668A1 (en) 2022-09-21 2024-03-28 Gilead Sciences, Inc. FOCAL IONIZING RADIATION AND CD47/SIRPα DISRUPTION ANTICANCER COMBINATION THERAPY

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013090776A1 (en) 2011-12-15 2013-06-20 The Board Of Trustees Of The Leland Stanford Junior University Inhibition of axl/gas6 signaling in the treatment of disease
CN102643347A (en) * 2012-03-29 2012-08-22 中国人民解放军第四军医大学 Leukemia stem cell targeting soluble protein TrxHis-hSIRP alpha
EP2931265B3 (en) 2012-12-14 2023-04-05 The Board of Trustees of the Leland Stanford Junior University Modified axl peptides and their use in inhibition of axl signaling in anti-metastatic therapy
US9873747B2 (en) 2013-01-31 2018-01-23 Thomas Jefferson University Fusion proteins that facilitate cancer cell destruction
AU2015204766B2 (en) * 2014-01-08 2020-08-13 The Board Of Trustees Of The Leland Stanford Junior University Targeted therapy for small cell lung cancer
JP6355032B2 (en) * 2014-03-24 2018-07-11 イミューンオンコ バイオファーマシューティカルズ (シャンハイ) カンパニー リミテッド Novel recombinant bifunctional fusion proteins, their preparation and use
WO2019061012A1 (en) * 2017-09-26 2019-04-04 南京凯地生物科技有限公司 Preparation of chimeric antigen receptor t-cell specifically targeting cd47 and application thereof
CN108484774B (en) * 2018-03-09 2021-11-05 上海高菲生物科技有限公司 SIRP alpha fusion protein and preparation method and application thereof
MA53493A (en) * 2018-08-31 2021-07-07 Alx Oncology Inc POLYPEPTIDES LURES
CN111087473B (en) * 2019-12-11 2022-06-14 上海百英生物科技有限公司 SIRPa-Fc-IL21 fusion protein and application thereof
CN110981942B (en) * 2019-12-11 2022-07-08 中国药科大学 Polypeptide RS-17 with anti-CD47 immune checkpoint antagonistic activity and application thereof
JP2023518835A (en) * 2020-03-26 2023-05-08 エショー バイオセラピー, インク. Bispecific fusion proteins for depletion of regulatory T cells

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999040940A1 (en) * 1998-02-16 1999-08-19 Marie Sarfati Ligands of the cd47 antigen, agents binding the ligands of the cd47 antigen and uses thereof
WO2001048020A1 (en) * 1999-12-24 2001-07-05 Medical Research Council Composition for inhibiting macrophage activity
WO2002092784A2 (en) * 2001-05-15 2002-11-21 Emory University POLYNUCLEOTIDES AND POLYPEPTIDES RELATING TO THE MODULATION OF SIRP α-CD47

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7534866B2 (en) * 2005-10-19 2009-05-19 Ibc Pharmaceuticals, Inc. Methods and compositions for generating bioactive assemblies of increased complexity and uses
EP2573112A1 (en) * 2007-10-11 2013-03-27 The Hospital For Sick Children Modulation of sirpa - cd47 interaction for increasing human hematopoietic stem cell engraftment and compounds therefor

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999040940A1 (en) * 1998-02-16 1999-08-19 Marie Sarfati Ligands of the cd47 antigen, agents binding the ligands of the cd47 antigen and uses thereof
WO2001048020A1 (en) * 1999-12-24 2001-07-05 Medical Research Council Composition for inhibiting macrophage activity
WO2002092784A2 (en) * 2001-05-15 2002-11-21 Emory University POLYNUCLEOTIDES AND POLYPEPTIDES RELATING TO THE MODULATION OF SIRP α-CD47

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
KATO HISASHI ET AL: "SHPS-1 negatively regulates integrin alphaIIbbeta3 function through CD47 without disturbing FAK phosphorylation.", JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH APR 2005, vol. 3, no. 4, April 2005 (2005-04-01), pages 763 - 774, XP009117242, ISSN: 1538-7933 *
KATZ H R: "Inhibitory receptors and allergy", CURRENT OPINION IN IMMUNOLOGY, ELSEVIER, OXFORD, GB, vol. 14, no. 6, 1 December 2002 (2002-12-01), pages 698 - 704, XP004390307, ISSN: 0952-7915 *
LIU ET AL: "Functional Elements on SIRPalpha IgV Domain Mediate Cell Surface Binding to CD47", JOURNAL OF MOLECULAR BIOLOGY, LONDON, GB, vol. 365, no. 3, 23 December 2006 (2006-12-23), pages 680 - 693, XP005733360, ISSN: 0022-2836 *
OKUZAWA C ET AL: "Resistance to collagen-induced arthritis in SHPS-1 mutant mice", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, ACADEMIC PRESS INC. ORLANDO, FL, US, vol. 371, no. 3, 4 July 2008 (2008-07-04), pages 561 - 566, XP022670742, ISSN: 0006-291X, [retrieved on 20080501] *
OLDENBORG P-A: "ROLE OF CD47 IN ERYTHROID CELLS AND IN AUTOIMMUNITY", LEUKEMIA AND LYMPHOMA, HARWOOD ACADEMIC PUBLISHERS, CHUR, CH, vol. 45, no. 7, 1 July 2004 (2004-07-01), pages 1319 - 1327, XP008060559, ISSN: 1042-8194 *
PICCIO LAURA ET AL: "Adhesion of human T cells to antigen-presenting cells through SIRPbeta2-CD47 interaction costimulates T-cell proliferation", BLOOD, AMERICAN SOCIETY OF HEMATOLOGY, US, vol. 105, no. 6, 15 March 2005 (2005-03-15), pages 2421 - 2427, XP002525575, ISSN: 0006-4971 *
SARFATI MARIKA ET AL: "CD47 in the immune response: role of thrombospondin and SIRP-alpha reverse signaling", CURRENT DRUG TARGETS, BENTHAM SCIENCE PUBLISHER, US, vol. 9, no. 10, 1 October 2008 (2008-10-01), pages 842 - 850, XP008105799, ISSN: 1389-4501 *
See also references of EP2379584A1 *
TANAKA KONAGI ET AL: "Inhibition of Src homology 2 domain-containing protein tyrosine phosphatase substrate-1 reduces the severity of collagen-induced arthritis", JOURNAL OF RHEUMATOLOGY, JOURNAL OF RHEUMATOLOGY PUBLISHING COMPANY, TORONTO, CA, vol. 35, no. 12, 1 December 2008 (2008-12-01), pages 2316 - 2324, XP008105798, ISSN: 0315-162X, [retrieved on 20081101] *
YU XIJUN ET AL: "Engagement of CD47 inhibits the contact hypersensitivity response via the suppression of motility and B7 expression by Langerhans cells", JOURNAL OF INVESTIGATIVE DERMATOLOGY, NATURE PUBLISHING GROUP, GB, vol. 126, no. 4, 1 April 2006 (2006-04-01), pages 797 - 807, XP002525574, ISSN: 0022-202X *

Cited By (119)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10907209B2 (en) 2009-05-15 2021-02-02 University Health Network Compositions and methods for treating hematological cancers targeting the SIRPα CD47 interaction
US10364287B2 (en) 2010-02-18 2019-07-30 Institut National De La Sante Et De La Recherche Medicale Anti-CD28 humanized antibodies
US8785604B2 (en) 2010-02-18 2014-07-22 Effimune Anti-CD28 humanized antibodies
US9562098B2 (en) 2010-02-18 2017-02-07 Ose Immunotherapeutics Anti-CD28 humanized antibodies
WO2011101791A1 (en) * 2010-02-18 2011-08-25 Tcl Pharma Anti-cd28 humanized antibodies
EP3428192A1 (en) * 2010-02-18 2019-01-16 OSE Immunotherapeutics Anti-cd28 humanized antibodies
US9566347B2 (en) 2011-02-07 2017-02-14 The Trustees Of The University Of Pennsylvania Peptides and methods using same
CN103635490A (en) * 2011-06-16 2014-03-12 诺瓦提斯公司 Soluble proteins for use as therapeutics
JP2020007329A (en) * 2012-01-17 2020-01-16 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー High-affinity sirp-alpha reagents
JP2020143104A (en) * 2012-01-17 2020-09-10 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー High affinity sirp-alpha reagents
US11208481B2 (en) 2012-01-17 2021-12-28 The Board Of Trustees Of The Leland Stanford Junior University High affinity SIRP-alpha reagents and methods of using
JP2021176873A (en) * 2012-01-17 2021-11-11 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー High affinity sirp-alpha reagents
JP2018021016A (en) * 2012-01-17 2018-02-08 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー High affinity sirp-alpha reagents
JP2015504899A (en) * 2012-01-17 2015-02-16 ザ ボード オブ トラスティーズ オブ ザ レランド スタンフォード ジュニア ユニバーシティー High affinity SIRP-alpha reagent
US10407665B2 (en) 2012-04-09 2019-09-10 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Methods for generation of pluripotent and multipotent cells
WO2013155109A1 (en) * 2012-04-09 2013-10-17 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods for generation of pluripotent and multipotent cells
WO2014094122A1 (en) 2012-12-17 2014-06-26 Trillium Therapeutics Inc. Treatment of cd47+ disease cells with sirp alpha-fc fusions
US9969789B2 (en) 2012-12-17 2018-05-15 Trillium Therapeutics Inc. Treatment of CD47+ disease cells with SIRP alpha-Fc fusions
EP3575326A1 (en) 2012-12-17 2019-12-04 Trillium Therapeutics Inc. Treatment of cd47+ disease cells with sirp alpha-fc fusions
EP4116331A1 (en) 2012-12-17 2023-01-11 PF Argentum IP Holdings LLC Treatment of cd47+ disease cells with sirp alpha-fc fusions
US10906954B2 (en) 2012-12-17 2021-02-02 Trillium Therapeutics Inc. Treatment of CD47+ disease cells with SIRPα-Fc fusions
US10329354B2 (en) 2013-09-18 2019-06-25 The Board Of Trustees Of The Leland Stanford Junior University Modulation of efferocytosis pathways for treatment of atherosclerotic disease
CN113621075A (en) * 2014-08-08 2021-11-09 Alx肿瘤生物技术公司 SIRP-alpha variant constructs and uses thereof
CN106535914A (en) * 2014-08-08 2017-03-22 阿列索治疗公司 Sirp-alpha variant constructs and uses thereof
US11021694B2 (en) 2014-08-15 2021-06-01 Merck Patent Gmbh SIRP-α immunoglobulin fusion proteins
EP3180363B2 (en) 2014-08-15 2022-09-14 Merck Patent GmbH Sirp-alpha immunoglobulin fusion proteins
EP3180363B1 (en) 2014-08-15 2019-09-25 Merck Patent GmbH Sirp-alpha immunoglobulin fusion proteins
US10822411B2 (en) * 2014-09-15 2020-11-03 The Board Of Trustees Of The Leland Stanford Junior University Targeting aneurysm disease by modulating phagocytosis pathways
US20170291945A1 (en) * 2014-09-15 2017-10-12 The Board Of Trustees Of The Leland Stanford Junior University Targeting aneurysm disease by modulating phagocytosis pathways
US11253588B2 (en) 2015-02-27 2022-02-22 The Board Of Trustees Of The Leland Stanford Junior University Combination therapy for treatment of coronary artery disease
EP3811972A1 (en) * 2015-02-27 2021-04-28 The Board of Trustees of the Leland Stanford Junior University Combination therapy for treatment of atherosclerosis
EP4218812A1 (en) * 2015-02-27 2023-08-02 The Board of Trustees of the Leland Stanford Junior University Combination therapy for treatment of atherosclerosis
EP3261671A4 (en) * 2015-02-27 2018-10-24 The Board of Trustees of The Leland Stanford Junior University Combination therapy for treatment of coronary artery disease
EP3287470A4 (en) * 2015-04-24 2018-10-31 Immuneonco Biopharmaceuticals (Shanghai) Co., Ltd Novel recombinant bi-functional fusion protein and preparation and application therefor
US10358472B2 (en) 2015-05-06 2019-07-23 The Board Of Trustees Of The Leland Stanford Junior University High affinity CD47 analogs
WO2016179399A1 (en) * 2015-05-06 2016-11-10 The Board Of Trustees Of The Leland Stanford Junior University High affinity cd47 analogs
US11639376B2 (en) 2015-08-07 2023-05-02 ALX Oncology Inc. Constructs having a SIRP-α domain or variant thereof
CN108350048B (en) * 2015-08-07 2024-02-09 Alx肿瘤生物技术公司 Constructs with SIRP-alpha domains or variants thereof
EP3913051A1 (en) * 2015-08-07 2021-11-24 ALX Oncology Inc. Constructs having a sirp-alpha domain or variant thereof
WO2017027422A1 (en) 2015-08-07 2017-02-16 Alexo Therapeutics Inc. Constructs having a sirp-alpha domain or variant thereof
US11208459B2 (en) 2015-08-07 2021-12-28 ALX Oncology Inc. Constructs having a SIRP-alpha domain or variant thereof
US10696730B2 (en) 2015-08-07 2020-06-30 ALX Oncology Inc. Constructs having a SIRP-alpha domain or variant thereof
CN108350048A (en) * 2015-08-07 2018-07-31 阿列索治疗公司 Construct with SIRP- αdomains or its variant
EP3128005A1 (en) * 2015-08-07 2017-02-08 Alexo Therapeutics Inc. Sirp-alpha variant constructs and uses thereof
AU2016210755B2 (en) * 2015-08-07 2021-05-13 ALX Oncology Inc. SIRP-alpha variant constructs and uses thereof
AU2021218004B2 (en) * 2015-08-07 2023-06-15 ALX Oncology Inc. Constructs having a SIRP-alpha domain or variant thereof
RU2740672C2 (en) * 2015-08-07 2021-01-19 ЭйЭлЭкс Онколоджи Инк. Structures having sirp-alpha domain or its version
EP3913050A1 (en) * 2015-08-07 2021-11-24 ALX Oncology Inc. Sirp-alpha variant constructs and uses thereof
EP3331902A4 (en) * 2015-08-07 2019-04-03 ALX Oncology Inc. Constructs having a sirp-alpha domain or variant thereof
AU2021215151B2 (en) * 2015-08-07 2023-05-18 ALX Oncology Inc. SIRP-alpha variant constructs and uses thereof
US10259859B2 (en) 2015-08-07 2019-04-16 ALX Oncology Inc. Constructs having a SIRP-α domain or variant thereof
US10188701B2 (en) * 2015-10-01 2019-01-29 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US11547742B1 (en) 2015-10-01 2023-01-10 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US10449233B2 (en) 2015-10-01 2019-10-22 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US10493128B2 (en) 2015-10-01 2019-12-03 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US11654180B2 (en) 2015-10-01 2023-05-23 Heat Biologies, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US10525102B2 (en) 2015-10-01 2020-01-07 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US20190151413A1 (en) * 2015-10-01 2019-05-23 Heat Biologics, Inc. Compositions and methods for adjoining type i and type ii extracellular domains as heterologous chimeric proteins
US10543253B2 (en) 2015-10-01 2020-01-28 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US10660936B2 (en) 2015-10-01 2020-05-26 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US10653748B2 (en) 2015-10-01 2020-05-19 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US10646545B2 (en) 2015-10-01 2020-05-12 Heat Biologics, Inc. Compositions and methods for adjoining type I and type II extracellular domains as heterologous chimeric proteins
US11142548B2 (en) 2016-05-10 2021-10-12 Sorbonne Universite Agents that activate CD47 and their use in the treatment of inflammation
WO2017194586A1 (en) * 2016-05-10 2017-11-16 Université Pierre Et Marie Curie - Paris 6 (Upmc) Agents that activate cd47 and their use in the treatment of inflammation
US11779631B2 (en) 2016-11-03 2023-10-10 Pfizer Inc. CD47 blockade therapy by HDAC inhibitors
WO2018081898A1 (en) 2016-11-03 2018-05-11 Trillium Therapeutics Inc. Improvements in cd47 blockade therapy by hdac inhibitors
US11446315B2 (en) 2016-11-03 2022-09-20 Pf Argentum Ip Holdings Llc Enhancement of CD47 blockade therapy by proteasome inhibitors
US11897937B2 (en) 2017-01-05 2024-02-13 Kahr Medical Ltd. SIRPalpha-41BBL fusion protein and methods of use thereof
US11702458B2 (en) 2017-01-05 2023-07-18 Kahr Medical Ltd. PD1-41BBL fusion protein and methods of use thereof
EP3585418A4 (en) * 2017-02-27 2020-11-25 Shattuck Labs, Inc. Tigit- and light-based chimeric proteins
US10513548B2 (en) 2017-02-27 2019-12-24 Shattuck Labs, Inc. CSF1R-based chimeric proteins
US11332509B2 (en) 2017-02-27 2022-05-17 Shattuck Labs, Inc. Methods of making and using extracellular domain-based chimeric proteins
US11267856B2 (en) 2017-02-27 2022-03-08 Shattuck Labs, Inc. CSF1R-CD40L chimeric proteins
US11267857B2 (en) 2017-02-27 2022-03-08 Shattuck Labs, Inc. CSF1R-based chimeric proteins
US10973212B2 (en) * 2017-03-31 2021-04-13 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Genetically modified non-human animal with human or chimeric SIRPa
US11723348B2 (en) 2017-03-31 2023-08-15 Biocytogen Pharmaceuticals (Beijing) Co., Ltd. Genetically modified mice expressing humanized CD47
US10918095B2 (en) 2017-03-31 2021-02-16 Beijing Biocytogen Co., Ltd Genetically modified mice expressing humanized CD47
US11753480B2 (en) 2017-07-26 2023-09-12 Forty Seven, Inc. Anti-SIRP-alpha antibodies and related methods
US10961318B2 (en) 2017-07-26 2021-03-30 Forty Seven, Inc. Anti-SIRP-α antibodies and related methods
US11401329B2 (en) 2017-08-02 2022-08-02 Phanes Therapeutics, Inc. Anti-CD47 antibodies and uses thereof
US11529425B2 (en) * 2017-09-07 2022-12-20 Dingfu Biotarget Co., Ltd. Immunoconjugates comprising signal regulatory protein alpha
US11771764B2 (en) 2017-11-06 2023-10-03 Pfizer Inc. CD47 blockade with radiation therapy
US11891423B2 (en) 2018-04-17 2024-02-06 Hangzhou Sumgen Biotech Co., Ltd. Fusion protein binding to CD47 protein and application thereof
RU2815515C2 (en) * 2018-07-11 2024-03-18 КАР Медикал Лтд. Embodiments of sirp-alpha-4-1bbl fusion protein and methods of use thereof
US10780121B2 (en) 2018-08-29 2020-09-22 Shattuck Labs, Inc. FLT3L-based chimeric proteins
US11896618B2 (en) 2018-08-29 2024-02-13 Shattuck Labs, Inc. FLT3L-based chimeric proteins
WO2020047651A1 (en) 2018-09-04 2020-03-12 Trillium Therapeutics Inc. Cd47 blockade with parp inhibition for disease treatment
US11613564B2 (en) 2019-05-31 2023-03-28 ALX Oncology Inc. Methods of treating cancer
WO2021011544A1 (en) 2019-07-16 2021-01-21 Gilead Sciences, Inc. Hiv vaccines and methods of making and using
EP4349413A2 (en) 2019-10-18 2024-04-10 Forty Seven, Inc. Combination therapies for treating myelodysplastic syndromes and acute myeloid leukemia
WO2021076908A1 (en) 2019-10-18 2021-04-22 Forty Seven, Inc. Combination therapies for treating myelodysplastic syndromes and acute myeloid leukemia
WO2021087064A1 (en) 2019-10-31 2021-05-06 Forty Seven, Inc. Anti-cd47 and anti-cd20 based treatment of blood cancer
WO2021130638A1 (en) 2019-12-24 2021-07-01 Carna Biosciences, Inc. Diacylglycerol kinase modulating compounds
WO2021163064A2 (en) 2020-02-14 2021-08-19 Jounce Therapeutics, Inc. Antibodies and fusion proteins that bind to ccr8 and uses thereof
US11692038B2 (en) 2020-02-14 2023-07-04 Gilead Sciences, Inc. Antibodies that bind chemokine (C-C motif) receptor 8 (CCR8)
WO2022190058A1 (en) 2021-03-12 2022-09-15 Dcprime B.V. Methods of vaccination and use of cd47 blockade
WO2022221304A1 (en) 2021-04-14 2022-10-20 Gilead Sciences, Inc. CO-INHIBITION OF CD47/SIRPα BINDING AND NEDD8-ACTIVATING ENZYME E1 REGULATORY SUBUNIT FOR THE TREATMENT OF CANCER
WO2022229818A1 (en) 2021-04-27 2022-11-03 Pf Argentum Ip Holdings Llc Enhancement of cd47 blockade therapy with dhfr inhibitors
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2023076983A1 (en) 2021-10-28 2023-05-04 Gilead Sciences, Inc. Pyridizin-3(2h)-one derivatives
WO2023073580A1 (en) 2021-10-29 2023-05-04 Pfizer Inc. Enhancement of cd47 blockade with taxanes for cd47+ cancer therapy
WO2023077030A1 (en) 2021-10-29 2023-05-04 Gilead Sciences, Inc. Cd73 compounds
WO2023079438A1 (en) 2021-11-08 2023-05-11 Pfizer Inc. Enhancement of cd47 blockade therapy with anti-vegf agents
WO2023122581A2 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023122615A1 (en) 2021-12-22 2023-06-29 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023147418A1 (en) 2022-01-28 2023-08-03 Gilead Sciences, Inc. Parp7 inhibitors
WO2023178181A1 (en) 2022-03-17 2023-09-21 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
EP4245756A1 (en) 2022-03-17 2023-09-20 Gilead Sciences, Inc. Ikaros zinc finger family degraders and uses thereof
WO2023183817A1 (en) 2022-03-24 2023-09-28 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
WO2023196784A1 (en) 2022-04-05 2023-10-12 Gilead Sciences, Inc. Combinations of antibody therapies for treating colorectal cancer
WO2023205719A1 (en) 2022-04-21 2023-10-26 Gilead Sciences, Inc. Kras g12d modulating compounds
WO2023228044A1 (en) 2022-05-25 2023-11-30 Pfizer Inc. Dosing regimens of sirp alpha fusion proteins for treatment of cancer
WO2024006929A1 (en) 2022-07-01 2024-01-04 Gilead Sciences, Inc. Cd73 compounds
WO2024015741A1 (en) 2022-07-12 2024-01-18 Gilead Sciences, Inc. Hiv immunogenic polypeptides and vaccines and uses thereof
WO2024040151A1 (en) 2022-08-18 2024-02-22 Pfizer Inc. Sirp alpha fusion protein and anti-cd38 antibody combination therapies
WO2024064668A1 (en) 2022-09-21 2024-03-28 Gilead Sciences, Inc. FOCAL IONIZING RADIATION AND CD47/SIRPα DISRUPTION ANTICANCER COMBINATION THERAPY

Also Published As

Publication number Publication date
AU2009327058A1 (en) 2011-06-30
KR20110112299A (en) 2011-10-12
EA201100947A1 (en) 2012-02-28
MX2011006621A (en) 2011-07-12
BRPI0923442A2 (en) 2016-01-12
EP2379584A1 (en) 2011-10-26
JP2012512640A (en) 2012-06-07
CN102257001A (en) 2011-11-23
US20110237498A1 (en) 2011-09-29
CA2747678A1 (en) 2010-06-24

Similar Documents

Publication Publication Date Title
EP2379584A1 (en) Soluble polypeptides for use in treating autoimmune and inflammatory disorders
JP5801197B2 (en) Drugs and methods for the treatment of pain
US11926671B2 (en) Antibodies and polypeptides directed against CD127
US20130011401A1 (en) Soluble proteins for use as therapeutics
JP6917368B2 (en) Single-stranded CD27 receptor agonist protein
CN104394880B (en) Methods and compositions for modulating TOSO activity
CZ20023517A3 (en) Peptide modulating thrombopoietin receptor
KR20230086809A (en) Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
TW202012015A (en) Methods of use of cd24 for the prevention and treatment of leukemia relapse
JP2018529729A (en) Treatment of bile acid disorders
JP2009543579A (en) WSX-1 / p28 as a target for anti-inflammatory response
EP3716991A1 (en) Trpv6 inhibitors and combination therapies for treating cancers
US9617325B2 (en) Treatment of IgE-mediated disease
EP4230217A1 (en) Pharmaceutical composition for preventing or treating lupus-related diseases comprising gip derivative or long-acting conjugate thereof
EP4342487A2 (en) Pharmaceutical composition comprising gip derivative or long-acting conjugate thereof for preventing or treating pulmonary disease
EP4230219A1 (en) Pharmaceutical composition comprising glucagon/glp-1/gip triple agonist or long-acting conjugate thereof for preventing or treating lupus-related diseases
US20210046153A1 (en) Oca-b peptide conjugates and methods of treatment
WO2012116210A2 (en) Peptide for the induction of immune tolerance as treatment for systemic lupus erythematosus
TW202311293A (en) Combinations of immunotherapies and uses thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980151137.4

Country of ref document: CN

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

Ref document number: 09796708

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 3936/DELNP/2011

Country of ref document: IN

Ref document number: 2009796708

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13132400

Country of ref document: US

ENP Entry into the national phase

Ref document number: 20117014013

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011541441

Country of ref document: JP

Ref document number: 2747678

Country of ref document: CA

Ref document number: MX/A/2011/006621

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2009327058

Country of ref document: AU

Date of ref document: 20091217

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201100947

Country of ref document: EA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI0923442

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI0923442

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20110617