WO2011008517A2 - Immunoglobulin fc polypeptides - Google Patents
Immunoglobulin fc polypeptides Download PDFInfo
- Publication number
- WO2011008517A2 WO2011008517A2 PCT/US2010/040304 US2010040304W WO2011008517A2 WO 2011008517 A2 WO2011008517 A2 WO 2011008517A2 US 2010040304 W US2010040304 W US 2010040304W WO 2011008517 A2 WO2011008517 A2 WO 2011008517A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- polypeptide
- domain
- binding
- aglycosylated
- fcr
- Prior art date
Links
Classifications
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/569—Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
- G01N33/56911—Bacteria
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P37/00—Drugs for immunological or allergic disorders
- A61P37/02—Immunomodulators
- A61P37/04—Immunostimulants
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/005—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies constructed by phage libraries
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/32—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6854—Immunoglobulins
- G01N33/6857—Antibody fragments
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/40—Immunoglobulins specific features characterized by post-translational modification
- C07K2317/41—Glycosylation, sialylation, or fucosylation
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/72—Increased effector function due to an Fc-modification
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/73—Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
- C07K2317/732—Antibody-dependent cellular cytotoxicity [ADCC]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/75—Agonist effect on antigen
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/76—Antagonist effect on antigen, e.g. neutralization or inhibition of binding
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2500/00—Screening for compounds of potential therapeutic value
- G01N2500/10—Screening for compounds of potential therapeutic value involving cells
Definitions
- the present invention relates generally to the field of protein engineering. More particularly, it concerns improved methods and compositions for the screening of combinatorial antibody Fc libraries expressed in bacteria.
- mAbs Monoclonal antibodies
- Monoclonal antibodies comprise the majority of recombinant proteins currently in the clinic, with more than 150 products in studies sponsored by companies located worldwide (Pavlou and Belsey, 2005).
- the mAb market is heavily focused on oncology and arthritis, immune and inflammatory disorders, and products within these therapeutic areas are set to continue to be the key growth drivers over the forecast period.
- genetically engineered mAbs generally have higher probability of FDA approval success than small-molecule drugs. At least 50 biotechnology companies and all the major pharmaceutical companies have active antibody discovery programs in place.
- PCR Polymerase Chain Reaction
- Anchored Periplasmic Expression is based on anchoring the antibody fragment on the periplasmic face of the inner membrane of E.coli followed by disruption of the outer membrane, incubation with fluorescently labeled target and sorting of the spheroplasts (U.S. Patent 7,094,571).
- APEx was used for the affinity maturation of antibody fragments (Harvey et al, 2004; Harvey et al, 2006). In one study over 200-fold affinity improvement was obtained after only two rounds of screening.
- Fc region of an antibody is crucial for recruitment of immunological cells and antibody dependent cytotoxicity (ADCC).
- ADCC antibody dependent cytotoxicity
- FcRs Fc receptors
- humans contain five different classes of Fc receptors.
- haplotypes, or genetic variants of different FcRs belonging to a particular class are known.
- the binding of an antibody to FcRs determines its ability to recruit other immunological cells and the type of cell recruited.
- mammalian antibodies with engineered Fc regions display increased binding to a particular FcR of interest but in addition they are still capable of binding to other FcRs with normal affinity.
- Such antibodies are more selective than the molecules naturally produced by the immune system they can nonetheless still mediate undesirable immunological responses.
- E.coli possesses a reducing cytoplasm that is unsuitable for the folding of proteins with disulfide bonds which accumulate in an unfolded or incorrectly folded state (Baneyx and Mujacic, 2004).
- the periplasm of E. coli is maintained in an oxidized state that allows the formation of protein disulfide bonds.
- periplasmic expression has been employed successfully for the expression of antibody fragments such as Fvs, scFvs, Fabs or F(ab')2s (Kipriyanov and Little, 1999). These fragments can be made relatively quickly in large quantities with the retention of antigen binding activity.
- antibody fragments lack the Fc domain, they do not bind the FcRn receptor and are cleared quickly; thus, they are only occasionally suitable as therapeutic proteins (Knight et al, 1995).
- full-length antibodies could only be expressed in E. coli as insoluble aggregates and then refolded in vitro (Boss et al, 1984; Cabilly et al, 1984).
- This approach is not amenable to the high throughput screening of antibody libraries since with the current technology it is not possible to refold millions or tens of millions of antibodies individually.
- a further problem is that since E. coli expressed antibodies are not glycosylated, they fail to bind to complement factor Iq (CIq) or Fc and many other Fc receptors.
- CIq complement factor Iq
- aglycosylated Fc domains can bind to the neonatal Fc receptor efficiently (FcRn). Consequently bacterially expressed aglycosylated antibodies do exhibit serum persistence and pharmacokinetics similar to those of fully glycosylated IgGs produced in human cells. Nonetheless, since the aglycosylated antibodies fail to elicit complement activation and can not mediate the recruitment of immune cells such as macrophages, they have previously been ineffective for many therapeutic applications.
- Fc domains [0011] Moreover, some studies have reported that binding of some Fc receptors by Fc domains can have an activating effect while others have an inhibitory one (Boruchov et al 2005; Kalergis et al, 2002).
- Different Fc ⁇ R effector functions include (antibody-dependent cell-mediated cytotoxicity (ADCC), cytokine release, phagocytosis, and maturation. Fc domains engineered to have selective effector functions could provide physiological benefits.
- ADCC antibody-dependent cell-mediated cytotoxicity
- cytokine release cytokine release
- phagocytosis phagocytosis
- maturation Fc domains engineered to have selective effector functions could provide physiological benefits.
- This disclosure provides compounds and methods involving aglycosylated antibody Fc domains that bind to Fc receptors.
- compositions involving a polypeptide that has an aglycosylated Fc domain from an antibody (“antibody Fc domain”).
- the aglycosylated Fc domain is a variant of a wild-type Fc domain such that the variation allows the Fc domain to specifically bind to one or more Fc receptors.
- a polypeptide with an aglycosylated Fc domain variant is able to bind only a subset of Fc receptors that a polypeptide with glycosylated version of the wild-type Fc domain (“glycosylated wild-type Fc domain”) can bind.
- the polypeptide with an aglycosylated Fc domain variant can specifically bind Fc ⁇ RI; in some cases, it has the affinity or binding ability that is within 2-fold of a polypeptide having a glycosylated wild-type Fc domain. In other embodiments, additionally or alternatively, the polypeptide with an aglycosylated Fc domain variant has significantly reduced affinity or binding ability (50-fold or greater reduction) compared to a polypeptide having a glycosylated wild-type Fc domain. In certain embodiments, the polypeptide with an aglycosylated Fc domain variant has a significantly reduced affinity to or ability to bind Fc ⁇ RIIb.
- a polypeptide may have an affinity or binding ability for Fc ⁇ RI that is comparable (within 2-fold), as well as significantly reduced affinity or binding ability for Fc ⁇ RIIB, both as compared to a polypeptide having a glycosylated wild- type Fc domain.
- affinity refers to the equilibrium constant for the reversible binding of two agents and is expressed as Kd.
- Affinity of a binding domain to its target can be, for example, from about 100 nanomolar (nM) to about 0.1 nM, from about 100 nM to about 1 picomolar (pM), or from about 100 nM to about 1 femtomolar (fM); alternatively, it can be between 100 nM and 1 nM or between 0.1 nM and 10 nM.
- agents specifically bind when there is an affinity between the two agents that is in the affinity ranges discussed above.
- An antibody Fc domain may be the Fc domain of an IgA, IgM, IgE, IgD or IgG antibody or a variant thereof.
- the domain is an IgG antibody Fc domain such as an IgGl, IgG2a, IgG2b, IgG3 or IgG4 antibody Fc domain.
- the antibody Fc domain may be defined as a human Fc domain, in which case it specifically binds one or more human Fc receptors.
- the Fc domain may be an IgGl Fc domain, such as the Fc domain of an anti-HER2 antibody, more specifically, the Fc domain of trastuzumab.
- an entire polypeptide is aglycosylated or that in other embodiments only a portion of the polypeptide is aglycosylated, such as the Fc domain.
- a polypeptide may contain one or more regions from an antibody in addition to the Fc domain.
- a polypeptide may contain an antigen binding domaine from an antibody.
- multiple polypeptides may form an antibody or antibody-like protein.
- embodiments include a polypeptide with a human Fc domain having an amino acid substitution at amino acids 382 and 428 and at least one additional substitution of any of the following amino acids: 224, 241, 251, 266, 269, 276, 279, 286, 295, 297, 300, 315, 325, 328, 330, 331, 332, 338, 340, 341, 348, 369, 378, 382, 392, 424, 426, 428 and/or 434.
- the amino acid at 329 of the human Fc domain is the wild-type sequence, that is, a proline.
- a polypeptide has a human Fc domain substitution at amino acid 382 that is a valine (V) instead of glutamic acid (E) (E382V).
- V valine
- E glutamic acid
- Conventional single letter abbreviations for amino acids are employed herein.
- the polypeptide has a human Fc domain substitution at amino acid 428 that is an isoleucine (M428I).
- a polypeptide has both the substitution at amino acid 382 that is a valine (E382V) and the substitution at amino acid 428 that is an isoleucine (M428I) in the human Fc domain.
- a polypeptide has a substitution in the human Fc domain at amino acid 328 that is a tryptophan (L328W).
- a polypeptide has a human Fc domain substitution at amino acid 332 that is a tyrosine (I332Y).
- Other polypeptides include those having a human Fc domain substitution at least at amino acids 328 and 332.
- the substitution at amino acid 328 may be a tryptophan (L328W) and the substitution at amino acid 332 may be a tyrosine (I332Y).
- a polypeptide has a human Fc domain substitution at amino acid 341, which is a valine (G341V) in further embodiments.
- More embodiments involve a polypeptide with a human Fc domain substitution at 382 and 428 and at least one additional substitutionin the domain in the following group: H224R/Y, F241L, K251F, V266M, E269K, N276D, V279M, N286D, Q295R, N297D, Y300C, N315D, N325S, L328W, A330V/E/I, P331A/S/E, I332Y, K338I/R, K340N/Q, G341V, V348M, V369A, A378D, K392E, S424L, S426I, or N434S/D.
- a polypeptide has an aglycosylated human Fc domain with a substitition in amino acids 382 and 428 and also has at least one additional substitution in the upper CH2 region.
- Some embodiments involve a polypeptide having at least one additional human Fc domain substitution that is of an amino acid in the following part of the upper CH2 region: 234L-239S; 264V-268H; 297N-299T; or 328L-332I.
- a polypeptide does not have the additional substitution in the human Fc domain of G341V and/or K338R.
- the Fc domain may have a substitution of G341V and at least one other substitution selected from the group consisting of: H224Y, F241L, E269K, N276D, N286D, Y300C, N325S, K338R, V348M, V369A, K392E, S424L, and N434D/S.
- the Fc domain has multiple other substitution selected from the group.
- a polypeptide may have an Fc domain substitution that includes a K338R substitution.
- polypeptides with a human Fc domain that has a set of substitutions selected from the group consisting of a) K338R and G341V; b) N297D, N315D, and K340N, c) K340N, d) K338I and K340N, e) K340Q and A378D; f) N325S and K340N, g) H224Y, E269K, N325S, and G341V, h)G341V and K392E, i) K338R, G341V, S424L and N434D, j) F241L and G341V, k) G341V, 1) N276D and G341V, m) G341V and V369A, n)N286D, G341V and N434S, o) N325S and G341V, p)Y300C and G341V;
- Embodiments involve a polypeptide having an aglycosylated Fc domain that is capable of specifically binding one or more particular human FcR polypeptides.
- the aglycosylated Fc domain has been mutated so that it can bind one or more of Fc ⁇ RIa, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa, Fc ⁇ RIIIb, or Fc ⁇ RI.
- the binding to one or more of these particular human FcR polypeptides is within 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% (or any range derivable therein) of the binding seen with a glycosylated Fc region or that the binding is altered (increased or decreased) by at least or at most 50, 60, 70, 80, 90, or 100 % (or any range derivable therein) relative to a wild-type glycosylated Fc domain.
- relative binding capabilities between polypeptides having a mutated and aglycosylated Fc domain and polypeptides having a glycosylated and wild-type Fc domain may be expressed in terms of X-fold differences (increased or decreased). For example, there may be at least or at most at least 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, or 10-fold difference, or any range derivable therein).
- a polypeptide with a mutated aglycosylated Fc domain is capable of specifically binding an Fc ⁇ RI polypeptide. In some cases, it binds at a level within 2-fold of the level of binding by a polypeptide having a glycosylated and wild-type Fc domain. In other embodiments, the level of binding is within at least 2-, 3-, A-, 5-, 6-, 7-, 8-, 9-, or 10-fold a glycosylated and wild-type Fc domain.
- the K D value for a particular Fc receptor and either a polypeptide with the aglycosylated Fc domain variant or a polypeptide with a glycosylated and wild-type Fc domain is within at least 2- or 3 -fold in embodiments described herein.
- a polypeptide has at least a 2-fold reduction in pH-dependent FcRn binding compared to polypeptide with an aglycosylated wild- type antibody Fc domain.
- Polypeptides described herein may include a linker in some embodiments.
- the linker is a conjugatable linker.
- the polypeptide contains an Fc domain from an antibody. It may contain other regions from an antibody, such as another binding domain. The additional binding domain is not an FcR binding domain in certain embodiments. In some embodiments, it may contain an antigen binding site or domain from an antibody. This would include all or part of the variable region from an antibody.
- a polypeptide contains an Fc domain from an antibody but another binding domain that is a non-FcR binding domain. In some embodiments, the non-Fc binding region is not an antigen binding site of an antibody but specifically binds a cell-surface protein.
- a cell-surface protein that the non-Fc binding region recognizes is a receptor.
- a cell-surface receptor is a tyrosine kinase.
- a polypeptide has a non-Fc binding region capable of binding multiple tyrosine kinase receptors.
- such a non-Fc binding region is capable of binding one or more of VEGF receptors, PDGF receptors, EGFR receptors, ErbB-2 receptors, EGF receptors, HGF receptors, and other Src-like tyrosine kinase receptors, or a combination thereof. It is also specifically contemplated that polypeptides have an antigen binding region that recognizes one or more of these receptor tyrosine kinases.
- polypeptides include those having an aglycosylated Fc domain capable of binding an FcR ⁇ l polypeptide and a second binding domain, wherein the second binding domain is capable of specifically binding a cell-surface molecule.
- the second binding domain is an antigen binding domain of an antibody ("antibody antigen binding domain"). In some cases, the second binding domain is not an antibody antigen binding domain.
- the second binding domain is capable of specifically binding a cell-surface molecule that is a proteinaceous molecule.
- the second binding domain may be a ligand for a cell- surface receptor or it may be a receptor for a cell-surface ligand.
- Embodiments also concern a nucleic acid that encodes any of the polypeptides discussed herein.
- the nucleic acid may be isolated and/or recombinant. It may be a nucleic acid segment that is isolated and/or recombinant.
- the nucleic acid is DNA while in others it is RNA.
- the nucleic acid is a DNA segment.
- the nucleic acid is an expression vector that is capable of expressing any of the polypeptides having an Fc binding domain with one or more substitutions that specifically binds a human FcR polypeptide.
- a nucleic acid may encode one or more polypeptides discussed above, which, depending on how the polypeptide is produced may or may not be glycosylated.
- nucleic acids encoding a polypeptide with an Fc domain capable of specifically binding a human FcR polypeptide.
- the nucleic acid may be placed in a host cell that can express the polypeptide, particularly an aglycosylated version of the polypeptide.
- the host cell may be a prokaryotic cell, such as a bacterial cell.
- the host cell may be an eukaryotic cell, such as a mammalian cell.
- a host cell contains a first expression vector, though it may comprises a second expression vector as well. Because some antibodies are made of multiple polypeptides, a host cell that expresses these polypeptides is contemplated in some embodiments.
- the population contains a plurality of host cells that express polypeptides having different Fc domains. It is contemplated that the amino acid sequence of any two different Fc domains differs in identity by less than 20%, 15%, 10%, 5% or less.
- polypeptides described herein polypeptides having an aglycosylated Fc region
- methods of using these polypeptides Any of these methods may be implemented with respect to any of the polypeptides described herein.
- an aglycosylated polypeptide comprising: a) obtaining a host cell capable of expressing an aglycosylated antibody comprising an Fc domain capable of binding an FcR polypeptide, wherein the Fc domain comprises an amino acid substitution at amino acids 382 and 428 and at least one additional substitution of any of the following amino acids: 224, 241, 251, 266, 269, 276, 279, 286, 295, 297, 300, 315, 325, 328, 330, 331, 332, 338, 340, 341, 348, 369, 378, 382, 392, 424, 426, 428 and/or 434; b) incubating the host cell in culture under conditions to promote expression of the aglycosylated antibody; and, c) purifying expressed antibody from the host cell.
- the host cell is a prokaryotic cell, such as a bacterial cell.
- the host cell is a eukaryotic cell and the polypeptide comprises a N297D substitution.
- methods involve collecting expressed antibody from the supernatant, which may be done prior to purification.
- methods involve purifying the antibody from the supernatant. This may involve subjecting the antibodies from the supernatant to filtration, HPLC, anion or cation exchange, high performance liquid chromatography (HPLC), affinity chromatography or a combination thereof. In some embodiments, methods involve affinity chromatography using staphylococcal Protein A, which binds the IgG Fc region. Other purification methods are well known to those of ordinary skill in the art.
- polypeptides having an aglycosylated Fc domain capable of binding an FcR polypeptide may be implemented in such methods.
- an antibody that is aglycosylated and that has an Fc domain capable of binding an Fc ⁇ RI polypeptide is prescribed or administered to a subject.
- methods may involve treating asubject with such an antibody. Any of the polypeptides described herein may be used.
- Certain embodiments involve a polypeptide having an aglycosylated human Fc domain that comprises an amino acid substitution at amino acids 382 and 428 and at least one additional substitution of any of the following amino acids: 224, 241, 251, 266, 269, 276, 279, 286, 295, 297, 300, 315, 325, 328, 330, 331, 332, 338, 340, 341, 348, 369, 378, 382, 392, 424, 426, 428 and/or 434.
- the aglycosylated polypeptide or antibody is capable of specifically binding an activating FcR polypeptide, which refers to an FcR polypeptide that activates one or more immune cells.
- Activating polypeptides include Fc ⁇ RI, Ha, Ilia, lib, and IHc.
- Fc ⁇ RIIb is an inhibitory FcR polypeptide.
- the aglycosylated polypeptide or antibody no longer binds an inhibitory FcR polypeptide at a level comparable to a glycosylated, wild-type Fc domain.
- an aglycosylated polypeptide or antibody specifically binds an Fc ⁇ RI polypeptide.
- the aglycosylated polypeptide or antibody has a reduced capability to bind an Fc ⁇ RIIb polypeptide, wherein its affinity is at least 50-fold less than a glycosylated, wild-type version of the polypeptide or antibody.
- the aglycosylated antibody is an aglycosylated version of a therapeutic antibody, which refers to an antibody used in therapy or treatment for a disease or condition. Any antibody or polypeptide discussed herein, including those discussed above, may be used in implementing methods for inducing an immune response. An example of a therapeutic antibody is trastuzumab.
- there are methods of inducing dendritic cell- (DC) mediated cell killing against a target cell expressing a targeted cell surface polypeptide comprising: a) contacting the target cell with a polypeptide comprising a i) mutated and aglycosylated Fc domain capable of specifically binding at least a dendritic-cell activating FcR and ii) a second binding domain that binds the targeted cell surface polypeptide; and b) exposing the target cell to dendritic cells under conditions that promote killing of the target cell.
- the activating FcR is an Fc ⁇ RI polypeptide.
- polypeptide with an aglycosylated Fc domain specifically binds to an Fc ⁇ RIIB polypeptide at a level that is reduced compared to a polypeptide having a glycosylated wild-type Fc domain.
- polypeptides have an Fc domain that comprises at least one amino acid substitution in the following amino acids: 224, 241, 251, 266, 269, 276, 279, 286, 295, 297, 300, 315, 325, 328, 330, 331, 332, 340, 348, 369, 378, 382, 392, 424, 426, 428 and/or 434. Additional polypeptides are discussed above and throughout this application.
- the target cell is a cancer cell. Consequently, methods of treating cancer using aglycosylated and mutated Fc domains in place of a glycosylated and wild-type Fc domain in an antibody therapy are contemplated. Treatment of other diseases or conditions involving antibodies that use glycosylated and wild-type Fc domains can be similarly implemented with aglycosylated and Fc variant polypeptides described herein.
- Other embodiments concern methods for screening for an aglycosylated polypeptide having an Fc domain that binds a one or more specific FcR polypeptides comprising: a) obtaining a population of Gram negative bacterial cells, cells of which population express a aglycosylated polypeptide comprising an Fc domain in their periplasm, wherein the population expresses a plurality of different Fc domains; b) contacting the bacterial cells with a first FcR polypeptide under conditions to allow contact between the FcR polypeptide and the aglycosylated Fc domains, wherein the FcR polypeptide is Fc ⁇ RIa, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa, Fc ⁇ RIIIb, or Fc ⁇ RI; and, c) selecting at least one bacterial cell based on binding of the aglycosylated Fc domain to the first FcR polypeptide.
- Methods may further involve identifying or isolating the aglycosylated polypeptide from the selected bacterial cell. Also, methods may involve determining whether an aglycosylated polypeptide in selected bacterial cells can bind to other FcR polypeptides. In some embodiments, determining whether an aglycosylated polypeptide in selected bacterial cells can bind to other FcR polypeptides comprises repeating steps a)-c) with a second FcR polypeptide to determine whether the aglycosylated polypeptide also binds the second FcR polypeptide. It is contemplated that steps a)-c) may be repeated with more than two different FcR polypeptides. In some embodiments, the aglycosylated polypeptide binds multiple FcR polypeptides.
- methods involve bacterial cells that are E. coli cells.
- the Fc domain is an IgG, IgA or IgE Fc domain.
- the population of Gram negative bacterial cells comprise a plurality of nucleic acids encoding the plurality of aglycosylated Fc domains.
- the plurality of nucleic acids further encodes a membrane secretion signal fused to the plurality of aglycosylated Fc domains.
- a membrane secretion signal may be PeIB or DsbA.
- the aglycosylated Fc domain may include a hinge, CH2 and CH3 region.
- the aglycosylated polypeptide comprises an eukaryotic FcR domain.
- the Fc domain binds human Fc ⁇ RIa, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa, Fc ⁇ RIIIb, Fc ⁇ RI or CIq.
- it has reduced binding affinity for Fc ⁇ RIIb relative to a glycosylated and wild-type version of the Fc domain. Specific methods are disclosed in WO 2008/137475, which is hereby incorporated by reference.
- Other embodiments involve methods for optimizing Fc binding to one or more specific FcR polypeptides of an aglycosylated polypeptide having an Fc domain comprising: a) obtaining a population of Gram negative bacterial cells, cells of which population express a aglycosylated polypeptide comprising an Fc domain in their periplasm, wherein the population expresses a plurality of different polypeptides expressing different mutated Fc domains; b) contacting the bacterial cells with a first FcR polypeptide under conditions to allow contact between the FcR polypeptide and the aglycosylated Fc domains, wherein the FcR polypeptide is Fc ⁇ RIa, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa, Fc ⁇ RIIIb, or Fc ⁇ RI; and c) selecting at least one bacterial cell based on binding of the aglycosylated Fc domain to the first FcR
- Embodiments discussed in the context of a methods and/or composition of the invention may be employed with respect to any other method or composition described herein. Thus, an embodiment pertaining to one method or composition may be applied to other methods and compositions of the invention as well.
- FIG. 1 Mutation points of isolated aglycosylated Fc5 (382E and 428M) represented on the 3D structure of glycosylated IgGl Fc (PBD Code: IFCl).
- FIG. 2 Two beta sheets including 382E in ⁇ -sheet C and 428M in ⁇ - sheet C of CH3 domain represented on the crystal structure of glycosylated IgG. (PBD Code: IFCl).
- FIG. 3 Error prone PCR library for engineering aglycosylated Fc5 domains.
- FIG. 4 Fluorescence histogram of spheroplasts from different rounds of sorting labeled with Fc ⁇ RIa-FITC.
- FIG. 5 DNA sequences of isolated Fc mutant clones exhibiting higher affinity to Fc ⁇ RIa than Fc5. Mean fluorescence values for the respective clones labeled with Fc ⁇ RIa are shown in parenthesis.
- FIG. 6 Mutation points of isolated aglycosylated Fc601-619 represented on the 3D structure of glycosylated IgGl Fc (PBD Code: IFCl).
- FIG. 7 Fluorescence histogram of spheroplasts for wild type Fc, Fc5, and Fc601 labeled with 30 nM of Fc ⁇ RIa-FITC. M: Mean fluorescence intensity.
- FIG. 8 Mutation points of isolated aglycosylated Fc 601 (K338R, G341V, E382V, M428I) represented on the 3D structure of glycosylated IgGl Fc (PBD Code: IFCl).
- FIG. 9 Map of plasmid pSTJ4-Herceptin IgGl.
- FIG. 10 Kinetic rates and equilibrium dissociation constants of aglycosylated trastuzumab, trastuzumab-Fc5, trastuzumab-Fc601, and glycosylated trastuzumab determined by BIACore analysis for binding to Fc ⁇ RIa.
- FIG. 11 ELISA assays for binding of trastuzumab antibodies to Fc ⁇ RIIa-GST.
- FIG. 12 ELISA assays for binding of trastuzumab antibodies to Fc ⁇ RIIb-GST.
- FIG. 13 ELISA assays for binding of trastuzumab antibodies to Fc ⁇ RIIIa.
- FIG. 14 ELISA assays for pH dependent binding to FcRn at pH 7.4 and 6.0. Plates were coated with aglycosylated trastuzumab, trastuzumab-Fc5, trastuzumab-Fc601 or commercial glycosylated trastuzumab and the binding of FcRn was detected using anti-GST-HRP.
- FIG. 15 Library for higher affinity to Fc ⁇ RIa than Fc5 and for pH dependent FcRn binding.
- FIG. 16 Gene assembly PCR for the construction of 4 sub-libraries that randomized upper CH2 region.
- FIG. 17 DNA sequences of isolated Fc mutant clones exhibiting higher affinity to Fc ⁇ RIa than Fc5. Mean fluorescence values for the respective clones labeled with Fc ⁇ RIa are shown in parenthesis.
- FIG. 18 Summary of mutations in Fc701 - 709.
- FIG. 19 Fluorescence histogram of spheroplasted cells for wild type Fc, Fc5, Fc701, and Fc702 labeled with 1 nM of Fc ⁇ RIa-FITC. M: Mean fluorescence intensity.
- FIG. 20 Fluorescence histogram of spheroplasted cells for wild type Fc, Fc5, Fc601, and Fc701 labeled with 1 nM of Fc ⁇ RIa-FITC.
- M Mean fluorescence intensity.
- FIG. 21 Mutation points of isolated aglycosylated Fc5 (382E and 428M) represented on the 3D structure of glycosylated IgGl Fc (PBD Code: IFCl)
- FIG. 22 Kinetic rates and equilibrium dissociation constants of aglycosylated trastuzumab, trastuzumab-Fc5, trastuzumab-Fc601, trastuzumab-Fc701 and glycosylated trastuzumab determined by BIACore analysis for binding to Fc ⁇ RI.
- FIG. 23 ELISA assays for pH dependent binding to FcRn at pH 7.4 and 6.0. Plates were coated with aglycosylated trastuzumab, trastuzumab-Fc5, trastuzumab-Fc601, trasutuzumab-Fc701 or commercial glycosylated trastuzumab and the binding of FcRn was detected using anti-GST-HRP.
- FIG. 24 Covalently anchored full length IgG display system.
- FIG. 25 Comparison of FACS signals between 2 plasmid covalently anchored full length IgG display system and dicistronic system. Trastuzumab full length IgGs were expressed using either the 2 plasmid anchored full length IgG display system or dicistronic full length IgG display system.
- M Mean fluorescence intensity. Spheroplasts were incubated with 30 nM Fc ⁇ RI-FITC probe for detection.
- FIG. 26 Comparison of FACS signals between the 2 plasmids covalently anchored full length IgG display system and dicistronic system. Trastuzumab full length IgGs were expressed using either the 2 plasmids anchored full length IgG display system or dicistronic full length IgG display system.
- M Mean fluorescence intensity. Spheroplasts were incubated with 30 nM Fc ⁇ RIIa-GST and labeled with polyclonal anti-GST-FITC (1 :200) probe for detection.
- FIG. 27 FACS analysis of trastuzumab full length IgG using 2 plasimids covalently anchored full length IgG display system and dicistronic system. Spheroplasts expressing trastuzumab full length IgGs were incubated with 30 nM Fc ⁇ RI-FITC probe for detection. M: Mean fluorescence intensity.
- FIG. 28 FACS analysis of trastuzumab full length IgG using 2 plasimids covalently anchored full length IgG display system and dicistronic system. Spheroplasts expressing trastuzumab full length IgGs were incubated with incubated with 30 nM Fc ⁇ RIIa-GST and labeled with polyclonal anti-GST-FITC (1 :200) probe for detection. M: Mean fluorescence intensity.
- FIG. 29 Library for randomization of upper CH2 region.
- FIG. 30 ADCC assays with PBMC as effector cells and SkBr3 as the target cell. *, P ⁇ 0.05.
- FIG. 31 ADCC assays with mDCs as effector cells and SkBr3 as the target cell. *, P ⁇ 0.05 ; **, P ⁇ 0.01.
- polypeptide comprising an antibody Fc domain may be expressed in the periplasmic space of a gram negative bacteria.
- an antibody Fc domain may be anchored to the periplasmic face of the inner membrane.
- an Fc domain may be directly fused to a membrane spanning or membrane bound polypeptide or may interact (e.g., via protein-protein interactions) with a membrane spanning or membrane bound polypeptide.
- Such a technique may be termed "Anchored Periplasmic Expression" or "APEx”.
- the periplasmic compartment is contained between the inner and outer membranes of Gram negative cells (see, e.g., Oliver, 1996). As a sub-cellular compartment, it is subject to variations in size, shape and content that accompany the growth and division of the cell.
- Within a framework of peptidoglycan heteroploymer is a dense mileau of periplasmic proteins and little water, lending a gel-like consistency to the compartment (Hobot et ah, 1984; van Wielink and Duine, 1990).
- the peptidoglycan is polymerized to different extents depending on the proximity to the outer membrane, close-up it forms the murein sacculus that affords cell shape and resistance to osmotic lysis.
- the outer membrane (see Nikaido, 1996) is composed of phospholipids, porin proteins and, extending into the medium, lipopolysaccharide (LPS).
- LPS lipopolysaccharide
- the molecular basis of outer membrane integrity resides with LPS ability to bind divalent cations (Mg 2+ and Ca 2+ ) and link each other electrostatically to form a highly ordered quasi-crystalline ordered "tiled roof on the surface (Labischinski et ah, 1985).
- the membrane forms a very strict permeability barrier allowing passage of molecules no greater than around 650 Da (Burman et ah, 1972; Decad and Nikaido, 1976) via the porins.
- the large water filled porin channels are primarily responsible for allowing free passage of mono and disaccharides, ions and amino acids in to the periplasm compartment (Nikaido and Nakae, 1979; Nikaido and Vaara, 1985). With such strict physiological regulation of access by molecules to the periplasm it may appear, at first glance, inconceivable that large ligands (i.e., larger than the 650 Da exclusion limit) could be employed in screening methods. However, the inventors have shown that ligands greater than 2000 Da in size can diffuse into the periplasm without disruption of the periplasmic membrane. Such diffusion can be aided by one or more treatments of a bacterial cell, thereby rendering the outer membrane more permeable, as is described herein below.
- a gram negative bacterial cell of the invention may be defined as an E. coli cell.
- a Gram negative bacterial cell may be defined as a genetically engineered bacterial cell such as a Jude-1 strain of E. coli.
- methods involve disrupting, permeablizing or removing the outer membrane of bacteria are well known in the art, for example, see U.S. Patent 7,094,571.
- the outer membrane of the bacterial cell may be treated with hyperosmotic conditions, physical stress, lysozyme, EDTA, a digestive enzyme, a chemical that disrupts the outer membrane, or by infecting the bacterium with a phage or a combination of the foregoing methods.
- the outer membrane may be disrupted by lysozyme and EDTA treatment.
- the bacterial outer membrane may be removed entirely.
- methods are employed for increasing the permeability of the outer membrane to one or more labeled ligands. This can allow screening access of labeled ligands otherwise unable to cross the outer membrane.
- certain classes of molecules for example, hydrophobic antibiotics larger than the 650 Da exclusion limit, can diffuse through the bacterial outer membrane itself, independent of membrane porins (Farmer et ah, 1999). The process may actually permeabilize the membrane on so doing (Jouenne and Junter, 1990).
- Such a mechanism has been adopted to selectively label the periplasmic loops of a cytoplasmic membrane protein in vivo with a polymyxin B nonapeptide (Wada et at., 1999).
- certain long chain phosphate polymers 100 Pi
- appear to bypass the normal molecular sieving activity of the outer membrane altogether Rao and Torriani, 1988).
- cells expressing binding proteins anchored to the outer (periplasmic) face of the cytoplasmic membrane can be fluorescently labeled simply by incubating with a solution of fluorescently labeled ligand in cells that either have a partially permeabilized membrane or a nearly completely removed outer membrane.
- the permeability of the outer membrane of different strains of bacterial hosts can vary widely. It has been shown previously that increased permeability due to OmpF overexpression was caused by the absence of a histone like protein resulting in a decrease in the amount of a negative regulatory mRNA for OmpF translation (Painbeni et al., 1997). Also, DNA replication and chromosomal segregation is known to rely on intimate contact of the replisome with the inner membrane, which itself contacts the outer membrane at numerous points.
- a preferred host for library screening applications is E. coli ABLEC strain, which additionally has mutations that reduce plasmid copy number.
- Treatments such as hyperosmotic shock can improve labeling significantly. It is known that many agents including, calcium ions (Bukau et al., 1985) and even Tris buffer (Irvin et al., 1981) alter the permeability of the outer- membrane. Further, phage infection stimulates the labeling process. Both the filamentous phage inner membrane protein pill and the large multimeric outer membrane protein pIV can alter membrane permeability (Boeke et al., 1982) with mutants in pIV known to improve access to maltodextrins normally excluded (Marciano et al., 1999).
- a high degree of permeability may be achieved (Daugherty et al., 1999).
- Cells comprising anchored or periplasm-associated polypeptides bound to fluorescently labeled ligands can then be easily isolated from cells that express binding proteins without affinity for the labeled ligand using flow cytometry or other related techniques.
- flow cytometry or other related techniques it will be desired to use less disruptive techniques in order to maintain the viability of cells.
- EDTA and Lysozyme treatments may also be useful in this regard.
- an Fc domain is engineered to bind one or more specific Fc receptors. Additionally or alternatively, an Fc domain may be engineered so that it does not specifically bind one or more specific Fc receptors.
- compositions comprising a proteinaceous molecule that has been modified relative to a native or wild-type protein.
- proteinaceous compound has been deleted of amino acid residues; in other embodiments, amino acid residues of the proteinaceous compound have been replaced, while in still further embodiments both deletions and replacements of amino acid residues in the proteinaceous compound have been made.
- a proteinaceous compound may include an amino acid molecule comprising more than one polypeptide entity.
- a "proteinaceous molecule,” “proteinaceous composition,” “proteinaceous compound,” “proteinaceous chain” or “proteinaceous material” generally refers, but is not limited to, a protein of greater than about 200 amino acids or the full length endogenous sequence translated from a gene; a polypeptide of 100 amino acids or greater; and/or a peptide of 3 to 100 amino acids. All the “proteinaceous” terms described above may be used interchangeably herein; however, it is specifically contemplated that embodiments may be limited to a particular type of proteinaceous compound, such as a polypeptide. Furthermore, these terms may be applied to fusion proteins or protein conjugates as well.
- a protein may include more than one polypeptide.
- an IgG antibody for example, has two heavy chain polypeptides and two light chain polypeptides, which are joined to each other through disulfide bonds.
- a "distinct Fc domain” may be defined as a domain that differs from another Fc by as little as one amino acid.
- Methods for making a library of distinct antibody Fc domains or nucleic acids that encode antibodies are well known in the art and exemplified herein.
- Fc domains may be amplified by error prone PCR as exemplified herein.
- a plurality of antibody Fc domains may comprise a stretch (1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) amino acids that have been randomized.
- residues that are normally glycosylated in an antibody Fc domain may be mutated.
- residues that are normally glycosylated (or adjacent residues) may be used as a site for an insertion of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids.
- An amino acid insertion may be made at, or adjacent to, a residue corresponding to amino acid 384 of the IgGl Fc (SEQ ID NO:2).
- a population of gram negative bacteria according to the invention may be defined as comprising at least about IxIO 3 , IxIO 4 , IxIO 5 , IxIO 6 , IxIO 7 , IxIO 8 , or more distinct antibodies Fc domains.
- a population of Gram negative bacterial cells may be produced by a method comprising the steps of: (a) preparing a plurality of nucleic acid sequences encoding a plurality of distinct antibody Fc domains; and (b) transforming a population of Gram negative bacteria with said nucleic acids wherein the Gram negative bacteria comprise a plurality of antibody Fc domains expressed in the periplasm.
- an FcR may have specificity for a particular type or subtype of Ig, such as IgA, IgM, IgE or IgG (e.g., IgGl, IgG2a, IgG2b, IgG3 or IgG4).
- the antibody-binding domain may be defined as an IgG binding domain.
- the FcR polypeptide may compries an eukaryotic, prokaryotic, or synthetic FcR domain.
- an antibody Fc-binding domain may be defined as a mammalian, bacterial or synthetic binding domain.
- Some Fc- binding domains for use in the invention include but are not limited to a binding domain from one of the polypeptides of Table 1.
- an Fc-binding polypeptide may be encoded by an FCGR2A, FCGR2B, FCGR2C, FCGR3A, FCGR3B, FCGRlA, Fcgrl, FCGR2, FCGR2, Fcgr2, Fcgr2, FCGR3, FCGR3, Fcgr3, FCGR3, Fcgr3, FCGRT, mrp4, spa or spg gene.
- an FcR polypeptide for use according to the invention may be an Fc binding region from human Fc ⁇ RIa, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa, Fc ⁇ RIIIb, Fc ⁇ RI or CIq.
- an Fc polypeptide may be anchored to the inner membrane of a Gram negative bacteria.
- Methods and compositions for the anchoring of polypeptides to the inner membrane of Gram negative bacterial have previously been described (U.S. Patent 7,094,571 and U.S. Patent Publ. 20050260736).
- an Fc domain may be fused to a polypeptide that is associated with or integrated in a bacterial inner membrane.
- Such a fusion protein may comprise an N terminal or C terminal fusion with an Fc domain and in some case may comprise additional linker amino acids between the membrane anchoring polypeptide and the Fc domain.
- a membrane anchoring polypeptide may be the first six amino acids encoded by the E.
- a membrane anchoring polypeptide may be an inner membrane lipoprotein or fragment thereof such as from AraH, MgIC, MaIF, MaIG, MaIC, MaID, RbsC, RbsC, ArtM, ArtQ, GInP, ProW, HisM, HisQ, LivH, LivM, LivA, LivE, DppB, DppC, OppB, AmiC, AmiD, BtuC, ThuD, FecC, FecD, FecR, FepD, MkB, MkC, CysT, CysW, UgpA, UgpE, PstA, PstC, PotB, PotC, PotH, Pod, ModB, NosY, PhnM,
- an FcR may be immobilized on a column or bead ⁇ e.g., a magnetic bead) and the bacterial cell binding to the FcR separated by repeated washing of the bead ⁇ e.g., magnetic separation) or column.
- a target ligand may be labeled such as with a fluorophor, a radioisotope or an enzyme.
- bacterial cells may, in some cases, be selected by detecting a label on a bound FcR.
- a fluorophore may be used to select cells using fluorescence activated cell sorting (FACS).
- FACS fluorescence activated cell sorting
- bacterial cells may be selected based on binding or lack of binding two or more FcR polypeptides.
- bacteria may be selected that display antibodies that bind to two FcR polypeptides, wherein each FcR is used to select the bacterial sequentially.
- bacteria may be selected that display antibody Fc domains that bind to one FcR (such as an FcR comprising a first label) but not to a second FcR (e.g., comprising a second label).
- the foregoing method maybe used, for example, to identify antibody Fc domains that bind to a specific FcR but not a second specific FcR.
- the size of the at least one proteinaceous molecule may comprise, but is not limited to, about or at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 275, 300, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, 950, 1000 or greater amino molecule residues, and any range derivable therein.
- Compounds may include the above-mentioned number of contiguous amino acids from SEQ ID NO:2 (human IgG Fc polypeptide) or from SEQ ID NOs 4-31 and these may be further qualified as having a percent identity or homology to SEQ ID NO:2 or any of SEQ ID NO:4-31 (discussed below). It is contemplated that embodiments with respect to SEQ ID NO:2 may be employed with respect to any other amino acid sequences described herein, and vice versa, if appropriate.
- an "amino molecule” refers to any amino acid, amino acid derivative or amino acid mimic as would be known to one of ordinary skill in the art.
- the residues of the proteinaceous molecule are sequential, without any non-amino molecule interrupting the sequence of amino molecule residues.
- the sequence may comprise one or more non-amino molecule moieties.
- the sequence of residues of the proteinaceous molecule may be interrupted by one or more non-amino molecule moieties.
- Embodiments concerns modified proteins and polypeptides, particularly a modified protein or polypeptide that exhibits at least one functional activity that is comparable to the unmodified version, yet the modified protein or polypeptide possesses an additional advantage over the unmodified version, such as provoking ADCC, easier or cheaper to produce, eliciting fewer side effects, and/or having better or longer efficacy or bioavailability.
- modified protein or a “modified polypeptide”
- a protein or polypeptide that 1) performs at least one of the same activities or has at least one of the same specificities as the unmodified protein or polypeptide, but that may have a different level of another activity or specificity; and 2) possesses an additional advantage over the unmodified protein or polypeptide. Determination of activity may be achieved using assays familiar to those of skill in the art, particularly with respect to the protein's activity, and may include for comparison purposes, for example, the use of native and/or recombinant versions of either the modified or unmodified protein or polypeptide.
- embodiments concerning a "modified protein” may be implemented with respect to a “modified polypeptide,” and vice versa.
- embodiments may involve domains, polypeptides, and proteins described in WO 2008/137475, which is hereby specifically incorporated by reference.
- Modified proteins may possess deletions and/or substitutions of amino acids; thus, a protein with a deletion, a protein with a substitution, and a protein with a deletion and a substitution are modified proteins. In some embodiments these modified proteins may further include insertions or added amino acids, such as with fusion proteins or proteins with linkers, for example.
- a "modified deleted protein” lacks one or more residues of the native protein, but possesses the specificity and/or activity of the native protein.
- a "modified deleted protein” may also have reduced immunogenicity or antigenicity.
- An example of a modified deleted protein is one that has an amino acid residue deleted from at least one antigenic region-that is, a region of the protein determined to be antigenic in a particular organism, such as the type of organism that may be administered the modified protein.
- Substitutional or replacement variants typically contain the exchange of one amino acid for another at one or more sites within the protein and may be designed to modulate one or more properties of the polypeptide, particularly its effector functions and/or bioavailability. Substitutions may or may not be conservative, that is, one amino acid is replaced with one of similar shape and charge.
- Conservative substitutions are well known in the art and include, for example, the changes of: alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalanine; and valine to isoleucine or leucine.
- a modified protein may possess an insertion of residues, which typically involves the addition of at least one residue in the polypeptide. This may include the insertion of a targeting peptide or polypeptide or simply a single residue. Terminal additions, called fusion proteins, are discussed below.
- biologically functional equivalent is well understood in the art and is further defined in detail herein. Accordingly, sequences that have between about 70% and about 80%, or between about 81% and about 90%, or even between about 91% and about 99% of amino acids that are identical or functionally equivalent to the amino acids of a native polypeptide are included, provided the biological activity of the protein is maintained.
- a modified protein may be biologically functionally equivalent to its native counterpart.
- amino acid and nucleic acid sequences may include additional residues, such as additional N- or C -terminal amino acids or 5' or 3' sequences, and yet still be essentially as set forth in one of the sequences disclosed herein, so long as the sequence meets the criteria set forth above, including the maintenance of biological protein activity where protein expression is concerned.
- the addition of terminal sequences particularly applies to nucleic acid sequences that may, for example, include various non-coding sequences flanking either of the 5' or 3' portions of the coding region or may include various internal sequences, i.e., introns, which are known to occur within genes.
- amino acids of a protein may be substituted for other amino acids in a protein structure with or without appreciable loss of interactive binding capacity with structures such as, for example, binding sites to substrate molecules. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid substitutions can be made in a protein sequence, and in its underlying DNA coding sequence, and nevertheless produce a protein with like properties. It is thus contemplated by the inventors that various changes may be made in the DNA sequences of genes without appreciable loss of their biological utility or activity, as discussed below.
- a proteinaceous molecule has "homology” or is considered “homologous” to a second proteinaceous molecule if one of the following "homology criteria" is met: 1) at least 30% of the proteinaceous molecule has sequence identity at the same positions with the second proteinaceous molecule; 2) there is some sequence identity at the same positions with the second proteinaceous molecule and at the nonidentical residues, at least 30% of them are conservative differences, as described herein, with respect to the second proteinaceous molecule; or 3) at least 30% of the proteinaceous molecule has sequence identity with the second proteinaceous molecule, but with possible gaps of nonidentical residues between identical residues.
- homologous may equally apply to a region of a proteinaceous molecule, instead of the entire molecule. If the term “homology” or “homologous” is qualified by a number, for example, “50% homology” or “50% homologous,” then the homology criteria, with respect to 1), 2), and 3), is adjusted from “at least 30%” to "at least 50%.” Thus it is contemplated that there may homology of at least 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or more between two proteinaceous molecules or portions of proteinaceous molecules.
- a modified polypeptide may be characterized as having a certain percentage of identity to an unmodified polypeptide or to any polypeptide sequence disclosed herein, including SEQ ID NO:2 or any of SEQ ID NOs:4-31.
- the percentage identity may be at most or at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% (or any range derivable therein) between two proteinaceous molecules or portions of proteinaceous molecules. It is contempated that percentage of identity discussed above may relate to a particular region of a polypeptide compared to an umodified region of a polypeptide.
- a polypeptide may contain a modified or mutant Fc domain that can be characterized based on the identity of the amino acid sequence of the modified or mutant Fc domain to an unmodified or mutant Fc domain from the same species.
- a modified or mutant human Fc domain characterized, for example, as having 90% identity to an unmodified Fc domain means that 90% of the amino acids in that domain are identical to the amino acids in the unmodified human Fc domain (SEQ ID NO:2).
- the hydropathic index of amino acids may be considered.
- the importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte & Doolittle, 1982). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
- Patent 4,554,101 the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ⁇ 1); glutamate (+3.0 ⁇ 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5 ⁇ 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5); tryptophan (-3.4).
- amino acid substitutions generally are based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
- substitutions that take into consideration the various foregoing characteristics are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
- Fc receptors to which Fc domains bind are well known in the art and some examples of receptors are listed below in Table 1.
- a polypeptide may comprise an aglycosylated antibody Fc domain capable of binding an FcR polypeptide.
- the aglycosylated Fc domain may be further defined as having a specific affinity for an FcR polypeptide under physiological conditions. For instance an Fc domain may have an equilibrium dissociation constant between about 10 "6 M to about 10 "9 M under physiological conditions.
- an aglycosylated Fc domain may be defined as comprising one or more amino acid substitution or insertion relative to a wild-type sequence, such as a human wild-type sequence.
- Means of preparing such a polypeptide include those discussed in WO 2008/137475, which is hereby incorporated by reference.
- the inventors have identified as particularly preferred substitutions for achieving such improved FcR binding as those at positions 331, 382 and/or 428 of the Fc domain (for example, see Nagaoka and Akaike 2003; such as P331, E382 and/or M428 of the human IgG Fc domain sequence as shown U.S. Patent Publ.
- a polypeptide may have a substitution that includes one or more of 426, 229, 322, 350, 361, 372, 442, 402, 224, 430, 238, 436, 310, 313, 384, 372, 380 or 331 of the Fc domain, such as S426, C229, K322, T350, N361, F372, S442, G402, H224, E430, P238, Y436, H310, W313, N384, F372, E380 or P331 of the human IgG Fc domain, with the specific preferred examples being a) E382 and M428; b) N361, E382 and M428; c) N361, F372, E382 and M428; d) H310, K32
- substitutions include a) E382V and M428I; b) E382V; c) N361D, E382V and M428I; d) N361D, F372L, E382V and M428I; e) H310Y, K322R, T350A, E382V, S426T and S442P; f) C229R, E382V and M428I; g) W313R and M428I; h) E382T, N384D and M428I; i) E380R, E382M and N384E; j) N361S, E382V and M428I; k) E382V, M428I and Y436A; 1) P238S, E382V, S426V, M428L and E430H; m) E380D, E382V, N384R, S426V, M428L and E430D
- Insertions of 5 to 15 amino acids are contemplated. In some embodiments, 10 amino acids are inserted, such as between amino acids N297 and S298 of an Fc domain, such as a human IgG Fc domain.
- Particular insertions at this position include a) RTETPVYMVM (SEQ ID NO:79); b) WQVFNKYTKP (SEQ ID NO:80); c) LGDGSPCKAN (SEQ ID NO:81); d) EVPLVWMWVS (SEQ ID NO: 82) together with F241L and K326E; and e) EQ WGSQFGCG (SEQ ID NO: 83) together with V282A.
- the Fc domain may be a human IgG Fc that comprises an amino acid substitution at an amino acid residue corresponding to E382 of the IgG Fc domain.
- an aglycosylated Fc domain may comprise an amino acid sequence insertion (e.g., about 1 to 5 amino acids) adjacent to an amino acid residue corresponding to E382 of the IgG Fc domain.
- an Fc domain may comprise a hydrophobic amino acid substitution at E382 such as an E to V substitution.
- an Fc domain of the invention may comprise an amino acid substitution at a residue corresponding to M428 (e.g., M428 to I), S426, C229, H310, K322, T350, N361, F372 or S442 of the human IgG Fc.
- an aglycosylated Fc domain may comprise an amino acid substitution corresponding to those found in the Fel l as described in WO 2008/137475, which is hereby incorporated by reference.
- an aglycosylated Fc domain may comprise the amino acid sequence of SEQ ID NO:2 (Fc5).
- an aglycosylated Fc domain comprises a specific binding affinity for an FcR such as human Fc ⁇ RIa, Fc ⁇ RIIa, Fc ⁇ RIIb, Fc ⁇ RIIc, Fc ⁇ RIIIa, Fc ⁇ RIIIb, Fc ⁇ RI or CIq.
- an aglycosylated Fc domain of the invention is defined as an Fc domain with a specific affinity for Fc ⁇ RIa.
- such an Fc domain may be defined as having an equilibrium dissociation constant, with respect to Fc ⁇ RIa binding, of about 10 "6 M to about 10 "9 M under physiological conditions.
- Embodiments concern a proteinaceous compound that may include amino acid sequences from more than one naturally occuring or native polypeptides or proteins. Embodiments discussed above are contemplated to apply to this section, and vice versa.
- a modified antibody is one that contains a modified Fc domain with an antigen binding domain.
- the antibody may have two different antigen binding regions, such as a different region on each of the two heavy chains.
- there are polypeptides comprising multiple heterologous peptides and/or polypeptides (“heterologous" meaning they are not derived from the same polypeptide).
- a proteinaceous compound or molecule could include a modified Fc domain with a protein binding region that is not from an antibody.
- polypeptides comprising a modified Fc domain with a protein binding region that binds a cell-surface receptor.
- These proteinaceous molecule comprising multiple functional domains may be two or more domains chemically conjugated to one another or it may be a fusion protein of two or more polypeptides encoded by the same nucleic acid molecule. It is contemplated that proteins or polypeptides may include all or part of two or more heterologous polypeptides.
- a multipolypeptide proteinaceous compound may be comprised of all or part of a first polypeptide and all or part of a second polypeptide, a third polypeptide, a fourth polypeptide, a fifth polypeptide, a sixth polypeptide, a seventh polypeptide, an eight polypeptide, a ninth polypeptide, a tenth polypeptide, or more polypeptides.
- Polypeptides or proteins (including antibodies) having an antigen binding domain or region of an antibody and an aglycosylated Fc domain can be used against any antigen or epitope, including but not limited to proteins, subunits, domains, motifs, and/or epitopes belonging to the following list of targets: 17-IA, A- IBB, 4Dc, 6-keto-PGFla, 8-iso-PGF2a, 8-oxo-dG, Al Adenosine Receptor, A33, ACE, ACE-2, Activin, Activin A, Activin AB, Activin B, Activin C, Activin RIA, Activin RIA ALK-2, Activin RIB ALK-4, Activin RIIA, Activin RIIB, ADAM, ADAMlO, ADAM12, ADAM15, ADAM17/TACE, ADAM8, ADAM9, ADAMTS, ADAMTS4, ADAMTS5, Addressin
- Fc domains can bind to an FcR, however, it is contemplated that ADCC can be directed not only through an antigen binding domain on the polypeptide containing the Fc domain, but through some other protein binding domain. Consequently, embodiments concern an Fc domain and a heterologous non- antigen binding domain. In certain embodiments, the non-antigen binding domain bind to the cell surface. Therefore, these agents require either chemical conjugation to or fusion with agents/proteins which are capable of binding to specific target cells. Embodiments further include adjoining all or part of an aglycosylated Fc domain to all or part of any of the proteins listed in Table 2. It is contemplated that embodiments include, but are not limited to, the examples provided in Table 2 and the description herein.
- a ligand for receptor may be employed to target a cell expressing on its surface the receptor for the ligand.
- Ligands also include, for instance, CD95 ligand, TRAIL, TNF (such as TNF- ⁇ . or TNF- ⁇ ), growth factors, including those discussed above, such as VEGF and cytokines, such as interferons or interleukins and variants thereof.
- VEGF Trap fusion protein that includes the second extracellular domain of the VEGF receptor 1 (FIt-I) with the third domain of the VEGF receptor 2 (KDR/FIK-1) and an IgG Fc region.
- a specialized kind of insertional variant is the fusion protein.
- This molecule generally has all or a substantial portion of the native molecule, linked at the N- or C-terminus, to all or a portion of a second polypeptide.
- Embodiments also concern conjugated polypeptides, such as translated proteins, polypeptides and peptides, that are linked to at least one agent to form a modified protein or polypeptide.
- conjugated polypeptides such as translated proteins, polypeptides and peptides
- it is conventional to link or covalently bind or complex at least one desired molecule or moiety.
- a molecule or moiety may be, but is not limited to, at least one effector or reporter molecule.
- Effector molecules comprise molecules having a desired activity, e.g., cytotoxic activity.
- Non- limiting examples of effector molecules which have been attached to antibodies include toxins, anti-tumor agents, therapeutic enzymes, radio-labeled nucleotides, antiviral agents, chelating agents, cytokines, growth factors, and oligo- or poly-nucleotides.
- a reporter molecule is defined as any moiety that may be detected using an assay.
- Non-limiting examples of reporter molecules which have been conjugated to antibodies include enzymes, radiolabels, haptens, fluorescent labels, phosphorescent molecules, chemiluminescent molecules, chromophores, luminescent molecules, photoaffinity molecules, colored particles or ligands, such as biotin.
- Any antibody of sufficient selectivity, specificity or affinity may be employed as the basis for an antibody conjugate. Such properties may be evaluated using conventional immunological screening methodology known to those of skill in the art.
- Sites for binding to biological active molecules in the antibody molecule include sites that reside in the variable domain that can bind pathogens, B-cell superantigens, the T cell co- receptor CD4 and the HIV-I envelope (Sasso et al, 1989; Shorki et ⁇ /., 1991; Silvermann et al, 1995; Cleary et al, 1994; Lenert et al, 1990; Berberian et al, 1993; Kreier et ⁇ /., 1991).
- the variable domain is involved in antibody self-binding (Kang et al., 1988), and contains epitopes (idiotopes) recognized by anti- antibodies (Kohler et al, 1989).
- antibody conjugates are those conjugates in which the antibody is linked to a detectable label.
- Detectable labels are compounds and/or elements that can be detected due to their specific functional properties, and/or chemical characteristics, the use of which allows the antibody to which they are attached to be detected, and/or further quantified if desired.
- Another such example is the formation of a conjugate comprising an antibody linked to a cytotoxic or anti-cellular agent, and may be termed "immunotoxins.”
- Amino acids such as selectively-cleavable linkers, synthetic linkers, or other amino acid sequences may be used to separate proteinaceous moieties.
- embodiments may involve methods and processes for purifying proteins, including modified proteins and recombinant proteins.
- these techniques involve, at one level, the crude fractionation of the cellular milieu to polypeptide and non- polypeptide fractions. Having separated the polypeptide from other proteins, the polypeptide of interest may be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or purification to homogeneity).
- Analytical methods particularly suited to the preparation are ion- exchange chromatography, exclusion chromatography; polyacrylamide gel electrophoresis; isoelectric focusing.
- a particularly efficient method of purifying peptides is fast protein liquid chromatography or even HPLC.
- the conditions under which such techniques are executed may be affect characteristics, such as functional activity, of the purified molecules.
- Certain aspects concern the purification, and in particular embodiments, the substantial purification, of an encoded protein or peptide.
- the term "purified protein or peptide” as used herein, is intended to refer to a composition, isolatable from other components, wherein the protein or peptide is purified to any degree relative to its naturally-obtainable state.
- a purified protein or peptide therefore also refers to a protein or peptide, free from the environment in which it may naturally occur.
- purified will refer to a protein or peptide composition that has been subjected to fractionation to remove various other components, and which composition substantially retains its expressed biological activity. Where the term “substantially purified” is used, this designation will refer to a composition in which the protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, about 99.2%, about 99.4%, about 99.6%, about 99.8%, about 99.9% or more of the proteins in the composition.
- Various methods for quantifying the degree of purification of the protein or peptide will be known to those of skill in the art in light of the present disclosure. These include, for example, determining the specific activity of an active fraction, or assessing the amount of polypeptides within a fraction by SDS/PAGE analysis.
- a preferred method for assessing the purity of a fraction is to calculate the specific activity of the fraction, to compare it to the specific activity of the initial extract, and to thus calculate the degree of purity, herein assessed by a "-fold purification number.”
- the actual units used to represent the amount of activity will, of course, be dependent upon the particular assay technique chosen to follow the purification and whether or not the expressed protein or peptide exhibits a detectable activity.
- a tag takes advantage of an interaction between two polypeptides.
- a portion of one of the polypeptides that is involved in the interaction may be used as a tag.
- GST glutathione S transferase
- An epitope tag which an amino acid region recognized by an antibody or T cell receptor, may be used.
- the tag may be encoded by a nucleic acid segment that is operatively linked to a nucleic acid segment encoding a modified protein such that a fusion protein is encoded by the nucleic acid molecule.
- Other suitable fusion proteins are those with ⁇ -galactosidase, ubiquitin, hexahistidine (6xHis), or the like.
- Examples of techniques that could be employed in conjunction with embodiments for creation of diverse antibody Fc domains and/or antibodies comprising such domains may employ techniques similar to those for expression of immunoglobulin heavy chain libraries described in U.S. Patent 5,824,520. Previously employed Fc libraries are discussed in WO 2008/137475, which is specifically incorporated by reference. V. Screening Antibody Fc Domains
- the binding polypeptides screened may comprise a large library of diverse candidate Fc domains, or, alternatively, may comprise particular classes of Fc domains (e.g., engineered point mutations or amino acid insertions) selected with an eye towards structural attributes that are believed to make them more likely to bind the target ligand.
- the candidate binding protein is an intact antibody, or a fragment or portion thereof comprising an Fc domain.
- a candidate Fc domain capable of binding a target ligand one may carry out the steps of: providing a population of Gram negative bacterial cells that express a distinct antibody Fc domain; admixing the bacteria or phages and at least a first labeled or immobilized target ligand (FcR polypeptide) capable of contacting the antibody and identifying at least a first bacterium expressing a molecule capable of binding the target ligand.
- FcR polypeptide first labeled or immobilized target ligand
- the binding between antibody Fc domain and a labeled FcR polypeptide will prevent diffusing out of a bacterial cell.
- molecules of the labeled ligand can be retained in the periplasm of the bacterium comprising a permeablized outer membrane.
- the periplasm can be removed, whereby the Fc domain will cause retention of the bound candidate molecule since Fc domains are shown to associate with the inner membrane.
- the labeling may then be used to isolate the cell expressing a binding polypeptide capable of binding the FcR polypeptide, and in this way, the gene encoding the Fc domain polypeptide isolated.
- the molecule capable of binding the target ligand may then be produced in large quantities using in vivo or ex vivo expression methods, and then used for any desired application, for example, for diagnostic or therapeutic applications.
- isolated antibody Fc domains identified may be used to construct an antibody fragment or full- length antibody comprising an antigen binding domain.
- methods for producing bacteria of the invention may comprise at least two rounds of selection (step c) wherein the sub- population of bacterial cells obtained in the first round of selection is subjected to at least a second round of selection based on the binding of the candidate antibody Fc domain to an FcR.
- the sub-population of bacterial cells obtained in the first round of selection may be grown under permissive conditions prior to a second selection (to expand the total number of cells).
- methods may comprise 2, 3, 4, 5, 6, 7, 8, 9, 10 or more rounds of selection.
- a sub-population of bacterial cells obtained from each round of selection will be grown under permissive conditions before a subsequent round of selection.
- Cells isolated following one or more such rounds of selection may be subjected to additional rounds of mutagenesis.
- selection will be performed after removing FcR polypeptide that is not bound to the antibody.
- the stringency of selection may be modified by adjusting the pH, salt concentration, or temperature of a solution comprising bacteria that display antibodies.
- a bacterial cell of the invention is grown at a sub-physiological temperature such as at about 25 0 C.
- a method of producing a bacterial cell according to the invention may be further defined as a method of producing a nucleic acid sequence encoding an Fc domain that binds to at least a first FcR.
- a bacterial cell produced by the methods herein may be used to clone a nucleic acid sequence encoding the Fc domain having a specific affinity for an FcR polypeptide.
- Methods for isolating and amplifying such a nucleic acid from a cell for example by PCR are well known in the art and further described below.
- a nucleic acid sequence produced by the forgoing methods is included as part of the instant invention.
- the invention provides a method for producing an Fc domain having a specific affinity for an FcR.
- the invention includes antibody Fc domains produced by the methods of the invention. It will be understood however that the antibody Fc domains produced by such a screen may be combine with antibody variable regions that have an affinity for a particular target ligand and these antibodies are also included as part of the invention.
- binding affinity of an antibody Fc or other binding protein can, for example, be determined by the Scatchard analysis of Munson & Pollard (1980). Alternatively, binding affinity can be determined by surface plasmon resonance or any other well known method for determining the kinetics and equilibrium constants for protein:protein interactions. After a bacterial cell is identified that produces molecules of the desired specificity, affinity, and/or activity, the corresponding coding sequence may be cloned. In this manner, DNA encoding the molecule can be isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the antibody or binding protein).
- the antibody Fc domain DNA may be placed into expression vectors, which can then transfected into host cells such as bacteria.
- the DNA also may be modified, for example, by the addition of sequence for human heavy and light chain variable domains, or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non- immunoglobulin polypeptide.
- “chimeric” or “hybrid” binding proteins are prepared to have the desired binding specificity.
- an identified antibody Fc domain may be fused to a therapeutic polypeptide or a toxin and used to target cells (in vitro or in vivo) that express a particular FcR.
- Chimeric or hybrid Fc domains also may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents.
- targeted-toxins may be constructed using a disulfide exchange reaction or by forming a thioether bond.
- suitable reagents for this purpose include iminothiolate and methyl-4-mercaptobutyrimidate.
- nucleic acids may be cloned from viable or inviable cells.
- inviable cells for example, it may be desired to use amplification of the cloned DNA, for example, using PCR. This may also be carried out using viable cells either with or without further growth of cells.
- an Fc domain is isolated which has affinity for a labeled FcR polypeptide.
- labeled ligands of potentially any size may be screened. In the absence of removal of the periplasmic membrane, it will typically be preferable that the labeled ligand is less that 50,000 Da in size in order to allow efficient diffusion of the ligand across the bacterial periplasmic membrane.
- FcR polypeptide which has been labeled with one or more detectable agent(s). This can be carried out, for example, by linking the ligand to at least one detectable agent to form a conjugate. For example, it is conventional to link or covalently bind or complex at least one detectable molecule or moiety.
- a "label” or “detectable label” is a compound and/or element that can be detected due to specific functional properties, and/or chemical characteristics, the use of which allows the ligand to which it is attached to be detected, and/or further quantified if desired.
- labels examples include, but are not limited to, enzymes, radiolabels, haptens, fluorescent labels, phosphorescent molecules, chemiluminescent molecules, chromophores, luminescent molecules, photoaff ⁇ nity molecules, colored particles or ligands, such as biotin.
- a visually-detectable marker is used such that automated screening of cells for the label can be carried out.
- fluorescent labels are beneficial in that they allow use of flow cytometry for isolation of cells expressing a desired binding protein or antibody.
- agents that may be detected by visualization with an appropriate instrument are known in the art, as are methods for their attachment to a desired ligand (see, e.g., U.S. Patents 5,021,236; 4,938,948; and 4,472,509, each incorporated herein by reference).
- agents can include paramagnetic ions; radioactive isotopes; fluorochromes; NMR-detectable substances and substances for X-ray imaging.
- FcR conjugate is where the ligand is linked to a secondary binding molecule and/or to an enzyme (an enzyme tag) that will generate a colored product upon contact with a chromogenic substrate.
- enzymes include urease, alkaline phosphatase, (horseradish) hydrogen peroxidase or glucose oxidase. I n such instances, it will be desired that cells selected remain viable.
- Preferred secondary binding ligands are biotin and/or avidin and streptavidin compounds. The use of such labels is well known to those of skill in the art and are described, for example, in U.S. Patents 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149 and 4,366,241; each incorporated herein by reference.
- Molecules containing azido groups also may be used to form covalent bonds to proteins through reactive nitrene intermediates that are generated by low intensity ultraviolet light (Potter & Haley, 1983).
- 2- and 8-azido analogues of purine nucleotides have been used as site-directed photoprobes to identify nucleotide -binding proteins in crude cell extracts (Owens & Haley, 1987; Atherton et al., 1985).
- the 2- and 8-azido nucleotides have also been used to map nucleotide-binding domains of purified proteins (Khatoon et al., 1989; King et al., 1989; and Dholakia et al., 1989) and may be used as ligand binding agents.
- Labeling can be carried out by any of the techniques well known to those of skill in the art.
- FcR polypeptides can be labeled by contacting the ligand with the desired label and a chemical oxidizing agent such as sodium hypochlorite, or an enzymatic oxidizing agent, such as lactoperoxidase.
- a ligand exchange process could be used.
- direct labeling techniques may be used, e.g., by incubating the label, a reducing agent such as SNCl 2 , a buffer solution such as sodium-potassium phthalate solution, and the ligand.
- Intermediary functional groups on the ligand could also be used, for example, to bind labels to a ligand in the presence of diethylenetriaminepentaacetic acid (DTPA) or ethylene diaminetetracetic acid (EDTA).
- DTPA diethylenetriaminepentaacetic acid
- EDTA ethylene diaminetetracetic acid
- chelating agent such as diethylenetriaminepentaacetic acid anhydride (DTPA); ethylenetriaminetetraacetic acid; N-chloro-p-toluenesulfonamide; and/or tetrachloro-3 ⁇ -6 ⁇ -diphenylglycouril-3 attached to the ligand (U.S. Patents 4,472,509 and 4,938,948, each incorporated herein by reference).
- FcR polypeptides also may be reacted with an enzyme in the presence of a coupling agent such as glutaraldehyde or periodate.
- Conjugates with fluorescein markers can be prepared in the presence of these coupling agents or by reaction with an isothiocyanate.
- imaging of breast tumors is achieved using monoclonal antibodies and the detectable imaging moieties are bound to the antibody using linkers such as methyl-p-hydroxybenzimidate or N-succinimidyl-3-(4-hydroxyphenyl)propionate.
- linkers such as methyl-p-hydroxybenzimidate or N-succinimidyl-3-(4-hydroxyphenyl)propionate.
- an FcR polypeptide may be fused to a reporter protein such as an enzyme as described supra or a fluorescence protein.
- the ability to specifically label periplasmic expressed proteins with appropriate fluorescent ligands also has applications other than library screening. Specifically labeling with fluorescent ligands and flow cytometry can be used for monitoring production of Fc domains during protein manufacturing.
- an Fc domain may be desired to link the molecule to at least one agent to form a conjugate to enhance the utility of that molecule.
- it is conventional to link or covalently bind or complex at least one desired molecule or moiety.
- a molecule or moiety may be, but is not limited to, at least one effector or reporter molecule.
- Effecter molecules comprise molecules having a desired activity, e.g. , cytotoxic activity.
- Non- limiting examples of effector molecules which have been attached to antibodies include toxins, anti-tumor agents, therapeutic enzymes, radio-labeled nucleotides, antiviral agents, chelating agents, cytokines, growth factors, and oligo- or polynucleotides.
- a reporter molecule is defined as any moiety which may be detected using an assay. Techniques for labeling such a molecule are known to those of skill in the art and have been described herein above.
- Labeled binding proteins such as Fc domains which have been prepared in accordance with the invention may also then be employed, for example, in immunodetection methods for binding, purifying, removing, quantifying and/or otherwise generally detecting biological components such as protein(s), polypeptide(s) or peptide(s).
- immunodetection methods include enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoradiometric assay, fluoroimmunoassay, chemiluminescent assay, bioluminescent assay, and Western blot to mention a few.
- the Fc domain molecules may be used, for example, in conjunction with both fresh-frozen and/or formalin-fixed, paraffin- embedded tissue blocks prepared for study by immunohistochemistry (IHC).
- IHC immunohistochemistry
- the method of preparing tissue blocks from these particulate specimens has been successfully used in previous IHC studies of various prognostic factors, and/or is well known to those of skill in the art (Abbondanzo et al., 1990).
- FACS fluorescence activated cell sorting
- Instruments for carrying out flow cytometry are known to those of skill in the art and are commercially available to the public. Examples of such instruments include FACS Star Plus, FACScan and FACSort instruments from Becton Dickinson (Foster City, Calif.) Epics C from Coulter Epics Division (Hialeah, FIa.) and MOFLOTM from Cytomation (Colorado Springs, Co).
- Flow cytometric techniques in general involve the separation of cells or other particles in a liquid sample.
- the purpose of flow cytometry is to analyze the separated particles for one or more characteristics thereof, for example, presence of a labeled ligand or other molecule.
- the basis steps of flow cytometry involve the direction of a fluid sample through an apparatus such that a liquid stream passes through a sensing region.
- the particles should pass one at a time by the sensor and are categorized base on size, refraction, light scattering, opacity, roughness, shape, fluorescence, etc.
- Quantitative analysis of multiparameter flow cytometric data for rapid cell detection consists of two stages: cell class characterization and sample processing.
- cell class characterization partitions the cell feature into cells of interest and not of interest.
- sample processing each cell is classified in one of the two categories according to the region in which it falls. Analysis of the class of cells is very important, as high detection performance may be expected only if an appropriate characteristic of the cells is obtained.
- an important aspect of flow cytometry is that multiple rounds of screening can be carried out sequentially.
- Cells may be isolated from an initial round of sorting and immediately reintroduced into the flow cytometer and screened again to improve the stringency of the screen.
- Another advantage known to those of skill in the art is that nonviable cells can be recovered using flow cytometry. Since flow cytometry is essentially a particle sorting technology, the ability of a cell to grow or propagate is not necessary. Techniques for the recovery of nucleic acids from such non-viable cells are well known in the art and may include, for example, use of template-dependent amplification techniques including PCR.
- Nucleic acid-based expression systems may find use, in certain embodiments of the invention, for the expression of recombinant proteins.
- one embodiment of the invention involves transformation of Gram negative bacteria with the coding sequences for an antibody Fc domain, or preferably a plurality of distinct Fc domains.
- Nucleic acid-based expression systems may find use, in certain embodiments of the invention, for the expression of recombinant proteins.
- one embodiment of the invention involves transformation of Gram negative bacteria with the coding sequences for an antibody Fc domain, or preferably a plurality of distinct Fc domains.
- Certain aspects of the invention may comprise delivery of nucleic acids to target cells (e.g., gram negative bacteria).
- target cells e.g., gram negative bacteria
- bacterial host cells may be transformed with nucleic acids encoding candidate Fc domains potentially capable binding an FcR.
- it may be desired to target the expression to the periplasm of the bacteria. Transformation of eukaryotic host cells may similarly find use in the expression of various candidate molecules identified as capable of binding a target ligand.
- Suitable methods for nucleic acid delivery for transformation of a cell are believed to include virtually any method by which a nucleic acid (e.g., DNA) can be introduced into such a cell, or even an organelle thereof.
- a nucleic acid e.g., DNA
- Such methods include, but are not limited to, direct delivery of DNA such as by injection (U.S. Patents 5,994,624, 5,981,274, 5,945,100, 5,780,448, 5,736,524, 5,702,932, 5,656,610, 5,589,466 and 5,580,859, each incorporated herein by reference), including microinjection (Harland and Weintraub, 1985; U.Sf. Patent 5,789,215, incorporated herein by reference); by electroporation (U.S.
- Patent 5,384,253, incorporated herein by reference by calcium phosphate precipitation (Graham and Van Der Eb, 1973; Chen and Okayama, 1987; Rippe et al, 1990); by using DEAE-dextran followed by polyethylene glycol (Gopal, 1985); by direct sonic loading (Fechheimer et al, 1987); by liposome mediated transfection (Nicolau and Sene, 1982; Fraley et al, 1979; Nicolau et al, 1987; Wong et al, 1980; Kaneda et al, 1989; Kato et al, 1991); by microprojectile bombardment (PCT Application Nos. WO 94/09699 and 95/06128; U.S.
- Vectors may find use with the current invention, for example, in the transformation of a Gram negative bacterium with a nucleic acid sequence encoding a candidate Fc domain which one wishes to screen for ability to bind a target FcR.
- an entire heterogeneous "library" of nucleic acid sequences encoding target polypeptides may be introduced into a population of bacteria, thereby allowing screening of the entire library.
- vector is used to refer to a carrier nucleic acid molecule into which a nucleic acid sequence can be inserted for introduction into a cell where it can be replicated.
- a nucleic acid sequence can be "exogenous,” or “heterologous”, which means that it is foreign to the cell into which the vector is being introduced or that the sequence is homologous to a sequence in the cell but in a position within the host cell nucleic acid in which the sequence is ordinarily not found.
- Vectors include plasmids, cosmids and viruses (e.g., bacteriophage).
- plasmids include plasmids, cosmids and viruses (e.g., bacteriophage).
- viruses e.g., bacteriophage
- expression vector refers to a vector containing a nucleic acid sequence coding for at least part of a gene product capable of being transcribed. In some cases, RNA molecules are then translated into a protein, polypeptide, or peptide.
- Expression vectors can contain a variety of "control sequences,” which refer to nucleic acid sequences necessary for the transcription and possibly translation of an operably linked coding sequence in a particular host organism. In addition to control sequences that govern transcription and translation, vectors and expression vectors may contain nucleic acid sequences that serve other functions as well and are described infra.
- a “promoter” is a control sequence that is a region of a nucleic acid sequence at which initiation and rate of transcription are controlled. It may contain genetic elements at which regulatory proteins and molecules may bind such as RNA polymerase and other transcription factors.
- the phrases "operatively positioned,” “operatively linked,” “under control,” and “under transcriptional control” mean that a promoter is in a correct functional location and/or orientation in relation to a nucleic acid sequence to control transcriptional initiation and/or expression of that sequence.
- a promoter may or may not be used in conjunction with an “enhancer,” which refers to a cis-acting regulatory sequence involved in the transcriptional activation of a nucleic acid sequence.
- a promoter may be one naturally associated with a gene or sequence, as may be obtained by isolating the 5' non-coding sequences located upstream of the coding segment and/or exon. Such a promoter can be referred to as "endogenous.”
- an enhancer may be one naturally associated with a nucleic acid sequence, located either downstream or upstream of that sequence.
- certain advantages will be gained by positioning the coding nucleic acid segment under the control of a recombinant or heterologous promoter, which refers to a promoter that is not normally associated with a nucleic acid sequence in its natural environment.
- a recombinant or heterologous enhancer refers also to an enhancer not normally associated with a nucleic acid sequence in its natural environment.
- Such promoters or enhancers may include promoters or enhancers of other genes, and promoters or enhancers isolated from any other prokaryotic cell, and promoters or enhancers not "naturally occurring," i.e., containing different elements of different transcriptional regulatory regions, and/or mutations that alter expression.
- sequences may be produced using recombinant cloning and/or nucleic acid amplification technology, including PCRTM, in connection with the compositions disclosed herein (see U.S. Patent 4,683,202, U.S. Patent 5,928,906, each incorporated herein by reference).
- promoter and/or enhancer that effectively directs the expression of the DNA segment in the cell type chosen for expression.
- promoter that may be used with the invention is the E. coli arabinose or T7 promoter.
- the promoters employed may be constitutive, tissue-specific, inducible, and/or useful under the appropriate conditions to direct high level expression of the introduced DNA segment, such as is advantageous in the large-scale production of recombinant proteins and/or peptides.
- the promoter may be heterologous or endogenous.
- a specific initiation signal also may be required for efficient translation of coding sequences. These signals include the ATG initiation codon or adjacent sequences. Exogenous translational control signals, including the ATG initiation codon, may need to be provided. One of ordinary skill in the art would readily be capable of determining this and providing the necessary signals. It is well known that the initiation codon must be "in-frame" with the reading frame of the desired coding sequence to ensure translation of the entire insert. The exogenous translational control signals and initiation codons can be either natural or synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements.
- Vectors can include a multiple cloning site (MCS), which is a nucleic acid region that contains multiple restriction enzyme sites, any of which can be used in conjunction with standard recombinant technology to digest the vector (see Carbonelli et al., 1999, Levenson et al., 1998, and Cocea, 1997, incorporated herein by reference.)
- MCS multiple cloning site
- Restriction enzyme digestion refers to catalytic cleavage of a nucleic acid molecule with an enzyme that functions only at specific locations in a nucleic acid molecule. Many of these restriction enzymes are commercially available. Use of such enzymes is understood by those of skill in the art.
- a vector is linearized or fragmented using a restriction enzyme that cuts within the MCS to enable exogenous sequences to be ligated to the vector.
- "Ligation” refers to the process of forming phosphodiester bonds between two nucleic acid fragments, which may or may not be contiguous with each other. Techniques involving restriction enzymes and ligation reactions are well known to those of skill in the art of recombinant technology.
- the vectors or constructs prepared in accordance with the present invention will generally comprise at least one termination signal.
- a “termination signal” or “terminator” is comprised of the DNA sequences involved in specific termination of an RNA transcript by an RNA polymerase.
- a termination signal that ends the production of an RNA transcript is contemplated.
- a terminator may be necessary in vivo to achieve desirable message levels.
- Terminators contemplated for use in the invention include any known terminator of transcription described herein or known to one of ordinary skill in the art, including but not limited to, for example, rhp dependent or rho independent terminators.
- the termination signal may be a lack of transcribable or translatable sequence, such as due to a sequence truncation.
- a vector in a host cell may contain one or more origins of replication sites (often termed "ori"), which is a specific nucleic acid sequence at which replication is initiated.
- ori origins of replication sites
- cells containing a nucleic acid construct of the present invention may be identified in vitro or in vivo by including a marker in the expression vector.
- markers would confer an identifiable change to the cell permitting easy identification of cells containing the expression vector.
- a selectable marker is one that confers a property that allows for selection.
- a positive selectable marker is one in which the presence of the marker allows for its selection, while a negative selectable marker is one in which its presence prevents its selection.
- An example of a positive selectable marker is a drug resistance marker.
- a drug selection marker aids in the cloning and identification of transformants, for example, genes that confer resistance to neomycin, puromycin, hygromycin, DHFR, GPT, zeocin and histidinol are useful selectable markers.
- markers conferring a phenotype that allows for the discrimination of transformants based on the implementation of conditions other types of markers including screenable markers such as GFP, whose basis is colorimetric analysis, are also contemplated.
- screenable enzymes such as chloramphenicol acetyltransferase (CAT) may be utilized.
- CAT chloramphenicol acetyltransferase
- One of skill in the art would also know how to employ immunologic markers, possibly in conjunction with FACS analysis. The marker used is not believed to be important, so long as it is capable of being expressed simultaneously with the nucleic acid encoding a gene product. Further examples of selectable and screenable markers are well known to one of skill in the art.
- host cell refers to a prokaryotic cell, and it includes any transformable organism that is capable of replicating a vector and/or expressing a heterologous gene encoded by a vector.
- a host cell can, and has been, used as a recipient for vectors.
- a host cell may be "transfected” or “transformed,” which refers to a process by which exogenous nucleic acid is transferred or introduced into the host cell.
- a transformed cell includes the primary subject cell and its progeny.
- a host cell is a Gram negative bacterial cell.
- Gram negative bacteria are suited for use with the invention in that they posses a periplasmic space between the inner and outer membrane and, particularly, the aforementioned inner membrane between the periplasm and cytoplasm, which is also known as the cytoplasmic membrane.
- any other cell with such a periplasmic space could be used in accordance with the invention.
- Gram negative bacteria that may find use with the invention may include, but are not limited to, E.
- the Gram negative bacterial cell may be still further defined as bacterial cell which has been transformed with the coding sequence of a fusion polypeptide comprising a candidate binding polypeptide capable of binding a selected ligand.
- the polypeptide is anchored to the outer face of the cytoplasmic membrane, facing the periplasmic space, and may comprise an antibody coding sequence or another sequence.
- One means for expression of the polypeptide is by attaching a leader sequence to the polypeptide capable of causing such directing.
- a plasmid or cosmid for example, can be introduced into a prokaryote host cell for replication of many vectors.
- Bacterial cells used as host cells for vector replication and/or expression include DH5 ⁇ , JM 109, and KC8, as well as a number of commercially available bacterial hosts such as SURE ® Competent Cells and SOLOPACKTM Gold Cells (STRATAGENE ® , La Jolla).
- bacterial cells such as E. coli LE392 could be used as host cells for bacteriophage.
- a viral vector may be used in conjunction with a prokaryotic host cell, particularly one that is permissive for replication or expression of the vector.
- Some vectors may employ control sequences that allow it to be replicated and/or expressed in both prokaryotic and eukaryotic cells.
- One of skill in the art would further understand the conditions under which to incubate all of the above described host cells to maintain them and to permit replication of a vector. Also understood and known are techniques and conditions that would allow large-scale production of vectors, as well as production of the nucleic acids encoded by vectors and their cognate polypeptides, proteins, or peptides. D. Expression Systems
- compositions discussed above could be used, for example, for the production of a polypeptide product identified in accordance with the invention as capable of binding a particular ligand.
- Prokaryote -based systems can be employed for use with the present invention to produce nucleic acid sequences, or their cognate polypeptides, proteins and peptides. Many such systems are commercially and widely available.
- Other examples of expression systems comprise of vectors containing a strong prokaryotic promoter such as T7, Tac, Trc, BAD, lambda pL, Tetracycline or Lac promoters, the pET Expression System and an E. coli expression system.
- antibody Fc domains are expressed on the cytoplasmic or in the periplasmic space membrane of a host bacterial cell. By expression of a heterogeneous population of such Fc domains, those polypeptides having a high affinity for a target ligand (FcR) may be identified. The identified Fc domains may then be used in various diagnostic or therapeutic applications, as described herein.
- FcR target ligand
- Fc domain is intended to refer broadly to any immunoglobulin Fc region such as an IgG, IgM, IgA, IgD or IgE Fc.
- the techniques for preparing and using various antibody-based constructs and fragments are well known in the art. Means for preparing and characterizing antibodies are also well known in the art (See, e.g., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988; incorporated herein by reference).
- the Fc domain may be purified, if desired, using filtration, centrifugation and various chromatographic methods such as HPLC or affinity chromatography.
- Fc domains, or polypeptides and peptides more generally, can be synthesized using an automated peptide synthesizer.
- the amplified PCR fragments were ligated into Sfil digested pPelBFLAG.
- the resulting plasmids were transformed into E. coli Jude-1(F' [TnIO(TQf) pro AB + lacP A(lacZ)Ml5] mcrA Umrr-hsdRMS-mcrBC) ⁇ 80d/ ⁇ cZ ⁇ M15 MacX74 deoR recAl araD139 A(ara leu)1691 galU galK rpsL endAl nupG) (Kawarasaki et al., 2003). Based on the sequence of 20 library clones randomly selected, the library was 7 x 108 individual transformats with 0.264% error rate per gene (FIG. 3).
- the washed cells were incubated in 1 ml of Solution A with 1 mg/ml of hen egg lysozyme at 37°C for 15 min. After centrifugation at 12,000 x g for 1 min and the resulting spheroplasts pellets were resuspended in 1 ml of cold PBS.
- extracellular domain of recombinant glycosylated Fc ⁇ PJa/CD64 was labeled with FITC using FITC protein labeling kit (Invitrogen, Carlsbad, CA).
- FITC protein labeling kit Invitrogen, Carlsbad, CA.
- the affinity of FITC labeled Fc ⁇ RI for human IgG Fc was confirmed by fluorescent ELISA displaying high fluorescence in the Fc glycosylated human IgG-Fc coated wells comparing in the BSA coated wells.
- Spheroplasts were labeled with 30 nM of Fc ⁇ RI-FITC.
- Fc ⁇ RIa-FITC reduced concentration of Fc ⁇ RIa-FITC (10, 3, and 1 nM for the 2 nd , 3 rd , and 4 th round sorting, respectively) were used for labeling of spheroplasts. More than 4 x 10 8 spheroplasts were sorted by MoFIo (Dako Cytomation, Fort Collins, CO) equipped with a 488 nm argon laser for excitation. In each round the top 3% of the population showing the highest fluorescence due to Fc ⁇ RIa-FITC binding was isolated by sorting and resorting immediately after the initial sorting.
- MoFIo Deoxyfluorescence
- Fc genes in the spheroplasts were rescued by PCR amplification using two specific primers (STJ#16 and STJ#220), ligated into pPelBFLAG-Fc using S ⁇ l restriction enzyme site, and transformed in electrocompetent E. coli Jude-1 cells. The resulting transformants were selected on chloramphenicol containing media and then grown, spheroplasted as above in preparation for the next round of sorting. High fluorescent clones were enriched as sorting rounds go on (FIG. 4). After the 5 th round of sorting, 19 individual clones exhibiting higher fluorescence than Fc5 were isolated (FIG. 5).
- Fc5 Nucleotide Sequence #2 and Protein Sequence #2
- Fc5 have 2 mutations (E382V and M428I) in the sequence of wild type IgGl-Fc (Nucleotide Sequence #1 and Protein Sequence #1).
- the engineered Fc mutants exhibiting higher affinity to Fc ⁇ RIa than Fc5 have substitution mutations in the sequence of Fc5.
- Isolated Fc mutants, Fc601-Fc619 Protein Sequence #3 ⁇ #21), have substitution mutations in the sequence of Fc5.
- the isolated mutants are summarized in Table 5.
- trastuzumab (HerceptinTM) has been clinically used for the treatment of breast metastatic carcinoma that overexpress HER2/neu (Erb2) (Sergina and Moasser, 2007).
- trastuzumab antibodies recognize HER2/neu (Erb2) and interact with surface Fc ⁇ Rs of immune cells leading to antibody-dependent cell-mediated cytotoxicity (ADCC), an essential effector function mechanism for therapeutic action (Lazar et al., 2006; Sergina and Moasser, 2007).
- Fc fragment genes engineered for high Fc ⁇ Rs affinity were incorporated to full length trastuzumab antibodies.
- pSTJ4- Herceptin IgGl E.
- V L and V H domains of humanized 4D5 were synthesized by total gene synthesis with overlap extension PCR using 12 oligonucleotides that included 2 external primers (STJ#302 and STJ#313) and 10 internal primers (STJ#303-312) for V L and 14 primers total 2 external primers (STJ#314 and STJ#327) and 12 internal primers (STJ#315-326) for V H, respectively.
- Fc5 and Fc2a mutant genes were amplified using the primers (STJ#290 and STJ#291) and the templates, pPelBFLAG-Fc5 or pPelBFLAG-Fc601, ligated into pSTJ4-Herceptin IgGl digested using Sail I EcoKV ' .
- pSTJ4-Herceptin-Fc2a-IgGl For pSTJ4- Herceptin-Fc2a-IgGl and pSTJ4-Herceptin-Fc5-IgGl, Fc5 and Fc2a mutant genes were amplified using the primers (STJ#290 and STJ#291) and the templates, pPelBFLAG-Fc5 or pPelBFLAG-Fc601, ligated into pSTJ4-Herceptin IgGl digested using Sail I EcoKV ' .
- coli dicistronic plasmids, pSTJ4- Herceptin-IgGl, pSTJ4-Herceptin-Fc5-IgGl, and pSTJ4-Herceptin-Fc601-IgGl were constructed. These plasmids are under the control of lac promter in a dicistronic operon with PeIB leader peptide fusions to both heavy and light chains (FIG. 9). [00190] After transformation of the plasmids into E.
- E. coli BL21(DE3) harboring pSTJ4-Herceptin-IgGl, pSTJ4-Herceptin-IgGl-Fc5, or pSTJ4-Herceptin-IgGl-Fc601 were cultured in 500 mL baffled-flask with 120 ml R/2 media at 30 0 C at 250 rpm for 8 h and then inoculated to 3.3L BioFlo 310 fermentor (New Brunswick Scientific Co., Edison, NJ) with 1.2 L R/2 medium. Fed-batch fermentation was performed at 30 0 C using pH-stat glucose feeding strategy.
- the dissolved oxygen (DO) concentration was maintained at 40% of air saturation using automatic cascade control by increasing agitation speed from 100 rpm to 1000 rpm, air flow rate from 1 to 3 SLPM (Standard liquid per minute) and pure oxygen flow rate from 0 to 1.5 SLPM when required.
- the initial pH was adjusted to 6.8 and controlled by the addition of 30% (v/v) ammonium hydroxide when it decreased to less than 6.75 and by the supply of feeding solutions, (700 g/L of glucose and 10 g/L of MgSO47H2O; before induction) and (500 g/L glucose, 10 g/L of MgSO47H2O, and 100 g/L of yeast extract; after induction), when it increased to more than 6.9.
- wild type aglycosylated trastuzumab, aglycosylated trastuzumab-Fc5, and aglycosylated trastuzumab-Fc601 were eluted from the resin using 15 ml of 0.1 M glycine (pH 3.0) and neutralized immediately with IM Tris (pH 8.0) solution.
- the eluted samples were concentrated by ultrafiltration through a 10 kDa MW cutoff membrane and the retentate was applied to a Superdex 200 gel filtration column developed with PBS (pH 7.4).
- Example 5 Affinity of aglycosylated trastuzumab-Fc601 to Fc receptors
- Affinity of full assembled aglycosylated trastuzumab antibodies to Fc ⁇ RIa was measured by immobilizing glycosylated trastuzumab (Clinical grade, Fox Chase Cancer Center Pharmacy), aglycosylated trastuzumab, and aglycosylated trastuzumab-Fc5, aglycosylated trastuzumab-Fc601 individually on the CM-5 sensor chip.
- the soluble monomeric Fc ⁇ RIa in HBS-EP (10 mM HEPES pH 7.4, 150 mM NaCl, 3.4 mM EDTA, and 0.005% P20 surfactant) buffer was injected at flow rate of 30 ⁇ l/min for 60 s with dissociation time 300 s. Regeneration of the ligand was performed by single injection of 100 mM citric acid, pH 3.0.
- Affinities of the soluble monomeric Fc ⁇ RIa with glycosylated trastuzumab, aglycosylated trastuzumab, trastuzumab-Fc5, and trastuzumab-Fc601 were obtained by injection of soluble Fc ⁇ RIa in duplicate at concentrations of 0, 25, 50, 100, 200 nM for 60 s at a flow rate of 30 ⁇ l/min over immobilized glycosylated trastuzumab, trastuzumab, trastuzumab- Fc5, and trastuzumab Fc601.
- Affinity of Fc ⁇ RIa toward wild type aglycosylated trastuzumab was obtained by Fc ⁇ RIa injections in duplicates at concentrations 0, 200, 300, 400, 500, and 600 nM for 60 s at a flow rate 30 ⁇ l/min over immobilized aglycosylated trastuzumab. Binding curve at zero concentration was subtracted as a blank. Equilibrium dissociation constants (K D ) were determined by fitting of equilibrium responses to steady-state affinity model provided by BIAevaluation 3.0 software. As shown in FIG.
- trastuzumab-Fc601 bound to Fc ⁇ RIa with similar affinity with commercial-grade glycosylated trastuzumab from CHO cells and over 130 fold increased affinity compared with wild type aglycosylated trastuzumab.
- the aglycosylated tratuzumab exhibited low affinity to Fc ⁇ RIIa or Fc ⁇ RIIb (ECso ⁇ 1000- fold and 100-fold higher for GST fused Fc ⁇ RIIa and Fc ⁇ RIIb, respectively, FIG. 3 C and D) (FIG. 11 and FIG. 12), Fc ⁇ RIIIa (FIG. 13).
- Trastuzumab-Fc601 antibody exhibited only slightly higher affinity for Fc ⁇ RIIb.
- the neonatal FcRn receptor binding to the interface of CH2 domain and the CH3 domain is responsible for the endosomal recycling of IgG in plasma (Ghetie and Ward, 2000).
- trastuzumab-Fc5 did show its pH-dependent binding (high affinity binding at pH pH 6.0 and its low binding at pH 7.4) to the neonatal FcRn.
- trastuzumab-Fc601 exhibited much reduced binding affinity to FcRn at pH 6.0 (FIG. 14).
- Human FcRn has high affinity to human IgG under slightly acidic pH condition and low affinity at neutral or basic pH (Ober et al., 2004a; Ober et al., 2004b; Raghavan and Bjorkman, 1996; Rodewald, 1976).
- the FcRn binding sites are located at the interface of CH2 and CH3 domains, similar binding sites for staphylococcal protein A (SpA) (Kim et al., 1994; Shields et al., 2001).
- SpA staphylococcal protein A
- Fc601 showed improved Fc ⁇ RIa binding affinity than Fc5.
- the N-terminal sequence extension of the PCR amplified fragments using the primer STJ#473 generated the sub-library replacing 5 amino acids in the region 234L-239S with random amino acids.
- Gene assembly PCR products using DNA fragments amplified by the primers (STJ#467 and STJ#220) and DNA fragments amplified by the primers (STJ#473 and STJ#468) generated the second sub-library that randomized 5 amino acid residues for 264V-268H.
- the third sub-library randomized 297N-299T was generated using the primer pairs (STJ#473/STJ#470 and STJ#469/STJ#220) and the fourth sub-library (328L-332I) was generated using the primer pairs (STJ#473/STJ#470 and STJ#469/STJ#220) using the same PCR template plasmid, pPelBFLAG-Fc5.
- the same amount of DNA from three sub-libraries (234L-239S; 264V-268H; 328L-332I) that randomized 5 amino acid residues were mixed with and 20 3 /20 5 fold amount of DNA from the third sub-library (297N-299T) that randomized 3 amino acid residues.
- the library cells composed of 4 sub-libraries were converted to spheroplasts by the methods described in EXAMPLE 2. Over 4x10 8 spheroplasts were sorted by MoFIo flow cytometry (Dako Cytomation, Fort Collins, CO) equipped with an argon laser. Following labeling of 10 nM (3 nM for the 2 nd round, 1 nM for the 3 rd round, 0.3 nM for the 4 th round) of Fc ⁇ RIa-FITC for 1 hr at room temperature, spheroplasts were sorted with selectively gating the top 3% of the population showing the highest fluorescence due to Fc ⁇ RIa-FITC binding.
- the amino acid residue 329P was well conserved suggesting the critical role of the specific amino acid residue in the binding of Fc ⁇ RIa (FIG. 18).
- the highest fluorescent clone was Fc701 that have L328W, A330V, P331A, I332Y mutations in 328L-332I region and one additional Q295R mutation (FIG. 19-21).
- Example 8 Sequences of selected clones displaying high affinity binding to
- the engineered Fc mutants exhibiting higher affinity to Fc ⁇ RIa than Fc5 have substitution mutations in the sequence of Fc5.
- Isolated Fc mutants Fc701- Fc708 (Protein Sequence #22 ⁇ #29) have mutations in the sequence of Fc5.
- Isolated mutant showing higher affinity to Fc ⁇ RI than Fc5 are summarized in Table 6.
- trastuzumab-Fc701 IgGl was produced using the fed batch fermentation and purified using Protein A affinity chromatography followed by gel filtration chromatography as described in EXAMPLE 4. To obtain kinetic rate constants for the binding of full length trastuzumab-Fc701 to Fc ⁇ RIa, purified trastuzuma-Fc701 was immobilized on CM5 sensor chip using amine coupling method. The interaction between trastuzumab-Fc701 and Fc ⁇ RIa was analyzed using the condition described in EXAMPLE 5. Trastuzumab-Fc701 bound to Fc ⁇ RIa with similar affinity with trastuzumab Fc601 (FIG. 22).
- trastuzumab Fc701 showed higher affinity binding to FcRn at pH 6.0 than wild type aglycosylated or glycosylated trasutuzumab antibodies (FIG. 23).
- STJ#311 45 TGGCGGGGTGGTGTAGTGCTGCTGACAATAATAGGTGGCGAAATCCTCCG
- STJ#312 46 ACTACACCACCCCGCCAACCTTTGGCCAGGGTACGAAAGTGGAGATTAAA
- STJ#315 49 ACCAGACCACCGCCAGATTCCACTAATTGAACCTCCGCCATTGCCGGCTG
- STJ#326 60 GGTTTCTACGCGATGGACTACTGGGGTCAGGGTACGCTGGTCACGGTCAG
- STJ#468 65 CCAGTTGAACTTGACCTCAGGGTCTTCSNNSNNSNNSNNSNNACCACGCATGTGACCTCAGG
- Example 10 Detailed Construction of plasmids for covalently anchored full length IgG display system.
- PeIB leader peptide fused trastuzumab VL-Ck was amplified using the primers (STJ#16 and STJ#340) and the template (pSTJ4-Herceptin IgGl), digested by Xbal I HmdIII endonucleases and ligated into pBAD-NlpA-VL-Ck- ⁇ is digested with same endonucleases to generate pBADPelB-VL-Ck.
- pBADPelB-VL-Ck-NlpA-VL- Ck- ⁇ is was constructed by ligating Xbal digested PCR fragments amplified using the primers (STJ#70 and STJ#332) and the template (pBADPelB-VL-Ck) into pBADNlpA-VL-Ck- ⁇ is digested using the same endonuclease.
- Trastuzumab heavy chains were amplified using the primers (STJ#474 and STJ#67) and the template pSTJ4- ⁇ erceptin IgGl for pPelB-Herceptin(H)-FLAG, pPelB-Herceptin(H)-Fc5- FLAG, and pPelB-Herceptin(H)-Fc2a-FLAG, respectively.
- Fc2a is an aglycosylated antibody variants optimized for Fc ⁇ RII binding by two mutations (S298G/T299A) in the upper CH2 region; it has been reported that IgG containing Fc2a displays Fc ⁇ RIIa binding and effector functions comparable to those of glycosylated antibodies (Sazinsky et al, 2008).
- the plasmids pDsbA-Fc-FLAG and pDsbA-Fc2a-FLAG were constructed for the export of Fc via the DsbA signal peptide.
- PCR amplified fragments were digested with Sfil, ligated into pPelBFLAG digested with the same endonuclease to generate pPelB-Herceptin(H)- FLAG, pPelB-Herceptin(H)-Fc5-FLAG and pPelB-Herceptin(H)-Fc2a-FLAG.
- Tables 7 and 8 summarize the plasmids and primers used for Examples 10-14.
- Example 11 Preparation of spheroplasts and FACS analysis for the covalently anchored full length IgG display system to engineer IgG heavy chain
- IgG heavy chains and light chains must be well expressed.
- the heavy and light chains should be assembled well in E. coli.
- binding ligands should be accessible to the full length IgG in bacterial cells.
- the anchoring of the displayed full length IgG should be robust during library screening.
- Two plasmid co-expression plasmids were used for stable, covalent anchoring of full length IgG (FIG. 24).
- the pBADPelB-VL-Ck-NlpA-VL-Ck-His plasmid enables the expression of the NIpA leader peptide fused IgG light chain (VL- Ck) and the PeIB leader peptide fused IgG light chain (VL-Ck).
- VL- Ck the NIpA leader peptide fused IgG light chain
- VL-Ck PeIB leader peptide fused IgG light chain
- pPelB- Herceptin(H)-FLAG is a high copy number plasmid encoding the IgG heavy chain under the control of the lac promoter.
- the plasmid pBADPelB-VL-Ck-NlpA-VL-Ck- His was transformed with pPelB-Herceptin(H)-FLAG, pPelB-Herceptin(H)-Fc5- FLAG, or pPelB-Herceptin(H)-Fc2a-FLAG for wild type trastuzumab, traszumab- Fc5, or trastuzumab-Fc2a, respectively into E.
- coli cells were cultured overnight at 37 0 C with 250 rpm shaking in Terrific Broth (Becton Dickinson Diagnostic Systems DifcoTM, Sparks, MD) with 2% (wt/vol) glucose supplemented with chloramphenicol (5o ⁇ g/ml) and kanamycin (50 g/ml).
- the overnight cultured cells were diluted 1 :100 in fresh 7 ml of TB medium with chloramphenicol (5o ⁇ g/ml) and kanamycin (50 g/ml) in 125 ml Erlenmeyer flask. After incubation at 37°C for 2 h and cooling at 25°C for 20 min with 250 rpm shaking, protein expression was induced with 1 mM of isopropyl-1- thio-D-galactopyranoside (IPTG). 20 h after IPTG induction, 6 ml of the culture broth was harvested by centrifugation and washed two times in 1 ml of cold 10 mM Tris- HCl (pH 8.0).
- IPTG isopropyl-1- thio-D-galactopyranoside
- the resulting spheroplast pellets were resuspended in 1 ml of cold PBS. 300 ⁇ l of the spheroplasts were further diluted in 700 ⁇ l of PBS was labeled with 30 nM Fc ⁇ RI-FITC to analyze the binding of Fc ⁇ RIa.
- spheroplasts were incubated with 90 nM Fc ⁇ RIIa C-terminal fused to GST (Berntzen et al, 2005), washed in 1 ml of PBS, and labeled with polyclonal goat anti-GST-FITC (Abeam, Cambridge, MA) diluted 1 :200 in 1 ml of PBS. After incubation for 1 h with vigorous shaking at 25°C in dark condition, the mixture was pelleted by centrifugation at centrifuged at 12,000 x g for 1 min and resuspended in 1 ml of PBS. The fluorescently labeled spheroplasts were diluted in 2.5 ml of PBS and analyzed on BD FACSCalibur (BD Bioscience, San Jose, CA).
- Spheroplasts generated from trastuzumab-Fc5 displaying cells using the 2 plasmids covalently anchored full length IgG display system cultured at 25 0 C exhibited much higher fluorescence and improved CV upon labeling with Fc ⁇ RI-FITC relative to spheroplasts expressing a wild type trastuzumab (FIG. 25). Also, in the FACS analysis to measure the affinity of spheroplasts for Fc ⁇ RIIa-GST (FIG. 26), 2 plasmids covalently anchored full length IgG display system cultured at 25 0 C in TB showed surprisingly improved signal intensity and CV providing a selective display system for real affinity maturation of full length IgG (FIG. 27 and FIG. 28).
- An error prone PCR library of the CH2-CH3 region in anchored IgG was constructed by standard error prone PCR (Fromant et al., 1995) using the wt Fc as the template and two primers (ST J# 196 and STJ#197). The amplified PCR fragments were ligated into pPelBFLAG with Sfil restriction sites for error prone PCR library. The library Fc fragments were amplified using the primers (STJ#479 and STJ#67).
- VH-CH 1-Hinge-CH2-CH3 For trastuzumab heavy chain (VH-CH 1-Hinge-CH2-CH3) library with randomized Fc region, VH-CHl fragments were amplified using the primers (STJ#474 and STJ#480) from the template, pSTJ4-Herceptin IgG.
- Gene assembly PCR from 2 fragments, Hinge-CH2-CH3 regions and VH-CHl regions using the primer (STJ#474 and STJ#67) generated trastuzumab heavy chain (VH-CH 1-Hinge- CH2-CH3) library that randomized Fc region.
- the gene assembled PCR fragments were ligated into pPelBFLAG with Sfil restriction sites. The resulting plasmids were transformed into E.
- VH-CH 1-Hinge-CH2-CH3 For trastuzumab heavy chain (VH-CH 1-Hinge-CH2-CH3) library that randomized upper CH2 region, VHl-CHl fragments were amplified using the primers (STJ#474 and STJ#480) from the template, pSTJ4-Herceptin IgG.
- Gene assembly PCR from 2 fragments, Hinge-CH2-CH3 regions and VHl-CHl regions using the primer (STJ#474 and STJ#67) generated trastuzumab heavy chain (VH-CH 1-Hinge- CH2-CH3) library that randomized upper CH2 region.
- the gene assembled PCR fragments were ligated into pPelBFLAG with Sfil restriction sites.
- the resulting plasmids were transformed into E. coli Jude-1.
- the constructed library size was over 3 x 10 individual transformants based on the sequence of 20 library clones randomly selected.
- Plasmids Relevant characteristics Reference or source pMoPacl Cm 1 , lac promoter, tetA gene, C- (Hayhurst et al., terminal polyhistidine tag and c-myc 2003) tag
- pMoPacl2 Ap r lac promoter, tetA gene, skp (Hayhurst et al., gene, C-terminal polyhistidine tag 2003) and c-myc tag
- Example 15 Isolation and differentation of human monocyte-derived dendritic cells (mDCs) for antibody-dependent cytotoxicity assays
- Buffy coats (Gulf Coast Blood Center, Galveston, TX) was added to histopaque solution (Sigma) at 1 :1 volume, avoiding mixing of the contents.
- the blood-histopaque solution was centrifuged at 1600 RPM for 30 minutes at 23 0 C without centrifugation braking.
- the peripheral blood mononuclear cell layer was isolated following gradient centrifugation, and washed twice through centrifugation with wash buffer (PBS, 2.5% Fetal Bovine Serum (FBS), ImM ethylenediaminetetraacetic acid (EDTA)).
- PBS wash buffer
- FBS Fetal Bovine Serum
- EDTA ImM ethylenediaminetetraacetic acid
- PBMCs from a 50 ml volume of blood was resuspended in 24 ml of IMDM, and plated at 1 ml/well. Media and non-adherent cells were then aspirated and adherent cells were washed 5 times with wash buffer.
- IMDM Iscove's Modified Dulbecco's Medium
- IL-4 Interleukin-4
- GM-CSF granulocyte macrophage colony stimulating factor
- the breast cancer cell line SkBr3 that expresses high levels of Her2 was used as the target for ADCC assays.
- Cells were labeled with the isotope Na 51 CrO 4 (Perkin Elmer Life Sciences) at 100 uCi/10 6 cells for 1 h at 37 0 C. Cells were then washed twice with PBS and resuspended in Roswell Park Memorial Institute medium- 1640 with glutamax (RPMI) and added to a 96 well plate at 10 4 cells/well.
- RPMI Roswell Park Memorial Institute medium- 1640 with glutamax
- Effector cells either fully differentiated mDCs (day 7) or freshly isolated PBMCs, were resuspended in RPMI, 2% low IgG FBS (Invitrogen), lipopolysaccharide (LPS) at 250 ng/10 6 cells and added to the wells at various ratios.
- Target cells and mDCs were incubated at 37 0 C for 24h. The isotope levels present in cell media were then measured in a liquid scinitillation counter for chromium 51. Incubation of target cells with SDS was used as a positive control for maximum lysis and incubation with no effector cells was used as background lysis.
- trastuzumab-Fc5 and trastuzumab-601 show very high levels of ADCC and glycosylated trastuzumab induces very low ADCC (FIG. 31). Presumably this is because Fc-601 and Fc-5 bind only to Fc ⁇ RI and not to the inhibitor receptor Fc ⁇ RIIb which is also expressed on the surface of monocyted derived DCs.
- Herceptin displays binding to all Fc ⁇ R receptors including Fc ⁇ RIIb and binding to the latter receptor likely inhibits target cell activation and killing.
- glycosylated trastuzumab that can engage all the Fc receptors and can activate NK cells demonstrates high ADCC.
- aglycosylated trastuzumab-Fc5 and trastuzumab-601 show low ADCC (FIG. 30).
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Molecular Biology (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Biochemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biomedical Technology (AREA)
- Hematology (AREA)
- Urology & Nephrology (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- General Physics & Mathematics (AREA)
- Analytical Chemistry (AREA)
- Physics & Mathematics (AREA)
- Food Science & Technology (AREA)
- Microbiology (AREA)
- Pathology (AREA)
- Virology (AREA)
- Biotechnology (AREA)
- Cell Biology (AREA)
- Oncology (AREA)
- Tropical Medicine & Parasitology (AREA)
- Public Health (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Pharmacology & Pharmacy (AREA)
- Veterinary Medicine (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Animal Behavior & Ethology (AREA)
- Peptides Or Proteins (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
- Investigating Or Analysing Biological Materials (AREA)
- Preparation Of Compounds By Using Micro-Organisms (AREA)
Abstract
Description
Claims
Priority Applications (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU2010273763A AU2010273763B2 (en) | 2009-06-30 | 2010-06-29 | Immunoglobulin Fc polypeptides |
CN201080038758.4A CN102549016B (en) | 2009-06-30 | 2010-06-29 | Immunoglobulin FC polypeptides |
EP10800284A EP2448972A4 (en) | 2009-06-30 | 2010-06-29 | Immunoglobulin fc polypeptides |
CA2766065A CA2766065C (en) | 2009-06-30 | 2010-06-29 | Immunoglobulin fc polypeptides |
JP2012517843A JP5683581B2 (en) | 2009-06-30 | 2010-06-29 | Immunoglobulin Fc polypeptide |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US22199909P | 2009-06-30 | 2009-06-30 | |
US61/221,999 | 2009-06-30 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2011008517A2 true WO2011008517A2 (en) | 2011-01-20 |
WO2011008517A3 WO2011008517A3 (en) | 2011-07-21 |
Family
ID=43381018
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2010/040304 WO2011008517A2 (en) | 2009-06-30 | 2010-06-29 | Immunoglobulin fc polypeptides |
Country Status (7)
Country | Link |
---|---|
US (2) | US8679493B2 (en) |
EP (1) | EP2448972A4 (en) |
JP (1) | JP5683581B2 (en) |
CN (1) | CN102549016B (en) |
AU (1) | AU2010273763B2 (en) |
CA (1) | CA2766065C (en) |
WO (1) | WO2011008517A2 (en) |
Cited By (14)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2012109133A1 (en) * | 2011-02-07 | 2012-08-16 | Research Development Foundation | Engineered immunoglobulin fc polypeptides |
CN103524625A (en) * | 2013-09-24 | 2014-01-22 | 众森源生物技术(江苏)有限公司 | Method for generating novel induction antibody |
US10457737B2 (en) | 2015-02-09 | 2019-10-29 | Research Development Foundation | Engineered immunoglobulin Fc polypeptides displaying improved complement activation |
US11053308B2 (en) | 2016-08-05 | 2021-07-06 | Chugai Seiyaku Kabushiki Kaisha | Method for treating IL-8-related diseases |
WO2021136256A1 (en) * | 2019-12-31 | 2021-07-08 | 周易 | Method for obtaining high-purity heterologous antibody |
US11180548B2 (en) | 2015-02-05 | 2021-11-23 | Chugai Seiyaku Kabushiki Kaisha | Methods of neutralizing IL-8 biological activity |
US11267868B2 (en) | 2013-04-02 | 2022-03-08 | Chugai Seiyaku Kabushiki Kaisha | Fc region variant |
RU2772771C1 (en) * | 2012-09-28 | 2022-05-25 | Чугаи Сейяку Кабусики Кайся | Antigen-binding molecule for accelerating antigen loss |
US11359009B2 (en) | 2015-12-25 | 2022-06-14 | Chugai Seiyaku Kabushiki Kaisha | Anti-myostatin antibodies and methods of use |
US11454633B2 (en) | 2014-12-19 | 2022-09-27 | Chugai Seiyaku Kabushiki Kaisha | Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use |
US11718678B2 (en) | 2011-02-25 | 2023-08-08 | Chugai Seiyaku Kabushiki Kaisha | Method for altering plasma retention and immunogenicity of antigen-binding molecule |
US11820793B2 (en) | 2011-11-30 | 2023-11-21 | Chugai Seiyaku Kabushiki Kaisha | Drug containing carrier into cell for forming immune complex |
US11827699B2 (en) | 2011-09-30 | 2023-11-28 | Chugai Seiyaku Kabushiki Kaisha | Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities |
US12084513B2 (en) | 2017-11-14 | 2024-09-10 | Chugai Seiyaku Kabushiki Kaisha | Anti-C1S antibodies and methods of use |
Families Citing this family (36)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US8008453B2 (en) * | 2005-08-12 | 2011-08-30 | Amgen Inc. | Modified Fc molecules |
US8962803B2 (en) | 2008-02-29 | 2015-02-24 | AbbVie Deutschland GmbH & Co. KG | Antibodies against the RGM A protein and uses thereof |
ES2562832T3 (en) | 2009-12-08 | 2016-03-08 | Abbvie Deutschland Gmbh & Co Kg | Monoclonal antibodies against the RGM protein for use in the treatment of degeneration of the retinal nerve fiber layer |
EP2699256B1 (en) | 2011-04-21 | 2017-09-27 | The Regents of the University of Colorado, a body corporate | Compositions and methods for the treatment of neuromyelitis optica |
CN110680920A (en) * | 2011-09-30 | 2020-01-14 | 中外制药株式会社 | Antigen binding molecules that induce an immune response against a target antigen |
CN107880124B (en) | 2012-01-27 | 2021-08-13 | 艾伯维德国有限责任两合公司 | Compositions and methods for diagnosing and treating diseases associated with neural mutations |
RU2727639C2 (en) * | 2014-01-15 | 2020-07-22 | Ф.Хоффманн-Ля Рош Аг | Variants of fc-region with modified ability to bind to fcrn and with preserved ability to bind with protein a |
EP3134115A4 (en) | 2014-04-25 | 2017-10-25 | The Trustees Of The University Of Pennsylvania | Methods and compositions for treating metastatic breast cancer and other cancers in the brain |
WO2015168643A2 (en) | 2014-05-02 | 2015-11-05 | Momenta Pharmaceuticals, Inc. | Compositions and methods related to engineered fc constructs |
DK3250610T3 (en) | 2015-01-30 | 2023-10-30 | Momenta Pharmaceuticals Inc | FCRN ANTIBODIES AND METHODS OF USING THEREOF |
JP6665485B2 (en) * | 2015-10-29 | 2020-03-13 | 東ソー株式会社 | Antibody analysis method based on antibody-dependent cytotoxicity |
CA3021086C (en) * | 2016-04-15 | 2023-10-17 | Bioatla, Llc | Anti-axl antibodies, antibody fragments and their immunoconjugates and uses thereof |
CN109963869A (en) | 2016-05-23 | 2019-07-02 | 动量制药公司 | Composition relevant to modified Fc construct and method |
EP3481864A1 (en) | 2016-07-08 | 2019-05-15 | Staten Biotechnology B.V. | Anti-apoc3 antibodies and methods of use thereof |
DK3491025T3 (en) | 2016-07-29 | 2024-01-15 | Momenta Pharmaceuticals Inc | FCRN ANTIBODIES AND METHODS OF USING THEREOF |
RU2019114175A (en) | 2016-10-14 | 2020-11-16 | Ксенкор, Инк. | BISPECIFIC HETERODYMERIC FUSION PROTEINS CONTAINING IL-15 / IL-15RA FC-FUSION PROTEINS AND ANTIBODY FRAGMENTS TO PD-1 |
CN110114369A (en) * | 2016-10-17 | 2019-08-09 | 威隆股份公司 | The antibody constant region of modification |
JP7146771B2 (en) | 2017-01-06 | 2022-10-04 | モメンタ ファーマシューティカルズ インコーポレイテッド | Compositions and methods for engineered Fc constructs |
MA50958A (en) | 2017-04-21 | 2020-10-14 | Staten Biotechnology B V | ANTI-APOC3 ANTIBODIES AND THEIR METHODS OF USE |
WO2019006472A1 (en) | 2017-06-30 | 2019-01-03 | Xencor, Inc. | Targeted heterodimeric fc fusion proteins containing il-15/il-15ra and antigen binding domains |
US11059892B2 (en) | 2017-08-11 | 2021-07-13 | Research Development Foundation | Engineered antibody Fc variants for enhanced serum half life |
CN111315772A (en) | 2017-10-31 | 2020-06-19 | 斯塔顿生物技术有限公司 | anti-APOC 3 antibodies and methods of use thereof |
US10538583B2 (en) | 2017-10-31 | 2020-01-21 | Staten Biotechnology B.V. | Anti-APOC3 antibodies and compositions thereof |
US11795579B2 (en) | 2017-12-11 | 2023-10-24 | Abalone Bio, Inc. | Yeast display of proteins in the periplasmic space |
CN111712259A (en) | 2017-12-13 | 2020-09-25 | 动量制药公司 | FCRN antibodies and methods of use thereof |
CN108676094B (en) * | 2017-12-31 | 2021-07-20 | 武汉班科生物技术股份有限公司 | Human IgG antibody Fc segment CH2 structural domain mutant and preparation method and application thereof |
JP7551499B2 (en) * | 2018-03-21 | 2024-09-17 | デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド | Fc variant compositions and methods of use thereof |
JP2021520829A (en) | 2018-04-18 | 2021-08-26 | ゼンコア インコーポレイテッド | TIM-3 targeted heterodimer fusion protein containing IL-15 / IL-15RA Fc fusion protein and TIM-3 antigen binding domain |
MX2020010910A (en) | 2018-04-18 | 2021-02-09 | Xencor Inc | Pd-1 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and pd-1 antigen binding domains and uses thereof. |
CN113164590A (en) * | 2018-07-11 | 2021-07-23 | 动量制药公司 | Compositions and methods relating to engineered Fc-antigen binding domain constructs targeting CCR4 |
WO2020077297A1 (en) * | 2018-10-11 | 2020-04-16 | Valerion Therapeutics, Llc | Methods and compositions for treatment of protein aggregation-based disorders |
MA53862A (en) | 2018-10-12 | 2022-01-19 | Xencor Inc | FC FUSION PROTEINS OF IL-15/IL-15RALPHA TARGETTING PD-1 AND USES IN COMBINATION THERAPIES INVOLVING THE SAME |
US11618776B2 (en) | 2018-12-20 | 2023-04-04 | Xencor, Inc. | Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15RA and NKG2D antigen binding domains |
AU2020248645A1 (en) * | 2019-03-27 | 2021-10-28 | Tigatx, Inc. | Engineered IgA antibodies and methods of use |
TW202128757A (en) | 2019-10-11 | 2021-08-01 | 美商建南德克公司 | Pd-1 targeted il-15/il-15ralpha fc fusion proteins with improved properties |
IL302589A (en) * | 2020-11-12 | 2023-07-01 | Bioatla Inc | Methods of treating axl-expressing cancers with anti-axl antibodies, antibody fragments and their immunoconjugates |
Family Cites Families (53)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4816567A (en) | 1983-04-08 | 1989-03-28 | Genentech, Inc. | Recombinant immunoglobin preparations |
US5618920A (en) | 1985-11-01 | 1997-04-08 | Xoma Corporation | Modular assembly of antibody genes, antibodies prepared thereby and use |
US5576195A (en) | 1985-11-01 | 1996-11-19 | Xoma Corporation | Vectors with pectate lyase signal sequence |
JP3101690B2 (en) | 1987-03-18 | 2000-10-23 | エス・ビィ・2・インコーポレイテッド | Modifications of or for denatured antibodies |
US5223409A (en) | 1988-09-02 | 1993-06-29 | Protein Engineering Corp. | Directed evolution of novel binding proteins |
EP0368684B2 (en) | 1988-11-11 | 2004-09-29 | Medical Research Council | Cloning immunoglobulin variable domain sequences. |
US5578464A (en) | 1989-10-31 | 1996-11-26 | Schering Corporation | E. coli secretory strains |
US6172197B1 (en) | 1991-07-10 | 2001-01-09 | Medical Research Council | Methods for producing members of specific binding pairs |
US6916605B1 (en) | 1990-07-10 | 2005-07-12 | Medical Research Council | Methods for producing members of specific binding pairs |
US5858657A (en) | 1992-05-15 | 1999-01-12 | Medical Research Council | Methods for producing members of specific binding pairs |
US5994514A (en) | 1991-08-14 | 1999-11-30 | Genentech, Inc. | Immunoglobulin variants |
ES2313867T3 (en) | 1991-12-02 | 2009-03-16 | Medical Research Council | ANTI-AUTO ANTIBODY PRODUCTION OF ANTIBODY SEGMENT REPERTORIES EXPRESSED ON THE PAYMENT SURFACE. |
US5702892A (en) | 1995-05-09 | 1997-12-30 | The United States Of America As Represented By The Department Of Health And Human Services | Phage-display of immunoglobulin heavy chain libraries |
US6828422B1 (en) | 1995-08-18 | 2004-12-07 | Morphosys Ag | Protein/(poly)peptide libraries |
FR2739629B1 (en) | 1995-10-06 | 1997-12-26 | Systems Bio Ind | USE OF A DRY-DEPENDENT SECRETION SYSTEM FOR SECRETING PROTEIN NORMALLY SECRETED BY A DRY-INDEPENDENT SECRETION SYSTEM, BACTERIA CONTAINING THEM AND THEIR USE |
DE19628805A1 (en) * | 1996-07-17 | 1998-01-22 | Basf Ag | Process for the production of caprolactam from 6-aminocapronitrile |
GB9701425D0 (en) | 1997-01-24 | 1997-03-12 | Bioinvent Int Ab | A method for in vitro molecular evolution of protein function |
WO1999006587A2 (en) | 1997-08-01 | 1999-02-11 | Morphosys Ag | Novel method and phage for the identification of nucleic acid sequences encoding members of a multimeric (poly)peptide complex |
US6528624B1 (en) | 1998-04-02 | 2003-03-04 | Genentech, Inc. | Polypeptide variants |
US6194551B1 (en) | 1998-04-02 | 2001-02-27 | Genentech, Inc. | Polypeptide variants |
US6737056B1 (en) | 1999-01-15 | 2004-05-18 | Genentech, Inc. | Polypeptide variants with altered effector function |
KR101155191B1 (en) | 1999-01-15 | 2012-06-13 | 제넨테크, 인크. | Polypeptide Variants with Altered Effector Function |
US7183387B1 (en) | 1999-01-15 | 2007-02-27 | Genentech, Inc. | Polypeptide variants with altered effector function |
KR100922809B1 (en) | 1999-05-06 | 2009-10-21 | 웨이크 포리스트 유니버시티 | Compositions And Methods For Identifying Antigens Which Elicit An Immune Response |
EP1077263A1 (en) | 1999-07-29 | 2001-02-21 | F.Hoffmann-La Roche Ag | Process for producing natural folded and secreted proteins by co-secretion of chaperones |
KR100615887B1 (en) * | 2000-04-07 | 2006-08-25 | 삼성전자주식회사 | Wireless communication system with feedback and method thereof |
AU2002243205A1 (en) | 2000-10-10 | 2002-07-24 | Genencor International, Inc. | Enhanced secretion of a polypeptide by a microorganism |
US7094571B2 (en) | 2000-10-27 | 2006-08-22 | The Board Of Regents Of The University Of Texas System | Combinatorial protein library screening by periplasmic expression |
US6979556B2 (en) | 2000-12-14 | 2005-12-27 | Genentech, Inc. | Separate-cistron contructs for secretion of aglycosylated antibodies from prokaryotes |
AU2002238449A1 (en) | 2000-12-20 | 2002-07-01 | Altana Pharma Ag | Process for the production and purification of secreted proteins ans the production of arrays of proteins |
US7419783B2 (en) | 2001-11-05 | 2008-09-02 | Research Development Foundation | Engineering of leader peptides for the secretion of recombinant proteins in bacteria |
US20040002587A1 (en) | 2002-02-20 | 2004-01-01 | Watkins Jeffry D. | Fc region variants |
US20040132101A1 (en) | 2002-09-27 | 2004-07-08 | Xencor | Optimized Fc variants and methods for their generation |
US7317091B2 (en) | 2002-03-01 | 2008-01-08 | Xencor, Inc. | Optimized Fc variants |
US8093357B2 (en) | 2002-03-01 | 2012-01-10 | Xencor, Inc. | Optimized Fc variants and methods for their generation |
US8188231B2 (en) | 2002-09-27 | 2012-05-29 | Xencor, Inc. | Optimized FC variants |
WO2003083056A2 (en) | 2002-03-23 | 2003-10-09 | Research Development Foundation | Secretion of proteins with multiple disulfide bonds in bacteria and uses thereof |
US7611866B2 (en) | 2002-07-15 | 2009-11-03 | Board Of Regents, The University Of Texas System | Selection of bacterial inner-membrane anchor polypeptides |
EP2364996B1 (en) * | 2002-09-27 | 2016-11-09 | Xencor Inc. | Optimized FC variants and methods for their generation |
US20060235208A1 (en) | 2002-09-27 | 2006-10-19 | Xencor, Inc. | Fc variants with optimized properties |
US7960512B2 (en) | 2003-01-09 | 2011-06-14 | Macrogenics, Inc. | Identification and engineering of antibodies with variant Fc regions and methods of using same |
ES2897506T3 (en) | 2003-01-09 | 2022-03-01 | Macrogenics Inc | Identification and modification of antibodies with variant Fc regions and methods of using them |
US8084582B2 (en) | 2003-03-03 | 2011-12-27 | Xencor, Inc. | Optimized anti-CD20 monoclonal antibodies having Fc variants |
WO2005077981A2 (en) | 2003-12-22 | 2005-08-25 | Xencor, Inc. | Fc POLYPEPTIDES WITH NOVEL Fc LIGAND BINDING SITES |
US7276585B2 (en) | 2004-03-24 | 2007-10-02 | Xencor, Inc. | Immunoglobulin variants outside the Fc region |
US7229792B2 (en) | 2004-05-03 | 2007-06-12 | Vinod Pandiripally | Method of producing recombinant proteins |
CA2587766A1 (en) | 2004-11-10 | 2007-03-01 | Macrogenics, Inc. | Engineering fc antibody regions to confer effector function |
AU2005304624B2 (en) | 2004-11-12 | 2010-10-07 | Xencor, Inc. | Fc variants with altered binding to FcRn |
RU2412200C2 (en) * | 2004-11-12 | 2011-02-20 | Ксенкор, Инк. | Fc-VERSIONS WITH CHANGED BINDING WITH FcRn |
US7875278B2 (en) * | 2005-02-18 | 2011-01-25 | Medarex, Inc. | Monoclonal antibodies against prostate specific membrane antigen (PSMA) lacking in fucosyl residues |
GB0503927D0 (en) * | 2005-02-25 | 2005-04-06 | Nokia Corp | Location services in a communication system |
WO2007021841A2 (en) | 2005-08-10 | 2007-02-22 | Macrogenics, Inc. | Identification and engineering of antibodies with variant fc regions and methods of using same |
US8629245B2 (en) * | 2007-05-01 | 2014-01-14 | Research Development Foundation | Immunoglobulin Fc libraries |
-
2010
- 2010-06-29 WO PCT/US2010/040304 patent/WO2011008517A2/en active Application Filing
- 2010-06-29 CN CN201080038758.4A patent/CN102549016B/en not_active Expired - Fee Related
- 2010-06-29 CA CA2766065A patent/CA2766065C/en active Active
- 2010-06-29 JP JP2012517843A patent/JP5683581B2/en not_active Expired - Fee Related
- 2010-06-29 AU AU2010273763A patent/AU2010273763B2/en not_active Ceased
- 2010-06-29 EP EP10800284A patent/EP2448972A4/en not_active Withdrawn
- 2010-06-30 US US12/827,386 patent/US8679493B2/en active Active
-
2014
- 2014-02-14 US US14/181,594 patent/US20140235482A1/en not_active Abandoned
Non-Patent Citations (1)
Title |
---|
See references of EP2448972A4 * |
Cited By (20)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9771426B2 (en) | 2011-02-07 | 2017-09-26 | Research Development Foundation | Engineered immunoglobulin FC polypeptides |
WO2012109133A1 (en) * | 2011-02-07 | 2012-08-16 | Research Development Foundation | Engineered immunoglobulin fc polypeptides |
JP2014510519A (en) * | 2011-02-07 | 2014-05-01 | リサーチ ディベロップメント ファウンデーション | Engineered immunoglobulin Fc polypeptides |
US8952132B2 (en) | 2011-02-07 | 2015-02-10 | Research Development Foundation | Engineered immunoglobulin FC polypeptides |
EP2673299B1 (en) * | 2011-02-07 | 2017-05-10 | Research Development Foundation | Engineered immunoglobulin fc polypeptides |
US11718678B2 (en) | 2011-02-25 | 2023-08-08 | Chugai Seiyaku Kabushiki Kaisha | Method for altering plasma retention and immunogenicity of antigen-binding molecule |
US11827699B2 (en) | 2011-09-30 | 2023-11-28 | Chugai Seiyaku Kabushiki Kaisha | Methods for producing antibodies promoting disappearance of antigens having plurality of biological activities |
US11820793B2 (en) | 2011-11-30 | 2023-11-21 | Chugai Seiyaku Kabushiki Kaisha | Drug containing carrier into cell for forming immune complex |
RU2772771C1 (en) * | 2012-09-28 | 2022-05-25 | Чугаи Сейяку Кабусики Кайся | Antigen-binding molecule for accelerating antigen loss |
US11267868B2 (en) | 2013-04-02 | 2022-03-08 | Chugai Seiyaku Kabushiki Kaisha | Fc region variant |
CN103524625A (en) * | 2013-09-24 | 2014-01-22 | 众森源生物技术(江苏)有限公司 | Method for generating novel induction antibody |
US11454633B2 (en) | 2014-12-19 | 2022-09-27 | Chugai Seiyaku Kabushiki Kaisha | Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use |
US11180548B2 (en) | 2015-02-05 | 2021-11-23 | Chugai Seiyaku Kabushiki Kaisha | Methods of neutralizing IL-8 biological activity |
US11332538B2 (en) | 2015-02-09 | 2022-05-17 | Research Development Foundation | Engineered immunoglobulin Fc polypeptides displaying improved complement activation |
US10457737B2 (en) | 2015-02-09 | 2019-10-29 | Research Development Foundation | Engineered immunoglobulin Fc polypeptides displaying improved complement activation |
US11359009B2 (en) | 2015-12-25 | 2022-06-14 | Chugai Seiyaku Kabushiki Kaisha | Anti-myostatin antibodies and methods of use |
US11780912B2 (en) | 2016-08-05 | 2023-10-10 | Chugai Seiyaku Kabushiki Kaisha | Composition for prophylaxis or treatment of IL-8 related diseases |
US11053308B2 (en) | 2016-08-05 | 2021-07-06 | Chugai Seiyaku Kabushiki Kaisha | Method for treating IL-8-related diseases |
US12084513B2 (en) | 2017-11-14 | 2024-09-10 | Chugai Seiyaku Kabushiki Kaisha | Anti-C1S antibodies and methods of use |
WO2021136256A1 (en) * | 2019-12-31 | 2021-07-08 | 周易 | Method for obtaining high-purity heterologous antibody |
Also Published As
Publication number | Publication date |
---|---|
CA2766065C (en) | 2020-07-21 |
US20100330076A1 (en) | 2010-12-30 |
EP2448972A2 (en) | 2012-05-09 |
CA2766065A1 (en) | 2011-01-20 |
CN102549016B (en) | 2015-05-06 |
EP2448972A4 (en) | 2012-11-28 |
JP2012531897A (en) | 2012-12-13 |
WO2011008517A3 (en) | 2011-07-21 |
AU2010273763B2 (en) | 2013-11-21 |
US20140235482A1 (en) | 2014-08-21 |
US8679493B2 (en) | 2014-03-25 |
JP5683581B2 (en) | 2015-03-11 |
AU2010273763A1 (en) | 2012-01-19 |
CN102549016A (en) | 2012-07-04 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CA2766065C (en) | Immunoglobulin fc polypeptides | |
US9771426B2 (en) | Engineered immunoglobulin FC polypeptides | |
US20220242934A1 (en) | Antigen-binding molecule containing modified fc region | |
US20220324966A1 (en) | Antigen-binding molecule for promoting clearance from plasma of antigen comprising sugar chain receptor-binding domain | |
US11332538B2 (en) | Engineered immunoglobulin Fc polypeptides displaying improved complement activation | |
AU2017364817B2 (en) | Antigen-binding domain, and polypeptide including conveying section | |
US10526408B2 (en) | Engineered antibody FC variants | |
US20220315909A1 (en) | Protease substrate, and polypeptide including protease cleavage sequence | |
AU2018314257A1 (en) | Engineered antibody Fc variants for enhanced serum half life | |
US20240010742A1 (en) | Engineered fcriib selective igg1 fc variants and uses thereof | |
GEORGIOU et al. | Patent 2766065 Summary | |
TW202434628A (en) | A polypeptide complex whose antigen-binding activity varies depending on the concentration of a plasma protein | |
TW202323284A (en) | Immunosilencing fc variants | |
CN118475619A (en) | Antibody containing IgG type Fc region variant and application thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
WWE | Wipo information: entry into national phase |
Ref document number: 201080038758.4 Country of ref document: CN |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 10800284 Country of ref document: EP Kind code of ref document: A2 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2010273763 Country of ref document: AU |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2012517843 Country of ref document: JP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2766065 Country of ref document: CA |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
REEP | Request for entry into the european phase |
Ref document number: 2010800284 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2010800284 Country of ref document: EP |
|
ENP | Entry into the national phase |
Ref document number: 2010273763 Country of ref document: AU Date of ref document: 20100629 Kind code of ref document: A |