WO2017222556A2 - Fragments variables insérables d'anticorps et domaines a1-a2 modifiés de ligands nkg2d - Google Patents

Fragments variables insérables d'anticorps et domaines a1-a2 modifiés de ligands nkg2d Download PDF

Info

Publication number
WO2017222556A2
WO2017222556A2 PCT/US2016/039281 US2016039281W WO2017222556A2 WO 2017222556 A2 WO2017222556 A2 WO 2017222556A2 US 2016039281 W US2016039281 W US 2016039281W WO 2017222556 A2 WO2017222556 A2 WO 2017222556A2
Authority
WO
WIPO (PCT)
Prior art keywords
domain
molecule
modified
nkg2d
ifv
Prior art date
Application number
PCT/US2016/039281
Other languages
English (en)
Other versions
WO2017222556A3 (fr
Inventor
Kyle LANDGRAF
Daniel P. Steiger
Steven R. Williams
David W. Martin
Dana Gebhart
Tarah Baron
Original Assignee
Avidbiotics Corp.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Avidbiotics Corp. filed Critical Avidbiotics Corp.
Priority to JP2019519951A priority Critical patent/JP2019523016A/ja
Priority to EP16734162.7A priority patent/EP3475300A2/fr
Priority to PCT/US2016/039281 priority patent/WO2017222556A2/fr
Priority to KR1020187037547A priority patent/KR20190019091A/ko
Priority to AU2016410294A priority patent/AU2016410294A1/en
Priority to CA3027908A priority patent/CA3027908A1/fr
Publication of WO2017222556A2 publication Critical patent/WO2017222556A2/fr
Publication of WO2017222556A3 publication Critical patent/WO2017222556A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70539MHC-molecules, e.g. HLA-molecules
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif

Definitions

  • This application relates generally to the production of polypeptides having specific antigen-binding properties of Fv domains, for example, insertable variable fragments of antibodies, and modified al-a2 domains of NKG2D ligands.
  • Each tip of the two arms of the "Y" of an antibody contains an antigen binding site, or a paratope, (a structure analogous to a lock) that is specific for one particular epitope (similarly analogous to a key) of an antigen, allowing these two structures to bind together with precision.
  • an antibody can tag a microbe or an infected cell for attack by other parts of the immune system or can neutralize its target directly, for example, by blocking a part of a microbe that is essential for its invasion and survival.
  • the production of antibodies is the main function of the humoral, or "adaptive", immune system. Antibodies are secreted by plasma cells.
  • Antibodies in nature can occur in two physical forms, a soluble form that is secreted from the cell, and a membrane-bound form that is attached to the surface of a B cell via the "stem" of the Y.
  • Antibodies are glycoproteins belonging to the immunoglobulin superfamily and are typically made of basic structural units— each with two large heavy chains and two small light chains. There are several different types of antibody heavy chains, and several different kinds of antibodies, which are grouped into different isotypes based on which heavy chain they possess. Five different antibody isotypes are known in mammals (Market E, Papavasiliou FN (October 2003). "V(D)J recombination and the evolution of the adaptive immune system”. PLoS Biol. 1 (1): E16.
  • the natural "Y"-shaped Ig molecule consists of four polypeptide chains; two identical heavy chains and two identical light chains connected by disulfide bonds, Figure 1. Each heavy chain has two major regions, the constant region (CH) and the variable region (VH). The constant region is essentially identical in all antibodies of the same isotype, but differs in antibodies of different isotypes. A light chain also has two successive domains: a smaller constant region (CL) and the variable region (VL) (Woof J, Burton D (2004). "Human antibody- Fc receptor interactions illuminated by crystal structures.” Nat Rev Immunol 4 (2): 89-99.
  • Y contains the sites that can bind to antigens and, therefore, recognize specific foreign objects.
  • This region of the antibody is called the Fv (fragment, variable) region. It is composed of one variable domain from the heavy chain (VH) and one variable region from the light chain (VL) of the antibody(Hochman J, Inbar D, Givol D (1973).
  • An active antibody fragment (Fv) composed of the variable portions of heavy and light chains.
  • the paratope is shaped at one end of the Fv and is the region for binding to antigens.
  • CDRs complementarity determining regions
  • Useful polypeptides that possess specific antigen binding function can be derived from the CDRs of the variable regions of antibodies. These two antibody variable domains, one of the light chain(VL) and one from the heavy chain (VH), each with 3 CDRs can be fused in tandem, in either order, using a single, short linker peptide of 10 to about 25 amino acids to create a linear single-chain variable fragment (scFv) polypeptide comprising one each of heavy and light chain variable domains, Figure 3 (Bird, R. E., Hardman, K. D., Jacobson, J. W., Johnson, S., Kaufman, B. M., Lee, S. M., Lee, T., Pope, S.
  • the linker is usually rich in glycine for flexibility, as well as serine, threonine, or charged amino acids for solubility, and can either connect the N-terminus of the VH with the C- terminus of the VL, or vice versa. This protein retains the specificity of the original
  • the present disclosure relates to modified al-a2 domains of NKG2D ligands attached to polypeptides, in some embodiments antibodies or fragments of antibodies.
  • the present disclosure relates to antigen-binding peptides derived from light and heavy chain antibody variable domains, which contain two linker regions and a split variable domain.
  • the patent or application file contains at least one drawing executed in color.
  • Figure 1 A cartoon of a typical mammalian antibody showing its Y-shaped structure and structural components.
  • Figure 2 A cartoon of the structure of an Fv region of a natural mammalian antibody showing the 3 labeled (Complementarity Determining Regions) CDRs of the VH and the 3 unlabeled loops of the VL CDRs, which form the paratope or antigen binding site.
  • Figure 3 A cartoon of the two possible structures of a single-chain variable fragment (scFv), with the antigen binding sites including the N-termini on the left and the C- termini on the right.
  • the single linker region, or linker peptide, in each scFv is shown as an arrow.
  • FIG. 1 Polyvalent single-chain variable fragments (scFv's). Structure of divalent (top) and trivalent (bottom) scFvs, tandem (left) and di-/trimerization format (right). Note that each has 2 or more spatially distant free termini.
  • FIGS 5A and 5B Diagram of an insertable variable fragment, iFv.
  • Figure 5A shows the structure of variable light (VL) and variable heavy (VH) domains from FGFR3 -binding antibody showing the domain topology of the iFv format.
  • Grey arrows represent the 2 linker regions (LR), one and only one of which is used traditionally to connect the termini of VL and VH to create an scFv.
  • the LR with a dotted border connected the C-terminus of VL to the N-terminus of VH (visible behind the molecule).
  • the LR with a solid border connected the C-terminus of VH to the N-terminus of VL.
  • Segments of the split VL domain are labeled Nt and Ct as described in text.
  • the VL has been divided into an N-terminal segment (VLN) and a C-terminal segment (VLC).
  • the 6 CDRs of VL and VH are represented as the loops at the top of the figure.
  • Figure 5B shows the scheme of the domain layout for inserting an iFv into loop 1 (LI) of MICA-a3 with or without a spacer region (SR).
  • An iFv could also be similarly inserted into loop 2 (L2) and/or loop 3 (L3).
  • FIG. 6 Titration curves for the modified sMICA molecules binding to FGFR3 coated wells. Bound sMICA was detected by ELISA using NKG2D-Fc to confirm the bispecific activity. Both versions of the inserted variable fragments (MICA-oc3-iFv. l and MICA-a3-iFv.2) bound FGFR3 comparably to the C-terminal fusion of an scFv (MICA-scFv).
  • FIGS 7A and 7B Thermal stability of MICA-a3-iFv.2.
  • the MICA-a3- iFv exhibited strong binding to FGFR3 after exposure to 80°C, whereas MICA-scFv lost significant activity after exposure to 70°C.
  • FIG. 8 NK-mediated target cell lysis assays.
  • NKL effector cells were co- incubated with calcein-loaded, FGFR3 -expressing P815 target cells at a effector:target ratio of 15: 1.
  • Increasing concentrations of a negative control MICA (sMIC A) had no effect on target cell lysis, whereas the indicated FGFR3-binding MICA-a3-iFv variants stimulated target cell lysis.
  • both MICA- 3-iFv variants directed greater target cell lysis.
  • FIGS 9A and 9B Target binding and cell lysis activity of a CD20-specific sMICA variant.
  • MICA-a3-iFv.3 exhibited titratable binding to CD20-coated wells in an ELISA ( Figure 9A), and also enhanced NK-mediated cell lysis of CD20-expressing Ramos cells ( Figure 9B).
  • NKL effector cells were co-incubated with calcein- loaded CD20-expressing Ramos cells at a effector: target ratio of 15: 1, and increasing
  • FIG. 11 NK-mediated target cell lysis assays.
  • NKL effector cells were co- incubated with calcein-loaded, FGFR3 -expressing P815 target cells at a effector:target ratio of 15: 1.
  • Increasing concentrations of a negative control MICA (sMICA) had no effect on target cell lysis, whereas each indicated NKG2DL-a3-iFv.2 protein stimulated target cell lysis.
  • Figures 12A and 12B Structure-directed mutagenesis of the ⁇ 1- ⁇ 2 domain of
  • FIG 12A shows the structure of the al -a2 domain of MICA (PDB IHYR) with the NKG2D-binding surface mapped to 57 residues colored dark grey.
  • Figure 12B shows six positions that were identified as key sites for NKG2D affinity mutations. The wild-type amino acid residues are labeled and their side chains shown in dark grey spheres.
  • Figures 13A and 13B NKG2D-Fc competition ELISAs to affinity rank al-a2 variants.
  • Figure 13A shows titration data for a panel of al-a2 affinity variants (15-18), wild-type (WT), or WED soluble MICA proteins inhibiting human NKG2D-Fc binding to plate-coated MICA.
  • Figure 13B shows the same set of proteins in Figure 13 A titrated against mouse NKG2D- Fc. In both assays variants 15, 16, 17, and 18 display IC 5 o values significantly less than both WT and WED proteins. The equilibrium IC 5 0 values are shown in Table 3. [0023] Figure 14.
  • Figure 15 NK-mediated target cell killing assay for the al-a2 variants targeting
  • FGFR3 -expressing target cells NKL effector cells were co-incubated with calcein-loaded, FGFR3 -expressing P815 target cells at a effector: target ratio of 15: 1.
  • Increasing concentrations of a negative control MICA (sMICA) had no effect on target cell lysis, whereas the indicated al- a2 variants stimulated target cell lysis.
  • Relative to WT and WED-MICA, variants 16, 17, and 18 exhibited significantly increased killing at low concentrations.
  • Figure 16 Analysis of the association and dissociation kinetics for al-a2 variants 20, 25, and 48 binding to NKG2D, as measured by biolayer interferometry on an Octet instrument. The association and dissociation phases were fit using a single exponential 1 : 1 binding equation, and on- and off-rate constants derived from the fits are shown in Table 5,
  • Figure 17 NK-mediated target cell killing, calcein-based assay for al - 2 variants 16, 25 and WED targeting FGFR3 -expressing P815 target cells.
  • FIG. Protein sequence alignment of al-a2 domains from MICA and
  • ULBPs (SEQ ID NOs: 77-83). Amino acids highlighted in grey were selected for NNK mutagenesis in ULBP2 (60 amino acids) and ULBP3 (36 amino acids). Residues highlighted in black were identified as key positions for selected and identified as mutations that modulate binding affinity to NKG2D (Tables 6 and 7). [0028] Figures 19A and 19B. Phage ELISA titrations of ULBP variants binding to
  • Figure 19A depicts experiments in which ULBP2 variants displayed on phage were titrated against NKG2D and relative binding affinities were measured relative to native ULBP2 (WT, black circles).
  • Figure 19B depicts experiments in which ULBP3 variants displayed on phage were titrated against NKG2D and relative binding affinities were measured relative to native ULBP3 (WT, black circles).
  • Figures 20A-D Fusions of native (WT), modified variant WED, 25 or 48 ⁇ l -o2 domains to heavy chain ( Figure 20A) or light chain ( Figure 20B) of an FGFR3 -specific antibody affected NK-dependent target cell killing. Fusions of variants 25 and 48 to either heavy chain ( Figure 20C) or light chain ( Figure 20D) significantly enhanced the extent of killing and the potency of killing compared to the WED variant and to the native (WT) fusions.
  • Figures 21A-C Fusions of variant 25 al-a2 domain to the heavy chains or light chains of antibodies targeting human EGFR (Figure 21 A), HER2 ( Figure 2 IB), or PDLl ( Figure 21 C) each enhanced NKL cell-mediated target cell killing The poor or absent killing by the respective parent antibodies, cetuximab (Figure 21 A), trastuzumab (Figure 2 IB), and anti-PDLl ( Figure 21 C) are shown.
  • FIGS 22A and 22B Trastuzumab-based fusions of variant 25 ⁇ 1 - ⁇ 2 domain arm NK cells in vivo.
  • Parent trastuzumab, trastuzumab HC_25 fusion, and trastuzumab LC_25 fusion were conjugated with Alexa Flour.
  • Groups of three C57BL/6 mice were injected with a single dose of 100 ⁇ g of parent, HC fusion or LC fusion; and blood was drawn from each animal at indicated times for plasma PK ELISAs ( Figure 22A) and flow cytometric analyses of the fluorescently labeled molecules bound to peripheral NK cells ( Figure 22B).
  • Figures 23A-C Anti-drug antibodies raised in the same animals described in
  • the control (Ctrl) plasma was from a mouse not administered any antibody-containing agent.
  • Figures 24A and 24B Antibodies generated in animals administered variant 25 al-a2 domain fusions to trastuzumab-HC and -LC, as described in Example 7 and Figures 21- 22, bound to both the parent antibody (Figure 24A) and to the al-a2 domain ( Figure 24B).
  • Figure 25 Anti -tumor activity of an anti-PDLl fusion to variant 25. Syngeneic
  • MC38 tumors were implanted subcutaneously in C57BL/6 mice, and tumors grew to an average of 100 mm 3 before the initiation of treatment.
  • four cohorts of 10 mice per group were treated parenterally with vehicle, anti-CTLA4 (100 ug i.p.), parent anti- PDLl (300 ug i.v.), or anti-PDLl HC_25 fusion (300 ug i.v.) on days 1, 4, and 7.
  • Tumor volumes (cubic mm) were measured in each animal at the indicated times.
  • FIGS 26 A and 26B Fusions of ULBP2 and ULBP3 al-a2 domain variants to the heavy chain of a HER2-specific antibody showed enhanced NKG2D binding affinity.
  • Modified ULBP2 al -a2 domain variants R80W (SEQ ID NO: 84) and V151D (SEQ ID NO: 85) displayed enhanced NKG2D binding relative to the natural ULBP2 (SEQ ID NO: 16) fusion (WT) ( Figure 26A).
  • FIGS 27A and 27B Fusions of ULBP2 and ULBP3 al-a2 domain variants to the heavy chain of a HER2-specific antibody showed specific lysis of SKBR3 target cells by NKL cells.
  • Modified ULBP2 l-a2 domain variants R80W (SEQ ID NO: 84) and VI 5 ID (SEQ ID NO: 85) displayed enhanced target cell killing relative to the natural ULBP2 (SEQ ID.: 16) fusion (WT) (Figure 27A).
  • Modified ULBP3 variant R162G SEQ ID NO: 86
  • the present invention relates to insertable variable fragment (iFv) peptides. Because the C-terminus and N-terminus of scFv molecules including polyvalent scFv structures are far apart spatially, scFv structures cannot be inserted into a loop region embedded within a protein fold of a parent or recipient protein without disrupting or destabilizing its fold(s) and/or without disrupting the Fv framework required to properly position the CDRs or hypervariable regions to retain their antigen-binding properties.
  • iFv insertable variable fragment
  • variable fragment of an antibody containing up to 6 CDRs into one or more loop regions of a nascent parent protein molecule without disrupting structural folds of the variable fragment or of the parent protein
  • new structures contained two linker regions, rather than the traditional single linker of scFv structures, plus a split variable domain.
  • VL variable light
  • VH variable heavy
  • That circular peptide structure containing all 6 CDRs of the Fv can then conceptually be split at one of several possible novel sites to create an insertable Fv (iFv).
  • the non-natural split site can be created within either the light or the heavy chain variable domain at or near the apex or turn of a loop to create new, unique N- and C-termini spatially positioned proximal to each other, preferably within 0.5 to 1.5 2016/039281 tun, so as to be insertable into loops of other (parent or recipient) proteins or polypeptides without disrupting the structure, stability, or desirable function.
  • This new class of peptides is called an insertable variable fragment (iFv).
  • the binding or targeting specificity conveyed by an iFv to a recipient molecule can be changed by inserting into the recipient another or different iFV based on a different antibody or scFv or by replacing 1 or more of the CDRs of an existing insertable iFv.
  • iFv polypeptides exhibiting specific antigen-binding properties of Fv domains into other proteins and thereby imparting novel binding properties will have multiple utilities. Such uses include but are not limited to enabling the parent protein to bind the specific antigen, target the antigen, detect the presence of antigen, remove the antigen, contact or draw near the antigen, to deliver a payload to the antigen or antigen-expressing cell, recruit the antigen, and image the presence of the antigen.
  • a payload could be conjugated directly to one or both the amino-terminus and carboxy-terminus of an iFv or indirectly to an iFv via a parent protein or peptide.
  • payloads include but are not limited to a chromophore, a fluorophore, a pharmacophore, an atom, a heavy or radioactive isotope, an imaging agent, a chemotherapeutic agent, or a toxin.
  • a payloaded iFv can be used to locate or identify the presence of a target molecule to which the iFv specifically binds and as such can serve as in vitro or in vivo imaging agents or diagnostic agents that are small and stable.
  • a chemotherapeutic agent or toxic molecule can be conjugated in order to create an iFv-drug conjugate, for example, as treatment for a malignancy or infection.
  • a single payload may be conjugated to both the amino-terminus and the carboxy-terminus of an iFv peptide so as to span or connect the two termini; such spanning may further stabilize the iFv by blocking the termini from exopeptidase degradation or protecting the iFv from denaturation or unfolding.
  • parent or recipient proteins or polypeptides that are candidates for insertions of iFv peptides include but are not limited to antibodies, proteins comprised of Ig folds or Ig domains, globulins, albumens, fibronectins and fibronectin domains, integrins, fluorescent proteins, enzymes, outer membrane proteins, receptor proteins, T-cell receptors, chimeric antigen receptors, viral antigens, virus capsids, viral ligands for cell receptors, high molecular weight bacteriocins, histones, hormones, knottins, cyclic peptides or polypeptides, major
  • MHC histocompatibility family proteins
  • MIC proteins lectins
  • ligands for lectins It is also possible to insert iFv structures into non-protein recipient molecules such a polysaccharides, dendrimers, polyglycols, peptidoglycans, antibiotics, and polyketides.
  • NK cells and certain T-cells of the immunity system have important roles in humans and other mammals as first-line, innate defense against neoplastic and virus-infected cells (Cerwenka, A., and L.L. Lanier. 2001. NK cells, viruses and cancer. Nat. Rev. Immunol. 1 :41-49).
  • NK cells and certain T-cells exhibit on their surfaces NKG2D, a prominent, homodimeric, surface immunoreceptor responsible for recognizing a target cell and activating the innate defense against the pathologic cell (Lanier, LL, 1998. NK cell receptors. Ann. Rev. Immunol.
  • the human NKG2D molecule possesses a C-type lectin-like extracellular domain that binds to its cognate ligands, the 84% sequence identical or homologous, monomeric MICA and MICB, polymorphic analogs of the Major Histocompatibility Complex (MHC) Class I chain-related glycoproteins (MIC) (Weis et al. 1998. The C-type lectin superfamily of the immune system. Immunol. Rev. 163 : 19-34; Bahram et al. 1994. A second lineage of mammalian MHC class I genes. PNAS 91 :6259-6263; Bahram et al. 1996a. Nucleotide sequence of the human MHC class I MICA gene.
  • Non-pathologic expression of MICA and MICB is restricted to intestinal epithelium, keratinocytes, endothelial cells and monocytes, but aberrant surface expression of these MIC proteins occurs in response to many types of cellular stress such as proliferation, oxidation and heat shock and marks the cell as pathologic (Groh et al. 1996. Cell stress-regulated human MHC class I gene expressed in GI epithelium. PNAS 93: 12445-12450; Groh et al. 1998.
  • NKG2D ligands such as the polymorphic MICA and MICB
  • the differential regulation of NKG2D ligands is important to provide the immunity system with a means to identify and respond to a broad range of emergency cues while still protecting healthy cells from unwanted attack (Stephens HA, (2001) MICA and MICB genes: can the enigma of their polymorphism be resolved? Trends Immunol. 22: 378-85; Spies, T. 2008. Regulation of NKG2D ligands: a purposeful but delicate affair. Nature Immunol. 9: 1013-1015).
  • Viral infection is a common inducer of MIC protein expression and identifies the viral-infected cell for NK or T-cell attack (Groh et al. 1998; Groh et al. 2001. Co-stimulation of CD8+ ⁇ -cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat.
  • the 3-dimensional structures of the human MICA and MICB a3 domains are nearly identical (root-mean square distance ⁇ 1 A on 94 C-aa's) and functionally interchangeable (Holmes et al. 2001. Structural Studies of Allelic Diversity of the MHC Class I Homolog MICB, a Stress-Inducible Ligand for the Activating Immunoreceptor NKG2D. J Immunol. 169: 1395-1400).
  • soluble MIC protein refers to a MIC protein containing the al , a2, and a3 domains of the MIC protein but without the transmembrane or intracellular domains.
  • the al -a2 platform domain of a soluble MIC protein is tethered to the a3 domain and is diffusible in the intercellular or intravascular space of the mammal.
  • the ⁇ xl-a2 platform domains of the non-natural MIC proteins of the invention are at least 80% identical or homologous to a native or natural al-a2 domain of a human MICA or MICB protein and bind NKG2D.
  • the al-a2 platform domain is 85% identical to a native or natural ⁇ 1- ⁇ 2 platform domain of a human MICA or MICB protein and binds NKG2D.
  • the al-a2 platform domain is 90%, 95%, 96%, 97%, 98%, or 99% identical to a native or natural al-a2 platform domain of a human MICA or MICB protein and binds NKG2D.
  • a heterologous peptide tag may be fused to the N-terminus or C-terminus of an al-a2 domain or a soluble MIC protein to aid in the purification of the soluble MIC protein.
  • Tag sequences include peptides such as a poly-histidine, myc-peptide or a FLAG tag. Such tags may be removed after isolation of the MIC molecule by methods known to one skilled in the art.
  • peptide As used herein "peptide”, “polypeptide”, and “protein” are used interchangeably; and a “heterologous molecule”, “heterologous peptide”, “heterologous sequence” or
  • heterologous atom is a molecule, peptide, nucleic acid or amino acid sequence, or atom, respectively, that is not naturally or normally found in physical conjunction with the subject molecule.
  • Example 1 (iFv). As specific examples, we synthesized a 1 126 bp and a 1 144 bp
  • DNA fragment (SEQ ID NO: 1 and 2, respectively) encoding in the following order: the a3 domain of human MICA (as a parent peptide) amino acid 182 to amino acid 194 (the beginning of loop 1 of the a3 domain), no spacer or a GGS amino acid spacer region (SR), an iFv peptide based on the structure of a Fibroblast Growth Factor Receptor 3 (FGFR3)-binding antibody (MAbR3;Qing, J., Du, X., Chen, Y., Chan, P., Li, H., Wu, P., Marsters, S., Stawicki, S., Tien, J., Totpal, K., Ross, S., Stinson, S., Dornan, D., French, D., Wang, Q.
  • FGFR3 Fibroblast Growth Factor Receptor 3
  • each synthetic, double stranded DNA polynucleotide then encoded a polypeptide that contained 6 CDRs in the form of an iFv inserted into loop 1 of the a3 domain of MICA.
  • This iFv peptide itself (SEQ ID NO: 3), encoded by SEQ ID NO: 4, contained two identical, typical linker regions (LR) corresponding to residues GGSSRSSSSGGGGSGGGG (SEQ ID NO: 5) (Andris-Widhopf, J., Steinberger, P., Fuller, R., Rader, C, and Barbas, C. F., 3rd. (201 1) Generation of human Fab antibody libraries: PCR amplification and assembly of light- and heavy-chain coding sequences, Cold Spring Harbor protocols 2011).
  • a baculoviral expression vector to accommodate the DNA sequences (SEQ ID NOs: 1 and 2) encoding the a3-iFv. l (SEQ ID NO: 6) and ⁇ x3-iFv.2 (SEQ ID NO: 7), respectively.
  • the DNA fragments were amplified by PCR, digested using Ncol and EcoRI restriction enzymes, and subcloned into the baculoviral expression vector, SW403, replacing the wild-type a3 domain.
  • SW403 is a baculoviral expression vector derived from pVL1393 (Invitrogen, Inc.) into which wild-type sMICA (residues 1-276) had previously been cloned using 5' BamHI and 3' EcoRI sites.
  • the new expression vector was co-transfected with baculoviral DNA into SF9 insect cells, and baculovirus was grown for two amplification cycles and used to express the His-tagged MICA-a3-iFv proteins in T.ni insect cells according to manufacturer's protocol (Invitrogen). The expression was carried out in a 100 mL volume for three days and the growth medium was harvested for purification of the secreted soluble protein using Ni-affmity chromatography. Monomeric MICA- 3-iFv was purified to >90% purity with the expected molecular weight of 60.9 kDa as determined by SDS-PAGE. Functional characterization was carried out using binding ELISAs and in vitro target cell killing assays
  • MICA-a3-iFv proteins were tested in a FGFR3-binding ELISA to confirm simultaneous binding to the FGFR3 target and the NKG2D receptor.
  • FGFR3 in phosphate buffered saline (PBS) was coated onto Maxisorp plates at 2 ug/ml concentration.
  • PBS phosphate buffered saline
  • Each MICA protein was titrated, allowed to bind FGFR3 for 1 hour, and washed to remove unbound sMICA protein.
  • Bound MICA-a3-iFv protein was detected using NKG2D-Fc and anti- Fc-HRP conjugate.
  • Figure 6 shows that the binding of both MICA-oo3-iFv.
  • MICA-a3-iFv The ability of MICA-a3-iFv to redirect NK cell-mediated lysis of FGFR3- expressing target cells was demonstrated in vitro in a calcein-release assay.
  • the Natural Killer (NK) cell line, NKL was co-cultured with calcein-loaded P815 target cells ectopically expressing FGFR3.
  • the results in Figure 8 showed that the two MICA-a3-iFv molecules induced significantly greater NK-mediated lysis compared to the traditional MICA-scFv fusion, while the non-targeted soluble MICA control had no killing activity.
  • Figures 9A and 9B show that MICA-a3-iFv.3 was able to specifically bind wells coated with CD20 in a plate-based ELIS A as described above and also induced NK-mediated lysis of Ramos cells expressing CD20 in a calcein-release assay.
  • Example 2 Modified al- a2 Domains of NKG2D Ligands.
  • Human proteins designated ULBP-1 through ULBP-6 are, like MICA and MICB, naturally occurring, stress- induced, cell surface ligands that bind NKG2D receptors on and activate human NK cells and certain T-cells (15; Cerwenka A, Lanier LL (2004).
  • NKG2D ligands unconventional MHC class I-like molecules exploited by viruses and cancer.
  • cowpox virus protein OMCP is a secreted domain that like the al-a2 domain of MIC proteins binds NKG2D.
  • OMCP exhibits a very high affinity for NKG2D, apparently in order to block NKG2D's recognition of the natural stress ligands induced by the virus on its infected host cell (Eric Lazear, Lance W. Peterson, Chris A. Nelson, David H. Fremont. J Virol. 2013 January; 87(2): 840-850.
  • a baculoviral expression vector to accommodate the DNA fragments (SEQ ID NOs: 9-14) that encoded the different al-a2 domains of ULBP-1 , ULBP-2, ULBP-3, ULBP-4, ULBP-6, and OMCP (SEQ ID NOs: 15-20, respectively).
  • the DNA fragments were amplified by PCR, digested using Blpl and Ncol restriction enzymes, and individually subcloned into the baculoviral expression vector, KLM44, replacing the MICA al -a2 domain.
  • KLM44 was a baculoviral expression vector derived from SW403 into which MICA-a3-iFv.2 had previously been cloned (example 1).
  • the new NKG2DL-a3-iFv.2 constructs, containing the ULBPs and OMCP al-a2 domain fusions to a3-iFv.2 (ULBPl -a3-iFv.2, ULBP2-a3-iFv.2, ULBP3-a3-iFv.2, ULBP4-a3-iFv.2, ULBP6-a3-iFv.2, and OMCP-a3-iFv.2; SEQ ID NOs: 21 -26, respectively), were co-transfected with baculoviral DNA into SF9 insect cells.
  • Baculovirus was grown for two amplification cycles and used to express these His-tagged N G2DL-a3-iFv.2 proteins in T.ni insect cells according to manufacturer's protocol (Invitrogen). The expression was carried out in a 100 mL volume for three days and the growth medium was harvested for purification of the secreted soluble protein using Ni-affinity chromatography. Monomeric proteins of correct molecular weight were purified to >90% purity as determined by SDS-PAGE. Functional characterization was carried out using binding ELISAs and in vitro target cell killing assays.
  • ELISA to confirm simultaneous binding to the FGFR3 target and the NKG2D receptor.
  • FGFR3 in phosphate buffered saline (PBS) was coated onto Maxisorp plates at 2 ug/ml concentration.
  • PBS phosphate buffered saline
  • Each NKG2DL-a3-iFv.2 protein was titrated, allowed to bind FGFR3 for 1 hour, and washed to remove unbound protein.
  • the bound NKG2DL-a3-iFv.2 protein was detected using NKG2D-Fc and anti-Fc-HRP conjugate.
  • Figure 10 shows that all 6 NKG2DL-a3-iFv.2 proteins bound potently to FGFR3, as expected, through interaction with the iFv.2 domain, and the NKG2D binding activity was retained by the attached NKG2DL al-a2 domains, which demonstrated that the attached cc3-iFv domain imparted functional FGFR3 binding activity to the ULBP and OMPC proteins that, like MIC proteins, bind NKG2D.
  • Example 3 Modified al-a2 Domains of NKG2D Ligands. These are examples of attaching polypeptides to NKG2DLs which were modified to significantly enhance their binding affinity to the human NKG2D receptor.
  • the al-a2 domain of MIC proteins is an NKG2DL for the NKG2D receptor. This affinity is sufficient for physiologic activation of NK cells and stimulating lysis of cells expressing native full-length MIC proteins irreversibly tethered to the two-dimensional plasma membrane surface of a "target cell” (Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, Spies T., Science. 1999 Jul 30;285(5428):727-9.).
  • engineered soluble MIC proteins of the instant invention reversibly bind specific target antigens on the surface of a target cell
  • the binding affinity of the engineered soluble MIC protein to NKG2D will directly affect the stability of the soluble MIC-dependent complex formed between NK cells and cells expressing target antigens.
  • the affinity between sMICA and NKG2D is increased by a substantially slower dissociation rate or off-rate of the modified sMICA from NKG2D, the NK cell-based killing would be expected to be greater at lower densities of soluble MIC molecules bound to a target cell.
  • This example of the instant invention relates to modifying the NKG2D binding affinity of soluble MIC proteins through engineering specific mutations at selected amino acid positions within the al-a2 domain that influence the off- rate binding kinetics and thereby alter the NK cell-mediated killing activity of the invented non-natural, targeted MIC molecules.
  • the l-a2 phage libraries were sorted for increased binding affinity using recombinant biotinylated NKG2D as the target antigen and cycled through iterative rounds of intentionally prolonged binding, prolonged washing, and eluting of the phage clones in order to select high affinity variants enriched for slow dissociation- or off-rates.
  • a set of specific amino acid mutations occurred at high frequencies at 6 positions in l - 2 and were selected as preferred amino acid substitutions with enhanced NKG2D binding affinity (Figure 12B, Table 1).
  • Table 1 Selected affinity mutations at the indicated 6 amino acid positions of the al-a2 domain of MIC.
  • the amino acids of SEQ ID NO: 35 at each of the 6 positions are shown in bold in the first row of the table.
  • the identified affinity mutations are listed in decreasing frequency from top to bottom. All amino acids are represented by the single letter IUPAC abbreviations.
  • heterologous molecules such as a polypeptide to each of these 4 modified al-a2 NKG2DLs using a linker peptide.
  • His-tagged proteins SEQ ID NOs: 31-34 consisting of modified NKG2DLs with attached heterologous molecules were expressed in insect cells and purified to characterize their N G2D binding affinities and kinetic binding parameters.
  • a competitive binding ELISA we determined the relative NKG2D binding affinities of the 4 modified l-a2 variants.
  • a soluble wild type (WT) NKG2DL, sMICA protein was coated in all wells of a maxisorp ELISA plate to provide a binding partner for the human NKG2D-Fc reagent.
  • Solutions of the four al-a2 variants as well as WT and WED- al-cc2 domains (SEQ ID NO: 35) were titrated in the ELISA wells and allowed to competitively inhibit 2nM human NKG2D-Fc binding to the WT sMICA coated on the plate.
  • the level of human NKG2D-Fc that bound to the WT NKG2DL on the plate was detected using an anti-Fc-HRP antibody.
  • Figure 13A shows variants 16, 17, and 18 exhibited IC50 values of 0.7, 0.6, 0.5 nM while variant 15 exhibited an IC50 value of 1.7 nM, all possessing significantly better binding to NKG2D, 27, 32-, 38- and 1 1 -fold better, than WT N G2DL, respectively, as well as substantially better than WED-MICA (Table 3).
  • variant 15 displayed a similar slow off-rate as did 16, 17, and 18, its on-rate was 2016/039281 decreased, resulting in an affinity stronger than WT but weaker variants 16, 17 and 18. Because the only difference between variant 15 (SEQ ID NO: 31) and 16 (SEQ ID NO: 32) was K125N versus K125L, the mutation at position 125 clearly altered the on-rate while the decreased off- rate was attributed to the HI 61R mutation.
  • FGFR3 -expressing target cells was demonstrated in vitro in a calcein-release assay.
  • the human Natural Killer (NK) cell line, NKL was co-cultured with calcein-loaded P815 target cells ectopically expressing FGFR3 and titrated with soluble modified MIC proteins.
  • the results in Figure 15 showed that the killing activities of the FGFR3 -specific soluble MIC variants correlated with their engineered al-a2 affinities. Specifically, variants 16, 17, and 18 exhibited ⁇ 15-fold more killing than WT at 0.78 nM.
  • the WED-MICA SEQ ID NO: 35 was only slightly better than WT.
  • the invention describes amino acid substitutions within the al-a2 domain that increased the NKG2D binding affinity by reducing the off-rate of soluble MIC protein binding to human NKG2D and consequentially led to the predictably increased killing potency.
  • WED-MICA which exhibited somewhat greater affinity than WT MICA to NKG2D (Figure 13 A) by increasing on-rate rather than reducing off-rate (Figure 14), did not exhibit substantial improvement of target cell killing (Figure 15).
  • Figure 13B WED-MICA exhibited substantially poorer binding to murine NKG2D than even WT MICA, while variants 15, 16, 17, and 18 each exhibited greater affinity for both human and murine N G2D, Figures 13 A and 13B.
  • Example 4 Modified al-a2 Domains of NKG2D Ligands.
  • This embodiment of the instant invention relates to additional al-a2 NKG2DL affinity variants derived through engineering specific mutations at selected amino acid positions within the al-a2 domain of soluble MIC molecules, as described in Example 3 (Table 1), that also influence the off-rate binding kinetics and thereby alter the NK cell-mediated killing activity of the non-natural al-a2 domains. While variants 15-18 focused on specific mutations found at positions S20, G68, K125, and H161 , another set of variants were isolated with additional mutations at E152, H158, and Q166 (Table 4).
  • Table 4 Sequences of specific al-a2 domain variants. The specific amino acid substitutions for variants 20, 25, and 48 are listed relative to the amino acids of SEQ ID NO: 35, shown in bold in the first row of the table. All amino acids are represented by the single letter IUPAC abbreviations.
  • DNA polynucleotides (SEQ ID NOs: 36-38) encoding the al-a2 domains of 3 representative variants 20, 25, 48 (SEQ ID NOs: 39-41 , respectively) that contained different combinations of specific discovered mutations (Table 4), were synthesized.
  • heterologous molecules such as an FGFR3-binding polypeptide, were directly attached to each of these 3 modified al -a2 NKG2DLs using a linker peptide.
  • the constructs were cloned into the Xbal and BamHI sites of pD2509, a CMV-based mammalian cell expression vector.
  • Three His-tagged proteins consisting of modified NKG2DLs with attached heterologous molecules that bind to FGFR3, were transiently expressed in HEK293 cells using the Expi293 expression system according to the manufacturer's protocol (Life Technologies), and purified using Ni-affinity chromatography to obtain the isolated proteins for biochemical and activity-based analysis.
  • both the on-rates and off- rates for the three l -a2 variant NKG2DLs binding to surface-coated biotinylated human NKG2D were measured using biolayer interferometry (Octet). Binding titrations were performed for each protein using a titration range of 1-100 nM, and the kinetic data were fitted to obtain on-rates, off-rates, and equilibrium binding constants.
  • Variant 25 contains only the addition of the Q166S mutation relative to variant 16 (SEQ ID NO: 32) (Table 2), and exhibited a N G2D binding affinity of 62 pM largely due to decreased off-rate ( Figure 16 and Table 5). This represented an 8-fold enhancement in equilibrium binding affinity due to the Q166S mutation (compare Table 3 and Table 5), and demonstrated that specific mutations at Q 166 influenced binding affinity through decreased off-rate.
  • Variant 20 (SEQ ID NO: 39) contained the specific mutations G68A, El 52Q,
  • Variant 48 contained the K125L and H161R mutations found in variant 16 (Table 2); however the addition of mutations E152A, H158I, and Q166A (Table 4) significantly increased the off-rate, resulting in a 250-fold reduction in N G2D binding affinity ( Figure 16 and Table 5).
  • the Q166A mutation is not one of the favored affinity enhancement mutations selected for position Q166 (Table 1) and may have contributed to the reduction in off- rate observed.
  • the non-natural al-a2 affinity variants with attached polypeptides redirected NK cell-mediated lysis of FGFR3 -expressing target cells, as demonstrated in vitro in a calcein- release assay.
  • the human Natural Killer (NK) cell line, NKL was co-cultured with calcein- loaded P815 target cells ectopically expressing FGFR3, and titrated with soluble modified NKG2D ligand al -a2 proteins.
  • the results in Figure 17 showed that the killing potencies of the FGFR3 -targeted soluble MIC variants correlated with their engineered al-a2 affinities.
  • variant 25 exhibited ⁇ 3-fold greater killing than variant 16 at 0.2 nM, representing an ⁇ 5-fold improvement in the EC50 for cell killing.
  • the data clearly showed preferred killing activity across representative soluble MIC variants in the order of variant 25>16>WED ( Figure 17).
  • Example 5 Modified l-a2 Domains of NKG2D Ligands.
  • This embodiment relates to additional l-a2 N G2DL affinity variants derived through engineering the al-a2 domains of ULBP proteins.
  • ULBP proteins contain l-a2 domains, which are NKG2D ligands capable of binding to the NKG2D receptor (Cerwenka A, Lanier LL (2004).
  • N G2D ligands unconventional MHC class I-like molecules exploited by viruses and cancer. Tissue Antigens 61 (5): 335-43. doi: 10.1034/j. l399-0039.2003.00070.x. PMID 12753652).
  • N G2D ligands unconventional MHC class I-like molecules exploited by viruses and cancer.
  • engineered soluble al-a2 domains fused to heterologous polypeptides in certain embodiments of the instant invention reversibly bind specific target antigens on the surface of a target cell
  • the binding affinity of the engineered ULBP al -a2 domains to NKG2D will directly affect the stability of the artificial synapse formed between NK cells and cells expressing target antigens, as already shown by engineered soluble MIC proteins (Examples 2- 4).
  • the al-a2 phage display libraries were sorted for increased binding affinity to NKG2D using human NKG2D-Fc as the T U 2016/039281 target protein, and cycled through iterative rounds of intentionally prolonged binding, prolonged washing, and eluting of the phage clones in order to select high affinity variants enriched for slow dissociation- or off-rates.
  • specific amino acid mutations were found at high frequencies at positions R80, V151, V152, and A153 in al-a2, and were identified as preferred amino acid substitutions with enhanced NKG2D-binding affinity (Figure 19 A and Table 6).
  • Table 7 Selected affinity mutations at the indicated 2 amino acid positions of the al-a2 domain of ULBP3. The amino acids of SEQ ID NO: 17 at each of the 2 positions are shown in bold in the first row of the table. The identified affinity mutations are listed in decreasing frequency from top to bottom. All amino acids are represented by the single letter IUPAC abbreviations.
  • Example 6 Modified l-a2 Domains fused to antibody peptides. These are examples of attaching antibody polypeptides to NKG2DLs which were modified to significantly enhance their binding affinity to the human NKG2D receptor.
  • the al-a2 domain of MIC proteins is an NKG2DL for the NKG2D receptor.
  • Antibodies are highly stable glycoproteins made up of two large heavy chains and two small light chains ( Figure 1). The large amount of diversity that can be generated within the CDR regions of the variable domains allows for specific antibodies to be generated to specific antigen targets (Hozumi N, Tonegawa S (1976). "Evidence for somatic rearrangement of immunoglobulin genes coding for variable and constant regions". Proc. Natl. Acad. Sci. U.S.A. 73 (10): 3628-3632. doi.T 0.1073/pnas.73.10.3628.
  • Antibodies have become a significant therapeutic platform for drug development and can mediate both target binding and neutralization, as well as modulate the immune system through complement and Fc receptor binding (Vidarsson, G., Dekkers, G., Rispens, T. (2014) IgG subclasses and allotypes: from structure to effector functions. Frontiers in Immunology 5, 520.). Prior to the present invention, there did not exist an IgG antibody format that can directly activate immune cells using non-natural al-a2 domains that bind more tightly than native NKG2DLs to the NKG2D receptor.
  • mice can be fused to an anti-Her2 antibody for use as an anti -tumor agent in mice (Cho, HM., Rosenblatt, JD., Tolba, K., Shin, SJ., Shin, D., Calfa, C, Zhang, Y., Shin, SU. (2010) Delivery of NKG2D ligand using and anti-Her2 antibody -NKG2D ligand fusion protein results in an enhanced innate and adaptive antitumor response. Cancer Research 70, 10121-30.).
  • mouse NKG2D ligands do not bind human NKG2D, and there are no natural human N G2D ligands with high affinity to human and mouse NKG2D.
  • Fusions between the engineered al -a2 NKG2D ligands of the instant invention and the heavy chain or light chain of IgG antibodies overcame these limitations and highlighted the versatility of fusions of modified al-a2 domains to heterologous proteins or peptides.
  • the DNA sequences encoding al-a2 domain for MIC WT, variants WED, 25, and 48 were synthesized and cloned as C-terminal fusions to either the heavy chain (HC_WT, HC_WED, HC_25, HC_48) or light chain (LC_WT, LC_WED, LC_25, LC_48) sequence from the FGFR3 -specific antibody (Qing, J., Du, X., Chen, Y., Chan, P., Li, H., Wu, P., Marsters, S., Stawicki, S., Tien, J., Totpal, K., Ross, S., Stinson, S., Dornan, D., French, D., Wang, Q.
  • Transient expressions were carried out in HEK293 cells using the Expi293 expression system according to the manufacturer's protocol (Life Technologies), and purified using standard protein A affinity chromatography.
  • the ability of the non-natural al-a2-antibody fusions to redirect N cell- mediated lysis of FGFR3 -expressing target cells was demonstrated in vitro in a calcein-release assay.
  • the human Natural Killer (NK) cell line, NKL was co-cultured with calcein-loaded P815 target cells ectopically expressing FGFR3 and titrated with the engineered antibody fusion proteins.
  • NK cell line The human Natural Killer (NK) cell line, NKL, was co-cultured with calcein-loaded A431 EGFR-expressing target cells, SKBR3 Her2-expressing target cells, or PDL1 -expressing B16 cells and titrated with the respective target-specific engineered antibody fusion proteins.
  • the results in Figures 21 A, 2 IB, and 21 C showed that the killing activities of the target-specific variant 25-antibody fusions were in all cases drastically improved over the non-fused parent antibody and very ' potent with sub-nanomolar EC50 values.
  • modified al-a2 variant-antibody fusions are a universal platform for allowing IgG antibodies to bind tightly to NKG2D and to direct antigen- specific cell lysis.
  • Example 7 (trastuzumab fusions to l- 2 variant 25 bind NK cells in vivo and elicit potent antigen presentation). Fusion proteins containing al-a2 domain variants that bind NKG2D with high affinity bound NK cells in vivo. Thus, antigen-specific antibodies containing modified al-a2 fusions bind NKG2D tightly and thereby effectively armed the surface of NK cells in vivo with antibodies to seek out target cells expressing a particular antigen. This activity was similar to engineered CAR cells (Gill, S., and June, CH. (2015) Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies.
  • trastuzumab and the corresponding heavy and light chain fusions of variant 25 were analyzed in vivo for serum pharmacokinetic (PK) profiles and the pharmacodynamics (PD) of NK cell labeling. All three antibodies: parent trastuzumab; trastuzumab HC_25 fusion; and trastuzumab LC_25 fusion, were conjugated with Alexa Flour 647 according to the
  • mice were injected with a single dose of 100 ⁇ g of each antibody, and blood was drawn at indicated time points for plasma PK ELISAs and flow cytometry of peripheral NK cells.
  • the PK profile of the parent trastuzumab antibody displayed typical alpha-phase distribution within 24-hrs and beta-phase elimination consistent with greater than a 1 week half-life of antibodies in mice ( Figure 22 A).
  • the initial alpha-phase showed a much greater volume of distribution relative to the parent antibody, consistent with an NKG2D- sink, while the beta-phase elimination was also consistent with typical antibody clearance in mice ( Figure 22 A).
  • Example 8 Antibody heavy chain fusion to l-a2 variant 25 exhibited antitumor activity in vivo.
  • an anti-PDLl antibody heavy chain fusion to variant 25 al-a2 was evaluated in a syngeneic MC38 tumor model. MC38 tumors were implanted sub-cutaneously in C57BL/6 mice and tumors grew to an average of 100 mm 3 before the initiation of treatment.
  • mice per group Upon initiation of treatment, four cohorts of 10 mice per group were treated with vehicle, anti-CTLA4 (100 ug i.p.), parent anti-PDLl (300 ug i.v.), or anti-PDLl HC_25 fusion (300 ug i.v.) on days 1, 4, and 7.
  • anti-CTLA4 100 ug i.p.
  • parent anti-PDLl 300 ug i.v.
  • anti-PDLl HC_25 fusion 300 ug i.v.
  • the anti-PDLl HC_25 treatment began to lose efficacy consistent with the occurrence of an ADA response as observed for trastuzumab fusions (Example 7).
  • the significant antitumor activity observed for the antibody heavy chain fusion to variant 25 relative to both the parent antibody and standard anti-CTLA4 treatments demonstrated the impressive therapeutic effect of antibody fusions to modified l-a2 domains that served as high affinity NKG2D ligands.
  • Example 9 (Binding and Cytolysis by Modified al- a2 Domains of ULBPs
  • the following example relates to attaching antibody polypeptides to NKG2DLs which were modified to significantly enhance their binding affinity to the human and murine NKG2D receptor.
  • the al-a2 domain of ULBP proteins is a natural ligand for the NKG2D receptor, i. e. an NKG2DL.
  • Antibodies are highly stable glycoproteins made up of two large heavy chains and two small light chains ( Figure 1). There did not exist in the art an IgG antibody format that can directly activate immune cells using non-natural ULBP l-a2 domains that bind more tightly than native ULBP domains to the NKG2D receptor.
  • the ULBP al -a2 domains provide alternative NKG2DLs to construct antibody fusions that may have differential in vivo properties relative to MICA al-a2 domains.
  • an in vivo antidrug antibody response to MICA al-a2 domains within an antibody fusion would likely not react to or interfere with modified ULBP l- 2 domains due to the low sequence homology between ULBP and MICA al-a2 domains ( Figure 18).
  • ULBP2 variant fusions HC_R80W and HC_V151D killed SKBR3 cells more effectively than antibody fusions containing either native l -a2 domain.
  • modified al-a2 variant-antibody fusions are a universal platform for enabling IgG molecules to bind tightly to NKG2D and to direct antigen-specific cell lysis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne de manière générale la production de polypeptides ayant des propriétés de liaison à l'antigène spécifiques de domaines Fv, par exemple, des fragments variables insérables d'anticorps et des domaines α1-α2 modifiés de ligands NKG2D. L'invention concerne également des domaines α1-α2 modifiés de ligands NKG2D attachés à des polypeptides, dans certains modes de réalisation, des anticorps ou des fragments d'anticorps. L'invention concerne en outre des peptides de liaison à l'antigène dérivés de domaines variables d'anticorps à chaîne légère et lourde, qui contiennent deux régions de liaison et un domaine variable divisé.
PCT/US2016/039281 2016-06-24 2016-06-24 Fragments variables insérables d'anticorps et domaines a1-a2 modifiés de ligands nkg2d WO2017222556A2 (fr)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP2019519951A JP2019523016A (ja) 2016-06-24 2016-06-24 抗体の挿入可能な可変フラグメントおよびnkg2dリガンドの改変型a1−a2ドメイン
EP16734162.7A EP3475300A2 (fr) 2016-06-24 2016-06-24 Fragments variables insérables d'anticorps et domaines a1-a2 modifiés de ligands nkg2d
PCT/US2016/039281 WO2017222556A2 (fr) 2016-06-24 2016-06-24 Fragments variables insérables d'anticorps et domaines a1-a2 modifiés de ligands nkg2d
KR1020187037547A KR20190019091A (ko) 2016-06-24 2016-06-24 항체의 삽입가능 가변 단편 및 nkg2d 리간드의 변형된 a1-a2 도메인
AU2016410294A AU2016410294A1 (en) 2016-06-24 2016-06-24 Insertable variable fragments of antibodies and modified a1-a2 domains of NKG2D ligands
CA3027908A CA3027908A1 (fr) 2016-06-24 2016-06-24 Fragments variables inserables d'anticorps et domaines a1-a2 modifies de ligands nkg2d

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2016/039281 WO2017222556A2 (fr) 2016-06-24 2016-06-24 Fragments variables insérables d'anticorps et domaines a1-a2 modifiés de ligands nkg2d

Publications (2)

Publication Number Publication Date
WO2017222556A2 true WO2017222556A2 (fr) 2017-12-28
WO2017222556A3 WO2017222556A3 (fr) 2018-04-19

Family

ID=60783281

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/039281 WO2017222556A2 (fr) 2016-06-24 2016-06-24 Fragments variables insérables d'anticorps et domaines a1-a2 modifiés de ligands nkg2d

Country Status (6)

Country Link
EP (1) EP3475300A2 (fr)
JP (1) JP2019523016A (fr)
KR (1) KR20190019091A (fr)
AU (1) AU2016410294A1 (fr)
CA (1) CA3027908A1 (fr)
WO (1) WO2017222556A2 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020013126A1 (fr) * 2018-07-09 2020-01-16 国立大学法人 熊本大学 Anticorps à chaîne unique cyclique
WO2021178896A1 (fr) 2020-03-06 2021-09-10 Go Therapeutics, Inc. Anticorps anti-glyco-cd44 et leurs utilisations
CN114072416A (zh) * 2019-02-18 2022-02-18 克里尔治疗股份有限公司 使用正痘病毒主要组织相容性复合物(mhc)i类样蛋白(omcp)和肿瘤特异性结合伴侣的双特异性融合蛋白
WO2022187591A1 (fr) 2021-03-05 2022-09-09 Go Therapeutics, Inc. Anticorps anti-glyco-cd44 et leurs utilisations
WO2023014863A1 (fr) 2021-08-05 2023-02-09 Go Therapeutics, Inc. Anticorps anti-glyco-muc4 et leurs utilisations
WO2023034571A1 (fr) 2021-09-03 2023-03-09 Go Therapeutics, Inc. Anticorps anti-glyco-lamp1 et leurs utilisations
WO2023034569A1 (fr) 2021-09-03 2023-03-09 Go Therapeutics, Inc. Anticorps anti-glyco-cmet et leurs utilisations
RU2815278C2 (ru) * 2018-03-27 2024-03-13 Зайфос Байосайенсиз Инк. МОДИФИЦИРОВАННЫЕ ДОМЕНЫ α1-α2 НЕПРИРОДНЫХ ЛИГАНДОВ NKG2D, КОТОРЫЕ СВЯЗЫВАЮТСЯ С НЕПРИРОДНЫМИ РЕЦЕПТОРАМИ NKG2D

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6217866B1 (en) 1988-09-15 2001-04-17 Rhone-Poulenc Rorer International (Holdings), Inc. Monoclonal antibodies specific to human epidermal growth factor receptor and therapeutic methods employing same
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8796420B2 (en) * 2009-12-31 2014-08-05 Avidbiotics Corp. Non-natural MIC proteins
US20150165065A1 (en) * 2009-12-31 2015-06-18 Avidbiotics Corp. Non-natural mic proteins
PL3227311T3 (pl) * 2014-12-05 2022-05-16 Xyphos Biosciences Inc. WSTAWIALNE ZMIENNE FRAGMENTY PRZECIWCIAŁ I ZMODYFIKOWANE DOMENY α1-α2 LIGANDÓW NKG2D
CN104592395B (zh) * 2015-01-27 2017-11-14 中国药科大学 Vegfr2单链抗体与mica融合蛋白的制备方法及用途
CA2993354A1 (fr) * 2015-08-04 2017-02-09 Avidbiotics Corp. Fragments d'anticorps variables et inserables et domaines a1-a2 modifies de ligands de nkg2d, et ligands non naturels de nkg2d qui se lient a des recepteurs nkg2d non naturels

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6217866B1 (en) 1988-09-15 2001-04-17 Rhone-Poulenc Rorer International (Holdings), Inc. Monoclonal antibodies specific to human epidermal growth factor receptor and therapeutic methods employing same
US20140341917A1 (en) 2011-11-28 2014-11-20 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof

Non-Patent Citations (55)

* Cited by examiner, † Cited by third party
Title
; BAHRAM; SPIES TA.: "Nucleotide sequence of human MHC class I MICB cDNA", IMMUNOGENETICS, vol. 43, 1996, pages 230 - 233
ANDRIS-WIDHOPF, J.; STEINBERGER, P.; FULLER, R.; RADER, C.; BARBAS, C. F., 3RD.: "Generation of human Fab antibody libraries: PCR amplification and assembly of light- and heavy-chain coding sequences", COLD SPRING HARBOR PROTOCOLS, 2011
ANDRIS-WIDHOPF, J.; STEINBERGER, P.; FULLER, R.; RADER, C; BARBAS, C. F., 3RD.: "Generation of human Fab antibody libraries: PCR amplification and assembly of light- and heavy-chain coding sequences", COLD SPRING HARBOR PROTOCOLS, 2011
BAHRAM ET AL.: "A second lineage of mammalian MHC class I genes.", PNAS, vol. 91, 1994, pages 6259 - 6263, XP002057254, DOI: doi:10.1073/pnas.91.14.6259
BAHRAM ET AL.: "Nucleotide sequence of the human MHC class I MICA gene", IMMUNOGENETICS, vol. 44, 1996, pages 80 - 81, XP002057253, DOI: doi:10.1007/s002510050092
BAUER S; GROH V; WU J; STEINLE A; PHILLIPS JH; LANIER LL; SPIES T., SCIENCE, vol. 285, no. 5428, 30 July 1999 (1999-07-30), pages 727 - 9
BAUER, S ET AL.: "Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA", SCIENCE, vol. 285, 1999, pages 727 - 730
BIRD, R. E., HARDMAN, K. D., JACOBSON, J. W.: "Single-chain antigen-binding proteins", SCIENCE, vol. 242, 1988, pages 423 - 426, XP000575094, DOI: doi:10.1126/science.3140379
BURGESS, SJ., IMMUNOL. RES, vol. 40, 2008, pages 18 - 34
BUSCHE, A ET AL.: "NK cell mediated rejection of experimental human lung cancer by genetic over expression of MHC class I chain-related gene A.", HUMAN GENE THERAPY, vol. 17, 2006, pages 135 - 146
CARTER, P.; PRESTA, L.; GORMAN, CM.; RIDGWAY, JB.; HENNER, D.; WONG, WL.; ROWLAND, AM.; KOTTS, C.; CARVER, ME.; SHEPARD, HM., PROC NATL ACAD SCI, vol. 15, 1992, pages 4285 - 9
CERWENKA A; LANIER LL: "NKG2D ligands: unconventional MHC class I-like molecules exploited by viruses and cancer", TISSUE ANTIGENS, vol. 61, no. 5, 2004, pages 335 - 43
CERWENKA A; LANIER LL: "NKG2D ligands: unconventional MHC class I-like molecules exploited by viruses and cancer.", TISSUE ANTIGENS, vol. 61, no. 5, 2004, pages 335 - 43
CERWENKA, A.; L.L. LANIER.: "NK cells, viruses and cancer", NAT. REV. IMMUNOL, vol. 1, 2001, pages 41 - 49, XP009029995, DOI: doi:10.1038/35095564
CHARLES JANEWAY: "Immunobiology", 2001, GARLAND PUBLISHING
CHO, HM.; ROSENBLATT, JD.; TOLBA, K.; SHIN, SJ.; SHIN, D.; CALFA, C.; ZHANG, Y.; SHIN, SU.: "Delivery of NKG2D ligand using and anti-Her2 antibody-NKG2D ligand fusion protein results in an enhanced innate and adaptive antitumor response.", CANCER RESEARCH, vol. 70, 2010, pages 10121 - 30
DOUBROVINA, ES; MM DOUBROVIN; E VIDER; RB SISSON; RJ O'REILLY; B DUPONT; YM VYAS: "Evasion from NK Cell Immunity by MHC Class I Chain-Related Molecules Expressing Colon Adenocarcinoma", J. IMMUNOLOGY, 2003, pages 6891 - 99, XP002507236
DU, J.; WANG, H.; ZHONG, C.; PENG, B.; ZHANG, M.; LI, B.; HUO, S.; GUO, Y.; DING, J.: "Structural basis for recognition of CD20 by therapeutic antibody Rituximab", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 282, 2007, pages 15073 - 15080, XP055094566, DOI: doi:10.1074/jbc.M701654200
ERIC LAZEAR; LANCE W. PETERSON; CHRIS A. NELSON; DAVID H. FREMONT., J VIROL., vol. 87, no. 2, January 2013 (2013-01-01), pages 840 - 850
FRIESE, M. ET AL.: "MICA/NKG2D-mediated immunogene therapy of experimental gliomas.", CANCER RESEARCH, vol. 63, 2003, pages 8996 - 9006, XP002472716
FUERTES, MB; MV GIRART; LL MOLINERO; CI DOMAICA; LE ROSSI; MM BARRIO; J MORDOH; GA RABINOVICH; NW ZWIRNER: "Intracellular Retention of the NKG2D Ligand MHC Class I Chain-Related Gene A in Human Melanomas Confers Immune Privilege and Prevents NK Cell-Mediated Cytotoxicity", J. IMMUNOLOGY, vol. 180, 2008, pages 4606 - 4614
GILL, S.; JUNE, CH.: "Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies", IMMUNOL REV, vol. 263, 2015, pages 68 - 89, XP002760559, DOI: doi:10.1111/imr.12243
GROH ET AL.: "Cell stress-regulated human MHC class I gene expressed in GI epithelium.", PNAS, vol. 93, 1996, pages 12445 - 12450, XP002057255, DOI: doi:10.1073/pnas.93.22.12445
GROH ET AL.: "Co-stimulation of CD8+ aPT-cells by NKG2D via engagement by MIC induced on virus-infected cells", NAT. IMMUNOL, vol. 2, 2001, pages 255 - 260
GROH ET AL.: "Recognition of stress-induced MHC molecules by intestinal 6T cells.", SCIENCE, vol. 279, 1998, pages 1737 - 1740
HENAGER, SAMUEL H.; MELISSA A. HALE; NICHOLAS J. MAURICE; ERIN C. DUNNINGTON; CARTER J. SWANSON; MEGAN J. PETERSON; JOSEPH J. BAN;: "Combining different design strategies for rational affinity maturation of the MICA-NKG2D interface.", PROTEIN SCIENCE, vol. 21, pages 1396 - 1402, XP055456979, DOI: doi:10.1002/pro.2115
HOCHMAN J; INBAR D; GIVOL D: "An active antibody fragment (Fv) composed of the variable portions of heavy and light chains", BIOCHEMISTRY, vol. 12, no. 6, 1973, pages 1130 - 1135
HOLMES ET AL.: "Structural Studies of Allelic Diversity of the MHC Class I Homolog MICB, a Stress-Inducible Ligand for the Activating Immunoreceptor NKG2D", J IMMUNOL., vol. 169, 2001, pages 1395 - 1400
HOUCHINS JP ET AL.: "DNA sequence analysis of NKG2, a family of related cDNA clones encoding type II integral membrane proteins on human NK cells.", J. EXP. MED., vol. 173, 1991, pages 1017 - 1020
HOZUMI N; TONEGAWA S: "Evidence for somatic rearrangement of immunoglobulin genes coding for variable and constant regions", PROC. NATL. ACAD. SCI. U.S.A., vol. 73, 1976, pages 3628 - 3632
HOZUMI N; TONEGAWA S: "Evidence for somatic rearrangement of immunoglobulin genes coding for variable and constant regions", PROC. NATL. ACAD. SCI. U.S.A., vol. 73, no. 10, 1976, pages 3628 - 3632
HUSTON, J. S.; LEVINSON, D; MUDGETT-HUNTER, M; TAI, M-S; NOVOTNY, J; MARGOLIES, M.N.; RIDGE, R.; BRUCCOLERI, RE.; HABER, E.; CREA,: "Protein engineering of antibody binding sites: Recovery of specific activity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli", PNAS, vol. 85, 1988, pages 5879 - 5883, XP000872837, DOI: doi:10.1073/pnas.85.16.5879
LANIER, LL: "NK cell receptors", ANN. REV. IMMUNOL., vol. 16, 1998, pages 359 - 393, XP002459728, DOI: doi:10.1146/annurev.immunol.16.1.359
LE GALL, F.; KIPRIYANOV, SM; MOLDENHAUER, G; LITTLE, M: "Di-, tri- and tetrameric single chain Fv antibody fragments against human CD19: effect of valency on cell binding", FEBS LETTERS, vol. 453, no. 1, 1999, pages 164 - 168, XP004259822, DOI: doi:10.1016/S0014-5793(99)00713-9
LENGYEL CS; WILLIS LJ; MANN P; BAKER D; KORTEMME T; STRONG RK; MCFARLAND BJ., J BIOL CHEM, vol. 282, no. 42, 8 August 2007 (2007-08-08), pages 30658 - 66
LI ET AL.: "Complex structure of the activating immunoreceptor NKG2D and its MHC class I-like ligand MICA.", NATURE IMMUNOL, vol. 2, 2001, pages 443 - 451, XP009153395, DOI: doi:10.1038/87757
LI P; MORRIS DL; WILLCOX BE; STEINLE A; SPIES T; STRONG RK, NAT IMMUNOL., vol. 2, no. 5, May 2001 (2001-05-01), pages 443 - 451
LODOEN, M.; K. OGASAWARA; J.A. HAMERMAN; H. ARASE; J.P. HOUCHINS; E.S. MOCARSKI; L.L. LANIER.: "NKG2D-mediated NK cell protection against cytomegalovirus is impaired by gp40 modulation of RAE-1 molecules", J. EXP. MED, vol. 197, 2003, pages 1245 - 1253, XP002359221, DOI: doi:10.1084/jem.20021973
MABR3;QING, J.; DU, X.; CHEN, Y.; CHAN, P.; LI, H.; WU, P.; MARSTERS, S.; STAWICKI, S.; TIEN, J.; TOTPAL, K.: "Antibody-based targeting of FGFR3 in bladder carcinoma and t(4;14)-positive multiple myeloma in mice", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 119, 2009, pages 1216 - 1229, XP002594035, DOI: doi:10.1172/JCI38017
MARKET E; PAPAVASILIOU FN: "V(D)J recombination and the evolution of the adaptive immune system", PLOS BIOL., vol. 1, no. 1, October 2003 (2003-10-01), pages E16
MILLER, B. R.; DEMAREST, S. J.; LUGOVSKOY, A.; HUANG, F.; WU, X.; SNYDER, W. B.; CRONER, L. J.; WANG, N.; AMATUCCI, A.; MICHAELSON: "Stability engineering of scFvs for the development of bispecific and multivalent antibodies", PROTEIN ENGINEERING, DESIGN & SELECTION : PEDS, vol. 23, 2010, pages 549 - 557, XP002690428, DOI: doi:10.1093/protein/gzq028
NORTH B; LEHMANN A; DUNBRACK RL: "A new clustering of antibody CDR loop conformations", J MOL BIOL, vol. 406, no. 2, 2010, pages 228 - 256, XP028129711, DOI: doi:10.1016/j.jmb.2010.10.030
QING, J.; DU, X.; CHEN, Y.; CHAN, P.; LI, H.; WU, P.; MARSTERS, S.; STAWICKI, S.; TIEN, J.; TOTPAL, K.: "Antibody-based targeting of FGFR3 in bladder carcinoma and t(4; 14)-positive multiple myeloma in mice", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 119, 2009, pages 1216 - 1229, XP002594035, DOI: doi:10.1172/JCI38017
RADAEV, S.; ROSTRO, B.; BROOKS, AG.; COLONNA, M.; SUN, PD.: "Conformational plasticity revealed by the cocrystal structure of NKG2D and its class I MHC-like Ligand ULBP3", IMMUNITY, vol. 15, 2001, pages 1039 - 49, XP055456733, DOI: doi:10.1016/S1074-7613(01)00241-2
RAVETCH, JV; LANIER LL.: "Immune Inhibitory Receptors", SCIENCE, vol. 290, 2000, pages 84 - 89, XP003012961, DOI: doi:10.1126/science.290.5489.84
SLAVULJICA, I A BUSCHE; M BABIC; M MITROVIC; I GASPAROVIC; Ð CEKINOVIC; E MARKOVA CAR; EP PUGEL; A CIKOVIC; VJ LISNIC; WJ BRITT: "Recombinant mouse cytomegalovirus expressing a ligand for the NKG2D receptor is attenuated and has improved vaccine properties", J. CLIN. INVEST, vol. 120, 2010, pages 4532 - 4545
SPIES, T.: "Regulation of NKG2D ligands: a purposeful but delicate affair", NATURE IMMUNOL, vol. 9, 2008, pages 1013 - 1015
STEPHENS HA: "MICA and MICB genes: can the enigma of their polymorphism be resolved?", TRENDS IMMUNOL., vol. 22, 2001, pages 378 - 85, XP004247293, DOI: doi:10.1016/S1471-4906(01)01960-3
STERN-GINOSSAR ET AL.: "Host immune system gene targeting by viral miRNA", SCIENCE, vol. 317, 2007, pages 376 - 381
STERN-GINOSSAR ET AL.: "Human microRNAs regulate stress-induced immune responses mediated by the receptor NKG2D", NATURE IMMUNOLOGY, vol. 9, 2008, pages 1065 - 73
VIDARSSON, G.; DEKKERS, G.; RISPENS, T.: "IgG subclasses and allotypes: from structure to effector functions", FRONTIERS IN IMMUNOLOGY, vol. 5, 2014, pages 520, XP055416330, DOI: doi:10.3389/fimmu.2014.00520
WEATHERILL, E. E.; CAIN, K. L.; HEYWOOD, S. P.; COMPSON, J. E.; HEADS, J. T.; ADAMS, R.; HUMPHREYS, D. P.: "Towards a universal disulphide stabilised single chain Fv format: importance of interchain disulphide bond location and vL-vH orientation", PROTEIN ENGINEERING, DESIGN & SELECTION : PEDS, vol. 25, 2012, pages 321 - 329, XP055170699, DOI: doi:10.1093/protein/gzs021
WEIS ET AL.: "The C-type lectin superfamily of the immune system", IMMUNOL. REV., vol. 163, 1998, pages 19 - 34, XP001024357, DOI: doi:10.1111/j.1600-065X.1998.tb01185.x
WOOF J; BURTON D: "Human antibody-Fc receptor interactions illuminated by crystal structures", NAT REV IMMUNOL, vol. 4, no. 2, 2004, pages 89 - 99, XP009051821, DOI: doi:10.1038/nri1266
ZWIRNER ET AL.: "Differential expression of MICA by endothelial cells, fibroblasts, keratinocytes and monocytes", HUMAN IMMUNOL, vol. 60, 1999, pages 323 - 330

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2815278C2 (ru) * 2018-03-27 2024-03-13 Зайфос Байосайенсиз Инк. МОДИФИЦИРОВАННЫЕ ДОМЕНЫ α1-α2 НЕПРИРОДНЫХ ЛИГАНДОВ NKG2D, КОТОРЫЕ СВЯЗЫВАЮТСЯ С НЕПРИРОДНЫМИ РЕЦЕПТОРАМИ NKG2D
WO2020013126A1 (fr) * 2018-07-09 2020-01-16 国立大学法人 熊本大学 Anticorps à chaîne unique cyclique
JPWO2020013126A1 (ja) * 2018-07-09 2021-08-02 国立大学法人 熊本大学 環状一本鎖抗体
CN114072416A (zh) * 2019-02-18 2022-02-18 克里尔治疗股份有限公司 使用正痘病毒主要组织相容性复合物(mhc)i类样蛋白(omcp)和肿瘤特异性结合伴侣的双特异性融合蛋白
JP2022520978A (ja) * 2019-02-18 2022-04-04 クーリエ セラピューティクス インコーポレイテッド オルソポックスウイルス主要組織適合性複合体(mhc)クラスi様タンパク質(omcp)と腫瘍特異的結合パートナーを用いた二重特異性融合タンパク質
WO2021178896A1 (fr) 2020-03-06 2021-09-10 Go Therapeutics, Inc. Anticorps anti-glyco-cd44 et leurs utilisations
WO2022187591A1 (fr) 2021-03-05 2022-09-09 Go Therapeutics, Inc. Anticorps anti-glyco-cd44 et leurs utilisations
WO2023014863A1 (fr) 2021-08-05 2023-02-09 Go Therapeutics, Inc. Anticorps anti-glyco-muc4 et leurs utilisations
WO2023034571A1 (fr) 2021-09-03 2023-03-09 Go Therapeutics, Inc. Anticorps anti-glyco-lamp1 et leurs utilisations
WO2023034569A1 (fr) 2021-09-03 2023-03-09 Go Therapeutics, Inc. Anticorps anti-glyco-cmet et leurs utilisations

Also Published As

Publication number Publication date
CA3027908A1 (fr) 2017-12-28
AU2016410294A1 (en) 2019-01-03
JP2019523016A (ja) 2019-08-22
KR20190019091A (ko) 2019-02-26
WO2017222556A3 (fr) 2018-04-19
EP3475300A2 (fr) 2019-05-01

Similar Documents

Publication Publication Date Title
AU2018223040B2 (en) Insertable variable fragments of antibodies and modified a1-a2 domains of NKG2D ligands
JP7446501B2 (ja) 抗体の挿入可能な可変フラグメント及びnkg2dリガンドの改変されたa1-a2ドメイン、及び非天然のnkg2d受容体に結合する非天然のnkg2dリガンド
US11117969B2 (en) Insertable variable fragments of antibodies and modified α1-α2 domains of NKG2D ligands
WO2017222556A2 (fr) Fragments variables insérables d'anticorps et domaines a1-a2 modifiés de ligands nkg2d
WO2022022709A1 (fr) PROTÉINE DE FUSION SIRPα-FC
CN116496396B (zh) 抗cd70纳米抗体及其用途
CN118119402A (zh) 工程化抗her2双特异性蛋白
CN118126184A (zh) 抗cd70纳米抗体及其用途

Legal Events

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

Ref document number: 16734162

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 3027908

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2019519951

Country of ref document: JP

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20187037547

Country of ref document: KR

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2016410294

Country of ref document: AU

Date of ref document: 20160624

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2016734162

Country of ref document: EP

Effective date: 20190124