WO2012002562A1 - Agents thérapeutiques protéiques modifiés - Google Patents

Agents thérapeutiques protéiques modifiés Download PDF

Info

Publication number
WO2012002562A1
WO2012002562A1 PCT/JP2011/065407 JP2011065407W WO2012002562A1 WO 2012002562 A1 WO2012002562 A1 WO 2012002562A1 JP 2011065407 W JP2011065407 W JP 2011065407W WO 2012002562 A1 WO2012002562 A1 WO 2012002562A1
Authority
WO
WIPO (PCT)
Prior art keywords
tnfrii
domains
cells
fusion protein
immunoglobulin
Prior art date
Application number
PCT/JP2011/065407
Other languages
English (en)
Inventor
Yasuhiko Masuho
Hiroaki Nagashima
Kaname Kaneko
Ayaka Yamanoi
Original Assignee
Tokyo University Of Science Educational Foundation Administrative Organization
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 Tokyo University Of Science Educational Foundation Administrative Organization filed Critical Tokyo University Of Science Educational Foundation Administrative Organization
Publication of WO2012002562A1 publication Critical patent/WO2012002562A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates to a fusion protein, a therapeutic agent and a therapeutic method.
  • Tumor necrosis factors include TNFa, and TNFP (also referred to as “lymphotoxin a”).
  • Tumor necrosis factor receptor 1 also referred to as, for example, “tumor necrosis factor receptor 1 A isoform”
  • tumor necrosis factor receptor 2 also referred to as, for example, “tumor necrosis factor receptor IB” are receptors of these tumor necrosis factors. Binding of TNF to these receptors causes cellular responses of immunity or inflammation.
  • Biological drugs of TNF-a antagonists include infliximab, adalimumab, etanercept, certolizumab pegol and golimumab.
  • etanercept is a kind of Fc fusion protein consisting of two extracellular domains of the tumor necrosis factor receptor 2 (TNFRII) linked to the Fc region of IgGl (see, for example, J Immunol (1993) 151 :1548-1561).
  • TNFRII tumor necrosis factor receptor 2
  • Etanercept is able to bind to a soluble form of TNF-a (sTNF-a) and a transmembrane TNF-a (tmTNF-a), and exerts a potent clinical effect on rheumatoid arthritis (RA) (see, for example, N Engl J Med (2000) 343:1586-1593).
  • sTNF-a soluble form of TNF-a
  • tmTNF-a transmembrane TNF-a
  • RA rheumatoid arthritis
  • infliximab, adalimumab, certolizumab pegol and golimumab are anti-TNFa antibodies.
  • etanercept which is a fusion protein
  • anti-TNFa antibodies etanercept differs from other antagonists in that etanercept lacks efficacy in Crohn disease (see, for example, Gastroenterology (2001) 121:1088-1094) and is less efficacious in psoriasis (see, for example, N Engl J Med (2003) 349:2014-2022).
  • infliximab and adalimumab induced antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) in tmTNF-a-transfected cells, whereas etanercept induced no or low levels of cytotoxicities (see, for example, Cytokine (2009) 45:124-131, ARTHRITIS & RHEUMATISM (2008) 58, 1248-1257 and Cancer Res (2008) 68:3863-3872).
  • Etanercept can bind to TNF-a in 1 : 1 molar ratio, whereas 3 molecules of other monoclonal antibody (mAb) agents bind to one molecule of TNF-a, and the dissociation rate of etanercept is more rapid than those of the others (see, for example, J Pharmacol Exp Ther (2002) 301 :418-426).
  • mAb monoclonal antibody
  • etanercept is reported to have a half-life of 70 hours to 100 hours in human blood (see, for example, J Pharm Pharmacol. (2005) ,57 (11), 1407-13), while infliximab and
  • adalimumab which are anti-TNFa antibodies, have retention time of about 3 weeks. As demonstrated above, the half-lives of infliximab and adalimumab in blood are known to be longer than that of etanercept.
  • Anti-CD20 mAbs having Fc domains connected in tandem bind to Fey receptors, FcyRIA, FcyRIIA, FcyRIIB and FcyRIIIA with higher avidities than the parental mAb, resulting in enhanced ADCC (see, for example, Mol Immunol (2008) 45:2752-2763 and International Publication WO2007/100083 pamphlet).
  • fusion proteins typified by etanercept
  • etanercept are desired to have enhanced ADCC activities and enhanced CDC activities, and an elongated half-lives in blood, so as to broaden the applications thereof to treatment of a wide range of diseases.
  • Anti-CD20 mAbs to which Fc domains are tandemly connected have been confirmed to have high affinity for Fey receptors compared to ordinary anti-CD20 mAbs, and have an increased ADCC activity. It has also been confirmed that CDC activity is not affected even in the case of a fusion protein of a Fc multimer and a variable region of an anti-CD20 mAb. Further, it has been confirmed that an antibody obtained by tandemly connecting two
  • CAMPATH-IH antibodies has a lower CDC activity than that of a single CAMPATH-IH antibody. That is, contribution to CDC activity varies depending on the type of antibody and the type of target site. Therefore, tandemly connected Fc regions do not necessarily increases ADCC activity and CDC activity, and further technical development has been requested with a view to enhancing ADCC activity and CDC activity.
  • An object of the present invention is provision of a fusion protein which has an enhanced ADCC activity and an enhanced CDC activity, an elongated half-life in blood, and which is capable of exerting high therapeutic effects against various diseases.
  • a fusion protein comprising : a first region comprising an extracellular domain of a cytokine receptor; and a second region comprising plural immunoglobulin Fc domains or plural single-chain immunoglobulin Fc domains, and fused with the first region,
  • immunoglobulin Fc domains comprise tandemly repeated immunoglobulin Fc domains or tandemly repeated single-chain immunoglobulin Fc domains.
  • cytokine receptor is a TNFa receptor.
  • ⁇ 3> The fusion protein according to ⁇ 2>, wherein the TNFa receptor is a TNFa receptor II.
  • ⁇ 4> The fusion protein according to any one of ⁇ 1> to ⁇ 3>, wherein the immunoglobulin is IgG.
  • ⁇ 5> The fusion protein according to ⁇ 4>, wherein the IgG is human IgGl .
  • ⁇ 6> The fusion protein according to any one of ⁇ 1> to ⁇ 5>, wherein adjacent domains in the tandemly repeated immunoglobulin Fc domains or the tandemly repeated single-chain immunoglobulin Fc domains are linked to each other via a linker peptide.
  • fusion protein according to ⁇ 1> to ⁇ 6>, wherein the linker peptide is a trimer of glycine-glycine-glycine-glycine-serine.
  • tandemly repeated immunoglobulin Fc domains or the tandemly repeated single-chain immunoglobulin Fc domains are composed of two or three immunoglobulin Fc domains that are connected in series, or of two or three single-chain immunoglobulin Fc domains that are connected in series.
  • a therapeutic agent for treating an inflammation-related disease comprising the fusion protein of any one of ⁇ 1> to ⁇ 8>.
  • ⁇ 10> The therapeutic agent according to ⁇ 9>, wherein the inflammation-related disease is Crohn disease.
  • ⁇ 11> A method for treating an inflammation-related disease, comprising administering the fusion protein of any one of ⁇ 1> to ⁇ 8> to a patient in need thereof.
  • ⁇ 12> The method for treating an inflammation-related disease according to ⁇ 11>, wherein the inflammation-related disease is Crohn disease.
  • a fusion protein which has an enhanced ADCC activity and an enhanced CDC activity, an elongated half-life in blood, and which is capable of exerting high therapeutic effects against various diseases, can be provided.
  • Fig. 1 A shows cDN A constructs of TNFRII-Fc and TNFRII-Fc-Fc.
  • Fig. 1 B shows schematic illustrations of TNFRII-Fc, TNFRII-Fc-Fc and
  • Figs. 2A, 2B, 2C and 2D show gel-filtration HPLC.
  • Figs. 2E and 2F show
  • FIG. 3 A shows binding activities of Fc fusion proteins to sTNF-a.
  • Fig. 3B shows neutralizing activities of Fc fusion proteins against sTNF-ot
  • FIGs. 4A and 4B show binding activities of Fc fusion proteins to tmTNF-a on CHO-DG44.
  • Figs. 5 A and 5B show CDC activities of Fc fusion proteins.
  • Figs. 6 A, 6B, 6C, 6D and 6E show binding activities of Fc fusion proteins to Fey receptors.
  • Figs. 7A, 7B and 7C show ADCC activities of Fc fusion proteins.
  • Fig. 8 shows the effect of fusion proteins on the body weight of BALB/c mice with
  • the fusion protein of the invention is a fusion protein including a first region including an extracellular domain of a cytokine receptor and a second region which is fused to the first region, and which includes tandemly repeated immunoglobulin Fc domains or tandemly repeated single-chain immunoglobulin Fc domains.
  • the fusion protein of the invention exhibits increased frequency and strength of binding to Clq, which is a component of a complement, and exhibits an enhanced ADCC activity and an enhanced CDC activity, due to fusion of the first region including an extracellular domain of a specific cytokine receptor and a second region having a specific structure including tandemly repeated immunoglobulin Fc domains or tandemly repeated single-chain immunoglobulin Fc domains. Further, it is presumed that an elongated half-life of the fusion protein of the invention in blood results from the fusion of the first region and the second region. Therefore, it is presumed that these are the reasons why the fusion protein of the invention produces high therapeutic effects against a wide range of inflammation-related diseases, to which conventional fusion proteins are ineffective.
  • the first region includes an extracellular domain of a cytokine receptor.
  • cytokine receptor examples include: interleukin receptors such as TNFa receptor, interleukine-1 receptor, IL-6 receptor and IL-17 receptor; G-CSF receptor; GM-CSF receptor; receptors for vascular endothelial growth factors (VEGF), epidermal growth factors (EGF), thrombopoetin, erythropoietin, CC chemokines, C chemokines, CXC chemokines and CX 3 C chemokines; cell adhesion molecules such as integrins; erythropoietin receptor, thrombopoietin receptor, epidermal growth factor receptor, platelet-derived growth factor receptor, vascular endothelial growth factors (VEGF) receptor and insulin receptor.
  • interleukin receptors such as TNFa receptor, interleukine-1 receptor, IL-6 receptor and IL-17 receptor
  • G-CSF receptor G-CSF receptor
  • GM-CSF receptor examples include receptors for vascular endotheli
  • the cytokine receptor include TNFa receptor, EGF receptor and VEGF receptor.
  • the cytokine receptor is particularly preferably TNFa receptor, from the viewpoint of suppressing inflammation.
  • TNFa receptor I and TNFa receptor II are involved in many TNF-a-related inflammations (Crit. Rev. Eukaryot. Gene. Expr. (2010) 20, 87-103). From this viewpoint, examples of TNFa receptors include TNFa receptor I and TNFa receptor II. Further, TNFa receptor II is most preferable from the viewpoint of the strength of the affinity for TNFa.
  • the cytokine receptor is preferably a cytokine receptor derived from human.
  • the cytokine receptor is not limited to a human-derived cytokine receptor, and known cytokine receptors having activities against the binding site comparable to the activities of human-derived cytokine receptors may be used.
  • the extracellular domain is not limited as long as the extracellular domain is an extracellular domain of the cytokine receptor described above.
  • the extracellular domain of TNFa receptor II is hereinafter sometimes abbreviated to "TNFRII".
  • the first region may consists of at least one (i.e., one, or more than one) extracellular domain of a cytokine receptor.
  • the second region includes tandemly repeated immunoglobulin Fc domains or tandemly repeated single-chain immunoglobulin Fc domains.
  • the immunoglobulin may be any of IgA, IgG, IgD, IgE or IgM, and is preferably IgG from the viewpoints of binding affinity for a Fc receptor, activation potency for complements and retention properties in the body.
  • the IgG may be any of human IgGl, human IgG2, human IgG3 or humanIgG4, and is preferably human IgGl from the viewpoints of purification by protein A, binding affinity for Fc receptors, activation potency for complements and retention properties in the body.
  • immunoglobulin Fc domains are immunoglobulin Fc domains derived from the same type of immunoglobulin.
  • the second region in the invention may include unmodified immunoglobulin Fc domains.
  • the second region may include single-chain immunoglobulin Fc domains.
  • single-chain immunoglobulin Fc domain refers to either one of the two chains constituting an immunoglobulin Fc domain, which may be obtainable by separation from the other chain by cleavage of disulfide bonds linking the two chains.
  • each of the linker peptides for connecting immunoglobulin Fc domains may be any of the linker peptides described below. That is, the amino acid sequence of the linker peptide may be any amino acid sequence as long as (i) the linker peptide has a flexible structure, (ii) the linker peptide has a certain length, and (iii) the linker peptide is not degraded in the blood.
  • linker peptide examples include a monomer of glycine-glycine-glycine-glycine-serine, a dimer of glycine-glycine-glycine-glycine-serine, a trimer of
  • the linker peptide is more preferably a trimer of glycine-glycine-glycine-glycine-serine (SEQ ID NO: 6). These linkers may be used also in a case in which the second region includes single-chain immunoglobulin Fc domains.
  • the number of the immunoglobulin Fc domains included in the protein of the invention is normally from two to five. It is preferable that the protein includes two or three tandemly repeated immunoglobulin Fc domains.
  • the number of the single-chain immunoglobulin Fc domains included in the protein of the invention is normally from two to five. It is preferable that the protein includes two or three tandemly repeated single-chain immunoglobulin Fc domains.
  • the tandemly repeated immunoglobulin Fc domains or the tandemly repeated single-chain immunoglobulin Fc domains include a hinge region of immunoglobulin for connecting the first region including an extracellular domain of a cytokine receptor and the second region.
  • the hinge region may be any hinge region of any human immunoglobulin (refer to, for example, J. Immunol. (2006), 177, 1129-1138), and is not limited to a hinge region having a naturally-occurring sequence as long as the function as a hinge region of a human immunoglobulin can be carried out.
  • the second region may consists of tandemly repeated immunoglobulin Fc domains or tandemly repeated single-chain immunoglobulin Fc domains (including the hinge region).
  • the fusion protein is a protein formed through transcription of a gene encoding the first region and a gene encoding the second region in an integrated manner, and an integrated expression of the first and second regions from the transcript.
  • the fusion protein of the invention is preferably a fusion protein including a first region including an extracellular domain of TNF a receptor, and a second region which is fused with the first region, and which includes two human IgG Fc domains of that are linked in series via linker peptides each including (G 4 S). It is also preferable that the fusion protein of the invention is a fusion protein including a first region including an extracellular domain of TNF a receptor, and a second region which is fused with the first region, and which includes two single-chain human IgG Fc domains that are linked in series via a linker peptide including (G 4 S).
  • the fusion protein of the invention is more preferably a fusion protein including a first region including an extracellular domain of TNFa receptor II, and a second region which is fused with the first region, and which includes two human IgGl Fc domains that are linked in series via linker peptides each including (G 4 S).
  • the fusion protein is more preferably a fusion protein including a first region including an extracellular domain of TNFa receptor II, and a second region which is fused with the first region, and which includes two single-chain human IgGl Fc domains that are linked in series via a linker peptide including (G 4 S).
  • the fusion protein can be prepared according to known methods, such as genetic recombination methods using a cDNA encoding the above-described protein, and chemical crosslinking methods for proteins.
  • the fusion protein may consist of the first and the second regions that are fused with each other.
  • the therapeutic agent of the invention is a therapeutic agent for treating a
  • inflammation-related disease which includes the above-described fusion protein.
  • the therapeutic method of the invention is a therapeutic method for treating an inflammation-related disease, the method including administering the above-described fusion protein to a patient in need thereof.
  • the inflammation-related disease include inflammatory diseases, autoimmune diseases and central demyelinating diseases.
  • the therapeutic agent and the therapeutic method are more suitable for treating Crohn disease, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, plaque psoriasis, polyarticular juvenile idiopathic arthritis, multiple sclerosis, juvenile idiopathic arthritis, Wegener's granulomatosis, or the like.
  • Examples of the Crohn disease include Crohn disease, fistulizing Crohn disease, moderate to severe Crohn disease and ulcerative colitis.
  • the therapeutic agent of the invention may further include a pharmaceutically acceptable carrier or additive such as a preservative, a stabilizer, a salt such as sodium chloride, or a buffer such as sodium phosphate. Specific examples thereof include sterile water, physiological saline, stabilizers, excipients, buffers, preservatives, detergents, binders and protein stabilizers such as polyethylene glycol. If necessary, the therapeutic agent of the invention may further include a suspending agent, a solubilizer, an isotonizing agent, an adsorption inhibitor, a diluent, a filler, a pH modifier, a soothing agent, a sulfur-containing reducing agent or an antioxidant.
  • the administration pathway of the therapeutic agent of the invention is not specifically limited, and is preferably via a parenteral route, for example, by injection (such as subcutaneous, intravenous or intramuscular injection).
  • the fusion protein of the invention may be purified by a known method, such as protein A affinity chromatography, ion-exchange chromatography, or gel filtration
  • a suitable dosage may be determined in accordance with the type of disease to be treated.
  • the dosage in terms of the fusion protein amount per week may be, for example, from 10 mg to 100 mg, and is preferably from 10 mg to 50 mg, and more preferably from 5 mg to 20 mg.
  • CHO-DG44 cells were purchased from Invitrogen (Carlsbad, CA) and cultured in IMDM containing 10% fetal bovine serum (FBS).
  • FBS fetal bovine serum
  • L929 cells and KHYG-1 cells were kindly provided by the Institute of Development, Aging and Cancer, Tohoku University (Sendai, Miyagi).
  • L929 cells were cultured in DMEM containing 10% FBS, 100 U/ml penicillin and 100 ⁇ g/ml streptomycin
  • KYHG-1 cells were cultured in RPMI1640 containing 10% FBS, 10 ng/ml IL-2 (Wako Pure Chemical Industries, Osaka), 100 U/ml penicillin and 100 ⁇ g/ml streptomycin.
  • the cDNAs encoding the TNFRII-Fc and the TNFRII-Fc-Fc were constructed using PCR and restriction enzymes.
  • the cDNA encoding the extracellular domain of TNFRII was assembled with the cDNA of the human IgGl Fc region (from hinge to CH3 domain) to construct the cDNA of TNFRII-Fc in pcDNA3.1/Zeo (Invitrogen, Carlsbad, CA) as shown in Fig. 1 A.
  • the cDNA of the tandemly repeated Fc was connected to the downstream of the cDNA encoding the extracellular domain of TNFRII.
  • the gene encoding the flexible peptide linker (G 4 S)3 was inserted between the two Fc cDNAs.
  • the linearized expression vectors were transfected into CHO-DG44 cells with TransFast
  • Transfection Reagent Promega, Madison, WI
  • the transfected cells were cultured at 37°C in IMDM containing 2% FBS and 100 ⁇ / ⁇ zeocin (Invitrogen) in flat-bottomed BD Falcon 96-well microplates (BD Biosciences, San Jose, CA) for a week. After the cells were cloned by limited dilution, the cloned cells were cultured in CD CHO medium (Invitrogen).
  • TNFRII-Fc and TNFRII-Fc-Fc were purified from the culture supernatants by a protein A-agarose (Santa Cruz Biotechnology, Santa Cruz, CA) column chromatography and subsequent gel-filtration HPLC on a TSK-GEL G3000SWXL (Tosoh, Tokyo) as previously described (Mol Immunol (2008) 45:2752-2763).
  • the purified Fc fusion proteins were concentrated by ultrafiltration using Amicon Ultra-4 50,000 MWCO (Millipore, Billerica, MA) in 20 mM phosphate buffer (pH7.4) containing 0.14 M NaCl (PBS). The purity of Fc fusion proteins were analyzed by SDS-PAGE under reducing and non-reducing conditions.
  • the biochemical properties of the Fc fusion proteins were analyzed by gel-filtration HPLC and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) as described previously (Mol Immunol (2008) 45:2752-2763). Briefly, the fusion proteins were analyzed by gel-filtration HPLC on a Protein Pak G3000SWXL column (Tosoh, Tokyo, Japan) at a flow rate of 1 ml/min in 0.1 M sodium phosphate (pH 6.8). In addition, the fusion proteins were analyzed by SDS-PAGE under reducing and non-reducing conditions on 10% and 6% polyacrylamide gels, respectively.
  • the number of free sulfhydryl residues in the individual fusion proteins was measured using 5,5'-dithiobis-(2-nitrobenzoic acid) (DTNB) according to the manufacturer's instructions.
  • the protein concentrations were determined by measurement of the absorbance at 280 nm.
  • the protein solutions were allowed to react with 4 mM DTNB in 0.1 M phosphate buffer (pH 8.0) containing 1 mM EDTA at 37°C for 15 min.
  • the number of free sulfhydryl residues was determined by measurement of the absorbance at 412 nm.
  • Human KH YG- 1 cells stably expressing human FcyRIIIA were developed by culturing of the cells with the culture supernatants of PL AT- A cells that were transfected with retroviral vector pMXs-puro containing the FcyRIIIA cDNA. Cells highly expressing FcyRIIIA were sorted by flow cytometry and were cultured in a similar manner to the parental KHYG-1 cells.
  • TNFRII-Fc and TNFRII-Fc-Fc are shown schematically in Fig. IB, and the calculated MWs are approximately 150kDa and 200kDa, respectively. The results are shown in Figs. 2A, 2B, 2C, 2D, 2E and 2F.
  • TNFRII-Fc(2A) is separated by gel-filtration HPLC.
  • TNFRII-Fc-Fc (Protein A-purified) proteins (2B) are purified by protein A-agarose and separated by gel-filtration HPLC.
  • TNFRII-Fc-Fc proteins (2C) and TNFRII-Fc-Fc (single) proteins (2D) are separated by gel-filtration HPLC. These proteins were
  • Lane 1 TNFRII-Fc
  • lane 2 TNFRII-Fc-Fc
  • lane 3 TNFRII-Fc-Fc (single).
  • TNFRII-Fc (Fig. 2A) and TNFRII-Fc-Fc (Fig. 2B) were purified from the culture supernatants of the respective transformed cells by means of protein A affinity
  • TNFRII-Fc-Fc proteins of different molecular weights (Fig. 2B); one larger and the other smaller than TNFRII-Fc. Those two TNFRII-Fc-Fc proteins were analyzed by SDS-PAGE. Under reducing conditions, both proteins showed a single band of 100 kDa, which is equal to the calculated value of a single chain of TNFRII-Fc-Fc (Fig. 2C). Under non-reducing conditions, the large and small TNFRII-Fc-Fc molecules showed a molecular weight of about 200 kDa and 80 kDa, respectively (Fig. 2D).
  • the smaller molecule is a half molecule of TNFRII-Fc-Fc without disulfide bonds between the two H chains at the hinge region.
  • the smaller molecule which is herein designated as "TNFRII-Fc-Fc (single)"
  • TNFRII-Fc-Fc single
  • the free sulfhydryl residues of these molecules were measured using DTNB, and TNFRII-Fc,
  • TNFRII-Fc-Fc and TNFRII-Fc-Fc had 0.64, 0.96 and 0.46 free sulfhydryl residues per molecule, respectively.
  • TNFRII-Fc-Fc Binding activities of the Fc fusion proteins to a soluble form of TNF-a (sTNF-a) were determined by ELISA using sTNF-a-coated plates (Fig. 3 A).
  • the dose response curve of TNFRII-Fc-Fc was very close to that of TNFRII-Fc, implying that multimerization of Fc domain does not affect its TNF-a binding activity.
  • TNFRII-Fc-Fc single gave an absorbance of 0.6 at the concentration that was 10 to 20 times higher than that of
  • Human Fc means a Fc fragment obtained by cleaving human IgG (available from Takara Bio Inc.) with papain, and the same applies hereinafter.
  • TNFRII-Fc-Fc was close to that of TNFRII-Fc.
  • TNFRII-Fc-Fc single gave 50% neutralizing activity at the concentration that was 10 to 20 times higher than that of TNFRII-Fc-Fc, suggesting its low neutralizing activity for sTNF-a. Therefore, these results indicate that the binding and neutralizing activities of TNFRII for sTNF-a is not influenced by Fc multimerization.
  • the CHO-DG44 stably expressing tmTNF-a was established by expression of tmTNF-a that has two mutations (R77T and S78T) to resist to TACE-mediated cleavage (J Immunol (2001) 166:130-136).
  • the cDNA encoding the mutated tmTNF-a was cloned into pcDNA3.1/Zeo expression vector and transfected into CHO-DG44 with TransFast
  • CHO-DG44/tmTNF-a was used to evaluate the binding activities of Fc fusion proteins to the tmTNF-a on the cell surface. 1.33 ⁇ 10 5 cells/ml CHO-DG44/tmTNF-a was blocked in Cytometry buffer (PBS, 0.4% BSA, 3 mM EDTA) at 4°C for 30 min. After the centrifugation at 600 ⁇ g at 4°C for 5 min, the supematants were discarded and the cells were allowed to react with 1 ⁇ g/ml Fc fusion proteins in 100 ⁇ of Cytometry buffer at 4°C for 30 min.
  • Cytometry buffer PBS, 0.4% BSA, 3 mM EDTA
  • CHO-DG44 cells that exhibit relatively high expression of tmTNF-a were used, and, in Fig. 4B, CHO-DG44 cells that exhibit relatively low expression of tmTNF-a were used.
  • TNFRII-Fc and TNFRII-Fc-Fc bound to tmTNF-a at the same intensities in both of the cell lines with high and low expressions of tmTNF-a.
  • the binding activity of TNFRII-Fc-Fc (single) to tmTNF-a was lower than that of TNFRII-Fc-Fc.
  • CDC assay was performed using CHO-DG44/tmTNF-a and a low-expressing CHO-DG44/tmTNF-a as target cells. After the CHO-DG44/tmTNF-a were centrifuged at 300 x g at room temperature for 5 min, the cells were then suspended at 10 x 10 5 cells/ml in CDC Buffer (RPMI1640 containing 0.1% BSA, 20 mM HEPES (pH7.2), 100 U/ml penicillin and 100 ⁇ g/ml streptomycin).
  • CDC Buffer RPMI1640 containing 0.1% BSA, 20 mM HEPES (pH7.2), 100 U/ml penicillin and 100 ⁇ g/ml streptomycin.
  • CDC cytotoxicity (%) 100 x (background fluorescence - sample fluorescence) / background fluorescence. The cytotoxic activities were measured in triplicate, and the results were expressed as the mean and SD.
  • CDC activities of Fc fusion proteins were determined using baby rabbit complement and tmTNF-a-expressing CHO-DG44 cells.
  • CHO-DG44 cells highly expressing tmTNF-a were used, and, in Fig. 5B, CHO-DG44 cells with a low expression of tmTNF-a were used.
  • TNFRII-Fc-Fc at 2.2 nM was equivalent to TNFRII-Fc at 6.6 nM, implying that TNFRII-Fc-Fc would be 3 times as potent as TNFRII-Fc in CDC.
  • TNFRII-Fc-Fc (single) was less active than TNFRII-Fc-Fc and close to TNFRII-Fc in CDC. Similar results were found with CHO-DG44 of lower tmTNF-a expression (Fig. 5B). These results suggest that Fc multimenzation enhances the CDC activity of Fc fusion protein.
  • Binding activities of Fc fusion proteins to individual Fey receptors were measured by ELISA as previously describe (Mol Immunol (2008) 45:2752-2763). Briefly, FcyRIA, FcyRIIA, FcyRIIB, FcyRIIIA (V158) and FcyRIIIA (F158) in PBS were coated on 96-well microplates in the forms of their extracellular domains fused with glutathione S-transferase, and HRP-conjugated goat anti-human IgG gamma chain was used as the secondary antibody. The enzyme activities were measured with 3,3',5,5'-tetramethylbenzidine.
  • Binding activities of Fc fusion proteins to Fey receptors were determined by ELISA to examine whether Fc multimenzation augments the binding activity (Figs. 6A to 6E).
  • the binding activity of TNFRII-Fc-Fc to FcyRIIIA (VI 58) (Fig. 6D) was about 20 times higher than that of TNFRII-Fc, and the binding activities of TNFRII-Fc-Fc to FcyRIA (Fig. 6 A), FcyRIIA (Fig. 6B), FcyRIIB (Fig. 6C) and FcyRIIIA (F158) (Fig. 6E) were about 10 times higher than those of TNFRII-Fc.
  • TNFRII-Fc-Fc (single) was weaker than TNFRII-Fc-Fc, and as potent as TNFRII-Fc in binding activities to all Fey receptors.
  • FcyRIIIA has two genotypes at amino acid 158 and that FcyRIIIA (VI 58) binds to human IgG more strongly than FcyRIIIA (F158) (J Immunol (1993) 151:6429-6439), the enhancement of binding activity to FcyRIIIA by Fc multimenzation has nothing to do with the genotype. Taking together, these results suggest that Fc multimerization augments the binding activities of an Fc fusion protein to all Fey receptors.
  • CHO-DG44/tmTNF-a cells were used as the target cells of ADCC.
  • the cells were suspended in HBSS-F (Hanks Balanced Salt Solution containing 10 mM HEPES and 5% FBS), and the cell suspension was centrifuged at 600 ⁇ g at 4°C for 5 min. The supernatant was discarded, and the cells were resuspended in 500 ⁇ of HBSS-F. 12.5 ⁇ of 1 mM Calcein-AM (Dojindo Molecular Technologies, Inc., Kumamoto) was then added to the cell suspension and the cells were incubated at 37°C for 30 min.
  • ADCC Buffer RPMI1640, 2 mM L-glutamine, 10 mM HEPES at pH 7.2, lOOU/ml penicillin and 100 ⁇ g/ml streptomycin
  • the cells were suspended in ADCC Buffer at 2 x 10 5 cells/ml.
  • 50 ⁇ CHO-DG44/tmTNF-a stained with Calcein-AM suspensions 0.1 10 5 cells/well
  • 50 ⁇ serially diluted Fc fusion proteins or human IgG Fc in the wells of round-bottomed BD Falcon 96-well microplates (BD Biosciences), and the plates were incubated at 37°C for 30 min.
  • KHYG-1 /FcyRIIIA cells were developed by transfection of human FcyRIIIA to human NK cell line KHYG-1 and selection of stably expressing cells (Kobayashi et al., 2009). 50 ⁇ of 50 ⁇ 10 5 , 25 10 5 or 12.5 ⁇ 10 5 cells/ml KH YG- 1 /FcyRIII A (2.5 ⁇ 10 5 , 1.25 ⁇ 10 5 or 0.625 ⁇ 10 5 cells/well) was added to the individual wells. The ratios of effector cells to target cells (E/T) were 25/1, 12.5/1 and 6.25/1.
  • the plates were incubated at 37°C for 4 hours and then centrifuged at 300 x g for 10 min.
  • the supernatants 50 ⁇
  • the supernatants 50 ⁇
  • the fluorescence intensities of the Calcein-AM released from the damaged CHO-DG44/tmTNF-a were measured using ARVO SX 1420 multilabel counter (Perkin Elmer, Waltham, MA) at the excitation of 485 nm and at the emission of 538 run.
  • ADCC cytotoxicities (%) were calculated according to the following formula.
  • the fluorescence intensity of the well treated with 1% Triton-XlOO was used as the maximum cytotoxicity.
  • ADCC cytotoxicity 100 x (sample fluorescence - background fluorescence) / (1% Triton-XlOO fluorescence - background fluorescence). ADCC activities were measurements in triplicate, and the results were expressed as the mean and SD.
  • ADCC activities of Fc fusion proteins were determined using KHYG-1 cells transfected FcyRIIIA (KH YG- 1 /FcyRIII A) and tmTNF-a-expressing CHO-DG44 cells (CHO-DG44/tmTNF-a).
  • Fig. 7A shows that the ADCC activities of three Fc fusion proteins against tmTNF-a highly expressing CHO-DG44 by KHYG-1 /FcyRIIIA at the E/T ratio of 25/1. Human IgG Fc alone did not bind to target cells and showed no significant
  • TNFRII-Fc-Fc was found to be cytotoxic at the concentrations of 0.74 nM and above, and TNFRII-Fc at the concentrations of 6.67 and 20 nM showed the cytotoxicity as much as TNFRII-Fc-Fc of 0.74 nM.
  • TNFRII-Fc-Fc (single) was as potent as TNFRII-Fc in ADCC. Also at the lower E/T ratios (12.5/1 and 6.25/1), the ADCC activity of
  • TNFRII-Fc-Fc was more potent than TNFRII-Fc at every concentration (Fig. 7B).
  • TNFRII-Fc-Fc would exert more potent ADCC activity than the ordinary TNFRII-Fc against tmTNF-a-expressing target cells.
  • mice blood collection from the mice was carried out 12 hours, 24hours, 48 hours (2 days), 72 hours (3 days), 96 hours (4 days), 120 hours (5 days), 168 hours (7 days), 192 hours (8 days), 216 hours (9 days) and 264 hours (11 days) after the administration of the Fc fusion protein, and the collected blood was processed in a manner similar to the above.
  • Goat anti human TNFRII polyclonal antibody in PBS (final 1.0 ⁇ ) was added, in an amount of 50 ⁇ , into the individual wells of an ELISA plate, and was allowed to stand at 4 °C overnight, thereby immobilizing the antibody.
  • TNFRII-Fc and TNFRII-Fc-Fc are compared, the half-life of TNFRII-Fc-Fc (single) is clearly elongated.
  • TNFRII-Fc-Fc has an enhanced CDC activity and an enhanced ADCC activity, as well as an elongated half-life. Therefore, TNFRII-Fc-Fc (single) is considered to be more effective as a therapeutic agent for inflammation-related diseases, as compared to TNFRII-Fc.
  • etanercept which is a TNFRII-Fc type fusion protein
  • etanercept has been reported to have a half-life of from about 70 hours to about 100 hours in human (J Pharm Pharmacol. (2005) ,57 (11), 1407-13). It is known that the half-life of drugs in human blood is longer than that in mouse blood, and it is estimated that the TNFRII-Fc-Fc (single) according to the invention has a half-life in human of from about 100 hours to about 200 hours. The high efficacy of the fusion protein of the invention is also demonstrated by this result.
  • TNF-a is known to be involved in ulcerative colitis induced by DSS administration (J. Clin. Invest. (2008) 118, 560-570). In view of this, a study was carried out using a
  • TNFRII-Fc -Fc and TNFRII-Fc -Fc which are fusion proteins according to the invention.
  • mice (BALB/c, male, 5-month age) were randomly divided into 14 groups, each group consisting of 5 mice. The weights thereof on Day 0 were measured. Thereafter, the weights of the mice were measured every 24 hours, and the recording of the weights was continued until Day 21. Each mouse was allowed to freely ingest potable water or potable water containing 5%(w/v) dextran sodium sulfate (DSS) dissolved therein, for 7 days.
  • DSS dextran sodium sulfate
  • Fig. 8 The results are shown in Fig. 8.
  • Six week-old BALB/c mice (five mice per group) received i.v. injections of PBS (trigona), 0.13 nmol/body of etanercept (square), 0.13 nmol/body of TNFRII-Fc-Fc (crosses) or 0.13 nmol/body of TNFRII-Fc-Fc (single) (round) five times over a 21 -day treatment period.
  • diamond marks indicate a change of the weight of mice that did not ingest DSS.
  • samples were analyzed in triplicate, and the mean values ⁇ S.D thereof are shown.
  • mice that did not ingest DSS exhibited an about 20% increase in body weight within the 21 days during which the experiment was carried out.
  • the mice which ingested DSS, and to which PBS was administered exhibited a decrease in body weight from around Day 5 of the DSS ingest, and exhibited the minimum weight on Day 11.
  • CDC assay is performed using mtTNF-a-transfected Jurkat cells or THP-1 cells activated with PMA (phorbol myristate acetate) as target cells. After these cells are centrifuged at 300 x g at room temperature for 5 min, the cells are then suspended at 10 ⁇ 10 5 cells/ml in CDC Buffer (RPMI1640 containing 0.1% BSA, 20 mM HEPES (pH7.2), 100 U/ml penicillin and 100 ⁇ g/ml streptomycin). 50 ⁇ of cell suspensions are dispensed into individual wells of uClear fluorescence black plates with transparent bottom and 50 ⁇ of serially diluted Fc fusion proteins are added to the cell suspensions.
  • CDC Buffer RPMI1640 containing 0.1% BSA, 20 mM HEPES (pH7.2), 100 U/ml penicillin and 100 ⁇ g/ml streptomycin.
  • CDC cytotoxicity (%) 100 ⁇ (background fluorescence - sample fluorescence) / background fluorescence. The cytotoxic activities are measured in triplicate, and the results are expressed as the means and Standard deviations.
  • CDC activities of Fc fusion proteins are determined using baby rabbit complement and tmTNF-a-expressing Jurkat cells or activated THP-1 cells [0111] (Example 10-2) ADCC assay using mtTNF-ct-transfected Jurkat cells or PMA-activated THP-1 cells
  • MtTNF-a-transfected Jurkat cells or PMA-activated THP-1 cells are used as the target cells of ADCC.
  • the cells are suspended in HBSS-F (Hanks Balanced Salt Solution containing 10 mM HEPES and 5% FBS), and the cell suspension is centrifuged at 600 ⁇ g at 4°C for 5 min. The supernatant is discarded, and the cells are resuspended in 500 ⁇ of HBSS-F. 12.5 ⁇ of 1 mM Calcein-AM (Dojindo Molecular Technologies, Inc., Kumamoto) is then added to the cell suspension and the cells are incubated at 37°C for 30 min.
  • ADCC Buffer RPMI1640, 2 mM L-glutamine, 10 mM HEPES at pH 7.2, lOOU/ml penicillin and 100 ⁇ g/ml streptomycin
  • the cells are suspended in ADCC Buffer at 2 x 10 5 cells/ml.
  • 50 ⁇ cell suspension of mtTNF-a-transfected Jurkat cells or PMA-activated THP-1 cells that are stained with Calcein-AM suspensions (0.1 x 10 5 cells/well) are mixed with 50 ⁇ serially diluted Fc fusion proteins or human IgG Fc in the wells of round-bottomed BD Falcon 96- well microplates, and the plates are incubated at 37°C for 30 min.
  • KH YG- 1 /FcyRIII A cells were developed by transfection of human FcyRIIIA to human NK cell line KHYG-1 and selection of stably expressing cells (Kobayashi et al., 2009). 50 ⁇ of 50 x 10 5 , 25 x 10 5 or 12.5 ⁇ 10 5 cells/ml KHYG-1 /FcyRIII A (2.5 10 5 , 1.25 10 5 or 0.625 x 10 5 cells/well) is added to the individual wells. The ratios of effector cells to target cells (E/T) were 25/1 , 12.5/1 and 6.25/1.
  • the plates are incubated at 37°C for 4 hours and then centrifuged at 300 x g for 10 min.
  • the supernatants (50 ⁇ ) are transferred into individual wells of uClear fluorescence black plates with transparent bottom (Greiner Bio-one, Frickenhausen, Germany).
  • the fluorescence intensities of the Calcein-AM released from the damaged tagete cells are measured using ARVO SX 1420 multilabel counter (Perkin Elmer, Waltham, MA) at the excitation of 485 nm and at the emission of 538 nm.
  • cytotoxicities (%) are calculated according to the following formula. The fluorescence intensity of the well treated with 1% Triton-XlOO is used as the maximum cytotoxicity.
  • ADCC cytotoxicity 100 x (sample fluorescence - background fluorescence) / (1% Triton-XlOO fluorescence - background fluorescence). ADCC activities are measurements in triplicate, and the results are expressed as the mean and SD.
  • ADCC activities of Fc fusion proteins are determined using KHYG-1 cells
  • FcyRIII A KHYG-1 /FcyRIII A
  • mtTNF-a-transfected Jurkat cells or PMA-activated THP-1 cells as target cells.
  • TNFRII-Fc -Fc and TNFRII-Fc -Fc exhibits cytotoxic activity against macrophages and T cells, each of which expresses tmTNFa and strongly induces inflammation, via CDC or ADCC. In immune inflammatory diseases, these T cells play a pivotal role in the pathogenecis (Rheumatology (2010), 49, 1215-1228).
  • TNFRII-Fc-Fc and TNFRII-Fc-Fc have strong activity, and are expected to not only simply neutralize secreted TNFa, but also to injure cells that produces inflammatory proteins such as TNFa, thereby suppressing production of inflammatory proteins.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Rheumatology (AREA)
  • Immunology (AREA)
  • Toxicology (AREA)
  • Pain & Pain Management (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne une protéine de fusion comprenant une première région comprenant un domaine extracellulaire d'un récepteur de cytokines, et une seconde région fusionnée à la première région, et comprenant des domaines Fc d'immunoglobulines qui sont répétés en tandem, ou des domaines Fc d'immunoglobulines à chaîne simple qui sont répétés en tandem.
PCT/JP2011/065407 2010-06-30 2011-06-24 Agents thérapeutiques protéiques modifiés WO2012002562A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US35982810P 2010-06-30 2010-06-30
US61/359,828 2010-06-30
US201061425773P 2010-12-22 2010-12-22
US61/425,773 2010-12-22

Publications (1)

Publication Number Publication Date
WO2012002562A1 true WO2012002562A1 (fr) 2012-01-05

Family

ID=45402259

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2011/065407 WO2012002562A1 (fr) 2010-06-30 2011-06-24 Agents thérapeutiques protéiques modifiés

Country Status (1)

Country Link
WO (1) WO2012002562A1 (fr)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000009560A2 (fr) * 1998-08-17 2000-02-24 Abgenix, Inc. Production de molecules modifiees avec demi-vie serique prolongee
WO2002072608A2 (fr) * 2001-03-09 2002-09-19 University Of Chicago Proteines hybrides d'immunoglobulines polymeriques ciblant des recepteurs fc$g(g) a faible affinite
WO2005035586A1 (fr) * 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Composition proteique hybride
WO2007005786A2 (fr) * 2005-06-30 2007-01-11 Centocor, Inc. Procedes et compositions a effet therapeutique ameliore
WO2007100083A1 (fr) * 2006-03-03 2007-09-07 Tokyo University Of Science anticorps modifie a bioactivite accrue
WO2008083150A2 (fr) * 2006-12-28 2008-07-10 Centocor Ortho Biotech Inc. Procédés et vecteurs de génération d'immunoglobulines asialylées
WO2009086320A1 (fr) * 2007-12-26 2009-07-09 Xencor, Inc Variants de fc avec une liaison altérée à fcrn

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000009560A2 (fr) * 1998-08-17 2000-02-24 Abgenix, Inc. Production de molecules modifiees avec demi-vie serique prolongee
WO2002072608A2 (fr) * 2001-03-09 2002-09-19 University Of Chicago Proteines hybrides d'immunoglobulines polymeriques ciblant des recepteurs fc$g(g) a faible affinite
WO2005035586A1 (fr) * 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Composition proteique hybride
WO2007005786A2 (fr) * 2005-06-30 2007-01-11 Centocor, Inc. Procedes et compositions a effet therapeutique ameliore
WO2007100083A1 (fr) * 2006-03-03 2007-09-07 Tokyo University Of Science anticorps modifie a bioactivite accrue
WO2008083150A2 (fr) * 2006-12-28 2008-07-10 Centocor Ortho Biotech Inc. Procédés et vecteurs de génération d'immunoglobulines asialylées
WO2009086320A1 (fr) * 2007-12-26 2009-07-09 Xencor, Inc Variants de fc avec une liaison altérée à fcrn

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ARORA T ET AL.: "Differences in binding and effector functions between classes of TNF antagonists", CYTOKINE, vol. 45, no. 2, February 2009 (2009-02-01), pages 124 - 131, XP025913408, DOI: doi:10.1016/j.cyto.2008.11.008 *
GREENWOOD J ET AL.: "Engineering multiple-domain forms of the therapeutic antibody CAMPATH-1H: effects on complement lysis", THER. IMMUNOL., vol. 1, no. 5, 1994, pages 247 - 255, XP009059035 *
NAGASHIMA H ET AL.: "Enhancement of Antibody-dependent Cellular Cytotoxicity by Tandem Fc Multimerization", YAKUGAKU ZASSHI, vol. 130, no. 1, January 2010 (2010-01-01), pages 49 - 54, XP055046840, DOI: doi:10.1248/yakushi.130.49 *
NAGASHIMA H ET AL.: "Tandemly repeated Fc domain augments binding avidities of antibodies for Fcgamma receptors, resulting in enhanced antibody-dependent cellular cytotoxicity", MOL. IMMUNOL., vol. 45, no. 10, 2008, pages 2752 - 2763, XP022605994, DOI: doi:10.1016/j.molimm.2008.02.003 *
NAGASHIMA H ET AL.: "TNF receptor II fusion protein with tandemly repeated Fc domains", J. BIOCHEM., vol. 149, no. 3, March 2011 (2011-03-01), pages 337 - 346 *
SANDBORN WJ ET AL.: "Etanercept for active Crohn's disease: a randomized, double-blind, placebo-controlled trial", GASTROENTEROLOGY, vol. 121, no. 5, 2001, pages 1088 - 1094 *
SHOJI-HOSAKA E ET AL.: "Enhanced Fc-dependent cellular cytotoxicity of Fc fusion proteins derived from TNF receptor II and LFA-3 by fucose removal from Asn-linked oligosaccharides", J. BIOCHEM., vol. 140, no. 6, 2006, pages 777 - 783, XP008079688, DOI: doi:10.1093/jb/mvj207 *
ZANMA A ET AL.: "Conjugates of superoxide dismutase with the Fc fragment of immunoglobulin G", J. BIOCHEM., vol. 110, no. 6, 1991, pages 868 - 872 *

Similar Documents

Publication Publication Date Title
AU2021218004B2 (en) Constructs having a SIRP-alpha domain or variant thereof
US20210301028A1 (en) Composition and methods for anti-tnfr2 antibodies
JP7148539B2 (ja) 免疫抱合体
CN107250157B (zh) 包含修饰的重链恒定区的抗体
AU2019274229A1 (en) Anti-CD33 antibodies, anti-CD33/anti-CD3 bispecific antibodies and uses thereof
JP2019520797A (ja) 二重特異性結合タンパク質およびその使用
US11618776B2 (en) Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains
KR20210091710A (ko) PD-1 표적화 IL-15/IL-15Rα Fc 융합 단백질 및 병용요법에서의 이의 용도
KR20220167342A (ko) 변형된 중쇄 불변 영역을 포함하는 항체
US20220056135A1 (en) Bifunctional anti-pd-1/sirpa molecule
US20090304715A1 (en) Modified antibodies with enhanced biological activities
US20190367611A1 (en) Monomeric human igg1 fc and bispecific antibodies
US20220227867A1 (en) ICOS TARGETED HETERODIMERIC FUSION PROTEINS CONTAINING IL-15/IL-15RA Fc-FUSION PROTEINS AND ICOS ANTIGEN BINDING DOMAINS
JP2019535282A (ja) Gitrおよびctla−4に対する二重特異性ポリペプチド
US20230303648A1 (en) Bifunctional molecules comprising an il-7 variant
ES2911442T3 (es) Inhibidor monovalente de la interacción de huTNFR1
WO2012002562A1 (fr) Agents thérapeutiques protéiques modifiés
CA3189257A1 (fr) Anticorps anti-il-2 priorisant cd25
US20240182572A1 (en) Scaffold for bifunctional molecules comprising pd-1 or cd28 and sirp binding domains
RU2817602C2 (ru) Молекулы антител, которые связывают cd137 и ox40
US20220332847A1 (en) RECOMBINANT IgG Fc MULTIMERS FOR THE TREATMENT OF IMMUNE COMPLEX-MEDIATED KIDNEY DISORDERS
WO2019023097A1 (fr) Anticorps bispécifiques asymétriques et leur utilisation
CA3213917A1 (fr) Nouvel echafaudage pour molecules bifonctionnelles presentant des proprietes ameliorees
WO2024028386A1 (fr) Molécule multifonctionnelle dirigée contre cd28
CN116783213A (zh) Cd25偏向的抗il‐2抗体

Legal Events

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

Ref document number: 11801022

Country of ref document: EP

Kind code of ref document: A1

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)
NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11801022

Country of ref document: EP

Kind code of ref document: A1