CD98HC antigen-binding protein and use thereofBACKGROUND OF THE INVENTIONThe blood-brain barrier (BBB) is a specialized barrier that separates the bloodstream from the brain tissue. It is composed of brain endothelial cells (BECs) , pericytes, and astrocytes. These cells work together to maintain the integrity of the continuous non-fenestrated capillaries through tight junctions. The primary function of the BBB is to regulate the exchange of substances between the blood and the brain, creating a stable and tightly controlled environment necessary for proper brain function. Its presence helps maintain the homeostasis of the central nervous system (CNS) by preventing the entry of harmful substances, pathogens, and large molecules into the brain. However, the BBB also poses challenges for delivering drugs to the brain, as it restricts the passage of many therapeutic agents. For instance, only a small fraction of circulating antibodies, approximately 0.1%, can cross the intact BBB. Moreover, peptides, antibody-drug conjugates, and proteins face similar limitations in crossing this barrier, substantially compromising the efficacy of treatments for CNS diseases. Some endogenous molecules in circulation can cross the BBB through specific receptors and transporters expressed on the luminal side of brain endothelial cells. These membrane protein pathways offer a promising route for delivering hardly penetrated molecules across the BBB using receptor-mediated transcytosis (RMT) .
CD98 heavy chain (CD98HC) , also known as solute carrier family 3 member 2 (SLC3A2) , is encoded by the SLC3A2 gene. CD98HC interacts with a light subunit from the SLC7 family to form heteromeric amino acid transporters capable of transporting 15 different amino acids across the BBB. CD98HC plays a crucial role in amino acid transport across the plasma membrane and is involved in various physiological and pathological processes, such as immune cell activation, cell adhesion, and tumor progression. CD98HC is highly enriched in brain capillaries and microvessels, located on the luminal and abluminal surfaces of brain endothelial cells, making it a suitable target to mediate hardly penetrated molecules across the BBB.
A single-domain antibody, also referred to as a Nanobody or VHH, is an antibody fragment composed of a monomeric variable antibody domain. Compared with conventional antibodies, their small size and single-domain nature offer several advantages, including enhanced tissue penetration and stability. Most importantly, it simplifies the construction of bispecific molecules and further facilitates brain delivery of antibodies, peptides, proteins, and nucleic acids.
Prior art has disclosed several CD98HC antibodies for delivery of drugs across BBB. However, there is still a need for the development of new drugs with high delivery efficiency and suitable for research in both animals and humans.
SUMMARY OF THE INVENTIONThe present disclosure provides an isolated antigen-binding protein, which is capable of binding to CD98HC. The antigen-binding protein exhibits cross-reactivity with the CD98HC of mice, rats and monkeys which confers an advantage for investigating the safety and efficacy of potential therapeutics in these animal models. The antigen-binding protein can also take the therapeutic entity across the blood-brain barrier. Taken Neurotensin which does not have the capability across BBB itself as an example, the CD98HC binding protein of the present application can take the Neurotensin across BBB.
In one aspect, the present application provides an isolated antigen-binding protein, which is capable of binding to CD98HC, said antigen-binding protein has one or more characteristics selected from the group consisting of: 1) is capable of binding to CD98HC with a KD of less that about 1×10-5M, as measured by Octet; 2) exhibit cross-reactivity with the CD98HC of mice, rats and monkeys; and 3) is capable of mediating therapeutical molecule across the BBB.
In some embodiments, the CD98HC comprises the extracellular domain (ECD) of CD98HC.
In some embodiments, the antigen-binding protein comprises an antibody or an antigen-binding fragment thereof.
In some embodiments, the antibody is selected from the group consisting of: a monoclonal antibody, a chimeric antibody, a humanized antibody, a fully human antibody, and a multi-specific antibody.
In some embodiments, the antigen-binding fragment is selected from the group consisting of: a Fab fragment, a Fab’ fragment, a F (ab) 2 fragment, a Fv fragment, a sdAb and an ScFv.
In some embodiments, the antigen-binding fragment is VHH.
In some embodiments, the antigen-binding protein comprises at least one CDR of a heavy chain variable region (VH) , wherein said VH comprises an amino acid sequence as set forth in SEQ ID NO: 92 or SEQ ID NO: 93.
In some embodiments, the antigen-binding protein comprises an HCDR3, wherein said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 90 or SEQ ID NO: 91.
In some embodiments, the antigen-binding protein comprises an HCDR3, wherein said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, or SEQ ID NO: 63.
In some embodiments, the antigen-binding protein comprises an HCDR2, wherein said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 88 or SEQ ID NO: 89.
In some embodiments, the antigen-binding protein comprises an HCDR2, wherein said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 55, SEQ ID NO: 56, or SEQ ID NO: 57.
In some embodiments, the antigen-binding protein comprises an HCDR1, wherein said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 86 or SEQ ID NO: 87.
In some embodiments, the antigen-binding protein comprises an HCDR1, wherein said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, or SEQ ID NO: 51.
In some embodiments, the antigen-binding protein comprises HCDRs 1-3, wherein said HCDR1-3 comprises the amino acid sequences selected from the group consisting of:
1) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 86, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 88, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 90; or
2) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 87, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 89, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 91.
In some embodiments, the antigen-binding protein comprises HCDRs 1-3, wherein said HCDR1-3 comprises the amino acid sequences selected from the group consisting of:
1) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 43, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 52, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 58;
2) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 44, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 52, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 59;
3) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 45, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 53, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 60;
4) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 47, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 55, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 62;
5) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 48, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 55, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63;
6) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 49, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 56, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63;
7) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 50, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 55, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63;
8) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 50, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 57, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63; and
9) said HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 51, said HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 55, said HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63.
In some embodiments, the antigen-binding protein comprises a heavy chain variable region (VH) , wherein said VH comprises an amino acid sequence as set forth in any one of SEQ ID NO: 92 or SEQ ID NO: 93.
In some embodiments, the antigen-binding protein comprises a heavy chain variable region (VH) , wherein said VH comprises an amino acid sequence as set forth in any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94.
In some embodiments, the antigen-binding protein is a VHH, and said VHH comprises an amino acid sequence as set forth in any one of SEQ ID NO: 92 or SEQ ID NO: 93.
In some embodiments, the antigen-binding protein is a VHH, and said VHH comprises an amino acid sequence as set forth in any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94.
In some embodiments, the antigen-binding protein comprises a Fc region.
In some embodiments, the Fc region is derived from IgG Fc region.
In another aspect, the present application provides a pharmaceutical molecule, comprising the antigen-binding protein of the present application.
In some embodiments, the pharmaceutical molecule further comprises a therapeutic entity.
In some embodiments, the therapeutic entity comprises a protein, a peptide, a nucleic acid molecule, and/or a compound.
In some embodiments, the pharmaceutical molecule is a fusion protein, a bispecific antibody, and/or an immunoconjugate.
In some embodiments, the therapeutic entity is linked to said antigen-binding protein directly or indirectly.
In some embodiments, the therapeutic entity is linked to said antigen-binding protein via a linker.
In another aspect, the present application provides an isolated nucleic acid molecule or molecules, encoding for the antigen-binding protein or the pharmaceutical molecule of the present application.
In another aspect, the present application provides vector or vectors, comprising the isolated nucleic acid molecule or molecules of the present application.
In another aspect, the present application provides a cell, comprising the isolated nucleic acid molecule or molecules, or the vector or vectors of the present application.
In another aspect, the present application provides a pharmaceutical composition, comprising the antigen-binding protein, the pharmaceutical molecule, the isolated nucleic acid molecule or molecules, the vector or vectors, and/or the cell, and optionally a pharmaceutically acceptable excipient.
In another aspect, the present application provides a use of the pharmaceutical molecule, the isolated nucleic acid molecule or molecules, the vector or vectors, the cell and/or the pharmaceutical composition in manufacture of a medicament for diagnosing, preventing and/or treating a disease.
In some embodiments, the disease is a CNS disease (central nervous system disease) .
Additional aspects and advantages of the present disclosure will become readily apparent to those skilled in this art from the following detailed description, wherein only illustrative embodiments of the present disclosure are shown and described. As will be realized, the present disclosure is capable of other and different embodiments, and its several details are capable of modifications in various obvious respects, all without departing from the disclosure. Accordingly, the drawings and description are to be regarded as illustrative in nature, and not as restrictive.
INCORPORATION BY REFERENCE
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
BRIEF DESCRIPTION OF THE DRAWING
The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are employed, and the accompanying drawings (also “figure” and “FIG. ” herein) , of which:
FIG. 1 illustrates the binding activity of VHH-Fc to rat CD98HC ECD.
FIG. 2 illustrates the binding activity of VHH-Fc to mouse CD98HC ECD.
FIG. 3 illustrates the FACS binding of VHH-Fc to human CD98 overexpression CHO-K1.
FIG. 4 illustrates the FACS binding of VHH-Fc to Cyno CD98 overexpression CHO-K1.
FIG. 5 illustrates neurotensin-VHH fusion protein-induced hypothermia effect in mice.
FIG. 6 illustrates the inhibition effect of CD98HC domain antibodies on amino acid uptake.
FIG. 7 illustrates neurotensin-humanized VHH fusion protein-induced hypothermia effect in mice.
FIG. 8 illustrates the IgG-VHH fusion protein concentration in the brain homogenate of human CD98HC knock-in mice at 72 hours after administration. As shown by fold change over isotype control.
FIG. 9 illustrates the IgG-VHH fusion protein concentration in brain parenchyma of human CD98HC knock-in mice at 72 hours after administration.
FIG. 10 illustrates the IgG-VHH fusion protein concentration in brain microvessel of human CD98HC knock-in mice at 72 hours after administration.
DETAILED DESCRIPTIONWhile various embodiments of the invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions may occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed.
The term “blood-brain barrier” or “BBB” generally refers to the physiological barrier between the peripheral circulation and the brain and spinal cord (i.e., the CNS) which is formed by tight junctions within the brain capillary endothelial plasma membranes, creating a tight barrier that restricts the transport of molecules into the brain, even very small molecules such as urea (60 Daltons) . The blood-brain barrier within the brain, the blood-spinal cord barrier within the spinal cord, and the blood-retinal barrier within the retina are contiguous capillary barriers within the CNS, and are herein collectively referred to as the blood-brain barrier or BBB. The BBB also encompasses the blood-CSF barrier (choroid plexus) where the barrier is comprised of ependymal cells rather than capillary endothelial cells.
The term “CD98HC (CD98 heavy chain) ” and “SLC3A2” can be used interchangeably, generally refers to any native CD98HC from any source, including mammals such as primates (e.g. humans) and rodents (e.g. mice and rats) , etc. The term may include full-length CD98HC or the fragment thereof. For example, the term may include the ECD of CD98HC. The term may also include the variants or analogues of CD98HC.
The term “antigen-binding protein” , as used herein, generally refers to the protein which is capable of binding to the antigen. The antigen-binding protein has the antigen-binding part. In the present application, the antigen-binding protein may comprise an antibody or the antigen-binding fragment thereof.
The term “antibody” , as used herein, generally refers to an immunoglobulin or an immunoglobulin-like molecule capable of specifically recognizing or binding to an antigen. An antibody may comprise a light chain (L) and/or a heavy chain (H) . The light chains of an antibody can be classified as к and λ light chains. The heavy chains can be classified as μ, δ, γ, α or ε, and the isotypes of an antibody are defined as IgM, IgD, IgG (e.g., IgGl, IgG2, IgG3 or IgG4 subtype) , IgA and IgE, respectively. Each heavy chain may comprise a heavy chain variable region (VH) and a heavy chain constant region (CH) . The heavy chain constant region may comprise three domains (CH1, CH2 and CH3) . Each light chain may comprise a light chain variable region (VL) and a light chain constant region (CL) . The light chain constant region may comprise a CL domain. The VH and/or VL regions can also be subdivided into regions with high variability known as complementarity determining regions (CDRs) interspersed with more conserved regions known as framework regions (FRs) . Each VH and/or VL consists of 3 CDRs and 4 FRs arranged from N-terminal to C-terminal in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions (VH and VL) of each heavy/light chain pair form the antibody binding site, respectively. Distribution of amino acids to regions or domains follows the definition of Kabat Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991) ) , or Chothia & Lesk (1987) J. Mol. Biol. 196: 901-917; Chothia et al. (1989) Nature 342: 878-883. The term “antibody” is not limited by any antibody-producing method. For example, it includes recombinant antibodies, monoclonal antibodies, and other forms of antibodies. In some cases, an antibody of the present disclosure is an isolated antibody.
In certain embodiments, an antibody heavy chain binds to an antigen in the absence of an antibody light chain. In certain embodiments, an antibody light chain binds to an antigen in the absence of an antibody heavy chain. In certain embodiments, an antibody binding region binds to an antigen in the absence of an antibody light chain. In certain embodiments, an antibody binding region binds to an antigen in the absence of an antibody heavy chain. In certain embodiments, an individual variable region specifically binds to an antigen in the absence of other variable regions.
In certain embodiments, definitive delineation of a CDR and identification of residues comprising the binding site of an antibody is accomplished by solving the structure of the antibody and/or solving the structure of the antibody-ligand complex. In certain embodiments, that can be accomplished by any of a variety of techniques known to those skilled in the art, such as X-ray crystallography. In certain embodiments, various methods of analysis can be employed to identify or approximate the CDR regions. Examples of such methods include, but are not limited to, the Kabat definition, the Chothia definition, the AbM definition and the contact definition.
The term “antigen-binding fragment” , as used herein, generally refers to one or more fragments of a full-length antibody that retain the ability to bind the same antigen to which the antibody binds (e.g., CD98HC) and/or competes against an intact antibody for an antigen-specific binding. Antigen-binding fragment can be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies. In some cases, the antigen-binding fragment includes Fab, Fab’, F (ab’) 2, F (ab) 2, VHH, Fd, Fv, dAb and complementarity determining region (CDR) fragments, single chain antibodies (e.g., ScFv) , chimeric antibodies, diabodies, and polypeptides that comprise at least a portion of an antibody that is sufficient to confer specific antigen-binding ability to the polypeptide.
The term “antigen” , as used herein, generally refers to a molecule or a portion of a molecule capable of being bound by a selective binding agent, such as an antigen binding protein (including, e.g., an antibody or immunological functional fragment thereof) . In some embodiments, the antigen is capable of being used in an animal to produce antibodies capable of binding to that antigen. An antigen can possess one or more epitopes that are capable of interacting with different antigen binding proteins, e.g., antibodies.
The term “binding specificity” , as used herein, generally refers to an ability of one substance to bind another substance specifically, and not substantially bind to any other substance at random. For example, one protein may bind to another protein specifically due to their specific structures. Binding specificity may be measured by, e.g., cross-competing assays or other binding assays known in the art.
The term “KD” , as used herein, generally refers to the dissociation constant, a specific type of equilibrium constant that measures the propensity of a larger object to separate (dissociate) reversibly into smaller components, as when a complex falls apart into its component molecules. The dissociation constant is the inverse of the association constant. In the specific case of antibodies (Ab) binding to antigen (Ag) , usually the term affinity constant refers to the association constant.
The term “Kon” , as used herein, generally refers to on rate constant for association of a binding protein (e.g., an antibody or an antigen-binding fragment thereof) to the antigen to form a bound complex (e.g., an antibody/antigen complex) . The term “Kon” also means “association rate constant” , or “ka” , as is used interchangeably herein. This value indicates the binding rate of a binding protein to its target antigen or the rate of complex formation between a binding protein, (e.g., an antibody) and the corresponding antigen.
The term “monoclonal antibody” , as used herein, generally refers to antibodies that are made by identical immune cells that are all clones of a unique parent cell. Monoclonal antibodies can have a monovalent affinity, in that they bind to the same epitope (the part of an antigen that is recognized by the antibody) . Sometimes, monoclonal antibodies may also be multi specific, such as bispecific or tri specific. It has become an important tool in biochemistry, molecular biology, and medicine. Several monoclonal antibody technologies had been developed recently, such as phage display, single B cell culture, single cell amplification from various B cell populations and single plasma cell interrogation technologies.
The term “chimeric antibody” , as used herein, generally refers to an antibody in which the Variable (V) region of light and heavy chains is of mouse origin, while the Constant (C) region is of human origin. In general, the chimeric antibody may retain the specificity and affinity of the original mouse monoclonal antibody, but Human Anti Mouse Antibody (HAMA) response may be significantly reduced.
The term “humanized antibody” , as used herein, generally refers to antibodies from non-human species whose protein sequences have been modified to increase their similarity to antibody variants produced naturally in humans. The amino acid sequence of a humanized antibody may be essentially identical to that of a human variant, despite the non-human origin of some of its complementarity-determining region (CDR) segments responsible for the ability of the antibody to bind to its target antigen.
The term “fully human antibody” and “human antibody” are used interchangeably herein, and generally refers to an antibody that comprises a human variable region and, most preferably a human constant region. In specific embodiments, the terms refer to an antibody that comprises a variable region and constant region of human origin. The term “fully human antibody” includes antibodies having variable and constant regions corresponding to human germline immunoglobulin sequences as described by Kabat et al. (See Kabat et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242) .
The term “Fab fragment” , as used herein, generally refers to a portion (such as an antigen-binding fragment) of an immunoglobulin molecule. A Fab fragment may comprise one light chain and part of a heavy chain with a single antigen-binding site. A Fab fragment may be obtained by papain digestion of an immunoglobulin molecule. For example, a Fab fragment may be composed of one constant and one variable domain of each of the heavy and the light chain. The variable domain may contain the paratope (the antigen-binding site) , comprising a set of the complementarity determining regions, at the amino-terminal end of the immunoglobulin molecule. The enzyme papain may be used to cleave an immunoglobulin molecule into two Fab fragments and one Fc fragment. The enzyme pepsin cleaves below the hinge region, so a F (ab') 2 fragment and a pFc' fragment is formed. Divalent F (ab) 2 or F (ab') 2 fragments have two antigen binding regions that are linked by disulfide bonds. Reduction of F (ab) 2 or F (ab') 2 fragments produce 2 monovalent Fab or Fab' fragments, which have a free sulfhydryl group that is useful for conjugation to other molecules.
The term “Fv fragment” , as used herein, generally refers to the smallest fragment made from enzymatic cleavage of IgG and IgM class antibodies. Fv fragments have the antigen-binding site made of the VH and VL regions, but they lack the CH1 and CL regions. The VH and VL chains may be held together in Fv fragments by non-covalent interactions.
The term “scFv” , as used herein, generally refers to a single-chain antibody fragment. An scFv may be a recombinant single chain polypeptide molecule in which VH and VL of an antibody are connected, either directly or via a peptide linker. Single chain antibodies (scFv) generally do not include portions of the Fc region of antibody, although methods are known for adding such regions to known scFv molecules if desired. See Helfrich et al., A rapid and versatile method for harnessing scFv antibody fragments with various biological functions. J Immunol Methods 237: 131-145 (2000) and de Haard et al., Creating and engineering human antibodies for immunotherapy. Advanced Drug Delivery Reviews 31: 5-31 (1998) .
The term “fusion protein” , as used herein, generally refers to a polypeptide that comprises, or alternatively consists of, an amino acid sequence of a polypeptide fused directly or indirectly (e.g., via a linker) to an amino acid sequence of a heterologous polypeptide (i.e., a polypeptide of a different origin, sequence or structure) .
The term “protein conjugate” , as used herein, generally refers to a conjugate comprising a protein (e.g., an antibody or a functional fragment thereof) conjugated to one or more additional moieties, such as cytotoxic agents, e.g., a chemotherapeutic agent, a drug, a growth inhibitory agent, a toxin (e.g., a protein toxin, an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof) , a label (e.g., a fluorescent label) and/or a radioactive isotope (i.e., a radio-conjugate) .
A number of CDR definitions are in use and are encompassed herein. The Kabat definition is based on sequence variability and is the most commonly used (Kabat EA et al., ibid. ) . Chothia definition is based on the location of the structural loops (Chothia & Lesk J. (1987) Mol. Biol. 196: 901-917) . The AbM definition is a compromise between the Kabat and the Chothia definitions and is used by Oxford Molecular's AbM antibody modelling software (Martin ACR et al., (1989) PNAS USA 86: 9268-9272; Martin ACR et al., (1991) Methods Enzymol. 203: 121-153; Pedersen JT et al., (1992) Immunomethods 1: 126-136; Rees AR et al., (1996) In Sternberg M. J. E. (ed. ) , Protein Structure Prediction. Oxford University Press, Oxford, 141-172) . The contact definition has been recently introduced (MacCallum RM et al., (1996) J. Mol. Biol. 262: 732-745) and is based on an analysis of the available complex structures available in the Protein Databank. The definition of the CDR by the international ImMunoGeneTics information (http: //www. imgt. org) is based on the IMGT numbering for all immunoglobulin and T cell receptor V-REGIONs of all species (the international ImMunoGeneTics informationLefranc MP et al., (1999) Nucleic Acids Res. 27 (1) : 209-12; Ruiz M et al., (2000) Nucleic Acids Res. 28 (1) : 219-21; Lefranc MP (2001) Nucleic Acids Res. 29 (1) : 207-9; Lefranc MP (2003) Nucleic Acids Res. 31 (1) : 307-10; Lefranc MP et al., (2005) Dev. Comp. Immunol. 29 (3) : 185-203; Kaas Q et al., (2007) Briefings in Functional Genomics & Proteomics, 6 (4) : 253-64) .
The term “isolated nucleic acid molecule or molecules” as used herein, generally refers to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof, isolated from its native environment, or that is artificially synthesized.
The term “vector or vectors” as used herein, generally refers to a nucleic acid vehicle into which a polynucleotide encoding a protein can be inserted and expressed. The genetic material elements carried in the vector can be expressed in a host cell by transforming, transducing, or transfecting the host cell with the vector. A vector may contain a variety of elements that control expression, including promoter sequences, transcriptional initiation sequences, enhancer sequences, selection elements, and reporter genes. In addition, the vector may also contain an origin of replication. It is also possible that the vector may include components that assist its entry into the cell, such as viral particles, liposomes or protein shells, but not only these substances.
The term “cell” as used herein, generally refers to a cell that may be used to carry the vector or vectors of the present disclosure, or be used to express or produce the antibody, the antigen-binding fragment of the present disclosure. A cell of the present disclosure may be a host cell.
The terms “disease” and “disorder” may be used interchangeably herein, and generally refer to any condition that impairs the normal functioning of the body.
As used herein, the term "subject" includes any human or non-human animal. The term "non-human animal" includes all vertebrates, e.g., mammals and non-mammals, such as primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc. For example, the subject may be human.
The term “pharmaceutically acceptable excipient” , as used herein, generally refers to any and all solvents, dispersion media, coatings, isotonic and absorption delaying agents, etc., that are compatible with pharmaceutical administration.
The term “about” , as used herein, generally refers to an approximation to a given value that would reasonably be inferred based on the ordinary skill in the art, including equivalents and approximations due to the experimental and/or measurement conditions for such given value. For example, it may refer to a value that is no more than 10%above or below the value being modified by the term.
The term “isolated protein” (such as isolated antibody) , as used herein, generally refers to a subject protein (1) is free of at least some other proteins with which it would normally be found, (2) is essentially free of other proteins from the same source, e.g., from the same species, (3) is expressed by a cell from a different species, (4) has been separated from at least about 50 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is associated in nature, (5) is operably associated (by covalent or noncovalent interaction) with a polypeptide with which it is not associated in nature, or (6) does not occur in nature. Typically, an “isolated protein” constitutes at least about 5%, at least about 10%, at least about 25%, or at least about 50%of a given sample. Genomic DNA, cDNA, mRNA or other RNA, of synthetic origin, or any combination thereof can encode such an isolated protein. Preferably, the isolated protein is substantially free from proteins or polypeptides or other contaminants that are found in its natural environment that would interfere with its therapeutic, diagnostic, prophylactic, research or other use.
The term “treat” and “treatment” includes therapeutic treatments, prophylactic treatments, and applications in which one reduces the risk that a subject will develop a disorder or other risk factor. Treatment does not require the complete curing of a disorder and encompasses embodiments in which one reduces symptoms or underlying risk factors.
The term “prevent” does not require the 100%elimination of the possibility of an event. Rather, it denotes that the likelihood of the occurrence of the event has been reduced in the presence of the compound or method.
Standard techniques can be used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection) . Enzymatic reactions and purification techniques can be performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures can be generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See, e.g., Sambrook et al, Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989) ) , which is incorporated herein by reference for any purpose. Unless specific definitions are provided, the nomenclatures utilized in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well-known and commonly used in the art. Standard techniques can be used for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients.
Antigen-binding protein
In one aspect, the present application provides an antigen-binding protein which binds to CD98HC. The antigen-binding protein may specifically bind to CD98HC.
In some embodiments, the CD98HC may comprise CD98HC or fragment thereof. For example, the CD98HC may comprise the extracellular domain (ECD) of CD98HC. For example, the ECD of CD98HC may comprise an amino acid sequence as set forth in SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4. For example, the CD98HC may be derived from human, mouse, rat or monkey.
The antigen-binding protein may bind to CD98HC with a KD of less than about 1.0 ×10-5M, as measured by Octet. For example, with a KD of less than about 5 ×10-6M, less than about 1 ×10-6M, less than about 5 ×10-7M, less than about 4 ×10-7M, less than about 3 ×10-7M, less than about 2×10-7M, less than about 1 ×10-7M, less than about 5 ×10-8M, less than about 4 ×10-8M, less than about 3 ×10-8M, less than about 2×10-8M, less than about 1 ×10-8M, less than about 5 ×10-9M, less than about 4 ×10-9M, less than about 3.5 ×10-9M, less than about 3×10-9M, less than about 2.8 ×10-9M, less than about 2.7×10-9M, less than about 2.5×10-9M, less than about 2×10-9M, less than about 1.5×10-9M, less than about 1×10-9M, less than about 8×10-10M, less than about 5×10-10M, less than about 5×10-10M, less than about 4.5×10-10M, less than about 4×10-10M, less than about 3.5×10-10M, less than about 3×10-10M, less than about 2.5×10-10M, less than about 2×10-10M, less than about 1.5×10-10M, less than about 1×10-10M, less than about 1×10-11M, less than about 1×10-12M, or less than about 1×10-13M, or a KD value that is even smaller.
In some embodiments, the antigen-binding protein comprises one or more CDRs (e.g., 1, 2, or 3 CDRs) . In some embodiments, the antigen-binding protein comprises (a) a polypeptide structure and (b) one or more CDRs that are inserted into and/or joined to the polypeptide structure. The polypeptide structure can take a variety of different forms. For example, it can be, or comprise, the framework of a naturally occurring antibody, or fragment or variant thereof, or can be completely synthetic in nature.
In certain embodiments, the polypeptide structure of the antigen-binding protein thereof is an antibody or is derived from an antibody, including, but not limited to, monoclonal antibodies, bispecific antibodies, minibodies, domain antibodies, synthetic antibodies (sometimes referred to herein as “antibody mimetics” ) , chimeric antibodies, humanized antibodies, antibody fusions (sometimes referred to as “antibody conjugates” ) , and portions or fragments of each, respectively. In some instances, the antigen-binding protein is an immunological fragment of an antibody (e.g., a VHH) .
Variable regions of antigen-binding protein generally exhibit the same overall structure, comprising relatively conserved framework regions (FR) joined by three hypervariable regions, more often called “complementarity determining regions” or CDRs. The CDRs from the variable regions typically are aligned by the framework regions to form a structure that binds specifically with a specific epitope on the target protein (e.g., CD98HC) . From N-terminal to C-terminal, naturally occurring VH and VL both typically conform with the following order of these elements: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. A numbering system has been devised for assigning numbers to amino acids that occupy positions in each of these domains. This numbering system is defined in Kabat Sequences of Proteins of Immunological Interest (1987 and 1991, NIH, Bethesda, MD) , or Chothia & Lesk, 1987, J. MoL Biol. 196: 901-917; Chothia et al., 1989, Nature 342: 878-883.
In the present application, the antigen-binding protein may comprise a heavy chain variable region. The amino acid sequence of the heavy chain variable region is as set forth in SEQ ID NO: 92 or SEQ ID NO: 93. For example, the amino acid sequence of the heavy chain variable region is as set forth in any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94.
As an example, an antigen-binding protein can include a heavy chain or two heavy chains. In some embodiments, the antigen-binding protein comprises (and/or consists of) 1, 2, and/or 3 heavy CDRs from at least one of the sequences as set forth in any one of SEQ ID NO: 86, SEQ ID NO: 87, SEQ ID NO: 88, SEQ ID NO: 89, SEQ ID NO: 90, and SEQ ID NO: 91.
Examples of the CDRs of the antigen-binding protein are listed below (determined according to the IMGT method) in Table 1.
Table 1 (IMGT)
As described herein, the CD98HC antigen-binding protein can comprise a humanized antibody and/or part thereof. An important practical application of such a strategy is the “humanization” of the mouse humoral immune system. In certain embodiments, a humanized antibody is substantially non-immunogenic in humans. In certain embodiments, a humanized antibody has substantially the same affinity for a target as an antibody from another species from which the humanized antibody is derived.
In certain embodiments, amino acids of an antibody variable domain that can be modified without diminishing the native affinity of the antigen binding domain while reducing its immunogenicity are identified.
In certain embodiments, modification of an antibody by methods known in the art is typically designed to achieve increased binding affinity for a target and/or to reduce immunogenicity of the antibody in the recipient. In certain embodiments, humanized antibodies are modified to eliminate glycosylation sites in order to increase affinity of the antibody for its cognate antigen. See, e.g., Co et al., MoI. Immunol., 30: 1361-1367 (1993) . In certain embodiments, techniques such as “reshaping” , “hyperchimerization” or “veneering/resurfacing” are used to produce humanized antibodies. See, e.g., Vaswami et al., Annals of Allergy, Asthma, & Immunol. 81: 105 (1998) ; Roguska et al, Prot. Engineer., 9: 895-904 (1996) ; and U.S. Patent No. 6,072,035. In certain such embodiments, such techniques typically reduce antibody immunogenicity by reducing the number of foreign residues, but do not prevent anti-idiotypic and anti-allotypic responses following repeated administration of the antibodies.
In certain instances, humanizing antibodies results in a loss of antigen binding capacity. In certain embodiments, humanized antibodies are “back mutated” . In certain such embodiments, the humanized antibody is mutated to include one or more of the amino acid residues found in the donor antibody. See, e.g., Saldanha et ai, MoI Immunol 36: 709-19 (1999) .
In certain embodiments the complementarity determining regions (CDRs) of the VH of an antibody to CD98HC can be grafted to framework regions (FRs) from the same, or another, species. In certain embodiments, the CDRs of the VH of an antibody to CD98HC can be grafted to consensus human FRs. To create consensus human FRs, in certain embodiments, FRs from several human heavy chain or light chain amino acid sequences are aligned to identify a consensus amino acid sequence. In certain embodiments, the FRs of an antibody heavy chain or light chain are replaced with the FRs from a different heavy chain or light chain. In certain embodiments, rare amino acids in the FRs of the heavy of an antibody are not replaced, while the rest of the FR amino acids are replaced. Rare amino acids are specific amino acids that are in positions in which they are not usually found in FRs. In certain embodiments, the grafted variable regions from an antibody can be used with a constant region that is different from the constant region of an antibody.
As described herein, the antigen-binding protein that binds to CD98HC can comprise a human (i.e., fully human) antibody and/or part thereof. In certain embodiments, sequences corresponding to complementarity determining regions (CDRs) , specifically from CDR1 through CDR3, are provided.
Other antibodies that are provided are variants of the antigen-binding protein comprising variable heavy chains that each have at least 50%, 50%-60%, 60%-70%, 70%-80%, 80%-85%, 85%-90%, 90%-95%, 95%-97%, 97%-99%, or above 99%identity to the amino acid sequences of the sequences as set forth in any one of any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94 (either the entire sequence or a subpart of the sequence, e.g., one or more CDR) . In some embodiments, sequence comparison can be used in order to identify sections of the antibodies that can be modified by observing those variations that impact binding and those variations that do not appear to impact binding. For example, by comparing similar sequences, one can identify those sections (e.g., particular amino acids) that can be modified and how they can be modified while still retaining (or improving) the functionality of the antigen-binding protein. In some embodiments, variants of the antigen-binding protein include consensus groups and sequences between alternatives, as described above. The CDRs shown in Table 1 are defined based upon the IMGT method (based on sequence variability, see, e.g., Sequences of Proteins of Immunological Interest, Fifth Edition. NIH Publication No. 91-3242, Kabat et al., (1991) ) .
In certain embodiments, the antigen-binding protein comprises a heavy chain comprising a variable region comprising an amino acid sequence at least 90%identical to an amino acid sequence selected from at least one of the sequences of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94. In certain embodiments, the antibody or its antigen-binding fragment comprises a heavy chain comprising a variable region comprising an amino acid sequence at least 95%identical to an amino acid sequence selected from at least one of the sequences of any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94. In certain embodiments, the antibody or its antigen-binding fragment comprises a heavy chain comprising a variable region comprising an amino acid sequence at least 99%identical to an amino acid sequence selected from at least one of the sequences of any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94.
In some embodiments, the antigen-binding protein comprises a sequence that is at least 90%, 90-95%, and/or 95-99%identical to one or more CDRs from the CDRs in at least one of sequences of SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 46, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, SEQ ID NO: 51, SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 54, SEQ ID NO: 55, SEQ ID NO: 56, SEQ ID NO: 57, SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 61, SEQ ID NO: 62, and SEQ ID NO: 63. In some embodiments, 1, 2, or 3 CDR (each being at least 90%, 90-95%, and/or 95-99%identical to the above sequences) is present.
In light of the present disclosure, a skilled artisan will be able to determine suitable variants of the antigen-binding protein as set forth herein using well-known techniques. In certain embodiments, one skilled in the art can identify suitable areas of the molecule that may be changed without destroying activity by targeting regions not believed to be important for activity. In certain embodiments, one can identify residues and portions of the molecules that are conserved among similar polypeptides. In certain embodiments, even areas that can be important for biological activity or for structure can be subject to conservative amino acid substitutions without destroying the biological activity or without adversely affecting the polypeptide structure.
Additionally, one skilled in the art can review structure-function studies identifying residues in similar polypeptides that are important for activity or structure. In view of such a comparison, one can predict the importance of amino acid residues in a protein that correspond to amino acid residues which are important for activity or structure in similar proteins. One skilled in the art can opt for chemically similar amino acid substitutions for such predicted important amino acid residues.
One skilled in the art can also analyze the three-dimensional structure and amino acid sequence in relation to that structure in similar antibodies. In view of such information, one skilled in the art can predict the alignment of amino acid residues of an antibody with respect to its three-dimensional structure. In certain embodiments, one skilled in the art can choose not to make radical changes to amino acid residues predicted to be on the surface of the protein, since such residues can be involved in important interactions with other molecules. Moreover, one skilled in the art can generate test variants containing a single amino acid substitution at each desired amino acid residue. The variants can then be screened using activity assays known to those skilled in the art. Such variants can be used to gather information about suitable variants. For example, if one discovered that a change to a particular amino acid residue resulted in destroyed, undesirably reduced, or unsuitable activity, variants with such a change can be avoided. In other words, based on information gathered from such routine experiments, one skilled in the art can readily determine the amino acids where further substitutions should be avoided either alone or in combination with other mutations.
In certain embodiments, antigen-binding protein variants include glycosylation variants wherein the number and/or type of glycosylation site has been altered compared to the amino acid sequences of a parent polypeptide. In certain embodiments, protein variants comprise a greater or a lesser number of N-linked glycosylation sites than the native protein. An N-linked glycosylation site is characterized by the sequence: Asn-X-Ser or Asn-X-Thr, wherein the amino acid residue designated as X can be any amino acid residue except proline. The substitution of amino acid residues to create this sequence provides a potential new site for the addition of an N-linked carbohydrate chain. Alternatively, substitutions which eliminate this sequence will remove an existing N-linked carbohydrate chain. Also provided is a rearrangement of N-linked carbohydrate chains wherein one or more N-linked glycosylation sites (typically those that are naturally occurring) are eliminated and one or more new N-linked sites are created. Additional preferred antibody variants include cysteine variants wherein one or more cysteine residues are deleted from or substituted for another amino acid (e.g., serine) as compared to the parent amino acid sequence. Cysteine variants can be useful when antibodies must be refolded into a biologically active conformation such as after the isolation of insoluble inclusion bodies. Cysteine variants generally have fewer cysteine residues than the native protein, and typically have an even number to minimize interactions resulting from unpaired cysteines.
According to certain embodiments, amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and/or (4) confer or modify other physicochemical or functional properties on such polypeptides. According to certain embodiments, single or multiple amino acid substitutions (in certain embodiments, conservative amino acid substitutions) can be made in the naturally occurring sequence (in certain embodiments, in the portion of the polypeptide outside the domain (s) forming intermolecular contacts) . In certain embodiments, a conservative amino acid substitution typically may not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence or disrupt other types of secondary structure that characterizes the parent sequence) . Examples of art-recognized polypeptide secondary and tertiary structures are described in Proteins, Structures and Molecular Principles (Creighton, Ed., W.H. Freeman and Company, New York (1984) ) ; Introduction to Protein Structure (C. Branden & J. Tooze, eds., Garland Publishing, New York, N.Y. (1991) ) ; and Thornton et al, Nature, 354: 105 (1991) , which are each incorporated herein by reference.
In certain embodiments, the antigen-binding protein are produced by immunization with an antigen (e.g., CD98HC) . In certain embodiments, antibodies can be produced by immunization with full-length CD98HC, a soluble form of CD98HC, the extracellular domain alone, a splice variant form of CD98HC, or a fragment thereof. In certain embodiments, the antibodies of the present disclosure can be polyclonal or monoclonal, and/or can be recombinant antibodies.
Epitopes to which anti-CD98HC antibodies bind are provided. In some embodiments, epitopes that are bound by the presently disclosed antibodies are particularly useful. In some embodiments, the antigen-binding protein that binds to any of the epitopes that are bound by the antibodies described herein are useful. In some embodiments, the epitopes bound by any of the antibodies of the present application are especially useful. In some embodiments, the epitope is on the extracellular domain of CD98HC.
In the present application, the antigen-binding protein may capable of competing with a reference antibody for binding to the CD98HC, wherein the reference antibody comprises HCDR1-3, wherein the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 86, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 88, and the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 90.
In the present application, the antigen-binding protein may capable of competing with a reference antibody for binding to the CD98HC, wherein the reference antibody comprises HCDR1-3, wherein the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 87, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 89, and the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 91.
In the present application, the antigen-binding protein may capable of competing with a reference antibody for binding to the CD98HC, wherein the reference antibody comprises a VH, and the VH comprises an amino acid sequence as set forth in any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94.
In some cases, the antigen-binding protein may comprise HCDR3, and the HCDR3 may comprise an amino acid sequence as set forth in SEQ ID NO: 90 or SEQ ID NO: 91.
In some cases, the antigen-binding protein may comprise HCDR3, and the HCDR3 may comprise an amino acid sequence as set forth in SEQ ID NO: 58, SEQ ID NO: 59, SEQ ID NO: 60, SEQ ID NO: 62, or SEQ ID NO: 63.
In some cases, the antigen-binding protein may comprise HCDR2, and the HCDR2 may comprise an amino acid sequence as set forth in SEQ ID NO: 88 or SEQ ID NO: 89.
In some cases, the antigen-binding protein may comprise HCDR2, and the HCDR2 may comprise an amino acid sequence as set forth in SEQ ID NO: 52, SEQ ID NO: 53, SEQ ID NO: 55, SEQ ID NO: 56, or SEQ ID NO: 57.
In some cases, the antigen-binding protein may comprise HCDR1, and the HCDR1 may comprise an amino acid sequence as set forth in SEQ ID NO: 86 or SEQ ID NO: 87.
In some cases, the antigen-binding protein may comprise HCDR1, and the HCDR1 may comprise an amino acid sequence as set forth in SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, SEQ ID NO: 47, SEQ ID NO: 48, SEQ ID NO: 49, SEQ ID NO: 50, or SEQ ID NO: 51.
In some cases, the antigen-binding protein may comprise HCDR1, HCDR2 and HCDR3, the HCDR1 may comprise an amino acid sequence as set forth in SEQ ID NO: 86, the HCDR2 may comprise an amino acid sequence as set forth in SEQ ID NO: 88, and the HCDR3 may comprise an amino acid sequence as set forth in SEQ ID NO: 90.
In some cases, the antigen-binding protein may comprise HCDR1, HCDR2 and HCDR3, the HCDR1 may comprise an amino acid sequence as set forth in SEQ ID NO: 87, the HCDR2 may comprise an amino acid sequence as set forth in SEQ ID NO: 89, and the HCDR3 may comprise an amino acid sequence as set forth in SEQ ID NO: 91.
In some cases, the antigen-binding protein may comprise HCDR1, HCDR2 and HCDR3.
For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 43, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 52, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 58. For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 44, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 52, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 59. For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 45, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 53, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 60. For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 47, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 55, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 62. For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 48, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 55, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63. For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 49, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 56, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63. For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 50, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 55, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63. For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 50, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 57, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63. For example, the HCDR1 comprises an amino acid sequence as set forth in SEQ ID NO: 51, the HCDR2 comprises an amino acid sequence as set forth in SEQ ID NO: 55, the HCDR3 comprises an amino acid sequence as set forth in SEQ ID NO: 63.
In some cases, the antigen-binding protein may comprise a VH, the VH may comprise an amino acid sequence as set forth in SEQ ID NO: 92 or SEQ ID NO: 93. For example, the VH may comprise an amino acid sequence as set forth in any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94.
In some cases, the antigen-binding protein may comprise a VHH, the VHH may comprise an amino acid sequence as set forth in any one of SEQ ID NO: 92 or SEQ ID NO: 93. For example, the VH may comprise an amino acid sequence as set forth in any one of SEQ ID NO: 64, SEQ ID NO: 65, SEQ ID NO: 66, SEQ ID NO: 67, SEQ ID NO: 69, SEQ ID NO: 70, SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73, and SEQ ID NO: 94.
In some cases, the antigen-binding protein may comprise a Fc region. For example, the Fc region mat comprises a human IgG Fc region (such as a human IgG1, IgG2, or IgG4 constant region) . In some cases, the Fc region may comprise an amino acid sequence as set forth in SEQ ID NO: 83, SEQ ID NO: 84, SEQ ID NO: 85, SEQ ID NO: 96 or SEQ ID NO: 103.
The antigen-binding protein may also encompass a homologue or a variant thereof having substantially the same function/property thereto. In some cases, the homologue or variant may have an amino acid sequence different from that of the antigen-binding protein of the present disclosure by at least one amino acid. For example, the homologue or variant may be a polypeptide different from the antibody or the antigen-binding fragment thereof by an addition, deletion or substitution of one or more amino acid, such as 1-50, 1-40, 1-30, 1-20, 1-15, 1-14, 1-13, 1-12, 1-11, 1-10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, or 1-2 amino acids. In some cases, the homologue or variant may be a polypeptide having a sequence identity of at least 80%with the antibody or antigen-binding fragment thereof. For example, the homologue or variant may be a polypeptide having a sequence identity of 80% (e.g., at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%or higher) to the antigen-binding protein.
The term “percent (%) sequence identity, ” as used in the context of polypeptide sequences identified herein, generally refers to the percentage of amino acid residues or nucleotides in a query sequence that are identical with the amino acid residues or nucleotides of a second, reference polypeptide sequence or a portion thereof, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid/nucleotide sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, NEEDLE or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. Percent identity may be measured over the length of an entire defined polypeptide/polynucleotide sequence, or may be measured over a shorter length, for example, over the length of a fragment taken from a larger, defined polypeptide/polynucleotide sequence. It is understood that any fragment length supported by the sequences shown herein, in the tables, figures or Sequence Listing, may be used to describe a length over which percentage identity may be measured.
Pharmaceutical molecule
In another aspect, the present application provides a pharmaceutical molecule, which comprises the antigen-binding protein of the present disclosure.
The pharmaceutical molecule can exist in various forms. For example, a fusion protein, a polypeptide, a bispecific antibody, a protein conjugate, and so on. The pharmaceutical molecule can comprise the antigen-binding protein linked to an antibody, a protein, a compound, a DNA or RNA.
In the present application, the pharmaceutical molecule may comprise the antigen-binding fragment thereof, and a therapeutic entity.
In the present application, the antigen-binding protein may take the therapeutic entity across the BBB. For example, the therapeutic entity may comprise any therapeutic entity which needs to be taken across the BBB.
In the present application, the therapeutic entity may be linked to the antigen-binding protein directly or indirectly. For example, the therapeutic entity may be linked to the antigen-binding protein via a linker. For example, the linker may be a peptide linker. For example, the linker may be a flexible linker. For example, the linker may be a chemical linker.
In some embodiments, the fusion protein may comprise a CD98HC antibody heavy chain variable region, a Fc region and a therapeutic entity linked to a Fc region. For example, the CD98HC heavy chain may comprise a Fc-knob. For example, the Fc region which linked to the therapeutic entity may comprise a Fc-hole.
In some embodiments, the pharmaceutical molecule may comprise two chains, wherein the first chain comprises a CD98HC VHH, and a Fc-Knob, the second chain comprise a therapeutic entity-Hole. For example, the first chain comprises 2LP-8-Knob (SEQ ID NO: 74) , the second chain comprise therapeutic entity-Hole. For example, the first chain comprises 2LP-14-Knob (SEQ ID NO: 75) , the second chain comprise therapeutic entity-Hole. For example, the first chain comprises 4CP-60-Knob (SEQ ID NO: 77) , the second chain comprise therapeutic entity-Hole. For example, the first chain comprises 4CP-255-Knob (SEQ ID NO: 78) , the second chain comprise therapeutic entity-Hole. For example, the first chain comprises 4CP-292-Knob (SEQ ID NO: 79) , the second chain comprise therapeutic entity-Hole. For example, the first chain comprises Hu4CP-60-Knob (SEQ ID NO: 102) , the second chain comprise therapeutic entity-Hole.
Nucleic acid, Vector, Cell, Preparation method, and Composition
In another aspect, the present disclosure provides isolated nucleic acid or molecules, encoding for the antigen-binding protein, or the pharmaceutical molecule.
The isolated nucleic acids may comprise one or more nucleic acid molecules, with each encoding for at least a part of the antigen-binding protein. In some cases, the isolated nucleic acids may encode for a fusion protein, a polypeptide or a bispecific antibody.
The isolated nucleic acid or isolated nucleic acids may be synthesized using recombinant techniques well known in the art. For example, the isolated nucleic acid or isolated nucleic acids may be synthesized with an automated DNA synthesizer. Standard recombinant DNA and molecular cloning techniques include those described by Sambrook, J., Fritsch, E. F. and Maniatis, T. Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory Press: Cold Spring Harbor, (1989) (Maniatis) and by T.J. Silhavy, M.L. Bennan, and L.W. Enquist, Experiments with Gene Fusions, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1984) and by Ausubel, F.M. et al., Current Protocols in Molecular Biology, pub. by Greene Publishing Assoc. and Wiley-Interscience (1987) . Briefly, the subject nucleic acids may be prepared from genomic DNA fragments, cDNAs, and RNAs, all of which may be extracted directly from a cell or recombinantly produced by various amplification processes including but not limited to PCR and RT-PCR.
Direct chemical synthesis of nucleic acids typically involves sequential addition of 3’-blocked and 5’-blocked nucleotide monomers to the terminal 5’-hydroxyl group of a growing nucleotide polymer chain, wherein each addition is affected by nucleophilic attack of the terminal 5’-hydroxyl group of the growing chain on the 3’-position of the added monomer, which is typically a phosphorus derivative, such as a phosphotriester, phosphoramidite, or the like. See for example, Matteuci et al., Tet. Lett. 521: 719 (1980) ; U.S. Pat. No. 4,500,707 to Caruthers et al.; and U.S. Pat. Nos. 5,436,327 and 5,700,637 to Southern et al.
In another aspect, the present disclosure provides a vector or vectors, comprising the isolated nucleic acid molecule or molecules.
The vector may be any linear nucleic acids, plasmids, phagemids, cosmids, RNA vectors, viral vectors and the like. Non-limiting examples of a viral vector may include a retrovirus, an adenovirus and an adeno-associated virus. In some cases, the vector is an expression vector, e.g., a phage display vector.
An expression vector may be suitable for use in particular types of host cells and not others. For example, the expression vector can be introduced into the host organism, which is then monitored for viability and expression of any genes/polynucleotides contained in the vector.
The expression vector may also contain one or more selectable marker genes that, upon expression, confer one or more phenotypic traits useful for selecting or otherwise identifying host cells that carry the expression vector. Non-limiting examples of suitable selectable markers for eukaryotic cells include dihydrofolate reductase and neomycin resistance.
The subject vectors can be introduced into a host cell stably or transiently by a variety of established techniques. For example, one method involves a calcium chloride treatment wherein the expression vector is introduced via a calcium precipitate. Other salts, for example calcium phosphate, may also be used following a similar procedure. In addition, electroporation (that is, the application of current to increase the permeability of cells to nucleic acids) may be used. Other examples of transformation methods include microinjection, DEAE dextran mediated transformation, and heat shock in the presence of lithium acetate. Lipid complexes, liposomes, and dendrimers may also be employed to transfect the host cells.
In another aspect, the present disclosure provides a cell (e.g., an isolated cell, such as a host cell) , comprising the isolated nucleic acid molecule or molecules of the present disclosure or the vector or vectors of the present disclosure.
The cell may express the antigen-binding protein of the present disclosure, or the pharmaceutical molecule of the present disclosure. The cell may be a eukaryotic cell or a prokaryotic cell. An appropriate cell may be transformed or transfected with the nucleic acid (s) or vector (s) of the present disclosure and utilized for the expression and/or secretion of the antibody, the antigen-binding fragment thereof, or the fusion protein. For example, the cell may be E. coli cells, other bacterial host cells, yeast cells, or various higher eukaryotic cells.
In another aspect, the present disclosure provides a method for producing the antigen-binding fragment, or the pharmaceutical molecule of the present disclosure, comprising culturing the cell of the present disclosure under conditions enabling expression of the antigen-binding protein, or the pharmaceutical molecule.
The method optionally may further comprise harvesting the antigen-binding protein, or the pharmaceutical molecule of the present disclosure.
In another aspect, the present disclosure provides a pharmaceutical composition, comprising the antigen-binding protein, the pharmaceutical molecule, the isolated nucleic acid molecule or molecules, the vector or vectors, and/or the cell of the present disclosure, and optionally a pharmaceutically acceptable excipient.
In some cases, the pharmaceutically acceptable excipient may comprise a buffer. In some cases, the pharmaceutically acceptable excipient may comprise an amino acid.
In some embodiments, the pH of the pharmaceutical composition may be 1-13, for example, the pH may be about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13.
In some cases, the pharmaceutical composition may further comprise an effective amount of an additional therapeutically active component, for example, an additional therapeutically active component for treating a disease or a disorder. Each of the active components may be present in the pharmaceutical composition in a pharmaceutically active amount. In the composition, the antibody, the fragment thereof of the present application may or may not be associated with the additional active component.
Described below are non-limiting exemplary pharmaceutical compositions and methods for preparing the same. The pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages. In some cases, the pharmaceutical composition may be a liquid pharmaceutical composition.
Pharmaceutical compositions of the disclosure can be presented as discrete dosage forms, with each dosage containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or non-aqueous liquid. Such dosage forms can be prepared by any of the methods known to a skilled person, for example, it may include the step of bringing the active ingredient into association with the carrier, which constitutes one or more other ingredients. In general, the compositions are prepared by uniformly and intimately mixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation.
The antigen-binding protein, or the pharmaceutical molecule of the present disclosure can be combined in an intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending on the form of preparation desired for administration.
The pharmaceutical composition can further include one or more pharmaceutically acceptable additives and excipients. Such additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and/or mixtures thereof.
The pharmaceutical compositions of the present disclosure may comprise a therapeutically effective amount of the active agent (e.g., the antibody, the antigen-binding fragment thereof, or the fusion protein of the present disclosure) . A therapeutically effective amount is an amount of the subject pharmaceutical composition capable of preventing and/or curing (at least partially) a condition or disorder and/or any complications thereof in a subject suffering from or having a risk of developing said condition or disorder. The specific amount/concentration of the active agent comprised may vary according to the method of administration and the need of a patient, and can be determined based on e.g., volume, viscosity, and/or body weight of a patient etc. It shall be understood that these specific doses may be conveniently adjusted by a skilled person in the art (e.g., a doctor or a pharmacist) based on conditions of a specific patient, formulation, and/or disease.
Medical Use and Methods of Treatment
In another aspect, the present application provides a method of taking a therapeutic entity across BBB, comprising using the antigen-binding protein of the present disclosure.
In another aspect, the present application provides a use of the antigen-binding protein of the present disclosure.
In another aspect, the present application provides a use of the antigen-binding protein, the pharmaceutical molecule, the isolated nucleic acid molecule or molecules, the vector or vectors, the cell, and/or the pharmaceutical composition of the present disclosure in the manufacture of a medicament for diagnosing, preventing and/or treating a disease or disorder.
In another aspect, the present application provides a method for diagnosing, preventing and/or treating a disease or disorder, comprising administering the antigen-binding protein, the pharmaceutical molecule, the isolated nucleic acid molecule or molecules, the vector or vectors, the cell, and/or the pharmaceutical composition to a subject in need thereof.
In another aspect, the present application provides the antigen-binding protein, the pharmaceutical molecule, the isolated nucleic acid molecule or molecules, the vector or vectors, the cell, and/or the pharmaceutical composition for use in diagnosing, preventing and/or treating a disease or disorder.
In some embodiments, the disease or disorder is determined by the kind of the therapeutic entity which the antibody or the antigen-binding fragment linked to.
For example, the fusion protein comprising the antibody or the antigen-binding fragment thereof may be used for diagnosing, preventing and/or treating a disease.
In some embodiments, the disease is a CNS disease (central nervous system disease) . In some embodiments, the CNS disease may comprise brain tumor.
Examples
The following examples are set forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g. amounts, temperature, etc. ) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Celsius, and pressure is at or near atmospheric. Standard abbreviations may be used, e.g., bp, base pair (s) ; kb, kilobase (s) ; pl, picoliter (s) ; s or sec, second (s) ; min, minute (s) ; h or hr, hour (s) ; aa, amino acid (s) ; nt, nucleotide (s) ; i.m., intramuscular (ly) ; i.p., intraperitoneal (ly) ; s.c., subcutaneous (ly) ; and the like.
Example 1 CD98HC-ECD recombinant protein preparation
To construct the expression plasmid, the gene of human CD98HC extracellular domain (ECD) consisting of the amino acid (Aa) 206 –630 (SEQ ID NO: 1) , cynomolgus monkey CD98HC ECD consisting of Aa 206 -630 (SEQ ID NO: 2) , mouse CD98HC ECD consisting of Aa 100 -526 (SEQ ID NO: 3) and rat CD98HC ECD consisting of Aa 100-526 (SEQ ID NO: 4) were synthesized and subcloned into pcDNA3.4 vector with N-terminus fused to an 8 x his tag. Subsequently, the plasmid was transiently transfected into 293F or CHO-K1 cells and cultured for 5-7 days in a CO2 incubator. Following incubation, the supernatant was harvested and purified through one-step immobilized metal affinity chromatography. The purified protein was used for immunization and antibody screening.
Example 2 Generation of domain antibodies
To generate domain antibodies, two healthy alpacas were immunized with recombinant human CD98HC ECD and mouse CD98HC ECD protein. In brief, human and mouse CD98HC ECD proteins were mixed with GERBU/Adjuvant Fama (Gerbu, 3030) in 1: 1 ratio to create a stable oil-in-water emulsion. Subsequently, the emulsion was injected into the alpaca via subcutaneous routes for 4 times.
Following immunization, serum was collected and PBMC was isolated through Ficoll-Paque PLUS density gradient media (GE, 17-1440-03) . Total cellular RNA was extracted using Trizol (Life Tech, 15596018) according to the manufacturer’s instructions. For the generation of cDNA templates, purified RNA was reverse transcribed using PrimeScriptTMII Reverse Transcriptase Kit (Takara, 2690A) . 450 bp VHH sequences were amplified from cDNA through two round nested PCR and ligated to phagemid plasmid. The ligation product was transformed into E. coil SS320 and generated domain antibody phage libraries. The library size was estimated at 3 -5 x 109 p. f. u with higher than 90%clones in frame through sequencing 48 randomly picked single clones.
The phage library was used to screen domain antibodies that bind with human CD98HC and mouse CD98HC. In brief, human CD98HC ECD or mouse CD98HC ECD protein were either directly coated to immune tubes (Thermo fisher, 341886) or captured by streptavidin beads (Thermo, 11206D) after biotinylation. The phage library was incubated with coated proteins overnight at 4℃ followed by 15 ~ 20 times washing by PBST (0.05%Tween20 in PBS, pH7.4) . The binder was eluted using Trypsin-EDTA (0.25%) from immune tubes or streptavidin beads and infected SS320 for the next round of screening. This process was performed for 3 ~4 rounds.
Individual phage clones from rounds 2-4 were prepared and tested for their binding activity to human and mouse CD98HC ECD protein. Briefly, the single clone was cultured in LB medium to log phase at 37℃. Then, IPTG was added to a final concentration of 0.5 mM and cultured overnight at 30 ℃. The collected cell pellet was lysed by lysis buffer (50 mM Tris-HCl pH 7.4; 150 mM NaCl; 1 mM PMSF, 1 mM EDTA, 5 μg/ml Aprotinin, 5 μg/ml Leupeptin, 1%Triton x-100, 1%Sodium deoxycholate, 0.1%SDS) . 100 μl cell lysate was applied to microplate coated with human or mouse CD98HC ECD protein and incubated for 1 h at room temperature. The binding signal was detected by an anti-flag HRP (Sigma, A8592) antibody. Finally, 9 unique clones were selected for further Fc-fusion expression and confirmation.
Example 3 Expression and purification of recombinant Fc fusion VHH
For transient transfection of HEK293 cells to express recombinant Fc fusion VHH (VHH-Fc) , the VHH were subcloned into the pCDNA3.4 vector along with human IgG1 Fc (SEQ ID NO: 83) . The subcloned constructs were verified by DNA sequencing. A total of 30 μg of plasmids were diluted in 1 ml ofI Reduced Serum Medium (GIBCO, 31985-070) and transfected into human 293F cells using 20 μg of PEI. The cells were cultured in serum-free medium (Shanghai opmbiosciences, OPM-293CD03) at 5%CO2, 37℃, and 125 rpm/min. The culture medium was replenished on day 1 and cultured for another 4~7 days.
The VHH-Fc were purified using Protein A MagBeads (GenScript, L00273) according to the manufacturer's instructions. Briefly, the clarified supernatant was incubated with the MagBeads at room temperature with gentle rocking for 2 hours. Subsequently, the MagBeads were washed three times with PBS buffer. The purified antibodies were eluted from the beads using Elution Buffer (0.1 M glycine, pH 3.0) and neutralized using Neutralization Buffer (1 M Tris, pH 8.5) . Finally, the buffer was exchanged to 1 x PBS.
The variable region sequences of VHH are as follows.
>2LP-37
>2LP-8
>2LP-62
>2LP-14
>3LP-89
>4CP-56
>4CP-60
>4CP-255
>4CP-292
Example 4 Determining the affinity of the antigen-binding protein to human and cyno CD98HC ECD
To evaluate the affinity of VHH-Fc with human and cyno CD98HC, we employed the bio-layer interferometry (BLI) method using the Octet Red384 instrument. Briefly, Anti-human Fc antibody-coated biosensor AHC tips (ForteBio, 18-5060) were firstly pre-wet in an assay buffer consisting of PBS with 0.1%w/v bovine serum albumin and 0.05%Tween-20 (assay buffer) for at least 10 minutes in the pre-wetting plate. The purified antibodies, at a concentration of 100 nM in assay buffer, were then immobilized onto AHC biosensors to achieve capture levels of approximately 1 nm.
A series of 2-fold dilutions (ranging from 3.125 nM to 200 nM) of the human or cyno CD98HC ECD solution was used to determine the association and dissociation rates. All measurements were referenced against a control sensor exposed solely to the assay buffer to correct for baseline drift. By applying a 1: 1 interaction model (fitting local, full) through the ForteBio data analysis software, association rate constants (kon) , dissociation rate constants (koff) , and KD values for each antibody were calculated. The binding affinities of the antigen-binding protein to human and cyno CD98HC can be found in Table 2 and 3.
Table 2 The binding affinities of the antigen-binding protein to human CD98HC ECD
Table 3 The binding affinities of the antigen-binding protein to cyno CD98HC ECD
Example 5 The binding activity of VHH-Fc to mouse and rat CD98HC ECD
The binding activity of VHH-Fc to mouse and rat CD98HC ECD proteins was tested by ELISA. In brief, mouse and rat CD98HC ECD proteins at 2 μg/ml were coated to 96 well micro-plate (Corning, 9018) . 200 nM Fc fusion VHHs were incubated with the plate at room temperature for 1 hour. After washing, the binding signal was detected by goat pAb to human IgG (Abcam, ab97225) . The raw data was processed and analyzed by the software GraphPad prism 9 (FIG. 1 and FIG. 2) .
Example 6 The binding activity of VHH-Fc to human and cyno CD98HC overexpression cells
The binding activity of VHH-Fc to human and cyno CD98HC overexpression cells was tested by FACS. Briefly, human and cyno CD98HC overexpression cells were collected and washed once with PBS at room temperature. Then, cells were resuspended in pre-chilled PBS and seeded 200,000 cells/well in a 96-well V-bottom plate (200 μL/well) . Series diluted VHH-Fc were incubated at 4℃for 1 hour. After washing cells with pre-chilled PBS (250 μL/well) three times, secondary antibody Goat Anti-Human IgG-PE (SouthernBiotech, Cat#2040-09) was added and incubated at 4℃ in the dark for 30 minutes followed three times washing with pre-chilled PBS (250 μL/well) . Cells were resuspended in pre-chilled 2%BSA-PBS (150 μL/well) and analyzed using a flow cytometer. The raw data were processed and analyzed using GraphPad Prism9 software (FIG. 3 and FIG. 4) .
Example 7: Evaluation of neurotensin-CD98HC VHH fusion proteins induced hypothermia effect in wild-type mice
To demonstrate the capability of VHH crossing BBB, neurotensin (8-13) -ashort peptide that could not cross BBB but can induce the hypothermia effect when entered into the brain, was fused to VHH and produced neurotensin-CD98HC VHH fusion proteins. The Neurotensin-CD98HC VHH fusion proteins induced hypothermia effect was evaluated in wild-type mice. In brief, the animals were housed in a controlled environment within a barrier system, adhering to the animal facility's Standard Operating Procedures (SOP) .
To generate Neurotensin-VHH fusion proteins, the variable region of VHH was combined with human IgG4 Fc with mutation S228P, F234A, L235A, F296Y, R409K, and K439E (SEQ ID NO: 84, Eu numbering) . This resulted in the production of the heavy chain 1. Furthermore, the neurotensin (8-13) (SEQ ID NO: 82) peptide with the linker (SEQ ID NO: 81) was fused with the C terminal of human IgG4 Fc with mutation S228P, F234A, L235A, F296Y, E356K, R409K and H435R (SEQ ID NO: 85, Eu numbering) , resulting in the production of the heavy chain 2. The two heavy chains were co-expressed in the HEK293 or CHO-K1 cells and purified through Protein A chromatography followed by Size Exclusion Chromatography or Ion Exchange Chromatography, depending on the protein purity (Table 4) . This process allowed us to obtain neurotensin-CD98HC VHH fusion proteins.
Table 4. The composition of neurotensin-CD98HC VHH fusion proteins
The fusion proteins were diluted to the desired experimental concentrations using 1X PBS and administered intravenously at the dose of 100-500 nmol/kg. The core temperature of animals was measured using an anus thermometer (Lab Animal Technology Develop Co., LAT-212) . Briefly, the animal was fixed, and the metal part of the anal thermometer probe was lubricated by petroleum jelly and submerged into the anus. The reading body temperature was recorded when the reading was stable and lasted more than 5 seconds. The basal core temperature was measured at 15-30 min before administration. After administration, the core temperature was monitored at 3 and 6 hours respectively. As shown in FIG. 5, 2LP-8-NT、4CP-60-NT、4CP-255-NT and 4CP-292-NT induced strong hypothermia effect and 2LP-14-NT induced weak hypothermia effect in wild-type mice. Meanwhile, 2LP-28-NT did not induce hypothermia effect.
Example 8 The impact of CD98HC VHHs on Amino Acid Transport of endothelial cells
To assess the effect of CD98HC VHHs on amino acid transport in cells, human CD98 overexpressed CHO-K1 cells were plated at a density of 100,000 cells/ml in a 96-deep well plate, with 500 μL per well. Next, the cells were washed three times with pre-warmed HBSS. Subsequently, 200 nM CD98HC VHHs and 10 mM 2-Amino-2-norbornanecarboxylic acid (BCH) (MCE, Catalog No. HY-108540) diluted with pre-warmed HBSS were added to the wells and incubated at 37℃/5%CO2 for 30 minutes. After the incubation, the supernatant was replaced with 500 μL of pre-warmed Boronophenylalanine (BPA) uptake solution (Tongren Chemical, Catalog No. UP04) and the cells were incubated at 37 ℃/5%CO2 for 5 minutes, followed by three washes with pre-warmed HBSS. Subsequently, 500 μL of pre-warmed working solution was added to each well, and the cells were incubated at 37℃/5%CO2 for 5 minutes. After the incubation, the fluorescence intensity was directly measured using flow cytometry (Ex/Em=405/450 nm) .
As shown in Fig. 6, compared with BCH, which significantly interferes with the amino acid uptake, two anti-CD98HC VHHs, 2LP-8 and 4CP-60, do not interfere with cells' normal amino acid transportation function.
Example 9 Humanization of CD98HC VHH
4CP-60 was chosen for humanization due to its high brain penetration efficacy. The humanization of VHH was performed using a CDR grafting approach. In brief, germline genes were selected based on the homology between the parental domain antibody and human germline V and J genes. Human heavy chain germline genes IGHV3-2304 and IGHJ401 were selected as framework region (FR) donors to humanize 4CP-60. The framework of the domain antibody was then replaced with those of the selected human germline V and J genes, resulting in the generation of the humanized domain antibody Hu4CP-60.
Hu4CP-60
The corresponding encoding nucleic acid sequences of humanized domain antibody were synthesized and subcloned into a pCDNA3.4 vector. All recombinant antibodies were expressed and purified as described in Example 3. The affinity of the humanized antibody was determined as described in Example 4. As shown in Table 5, the affinity of humanized Fc fusion Hu4CP-60 is similar to chimeric Fc fusion 4CP-60.
Table 5 The binding affinities of the humanized CD98HC VHH
Example 10 Evaluation of neurotensin-humanized CD98HC VHH fusion proteins induced hypothermia effect in CD98HC KI mice
To further demonstrate the capability of Hu4CP-60 to deliver a functional peptide into the mouse brain. Neurotensin (8-13) was fused with Hu4CP-60 to produce neurotensin-humanized CD98HC VHH fusion protein. The neurotensin-humanized CD98HC VHH fusion protein-induced hypothermia effect was evaluated in human CD98HC KI mice.
Neurotensin-humanized CD98HC VHH fusion proteins were generated as described in Example 7.
Briefly, the variable region of Hu4CP-60 was combined with human IgG4 Fc with mutation S228P, F234A, L235A, F296Y, R409K, and K439E (SEQ ID NO: 84, Eu numbering) . This resulted in the production of Hu4CP-60-Hole (SEQ ID NO: 95) . Furthermore, the neurotensin (8-13) (SEQ ID NO: 82) peptide with the linker (SEQ ID NO: 81) was fused with the C terminal of human IgG4 Fc with mutation S228P, F234A, L235A, F296Y, E356K, R409K and H435R (SEQ ID NO: 85, Eu numbering) , resulting in the production of NT-Hole (SEQ ID NO: 80) . Two chains were co-expressed in the HEK293 or CHO-K1 cells and purified through Protein A chromatography followed by Size Exclusion Chromatography or Ion Exchange Chromatography, depending on the protein purity. This process generated neurotensin-humanized CD98HC VHH fusion protein Hu4CP-60-NT. An isotype control-NT (A613-NbHuL6) was similarly prepared using an anti-human lysozyme VHH (Vincke C, Loris R, Saerens D, Martinez-Rodriguez S, Muyldermans S, Conrath K. General strategy to humanize a camelid single-domain antibody and identification of a universal humanized nanobody scaffold. J Biol Chem. 2009 Jan 30; 284 (5) : 3273-3284) through the same method.
>Hu4CP-60-Hole (SEQ ID NO: 95)
>NT-Hole (SEQ ID NO: 80)
The hypothermia effect of neurotensin-VHH fusion proteins was measured as described in Example 7. As shown in Figure 7, PBS and isotype control-NT cannot induce a decrease in the body temperature in mice. In contrast, Hu4CP-60-NT significantly decreased the body temperature in mice in a concentration-dependent manner.
Example 11 The PK study of IgG-humanized CD98HC VHH fusion protein in human CD98HC knock-in mice
To demonstrate the capability of humanized CD98HC VHH to deliver large molecules, such as IgG, into the mouse brain. An anti-trinitrophenol (TNP) antibody was fused with Hu4CP-60 to produce IgG-humanized VHH fusion proteins. The PK of the IgG-humanized VHH fusion proteins was evaluated in human CD98HC KI mice.
Fusion protein preparation: Two IgG-VHH fusion proteins were prepared. To generate the first IgG-VHH fusion protein, Anti-TNP-VHH-01, the variable region of Hu4CP-60 was combined with human IgG1 Fc with mutation C230A, T366S, T368A and V407Y (SEQ ID NO: 96, Eu numbering) . This resulted in the production of Hu4CP-60-Knob (SEQ ID NO: 102) . Furthermore, the variable region of anti-TNP was fused with human IgG4 CH1, hinge, and CH2-CH3 region with mutation T366W (SEQ ID NO: 97, Eu numbering) , resulting in the production of Anti-TNP-Hole (SEQ ID NO: 98) . The two heavy chains, along with the associated anti-CD98 light chain, were co-expressed in the HEK293 or CHO-K1 cells and purified through Protein A chromatography followed by Size Exclusion Chromatography or Ion Exchange Chromatography, depending on the protein purity (Table 4) . To generate the second IgG-fusion protein, Anti-TNP-VHH-02, the variable region of Hu4CP-60 was combined with the light chain of Anti-TNP (SEQ ID NO: 99) and co-expressed with Anti-TNP heavy chain (SEQ ID NO: 100) in the HEK293 or CHO-K1 cells and purified as described above.
>Hu4CP-60-Knob (SEQ ID NO: 102)
>Anti-TNP-Hole (SEQ ID NO: 98)
>Anti-TNP light chain (SEQ ID NO: 99)
>Anti-TNP heavy chain (SEQ ID NO: 100)
>Anti-TNP light chain-Hu4CP-60 (SEQ ID NO: 101)
The PK study: The animals were housed in a controlled environment within a barrier system, adhering to the animal facility's Standard Operating Procedures (SOP) . The antibodies were diluted to the desired experimental concentrations using 1X PBS and administered intravenously at a 100 nmol/kg dosage, 3 or 4 animals included in each group. At 72 hours after administration, the animals were euthanized according to the experimental design and subsequently perfused with physiological saline. Blood and tissue samples were collected based on the experimental design.
To prepare the brain homogenate, the collected brain tissues were rinsed with ice-cold saline first and then mixed with lysis buffer (NP-40 (Beyotime, P0013F) containing 1x Protease Inhibitor Cocktail (Beyotime, P1006-1) ) at a ratio of 1 mg brain tissue to 5 μL lysis buffer. Next, the mixture was homogenized using a tissue homogenizer, followed by centrifugation at 14,000 rpm, 4℃ for 20 minutes to obtain brain homogenate.
To isolate the brain parenchyma and microvessels, mouse brains were homogenized using a Tissue Homogenizer. Briefly, the brain tissue was mixed with Homogenization Buffer A (containing 10 mM HEPES, 141 mM NaCl, 4 mM KCl, 2.8 mM CaCl2, 1 mM MgSO4·7H2O, 1 mM NaH2PO4·7H2O, and 10 mM D-glucose) at a 1: 3 ratio (brain tissue weight: buffer volume) and homogenized for 8-10 rotations. Next, Homogenization Buffer B (Homogenization Buffer A supplemented with 26%Dextran) was added at a 1: 4 ratio (brain tissue weight: buffer volume) , followed by 3 additional rotations. After centrifugation at 5200 x g for 15 minutes, the mixture separated into the supernatant and the pellet. Triton X-100 at a final concentration of 1%and PMSF at a final concentration of 1 mM were added to the supernatant and centrifuged at 11,000 rpm for 15 minutes to obtain solution with brain parenchyma. The pellet was resuspended in a solution with 5 ml of Homogenization Buffer A, 5 ml of Homogenization Buffer B, and 100 μL of Triton X-100 and centrifuged at 11,000 rpm for 15 minutes to obtain solution with brain microvessels.
To measure the concentration of antibodies in the brain homogenate, antibodies were captured using Anti-Human IgG (Fab specific) (Sigma, I5260) and detected with Goat Anti-Human IgG Fc (Biotin) preadsorbed (Abcam, ab98618) and Streptavidin HRP (BD, 554066) .
As shown in Figure 8, the brain concentration of Anti-TNP-VHH-01 is 4.9 times, and Anti-TNP-VHH-02 is 4.4 times higher than that of anti-TNP at 72 hours after administration. Meanwhile, Anti-TNP-VHH-01 enriched on parenchyma (Figure 9) and microvessel (Figure 10) .
While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. It is not intended that the invention be limited by the specific examples provided within the specification. While the invention has been described with reference to the aforementioned specification, the descriptions and illustrations of the embodiments herein are not meant to be construed in a limiting sense. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. Furthermore, it shall be understood that all aspects of the invention are not limited to the specific depictions, configurations or relative proportions set forth herein which depend upon a variety of conditions and variables. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is therefore contemplated that the invention shall also cover any such alternatives, modifications, variations or equivalents. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.