WO2023149977A1 - Methods for treating cancer - Google Patents

Methods for treating cancer Download PDF

Info

Publication number
WO2023149977A1
WO2023149977A1 PCT/US2022/080568 US2022080568W WO2023149977A1 WO 2023149977 A1 WO2023149977 A1 WO 2023149977A1 US 2022080568 W US2022080568 W US 2022080568W WO 2023149977 A1 WO2023149977 A1 WO 2023149977A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
tigit
light chain
weeks
Prior art date
Application number
PCT/US2022/080568
Other languages
French (fr)
Inventor
Suba KRISHNAN
John Lewicki
Ann KAPOUN
Brian Schwartz
Original Assignee
Mereo Biopharma 5, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mereo Biopharma 5, Inc. filed Critical Mereo Biopharma 5, Inc.
Publication of WO2023149977A1 publication Critical patent/WO2023149977A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present invention relates to methods for treating cancer and in particular treating soft tissue sarcomas, testicular germ cell tumors and uveal melanomas.
  • the basis for immunotherapy is the manipulation and/or modulation of the immune system, including both innate immune responses and adaptive immune responses.
  • the general aim of immunotherapy is to treat diseases by controlling the immune response to a “foreign agent”, for example a pathogen or a tumor cell.
  • the immune system is a highly complex system made up of a great number of cell types, including but not limited to, T-cells, B-cells, natural killer cells, antigen-presenting cells, dendritic cells, monocytes, and macrophages. These cells possess complex and subtle systems for controlling their interactions and responses.
  • the cells utilize both activating and inhibitory mechanisms and feedback loops to keep responses in check and not allow negative consequences of an uncontrolled immune response (e.g., autoimmune diseases).
  • cancer immunosurveillance is based on the theory that the immune system can recognize tumor cells, mount an immune response, and suppress the development and/or progression of a tumor.
  • cancerous cells have developed mechanisms to evade the immune system which can allow for uninhibited growth of tumors.
  • Cancer/tumor immunotherapy focuses on the development of new and novel agents that can activate and/or boost the immune system to achieve a more effective attack against tumor cells resulting in increased killing of tumor cells and/or inhibition of tumor growth.
  • an anti-TIGIT antibody and an anti-PD-1 antibody can be used to treat rare cancers including soft tissue sarcomas, testicular germ cell tumours and uveal melanomas.
  • the invention provides a method of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti- PD-1 antibody, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID N0:5), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumour and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR
  • the anti-TIGIT antibody comprises a heavy chain variable region having at least 90% sequence identity to SEQ ID NO:7 and a light chain variable region having at least 90% sequence identity to SEQ ID NO: 8. In certain embodiments, the anti-TIGIT antibody comprises a heavy chain variable region comprising SEQ ID NO:7 and a light chain variable region comprising SEQ ID NO:8. In some embodiments, the anti-TIGIT antibody comprises a heavy chain having at least 90% sequence identity to SEQ ID NO:9 and a light chain having at least 90% sequence identity to SEQ ID NO: 10. In certain embodiments, the anti-TIGIT antibody comprises a heavy chain comprising SEQ ID NO:9 and a light chain comprising SEQ ID NO: 10.
  • the anti-TIGIT antibody is etigilimab.
  • the anti-TIGIT antibody can be a monoclonal antibody, a humanized antibody, a human antibody, a chimeric antibody, a bispecific antibody, an IgGl antibody, an IgG2 antibody, an IgG4 antibody, or an antibody fragment comprising an antigen binding site.
  • the anti-PD-1 antibody is selected from the group consisting of pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo), dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab.
  • the anti-TIGIT antibody is administered prior to, concurrently with and/or subsequently to the administration of the anti-PD-1 antibody.
  • the anti-TIGIT antibody and the anti-PD-1 antibody are administered intravenously.
  • the anti-TIGIT antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks.
  • the anti-TIGIT antibody is administered once every two weeks.
  • the anti-PD-1 antibody is administered once every two weeks, once every three weeks, once every four weeks, once every five weeks or once every six weeks.
  • the anti-PD-1 antibody is administered once every two weeks, once every three weeks, once every four weeks or once every six weeks.
  • the anti-TIGIT antibody can be administered at a flat dose of 1000 mg. In some embodiments, the anti-TIGIT antibody is administered at a flat dose of 1000 mg once every two weeks.
  • the anti-PD-1 antibody can be administered at a flat dose of 200 mg- 1000 mg, for example the anti-PD-1 antibody is administered at a flat dose of 200 mg, 240 mg, 350 mg, 400mg, 500 mg or 1000 mg.
  • the human patient has not received prior treatment with an anti-cancer agent.
  • the human patient has been previously treated with an anti-cancer agent and there is tumour growth, progression, or recurrence during or after treatment with the anti-cancer agent.
  • the anti-cancer agent is a checkpoint inhibitor, a chemotherapeutic agent or a cell therapy.
  • the checkpoint inhibitor is an anti-PD-1 antibody, an anti-PD-Ll antibody or a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1.
  • the human patient has been previously been treated with a chemotherapeutic agent, and there is tumour growth, progression, or recurrence during or after treatment with the chemotherapeutic agent.
  • the soft tissue sarcoma is selected from the group consisting of leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, dedifferentiated liposarcoma, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid, malignant peripheral nerve sheath tumours (MPNST) and undifferentiated (pleomorphic sarcoma).
  • the soft tissue sarcoma is a dedifferentiated liposarcoma or an undifferentiated (pleomorphic sarcoma).
  • the testicular germ cell tumor is a seminomas testicular germ cell tumour or a non-seminomas testicular germ cell tumor.
  • the uveal melanoma is choroidal melanoma, ciliary body melanoma and iris melanoma.
  • the human patient is over 50 years old.
  • Figure 1 FACS analysis was conducted with anti-TIGIT antibodies 313M32, 313M26 and 313R19 to assess the ability of these antibodies to block PVR binding to human TIGIT.
  • Figure 2 Inhibition of tumor growth by anti-TIGIT antibodies 313R19 and 313M32 in a humanized mouse model.
  • Figure 3 Historical overall response rate with checkpoint inhibitors for ovarian cancer, cervical cancer, and uveal melanoma.
  • Figure 4 Scans of a cervical cancer tumor in a patient who was treated with an anti-TIGIT antibody and an anti-PD-1 antibody. The circles indicate the location of the tumor on the scan. The change in tumour volume resulted in a complete overall response after the patient was treated with the combination of an anti-TIGIT antibody and an anti-PD-1 antibody.
  • Figure 5 Heavy and light chain sequence of Camrelizumab (PD-1 antibody)
  • Figure 10 Heavy and light chain sequence of Socazolimab (PD-L1 antibody)
  • Figure 11 A summary showing the length of time that patients have been part of the Phase lb/2 clinical trial.
  • Figures 12-17 disclose pharmacodynamic analysis of biomarkers after treatment with the combination of etigilimab and nivolumab.
  • Figure 12 shows the change in Treg cells and CD8+ cells after treatment;
  • Figures 13A and 13B show the change in Ki-67+ TIGIT+ CD4 cells after treatment;
  • Figure 14 shows the change in the level of IFNy produced by CD4+ effector memory cells after treatment;
  • Figure 15 shows the change in the level of Ki-67+ CD8+ PD-1+ cells after treatment and
  • Figure 16 shows the change in Ki-67+ natural killer cells (NK cells) after treatment.
  • Figure 17 shows the change in T Progenitor Exhausted like (TPEX) cells after treatment.
  • TPEX T Progenitor Exhausted like
  • Figure 18 Scans of a cervical cancer tumor in a patient who was treated with an anti-TIGIT antibody and an anti-PD-1 antibody. The circles indicate the location of the tumor on the scan. The change in tumour volume resulted in a partial overall response after the patient was treated with the combination of an anti-TIGIT antibody and an anti-PD-1 antibody.
  • Figure 19 A summary showing the length of time that patients have been part of the Phase lb/2 clinical trial. Time on treatment and investigator-assessed RECIST 1.1 response.
  • Figure 20 A summary of the preliminary efficacy of the combination of etigilimab with an anti-PD-1 antibody (nivolumab).
  • PD disease progression
  • SD stable disease
  • cCR confirmed complete response
  • uCR unconfirmed complete response
  • cPR confirmed partial response
  • uPR unconfirmed partial response
  • TMB-H high tumor mutational burden
  • Figure 21 A waterfall plot showing the percentage change compared to the baseline for each subject.
  • T-cell immunoreceptor with Ig and ITIM domains is a type I transmembrane glycoprotein that contains an immunoglobulin variable (IgV) domain.
  • TIGIT belongs to the poliovirus receptor (PVR) family and binds to the poliovirus receptor (PVR; CD155) with high affinity and to PVRL-2 (CD112) and PVRL-3 (CD113) with a lower affinity.
  • PVR poliovirus receptor
  • PVRL-2 CD112
  • PVRL-3 CD113
  • TIGIT is expressed on T-cells, including regulatory T-cells (Tregs) and memory T-cells, as well as on NK cells and is upregulated following activation of naive CD4+ T-cells.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6).
  • the anti-TIGIT antibody is a monoclonal antibody, a humanized antibody, a human antibody, a recombinant antibody, a chimeric antibody, a bispecific antibody or a multi-specific antibody.
  • the antibody is monovalent.
  • the antibody is bivalent.
  • the antibody is monospecific.
  • the antibody is bispecific.
  • the anti-TIGIT antibody is an IgA, IgD, IgE, IgG, or IgM antibody.
  • the anti-TIGIT antibody is an IgGl antibody.
  • the anti- TIGIT antibody is an IgG2 antibody.
  • the anti-TIGIT antibody is an IgG4 antibody. In some embodiments, the anti-TIGIT antibody is conjugated to a cytotoxic moiety. In some embodiments, the anti-TIGIT antibody is isolated. In some embodiments, the anti- TIGIT antibody is substantially pure.
  • the anti-TIGIT antibody comprises a heavy chain variable region having at least 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO:7 and/or a light chain variable region having at least 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO:8.
  • the anti-TIGIT antibody comprises a heavy chain variable region comprising SEQ ID NO:7 and a light chain variable region comprising SEQ ID NO: 8.
  • the anti-TIGIT antibody comprises a heavy chain variable region consisting of SEQ ID NO:7 and a light chain variable region consisting of SEQ ID NOG.
  • the anti-TIGIT antibody comprises a heavy chain having at least 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO:9 and/or a light chain having at least 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 10.
  • the anti-TIGIT antibody comprises a heavy chain comprising SEQ ID NO:9 and a light chain comprising SEQ ID NO: 10.
  • the anti-TIGIT antibody comprises a heavy chain consisting of SEQ ID NO:9 and a light chain consisting of SEQ ID NOTO.
  • the anti-TIGIT antibody is etigilimab.
  • the heavy chain of etigilimab consists of SEQ ID NO:9 and the light chain of etigilimab consists of SEQ ID NOTO.
  • the anti-TIGIT antibody comprises the variable regions of etigilimab wherein the heavy chain variable region (SEQ ID NO: 7) and/or the light chain variable region (SEQ ID NO: 8) of etigilimab have been affinity-matured.
  • the anti-TIGIT antibody comprises the heavy chain and light chain of etigilimab (with or without the leader sequence).
  • the anti-TIGIT antibody is etigilimab.
  • the anti-TIGIT antibody comprises the heavy chain variable region of etigilimab as part of an IgGl, IgG2, or IgG4 heavy chain.
  • the anti-TIGIT antibody comprises the heavy chain variable region of the etigilimab as part of a human IgGl heavy chain. In certain embodiments, the anti-TIGIT antibody comprises the heavy chain variable region of the etigilimab as part of a human IgG2 heavy chain. In certain embodiments, the anti-TIGIT antibody comprises the heavy chain variable region of the etigilimab as part of a human IgG4 heavy chain.
  • the anti-TIGIT antibody is a monoclonal antibody.
  • Monoclonal antibodies can be prepared using hybridoma methods known to one of skill in the art. In some embodiments, using the hybridoma method, a mouse, rat, rabbit, hamster, or other appropriate host animal, is immunized as described above to elicit the production of antibodies that specifically bind the immunizing antigen. In some embodiments, lymphocytes can be immunized in vitro. In some embodiments, the immunizing antigen can be a human protein or a fragment thereof. In some embodiments, the immunizing antigen can be a mouse protein or a fragment thereof.
  • lymphocytes are isolated and fused with a suitable myeloma cell line using, for example, polyethylene glycol.
  • the hybridoma cells are selected using specialized media as known in the art and unfused lymphocytes and myeloma cells do not survive the selection process.
  • Hybridomas that produce monoclonal antibodies directed specifically against a chosen antigen may be identified by a variety of methods including, but not limited to, immunoprecipitation, immunoblotting, and in vitro binding assays (e.g., flow cytometry, FACS, ELISA, and radioimmunoassay).
  • the hybridomas can be propagated either in in vitro culture using standard methods or in vivo as ascites tumors in an animal.
  • the monoclonal antibodies can be purified from the culture medium or ascites fluid according to standard methods in the art including, but not limited to, affinity chromatography, ion-exchange chromatography, gel electrophoresis, and dialysis.
  • monoclonal antibodies can be made using recombinant DNA techniques as known to one skilled in the art.
  • the polynucleotides encoding a monoclonal antibody are isolated from mature B-cells or hybridoma cells, such as by RT-PCR using oligonucleotide primers that specifically amplify the genes encoding the heavy and light chains of the antibody, and their sequence is determined using standard techniques.
  • the isolated polynucleotides encoding the heavy and light chains are then cloned into suitable expression vectors which produce the monoclonal antibodies when transfected into host cells such as E.
  • recombinant monoclonal antibodies, or fragments thereof can be isolated from phage display libraries expressing variable domains or CDRs of a desired species.
  • Polynucleotide(s) encoding a monoclonal antibody can be modified, for example, by using recombinant DNA technology to generate alternative antibodies.
  • the constant domains of the light chain and heavy chain of, for example, a mouse monoclonal antibody can be substituted for constant regions of, for example, a human antibody to generate a chimeric antibody, or for a non-immunoglobulin polypeptide to generate a fusion antibody.
  • the constant regions are truncated or removed to generate a desired antibody fragment of a monoclonal antibody. Site-directed or high-density mutagenesis of the variable region(s) can be used to optimize specificity, affinity, etc. of a monoclonal antibody.
  • the anti-TIGIT antibody is a humanized antibody.
  • humanized antibodies are human immunoglobulins in which the amino acid residues of the CDRs are replaced by amino acid residues from CDRs of a non-human species (e.g., mouse, rat, rabbit, hamster, etc.) that have the desired specificity, affinity, and/or binding capability using methods known to one skilled in the art.
  • some of the framework variable region amino acid residues of a human immunoglobulin are replaced with corresponding amino acid residues in an antibody from a non-human species.
  • a humanized antibody can be further modified by the substitution of additional residues either in the framework variable region and/or within the replaced non-human residues to further refine and optimize antibody specificity, affinity, and/or capability.
  • a humanized antibody will comprise variable domain regions containing all, or substantially all, of the CDRs that correspond to the non-human immunoglobulin whereas all, or substantially all, of the framework regions are those of a human immunoglobulin sequence.
  • the framework regions are those of a human consensus immunoglobulin sequence.
  • a humanized antibody can also comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region or domain
  • such humanized antibodies are used therapeutically because they may reduce antigenicity and HAMA (human anti-mouse antibody) responses when administered to a human patient.
  • the anti-TIGIT antibody is a human antibody.
  • Human antibodies can be directly prepared using various techniques known in the art.
  • human antibodies may be generated from immortalized human B lymphocytes immunized in vitro or from lymphocytes isolated from an immunized individual. In either case, cells that produce an antibody directed against a target antigen can be generated and isolated.
  • the human antibody can be selected from a phage library, where that phage library expresses human antibodies.
  • phage display technology can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable domain gene repertoires from unimmunized donors. Techniques for the generation and use of antibody phage libraries are well known in the art.
  • human antibodies can be made in transgenic mice that contain human immunoglobulin loci. Upon immunization these mice are capable of producing the full repertoire of human antibodies in the absence of endogenous immunoglobulin production.
  • the anti-TIGIT antibody binds with TIGIT and interferes with the interaction of TIGIT with a second protein, for example PVR, PVRL2 or PVLR3. In some embodiments, the anti-TIGIT antibody binds TIGIT and disrupts binding of TIGIT to PVR, and/or disrupts PVR activation of TIGIT signalling.
  • the anti-TIGIT antibody is an antibody that specifically binds the extracellular domain of human TIGIT, or a fragment thereof. In some embodiments, the anti-TIGIT antibody is an antibody that specifically binds the Ig-like domain of human TIGIT. In some embodiments, the anti-TIGIT antibody is an antibody that specifically binds the IgV domain of human TIGIT. In some embodiments, the anti-TIGIT antibody is an antibody that binds within amino acids 22-141 of human TIGIT. In some embodiments, the anti-TIGIT antibody is an antibody that binds within amino acids 22-141 of SEQ ID NO: 12. In some embodiments, the antibody binds within amino acids 22-124 of human TIGIT.
  • the antibody binds within amino acids 22-124 of SEQ ID NO: 11. In certain embodiments, the anti-TIGIT antibody binds within SEQ ID NO: 12, or a fragment thereof. In some embodiments, the anti-TIGIT antibody is an antibody that binds within amino acids SO- 124 of human TIGIT. In some embodiments, the anti-TIGIT antibody is an antibody that binds within amino acids 50-124 of SEQ ID NOT E In certain embodiments, the anti-TIGIT antibody binds within SEQ ID NO: 12, or a fragment thereof.
  • the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids within SEQ ID NO: 13. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids within SEQ ID NO: 14. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids within SEQ ID NO: 13 and SEQ ID NO: 14. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62 and 1109 of SEQ ID NO: 11. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62 and T119 of SEQ ID NO: 11.
  • the anti- TIGIT antibody is an antibody that binds an epitope comprising amino acids Q64 and 1109 of SEQ ID NO: 11. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q64 and T119 of SEQ ID NO: 11.
  • the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, Q64, and 1109 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, Q64, and T119 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, 1109, and T119 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q64, 1109, and T119 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, Q64, 1109, and T119 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, Q64, 1109, and T119 of SEQ ID NOTE In some embodiments, the anti
  • the anti-TIGIT antibody binds TIGIT with a dissociation constant (KD) of about IpM or less, about lOOnM or less, about 40nM or less, about 20nM or less, about lOnM or less, about InM or less, about O.lnM or less, 50pM or less, lOpM or less, or IpM or less.
  • KD dissociation constant
  • an anti-TIGIT antibody binds TIGIT with a KD of about 20nM or less.
  • an anti-TIGIT antibody binds TIGIT with a KD of about lOnM or less.
  • an anti-TIGIT antibody binds TIGIT with a KD of about InM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about 0.5nM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about O.lnM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about 50pM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about 25pM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about lOpM or less.
  • an anti-TIGIT antibody binds TIGIT with a KD of about IpM or less.
  • the dissociation constant of the antibody to TIGIT is the dissociation constant determined using a TIGIT fusion protein comprising at least a portion of the extracellular domain of TIGIT protein immobilized on a Biacore chip.
  • the dissociation constant of the antibody to TIGIT is the dissociation constant determined using the antibody captured by an anti-human IgG antibody on a Biacore chip and a soluble TIGIT protein.
  • the anti-TIGIT antibody binds TIGIT with a KD of about 0.4nM or less determined using a TIGIT fusion protein comprising at least a portion of the extracellular domain of TIGIT protein immobilized on a Biacore chip. In certain embodiments, the anti-TIGIT antibody binds TIGIT with a KD of about 0.4nM or less determined using the antibody captured by an anti-human IgG antibody on a Biacore chip and a soluble TIGIT protein.
  • the anti-TIGIT antibody binds TIGIT with a half maximal effective concentration (EC50) of about I pM or less, about lOOnM or less, about 40nM or less, about 20nM or less, about lOnM or less, about InM or less, or about O.lnM or less.
  • the anti-TIGIT antibody binds to human TIGIT with a half maximal effective concentration (EC 50) of about I M or less, about lOOnM or less, about 40nM or less, about 20nM or less, about lOnM or less, about InM or less, or about O.lnM or less.
  • Heteroconjugate antibodies are also within the scope of the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune cells to unwanted cells. It is also contemplated that the heteroconjugate antibodies can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4- mercaptobutyrimidate.
  • modified antibodies can comprise any type of variable region that provides for the association of the antibody with the target (i.e., TIGIT).
  • the variable region may comprise or be derived from any type of mammal that can be induced to mount a humoral response and generate immunoglobulins against the desired antigen.
  • the variable region of the modified antibodies can be, for example, of human, murine, rat, rabbit, non-human primate (e.g. cynomolgus monkeys, macaques, etc.), or rabbit origin.
  • both the variable and constant regions of the modified immunoglobulins are human.
  • variable regions of compatible antibodies can be engineered or specifically tailored to improve the binding properties or reduce the immunogenicity of the molecule.
  • variable regions useful in the present invention can be humanized or otherwise altered through the inclusion of imported amino acid sequences.
  • variable domains in both the heavy and light chains are altered by at least partial replacement of one or more CDRs and, if necessary, by partial framework region replacement and sequence modification and/or alteration.
  • the CDRs may be derived from an antibody of the same class or even subclass as the antibody from which the framework regions are derived, it is envisaged that the CDRs may be derived from an antibody of different class and often from an antibody from a different species. It may not be necessary to replace all of the CDRs with all of the CDRs from the donor variable region to transfer the antigen binding capacity of one variable domain to another. Rather, it may only be necessary to transfer those residues that are required to maintain the activity of the antigenbinding site.
  • the modified antibodies of this invention will comprise antibodies (e.g., full- length antibodies or immunoreactive fragments thereof) in which at least a fraction of one or more of the constant region domains has been deleted or otherwise altered so as to provide desired biochemical characteristics such as increased tumor localization or increased serum half-life when compared with an antibody of approximately the same immunogenicity comprising a native or unaltered constant region.
  • the constant region of the modified antibodies will comprise a human constant region.
  • Modifications to the constant region compatible with this invention comprise additions, deletions or substitutions of one or more amino acids in one or more domains.
  • the modified antibodies disclosed herein may comprise alterations or modifications to one or more of the three heavy chain constant domains (CHI, CH2 or CH3) and/or to the light chain constant domain (CL).
  • one or more domains are partially or entirely deleted from the constant regions of the modified antibodies.
  • the modified antibodies will comprise domain deleted constructs or variants wherein the entire CH2 domain has been removed (ACH2 constructs).
  • the omitted constant region domain is replaced by a short amino acid spacer (e.g., 10 amino acid residues) that provides some of the molecular flexibility typically imparted by the absent constant region.
  • the modified antibodies are engineered to fuse the CH3 domain directly to the hinge region of the antibody.
  • a peptide spacer is inserted between the hinge region and the modified CH2 and/or CH3 domains.
  • constructs may be expressed wherein the CH2 domain has been deleted and the remaining CH3 domain (modified or unmodified) is joined to the hinge region with a 5-20 amino acid spacer.
  • a spacer may be added to ensure that the regulatory elements of the constant domain remain free and accessible or that the hinge region remains flexible.
  • amino acid spacers may, in some cases, prove to be immunogenic and elicit an unwanted immune response against the construct. Accordingly, in certain embodiments, any spacer added to the construct will be relatively non-immunogenic so as to maintain the desired biological qualities of the modified antibodies.
  • the modified antibodies may have only a partial deletion of a constant domain or substitution of a few or even a single amino acid.
  • the mutation of a single amino acid in selected areas of the CH2 domain may be enough to substantially reduce Fc binding.
  • the mutation of a single amino acid in selected areas of the CH2 domain may be enough to substantially reduce Fc binding and increase cancer cell localization and/or tumor penetration.
  • Such partial deletions of the constant regions may improve selected characteristics of the antibody (serum half-life) while leaving other desirable functions associated with the subject constant region domain intact.
  • the constant regions of the disclosed antibodies may be modified through the mutation or substitution of one or more amino acids that enhances the profile of the resulting construct.
  • the modified antibodies comprise the addition of one or more amino acids to the constant region to enhance desirable characteristics such as decreasing or increasing effector function or provide for more cytotoxin or carbohydrate attachment sites.
  • the constant region mediates several effector functions. For example, binding of the Cl component of complement to the Fc region of IgG or IgM antibodies (bound to antigen) activates the complement system. Activation of complement is important in the opsonization and lysis of cell pathogens. The activation of complement also stimulates the inflammatory response and can also be involved in autoimmune hypersensitivity.
  • the Fc region of an antibody can bind a cell expressing a Fc receptor (FcR).
  • Fc receptors There are a number of Fc receptors which are specific for different classes of antibody, including IgG (gamma receptors), IgE (epsilon receptors), IgA (alpha receptors) and IgM (mu receptors).
  • ADCC antibody-dependent cell cytotoxicity
  • the modified antibodies provide for altered effector functions that, in turn, affect the biological profile of the administered antibody.
  • the deletion or inactivation (through point mutations or other means) of a constant region domain may reduce Fc receptor binding of the circulating modified antibody.
  • the deletion or inactivation (through point mutations or other means) of a constant region domain may reduce Fc receptor binding of the circulating modified antibody thereby increasing cancer cell localization and/or tumor penetration.
  • the constant region modifications increase the serum half-life of the antibody.
  • the constant region modifications reduce the serum half-life of the antibody.
  • the constant region is modified to eliminate disulfide linkages or oligosaccharide moieties. Modifications to the constant region in accordance with this invention may easily be made using well known biochemical or molecular engineering techniques.
  • the anti-TIGIT antibody does not have one or more effector functions. For instance, in some embodiments, the antibody has no ADCC activity, and/or no complement-dependent cytotoxicity (CDC) activity. In certain embodiments, the antibody does not bind an Fc receptor and/or complement factors. In certain embodiments, the antibody has no effector function(s).
  • the anti-TIGIT antibody has one or more effector functions.
  • the antibody has ADCC activity, and/or complement-dependent cytotoxicity (CDC) activity.
  • the antibody binds an Fc receptor and/or complement factors.
  • the antibody has effector function(s).
  • the anti-TIGIT antibody binds the Fc receptor. In certain embodiments, the anti-TIGIT antibody activates ADCC. In some embodiments, the anti-TIGIT antibody recruits activated myeloid cells to the tumor. In some embodiments, the anti-TIGIT antibody activates T cells. In some embodiments, the anti-TIGIT antibody activates NK cells. In some embodiments, the anti-TIGIT antibody reduces the number of T-regulatory cells. In some embodiments, the anti-TIGIT antibody increases the CD8/Treg ratio. In some embodiments, the anti-TIGIT antibody increases the CD8/Treg ratio.
  • the present invention further embraces variants and equivalents which are substantially homologous to the recombinant, monoclonal, chimeric, humanized, and human antibodies, or antibody fragments thereof, described herein.
  • variants can contain, for example, conservative substitution mutations, i.e. the substitution of one or more amino acids by similar amino acids.
  • the antibodies described herein are isolated. In certain embodiments, the antibodies described herein are substantially pure.
  • the anti-TIGIT antibodies of the present invention can be assayed for specific binding by any method known in the art.
  • the immunoassays which can be used include, but are not limited to, competitive and non-competitive assay systems using techniques such as Biacore analysis, FACS analysis, immunofluorescence, immunocytochemistry, Western blot analysis, radioimmunoassay, ELISA, “sandwich” immunoassay, immunoprecipitation assay, precipitation reaction, gel diffusion precipitin reaction, immunodiffusion assay, agglutination assay, complement-fixation assay, immunoradiometric assay, fluorescent immunoassay, and protein A immunoassay.
  • screening for specific binding of an antibody to human TIGIT may be determined using ELISA.
  • An ELISA comprises preparing antigen (e.g., TIGIT or a fragment thereof), coating wells of a 96-well microtiter plate with antigen, adding the test antibodies conjugated to a detectable compound such as an enzymatic substrate (e.g.
  • test antibodies are not conjugated to a detectable compound, but instead a secondary antibody that recognizes the antibody (e.g., an anti-Fc antibody) and is conjugated to a detectable compound is added to the wells.
  • a secondary antibody that recognizes the antibody e.g., an anti-Fc antibody
  • the test antibodies instead of coating the well with the antigen, can be coated to the wells, the antigen (e.g., TIGIT) is added to the wells, followed by a secondary antibody conjugated to a detectable compound.
  • the specific binding of an antibody to TIGIT may be determined using FACS.
  • a FACS screening assay may comprise generating a cDNA construct that expresses an antigen as a full-length protein (TIGIT) or a fusion protein (e.g., TIGIT-CD4TM), transfecting the construct into cells, expressing the antigen on the surface of the cells, mixing the test antibodies with the transfected cells, and incubating for a period of time.
  • the cells bound by the test antibodies may be identified using a secondary antibody conjugated to a detectable compound (e.g., PE-conjugated anti-Fc antibody) and a flow cytometer.
  • a detectable compound e.g., PE-conjugated anti-Fc antibody
  • the binding affinity of an antibody and the off-rate of an antibody-antigen interaction can be determined by competitive binding assays.
  • a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 125I-TIGIT), or fragment or variant thereof, with the antibody of interest in the presence of increasing amounts of unlabeled antigen followed by the detection of the antibody bound to the labeled antigen.
  • the affinity of the antibody for the antigen and the binding off-rates can be determined from the data by Scatchard plot analysis.
  • Biacore kinetic analysis is used to determine the binding on and off rates of antibodies.
  • Biacore kinetic analysis comprises analyzing the binding and dissociation of antibodies from chips with immobilized antigen (e.g., TIGIT) on their surface. In some embodiments, Biacore kinetic analysis comprises analyzing the binding and dissociation of antigen (e.g., TIGIT) from chips with immobilized antibody (e.g., anti-TIGIT antibody) on their surface.
  • TIGIT antigen
  • anti-TIGIT antibody anti-TIGIT antibody
  • the proteins target the polypeptide to the endoplasmic reticulum and the proteins are sorted to their destinations, for example, to the inner space of an organelle, to an interior membrane, to the cell's outer membrane, or to the cell exterior via secretion.
  • Most signal sequences are cleaved from the protein by a signal peptidase after the proteins are transported to the endoplasmic reticulum. The cleavage of the signal sequence from the polypeptide usually occurs at a specific site in the amino acid sequence and is dependent upon amino acid residues within the signal sequence.
  • the polypeptides as described herein may comprise a mixture of polypeptides with different N-termini.
  • the N-termini differ in length by 1, 2, 3, 4, or 5 amino acids.
  • the polypeptide is substantially homogeneous, i.e., the polypeptides have the same N-terminus.
  • the signal sequence of the polypeptide comprises one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, etc.) amino acid substitutions and/or deletions as compared to a “native” or “parental” signal sequence.
  • the signal sequence of the polypeptide comprises amino acid substitutions and/or deletions that allow one cleavage site to be dominant, thereby resulting in a substantially homogeneous polypeptide with one N-terminus.
  • a signal sequence of the polypeptide affects the expression level of the polypeptide, e.g., increased expression or decreased expression.
  • the anti-TIGIT antibodies compete for specific binding to TIGIT with the anti- TIGIT antibody comprising: (a) a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), and a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG) and (b) a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6).
  • the anti-TIGIT antibodies competes for specific binding to human TIGIT with the anti-TIGIT antibody comprising a heavy chain variable region comprising SEQ ID NO:7 and a light chain variable region comprising SEQ ID NOG. In certain embodiments, the anti-TIGIT antibodies competes for specific binding to human TIGIT with the anti-TIGIT antibody comprising a heavy chain comprising SEQ ID NO:9 and a light chain comprising SEQ ID NO: 10.
  • the anti-TIGIT antibodies compete with etigilimab for specific binding to human TIGIT.
  • the anti-TIGIT antibody competes with a reference antibody for specific binding to human TIGIT, wherein the reference antibody is etigilimab.
  • the anti-TIGIT antibody binds the same epitope, or essentially the same epitope, on TIGIT as etigilimab.
  • the anti-TIGIT antibody binds an epitope on TIGIT that overlaps with the epitope on human TIGIT bound by etigilimab.
  • the anti-TIGIT antibody competes for binding to an epitope comprising amino acids within SEQ ID NO: 13 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids within SEQ ID NO: 14 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids within SEQ ID NO:13 and SEQ ID NO:14 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62 and 1109 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein.
  • the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62 and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding with an epitope comprising amino acids Q64 and 1109 of SEQ ID NO:11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q64 and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein.
  • the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62, Q64, and 1109 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62, Q64, and T119 of SEQ ID NO:11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62, 1109, and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein.
  • the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q64, 1109, and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein.
  • the anti- TIGIT antibody competes for binding to an epitope comprising amino acids Q62, Q64, 1109, and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein.
  • the anti-TIGIT antibody competes for binding to an epitope comprising at least one amino acid selected from the group consisting of: N58, E60, Q62, Q64, L65, F107, 1109, Hl 11, T117, T119, G120, and R121 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein.
  • the anti-TIGIT antibody binds TIGIT and modulates TIGIT activity.
  • the anti-TIGIT antibody is a TIGIT antagonist and decreases TIGIT activity.
  • the anti-TIGIT antibody inhibits TIGIT activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%.
  • the anti-TIGIT antibody inhibits TIGIT activity is etigilimab.
  • the anti-TIGIT antibody binds TIGIT and inhibits or reduces TIGIT signalling. In certain embodiments, the anti-TIGIT antibody inhibits TIGIT signalling by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%.
  • TIGIT is phosphorylated at its cytoplasmic tail after interaction with its counterreceptor PVR.
  • the phosphorylation of TIGIT is the beginning of a cascade that includes downstream events affecting other known signalling pathways. Therefore, evaluating TIGIT phosphorylation can give information about TIGIT activity and TIGIT signalling.
  • Phosphorylation assays are known to those of skill in the art and are commonly used to monitor protein activation and/or pathway activation. The assays may be used to monitor the effect of various treatments on activation of a target protein and/or a target pathway. For example, an in vitro phosphorylation assay can be used to evaluate the effect of a TIGIT antagonist on the PVR-induced activation of TIGIT.
  • the anti-TIGIT antibody inhibits binding of TIGIT to a receptor. In certain embodiments, the anti-TIGIT antibody inhibits binding of TIGIT to PVR. In some embodiments, the anti-TIGIT antibody inhibits binding of TIGIT to PVR-L2, PVR- L3, and/or PVR-L4. In certain embodiments, the inhibition of binding of the anti-TIGIT antibody to PVR is at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%.
  • the inhibition of binding of the anti-TIGIT antibody to PVR-L2, PVR-L3, and/or PVR-L4 is at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%.
  • the anti-TIGIT antibody that inhibits binding of TIGIT to PVR is etigilimab.
  • the anti-TIGIT antibody that inhibits binding of TIGIT to PVR-L2, PVR-L3, and/or PVR-L4 is etigilimab.
  • the anti-TIGIT antibody blocks binding of TIGIT to a receptor.
  • the anti-TIGIT antibody blocks binding of TIGIT to PVR. In certain embodiments, the blocking of binding of the anti-TIGIT antibody to PVR is at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In some embodiments, the anti-TIGIT antibody blocks binding of TIGIT to PVRL2, PVRL3, and/or PVRL4. In certain embodiments, the blocking of binding of the anti- TIGIT antibody to PVRL2, PVRL3, and/or PVRL4 is at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In certain embodiments, the anti-TIGIT antibody that blocks binding of TIGIT to PVR is etigilimab.
  • Binding assays are known to those of skill in the art and are described herein. Binding assays may be used to monitor the effect of a test agent on the interaction between a target protein and the protein’s binding partner (e.g., receptor or ligand). For example, an in vitro binding assay can be used to evaluate if a TIGIT antagonist blocks the interaction of TIGIT to PVR.
  • binding partner e.g., receptor or ligand
  • the anti-TIGIT antibodies described herein have one or more of the following effects: inhibit proliferation of tumor cells, inhibit tumor growth, reduce the tumorigenicity of a tumor, reduce the tumorigenicity of a tumor by reducing the frequency of cancer stem cells in the tumor, trigger cell death of tumor cells, enhance or boost the immune response, enhance or boost the anti-tumor response, increase cytolytic activity of immune cells, increase killing of tumor cells, increase killing of tumor cells by immune cells, induce cells in a tumor to differentiate, differentiate tumorigenic cells to a non-tumorigenic state, induce expression of differentiation markers in the tumor cells, prevent metastasis of tumor cells, decrease survival of tumor cells, increase cell contact-dependent growth inhibition, increase tumor cell apoptosis, reduce epithelial mesenchymal transition (EMT), or decrease survival of tumor cells.
  • EMT epithelial mesenchymal transition
  • the anti-TIGIT antibodies inhibit tumor growth.
  • the anti-TIGIT antibodies inhibit tumor growth in vivo (e.g., in a mouse model, and/or in a human having cancer).
  • tumor growth is inhibited at least about two-fold, about three-fold, about five-fold, about ten-fold, about 50-fold, about 100- fold, or about 1000-fold as compared to an untreated tumor.
  • the anti-TIGIT antibodies bind TIGIT and modulate an immune response.
  • the anti-TIGIT antibody activates and/or increases an immune response.
  • the anti-TIGIT antibody increases, promotes, or enhances cell-mediated immunity.
  • the anti-TIGIT antibody increases, promotes, or enhances innate cell-mediated immunity.
  • the anti-TIGIT antibody increases, promotes, or enhances adaptive cell-mediated immunity.
  • the anti-TIGIT antibody increases, promotes, or enhances T-cell activity.
  • the anti-TIGIT antibody increases, promotes, or enhances cytolytic T-cell (CTL) activity.
  • CTL cytolytic T-cell
  • the antibody increases the activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%.
  • the anti-TIGIT antibody increases, promotes, or enhances NK cell activity.
  • the antibody increases the activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%.
  • the anti-TIGIT antibody increases, promotes, or enhances lymphokine-activated killer cell (LAK) activity.
  • the anti-TIGIT antibody increases, promotes, or enhances tumorinfiltrating lymphocyte (TIL) activity.
  • the anti-TIGIT antibody inhibits or decreases Treg cell activity. In some embodiments, the anti-TIGIT antibody inhibits or decreases MDSC activity. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances tumor cell killing. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances the inhibition of tumor growth.
  • the anti-TIGIT antibody induces and/or enhances a Thl- type immune response.
  • a Th 1 -type immune response includes production of interferon-gamma (IFN-y), IL-2, and tumor necrosis factor-beta (TNF-P).
  • a Th2-type immune response generally includes production of IL-4, IL-5, IL-6, IL-9, IL-10, and IL- 13.
  • the anti-TIGIT antibody induces and/or increases cytokine or lymphokine production.
  • the induction and/or increase in cytokine or lymphokines production may be an indirect effect.
  • the anti-TIGIT antibody increases activation of NK cells. In certain embodiments, the anti-TIGIT antibody increases activation of T-cells. In certain embodiments, the activation of NK cells and/or T-cells by the anti-TIGIT antibody results in an increase in the level of activation of NK cells and/or T-cells of at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In certain embodiments, the anti-TIGIT antibody that increases activation of NK cells is etigilimab.
  • the anti-TIGIT antibody is an antagonist of regulatory T- cell (Treg) activity.
  • the anti-TIGIT antibody inhibits or decreases the activity of Tregs.
  • the inhibition of activity of Tregs by the anti-TIGIT antibody results in an inhibition of suppressive activity of a Treg cell of at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, at least about 95%, or about 100%.
  • the anti-TIGIT antibody that inhibits Treg activity is etigilimab.
  • the anti-TIGIT antibody is an antagonist of myeloid- derived suppressor cells (MDSCs). In certain embodiments, the anti-TIGIT antibody inhibits MDSC activity. In certain embodiments, the anti-TIGIT antibody inhibits MDSC activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody that inhibits MDSC activity is etigilimab.
  • the anti-TIGIT antibody increases natural killer (NK) cell activity. In certain embodiments, the anti-TIGIT antibody increases NK cell activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody that increases NK cell activity is etigilimab.
  • the anti-TIGIT antibody increases tumor-infiltrating lymphocyte (TIL) activity. In certain embodiments, the anti-TIGIT antibody increases TIL activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody that increases TIL cell activity is etigilimab.
  • the anti-TIGIT antibody increases or enhances lymphokines-activated killer cell (LAK) activity. In certain embodiments, the anti-TIGIT antibody increases LAK activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody that increases LAK cell activity is etigilimab.
  • a functional assay that detects T-cell activation may be used.
  • a functional assay that detects T-cell proliferation may be used.
  • a functional assay that detects NK activity may be used.
  • a functional assay that detects CTL activity may be used.
  • a functional assay that detects Treg activity may be used.
  • a functional assay that detects MDSC activity may be used.
  • a functional assay that detects production of cytokines or lymphokines or cells producing cytokines or lymphokines may be used.
  • an ELISpot assay is used to measure antigen-specific T-cell frequency.
  • an ELISpot assay is used to measure cytokine release/production and/or used to measure the number of cytokine producing cells.
  • cytokine assays are used to identify a Thl-type response.
  • cytokine assays are used to identify a Th2-type response.
  • cytokine assays are used to identify a Thl7-type response.
  • FACS analysis is used to measure activation markers on immune cells, including but not limited to, T-cells, B-cells, NK cells, macrophages, and/or myeloid cells.
  • the anti-TIGIT antibodies have a circulating half-life in humans of at least about 2 hours, at least about 5 hours, at least about 10 hours, at least about 24 hours, at least about 3 days, at least about 1 week, or at least about 2 weeks.
  • the anti-TIGIT antibody is an IgG (e.g., IgGl, IgG2, or IgG4) antibody that has a circulating half-life in humans of at least about 2 hours, at least about 5 hours, at least about 10 hours, at least about 24 hours, at least about 3 days, at least about 1 week, or at least about 2 weeks.
  • known methods of increasing the circulating halflife of IgG antibodies include the introduction of mutations in the Fc region which increase the pH-dependent binding of the antibody to the neonatal Fc receptor (FcRn) at pH 6.0.
  • Known methods of increasing the circulating half-life of antibody fragments lacking the Fc region include such techniques as PEGylation.
  • the anti-TIGIT antibodies are used in any one of a number of conjugated (i.e. an immunoconjugate or radioconjugate) or non-conjugated forms.
  • the antibodies can be used in a non-conjugated form to harness the patient’s natural defense mechanisms including complement-dependent cytotoxicity (CDC) and antibody dependent cellular cytotoxicity (ADCC) to eliminate malignant or cancer cells.
  • CDC complement-dependent cytotoxicity
  • ADCC antibody dependent cellular cytotoxicity
  • the anti-TIGIT antibody is conjugated to a cytotoxic agent.
  • the anti-TIGIT antibody is conjugated to a cytotoxic agent as an ADC (antibody-drug conjugate).
  • the cytotoxic agent is a chemotherapeutic agent including, but not limited to, methotrexate, adriamycin/doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin, pyrrolobenzodiazepines (PBDs), or other intercalating agents.
  • chemotherapeutic agent including, but not limited to, methotrexate, adriamycin/doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin, pyrrolobenzodiazepines (PBDs), or other intercalating agents.
  • the cytotoxic agent is an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof, including, but not limited to, diphtheria A chain, non-binding active fragments of diphtheria toxin, exotoxin A chain, ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), Momordica charantia inhibitor, curcin, crotin, Sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • diphtheria A chain non-binding active fragments of diphtheria toxin
  • exotoxin A chain exotoxin A chain
  • ricin A chain abrin A chain
  • modeccin A chain
  • the cytotoxic agent is a radioisotope to produce a radioconjugate or a radioconjugated antibody.
  • radionuclides are available for the production of radioconjugated antibodies including, but not limited to, 90Y, 1251, 1311, 1231, Ulin, 131In, 105Rh, 153Sm, 67Cu, 67Ga, 166Ho, 177Lu, 186Re, 188Re and 212Bi.
  • Conjugates of an antibody and one or more small molecule toxins, such as calicheamicins, maytansinoids, trichothenes, and CC1065, and the derivatives of these toxins that have toxin activity, can also be used.
  • Conjugates of an antibody and cytotoxic agent may be made using a variety of bifunctional protein-coupling agents such as N-succinimidyl- 3-(2-pyridyidithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis(p- azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)- ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as l,5-difluoro-2,4-dinitrobenzene).
  • SPDP N-succinimi
  • the invention provides a fragment of an anti-TIGIT antibody, wherein the antibody fragment comprises an antigen-binding site.
  • the antibody fragment comprises an IgG antibody, an IgGl antibody, an IgG2 antibody, or an IgG4 antibody.
  • Antibody fragments may have different functions or capabilities than intact antibodies; for example, antibody fragments can have increased tumor penetration.
  • Various techniques are known for the production of antibody fragments including, but not limited to, proteolytic digestion of intact antibodies.
  • antibody fragments include a F(ab')2 fragment produced by pepsin digestion of an antibody molecule.
  • antibody fragments include a Fab fragment generated by reducing the disulfide bridges of an F(ab')2 fragment. In other embodiments, antibody fragments include a Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent. In certain embodiments, antibody fragments are produced by recombinant methods. In some embodiments, antibody fragments include Fv or single chain Fv (scFv) fragments. Fab, Fv, and scFv antibody fragments can be expressed in and secreted from E. coli or other host cells, allowing for the production of large amounts of these fragments. In some embodiments, antibody fragments are isolated from antibody phage libraries as discussed herein.
  • antibody fragments are linear antibody fragments.
  • antibody fragments are monospecific or bispecific.
  • the anti-TIGIT antibody is a scFv.
  • Various techniques can be used for the production of single -chain antibodies specific to TIGIT.
  • an antibody is modified in order to alter (e.g., increase or decrease) its serum half-life. This can be achieved, for example, by incorporation of a salvage receptor binding epitope into the antibody fragment by mutation of the appropriate region in the antibody fragment or by incorporating the epitope into a peptide tag that is then fused to the antibody fragment at either end or in the middle (e.g., by DNA or peptide synthesis).
  • the anti-TIGIT antibody is a bispecific antibody.
  • this invention encompasses bispecific anti-TIGIT antibody that specifically recognize TIGIT and at least one additional target.
  • Bispecific antibodies are capable of specifically recognizing and binding at least two different antigens or epitopes.
  • the different epitopes can either be within the same molecule (e.g., two epitopes on TIGIT) or on different molecules (e.g., one epitope on TIGIT and one epitope on a different protein).
  • a bispecific antibody has enhanced potency as compared to an individual antibody or to a combination of more than one antibody.
  • a bispecific antibody has reduced toxicity as compared to an individual antibody or to a combination of more than one antibody. It is known to those of skill in the art that any therapeutic agent may have unique pharmacokinetics (PK) (e.g., circulating half-life).
  • PK pharmacokinetics
  • a bispecific antibody has the ability to synchronize the PK of two active binding agents wherein the two individual binding agents have different PK profiles.
  • a bispecific antibody has the ability to concentrate the actions of two agents in a common area (e.g., a tumor and/or tumor microenvironment).
  • a bispecific antibody has the ability to concentrate the actions of two agents to a common target (e.g., a tumor or a tumor cell).
  • a bispecific antibody has the ability to target the actions of two agents to more than one biological pathway or function. In some embodiments, a bispecific antibody has the ability to target two different cells and bring them closer together (e.g., an immune cell and a tumor cell).
  • the bispecific antibody is a monoclonal antibody. In some embodiments, the bispecific antibody is a humanized antibody. In some embodiments, the bispecific antibody is a human antibody. In some embodiments, the bispecific antibody is an IgGl antibody. In some embodiments, the bispecific antibody is an IgG2 antibody. In some embodiments, the bispecific antibody is an IgG4 antibody. In some embodiments, the bispecific antibody has decreased toxicity and/or side effects. In some embodiments, the bispecific antibody has decreased toxicity and/or side effects as compared to a mixture of the two individual antibodies or the antibodies as single agents. In some embodiments, the bispecific antibody has an increased therapeutic index. In some embodiments, the bispecific antibody has an increased therapeutic index as compared to a mixture of the two individual antibodies or the antibodies as single agents.
  • the bispecific antibodies can specifically recognize and bind TIGIT as well as a second antigen target, such as CD2, CD3, CD28, CTLA4, PD-1, PD- Ll, CD80, CD86, CD64, CD32, or CD16, TIM-3, LAG-3, OX-40, 4-1BB, or GITR.
  • the bispecific antibodies can specifically recognize and bind TIGIT as well as specifically recognizing and binding to PD-1 or PD-L1.
  • the antibodies can be used to direct cytotoxic agents to cells which express a particular target antigen. These antibodies possess an antigen-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA, or TETA.
  • the bispecific antibodies comprise heavy chain constant regions with modifications in the amino acids which are part of the interface between the two heavy chains.
  • the bispecific antibodies can be generated using a “knobs-into-holes” strategy. In some cases, the “knobs” and “holes” terminology is replaced with the terms “protuberances” and “cavities”.
  • the bispecific antibodies may comprise variant hinge regions incapable of forming disulfide linkages between the heavy chains.
  • the modifications may comprise changes in amino acids that result in altered electrostatic interactions. In some embodiments, the modifications may comprise changes in amino acids that result in altered hydrophobic/hydrophilic interactions.
  • Bispecific antibodies can be intact antibodies or antibody fragments comprising antigen-binding sites. Antibodies with more than two valencies are also contemplated. For example, trispecific antibodies can be prepared. Thus, in certain embodiments the antibodies to TIGIT are multispecific.
  • the bispecific antibodies bind both TIGIT and the second target with a KD of about lOOnM or less.
  • the anti-TIGIT antibody binds both TIGIT and the second target with a KD of about 50nM or less.
  • the anti-TIGIT antibody binds both TIGIT and the second target with a KD of about 20nM or less.
  • the anti-TIGIT antibody binds both TIGIT and the second target with a KD of about lOnM or less.
  • the anti-TIGIT antibody binds both TIGIT and the second target with a KD of about InM or less.
  • the affinity of one of the antigen-binding sites may be weaker than the affinity of the other antigen-binding site.
  • the KD of one antigen binding site may be about InM and the KD of the second antigen-binding site may be about lOnM.
  • the anti-TIGIT antibody is a heterodimeric agent or heterodimeric molecule.
  • a heterodimeric molecule comprises a first arm which binds human TIGIT and a second arm which binds a second target.
  • a heterodimeric molecule comprises a first arm that specifically binds human TIGIT and a second arm, wherein the first arm comprises an anti-TIGIT antibody.
  • a heterodimeric molecule comprises a first arm that binds human TIGIT and a second arm which comprises an antigen-binding site from an antibody that specifically binds a second target.
  • a heterodimeric molecule is a bispecific antibody.
  • a heterodimeric molecule comprises a first arm that binds human TIGIT and a second arm that specifically binds a tumor antigen. In some embodiments, a heterodimeric molecule comprises a first arm that binds human TIGIT and a second arm that specifically binds PD-1, PD-L1, CTLA-4, CD2, CD3, CD28, CD80, CD86, CD64, CD32, CD16, TIM-3, LAG-3, OX-40, 4-1BB, or GITR. In some embodiments, a heterodimeric molecule comprises a first arm that binds TIGIT and a second arm that comprises an immunotherapeutic agent.
  • the immunotherapeutic agent is selected from the group consisting of: granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF), interleukin 2 (IL-2), interleukin 3 (IL-3), interleukin 12 (IL-12), interleukin 15 (IL-15), B7-1 (CD80), B7-2 (CD86), 4-1BB ligand, GITRL, OX-40L, anti-CD3 antibody, anti-CTLA-4 antibody, anti-PD-1 antibody, anti-PD-Ll antibody, anti-4-lBB antibody, anti-GITR antibody, anti-OX-40 antibody, anti-LAG-3 antibody, and anti-TIM-3 antibody.
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • M-CSF macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • IL-2 interleukin 2
  • IL-3 interleuk
  • the invention provides methods of treating various cancers using a combination of an anti-TIGIT antibody and an anti-PD-1 antibody.
  • anti-PD-1 antibodies include pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo),dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab.
  • the anti-PD-1 antibody is selected from the group consisting of pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo), dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab.
  • the invention is exemplified with data based on the combination of etigilimab and the anti-PD-1 antibody nivolumab. Without wishing to be bound by theory, the inventors rationalise that the surprising technical effects achieved by this combination would also be seen with other anti-PD-1 antibodies described herein. The surprising technical effects of this combination are thought to be principally attributable to etigilimab and its functionality rather than the anti-PD-1 antibody component of the combination therapy.
  • PD-1 and its ligand PD-L1 perform a role in tumor progression and survival by escaping tumor neutralizing immune surveillance.
  • Anti-PD- 1 and anti-PD-Ll antibodies act by blocking the binding of PD-1 to its ligand PD-L1 which prevents cancer cells from evading the immune system.
  • the inventors hypothesize that the surprising technical effects achieved by nivolumab (anti-PD-1 antibody) in combination with etigilimab would also be seen with other anti-PD-1 and PD-L1 antibodies described herein as all of these antibodies act by blocking PD-1/PD-L1 signalling.
  • the heavy chain sequence of pembrolizumab is SEQ ID NO: 15.
  • the light chain sequence of pembrolizumab is SEQ ID NO: 16.
  • the heavy chain sequence of nivolumab is SEQ ID NO: 17.
  • the light chain sequence of nivolumab is SEQ ID NO: 18.
  • the heavy chain sequence of cemiplimab is SEQ ID NO: 19.
  • the light chain sequence of cemiplimab is SEQ ID NO:20.
  • the heavy chain sequence of dostarlimab is SEQ ID NO:21.
  • the light chain sequence of dostarlimab is SEQ ID NO:22.
  • the heavy chain sequence of Tislelizumab is SEQ ID NO:40.
  • the light chain sequence of Tislelizumab is SEQ ID NO:41.
  • the heavy chain sequence of Sintilimab is SEQ ID NO:42.
  • the light chain sequence of Sintilimab is SEQ ID NO:43.
  • the heavy chain sequence of Zimberelimab is SEQ ID NO:44.
  • the light chain sequence of Zimberelimab is SEQ ID NO:45.
  • the heavy chain sequence of Toripalimab is SEQ ID NO:46.
  • the light chain sequence of Toripalimab is SEQ ID NO:47.
  • the heavy chain sequence of Penpulimab is SEQ ID NO:48.
  • the light chain sequence of Penpulimab is SEQ ID NO:49.
  • the heavy chain sequence of Balstilimab is SEQ ID NO:50.
  • the light chain sequence of Balstilimab is SEQ ID N0:51.
  • the heavy chain sequence of Retifanlimab is SEQ ID NO:52.
  • the light chain sequence of Retifanlimab is SEQ ID NO:53.
  • the light chain sequence of Cetrelimab is SEQ ID NO:54.
  • the heavy chain sequence of Cetrelimab is SEQ ID NO:55.
  • the light chain sequence of Budigalimab is SEQ ID NO:56.
  • the light chain sequence of Budigalimab is SEQ ID NO:57.
  • the heavy chain sequence of Pimivalimab is SEQ ID NO:58.
  • the light chain sequence of Pimivalimab is SEQ ID NO:59.
  • the light chain sequence of Spartalizumab is SEQ ID NO:60.
  • the heavy chain sequence of Spartalizumab is SEQ ID N0:61.
  • the light chain sequence of Serplulimab is SEQ ID NO:62.
  • the light chain sequence of Serplulimab is SEQ ID NO:63.
  • the heavy chain sequence of Sasanlimab is SEQ ID NO:63.
  • the light chain sequence of Sasanlimab is SEQ ID NO:65.
  • the anti-PD-1 antibody comprises a heavy chain comprising SEQ ID NO: 15 and a light chain comprising SEQ ID NO: 16. In some embodiments, the anti-PD-1 antibody comprises a heavy chain comprising SEQ ID NO: 17 and a light chain comprising SEQ ID NO: 18. In some embodiments, the anti-PD-1 antibody comprises a heavy chain comprising SEQ ID NO: 19 and a light chain comprising SEQ ID NO:20. In some embodiments, the anti-PD-1 antibody comprises a heavy chain comprising SEQ ID NO:21 and a light chain comprising SEQ ID NO:22.
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and nivolumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSD
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and pembrolizumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and cemiplimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and dostarlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Tislelizumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYA
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Sintilimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYA
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Zimberelimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Toripalimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Penpulimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Balstilimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID N0:4), a light chain CDR2 comprising SASYRYT (SEQ ID NO:5), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising T
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Retifanlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Cetrelimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Budigalimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Pimivalimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSD
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and spartalizumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYA
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Camrelizumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Prolgolimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Pucotenlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising T
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and serplulimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and sasanlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOH), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSD
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and ezabenlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOH), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSD
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and genolimzumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD-Ll antibody, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSD
  • Non-limiting examples of anti-PD-Ll antibodies include atezolizumab (Tecentriq), avelumab (Bavencio),durvalumab (Imfinzi), Envafolimab, Sugemalimab, Socazolimab, Adebrelimab, Pacmilimab, Lodapolimab and Cosibelimab.
  • the anti-PD-Ll antibody is selected from the group consisting of atezolizumab (Tecentriq), avelumab (Bavencio), durvalumab (Imfinzi), Envafolimab, Sugemalimab, Socazolimab, Adebrelimab, Pacmilimab, Lodapolimab and Cosibelimab.
  • the heavy chain sequence of atezolizumab is SEQ ID NO:23.
  • the light chain sequence of atezolizumab is SEQ ID NO:24.
  • the heavy chain sequence of avelumab is SEQ ID NO:25.
  • the light chain sequence of avelumab is SEQ ID NO:26.
  • the heavy chain sequence of durvalumab is SEQ ID NO:27.
  • the light chain sequence of durvalumab is SEQ ID NO:28.
  • the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:23 and a light chain comprising SEQ ID NO:24. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:25 and a light chain comprising SEQ ID NO:26. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:27 and a light chain comprising SEQ ID NO:28. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:66 and a light chain comprising SEQ ID NO: 67.
  • the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO: 68 and a light chain comprising SEQ ID NO:69. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:70 and a light chain comprising SEQ ID NO:71. In some embodiments, the anti- PD-L1 antibody comprises a heavy chain comprising SEQ ID NO:72 and a light chain comprising SEQ ID NO:73. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:74 and a light chain comprising SEQ ID NO:75.
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD- 1 antibody, wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the invention provides methods of treating cancer in a human patient, comprising administering to the human patient an anti-TIGIT antibody and an anti-PD-1 antibody, wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the invention also provides an anti-TIGIT antibody and an anti-PD-1 antibody for use in treating cancer in a human patient, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR
  • the invention also provides the use of an anti-TIGIT antibody and an anti-PD-1 antibody in the preparation of a medicament for treating cancer in a human patient, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
  • the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor
  • Soft tissue sarcomas are a rare type of cancer that start in soft tissues (muscle, tendons, fat, lymph and blood vessels, and nerves). These cancers can develop anywhere in the body but are found mostly in the arms, legs, chest, and abdomen.
  • Types of soft tissue sarcoma include leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, fibrosarcomas, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid haemangioendothelioma, malignant peripheral nerve sheath tumors (MPNST), gastrointestinal Stromal Tumors (GISTs), follicular dendritic cell sarcoma, epithelioid sarcoma, myxofibrosarcoma, solitary fibrous tumor, undifferentiated (pleomorphic sarcoma), fibromatosis or dermatofibrosarcoma protuberans.
  • leiomyosarcoma inflammatory leiomyosarcoma, liposarcoma, fibrosarcomas, rhabdomyosarcomas, syn
  • the invention provides a method of treating a soft tissue sarcoma in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD-1 antibody, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6).
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ
  • the soft tissue sarcoma is selected from the group consisting of leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, dedifferentiated liposarcoma, fibrosarcomas, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid haemangioendothelioma, malignant peripheral nerve sheath tumours (MPNST), gastrointestinal Stromal Tumours (GISTs), follicular dendritic cell sarcoma, epithelioid sarcoma, myxofibrosarcoma, solitary fibrous tumor, undifferentiated (pleomorphic sarcoma), fibromatosis and dermatofibrosarcoma protuberans.
  • leiomyosarcoma inflammatory leiomyosarcoma, liposarcoma, dedifferentiated
  • the soft tissue sarcoma is selected from the group consisting of leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, dedifferentiated liposarcoma, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid, malignant peripheral nerve sheath tumours (MPNST) and undifferentiated (pleomorphic sarcoma).
  • the soft tissue sarcoma is a dedifferentiated liposarcoma or a undifferentiated (pleomorphic sarcoma).
  • testicular cancers About 95 percent of testicular cancers begin in germ cells. Two of the most common germ cell tumors are seminoma and nonseminoma. Seminomas make up about half of all germ cell tumors. They usually grow slowly. Nonseminomas are often more aggressive than seminomas, and more likely to spread beyond the testicle. These types occur about equally. Many testicular cancers contain both seminoma and non-seminoma cells. These mixed germ cell tumors are treated as non-seminomas because they grow and spread like non-seminomas.
  • the invention provides a method of treating a testicular germ cell tumor in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD-1 antibody, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG).
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO
  • the testicular germ cell tumor is a seminomas testicular germ cell tumor or a non-seminomas testicular germ cell tumor. In some embodiments, the testicular germ cell tumor comprises both seminoma and non-seminoma cells. In some embodiments, the testicular germ cell tumor patient has inadequate renal function for high dose chemotherapy. In some embodiments, the testicular germ cell tumor patient has inadequate stem cell collection for high dose chemotherapy.
  • Uveal melanoma is a rare cancer of the eye involving the iris, ciliary body, or choroid (collectively referred to as the uvea). Tumors arise from the pigment cells (melanocytes) that reside within the uvea and give color to the eye. These melanocytes are distinct from the retinal pigment epithelium cells underlying the retina that do not form melanomas. Uveal melanomas are sometimes referred to by their location, choroidal melanoma, ciliary body melanoma, or iris melanoma.
  • Iris and ciliary body melanomas can be described as anterior melanomas, whereas a choroidal melanoma behind the equator of the eye would be considered posterior uveal melanomas.
  • uveal melanomas include posterior uveal melanoma, choroidal melanoma, ciliary body melanoma and iris melanoma.
  • the invention provides a method of treating uveal melanoma in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti- TIGIT antibody and an anti-PD-1 antibody, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG).
  • the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG
  • the uveal melanoma is selected from the group consisting of a choroidal melanoma, a ciliary body melanoma or an iris melanoma. In some embodiments, the uveal melanoma is an anterior uveal melanoma or a posterior uveal melanomas.
  • the anti-TIGIT antibody can be administered prior to, concurrently with and/or subsequently to the administration of the anti-PD-1 antibody in order to treat the cancer in human patient.
  • Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously. It will be appreciated that the combination of an anti- TIGIT antibody and an anti-PD- 1 antibody may be administered in any order or concurrently.
  • the anti-TIGIT antibody and the anti-PD-1 antibody can be administered in any number of ways for either local or systemic treatment.
  • administration is (i) topical by epidermal or transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders; (ii) pulmonary by inhalation or insufflation of powders or aerosols, including by nebulizer, intratracheal, and intranasal; (iii) oral; or (iv) parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e.g., injection or infusion), or intracranial (e.g., intrathecal or intraventricular).
  • the anti-TIGIT antibodies and the anti-PD- 1 antibodies described herein are administered intravenously.
  • the anti-TIGIT antibodies and the anti-PD-1 antibodies described herein are administered subcutaneously.
  • the anti-TIGIT antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks. In certain embodiments, the anti-TIGIT antibody is administered once every two weeks. In some embodiments, the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks. In some embodiments, both the anti-TIGIT antibody and the anti-PD-1 antibody are administered once every week, once every two weeks, once every three weeks, or once every four weeks. In other embodiments, the anti-TIGIT antibody and the anti-PD-1 antibody are administered at different intervals.
  • the anti-TIGIT antibody is administered once every two weeks and the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks.
  • etigilimab is administered once every two weeks and the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks.
  • the anti-TIGIT antibody is administered once every two weeks and the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks, once every five weeks or once every six weeks.
  • etigilimab is administered once every two weeks and the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, once every four weeks, once every five weeks or once every six weeks.
  • etigilimab is administered once every two weeks and pembrolizumab is administered once every three weeks, or once every six weeks. In some embodiments, etigilimab is administered once every two weeks and nivolumab is administered once every two weeks, or once every four weeks. In some embodiments, etigilimab is administered once every two weeks and cemiplimab is administered once every three weeks. In some embodiments, etigilimab is administered once every two weeks and dostarlimab is administered once every three weeks, or once every six weeks.
  • the anti-TIGIT antibody is administered once every two weeks at a flat dose of 1000 mg.
  • the anti-PD-1 antibody is administered at a flat dose of 200 mg-1000 mg.
  • the anti-PD-1 antibody is administered at a flat dose of 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg or 1000 mg.
  • the -PD-1 antibody is administered at a flat dose of 200 mg, 240 mg, 350 mg, 400mg, 500 mg or 1000 mg.
  • etigilimab is administered once every two weeks at a flat dose of 1000 mg and pembrolizumab is administered once every three weeks at a flat dose of 200 mg, or once every six weeks at a flat dose of 400 mg. In some embodiments, etigilimab is administered once every two weeks at a flat dose of 1000 mg and nivolumab is administered once every two weeks at a flat dose of 240 mg, or once every four weeks at a flat dose of 480 mg. In some embodiments, etigilimab is administered once every two weeks at a flat dose of 1000 mg and cemiplimab is administered once every three weeks at a flat dose of 350 mg. In some embodiments, etigilimab is administered once every two weeks at a flat dose of 1000 mg and dostarlimab is administered once every three weeks at a flat dose of 500 mg, or once every six weeks at a flat dose of 1000 mg.
  • the human patient can be an infant (between the ages 0-1 years), a child (between the ages of 1-18 years) or an adult (with an age above 18 years). In some embodiments, the patient is aged above 18 years old, 20 years old, 30 years old or 40 years old. In certain embodiments, the human patient is aged above 50 years old, for example 55 years or older, 60 years or older, 65 years or older, 70 years or older, 75 years or older or 80 years or older. [00157]
  • the anti-TIGIT antibody and the anti-PD-1 antibody can be administered to patients that have previously undergone treatment with an anti-cancer agent.
  • the human patient has previously undergone treatment with one or more anti-cancer agents, such as with two or more or three or more anti-cancer agents.
  • the human patient has previously received one, two or three anti-cancer agents.
  • the human patient has been previously treated with one or more, two or more or three or more anti -cancer agents and there is tumour growth, progression, or recurrence during or after treatment with the one or more, two or more or three or more anti-cancer agents.
  • the anti-TIGIT antibody, the anti-PD-1 antibody and the anti-cancer agent agent will be administered substantially simultaneously or concurrently.
  • a patient may be given the anti-TIGIT antibody and the anti-PD-1 antibody while undergoing a course of treatment with an anti-cancer agent (e.g., chemotherapy).
  • an anti-cancer agent e.g., chemotherapy
  • the anti-TIGIT antibody and the anti-PD-1 antibody will be administered within 1 year of the treatment with the anti-cancer agent.
  • the anti-TIGIT antibody and the anti-PD-1 antibody will be administered within 10, 8, 6, 4, or 2 months of any treatment with the anti-cancer agent.
  • the anti-TIGIT antibody and the anti-PD-1 antibody will be administered within 4, 3, 2, or 1 weeks of any treatment with the anti-cancer agent.
  • the anti-TIGIT antibody and the anti-PD-1 antibody will be administered within 5, 4, 3, 2, or 1 days of any treatment with the anti-cancer agent. It will further be appreciated that the two (or more) anti-cancer agents or treatments may be administered to the patient within a matter of hours or minutes (i.e., substantially simultaneously).
  • the anti-cancer agent can be a checkpoint inhibitor, a chemotherapeutic agent or a cell therapy.
  • the human patient has not received prior treatment been treated with a checkpoint inhibitor. In some embodiments, the human patient is naive to checkpoint inhibitor therapy. In some embodiments, the human patient has not been previously treated with a checkpoint inhibitor. In some embodiments, the human patient has not received prior treatment/ been treated with an anti-PD-1 antibody therapy. In some embodiments, the human patient is naive to anti-PD-1 antibody therapy. In some embodiments, the human patient has not been previously treated with an anti-PD-1 antibody therapy. In some embodiments, the human patient has not received prior treatment/ been treated with an anti-PD-Ll antibody therapy. In some embodiments, the human patient is naive to anti-PD-Ll antibody therapy.
  • the human patient has not been previously treated with an anti-PD-Ll antibody therapy. In some embodiments, the human patient has not received prior treatment/ been treated with a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1. In some embodiments, the human patient is naive to a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1. In some embodiments, the human patient has not been previously treated with a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1.
  • the human patient has not received prior radiotherapy. In some embodiments, the human patient has not been previously treated with radiotherapy. In some embodiments, the human patient has not received prior chemotherapy. In some embodiments, the human patient has not been previously treated with chemotherapy.
  • the human patient has previously treated with a checkpoint inhibitor and there is tumor growth, progression, or recurrence during or after treatment with the checkpoint inhibitor.
  • the human patient has been previously treated with an anti-PD-1 antibody and there is tumor growth, progression, or recurrence during or after treatment with the PD-1 antibody.
  • the human patient has been previously treated with an anti-PD-Ll antibody and there is tumor growth, progression, or recurrence during or after treatment with the PD-L1 antibody.
  • the human patient has previously treated with a cell therapy and there is tumor growth, progression, or recurrence during or after treatment with the cell therapy.
  • the human patient has been previously treated with CAR T cell therapy and there is tumor growth, progression, or recurrence during or after treatment with the CAR T cell therapy.
  • the human patient has been previously treated with a TCR cell therapy and there is tumor growth, progression, or recurrence during or after treatment with the TCR cell therapy.
  • the human patient has previously treated with an immunotherapeutic agent and there is tumor growth, progression, or recurrence during or after treatment with the immunotherapeutic agent.
  • the human patient has been previously treated with tebentafusp and there is tumor growth, progression, or recurrence during or after treatment with tebentafusp.
  • the human patient has previously treated with a chemotherapeutic agent and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agent, for example a chemotherapeutic agent or in combination with a cocktail of chemotherapeutic agents.
  • Classes of therapeutic agents include, for example, anti-tubulin agents, auristatins, DNA minor groove binders, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cisplatin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and carboplatin), anthracyclines, antibiotics, anti-folates, antimetabolites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinols, purine antimetabolites, puromycins, radiation sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like.
  • alkylating agents e.g., platinum complexes such as cisplatin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and
  • Possible chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamime; nitrogen mustards such as chlorambucil, chlornaphazine, cyclophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmus
  • paclitaxel TAXOL
  • docetaxel TAXOTERE
  • chlorambucil gemcitabine
  • 6-thioguanine mercaptopurine
  • platinum analogs such as cisplatin and carboplatin
  • vinblastine platinum
  • etoposide VP-16
  • ifosfamide mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; ibandronate; CPT11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine (XELODA); and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • DMFO difluoromethylornithine
  • XELODA retinoic acid
  • esperamicins capecitabine
  • Chemotherapeutic agents also include anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (FARESTON); and antiandrogens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • the human patient has previously treated with a platinum-based therapies such as cisplatin or carboplatin.
  • the patient has previously been treated with Lenvatinib.
  • the patient has previously been treated with an anti-LAG3 antibody, for example Relatlimab, PRS-332, P13B02-3, LBL-007, Eftilagimod alpha (IMP321), LAG525 (IMP701), MK-4280, BI 754111, FS118, Tebotelimab, TSR-033, INCAGN2385, Sym022, XmAb22841 or REGN3767.
  • the patient has previously been treated with Relatlimab.
  • the patient has previously been treated with pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo), dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab.
  • pembrolizumab Keytruda
  • nivolumab Opdivo
  • cemiplimab Libtayo
  • dostarlimab Jemperli
  • Tislelizumab Tislelizumab, Sintilimab,
  • the patient has previously been treated with atezolizumab (Tecentriq), avelumab (Bavencio), durvalumab (Imfinzi), Envafolimab, Sugemalimab, Socazolimab, Adebrelimab, Pacmilimab, Lodapolimab and Cosibelimab.
  • Tecentriq avelumab
  • durvalumab Imfinzi
  • Envafolimab Sugemalimab
  • Socazolimab Adebrelimab
  • Pacmilimab Lodapolimab
  • Cosibelimab Cosibelimab
  • the chemotherapeutic agent is a topoisomerase inhibitor.
  • Topoisomerase inhibitors are chemotherapy agents that interfere with the action of a topoisomerase enzyme (e.g., topoisomerase I or II).
  • Topoisomerase inhibitors include, but are not limited to, doxorubicin HC1, daunorubicin citrate, mitoxantrone HC1, actinomycin D, etoposide, topotecan HC1, teniposide (VM-26), and irinotecan, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these.
  • the chemotherapeutic agent is irinotecan.
  • the chemotherapeutic agent is an anti-metabolite.
  • An anti-metabolite is a chemical with a structure that is similar to a metabolite required for normal biochemical reactions, yet different enough to interfere with one or more normal functions of cells, such as cell division.
  • Anti-metabolites include, but are not limited to, gemcitabine, fluorouracil, capecitabine, methotrexate sodium, ralitrexed, pemetrexed, tegafur, cytosine arabinoside, thioguanine, 5-azacytidine, 6-mercaptopurine, azathioprine, 6-thioguanine, pentostatin, fludarabine phosphate, and cladribine, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these.
  • the chemotherapeutic agent is gemcitabine.
  • the chemotherapeutic agent is an antimitotic agent, including, but not limited to, agents that bind tubulin.
  • the agent is a taxane.
  • the agent is paclitaxel or docetaxel, or a pharmaceutically acceptable salt, acid, or derivative of paclitaxel or docetaxel.
  • the agent is paclitaxel (TAXOL), docetaxel (TAXOTERE), albumin-bound paclitaxel (ABRAXANE®), DHA-paclitaxel, or PG-paclitaxel.
  • the antimitotic agent comprises a vinca alkaloid, such as vincristine, vinblastine, vinorelbine, or vindesine, or pharmaceutically acceptable salts, acids, or derivatives thereof.
  • the antimitotic agent is an inhibitor of kinesin Eg5 or an inhibitor of a mitotic kinase such as Aurora A or Plkl.
  • the chemotherapeutic agent is paclitaxel.
  • the chemotherapeutic agent is albumin-bound paclitaxel (ABRAXANE®).
  • the human patient has previously treated with 2 or more chemotherapeutic agents and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agents.
  • the human patient has previously treated with 2 or more, 3 or more, 4 or more, or 5 or more chemotherapeutic agents.
  • Known drugs that are approved to treat soft tissue sarcoma include Cosmegen (Dactinomycin), Doxorubicin Hydrochloride, Eribulin Mesylate, Gleevec (Imatinib Mesylate), Halaven (Eribulin Mesylate), Imatinib Mesylate, Pazopanib Hydrochloride, Tazemetostat Hydrobromide, Tazverik (Tazemetostat Hydrobromide), Trabectedin, Votrient (Pazopanib Hydrochloride) and Yondelis (Trabectedin).
  • Cosmegen Dactinomycin
  • Doxorubicin Hydrochloride Eribulin Mesylate
  • Gleevec Imatinib Mesylate
  • Halaven Eribulin Mesylate
  • Imatinib Mesylate Imatinib Mesylate
  • Pazopanib Hydrochloride Tazemetostat Hydrobromide
  • Tazverik Tazemetostat Hydro
  • the human patient has been previously treated with one of more chemotherapeutic agents, and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agent, wherein the chemotherapeutic agent is selected from the group consisting of Cosmegen (Dactinomycin), Doxorubicin Hydrochloride, Eribulin Mesylate, Gleevec (Imatinib Mesylate), Halaven (Eribulin Mesylate), Imatinib Mesylate, Pazopanib Hydrochloride, Tazemetostat Hydrobromide, Tazverik (Tazemetostat Hydrobromide), Trabectedin, Votrient (Pazopanib Hydrochloride) and Yondelis (Trabectedin).
  • Cosmegen Dactinomycin
  • Doxorubicin Hydrochloride Eribulin Mesylate
  • Gleevec Imatinib Mesylate
  • Halaven Eribulin Mesylate
  • a known combination of drugs that is approved to treat a soft tissue sarcoma is Vincristine Sulfate, Dactinomycin and Cyclophosphamide.
  • the human patient has previously treated with 3 or more chemotherapeutic agents and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agents, wherein the 3 or more chemotherapeutic agents are Vincristine Sulfate, Dactinomycin and Cyclophosphamide,
  • Known drugs that are approved to treat testicular germ cell tumor include Bleomycin Sulfate, Cisplatin, Cosmegen (Dactinomycin), Etopophos (Etoposide Phosphate), Etoposide, Etoposide Phosphate, Ifex (Ifosfamide) and Vinblastine Sulfate.
  • the human patient has been previously treated with one of more chemotherapeutic agents, and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agent, wherein the chemotherapeutic agent is selected from the group consisting of Bleomycin Sulfate, Cisplatin, Cosmegen (Dactinomycin), Etopophos (Etoposide Phosphate), Etoposide, Etoposide Phosphate, Ifex (Ifosfamide) and Vinblastine Sulfate.
  • the chemotherapeutic agent is selected from the group consisting of Bleomycin Sulfate, Cisplatin, Cosmegen (Dactinomycin), Etopophos (Etoposide Phosphate), Etoposide, Etoposide Phosphate, Ifex (Ifosfamide) and Vinblastine Sulfate.
  • Known combinations of drugs that are approved to treat testicular germ cell tumor include: (a) Bleomycin, Etoposide Phosphate and Cisplatin; (b) Bleomycin, Etoposide Phosphate and Carboplatin; (c) Vinblastine Sulfate, Ifosfamide and Cisplatin and (d) Etoposide, Ifosfamide and Cisplatin.
  • the human patient has previously treated with 3 or more chemotherapeutic agents and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agents, wherein the 3 or more chemotherapeutic agents are selected from the group consisting of (a) Bleomycin, Etoposide Phosphate and Cisplatin; (b) Bleomycin, Etoposide Phosphate and Carboplatin; (c) Vinblastine Sulfate, Ifosfamide and Cisplatin and (d) Etoposide, Ifosfamide and Cisplatin.
  • the 3 or more chemotherapeutic agents are selected from the group consisting of (a) Bleomycin, Etoposide Phosphate and Cisplatin; (b) Bleomycin, Etoposide Phosphate and Carboplatin; (c) Vinblastine Sulfate, Ifosfamide and Cisplatin and (d) Etoposide, Ifosfamide and Cisplatin
  • Known treatments for uveal melanoma include surgery, photocoagulation and thermotherapy.
  • the following types of surgery may be used: (a) Resection: Surgery to remove the tumor and a small amount of healthy tissue around it; (b) Enucleation: Surgery to remove the eye and part of the optic nerve. This is done if vision cannot be saved and the tumor is large, has spread to the optic nerve, or causes high pressure inside the eye. After surgery, the patient is usually fitted for an artificial eye to match the size and color of the other eye; and (c) Exenteration: Surgery to remove the eye and eyelid, and muscles, nerves, and fat in the eye socket. After surgery, the patient may be fitted for an artificial eye to match the size and color of the other eye or a facial prosthesis.
  • the human patient has previously undergone surgery. In some embodiments, the surgery was resection, enucleation or exenteration.
  • Photocoagulation is a procedure that uses laser light to destroy blood vessels that bring nutrients to the tumor, causing the tumor cells to die. Photocoagulation may be used to treat small tumors. This is also called light coagulation.
  • the human patient has previously received photocoagulation therapy.
  • the human patient has previously been treated with photocoagulation therapy.
  • Thermotherapy is the use of heat from a laser to destroy cancer cells and shrink the tumor.
  • the human patient has previously received thermo therapy.
  • the human patient has previously been treated with thermotherapy.
  • the anti-TIGIT and anti-PD-1 antibodies can be provided as a pharmaceutical composition.
  • the pharmaceutical composition may be formulated with a pharmaceutically acceptable carrier.
  • Formulations are prepared for storage and use by combining a purified antibody with a pharmaceutically acceptable vehicle (e.g., a carrier or excipient).
  • a pharmaceutically acceptable vehicle e.g., a carrier or excipient.
  • Pharmaceutically acceptable carriers include a sterile aqueous solution. Those of skill in the art generally consider pharmaceutically acceptable carriers, excipients, and/or stabilizers to be inactive ingredients of a formulation or pharmaceutical composition.
  • Suitable pharmaceutically acceptable vehicles include, but are not limited to, nontoxic buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives such as octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol, alkyl parabens, such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol; low molecular weight polypeptides (e.g., less than about 10 amino acid residues); proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • compositions of the present invention can be administered in any number of ways for either local or systemic treatment. Administration can be topical by epidermal or transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders; pulmonary by inhalation or insufflation of powders or aerosols, including by nebulizer, intratracheal, and intranasal; oral; or parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e.g., injection or infusion), or intracranial (e.g., intrathecal or intraventricular).
  • the pharmaceutical compositions comprising the anti-TIGIT and anti-PD-1 antibodies is administered intravenously.
  • kits that comprise the anti-TIGIT antibodies and anti-PD-1 antibodies as described herein and that can be used to perform the methods described herein.
  • a kit comprises at least one purified anti-TIGIT antibody in one or more containers.
  • the disclosed anti- TIGIT antibodies and anti-PD-1 antibodies can be readily incorporated into one of the established kit formats which are well known in the art.
  • composition “comprising” encompasses “including” as well as “consisting”, “consisting of’ and/or “consisting essentially of’ e.g. a composition “comprising” X may consist exclusively of X or may include something additional e.g. X + Y. It is also understood that wherever embodiments are described herein with the language “consisting essentially of’ otherwise analogous embodiments described in terms of “consisting of’ are also provided.
  • any reference to a method for treatment comprising administering an agent to a patient, also covers that agent for use in said method for treatment, as well as the use of the agent in said method for treatment, and the use of the agent in the manufacture of a medicament.
  • the terms “agonist” and “agonistic” as used herein refer to or describe an agent that is capable of, directly or indirectly, substantially inducing, activating, promoting, increasing, or enhancing the biological activity of a target and/or a pathway.
  • agonist is used herein to include any agent that partially or fully induces, activates, promotes, increases, or enhances the activity of a protein.
  • antagonists refer to or describe an agent that is capable of, directly or indirectly, partially or fully blocking, inhibiting, reducing, or neutralizing a biological activity of a target and/or pathway.
  • antagonist is used herein to include any agent that partially or fully blocks, inhibits, reduces, or neutralizes the activity of a protein.
  • modulation and “modulate” as used herein refer to a change or an alteration in a biological activity. Modulation includes, but is not limited to, stimulating an activity or inhibiting an activity. Modulation may be an increase or a decrease in activity, a change in binding characteristics, or any other change in the biological, functional, or immunological properties associated with the activity of a protein, a pathway, a system, or other biological targets of interest.
  • antibody refers to an immunoglobulin molecule that recognizes and specifically binds a target through at least one antigen-binding site.
  • the target may be a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or a combination of any of the foregoing.
  • the term encompasses intact polyclonal antibodies, intact monoclonal antibodies, antibody fragments (such as Fab, Fab', F(ab')2, and Fv fragments), single chain Fv (scFv) antibodies, multispecific antibodies, bispecific antibodies, monospecific antibodies, monovalent antibodies, chimeric antibodies, humanized antibodies, human antibodies, fusion proteins comprising an antigen-binding site of an antibody, and any other modified immunoglobulin molecule comprising an antigen-binding site as long as the antibodies exhibit the desired biological activity.
  • antibody fragments such as Fab, Fab', F(ab')2, and Fv fragments
  • scFv single chain Fv
  • An antibody can be any of the five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, or subclasses (isotypes) thereof (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2), based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu, respectively.
  • the different classes of immunoglobulins have different and well-known subunit structures and three-dimensional configurations.
  • Antibodies can be naked or conjugated to other molecules, including but not limited to, toxins and radioisotopes.
  • antibody fragment refers to a portion of an intact antibody and generally refers to the antigenic determining variable regions of an intact antibody.
  • antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, and Fv fragments, linear antibodies, single chain antibodies, and multispecific antibodies formed from antibody fragments.
  • Antibody fragment as used herein comprises an antigen-binding site or epitopebinding site.
  • variable region of an antibody refers to the variable region of an antibody light chain or the variable region of an antibody heavy chain, either alone or in combination.
  • variable region of a heavy chain or a light chain consists of four framework regions connected by three complementarity determining regions (CDRs), also known as “hypervariable regions”.
  • CDRs complementarity determining regions
  • the CDRs in each chain are held together in close proximity by the framework regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site(s) of the antibody.
  • CDRs There are at least two techniques for determining CDRs: (1) an approach based on cross-species sequence variability (i.e., Kabat et al., 1991, Sequences of Proteins of Immunological Interest, 5th Edition, National Institutes of Health, Bethesda MD.), and (2) an approach based on crystallographic studies of antigen-antibody complexes (Al Eazikani et al., 1997, J. Mol. Biol., 273:927-948). In addition, combinations of these two approaches are sometimes used in the art to determine CDRs.
  • the term “monoclonal antibody” as used herein refers to a homogenous antibody population involved in the highly specific recognition and binding of a single antigenic determinant or epitope. This is in contrast to polyclonal antibodies that typically include a mixture of different antibodies that recognize different antigenic determinants.
  • the term “monoclonal antibody” encompasses both intact and full-length monoclonal antibodies as well as antibody fragments (e.g., Fab, Fab', F(ab')2, Fv), single chain (scFv) antibodies, fusion proteins comprising an antibody fragment, and any other modified immunoglobulin molecule comprising an antigen-binding site.
  • “monoclonal antibody” refers to such antibodies made by any number of techniques, including but not limited to, hybridoma production, phage selection, recombinant expression, and transgenic animals.
  • humanized antibody refers to antibodies that are specific immunoglobulin chains, chimeric immunoglobulins, or fragments thereof that contain minimal non-human sequences.
  • humanized antibodies are human immunoglobulins in which amino acid residues of the CDRs are replaced by amino acid residues from the CDRs of a non-human species (e.g., mouse, rat, rabbit, or hamster) that have the desired specificity, affinity, and/or binding capability.
  • a non-human species e.g., mouse, rat, rabbit, or hamster
  • the framework variable region amino acid residues of a human immunoglobulin may be replaced with the corresponding amino acid residues in an antibody from a non-human species.
  • the humanized antibody can be further modified by the substitution of additional amino acid residues either in the framework variable region and/or within the replaced non-human amino acid residues to further refine and optimize antibody specificity, affinity, and/or binding capability.
  • the humanized antibody may comprise variable domains containing all or substantially all of the CDRs that correspond to the non- human immunoglobulin, whereas all or substantially all of the framework variable regions are those of a human immunoglobulin sequence.
  • the variable domains comprise the framework regions of a human immunoglobulin sequence.
  • the variable domains comprise the framework regions of a human immunoglobulin consensus sequence.
  • the humanized antibody can also comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
  • human antibody refers to an antibody produced by a human or an antibody having an amino acid sequence corresponding to an antibody produced by a human made using any of the techniques known in the art.
  • chimeric antibody refers to an antibody wherein the amino acid sequence of the immunoglobulin molecule is derived from two or more species.
  • the variable regions of the light and heavy chains correspond to the variable regions of an antibody derived from one species of mammal (e.g., mouse, rat, rabbit, etc.) with the desired specificity, affinity, and/or binding capability, while the constant regions are homologous to the sequence in an antibody derived from another species.
  • epitopes and “antigenic determinant” are used interchangeably herein and refer to that portion of an antigen or target capable of being recognized and specifically bound by a particular antibody.
  • epitopes can be formed both from contiguous amino acids and noncontiguous amino acids juxtaposed by tertiary folding of the protein.
  • Epitopes formed from contiguous amino acids also referred to as linear epitopes
  • epitopes formed by tertiary folding also referred to as conformational epitopes
  • An epitope typically includes at least 3, and more usually, at least 5, 6, 7, or 8-10 amino acids in a unique spatial conformation.
  • the terms “selectively binds” or “specifically binds” mean that an agent interacts more frequently, more rapidly, with greater duration, with greater affinity, or with some combination of the above to the epitope, protein, or target molecule than with alternative substances, including related and unrelated proteins.
  • “specifically binds” means, for instance, that an agent binds a protein or target with a KD of about O.lmM or less, but more usually less than about I p M.
  • “specifically binds” means that an agent binds a target with a KD of at least about O.lpM or less, at least about O.OlpM or less, or at least about InM or less.
  • specific binding can include an agent that recognizes a protein or target in more than one species (e.g., mouse TIGIT and human TIGIT).
  • specific binding can include an agent that recognizes more than one protein or target.
  • an agent that specifically binds a first target may or may not specifically bind a second target.
  • “specific binding” does not necessarily require (although it can include) exclusive binding, i.e. binding to a single target.
  • an agent may, in certain embodiments, specifically bind more than one target.
  • multiple targets may be bound by the same antigen-binding site on the agent.
  • an antibody may, in certain instances, comprise two identical antigen-binding sites, each of which specifically binds the same epitope on two or more proteins.
  • an antibody may be bispecific and comprise at least two antigen-binding sites with differing specificities.
  • reference to binding means specific binding.
  • selecting and “selected” in reference to a patient is used to mean that a particular patient is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria, e.g., the patient has a tumor with an elevated expression level of PVR and/or PVRL2.
  • selectively treating a patient having a tumor refers to providing treatment to a cancer patient that is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria, e.g., the patient has a tumor with an elevated expression level of PVR and/or PVRL2.
  • “selectively administering” refers to administering a drug to a cancer patient that is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria, e.g., the patient the patient has a tumor with an elevated expression level of PVR and/or PVRL2.
  • selectively treating and selectively administering it is meant that a patient is delivered a personalized therapy for cancer based on the patient's cancer biology, rather than being delivered a standard treatment regimen based solely on the patient having a cancer, such as CRC or NSCLC.
  • polypeptide and “peptide” and “protein” are used interchangeably herein and refer to polymers of amino acids of any length.
  • the polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids.
  • the terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component.
  • polypeptides containing one or more analogs of an amino acid including, for example, unnatural amino acids
  • a “polypeptide” can occur as a single chain or as two or more associated chains.
  • nucleic acid and nucleic acid molecule are used interchangeably herein and refer to polymers of nucleotides of any length, and include DNA and RNA.
  • the nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase.
  • nucleic acids or polypeptides refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity.
  • the percent identity may be measured using sequence comparison software or algorithms or by visual inspection.
  • Various algorithms and software that may be used to obtain alignments of amino acid or nucleotide sequences are well-known in the art. These include, but are not limited to, BLAST, ALIGN, Megalign, BestFit, GCG Wisconsin Package, and variants thereof.
  • two nucleic acids or polypeptides of the invention are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection.
  • identity exists over a region of the sequences that is at least about 10, at least about 20, at least about 40-60 nucleotides or amino acid residues, at least about 60-80 nucleotides or amino acid residues in length or any integral value there between.
  • identity exists over a longer region than 60-80 nucleotides or amino acid residues, such as at least about 80-100 nucleotides or amino acid residues, and in some embodiments the sequences are substantially identical over the full length of the sequences being compared, for example, the coding region of a nucleotide sequence.
  • a “conservative amino acid substitution” is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain.
  • Families of amino acid residues having similar side chains have been generally defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic
  • substitution of a phenylalanine for a tyrosine is considered to be a conservative substitution.
  • conservative substitutions in the sequences of polypeptides and/or antibodies of the invention do not abrogate the binding of the polypeptide or antibody containing the amino acid sequence, to the target binding site.
  • Methods of identifying nucleotide and amino acid conservative substitutions which do not eliminate binding are well- known in the art.
  • vector means a construct, which is capable of delivering, and usually expressing, one or more gene(s) or sequence(s) of interest in a host cell.
  • vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid, or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, and DNA or RNA expression vectors encapsulated in liposomes.
  • a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is “isolated” is a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is in a form not found in nature.
  • Isolated polypeptides, soluble proteins, antibodies, polynucleotides, vectors, cells, or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature.
  • a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is isolated is substantially pure.
  • substantially pure refers to material which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.
  • the term “immune response” as used herein includes responses from both the innate immune system and the adaptive immune system. It includes both cell-mediated and/or humoral immune responses. It includes, but is not limited to, both T-cell and B-cell responses, as well as responses from other cells of the immune system such as natural killer (NK) cells, monocytes, macrophages, etc.
  • NK natural killer
  • cancer and “cancerous” as used herein refer to or describe the physiological condition in mammals in which a population of cells are characterized by unregulated cell growth. Examples of cancer include, but are not limited to, carcinoma, blastoma, sarcoma, and hematologic cancers such as lymphoma and leukemia.
  • tumor and “neoplasm” as used herein refer to any mass of tissue that results from excessive cell growth or proliferation, either benign (non-cancerous) or malignant (cancerous) including pre-cancerous lesions.
  • metalastasis refers to the process by which a cancer spreads or transfers from the site of origin to other regions of the body with the development of a similar cancerous lesion at a new location.
  • a “metastatic” or “metastasizing” cell is one that loses adhesive contacts with neighboring cells and migrates via the bloodstream or lymph from the primary site of disease to secondary sites throughout the body.
  • cancer cell and “tumor cell” refer to the total population of cells derived from a cancer or tumor or pre-cancerous lesion, including both non-tumorigenic cells, which comprise the bulk of the cancer cell population, and tumorigenic stem cells (cancer stem cells).
  • cancer stem cells tumorigenic stem cells
  • the terms “cancer cell” or “tumor cell” will be modified by the term “non-tumorigenic” when referring solely to those cells lacking the capacity to renew and differentiate to distinguish those tumor cells from cancer stem cells.
  • subject refers to any animal (e.g., a mammal), including, but not limited to, humans, non-human primates, canines, felines, rabbits, rodents, and the like, which is to be the recipient of a particular treatment.
  • subject and patient are used interchangeably herein in reference to a human subject.
  • pharmaceutically acceptable refers to a substance approved or approvable by a regulatory agency of the Federal government or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
  • pharmaceutically acceptable excipient, carrier, or adjuvant refers to an excipient, carrier, or adjuvant that can be administered to a patient, together with at least one agent of the present disclosure, and which does not destroy the pharmacological activity thereof and is non-toxic when administered in doses sufficient to deliver a therapeutic effect.
  • pharmaceutically acceptable excipient, carrier, or adjuvant to be an inactive ingredient of any formulation.
  • an effective amount or “therapeutically effective amount” or “therapeutic effect” refer to an amount of an agent, an antibody, a polypeptide, a polynucleotide, a small organic molecule, or other drug effective to “treat” a disease or disorder in a subject such as, a mammal.
  • the therapeutically effective amount of an antibody has a therapeutic effect and as such can enhance or boost the immune response, enhance or boost the anti-tumor response, increase cytolytic activity of immune cells, increase killing of tumor cells, increase killing of tumor cells by immune cells, reduce the number of tumor cells; decrease tumorigenicity, tumorigenic frequency, or tumorigenic capacity; reduce the number or frequency of cancer stem cells; reduce the tumor size; reduce the cancer cell population; inhibit or stop cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibit and stop tumor or cancer cell metastasis; inhibit and stop tumor or cancer cell growth; relieve to some extent one or more of the symptoms associated with the cancer; reduce morbidity and mortality; improve quality of life; or a combination of such effects.
  • treating or “treatment” or “to treat” or “alleviating” or “to alleviate” refers to therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder.
  • a patient is successfully “treated” according to the methods of the present invention if the patient shows one or more of the following: an increased immune response, an increased antitumor response, increased cytolytic activity of immune cells, increased killing of tumor cells, increased killing of tumor cells by immune cells, a reduction in the number of or complete absence of cancer cells; a reduction in the tumor size; inhibition of or an absence of cancer cell infiltration into peripheral organs including the spread of cancer cells into soft tissue and bone; inhibition of or an absence of tumor or cancer cell metastasis; inhibition or an absence of cancer growth; relief of one or more symptoms associated with the specific cancer; reduced morbidity and mortality; improvement in quality of life; reduction in tumorigenicity; reduction in the number or frequency of cancer stem cells; or some combination of effects.
  • the term “and/or” as used in a phrase such as “A and/or B” herein is intended to include both A and B; A or B; A (alone); and B (alone).
  • the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
  • Flow cytometry analysis was conducted to assess the ability of the anti-TIGIT antibodies 313M32 (etigilimab), 313M26 and 313R19 to bind to various forms of TIGIT from different species.
  • the species tested were rat, guinea pig, rabbit, marmoset, pig, dog, rhesus monkey and human.
  • the antibody 313M26 is the parental murine anti-hTIGIT antibody of 313M32 and therefore 313M26 and 313M32 share both comprise the heavy chain and light chain CDRs of SEQ ID Nos: 1-6.
  • 313R19 is an anti-TIGIT antibody that does not share the same heavy and light chain CDRs.
  • This analysis utilized human embryonic kidney 293 cells (HEK-293, American Type Culture Collection, Manassas, VA) that were transiently transfected with cDNA expression vectors encoding a membrane-anchored derivative of TIGIT from the species listed above. These cDNA expression vectors encoded the extracellular domains of TIGIT from different species each fused to the transmembrane domain of human CD4 and an intracellular domain comprised of green fluorescent protein. Cells were then exposed to 313M26, 313M26 or 313R19 and allophycocyanin (APC)- labeled anti-human Fc secondary antibody and then examined by flow cytometry.
  • HEK-293 American Type Culture Collection, Manassas, VA
  • FACS analysis was conducted to assess the ability of anti-TIGIT antibodies 313M32, 313M26 and 313R19 to block PVR binding to human TIGIT.
  • a cell surface human TIGIT protein was generated by ligating amino acids 22- 141 of human TIGIT to the transmembrane domain of CD4 and a C-terminal GFP protein tag using standard recombinant DNA techniques (hTIGIT-CD4TM-GFP).
  • PVR-Fc constructs were generated using standard recombinant DNA techniques. Specifically, the extracellular domain of human PVR was ligated in-frame to a rabbit Fc region and the recombinant hPVR- rbFc protein was expressed in CHO cells. The fusion proteins were purified from cell culture medium using protein A chromatography.
  • HEK-293T cells were transiently transfected with the hTIGIT-CD4TM-GFP construct. After 16 hours, transfected cells were suspended in ice cold HBSS containing 2% FBS and heparin and incubated on ice with 0.5pg/ml hPVR-rbFc fusion protein in the presence of anti-TIGIT antibodies 313R19, 313M26, or 313M32 for 60 minutes. The antibodies were tested at concentrations of 10, 2, and 0.4ug/ml. Cells were incubated without antibody or without hPVR-rbFc as controls.
  • a second incubation with 100, u I PE-conjugated anti-rabbit Fc secondary antibody was performed to detect cells bound by the hPVR-rbFc fusion protein.
  • the cells were analyzed on a FACSCanto instrument (BD Biosciences) and the data was processed using Flow Jo software.
  • a humanized mouse model was used to study the efficacy of treatment with an anti-TIGIT antibody on a human tumor.
  • Tumor-bearing mice were treated with either a control antibody, anti-TIGIT antibody 313R19, or anti-TIGIT antibody 313M32. Mice were dosed every 5 days at Img/kg or 5mg/kg. Tumor growth was monitored and tumor volumes were measured with electronic calipers at the indicated time points.
  • Figure 2 demonstrates that etigilimab was able to significantly reduce tumor growth at a concentration of Img/kg compared to the control, whereas the anti-TIGIT antibody 313R19 was not able to significantly reduce tumor growth at the same concentration.
  • humanized mouse models bearing patient-derived xenografts can be used to study the anti-hTIGIT antibody OMP-313M32 (which only binds human TIGIT) in parallel with pre -clinical studies carried out with the anti-TIGIT antibodies 313R12 and 313R19 and murine tumor models.
  • Example 3 A phase lb/2 clinical trial evaluating the safety and efficacy of anti-TIGIT antibodies in combination with anti-PD-1 antibodies in the treatment of cancer
  • a phase lb/2 open-label basket study of etigilimab in combination with an anti- PD-1 antibody (nivolumab) was conducted in patients with locally advanced or metastatic solid tumors. Patient populations selected for evaluation in this study all had advanced or refractory disease. These patients therefore had a high unmet therapeutic need for which limited therapeutic options exist.
  • the types of cancer included in the clinical trial were endometrial cancer, head and neck cancer, cervical cancer, ovarian cancer, rare tumours, including soft tissue sarcomas, testicular germ cell tumours and uveal melanomas, high tumor mutational burden (TMB-H) and microsatellite stable (MSS) cancers. Twenty two patients were included in the trial which were broken down into the following cohorts:
  • the path mts were naive to checkpoint inhibitor therapy.
  • the patients were intravenously administered etigilimab every two weeks at a dose of 1000 mg and intravenously administered nivolumab every two weeks at a dose of 240 mg.
  • the objective response rate (which is a combination of complete response rates and partial response rates) was 13.0%.
  • the disease control rate (which is a combination of complete response rates, partial response rates and stable diseases) was 34.7%.
  • Figures 17a and 17b show the reduction in tumour size in a cervical cancer patient that occurred after treatment. These figures show that a partial response occurred as the tumour was no longer visible after the second scan.
  • the lesions that the patient had at the beginning of the trial and their response to treatment with the combination of etigilimab and an anti-PD- 1 antibody are summarised in the Table below.
  • FIG. 12 demonstrates that the combination treatment decreased the number of Treg cells in circulation but had no effect on the number of CD8+ T cells.
  • Figures 13A and 13B show that the combination treatment increased markers of cell proliferation in T cells.
  • Figure 14 demonstrates that the combination treatment increased IFNy production in CD4+ effector memory cells.
  • Figure 15 shows that the combination treatment increased the level of proliferating (Ki-67+) CD8+ PD-1+ cells.
  • Figure 16 shows that the combination treatment increased markers of cell proliferation in natural killer cells.
  • Figure 17 shows that the combination treatment reduced the number of exhausted T-cells.
  • Example 5 Updated clinical data from the phase lb/2 clinical trial evaluating the safety and efficacy of anti-TIGIT antibodies in combination with anti-PD-1 antibodies in the treatment of cancer
  • the patients were naive to checkpoint inhibitor therapy.
  • the patients were intravenously administered etigilimab every two weeks at a dose of 1000 mg and intravenously administered nivolumab every two weeks at a dose of 240 mg.
  • the preliminary efficacy data are shown in the Figures 19-21.
  • the objective response rate (which is a combination of complete response rates and partial response rates) was 15.8%.
  • the disease control rate (which is a combination of complete response rates, partial response rates and stable diseases) was 47.4%.
  • the median duration of stable disease was 3.7 months (0.0- 7.7 months).
  • the median progression-free survival was 3.0 months (1.7-4.6 months).
  • These data further support the ability of the combination of etigilimab and nivolumab to treat various cancers, including rare cancers, such as such as soft tissue sarcomas, testicular germ cell tumors and uveal melanomas.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention provides methods for treating cancer that involve a combination of anti-TIGIT and anti-PD-1 antibodies.

Description

METHODS FOR TREATING CANCER
TECHNICAL FIELD
[0001] The present invention relates to methods for treating cancer and in particular treating soft tissue sarcomas, testicular germ cell tumors and uveal melanomas.
BACKGROUND TO THE INVENTION
[0002] The basis for immunotherapy is the manipulation and/or modulation of the immune system, including both innate immune responses and adaptive immune responses. The general aim of immunotherapy is to treat diseases by controlling the immune response to a “foreign agent”, for example a pathogen or a tumor cell.
[0003] The immune system is a highly complex system made up of a great number of cell types, including but not limited to, T-cells, B-cells, natural killer cells, antigen-presenting cells, dendritic cells, monocytes, and macrophages. These cells possess complex and subtle systems for controlling their interactions and responses. The cells utilize both activating and inhibitory mechanisms and feedback loops to keep responses in check and not allow negative consequences of an uncontrolled immune response (e.g., autoimmune diseases).
[0004] The concept of cancer immunosurveillance is based on the theory that the immune system can recognize tumor cells, mount an immune response, and suppress the development and/or progression of a tumor. However, it is clear that many cancerous cells have developed mechanisms to evade the immune system which can allow for uninhibited growth of tumors. Cancer/tumor immunotherapy focuses on the development of new and novel agents that can activate and/or boost the immune system to achieve a more effective attack against tumor cells resulting in increased killing of tumor cells and/or inhibition of tumor growth.
SUMMARY OF THE INVENTION
[0005] The inventors have surprisingly shown that a combination of an anti-TIGIT antibody and an anti-PD-1 antibody can be used to treat rare cancers including soft tissue sarcomas, testicular germ cell tumours and uveal melanomas. Therefore, in some aspects the invention provides a method of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti- PD-1 antibody, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID N0:5), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumour and uveal melanoma.
[0006] In some embodiments, the anti-TIGIT antibody comprises a heavy chain variable region having at least 90% sequence identity to SEQ ID NO:7 and a light chain variable region having at least 90% sequence identity to SEQ ID NO: 8. In certain embodiments, the anti-TIGIT antibody comprises a heavy chain variable region comprising SEQ ID NO:7 and a light chain variable region comprising SEQ ID NO:8. In some embodiments, the anti-TIGIT antibody comprises a heavy chain having at least 90% sequence identity to SEQ ID NO:9 and a light chain having at least 90% sequence identity to SEQ ID NO: 10. In certain embodiments, the anti-TIGIT antibody comprises a heavy chain comprising SEQ ID NO:9 and a light chain comprising SEQ ID NO: 10. In a preferred embodiment, the anti-TIGIT antibody is etigilimab. [0007] The anti-TIGIT antibody can be a monoclonal antibody, a humanized antibody, a human antibody, a chimeric antibody, a bispecific antibody, an IgGl antibody, an IgG2 antibody, an IgG4 antibody, or an antibody fragment comprising an antigen binding site.
[0008] In some embodiments, the anti-PD-1 antibody is selected from the group consisting of pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo), dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab.
[0009] In some embodiments, the anti-TIGIT antibody is administered prior to, concurrently with and/or subsequently to the administration of the anti-PD-1 antibody. In some embodiments, the anti-TIGIT antibody and the anti-PD-1 antibody are administered intravenously. In some embodiments, the anti-TIGIT antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks. In certain embodiments, the anti-TIGIT antibody is administered once every two weeks. In some embodiments, the anti-PD-1 antibody is administered once every two weeks, once every three weeks, once every four weeks, once every five weeks or once every six weeks. In certain embodiments, the anti-PD-1 antibody is administered once every two weeks, once every three weeks, once every four weeks or once every six weeks.
[0010] The anti-TIGIT antibody can be administered at a flat dose of 1000 mg. In some embodiments, the anti-TIGIT antibody is administered at a flat dose of 1000 mg once every two weeks. The anti-PD-1 antibody can be administered at a flat dose of 200 mg- 1000 mg, for example the anti-PD-1 antibody is administered at a flat dose of 200 mg, 240 mg, 350 mg, 400mg, 500 mg or 1000 mg.
[0011] In some embodiments, the human patient has not received prior treatment with an anti-cancer agent. In alternative embodiments, the human patient has been previously treated with an anti-cancer agent and there is tumour growth, progression, or recurrence during or after treatment with the anti-cancer agent. In some embodiments, the anti-cancer agent is a checkpoint inhibitor, a chemotherapeutic agent or a cell therapy. In certain embodiments, the checkpoint inhibitor is an anti-PD-1 antibody, an anti-PD-Ll antibody or a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1.
[0012] In some embodiments, the human patient has been previously been treated with a chemotherapeutic agent, and there is tumour growth, progression, or recurrence during or after treatment with the chemotherapeutic agent.
[0013] In certain embodiments, the soft tissue sarcoma is selected from the group consisting of leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, dedifferentiated liposarcoma, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid, malignant peripheral nerve sheath tumours (MPNST) and undifferentiated (pleomorphic sarcoma). In certain embodiments, the soft tissue sarcoma is a dedifferentiated liposarcoma or an undifferentiated (pleomorphic sarcoma).
[0014] In certain embodiments, the testicular germ cell tumor is a seminomas testicular germ cell tumour or a non-seminomas testicular germ cell tumor.
[0015] In certain embodiments, the uveal melanoma is choroidal melanoma, ciliary body melanoma and iris melanoma.
[0016] In some embodiments, the human patient is over 50 years old.
BRIEF DESCRIPTION OF DRAWINGS
[0017] Figure 1: FACS analysis was conducted with anti-TIGIT antibodies 313M32, 313M26 and 313R19 to assess the ability of these antibodies to block PVR binding to human TIGIT.
[0018] Figure 2: Inhibition of tumor growth by anti-TIGIT antibodies 313R19 and 313M32 in a humanized mouse model.
[0019] Figure 3 : Historical overall response rate with checkpoint inhibitors for ovarian cancer, cervical cancer, and uveal melanoma.
[0020] Figure 4 : Scans of a cervical cancer tumor in a patient who was treated with an anti-TIGIT antibody and an anti-PD-1 antibody. The circles indicate the location of the tumor on the scan. The change in tumour volume resulted in a complete overall response after the patient was treated with the combination of an anti-TIGIT antibody and an anti-PD-1 antibody. [0021] Figure 5: Heavy and light chain sequence of Camrelizumab (PD-1 antibody)
[0022] Figure 6: Heavy and light chain sequence of Prolgolimab (PD-1 antibody)
[0023] Figure 7: Heavy and light chain sequence of Pucotenlimab (PD-1 antibody)
[0024] Figure 8: Heavy and light chain sequence of Ezabenlimab (PD-1 antibody)
[0025] Figure 9: Heavy and light chain sequence of Envafolimab (PD-L1 antibody)
[0026] Figure 10: Heavy and light chain sequence of Socazolimab (PD-L1 antibody) [0027] Figure 11: A summary showing the length of time that patients have been part of the Phase lb/2 clinical trial.
[0028] Figures 12-17 disclose pharmacodynamic analysis of biomarkers after treatment with the combination of etigilimab and nivolumab. Figure 12 shows the change in Treg cells and CD8+ cells after treatment; Figures 13A and 13B show the change in Ki-67+ TIGIT+ CD4 cells after treatment; Figure 14 shows the change in the level of IFNy produced by CD4+ effector memory cells after treatment; Figure 15 shows the change in the level of Ki-67+ CD8+ PD-1+ cells after treatment and Figure 16 shows the change in Ki-67+ natural killer cells (NK cells) after treatment. Figure 17 shows the change in T Progenitor Exhausted like (TPEX) cells after treatment.
[0029] Figure 18: Scans of a cervical cancer tumor in a patient who was treated with an anti-TIGIT antibody and an anti-PD-1 antibody. The circles indicate the location of the tumor on the scan. The change in tumour volume resulted in a partial overall response after the patient was treated with the combination of an anti-TIGIT antibody and an anti-PD-1 antibody.
[0030] Figure 19: A summary showing the length of time that patients have been part of the Phase lb/2 clinical trial. Time on treatment and investigator-assessed RECIST 1.1 response. [0031] Figure 20: A summary of the preliminary efficacy of the combination of etigilimab with an anti-PD-1 antibody (nivolumab). PD = disease progression; SD = stable disease; cCR = confirmed complete response; uCR = unconfirmed complete response; cPR = confirmed partial response; uPR = unconfirmed partial response; TMB-H = high tumor mutational burden; MSS = microsatellite stable (MSS); and HGSOC = high-grade serous ovarian cancer.
[0032] Figure 21: A waterfall plot showing the percentage change compared to the baseline for each subject. DISCLOSURE OF THE INVENTION
Anti-TIGIT antibodies
[0033] T-cell immunoreceptor with Ig and ITIM domains (TIGIT) is a type I transmembrane glycoprotein that contains an immunoglobulin variable (IgV) domain. TIGIT belongs to the poliovirus receptor (PVR) family and binds to the poliovirus receptor (PVR; CD155) with high affinity and to PVRL-2 (CD112) and PVRL-3 (CD113) with a lower affinity. TIGIT is expressed on T-cells, including regulatory T-cells (Tregs) and memory T-cells, as well as on NK cells and is upregulated following activation of naive CD4+ T-cells.
[0034] In one aspect, the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6).
[0035] In some embodiments, the anti-TIGIT antibody is a monoclonal antibody, a humanized antibody, a human antibody, a recombinant antibody, a chimeric antibody, a bispecific antibody or a multi-specific antibody. In some embodiments, the antibody is monovalent. In some embodiments, the antibody is bivalent. In some embodiments, the antibody is monospecific. In some embodiments, the antibody is bispecific. In some embodiments, the anti-TIGIT antibody is an IgA, IgD, IgE, IgG, or IgM antibody. In certain embodiments, the anti-TIGIT antibody is an IgGl antibody. In certain embodiments, the anti- TIGIT antibody is an IgG2 antibody. In some embodiments, the anti-TIGIT antibody is an IgG4 antibody. In some embodiments, the anti-TIGIT antibody is conjugated to a cytotoxic moiety. In some embodiments, the anti-TIGIT antibody is isolated. In some embodiments, the anti- TIGIT antibody is substantially pure.
[0036] In some embodiments, the anti-TIGIT antibody comprises a heavy chain variable region having at least 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO:7 and/or a light chain variable region having at least 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO:8. In some embodiments, the anti-TIGIT antibody comprises a heavy chain variable region comprising SEQ ID NO:7 and a light chain variable region comprising SEQ ID NO: 8. In some embodiments, the anti-TIGIT antibody comprises a heavy chain variable region consisting of SEQ ID NO:7 and a light chain variable region consisting of SEQ ID NOG. [0037] In some embodiments, the anti-TIGIT antibody comprises a heavy chain having at least 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO:9 and/or a light chain having at least 90%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID NO: 10. In some embodiments, the anti-TIGIT antibody comprises a heavy chain comprising SEQ ID NO:9 and a light chain comprising SEQ ID NO: 10. In some embodiments, the anti-TIGIT antibody comprises a heavy chain consisting of SEQ ID NO:9 and a light chain consisting of SEQ ID NOTO. In certain embodiments, the anti-TIGIT antibody is etigilimab. The heavy chain of etigilimab consists of SEQ ID NO:9 and the light chain of etigilimab consists of SEQ ID NOTO.
[0038] In some embodiments, the anti-TIGIT antibody comprises the variable regions of etigilimab wherein the heavy chain variable region (SEQ ID NO: 7) and/or the light chain variable region (SEQ ID NO: 8) of etigilimab have been affinity-matured. In certain embodiments, the anti-TIGIT antibody comprises the heavy chain and light chain of etigilimab (with or without the leader sequence). In certain embodiments, the anti-TIGIT antibody is etigilimab. In certain embodiments, the anti-TIGIT antibody comprises the heavy chain variable region of etigilimab as part of an IgGl, IgG2, or IgG4 heavy chain. In certain embodiments, the anti-TIGIT antibody comprises the heavy chain variable region of the etigilimab as part of a human IgGl heavy chain. In certain embodiments, the anti-TIGIT antibody comprises the heavy chain variable region of the etigilimab as part of a human IgG2 heavy chain. In certain embodiments, the anti-TIGIT antibody comprises the heavy chain variable region of the etigilimab as part of a human IgG4 heavy chain.
[0039] In some embodiments, the anti-TIGIT antibody is a monoclonal antibody. Monoclonal antibodies can be prepared using hybridoma methods known to one of skill in the art. In some embodiments, using the hybridoma method, a mouse, rat, rabbit, hamster, or other appropriate host animal, is immunized as described above to elicit the production of antibodies that specifically bind the immunizing antigen. In some embodiments, lymphocytes can be immunized in vitro. In some embodiments, the immunizing antigen can be a human protein or a fragment thereof. In some embodiments, the immunizing antigen can be a mouse protein or a fragment thereof.
[0040] Following immunization, lymphocytes are isolated and fused with a suitable myeloma cell line using, for example, polyethylene glycol. The hybridoma cells are selected using specialized media as known in the art and unfused lymphocytes and myeloma cells do not survive the selection process. Hybridomas that produce monoclonal antibodies directed specifically against a chosen antigen may be identified by a variety of methods including, but not limited to, immunoprecipitation, immunoblotting, and in vitro binding assays (e.g., flow cytometry, FACS, ELISA, and radioimmunoassay). The hybridomas can be propagated either in in vitro culture using standard methods or in vivo as ascites tumors in an animal. The monoclonal antibodies can be purified from the culture medium or ascites fluid according to standard methods in the art including, but not limited to, affinity chromatography, ion-exchange chromatography, gel electrophoresis, and dialysis.
[0041] In certain embodiments, monoclonal antibodies can be made using recombinant DNA techniques as known to one skilled in the art. The polynucleotides encoding a monoclonal antibody are isolated from mature B-cells or hybridoma cells, such as by RT-PCR using oligonucleotide primers that specifically amplify the genes encoding the heavy and light chains of the antibody, and their sequence is determined using standard techniques. The isolated polynucleotides encoding the heavy and light chains are then cloned into suitable expression vectors which produce the monoclonal antibodies when transfected into host cells such as E. coli, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin proteins. In certain other embodiments, recombinant monoclonal antibodies, or fragments thereof, can be isolated from phage display libraries expressing variable domains or CDRs of a desired species.
[0042] Polynucleotide(s) encoding a monoclonal antibody can be modified, for example, by using recombinant DNA technology to generate alternative antibodies. In some embodiments, the constant domains of the light chain and heavy chain of, for example, a mouse monoclonal antibody can be substituted for constant regions of, for example, a human antibody to generate a chimeric antibody, or for a non-immunoglobulin polypeptide to generate a fusion antibody. In some embodiments, the constant regions are truncated or removed to generate a desired antibody fragment of a monoclonal antibody. Site-directed or high-density mutagenesis of the variable region(s) can be used to optimize specificity, affinity, etc. of a monoclonal antibody.
[0043] In some embodiments, the anti-TIGIT antibody is a humanized antibody. Typically, humanized antibodies are human immunoglobulins in which the amino acid residues of the CDRs are replaced by amino acid residues from CDRs of a non-human species (e.g., mouse, rat, rabbit, hamster, etc.) that have the desired specificity, affinity, and/or binding capability using methods known to one skilled in the art. In some embodiments, some of the framework variable region amino acid residues of a human immunoglobulin are replaced with corresponding amino acid residues in an antibody from a non-human species. In some embodiments, a humanized antibody can be further modified by the substitution of additional residues either in the framework variable region and/or within the replaced non-human residues to further refine and optimize antibody specificity, affinity, and/or capability. In general, a humanized antibody will comprise variable domain regions containing all, or substantially all, of the CDRs that correspond to the non-human immunoglobulin whereas all, or substantially all, of the framework regions are those of a human immunoglobulin sequence. In some embodiments, the framework regions are those of a human consensus immunoglobulin sequence. In some embodiments, a humanized antibody can also comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. In certain embodiments, such humanized antibodies are used therapeutically because they may reduce antigenicity and HAMA (human anti-mouse antibody) responses when administered to a human patient.
[0044] In certain embodiments, the anti-TIGIT antibody is a human antibody. Human antibodies can be directly prepared using various techniques known in the art. In some embodiments, human antibodies may be generated from immortalized human B lymphocytes immunized in vitro or from lymphocytes isolated from an immunized individual. In either case, cells that produce an antibody directed against a target antigen can be generated and isolated. In some embodiments, the human antibody can be selected from a phage library, where that phage library expresses human antibodies. Alternatively, phage display technology can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable domain gene repertoires from unimmunized donors. Techniques for the generation and use of antibody phage libraries are well known in the art. Once antibodies are identified, affinity maturation strategies known in the art, including but not limited to, chain shuffling and site- directed mutagenesis, may be employed to generate higher affinity human antibodies. In some embodiments, human antibodies can be made in transgenic mice that contain human immunoglobulin loci. Upon immunization these mice are capable of producing the full repertoire of human antibodies in the absence of endogenous immunoglobulin production.
[0045] In some embodiments, the anti-TIGIT antibody binds with TIGIT and interferes with the interaction of TIGIT with a second protein, for example PVR, PVRL2 or PVLR3. In some embodiments, the anti-TIGIT antibody binds TIGIT and disrupts binding of TIGIT to PVR, and/or disrupts PVR activation of TIGIT signalling.
[0046] The full-length amino acid sequence for human TIGIT (UniProtKB No. Q495A1) is known in the art and are provided herein as SEQ ID NO: 11. As used herein, reference to amino acid positions refer to the numbering of full-length amino acid sequences including the signal sequence.
[0047] In certain embodiments, the anti-TIGIT antibody is an antibody that specifically binds the extracellular domain of human TIGIT, or a fragment thereof. In some embodiments, the anti-TIGIT antibody is an antibody that specifically binds the Ig-like domain of human TIGIT. In some embodiments, the anti-TIGIT antibody is an antibody that specifically binds the IgV domain of human TIGIT. In some embodiments, the anti-TIGIT antibody is an antibody that binds within amino acids 22-141 of human TIGIT. In some embodiments, the anti-TIGIT antibody is an antibody that binds within amino acids 22-141 of SEQ ID NO: 12. In some embodiments, the antibody binds within amino acids 22-124 of human TIGIT. In some embodiments, the antibody binds within amino acids 22-124 of SEQ ID NO: 11. In certain embodiments, the anti-TIGIT antibody binds within SEQ ID NO: 12, or a fragment thereof. In some embodiments, the anti-TIGIT antibody is an antibody that binds within amino acids SO- 124 of human TIGIT. In some embodiments, the anti-TIGIT antibody is an antibody that binds within amino acids 50-124 of SEQ ID NOT E In certain embodiments, the anti-TIGIT antibody binds within SEQ ID NO: 12, or a fragment thereof.
[0048] In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids within SEQ ID NO: 13. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids within SEQ ID NO: 14. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids within SEQ ID NO: 13 and SEQ ID NO: 14. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62 and 1109 of SEQ ID NO: 11. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62 and T119 of SEQ ID NO: 11. In some embodiments, the anti- TIGIT antibody is an antibody that binds an epitope comprising amino acids Q64 and 1109 of SEQ ID NO: 11. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q64 and T119 of SEQ ID NO: 11. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, Q64, and 1109 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, Q64, and T119 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, 1109, and T119 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q64, 1109, and T119 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising amino acids Q62, Q64, 1109, and T119 of SEQ ID NOTE In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope comprising at least one amino acid selected from the group consisting of: N58, E60, Q62, Q64, L65, F107, 1109, Hi ll, T117, T119, G120, and R121 of SEQ ID NOT E In some embodiments, the epitope is a conformational epitope. In some embodiments, the anti-TIGIT antibody is an antibody that binds an epitope which does not comprise amino acid V100 of SEQ ID NO:11.
[0049] In certain embodiments, the anti-TIGIT antibody binds TIGIT with a dissociation constant (KD) of about IpM or less, about lOOnM or less, about 40nM or less, about 20nM or less, about lOnM or less, about InM or less, about O.lnM or less, 50pM or less, lOpM or less, or IpM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about 20nM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about lOnM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about InM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about 0.5nM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about O.lnM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about 50pM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about 25pM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about lOpM or less. In some embodiments, an anti-TIGIT antibody binds TIGIT with a KD of about IpM or less. In some embodiments, the dissociation constant of the antibody to TIGIT is the dissociation constant determined using a TIGIT fusion protein comprising at least a portion of the extracellular domain of TIGIT protein immobilized on a Biacore chip. In some embodiments, the dissociation constant of the antibody to TIGIT is the dissociation constant determined using the antibody captured by an anti-human IgG antibody on a Biacore chip and a soluble TIGIT protein. In certain embodiments, the anti-TIGIT antibody binds TIGIT with a KD of about 0.4nM or less determined using a TIGIT fusion protein comprising at least a portion of the extracellular domain of TIGIT protein immobilized on a Biacore chip. In certain embodiments, the anti-TIGIT antibody binds TIGIT with a KD of about 0.4nM or less determined using the antibody captured by an anti-human IgG antibody on a Biacore chip and a soluble TIGIT protein.
[0050] In certain embodiments, the anti-TIGIT antibody binds TIGIT with a half maximal effective concentration (EC50) of about I pM or less, about lOOnM or less, about 40nM or less, about 20nM or less, about lOnM or less, about InM or less, or about O.lnM or less. In certain embodiments, the anti-TIGIT antibody binds to human TIGIT with a half maximal effective concentration (EC 50) of about I M or less, about lOOnM or less, about 40nM or less, about 20nM or less, about lOnM or less, about InM or less, or about O.lnM or less.
[0051] Non-limiting examples of anti-TIGIT antibodies are described in [1], [2], [3] and [4], which are hereby incorporated in their entirety herein by reference thereto. [0052] Heteroconjugate antibodies are also within the scope of the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune cells to unwanted cells. It is also contemplated that the heteroconjugate antibodies can be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this purpose include iminothiolate and methyl-4- mercaptobutyrimidate.
[0053] For the purposes of the present invention, it should be appreciated that modified antibodies can comprise any type of variable region that provides for the association of the antibody with the target (i.e., TIGIT). In this regard, the variable region may comprise or be derived from any type of mammal that can be induced to mount a humoral response and generate immunoglobulins against the desired antigen. As such, the variable region of the modified antibodies can be, for example, of human, murine, rat, rabbit, non-human primate (e.g. cynomolgus monkeys, macaques, etc.), or rabbit origin. In some embodiments, both the variable and constant regions of the modified immunoglobulins are human. In other embodiments, the variable regions of compatible antibodies (usually derived from a non-human source) can be engineered or specifically tailored to improve the binding properties or reduce the immunogenicity of the molecule. In this respect, variable regions useful in the present invention can be humanized or otherwise altered through the inclusion of imported amino acid sequences.
[0054] In certain embodiments, the variable domains in both the heavy and light chains are altered by at least partial replacement of one or more CDRs and, if necessary, by partial framework region replacement and sequence modification and/or alteration. Although the CDRs may be derived from an antibody of the same class or even subclass as the antibody from which the framework regions are derived, it is envisaged that the CDRs may be derived from an antibody of different class and often from an antibody from a different species. It may not be necessary to replace all of the CDRs with all of the CDRs from the donor variable region to transfer the antigen binding capacity of one variable domain to another. Rather, it may only be necessary to transfer those residues that are required to maintain the activity of the antigenbinding site.
[0055] Alterations to the variable region notwithstanding, those skilled in the art will appreciate that the modified antibodies of this invention will comprise antibodies (e.g., full- length antibodies or immunoreactive fragments thereof) in which at least a fraction of one or more of the constant region domains has been deleted or otherwise altered so as to provide desired biochemical characteristics such as increased tumor localization or increased serum half-life when compared with an antibody of approximately the same immunogenicity comprising a native or unaltered constant region. In some embodiments, the constant region of the modified antibodies will comprise a human constant region. Modifications to the constant region compatible with this invention comprise additions, deletions or substitutions of one or more amino acids in one or more domains. The modified antibodies disclosed herein may comprise alterations or modifications to one or more of the three heavy chain constant domains (CHI, CH2 or CH3) and/or to the light chain constant domain (CL). In some embodiments, one or more domains are partially or entirely deleted from the constant regions of the modified antibodies. In some embodiments, the modified antibodies will comprise domain deleted constructs or variants wherein the entire CH2 domain has been removed (ACH2 constructs). In some embodiments, the omitted constant region domain is replaced by a short amino acid spacer (e.g., 10 amino acid residues) that provides some of the molecular flexibility typically imparted by the absent constant region.
[0056] In some embodiments, the modified antibodies are engineered to fuse the CH3 domain directly to the hinge region of the antibody. In other embodiments, a peptide spacer is inserted between the hinge region and the modified CH2 and/or CH3 domains. For example, constructs may be expressed wherein the CH2 domain has been deleted and the remaining CH3 domain (modified or unmodified) is joined to the hinge region with a 5-20 amino acid spacer. Such a spacer may be added to ensure that the regulatory elements of the constant domain remain free and accessible or that the hinge region remains flexible. However, it should be noted that amino acid spacers may, in some cases, prove to be immunogenic and elicit an unwanted immune response against the construct. Accordingly, in certain embodiments, any spacer added to the construct will be relatively non-immunogenic so as to maintain the desired biological qualities of the modified antibodies.
[0057] In some embodiments, the modified antibodies may have only a partial deletion of a constant domain or substitution of a few or even a single amino acid. For example, the mutation of a single amino acid in selected areas of the CH2 domain may be enough to substantially reduce Fc binding. In some embodiments, the mutation of a single amino acid in selected areas of the CH2 domain may be enough to substantially reduce Fc binding and increase cancer cell localization and/or tumor penetration. Similarly, it may be desirable to simply delete the part of one or more constant region domains that control a specific effector function (e.g. complement Clq binding) to be modulated. Such partial deletions of the constant regions may improve selected characteristics of the antibody (serum half-life) while leaving other desirable functions associated with the subject constant region domain intact. Moreover, as alluded to above, the constant regions of the disclosed antibodies may be modified through the mutation or substitution of one or more amino acids that enhances the profile of the resulting construct. In this respect it may be possible to disrupt the activity provided by a conserved binding site (e.g., Fc binding) while substantially maintaining the configuration and immunogenic profile of the modified antibody. In certain embodiments, the modified antibodies comprise the addition of one or more amino acids to the constant region to enhance desirable characteristics such as decreasing or increasing effector function or provide for more cytotoxin or carbohydrate attachment sites.
[0058] It is known in the art that the constant region mediates several effector functions. For example, binding of the Cl component of complement to the Fc region of IgG or IgM antibodies (bound to antigen) activates the complement system. Activation of complement is important in the opsonization and lysis of cell pathogens. The activation of complement also stimulates the inflammatory response and can also be involved in autoimmune hypersensitivity. In addition, the Fc region of an antibody can bind a cell expressing a Fc receptor (FcR). There are a number of Fc receptors which are specific for different classes of antibody, including IgG (gamma receptors), IgE (epsilon receptors), IgA (alpha receptors) and IgM (mu receptors). Binding of antibody to Fc receptors on cell surfaces triggers a number of important and diverse biological responses including engulfment and destruction of antibody-coated particles, clearance of immune complexes, lysis of antibody-coated target cells by killer cells (called antibody-dependent cell cytotoxicity or ADCC), release of inflammatory mediators, placental transfer, and control of immunoglobulin production.
[0059] In certain embodiments, the modified antibodies provide for altered effector functions that, in turn, affect the biological profile of the administered antibody. For example, in some embodiments, the deletion or inactivation (through point mutations or other means) of a constant region domain may reduce Fc receptor binding of the circulating modified antibody. In some embodiments, the deletion or inactivation (through point mutations or other means) of a constant region domain may reduce Fc receptor binding of the circulating modified antibody thereby increasing cancer cell localization and/or tumor penetration. In other embodiments, the constant region modifications increase the serum half-life of the antibody. In other embodiments, the constant region modifications reduce the serum half-life of the antibody. In some embodiments, the constant region is modified to eliminate disulfide linkages or oligosaccharide moieties. Modifications to the constant region in accordance with this invention may easily be made using well known biochemical or molecular engineering techniques. [0060] In certain embodiments, the anti-TIGIT antibody does not have one or more effector functions. For instance, in some embodiments, the antibody has no ADCC activity, and/or no complement-dependent cytotoxicity (CDC) activity. In certain embodiments, the antibody does not bind an Fc receptor and/or complement factors. In certain embodiments, the antibody has no effector function(s).
[0061] In certain embodiments, the anti-TIGIT antibody has one or more effector functions. For instance, in some embodiments, the antibody has ADCC activity, and/or complement-dependent cytotoxicity (CDC) activity. In certain embodiments, the antibody binds an Fc receptor and/or complement factors. In certain embodiments, the antibody has effector function(s).
[0062] In certain embodiments, the anti-TIGIT antibody binds the Fc receptor. In certain embodiments, the anti-TIGIT antibody activates ADCC. In some embodiments, the anti-TIGIT antibody recruits activated myeloid cells to the tumor. In some embodiments, the anti-TIGIT antibody activates T cells. In some embodiments, the anti-TIGIT antibody activates NK cells. In some embodiments, the anti-TIGIT antibody reduces the number of T-regulatory cells. In some embodiments, the anti-TIGIT antibody increases the CD8/Treg ratio. In some embodiments, the anti-TIGIT antibody increases the CD8/Treg ratio.
[0063] The present invention further embraces variants and equivalents which are substantially homologous to the recombinant, monoclonal, chimeric, humanized, and human antibodies, or antibody fragments thereof, described herein. These variants can contain, for example, conservative substitution mutations, i.e. the substitution of one or more amino acids by similar amino acids.
[0064] In certain embodiments, the antibodies described herein are isolated. In certain embodiments, the antibodies described herein are substantially pure.
[0065] The anti-TIGIT antibodies of the present invention can be assayed for specific binding by any method known in the art. The immunoassays which can be used include, but are not limited to, competitive and non-competitive assay systems using techniques such as Biacore analysis, FACS analysis, immunofluorescence, immunocytochemistry, Western blot analysis, radioimmunoassay, ELISA, “sandwich” immunoassay, immunoprecipitation assay, precipitation reaction, gel diffusion precipitin reaction, immunodiffusion assay, agglutination assay, complement-fixation assay, immunoradiometric assay, fluorescent immunoassay, and protein A immunoassay. Such assays are routine and well-known in the art (see, e.g., Ausubel et al., Editors, 1994-present, Current Protocols in Molecular Biology, John Wiley & Sons, Inc., New York, NY). [0066] In a non-limiting example, screening for specific binding of an antibody to human TIGIT may be determined using ELISA. An ELISA comprises preparing antigen (e.g., TIGIT or a fragment thereof), coating wells of a 96-well microtiter plate with antigen, adding the test antibodies conjugated to a detectable compound such as an enzymatic substrate (e.g. horseradish peroxidase or alkaline phosphatase) to the well, incubating for a period of time, and detecting the presence of an antibody bound to the antigen. In some embodiments, the test antibodies are not conjugated to a detectable compound, but instead a secondary antibody that recognizes the antibody (e.g., an anti-Fc antibody) and is conjugated to a detectable compound is added to the wells. In some embodiments, instead of coating the well with the antigen, the test antibodies can be coated to the wells, the antigen (e.g., TIGIT) is added to the wells, followed by a secondary antibody conjugated to a detectable compound. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art.
[0067] In another non-limiting example, the specific binding of an antibody to TIGIT may be determined using FACS. A FACS screening assay may comprise generating a cDNA construct that expresses an antigen as a full-length protein (TIGIT) or a fusion protein (e.g., TIGIT-CD4TM), transfecting the construct into cells, expressing the antigen on the surface of the cells, mixing the test antibodies with the transfected cells, and incubating for a period of time. The cells bound by the test antibodies may be identified using a secondary antibody conjugated to a detectable compound (e.g., PE-conjugated anti-Fc antibody) and a flow cytometer. One of skill in the art would be knowledgeable as to the parameters that can be modified to optimize the signal detected as well as other variations of FACS that may enhance screening (e.g., screening for blocking antibodies).
[0068] The binding affinity of an antibody and the off-rate of an antibody-antigen interaction can be determined by competitive binding assays. One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3H or 125I-TIGIT), or fragment or variant thereof, with the antibody of interest in the presence of increasing amounts of unlabeled antigen followed by the detection of the antibody bound to the labeled antigen. The affinity of the antibody for the antigen and the binding off-rates can be determined from the data by Scatchard plot analysis. In some embodiments, Biacore kinetic analysis is used to determine the binding on and off rates of antibodies. In some embodiments, Biacore kinetic analysis comprises analyzing the binding and dissociation of antibodies from chips with immobilized antigen (e.g., TIGIT) on their surface. In some embodiments, Biacore kinetic analysis comprises analyzing the binding and dissociation of antigen (e.g., TIGIT) from chips with immobilized antibody (e.g., anti-TIGIT antibody) on their surface. [0069] Many proteins, including antibodies, contain a signal sequence that directs the transport of the proteins to various locations. Generally, signal sequences (also referred to as signal peptides or leader sequences) are located at the N-terminus of nascent polypeptides. They target the polypeptide to the endoplasmic reticulum and the proteins are sorted to their destinations, for example, to the inner space of an organelle, to an interior membrane, to the cell's outer membrane, or to the cell exterior via secretion. Most signal sequences are cleaved from the protein by a signal peptidase after the proteins are transported to the endoplasmic reticulum. The cleavage of the signal sequence from the polypeptide usually occurs at a specific site in the amino acid sequence and is dependent upon amino acid residues within the signal sequence. Although there is usually one specific cleavage site, more than one cleavage site may be recognized and/or may be used by a signal peptidase resulting in a non-homogenous N- terminus of the polypeptide. For example, the use of different cleavage sites within a signal sequence can result in a polypeptide expressed with different N-terminal amino acids. Accordingly, in some embodiments, the polypeptides as described herein may comprise a mixture of polypeptides with different N-termini. In some embodiments, the N-termini differ in length by 1, 2, 3, 4, or 5 amino acids. In some embodiments, the polypeptide is substantially homogeneous, i.e., the polypeptides have the same N-terminus. In some embodiments, the signal sequence of the polypeptide comprises one or more (e.g., one, two, three, four, five, six, seven, eight, nine, ten, etc.) amino acid substitutions and/or deletions as compared to a “native” or “parental” signal sequence. In some embodiments, the signal sequence of the polypeptide comprises amino acid substitutions and/or deletions that allow one cleavage site to be dominant, thereby resulting in a substantially homogeneous polypeptide with one N-terminus. In some embodiments, a signal sequence of the polypeptide affects the expression level of the polypeptide, e.g., increased expression or decreased expression.
[0070] Experiments determining the epitope of etigilimab are described in WO2016/191643 and are hereby incorporated herein by reference in their entirety. In certain embodiments, the anti-TIGIT antibodies compete for specific binding to TIGIT with the anti- TIGIT antibody comprising: (a) a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), and a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG) and (b) a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6).
[0071] In certain embodiments, the anti-TIGIT antibodies competes for specific binding to human TIGIT with the anti-TIGIT antibody comprising a heavy chain variable region comprising SEQ ID NO:7 and a light chain variable region comprising SEQ ID NOG. In certain embodiments, the anti-TIGIT antibodies competes for specific binding to human TIGIT with the anti-TIGIT antibody comprising a heavy chain comprising SEQ ID NO:9 and a light chain comprising SEQ ID NO: 10.
[0072] In certain embodiments, the anti-TIGIT antibodies compete with etigilimab for specific binding to human TIGIT. In some embodiments, the anti-TIGIT antibody competes with a reference antibody for specific binding to human TIGIT, wherein the reference antibody is etigilimab. In certain embodiments, the anti-TIGIT antibody binds the same epitope, or essentially the same epitope, on TIGIT as etigilimab.
[0073] In another embodiment, the anti-TIGIT antibody binds an epitope on TIGIT that overlaps with the epitope on human TIGIT bound by etigilimab.
[0074] In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids within SEQ ID NO: 13 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids within SEQ ID NO: 14 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids within SEQ ID NO:13 and SEQ ID NO:14 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62 and 1109 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62 and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding with an epitope comprising amino acids Q64 and 1109 of SEQ ID NO:11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q64 and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62, Q64, and 1109 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62, Q64, and T119 of SEQ ID NO:11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q62, 1109, and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising amino acids Q64, 1109, and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti- TIGIT antibody competes for binding to an epitope comprising amino acids Q62, Q64, 1109, and T119 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein. In some embodiments, the anti-TIGIT antibody competes for binding to an epitope comprising at least one amino acid selected from the group consisting of: N58, E60, Q62, Q64, L65, F107, 1109, Hl 11, T117, T119, G120, and R121 of SEQ ID NO: 11 with the anti-TIGIT antibody described herein.
[0075] In certain embodiments, the anti-TIGIT antibody binds TIGIT and modulates TIGIT activity. In some embodiments, the anti-TIGIT antibody is a TIGIT antagonist and decreases TIGIT activity. In certain embodiments, the anti-TIGIT antibody inhibits TIGIT activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody inhibits TIGIT activity is etigilimab.
[0076] In some embodiments, the anti-TIGIT antibody binds TIGIT and inhibits or reduces TIGIT signalling. In certain embodiments, the anti-TIGIT antibody inhibits TIGIT signalling by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%.
[0077] TIGIT is phosphorylated at its cytoplasmic tail after interaction with its counterreceptor PVR. The phosphorylation of TIGIT is the beginning of a cascade that includes downstream events affecting other known signalling pathways. Therefore, evaluating TIGIT phosphorylation can give information about TIGIT activity and TIGIT signalling.
[0078] Phosphorylation assays are known to those of skill in the art and are commonly used to monitor protein activation and/or pathway activation. The assays may be used to monitor the effect of various treatments on activation of a target protein and/or a target pathway. For example, an in vitro phosphorylation assay can be used to evaluate the effect of a TIGIT antagonist on the PVR-induced activation of TIGIT.
[0079] In certain embodiments, the anti-TIGIT antibody inhibits binding of TIGIT to a receptor. In certain embodiments, the anti-TIGIT antibody inhibits binding of TIGIT to PVR. In some embodiments, the anti-TIGIT antibody inhibits binding of TIGIT to PVR-L2, PVR- L3, and/or PVR-L4. In certain embodiments, the inhibition of binding of the anti-TIGIT antibody to PVR is at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In certain embodiments, the inhibition of binding of the anti-TIGIT antibody to PVR-L2, PVR-L3, and/or PVR-L4 is at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In certain embodiments, the anti-TIGIT antibody that inhibits binding of TIGIT to PVR is etigilimab. In certain embodiments, the anti-TIGIT antibody that inhibits binding of TIGIT to PVR-L2, PVR-L3, and/or PVR-L4 is etigilimab. [0080] In certain embodiments, the anti-TIGIT antibody blocks binding of TIGIT to a receptor. In certain embodiments, the anti-TIGIT antibody blocks binding of TIGIT to PVR. In certain embodiments, the blocking of binding of the anti-TIGIT antibody to PVR is at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In some embodiments, the anti-TIGIT antibody blocks binding of TIGIT to PVRL2, PVRL3, and/or PVRL4. In certain embodiments, the blocking of binding of the anti- TIGIT antibody to PVRL2, PVRL3, and/or PVRL4 is at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In certain embodiments, the anti-TIGIT antibody that blocks binding of TIGIT to PVR is etigilimab.
[0081] Binding assays are known to those of skill in the art and are described herein. Binding assays may be used to monitor the effect of a test agent on the interaction between a target protein and the protein’s binding partner (e.g., receptor or ligand). For example, an in vitro binding assay can be used to evaluate if a TIGIT antagonist blocks the interaction of TIGIT to PVR.
[0082] In certain embodiments, the anti-TIGIT antibodies described herein have one or more of the following effects: inhibit proliferation of tumor cells, inhibit tumor growth, reduce the tumorigenicity of a tumor, reduce the tumorigenicity of a tumor by reducing the frequency of cancer stem cells in the tumor, trigger cell death of tumor cells, enhance or boost the immune response, enhance or boost the anti-tumor response, increase cytolytic activity of immune cells, increase killing of tumor cells, increase killing of tumor cells by immune cells, induce cells in a tumor to differentiate, differentiate tumorigenic cells to a non-tumorigenic state, induce expression of differentiation markers in the tumor cells, prevent metastasis of tumor cells, decrease survival of tumor cells, increase cell contact-dependent growth inhibition, increase tumor cell apoptosis, reduce epithelial mesenchymal transition (EMT), or decrease survival of tumor cells.
[0083] In certain embodiments, the anti-TIGIT antibodies inhibit tumor growth. In certain embodiments, the anti-TIGIT antibodies inhibit tumor growth in vivo (e.g., in a mouse model, and/or in a human having cancer). In certain embodiments, tumor growth is inhibited at least about two-fold, about three-fold, about five-fold, about ten-fold, about 50-fold, about 100- fold, or about 1000-fold as compared to an untreated tumor.
[0084] In certain embodiments, the anti-TIGIT antibodies bind TIGIT and modulate an immune response. In some embodiments, the anti-TIGIT antibody activates and/or increases an immune response. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances cell-mediated immunity. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances innate cell-mediated immunity. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances adaptive cell-mediated immunity. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances T-cell activity. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances cytolytic T-cell (CTL) activity. In certain embodiments, the antibody increases the activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances NK cell activity. In certain embodiments, the antibody increases the activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances lymphokine-activated killer cell (LAK) activity. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances tumorinfiltrating lymphocyte (TIL) activity. In some embodiments, the anti-TIGIT antibody inhibits or decreases Treg cell activity. In some embodiments, the anti-TIGIT antibody inhibits or decreases MDSC activity. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances tumor cell killing. In some embodiments, the anti-TIGIT antibody increases, promotes, or enhances the inhibition of tumor growth.
[0085] In some embodiments, the anti-TIGIT antibody induces and/or enhances a Thl- type immune response. In general, a Th 1 -type immune response includes production of interferon-gamma (IFN-y), IL-2, and tumor necrosis factor-beta (TNF-P). In comparison, a Th2-type immune response generally includes production of IL-4, IL-5, IL-6, IL-9, IL-10, and IL- 13. In some embodiments, the anti-TIGIT antibody induces and/or increases cytokine or lymphokine production. In some embodiments, the induction and/or increase in cytokine or lymphokines production may be an indirect effect.
[0086] In certain embodiments, the anti-TIGIT antibody increases activation of NK cells. In certain embodiments, the anti-TIGIT antibody increases activation of T-cells. In certain embodiments, the activation of NK cells and/or T-cells by the anti-TIGIT antibody results in an increase in the level of activation of NK cells and/or T-cells of at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, or at least about 95%. In certain embodiments, the anti-TIGIT antibody that increases activation of NK cells is etigilimab.
[0087] In certain embodiments, the anti-TIGIT antibody is an antagonist of regulatory T- cell (Treg) activity. In certain embodiments, the anti-TIGIT antibody inhibits or decreases the activity of Tregs. In certain embodiments, the inhibition of activity of Tregs by the anti-TIGIT antibody results in an inhibition of suppressive activity of a Treg cell of at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 90%, at least about 95%, or about 100%. In certain embodiments, the anti-TIGIT antibody that inhibits Treg activity is etigilimab.
[0088] In certain embodiments, the anti-TIGIT antibody is an antagonist of myeloid- derived suppressor cells (MDSCs). In certain embodiments, the anti-TIGIT antibody inhibits MDSC activity. In certain embodiments, the anti-TIGIT antibody inhibits MDSC activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody that inhibits MDSC activity is etigilimab.
[0089] In certain embodiments, the anti-TIGIT antibody increases natural killer (NK) cell activity. In certain embodiments, the anti-TIGIT antibody increases NK cell activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody that increases NK cell activity is etigilimab.
[0090] In certain embodiments, the anti-TIGIT antibody increases tumor-infiltrating lymphocyte (TIL) activity. In certain embodiments, the anti-TIGIT antibody increases TIL activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody that increases TIL cell activity is etigilimab.
[0091] In certain embodiments, the anti-TIGIT antibody increases or enhances lymphokines-activated killer cell (LAK) activity. In certain embodiments, the anti-TIGIT antibody increases LAK activity by at least about 10%, at least about 20%, at least about 30%, at least about 50%, at least about 75%, at least about 90%, or about 100%. In certain embodiments, the anti-TIGIT antibody that increases LAK cell activity is etigilimab.
[0092] In vivo and in vitro assays for determining whether the anti-TIGIT antibody modulates an immune response are known in the art or are being developed. In some embodiments, a functional assay that detects T-cell activation may be used. In some embodiments, a functional assay that detects T-cell proliferation may be used. In some embodiments, a functional assay that detects NK activity may be used. In some embodiments, a functional assay that detects CTL activity may be used. In some embodiments, a functional assay that detects Treg activity may be used. In some embodiments, a functional assay that detects MDSC activity may be used. In some embodiments, a functional assay that detects production of cytokines or lymphokines or cells producing cytokines or lymphokines may be used. In some embodiments, an ELISpot assay is used to measure antigen-specific T-cell frequency. In some embodiments, an ELISpot assay is used to measure cytokine release/production and/or used to measure the number of cytokine producing cells. In some embodiments, cytokine assays are used to identify a Thl-type response. In some embodiments, cytokine assays are used to identify a Th2-type response. In some embodiments, cytokine assays are used to identify a Thl7-type response. In some embodiments, FACS analysis is used to measure activation markers on immune cells, including but not limited to, T-cells, B-cells, NK cells, macrophages, and/or myeloid cells.
[0093] In certain embodiments, the anti-TIGIT antibodies have a circulating half-life in humans of at least about 2 hours, at least about 5 hours, at least about 10 hours, at least about 24 hours, at least about 3 days, at least about 1 week, or at least about 2 weeks. In certain embodiments, the anti-TIGIT antibody is an IgG (e.g., IgGl, IgG2, or IgG4) antibody that has a circulating half-life in humans of at least about 2 hours, at least about 5 hours, at least about 10 hours, at least about 24 hours, at least about 3 days, at least about 1 week, or at least about 2 weeks. Methods of increasing (or decreasing) the half-life of agents such as polypeptides and antibodies are known in the art. For example, known methods of increasing the circulating halflife of IgG antibodies include the introduction of mutations in the Fc region which increase the pH-dependent binding of the antibody to the neonatal Fc receptor (FcRn) at pH 6.0. Known methods of increasing the circulating half-life of antibody fragments lacking the Fc region include such techniques as PEGylation.
[0094] In certain embodiments, the anti-TIGIT antibodies are used in any one of a number of conjugated (i.e. an immunoconjugate or radioconjugate) or non-conjugated forms. In certain embodiments, the antibodies can be used in a non-conjugated form to harness the patient’s natural defense mechanisms including complement-dependent cytotoxicity (CDC) and antibody dependent cellular cytotoxicity (ADCC) to eliminate malignant or cancer cells. [0095] In some embodiments, the anti-TIGIT antibody is conjugated to a cytotoxic agent. In some embodiments, the anti-TIGIT antibody is conjugated to a cytotoxic agent as an ADC (antibody-drug conjugate). In some embodiments, the cytotoxic agent is a chemotherapeutic agent including, but not limited to, methotrexate, adriamycin/doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin, pyrrolobenzodiazepines (PBDs), or other intercalating agents. In some embodiments, the cytotoxic agent is an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof, including, but not limited to, diphtheria A chain, non-binding active fragments of diphtheria toxin, exotoxin A chain, ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), Momordica charantia inhibitor, curcin, crotin, Sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. In some embodiments, the cytotoxic agent is a radioisotope to produce a radioconjugate or a radioconjugated antibody. A variety of radionuclides are available for the production of radioconjugated antibodies including, but not limited to, 90Y, 1251, 1311, 1231, Ulin, 131In, 105Rh, 153Sm, 67Cu, 67Ga, 166Ho, 177Lu, 186Re, 188Re and 212Bi. Conjugates of an antibody and one or more small molecule toxins, such as calicheamicins, maytansinoids, trichothenes, and CC1065, and the derivatives of these toxins that have toxin activity, can also be used. Conjugates of an antibody and cytotoxic agent may be made using a variety of bifunctional protein-coupling agents such as N-succinimidyl- 3-(2-pyridyidithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis(p- azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)- ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as l,5-difluoro-2,4-dinitrobenzene).
Fragments
[0096] In some embodiments, the invention provides a fragment of an anti-TIGIT antibody, wherein the antibody fragment comprises an antigen-binding site. In some embodiments, the antibody fragment comprises an IgG antibody, an IgGl antibody, an IgG2 antibody, or an IgG4 antibody. Antibody fragments may have different functions or capabilities than intact antibodies; for example, antibody fragments can have increased tumor penetration. Various techniques are known for the production of antibody fragments including, but not limited to, proteolytic digestion of intact antibodies. In some embodiments, antibody fragments include a F(ab')2 fragment produced by pepsin digestion of an antibody molecule. In some embodiments, antibody fragments include a Fab fragment generated by reducing the disulfide bridges of an F(ab')2 fragment. In other embodiments, antibody fragments include a Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent. In certain embodiments, antibody fragments are produced by recombinant methods. In some embodiments, antibody fragments include Fv or single chain Fv (scFv) fragments. Fab, Fv, and scFv antibody fragments can be expressed in and secreted from E. coli or other host cells, allowing for the production of large amounts of these fragments. In some embodiments, antibody fragments are isolated from antibody phage libraries as discussed herein. For example, methods can be used for the construction of Fab expression libraries to allow rapid and effective identification of monoclonal Fab fragments with the desired specificity for TIGIT or derivatives, fragments, analogs or homologs thereof. In some embodiments, antibody fragments are linear antibody fragments. In certain embodiments, antibody fragments are monospecific or bispecific. In certain embodiments, the anti-TIGIT antibody is a scFv. Various techniques can be used for the production of single -chain antibodies specific to TIGIT.
[0097] In some embodiments, especially in the case of antibody fragments, an antibody is modified in order to alter (e.g., increase or decrease) its serum half-life. This can be achieved, for example, by incorporation of a salvage receptor binding epitope into the antibody fragment by mutation of the appropriate region in the antibody fragment or by incorporating the epitope into a peptide tag that is then fused to the antibody fragment at either end or in the middle (e.g., by DNA or peptide synthesis).
Bispecific antibodies and heterodimeric agents
[0098] In some embodiments, the anti-TIGIT antibody is a bispecific antibody. Thus, this invention encompasses bispecific anti-TIGIT antibody that specifically recognize TIGIT and at least one additional target. Bispecific antibodies are capable of specifically recognizing and binding at least two different antigens or epitopes. The different epitopes can either be within the same molecule (e.g., two epitopes on TIGIT) or on different molecules (e.g., one epitope on TIGIT and one epitope on a different protein). In some embodiments, a bispecific antibody has enhanced potency as compared to an individual antibody or to a combination of more than one antibody. In some embodiments, a bispecific antibody has reduced toxicity as compared to an individual antibody or to a combination of more than one antibody. It is known to those of skill in the art that any therapeutic agent may have unique pharmacokinetics (PK) (e.g., circulating half-life). In some embodiments, a bispecific antibody has the ability to synchronize the PK of two active binding agents wherein the two individual binding agents have different PK profiles. In some embodiments, a bispecific antibody has the ability to concentrate the actions of two agents in a common area (e.g., a tumor and/or tumor microenvironment). In some embodiments, a bispecific antibody has the ability to concentrate the actions of two agents to a common target (e.g., a tumor or a tumor cell). In some embodiments, a bispecific antibody has the ability to target the actions of two agents to more than one biological pathway or function. In some embodiments, a bispecific antibody has the ability to target two different cells and bring them closer together (e.g., an immune cell and a tumor cell).
[0099] In some embodiments, the bispecific antibody is a monoclonal antibody. In some embodiments, the bispecific antibody is a humanized antibody. In some embodiments, the bispecific antibody is a human antibody. In some embodiments, the bispecific antibody is an IgGl antibody. In some embodiments, the bispecific antibody is an IgG2 antibody. In some embodiments, the bispecific antibody is an IgG4 antibody. In some embodiments, the bispecific antibody has decreased toxicity and/or side effects. In some embodiments, the bispecific antibody has decreased toxicity and/or side effects as compared to a mixture of the two individual antibodies or the antibodies as single agents. In some embodiments, the bispecific antibody has an increased therapeutic index. In some embodiments, the bispecific antibody has an increased therapeutic index as compared to a mixture of the two individual antibodies or the antibodies as single agents.
[00100] In some embodiments, the bispecific antibodies can specifically recognize and bind TIGIT as well as a second antigen target, such as CD2, CD3, CD28, CTLA4, PD-1, PD- Ll, CD80, CD86, CD64, CD32, or CD16, TIM-3, LAG-3, OX-40, 4-1BB, or GITR. In certain embodiments, the bispecific antibodies can specifically recognize and bind TIGIT as well as specifically recognizing and binding to PD-1 or PD-L1. In some embodiments, the antibodies can be used to direct cytotoxic agents to cells which express a particular target antigen. These antibodies possess an antigen-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA, or TETA.
[00101] Techniques for making bispecific antibodies are known by those skilled in the art. In some embodiments, the bispecific antibodies comprise heavy chain constant regions with modifications in the amino acids which are part of the interface between the two heavy chains. In some embodiments, the bispecific antibodies can be generated using a “knobs-into-holes” strategy. In some cases, the “knobs” and “holes” terminology is replaced with the terms “protuberances” and “cavities”. In some embodiments, the bispecific antibodies may comprise variant hinge regions incapable of forming disulfide linkages between the heavy chains. In some embodiments, the modifications may comprise changes in amino acids that result in altered electrostatic interactions. In some embodiments, the modifications may comprise changes in amino acids that result in altered hydrophobic/hydrophilic interactions.
[00102] Bispecific antibodies can be intact antibodies or antibody fragments comprising antigen-binding sites. Antibodies with more than two valencies are also contemplated. For example, trispecific antibodies can be prepared. Thus, in certain embodiments the antibodies to TIGIT are multispecific.
[00103] In some embodiments, the bispecific antibodies bind both TIGIT and the second target with a KD of about lOOnM or less. In some embodiments, the anti-TIGIT antibody binds both TIGIT and the second target with a KD of about 50nM or less. In some embodiments, the anti-TIGIT antibody binds both TIGIT and the second target with a KD of about 20nM or less. In some embodiments, the anti-TIGIT antibody binds both TIGIT and the second target with a KD of about lOnM or less. In some embodiments, the anti-TIGIT antibody binds both TIGIT and the second target with a KD of about InM or less. In some embodiments, the affinity of one of the antigen-binding sites may be weaker than the affinity of the other antigen-binding site. For example, the KD of one antigen binding site may be about InM and the KD of the second antigen-binding site may be about lOnM. In some embodiments, the difference in affinity between the two antigen-binding sites may be about 2-fold or more, about 3-fold or more, about 5-fold or more, about 8-fold or more, about 10-fold or more, about 15-fold or more, about 20- fold or more, about 30-fold or more, about 50-fold or more, or about 100-fold or more. Modulation of the affinities of the two antigen-binding sites may affect the biological activity of the bispecific antibody. For example, decreasing the affinity of the antigen-binding site for TIGIT or the second target, may have a desirable effect, for example decreased toxicity of the binding agent and/or increased therapeutic index.
[00104] In some embodiments, the anti-TIGIT antibody is a heterodimeric agent or heterodimeric molecule. In some embodiments, a heterodimeric molecule comprises a first arm which binds human TIGIT and a second arm which binds a second target. In some embodiments, a heterodimeric molecule comprises a first arm that specifically binds human TIGIT and a second arm, wherein the first arm comprises an anti-TIGIT antibody. In some embodiments, a heterodimeric molecule comprises a first arm that binds human TIGIT and a second arm which comprises an antigen-binding site from an antibody that specifically binds a second target. In some embodiments, a heterodimeric molecule is a bispecific antibody. In some embodiments, a heterodimeric molecule comprises a first arm that binds human TIGIT and a second arm that specifically binds a tumor antigen. In some embodiments, a heterodimeric molecule comprises a first arm that binds human TIGIT and a second arm that specifically binds PD-1, PD-L1, CTLA-4, CD2, CD3, CD28, CD80, CD86, CD64, CD32, CD16, TIM-3, LAG-3, OX-40, 4-1BB, or GITR. In some embodiments, a heterodimeric molecule comprises a first arm that binds TIGIT and a second arm that comprises an immunotherapeutic agent. In some embodiments, the immunotherapeutic agent is selected from the group consisting of: granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage colony stimulating factor (M-CSF), granulocyte colony stimulating factor (G-CSF), interleukin 2 (IL-2), interleukin 3 (IL-3), interleukin 12 (IL-12), interleukin 15 (IL-15), B7-1 (CD80), B7-2 (CD86), 4-1BB ligand, GITRL, OX-40L, anti-CD3 antibody, anti-CTLA-4 antibody, anti-PD-1 antibody, anti-PD-Ll antibody, anti-4-lBB antibody, anti-GITR antibody, anti-OX-40 antibody, anti-LAG-3 antibody, and anti-TIM-3 antibody.
Anti-PD-1 antibodies
[00105] As described above, the invention provides methods of treating various cancers using a combination of an anti-TIGIT antibody and an anti-PD-1 antibody. Non-limiting examples of anti-PD-1 antibodies include pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo),dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab. Therefore, in some embodiments the anti-PD-1 antibody is selected from the group consisting of pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo), dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab.
[00106] The invention is exemplified with data based on the combination of etigilimab and the anti-PD-1 antibody nivolumab. Without wishing to be bound by theory, the inventors rationalise that the surprising technical effects achieved by this combination would also be seen with other anti-PD-1 antibodies described herein. The surprising technical effects of this combination are thought to be principally attributable to etigilimab and its functionality rather than the anti-PD-1 antibody component of the combination therapy.
[00107] In the tumor microenvironment, PD-1 and its ligand PD-L1 perform a role in tumor progression and survival by escaping tumor neutralizing immune surveillance. Anti-PD- 1 and anti-PD-Ll antibodies act by blocking the binding of PD-1 to its ligand PD-L1 which prevents cancer cells from evading the immune system. The inventors hypothesize that the surprising technical effects achieved by nivolumab (anti-PD-1 antibody) in combination with etigilimab would also be seen with other anti-PD-1 and PD-L1 antibodies described herein as all of these antibodies act by blocking PD-1/PD-L1 signalling.
[00108] The heavy chain sequence of pembrolizumab is SEQ ID NO: 15. The light chain sequence of pembrolizumab is SEQ ID NO: 16. The heavy chain sequence of nivolumab is SEQ ID NO: 17. The light chain sequence of nivolumab is SEQ ID NO: 18. The heavy chain sequence of cemiplimab is SEQ ID NO: 19. The light chain sequence of cemiplimab is SEQ ID NO:20. The heavy chain sequence of dostarlimab is SEQ ID NO:21. The light chain sequence of dostarlimab is SEQ ID NO:22. The heavy chain sequence of Tislelizumab is SEQ ID NO:40. The light chain sequence of Tislelizumab is SEQ ID NO:41. The heavy chain sequence of Sintilimab is SEQ ID NO:42. The light chain sequence of Sintilimab is SEQ ID NO:43. The heavy chain sequence of Zimberelimab is SEQ ID NO:44. The light chain sequence of Zimberelimab is SEQ ID NO:45. The heavy chain sequence of Toripalimab is SEQ ID NO:46. The light chain sequence of Toripalimab is SEQ ID NO:47. The heavy chain sequence of Penpulimab is SEQ ID NO:48. The light chain sequence of Penpulimab is SEQ ID NO:49. The heavy chain sequence of Balstilimab is SEQ ID NO:50. The light chain sequence of Balstilimab is SEQ ID N0:51. The heavy chain sequence of Retifanlimab is SEQ ID NO:52. The light chain sequence of Retifanlimab is SEQ ID NO:53. The light chain sequence of Cetrelimab is SEQ ID NO:54. The heavy chain sequence of Cetrelimab is SEQ ID NO:55. The light chain sequence of Budigalimab is SEQ ID NO:56. The light chain sequence of Budigalimab is SEQ ID NO:57. The heavy chain sequence of Pimivalimab is SEQ ID NO:58. The light chain sequence of Pimivalimab is SEQ ID NO:59. The light chain sequence of Spartalizumab is SEQ ID NO:60. The heavy chain sequence of Spartalizumab is SEQ ID N0:61. The light chain sequence of Serplulimab is SEQ ID NO:62. The light chain sequence of Serplulimab is SEQ ID NO:63. The heavy chain sequence of Sasanlimab is SEQ ID NO:63. The light chain sequence of Sasanlimab is SEQ ID NO:65.
[00109] In some embodiments, the anti-PD-1 antibody comprises a heavy chain comprising SEQ ID NO: 15 and a light chain comprising SEQ ID NO: 16. In some embodiments, the anti-PD-1 antibody comprises a heavy chain comprising SEQ ID NO: 17 and a light chain comprising SEQ ID NO: 18. In some embodiments, the anti-PD-1 antibody comprises a heavy chain comprising SEQ ID NO: 19 and a light chain comprising SEQ ID NO:20. In some embodiments, the anti-PD-1 antibody comprises a heavy chain comprising SEQ ID NO:21 and a light chain comprising SEQ ID NO:22.
[00110] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and nivolumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00111] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and pembrolizumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma. [00112] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and cemiplimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00113] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and dostarlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00114] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Tislelizumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00115] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Sintilimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00116] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Zimberelimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00117] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Toripalimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00118] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Penpulimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00119] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Balstilimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID N0:4), a light chain CDR2 comprising SASYRYT (SEQ ID NO:5), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00120] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Retifanlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00121] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Cetrelimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00122] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Budigalimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00123] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Pimivalimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00124] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and spartalizumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00125] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Camrelizumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00126] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Prolgolimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00127] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and Pucotenlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00128] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and serplulimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00129] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and sasanlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOH), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00130] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and ezabenlimab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOH), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00131] In some embodiments, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and genolimzumab, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00132] In alternative aspects, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD-Ll antibody, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00133] Non-limiting examples of anti-PD-Ll antibodies include atezolizumab (Tecentriq), avelumab (Bavencio),durvalumab (Imfinzi), Envafolimab, Sugemalimab, Socazolimab, Adebrelimab, Pacmilimab, Lodapolimab and Cosibelimab. Therefore, in some embodiments, the anti-PD-Ll antibody is selected from the group consisting of atezolizumab (Tecentriq), avelumab (Bavencio), durvalumab (Imfinzi), Envafolimab, Sugemalimab, Socazolimab, Adebrelimab, Pacmilimab, Lodapolimab and Cosibelimab. The heavy chain sequence of atezolizumab is SEQ ID NO:23. The light chain sequence of atezolizumab is SEQ ID NO:24. The heavy chain sequence of avelumab is SEQ ID NO:25. The light chain sequence of avelumab is SEQ ID NO:26. The heavy chain sequence of durvalumab is SEQ ID NO:27. The light chain sequence of durvalumab is SEQ ID NO:28.
[00134] In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:23 and a light chain comprising SEQ ID NO:24. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:25 and a light chain comprising SEQ ID NO:26. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:27 and a light chain comprising SEQ ID NO:28. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:66 and a light chain comprising SEQ ID NO: 67. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO: 68 and a light chain comprising SEQ ID NO:69. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:70 and a light chain comprising SEQ ID NO:71. In some embodiments, the anti- PD-L1 antibody comprises a heavy chain comprising SEQ ID NO:72 and a light chain comprising SEQ ID NO:73. In some embodiments, the anti-PD-Ll antibody comprises a heavy chain comprising SEQ ID NO:74 and a light chain comprising SEQ ID NO:75.
[00135] Any of the aspects/ embodiments described herein in relation to anti-PD-1 antibodies apply equally to the anti-PD-Ll antibodies described above.
Methods of treatment
[00136] The invention provides methods of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD- 1 antibody, wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma. In some aspects, the invention provides methods of treating cancer in a human patient, comprising administering to the human patient an anti-TIGIT antibody and an anti-PD-1 antibody, wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00137] The invention also provides an anti-TIGIT antibody and an anti-PD-1 antibody for use in treating cancer in a human patient, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00138] The invention also provides the use of an anti-TIGIT antibody and an anti-PD-1 antibody in the preparation of a medicament for treating cancer in a human patient, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO: 1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumor and uveal melanoma.
[00139] Soft tissue sarcomas are a rare type of cancer that start in soft tissues (muscle, tendons, fat, lymph and blood vessels, and nerves). These cancers can develop anywhere in the body but are found mostly in the arms, legs, chest, and abdomen. Types of soft tissue sarcoma include leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, fibrosarcomas, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid haemangioendothelioma, malignant peripheral nerve sheath tumors (MPNST), gastrointestinal Stromal Tumors (GISTs), follicular dendritic cell sarcoma, epithelioid sarcoma, myxofibrosarcoma, solitary fibrous tumor, undifferentiated (pleomorphic sarcoma), fibromatosis or dermatofibrosarcoma protuberans.
[00140] The invention provides a method of treating a soft tissue sarcoma in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD-1 antibody, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NO:6).
[00141] In some embodiments, the soft tissue sarcoma is selected from the group consisting of leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, dedifferentiated liposarcoma, fibrosarcomas, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid haemangioendothelioma, malignant peripheral nerve sheath tumours (MPNST), gastrointestinal Stromal Tumours (GISTs), follicular dendritic cell sarcoma, epithelioid sarcoma, myxofibrosarcoma, solitary fibrous tumor, undifferentiated (pleomorphic sarcoma), fibromatosis and dermatofibrosarcoma protuberans. In certain embodiments, the soft tissue sarcoma is selected from the group consisting of leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, dedifferentiated liposarcoma, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid, malignant peripheral nerve sheath tumours (MPNST) and undifferentiated (pleomorphic sarcoma). In further embodiments, the soft tissue sarcoma is a dedifferentiated liposarcoma or a undifferentiated (pleomorphic sarcoma).
[00142] About 95 percent of testicular cancers begin in germ cells. Two of the most common germ cell tumors are seminoma and nonseminoma. Seminomas make up about half of all germ cell tumors. They usually grow slowly. Nonseminomas are often more aggressive than seminomas, and more likely to spread beyond the testicle. These types occur about equally. Many testicular cancers contain both seminoma and non-seminoma cells. These mixed germ cell tumors are treated as non-seminomas because they grow and spread like non-seminomas. [00143] The invention provides a method of treating a testicular germ cell tumor in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD-1 antibody, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG).
[00144] In some embodiments, the testicular germ cell tumor is a seminomas testicular germ cell tumor or a non-seminomas testicular germ cell tumor. In some embodiments, the testicular germ cell tumor comprises both seminoma and non-seminoma cells. In some embodiments, the testicular germ cell tumor patient has inadequate renal function for high dose chemotherapy. In some embodiments, the testicular germ cell tumor patient has inadequate stem cell collection for high dose chemotherapy.
[00145] Uveal melanoma is a rare cancer of the eye involving the iris, ciliary body, or choroid (collectively referred to as the uvea). Tumors arise from the pigment cells (melanocytes) that reside within the uvea and give color to the eye. These melanocytes are distinct from the retinal pigment epithelium cells underlying the retina that do not form melanomas. Uveal melanomas are sometimes referred to by their location, choroidal melanoma, ciliary body melanoma, or iris melanoma. Iris and ciliary body melanomas can be described as anterior melanomas, whereas a choroidal melanoma behind the equator of the eye would be considered posterior uveal melanomas. Examples of uveal melanomas include posterior uveal melanoma, choroidal melanoma, ciliary body melanoma and iris melanoma.
[00146] The invention provides a method of treating uveal melanoma in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti- TIGIT antibody and an anti-PD-1 antibody, wherein the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NOG), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NOG), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG). In some embodiments, the uveal melanoma is selected from the group consisting of a choroidal melanoma, a ciliary body melanoma or an iris melanoma. In some embodiments, the uveal melanoma is an anterior uveal melanoma or a posterior uveal melanomas. Mode of administration
[00147] The anti-TIGIT antibody can be administered prior to, concurrently with and/or subsequently to the administration of the anti-PD-1 antibody in order to treat the cancer in human patient. Combined administration can include co-administration, either in a single pharmaceutical formulation or using separate formulations, or consecutive administration in either order but generally within a time period such that all active agents can exert their biological activities simultaneously. It will be appreciated that the combination of an anti- TIGIT antibody and an anti-PD- 1 antibody may be administered in any order or concurrently. [00148] The anti-TIGIT antibody and the anti-PD-1 antibody can be administered in any number of ways for either local or systemic treatment. In some embodiments, administration is (i) topical by epidermal or transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders; (ii) pulmonary by inhalation or insufflation of powders or aerosols, including by nebulizer, intratracheal, and intranasal; (iii) oral; or (iv) parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e.g., injection or infusion), or intracranial (e.g., intrathecal or intraventricular). [00149] In certain embodiments, the anti-TIGIT antibodies and the anti-PD- 1 antibodies described herein are administered intravenously. In certain embodiments, the anti-TIGIT antibodies and the anti-PD-1 antibodies described herein are administered subcutaneously.
Dosing
[00150] In some embodiments, the anti-TIGIT antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks. In certain embodiments, the anti-TIGIT antibody is administered once every two weeks. In some embodiments, the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks. In some embodiments, both the anti-TIGIT antibody and the anti-PD-1 antibody are administered once every week, once every two weeks, once every three weeks, or once every four weeks. In other embodiments, the anti-TIGIT antibody and the anti-PD-1 antibody are administered at different intervals. For example, the anti-TIGIT antibody is administered once every two weeks and the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks. In certain embodiments, etigilimab is administered once every two weeks and the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks.
[00151] In some embodiments, the anti-TIGIT antibody is administered once every two weeks and the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks, once every five weeks or once every six weeks.
[00152] In some embodiments, etigilimab is administered once every two weeks and the anti-PD-1 antibody is administered once every week, once every two weeks, once every three weeks, once every four weeks, once every five weeks or once every six weeks.
[00153] In some embodiments, etigilimab is administered once every two weeks and pembrolizumab is administered once every three weeks, or once every six weeks. In some embodiments, etigilimab is administered once every two weeks and nivolumab is administered once every two weeks, or once every four weeks. In some embodiments, etigilimab is administered once every two weeks and cemiplimab is administered once every three weeks. In some embodiments, etigilimab is administered once every two weeks and dostarlimab is administered once every three weeks, or once every six weeks.
[00154] In some embodiments, the anti-TIGIT antibody is administered once every two weeks at a flat dose of 1000 mg. In some embodiments, the anti-PD-1 antibody is administered at a flat dose of 200 mg-1000 mg. In some embodiments, the anti-PD-1 antibody is administered at a flat dose of 200 mg, 250 mg, 300 mg, 350 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg or 1000 mg. In certain embodiments, the -PD-1 antibody is administered at a flat dose of 200 mg, 240 mg, 350 mg, 400mg, 500 mg or 1000 mg.
[00155] In some embodiments, etigilimab is administered once every two weeks at a flat dose of 1000 mg and pembrolizumab is administered once every three weeks at a flat dose of 200 mg, or once every six weeks at a flat dose of 400 mg. In some embodiments, etigilimab is administered once every two weeks at a flat dose of 1000 mg and nivolumab is administered once every two weeks at a flat dose of 240 mg, or once every four weeks at a flat dose of 480 mg. In some embodiments, etigilimab is administered once every two weeks at a flat dose of 1000 mg and cemiplimab is administered once every three weeks at a flat dose of 350 mg. In some embodiments, etigilimab is administered once every two weeks at a flat dose of 1000 mg and dostarlimab is administered once every three weeks at a flat dose of 500 mg, or once every six weeks at a flat dose of 1000 mg.
Patients
[00156] The human patient can be an infant (between the ages 0-1 years), a child (between the ages of 1-18 years) or an adult (with an age above 18 years). In some embodiments, the patient is aged above 18 years old, 20 years old, 30 years old or 40 years old. In certain embodiments, the human patient is aged above 50 years old, for example 55 years or older, 60 years or older, 65 years or older, 70 years or older, 75 years or older or 80 years or older. [00157] The anti-TIGIT antibody and the anti-PD-1 antibody can be administered to patients that have previously undergone treatment with an anti-cancer agent. For example, in some embodiments, the human patient has previously undergone treatment with one or more anti-cancer agents, such as with two or more or three or more anti-cancer agents. In some embodiments, the human patient has previously received one, two or three anti-cancer agents. In some embodiments, the human patient has been previously treated with one or more, two or more or three or more anti -cancer agents and there is tumour growth, progression, or recurrence during or after treatment with the one or more, two or more or three or more anti-cancer agents. [00158] In certain other embodiments, the anti-TIGIT antibody, the anti-PD-1 antibody and the anti-cancer agent agent will be administered substantially simultaneously or concurrently. For example, a patient may be given the anti-TIGIT antibody and the anti-PD-1 antibody while undergoing a course of treatment with an anti-cancer agent (e.g., chemotherapy). In certain embodiments, the anti-TIGIT antibody and the anti-PD-1 antibody will be administered within 1 year of the treatment with the anti-cancer agent. In certain alternative embodiments, the anti-TIGIT antibody and the anti-PD-1 antibody will be administered within 10, 8, 6, 4, or 2 months of any treatment with the anti-cancer agent. In certain other embodiments, the anti-TIGIT antibody and the anti-PD-1 antibody will be administered within 4, 3, 2, or 1 weeks of any treatment with the anti-cancer agent. In some embodiments, the anti-TIGIT antibody and the anti-PD-1 antibody will be administered within 5, 4, 3, 2, or 1 days of any treatment with the anti-cancer agent. It will further be appreciated that the two (or more) anti-cancer agents or treatments may be administered to the patient within a matter of hours or minutes (i.e., substantially simultaneously).
[00159] The anti-cancer agent can be a checkpoint inhibitor, a chemotherapeutic agent or a cell therapy.
[00160] In some embodiments, the human patient has not received prior treatment been treated with a checkpoint inhibitor. In some embodiments, the human patient is naive to checkpoint inhibitor therapy. In some embodiments, the human patient has not been previously treated with a checkpoint inhibitor. In some embodiments, the human patient has not received prior treatment/ been treated with an anti-PD-1 antibody therapy. In some embodiments, the human patient is naive to anti-PD-1 antibody therapy. In some embodiments, the human patient has not been previously treated with an anti-PD-1 antibody therapy. In some embodiments, the human patient has not received prior treatment/ been treated with an anti-PD-Ll antibody therapy. In some embodiments, the human patient is naive to anti-PD-Ll antibody therapy. In some embodiments, the human patient has not been previously treated with an anti-PD-Ll antibody therapy. In some embodiments, the human patient has not received prior treatment/ been treated with a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1. In some embodiments, the human patient is naive to a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1. In some embodiments, the human patient has not been previously treated with a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1.
[00161] In some embodiments, the human patient has not received prior radiotherapy. In some embodiments, the human patient has not been previously treated with radiotherapy. In some embodiments, the human patient has not received prior chemotherapy. In some embodiments, the human patient has not been previously treated with chemotherapy.
[00162] In some embodiments, the human patient has previously treated with a checkpoint inhibitor and there is tumor growth, progression, or recurrence during or after treatment with the checkpoint inhibitor. In certain embodiments, the human patient has been previously treated with an anti-PD-1 antibody and there is tumor growth, progression, or recurrence during or after treatment with the PD-1 antibody. In certain embodiments, the human patient has been previously treated with an anti-PD-Ll antibody and there is tumor growth, progression, or recurrence during or after treatment with the PD-L1 antibody.
[00163] In some embodiments, the human patient has previously treated with a cell therapy and there is tumor growth, progression, or recurrence during or after treatment with the cell therapy. In certain embodiments, the human patient has been previously treated with CAR T cell therapy and there is tumor growth, progression, or recurrence during or after treatment with the CAR T cell therapy. In certain embodiments, the human patient has been previously treated with a TCR cell therapy and there is tumor growth, progression, or recurrence during or after treatment with the TCR cell therapy.
[00164] In some embodiments, the human patient has previously treated with an immunotherapeutic agent and there is tumor growth, progression, or recurrence during or after treatment with the immunotherapeutic agent. In certain embodiments, the human patient has been previously treated with tebentafusp and there is tumor growth, progression, or recurrence during or after treatment with tebentafusp.
[00165] In some embodiments, the human patient has previously treated with a chemotherapeutic agent and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agent, for example a chemotherapeutic agent or in combination with a cocktail of chemotherapeutic agents. Classes of therapeutic agents include, for example, anti-tubulin agents, auristatins, DNA minor groove binders, DNA replication inhibitors, alkylating agents (e.g., platinum complexes such as cisplatin, mono(platinum), bis(platinum) and tri-nuclear platinum complexes and carboplatin), anthracyclines, antibiotics, anti-folates, antimetabolites, chemotherapy sensitizers, duocarmycins, etoposides, fluorinated pyrimidines, ionophores, lexitropsins, nitrosoureas, platinols, purine antimetabolites, puromycins, radiation sensitizers, steroids, taxanes, topoisomerase inhibitors, vinca alkaloids, or the like.
[00166] Possible chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamime; nitrogen mustards such as chlorambucil, chlornaphazine, cyclophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, caminomycin, carzinophilin, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5- fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercap topurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytosine arabinoside, dideoxyuridine, doxifluridine, enocitabine, floxuridine, 5-FU; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenishers such as folinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2- ethylhydrazide; procarbazine; PSK; razoxane; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (Ara-C); taxoids, e.g. paclitaxel (TAXOL) and docetaxel (TAXOTERE); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; ibandronate; CPT11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine (XELODA); and pharmaceutically acceptable salts, acids or derivatives of any of the above. Chemotherapeutic agents also include anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and toremifene (FARESTON); and antiandrogens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above. In certain embodiments, the human patient has previously treated with a platinum-based therapies such as cisplatin or carboplatin.
[00167] In some embodiments, the patient has previously been treated with Lenvatinib. In some embodiments, the patient has previously been treated with an anti-LAG3 antibody, for example Relatlimab, PRS-332, P13B02-3, LBL-007, Eftilagimod alpha (IMP321), LAG525 (IMP701), MK-4280, BI 754111, FS118, Tebotelimab, TSR-033, INCAGN2385, Sym022, XmAb22841 or REGN3767. In certain embodiments, the patient has previously been treated with Relatlimab.
[00168] In some embodiments, the patient has previously been treated with pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo), dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab. In some embodiments, the patient has previously been treated with atezolizumab (Tecentriq), avelumab (Bavencio), durvalumab (Imfinzi), Envafolimab, Sugemalimab, Socazolimab, Adebrelimab, Pacmilimab, Lodapolimab and Cosibelimab.
[00169] In certain embodiments, the chemotherapeutic agent is a topoisomerase inhibitor. Topoisomerase inhibitors are chemotherapy agents that interfere with the action of a topoisomerase enzyme (e.g., topoisomerase I or II). Topoisomerase inhibitors include, but are not limited to, doxorubicin HC1, daunorubicin citrate, mitoxantrone HC1, actinomycin D, etoposide, topotecan HC1, teniposide (VM-26), and irinotecan, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these. In some embodiments, the chemotherapeutic agent is irinotecan.
[00170] In certain embodiments, the chemotherapeutic agent is an anti-metabolite. An anti-metabolite is a chemical with a structure that is similar to a metabolite required for normal biochemical reactions, yet different enough to interfere with one or more normal functions of cells, such as cell division. Anti-metabolites include, but are not limited to, gemcitabine, fluorouracil, capecitabine, methotrexate sodium, ralitrexed, pemetrexed, tegafur, cytosine arabinoside, thioguanine, 5-azacytidine, 6-mercaptopurine, azathioprine, 6-thioguanine, pentostatin, fludarabine phosphate, and cladribine, as well as pharmaceutically acceptable salts, acids, or derivatives of any of these. In certain embodiments, the chemotherapeutic agent is gemcitabine.
[00171] In certain embodiments, the chemotherapeutic agent is an antimitotic agent, including, but not limited to, agents that bind tubulin. In some embodiments, the agent is a taxane. In certain embodiments, the agent is paclitaxel or docetaxel, or a pharmaceutically acceptable salt, acid, or derivative of paclitaxel or docetaxel. In certain embodiments, the agent is paclitaxel (TAXOL), docetaxel (TAXOTERE), albumin-bound paclitaxel (ABRAXANE®), DHA-paclitaxel, or PG-paclitaxel. In certain alternative embodiments, the antimitotic agent comprises a vinca alkaloid, such as vincristine, vinblastine, vinorelbine, or vindesine, or pharmaceutically acceptable salts, acids, or derivatives thereof. In some embodiments, the antimitotic agent is an inhibitor of kinesin Eg5 or an inhibitor of a mitotic kinase such as Aurora A or Plkl. In certain embodiments, the chemotherapeutic agent is paclitaxel. In certain embodiments, the chemotherapeutic agent is albumin-bound paclitaxel (ABRAXANE®).
[00172] In some embodiments, the human patient has previously treated with 2 or more chemotherapeutic agents and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agents. For example, the human patient has previously treated with 2 or more, 3 or more, 4 or more, or 5 or more chemotherapeutic agents. Known drugs that are approved to treat soft tissue sarcoma include Cosmegen (Dactinomycin), Doxorubicin Hydrochloride, Eribulin Mesylate, Gleevec (Imatinib Mesylate), Halaven (Eribulin Mesylate), Imatinib Mesylate, Pazopanib Hydrochloride, Tazemetostat Hydrobromide, Tazverik (Tazemetostat Hydrobromide), Trabectedin, Votrient (Pazopanib Hydrochloride) and Yondelis (Trabectedin). In some embodiments, the human patient has been previously treated with one of more chemotherapeutic agents, and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agent, wherein the chemotherapeutic agent is selected from the group consisting of Cosmegen (Dactinomycin), Doxorubicin Hydrochloride, Eribulin Mesylate, Gleevec (Imatinib Mesylate), Halaven (Eribulin Mesylate), Imatinib Mesylate, Pazopanib Hydrochloride, Tazemetostat Hydrobromide, Tazverik (Tazemetostat Hydrobromide), Trabectedin, Votrient (Pazopanib Hydrochloride) and Yondelis (Trabectedin).
[00173] A known combination of drugs that is approved to treat a soft tissue sarcoma is Vincristine Sulfate, Dactinomycin and Cyclophosphamide. In some embodiments, the human patient has previously treated with 3 or more chemotherapeutic agents and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agents, wherein the 3 or more chemotherapeutic agents are Vincristine Sulfate, Dactinomycin and Cyclophosphamide,
[00174] Known drugs that are approved to treat testicular germ cell tumor include Bleomycin Sulfate, Cisplatin, Cosmegen (Dactinomycin), Etopophos (Etoposide Phosphate), Etoposide, Etoposide Phosphate, Ifex (Ifosfamide) and Vinblastine Sulfate. In some embodiments, the human patient has been previously treated with one of more chemotherapeutic agents, and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agent, wherein the chemotherapeutic agent is selected from the group consisting of Bleomycin Sulfate, Cisplatin, Cosmegen (Dactinomycin), Etopophos (Etoposide Phosphate), Etoposide, Etoposide Phosphate, Ifex (Ifosfamide) and Vinblastine Sulfate.
[00175] Known combinations of drugs that are approved to treat testicular germ cell tumor include: (a) Bleomycin, Etoposide Phosphate and Cisplatin; (b) Bleomycin, Etoposide Phosphate and Carboplatin; (c) Vinblastine Sulfate, Ifosfamide and Cisplatin and (d) Etoposide, Ifosfamide and Cisplatin. In some embodiments, the human patient has previously treated with 3 or more chemotherapeutic agents and there is tumor growth, progression, or recurrence during or after treatment with the chemotherapeutic agents, wherein the 3 or more chemotherapeutic agents are selected from the group consisting of (a) Bleomycin, Etoposide Phosphate and Cisplatin; (b) Bleomycin, Etoposide Phosphate and Carboplatin; (c) Vinblastine Sulfate, Ifosfamide and Cisplatin and (d) Etoposide, Ifosfamide and Cisplatin.
[00176] Known treatments for uveal melanoma include surgery, photocoagulation and thermotherapy. The following types of surgery may be used: (a) Resection: Surgery to remove the tumor and a small amount of healthy tissue around it; (b) Enucleation: Surgery to remove the eye and part of the optic nerve. This is done if vision cannot be saved and the tumor is large, has spread to the optic nerve, or causes high pressure inside the eye. After surgery, the patient is usually fitted for an artificial eye to match the size and color of the other eye; and (c) Exenteration: Surgery to remove the eye and eyelid, and muscles, nerves, and fat in the eye socket. After surgery, the patient may be fitted for an artificial eye to match the size and color of the other eye or a facial prosthesis. In some embodiments, the human patient has previously undergone surgery. In some embodiments, the surgery was resection, enucleation or exenteration.
[00177] Photocoagulation is a procedure that uses laser light to destroy blood vessels that bring nutrients to the tumor, causing the tumor cells to die. Photocoagulation may be used to treat small tumors. This is also called light coagulation. In some embodiments, the human patient has previously received photocoagulation therapy. In some embodiments, the human patient has previously been treated with photocoagulation therapy. Thermotherapy is the use of heat from a laser to destroy cancer cells and shrink the tumor. In some embodiments, the human patient has previously received thermo therapy. In some embodiments, the human patient has previously been treated with thermotherapy.
Pharmaceutical compositions
[00178] The anti-TIGIT and anti-PD-1 antibodies can be provided as a pharmaceutical composition. The pharmaceutical composition may be formulated with a pharmaceutically acceptable carrier.
[00179] Formulations are prepared for storage and use by combining a purified antibody with a pharmaceutically acceptable vehicle (e.g., a carrier or excipient). Pharmaceutically acceptable carriers include a sterile aqueous solution. Those of skill in the art generally consider pharmaceutically acceptable carriers, excipients, and/or stabilizers to be inactive ingredients of a formulation or pharmaceutical composition. Suitable pharmaceutically acceptable vehicles include, but are not limited to, nontoxic buffers such as phosphate, citrate, and other organic acids; salts such as sodium chloride; antioxidants including ascorbic acid and methionine; preservatives such as octadecyldimethylbenzyl ammonium chloride, hexamethonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butyl or benzyl alcohol, alkyl parabens, such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol; low molecular weight polypeptides (e.g., less than about 10 amino acid residues); proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; carbohydrates such as monosaccharides, disaccharides, glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes such as Zn-protein complexes; and non-ionic surfactants such as TWEEN or polyethylene glycol (PEG) [5].
[00180] The pharmaceutical compositions of the present invention can be administered in any number of ways for either local or systemic treatment. Administration can be topical by epidermal or transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders; pulmonary by inhalation or insufflation of powders or aerosols, including by nebulizer, intratracheal, and intranasal; oral; or parenteral including intravenous, intraarterial, intratumoral, subcutaneous, intraperitoneal, intramuscular (e.g., injection or infusion), or intracranial (e.g., intrathecal or intraventricular). In certain embodiments, the pharmaceutical compositions comprising the anti-TIGIT and anti-PD-1 antibodies is administered intravenously.
Kits of the invention
[00181] The present invention provides kits that comprise the anti-TIGIT antibodies and anti-PD-1 antibodies as described herein and that can be used to perform the methods described herein. In certain embodiments, a kit comprises at least one purified anti-TIGIT antibody in one or more containers. One skilled in the art will readily recognize that the disclosed anti- TIGIT antibodies and anti-PD-1 antibodies can be readily incorporated into one of the established kit formats which are well known in the art.
Definitions
[00182] The practice of the present invention will employ, unless otherwise indicated, conventional methods of chemistry, biochemistry, molecular biology, immunology and pharmacology, within the skill of the art. Such techniques are explained fully in the literature. See, e.g., references [6-13], etc.
[00183] The term “comprising” encompasses “including” as well as “consisting”, “consisting of’ and/or “consisting essentially of’ e.g. a composition “comprising” X may consist exclusively of X or may include something additional e.g. X + Y. It is also understood that wherever embodiments are described herein with the language “consisting essentially of’ otherwise analogous embodiments described in terms of “consisting of’ are also provided.
[00184] The term “about” or “approximately” in relation to a numerical value x is optional and means, for example, x+10%.
[00185] The word “substantially” does not exclude “completely” e.g. a composition which is “substantially free” from Y may be completely free from Y. Where necessary, the word “substantially” may be omitted from the definition of the invention.
[00186] Unless specifically stated, a process or method comprising numerous steps may comprise additional steps at the beginning or end of the method, or may comprise additional intervening steps. Also, steps may be combined, omitted or performed in an alternative order, if appropriate.
[00187] Various embodiments of the invention are described herein. It will be appreciated that the features specified in each embodiment may be combined with other specified features, to provide further embodiments. In particular, embodiments highlighted herein as being suitable, typical or preferred may be combined with each other (except when they are mutually exclusive). [00188] All patent and literature references cited in the present specification are hereby incorporated by reference in their entirety.
[00189] Any reference to a method for treatment comprising administering an agent to a patient, also covers that agent for use in said method for treatment, as well as the use of the agent in said method for treatment, and the use of the agent in the manufacture of a medicament. [00190] The terms “agonist” and “agonistic” as used herein refer to or describe an agent that is capable of, directly or indirectly, substantially inducing, activating, promoting, increasing, or enhancing the biological activity of a target and/or a pathway. The term “agonist” is used herein to include any agent that partially or fully induces, activates, promotes, increases, or enhances the activity of a protein.
[00191] The terms “antagonist” and “antagonistic” as used herein refer to or describe an agent that is capable of, directly or indirectly, partially or fully blocking, inhibiting, reducing, or neutralizing a biological activity of a target and/or pathway. The term “antagonist” is used herein to include any agent that partially or fully blocks, inhibits, reduces, or neutralizes the activity of a protein.
[00192] The terms “modulation” and “modulate” as used herein refer to a change or an alteration in a biological activity. Modulation includes, but is not limited to, stimulating an activity or inhibiting an activity. Modulation may be an increase or a decrease in activity, a change in binding characteristics, or any other change in the biological, functional, or immunological properties associated with the activity of a protein, a pathway, a system, or other biological targets of interest.
[00193] The term “antibody” as used herein refers to an immunoglobulin molecule that recognizes and specifically binds a target through at least one antigen-binding site. The target may be a protein, polypeptide, peptide, carbohydrate, polynucleotide, lipid, or a combination of any of the foregoing. As used herein, the term encompasses intact polyclonal antibodies, intact monoclonal antibodies, antibody fragments (such as Fab, Fab', F(ab')2, and Fv fragments), single chain Fv (scFv) antibodies, multispecific antibodies, bispecific antibodies, monospecific antibodies, monovalent antibodies, chimeric antibodies, humanized antibodies, human antibodies, fusion proteins comprising an antigen-binding site of an antibody, and any other modified immunoglobulin molecule comprising an antigen-binding site as long as the antibodies exhibit the desired biological activity. An antibody can be any of the five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, or subclasses (isotypes) thereof (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2), based on the identity of their heavy-chain constant domains referred to as alpha, delta, epsilon, gamma, and mu, respectively. The different classes of immunoglobulins have different and well-known subunit structures and three-dimensional configurations. Antibodies can be naked or conjugated to other molecules, including but not limited to, toxins and radioisotopes.
[00194] The term “antibody fragment” refers to a portion of an intact antibody and generally refers to the antigenic determining variable regions of an intact antibody. Examples of antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, and Fv fragments, linear antibodies, single chain antibodies, and multispecific antibodies formed from antibody fragments. “Antibody fragment” as used herein comprises an antigen-binding site or epitopebinding site.
[00195] The term “variable region” of an antibody refers to the variable region of an antibody light chain or the variable region of an antibody heavy chain, either alone or in combination. Generally, the variable region of a heavy chain or a light chain consists of four framework regions connected by three complementarity determining regions (CDRs), also known as “hypervariable regions”. The CDRs in each chain are held together in close proximity by the framework regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site(s) of the antibody. There are at least two techniques for determining CDRs: (1) an approach based on cross-species sequence variability (i.e., Kabat et al., 1991, Sequences of Proteins of Immunological Interest, 5th Edition, National Institutes of Health, Bethesda MD.), and (2) an approach based on crystallographic studies of antigen-antibody complexes (Al Eazikani et al., 1997, J. Mol. Biol., 273:927-948). In addition, combinations of these two approaches are sometimes used in the art to determine CDRs.
[00196] The term “monoclonal antibody” as used herein refers to a homogenous antibody population involved in the highly specific recognition and binding of a single antigenic determinant or epitope. This is in contrast to polyclonal antibodies that typically include a mixture of different antibodies that recognize different antigenic determinants. The term “monoclonal antibody” encompasses both intact and full-length monoclonal antibodies as well as antibody fragments (e.g., Fab, Fab', F(ab')2, Fv), single chain (scFv) antibodies, fusion proteins comprising an antibody fragment, and any other modified immunoglobulin molecule comprising an antigen-binding site. Furthermore, “monoclonal antibody” refers to such antibodies made by any number of techniques, including but not limited to, hybridoma production, phage selection, recombinant expression, and transgenic animals.
[00197] The term “humanized antibody” as used herein refers to antibodies that are specific immunoglobulin chains, chimeric immunoglobulins, or fragments thereof that contain minimal non-human sequences. Typically, humanized antibodies are human immunoglobulins in which amino acid residues of the CDRs are replaced by amino acid residues from the CDRs of a non-human species (e.g., mouse, rat, rabbit, or hamster) that have the desired specificity, affinity, and/or binding capability. In some instances, the framework variable region amino acid residues of a human immunoglobulin may be replaced with the corresponding amino acid residues in an antibody from a non-human species. The humanized antibody can be further modified by the substitution of additional amino acid residues either in the framework variable region and/or within the replaced non-human amino acid residues to further refine and optimize antibody specificity, affinity, and/or binding capability. The humanized antibody may comprise variable domains containing all or substantially all of the CDRs that correspond to the non- human immunoglobulin, whereas all or substantially all of the framework variable regions are those of a human immunoglobulin sequence. In some embodiments, the variable domains comprise the framework regions of a human immunoglobulin sequence. In some embodiments, the variable domains comprise the framework regions of a human immunoglobulin consensus sequence. The humanized antibody can also comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin.
[00198] The term “human antibody” as used herein refers to an antibody produced by a human or an antibody having an amino acid sequence corresponding to an antibody produced by a human made using any of the techniques known in the art.
[00199] The term “chimeric antibody” as used herein refers to an antibody wherein the amino acid sequence of the immunoglobulin molecule is derived from two or more species. Typically, the variable regions of the light and heavy chains correspond to the variable regions of an antibody derived from one species of mammal (e.g., mouse, rat, rabbit, etc.) with the desired specificity, affinity, and/or binding capability, while the constant regions are homologous to the sequence in an antibody derived from another species.
[00200] The terms “epitope” and “antigenic determinant” are used interchangeably herein and refer to that portion of an antigen or target capable of being recognized and specifically bound by a particular antibody. When the antigen or target is a polypeptide, epitopes can be formed both from contiguous amino acids and noncontiguous amino acids juxtaposed by tertiary folding of the protein. Epitopes formed from contiguous amino acids (also referred to as linear epitopes) are typically retained upon protein denaturing, whereas epitopes formed by tertiary folding (also referred to as conformational epitopes) are typically lost upon protein denaturing. An epitope typically includes at least 3, and more usually, at least 5, 6, 7, or 8-10 amino acids in a unique spatial conformation.
[00201] The terms “selectively binds” or “specifically binds” mean that an agent interacts more frequently, more rapidly, with greater duration, with greater affinity, or with some combination of the above to the epitope, protein, or target molecule than with alternative substances, including related and unrelated proteins. In certain embodiments “specifically binds” means, for instance, that an agent binds a protein or target with a KD of about O.lmM or less, but more usually less than about I p M. In certain embodiments, “specifically binds” means that an agent binds a target with a KD of at least about O.lpM or less, at least about O.OlpM or less, or at least about InM or less. Because of the sequence identity between homologous proteins in different species, specific binding can include an agent that recognizes a protein or target in more than one species (e.g., mouse TIGIT and human TIGIT). Likewise, because of homology within certain regions of polypeptide sequences of different proteins, specific binding can include an agent that recognizes more than one protein or target. It is understood that, in certain embodiments, an agent that specifically binds a first target may or may not specifically bind a second target. As such, “specific binding” does not necessarily require (although it can include) exclusive binding, i.e. binding to a single target. Thus, an agent may, in certain embodiments, specifically bind more than one target. In certain embodiments, multiple targets may be bound by the same antigen-binding site on the agent. For example, an antibody may, in certain instances, comprise two identical antigen-binding sites, each of which specifically binds the same epitope on two or more proteins. In certain alternative embodiments, an antibody may be bispecific and comprise at least two antigen-binding sites with differing specificities. Generally, but not necessarily, reference to binding means specific binding.
[00202] As used herein, “selecting” and “selected” in reference to a patient is used to mean that a particular patient is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria, e.g., the patient has a tumor with an elevated expression level of PVR and/or PVRL2. Similarly, “selectively treating a patient having a tumor” refers to providing treatment to a cancer patient that is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria, e.g., the patient has a tumor with an elevated expression level of PVR and/or PVRL2. Similarly, “selectively administering” refers to administering a drug to a cancer patient that is specifically chosen from a larger group of patients on the basis of (due to) the particular patient having a predetermined criteria, e.g., the patient the patient has a tumor with an elevated expression level of PVR and/or PVRL2. By selecting, selectively treating and selectively administering, it is meant that a patient is delivered a personalized therapy for cancer based on the patient's cancer biology, rather than being delivered a standard treatment regimen based solely on the patient having a cancer, such as CRC or NSCLC.
[00203] The terms “polypeptide” and “peptide” and “protein” are used interchangeably herein and refer to polymers of amino acids of any length. The polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non-amino acids. The terms also encompass an amino acid polymer that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids), as well as other modifications known in the art. It is understood that, because the polypeptides of this invention may be based upon antibodies or other members of the immunoglobulin superfamily, in certain embodiments, a “polypeptide” can occur as a single chain or as two or more associated chains.
[00204] The terms “polynucleotide” and “nucleic acid” and “nucleic acid molecule” are used interchangeably herein and refer to polymers of nucleotides of any length, and include DNA and RNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase.
[00205] The terms “identical” or percent “identity” in the context of two or more nucleic acids or polypeptides, refer to two or more sequences or subsequences that are the same or have a specified percentage of nucleotides or amino acid residues that are the same, when compared and aligned (introducing gaps, if necessary) for maximum correspondence, not considering any conservative amino acid substitutions as part of the sequence identity. The percent identity may be measured using sequence comparison software or algorithms or by visual inspection. Various algorithms and software that may be used to obtain alignments of amino acid or nucleotide sequences are well-known in the art. These include, but are not limited to, BLAST, ALIGN, Megalign, BestFit, GCG Wisconsin Package, and variants thereof. In some embodiments, two nucleic acids or polypeptides of the invention are substantially identical, meaning they have at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, and in some embodiments at least 95%, 96%, 97%, 98%, 99% nucleotide or amino acid residue identity, when compared and aligned for maximum correspondence, as measured using a sequence comparison algorithm or by visual inspection. In some embodiments, identity exists over a region of the sequences that is at least about 10, at least about 20, at least about 40-60 nucleotides or amino acid residues, at least about 60-80 nucleotides or amino acid residues in length or any integral value there between. In some embodiments, identity exists over a longer region than 60-80 nucleotides or amino acid residues, such as at least about 80-100 nucleotides or amino acid residues, and in some embodiments the sequences are substantially identical over the full length of the sequences being compared, for example, the coding region of a nucleotide sequence. [00206] A “conservative amino acid substitution” is one in which one amino acid residue is replaced with another amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been generally defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). For example, substitution of a phenylalanine for a tyrosine is considered to be a conservative substitution. Generally, conservative substitutions in the sequences of polypeptides and/or antibodies of the invention do not abrogate the binding of the polypeptide or antibody containing the amino acid sequence, to the target binding site. Methods of identifying nucleotide and amino acid conservative substitutions which do not eliminate binding are well- known in the art.
[00207] The term “vector” as used herein means a construct, which is capable of delivering, and usually expressing, one or more gene(s) or sequence(s) of interest in a host cell. Examples of vectors include, but are not limited to, viral vectors, naked DNA or RNA expression vectors, plasmid, cosmid, or phage vectors, DNA or RNA expression vectors associated with cationic condensing agents, and DNA or RNA expression vectors encapsulated in liposomes.
[00208] A polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is “isolated” is a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is in a form not found in nature. Isolated polypeptides, soluble proteins, antibodies, polynucleotides, vectors, cells, or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature. In some embodiments, a polypeptide, soluble protein, antibody, polynucleotide, vector, cell, or composition which is isolated is substantially pure.
[00209] The term “substantially pure” as used herein refers to material which is at least 50% pure (i.e., free from contaminants), at least 90% pure, at least 95% pure, at least 98% pure, or at least 99% pure.
[00210] The term “immune response” as used herein includes responses from both the innate immune system and the adaptive immune system. It includes both cell-mediated and/or humoral immune responses. It includes, but is not limited to, both T-cell and B-cell responses, as well as responses from other cells of the immune system such as natural killer (NK) cells, monocytes, macrophages, etc. [00211] The terms “cancer” and “cancerous” as used herein refer to or describe the physiological condition in mammals in which a population of cells are characterized by unregulated cell growth. Examples of cancer include, but are not limited to, carcinoma, blastoma, sarcoma, and hematologic cancers such as lymphoma and leukemia.
[00212] The terms “tumor” and “neoplasm” as used herein refer to any mass of tissue that results from excessive cell growth or proliferation, either benign (non-cancerous) or malignant (cancerous) including pre-cancerous lesions.
[00213] The term “metastasis” as used herein refers to the process by which a cancer spreads or transfers from the site of origin to other regions of the body with the development of a similar cancerous lesion at a new location. Generally, a “metastatic” or “metastasizing” cell is one that loses adhesive contacts with neighboring cells and migrates via the bloodstream or lymph from the primary site of disease to secondary sites throughout the body.
[00214] The terms “cancer cell” and “tumor cell” refer to the total population of cells derived from a cancer or tumor or pre-cancerous lesion, including both non-tumorigenic cells, which comprise the bulk of the cancer cell population, and tumorigenic stem cells (cancer stem cells). As used herein, the terms “cancer cell” or “tumor cell” will be modified by the term “non-tumorigenic” when referring solely to those cells lacking the capacity to renew and differentiate to distinguish those tumor cells from cancer stem cells.
[00215] The term “subject” refers to any animal (e.g., a mammal), including, but not limited to, humans, non-human primates, canines, felines, rabbits, rodents, and the like, which is to be the recipient of a particular treatment. Typically, the terms “subject” and “patient” are used interchangeably herein in reference to a human subject.
[00216] The term “pharmaceutically acceptable” refers to a substance approved or approvable by a regulatory agency of the Federal government or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
[00217] The terms “pharmaceutically acceptable excipient, carrier, or adjuvant” or “acceptable pharmaceutical carrier” refer to an excipient, carrier, or adjuvant that can be administered to a patient, together with at least one agent of the present disclosure, and which does not destroy the pharmacological activity thereof and is non-toxic when administered in doses sufficient to deliver a therapeutic effect. In general, those of skill in the art and the U.S. FDA consider a pharmaceutically acceptable excipient, carrier, or adjuvant to be an inactive ingredient of any formulation.
[00218] The terms “effective amount” or “therapeutically effective amount” or “therapeutic effect” refer to an amount of an agent, an antibody, a polypeptide, a polynucleotide, a small organic molecule, or other drug effective to “treat” a disease or disorder in a subject such as, a mammal. In the case of cancer or a tumor, the therapeutically effective amount of an antibody has a therapeutic effect and as such can enhance or boost the immune response, enhance or boost the anti-tumor response, increase cytolytic activity of immune cells, increase killing of tumor cells, increase killing of tumor cells by immune cells, reduce the number of tumor cells; decrease tumorigenicity, tumorigenic frequency, or tumorigenic capacity; reduce the number or frequency of cancer stem cells; reduce the tumor size; reduce the cancer cell population; inhibit or stop cancer cell infiltration into peripheral organs including, for example, the spread of cancer into soft tissue and bone; inhibit and stop tumor or cancer cell metastasis; inhibit and stop tumor or cancer cell growth; relieve to some extent one or more of the symptoms associated with the cancer; reduce morbidity and mortality; improve quality of life; or a combination of such effects.
[00219] The terms “treating” or “treatment” or “to treat” or “alleviating” or “to alleviate” refers to therapeutic measures that cure, slow down, lessen symptoms of, and/or halt progression of a diagnosed pathologic condition or disorder. In the case of cancer or a tumor, a patient is successfully “treated” according to the methods of the present invention if the patient shows one or more of the following: an increased immune response, an increased antitumor response, increased cytolytic activity of immune cells, increased killing of tumor cells, increased killing of tumor cells by immune cells, a reduction in the number of or complete absence of cancer cells; a reduction in the tumor size; inhibition of or an absence of cancer cell infiltration into peripheral organs including the spread of cancer cells into soft tissue and bone; inhibition of or an absence of tumor or cancer cell metastasis; inhibition or an absence of cancer growth; relief of one or more symptoms associated with the specific cancer; reduced morbidity and mortality; improvement in quality of life; reduction in tumorigenicity; reduction in the number or frequency of cancer stem cells; or some combination of effects.
[00220] As used in the present disclosure and claims, the singular forms “a”, “an” and “the” include plural forms unless the context clearly dictates otherwise.
[00221] The term “and/or” as used in a phrase such as “A and/or B” herein is intended to include both A and B; A or B; A (alone); and B (alone). Likewise, the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
[00222] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to person skilled in the art and are to be included within the spirit and purview of this application.
[00223] All publications, patents, patent applications, internet sites, and accession numbers/database sequences including both polynucleotide and polypeptide sequences cited herein are hereby incorporated by reference herein in their entirety for all purposes to the same extent as if each individual publication, patent, patent application, internet site, or accession number/database sequence were specifically and individually indicated to be so incorporated by reference.
MODES FOR CARRYING OUT THE INVENTION
Example 1 - Binding properties of etigilimab
[00224] Flow cytometry analysis was conducted to assess the ability of the anti-TIGIT antibodies 313M32 (etigilimab), 313M26 and 313R19 to bind to various forms of TIGIT from different species. The species tested were rat, guinea pig, rabbit, marmoset, pig, dog, rhesus monkey and human. The antibody 313M26 is the parental murine anti-hTIGIT antibody of 313M32 and therefore 313M26 and 313M32 share both comprise the heavy chain and light chain CDRs of SEQ ID Nos: 1-6. 313R19 is an anti-TIGIT antibody that does not share the same heavy and light chain CDRs.
[00225] This analysis utilized human embryonic kidney 293 cells (HEK-293, American Type Culture Collection, Manassas, VA) that were transiently transfected with cDNA expression vectors encoding a membrane-anchored derivative of TIGIT from the species listed above. These cDNA expression vectors encoded the extracellular domains of TIGIT from different species each fused to the transmembrane domain of human CD4 and an intracellular domain comprised of green fluorescent protein. Cells were then exposed to 313M26, 313M26 or 313R19 and allophycocyanin (APC)- labeled anti-human Fc secondary antibody and then examined by flow cytometry.
[00226] The results of these experiments are summarized in the table below:
Figure imgf000058_0001
Figure imgf000059_0001
[00227] FACS analysis was conducted to assess the ability of anti-TIGIT antibodies 313M32, 313M26 and 313R19 to block PVR binding to human TIGIT.
[00228] A cell surface human TIGIT protein was generated by ligating amino acids 22- 141 of human TIGIT to the transmembrane domain of CD4 and a C-terminal GFP protein tag using standard recombinant DNA techniques (hTIGIT-CD4TM-GFP). PVR-Fc constructs were generated using standard recombinant DNA techniques. Specifically, the extracellular domain of human PVR was ligated in-frame to a rabbit Fc region and the recombinant hPVR- rbFc protein was expressed in CHO cells. The fusion proteins were purified from cell culture medium using protein A chromatography.
[00229] HEK-293T cells were transiently transfected with the hTIGIT-CD4TM-GFP construct. After 16 hours, transfected cells were suspended in ice cold HBSS containing 2% FBS and heparin and incubated on ice with 0.5pg/ml hPVR-rbFc fusion protein in the presence of anti-TIGIT antibodies 313R19, 313M26, or 313M32 for 60 minutes. The antibodies were tested at concentrations of 10, 2, and 0.4ug/ml. Cells were incubated without antibody or without hPVR-rbFc as controls. A second incubation with 100, u I PE-conjugated anti-rabbit Fc secondary antibody was performed to detect cells bound by the hPVR-rbFc fusion protein. The cells were analyzed on a FACSCanto instrument (BD Biosciences) and the data was processed using Flow Jo software.
[00230] The results of these experiments are summarised in Figure 1. In the absence of any anti-TIGIT antibody, hPVR-rbFc bound strongly to hTIGIT expressed on the surface of the HEK-293T cells. At the lower concentrations of 2pg/ml and 0.4pg/ml antibody, the 313R19 antibody did not block binding of hPVR-rbFc to hTIGIT. In contrast, 313M32 strongly blocked binding of hPVR-rbFc to hTIGIT at 2pg/ml and the level of blockade was higher than the parental mouse antibody 313M26.
Example 2 - In vivo tumor growth inhibition by etigilimab
[00231] A humanized mouse model was used to study the efficacy of treatment with an anti-TIGIT antibody on a human tumor. The humanized mice were obtained from Jackson Laboratories. These mice are created by injecting human hematopoietic stem cells (CD34+ cells) into irradiated NSG mice. After 15 weeks, the presence of mature human lymphocytes is confirmed by flow cytometry. Each mouse was injected subcutaneously with patient-derived melanoma tumor cells (0MP-M9, 75,000 cells/mouse). Tumors were allowed to grow 19 days until they had reached an average volume of approximately 50mm3. Tumor-bearing mice were randomized into groups (n = 8 mice per group). Tumor-bearing mice were treated with either a control antibody, anti-TIGIT antibody 313R19, or anti-TIGIT antibody 313M32. Mice were dosed every 5 days at Img/kg or 5mg/kg. Tumor growth was monitored and tumor volumes were measured with electronic calipers at the indicated time points.
[00232] Figure 2 demonstrates that etigilimab was able to significantly reduce tumor growth at a concentration of Img/kg compared to the control, whereas the anti-TIGIT antibody 313R19 was not able to significantly reduce tumor growth at the same concentration. In addition, these results demonstrated that humanized mouse models bearing patient-derived xenografts can be used to study the anti-hTIGIT antibody OMP-313M32 (which only binds human TIGIT) in parallel with pre -clinical studies carried out with the anti-TIGIT antibodies 313R12 and 313R19 and murine tumor models.
Example 3 - A phase lb/2 clinical trial evaluating the safety and efficacy of anti-TIGIT antibodies in combination with anti-PD-1 antibodies in the treatment of cancer
[00233] A phase lb/2 open-label basket study of etigilimab in combination with an anti- PD-1 antibody (nivolumab) was conducted in patients with locally advanced or metastatic solid tumors. Patient populations selected for evaluation in this study all had advanced or refractory disease. These patients therefore had a high unmet therapeutic need for which limited therapeutic options exist.
[00234] The types of cancer included in the clinical trial were endometrial cancer, head and neck cancer, cervical cancer, ovarian cancer, rare tumours, including soft tissue sarcomas, testicular germ cell tumours and uveal melanomas, high tumor mutational burden (TMB-H) and microsatellite stable (MSS) cancers. Twenty two patients were included in the trial which were broken down into the following cohorts:
• Cohort B - head and neck (N=l)
• Cohort C - Cervical cancer (N=l)
• Cohort E - TMB-H/MSS cancer (N=4)
• Cohort F - rare tumours (N=12 - N=6 uveal melanoma, N=5 soft tissue sarcomas, N=1 testicular germ cell tumours)
• Cohort G - Ovarian cancer (N=4)
[00235] The demographics of the patients tested are summarised in the table below. As shown in the data below, a number of the patients in the clinical trial had been heavily pretreated (greater than 3 previous therapies prior to treatment with etigilimab and an anti-PD-1 antibody).
Figure imgf000061_0001
[00236] The path mts were naive to checkpoint inhibitor therapy. The patients were intravenously administered etigilimab every two weeks at a dose of 1000 mg and intravenously administered nivolumab every two weeks at a dose of 240 mg.
[00237] 18 adverse events occurred in 10 of the patients and are summarised in the table below. The most common treatment-related adverse events were due to skin reactions, but none of them required treatment with systemic steroids. As shown in the table below, the adverse events were mostly low grade (Grades 1-2). These data demonstrate that the combination of etigilimab in combination with an anti-PD-1 antibody (nivolumab) is well tolerated in human patients.
Figure imgf000061_0002
Figure imgf000062_0002
[00238] The preliminary efficacy of the combination of etigilimab with an anti-PD-1 antibody (nivolumab) was assessed in 23 patients who had the following cancers after a minimum of one scan. The results are shown in the table below:
• Cohort C - Cervical cancer (N=3)
• Cohort E - TMB-H/MSS cancer (N=4)
• Cohort F - rare tumours (N=6 uveal melanomas, N=1 soft tissue sarcomas, N=3 testicular germ cell tumours)
• Cohort H - Ovarian cancer (N=6)
Evaluable Complete Partial Stable Disease
C h rt
Figure imgf000062_0001
Figure imgf000063_0002
[00239] The objective response rate (which is a combination of complete response rates and partial response rates) was 13.0%. The disease control rate (which is a combination of complete response rates, partial response rates and stable diseases) was 34.7%.
[00240] Historical overall response rates for treating ovarian cancer ([14] and [15]), cervical cancer ([16]) and uveal cancer ([17] and [18] are provided in Figure 3. An overall response rate of greater than 10% is therefore a promising result when compared to these historical data. These preliminary efficacy data support the use of the combination of etigilimab and an anti-PD-1 antibody to treat various cancers including cervical cancer, TMB-H/MSS cancers, ovarian cancers and rare cancers, such as uveal melanomas.
[00241] A summary of the cancers that resulted in a clinical benefit after treatment with the combination of etigilimab with an anti-PD-1 antibody is provided below:
Figure imgf000063_0001
[00242] A complete response was observed in the cervical cancer patient after two scans following treatment with the combination of etigilimab and an anti-PD-1 antibody (nivolumab). The lesions that the patient had at the beginning of the trial and their response to treatment with the combination of etigilimab and an anti-PD-1 antibody after the first and the second scan are summarised in the Table below. Figures 4a and 4b show the reduction in tumour size in a cervical cancer patient that occurred after treatment. These figures show that a complete response occurred as the tumour was no longer visible after the second scan.
Figure imgf000064_0001
[00243] Figures 17a and 17b show the reduction in tumour size in a cervical cancer patient that occurred after treatment. These figures show that a partial response occurred as the tumour was no longer visible after the second scan.
Figure imgf000064_0002
[00244] A partial response was observed in one of the ovarian cancer patients after one scan following treatment with the combination of etigilimab and an anti-PD-1 antibody (nivolumab). The lesions that the patient had at the beginning of the trial and their response to treatment with the combination of etigilimab and an anti-PD- 1 antibody are summarised in the Table below.
Figure imgf000064_0003
[00245] These data support the ability of the combination of etigilimab and nivolumab to treat various cancers, including cervical cancer, TMB-H/MSS cancers, ovarian cancers and rare cancers, such as uveal melanomas, as the combination was able to reduce tumour volume and prevent cancer progression.
[00246] Two of the patients with uveal melanomas had a stable disease after treatment with etigilimab and nivolumab. The remaining four patients experienced disease progression.
[00247] These data support the use of the combination of etigilimab and nivolumab to treat rare cancers, such as soft tissue sarcomas, testicular germ cell tumors and uveal melanomas, as the combination is able to prevent disease progression in patients.
Example 4 - Pharmacodynamic biomarkers
[00248] Pharmacodynamic biomarkers in the blood were analysed following etigilimab treatment in combination with nivolumab. Figure 12 demonstrates that the combination treatment decreased the number of Treg cells in circulation but had no effect on the number of CD8+ T cells. Figures 13A and 13B show that the combination treatment increased markers of cell proliferation in T cells. Figure 14 demonstrates that the combination treatment increased IFNy production in CD4+ effector memory cells. Figure 15 shows that the combination treatment increased the level of proliferating (Ki-67+) CD8+ PD-1+ cells. Figure 16 shows that the combination treatment increased markers of cell proliferation in natural killer cells. Figure 17 shows that the combination treatment reduced the number of exhausted T-cells.
[00249] The data presented in Figures 12-17 concur with the effects of etigilimab treatment observed in the Phase la/b clinical trial. These data confirm that the combination of etigilimab and nivolumab is robustly engaging with biological targets and activating downstream effects in human patients.
Example 5 - Updated clinical data from the phase lb/2 clinical trial evaluating the safety and efficacy of anti-TIGIT antibodies in combination with anti-PD-1 antibodies in the treatment of cancer
[00250] A phase lb/2 open-label basket study was conducted as described in Example 3. 50 patients were included in the trial and their baseline characteristics are summarised in the table below.
Figure imgf000065_0001
Figure imgf000066_0001
[00251] The patients were naive to checkpoint inhibitor therapy. The patients were intravenously administered etigilimab every two weeks at a dose of 1000 mg and intravenously administered nivolumab every two weeks at a dose of 240 mg.
[00252] The preliminary efficacy of the combination of etigilimab with an anti-PD-1 antibody (nivolumab) was assessed in 38 patients who had the following cancers after a minimum of one scan.
• Cohort A - Endometrial cancer CPI- naive (N=l)
• Cohort C - Cervical cancer (N=5)
• Cohort E - TMB-H/MSS tumor (N=6)
• Cohort F - rare tumours (N=6 uveal melanomas, N=7 soft tissue sarcomas, N=4 testicular germ cell tumours)
• Cohort H - Ovarian cancer (N=9)
[00253] The preliminary efficacy data are shown in the Figures 19-21. The objective response rate (which is a combination of complete response rates and partial response rates) was 15.8%. The disease control rate (which is a combination of complete response rates, partial response rates and stable diseases) was 47.4%. The median duration of stable disease was 3.7 months (0.0- 7.7 months). The median progression-free survival was 3.0 months (1.7-4.6 months). [00254] These data further support the ability of the combination of etigilimab and nivolumab to treat various cancers, including rare cancers, such as such as soft tissue sarcomas, testicular germ cell tumors and uveal melanomas.
[00255] Sequences
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
REFERENCES
[1] WO/2016/191643
[2] WO 2016/106302
[3] WO 2016/028656
[4] US 2013/0251720
[5] Remington: The Science and Practice of Pharmacy, 22st Edition, 2012, Pharmaceutical Press, London
[6] Gennaro (2000) Remington: The Science and Practice of Pharmacy. 20th edition, ISBN: 0683306472.
[7] Molecular Biology Techniques: An Intensive Laboratory Course, (Ream et al., eds., 1998, Academic Press).
[8] Methods In Enzymology (S. Colowick and N. Kaplan, eds., Academic Press, Inc.)
[9] Handbook of Experimental Immunology, Vols. I- IV (D.M. Weir and C.C. Blackwell, eds, 1986, Blackwell Scientific Publications)
[10] Sambrook et al. (2001) Molecular Cloning: A Laboratory Manual, 3rd edition (Cold Spring Harbor Laboratory Press).
[11] Handbook of Surface and Colloidal Chemistry (Birdi, K.S. ed., CRC Press, 1997)
[12] Ausubel et al. (eds) (2002) Short protocols in molecular biology, 5th edition (Current Protocols).
[13] PCR (Introduction to Biotechniques Series), 2nd ed. (Newton & Graham eds., 1997, Springer Verlag)
[14] Hamanishi et al. (2021) Journal of Clinical Oncology, 39, 33, 3671-3681
[15] Matulonis et al. (2019) annals of Oncology 30: 1080-1087
[16] Chung, et al. (2019) Journal of Clinical Oncology 37:17, 1470-1478 [17] Pelster et al. (2020) Journal of Clinical Oncology; 39(6):599-607
[18] JM Piulats et al. (2021) Journal of Clinical Oncology; 39(6):586-598

Claims

Claims
1. A method of treating cancer in a human patient, comprising administering to the human patient a therapeutically effective amount of an anti-TIGIT antibody and an anti-PD-1 antibody, wherein: the anti-TIGIT antibody comprises a heavy chain CDR1 comprising TSDYAWN (SEQ ID NO:1), a heavy chain CDR2 comprising YISYSGSTSYNPSLRS (SEQ ID NO:2), a heavy chain CDR3 comprising ARRQVGLGFAY (SEQ ID NOG), a light chain CDR1 comprising KASQDVSTAVA (SEQ ID NO:4), a light chain CDR2 comprising SASYRYT (SEQ ID NOG), and a light chain CDR3 comprising QQHYSTP (SEQ ID NOG), and wherein the cancer is selected from the group consisting of a soft tissue sarcoma, a testicular germ cell tumour and uveal melanoma.
2. The method of claim 1, wherein the anti-TIGIT antibody comprises a heavy chain variable region having at least 90% sequence identity to SEQ ID NO:7 and a light chain variable region having at least 90% sequence identity to SEQ ID NO:8.
3. The method of claim 1 or claim 2, wherein the anti-TIGIT antibody comprises a heavy chain variable region comprising SEQ ID NO:7 and a light chain variable region comprising SEQ ID NOG.
4. The method of any one of claims 1-3, wherein the anti-TIGIT antibody is a monoclonal antibody, a humanized antibody, a human antibody, a chimeric antibody, a bispecific antibody, an IgGl antibody, an IgG2 antibody, an IgG4 antibody, or an antibody fragment comprising an antigen binding site
5. The method of any one of claims 1-4, wherein the anti-TIGIT antibody comprises a heavy chain having at least 90% sequence identity to SEQ ID NO:9 and a light chain having at least 90% sequence identity to SEQ ID NO: 10.
6. The method of any one of claims 1-5, wherein the anti-TIGIT antibody comprises a heavy chain comprising SEQ ID NO:9 and a light chain comprising SEQ ID NO: 10.
7. The method of any one of claims 1-6, wherein the anti-TIGIT antibody is etigilimab. The method according to any one of claims 1-7, wherein the anti-PD-1 antibody is selected from the group consisting of pembrolizumab (Keytruda), nivolumab (Opdivo), cemiplimab (Libtayo), dostarlimab (Jemperli), Tislelizumab, Sintilimab, Zimberelimab, Toripalimab, Penpulimab, Balstilimab, Retifanlimab, Cetrelimab, Budigalimab, Pimivalimab, Spartalizumab, Serplulimab, Sasanlimab, Camrelizumab, Prolgolimab, Pucotenlimab, Ezabenlimab and Genolimzumab. The method according to any one of claims 1-8, wherein the anti-TIGIT antibody is administered prior to, concurrently with and/or subsequently to the administration of the anti-PD-1 antibody. The method according to any one of claims 1-9, wherein the anti-TIGIT antibody and the anti-PD-1 antibody are administered intravenously. The method according to any one of claims 1-10, wherein the anti-TIGIT antibody is administered once every week, once every two weeks, once every three weeks, or once every four weeks. The method according to claim 11, wherein the anti-TIGIT antibody is administered once every two weeks. The method according to any one of claims 1-12, wherein the anti-PD-1 antibody is administered once every two weeks, once every three weeks, once every four weeks, once every five weeks or once every six weeks. The method according to claim 13, wherein the anti-PD-1 antibody is administered once every two weeks, once every three weeks, once every four weeks or once every six weeks. The method according to any one of claims 1-14, wherein the anti-TIGIT antibody is administered at a flat dose of 1000 mg. The method according to any one of claims 1-15, wherein the anti-TIGIT antibody is administered at a flat dose of 1000 mg once every two weeks. The method according to any one of claims 1-16, wherein the anti-PD-1 antibody is administered at a flat dose of 200 mg- 1000 mg, optionally wherein the anti-PD-1 antibody is administered at a flat dose of 200 mg, 240 mg, 350 mg, 400mg, 500 mg or 1000 mg. The method according to any one of claims 1-17, wherein the human patient has not received prior treatment with an anti-cancer agent. . The method according to claim 18, wherein the anti-cancer agent is a checkpoint inhibitor, a chemotherapeutic agent or a cell therapy. The method according to any one of claims 1-17, wherein the human patient has been previously treated with an anti-cancer agent and there is tumour growth, progression, or recurrence during or after treatment with the anti-cancer agent. The method according to claim 20, wherein the anti-cancer agent is a checkpoint inhibitor, a chemotherapeutic agent or a cell therapy. The method according to claim 21, wherein the checkpoint inhibitor is an anti-PD-1 antibody, an anti-PD-Ll antibody or a bispecific antibody that comprises a first arm that binds to PD-1 or PD-L1. The method according to any one of claims 1-22, wherein the cancer is soft tissue sarcoma; optionally wherein the soft tissue sarcoma is selected from the group consisting of leiomyosarcoma, inflammatory leiomyosarcoma, liposarcoma, dedifferentiated liposarcoma, rhabdomyosarcomas, synovial sarcomas, angiosarcoma, Kaposi’s sarcoma, epithelioid, malignant peripheral nerve sheath tumours (MPNST) and undifferentiated (pleomorphic sarcoma). The method of claim 23, wherein the soft tissue sarcoma is a dedifferentiated liposarcoma or an undifferentiated (pleomorphic sarcoma). The method according to any one of claims 1-22, wherein the cancer is testicular germ cell tumour; optionally wherein the testicular germ cell tumor is a seminomas testicular germ cell tumour or a non-seminomas testicular germ cell tumor. The method according to any one of claims 1-22, wherein the cancer is uveal melanoma; optionally wherein the uveal melanoma is choroidal melanoma, ciliary body melanoma and iris melanoma. The method according to any one of claims 1-26, wherein human patient is over 50 years old.
PCT/US2022/080568 2022-02-04 2022-11-29 Methods for treating cancer WO2023149977A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263306837P 2022-02-04 2022-02-04
US63/306,837 2022-02-04
US202263345611P 2022-05-25 2022-05-25
US63/345,611 2022-05-25

Publications (1)

Publication Number Publication Date
WO2023149977A1 true WO2023149977A1 (en) 2023-08-10

Family

ID=87552745

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/080568 WO2023149977A1 (en) 2022-02-04 2022-11-29 Methods for treating cancer

Country Status (1)

Country Link
WO (1) WO2023149977A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100316637A1 (en) * 2006-09-29 2010-12-16 Oncomed Pharmaceuticals, Inc. Compositions and Methods for Diagnosing and Treating Cancer
WO2016191643A2 (en) * 2015-05-28 2016-12-01 Oncomed Pharmaceuticals, Inc. Tigit-binding agents and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100316637A1 (en) * 2006-09-29 2010-12-16 Oncomed Pharmaceuticals, Inc. Compositions and Methods for Diagnosing and Treating Cancer
WO2016191643A2 (en) * 2015-05-28 2016-12-01 Oncomed Pharmaceuticals, Inc. Tigit-binding agents and uses thereof

Similar Documents

Publication Publication Date Title
JP7203446B2 (en) TIGIT binding agents and methods of use thereof
US11136384B2 (en) Methods for treatment of cancer comprising TIGIT-binding agents
US11046760B2 (en) Combination therapy for treatment of disease
US20180346571A1 (en) Pd-l1-binding agents and uses thereof
WO2018017864A2 (en) Pvrig-binding agents and uses thereof
US20190048094A1 (en) Redirecting immune responses
US20170044268A1 (en) Immunotherapy with Binding Agents
WO2017040660A1 (en) Combination therapy for treatment of disease
WO2023149977A1 (en) Methods for treating cancer
US20170267758A1 (en) Immunotherapy with binding agents
WO2023149978A1 (en) Cancer biomarkers and cancer treatments
OA18684A (en) Tigit-Binding Agents and Uses Thereof
EA039540B1 (en) Tigit-binding agents and uses thereof

Legal Events

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

Ref document number: 22925216

Country of ref document: EP

Kind code of ref document: A1