The present disclosure generally relates to the technical field of biologic therapeutics, and more particularly relates to making and using bispecific antibodies. All references are incorporated herein by its entirety.
Cancer cells develop various strategies to evade immunosurveillance. Absence of specific tumor antigens and loss of expression of major histocompatibility complex (MHC) molecules hinder the recognition of cancer cells by T lymphocytes. Immunosuppressive tumor microenvironment also contributes to the reduced recognition of tumor cells by the immune system. The tumor microenvironment is presented by immunosuppressive cellular populations composed of regulatory T cells, myeloid derived suppressor cells, tumor associated macrophages, suppressive B cells, immunosuppressive cytokines produced by tumor or stroma cells such as TGF-beta or IL-10, and immune checkpoint molecules that regulate T cell function [Marshall H T et al., Front Oncol 2018, 8:315].
Engaging a patient own immune system has been shown to be effective at controlling tumor growth and specific elimination of tumor cells while leaving normal tissue intact. Immunotherapy has provided an additional angle to treating cancer [Khalil D N et al., Adv Cancer Res 2015, 128:1-68].
Combining multiple modulators of the immune system is a new rapidly developing area of the immuno-oncology field. New therapeutic agents that can modulate immune response to tumor cells via multiple pathways can be greatly beneficial for cancer patient by increasing the patient response rate and in some cases decreasing toxicity.
Combination therapy with more than one monoclonal antibody targeting the immune system have been shown to be more efficacious in the treatment of cancer than treatments with single agents [Hellman M D et al., Adv Immunol 2016, 130: 251-77]. In addition to the increased efficacy and response rate, the combination therapy often has greater toxicity than a single agent treatment. Bispecific agents that modulate the immune system can be less toxic to patients and/or more potent, have additional mechanisms of action than treatments comprised of a combination of monoclonal antibodies with identical specificities.
The current disclosure relates to the bispecific antibodies, specifically, the bispecific antibodies that contain an IgG component therefore overcome fast clearance of BiTE molecule, having an advantage over CAR-T cell therapy as an off-the-shelf therapy that does not require ex vivo expansion of patients' immune cells. Another advantage of the bispecific antibodies is the enhanced ability to overcome suppressive tumor microenvironment by simultaneous engagement of two checkpoint receptors.
The bispecific antibodies in the current disclosure can be combined with other agents, for instance T-cell engagers, and further enhance their activity.
The present disclosure relates to bispecific antibodies that bind to two distinct targets expressed on immune and tumor cells. Both targets may be checkpoint antigens. In one embodiment, both targets may be checkpoint antigens on immune cells. In one embodiment, both targets may be checkpoint antigens on tumor cells. In one embodiment, one target is a checkpoint antigen on immune cells and another target is a checkpoint antigen on tumor cells. In one embodiment, the checkpoint antigen may be selected from PD-1, PD-L1 and CTLA4. In one embodiment, the targets may include any combination of PD-1, PD-L1 and CTLA4.
The disclosure further provides the composition of the bispecific agents and their therapeutic use for treatment of cancer and autoimmune deficiencies.
In one embodiment, the application discloses a bispecific antibody comprising IgG heavy chains and light chains, and two scFv components being connected to either C terminal of the heavy chains or N terminal of the light chains, wherein the IgG has the binding specificity to a first antigen, wherein the scFv components have the binding specificity to a second antigen, and wherein the first antigen and the second antigen are different and are independently selected from α-CTLA4, α-PD-1, and α-PD-L1.
In one embodiment, the bispecific antibody has the two scFv components connected to the C terminal of the heavy chain. In one embodiment, the first antigen comprises α-CTLA4 and the second antigen comprises α-PD-1 or α-PD-L1. In another embodiment, the first antigen comprises α-PD-1 or α-PD-L1 and the second antigen comprises α-CTLA4.
In one embodiment, the bispecific antibody has the two scFv components connected to the N terminal of the light chain. In one embodiment, the first antigen comprises α-PD-1 or α-PD-L1 and the second antigen comprises α-CTLA4. In another embodiment, the first antigen comprises α-CTLA4 and the second antigen comprises α-PD-1 or α-PD-L1.
In one embodiment, the bispecific antibody is an isolated monoclonal antibody.
In one embodiment, the bispecific antibody comprises an antigenic peptide sequence having a sequence as disclosed herein. In one embodiment, the bispecific antibody may have an antigenic peptide sequence having at least 70%, 80%, 90%, 95%, 98%, or 99% similarity with the disclosed amino acid sequences.
In one embodiment, the bispecific antibody comprises an antigen-binding fragment having a sequence as disclosed herein. In one embodiment, the bispecific antibody may have an antigen-binding fragment having a sequence with at least 70%, 80%, 90%, 98%, or 99% similarity with the disclosed antibody sequences.
In one embodiment, the bispecific antibody may have a binding affinity to α-CTLA4, α-PD-1 or α-PD-L1 with a Kd not greater than 70 nM, 50 nM, 40 nM, 30 nM, 20 nM, 10 nM, or 5 nM.
In one embodiment, the bispecific antibody may have a binding affinity to α-CTLA4 and α-PD-1 with a Kd not greater than 70 nM, 50 nM, 40 nM, 30 nM, 20 nM, 10 nM, or 5 nM.
In one embodiment, the bispecific antibody may have a binding affinity to α-CTLA4 and α-PD-L1 with a Kd not greater than 70 nM, 50 nM, 40 nM, 30 nM, 20 nM, 10 nM, or 5 nM.
In one embodiment, the bispecific antibody may have a binding affinity to two of α-CTLA4, α-PD-1, or α-PD-L1 with a Kd not greater than 70 nM, 50 nM, 40 nM, 30 nM, 20 nM, 10 nM, or 5 nM.
In one embodiment, the bispecific antibody may exhibit one or more functional properties selected from high affinity binding to α-CTLA4, α-PD-1, or α-PD-L1, inhibiting binding of PD-L1 to PD-1, enhancing T cell activation, the ability to stimulate antibody responses and/or the ability to reverse the suppressive function of immunosuppressive cells, such as T regulatory cells.
In one embodiment, enhancing T-cell activation comprises T-cell proliferation, IFN-γ and/or IL-2 secretion, or a combination thereof.
In one embodiment, the bispecific antibody comprising a human framework region.
In one embodiment, the bispecific antibody may be a humanized antibody, a chimeric antibody, or a recombinant antibody.
In one embodiment, the bispecific antibody comprises an IgG1 constant region to extend the circulating half-life of the bispecific molecules. In one embodiment, the IgG1 constant region of the bispecific antibody comprises an amino acid sequence having at least 98% similarity with SEQ ID No. 136.
In one embodiment, the application discloses an isolated bispecific antibody selected from the group consisting of those clones described or having the described characteristics as disclosed herein.
In one embodiment, the application discloses an IgG1 heavy chains for the bispecific antibody, comprising an amino acid sequence selected from sequences as disclosed herein. In one embodiment, the IgG1 heavy chains may have an amino acid sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with SEQ ID NO. 02, 06, 08, 10, 12, 14, 16, 18, 20, 22, 26, 30, 34, 38, 42, 46, 50, 54, 58, 62, 72, 92, 96, 100, 104, 108, 112, 116, 120, 124, 128, or 132.
In one embodiment, the application discloses a kappa light chain for the bispecific antibody. In one embodiment, the kappa light chain may have an amino acid sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with SEQ ID NO. 04, 28, 32, 36, 40, 44, 48, 52, 56, 60, and 64.
In one embodiment, the application discloses a variable light chain for the bispecific antibody, comprising an amino acid sequence as disclosed herein. In one embodiment, the variable light chain may have an amino acid sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with SEQ ID NO. 94, 98, 102, 106, 110, 114, 118, 122, 126, 130, or 134.
In one embodiment, the application discloses a variable heavy chain for the bispecific antibody, comprising an amino acid sequence as disclosed herein. In one embodiment, the variable heavy chain may have an amino acid sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with SEQ ID NO. 92, 96, 100, 104, 108, 112, 116, 120, 124, 128, or 132.
In one embodiment, the application discloses an isolated nucleic acid encoding the bispecific antibody, comprising the IgG1 heavy chain disclosed herein, the kappa light chain disclosed herein, the variable light chain disclosed herein, or the variable heavy chain disclosed herein. In one embodiment, the application discloses an isolated nucleic acid encoding the bispecific antibody, comprising the IgG1 heavy chain having an amino acid sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with SEQ ID NO. 02, 06, 08, 10, 12, 14, 16, 18, 20, 22, 26, 30, 34, 38, 42, 46, 50, 54, 58, 62, 72, 92, 96, 100, 104, 108, 112, 116, 120, 124, 128, or 132, the kappa light chain having an amino acid sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with SEQ ID NO. 04, 28, 32, 36, 40, 44, 48, 52, 56, 60, and 64, the variable light chain having an amino acid sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with SEQ ID NO. 94, 98, 102, 106, 110, 114, 118, 122, 126, 130, or 134, or the variable heavy chain having an amino acid sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with SEQ ID NO. 92, 96, 100, 104, 108, 112, 116, 120, 124, 128, or 132.
In one embodiment, the application discloses an expression vector comprising the isolated nucleic acid disclosed herein. In one embodiment, the expression vector comprises an isolated nucleic acid having a sequence having at least 70%, 80%, 90%, 98%, or 99% similarity with the nucleic acid sequence disclosed herein.
In one embodiment, the expression vector is expressible in a cell.
In one embodiment, the application discloses a host cell comprising the nucleic acid of disclosed herein.
In one embodiment, the application discloses a host cell comprising the expression vector.
In some embodiments, the application discloses the host cell, wherein the host cell is a prokaryotic cell or a eukaryotic cell.
In some embodiments, the application discloses a method of producing an antibody comprising culturing the host cell provided thereof so that the antibody is produced.
In some embodiments, the application discloses an immunoconjugate comprising the bispecific antibody and a cytotoxic agent. In some embodiments, the cytotoxic agent is a chemotherapeutic agent, a growth inhibitory agent, a toxin, or a radioactive isotope.
In one embodiment, the application discloses a pharmaceutical composition, comprising the bispecific antibody and a pharmaceutically acceptable carrier. In one embodiment, the application discloses a pharmaceutical composition, comprising the immunoconjugate and a pharmaceutically acceptable carrier. In one embodiment, the pharmaceutical composition further comprises radioisotope, radionuclide, a toxin, a therapeutic agent, a chemotherapeutic agent or a combination thereof.
In one embodiment, the application discloses a method of treating a subject with a cancer, comprising administering to the subject an effective amount of the bispecific antibody disclosed herein. In one embodiment, the cancer comprises breast cancer, colorectal cancer, pancreatic cancer, head and neck cancer, melanoma, ovarian cancer, prostate cancer, non-small lung cell cancer, small cell lung cancer, glioma, esophageal cancer, nasopharyngeal cancer, kidney cancer, gastric cancer, liver cancer, bladder cancer, cervical cancer, brain cancer, lymphoma, leukaemia, myeloma.
In one embodiment, the application discloses the method of treating a subject with a cancer, wherein the cancer comprises cells expressing PD-L1.
In one embodiment, the application discloses the method of treating a subject with a cancer, further comprising co-administering an effective amount of a therapeutic agent. In some embodiments, the therapeutic agent comprises an antibody, a chemotherapy agent, an enzyme, or a combination thereof.
In some embodiments, the therapeutic agent comprises capecitabine, cisplatin, trastuzumab, fulvestrant, tamoxifen, letrozole, exemestane, anastrozole, aminoglutethimide, testolactone, vorozole, formestane, fadrozole, letrozole, erlotinib, lafatinib, dasatinib, gefitinib, imatinib, pazopinib, lapatinib, sunitinib, nilotinib, sorafenib, nab-palitaxel, a derivative or a combination thereof.
In one embodiment, the application discloses the method of treating a subject with a cancer, wherein the subject is a human.
In one embodiment, the application discloses a solution comprising an effective concentration of the bispecific antibody disclosed herein, wherein the solution is blood plasma in a subject.
Still other embodiments of the present application will become readily apparent to those skilled in the art from the following detailed description, wherein is described embodiments of the application by way of illustrating the best mode contemplated for carrying out the application. As will be realized, the application is capable of other and different embodiments and its several details are capable of modifications in various obvious respects, all without departing from the spirit and the scope of the present application. Accordingly, the drawings and detailed description are to be regarded as illustrative in nature and not as restrictive.
The foregoing and other features of this disclosure may become more fully apparent from the following description and appended claims, taken in conjunction with the accompanying drawings. Understanding that these drawings depict only several embodiments arranged in accordance with the disclosure and are, therefore, not to be considered limiting of its scope, the disclosure may be described with additional specificity and detail through use of the accompanying drawings, in which:
In the following detailed description of embodiments of the application, reference is made to the accompanying drawings in which like references indicates similar elements, and in which is shown by way of illustration, specific embodiments in which the application may be practiced. These embodiments are described in sufficient detail to enable those skilled in the art to practice the application. In other instances, well-known circuits, structures, and techniques have not been shown in detail in order not to obscure the understanding of this description. The following detailed description is, therefore, not to be taken in a limiting sense, and the scoop of the application is defined only by the appended claims.
The disclosure relates to bispecific antibodies that specifically bind to human CTLA4, PD-1 or PD-L1. In some embodiments, the bispecific antibody comprises of a first arm that binds to CTLA4 and a second arm that binds to PD-1 or PD-L1. In some embodiments, the bispecific antibody comprises of a first arm that binds to PD-1 or PD-L1 and a second arm that binds to CTLA4. Examples of domains that comprise the arms include, but are not limited to, Fab and scFv domains. Each arm contains two antigen-binding domains and is connected to another arm via Fc domain. The Fc domain can be of human IgG1, IgG2, IgG3, IgG4 or an engineered isotype.
The bispecific antibodies of this application (
In some embodiments, scFv molecules described herein contain a 20-amino acid flexible gly-gly-gly-gly-ser (G4S) X4 linker that operably links the VH and VL, regardless of the V-region orientation (LH or HL). The remaining positions in the bispecific antibody may be consist of a human IgG1 Fc or IgG1 null Fc heavy chain, VH-CH1-Hinge-CH2-CH3, and its corresponding kappa light chain, VL-CL. scFv domains were genetically linked through a 10-amino acid (G4S)×2 linker to either N-terminal or C-terminal of IgG1 heavy chain, resulting in a contiguous ˜100 kDa heavy chain monomer peptide. When co-transfected with the appropriate light chain, the final symmetric bispecific molecule may be purified through the human IgG1 Fc (Protein A) and assayed to assess functional activity.
Heavy and light chain gene “cassettes” were previously constructed such that V-regions could be cloned using either restriction enzyme sites (
Bispecific antibodies are produced through a process that involves design of the intact molecule, synthesis and cloning of the nucleotide sequences for each domain, expression in mammalian cells and purification of the final product. Nucleotide sequences were assembled using the Geneious 10.2.3 software package (Biomatters, Auckland, NZ) and broken up into their component domains for gene synthesis (Genewiz, South Plainsfield, N.J.). In one example (
The light chain of CT4×PD224D1 IgG1 null consists of the anti-CTLA4 VL domain fused to the human C kappa domain. A synthesized gene fragment was digested with the restriction enzymes HindIII and BsiWI and was then ligated in-frame with the human C kappa domain. For both constructs, a small aliquot was transformed into E. coli DH10b (Invitrogen, Carlsbad, Calif.) and plated on TB+carbenicillin 100 ug/ml plates (Teknova, Hollister, Calif.) and incubated at 37 C overnight. Resultant colonies were selected and 2 ml overnight cultures inoculated in TB+carbenicillin. DNA was prepared (Thermo-Fisher, Carlsbad, Calif.) from overnight cultures and subsequently sequenced (Genewiz, South Plainsfield, N.J.) using sequencing primers (Sigma, St. Louis, Mo.) flanking each domain. All DNA sequences were assembled and analysed in Geneious.
Binding affinities and kinetics of anti-PD-L1 antibodies to PD-L1 recombinant protein were assessed via Surface Plasmon Resonance on ForteBio Octet RED96 instrument. The antigen was immobilized on the sensor chip surface and the tested antibodies were flown over the immobilized antigens. All molecules showed strong binding to the antigen as shown in Table 1 for examples.
Binding of the bispecific antibodies and their components to PD-1 antigen expressed on the surface of CHO cell line was assessed using FACS method. The bispecific antibodies were incubated with CHO cell line expressing PD-1 antigen and then detected with secondary anti-human antibodies directly conjugated to Alexa Fluor 647 fluorochrome. Cellular binding of the test antibodies was analyzed on a flow cytometer BD LSRFortessa. All tested antibodies bound to the antigen with a KD in a single digit nanomolar range (
Ability of the bispecific antibodies to block the interaction between CTLA4 and its ligand CD80 was tested in a biochemical interaction assay (Cisbio). Briefly, a bispecific antibody was incubated with CTLA4 and CD80 proteins. Detection antibodies recognizing CTLA4 and CD80 proteins and labeled with HTRF donor/acceptor fluorescent pair then were added to the mixture. The interaction between CTLA4 and CD80 was assessed via FRET efficiency. All bispecific antibodies tested were able to block the interaction between CTLA4 and CD80 (
Ability of the bispecific antibodies to enhance cytokine release from human Peripheral Blood Mononuclear Cells (PBMC) after stimulation with superantigen SEB was assessed. All bispecific molecules were able to significantly enhance the production of IL-2 by PBMC upon stimulation with SEB, as shown in
The bispecific antibodies were tested for their ability to block PD-1/PD-L1 pathway. Briefly, the test molecules were incubated with Jurkat reporter cell line expressing PD-1 receptor and luciferase NFAT reporter and CHO-PD-L1 cell line (Promega). The ability of the test antibodies to block the signaling through the PD-1/PD-L1 pathway was assessed via an increase in NFAT signaling. The NFAT signaling in turn was monitored via activity of luciferase reporter gene. The assay was read on a plate reader (Clariostar, BMG). All tested bispecific antibodies and monoclonal antibody controls were able to block PD-1/PD-L1 signaling (
The bispecific antibodies were tested for their ability to enhance Dendritic cell Mixed Leucocyte Reaction (MLR). The test molecules were incubated for 6 days with dendritic cells from one donor and T cells isolated from another donor. Dendritic cells were differentiated in vitro from monocytes in the presence of GM-CSF and IL-4. Monocytes and T cell populations were isolated from PBMC with StemCell isolation kits. The ability of the test molecules to enhance MLR was assessed via secreted IFNγ. All tested bispecific antibodies were able to augment production of IFNγ as shown on
The bispecific antibodies were tested for their ability to block suppressive effect of regulatory T cells on effector CD8 T cell proliferation and cytokine production. CD8 T cells were isolated with StemCell isolation kit and labeled with CellTrace dye (ThermoFisher). Dendritic cells were prepared as described earlier in the MLR study. Regulatory T cells were isolated from PBMC with StemCell isolation kit and expanded in vitro. The bispecific antibody was incubated with effector CD8 T cells, dendritic cells and regulatory T cells for 4 days. The ability of the bispecific antibody to rescue effector CD8 T cell function in the presence of regulatory T cells was assessed via proliferation of effector CD8 T cells (
Ability of bispecific molecules to have an effect on cytotoxic CD8 T cells was assessed in this study. Briefly, CD8 T cells were purified with StemCell isolation kit and stimulated with CEFT peptide pool (JPT Peptide Technologies) in the presence of the bispecific test molecules. The media was supplemented with IL-7 and IL-21. On day 11 CD8 T cells were re-stimulated with the peptides in the presence of Brefeldin and Monensin and anti-CD107a antibody directly labeled with a fluorochrome. 24 hours later CD8 T cells were stained with anti-IFNγ antibodies directly conjugated to a fluorochrome and assessed on a flow cytometer BD LSRFortessa. As shown on
Humanized mouse model was used to assess the ability of bispecific antibodies of this class to inhibit tumor growth in vivo. Briefly, NOG mice were reconstituted with human PBMC (5×106 cells per mouse). On day 3 the animals were subcutaneously inoculated with a human lung cancer cell line HCC827 (0.5E6 cells/animal) and started on a biweekly treatment with a bispecific antibody and control antibodies. Tumor volumes were measured every 2-3 days. Animal weight was monitored. The tested bispecific antibody was able to inhibit tumor growth better than the control antibodies (
Ability of the bispecific antibodies to augment proliferation of antigen specific CD8 T cells was assessed in this study. CD8 T cells were purified from PBMC with StemCell isolation kit, pulsed with influenza specific peptides (JPT Peptide Technologies) and incubated in the presence of the bispecific antibodies for 14 days. The media was supplemented with IL-7 and IL-21. On day 15 the cells were stained with a peptide specific MHC dextramers (Immudex) and assessed on a flow cytometer BD LSRFortessa. All tested bispecific antibodies were able to increase the number of antigen specific CD8 T cells (
Ability of the bispecific antibodies to augment T cell memory response was assessed. Briefly, PBMC were incubated for 4-5 days in the presence of peptides specific for CMV, EBV, Influenza and Tetanus (JPT Peptide Technologies). The amount of secreted IFNγ was quantified. The bispecific CT4×PD224D1, shown on
The bispecific antibodies were tested for their ability to enhance Redirected T cell Cytotoxicity (RTCC) against a tumor cell line target. The tumor cell line was stably expressing nucleus-localized Red Fluorescent Protein (RFP) delivered via lentiviral transduction (Sartorius). The tumor cells were co-cultured with PBMC and a T cell engager molecule specific for the given tumor cell line. The bispecific antibodies were added to the co-cultures. Lysis of tumor cells was assessed by counting RFP labeled tumor cell nuclei. Images were acquired on live cell imager IncuCyte (Sartorius). Activity of the antibodies were assessed after 96 hours of incubation. Four PBMC donors were tested in this study. All bispecific antibodies tested were able to enhance RTCC activity in at least one PBMC donor tested (
While the present disclosure has been described with reference to particular embodiments or examples, it may be understood that the embodiments are illustrative and that the disclosure scope is not so limited. Alternative embodiments of the present disclosure may become apparent to those having ordinary skill in the art to which the present disclosure pertains. Such alternate embodiments are considered to be encompassed within the scope of the present disclosure. Accordingly, the scope of the present disclosure is defined by the appended claims and is supported by the foregoing description. All references cited or referred to in this disclosure are hereby incorporated by reference in their entireties.
MSWVRQAPGKGLEYIGYIGDTTGIAYASWANGRFTISKDNTKNTVDLQMNSLRAEDTAVYYCARGWSYLD
IWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSALVMTQSPSSLSASVGDRVTITCQASQNIYSNLAWYQQ
IWVRQAPGKGLEWVGIITYSGSRYYANWAKGRFTISKDNTKNTVYLQMNSLRAEDTAVYYCARDYMSGSH
LWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSAYDMTQSPSSVSASVGDRVTIKCQASEDIYSFLAWYQQ
DMCWVRQAPGKGLEWIACIAAGSAGITYDANWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARSAF
SFDYAMDLWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSDIQMTQSPSTLSASVGDRVTITCQASQSISS
HLNWYQQKPGKAPKLLIYKASTLASGVPSRFSGSGSGTEFTLTISSLQPDDFATYYCQQGYSWGNVDNVF
MIWVRQAPGKGLEWVGIISYVGNTYYASWAKGRFTLSKDNTSTTVDLQMNSLRAEDTAVYYCARDFISGS
HLWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSAYDMTQSPSSVSASVGDRVTINCQASESISSFLSWYQ
MSWVRQAPGKGLEWIGVIDINVYIYYANWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARYVGNND
DYINLWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSDIQMTQSPSTLSASVGDRVTITCQSSQSVYNGYW
LSWYQQKPGKAPKLLIYGASTLASGVPSRFSGSGSGTEFTLTISSLQPDDFATYYCLGSYTSSTENSEGG
MGWVRQAPGKGLEWIGTITYVGNTYYASWAKGRFTISKTSTTVDLKITSPTTEDTAVYYCARESGTIYYS
YFNLWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSAFELTQSPSSLSASVGDRVTIKCQASESISNYLSW
WICWVRQAPGKGLELIACIYAGTSGSTSYASWARGRFTISETSKNIVTLQMNSLRAEDSAVYYCARNLYT
YNSLWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSDIQMTQSPSTLSASVGDRVTITCQSSQSVYDNNWL
AWYQQKPGKAPKLLIYTVSTLASGVPSRFSGSGSGTEFTLTISSLQPDDFATYYCQGTYYSSGWNFAFGG
MCWVRQAPGKGLEWIGCITTGSGSTYYASWAKRRFTISKDNSKNTVTLQMNSLRAEDTAVYYCTRAFDLW
VSWVRQAPGKGLEWVALIFPGIGFKDYASWVNGRFTLSSDNAQNTVELQMNSLRAEDTAVYYCARDLDLW
WICWVRQAPGKGLEWIACIYIGSIDVFYYASWAKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARAAN
TDTTYFNLWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSAYDMTQSPSSLSASVGDRVTINCQASQSINN
QLSWYQQKPGKVPKLLIYGASTLASGVPSRFTGSGSGTDFTLTISSLQPEDVATYYCHVHYCSGGSCFWA
YWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGERATLSCRASQSVGSSYLAWYQ
YWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGERATLSCRASQSVGSSYLAWYQ
SSYTMHWVRQAPGKGLEWVTFISYDGNNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCART
GWLGPFDYWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGERATLSCRASQSVGS
SYLAWYQQKPGQAPRLLIYGAFSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSSPWTFGQ
DYWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGERATLSCRASQSVGSSYLAWY
TMHWVRQAPGKGLEWVTFISYDGNNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCARTGWL
GPFDYWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGERATLSCRASQSVGSSYL
AWYQQKPGQAPRLLIYGAFSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSSPWTFGQGTK
MHWVRQAPGKGLEWVTFISYDGNNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCARTGWLG
PFDYWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGERATLSCRASQSVGSSYLA
MHWVRQAPGKGLEWVTFISYDGNNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCARTGWLG
PFDYWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGERATLSCRASQSVGSSYLA
SSYTMHWVRQAPGKGLEWVTFISYDGNNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCART
GWLGPFDYWGQGTLVTVSSGGGGSGGGGSGGGGSGGGGSEIVLTQSPGTLSLSPGERATLSCRASQSVGS
SYLAWYQQKPGQAPRLLIYGAFSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSSPWTFGQ
LAWYQQKPGKVPKLLIYQASTLASGVPSRFSGSGYGTDFTLTISSLQPEDVATYYCQGGYYSAALNTFGGGTKVEIK
This application claims the benefit of U.S. Provisional Patent Application No. 62/580,845 filed on Nov. 2, 2017, titled “BISPECIFIC ANTIBODIES AND METHODS OF MAKING AND USING THEREOF”, which are expressly incorporated herein by reference in its entirety.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2018/058810 | 11/2/2018 | WO | 00 |
Number | Date | Country | |
---|---|---|---|
62580845 | Nov 2017 | US |