SECRETABLE VARIANT IMMUNOMODULATORY PROTEINS AND ENGINEERED CELL THERAPY

Abstract
Provided are immunomodulatory proteins, nucleic acids encoding such immunomodulatory proteins, cells engineered to express the immunomodulatory proteins and infections agents containing nucleic acid encoding the immunomodulatory proteins. In some embodiments, the immunomodulatory proteins are secretable. In some embodiments, the immunomodulatory proteins are transmembrane proteins that are surface expressed. The immunomodulatory proteins, engineered cells and infectious agents provide therapeutic utility for a variety of immunological and oncological conditions. Compositions and methods for making and using such proteins are provided.
Description
INCORPORATION BY REFERENCE OF SEQUENCE LISTING

The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled 761612001440SeqList.TXT, created Oct. 19, 2017 which is 3,524,332 bytes in size. The information in the electronic format of the Sequence Listing is incorporated by reference in its entirety.


FIELD

The present disclosure provides immunomodulatory proteins, nucleic acids encoding such immunomodulatory proteins, cells engineered to express the immunomodulatory proteins and infections agents containing nucleic acid encoding the immunomodulatory proteins. In some embodiments, the immunomodulatory proteins are secretable. In some embodiments, the immunomodulatory proteins are transmembrane proteins that are surface expressed. The immunomodulatory proteins, engineered cells and infectious agents provide therapeutic utility for a variety of immunological and oncological conditions. Compositions and methods for making and using such proteins are provided.


BACKGROUND

Modulation of the immune response by intervening in the processes that occur in the immunological synapse (IS) formed by and between antigen-presenting cells (APCs) or target cells and lymphocytes is of increasing medical interest. Currently, biologics used to enhance or suppress immune responses have generally been limited to immunoglobulins (e.g., anti-PD-1 mAbs) or soluble receptors (e.g., Fc-CTLA4). Soluble receptors, in some cases, suffer from a number of deficiencies. While useful for antagonizing interactions between proteins, soluble receptors often lack the ability to agonize such interactions. Antibodies have proven less limited in this regard and examples of both agonistic and antagonistic antibodies are known in the art. Nevertheless, both soluble receptors and antibodies lack important attributes that are critical to function in the IS. Mechanistically, cell surface proteins in the IS can involve the coordinated and often simultaneous interaction of multiple protein targets with a single protein to which they bind. IS interactions occur in close association with the junction of two cells, and a single protein in this structure can interact with both a protein on the same cell (cis) as well as a protein on the associated cell (trans), likely at the same time. Although some agents are known that can modulate the IS, improved therapeutics are needed. Provided are embodiments that meet such needs.


SUMMARY

In one aspect, there is provided an immunomodulatory protein comprising at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; the immunomodulatory protein does not comprise a transmembrane domain; and the immunomodulatory protein is not conjugated to a half-life extending moiety. In some embodiments, the half-life extending moiety is a multimerization domain. In some embodiments, the half-life extending moiety is an Fc domain.


In some embodiments, the immunomodulatory protein further comprises a signal peptide. In some embodiments, the signal peptide is a native signal peptide from the corresponding wild-type IgSF member. In some embodiments, the signal peptide is a non-native signal peptide. In some embodiments, the signal peptide is a signal peptide from an immunoglobulin antibody molecule (e.g. an IgG-kappa signal peptide), an IL-2 signal peptide, or a CD33 signal peptide or other signal peptide known or described. Exemplary signal peptides are set forth in any of SEQ ID NOS: 413-430.


In some embodiments of the immunomodulatory protein, the at least one cell surface cognate binding partner is expressed on a mammalian cell. In some embodiments, the mammalian cell is an antigen presenting cell (APC), a tumor cell, or a lymphocyte. In some embodiments, the mammalian cell is a T-cell. In some embodiments, the mammalian cell is a mouse, rat, cynomolgus monkey, or human cell.


In some embodiments of the immunomodulatory protein, at least one affinity modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the wild-type IgSF domain.


In some embodiments of the immunomodulatory protein, specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain modulates immunological activity of the mammalian cell compared to the wild-type IgSF domain. In some embodiments, specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain increases immunological activity of the mammalian cell compared to the wild-type IgSF domain. In some embodiments, specific binding of the immunomodulatory protein attenuates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


In some embodiments of the immunomodulatory protein, the wild-type IgSF domain is from an IgSF family member of a family selected from the group consisting of Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family, and Killer-cell immunoglobulin-like receptors (KIR) family. In some embodiments, the wild-type IgSF domain is from an IgSF member selected from the group consisting of CD80, CD86, PD-L1, PD-L2, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAGS, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R, NKp30, VISTA, VSIG3, and VSIG8. In some embodiments, the wild-type IgSF domain is a human IgSF domain. In some embodiments, at least one affinity modified IgSF domain has at least 90% sequence identity to a wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27 and 408. In some embodiments, the immunomodulatory protein has at least 90% sequence identity to the amino acid sequence selected from any of SEQ ID NOS:28-54 and 410 or to a specific binding fragment thereof containing an IgSF domain. In some embodiments, the wild-type IgSF domain is a member of the B7 family. In some embodiments, the wild-type IgSF domain is a domain of CD80, CD86 or ICOSL.


In some embodiments of the immunomodulatory protein, the at least one cell surface cognate binding partner is a stimulatory receptor expressed on a T-cell, and the at least one affinity-modified IgSF domain has increased binding affinity to the stimulatory receptor compared to the binding affinity of the wild-type IgSF domain to the stimulatory receptor. In some embodiments, binding of the affinity-modified IgSF domain to the stimulatory receptor, such as when secreted from a cell, increases immunological activity of the T-cell. In some embodiments, binding of the affinity-modified IgSF domain to the stimulatory receptor, such as when secreted from a cell, decreases immunological activity of the T-cell. In some embodiments, the stimulatory receptor is CD28, ICOS, or CD226. In some embodiments, the at least one affinity-modified IgSF domain is an affinity-modified ICOSL IgSF domain that has increased binding affinity to at least one of: ICOS and CD28. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is ICOS. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is CD28. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified CD80 IgSF domain and the stimulatory receptor is CD28. In some embodiments, the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4 or exhibits decreased binding affinity to CTLA-4 compared to the binding affinity of wild-type IgSF domain to CTLA-4.


In some embodiments of the immunomodulatory protein, the at least one cell surface cognate binding partner is an inhibitory receptor expressed on a T-cell, and the at least one affinity-modified IgSF domain has increased binding affinity to the inhibitor receptor compared to the binding affinity of the wild-type IgSF domain to the inhibitor receptor. In some embodiments, binding of the affinity-modified IgSF domain to the inhibitory receptor, such as when secreted from a cell, increases immunological activity of the T-cell. In some embodiments, binding of the affinity-modified IgSF domain to the inhibitor receptor, such as when secreted from a cell, decreases immunological activity of the T-cell. In some embodiments of the immunomodulatory protein, the inhibitory receptor comprises an ITIM signaling domain. In some embodiments, the inhibitory receptor is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA VSIG3 or VSIG8 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3 or VSIG8, respectively. In some embodiments, the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA, respectively. In some embodiments, the inhibitory receptor is PD-1 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of PD-L1 or an affinity-modified IgSF of PD-L2. In some embodiments, the inhibitory receptor is TIGIT and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of CD112 or CD155.


In some embodiments of the immunomodulatory protein, the at least one affinity-modified IgSF domain specifically binds to no more than one cell surface cognate binding partner. In some embodiments, the immunomodulatory protein specifically binds to no more than one cell surface cognate binding partner.


In some embodiments of the immunomodulatory protein, the at least one affinity-modified domain specifically binds to at least two cell surface cognate binding partners. In some embodiments, the first cell surface cognate binding partner is a stimulatory receptor expressed on a T cell; and the second cell surface cognate binding partner is an inhibitory ligand of an inhibitory receptor, wherein the inhibitory receptor is expressed on a T-cell. In some embodiments, binding of the affinity-modified domain to the inhibitory ligand competitively inhibits binding of the inhibitory ligand to the inhibitory receptor. In some embodiments, the inhibitory receptor is PD-1, CTLA-4, LAG-3, TIGIT, CD96, CD112R, BTLA, CD160, TIM-3 VSIG3, or VSIG8; or the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, HVEM, MHC class II, PVR, CEACAM-1, GAL9 or VISTA. In some embodiments, the affinity modified IgSF domain is an affinity modified CD80 domain and the stimulatory receptor is CD28. In some embodiments, the inhibitory ligand is PD-L1 and the inhibitory receptor is PD-1. In some embodiments, the affinity-modified IgSF domain exhibits decreased binding affinity to CTLA-4 compared to the wild-type IgSF domain. In some embodiments, the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4.


In some embodiments of the immunomodulatory protein, the affinity modified IgSF domain is an affinity modified CD155 IgSF domain or an affinity modified CD112 IgSF domain and the at least one cell surface cognate binding partner is CD226, TIGIT or CD112R. In some embodiments, the affinity-modified IgSF domain exhibits decreased binding affinity to CD226 compared to the binding affinity of the wild-type IgSF domain to CD226. In some embodiments, the affinity-modified IgSF domain retains or exhibits increased binding to TIGIT or CD112R compared to the binding affinity of the wild-type IgSF domain to TIGIT or CD112R.


In some embodiments of the immunomodulatory protein, the at least one affinity-modified IgSF domain specifically binds to a cell surface cognate binding partner that is a tumor specific antigen. In some embodiments, the tumor specific antigen is B7-H6. In some embodiments, the affinity-modified IgSF domain is an affinity modified NKp30 IgSF domain.


In some embodiments of the immunomodulatory protein, the at least one affinity-modified IgSF domain comprises a first affinity-modified IgSF domain and a second affinity-modified IgSF domain. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain are different. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more different amino acid substitutions in the same wild-type IgSF domain. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more amino acid substitutions in a different wild-type IgSF domain.


In some embodiments of the immunomodulatory protein, the wild-type IgSF domain is from an IgSF member that is a ligand of an inhibitory receptor in which the inhibitory receptor comprises an ITIM signaling domain. In some embodiments, the inhibitory receptor is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8, respectively. In some embodiments, the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA, respectively. In some embodiments, the inhibitory receptor is PD-1 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of PD-L1 or PD-L2. In some embodiments, the inhibitory receptor is TIGIT and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of CD112 or CD155.


In some embodiments of the immunomodulatory proteins, the affinity modified IgSF domain differs by no more than ten amino acid substitutions from the wildtype IgSF domain. In some embodiments, the affinity modified IgSF domain differs by no more than five amino acid substitutions from the wildtype IgSF domain.


In some embodiments of the immunomodulatory protein, the one or more affinity-modified IgSF domain is or comprises an affinity modified IgV domain, affinity modified IgC1 domain, or an affinity modified IgC2 domain, or is a specific binding fragment thereof comprising the one or more amino acid substitutions.


In some embodiments of the immunomodulatory protein, the immunomodulatory protein further comprises one or more non-affinity modified IgSF domains.


In some embodiments of the immunomodulatory protein, the immunomodulatory protein has been or is capable of being secreted from an engineered cell. In some embodiments, the engineered cell is an immune cell. In some embodiments, the engineered cell is a primary cell.


In another aspect, there is provided a recombinant nucleic acid encoding an immunomodulatory protein. In some embodiments, the nucleic acid molecule further comprises at least one promoter operably linked to control expression of the immunomodulatory protein. In some embodiments, the promoter is a constitutively active promoter. In some embodiments, the promoter is an inducible promoter. In some embodiments, the promoter is responsive to an element responsive to T-cell activation signaling. In some embodiments, the promoter comprises a binding site for NFAT or a binding site for NF-κB. In another aspect, there is provided a recombinant expression vector comprising the nucleic acid described.


In a further aspect, there is provided a recombinant expression vector comprising a nucleic acid encoding an immunomodulatory protein under the operable control of a signal sequence for secretion, wherein: the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; and the encoded immunomodulatory protein is secreted when expressed from a cell. In some embodiments, the immunomodulatory protein does not comprise a transmembrane domain. In some embodiments, the immunomodulatory protein is not conjugated to a half-life extending moiety. In some embodiments, the half-life extending moiety is a multimerization domain. In some embodiments, the half-life extending moiety is an Fc domain. In some embodiments, the signal sequence for secretion encodes a secretory signal peptide. In some embodiments, the signal peptide is a native signal peptide from the corresponding wild-type IgSF member. In some embodiments, the signal peptide is a non-native signal peptide. In some embodiments, the signal peptide is an IgG-kappa signal peptide, an IL-2 signal peptide, or a CD33 signal peptide or other signal peptides known or described. Exemplary signal peptides are set forth in any of SEQ ID NOS: 413-430.


In some embodiments of the expression vector, the nucleic acid molecule further comprises at least one promoter operably linked to control expression of the immunomodulatory protein. In some embodiments, the promoter is a constitutively active promoter. In some embodiments, the promoter is an inducible promoter. In some embodiments, the promoter is responsive to an element responsive to T-cell activation signaling. In some embodiments, the promoter comprises a binding site for NFAT or a binding site for NF-κB.


In some embodiments of the expression vector, the vector is a viral vector. In some embodiments, the viral vector is a retroviral vector. In some embodiments, the viral vector is a lentiviral vector or a gammaretroviral vector.


In some embodiments of the expression vector, the at least one affinity-modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the wild-type IgSF domain.


In some embodiments of the expression vector, the at least one cell surface cognate binding partner is expressed on a mammalian cell. In some embodiments, the mammalian cell is an antigen presenting cell (APC), a tumor cell, or a lymphocyte. In some embodiments, the mammalian cell is a T-cell. In some embodiments, the mammalian cell is a mouse, rat, cynomolgus monkey, or human cell.


In some embodiments of the expression vector, specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain modulates immunological activity of the mammalian cell compared to the wild-type IgSF domain. In some embodiments, specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain increases immunological activity of the mammalian cell compared to the wild-type IgSF domain. In some embodiments, specific binding of the immunomodulatory protein attenuates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


In some embodiments of the expression vector, the wild-type IgSF domain is from an IgSF family member of a family selected from the group consisting of Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family, and Killer-cell immunoglobulin-like receptors (KIR) family. In some embodiments, the wild-type IgSF domain is from an IgSF member selected from the group consisting of CD80, CD86, PD-L1, PD-L2, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAG3, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R, NKp30, VISTA, VSIG3, and VSIG8. In some embodiments, the wild-type IgSF domain is a human IgSF domain. In some embodiments, the at least one affinity modified IgSF domain has at least 90% sequence identity to a wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27 and 408. In some embodiments, the immunomodulatory protein has at least 90% sequence identity to the amino acid sequence selected from any of SEQ ID NOS: 28-54 and 410. In some embodiments, the wild-type IgSF domain is a member of the B7 family. In some embodiments, the wild-type IgSF domain is a domain of CD80, CD86 or ICOSL.


In some embodiments of the expression vector, the at least one cell surface cognate binding partner is a stimulatory receptor expressed on a T-cell and the at least one affinity-modified IgSF domain has increased binding affinity to the stimulatory receptor compared to the binding affinity of the wild-type IgSF domain to the stimulatory receptor. In some embodiments, binding of the affinity-modified IgSF domain to the stimulatory receptor, such as when expressed from a cell containing the expression vector, increases immunological activity of the T-cell. In some embodiments, binding of the affinity-modified IgSF domain to the stimulatory receptor, such as when expressed from a cell containing the expression vector, decreases immunological activity of the T-cell. In some embodiments, the stimulatory receptor is CD28, ICOS, or CD226. In some embodiments, the at least one affinity-modified IgSF domain is an affinity-modified ICOSL IgSF domain that has increased binding affinity to at least one of: ICOS and CD28. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is ICOS. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is CD28. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified CD80 IgSF domain and the stimulatory receptor is CD28. In some embodiments, the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4 or exhibits decreased binding affinity to CTLA-4 compared to the binding affinity of wild-type IgSF domain to CTLA-4.


In some embodiments of the expression vector, the at least one affinity-modified IgSF domain specifically binds to no more than one cell surface cognate binding partner. In some embodiments, the immunomodulatory protein specifically binds to no more than one cell surface cognate binding partner.


In some embodiments of the expression vector, the at least one affinity-modified domain specifically binds to at least two cell surface cognate binding partners. In some embodiments, the first cell surface cognate binding partner is a stimulatory receptor expressed on a T cell; and the second cell surface cognate binding partner is an inhibitory ligand of an inhibitory receptor, wherein the inhibitory receptor is expressed on a T-cell. In some embodiments, binding of the affinity-modified IgSF domain to the inhibitory ligand competitively inhibits binding of the inhibitory ligand to the inhibitory receptor. In some embodiments, the inhibitory receptor is PD-1, CTLA-4, LAG-3, TIGIT, CD96, CD112R, BTLA, CD160, TIM-3, VSIG3, or VSIG8; or the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, HVEM, MHC class II, PVR, CEACAM-1, GAL9 or VISTA.


In some embodiments of the expression vector, the affinity modified IgSF domain is an affinity modified CD80 domain and the stimulatory receptor is CD28. In some embodiments, the affinity-modified IgSF domain exhibits decreased binding affinity to CTLA-4 compared to the wild-type IgSF domain. In some embodiments, the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4. In some embodiments, the inhibitory ligand is PD-L1 and the inhibitory receptor is PD-1. In some embodiments, the inhibitory ligand is PD-L2 and the inhibitory receptor is PD-1.


In some embodiments of the expression vector, the affinity-modified IgSF domain is an affinity modified CD155 IgSF domain or an affinity modified CD112 IgSF domain and the at least one cell surface cognate binding partner is CD226, TIGIT or CD112R. In some embodiments, the affinity-modified IgSF domain exhibits decreased binding affinity to CD226 compared to the binding affinity of the wild-type IgSF domain to CD226. In some embodiments, the affinity-modified IgSF domain retains or exhibits increased binding to TIGIT (T-cell immunoreceptor with Ig and ITIM domains) or CD112R compared to the binding affinity of the wild-type IgSF domain for TIGIT or CD112R.


In some embodiments of the expression vector, the at least one affinity-modified IgSF domain specifically binds to a cell surface cognate binding partner that is a tumor specific antigen. In some embodiments, the tumor specific antigen is B7-H6. In some embodiments, the affinity-modified IgSF domain is an affinity modified NKp30 IgSF domain.


In some embodiments of the expression vector, the at least one affinity-modified IgSF domain comprises a first affinity-modified IgSF domain and a second affinity-modified IgSF domain. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain are different. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more different amino acid substitutions in the same wild-type IgSF domain. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more amino acid substitutions in a different wild-type IgSF domain.


In some embodiments of the expression vector, the wild-type IgSF domain is from an IgSF member that is a ligand of an inhibitory receptor in which the inhibitory receptor comprises an ITIM signaling domain. In some embodiments, the inhibitory receptor is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8, respectively. In some embodiments, the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA, respectively. In some embodiments, the inhibitory receptor is PD-1 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of PD-L1 or PD-L2. In some embodiments, the affinity-modified IgSF domain has increased binding affinity for a trans surface cognate binding partner compared to the wildtype IgSF domain, whereby the increased binding affinity competitively inhibits binding of the trans surface cognate binding partner to the inhibitory receptor.


In some embodiments of the expression vector, the affinity modified IgSF domain differs by no more than ten amino acid substitutions from the wildtype IgSF domain. In some embodiments, the affinity modified IgSF domain differs by no more than five amino acid substitutions from the wildtype IgSF domain.


In some embodiments of the expression vector, the one or more affinity-modified IgSF domain is or comprises an affinity modified IgV domain, affinity modified IgC1 domain, or an affinity modified IgC2 domain, or is a specific binding fragment thereof, comprising the one or more amino acid substitutions.


In some embodiments of the expression vector, the immunomodulatory protein further comprises one or more non-affinity modified IgSF domains.


In another aspect, there is provided an engineered cell comprising any one or more of the above nucleic acid or the above expression vector. In another aspect, there is provided an engineered cell comprising any one or more of the immunomodulatory protein. In another aspect, there is provided an engineered cell that secretes any one or more of the immunomodulatory protein. In some embodiments, the engineered cell is an immune cell.


In another aspect, there is provided an engineered immune cell comprising a nucleic acid molecule that encodes an immunomodulatory protein, wherein the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; and the engineered cell expresses and secretes the immunomodulatory protein. In some embodiments, the immunomodulatory protein does not comprise a transmembrane domain. In some embodiments, the immunomodulatory protein is not conjugated to a half-life extending moiety. In some embodiments, the half-life extending moiety is a multimerization domain. In some embodiments, the half-life extending moiety is an Fc domain.


In some embodiments of the engineered cell, the nucleic acid molecule comprises a sequence encoding a secretory signal peptide operably linked to the sequence encoding the immunomodulatory protein. In some embodiments, the signal peptide is the native signal peptide from the corresponding wild-type IgSF member. In some embodiments, the signal peptide is a non-native signal sequence. In some embodiments, the signal peptide is an IgG-kappa signal peptide, an IL-2 signal peptide, or a CD33 signal peptide or any other signal peptide known in the art. Exemplary signal peptides are set forth in any of SEQ ID NOS: 413-430.


In some embodiments of the engineered cell, the nucleic acid molecule further comprises at least one promoter operably linked to control expression of the immunomodulatory protein. In some embodiments, the promoter is a constitutively active promoter. In some embodiments, the promoter is an inducible promoter. In some embodiments, the promoter is responsive to an element responsive to T-cell activation signaling. In some embodiments, the promoter comprises a binding site for NFAT or a binding site for NF-κB. In some embodiments, the immunomodulatory protein is expressed and secreted by the engineered cell after the engineered cell is contacted with an inducing agent or after induction of T cell activation signaling, which optionally is induced upon binding of an antigen to a chimeric antigen receptor (CAR) or engineered T-cell receptor (TCR) expressed by the engineered cell. In some embodiments, the cell is a lymphocyte. In some embodiments, the lymphocyte is a T cell, a B cell or an NK cell. In some embodiments, the cell is a T cell. In some embodiments, the T cell is CD4+ or CD8+. In some embodiments, the cell is an antigen presenting cell. In some embodiments, the cell is a primary cell obtained from a subject. In some embodiments, the subject is a human subject.


In some embodiments of the engineered cell, the at least one affinity modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the wild-type IgSF domain. In some embodiments, the at least one cell surface cognate binding partner is expressed on a mammalian cell. In some embodiments, the mammalian cell is an antigen presenting cell (APC), a tumor cell, or a lymphocyte. In some embodiments, the mammalian cell is a T-cell. In some embodiments, the mammalian cell is a mouse, rat, cynomolgus monkey, or human cell.


In some embodiments of the engineered cell, specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain modulates immunological activity of the mammalian cell compared to the wild-type IgSF domain. In some embodiments, specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain increases immunological activity of the mammalian cell compared to the wild-type IgSF domain. In some embodiments, specific binding of the immunomodulatory protein attenuates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


In some embodiments of the engineered cell, the wild-type IgSF domain is from an IgSF family member of a family selected from the group consisting of Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family, and Killer-cell immunoglobulin-like receptors (KIR) family. In some embodiments, the wild-type IgSF domain is from an IgSF member selected from the group consisting of CD80, CD86, PD-L1, PD-L2, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAG3, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R, NKp30, VISTA, VSIG3, and VSIG8. In some embodiments, the wild-type IgSF domain is a human IgSF domain. In some embodiments, the at least one affinity modified IgSF domain has at least 90% sequence identity to a wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27 and 408. In some embodiments, the immunomodulatory protein has at least 90% sequence identity to the amino acid sequence selected from any of SEQ ID NOS: 28-54 and 410.


In some embodiments of the engineered cell, the at least one cell surface cognate binding partner is a stimulatory receptor expressed on a T-cell and the at least one affinity-modified IgSF domain has increased binding affinity to the stimulatory receptor compared to the binding affinity of the wild-type IgSF domain to the stimulatory receptor. In some embodiments, binding of the affinity-modified IgSF domain to the stimulatory receptor, such as when secreted from the cell, increases immunological activity of the T-cell. In some embodiments, binding of the affinity-modified IgSF domain to the stimulatory receptor, such as when secreted from the cell, decreases immunological activity of the T-cell. In some embodiments, the stimulatory receptor is CD28, ICOS, or CD226. In some embodiments, the at least one affinity-modified IgSF domain is an affinity-modified ICOSL IgSF domain that has increased binding affinity to at least one of: ICOS and CD28. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is ICOS. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is CD28. In some embodiments, the at least one affinity-modified IgSF domain is an affinity modified CD80 IgSF domain and the stimulatory receptor is CD28. In some embodiments, the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4 or exhibits decreased binding affinity to CTLA-4 compared to the binding affinity of wild-type IgSF domain to CTLA-4.


In some embodiments of the engineered cell, the at least one affinity-modified IgSF domain specifically binds to no more than one cell surface cognate binding partner. In some embodiments, the immunomodulatory protein specifically binds to no more than one cell surface cognate binding partner.


In some embodiments of the engineered cell, the at least one affinity-modified domain specifically binds to at least two cell surface cognate binding partners. In some embodiments, the first cell surface cognate binding partner is a stimulatory receptor expressed on a T cell; and the second cell surface cognate binding partner is an inhibitory ligand of an inhibitory receptor, wherein the inhibitory receptor is expressed on a T-cell. In some embodiments, binding of the affinity-modified domain to the inhibitory ligand competitively inhibits binding of the inhibitory ligand to the inhibitory receptor. In some embodiments, the inhibitory receptor is PD-1, CTLA-4, LAG-3, TIGIT, CD96, CD112R, BTLA, CD160, TIM-3, VSIG3, or VSIG8; or the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, HVEM, MHC class II, PVR, CEACAM-1, GAL9 or VISTA. In some embodiments, the affinity modified IgSF domain is an affinity modified CD80 domain and the stimulatory receptor is CD28. In some embodiments, the inhibitory ligand is PD-L1 and the inhibitory receptor is PD-1. In some embodiments, the inhibitory ligand is PD-L2 and the inhibitory receptor is PD-1. In some embodiments, the affinity-modified IgSF domain exhibits decreased binding affinity to CTLA-4 compared to the wild-type IgSF domain. In some embodiments, the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4.


In some embodiments of the engineered cell, the affinity modified IgSF domain is an affinity modified CD155 IgSF domain or an affinity modified CD112 IgSF domain and the at least one cell surface cognate binding partner is CD226, TIGIT or CD112R. In some embodiments, the affinity-modified IgSF domain exhibits decreased binding affinity to CD226 compared to the binding affinity of the wild-type IgSF domain to CD226. In some embodiments, the affinity-modified IgSF domain retains or exhibits increased binding to TIGIT (T-cell immunoreceptor with Ig and ITIM domains) or CD112R compared to the binding affinity of the wild-type IgSF domain for TIGIT or CD112R.


In some embodiments of the engineered cell, the at least one affinity-modified IgSF domain specifically binds to a cell surface cognate binding partner that is a tumor specific antigen. In some embodiments, the tumor specific antigen is B7-H6. In some embodiments, the affinity-modified IgSF domain is an affinity modified NKp30 IgSF domain.


In some embodiments of the engineered cell, the at least one affinity-modified IgSF domain comprises a first affinity-modified IgSF domain and a second affinity-modified IgSF domain. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain are different. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprises one or more different amino acid substitutions in the same wild-type IgSF domain. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more amino acid substitutions in a different wild-type IgSF domain.


In some embodiments of the engineered cell, the wild-type IgSF domain is from an IgSF member that is a ligand of an inhibitory receptor in which the inhibitory receptor comprises an ITIM signaling domain. In some embodiments, the inhibitory receptor is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8, respectively. In some embodiments, the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA, respectively. In some embodiments, the inhibitory receptor is PD-1 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of PD-L1 or PD-L2. In some embodiments, the inhibitory receptor is TIGIT and the affinity-modified IgSF domain is an affinity-modified IgSF domain of CD155 or CD112.


In some embodiments of the engineered cell, the affinity modified IgSF domain differs by no more than ten amino acid substitutions from the wildtype IgSF domain. In some embodiments, the affinity modified IgSF domain differs by no more than five amino acid substitutions from the wildtype IgSF domain.


In some embodiments of the engineered cell, the one or more affinity-modified IgSF domain is or comprises an affinity modified IgV domain, affinity modified IgC1 domain, or an affinity modified IgC2 domain, or is a specific binding fragment thereof comprising the one or more amino acid substitutions.


In some embodiments of the engineered cell, the immunomodulatory protein further comprises one or more non-affinity modified IgSF domains.


In some embodiments of the engineered cell further comprises a chimeric antigen receptor (CAR) or an engineered T-cell receptor (TCR).


In another aspect, there is provided a pharmaceutical composition comprising the above engineered cell and a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition is sterile.


In another aspect, there is provided a method of introducing an immunomodulatory protein into a subject, comprising administering any of the provided engineered cells or the pharmaceutical composition containing the engineered cells to the subject.


In another aspect, there is provided a method of modulating an immune response in a subject, comprising administering any of the provided engineered cells or a pharmaceutical composition containing any of the provided engineered cells to the subject. In some embodiments, modulating the immune response treats a disease or disorder in the subject. In some embodiments, the modulated immune response is increased. In some embodiments, the disease or disorder is a tumor. In some embodiments, the disease or disorder is a cancer. In some embodiments, the disease or disorder is melanoma, lung cancer, bladder cancer, or a hematological malignancy. In some embodiments, the modulated immune response is decreased. In some embodiments, the disease or disorder is an inflammatory disease or condition. In some embodiments, the disease or condition is Crohn's disease, ulcerative colitis, multiple sclerosis, asthma, rheumatoid arthritis, or psoriasis.


In some embodiments of the methods, the subject is human. In some embodiments, the cell is autologous to the subject. In some embodiments, the cell is allogenic to the subject. In some embodiments, the engineered cell expresses and secretes the immunomodulatory protein. In some embodiments, the immunomodulatory protein is constitutively expressed by the engineered cell. In some embodiments, the immunomodulatory protein is expressed and secreted by the engineered cell after the engineered cell is contacted with an inducing agent. In some embodiments, the immunomodulatory protein is expressed and secreted by the engineered cell upon T cell activation signaling. In some embodiments, the engineered cell expresses a chimeric antigen receptor (CAR) or an engineered T-cell receptor (TCR) and T cell activation signaling is induced upon binding of an antigen by the CAR or TCR.


Also provided are any of the infectious agents as described containing a nucleic acid molecule encoding a secretable immunomodulatory protein or a transmembrane immunomodulatory protein.


Provided are infectious agents containing a nucleic acid molecule encoding a transmembrane immunomodulatory protein (TIP) containing an ectodomain comprising at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitution(s) in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; and a transmembrane domain.


In some embodiments, the at least one affinity modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the reference wild-type IgSF domain.


In some of any such embodiments, the wild-type IgSF domain is from an IgSF family member of a family selected from Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family or Killer-cell immunoglobulin-like receptors (KIR) family. In some embodiments, the wild-type IgSF domain is from an IgSF member selected from CD80, CD86, PD-L1, PD-L2, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAGS, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R, NKp30, VISTA, VSIG3, and VSIG8.


In some of any such embodiments, the wild-type IgSF domain is a human IgSF member. In some embodiments, the transmembrane immunomodulatory protein has at least 90% sequence identity to the amino acid sequence selected from any of SEQ ID NOS: 381-407 and 409 or to a contiguous portion thereof containing the affinity-modified IgSF domain and a transmembrane domain.


In some embodiments, the transmembrane immunomodulatory protein is a chimeric receptor, wherein the endodomain is not the endodomain from the wild-type IgSF member comprising the wild-type IgSF domain. In some cases, the endodomain contains at least one ITAM (immunoreceptor tyrosine-based activation motif)-containing signaling domain. In some instances, the endodomain contains a CD3-zeta signaling domain. In some aspects, the endodomain further contains at least one of: a CD28 costimulatory domain, an ICOS signaling domain, an OX40 signaling domain, and a 41BB signaling domain.


In some of any such embodiments, the affinity modified IgSF domain differs by no more than ten amino acid substitutions or no more than five amino acid substitutions from the wildtype IgSF domain. In some embodiments, the affinity-modified IgSF domain is or contains an affinity modified IgV domain, affinity modified IgC1 domain or an affinity modified IgC2 domain or is a specific binding fragment thereof comprising the one or more amino acid substitutions.


In some of any such embodiments, the transmembrane domain is the native transmembrane domain from the corresponding wild-type IgSF member. In some cases, the transmembrane domain is not the native transmembrane domain from the corresponding wild-type IgSF member. In some examples, the transmembrane protein is a transmembrane protein derived from CD8.


In some of any such embodiments, the infectious agent is a bacteria or a virus. In some cases, the virus is an oncolytic virus. In some aspects, the oncolytic virus is an adenovirus, adeno-associated virus, herpes virus, Herpes Simplex Virus, Vesticular Stomatic virus, Reovirus, Newcastle Disease virus, parvovirus, measles virus, vesticular stomatitis virus (VSV), Coxsackie virus or a Vaccinia virus. In some cases, the virus specifically targets dendritic cells (DCs) and/or is dendritic cell-tropic. In some embodiments, the virus is a lentiviral vector that is pseudotyped with a modified Sindbis virus envelope product.


In some of any such embodiments, the infectious agent further contains a nucleic acid molecule encoding a further gene product that results in death of a target cell or that can augment or boost an immune response. In some cases, the further gene product is selected from an anticancer agent, anti-metastatic agent, an antiangiogenic agent, an immunomodulatory molecule, an immune checkpoint inhibitor, an antibody, a cytokine, a growth factor, an antigen, a cytotoxic gene product, a pro-apoptotic gene product, an anti-apoptotic gene product, a cell matrix degradative gene, genes for tissue regeneration or a reprogramming human somatic cells to pluripotency.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A depicts results of a competition binding assay for binding of biotinylated recombinant CD28-Fc fusion protein (rCD28.Fc) to immobilized CD80 variant A91G ECD-Fc fusion molecule in the presence of unlabeled recombinant human PD-L1-His, human CTLA-4-His, or human-PD-L2-Fc fusion protein.



FIG. 1B depicts results of a competition binding assay for binding of biotinylated recombinant human PD-L1-his monomeric protein to immobilized CD80 variant A91G ECD-Fc fusion molecule in the presence of unlabeled recombinant human rCD28.Fc, human CTLA-4.Fc or human PD-L2.Fc



FIGS. 2A and 2B depicts the detection of PD-L2 SIP in supernatant of transduced CD19 CAR T cells and in HEK-293 cells, respectively.



FIG. 3A depicts the proliferation studies for T cells transduced with exemplary tested variant PD-L2 SIP.



FIG. 3B depicts levels of IFN-gamma in the supernatant released by T cells transduced with exemplary tested variant PD-L2 SIP as measured by ELISA on day 5 after re-stimulation.



FIG. 3C depicts proliferation of T cells co-transduced with a CAR and an exemplary variant PD-L2 SIP or a wild-type PD-L2 SIP following stimulation with target cells.



FIG. 4A depicts a secreted immunomodulatory protein (SIP) in which a variant IgSF domain (vIgD) is secreted from a cell, such as a first T cell (e.g. CAR T cell). In an exemplary embodiment, the cognate binding partner of the secreted vIgD is an activating receptor, which can be expressed on the first cell (e.g. T cell) and/or on a second cell (e.g. T cell; either endogenous or engineered, such as a CAR T cell). Upon binding of the SIP with its cognate binding partner, signaling via the activating receptor is blocked. In all cases, the vIgD can be a V-domain (IgV) only, the combination of the V-domain (IgV) and C-domain (IgC), including the entire extracellular domain (ECD), or any combination of Ig domains of the IgSF superfamily member.



FIG. 4B depicts a secreted immunomodulatory protein (SIP) in which a variant IgSF domain (vIgD) is secreted from a cell, such as a first T cell (e.g. CAR T cell). In an exemplary embodiment, the cognate binding partner of the secreted vIgD is an inhibitory receptor, which can be expressed on the first cell (e.g. CAR T cell) and/or on a second cell (e.g. T cell; either endogenous or engineered, such as a CAR T cell). Upon binding of the SIP with its cognate binding partner, the SIP antagonizes or blocks the negative signaling via the inhibitory receptor, thereby resulting in an activated T cell or effector T cell is blocked. In all cases, the vIgD can be a V-domain (IgV) only, the combination of the V-domain (IgV) and C-domain (IgC), including the entire extracellular domain (ECD), or any combination of Ig domains of the IgSF superfamily member.



FIG. 5 depicts proliferation studies for T cells transduced with exemplary tested variant PD-L1 SIP.



FIG. 6 depicts results of a cell-based assay for detection of SIPs, including variant or wild-type PD-L1 and PD-L2 SIPS, in supernatant of transduced HEK-293 cells.





DETAILED DESCRIPTION

Provided herein are secretable immunomodulatory proteins that can be secreted when expressed in a cell, nucleic acids and vectors encoding the same, and cells, such as immune cells or infectious agents, engineered to express and secrete such immunomodulatory proteins. The immunomodulatory protein contains an affinity-modified IgSF domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain. In some embodiments, the immunomodulatory protein does not include a transmembrane domain and/or a half-life extending moiety.


Among the provided embodiments are cells, such as immune cells (e.g. T cell), engineered to express and secrete the immunomodulatory proteins, for example by engineering the cells to include an expression vector that encodes the immunomodulatory protein. In some embodiments, the immunomodulatory protein encoded by the expression vector is under the operable control of a signal sequence for secretion, such that when the cell expresses the immunomodulatory protein, the immunomodulatory protein is secreted by the engineered cell. In some embodiments, expression of the immunomodulatory protein is under the operable control of a promotor. The promotor can be, for example, an inducible promoter. In some embodiments, the inducible promoter is responsive to an element responsive to T-cell activation signaling. In some embodiments, the engineered cell (e.g. T cell) also is engineered to express on its surface an antigen receptor, such as a T cell receptor (TCR) or chimeric antigen receptor (CAR), containing an intracellular signaling domain capable of or that does induce or mediate T-cell signaling upon antigen binding. In some aspects, the intracellular signaling domain comprises an immunoreceptor tyrosine-based activation motif (ITAM). Thus, in some embodiments, an engineered immune cell expresses and secretes an immunomodulatory protein only in response to activation of the immune cell, including, in some aspects, in response to recognition of antigen by an engineered antigen receptor.


Also provided herein are methods of introducing an immunomodulatory protein into a subject, the method comprising administering an engineered cell or infectious agent (or a pharmaceutical composition comprising an engineered cell or infectious agent) into the subject. In another aspect, there is provided a method of modulating an immune response in a subject, the method comprising administering an engineered cell (or a pharmaceutical composition comprising an engineered cell or infectious agent) into the subject. In another aspect, there is provided a method of treating a subject in need of treatment (such as a subject with a disease, such as cancer), the method comprising administering an effective amount of an engineered cell or infectious agent (or a pharmaceutical composition comprising an engineered cell or infectious agent) into the subject. The engineered cell can be a population of cells, such as a cell culture or a plurality of separately engineered cells.


In some embodiments, the provided engineered cells are administered to a subject and, in response to an induction signal (for example, immune cell activation or administration of an inducing agent) the engineered cells administered to the subject express and secrete the immunomodulatory protein. In some embodiments, the engineered cells constitutively express and secrete the immunomodulatory protein. In some embodiments, the engineered cells localize to a diseased cell or lesion, such as a cell present in a tumor microenvironment (e.g. tumor cell), which, in some aspects, is mediated by recognition of an antigen by an engineered antigen receptor expressed by the engineered cells, thereby targeting the secreted immunomodulatory proteins to the site of the disease or lesion, e.g. tumor microenvironment.


In some embodiments, the cognate binding partner of the immunomodulatory protein is a cell surface protein expressed by immune cells that engage with one or more other immune receptors (e.g. on lymphocytes) to induce inhibitory or activating signals. For example, the interaction of certain receptors on lymphocytes with their cell surface cognate binding partners to form an immunological synapse (IS) between antigen-presenting cells (APCs) or target cells and lymphocytes can provide costimulatory or inhibitory signals that can regulate the immune system. In some aspects, the immunomodulatory proteins provided herein (which can be expressed and secreted by an engineered cell) can alter the interaction of cell surface protein ligands with their receptors to modulate immune cells, such as T cell, activity. In some embodiments, the binding of the immunomodulatory protein to a ligand (cognate binding partner) modulates, e.g. induces, enhances or suppresses, immunological immune responses of a cell to which the immunomodulatory protein specifically binds. In some embodiments, the secretable immunomodulatory protein as provided, such as is secreted by the engineered cells, suppresses, such as inhibits or antagonizes, its cognate binding partner.


In some embodiments, under normal physiological conditions, the T cell-mediated immune response is initiated by antigen recognition by the T cell receptor (TCR) and is regulated by a balance of co-stimulatory and inhibitory signals (i.e., immune checkpoint proteins). The immune system relies on immune checkpoints to prevent autoimmunity (i.e., self-tolerance) and to protect tissues from excessive damage during an immune response, for example during an attack against a pathogenic infection. In some cases, however, these immunomodulatory proteins can be dysregulated in diseases and conditions, including tumors, as a mechanism for evading the immune system.


Thus, in some aspects, immunotherapy that alters immune cell activity, such as T cell activity, can treat certain diseases and conditions in which the immune response is dysregulated. Therapeutic approaches that seek to modulate interactions in the IS would benefit from the ability to bind multiple IS targets simultaneously and in a manner that is sensitive to temporal sequence and spatial orientation. Current therapeutic approaches fall short of this goal. Wild-type receptors and ligands possess low affinities for cognate binding partners, which can preclude their use as soluble therapeutics. Additionally, soluble receptors and antibodies, in some aspects, typically bind no more than a single target protein at a time and/or bind competitively to their targets (e.g., to no more than one target species at a time), and therefore lack the ability to simultaneously bind multiple targets. And while bispecific antibodies, as well as modalities comprising dual antigen binding regions, can bind to more than one target molecule simultaneously, the three-dimensional configuration typical of these modalities often precludes them intervening in key processes occurring in the IS in a manner consistent with their temporal and spatial requirements.


What is needed is an entirely new class of therapeutic molecules that have the specificity and affinity of antibodies, but also maintain the size, volume, and spatial orientation constraints required in the IS. Further, such therapeutics would have the ability to bind to their targets non-competitively as well as competitively. A molecule with these properties would therefore have novel function in the ability to integrate into multi-protein complexes at IS and generate the desired binding configuration and resulting biological activity.


To this end, emerging immuno-oncology therapeutic regimes need to safely break tumor-induced T cell tolerance. Current state-of-the-art immuno-therapeutics block PD-1 or CTLA4, central inhibitory molecules of the B7/CD28 family that are known to limit T cell effector function. While antagonistic antibodies against such single targets function to disrupt immune synapse checkpoint signaling complexes, they fall short of simultaneously activating T cells. Conversely, bispecific antibody approaches activate T cells, but fall short of simultaneously blocking inhibitory ligands that regulate the induced signal.


To address these shortcomings, provided are immunotherapies, such as cell therapies, that can modulate immune cell activities. In some embodiments, the provided immunotherapies can enhance immune cells signaling, such as T-cell activation signaling, and/or can block inhibitory regulation, which, in some cases, can occur simultaneously. In some embodiments, the provided immunotherapies relate to immunoglobulin superfamily (IgSF) components of the immune synapse that are known to have a dual role in both T-cell activation and blocking of inhibitory ligands. In some aspects, IgSF based-cell therapies engineered from immune system ligands, such as human immune system ligands themselves are more likely to retain their ability to normally assemble into key pathways of the immune synapse and maintain normal interactions and regulatory functions in ways that antibodies or next-generation bi-specific reagents cannot. This is due to the relatively large size of antibodies as well as from the fact they are not natural components of the immune synapse. These unique features of human immune system ligands, and cells engineered to express affinity-modified variants of such ligands, promise to provide a new level of immunotherapeutic efficacy and safety. In particular aspects, the provided engineered cells provide a secretable immunomodulatory platform using affinity-modified native immune ligands to generate immunotherapy biologics that bind with tunable affinities to one or more of their cognate immune receptors in the treatment of a variety of oncological and immunological indications.


All publications, including patents, patent applications scientific articles and databases, mentioned in this specification are herein incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, including patent, patent application, scientific article or database, were specifically and individually indicated to be incorporated by reference. If a definition set forth herein is contrary to or otherwise inconsistent with a definition set forth in the patents, applications, published applications and other publications that are herein incorporated by reference, the definition set forth herein prevails over the definition that is incorporated herein by reference.


The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.


I. Definitions

Unless defined otherwise, all terms of art, notations and other technical and scientific terms or terminology used herein are intended to have the same meaning as is commonly understood by one of ordinary skill in the art to which the claimed subject matter pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art.


The terms used throughout this specification are defined as follows unless otherwise limited in specific instances. Unless defined otherwise, all technical and scientific terms, acronyms, and abbreviations used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Unless indicated otherwise, abbreviations and symbols for chemical and biochemical names are per IUPAC-IUB nomenclature. Unless indicated otherwise, all numerical ranges are inclusive of the values defining the range as well as all integer values in-between.


As used in the specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly indicates otherwise.


The term “affinity-modified” as used in the context of an immunoglobulin superfamily domain, means a mammalian immunoglobulin superfamily (IgSF) domain having an altered amino acid sequence (relative to the corresponding wild-type parental or unmodified IgSF domain) such that it has an increased or decreased binding affinity or avidity to at least one of its cognate binding partners (alternatively “counter-structures”) compared to the parental wild-type or unmodified (i.e., non-affinity modified) IgSF control domain. In some embodiments, the affinity-modified IgSF domain can contain 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more amino acid differences, such as amino acid substitutions, in a wild-type or unmodified IgSF domain. An increase or decrease in binding affinity or avidity can be determined using well known binding assays such as flow cytometry. Larsen et al., American Journal of Transplantation, Vol 5: 443-453 (2005). See also, Linsley et al., Immunity, 1: 7930801 (1994). An increase in a protein's binding affinity or avidity to its cognate binding partner(s) is to a value at least 10% greater than that of the wild-type IgSF domain control and in some embodiments, at least 20%, 30%, 40%, 50%, 100%, 200%, 300%, 500%, 1000%, 5000%, or 10000% greater than that of the wild-type IgSF domain control value. A decrease in a protein's binding affinity or avidity to at least one of its cognate binding partner is to a value no greater than 90% of the control but no less than 10% of the wild-type IgSF domain control value, and in some embodiments no greater than 80%, 70% 60%, 50%, 40%, 30%, or 20% but no less than 10% of the wild-type IgSF domain control value. An affinity-modified protein is altered in primary amino acid sequence by substitution, addition, or deletion of amino acid residues. The term “affinity-modified IgSF domain” is not be construed as imposing any condition for any particular starting composition or method by which the affinity-modified IgSF domain was created. Thus, the affinity-modified IgSF domains of the present invention are not limited to wild-type IgSF domains that are then transformed to an affinity-modified IgSF domain by any particular process of affinity modification. An affinity-modified IgSF domain polypeptide can, for example, be generated starting from wild-type mammalian IgSF domain sequence information, then modeled in silico for binding to its cognate binding partner, and finally recombinantly or chemically synthesized to yield the affinity-modified IgSF domain composition of matter. In but one alternative example, an affinity-modified IgSF domain can be created by site-directed mutagenesis of a wild-type IgSF domain. Thus, affinity modified IgSF domain denotes a product and not necessarily a product produced by any given process. A variety of techniques including recombinant methods, chemical synthesis, or combinations thereof, may be employed.


The term “allogeneic” as used herein means a cell or tissue that is removed from one organism and then infused or adoptively transferred into a genetically dissimilar organism of the same species.


The term “autologous” as used herein means a cell or tissue that is removed from the same organism to which it is later infused or adoptively transferred. An autologous cell or tissue can be altered by, for example, recombinant DNA methodologies, such that it is no longer genetically identical to the native cell or native tissue which is removed from the organism. For example, a native autologous T-cell can be genetically engineered by recombinant DNA techniques to become an autologous engineered cell expressing a immunomodulatory protein (which can be secreted from the engineered cell) and/or chimeric antigen receptor (CAR), which in some cases involves engineering a T-cell or TIL (tumor infiltrating lymphocyte). The engineered cell can then be infused into a patient from which the native T-cell was isolated. In some embodiments, the organism is human or murine.


The terms “binding affinity,” and “binding avidity” as used herein means the specific binding affinity and specific binding avidity, respectively, of a protein for its cognate binding partner (i.e., its counter-structure) under specific binding conditions. In biochemical kinetics avidity refers to the accumulated strength of multiple affinities of individual non-covalent binding interactions, such as between an IgSF domain and its cognate binding partner (i.e., its counter-structure). As such, avidity is distinct from affinity, which describes the strength of a single interaction. An increase or attenuation in binding affinity of an affinity-modified IgSF domain to its cognate binding partner is determined relative to the binding affinity of the unmodified IgSF domain (e.g., the native or wild-type IgSF domain). Methods for determining binding affinity or avidity are known in art. See, for example, Larsen et al., American Journal of Transplantation, vol. 5: 443-453 (2005). The term “cell surface counter-structure” (alternatively “cognate cell surface binding partner”) as used herein is a counter-structure (alternatively is a cognate binding partner) expressed on a mammalian cell. Typically, the cell surface cognate binding partner is a transmembrane protein. In some embodiments, the cell surface cognate binding partner is a receptor.


The term “chimeric antigen receptor” or “CAR” as used herein refers to an artificial (i.e., man-made) transmembrane protein expressed on a mammalian cell comprising at least an ectodomain, a transmembrane, and an endodomain. Optionally, the CAR protein includes a “spacer” which covalently links the ectodomain to the transmembrane domain. A spacer is often a polypeptide linking the ectodomain to the transmembrane domain via peptide bonds. The CAR is typically expressed on a mammalian lymphocyte. In some embodiments, the CAR is expressed on a mammalian cell such as a T-cell or a tumor infiltrating lymphocyte (TIL). A CAR expressed on a T-cell is referred to herein as a CAR T-cell or “CAR-T.” In some embodiments the CAR-T is a T helper cell, a cytotoxic T-cell, a natural killer T-cell, a memory T-cell, a regulatory T-cell, or a gamma delta T-cell. When used clinically in, e.g. adoptive cell transfer, a CAR with antigen binding specificity to the patient's tumor is typically engineered to be expressed on a native lymphocyte obtained from the patient. The engineered lymphocyte expressing the CAR is then infused back into the patient. The lymphocyte is thus often an autologous T-cell although allogeneic T-cells are included within the scope of the invention. The ectodomain of a CAR comprises an antigen binding region, such as an antibody or antigen binding fragment thereof (e.g. scFv), that specifically binds under physiological conditions with an antigen, such as a tumor specific antigen. Upon specific binding a biochemical chain of events (i.e., signal transduction) results in modulation of the immunological activity of the cell on which the CAR is expressed. Thus, for example, upon specific binding by the antigen binding region of the CAR-T to its antigen can lead to changes in the immunological activity of the T-cell activity as reflected by changes in cytotoxicity, proliferation or cytokine production. Signal transduction upon CAR activation is achieved in some embodiments by the CD3-zeta chain (“CD3-z”) which is involved in signal transduction in native mammalian T-cells. CARs can further comprise multiple signaling domains such as CD28, ICOS, 41BB or OX40, to further modulate immunomodulatory response of the T-cell. CD3-z comprises a conserved motif known as an immunoreceptor tyrosine-based activation motif (ITAM) which is involved in T-cell receptor signal transduction.


The terms “cognate binding partner” or “counter-structure” in reference to a protein, such as an IgSF domain or an affinity-modified IgSF domain, refers to at least one molecule (typically a native mammalian protein) to which the referenced protein specifically binds under specific binding conditions. In some aspects, an affinity-modified IgSF domain specifically binds to the cognate binding partner of the corresponding native or wild-type IgSF domain but with increased or attenuated affinity. A species of ligand recognized and specifically binding to its cognate receptor under specific binding conditions is an example of a counter-structure or cognate binding partner of that receptor. A receptor, to which a native ligand recognizes and specifically binds to under specific binding conditions, is an example of a cognate binding partner of that ligand. In turn, the native ligand is the cognate binding partner of the receptor. For example, ICOSL specifically binds to CD28 and ICOS and thus these proteins are cognate binding partners of ICOSL. In another example, a tumor specific antigen and an affinity-modified IgSF domain to which it specifically binds are each cognate binding partners of the other. In the present invention a “cell surface molecular species” is a cognate binding partner of ligands of the immunological synapse (IS), expressed on and by cells, such as mammalian cells, forming the immunological synapse, for example immune cells.


The term “competitive binding” as used herein means that a protein is capable of specifically binding to at least two cognate binding partners but that specific binding of one cognate binding partner inhibits, such as prevents or precludes, simultaneous binding of the second cognate binding partner. Thus, in some cases, it is not possible for a protein to bind the two cognate binding partners at the same time. Generally, competitive binders contain the same or overlapping binding site for binding but this is not a requirement. In some embodiments, competitive binding causes a measurable inhibition (partial or complete) of specific binding of a protein to one of its cognate binding partner due to specific binding of a second cognate binding partner. A variety of methods are known to quantify competitive binding such as ELISA (enzyme linked immunosorbent assay) or Forte-Bio Octet experimental systems.


The term “conservative amino acid substitution” as used herein means an amino acid substitution in which an amino acid residue is substituted by another amino acid residue having a side chain R group with similar chemical properties (e.g., charge or hydrophobicity). Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine, and isoleucine; 2) aliphatic-hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartic acid and glutamic acid; and 7) sulfur-containing side chains: cysteine and methionine. Conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine.


The term, “corresponding to” with reference to positions of a protein, such as recitation that nucleotides or amino acid positions “correspond to” nucleotides or amino acid positions in a disclosed sequence, such as set forth in the Sequence Listing, refers to nucleotides or amino acid positions identified upon alignment with the disclosed sequence based on structural sequence alignment or using a standard alignment algorithm, such as the GAP algorithm. By aligning the sequences, one skilled in the art can identify corresponding residues, for example, using conserved and identical amino acid residues as guides.


The term “cytokine” includes, e.g., but is not limited to, interleukins, interferons (IFN), chemokines, hematopoietic growth factors, tumor necrosis factors (TNF), and transforming growth factors. In general, these are small molecular weight proteins that regulate maturation, activation, proliferation, and differentiation of cells of the immune system.


The terms “derivatives” or “derivatized” refer to modification of an immunomodulatory protein by covalently linking it, directly or indirectly, so as to alter such characteristics as half-life, bioavailability, immunogenicity, solubility, toxicity, potency, or efficacy while retaining or enhancing its therapeutic benefit. Derivatives can be made by glycosylation, pegylation, lipidation, or Fc-fusion. In some embodiments, the immunomodulatory protein is not derivatized. In some embodiments, the immunomodulatory protein is not conjugated to a half-life extending moiety, such as an Fc domain.


As used herein, “domain” (typically a sequence of three or more, generally 5 or 7 or more amino acids, such as 10 to 200 amino acid residues) refers to a portion of a molecule, such as a protein or encoding nucleic acid, that is structurally and/or functionally distinct from other portions of the molecule and is identifiable. For example, domains include those portions of a polypeptide chain that can form an independently folded structure within a protein made up of one or more structural motifs and/or that is recognized by virtue of a functional activity, such as binding activity. A protein can have one, or more than one, distinct domains. For example, a domain can be identified, defined or distinguished by homology of the primary sequence or structure to related family members, such as homology to motifs. In another example, a domain can be distinguished by its function, such as an ability to interact with a biomolecule, such as a cognate binding partner. A domain independently can exhibit a biological function or activity such that the domain independently or fused to another molecule can perform an activity, such as, for example binding. A domain can be a linear sequence of amino acids or a non-linear sequence of amino acids. Many polypeptides contain a plurality of domains. Such domains are known, and can be identified by those of skill in the art. For exemplification herein, definitions are provided, but it is understood that it is well within the skill in the art to recognize particular domains by name. If needed appropriate software can be employed to identify domains. It is understood that reference to amino acids, including to a specific sequence set forth as a SEQ ID NO used to describe domain organization of an IgSF domain are for illustrative purposes and are not meant to limit the scope of the embodiments provided. It is understood that polypeptides and the description of domains thereof are theoretically derived based on homology analysis and alignments with similar molecules. Thus, the exact locus can vary, and is not necessarily the same for each protein. Hence, the specific IgSF domain, such as specific IgV domain or IgC domain, can be several amino acids (one, two, three or four) longer or shorter.


The term “ectodomain,” “extracellular domain,” or “ECD” as used herein refers to the region of a membrane protein, such as a transmembrane protein, that lies outside the vesicular membrane (e.g., the space outside of a cell). Ectodomains often interact with specific ligands or specific cell surface receptors, such as via a binding domain that specifically binds to the ligand or cell surface receptor.


The terms “effective amount” or “therapeutically effective amount” refer to a quantity and/or concentration of a therapeutic composition of the invention, such as composition containing engineered cells, that when administered ex vivo (by contact with a cell from a patient) or in vivo (by administration into a patient, such as by adoptive transfer) either alone (i.e., as a monotherapy) or in combination with additional therapeutic agents, yields a statistically significant inhibition of disease progression as, for example, by ameliorating or eliminating symptoms and/or the cause of the disease. An effective amount for treating an immune system disease or disorder may be an amount that relieves, lessens, or alleviates at least one symptom or biological response or effect associated with the disease or disorder, prevents progression of the disease or disorder, or improves physical functioning of the patient. In the case of cell therapy, the effective amount is an effective dose or number of cells administered to a patient. In some embodiments the patient is a human patient.


The term “endodomain” as used herein refers to the region found in some membrane proteins, such as transmembrane proteins, that extends into the interior space defined by the cell surface membrane. In mammalian cells, the endodomain is the cytoplasmic region of the membrane protein. In cells, the endodomain interacts with intracellular constituents and can be play a role in signal transduction and thus, in some cases, can be an intracellular signaling domain. The endodomain of a cellular transmembrane protein is alternately referred to as a cytoplasmic domain, which, in some cases, can be a cytoplasmic signaling domain.


The term “enhanced” or “increased” as used herein in the context of increasing immunological activity of a mammalian lymphocyte means to increase one or more activities of the lymphocyte. An increased activity can be one or more of an increase cell survival, cell proliferation, cytokine production, or T-cell cytotoxicity, such as by a statistically significant amount. In some embodiments, reference to increased immunological activity means to increase interferon gamma (IFN-gamma) production, such as by a statistically significant amount. Methods of assessing activities of lymphocytes are known in the art, including any assay as described herein. In some embodiments an enhancement can be an increase of at least 10%, 20%, 30%, 40%, 50%, 75%, 100%, 200%, 300%, 400%, or 500% greater than a non-zero control value.


The term “engineered cell” as used herein refers to a mammalian cell that has been genetically modified by human intervention such as by recombinant DNA methods or viral transduction. In some embodiments, the engineered cell is an immune cell, such as a lymphocyte (e.g. T cell, B cell, NK cell) or an antigen presenting cell (e.g. dendritic cell). The cell can be a primary cell from a patient or can be a cell line. In some embodiments, an engineered cell is capable of expressing and secreting an immunomodulatory protein as described herein. In some embodiments, the engineered cell further expresses or contains an engineered T-cell receptor (TCR) or chimeric antigen receptor (CAR).


The term “engineered T-cell” as used herein refers to a T-cell such as a T helper cell, cytotoxic T-cell (alternatively, cytotoxic T lymphocyte or CTL), natural killer T-cell, regulatory T-cell, memory T-cell, or gamma delta T-cell, that has been genetically modified by human intervention such as by recombinant DNA methods. In some embodiments, an engineered T-cell is capable of expressing and secreting an immunomodulatory protein as described herein.


The term “engineered T-cell receptor” or “engineered TCR” refers to a T-cell receptor (TCR) engineered to specifically bind with a desired affinity to a major histocompatibility complex (MHC)/peptide target antigen that is selected, cloned, and/or subsequently introduced into a population of T-cells, often used for adoptive immunotherapy. In contrast to engineered TCRs, CARs are engineered to bind target antigens in a MHC independent manner.


A protein “expressed on” a cell is used herein to reference to a protein expressed by a cell and in which at least a portion of the protein is present on the surface of the cell. Proteins expressed on a cell include transmembrane proteins or cell surface receptors. In some embodiments, the protein is conjugated to a small molecule moiety such as a drug or detectable label.


The term “half-life extending moiety” refers to a moiety of a polypeptide fusion or chemical conjugate that extends the half-life of a protein circulating in mammalian blood serum compared to the half-life of the protein that is not so conjugated to the moiety. In some embodiments, half-life is extended by greater than or greater than about 1.2-fold, 1.5-fold, 2.0-fold, 3.0-fold, 4.0-fold, 5.0-fold, or 6.0-fold. In some embodiments, half-life is extended by more than 6 hours, more than 12 hours, more than 24 hours, more than 48 hours, more than 72 hours, more than 96 hours or more than 1 week after in vivo administration compared to the protein without the half-life extending moiety. The half-life refers to the amount of time it takes for the protein to lose half of its concentration, amount, or activity. Half-life can be determined for example, by using an ELISA assay or an activity assay. Exemplary half-life extending moieties include an Fc domain, a multimerization domain, polyethylene glycol (PEG), hydroxyethyl starch (HES), XTEN (extended recombinant peptides; see, WO2013130683), human serum albumin (HSA), bovine serum albumin (BSA), lipids (acylation), and poly-Pro-Ala-Ser (PAS), and polyglutamic acid (glutamylation).


The term “host cell” refers to any cell that can be used to express a protein encoded by a recombinant expression vector. A host cell can be a prokaryote, for example, E. coli, or it can be a eukaryote, for example, a single-celled eukaryote (e.g., a yeast or other fungus), a plant cell (e.g., a tobacco or tomato plant cell), an animal cell (e.g., a human cell, a monkey cell, a hamster cell, a rat cell, a mouse cell, or an insect cell) or a hybridoma. Examples of host cells include Chinese hamster ovary (CHO) cells or their derivatives such as Veggie CHO and related cell lines which grow in serum-free media or CHO strain DX-B11, which is deficient in DHFR.


The term “immunological synapse” or “immune synapse” (abbreviated “IS”) as used herein means the interface between a mammalian cell that expresses MHC I (major histocompatibility complex) or MHC II, such as an antigen-presenting cell or tumor cell, and a mammalian lymphocyte such as an effector T cell or Natural Killer (NK) cell.


The term “immunoglobulin” (abbreviated “Ig”) as used herein is synonymous with the term “antibody” (abbreviated “Ab”) and refers to a mammalian immunoglobulin protein including any of the five human classes: IgA (which includes subclasses IgA1 and IgA2), IgD, IgE, IgG (which includes subclasses IgG1, IgG2, IgG3, and IgG4), and IgM. The term is also inclusive of immunoglobulins that are less than full-length, whether wholly or partially synthetic (e.g., recombinant or chemical synthesis) or naturally produced, such as antigen binding fragment (Fab), variable fragment (Fv) containing VH and VL, the single chain variable fragment (scFv) containing VH and VL linked together in one chain, as well as other antibody V region fragments, such as Fab′, F(ab)2, F(ab′)2, dsFv diabody, Fc, and Fd polypeptide fragments. Bispecific antibodies, homobispecific and heterobispecific, are included within the meaning of the term.


An Fc (fragment crystallizable) region or domain of an immunoglobulin molecule (also termed an Fc polypeptide) corresponds largely to the constant region of the immunoglobulin heavy chain, and is responsible for various functions, including the antibody's effector function(s). An immunoglobulin Fc fusion (“Fc-fusion”) is a molecule comprising one or more polypeptides (or one or more small molecules) operably linked to an Fc region of an immunoglobulin. An Fc-fusion may comprise, for example, the Fc region of an antibody (which, in some cases, facilitates effector functions and pharmacokinetics) and the IgSF domain of a wild-type or affinity-modified immunoglobulin superfamily domain (“IgSF”), or other protein or fragment thereof. In some embodiments, the Fc is a variant Fc that exhibits reduced (e.g. reduced greater than 30%, 40%, 50%, 60%, 70%, 80%, 90% or more) activity to facilitate an effector function. The IgSF domain mediates recognition of the cognate binding partner (comparable to that of antibody variable region of an antibody for an antigen). An immunoglobulin Fc region may be linked indirectly or directly to one or more polypeptides or small molecules (fusion partners). Various linkers are known in the art and can be used to link an Fc to a fusion partner to generate an Fc-fusion. An Fc-fusion protein can comprise an immunoglobulin Fc region covalently linked, directly or indirectly, to at least one affinity modified IgSF domain. Fc-fusions of identical species can be dimerized to form Fc-fusion homodimers, or using non-identical species to form Fc-fusion heterodimers. In some embodiments, the immunomodulatory protein is not conjugated to an Fc domain or any portion thereof.


The term “immunoglobulin superfamily” or “IgSF” as used herein means the group of cell surface and soluble proteins that are involved in the recognition, binding, or adhesion processes of cells. Molecules are categorized as members of this superfamily based on shared structural features with immunoglobulins (i.e., antibodies); they all possess a domain known as an immunoglobulin domain or fold. Members of the IgSF include cell surface antigen receptors, co-receptors and co-stimulatory molecules of the immune system, molecules involved in antigen presentation to lymphocytes, cell adhesion molecules, certain cytokine receptors and intracellular muscle proteins. They are commonly associated with roles in the immune system. Proteins in the immunological synapse are often members of the IgSF. IgSF can also be classified into “subfamilies” based on shared properties such as function. Such subfamilies typically consist of from 4 to 30 IgSF members.


The terms “IgSF domain” or “immunoglobulin domain” or “Ig domain” as used herein refers a structural domain of IgSF proteins. Ig domains are named after the immunoglobulin molecules. They contain about 70-110 amino acids and are categorized according to their size and function. Ig-domains possess a characteristic Ig-fold, which has a sandwich-like structure formed by two sheets of antiparallel beta strands. Interactions between hydrophobic amino acids on the inner side of the sandwich and highly conserved disulfide bonds formed between cysteine residues in the B and F strands, stabilize the Ig-fold. One end of the Ig domain has a section called the complementarity determining region that is important for the specificity of antibodies for their ligands. The Ig like domains can be classified (into classes) as: IgV, IgC1, IgC2, or IgI. Most Ig domains are either variable (IgV) or constant (IgC). IgV domains with 9 beta strands are generally longer than IgC domains with 7 beta strands. Ig domains of some members of the IgSF resemble IgV domains in the amino acid sequence, yet are similar in size to IgC domains. These are called IgC2 domains, while standard IgC domains are called IgC1 domains. T-cell receptor (TCR) chains contain two Ig domains in the extracellular portion; one IgV domain at the N-terminus and one IgC1 domain adjacent to the cell membrane.


The term “IgSF species” as used herein means an ensemble of IgSF member proteins with identical or substantially identical primary amino acid sequence. Each mammalian immunoglobulin superfamily (IgSF) member defines a unique identity of all IgSF species that belong to that IgSF member. Thus, each IgSF family member is unique from other IgSF family members and, accordingly, each species of a particular IgSF family member is unique from the species of another IgSF family member. Nevertheless, variation between molecules that are of the same IgSF species may occur owing to differences in post-translational modification such as glycosylation, phosphorylation, ubiquitination, nitrosylation, methylation, acetylation, and lipidation. Additionally, minor sequence differences within a single IgSF species owing to gene polymorphisms constitute another form of variation within a single IgSF species as do wild type truncated forms of IgSF species owing to, for example, proteolytic cleavage. A “cell surface IgSF species” is an IgSF species expressed on the surface of a cell, generally a mammalian cell.


The term “immunological activity” as used herein in the context of mammalian lymphocytes refers to one or more cell survival, cell proliferation, cytokine production (e.g. interferon-gamma), or T-cell cytotoxicity activities. Methods to assay the immunological activity of engineered cells, including to evaluate the activity of the immunomodulatory protein, are known in the art and include, but are not limited to, the ability to expand T cells following antigen stimulation, sustain T cell expansion in the absence of re-stimulation, and anti-cancer activities in appropriate animal models. Assays also include assays to assess cytotoxicity, including a standard 51Cr-release assay (see e.g. Milone et al., (2009) Molecular Therapy 17: 1453-1464) or flow based cytotoxicity assays, or an impedance based cytotoxicity assay (Peper et al. (2014) Journal of Immunological Methods, 405:192-198). Assays to assess immunological activity of engineered cells can be compared to control non-engineered cells or to cells containing one or more other engineered recombinant receptor (e.g. antigen receptor) with a known activity.


An “immunomodulatory protein” or “immunomodulatory polypeptide” is a protein that modulates immunological activity. By “modulation” or “modulating” an immune response is meant that immunological activity is either enhanced or suppressed. An immunomodulatory protein can be a single polypeptide chain or a multimer (dimers or higher order multimers) of at least two polypeptide chains covalently bonded to each other by, for example, interchain disulfide bonds. Thus, monomeric, dimeric, and higher order multimeric proteins are within the scope of the defined term. Multimeric proteins can be homomultimeric (of identical polypeptide chains) or heteromultimeric (of different polypeptide chains). Secretable immunomodulatory proteins are a type of immunomodulatory protein.


The term “increase” as used herein means to increase by a statistically significant amount. An increase can be at least 5%, 10%, 20%, 30%, 40%, 50%, 75%, 100%, or greater than a non-zero control value.


The term “lymphocyte” as used herein means any of three subtypes of white blood cell in a mammalian immune system. They include natural killer cells (NK cells) (which function in cell-mediated, cytotoxic innate immunity), T cells (for cell-mediated, cytotoxic adaptive immunity), and B cells (for humoral, antibody-driven adaptive immunity). T cells include T helper cells, cytotoxic T-cells, natural killer T-cells, memory T-cells, regulatory T-cells, or gamma delta T-cells. Innate lymphoid cells (ILC) are also included within the definition of lymphocyte.


An “inhibitory counter-structure” or “inhibitory cognate binding partner” is a cell membrane protein, often a receptor, which when proximally bound near a separate activating receptor leads to an attenuation in the frequency, duration, magnitude, or intensity of the activating signaling cascade and phenotype mediated by the activating receptor. Examples of inhibitory receptors include PD-1, CTLA-4, LAG-3, TIGIT, CD96, CD112R, BTLA, CD160, TIM-3, and VSIG8. The term “stimulatory counter-structure” or “stimulatory cognate binding partner” is a cell membrane protein, often a receptor, which when activated and signal transduction is thereby induced, leads to an increase in the frequency, duration, or intensity of the phenotype mediated by that receptor. Examples of stimulatory receptors include CD28, ICOS, and CD226.


The term “lymphocyte” as used herein means any of three subtypes of white blood cell in a mammalian immune system. They include natural killer cells (NK cells) (which function in cell-mediated, cytotoxic innate immunity), T cells (for cell-mediated, cytotoxic adaptive immunity), and B cells (for humoral, antibody-driven adaptive immunity). T cells include: T helper cells, cytotoxic T-cells, natural killer T-cells, memory T-cells, regulatory T-cells, or gamma delta T-cells. Innate lymphoid cells (ILC) are also included within the definition of lymphocyte.


The terms “mammal,” “subject,” or “patient” specifically includes reference to at least one of a: human, chimpanzee, rhesus monkey, cynomolgus monkey, dog, cat, mouse, or rat.


The term “membrane protein” as used herein means a protein that, under physiological conditions, is attached directly or indirectly to a lipid bilayer. A lipid bilayer that forms a membrane can be a biological membrane such as a eukaryotic (e.g., mammalian) cell membrane or an artificial (i.e., man-made) membrane such as that found on a liposome. Attachment of a membrane protein to the lipid bilayer can be by way of covalent attachment, or by way of non-covalent interactions such as hydrophobic or electrostatic interactions. A membrane protein can be an integral membrane protein or a peripheral membrane protein. Membrane proteins that are peripheral membrane proteins are non-covalently attached to the lipid bilayer or non-covalently attached to an integral membrane protein. A peripheral membrane protein forms a temporary attachment to the lipid bilayer such that under the range of conditions that are physiological in a mammal, peripheral membrane protein can associate and/or disassociate from the lipid bilayer. In contrast to peripheral membrane proteins, integral membrane proteins form a substantially permanent attachment to the membrane's lipid bilayer such that under the range of conditions that are physiological in a mammal, integral membrane proteins do not disassociate from their attachment to the lipid bilayer. A membrane protein can form an attachment to the membrane by way of one layer of the lipid bilayer (monotopic), or attached by way of both layers of the membrane (polytopic). An integral membrane protein that interacts with only one lipid bilayer is an “integral monotopic protein”. An integral membrane protein that interacts with both lipid bilayers is an “integral polytopic protein” alternatively referred to herein as a “transmembrane protein”.


The terms “modulating” or “modulate” as used herein in the context of an immune response, such as a mammalian immune response, refer to any alteration, such as an increase or decrease, of an existing or potential immune responses that occurs as a result of administration of an immunomodulatory protein or as a result of administration of engineered cells expressing an immunomodulatory protein, such as a secretable immunomodulatory protein of the present invention. Such modulation includes any induction, or alteration in degree or extent, or suppression of immunological activity of an immune cell. Immune cells include B cells, T cells, NK (natural killer) cells, NK T cells, professional antigen-presenting cells (APCs), and non-professional antigen-presenting cells, and inflammatory cells (neutrophils, macrophages, monocytes, eosinophils, and basophils). Modulation includes any change imparted on an existing immune response, a developing immune response, a potential immune response, or the capacity to induce, regulate, influence, or respond to an immune response. Modulation includes any alteration in the expression and/or function of genes, proteins and/or other molecules in immune cells as part of an immune response. Modulation of an immune response or modulation of immunological activity includes, for example, the following: elimination, deletion, or sequestration of immune cells; proliferation, induction, survival or generation of immune cells that can modulate the functional capacity of other cells such as autoreactive lymphocytes, antigen presenting cells, or inflammatory cells; induction of an unresponsive state in immune cells (i.e., anergy); enhancing or suppressing the activity or function of immune cells, including but not limited to altering the pattern of proteins expressed by these cells. Examples include altered production and/or secretion of certain classes of molecules such as cytokines, chemokines, perforins, granzymes, growth factors, transcription factors, kinases, costimulatory molecules, or other cell surface receptors or any combination of these modulatory events. Modulation can be assessed, for example, by an alteration of an immunological activity of engineered cells, such as an alteration in in cytotoxic activity of engineered cells or an alteration in cytokine secretion of engineered cells relative to cells engineered with the wild-type IgSF protein.


The term “molecular species” as used herein means an ensemble of proteins with identical or substantially identical primary amino acid sequence. Each mammalian immunoglobulin superfamily (IgSF) member defines a collection of identical or substantially identical molecular species. Thus, for example, human CD80 is an IgSF member and each human CD80 molecule is a species of CD80. Variation between molecules that are of the same molecular species may occur owing to differences in post-translational modification such as glycosylation, phosphorylation, ubiquitination, nitrosylation, methylation, acetylation, and lipidation. Additionally, minor sequence differences within a single molecular species owing to gene polymorphisms constitute another form of variation within a single molecular species as do wild type truncated forms of a single molecular species owing to, for example, proteolytic cleavage. A “cell surface molecular species” is a molecular species expressed on the surface of a mammalian cell. Two or more different species of protein, each of which is present exclusively on one or exclusively the other (but not both) of the two mammalian cells forming the IS, are said to be in “cis” or “cis configuration” with each other. Two different species of protein, the first of which is exclusively present on one of the two mammalian cells forming the IS and the second of which is present exclusively on the second of the two mammalian cells forming the IS, are said to be in “trans” or “trans configuration.” Two different species of protein each of which is present on both of the two mammalian cells forming the IS are in both cis and trans configurations on these cells.


The term “non-competitive binding” as used herein means the ability of a protein to specifically bind simultaneously to at least two cognate binding partners. In some embodiments, the binding occurs under specific binding conditions. Thus, the protein is able to bind to at least two different cognate binding partners at the same time although the binding interaction need not be for the same duration such that, in some cases, the protein is specifically bound to only one of the cognate binding partners. In some embodiments, the simultaneous binding is such that binding of one cognate binding partner does not substantially inhibit simultaneous binding to a second cognate binding partner. In some embodiments, non-competitive binding means that binding a second cognate binding partner to its binding site on the protein does not displace the binding of a first cognate binding partner to its binding site on the protein. Methods of assessing non-competitive binding are well known in the art such as the method described in Perez de La Lastra et al., Immunology, 1999 April: 96(4): 663-670. In some cases, in non-competitive interactions, the first cognate binding partner specifically binds at an interaction site that does not overlap with the interaction site of the second cognate binding partner such that binding of the second cognate binding partner does not directly interfere with the binding of the first cognate binding partner. Thus, any effect on binding of the cognate binding partner by the binding of the second cognate binding partner is through a mechanism other than direct interference with the binding of the first cognate binding partner. For example, in the context of enzyme-substrate interactions, a non-competitive inhibitor binds to a site other than the active site of the enzyme. Non-competitive binding encompasses uncompetitive binding interactions in which a second cognate binding partner specifically binds at an interaction site that does not overlap with the binding of the first cognate binding partner but binds to the second interaction site only when the first interaction site is occupied by the first cognate binding partner.


The terms “nucleic acid” and “polynucleotide” are used interchangeably to refer to a polymer of nucleic acid residues (e.g., deoxyribonucleotides or ribonucleotides) in either single- or double-stranded form. Unless specifically limited, the terms encompass nucleic acids containing known analogues of natural nucleotides and that have similar binding properties to it and are metabolized in a manner similar to naturally-occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary nucleotide sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues. The term nucleic acid or polynucleotide encompasses cDNA or mRNA encoded by a gene.


The term “pharmaceutical composition” refers to a composition suitable for pharmaceutical use in a mammalian subject, often a human. A pharmaceutical composition typically comprises an effective amount of an active agent (e.g., an immunomodulatory protein or engineered cells expressing and/or secreting an immunomodulatory protein of the present invention) and a carrier, excipient, or diluent. The carrier, excipient, or diluent is typically a pharmaceutically acceptable carrier, excipient or diluent, respectively.


The terms “polypeptide” and “protein” are used interchangeably herein and refer to a molecular chain of two or more amino acids linked through peptide bonds. The terms do not refer to a specific length of the product. Thus, “peptides,” and “oligopeptides,” are included within the definition of polypeptide. The terms include post-translational modifications of the polypeptide, for example, glycosylations, acetylations, phosphorylations and the like. The terms also include molecules in which one or more amino acid analogs or non-canonical or unnatural amino acids are included as can be synthesized, or expressed recombinantly using known protein engineering techniques. In addition, proteins can be derivatized as described herein by well-known organic chemistry techniques.


The term “primary T-cell assay” as used herein refers to an in vitro assay to measure interferon-gamma (“IFN-gamma”) expression. A variety of such primary T-cell assays are known in the art such as that described in Example 6. In a preferred embodiment, the assay used is anti-CD3 coimmobilization assay. In this assay, primary T cells are stimulated by anti-CD3 immobilized with or without additional recombinant proteins. Culture supernatants are harvested at timepoints, usually 24-72 hours. In another embodiment, the assay used is a mixed lymphocyte reaction (MLR). In this assay, primary T cells are simulated with allogenic APC. Culture supernatants are harvested at timepoints, usually 24-72 hours. Human IFN-gamma levels are measured in culture supernatants by standard ELISA techniques. Commercial kits are available from vendors and the assay is performed according to manufacturer's recommendation.


The term “purified” as applied to nucleic acids, such as encoding a secretable immunomodulatory proteins, or proteins (e.g. immunomodulatory proteins) generally denotes a nucleic acid or polypeptide that is substantially free from other components as determined by analytical techniques well known in the art (e.g., a purified polypeptide or polynucleotide forms a discrete band in an electrophoretic gel, chromatographic eluate, and/or a media subjected to density gradient centrifugation). For example, a nucleic acid or polypeptide that gives rise to essentially one band in an electrophoretic gel is “purified.” A purified nucleic acid, or protein is at least about 50% pure, usually at least about 75%, 80%, 85%, 90%, 95%, 96%, 99% or more pure (e.g., percent by weight or on a molar basis).


The term “recombinant” indicates that the material (e.g., a nucleic acid or a polypeptide) has been artificially (i.e., non-naturally) altered by human intervention. The alteration can be performed on the material within, or removed from, its natural environment or state. For example, a “recombinant nucleic acid” is one that is made by recombining nucleic acids, e.g., during cloning, affinity modification, DNA shuffling or other well-known molecular biological procedures. A “recombinant DNA molecule,” is comprised of segments of DNA joined together by means of such molecular biological techniques. The term “recombinant protein” or “recombinant polypeptide” as used herein refers to a protein molecule (e.g., an immunomodulatory protein) which is expressed using a recombinant DNA molecule. A “recombinant host cell” is a cell that contains and/or expresses a recombinant nucleic acid or that is otherwise altered by genetic engineering, such as by introducing into the cell a nucleic acid molecule encoding a recombinant protein, such as a immunomodulatory protein provided herein. Transcriptional control signals in eukaryotes comprise “promoter” and “enhancer” elements. Promoters and enhancers consist of short arrays of DNA sequences that interact specifically with cellular proteins involved in transcription. Promoter and enhancer elements have been isolated from a variety of eukaryotic sources including genes in yeast, insect and mammalian cells and viruses (analogous control elements, i.e., promoters, are also found in prokaryotes). The selection of a particular promoter and enhancer depends on what cell type is to be used to express the protein of interest. The terms “in operable combination,” “in operable order” and “operably linked” as used herein refer to the linkage of nucleic acid sequences in such a manner or orientation that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced. The term also refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced and/or transported.


The term “recombinant expression vector” as used herein refers to a DNA molecule containing a desired coding sequence (e.g., encoding an immunomodulatory protein) and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular cell. Nucleic acid sequences necessary for expression in prokaryotes include a promoter, optionally an operator sequence, a ribosome binding site and possibly other sequences. Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals. A secretory signal peptide sequence can also, optionally, be encoded by the recombinant expression vector, operably linked to the coding sequence so that the expressed protein can be secreted by the recombinant host cell, for more facile isolation of the fusion protein from the cell, if desired. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Among the vectors are viral vectors, such as lentiviral vectors.


The term “selectivity” refers to the preference of a subject protein, or polypeptide, for specific binding of one substrate, such as one cognate binding partner, compared to specific binding for another substrate, such as a different cognate binding partner of the subject protein. Selectivity can be reflected as a ratio of the binding activity (e.g. binding affinity) of a subject protein and a first substrate, such as a first cognate binding partner, (e.g., Kd1) and the binding activity (e.g. binding affinity) of the same subject protein with a second cognate binding partner (e.g., Kd2).


The term “sequence identity” as used herein refers to the sequence identity between genes or proteins at the nucleotide or amino acid level, respectively. “Sequence identity” is a measure of identity between proteins at the amino acid level and a measure of identity between nucleic acids at nucleotide level. The protein sequence identity may be determined by comparing the amino acid sequence in a given position in each sequence when the sequences are aligned. Similarly, the nucleic acid sequence identity may be determined by comparing the nucleotide sequence in a given position in each sequence when the sequences are aligned. Methods for the alignment of sequences for comparison are well known in the art, such methods include GAP, BESTFIT, BLAST, FASTA and TFASTA. The BLAST algorithm calculates percent sequence identity and performs a statistical analysis of the similarity between the two sequences. The software for performing BLAST analysis is publicly available through the National Center for Biotechnology Information (NCBI) website.


The term “soluble” as used herein in reference to proteins, means that the protein is not a membrane protein. In general, a soluble protein contains only the extracellular domain of an IgSF family member receptor, or a portion thereof containing an IgSF domain or domains or specific-binding fragments thereof.


The term “species” as used herein in the context of a nucleic acid sequence or a polypeptide sequence refers to an identical collection of such sequences. Slightly truncated sequences that differ (or encode a difference) from the full length species at the amino-terminus or carboxy-terminus by no more than 1, 2, or 3 amino acid residues are considered to be of a single species. Such microheterogeneities are a common feature of manufactured proteins.


The term “specifically binds” as used herein means the ability of a protein, under specific binding conditions, to bind to a target protein such that its affinity or avidity is at least 10 times as great, but optionally 50, 100, 250 or 500 times as great, or even at least 1000 times as great as the average affinity or avidity of the same protein to a collection of random peptides or polypeptides of sufficient statistical size. A specifically binding protein need not bind exclusively to a single target molecule (e.g., its cognate binding partner) but may specifically bind to a non-target molecule due to similarity in structural conformation between the target and non-target (e.g., paralogs or orthologs). Those of skill will recognize that specific binding to a molecule having the same function in a different species of animal (i.e., ortholog) or to a non-target molecule having a substantially similar epitope as the target molecule (e.g., paralog) is possible and does not detract from the specificity of binding which is determined relative to a statistically valid collection of unique non-targets (e.g., random polypeptides). Thus, an affinity-modified polypeptide of the invention may specifically bind to more than one distinct species of target molecule due to cross-reactivity. Generally, such off-target specific binding is mitigated by reducing affinity or avidity for undesired targets. Solid-phase ELISA immunoassays or Biacore measurements can be used to determine specific binding between two proteins. Generally, interactions between two binding proteins have dissociation constants (Kd) less than about 1×10−5 M, and often as low as about 1×10−12 M. In certain aspects of the present disclosure, interactions between two binding proteins have dissociation constants of less than about 1×10−6 M, 1×10−7 M, 1×10−8 M, 1×10−9 M, 1×10−10 M, or 1×10−11 M.


The term “specific binding fragment” or “fragment” as used herein in reference to a mature (i.e., absent the signal peptide) wild-type IgSF domain, means a polypeptide that is shorter than the full-length mature IgSF domain and that specifically binds in vitro and/or in vivo to the wild-type IgSF domain's native cognate binding partner. In some embodiments, the specific binding fragment is at least 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% the sequence length of the full-length mature wild-type sequence. The specific binding fragment can be altered in sequence to form an affinity modified IgSF domain of the invention. In some embodiments, the specific binding fragment modulates immunological activity of a lymphocyte. The terms “suppress” or “attenuate” or “decrease” as used herein means to decrease by a statistically significant amount. In some embodiments suppression can be a decrease of at least 10%, and up to 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%.


As used herein, “synthetic,” with reference to, for example, a synthetic nucleic acid molecule or a synthetic gene or a synthetic peptide refers to a nucleic acid molecule or polypeptide molecule that is produced by recombinant methods and/or by chemical synthesis methods.


The term “transmembrane protein” as used herein means a membrane protein that substantially or completely spans a lipid bilayer such as those lipid bilayers found in a biological membrane such as a mammalian cell, or in an artificial construct such as a liposome. The transmembrane protein comprises a transmembrane domain (“transmembrane domain”) by which it is integrated into the lipid bilayer and by which the integration is thermodynamically stable under physiological conditions. Transmembrane domains are generally predictable from their amino acid sequence via any number of commercially available bioinformatics software applications on the basis of their elevated hydrophobicity relative to regions of the protein that interact with aqueous environments (e.g., cytosol, extracellular fluid). A transmembrane domain is often a hydrophobic alpha helix that spans the membrane. A transmembrane protein can pass through the both layers of the lipid bilayer once or multiple times.


The terms “treating,” “treatment,” or “therapy” of a disease or disorder as used herein mean slowing, stopping or reversing the disease or disorders progression, as evidenced by decreasing, cessation or elimination of either clinical or diagnostic symptoms, by administration of an immunomodulatory protein or engineered cells expressing a transmembrane immunomodulatory protein of the present invention either alone or in combination with another compound as described herein. “Treating,” “treatment,” or “therapy” also means a decrease in the severity of symptoms in an acute or chronic disease or disorder or a decrease in the relapse rate as for example in the case of a relapsing or remitting autoimmune disease course or a decrease in inflammation in the case of an inflammatory aspect of an autoimmune disease. As used herein in the context of cancer, the terms “treatment” or, “inhibit,” “inhibiting” or “inhibition” of cancer refers to at least one of: a statistically significant decrease in the rate of tumor growth, a cessation of tumor growth, or a reduction in the size, mass, metabolic activity, or volume of the tumor, as measured by standard criteria such as, but not limited to, the Response Evaluation Criteria for Solid Tumors (RECIST), or a statistically significant increase in progression free survival (PFS) or overall survival (OS). “Preventing,” “prophylaxis,” or “prevention” of a disease or disorder as used in the context of this invention refers to the administration of an immunomodulatory protein or engineered cells expressing an immunomodulatory protein of the present invention, either alone or in combination with another compound, to prevent the occurrence or onset of a disease or disorder or some or all of the symptoms of a disease or disorder or to lessen the likelihood of the onset of a disease or disorder.


The term “tumor specific antigen” or “TSA” as used herein refers to a an antigen that is present primarily on tumor cells of a mammalian subject but generally not found on normal cells of the mammalian subject. In some cases, a tumor specific antigen is a counter structure or cognate binding partner of an IgSF member. A tumor specific antigen need not be exclusive to tumor cells but the percentage of cells of a particular mammal that have the tumor specific antigen is sufficiently high or the levels of the tumor specific antigen on the surface of the tumor are sufficiently high such that it can be targeted by anti-tumor therapeutics and provide prevention or treatment of the mammal from the effects of the tumor. In some embodiments, in a random statistical sample of cells from a mammal with a tumor, at least 50% of the cells displaying a TSA are cancerous. In other embodiments, at least 60%, 70%, 80%, 85%, 90%, 95%, or 99% of the cells displaying a TSA are cancerous.


The term “wild-type” or “natural” or “native” as used herein is used in connection with biological materials such as nucleic acid molecules, proteins, IgSF members, host cells, and the like, refers to those which are found in nature and not modified by human intervention.


II. Immunomodulatory Proteins Containing Affinity-Modified Domains

In some embodiments, the immunomodulatory protein, including secretable immunomodulatory proteins (SIPs) or transmembrane immunomodulatory proteins (TIPs), includes at least one affinity-modified IgSF domain compared to an IgSF domain of a wild-type mammalian IgSF member. The wild-type mammalian IgSF member excludes antibodies (i.e., immunoglobulins) such as those that are mammalian or may be of mammalian origin. Thus, the present invention relates to IgSF domains that are non-immunoglobulin (i.e., non-antibody) IgSF domains. Wild-type mammalian IgSF family members that are not immunoglobulins (i.e. antibodies) are known in the art as are their nucleic and amino acid sequences. Affinity-modified IgSF domains of a wild-type IgSF domain of all non-immunoglobulin mammalian IgSF family members are included within the scope of the invention.


In some embodiments, immunomodulatory proteins as provided in their various embodiments comprise at least one affinity-modified mammalian IgSF domain, such as at least one affinity modified non-immunoglobulin mammalian IgSF domain. In some embodiments, the non-immunoglobulin IgSF family members, and the corresponding IgSF domains present therein, are of mouse, rat, cynomolgus monkey, or human origin. In some embodiments, the IgSF family members are members from at least or exactly one, two, three, four, five, or more IgSF subfamilies such as: Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, or Killer-cell immunoglobulin-like receptors (KIR) family. In some embodiments, the at least one IgSF domain is derived from an IgSF protein that is any of CD80(B7-1), CD86(B7-2), CD274 (PD-L1, B7-H1), PDCD1LG2(PD-L2, CD273), ICOSLG(B7RP1, CD275, ICOSL, B7-H2), CD276(B7-H3), VTCN1(B7-H4), CD28, CTLA4, PDCD1(PD-1), ICOS, BTLA(CD272), CD4, CD8A(CD8-alpha), CD8B(CD8-beta), LAG3, HAVCR2(TIM-3), CEACAM1, TIGIT, PVR(CD155), PVRL2(CD112), CD226, CD2, CD160, CD200, CD200R1(CD200R), NC R3 (NKp30), VISTA, VSIG3, and VSIG8.


In some embodiments, the immunomodulatory protein contains at least one affinity-modified IgSF domain. In some embodiments, the at least one affinity-modified IgSF domain is affinity-modified compared to a corresponding IgSF domain of a non-immunoglobulin IgSF family member that is a mammalian IgSF member. In some embodiments, the mammalian IgSF member is one of the IgSF members or comprises an IgSF domain from one of the IgSF members as indicated in Table 1 including any mammalian orthologs thereof. Orthologs are genes in different species that evolved from a common ancestral gene by speciation. Normally, orthologs retain the same function in the course of evolution. In some embodiments, the affinity modified IgSF domain is an affinity modified IgV or IgC domain, including IgC1 or IgC2 domain.


In some embodiments, the immunomodulatory protein of the present invention comprises the sequence of the extracellular domain of a wild-type mammalian non-immunoglobulin (i.e., non-antibody) IgSF family member but wherein at least one IgSF domain therein is affinity modified. Solely by way of example, in some embodiments, a wild-type mammalian non-immunoglobulin IgSF family member comprises a first IgSF domain and a second IgSF domain, and an immunomodulatory protein comprises the first IgSF domain and the second IgSF domain of the wild-type IgSF family member, except at least the first IgSF domain or the second IgSF domain is affinity-modified. Immunomodulatory proteins comprising the sequence of the extracellular domain of a wild-type mammalian immunoglobulin (i.e., non-antibody) IgSF family member, but wherein at least one IgSF domain is affinity modified can be referred to as “Type I” immunomodulatory proteins. Additional domains present within the IgSF family can be affinity modified, such as at least two, three, four, or five IgSF domains and, in some embodiments, exactly two, three, four, or five IgSF domains. In some embodiments of an immunomodulatory protein of the invention, the mammalian IgSF member will be one of the IgSF members as indicated in Table 1 including any mammalian orthologs thereof


The first column of Table 1 provides the name and, optionally, the name of some possible synonyms for that particular IgSF member. The second column provides the protein identifier of the UniProtKB database, a publicly available database accessible via the internet at uniprot.org. The Universal Protein Resource (UniProt) is a comprehensive resource for protein sequence and annotation data. The UniProt databases include the UniProt Knowledgebase (UniProtKB). UniProt is a collaboration between the European Bioinformatics Institute (EMBL-EBI), the SIB Swiss Institute of Bioinformatics and the Protein Information Resource (PR) and supported mainly by a grant from the U.S. National Institutes of Health (NIH). The third column provides the region where the indicated IgSF domain is located. The region is specified as a range where the domain is inclusive of the residues defining the range. Column 3 also indicates the IgSF domain class for the specified IgSF region. Column 4 provides the region where the indicated additional domains are located (signal peptide, S; extracellular domain, E; transmembrane domain, T; cytoplasmic domain, C). Column 5 indicates for some of the listed IgSF members, some of its cognate cell surface binding partners.


Typically, the affinity-modified IgSF domain of the immunomodulatory protein of the provided embodiments is a human or murine affinity modified IgSF domain.









TABLE 1







IgSF members according to the present disclosure.













NCBI







Protein



Accession



IgSF Member Amino Acid



Number/


Cognate Cell
Sequence (SEQ ID NO)














IgSF
UniProtKB
IgSF Region

Surface
Precursor




Member
Protein
& Domain
Other
Binding
(mature


(Synonyms)
Identifier
Class
Domains
Partners
residues)
Mature
ECD


CD80
NP_005182.1
35-135, 35-138,
S: 1-34,
CD28,
SEQ ID NO: 1
SEQ ID
SEQ ID





(B7-1)
P33681
35-141
E: 35-242,
CTLA4, PD-
(35-288)
NO: 381
NO: 28




or 37-138
T: 243-263,
L1




IgV,
C: 264-288




145-230 or




154-232 IgC


CD86
P42081.2
33-131 IgV,
S: 1-23,
CD28, CTLA4
SEQ ID NO: 2
SEQ ID
SEQ ID


(B7-2)

150-225 IgC2
E: 24-247,

(24-329)
NO: 382
NO: 29





T: 248-268,





C: 269-329


CD274
Q9NZQ7.1
24-130 IgV,
S: 1-18,
PD-1, B7-1
SEQ ID NO: 3
SEQ ID
SEQ ID


(PD-L1, B7-

133-225 IgC2
E: 19-238,

(19-290)
NO: 383
NO: 30


H1)


T: 239-259,





C: 260-290


PDCD1LG2
Q9BQ51.2
21-118 IgV,
S: 1-19,
PD-1, RGMb
SEQ ID NO: 4
SEQ ID
SEQ ID


(PD-L2,

122-203 IgC2
E: 20-220,

(20-273)
NO: 384
NO: 31


CD273)


T: 221-241,





C: 242-273


ICOSLG
O75144.2
19-129 IgV,
S: 1-18,
ICOS, CD28,
SEQ ID NO: 5
SEQ ID
SEQ ID


(B7RP1,

141-227 IgC2
E: 19-256,
CTLA4
(19-302)
NO: 385
NO: 32


CD275,


T: 257-277,


ICOSL, B7-


C: 278-302


H2)


CD276
Q5ZPR3.1
29-139 IgV,
S: 1-28,

SEQ ID NO: 6
SEQ ID
SEQ ID


(B7-H3)

145-238
E: 29-466,

(29-534)
NO: 386
NO: 33




IgC2,
T: 467-487,




243-357
C: 488-534




IgV2, 363-456,




367-453




IgC2


VTCN1
Q7Z7D3.1
35-146 IgV,
S: 1-24,

SEQ ID NO: 7
SEQ ID
SEQ ID


(B7-H4)

153-241 IgV
E: 25-259,

(25-282)
NO: 387
NO: 34





T: 260-280,





C: 281-282


CD28
P10747.1
28-137 IgV
S: 1-18,
B7-1, B7-2,
SEQ ID NO: 8
SEQ ID
SEQ ID





E: 19-152,
B7RP1
(19-220)
NO: 388
NO: 35





T: 153-179,





C: 180-220


CTLA4
P16410.3
39-140 IgV
S: 1-35,
B7-1, B7-2,
SEQ ID NO: 9
SEQ ID
SEQ ID





E: 36-161,
B7RP1
(36-223)
NO: 389
NO: 36





T: 162-182,





C: 183-223


PDCD1
Q15116.3
35-145 IgV
S: 1-20,
PD-L1, PD-L2
SEQ ID NO: 10
SEQ ID
SEQ ID


(PD-1)


E: 21-170,

(21-288)
NO: 390
NO: 37





T: 171-191,





C: 192-288


ICOS
Q9Y6W8.1
30-132 IgV
S: 1-20,
B7RP1
SEQ ID NO: 11
SEQ ID
SEQ ID





E: 21-140,

(21-199)
NO: 391
NO: 38





T: 141-161,





C: 162-199


BTLA
Q7Z6A9.3
31-132 IgV
S: 1-30,
HVEM
SEQ ID NO: 12
SEQ ID
SEQ ID


(CD272)


E: 31-157,

(31-289)
NO: 392
NO: 39





T: 158-178,





C: 179-289


CD4
P01730.1
26-125 IgV,
S: 1-25,
MHC class II
SEQ ID NO: 13
SEQ ID
SEQ ID




126-203
E: 26-396,

(26-458)
NO: 393
NO: 40




IgC2, 204-317
T: 397-418,




IgC2,
C: 419-458




317-389, 318-374




IgC2


CD8A
P01732.1
22-135 IgV
S: 1-21, E:
MHC class I
SEQ ID NO: 14
SEQ ID
SEQ ID


(CD8-alpha)


22-182, T:

(22-235)
NO: 394
NO: 41





183-203, C:





204-235


CD8B
P10966.1
22-132 IgV
S: 1-21,
MHC class I
SEQ ID NO: 15
SEQ ID
SEQ ID


(CD8-beta)


E: 22-170,

(22-210)
NO: 395
NO: 42





T: 171-191,





C: 192-210


LAG3
P18627.5
37-167 IgV,
S: 1-28,
MHC class II
SEQ ID NO: 16
SEQ ID
SEQ ID




168-252
E: 29-450,

(29-525)
NO: 396
NO: 43




IgC2,
T: 451-471,




265-343
C: 472-525




IgC2, 349-419




IgC2


HAVCR2
Q8TDQ0.3
22-124 IgV
S: 1-21,
CEACAM-1,
SEQ ID NO: 17
SEQ ID
SEQ ID


(TIM-3)


E: 22-202,
phosphatidyl
(22-301)
NO: 397
NO: 44





T: 203-223,
serine,





C: 224-301
Galectin-9,






HMGB1


CEACAM1
P13688.2
35-142 IgV,
S: 1-34,
TIM-3
SEQ ID NO: 18
SEQ ID
SEQ ID




145-232
E: 35-428,

(35-526)
NO: 398
NO: 45




IgC2, 237-317
T: 429-452,




IgC2,
C: 453-526




323-413 IgC2


TIGIT
Q495A1.1
22-124 IgV
S: 1-21,
CD155,
SEQ ID NO: 19
SEQ ID
SEQ ID





E: 22-141,
CD112
(22-244)
NO: 399
NO: 46





T: 142-162,





C: 163-244


PVR
P15151.2
24-139 IgV,
S: 1-20,
TIGIT,
SEQ ID NO: 20
SEQ ID
SEQ ID


(CD155)

145-237
E: 21-343,
CD226,
(21-417)
NO: 400
NO: 47




IgC2, 244-328
T: 344-367,
CD96,




IgC2
C: 368-417
poliovirus


PVRL2
Q92692.1
32-156 IgV,
S: 1-31,
TIGIT,
SEQ ID NO: 21
SEQ ID
SEQ ID


(CD112)

162-256
E: 32-360,
CD226,
(32-538)
NO: 401
NO: 48




IgC2, 261-345
T: 361-381,
CD112R




IgC2
C: 382-538


CD226
Q15762.2
19-126 IgC2,
S: 1-18,
CD155,
SEQ ID NO: 22
SEQ ID
SEQ ID




135-239 IgC2
E: 19-254,
CD112
(19-336)
NO: 402
NO: 49





T: 255-275,





C: 276-336


CD2
P06729.2
25-128 IgV,
S: 1-24,
CD58
SEQ ID NO: 23
SEQ ID
SEQ ID




129-209 IgC2
E: 25-209,

(25-351)
NO: 403
NO: 50





T: 210-235,





C: 236-351


CD160
O95971.1
27-122 IgV
N/A
HVEM, MHC
SEQ ID NO: 24
SEQ ID
SEQ ID






family of
(27-159)
NO: 404
NO: 51






proteins


CD200
P41217.4
31-141 IgV,
S: 1-30,
CD200R
SEQ ID NO: 25
SEQ ID
SEQ ID




142-232 IgC2
E: 31-232,

(31-278)
NO: 405
NO: 52





T: 233-259,





C: 260-278


CD200R1
Q8TD46.2
53-139 IgV,
S: 1-28,
CD200
SEQ ID NO: 26
SEQ ID
SEQ ID


(CD200R)

140-228 IgC2
E: 29-243,

(29-325)
NO: 406
NO: 53





T: 244-264,





C: 265-325


NCR3
O14931.1
19-126 IgC-
S: 1-18,
B7-H6
SEQ ID NO: 27
SEQ ID
SEQ ID


(NKp30)

like
E: 19-135,

(19-201)
NO: 407
NO: 54





T: 136-156,





C: 157-201


VSIG8
Q5VU13
22-141 IgV1,
S: 1-21
VISTA
SEQ ID NO: 408
SEQ ID
SEQ ID




146-257
E: 22-263

(22-414)
NO: 409
NO:




IgV2
T: 264-284



410





C: 285-414









In some embodiments, the immunomodulatory protein contains at least one affinity-modified domain and at least one non-affinity modified IgSF domain (e.g. unmodified or wild-type IgSF domain). In some embodiments, the immunomodulatory protein contains at least two affinity modified domains. In some embodiments, the immunomodulatory protein contains a plurality of non-affinity modified IgSF domains and/or affinity-modified IgSF domains, such as 1, 2, 3, 4, 5, or 6 non-affinity modified IgSF and/or affinity modified IgSF domains.


In some embodiments, the immunomodulatory protein comprises a combination (a “non-wild-type combination”) and/or arrangement (a “non-wild type arrangement” or “non-wild-type permutation”) of an affinity-modified and/or non-affinity modified IgSF domain sequences that are not found in wild-type IgSF family members (“Type II” immunomodulatory proteins). The sequences of the IgSF domains which are non-affinity modified (e.g., wild-type) or have been affinity modified can be mammalian, such as from mouse, rat, cynomolgus monkey, or human origin, or combinations thereof. In some embodiments, the sequence of the non-affinity modified domain is any IgSF domain set forth in Table 1. The number of such non-affinity modified or affinity modified IgSF domains present in these embodiments of a Type II immunomodulatory protein (whether non-wild type combinations or non-wild type arrangements) is at least 2, 3, 4, or 5 and in some embodiments exactly 2, 3, 4, or 5 IgSF domains.


In some embodiments, at least two of the affinity modified IgSF domains are identical affinity modified IgSF domains. In some embodiments, the affinity modified IgSF domains are non-identical (i.e., different) IgSF domains. Non-identical affinity modified IgSF domains specifically bind, under specific binding conditions, different cognate binding partners and are “non-identical” irrespective of whether or not the wild-type IgSF domains from which they are designed was the same. Thus, for example, a combination of at least two non-identical IgSF domains in the immunomodulatory protein of the present invention can comprise at least one IgSF domain sequence whose origin is from and unique to one IgSF family member and at least one of a second IgSF domain sequence whose origin is from and unique to another IgSF family member wherein the IgSF domains of the immunomodulatory protein are in affinity modified form. However, in alternative embodiments, the two non-identical IgSF domains originate from the same IgSF domain sequence but are affinity modified differently such that they specifically bind to different cognate binding partners. In some embodiments, the number of non-identical affinity modified IgSF domains present in the immunomodulatory protein of the invention is at least 2, 3, 4, or 5 and in some embodiments exactly 2, 3, 4, or 5 non-identical affinity modified IgSF domains. In some embodiments, the non-identical IgSF domains are combinations from at least two IgSF members indicated in Table 1, and in some embodiments at least three or four IgSF members of Table 1.


In other embodiments an immunomodulatory protein provided herein comprises at least two IgSF domains from a single IgSF member but in a non-wild-type arrangement. One illustrative example of a non-wild type arrangement or permutation is an immunomodulatory protein of the present invention comprising a non-wild type order of affinity modified IgSF domain sequences relative to those found in the wild-type mammalian IgSF family member whose IgSF domain sequences served as the source of the affinity modified IgSF domains. The mammalian wild-type IgSF member in the preceding embodiment specifically includes those listed in Table 1. Thus, in one example, if the wild-type family member comprises an IgC1 domain proximal to the N-terminus of the protein and an IgV domain distal to the N-terminus, then the immunomodulatory protein provided herein can comprise an IgV proximal and an IgC1 distal to the N-terminus, albeit in affinity modified form. The presence, in an immunomodulatory protein, of both non-wild type combinations and non-wild type arrangements of affinity modified IgSF domains is also within the scope of the present invention. A plurality of affinity-modified IgSF domains in an immunomodulatory protein's polypeptide chain need not be covalently linked directly to one another. In some embodiments, an intervening span of one or more amino acid residues indirectly covalently bonds the affinity-modified IgSF domains to each other. Such “peptide linkers” can be a single amino acid residue or greater in length.


In some embodiments, the affinity modified IgSF domain can be affinity modified to specifically bind to a single (e.g., 1) or multiple (e.g., 2, 3, 4, or more) counter-structures (also called a “cognate binding partner”) expressed on a mammalian cell. Typically, the cognate binding partner is a native cognate binding partner of the wild-type IgSF domain that has been affinity modified. In some embodiments, the cognate binding partner is an IgSF member. In some embodiments the cognate binding partner e is a non-IgSF family member. For example, in some embodiments the cognate binding partner of an affinity-modified IgSF domain such as BTLA (B- and T-lymphocyte attenuation) is the non-IgSF member cognate binding partner HVEM (herpes virus entry mediator). BTLA-HVEM complexes negatively regulate T-cell immune responses. Each IgSF domain present in an immunomodulatory protein can be affinity modified to independently increase or attenuate specific binding affinity or avidity to each of the single or multiple cognate binding partners to which it binds. By this method, specific binding to each of multiple cognate binding partners is independently tuned to a particular affinity or avidity.


In some embodiments, the cognate binding partner of an IgSF domain is at least one, and sometimes at least two or three of the counter-structures (cognate binding partners) of the wild-type IgSF domain, such as those listed in Table 1.


The sequence of the IgSF domain, such as mammalian IgSF domain, is affinity-modified by altering its sequence with at least one substitution, addition, or deletion. Alteration of the sequence can occur at the binding site for the cognate binding partner or at an allosteric site. In some embodiments, a nucleic acid encoding an IgSF domain, such as a mammalian IgSF domain, is affinity modified by substitution, addition, deletion, or combinations thereof, of specific and pre-determined nucleotide sites to yield a nucleic acid encoding an immunomodulatory protein of the invention. In some contrasting embodiments, a nucleic acid encoding an IgSF domain, such as a mammalian IgSF domain, is affinity modified by substitution, addition, deletion, or combinations thereof, at random sites within the nucleic acid. In some embodiments, a combination of the two approaches (pre-determined and random) is utilized. In some embodiments, design of the affinity modified IgSF domains is performed in silico.


In some embodiments, the affinity-modified IgSF domain of the immunomodulatory protein contains one or more amino acid substitutions (alternatively, “mutations” or “replacements”) relative to a wild-type or unmodified polypeptide or a portion thereof containing an immunoglobulin superfamily (IgSF) domain. In some embodiments, the IgSF domain is an IgV domain or an IgC domain or specific binding fragment of the IgV domain or the IgC domain. In some embodiments, the immunomodulatory protein comprises an affinity modified IgSF domain that contains an IgV domain or an IgC domain or specific binding fragments thereof in which the at least one of the amino acid substitutions is in the IgV domain or IgC domain or a specific binding fragment thereof. In some embodiments, by virtue of the altered binding activity or affinity, the IgV domain or IgC domain is an affinity-modified IgSF domain.


In some embodiments, the IgSF domain, such as a mammalian IgSF domain, is affinity-modified in sequence with no more than a total of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions, additions, deletions, or combinations thereof. In some embodiments, the IgSF domain, such as a mammalian IgSF domain, is affinity-modified in sequence with at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions, additions, deletions, or combinations thereof. In some embodiments, the IgSF domain, such as a mammalian IgSF domain, is affinity-modified in sequence with between 1 (or 2, 3, 4, 5, 6, 7, 8, or 9) and 10 (or 9, 8, 7, 6, 5, 4, 3, or 2) amino acid substitutions, additions, deletions, or combinations thereof. In some embodiments, the IgSF domain, such as a mammalian IgSF domain, is affinity-modified in sequence with no more than a total of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions. In some embodiments, the IgSF domain, such as a mammalian IgSF domain, is affinity-modified in sequence with at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid substitutions. In some embodiments, the IgSF domain, such as a mammalian IgSF domain, is affinity-modified in sequence with between 1 (or 2, 3, 4, 5, 6, 7, 8, or 9) and 10 (or 9, 8, 7, 6, 5, 4, 3, or 2) amino acid substitutions. In some embodiments, the substitutions are conservative substitutions. In some embodiments, the substitutions are non-conservative. In some embodiments, the substitutions are a combination of conservative and non-conservative substitutions. In some embodiments, the modification in sequence is made at the binding site of the IgSF domain for its cognate binding partner.


In some embodiments, the wild-type or unmodified IgSF domain is a mammalian IgSF domain. In some embodiments, the wild-type or unmodified IgSF domain can be an IgSF domain that includes, but is not limited to, human, mouse, cynomolgus monkey, or rat. In some embodiments, the wild-type or unmodified IgSF domain is human.


In some embodiments, the wild-type or unmodified IgSF domain is or comprises an extracellular domain of an IgSF family member or a portion thereof containing an IgSF domain (e.g. IgV domain or IgC domain). In some cases, the extracellular domain of an unmodified or wild-type IgSF domain can comprise more than one IgSF domain, for example, an IgV domain and an IgC domain. However, the affinity modified IgSF domain need not comprise both the IgV domain and the IgC domain. In some embodiments, the affinity modified IgSF domain comprises or consists essentially of the IgV domain or a specific binding fragment thereof. In some embodiments, the affinity modified IgSF domain comprises or consists essentially of the IgC domain or a specific binding fragment thereof. In some embodiments, the affinity modified IgSF domain comprises the IgV domain or a specific binding fragment thereof, and the IgC domain or a specific binding fragment thereof.


In some embodiments, the one or more amino acid substitutions of the affinity modified IgSF domain can be located in any one or more of the IgSF polypeptide domains. For example, in some embodiments, one or more amino acid substitutions are located in the extracellular domain of the IgSF polypeptide. In some embodiments, one or more amino acid substitutions are located in the IgV domain or specific binding fragment of the IgV domain. In some embodiments, one or more amino acid substitutions are located in the IgC domain or specific binding fragment of the IgC domain.


In some embodiments, the wild-type or unmodified IgSF domain is an IgSF domain or specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS:1-27 and 408 or contained in a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any of SEQ ID NOS:1-27 and 408. In some embodiments, the IgSF domain is an IgV domain or IgC domain contained therein or specific binding fragments thereof. Table 1 identifies the IgSF domains contained in each of SEQ ID NOS: 1-27 and 408.


In some embodiments, the unmodified or wild-type IgSF domain comprises the extracellular domain (ECD) or a portion comprising an IgSF domain (e.g. IgV domain or IgC domain) of an IgSF member, such as a mammalian IgSF member. In some embodiments, the unmodified or wild-type IgSF domain comprises (i) the sequence of amino acids set forth in any of SEQ ID NOS:28-54 and 410, (ii) a sequence of amino acids that has at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to any of SEQ ID NOS: 28-54 and 410, or (iii) is a specific binding fragment of (i) or (ii) comprising an IgV domain or an IgC domain.


In some embodiments, at least one IgSF domain, such as at least one mammalian IgSF domain, of an immunomodulatory protein of the present invention is independently affinity modified in sequence to have at least 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86% 85%, or 80% sequence identity with the corresponding wild-type IgSF domain or specific binding fragment thereof contained in a wild-type or unmodified IgSF protein, such as, but not limited to, those disclosed in Table 1 as SEQ ID NOS: 1-27 and 408.


In some embodiments, the affinity-modified IgSF domain of an immunomodulatory protein provided herein is a specific binding fragment of a wild-type or unmodified IgSF domain contained in a wild-type or unmodified IgSF protein, such as but not limited to, those disclosed in Table 1 in SEQ ID NOS: 1-27 and 408. In some embodiments, the specific binding fragment can have an amino acid length of at least 50 amino acids, such as at least 60, 70, 80, 90, 100, or 110 amino acids. In some embodiments, the specific binding fragment of the IgV domain contains an amino acid sequence that is at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% of the length of the wild-type or unmodified IgV domain. In some embodiments, the specific binding fragment of the IgC domain comprises an amino acid sequence that is at least about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% of the length of the wild-type or unmodified IgC domain. In some embodiments, the specific binding fragment modulates immunological activity. In more specific embodiments, the specific binding fragment of an IgSF domain increases immunological activity. In alternative embodiments, the specific binding fragment decreases immunological activity.


In some embodiments, to determine the percent identity of two nucleic acid sequences or of two amino acids, the sequences are aligned for optimal comparison purposes (e.g., gaps may be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity=# of identical positions/total # of positions (e.g., overlapping positions)×100). In one embodiment the two sequences are the same length. One may manually align the sequences and count the number of identical nucleic acids or amino acids. Alternatively, alignment of two sequences for the determination of percent identity may be accomplished using a mathematical algorithm. Such an algorithm is incorporated into the NBLAST and XBLAST programs. BLAST nucleotide searches may be performed with the NBLAST program, score=100, wordlength=12, to obtain nucleotide sequences homologous to a nucleic acid molecules of the invention. BLAST protein searches may be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to a protein molecule of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST may be utilized. Alternatively, PSI-Blast may be used to perform an iterated search which detects distant relationships between molecules. When utilizing the NBLAST, XBLAST, and Gapped BLAST programs, the default parameters of the respective programs may be used such as those available on the NCBI website. Alternatively, sequence identity may be calculated after the sequences have been aligned e.g. by the BLAST program in the NCBI database. Generally, the default settings with respect to e.g. “scoring matrix” and “gap penalty” may be used for alignment. In the context of the present invention, the BLASTN and PSI BLAST NCBI default settings may be employed.


The means by which the affinity-modified IgSF domains of the immunomodulatory proteins are designed or created is not limited to any particular method. In some embodiments, however, wild-type IgSF domains are mutagenized (site specific, random, or combinations thereof) from wild-type IgSF genetic material and screened for altered binding according to the methods disclosed in the Examples. Methods or mutagenizing nucleic acids is known to those of skill in the art. In some embodiments, the affinity modified IgSF domains are synthesized de novo utilizing protein or nucleic acid sequences available at any number of publicly available databases and then subsequently screened. The National Center for Biotechnology Information provides such information and its website is publicly accessible via the internet as is the UniProtKB database as discussed previously.


In some embodiments, at least one non-affinity modified IgSF domain and/or one affinity modified IgSF domain present in the immunomodulatory protein provided herein specifically binds to at least one cell surface molecular species expressed on mammalian cells forming the immunological synapse (IS). In some embodiments, an immunomodulatory protein provided herein can comprise a plurality of non-affinity modified IgSF domains and/or affinity modified IgSF domains such as 1, 2, 3, 4, 5, or 6 non-affinity modified IgSF and/or affinity modified IgSF domains. One or more of these non-affinity modified IgSF domains and/or affinity modified IgSF domains can independently specifically bind to either one or both of the mammalian cells forming the IS.


Often, the cell surface molecular species to which the affinity-modified IgSF domain of the immunomodulatory protein specifically binds will be the cognate binding partner of the wild type IgSF family member or wild type IgSF domain that has been affinity modified. In some embodiments, the cell surface molecular species is a mammalian IgSF member. In some embodiments, the cell surface molecular species is a human IgSF member. In some embodiments, the cell surface molecular species will be the cell surface cognate binding partners as indicated in Table 1. In some embodiments, the cell surface molecular species will be a viral protein, such as a poliovirus protein, on the cell surface of a mammalian cell such as a human cell.


In some embodiments, at least one non-affinity modified and/or affinity modified IgSF domain of the immunomodulatory protein provided herein binds to at least two or three cell surface molecular species present on mammalian cells forming the IS. The cell surface molecular species to which the non-affinity modified IgSF domains and/or the affinity modified IgSF domains of the immunomodulatory protein specifically bind to can exclusively be on one or the other of the two mammalian cells (i.e. in cis configuration) forming the IS or, alternatively, the cell surface molecular species can be present on both.


In some embodiments, the affinity modified IgSF domain specifically binds to at least two cell surface molecular species wherein one of the molecular species is present on one of the two mammalian cells forming the IS and the other molecular species is present on the second of the two mammalian cells forming the IS. In such embodiments, the cell surface molecular species is not necessarily present solely on one or the other of the two mammalian cells forming the IS (i.e., in a trans configuration) although in some embodiments it is. Thus, embodiments provided herein include those wherein each cell surface molecular species is exclusively on one or the other of the mammalian cells forming the IS (cis configuration) as well as those where the cell surface molecular species to which each affinity modified IgSF binds is present on both of the mammalian cells forming the IS (i.e., cis and trans configuration).


Those of skill will recognize that antigen presenting cells (APCs) and tumor cells form an immunological synapse with lymphocytes. Thus, in some embodiments at least one non-affinity modified IgSF domain and/or at least one affinity modified IgSF domain of the immunomodulatory protein specifically binds to only cell surface molecular species present on a cancer cell, wherein the cancer cell in conjunction with a lymphocyte forms the IS. In other embodiments, at least one non-affinity modified IgSF domain and/or at least one affinity modified IgSF domain of the immunomodulatory protein specifically binds to only cell surface molecular species present on a lymphocyte, wherein the lymphocyte in conjunction with an APC or tumor cell forms the IS. In some embodiments, the non-affinity modified IgSF domain and/or affinity modified IgSF domain bind to cell surface molecular species present on both the target cell (or APC) and the lymphocyte forming the IS.


Embodiments of the invention include those in which an immunomodulatory protein provided herein comprises at least one affinity modified IgSF domain with an amino acid sequence that differs from a wild-type or unmodified IgSF domain (e.g. a mammalian IgSF domain) such that the binding affinity (or avidity if in a multimeric or other relevant structure) of the affinity-modified IgSF domain, under specific binding conditions, to at least one of its cognate binding partners is either increased or decreased relative to the unaltered wild-type or unmodified IgSF domain control. In some embodiments, an affinity modified IgSF domain has a binding affinity for a cognate binding partner that differs from that of a wild-type or unmodified IgSF control sequence as determined by, for example, solid-phase ELISA immunoassays, flow cytometry or Biacore assays. In some embodiments, the affinity modified IgSF domain has an increased binding affinity for one or more cognate binding partners, relative to a wild-type or unmodified IgSF domain. In some embodiments, the affinity modified IgSF domain has a decreased binding affinity for one or more cognate binding partners, relative to a wild-type or unmodified IgSF domain. In some embodiments, the cognate binding partner can be a mammalian protein, such as a human protein or a murine protein.


Binding affinities for each of the cognate binding partners are independent; that is, in some embodiments, an affinity modified IgSF domain has an increased binding affinity for one, two or three different cognate binding partners, and a decreased binding affinity for one, two or three of different cognate binding partners, relative to a wild-type or unmodified polypeptide.


In some embodiments, the immunomodulatory protein provided herein comprises at least one affinity modified domain in which the binding affinity or avidity of the affinity modified IgSF domain is increased at least 10%, 20%, 30%, 40%, 50%, 100%, 200%, 300%, 400%, 500%, 1000%, 5000%, or 10,000% relative to the wild type or unmodified control IgSF domain. In some embodiments, the increase in binding affinity relative to the wild-type or unmodified IgSF domain is more than 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold 40-fold or 50-fold.


In some embodiments, the immunomodulatory protein provided herein comprises at least one affinity modified domain in which the binding affinity or avidity of the affinity modified IgSF domain is decreased at least 10%, and up to 20%, 30%, 40%, 50%, 60%, 70%, 80% or up to 90% relative to the wild-type or unmodified control IgSF domain. In some embodiments, the decrease in binding affinity relative to the wild-type or unmodified IgSF domain is more than 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold 40-fold or 50-fold.


In some embodiments, the immunomodulatory protein provided herein comprises at least one affinity modified domain in which the selectivity of the affinity modified IgSF domain for a particular cognate binding partner is increased compared to the wild-type or unmodified control IgSF domain. In some embodiments, the selectivity is represented as a ratio for binding of the particular cognate binding partner compared to one or more other cognate binding partner. In some embodiments, the selectivity ratio for binding a particular cognate binding partner is greater than or greater than about 1.2-fold, 1.5-fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 20-fold, 30-fold 40-fold or 50-fold. In some embodiments, the selectivity of the affinity modified IgSF domain is increased at least 10%, 20%, 30%, 40%, 50%, 100%, 200%, 300%, 400%, 500%, 1000%, 5000%, or 10,000% relative to the wild type or unmodified control IgSF domain.


In some embodiments, the immunomodulatory protein provided herein comprises at least one affinity modified domain in which its specific binding affinity to a cognate binding partner can be less than or less than about 1×10−5 M, 1×10−6 M, 1×10−7 M, 1×10−8 M, 1×10−9 M, 1×10−10 M or 1×10−11 M, or 1×10−12 M.


In some embodiments, the immunomodulatory protein provided comprises at least two IgSF domains in which at least one of the IgSF domain is affinity modified while in some embodiments both are affinity modified, and wherein at least one of the affinity modified IgSF domains has increased affinity (or avidity) to its cognate binding partner and at least one affinity modified IgSF domain has a decreased affinity (or avidity) to its cognate binding partner. Functionally, and irrespective of whether specific binding to its cognate binding partner is increased or decreased, the immunomodulatory protein comprising one or more affinity-modified IgSF domains acts to enhance or suppress immunological activity of engineered immune cells, such as lymphocytes or antigen presenting cells, relative to engineered immune cells expressing the wild-type, parental molecule under the appropriate assay controls. In some embodiments, an immunomodulatory protein comprising an at least two affinity modified IgSF domains is one in which at least one of the affinity-modified IgSF domains agonizes an activating receptor and at least one affinity modified IgSF domain acts to antagonize an inhibitory receptor. In some embodiments, an enhancement of immunological activity can be an increase of at least 10%, 20%, 30%, 40%, 50%, 75%, 100%, 200%, 300%, 400%, or 500% greater than a non-zero control value such as in a cytotoxic activity assay, an assay for assessing cellular cytokines or a cell proliferation assay. In some embodiments, suppression of immunological activity can be a decrease of at least 10%, and up to 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%.


The affinity modified IgSF domains of the immunomodulatory proteins of the invention can in some embodiments specifically bind competitively to its cognate binding partner. In other embodiments the affinity modified IgSF domains of the present invention specifically bind non-competitively to its cognate binding partner.


In some embodiments, the immunomodulatory protein provided herein contains an IgSF domain that otherwise binds to multiple cell surface molecular species but is affinity modified such that it substantially no longer specifically binds to one of its cognate cell surface molecular species. Thus, in these embodiments the specific binding to one of its cognate cell surface molecular species is reduced to specific binding of no more than 90% of the wild type level, such as no more than 80%, 70%, 60%, 50%, 40%, 30%, 20% or less. In some embodiments, the specific binding to one of its cognate cell surface molecular species is reduced to specific binding of no more than 10% of the wild type level and often no more than 7%, 5%, 3%, 1%, or no detectable or statistically significant specific binding.


In some embodiments, a specific binding site on a mammalian IgSF domain is inactivated or substantially inactivated with respect to at least one of the cell surface molecular species. Thus, for example, if a wild type IgSF domain specifically binds to exactly two cell surface molecular species then in some embodiments it is affinity modified to specifically bind to exactly one cell surface molecular species. And, if a wild type IgSF domain specifically binds to exactly three cell surface molecular species then in some embodiments it is affinity modified to specifically bind to exactly two cell surface molecular species. The IgSF domain that is affinity modified to substantially no longer specifically bind to one of its cognate cell surface molecular species can be an IgSF domain that otherwise specifically binds competitively or non-competitively to its cell surface molecular species. An illustrative example concerns native CD80 (B7-1) which specifically binds cognate binding partners: CD28, PD-L1, and CTLA4. In some embodiments, CD80 can be IgSF affinity modified to increase or attenuate its specific binding to CD28 and/or PD-L1 but not to specifically bind to any physiologically significant extent to CTLA4. The IgSF domain that is affinity modified to substantially no longer specifically bind to one of its cell surface cognate binding partners can be an IgSF domain that otherwise specifically binds competitively or non-competitively to its cognate binding partner. Those of skill will appreciate that a wild-type IgSF domain that competitively binds to two cognate binding partners can nonetheless be inactivated with respect to exactly one of them if, for example, their binding sites are not precisely coextensive but merely overlap such that specific binding of one inhibits binding of the other cognate binding partner and yet both competitive binding sites are distinct.


The non-affinity modified IgSF domains and/or affinity modified IgSF domains of the immunomodulatory proteins provided herein can, in some embodiments, specifically bind competitively to its cognate cell surface molecular species. In other embodiments the non-affinity-modified IgSF domain(s) and/or affinity-modified IgSF domain(s) of the immunomodulatory protein provided herein specifically bind non-competitively to its cognate cell surface molecular species. Any number of the non-affinity modified IgSF domains and/or affinity modified IgSF domains present in the immunomodulatory protein provided herein can specifically bind competitively or non-competitively.


In some embodiments, the immunomodulatory protein provided herein comprises at least two non-affinity modified IgSF domains, or at least one non-affinity modified IgSF domain and at least one affinity modified IgSF domain, or at least two affinity modified IgSF domains wherein one IgSF domain specifically binds competitively and a second IgSF domain binds non-competitively to its cognate cell surface molecular species. More generally, the immunomodulatory protein provided herein can comprise 1, 2, 3, 4, 5, or 6 competitive or 1, 2, 3, 4, 5, or 6 non-competitive binding non-affinity modified IgSF and/or affinity modified IgSF domains or any combination thereof. Thus, the immunomodulatory protein provided herein can have the number of non-competitive and competitive binding IgSF domains, respectively, of: 0 and 1, 0 and 2, 0 and 3, 0 and 4, 1 and 0, 1 and 1, 1 and 2, 1 and 3, 2 and 0, 2 and 1, 2 and 2, 2 and 3, 3 and 0, 3 and 1, 3 and 2, 3 and 3, 4 and 0, 4 and 1, and, 4 and 2.


In some embodiments in which the immunomodulatory protein contains a plurality of IgSF domains, the plurality of non-affinity modified and/or affinity modified IgSF domains of the immunomodulatory protein provided herein need not be covalently linked directly to one another. In some embodiments, an intervening span of one or more amino acid residues indirectly covalently bonds the non-affinity modified and/or affinity modified IgSF domains to each other. The linkage can be via the N-terminal to C-terminal residues.


In some embodiments, the linkage can be made via side chains of amino acid residues that are not located at the N-terminus or C-terminus of the non-affinity modified or affinity-modified IgSF domain. Thus, linkages can be made via terminal or internal amino acid residues or combinations thereof.


The “peptide linkers” that link the non-affinity modified and/or affinity modified IgSF domains can be a single amino acid residue or greater in length. In some embodiments, the peptide linker has at least one amino acid residue but is no more than 20, 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 amino acid residues in length. In some embodiments, the linker is three alanines (AAA). In some embodiments, the linker is (in one-letter amino acid code): GGGGS (“4GS”; SEQ ID NO: 1870) or multimers of the 4GS linker, such as repeats of 2, 3, 4, or 5 4GS linkers, such as set forth in SEQ ID NO: 229 (2×GGGGS) or SEQ ID NO: 228 (3×GGGGS). In some embodiments, the linker (in one-letter amino acid code) is GSGGGGS (SEQ ID NO:1869).


III. Exemplary Affinity-Modified Domains and Immunomodulatory Proteins

In some embodiments, the immunomodulatory protein contains an affinity-modified IgSF domain that has one or more amino acid substitutions in an IgSF domain of a wild-type or unmodified IgSF protein, such as set forth in Table 1 above. In some embodiments, the one or more amino acid substitutions are in the IgV domain or specific binding fragment thereof. In some embodiments, the one or more amino acid substitutions are in the IgC domain or specific binding fragment thereof. In some embodiments, one or more amino acid substitutions are in the IgV domain or a specific binding fragment thereof, and some of the one or more amino acid substitutions are in the IgC domain or a specific binding fragment thereof.


The wild-type or unmodified IgSF domain sequence does not necessarily have to be used as a starting composition to generate variant IgSF domain polypeptides described herein. Therefore, use of the term “modification”, such as “substitution” does not imply that the present embodiments are limited to a particular method of making variant IgSF protein. Variant IgSF polypeptides can be made, for example, by de novo peptide synthesis and thus does not necessarily require a modification, such as a “substitution” in the sense of altering a codon to encode for the modification, e.g. substitution. This principle also extends to the terms “addition” and “deletion” of an amino acid residue which likewise do not imply a particular method of making. The means by which the variant IgSF polypeptides are designed or created is not limited to any particular method. In some embodiments, however, a wild-type or unmodified IgSF polypeptide encoding nucleic acid is mutagenized from wild-type or unmodified genetic material and screened for desired specific binding affinity and/or induction of IFN-gamma expression or other functional activity. In some embodiments, a variant IgSF polypeptide is synthesized de novo utilizing protein or nucleic acid sequences available at any number of publicly available databases and then subsequently screened. The National Center for Biotechnology Information provides such information and its website is publicly accessible via the internet as is the UniProtKB database as discussed previously.


Unless stated otherwise, as indicated throughout the present disclosure, the amino acid substitution(s) are designated by amino acid position number corresponding to the numbering of positions of the unmodified ECD sequences set forth in Table 1.


It is within the level of a skilled artisan to identify the corresponding position of a modification, e.g. amino acid substitution, in an affinity-modified IgSF domain, including portion thereof containing an IgSF domain (e.g. IgV) thereof, such as by alignment of a reference sequence. In the listing of modifications throughout this disclosure, the amino acid position is indicated in the middle, with the corresponding unmodified (e.g. wild-type) amino acid listed before the number and the identified variant amino acid substitution listed after the number. If the modification is a deletion of the position a “del” is indicated and if the modification is an insertion at the position an “ins” is indicated. In some cases, an insertion is listed with the amino acid position indicated in the middle, with the corresponding unmodified (e.g. wild-type) amino acid listed before and after the number and the identified variant amino acid insertion listed after the unmodified (e.g. wild-type) amino acid.


In some embodiments, the affinity-modified IgSF domain has up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid substitutions. The substitutions can be in the IgV domain or the IgC domain. In some embodiments, the affinity modified IgSF domain has up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid substitutions in the IgV domain or specific binding fragment thereof. In some embodiments, the affinity modified IgSF domain has up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid substitutions in the IgC domain or specific binding fragment thereof. In some embodiments, the affinity modified IgSF domain has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the wild-type or unmodified IgSF domain or specific binding fragment thereof, such as an IgSF domain contained in the IgSF protein set forth in any of SEQ ID NOS: 1-27 and 408.


In some embodiments, the immunomodulatory protein contains at least one affinity-modified IgSF domain containing one or more amino acid substitutions in a wild-type or unmodified IgSF domain of a B7 IgSF family member. In some embodiments, the B7 IgSF family member is CD80, CD86 or ICOS Ligand (ICOSL). In some embodiments, the affinity modified IgSF domain of the immunomodulatory protein has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the wild-type or unmodified IgSF domain or specific binding fragment thereof of CD80, CD86 or ICOS Ligand (ICOSL), such as the IgSF domain (e.g. IgV or IgC) contained in the IgSF protein set forth in any of SEQ ID NOS: 1, 2 or 5. Exemplary affinity modified IgSF domains of CD80 are set forth in Table 2 and Table 3. In some embodiments, the affinity modified IgSF domain of CD80 exhibits increased binding affinity or increased binding selectivity to CD28. In some embodiments, the affinity modified IgSF domain of CD80 exhibits increased binding affinity or increased binding selectivity to CTLA-4. Exemplary affinity modified IgSF domains of ICOSL are set forth in Table 4. In some embodiments, the affinity modified IgSF domain of ICOSL exhibits increased binding affinity or increased binding selectivity to CD28 or ICOS. Exemplary affinity modified IgSF domains of CD86 are set forth in Table 5.









TABLE 2







Exemplary variant CD80 polypeptides










ECD
IgV



SEQ
SEQ ID


Mutation(s)
ID NO
NO





Wild-type
28
152, 504,




578


L70Q/A91G
55
153


L70Q/A91G/T130A
56



L70Q/A91G/I118A/T120S/T130A
57



V4M/L70Q/A91G/T120S/T130A
58
154


L70Q/A91G/T120S/T130A
59



V20L/L70Q/A91S/T120S/T130A
60
155


S44P/L70Q/A91G/T130A
61
156


L70Q/A91G/E117G/T120S/T130A
62



A91G/T120S/T130A
63
157


L70R/A91G/T120S/T130A
64
158


L70Q/E81A/A91G/T120S/I127T/T130A
65
159


L70Q/Y87N/A91G/T130A
66
160


T28S/L70Q/A91G/E95K/T120S/T130A
67
161


N63S/L70Q/A91G/T120S/T130A
68
162


K36E/I67T/L70Q/A91G/T120S/T130A/N152T
69
163


E52G/L70Q/A91G/T120S/T130A
70
164


K37E/F59S/L70Q/A91G/T120S/T130A
71
165


A91G/S103P
72



K89E/T130A
73
166


A91G
74



D60V/A91G/T120S/T130A
75
167


K54M/A91G/T120S
76
168


M38T/L70Q/E77G/A91G/T120S/T130A/N152T
77
169


R29H/E52G/L70R/E88G/A91G/T130A
78
170


Y31H/T41G/L70Q/A91G/T120S/T130A
79
171


V68A/T110A
80
172


S66H/D90G/T110A/F116L
81
173


R29H/E52G/T120S/T130A
82
174


A91G/L102S
83



I67T/L70Q/A91G/T120S
84
175


L70Q/A91G/T110A/T120S/T130A
85



M38V/T41D/M43I/W50G/D76G/V83A/K89E/
86
176


T120S/T130A




V22A/L70Q/S121P
87
177


A12V/S15F/Y31H/T41G/T130A/P137L/N152T
88
178


I67F/L70R/E88G/A91G/T120S/T130A
89
179


E24G/L25P/L70Q/T120S
90
180


A91G/F92L/F108L/T120S
91
181


R29D/Y31L/Q33H/K36G/M38I/T41A/M43R/
92
182


M47T/E81V/L85R/K89N/A91T/F92P/




K93V/R94L/I118T/N149S




R29D/Y31L/Q33H/K36G/M38I/T41A/M43R/
93



M47T/E81V/L85R/K89N/A91T/F92P/




K93V/R94L/N144S/N149S




R29D/Y31L/Q33H/K36G/M38I/T41A/M42T/
94
183


M43R/M47T/E81V/L85R/K89N/A91T/




F92P/K93V/R94L/L148S/N149S




E24G/R29D/Y31L/Q33H/K36G/M38I/T41A/
95
184


M43R/M47T/F59L/E81V/L85R/K89N/A91T/




F92P/K93V/R94L/H96R/N149S/C182S




R29D/Y31L/Q33H/K36G/M38I/T41A/M43R/
96



M47T/E81V/L85R/K89N/A91T/




F92P/K93V/R94L/N149S




R29V/M43Q/E81R/L85I/K89R/D90L/A91E/
97
185


F92N/K93Q/R94G




T41I/A91G
98
186


K89R/D90K/A91G/F92Y/K93R/N122S/N177S
99
187


K89R/D90K/A91G/F92Y/K93R
100



K36G/K37Q/M38I/F59L/E81V/L85R/K89N/
101
188


A91T/F92P/K93V/R94L/E99G/T130A/N149S




E88D/K89R/D90K/A91G/F92Y/K93R
102
189


K36G/K37Q/M38I/L40M
103
190


K36G
104
191


R29H/Y31H/T41G/Y87N/E88G/K89E/D90N/
105
192


A91G/P109S




A12T/H18L/M43V/F59L/E77K/P109S/I118T
106
193


R29V/Y31F/K36G/M38L/M43Q/E81R/V83I/
107
194


L85I/K89R/D90L/A91E/F92N/K93Q/R94G




V68M/L70P/L72P/K86E
108
195
















TABLE 3







Exemplary variant CD80 polypeptides










ECD
IgV



SEQ
SEQ ID


Mutation(s)
ID NO
NO





Wild-type

152, 504,




578


L70P
431
505, 579


I30F/L70P
432
506, 580


Q27H/T41S/A71D
433
507, 581


I30T/L70R
434
508, 582


T13R/C16R/L70Q/A71D
435
509, 583


T57I
436
510, 584


M43I/C82R
437
511, 585


V22L/M38V/M47T/A71D/L85M
438
512, 586


I30V/T57I/L70P/A71D/A91T
439
513, 587


V22I/L70M/A71D
440
514, 588


N55D/L70P/E77G
441
515, 589


T57A/I69T
442
516, 590


N55D/K86M
443
517, 591


L72P/T79I
444
518, 592


L70P/F92S
445
519, 593


T79P
446
520, 594


E35D/M47I/L65P/D90N
447
521, 595


L25S/E35D/M47I/D90N
448
522, 596


A71D
450
524, 598


E81K/A91S
452
526, 600


A12V/M47V/L70M
453
527, 601


K34E/T41A/L72V
454
528, 602


T41S/A71D/V84A
455
529, 603


E35D/A71D
456
530, 604


E35D/M47I
457
531, 605


K36R/G78A
458
532, 606


Q33E/T41A
459
533, 607


M47V/N48H
460
534, 608


M47L/V68A
461
535, 609


S44P/A71D
462
536, 610


Q27H/M43I/A71D/R73S
463
537, 611


E35D/T57I/L70Q/A71D
465
539, 613


M47I/E88D
466
540, 614


M42I/I61V/A71D
467
541, 615


P51A/A71D
468
542, 616


H18Y/M47I/T57I/A71G
469
543, 617


V20I/M47V/T57I/V84I
470
544, 618


V20I/M47V/A71D
471
545, 619


A71D/L72V/E95K
472
546, 620


V22L/E35G/A71D/L72P
473
547, 621


E35D/A71D
474
548, 622


E35D/I67L/A71D
475
549, 623


Q27H/E35G/A71D/L72P/T79I
476
550, 624


T13R/M42V/M47I/A71D
477
551, 625


E35D
478
552, 626


E35D/M47I/L70M
479
553, 627


E35D/A71D/L72V
480
554, 628


E35D/M43L/L70M
481
555, 629


A26P/E35D/M43I/L85Q/E88D
482
556, 630


E35D/D46V/L85Q
483
557, 631


Q27L/E35D/M47I/T57I/L70Q/E88D
484
558, 632


M47V/I69F/A71D/V83I
485
559, 633


E35D/T57A/A71D/L85Q
486
560, 634


H18Y/A26T/E35D/A71D/L85Q
487
561, 635


E35D/M47L
488
562, 636


E23D/M42V/M43I/I58V/L70R
489
563, 637


V68M/L70M/A71D/E95K
490
564, 638


N55I/T57I/I69F
491
565, 639


E35D/M43I/A71D
492
566, 640


T41S/T57I/L70R
493
567, 641


H18Y/A71D/L72P/E88V
494
568, 642


V20I/A71D
495
569, 643


E23G/A26S/E35D/T62N/A71D/L72V/L85M
496
570, 644


A12T/E24D/E35D/D46V/I61V/L72P/E95V
497
571, 645


V22L/E35D/M43L/A71G/D76H
498
572, 646


E35G/K54E/A71D/L72P
499
573, 647


L70Q/A71D
500
574, 648


A26E/E35D/M47L/L85Q
501
575, 649


D46E/A71D
502
576, 650


Y31H/E35D/T41S/V68L/K93R/R94W
503
577, 651
















TABLE 4







Exemplary variant ICOSL polypeptides










ECD
IgV



SEQ ID
SEQ ID


Mutation(s)
NO
NO












Wild-type
32
196


N52S
109
197


N52H
110
198


N52D
111
199


N52Y/N57Y/F138L/L203P
112
200


N52H/N57Y/Q100P
113
201


N52S/Y146C/Y152C
114



N52H/C198R
115



N52H/C140D/T225A
116



N52H/C198R/T225A
117



N52H/K92R
118
202


N52H/S99G
119
203


N52Y
120
204


N57Y
121
205


N57Y/Q100P
122
206


N52S/S130G/Y152C
123



N52S/Y152C
124



N52S/C198R
125



N52Y/N57Y/Y152C
126



N52Y/N57Y/129P/C198R
127



N52H/L161P/C198R
128



N52S/T113E
129



S54A
130
207


N52D/S54P
131
208


N52K/L208P
132
209


N52S/Y152H
133



N52D/V151A
134



N52H/I143T
135



N52S/L80P
136
210


F120S/Y152H/N201S
137



N52S/R75Q/L203P
138
211


N52S/D158G
139



N52D/Q133H
140



N52S/N57Y/H94D/L96F/L98F/Q100R
141
212


N52S/N57Y/H94D/L96F/L98F/Q100R/G103E/F120S
142
213


N52S/G103E
239
240


N52H/C140delta/T225A
1563



N52S/S54P
1564
1565
















TABLE 5







Exemplary variant CD86 polypeptides












ECD
IgV




SEQ ID
SEQ ID



Mutation(s)
NO
NO














Wild-type
29
220



Q35H/H90L/Q102H
148
221



Q35H
149
222



H90L
150
223



Q102H
151
224









In some embodiments, the affinity-modified IgSF domain binds, in some cases with higher binding affinity or selectivity, to an inhibitory receptor. In some embodiments, the affinity-modified IgSF domain contains one or more amino acid modifications (e.g. substitutions, deletions or additions) in a wild-type or unmodified IgSF domain (e.g. IgV) of an IgSF family member that binds to an inhibitory receptor. Exemplary of such inhibitory receptors are PD-1, CTLA-4, LAGS, TIGIT, TIM-3, or BTLA.


In some embodiments, the immunomodulatory protein contains at least one affinity modified IgSF domain containing one or more amino acid substitutions in a wild-type or unmodified IgSF domain of a poliovirus receptor IgSF family member. In some embodiments, the poliovirus IgSF family member is CD155 (PVR) or CD122 (PRR-2). In some embodiments, the affinity modified IgSF domain of the immunomodulatory protein has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the wild-type or unmodified IgSF domain or specific binding fragment thereof of CD155 or CD112, such as an IgSF domain (e.g. IgV or IgC) contained in the IgSF protein set forth in any of SEQ ID NO:20 or 21. Exemplary affinity modified IgSF domains of CD155 are set forth in Table 6. Exemplary affinity modified IgSF domains of CD112 are set forth in Table 7. In some embodiments, the affinity modified IgSF domain of CD155 or CD122 exhibits increased binding affinity or increased binding selectivity to TIGIT.









TABLE 6







Exemplary variant CD155 polypeptides










ECD




SEQ




ID
IgV SEQ


Mutation(s)
NO
ID NO












Wild-type
47
241, 652


P18S, P64S, F91S
242
264, 653


P18S, F91S, L104P
243
265, 654


L44P
244
266, 655


A56V
245
267, 656


P18L, L79V, F91S
246
268, 657


P18S, F91S
247
269, 658


P18T, F91S
248
270, 659


P18T, S42P, F91S
249
271, 660


G7E, P18T, Y30C, F91S
250
272, 661


P18T, F91S, G111D
251
273, 662


P18S, F91P
252
274, 663


P18T, F91S, F108L
253
275, 664


P18T, T45A, F91S
255
277, 665


P18T, F91S, R94H
256
278, 666


P18S, Y30C, F91S
257
279, 667


A81V, L83P
258
280, 668


L88P
259
281, 669


R94H
260
282, 670


A13E, P18S, A56V, F91S
261
283, 671


P18T, F91S, V115A
262
284, 672


P18T, Q60K
263
285, 673


S52M
674
771, 868


T45Q, S52L, L104E, G111R
675
772, 869


S42G
676
773, 870


Q62F
677
774, 871


S52Q
678
775, 872


S42A, L104Q, G111R
679
776, 873


S42A, S52Q, L104Q, G111R
680
777, 874


S52W, L104E
681
778, 875


S42C
682
779, 876


S52W
683
780, 877


S52M, L104Q
684
781, 878


S42L, S52L, Q62F, L104Q
685
782, 879


S42W
686
783, 880


S42Q
687
784, 881


S52L
688
785, 882


S52R
689
786, 883


L104E
690
787, 884


G111R
691
788, 885


S52E
692
789, 886


Q62Y
693
790, 887


T45Q, S52M, L104E
694
791, 888


S42N, L104Q, G111R
695
792, 889


S52M, V57L
696
793, 890


S42N, S52Q, Q62F
697
794, 891


S42A, S52L, L104E, G111R
698
795, 892


S42W, S52Q, V57L, Q62Y
699
796, 893


L104Q
700
797, 894


S42L, S52Q, L104E
701
798, 895


S42C, S52L
702
799, 896


S42W, S52R, Q62Y, L104Q
703
800, 897


T45Q, S52R, L104E
704
801, 898


S52R, Q62F, L104Q, G111R
705
802, 899


T45Q, S52L, V57L, L104E
706
803, 900


S52M, Q62Y
707
804, 901


Q62F, L104E, G111R
708
805, 902


T45Q, S52Q
709
806, 903


S52L, L104E
710
807, 904


S42V, S52E
711
808, 905


T45Q, S52R, G111R
712
809, 906


S42G, S52Q, L104E, G111R
713
810, 907


S42N, S52E, V57L, L104E
714
811, 908


S42C, S52M, Q62F
715
812, 909


S42L
716
813, 910


S42A
717
814, 911


S42G, S52L, Q62F, L104Q
718
815, 912


S42N
719
816, 913


P18T, S65A, S67V, F91S
720
817, 914


P18F, T39A, T45Q, T61R, S65N, S67L, E73G, R78G
721
818, 915


P18T, T45Q, T61R, S65N, S67L
722
819, 916


P18F, S65A, S67V, F91S
723
820, 917


P18F, T45Q, T61R, S65N, S67L, F91S, L104P
724
821, 918


P18S, L79P, L104M
725
822, 919


P18S, L104M
726
823, 920


L79P, L104M
727
824, 921


P18T, T45Q, L79P
728
825, 922


P18T, T45Q, T61R, S65H, S67H
729
826, 923


P18T, A81E
730
827, 924


P18S, D23Y, E37P, S52G, Q62M, G80S, A81P, G99Y, S112N
731
828, 925


A13R, D23Y, E37P, S42P, Q62Y, A81E
732
829, 926


A13R, D23Y, E37P, G99Y, S112N
733
830, 927


A13R, D23Y, E37P, Q62M, A77V, G80S, A81P, G99Y
734
831, 928


P18L, E37S, Q62M, G80S, A81P, G99Y, S112N
735
832, 929


P18S, L104T
736
833, 930


P18S, Q62H, L79Q, F91S
737
834, 931


T45Q, S52K, Q62F, L104Q, G111R
738
835, 932


T45Q, S52Q, Q62Y, L104Q, G111R
739
836, 933


T45Q, S52Q, Q62Y, L104E, G111R
740
837, 934


V57A, T61M, S65W, S67A, E96D, L104T
741
838, 935


P18L, V57T, T61S, S65Y, S67A, L104T
742
839, 936


P18T, T45Q
743
840, 937


P18L, V57A, T61M, S65W, S67A, L104T
744
841, 938


T61M, S65W, S67A, L104T
745
842, 939


P18S, V41A, S42G, T45G, L104N
746
843, 940


P18H, S42G, T45I, S52T, G53R, S54H, V57L, H59E, T61S, S65D,
747
844, 941


E68G, L104N




P18S, S42G, T45V, F58L, S67W, L104N
748
845, 942


P18S, T45I, L104N
749
846, 943


P18S, S42G, T45G, L104N, V106A
750
847, 944


P18H, H40R, S42G, T45I, S52T, G53R, S54H, V57L, H59E, T61S,
751
848, 945


S65D, E68G, L104Y, V106L, F108H




E37V, S42G, T45G, L104N
752
849, 946


P18S, T45Q, L79P, L104T
753
850, 947


P18L, Q62R
754
851, 948


A13R, D23Y, E37P, S42L, S52G, Q62Y, A81E
755
852, 949


P18L, H49R, L104T, D116N
756
853, 950


A13R, D23Y, E37P, Q62M, G80S, A81P, L104T
757
854, 951


S65T, L104T
758
855, 952


A13R, D23Y, E37P, S52G, V57A, Q62M, K70E, L104T
759
856, 953


P18L, A47V, Q62Y, E73D, L104T
760
857, 954


H40T, V41M, A47V, S52Q, Q62L, S65T, E73R, D97G, E98S, L104T,
761
858, 955


D116N




P18L, S42P, T45Q, T61G, S65H, S67E, L104T, D116N
762
859, 956


P18S, H40T, V41M, A47V, S52Q, Q62L, S65T, E73R, L104M, V106A
763
860, 957


H40T, V41M, A47V, S52Q, Q62L, S65T, E68G, E73R, D97G, E98S,
764
861, 958


L104T




T45Q, S52E, L104E
765
862, 959


T45Q, S52E, Q62F, L104E
766
863, 960


P18F, T26M, L44V, Q62K, L79P, F91S, L104M, G111D
767
864, 961


P18S, T45S, T61K, S65W, S67A, F91S, G111R
768
865, 962


P18S, L79P, L104M, T107M
769
866, 963


P18S, S65W, S67A, M90V, V95A, L104Q, G111R
770
867, 964


P18S, A47G, L79P, F91S, L104M, T107A, R113W
1701
1655, 1678


P18T, D23G, S24A, N35D, H49L, L79P, F91S, L104M, G111R
1702
1656, 1679


V9L, P18S, Q60R, V75L, L79P, R89K, F91S, L104E, G111R
1703
1657, 1680


P18S, H49R, E73D, L79P, N85D, F91S, V95A, L104M, G111R
1704
1658, 1681


V11A, P18S, L79P, F91S, L104M, G111R
1705
1659, 1682


V11A, P18S, S54R, Q60P, Q62K, L79P, N85D, F91S, T107M
1706
1660, 1683


P18T, S52P, S65A, S67V, L79P, F91S, L104M, G111R
1707
1661, 1684


P18T, M36T, L79P, F91S, G111R
1708
1662, 1685


D8G, P18S, M36I, V38A, H49Q, A76E, F91S, L104M, T107A, R113W
1709
1663, 1686


P18S, S52P, S65A, S67V, L79P, F91S, L104M, T107S, R113W
1710
1664, 1687


T15I, P18T, L79P, F91S, L104M, G111R
1711
1665, 1688


P18F, T26M, L44V, Q62K, L79P, E82D, F91S, L104M, G111D
1712
1666, 1689


P18T, E37G, G53R, Q62K, L79P, F91S, E98D, L104M, T107M
1713
1667, 1690


P18L, K70E, L79P, F91S, V95A, G111R
1714
1668, 1691


V9I, Q12K, P18F, S65A, S67V, L79P, L104T, G111R, S112I
1715
1669, 1692


P18F, S65A, S67V, F91S, L104M, G111R
1716
1670, 1693


V9I, V10I, P18S, F20S, T45A, L79P, F91S, L104M, F108Y, G111R,
1717
1671, 1694


S112V




V9L, P18L, L79P, M90I, F91S, T102S, L104M, G111R
1718
1672, 1695


P18C, T26M, L44V, M55I, Q62K, L79P, F91S, L104M, T107M
1719
1673, 1696


V9I, P18T, D23G, L79P, F91S, G111R
1720
1674, 1697


P18F, L79P, M90L, F91S, V95A, L104M, G111R
1721
1675, 1698


P18T, M36T, S65A, S67E, L79Q, A81T, F91S, G111R
1722
1676, 1699


V9L, P18T, Q62R, L79P, F91S, L104M, G111R
1723
1677, 1700


P18S, S65W, S67A, L104Q, G111R
1724
1725, 1726


P18T, G19D, M36T, S54N, L79P, L83Q, F91S, T107M, F108Y
1727
1773, 1819


V9L, P18L, M55V, S69L, L79P, A81E, F91S, T107M
1728
1774, 1820


P18F, H40Q, T61K, Q62K, L79P, F91S, L104M, T107V
1729
1775, 1821


P18S, Q32R, Q62K, R78G, L79P, F91S, T107A, R113W
1730
1776, 1822


Q12H, P18T, L21S, G22S, V57A, Q62R, L79P, F91S, T107M
1731
1777, 1823


V9I, P18S, S24P, H49Q, F58Y, Q60R, Q62K, L79P, F91S, T107M
1732
1778, 1824


P18T, W46C, H49R, S65A, S67V, A76T, L79P, S87T, L104M
1733
1779, 1825


P18S, S42T, E51G, L79P, F91S, G92W, T107M
1734
1780, 1826


V10F, T15S, P18L, R48Q, L79P, F91S, T107M, V115M
1735
1781, 1827


P18S, L21M, Y30F, N35D, R84W, F91S, T107M, D116G
1736
1782, 1828


P18F, E51V, S54G, Q60R, L79Q, E82G, S87T, M90I, F91S, G92R,
1737
1783, 1829


T107M




Q16H, P18F, F91S, T107M
1738
1784, 1830


P18T, D23G, Q60R, S67L, L79P, F91S, T107M, V115A
1739
1785, 1831


D8G, V9I, V11A, P18T, T26M, S52P, L79P, F91S, G92A, T107L,
1740
1786, 1832


V115A




V9I, P18F, A47E, G50S, E68G, L79P, F91S, T107M
1741
1787, 1833


P18S, M55I, Q62K, S69P, L79P, F91S, T107M
1742
1788, 1834


P18T, T39S, S52P, S54R, L79P, F91S, T107M
1743
1789, 1835


P18S, D23N, L79P, F91S, T107M, S114N
1744
1790, 1836


P18S, P34S, E51V, L79P, F91S, G111R
1745
1791, 1837


P18S, H59N, V75A, L79P, A81T, F91S, L104M, T107M
1746
1792, 1838


P18S, W46R, E68D, L79P, F91S, T107M, R113G
1747
1793, 1839


V9L, P18F, T45A, S65A, S67V, R78K, L79V, F91S, T107M, S114T
1748
1794, 1840


P18T, M55L, T61R, L79P, F91S, V106I, T107M
1749
1795, 1841


T15I, P18S, V33M, N35F, T39S, M55L, R78S, L79P, F91S, T107M
1750
1796, 1842


P18S, Q62K, K70E, L79P, F91S, G92E, R113W
1751
1797, 1843


P18F, F20I, T26M, A47V, E51K, L79P, F91S
1752
1798, 1844


P18T, D23A, Q60H, L79P, M90V, F91S, T107M
1753
1799, 1845


P18S, D23G, C29R, N35D, E37G, M55I, Q62K, S65A, S67G, R78G,
1754
1800, 1846


L79P, F91S, L104M, T107M, Q110R




A13E, P18S, M36R, Q62K, S67T, L79P, N85D, F91S, T107M
1755
1801, 1847


V9I, P18T, H49R, L79P, N85D, F91S, L104T, T107M
1756
1802, 1848


V9A, P18F, T61S, Q62L, L79P, F91S, G111R
1757
1803, 1849


D8E, P18T, T61A, L79P, F91S, T107M
1758
1804, 1850


P18S, V41A, H49R, S54C, L79S, N85Y, L88P, F91S, L104M, T107M
1759
1805, 1851


V11E, P18H, F20Y, V25E, N35S, H49R, L79P, F91S, T107M, G111R
1760
1806, 1852


V11A, P18F, D23A, L79P, G80D, V95A, T107M
1761
1807, 1853


P18S, K70R, L79P, F91S, G111R
1762
1808, 1854


V9L, V11M, P18S, N35S, S54G, Q62K, L79P, L104M, T107M,
1763
1809, 1855


V115M




V9L, P18Y, V25A, V38G, M55V, A77T, L79P, M90I, F91S, L104M
1764
1810, 1856


V10G, P18T, L72Q, L79P, F91S, T107M
1765
1811, 1857


P18S, H59R, A76G, R78S, L79P
1766
1812, 1858


V9A, P18S, M36T, S65G, L79P, F91S, L104T, G111R, S112I
1767
1813, 1859


P18T, S52A, V57A, Q60R, Q62K, S65C, L79P, F91T, N100Y, T107M
1768
1814, 1860


V11A, P18F, N35D, A47E, Q62K, L79P, F91S, G99D, T107M, S114N
1769
1815, 1861


V11A, P18T, N35S, L79P, S87T, F91S
1770
1816, 1862


V9D, V11M, Q12L, P18S, E37V, M55I, Q60R, K70Q, L79P, F91S,
1771
1817, 1863


L104M, T107M




T15S, P18S, Y30H, Q32L, Q62R, L79P, F91S, T107M
1772
1818, 1864
















TABLE 7







Exemplary variant CD112 polypeptides










ECD




SEQ ID
IgV SEQ


Mutation(s)
NO
ID NO












Wild-type
48
286, 965


Y33H, A112V, G117D
287
334, 966


V19A, Y33H, S64G, S80G, G98S, N106Y,
288
335, 967


A112V




L32P, A112V
289
336, 968


A95V, A112I
290
337, 969


P28S, A112V
291
338, 970


P27A, T38N, V101A, A112V
292
339, 971


S118F
293
340, 972


R12W, H48Y, F54S, S118F
294
341, 973


R12W, Q79R, S118F
295
342, 974


T113S, S118Y
296
343, 975


S118Y
297
344, 976


N106I, S118Y
298
345, 977


N106I, S118F
299
346, 978


A95T, L96P, S118Y
300
347, 979


Y33H, P67S, N106Y, A112V
301
348, 980


N106Y, A112V
302
349, 981


T18S, Y33H, A112V
303
350, 982


P9S, Y33H, N47S, A112V
304
351, 983


P42S, P67H, A112V
305
352, 984


P27L, L32P, P42S, A112V
306
353, 985


G98D, A112V
307
354, 986


Y33H, S35P, N106Y, A112V
308
355, 987


L32P, P42S, T100A, A112V
309
356, 988


P27S, P45S, N106I, A112V
310
357, 989


Y33H, N47K, A112V
311
358, 990


Y33H, N106Y, A112V
312
359, 991


K78R, D84G, A112V, F114S
313
360, 992


Y33H, N47K, F54L, A112V
314
361, 993


Y33H, A112V
315
362, 994


A95V, A112V
316
363, 995


R12W, A112V
317
364, 996


R12W, P27S, A112V
318
365, 997


Y33H, V51M, A112V
319
366, 998


Y33H, A112V, S118T
320
367, 999


Y33H, V101A, A112V, P115S
321
 368, 1000


H24R, T38N, D43G, A112V
322
 369, 1001


A112V
323
 370, 1002


P27A, A112V
324
 371, 1003


A112V, S118T
325
 372, 1004


R12W, A112V, M122I
326
 373, 1005


Q83K, N106Y, A112V
327
 374, 1006


R12W, P27S, A112V, S118T
328
 375, 1007


P28S, Y33H, A112V
329
 376, 1008


P27S, Q90R, A112V
330
 377, 1009


L15V, P27A, A112V, S118T
331
 378, 1010


Y33H, N106Y, T108I, A112V
332
 379, 1011


Y33H, P56L, V75M, V101M, A112V
333
 380, 1012


N47K, Q79R, S118F
1013
1054, 1095


Q40R, P60T, A112V, S118T
1014
1055, 1096


F114Y, S118F
1015
1056, 1097


Y33H, K78R, S118Y
1016
1057, 1098


R12W, A46T, K66M, Q79R, N106I,
1017
1058, 1099


T113A, S118F




Y33H, A112V, S118F
1018
1059, 1100


R12W, Y33H, N106I, S118F
1019
1060, 1101


L15V, Q90R, S118F
1020
1061, 1102


N47K, D84G, N106I, S118Y
1021
1062, 1103


L32P, S118F
1022
1063, 1104


Y33H, Q79R, A112V, S118Y
1023
1064, 1105


T18A, N106I, S118T
1024
1065, 1106


L15V, Y33H, N106Y, A112V, S118F
1025
1066, 1107


V37M, S118F
1026
1067, 1108


N47K, A112V, S118Y
1027
1068, 1109


A46T, A112V
1028
1069, 1110


P28S, Y33H, N106I, S118Y
1029
1070, 1111


P30S, Y33H, N47K, V75M, Q79R,
1030
1071, 1112


N106I, S118Y




V19A, N47K, N106Y, K116E, S118Y
1031
1072, 1113


Q79R, T85A, A112V, S118Y
1032
1073, 1114


V101M, N106I, S118Y
1033
1074, 1115


Y33H, Q79R, N106I, A112V, S118T
1034
1075, 1116


Q79R, A112V
1035
1076, 1117


Y33H, A46T, Q79R, N106I, S118F
1036
1077, 1118


A112V, G121S
1037
1078, 1119


Y33H, Q79R, N106I, S118Y
1038
1079, 1120


Y33H, N106I, A112V
1039
1080, 1121


Y33H, A46T, V101M, A112V, S118T
1040
1081, 1122


L32P, L99M, N106I, S118F
1041
1082, 1123


L32P, T108A, S118F
1042
1083, 1124


R12W, Q79R, A112V
1043
1084, 1125


Y33H, N106Y, E110G, A112V
1044
1085, 1126


Y33H, N106I, S118Y
1045
1086, 1127


Q79R, S118F
1046
1087, 1128


Y33H, Q79R, G98D, V101M, A112V
1047
1088, 1129


N47K, T81S, V101M, A112V, S118F
1048
1089, 1130


G82S, S118Y
1049
1090, 1131


Y33H, A112V, S118Y
1050
1091, 1132


Y33H, N47K, Q79R, N106Y, A112V
1051
1092, 1133


Y33H, S118T
1052
1093, 1134


R12W, Y33H, Q79R, V101M, A112V
1053
1094, 1135


Y33H, Q83K, A112V, S118T
1583
1607, 1631


V29M, Y33H, N106I, S118F
1584
1608, 1632


Y33H, A46T, A112V
1585
1609, 1633


Y33H, Q79R, S118F
1586
1610, 1634


Y33H, N47K, F74L, S118F
1587
1611, 1635


R12W, V101M, N106I, S118Y
1588
1612, 1636


A46T, V101A, N106I, S118Y
1589
1613, 1637


N106Y, A112V, S118T
1590
1614, 1638


S76P, T81I, V101M, N106Y, A112V, S118F
1591
1615, 1639


P9R, L21V, P22L, I34M, S69F, F74L, A87V,
1592
1616, 1640


A112V, L125A




Y33H, V101M, A112V
1593
1617, 1641


V29A, L32P, S118F
1594
1618, 1642


Y33H, V101M, N106I, A112V
1595
1619, 1643


R12W, Y33H, N47K, Q79R, S118Y
1596
1620, 1644


Y33H, A46T, A112V, S118T
1597
1621, 1645


Y33H, A112V, F114L, S118T
1598
1622, 1646


Y33H, T38A, A46T, V101M, A112V
1599
1623, 1647


P28S, Y33H, S69P, N106I, A112V, S118Y
1600
1624, 1648


Y33H, P42L, N47K, V101M, A112V
1601
1625, 1649


Y33H, N47K, F74S, Q83K, N106I, F111L,
1602
1626, 1650


A112V, S118T




Y33H, A112V, S118T, V119A
1603
1627, 1651


Y33H, N106I, A112V, S118F
1604
1628, 1652


Y33H, K66M, S118F, W124L
1605
1629, 1653


N106I, A112V
1606
1630, 1654









In some embodiments, the immunomodulatory protein contains at least one affinity modified IgSF domain containing one or more amino acid substitutions in a wild-type or unmodified IgSF domain of PD-L1 or PD-L2. In some embodiments, the affinity modified IgSF domain of the immunomodulatory protein has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the wild-type or unmodified IgSF domain or specific binding fragment thereof of PD-L1 or PD-L2, such as an IgSF domain (e.g. IgV or IgC) contained in the IgSF protein set forth in SEQ ID NO:3 or 4. Exemplary affinity modified IgSF domains of PD-L1 are set forth in Table 8. Exemplary affinity modified IgSF domains of PD-L2 are set forth in Table 9. In some embodiments, the affinity modified IgSF domain of PD-L1 or PD-L2 exhibits increased binding affinity or increased binding selectivity to PD-1.









TABLE 8







Exemplary variant PD-L1 polypeptides











IgV



ECD SEQ
SEQ ID


Mutation(s)
ID NO
NO





Wild-type
 30, 1874
1136, 1332


K28N/M41V/N45T/H51N/K57E
1137, 2087
1202, 1267


I20L/I36T/N45D/I47T
1138, 2088
1203, 1268


I20L/M41K/K44E
1139, 2089
1204, 1269


P6S/N45T/N78I/I83T
1140, 2090
1205, 1270


N78I
1141, 2091
1206, 1271


M41K/N78I
1142, 2092
1207, 1272


N45T/N78I
1143, 2093
1208, 1273


I20L/N45T
1144, 2094
1209, 1274


N45T
1145, 2095
1210, 1275


M41K
1146, 2096
1211, 1276


I20L/I36T/N45D
1147, 2097
1212, 1277


N17D/N45T/V50A/D72G
1148, 2098
1213, 1278


I20L/F49S
1149, 2099
1214, 1279


N45T/V50A
1150, 2100
1215, 1280


I20L/N45T/N78I
1151, 2101
1216, 1281


I20L/N45T/V50A
1152, 2102
1217, 1282


M41V/N45T
1153, 2103
1218, 1283


M41K/N45T
1154, 2104
1219, 1284


A33D/S75P/D85E
1155, 2105
1220, 1285


M18I/M41K/D43G/H51R/N78I
1156, 2106
1221, 1286


V11E/I20L/I36T/N45D/H60R/S75P
1157, 2107
1222, 1287


A33D/V50A
1158, 2108
1223, 1288


S16G/A33D/K71E/S75P
1159, 2109
1224, 1289


E27G/N45T/M97I
1160, 2110
1225, 1290


E27G/N45T/K57R
1161, 2111
1226, 1291


A33D/E53V
1162, 2112
1227, 1292


D43G/N45D/V58A
1163, 2113
1228, 1293


E40G/D43V/N45T/V50A
1164, 2114
1229, 1294


Y14S/K28E/N45T
1165, 2115
1230, 1295


A33D/N78S
1166, 2116
1231, 1296


A33D/N78I
1167, 2117
1232, 1297


A33D/N45T
1168, 2118
1233, 1298


A33D/N45T/N78I
1169, 2119
1234, 1299


E27G/N45T/V50A
1170, 2120
1235, 1300


N45T/V50A/N78S
1171, 2121
1236, 1301


I20L/N45T/V110M
1172, 2122
1237, 1302


I20L/I36T/N45T/V50A
1173, 2123
1238, 1303


N45T/L74P/S75P
1174, 2124
1239, 1304


N45T/S75P
1175, 2125
1240, 1305


S75P/K106R
1176, 2126
1241, 1306


S75P
1177, 2127
1242, 1307


A33D/S75P
1178, 2128
1243, 1308


A33D/S75P/D104G
1179, 2129
1244, 1309


A33D/S75P
1180, 2130
1245, 1310


I20L/E27G/N45T/V50A
1181, 2131
1246, 1311


I20L/E27G/D43G/N45D/V58A/N78I
1182, 2132
1247, 1312


I20L/D43G/N45D/V58A/N78I
1183, 2133
1248, 1313


I20L/A33D/D43G/N45D/V58A/N78I
1184, 2134
1249, 1314


I20L/D43G/N45D/N78I
1185, 2135
1250, 1315


E27G/N45T/V50A/N78I
1186, 2136
1251, 1316


N45T/V50A/N78I
1187, 2137
1252, 1317


V11A/I20L/E27G/D43G/N45D/H51Y/S99G
1188, 2138
1253, 1318


I20L/E27G/D43G/N45T/V50A
1189, 2139
1254, 1319


I20L/K28E/D43G/N45D/V58A/Q89R/
1190, 2140
1255, 1320


G101G-ins (G101GG)




I20L/I36T/N45D
1191, 2141
1256, 1321


I20L/K28E/D43G/N45D/E53G/V58A/N78I
1192, 2142
1257, 1322


A33D/D43G/N45D/V58A/S75P
1193, 2143
1258, 1323


K23R/D43G/N45D
1194, 2144
1259, 1324


I20L/D43G/N45D/V58A/N78I/D90G/G101D
1195, 2145
1260, 1325


D43G/N45D/L56Q/V58A/
1196, 2146
1261, 1326


G101G-ins (G101GG)




I20L/K23E/D43G/N45D/V58A/N78I
1197, 2147
1262, 1327


I20L/K23E/D43G/N45D/V50A/N78I
1198, 2148
1263, 1328


T19I/E27G/N45I/V50A/N78I/M97K
1199, 2149
1264, 1329


I20L/M41K/D43G/N45D
1200, 2150
1265, 1330


K23R/N45T/N78I
1201, 2151
1266, 1331


I20L/K28E/D43G/N45D/V58A/Q89R/
1871, 2152
1754, 1755


G101G-ins (G101GG)




K57R/S99G
1875, 1965
2054, 2069


K57R/S99G/F189L
1876, 1966



M18V/M97L/F193S/R195G/E200K/H202Q
1877, 1967



I36S/M41K/M97L/K144Q/R195G/E200K/
1878, 1968



H202Q/L206F




C22R/Q65L/L124S/K144Q/R195G/
1879



E200N/H202Q/T221L




M18V/I98L/L124S/P198T/L206F
1880, 1969



S99G/N117S/I148V/K171R/R180S
1881, 1970



I36T/M97L/A103V/Q155H
1882, 1971



K28I/S99G
1883, 1972
2055, 2070


R195S
1884, 1973



A79T/S99G/T185A/R195G/E200K/
1885, 1974



H202Q/L206F




K57R/S99G/L124S/K144Q
1886, 1975



K57R/S99G/R195G
1887, 1976



D55V/M97L/S99G
1888, 1977
2056, 2071


E27G/I36T/D55N/M97L/K111E
1889, 1978
2057, 2072


E54G/M97L/S99G
1890, 1979
2058, 2073


G15A/I36T/M97L/K111E/H202Q
1891, 1980



G15A/I36T/V129D
1892, 1981



G15A/I36T/V129D/R195G
1893, 1982



G15A/V129D
1894, 1983



I36S/M97L
1895, 1984
2059, 2074


I36T/D55N/M97L/K111E/A204T
1896, 1985



I36T/D55N/M97L/K111E/V129A/F173L
1897, 1986



I36T/D55S/M97L/K111E/I148V/R180S
1898, 1987



I36T/G52R/M97L/V112A/K144E/
1899, 1988



V175A/P198T




I36T/I46V/D55G/M97L/K106E/K144E/
1900, 1989



T185A/R195G




I36T/I83T/M97L/K144E/P198T
1901, 1990



I36T/M97L/K111E
1902, 1991
2060, 2075


I36T/M97L/K144E/P198T
1903, 1992



I36T/M97L/Q155H/F193S/N201Y
1904, 1993



I36T/M97L/V129D
1905, 1994



L35P/I36S/M97L/K111E
1906, 1995
2061, 2076


M18I/I36T/E53G/M97L/K144E/E199G/
1907, 1996



V207A




M18T/I36T/D55N/M97L/K111E
1908, 1997
2062, 2077


M18V/M97L/T176N/R195G
1909, 1998



M97L/S99G
1910, 1999
2063, 2078


N17D/M97L/S99G
1911, 2000
2064, 2079


S99G/T185A/R195G/P198T
1912, 2001



V129D/H202Q
1913, 2002



V129D/P198T
1914, 2003



V129D/T150A
1915, 2004



V93E/V129D
1916, 2005



Y10F/M18V/S99G/Q138R/T203A
1917, 2006



N45D
1918, 2007
2065, 2080


K160M/R195G
1919, 2008



N45D/K144E
1920, 2009



N45D/P198S
1921, 2010



N45D/P198T
1922, 2011



N45D/R195G
1923, 2012



N45D/R195S
1924, 2013



N45D/S131F
1925, 2014



N45D/V58D
1926, 2015
2066, 2081


V129D/R195S
1927, 2016



I98T/F173Y/L196S
1928, 2017



N45D/E134G/L213P
1929, 2018



N45D/F173I/S177C
1930, 2019



N45D/I148V/R195G
1931, 2020



N45D/K111T/R195G
1932, 2021



N45D/N113Y/R195S
1933, 2022



N45D/N165Y/E170G
1934, 2023



N45D/Q89R/I98V
1935, 2024
2067, 2082


N45D/S131F/P198S
1936, 2025



N45D/S75P/P198S
1937, 2026



N45D/V50A/R195T
1938, 2027



E27D/N45D/T183A/I188V
1939, 2028



F173Y/T183I/L196S/T203A
1940, 2029



K23N/N45D/S75P/N120S
1941, 2030



N45D/G102D/R194W/R195G
1942, 2031



N45D/G52V/Q121L/P198S
1943, 2032



N45D/I148V/R195G/N201D
1944, 2033



N45D/K111T/T183A/I188V
1945, 2034



N45D/Q89R/F189S/P198S
1946, 2035



N45D/S99G/C137R/V207A
1947, 2036



N45D/T163I/K167R/R195G
1948, 2037



N45D/T183A/T192S/R194G
1949, 2038



N45D/V50A/I119T/K144E
1950, 2039



T19A/N45D/K144E/R195G
1951, 2040



V11E/N45D/T130A/P198T
1952, 2041



V26A/N45D/T163I/T185A
1953, 2042



K23N/N45D/L124S/K167T/R195G
1954, 2043



K23N/N45D/Q73R/T163I
1955, 2044



K28E/N45D/W149R/S158G/P198T
1956, 2045



K28R/N45D/K57E/I98V/R195S
1957, 2046



K28R/N45D/V129D/T163N/R195T
1958, 2047



M41K/D43G/N45D/R64S/R195G
1959, 2048



M41K/D43G/N45D/R64S/S99G
1960, 2049
2068, 2083


N45D/R68L/F173L/D197G/P198S
1961, 2050



N45D/V50A/I148V/R195G/N201D
1962, 2051



M41K/D43G/K44E/N45D/R195G/N201D
1963, 2052



N45D/V50A/L124S/K144E/L179P/R195G
1964, 2053
















TABLE 9







Exemplary variant PD-L2 polypeptides










ECD
IgV



SEQ
SEQ ID


Mutation(s)
ID NO
NO












Wild-type
31
1333, 1393


H15Q
1334
1411, 1487


N24D
1335
1412, 1488


E44D
1336
1413, 1489


V89D
1337
1414, 1490


Q82R/V89D
1338
1415, 1491


E59G/Q82R
1339
1416, 1492


S39I/V89D
1340
1417, 1493


S67L/V89D
1341
1418, 1494


S67L/I85F
1342
1419, 1495


S67L/I86T
1343
1420, 1496


H15Q/K65R
1344
1421, 1497


H15Q/Q72H/V89D
1345
1422, 1498


H15Q/S67L/R76G
1346
1423, 1499


H15Q/R76G/I85F
1347
1424, 1500


H15Q/T47A/Q82R
1348
1425, 1501


H15Q/Q82R/V89D
1349
1426, 1502


H15Q/C23S/I86T
1350
1427, 1503


H15Q/S39I/I86T
1351
1428, 1504


H15Q/R76G/I85F
1352
1429, 1505


E44D/V89D/W91R
1353
1430, 1506


I13V/S67L/V89D
1354
1431, 1507


H15Q/S67L/I86T
1355
1432, 1508


I13V/H15Q/S67L/I86T
1356
1433, 1509


I13V/H15Q/E44D/V89D
1357
1434, 1510


I13V/S39I/E44D/Q82R/V89D
1358
1435, 1511


I13V/E44D/Q82R/V89D
1359
1436, 1512


I13V/Q72H/R76G/I86T
1360
1437, 1513


I13V/H15Q/R76G/I85F
1361
1438, 1514


H15Q/S39I/R76G/V89D
1362
1439, 1515


H15Q/S67L/R76G/I85F
1363
1440, 1516


H15Q/T47A/Q72H/R76G/I86T
1364
1441, 1517


H15Q/T47A/Q72H/R76G
1365
1442, 1518


I13V/H15Q/T47A/Q72H/R76G
1366
1443, 1519


H15Q/E44D/R76G/I85F
1367
1444, 1520


H15Q/S39I/S67L/V89D
1368
1445, 1521


H15Q/N32D/S67L/V89D
1369
1446, 1522


N32D/S67L/V89D
1370
1447, 1523


H15Q/S67L/Q72H/R76G/V89D
1371
1448, 1524


H15Q/Q72H/Q74R/R76G/I86T
1372
1449, 1525


G28V/Q72H/R76G/I86T
1373
1450, 1526


I13V/H15Q/S39I/E44D/S67L
1374
1451, 1527


E44D/S67L/Q72H/Q82R/V89D
1375
1452, 1528


H15Q/V89D
1376
1453, 1529


H15Q/T47A
1377
1454, 1530


I13V/H15Q/Q82R
1378
1455, 1531


I13V/H15Q/V89D
1379
1456, 1532


I13V/S67L/Q82R/V89D
1380
1457, 1533


I13V/H15Q/Q82R/V89D
1381
1458, 1534


H15Q/V31M/S67L/Q82R/V89D
1382
1459, 1535


I13V/H15Q/T47A/Q82R
1383
1460, 1536


I13V/H15Q/V31A/N45S/Q82R/V89D
1384
1461, 1537


I13V/T20A/T47A/K65X/Q82R/V89D
1385
1462, 1538


H15Q/T47A/H69L/Q82R/V89D
1386
1463, 1539


I13V/H15Q/T47A/H69L/R76G/V89D
1387
1464, 1540


I12V/I13V/H15Q/T47A/Q82R/V89D
1388
1465, 1541


I13V/H15Q/R76G/D77N/Q82R/V89D
1389
1466, 1542


I13V/H15Q/T47A/R76G/V89D
1390
1467, 1543


I13V/H15Q/T47A/Q82R/V89D
1391
1468, 1544


I13V/H15Q/N24D/Q82R/V89D
1392
1469, 1545


I13V/H15Q/I36V/T47A/S67L/V89D
1394
1470, 1546


H15Q/T47A/K65R/S67L/Q82R/V89D
1395
1471, 1547


H15Q/L33P/T47A/S67L/P71S/V89D
1396
1472, 1548


I13V/H15Q/Q72H/R76G/I86T
1397
1473, 1549


H15Q/T47A/S67L/Q82R/V89D
1398
1474, 1550


F2L/H15Q/D46E/T47A/Q72H/R76G/Q82R/V89D
1399
1475, 1551


I13V/H15Q/L33F/T47A/Q82R/V89D
1400
1476, 1552


I13V/H15Q/T47A/E58G/S67L/Q82R/V89D
1401
1477, 1553


H15Q/N24S/T47A/Q72H/R76G/V89D
1402
1478, 1554


I13V/H15Q/E44V/T47A/Q82R/V89D
1403
1479, 1555


H15Q/N18D/T47A/Q72H/V73A/R76G/I86T/
1404
1480, 1556


V89D




I13V/H15Q/T37A/E44D/S48C/S67L/Q82R/V89D
1405
1481, 1557


H15Q/L33H/S67L/R76G/Q82R/V89D
1406
1482, 1558


I13V/H15Q/T47A/Q72H/R76G/I86T
1407
1483, 1559


H15Q/S39I/E44D/Q72H/V75G/R76G/Q82R/
1408
1484, 1560


V89D




H15Q/T47A/S67L/R76G/Q82R/V89D
1409
1485, 1561


I13V/H15Q/T47A/S67L/Q72H/R76G/Q82R/
1410
1486, 1562


V89D









In some embodiments, the immunomodulatory protein contains an affinity modified IgSF domain containing one or more amino acid substitutions in a wild-type or unmodified IgSF domain of an NKp30 family member. In some embodiments, the affinity modified IgSF domain has at least about 85%, 86%, 86%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity with the wild-type or unmodified IgSF domain or specific binding fragment thereof of an NKp30 family member, such as the IgSF domain (e.g. IgC) contained in the IgSF protein set forth in SEQ ID NO: 27. Table 10 provides exemplary affinity modified NKp30 IgSF domains.









TABLE 10







Exemplary variant NKp30 polypeptides













IgC-like




ECD
domain




SEQ ID
SEQ ID



Mutation(s)
NO
NO















Wild-type
54
214



L30V/A60V/S64P/S86G
143
215



L30V
144
216



A60V
145
217



S64P
146
218



S86G
147
219










IV. Secretable Immunomodulatory Proteins and Engineered Cells

Provided herein are immunomodulatory proteins, such as any described above, which can be expressed and secreted by engineered cells. Secretable immunomodulatory proteins (SIPs), and engineered cells expressing and secreting such secretable immunomodulatory proteins, can have therapeutic utility by modulating immunological activity in a mammal with a disease or disorder in which modulation of the immune system response is beneficial.


In some embodiments, the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain. In some embodiments, the affinity-modified IgSF domain can include an affinity-modified IgSF domain of an Ig super family member, such as any described herein. In some embodiments, the affinity-modified IgSF domain is in an IgSF family member of the B7 family of proteins. In some embodiments, the affinity-modified IgSF domain is in an IgSF family member that binds an inhibitory receptor. In some embodiments, the affinity-modified IgSF domain comprises any one or more amino acid substitution in an IgSF domain, such as any one or more amino acid substitution(s) set forth in any of Tables 2-10.


In some embodiments, the immunomodulatory protein does not comprise a transmembrane domain. In some embodiments, the immunomodulatory protein is not conjugated to a half-life extending moiety (such as an Fc domain or a multimerization domain). In some embodiments, the immunomodulatory protein comprises a signal peptide, e.g. an antibody signal peptide or other efficient signal sequence to get domains outside of cell. When the immunomodulatory protein comprises a signal peptide and is expressed by an engineered cell, the signal peptide causes the immunomodulatory protein to be secreted by the engineered cell. Generally, the signal peptide, or a portion of the signal peptide, is cleaved from the immunomodulatory protein with secretion. The immunomodulatory protein can be encoded by a nucleic acid (which can be part of an expression vector). In some embodiments, the immunomodulatory protein is expressed and secreted by a cell (such as an immune cell, for example a primary immune cell).


A. Signal Peptide


In some embodiments, the immunomodulatory proteins provided herein further comprises a signal peptide. In some embodiments, provided herein is a nucleic acid molecule encoding the immunomodulatory protein operably connected to a secretion sequence encoding the signal peptide.


A signal peptide is a sequence on the N-terminus of an immunomodulatory protein that signals secretion of the immunomodulatory protein from a cell. In some embodiments, the signal peptide is about 5 to about 40 amino acids in length (such as about 5 to about 7, about 7 to about 10, about 10 to about 15, about 15 to about 20, about 20 to about 25, or about 25 to about 30, about 30 to about 35, or about 35 to about 40 amino acids in length).


In some embodiments, the signal peptide is a native signal peptide from the corresponding wild-type IgSF family member. For example, in some embodiments, the immunomodulatory protein comprises at least one non-immunoglobulin affinity modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain from a wild-type IgSF family member, and a signal peptide from the wild-type IgSF family member. Exemplary signal peptides from IgSF family members are identified in Table 1.


In some embodiments, the signal peptide is a non-native signal peptide. For example, in some embodiments, the non-native signal peptide is a mutant native signal peptide from the corresponding wild-type IgSF family member, and can include one or more (such as 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more) substitutions insertions or deletions. In some embodiments, the non-native signal peptide is a signal peptide or mutant thereof of a family member from the same IgSF family as the wild-type IgSF family member. In some embodiments, the non-native signal peptide is a signal peptide or mutant thereof from an IgSF family member from a different IgSF family that the wild-type IgSF family member. In some embodiments, the signal peptide is a signal peptide or mutant thereof from a non-IgSF protein family, such as a signal peptide from an immunoglobulin (such as IgG heavy chain or IgG-kappa light chain), a cytokine (such as interleukin-2 (IL-2), or CD33), a serum albumin protein (e.g. HSA or albumin), a human azurocidin preprotein signal sequence, a luciferase, a trypsinogen (e.g. chymotrypsinogen or trypsinogen) or other signal peptide able to efficiently secrete a protein from a cell. Exemplary signal peptides include any described in the Table 11.









TABLE 11







Exemplary Signal Peptides









SEQ ID NO
Signal Peptide
Peptide Sequence





SEQ ID NO: 413
HSA signal peptide
MKWVTFISLLFLFSSAYS





SEQ ID NO: 414
Ig kappa light chain
MDMRAPAGIFGFLLVLFPGYR




S





SEQ ID NO: 415
human azurocidin preprotein
MTRLTVLALLAGLLASSRA



signal sequence






SEQ ID NO: 416
IgG heavy chain signal peptide
MELGLSWIFLLAILKGVQC





SEQ ID NO: 417
IgG heavy chain signal peptide
MELGLRWVFLVAILEGVQC





SEQ ID NO: 418
IgG heavy chain signal peptide
MKHLWFFLLLVAAPRWVLS





SEQ ID NO: 419
IgG heavy chain signal peptide
MDWTWRILFLVAAATGAHS





SEQ ID NO: 420
IgG heavy chain signal peptide
MDWTWRFLFVVAAATGVQS





SEQ ID NO: 421
IgG heavy chain signal peptide
MEFGLSWLFLVAILKGVQC





SEQ ID NO: 422
IgG heavy chain signal peptide
MEFGLSWVFLVALFRGVQC





SEQ ID NO: 423
IgG heavy chain signal peptide
MDLLHKNMKHLWFFLLLVAA




PRWVLS





SEQ ID NO: 424
IgG Kappa light chain signal
MDMRVPAQLLGLLLLWLSGA



sequences:
RC





SEQ ID NO: 425
IgG Kappa light chain signal
MKYLLPTAAAGLLLLAAQPAM



sequences:
A





SEQ ID NO: 426
Gaussia luciferase
MGVKVLFALICIAVAEA





SEQ ID NO: 427
Human albumin
MKWVTFISLLFLFSSAYS





SEQ ID NO: 428
Human chymotrypsinogen
MAFLWLLSCWALLGTTFG





SEQ ID NO: 429
Human interleukin-2
MQLLSCIALILALV





SEQ ID NO: 430
Human trypsinogen-2
MNLLLILTFVAAAVA









In some embodiments, the immunomodulatory protein comprises a signal peptide when expressed, and the signal peptide (or a portion thereof) is cleaved from the immunomodulatory protein upon secretion.


B. Nucleic Acid Molecules and Expression Vectors


Provided herein are isolated or recombinant nucleic acids collectively referred to as “nucleic acids” which encode any of the various provided embodiments of the immunomodulatory polypeptides of the invention. In one aspect, there is provided a nucleic acid encoding an immunomodulatory protein comprising at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain. In some embodiments, the immunomodulatory protein encoded by the nucleic acid molecule does not comprise a transmembrane domain. In some embodiments, the immunomodulatory protein encoded by the nucleic acid molecule does not comprise a half-life extending moiety (such as an Fc domain or a multimerization domain). In some embodiments, the immunomodulatory protein encoded by the nucleic acid molecule comprises a signal peptide. In some embodiments the nucleic acid molecule further comprises at least one promoter operably linked to control expression of the immunomodulatory protein.


Nucleic acids provided herein, including all described below, are useful in recombinant expression of the immunomodulatory proteins, including for engineering cells. The nucleic acids provided herein can be in the form of RNA or in the form of DNA, and include mRNA, cRNA, recombinant or synthetic RNA and DNA, and cDNA. The nucleic acids of the invention are typically DNA molecules, and usually double-stranded DNA molecules. However, single-stranded DNA, single-stranded RNA, double-stranded RNA, and hybrid DNA/RNA nucleic acids or combinations thereof comprising any of the nucleotide sequences of the invention also are provided.


Also provided herein are expression vectors useful in engineering cells to express the immunomodulatory proteins of the present invention. In one aspect, there is provided a recombinant expression vector comprising a nucleic acid encoding an immunomodulatory protein under the operable control of a signal sequence for secretion, wherein the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain. In some embodiments, the encoded immunomodulatory protein is secreted when expressed from a cell. In some embodiments, the immunomodulatory protein encoded by the nucleic acid molecule does not comprise a transmembrane domain. In some embodiments, the immunomodulatory protein encoded by the nucleic acid molecule does not comprise a half-life extending moiety (such as an Fc domain or a multimerization domain). In some embodiments, the immunomodulatory protein encoded by the nucleic acid molecule comprises a signal peptide.


The nucleic acids encoding the immunomodulatory polypeptides provided herein can be introduced into cells using recombinant DNA and cloning techniques. To do so, a recombinant DNA molecule encoding a immunomodulatory polypeptide is prepared. Methods of preparing such DNA molecules are well known in the art. For instance, sequences coding for the peptides could be excised from DNA using suitable restriction enzymes. Alternatively, the DNA molecule could be synthesized using chemical synthesis techniques, such as the phosphoramidite method. Also, a combination of these techniques could be used. In some instances, a recombinant or synthetic nucleic acid may be generated through polymerase chain reaction (PCR).


In some embodiments, a DNA insert can be generated encoding one or more affinity-modified IgSF domains and, in some embodiments, a signal peptide. This DNA insert can be cloned into an appropriate transduction/transfection vector as is known to those of skill in the art. Also provided are expression vectors containing the nucleic acid molecules.


In some embodiments, the expression vectors are capable of expressing the immunomodulatory proteins in an appropriate cell under conditions suited to expression of the protein. In some aspects, nucleic acid molecule or an expression vector comprises the DNA molecule that encodes the immunomodulatory protein operatively linked to appropriate expression control sequences. Methods of affecting this operative linking, either before or after the DNA molecule is inserted into the vector, are well known. Expression control sequences include promoters, activators, enhancers, operators, ribosomal binding sites, start signals, stop signals, cap signals, polyadenylation signals, and other signals involved with the control of transcription or translation. In some embodiments, a nucleic acid of the invention further comprises nucleotide sequence that encodes a secretory or signal peptide operably linked to the nucleic acid encoding the immunomodulatory protein, thereby allowing for secretion of the immunomodulatory protein.


In some embodiments, expression of the immunomodulatory protein is controlled by a promoter to enhance to control or regulate expression. The promoter is operably linked to the portion of the nucleic acid molecule encoding the immunomodulatory protein. In some embodiments, the promotor is a constitutively active promotor (such as a tissue-specific constitutively active promotor or other constitutive promotor). In some embodiments, the promotor is an inducible promotor, which may be responsive to an inducing agent (such as a T cell activation signal).


In some embodiments, a constitutive promoter is operatively linked to the nucleic acid molecule encoding the immunomodulatory protein. Exemplary constitutive promoters include the Simian vacuolating virus 40 (SV40) promoter, the cytomegalovirus (CMV) promoter, the ubiquitin C (UbC) promoter, and the EF-1 alpha (EF1a) promoter. In some embodiments, the constitutive promoter is tissue specific. For example, in some embodiments, the promoter allows for constitutive expression of the immunomodulatory protein in specific tissues, such as immune cells, lymphocytes, or T cells. Exemplary tissue-specific promoters are described in U.S. Pat. No. 5,998,205, including, for example, a fetoprotein, DF3, tyrosinase, CEA, surfactant protein, and ErbB2 promoters.


In some embodiments, an inducible promoter is operatively linked to the nucleic acid molecule encoding the immunomodulatory protein such that expression of the nucleic acid is controllable by controlling the presence or absence of the appropriate inducer of transcription. For example, the promoter can be a regulated promoter and transcription factor expression system, such as the published tetracycline-regulated systems or other regulatable systems (see, e.g. published International PCT Appl. No. WO 01/30843), to allow regulated expression of the encoded polypeptide. An exemplary regulatable promoter system is the Tet-On (and Tet-Off) system available, for example, from Clontech (Palo Alto, Calif.). This promoter system allows the regulated expression of the transgene controlled by tetracycline or tetracycline derivatives, such as doxycycline. Other regulatable promoter systems are known (see e.g., published U.S. Application No. 2002-0168714, entitled “Regulation of Gene Expression Using Single-Chain, Monomeric, Ligand Dependent Polypeptide Switches,” which describes gene switches that contain ligand binding domains and transcriptional regulating domains, such as those from hormone receptors).


In some embodiments, the promotor is responsive to an element responsive to T-cell activation signaling. Solely by way of example, in some embodiments, an engineered T cell comprises an expression vector encoding the immunomodulatory protein and a promotor operatively linked to control expression of the immunomodulatory protein. The engineered T cell can be activated, for example by signaling through an engineered T cell receptor (TCR) or a chimeric antigen rector (CAR), and thereby triggering expression and secretion of the immunomodulatory protein through the responsive promotor.


In some embodiments, an inducible promoter is operatively linked to the nucleic acid molecule encoding the immunomodulatory protein such that the immunomodulatory protein is expressed in response to a nuclear factor of activated T-cells (NFAT) or nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB). For example, in some embodiments, the inducible promoter comprises a binding site for NFAT or NF-κB. For example, in some embodiments, the promoter is an NFAT or NF-κB promoter or a functional variant thereof. Thus, in some embodiments, the nucleic acids make it possible to control the expression of immunomodulatory protein while also reducing or eliminating the toxicity of the immunomodulatory protein. In particular, engineered immune cells comprising the nucleic acids of the invention express and secrete the immunomodulatory protein only when the cell (e.g., a T-cell receptor (TCR) or a chimeric antigen receptor (CAR) expressed by the cell) is specifically stimulated by an antigen and/or the cell (e.g., the calcium signaling pathway of the cell) is non-specifically stimulated by, e.g., phorbol myristate acetate (PMA)/Ionomycin. Accordingly, the expression and secretion of immunomodulatory protein can be controlled to occur only when and where it is needed (e.g., in the presence of an infectious disease-causing agent, cancer, or at a tumor site), which can decrease or avoid undesired immunomodulatory protein interactions.


In some embodiments, the nucleic acid encoding an immunomodulatory protein described herein comprises a suitable nucleotide sequence that encodes a NFAT promoter, NF-κB promoter, or a functional variant thereof. “NFAT promoter” as used herein means one or more NFAT responsive elements linked to a minimal promoter. “NF-κB promoter” refers to one or more NF-κB responsive elements linked to a minimal promoter. In some embodiments, the minimal promoter of a gene is a minimal human IL-2 promoter or a CMV promoter. The NFAT responsive elements may comprise, e.g., NFAT1, NFAT2, NFAT3, and/or NFAT4 responsive elements. The NFAT promoter, NF-κB promoter, or a functional variant thereof may comprise any number of binding motifs, e.g., at least two, at least three, at least four, at least five, or at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, or up to twelve binding motifs.


In some embodiments, the resulting expression vector having the DNA molecule thereon is used to transform, such as transduce, an appropriate cell. The introduction can be performed using methods well known in the art. Exemplary methods include those for transfer of nucleic acids encoding the receptors, including via viral, e.g., retroviral or lentiviral, transduction, transposons, and electroporation. In some embodiments, the expression vector is a viral vector. In some embodiments, the nucleic acid is transferred into cells by lentiviral or retroviral transduction methods.


C. Exemplary Immunomodulatory Proteins and Encoding Nucleic Acid Molecules


Provided herein is a immunomodulatory protein, e.g. secretable immunomodulatory protein, in accord with the above description that comprises a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to any of SEQ ID NOS: 28-54 and 410 or to a specific binding fragment thereof containing an IgSF domain (e.g. IgV domain or IgC domain), and that contains at least one affinity-modified IgSF domain as described containing one or more amino acid modifications, e.g. amino acid substitutions, in an IgSF domain. In some embodiments, the immunomodulatory protein e.g. secretable immunomodulatory protein, comprises a sequence of amino acids that comprises the IgV domain or a specific fragment thereof contained within the sequence of amino acids set forth in any of SEQ ID NOS: 28-54 and 410 (see e.g. Table 1) but in which is contained therein one or more amino acid modifications, e.g. amino acid substitutions, such that the IgV domain exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to the corresponding IgV domain contained in any of SEQ ID NOS: 28-54 and 410. In some embodiments, the immunomodulatory protein contains 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid modifications, e.g. amino acid substitutions. In some embodiments, the immunomodulatory protein further comprises a signal peptide as described. In some embodiments, the signal peptide is the native signal peptide of the corresponding wild-type IgSF member (see e.g. Table 1). In some embodiments, the signal peptide is a non-native signal peptide, such as any as described, e.g. Table 11.


Also provided herein is a nucleic acid molecule encoding an immunomodulatory protein comprising a nucleotide sequence that encodes a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to any of SEQ ID NOS: 28-54 and 410 or to a specific binding fragment thereof containing an IgSF domain (e.g. IgV domain or IgC domain) and that contains at least one affinity-modified IgSF domain as described containing one or more amino acid modifications, e.g. amino acid substitutions. In some embodiments, the immunomodulatory protein e.g. secretable immunomodulatory protein, comprises a nucleotide sequence that encodes a sequence of amino acids that comprises the IgV domain or a specific fragment thereof contained within the sequence of amino acids set forth in any of SEQ ID NOS: 28-54 and 410 (see e.g. Table 1) but in which is contained therein one or more amino acid modifications, e.g. amino acid substitutions, such that the IgV domain exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to the corresponding IgV domain contained in any of SEQ ID NOS: 28-54 and 410. In some embodiments, the nucleic acid molecule further comprises a sequence of nucleotides encoding a signal peptide. In some embodiments, the signal peptide is the native signal peptide of the corresponding wild-type IgSF member (see e.g. Table 1). In some embodiments, the signal peptide is a non-native signal peptide, such as any as described, e.g. Table 11.


In some embodiments, the immunomodulatory protein has the sequence of amino acids set forth in any of the SEQ ID NOS of an ECD or an IgV set forth in any of Tables 2-10, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% to any of the SEQ ID NOS of an ECD or an IgV set forth in any of Tables 2-10 and that contains the recited amino acid modifications (e.g. amino acid substitutions). In some embodiments, the immunomodulatory protein further comprises a signal peptide as described. In some embodiments, the signal peptide is the native signal peptide of the corresponding wild-type IgSF member (see e.g. Table 1). In some embodiments, the signal peptide is a non-native signal peptide, such as any as described, e.g. Table 11.


In some embodiments, the nucleic acid molecule encodes an immunomodulatory protein that has the sequence of amino acids set forth in any of the SEQ ID NOS of an ECD or an IgV set forth in any of Tables 2-10, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% to any of the SEQ ID NOS of an ECD or an IgV set forth in any of Tables 2-10 and that contains the recited amino acid modifications (e.g. amino acid substitutions). In some embodiments, the nucleic acid molecule further comprises a sequence of nucleotides encoding a signal peptide. In some embodiments, the signal peptide is the native signal peptide of the corresponding wild-type IgSF member (see e.g. Table 1). In some embodiments, the signal peptide is a non-native signal peptide, such as any as described, e.g. Table 11.


V. Transmembrane Immunomodulatory Protein

Provided herein are immunomodulatory proteins that are transmembrane proteins (“transmembrane immunomodulatory proteins”). Transmembrane immunomodulatory proteins, and engineered cells expressing such transmembrane immunomodulatory proteins, generally have therapeutic utility by modulating immunological activity in a mammal with a disease or disorder in which modulation of the immune system response is beneficial. A transmembrane immunomodulatory protein of the present invention comprises an ectodomain, a transmembrane, and in some embodiments, an endodomain, such as a cytoplasmic signaling domain.


A. Ectodomain


In some embodiments, the provided transmembrane immunomodulatory proteins include an ectodomain comprising at least one affinity modified IgSF domain compared to an IgSF domain of a wild-type mammalian IgSF member. In some embodiments, the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain. In some embodiments, the affinity-modified IgSF domain can include an affinity-modified IgSF domain of an Ig super family member, such as any described herein In some embodiments, the affinity-modified IgSF domain is in an IgSF family member of the B7 family of proteins. In some embodiments, the affinity-modified IgSF domain is in an IgSF family member that binds an inhibitory receptor. In some embodiments, the affinity-modified IgSF domain comprises any one or more amino acid substitution in an IgSF domain described herein, such as any one or more amino acid substitution(s) set forth in any of Tables 2-10.


B. Transmembrane Domain


The transmembrane immunomodulatory proteins provided herein further contain a transmembrane domain linked to the ectodomain. In some embodiments, the transmembrane domain results in an encoded protein for cell surface expression on a cell. In some embodiments, the transmembrane domain is linked directly to the ectodomain. In some embodiments, the transmembrane domain is linked indirectly to the ectodomain via one or more linkers or spacers. In some embodiments, the transmembrane domain contains predominantly hydrophobic amino acid residues, such as leucine and valine.


In some embodiments, a full length transmembrane anchor domain can be used to ensure that the TIPs will be expressed on the surface of the engineered cell, such as engineered T cell. Conveniently, this could be from a particular native protein that is being affinity modified (e.g. CD80 or ICOSL or other native IgSF protein), and simply fused to the sequence of the first membrane proximal domain in a similar fashion as the native IgSF protein (e.g. CD80 or ICOSL). In some embodiments, the transmembrane immunomodulatory protein comprises a transmembrane domain of the corresponding wild-type or unmodified IgSF member, such as a transmembrane domain contained in the sequence of amino acids set forth in any of SEQ ID NOs:1-27 and 408 (see Table 1).


In some embodiments, the transmembrane domain is a non-native transmembrane domain that is not the transmembrane domain of the wild-type IgSF member. In some embodiments, the transmembrane domain is derived from a transmembrane domain from another non-IgSF family member polypeptide that is a membrane-bound or is a transmembrane protein. In some embodiments, a transmembrane anchor domain from another protein on T cells can be used. In some embodiments, the transmembrane domain is derived from CD8. In some embodiments, the transmembrane domain can further contain an extracellular portion of CD8 that serves as a spacer domain. An exemplary CD8 derived transmembrane domain is set forth in SEQ ID NO: 1574 or a portion thereof containing the CD8 transmembrane domain. In some embodiments, the transmembrane domain is a synthetic transmembrane domain.


C. Endodomain


In some embodiments, the transmembrane immunomodulatory protein further contains an endodomain, such as a cytoplasmic signaling domain, linked to the transmembrane domain. In some embodiments, the cytoplasmic signaling domain induces cell signaling. In some embodiments, the endodomain of the transmembrane immunomodulatory protein comprises the cytoplasmic domain of the corresponding wild-type or unmodified polypeptide, such as a cytoplasmic domain contained in the sequence of amino acids set forth in any of SEQ ID NOS:1-27 and 408 (see Table 1).


In some embodiments, provided are CAR-related transmembrane immunomodulatory proteins in which the endodomain of a transmembrane immunomodulatory protein comprises a cytoplasmic signaling domain that comprises at least one ITAM (immunoreceptor tyrosine-based activation motif)-containing signaling domain. ITAM is a conserved motif found in a number of protein signaling domains involved in signal transduction of immune cells, including in the CD3-zeta chain (“CD3-z”) involved in T-cell receptor signal transduction. In some embodiments, the endodomain comprises at CD3-zeta signaling domain. In some embodiments, the CD3-zeta signaling domain comprises the sequence of amino acids set forth in SEQ ID NO: 1575 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% to SEQ ID NO:1575 and retains the activity of T cell signaling. In some embodiments, the endodomain of a CAR-related transmembrane immunomodulatory protein can further comprise a costimulatory signaling domain to further modulate immunomodulatory responses of the T-cell. In some embodiments, the costimulatory signaling domain is CD28, ICOS, 41BB or OX40. In some embodiments, the provided CAR-related transmembrane immunomodulatory proteins have features of CARs to stimulate T cell signaling upon binding of an affinity modified IgSF domain to a cognate binding partner or counter structure. In some embodiments, upon specific binding by the affinity-modified IgSF domain to its counter structure can lead to changes in the immunological activity of the T-cell activity as reflected by changes in cytotoxicity, proliferation or cytokine production.


In some embodiments, a CAR-related transmembrane immunomodulatory protein comprises an antigen binding region that is engineered to specifically bind to a desired counter-structure. In some embodiments, an affinity modified IgSF domain specifically binds its native counter-structure. In some embodiments the counter-structure is an IgSF family member. In some embodiments, provided is a CAR-related transmembrane immunomodulatory protein that specifically binds to a tumor specific IgSF counter-structure. In some embodiments, the antigen binding region (ectodomain) is an affinity modified IgSF domain NKp30. In some embodiments, the affinity modified IgSF domain specifically binds the tumor specific antigen NKp30 ligand B7-H6 (see, Levin et al., The Journal of Immunology, 2009, 182, 134.20). In examples of such embodiments, the endodomain comprises at least one ITAM (immunoreceptor tyrosine-based activation motif) containing signaling domain, such as a CD3-zeta signaling domain. In some embodiments, the endodomain can further comprises at least one of: a CD28 costimulatory domain, an OX40 signaling domain, and a 41BB signaling domain.


In some embodiments, the transmembrane immunomodulatory protein does not contain an endodomain capable of mediating cytoplasmic signaling. In some embodiments, the transmembrane immunomodulatory protein lacks the signal transduction mechanism of the wild-type or unmodified polypeptide and therefore does not itself induce cell signaling. In some embodiments, the transmembrane immunomodulatory protein lacks an intracellular (cytoplasmic) domain or a portion of the intracellular domain of the corresponding wild-type or unmodified polypeptide, such as a cytoplasmic signaling domain contained in the sequence of amino acids set forth in any of SEQ ID NOS:1-27 and 408 (see Table 1). In some embodiments, the transmembrane immunomodulatory protein does not contain an ITIM (immunoreceptor tyrosine-based inhibition motif), such as contained in certain inhibitory receptors, including inhibitory receptors of the IgSF family (e.g. PD-1 or TIGIT). Thus, in some embodiments, the transmembrane immunomodulatory protein only contains the ectodomain and the transmembrane domain, such as any as described.


D. Nucleic Acid Molecules and Vectors


Provided herein are isolated or recombinant nucleic acids collectively referred to as “nucleic acids” which encode any of the various provided embodiments of the transmembrane immunomodulatory polypeptides of the invention. Nucleic acids provided herein, including all described below, are useful in recombinant expression of the transmembrane immunomodulatory proteins, including for engineering cells. The nucleic acids provided herein can be in the form of RNA or in the form of DNA, and include mRNA, cRNA, recombinant or synthetic RNA and DNA, and cDNA. The nucleic acids of the invention are typically DNA molecules, and usually double-stranded DNA molecules. However, single-stranded DNA, single-stranded RNA, double-stranded RNA, and hybrid DNA/RNA nucleic acids or combinations thereof comprising any of the nucleotide sequences of the invention also are provided.


In some embodiments, expression of the transmembrane immunomodulatory protein is controlled by a promoter to enhancer to control or regulate expression, such as described herein for the alternative secretable immunomodulatory proteins. The promoter is operably linked to the portion of the nucleic acid molecule encoding the immunomodulatory protein. In some embodiments, the promotor is a constitutively active promotor (such as a tissue-specific constitutively active promotor or other constitutive promotor). In some embodiments, the promotor is an inducible promotor, which may be responsive to an inducing agent (such as a T cell activation signal). Any promoter or element for enhancing, controlling or regulating expression, either constitutively or inducibly, can be employed, include any described herein. In particular aspects, the promoter is a promoter that is responsive to an element responsive to T-cell activation signaling, for example, by signaling through an engineered T cell receptor (TCR) or a chimeric antigen receptor (CAR). Exemplary of such promoters are those containing a binding site for NFAT or NF-κB as described. For example, in some embodiments, the promoter is an NFAT or NF-κB promoter or a functional variant thereof.


Also provided herein are expression vectors useful in engineering cells to express the transmembrane immunomodulatory proteins of the present invention. The immunomodulatory polypeptides provided herein can be introduced into cells using recombinant DNA techniques. To do so, a recombinant DNA molecule encoding a transmembrane immunomodulatory polypeptide is prepared. Methods of preparing such DNA molecules are well known in the art. For instance, sequences coding for the peptides could be excised from DNA using suitable restriction enzymes. Alternatively, the DNA molecule could be synthesized using chemical synthesis techniques, such as the phosphoramidite method. Also, a combination of these techniques could be used. In some instances, a recombinant or synthetic nucleic acid may be generated through polymerase chain reaction (PCR).


In some embodiments, a full length DNA insert can be generated comprising an optional endodomain (i.e., cytoplasmic domain), a transmembrane anchor domain, an optional spacer domain, an optional epitope tag, and finally one or more extracellular affinity modified IgSF domains. This DNA insert can be cloned into an appropriate T cell transduction/transfection vector as is known to those of skill in the art. Also provided are vectors containing the nucleic acid molecules.


In some embodiments, the expression vectors are capable of expressing the transmembrane immunomodulatory proteins in an appropriate cell under conditions suited to expression of the protein. In some aspects, an expression vector comprises the DNA molecule that codes for the transmembrane immunomodulatory protein operatively linked to appropriate expression control sequences. Methods of affecting this operative linking, either before or after the DNA molecule is inserted into the vector, are well known. Expression control sequences include promoters, activators, enhancers, operators, ribosomal binding sites, start signals, stop signals, cap signals, polyadenylation signals, and other signals involved with the control of transcription or translation. In some embodiments, a nucleic acid of the invention further comprises nucleotide sequence that encodes a secretory or signal peptide operably linked to the nucleic acid encoding the transmembrane immunomodulatory protein.


In some embodiments, the resulting expression vector having the DNA molecule thereon is used to transform, such as transduce, an appropriate cell. The introduction can be performed using methods well known in the art. Exemplary methods include those for transfer of nucleic acids encoding the receptors, including via viral, e.g., retroviral or lentiviral, transduction, transposons, and electroporation. In some embodiments, the expression vector is a viral vector. In some embodiments, the nucleic acid is transferred into cells by lentiviral or retroviral transduction methods.


E. Exemplary Transmembrane Immunomodulatory Proteins and Encoding Nucleic Acid Molecules


Provided herein is a transmembrane immunomodulatory protein in accord with the above description that comprises a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to any of SEQ ID NOS: 381-407 and 409 or to a portion thereof that contains an ectodomain comprising at least one affinity-modified IgSF domain as described and a transmembrane domain. In some embodiments, the transmembrane immunomodulatory protein has an ecotodomain containing the sequence of amino acids set forth in any of the SEQ ID NOS of an ECD or an IgV set forth in any of Tables 2-10, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% to any of the SEQ ID NOS of an ECD or an IgV set forth in any of Tables 2-10 and that contains the recited amino acid modifications (e.g. amino acid substitutions). In some embodiments, the encoded immunomodulatory protein contains 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid modifications, e.g. amino acid substitutions. In some embodiments, the transmembrane immunomodulatory protein can further comprise a cytoplasmic domain as described, such as a cytoplasmic domain of any of SEQ ID NOS: 381-407 and 409 as set forth in Table 1. In some embodiments, the transmembrane immunomodulatory protein can further contain a signal peptide. In some embodiments, the signal peptide is the native signal peptide of the corresponding wild-type IgSF member (see e.g. Table 1). In some embodiments, the signal peptide is a non-native signal peptide, such as any as described, e.g. Table 11.


Also provided herein is a nucleic acid molecule encoding a transmembrane immunomodulatory protein comprising a nucleotide sequence that encodes a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to any of SEQ ID NOS: 381-407 and 409 or to a portion thereof that contains an ectodomain comprising at least one affinity-modified IgSF domain as described, a transmembrane domain and, optionally, a cytoplasmic domain. In some embodiments, the nucleic acid encodes an immunomodulatory protein that has the sequence of amino acids set forth in any of the SEQ ID NOS of an ECD or an IgV set forth in any of Tables 2-10, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% to any of the SEQ ID NOS of an ECD or an IgV set forth in any of Tables 2-10 and that contains the recited amino acid modifications (e.g. amino acid substitutions). In some embodiments, the encoded immunomodulatory protein contains 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more amino acid modifications, e.g. amino acid substitutions. In some embodiments, the nucleic acid molecule can further comprise a sequence of nucleotides encoding a signal peptide. In some embodiments, the signal peptide is the native signal peptide of the corresponding wild-type IgSF member (see e.g. Table 1). In some embodiments, the signal peptide is a non-native signal peptide, such as any as described, e.g. Table 11.


VI. Engineered Cells

Provided herein are engineered cells that comprise an immunomodulatory protein or a nucleic acid (such as an expression vector) that encodes an immunomodulatory protein as described herein.


In some embodiments, the engineered cells contain a secretable immunomodulatory protein or a nucleic acid molecule encoding a secretable immunomodulatory protein, such as any as described. In some embodiments, the engineered cell expresses and secretes the immunomodulatory protein. In some embodiments, the engineered cells express and are capable of or are able to secrete the immunomodulatory protein from the cells under conditions suitable for secretion of the protein.


In one aspect, there is provided an engineered immune cell comprising a nucleic acid molecule that encodes an immunomodulatory protein, wherein the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid modifications (e.g., substitutions) in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; and the engineered cell is configured to express and secrete the immunomodulatory protein. In some embodiments, the immunomodulatory protein does not comprise a transmembrane domain. In some embodiments, the immunomodulatory protein is not conjugated to half-life extending moiety (such as a multimerization domain or an Fc domain). In some embodiments, the nucleic acid molecule comprises a sequence encoding a secretory signal peptide operably linked to the sequence encoding the immunomodulatory protein.


In some embodiments, the engineered cells contain a transmembrane immunomodulatory protein or a nucleic acid molecule encoding a transmembrane immunomodulatory protein, such as any as described. In some embodiments, the engineered cells express the immunomodulatory protein on its surface.


In some embodiments, the engineered cell is an immune cell, such as a lymphocyte (for example, a tumor infiltrating lymphocyte (TIL), T-cell or NK cell) or on a myeloid cell. In some embodiments, the engineered cell is an antigen presenting cell (APC). In some embodiments, the engineered cells are engineered mammalian T cells or engineered mammalian antigen presenting cells (APCs). In some embodiments, the engineered immune cells or APCs are human or murine cells.


In some embodiments, engineered T cells include, but are not limited to, T helper cell, cytotoxic T-cell (alternatively, cytotoxic T lymphocyte or CTL), natural killer T-cell, regulatory T-cell, memory T-cell, or gamma delta T-cell. In some embodiments, the engineered T cells are CD4+ or CD8+. In addition to the signal of the MHC, engineered T-cells also require a co-stimulatory signal which in some embodiments is provided by the immunomodulatory proteins as discussed herein.


In some embodiments, the engineered APCs include, for example, MHC II expressing APCs such as macrophages, B cells, and dendritic cells, as well as artificial APCs (aAPCs) including both cellular and acellular (e.g., biodegradable polymeric microparticles) aAPCs. Artificial APCs (aAPCs) are synthetic versions of APCs that can act in a similar manner to APCs in that they present antigens to T cells as well as activate them. Antigen presentation is performed by the MHC (Class I or Class II). In some aspects, in engineered APCs such as aAPCs, the antigen that is loaded onto the MHC is, in some embodiments, a tumor specific antigen or a tumor associated antigen. The antigen loaded onto the MHC is recognized by a T-cell receptor (TCR) of a T cell, which, in some cases, can express one or more cognate binding partners or other molecule recognized by the affinity modified IgSF domain of the immunomodulatory proteins provided herein.


In some embodiments a cellular aAPC can be engineered to contain a SIP or TIP and TCR agonist which is used in adoptive cellular therapy. In some embodiments, a cellular aAPC can be engineered to contain a SIP or TIP and TCR agonist which is used in ex vivo expansion of human T cells, such as prior to administration, e.g., for reintroduction into the patient. In some aspects, the aAPC may include expression of at least one anti-CD3 antibody clone, e.g. such as, for example, OKT3 and/or UCHT1. In some aspects, the aAPCs may be inactivated (e.g. irradiated). In some embodiment, the SIP or TIP can include any variant or affinity-modified IgSF domain that exhibits binding affinity for a cognate binding partner on a T cell.


In some embodiments, a immunomodulatory protein provided herein, such as a transmembrane immunomodulatory protein or a secretable immunomodulatory protein, is co-expressed or engineered into a cell that expresses an antigen-binding receptor, such as a recombinant receptor, such as a chimeric antigen receptor (CAR) or T cell receptor (TCR). In some embodiments, the engineered cell, such as an engineered T cell, recognizes a desired antigen associated with cancer, inflammatory and autoimmune disorders, or a viral infection. In specific embodiments, the antigen-binding receptor contains an antigen-binding moiety that specifically binds a tumor specific antigen or a tumor associated antigen. In some embodiments, the engineered T-cell is a CAR (chimeric antigen receptor) T-cell that contains an antigen-binding domain (e.g. scFv) that specifically binds to an antigen, such as a tumor specific antigen or tumor associated antigen. In another embodiment, the engineered T-cell possesses a TCR, including a recombinant or engineered TCR. In some embodiments, the TCR can be a native TCR. Those of skill in the art will recognize that generally native mammalian T-cell receptors comprise an alpha and a beta chain (or a gamma and a delta chain) involved in antigen specific recognition and binding. In some embodiments, the TCR is an engineered TCR that is modified. In some embodiments, the TCR of an engineered T-cell specifically binds to a tumor associated or tumor specific antigen presented by an APC. Thus, in some embodiments, the immunomodulatory protein is expressed and secreted in an engineered T-cell receptor cell or and engineered chimeric antigen receptor cell. In such embodiments, the engineered cell co-expresses the immunomodulatory protein and the CAR or TCR. In some embodiments, the SIP protein is expressed in an engineered T-cell receptor cell or an engineered chimeric antigen receptor cell. In such embodiments, the engineered cell co-expresses the SIP and the CAR or TCR.


Chimeric antigen receptors (CARs) are recombinant receptors that include an antigen-binding domain (ectodomain), a transmembrane domain and an intracellular signaling region (endodomain) that is capable of inducing or mediating an activation signal to the T cell after the antigen is bound. In some examples, CAR-expressing cells are engineered to express an extracellular single chain variable fragment (scFv) with specificity for a particular tumor antigen linked to an intracellular signaling part comprising an activating domain and, in some cases, a costimulatory domain. The costimulatory domain can be derived from, e.g., CD28, OX-40, 4-1BB/CD137, inducible T cell costimulator (ICOS). The activating domain can be derived from, e.g., CD3, such as CD3 zeta, epsilon, delta, gamma, or the like. In certain embodiments, the CAR is designed to have two, three, four, or more costimulatory domains. The CAR scFv can be designed to target an antigen expressed on a cell associated with a disease or condition, e.g. a tumor antigen, such as, for example, CD19, which is a transmembrane protein expressed by cells in the B cell lineage, including all normal B cells and B cell malignances, including but not limited to NHL, CLL, and non-T cell ALL. Example CAR+ T cell therapies and constructs are described in U.S. Patent Publication Nos. 2013/0287748, 2014/0227237, 2014/0099309, and 2014/0050708, and these references are incorporated by reference in their entirety.


In some aspects, the antigen-binding domain is an antibody or antigen-binding fragment thereof, such as a single chain fragment (scFv). In some embodiments, the antigen is expressed on a tumor or cancer cell. Exemplary of an antigen is CD19. Exemplary of a CAR is an anti-CD19 CAR, such as a CAR containing an anti-CD19 scFv set forth in SEQ ID NO:1576 or SEQ ID NO:1577.


In some embodiments, the CAR further contains a spacer, a transmembrane domain, and an intracellular signaling domain or region comprising an ITAM signaling domain, such as a CD3zeta signaling domain.


In some embodiments, the CAR further includes a costimulatory signaling domain. In some embodiments, the spacer and transmembrane domain are the hinge and transmembrane domain derived from CD8, such as having an exemplary sequence set forth in SEQ ID NO: 1574, 1578, 2158 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO:1574, 1578, 2158. In some embodiments, the endodomain comprises at CD3-zeta signaling domain. In some embodiments, the CD3-zeta signaling domain comprises the sequence of amino acids set forth in SEQ ID NO: 1575 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to SEQ ID NO:1575 and retains the activity of T cell signaling. In some embodiments, the endodomain of a CAR can further comprise a costimulatory signaling domain or region to further modulate immunomodulatory responses of the T-cell. In some embodiments, the costimulatory signaling domain is CD28, ICOSL, 41BB or OX40. In some embodiments, the costimulatory signaling domain is a derived from CD28 or 4-1BB and comprises the sequence of amino acids set forth in any of SEQ ID NOS:1579-1582 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to SEQ ID NO:1579-1582 and retains the activity of T cell costimulatory signaling.


In some embodiments, the construct encoding the CAR further encodes a second protein, such as a marker, e.g. detectable protein, separated from the CAR by a self-cleaving peptide sequence. In some embodiments, the self-cleaving peptide sequence is an F2A, T2A, E2A or P2A self-cleaving peptide. Exemplary sequences of a T2A self-cleaving peptide are set for the in any one of SEQ ID NOS: 2165, 2169, 2177 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to any of SEQ ID NOS: 2165, 2169, 2177. In some embodiments, the T2A is encoded by the sequence of nucleotides set forth in SEQ ID NO:2176 or a sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to any of SEQ ID NO: 2176. An exemplary sequence of a P2A self-cleaving peptide is set in SEQ ID NO: 2178 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to SEQ ID NOS: 2178.


In some embodiments, the marker is a detectable protein, such as a fluorescent protein, e.g. a green fluorescent protein (GFP) or blue fluorescent protein (BFP). Exemplary sequences of a fluorescent protein marker are set forth in SEQ ID NO: 2164, 2170, 2179, 2180, or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to SEQ ID NO: 2164 or 2170.


In some embodiments, the CAR has the sequence of amino acids set forth in any of SEQ ID NOS: 2166, 2171, 2172, 2173, 2159, 2160, 2162, or 2163 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to any one of SEQ ID NOS: 2166, 2171, 2172, 2173, 2159, 2160, 2162 or 2163. In some embodiments, the CAR is encoded by a sequence of nucleotides set forth in SEQ ID NO: 2175 or 2161 or a sequence of amino acids that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% or more sequence identity to any one of SEQ ID NO: 2175 or 2161.


In another embodiment, the engineered T-cell possesses a TCR, including a recombinant or engineered TCR. In some embodiments, the TCR can be a native TCR. Those of skill in the art will recognize that generally native mammalian T-cell receptors comprise an alpha and a beta chain (or a gamma and a delta chain) involved in antigen specific recognition and binding. In some embodiments, the TCR is an engineered TCR that is modified. In some embodiments, the TCR of an engineered T-cell specifically binds to a tumor associated or tumor specific antigen presented by an APC.


In some embodiments, nucleic acids encoding the immunomodulatory protein, such as transmembrane immunomodulatory polypeptides or secretable immunomodulatory polypeptides, are incorporated into engineered cells, such as engineered T cells or engineered APCs, by a variety of strategies such as those employed for recombinant host cells. A variety of methods to introduce a DNA construct into primary T cells are known in the art. In some embodiments, viral transduction or plasmid electroporation are employed. In typical embodiments, the nucleic acid molecule encoding the immunomodulatory protein, or the expression vector, comprises a signal peptide that causes the expressed immunomodulatory proteins to be secreted from the cell. In some embodiments, a nucleic acid encoding an immunomodulatory protein of the invention is sub-cloned into a viral vector, such as a retroviral vector, which allows expression in the host mammalian cell. The expression vector can be introduced into a mammalian host cell and, under host cell culture conditions, the immunomodulatory protein is expressed and secreted.


In an exemplary example, primary T cells can be purified ex vivo (CD4 cells or CD8 cells or both) and stimulated with an activation protocol consisting of various TCR/CD28 agonists, such as anti-CD3/anti-CD28 coated beads. After a 2 or 3 day activation process, the DNA vector encoding the immunomodulatory protein of the present invention can be stably introduced into the primary T cells through art standard lentiviral or retroviral transduction protocols or plasmid electroporation strategies. Cells can be monitored for immunomodulatory protein expression and secretion by, for example, using anti-epitope tag or antibodies that cross-react with native parental molecule and affinity modified variant.


Upon immunomodulatory protein expression and secretion, the engineered T cell can be assayed for improved function by a variety of means. The engineered CAR or TCR co-expression can be validated to show that this part of the engineered T cell was not significantly impacted by the expression and secretion of the immunomodulatory construct. Once validated, standard in vitro cytotoxicity, proliferation, or cytokine assays can be used to assess the function of the engineered cells. Exemplary standard endpoints are percent lysis of the tumor line, proliferation of the engineered T-cell, or IFN-gamma protein expression in culture supernatants. An engineered construct which results in statistically significant increased lysis of tumor line, increased proliferation of the engineered T-cell, or increased IFN-gamma expression over the control construct can be selected for. Additionally, non-engineered cells, such as native primary or endogenous T-cells, could also be incorporated into the same in vitro assay to measure the ability of the immunomodulatory protein construct expressed and secreted by the engineered cells, such as engineered T-cells, to modulate activity, including, in some cases, to activate and generate effector function in bystander, native T-cells. Increased expression of activation markers such as CD69, CD44, or CD62L could be monitored on endogenous T cells, and increased proliferation and/or cytokine production could indicate desired activity of the immunomodulatory protein expressed and secreted by the engineered T cells.


In some embodiments, the similar assays can be used to compare the function of engineered T cells containing the CAR or TCR alone to those containing the CAR or TCR and a expressing and secreting an immunomodulatory protein. Typically, these in vitro assays are performed by plating various ratios of the engineered T cell and a “tumor” cell line containing the cognate CAR or TCR antigen together in culture. Standard endpoints are percent lysis of the tumor line, proliferation of the engineered T cell, or IFN-gamma production in culture supernatants. An engineered cell which resulted in statistically significant increased lysis of tumor line, increased proliferation of the engineered T cell, or increased IFN-gamma production over the same TCR or CAR construct alone can be selected for. Engineered human T cells can be analyzed in immunocompromised mice, like the NSG strain, which lacks mouse T, NK, and B cells. Engineered human T cells in which the CAR or TCR binds a target cognate binding partner on the xenograft and is co-expressed with the immunomodulatory protein containing an affinity-modified IgSF domain (which is also secreted) can be adoptively transferred in vivo at different cell numbers and ratios compared to the xenograft. For example, engraftment of CD19+ leukemia tumor lines containing a luciferase/GFP vector can be monitored through bioluminescence or ex vivo by flow cytometry. In a common embodiment, the xenograft is introduced into the murine model, followed by the engineered T cells several days later. Engineered T cells that express and secrete an immunomodulatory protein can be assayed for increased survival, tumor clearance, or expanded engineered T cells numbers relative to engineered T cells containing the CAR or TCR alone. As in the in vitro assay, endogenous, native (i.e., non-engineered) human T cells could be co-adoptively transferred to look for successful epitope spreading in that population, resulting in better survival or tumor clearance.


A. Exemplary Functional Activities and Features


In some aspects, engineered cells, such as engineered lymphocytes (e.g. tumor infiltrating lymphocytes, T cells or NK cells) or myeloid cells (e.g. antigen presenting cells), exhibit one or more desirable features or activities.


In some embodiments, when expressed on or from cells, the affinity-modified IgSF domain of the immunomodulatory protein specifically binds to at least one cognate binding partner expressed on a mammalian cell. In some embodiments, the mammalian cell is an autologous or allogeneic mouse, rat, cynomolgus monkey, or human cell. In some aspects, the mammalian cell can include such embodiments as an antigen presenting cell (APC), a tumor cell, or a T-cell. In some embodiments, the tumor cell is a mouse, rat, cynomolgus monkey, or human tumor cell.


An immunomodulatory protein can comprise one or multiple (e.g., 2, 3, or 4) affinity modified IgSF domains. Thus, in some embodiments the immunomodulatory protein binds to no more than one cognate binding partner on the mammalian cell. In some embodiments, an affinity-modified IgSF domain of an immunomodulatory protein specifically binds to no more than one cognate binding partner on the mammalian cell. Alternatively, in some embodiments, an immunomodulatory protein specifically binds to at least two, three or four, or exactly two, three, or four, cognate binding partners expressed on a mammalian cell. In some embodiments, the immunomodulatory protein specifically binds to no more than one cell surface cognate binding partners. Specific binding of the immunomodulatory protein containing one or more affinity-modified IgSF domains to the cognate binding partner on a mammalian cell modulates immunological activity of the mammalian cell. Specific binding by and between an affinity-modified IgSF domain and a mammalian cell cognate binding partner can be specific binding in cis arrangement (i.e., specific binding on the same cell) or in trans arrangement (i.e., specific binding on different cells) or in both cis and trans arrangement. Immunological activity of the cell can be increased as evidenced by increased, e.g., cell survival, cell proliferation, cytokine production, or T-cell cytotoxicity. In alternative embodiments, the immunological activity of the cell is attenuated as evidenced by a decrease of cell survival, cell proliferation, cytokine production, or T-cell cytotoxicity.


In some embodiments, at least one affinity-modified IgSF domain present in an immunomodulatory protein specifically binds to at least one cell surface cognate binding partner expressed on a mammalian cell and in which modulation of immunological activity is desired. In some embodiments, the cognate binding partner to which the affinity modified IgSF domain specifically binds is the native cognate binding partner of the wild-type IgSF family member or wild-type IgSF domain that has been affinity modified. In some embodiments, the specific binding increases and/or attenuates activity a mammalian cell expressing a cognate binding partner to which the affinity modified IgSF domain exhibits improved binding. Thus, by increasing specific binding affinity, the provided immunomodulatory proteins can either increase or attenuate immunological activity of a mammalian cell. In some embodiments, the specific binding modulates, such as increases, immunological activity of the engineered cell with the immunomodulatory protein.


In some embodiments, the cognate binding partner expressed on the mammalian cell is a mammalian IgSF member. The mammalian cell is, in some embodiments, an antigen presenting cell (APC), a lymphocyte, or a tumor cell. In some embodiments, the lymphocyte is a tumor infiltrating lymphocyte (TIL), an engineered or native T-cell, or an engineered or native NK cell. In some embodiments, the cognate binding partner of the affinity modified IgSF domain is a native human IgSF member. In some embodiments, the cognate binding partner is a “cell surface cognate binding partner” as indicated in Table 1.


In some embodiments, an immunomodulatory protein comprising an affinity-modified IgSF when expressed and secreted by an immune cell (e.g. a lymphocyte such as a T-cell) can specifically bind at least one cognate binding partner expressed on a second immune cells, e.g. a lymphocyte such as a T-cell. The cognate binding partner on the second immune cells, e.g. second T-cell, can be an inhibitory cognate binding partner or a stimulatory cognate binding partner. Exemplary cognate binding partners include cell surface receptors or ligands. Examples of inhibitory receptors/ligands include PD-1/PD-L1, PD-L2, CTLA-4/B7-1/B7-2, BTLA/HVEM, LAGS/MHC class II, TIGIT/PVR, TIM-3/CEACAM-1/GAL9 and VSIG8/VISTA. Examples of stimulatory receptors/ligands include CD28/B7-1/B7-2, ICOS/ICOSL, and CD226/PVR.


In a particular embodiment, the immunomodulatory protein is expressed and secreted by a T cell and comprises an affinity modified IgSF domain that specifically binds to a cognate binding partner expressed on a T-cell. In some embodiments the first and second T-cells are separate T-cells and in this embodiment the immunomodulatory protein and cognate binding partner are in trans to each other. In some embodiments, the immunomodulatory protein and cognate binding partner are expressed on the same T-cell (wherein the immunomodulatory protein is secreted) and are cis to each other. In some embodiments, at least one of the T-cells is a native T-cell or an engineered T-cell. In some embodiments, the engineered T-cell is a chimeric antigen receptor (CAR) T-cell or a T-cell receptor (TCR) engineered T-cell.


In some embodiments, a immunomodulatory protein comprises an affinity modified IgSF domain with increased affinity to a cell surface receptor to stimulate an increase in receptor signal transduction. Stimulating an increase in receptor signaling can in some embodiments increase immunological activity of that cell if, for example, the receptor is a stimulatory receptor that works to mediate those effects. In some cases, the inflammatory activity of the cell in which receptor signaling is stimulated is increased. In some embodiments, the immunomodulatory protein increases the activity of a stimulatory receptor. In such examples, an IgSF domain of an immunomodulatory protein can be affinity modified to increase the specific binding affinity to the native cognate binding partner on a mammalian cell, which, in some cases, is a stimulatory receptor. In some embodiments, the stimulatory receptor is expressed on T cells. In certain embodiments, the affinity modified IgSF domain of the immunomodulatory protein, such as is expressed and secreted by an engineered cell (e.g. a first T cell), specifically binds to a stimulatory cognate binding partner expressed on a T cell (e.g. a second T cell) with increased affinity (relative to the non-affinity modified IgSF domain as a control). In certain embodiments, the affinity modified IgSF domain of the immunomodulatory protein specifically binds to a stimulatory cognate binding partner expressed on a T cell (e.g. second T cell) and increases immunomodulatory activity of the T-cell. In some embodiments, the affinity modified IgSF domain of an immunomodulatory protein binds to a stimulatory cognate binding partner on the T cell (e.g. second T-cell) with increased affinity and increases immunomodulatory activity of the T-cell.


In some embodiments, the stimulatory receptor is CD28, ICOS or CD226 and the immunomodulatory protein containing an affinity-modified IgSF domain that exhibits increased binding affinity to one of CD28, ICOS or CD226 compared to a protein containing a wild-type IgSF domain. In some embodiments, the affinity modified IgSF domain is an affinity modified domain of CD80 (B7-1). In some embodiments, an affinity modified CD80 (B7-1) IgSF domain of an immunomodulatory protein of the present invention is expressed on a first T-cell and is affinity modified to bind with increased affinity to the stimulatory cognate binding partner CD28 on the second T-cell. In some embodiments, the affinity modified IgSF domain is an affinity modified domain of ICOSL. In specific embodiments, the affinity modified IgSF domain is an affinity modified ICOSL (inducible costimulator ligand) domain and the stimulatory cognate binding partner is at least one of: ICOS (inducible costimulator) or CD28. In some embodiments, the ICOSL domain is affinity-modified to specifically bind to both ICOS and CD28. In some embodiments, ICOSL is affinity modified to specifically bind to either ICOS or to CD28 but not both. In some embodiments, binding affinity to one of ICOS or CD28 is increased while binding affinity to the other is attenuated. In some embodiments, the affinity modified IgSF domain is an affinity modified CD155 and the activating cognate ligand is CD226. In some embodiment, the affinity modified IgSF domain is an affinity modified CD112 and the activating cognate ligand is CD226.


In some methods of the present invention, the immunomodulatory protein attenuates the activity of an inhibitory receptor. In some cases, the increased binding affinity of the immunomodulatory protein to a cognate cell surface molecule results in inhibition of specific binding between native cognate binding partners on mammalian cells. The greater affinity for that native cognate binding partner (relative to the competing affinity of the native IgSF member) attenuates specific binding affinity of native molecule to its cognate binding partner. Those of skill in the art will appreciate that antagonizing an inhibitory receptor signaling can in some embodiments attenuate immunological activity of that cell if, for example, the receptor is an inhibitory receptor that serves to cause those cellular effects. In some embodiments, one or more activities between the inhibitory receptor and its ligand from among CD155/TIGIT, CD112/TIGIT, CD80/CTLA-4, ICOSL/CTLA-4. PD-L1/PD-1 or PD-:2/PD-lis blocked by the immunomodulatory protein.


Thus, in some embodiments, an immunomodulatory protein can be used to stimulate a cell for which the immunomodulatory protein is not expressed and secreted (i.e., the trans cell) while attenuating inhibition of the cell by which the immunomodulatory protein is expressed and secreted (the cis cell). For example, in some embodiments, the immunomodulatory protein comprises at least one affinity-modified IgSF domain, and in some cases at least two affinity modified domains, that results in increased binding affinity to at least two cell surface cognate binding partners. In some embodiments, a first cognate binding partner is a stimulatory receptor and the second cell surface cognate binding partner is an inhibitory ligand of an inhibitory receptor. In some embodiments, binding of the affinity-modified domain to the inhibitory ligand competitively inhibits binding of the inhibitory ligand to the inhibitory receptor. In some embodiments, the stimulatory receptor and inhibitory receptor can independently be expressed on immune cells, such as T cells or antigen presenting cells. In some embodiments the stimulatory receptor is expressed on lymphocytes, such as T cells. In some embodiments, the stimulatory receptor is CD28, ICOS, or CD226 and/or the ligand of the stimulatory receptor is ICOSL. In some embodiments, the inhibitory receptor is expressed on the engineered cells, such as an engineered T-cell. In some embodiments, the inhibitory receptor is PD-1, CTLA-4, LAG-3, TIGIT, CD96, CD112R, BTLA, CD160, TIM-3, VSIG3, or VSIG8 and/or the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, CD112, CD155, HVEM, MHC class II, PVR, CEACAM-1, GAL9 or VISTA (see e.g. Table 1). In some embodiments, the inhibitory cognate binding partner is PD-L1 or PD-L2.


In some embodiments, an immunomodulatory protein can be used to attenuate inhibition of the cell that expresses and secretes the immunomodulatory protein, such as a T cell that expresses and secretes the immunomodulatory protein. For example, an immunomodulatory protein expressed and secreted by a T cell (e.g. first T cell) can comprise an affinity-modified IgSF domain that inhibits specific binding between a cognate binding partner on a second T cell. In some embodiments, a ligand of an inhibitory receptor is affinity-modified and, when secreted from an engineered cell, antagonizes or inhibits its inhibitory receptor. In some embodiments, the affinity-modified ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, HVEM, MHC class II, PVR (e.g. CD112 or CD155), CEACAM-1, GAL9 or VISTA (see e.g. Table 1). In some embodiments, the affinity-modified domain that is a ligand of the inhibitory receptor retains or exhibits increased binding for another receptor, such as a stimulatory receptor. For example, the affinity-modified domain can be CD155 or CD112 that is affinity-modified to exhibit reduced binding to CD226 but that retains or exhibits increased binding to TIGIT, and, in some cases, CD112R. In such embodiments, the affinity-modified CD155 or CD112 can antagonize TIGIT when expressed and secreted from a cell.


In some cases, this embodiment can be used independently or in conjunction with embodiments wherein an affinity-modified IgSF domain of the invention is expressed and secreted by a first T cell and specifically binds at least one stimulatory cognate binding partner expressed on a second T cell and increases immunological activity in the second T cell. By this mechanism, an increased immunomodulatory response is generated in the second T cell by specific binding of the immunomodulatory protein expressed on the first T cell to a stimulatory cognate binding partner on the second cell; and the second T cell is inhibited from attenuating the immunomodulatory activity of the first T cell by specific binding of an affinity modified IgSF domain expressed on the first T cell that inhibits specific binding by and between a cognate binding partner on the second T cell and an inhibitory cognate binding partner expressed on the first T cell. The T cells used in this and the preceding embodiments are generally murine or human T-cells although other mammalian T-cells can be employed. Often, cytotoxic T-cells (CTL) are used.


As previously noted, in some embodiments an immunomodulatory protein of the present invention is expressed on a first T-cell and comprises an affinity-modified IgSF domain that specifically binds to a stimulatory cognate binding partner (e.g. stimulatory receptor) on a second T cell while also inhibiting specific binding between a native cognate binding partner (e.g. inhibitory ligand) on the second T-cell to its inhibitory native cognate binding partner (e.g. inhibitory receptor) on the first T-cell. Inhibition of specific binding between the cognate binding partner on the second T-cell to the cognate binding partner on the first T-cell can be achieved by competitive binding of an affinity-modified IgSF domain with at least one of the two native cognate binding partners such that their mutual binding is interfered with. Typically, the IgSF domain is affinity modified to have a higher binding affinity to its cognate binding partner than the native cognate binding partners have to each other. In some embodiments of this design, an immunomodulatory protein can comprise an affinity-modified IgSF domain that binds to both the inhibitory and stimulatory cognate binding partners. Thus, in this embodiment the affinity-modified IgSF has dual binding capability. In some embodiments, an immunomodulatory protein comprises a first affinity-modified IgSF domain that binds a cognate binding partner on the first T-cell and a second affinity-modified IgSF domain that inhibits specific binding by and between the cognate binding partners on the first and second T-cells.


In yet another embodiment, the affinity-modified IgSF domain that binds to the stimulatory cognate binding partner on the first T-cell is on a first immunomodulatory protein and the affinity-modified IgSF domain that inhibits specific binding by and between the cognate binding partners on the first and second T cells is on a second immunomodulatory protein. In this embodiment, the first and second immunomodulatory proteins are different polypeptide chains. In some embodiments, the first affinity-modified IgSF domain and the second affinity-modified IgSF domain are the identical affinity-modified IgSF domain. For example, in a specific embodiment the ICOSL (inducible costimulator ligand) IgSF domain (e.g. affinity modified IgV domain) is affinity modified to specifically bind with increased affinity to both ICOS and CD28. In some embodiments, the affinity modified IgSF domain is an affinity modified ICOSL IgSF domain (e.g. affinity modified IgV domain) with increased affinity to both ICOS and CD28, or decreased affinity to one of or both of: ICOS and CD28.


In some embodiments, the immunomodulatory protein results in inhibition of specific binding by and between native cognate binding partners. In some embodiments, this can be achieved by an affinity-modified IgSF domain having greater affinity for one or both native cognate binding partners thereby competitively inhibiting the specific binding by and between these cognate binding partners.


In some embodiments, the immunomodulatory protein comprises an affinity-modified IgSF domain that is an affinity-modified CD155 IgSF domain with increased affinity to CD226 and attenuated affinity to TIGIT (T-cell immunoreceptor with Ig and ITIM domains).


In some embodiments, the immunomodulatory protein (e.g. expressed on a first T cell) comprises an affinity-modified CD80 (B7-1) IgSF domain that is affinity modified to bind with increased affinity to the stimulatory cognate binding partner CD28 (e.g. on a second T-cell). Additionally, in this embodiment, the affinity-modified CD80 (B7-1) IgSF domain can bind with increased affinity to PD-L1 (e.g. expressed on the second T-cell) and inhibit specific binding to its cognate binding partner PD-1 (e.g. expressed on the first T-cell). In yet a further addition either of the preceding embodiments, the affinity-modified CD80 (B7-1) IgSF domain can be affinity modified such that it does not substantially specifically bind to CTLA-4 or binds with attenuated affinity and therefore is not significantly inhibited in its specific binding to the stimulatory cognate binding partner CD28 by CTLA-4.


In some embodiments, an immunomodulatory protein is used as a decoy cognate binding partner to inhibit specific binding by and between native cognate binding partners, at least one of which comprises an IgSF family member. In some cases, specific binding of an immunomodulatory protein comprising an affinity-modified IgSF domain with one of the native cognate binding partners inhibits mutual specific binding by and between the native cognate binding partners (e.g. native receptor and ligand pairs). Thus, in some embodiments, immunomodulatory proteins can attenuate specific binding by means of competitive or non-competitive binding. In some embodiments, the native cognate binding partner is a cell surface receptor, which can be a stimulatory receptor or an inhibitory receptor. Embodiments wherein specific binding of the affinity modified IgSF domain of a cognate binding partner increases or attenuates immunological activity of the T-cell are included within the scope of the invention.


In some embodiments, a native cognate binding partner is an inhibitory cognate binding partner that acts to attenuate immunological activity when specifically bound by its native cognate binding partner. For example, a native cell surface cognate binding partner expressed on an antigen presenting cell (APC) or a mammalian tumor cell can specifically bind a native inhibitory cognate binding partner on an NK cell or a lymphocyte such as a T-cell. Specific binding to the inhibitory cognate binding partner acts to attenuate immunomodulatory activity of the NK cell or lymphocyte on which the inhibitory cognate binding partner is expressed.


In some embodiments, an inhibitory cognate binding partner is an inhibitory receptor. In some embodiments, the inhibitory cognate binding partner is an ITIM (immunoreceptor tyrosine-based inhibition motif) containing inhibitory cognate binding partner. The ITIM motif is found in the endodomain of many inhibitory receptors of the immune system (Cell Signal, 16 (4): 435-456, 2004). In some embodiments, the affinity-modified IgSF domain is an affinity-modified form of a wild-type inhibitory receptor IgSF domain that results in greater affinity of the affinity-modified IgSF domain of the immunomodulatory protein for its native cognate binding partner than the wild-type inhibitory receptor for the native cognate binding partner. Thus, in these embodiments a immunomodulatory protein can attenuate the inhibitory response of ITIM motif receptors by specific binding of the immunomodulatory protein affinity-modified IgSF domain to its native IgSF domain cognate binding partner, such as specific binding of the immunomodulatory protein affinity-modified IgSF domain to the ITIM containing inhibitory receptor. As an example, an ITIM containing cognate binding partner is PD-1. Typically, PD-1 is the inhibitory receptor that is specifically bound to the inhibitory ligand PD-1. Upon specific binding of PD-L1 to PD-1, PD-1 is involved in inhibiting T-cell activation via signal transduction from the ITIM domain.


In some embodiments, the inhibitory receptor cognate binding partner is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8. In some embodiments, the immunomodulatory protein contains an affinity-modified IgSF domain that is an affinity-modified IgSF domain of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, or BTLA that binds with greater affinity to the native inhibitory ligand of the inhibitory receptor than the wild-type inhibitory receptor (see Table 1 for ligand binding partners of exemplary inhibitory receptors). In some embodiments, an immunomodulatory protein can comprise an affinity-modified PD-1 IgSF domain that binds with greater affinity to PD-L1 than wild-type PD-1. Specific binding can be achieved by competitive or non-competitive binding and are specific embodiments of the invention. Competitive binding by and between the affinity-modified IgSF domain and the cognate binding partner (i.e. inhibitory receptor, e.g. PD-1) inhibits its binding to its native ligand cognate binding partner (e.g., PD-L1). In some embodiments, the immunomodulatory protein of this embodiment substantially lacks the signal transduction mechanism of the wild-type inhibitory receptor and therefore does not itself induce an inhibitory response.


VII. Infectious Agents

Also provided are infectious agents that contain nucleic acids encoding any of the immunomodulatory proteins containing an affinity-modified IgSF domain, including secretable or transmembrane immunomodulatory proteins described herein. In some embodiments, such infectious agents can deliver the nucleic acids encoding the immunomodulatory polypeptides described herein to a target cell in a subject, e.g., immune cell and/or antigen-presenting cell (APC) or tumor cell in a subject. Also provided are nucleic acids contained in such infectious agents, and/or nucleic acids for generation or modification of such infectious agents, such as vectors and/or plasmids, and compositions containing such infectious agents.


In some embodiments, the infectious agent is a microorganism or a microbe. In some embodiments, the infectious agent is a virus or a bacterium. In some embodiments, the infectious agent is a virus. In some embodiments, the infectious agent is a bacterium. In some embodiments, such infectious agents can deliver nucleic acid sequences encoding any of the immunomodulatory proteins, including secretable or transmembrane immunomodulatory proteins, described herein. Thus, in some embodiments, the cell in a subject that is infected or contacted by the infectious agents can be rendered to express on the cell surface or secrete, the immunomodulatory polypeptides. In some embodiments, the infectious agent can also deliver one or more other therapeutics or nucleic acids encoding other therapeutics to the cell and/or to an environment within the subject. In some embodiments, other therapeutics that can be delivered by the infectious agents include cytokines or other immunomodulatory molecules.


In some embodiments, the infectious agent, e.g., virus or bacteria, contains nucleic acid sequences that encode any of the immunomodulatory proteins, including secretable or transmembrane immunomodulatory proteins, described herein, and by virtue of contact and/or infection of a cell in the subject, the cell expresses the immunomodulatory proteins, including secretable or transmembrane immunomodulatory proteins, encoded by the nucleic acid sequences contained in the infectious agent. In some embodiments, the infectious agent can be administered to the subject. In some embodiments, the infectious agent can be contacted with cells from the subject ex vivo.


In some embodiments, the immunomodulatory protein is a transmembrane immunomodulatory proteins that is expressed by the cell infected by the infectious agent and is surface expressed. In some embodiments, the immunomodulatory protein is a secretable immunomodulatory protein that is expressed by the cell infected by the infectious agent and is expressed and secreted from the cell. The transmembrane immunomodulatory protein or secreted immunomodulatory protein can be any described herein.


In some embodiments, the cells in the subject that are targeted by the infectious agent include a tumor cell, an immune cell, and/or an antigen-presenting cell (APC). In some embodiments, the infectious agent targets a cell in the tumor microenvironment (TME). In some embodiments, the infectious agent delivers the nucleic acids encoding the immunomodulatory protein, including secretable or transmembrane immunomodulatory proteins, to an appropriate cell (for example, an APC, such as a cell that displays a peptide/MHC complex on its cell surface, such as a dendritic cell) or tissue (e.g., lymphoid tissue) that will induce and/or augment the desired effect, e.g., immunomodulation and/or a specific cell-medicated immune response, e.g., CD4 and/or CD8 T cell response, which CD8 T cell response may include a cytotoxic T cell (CTL) response. In some embodiments, the infectious agent targets an APC, such as a dendritic cell (DC). In some embodiments, the nucleic acid molecule delivered by the infectious agents described herein include appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequences encoding the variant immunomodulatory polypeptides, in a particular target cell, e.g., regulatory elements such as promoters.


In some embodiments, the infectious agent that contains nucleic acid sequences encoding the immunomodulatory polypeptides can also contain nucleic acid sequences that encode one or more additional gene products, e.g., cytokines, prodrug converting enzymes, cytotoxins and/or detectable gene products. For example, in some embodiments, the infectious agent is an oncolytic virus and the virus can include nucleic acid sequences encoding additional therapeutic gene products (see, e.g., Kirn et al., (2009) Nat Rev Cancer 9:64-71; Garcia-Aragoncillo et al., (2010) Curr Opin Mol Ther 12:403-411; see U.S. Pat. Nos. 7,588,767, 7,588,771, 7,662,398 and 7,754,221 and U.S. Pat. Publ. Nos. 2007/0202572, 2007/0212727, 2010/0062016, 2009/0098529, 2009/0053244, 2009/0155287, 2009/0117034, 2010/0233078, 2009/0162288, 2010/0196325, 2009/0136917 and 2011/0064650. In some embodiments, the additional gene product can be a therapeutic gene product that can result in death of the target cell (e.g., tumor cell) or gene products that can augment or boost or regulate an immune response (e.g., cytokine). Exemplary gene products also include among an anticancer agent, an anti-metastatic agent, an antiangiogenic agent, an immunomodulatory molecule, an immune checkpoint inhibitor, an antibody, a cytokine, a growth factor, an antigen, a cytotoxic gene product, a pro-apoptotic gene product, an anti-apoptotic gene product, a cell matrix degradative gene, genes for tissue regeneration and reprogramming human somatic cells to pluripotency, and other genes described herein or known to one of skill in the art. In some embodiments, the additional gene product is Granulocyte-macrophage colony-stimulating factor (GM-CSF).


A. Viruses


In some embodiments, the infectious agent is a virus. In some embodiments, the infectious agent is an oncolytic virus, or a virus that targets particular cells, e.g., immune cells. In some embodiments, the infectious agent targets a tumor cell and/or cancer cell in the subject. In some embodiments, the infectious agent targets an immune cell or an antigen-presenting cell (APC).


In some embodiments, the infectious agent is an oncolytic virus. Oncolytic viruses are viruses that accumulate in tumor cells and replicate in tumor cells. By virtue of replication in the cells, and optional delivery of nucleic acids encoding immunomodulatory polypeptides described herein, tumor cells are lysed, and the tumor shrinks and can be eliminated. Oncolytic viruses can also have a broad host and cell type range. For example, oncolytic viruses can accumulate in immunoprivileged cells or immunoprivileged tissues, including tumors and/or metastases, and also including wounded tissues and cells, thus allowing the delivery and expression of nucleic acids encoding the immunomodulatory polypeptides described herein in a broad range of cell types. Oncolytic viruses can also replicate in a tumor cell specific manner, resulting in tumor cell lysis and efficient tumor regression.


Exemplary oncolytic viruses include adenoviruses, adeno-associated viruses, herpes viruses, Herpes Simplex Virus, Vesticular Stomatic virus, Reovirus, Newcastle Disease virus, parvovirus, measles virus, vesticular stomatitis virus (VSV), Coxsackie virus and Vaccinia virus. In some embodiments, oncolytic viruses can specifically colonize solid tumors, while not infecting other organs, and can be used as an infectious agent to deliver the nucleic acids encoding the immunomodulatory polypeptides described herein to such solid tumors.


Oncolytic viruses for use in delivering the nucleic acids encoding variant ICOSL polypeptides or immunomodulatory polypeptides described herein, can be any of those known to one of skill in the art and include, for example, vesicular stomatitis virus, see, e.g., U.S. Pat. Nos. 7,731,974, 7,153,510, 6,653,103 and U.S. Pat. Pub. Nos. 2010/0178684, 2010/0172877, 2010/0113567, 2007/0098743, 20050260601, 20050220818 and EP Pat. Nos. 1385466, 1606411 and 1520175; herpes simplex virus, see, e.g., U.S. Pat. Nos. 7,897,146, 7,731,952, 7,550,296, 7,537,924, 6,723,316, 6,428,968 and U.S. Pat. Pub. Nos., 2014/0154216, 2011/0177032, 2011/0158948, 2010/0092515, 2009/0274728, 2009/0285860, 2009/0215147, 2009/0010889, 2007/0110720, 2006/0039894, 2004/0009604, 2004/0063094, International Patent Pub. Nos., WO 2007/052029, WO 1999/038955; retroviruses, see, e.g., U.S. Pat. Nos. 6,689,871, 6,635,472, 5,851,529, 5,716,826, 5,716,613 and U.S. Pat. Pub. No. 20110212530; vaccinia viruses, see, e.g., 2016/0339066, and adeno-associated viruses, see, e.g., U.S. Pat. Nos. 8,007,780, 7,968,340, 7,943,374, 7,906,111, 7,927,585, 7,811,814, 7,662,627, 7,241,447, 7,238,526, 7,172,893, 7,033,826, 7,001,765, 6,897,045, and 6,632,670.


Oncolytic viruses also include viruses that have been genetically altered to attenuate their virulence, to improve their safety profile, enhance their tumor specificity, and they have also been equipped with additional genes, for example cytotoxins, cytokines, prodrug converting enzymes to improve the overall efficacy of the viruses (see, e.g., Kim et al., (2009) Nat Rev Cancer 9:64-71; Garcia-Aragoncillo et al., (2010) Curr Opin Mol Ther 12:403-411; see U.S. Pat. Nos. 7,588,767, 7,588,771, 7,662,398 and 7,754,221 and U.S. Pat. Publ. Nos. 2007/0202572, 2007/0212727, 2010/0062016, 2009/0098529, 2009/0053244, 2009/0155287, 2009/0117034, 2010/0233078, 2009/0162288, 2010/0196325, 2009/0136917 and 2011/0064650). In some embodiments, the oncolytic viruses can be those that have been modified so that they selectively replicate in cancerous cells, and, thus, are oncolytic. For example, the oncolytic virus is an adenovirus that has been engineered to have modified tropism for tumor therapy and also as gene therapy vectors. Exemplary of such is ONYX-015, H101 and Ad5ACR (Hallden and Portella (2012) Expert Opin Ther Targets, 16:945-58) and TNFerade (McLoughlin et al. (2005) Ann. Surg. Oncol., 12:825-30), or a conditionally replicative adenovirus Oncorine®.


In some embodiments, the infectious agent is a modified herpes simplex virus. In some embodiments, the infectious agent is a modified version of Talimogene laherparepvec (also known as T-Vec, Imlygic or OncoVex GM-CSF), that is modified to contain nucleic acids encoding any of the immunomodulatory polypeptides described herein. In some embodiments, the infectious agent is a modified herpes simplex virus that is described, e.g., in WO 2007/052029, WO 1999/038955, US 2004/0063094, US 2014/0154216, or, variants thereof.


In some embodiments, the infectious agent is a virus that targets a particular type of cells in a subject that is administered the virus, e.g., a virus that targets immune cells or antigen-presenting cells (APCs). Dendritic cells (DCs) are essential APCs for the initiation and control of immune responses. DCs can capture and process antigens, migrate from the periphery to a lymphoid organ, and present the antigens to resting T cells in a major histocompatibility complex (MHC)-restricted fashion. In some embodiments, the infectious agent is a virus that specifically can target DCs to deliver nucleic acids encoding the immunomodulatory polypeptides for expression in DCs. In some embodiments, the virus is a lentivirus or a variant or derivative thereof, such as an integration-deficient lentiviral vector. In some embodiments, the virus is a lentivirus that is pseudotyped to efficiently bind to and productively infect cells expressing the cell surface marker dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), such as DCs. In some embodiments, the virus is a lentivirus pseudotyped with a Sindbis virus E2 glycoprotein or modified form thereof, such as those described in WO 2013/149167. In some embodiments, the virus allows for delivery and expression of a sequence of interest (e.g., a nucleic acid encoding any of the immunomodulatory polypeptides described herein) to a DC. In some embodiments, the virus includes those described in WO 2008/011636, US 2011/0064763, Tareen et al. (2014) Mol. Ther., 22:575-587, or variants thereof. Exemplary of a dendritic cell-tropic vector platform is ZVex™.


B. Bacteria


In some embodiments, the infectious agent is a bacterium. For example, in some embodiments, the bacteria can deliver nucleic acids encoding any of the immunomodulatory polypeptides described herein to a target cell in the subject, such as a tumor cell, an immune cell, an antigen-presenting cell and/or a phagocytic cell. In some embodiments, the bacterium can be preferentially targeted to a specific environment within a subject, such as a tumor microenvironment (TME), for expression and/or secretion of the immunomodulatory polypeptides and/or to target specific cells in the environment for expression of the variant immunomodulatory polypeptides.


In some embodiments, the bacterium delivers the nucleic acids to the cells via bacterial-mediated transfer of plasmid DNA to mammalian cells (also referred to as “bactofection”). For example, in some embodiments, delivery of genetic material is achieved through entry of the entire bacterium into target cells. In some embodiments, spontaneous or induced bacterial lysis can lead to the release of plasmid for subsequent eukaryotic cell expression. In some embodiments, the bacterium can deliver nucleic acids to non-phagocytic mammalian cells (e.g., tumor cells) and/or to phagocytic cells, e.g., certain immune cells and/or APCs. In some embodiments, the nucleic acids delivered by the bacterium can be transferred to the nucleus of the cell in the subject for expression. In some embodiments, the nucleic acids also include appropriate nucleic acid sequences necessary for the expression of the operably linked sequences encoding the variant immunomodulatory polypeptides in a particular host cell, e.g., regulatory elements such as promoters or enhancers. In some embodiments, the infectious agent that is a bacterium can deliver nucleic acids encoding the immunomodulatory proteins in the form of an RNA, such as a pre-made translation-competent RNA delivered to the cytoplasm of the target cell for translation by the target cell's machinery.


In some embodiments, the bacterium can replicate and lyse the target cells, e.g., tumor cells. In some embodiments, the bacterium can contain and/or release nucleic acid sequences and/or gene products in the cytoplasm of the target cells, thereby killing the target cell, e.g., tumor cell. In some embodiments, the infectious agent is bacterium that can replicate specifically in a particular environment in the subject, e.g., tumor microenvironment (TME). For example, in some embodiments, the bacterium can replicate specifically in anaerobic or hypoxic microenvironments. In some embodiments, conditions or factors present in particular environments, e.g., aspartate, serine, citrate, ribose or galactose produced by cells in the TME, can act as chemoattractants to attract the bacterium to the environment. In some embodiments, the bacterium can express and/or secrete the immunomodulatory proteins described herein in the environment, e.g., TME.


In some embodiments, the infectious agent is a bacterium that is a Listeria sp., a Bifidobacterium sp., an Escherichia sp., a Clostridium sp., a Salmonella sp., a Shigella sp., a Vibrio sp. or a Yersinia sp. In some embodiments, the bacterium is selected from among one or more of Listeria monocytogenes, Salmonella typhimurium, Salmonella choleraesuis, Escherichia coli, Vibrio cholera, Clostridium perfringens, Clostridium butyricum, Clostridium novyi, Clostridium acetobutylicum, Bifidobacterium infantis, Bifidobacterium longum and Bifidobacterium adolescentis. In some embodiments, the bacterium is an engineered bacterium. In some embodiments, the bacterium is an engineered bacterium such as those described in, e.g., Seow and Wood (2009) Molecular Therapy 17(5):767-777; Baban et al. (2010) Bioengineered Bugs 1:6, 385-394; Patyar et al. (2010) J Biomed Sci 17:21; Tangney et al. (2010) Bioengineered Bugs 1:4, 284-287; van Pijkeren et al. (2010) Hum Gene Ther. 21(4):405-416; WO 2012/149364; WO 2014/198002; U.S. Pat. Nos. 9,103,831; 9,453,227; US 2014/0186401; US 2004/0146488; US 2011/0293705; US 2015/0359909 and EP 3020816. The bacterium can be modified to deliver nucleic acid sequences encoding any of the immunomodulatory polypeptides, conjugates and/or fusions provided herein, and/or to express such immunomodulatory polypeptides in the subject.


VIII. Compositions, Methods, and Therapeutic Applications

Provided herein are compositions and methods relating to the provided immunomodulatory proteins, engineered cells and infectious agents described herein for use in modulating immunological activity of a mammalian cell. The compositions can be used in associated methods to, for example, modulate immunological activity in an immunotherapy approach to the treatment of, for example, a mammalian cancer or, in other embodiments the treatment of autoimmune disorders. In some embodiments, the method comprises contacting an immunomodulatory protein (which may be secreted by an engineered cell) of the present invention with a mammalian cell under conditions that are permissive to specific binding of the affinity-modified IgSF domain and modulation of the immunological activity of the mammalian cell. The methods can be employed ex vivo or in vivo.


In some embodiments, the method of modulating immunological activity is achieved by expression and secretion of an immunomodulatory protein of the present invention by an immune cell, such as a lymphocyte (e.g., a T-cell or TIL) or NK cell engineered or infected to express and secrete the immunomodulatory protein. In some embodiments, the method of modulating immunological activity is achieved by expression and surface expression of an immunomodulatory protein of the present invention by an immune cell, such as a lymphocyte (e.g., a T-cell or TIL) or NK cell engineered or infected to express on their surface a transmembrane immunomodulatory protein. In some embodiments, a tumor cell can be infected, e.g. with an oncolytic virus, to express a secretable immunomodulatory protein or transmembrane immunomodulatory protein modulation of immune cells in the tumor environment. In some embodiments, the cell expressing and secreting the immunomodulatory protein is contacted with a mammalian cell such as an APC, a second lymphocyte or tumor cell under conditions that are permissive of specific binding of the affinity modified IgSF domain to a cognate binding partner on the mammalian cell such that immunological activity can be modulated in the mammalian cell.


In some embodiments, the method is conducted by adoptive cell transfer of engineered cells expressing and secreting the immunomodulatory protein (e.g., a T-cell) are infused back into the patient. In some embodiments, the method is conducted by adoptive cell transfer of engineered cells (e.g. T cells) expressing on their surface a transmembrane immunomodulatory protein.


Provided herein are methods of administering an effective amount of engineered cells configured to express and secrete or surface express an immunomodulatory proteins to a subject in need (for example a subject having a disease or disorder). The pharmaceutical compositions described herein can be used in a variety of therapeutic applications, such as the treatment of a disease. For example, in some embodiments the pharmaceutical composition is used to treat inflammatory or autoimmune disorders, cancer, organ transplantation, viral infections, and/or bacterial infections in a mammal. The pharmaceutical composition can modulate an immune response to treat the disease. For example, in some embodiments, the pharmaceutical composition stimulates the immune response, which can be useful, for example, in the treatment of cancer, viral infections, or bacterial infections. In some embodiments, the pharmaceutical composition suppresses an immune response, which can be useful in the treatment of inflammatory or autoimmune disorders, or organ transplantation.


The provided methods are believed to have utility in a variety of applications, including, but not limited to, e.g., in prophylactic or therapeutic methods for treating a variety of immune system diseases or conditions in a mammal in which modulation or regulation of the immune system and immune system responses is beneficial. For example, suppressing an immune response can be beneficial in prophylactic and/or therapeutic methods for inhibiting rejection of a tissue, cell, or organ transplant from a donor by a recipient. In a therapeutic context, the mammalian subject is typically one with an immune system disease or condition, and administration is conducted to prevent further progression of the disease or condition.


Cell compositions engineered to express and secrete immunomodulatory proteins of the present invention and associated methods can be used in immunotherapy applications. In some embodiments, cells isolated from a mammal, such as a mouse or human, and can be engineered to express and secrete or surface express an immunomodulatory protein. In some embodiments, the mammalian cell serving as a host cell for expression and secretion or surface expression of an immunomodulatory protein is a lymphocyte such as a tumor infiltrating lymphocyte (TIL), a natural killer (NK) cell, or a T-cell such as a CD8+ cytotoxic T lymphocyte or a CD4+ helper T lymphocyte. In some embodiments, the cells are autologous cells. In aspects of the provided method, the engineered cells are contacted, generally under physiological conditions, with a mammalian cell in which modulation of immunological activity is desired. For example, the mammalian cell can be a murine or human cell such as an antigen presenting cell or tumor cell. In some embodiments, the engineered cells are autologous cells. In other embodiments, the cells are allogeneic. Cells can be contacted in vivo or ex vivo. In some embodiments, the engineered cells are administered to the subject, such as by infusion. Thus, composition and methods can be used in adoptive cell transfer immunotherapy.


In some embodiments, the method is conducted by administration of a pharmaceutical compositions containing infectious agent containing a nucleic acid molecule encoding the immunomodulatory protein, either secretable immunomodulatory protein or transmembrane immunomodulatory protein. In some embodiments, the pharmaceutical composition contains a dose of infectious agents suitable for administration to a subject that is suitable for treatment. In some embodiments, the pharmaceutical composition contains an infectious agent that is a virus, at a single or multiple dosage amount, of between about between or between about 1×105 and about 1×1012 plaque-forming units (pfu), 1×106 and 1×1010 pfu, or 1×107 and 1×1010 pfu, each inclusive, such as at least or at least about or at about 1×106, 1×107, 1×108, 1×109, 2×109, 3×109, 4×109, 5×109 pfu or about 1×1010 pfu. In some embodiments, the pharmaceutical composition can contain a virus concentration of from or from about 105 to about 1010 pfu/mL, for example, 5×106 to 5×109 or 1×107 to 1×109 pfu/mL, such as at least or at least about or at about 106 pfu/mL, 107 pfu/mL, 108 pfu/mL or 109 pfu/mL. In some embodiments, the pharmaceutical composition contains an infectious agent that is a bacterium, at a single or multiple dosage amount, of between about between or between about 1×103 and about 1×109 colony-forming units (cfu), 1×104 and 1×109 cfu, or 1×105 and 1×107 cfu, each inclusive, such as at least or at least about or at about 1×104, 1×105, 1×106, 1×107, 1×108 or 1×109 cfu. In some embodiments, the pharmaceutical composition can contain a bacterial concentration of from or from about 103 to about 108 cfu/mL, for example, 5×105 to 5×107 or 1×106 to 1×107 cfu/mL, such as at least or at least about or at about 105 cfu/mL, 106 cfu/mL, 107 cfu/mL or 108 cfu/mL


In some embodiments, an effective amount of a pharmaceutical composition is administered to inhibit, halt, or reverse progression of cancers that are sensitive to modulation of immunological activity by immunomodulatory proteins of the present invention. In some embodiments, the methods of the invention are used in the treatment of a mammalian patient of cancers such as lymphoma, lymphoid leukemia, myeloid leukemia, cervical cancer, neuroblastoma, or multiple myeloma. Other cancers which can be treated by the methods of the invention include, but are not limited to, melanoma, bladder cancer, hematological malignancies (leukemia, lymphoma, myeloma), liver cancer, brain cancer, renal cancer, breast cancer, pancreatic cancer (adenocarcinoma), colorectal cancer, lung cancer (small cell lung cancer and non-small-cell lung cancer), spleen cancer, cancer of the thymus or blood cells (i.e., leukemia), prostate cancer, testicular cancer, ovarian cancer, uterine cancer, gastric carcinoma, or Ewing's sarcoma.


Human cancer cells can be treated in vivo, or ex vivo. In ex vivo treatment of a human patient, tissue or fluids containing cancer cells are treated outside the body and then the tissue or fluids are reintroduced back into the patient. In some embodiments, the cancer is treated in a human patient in vivo by administration of the therapeutic composition into the patient. Thus, the present invention provides ex vivo and in vivo methods to inhibit, halt, or reverse progression of the tumor, or otherwise result in a statistically significant increase in progression-free survival (i.e., the length of time during and after treatment in which a patient is living with cancer that does not get worse), or overall survival (also called “survival rate;” i.e., the percentage of people in a study or treatment group who are alive for a certain period of time after they were diagnosed with or treated for cancer) relative to treatment with a control.


In some embodiments, a pharmaceutical composition of the invention can also be used to inhibit growth of mammalian, particularly human, cancer cells as a monotherapy (i.e., as a single agent), in combination with at least one chemotherapeutic agent (i.e., a combination therapy), in combination with a cancer vaccine, in combination with an immune checkpoint inhibitor and/or in combination with radiation therapy. In some aspects of the present disclosure, the immune checkpoint inhibitor is nivolumab, tremelimumab, pembrolizumab, ipilimumab, or the like.


In some embodiments, the provided compositions can attenuate an immune response, such as, for example, where the immunomodulatory protein comprises an affinity modified IgSF domain of an inhibitory ligand. In some embodiments, the compositions can be used to treat an autoimmune disease. In some embodiments, the administration of a therapeutic composition of the invention to a subject suffering from an immune system disease (e.g., autoimmune disease) can result in suppression or inhibition of such immune system attack or biological responses associated therewith. By suppressing this immune system attack on healthy body tissues, the resulting physical symptoms (e.g., pain, joint inflammation, joint swelling or tenderness) resulting from or associated with such attack on healthy tissues can be decreased or alleviated, and the biological and physical damage resulting from or associated with the immune system attack can be decreased, retarded, or stopped. In a prophylactic context, the subject may be one with, susceptible to, or believed to present an immune system disease, disorder or condition, and administration is typically conducted to prevent progression of the disease, disorder or condition, inhibit or alleviate symptoms, signs, or biological responses associated therewith, prevent bodily damage potentially resulting therefrom, and/or maintain or improve the subject's physical functioning.


In some embodiments, the inflammatory or autoimmune disorder is antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis, a vasculitis, an autoimmune skin disease, transplantation, a Rheumatic disease, an inflammatory gastrointestinal disease, an inflammatory eye disease, an inflammatory neurological disease, an inflammatory pulmonary disease, an inflammatory endocrine disease, or an autoimmune hematological disease.


In some embodiments, the pharmaceutical compositions comprising cells engineered to express and secrete immunomodulatory proteins can be used to treat one or more other immune disease or disorder in the subject. The immune system disease or disorder of the patient may be or involve, e.g., but is not limited to, Addison's Disease, Allergy, Alopecia Areata, Alzheimer's, Antineutrophil cytoplasmic antibodies (ANCA)-associated vasculitis, Ankylosing Spondylitis, Antiphospholipid Syndrome (Hughes Syndrome), arthritis, Asthma, Atherosclerosis, Atherosclerotic plaque, autoimmune disease (e.g., lupus, RA, MS, Graves' disease, etc.), Autoimmune Hemolytic Anemia, Autoimmune Hepatitis, Autoimmune inner ear disease, Autoimmune Lymphoproliferative syndrome, Autoimmune Myocarditis, Autoimmune Oophoritis, Autoimmune Orchitis, Azoospermia, Behcet's Disease, Berger's Disease, Bullous Pemphigoid, Cardiomyopathy, Cardiovascular disease, Celiac Sprue/Coeliac disease, Chronic Fatigue Immune Dysfunction Syndrome (CFIDS), Chronic idiopathic polyneuritis, Chronic Inflammatory Demyelinating, Polyradicalneuropathy (CIPD), Chronic relapsing polyneuropathy (Guillain-Barré syndrome), Churg-Strauss Syndrome (CSS), Cicatricial Pemphigoid, Cold Agglutinin Disease (CAD), COPD, CREST syndrome, Crohn's disease, Dermatitis, Herpetiformus, Dermatomyositis, diabetes, Discoid Lupus, Eczema, Epidermolysis bullosa acquisita, Essential Mixed Cryoglobulinemia, Evan's Syndrome, Exopthalmos, Fibromyalgia, Goodpasture's Syndrome, graft-related disease or disorder, Graves' Disease, GVHD, Hashimoto's Thyroiditis, Idiopathic Pulmonary Fibrosis, Idiopathic Thrombocytopenia Purpura (ITP), IgA Nephropathy, immunoproliferative disease or disorder (e.g., psoriasis), Inflammatory bowel disease (IBD), Insulin Dependent Diabetes Mellitus (IDDM), Interstitial lung disease, juvenile diabetes, Juvenile Arthritis, juvenile idiopathic arthritis (JIA), Kawasaki's Disease, Lambert-Eaton Myasthenic Syndrome, Lichen Planus, lupus, Lupus Nephritis, Lymphoscytic Lypophisitis, Ménière's Disease, Miller Fish Syndrome/acute disseminated encephalomyeloradiculopathy, Mixed Connective Tissue Disease, Multiple Sclerosis (MS), muscular rheumatism, Myalgic encephalomyelitis (ME), Myasthenia Gravis, Ocular Inflammation, Pemphigus Foliaceus, Pemphigus Vulgaris, Pernicious Anaemia, Polyarteritis Nodosa, Polychondritis, Polyglandular Syndromes (Whitaker's syndrome), Polymyalgia Rheumatica, Polymyositis, Primary Agammaglobulinemia, Primary Biliary Cirrhosis/Autoimmune cholangiopathy, Psoriasis, Psoriatic arthritis, Raynaud's Phenomenon, Reiter's Syndrome/Reactive arthritis, Restenosis, Rheumatic Fever, rheumatic disease, Rheumatoid Arthritis, Sarcoidosis, Schmidt's syndrome, Scleroderma, Sjörgen's Syndrome, Solid-organ transplant rejection (kidney, heart, liver, lung, etc.), Stiff-Man Syndrome, Systemic Lupus Erythematosus (SLE), systemic scleroderma, Takayasu Arteritis, Temporal Arteritis/Giant Cell Arteritis, Thyroiditis, Type 1 diabetes, Type 2 diabetes, Ulcerative colitis, Uveitis, Vasculitis, Vitiligo, Wegener's Granulomatosis, and preventing or suppressing an immune response associated with rejection of a donor tissue, cell, graft, or organ transplant by a recipient subject. Graft-related diseases or disorders include graft versus host disease (GVDH), such as associated with bone marrow transplantation, and immune disorders resulting from or associated with rejection of organ, tissue, or cell graft transplantation (e.g., tissue or cell allografts or xenografts), including, e.g., grafts of skin, muscle, neurons, islets, organs, parenchymal cells of the liver, etc. With regard to a donor tissue, cell, graft or solid organ transplant in a recipient subject, it is believed that a therapeutic composition of the invention disclosed herein may be effective in preventing acute rejection of such transplant in the recipient and/or for long-term maintenance therapy to prevent rejection of such transplant in the recipient (e.g., inhibiting rejection of insulin-producing islet cell transplant from a donor in the subject recipient suffering from diabetes).


In some embodiments, a therapeutic amount of the pharmaceutical composition is administered. Typically, precise amount of the compositions of the present invention to be administered can be determined by a physician with consideration of individual differences in age, weight, tumor size, extent of infection or metastasis, and condition of the patient (subject). It can generally be stated that a pharmaceutical composition comprising engineered cells, e.g. T cells, as described herein may be administered at a dosage of 104 to 109 cells/kg body weight, such as 105 to 106 cells/kg body weight, including all integer values within those ranges. Engineered cell compositions, such as T cell compositions, may also be administered multiple times at these dosages. The cells can be administered by using infusion techniques that are commonly known in immunotherapy (see, e.g., Rosenberg et al, New Eng. J. of Med. 319: 1676, 1988). The optimal dosage and treatment regime for a particular patient can readily be determined by one skilled in the art of medicine by monitoring the patient for signs of disease and adjusting the treatment accordingly.


The administration of the subject compositions may be carried out in any convenient manner, including by aerosol inhalation, injection, ingestion, transfusion, implantation or transplantation. The compositions described herein may be administered to a patient subcutaneously, intradermally, intratumorally, intranodally, intramedullary, intramuscularly, by intravenous (i.v.) injection, or intraperitoneally. In one embodiment, the therapeutic composition is administered to a patient by intradermal or subcutaneous injection. In another embodiment, the therapeutic composition is administered by i.v. injection. In some cases, the cell compositions may be injected directly into a tumor, lymph node, or site of infection.


A. Pharmaceutical Compositions


Provided are pharmaceutical compositions containing the immunomodulatory protein, engineered cells configured to express and secrete or surface express such immunomodulatory proteins or infectious agents. In some embodiments, the pharmaceutical compositions and formulations include one or more optional pharmaceutically acceptable carrier or excipient.


Such compositions may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives. Compositions of the present invention are preferably formulated for intravenous administration.


Pharmaceutical compositions of the present invention may be administered in a manner appropriate to the disease to be treated (or prevented). The quantity and frequency of administration will be determined by such factors as the condition of the patient, and the type and severity of the patient's disease, although appropriate dosages may be determined by clinical trials.


Such a formulation may, for example, be in a form suitable for intravenous infusion. A pharmaceutically acceptable carrier may be a pharmaceutically acceptable material, composition, or vehicle that is involved in carrying or transporting cells of interest from one tissue, organ, or portion of the body to another tissue, organ, or portion of the body. For example, the carrier may be a liquid or solid filler, diluent, excipient, solvent, or encapsulating material, or some combination thereof. Each component of the carrier is “pharmaceutically acceptable” in that it must be compatible with the other ingredients of the formulation. It also must be suitable for contact with any tissue, organ, or portion of the body that it may encounter, meaning that it must not carry a risk of toxicity, irritation, allergic response, immunogenicity, or any other complication that excessively outweighs its therapeutic benefits.


In some embodiments, the pharmaceutical composition is sterile. In some embodiments, the pharmaceutical composition is free or essentially free of bacteria or viruses. In some embodiments, the pharmaceutical composition is free or essentially free of cells other than the engineered cells described herein.


An effective amount of a pharmaceutical composition to be employed therapeutically will depend, for example, upon the therapeutic context and objectives. One skilled in the art will appreciate that the appropriate dosage levels for treatment will thus vary depending, in part, upon the molecule delivered, the indication for which the binding agent molecule is being used, the route of administration, and the size (body weight, body surface or organ size) and condition (the age and general health) of the patient. Accordingly, the clinician may titer the dosage and modify the route of administration to obtain the optimal therapeutic effect. The pharmaceutical composition of the invention can be administered parentally, subcutaneously, or intravenously, or as described elsewhere herein. The pharmaceutical composition of the invention may be administered in a therapeutically effective amount one, two, three or four times per month, two times per week, biweekly (every two weeks), or bimonthly (every two months). Administration may last for a period of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or longer (e.g., one, two, three, four or more years, including for the life of the subject).


For any composition, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models such as mice, rats, rabbits, dogs, pigs, or monkeys. An animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans. The exact dosage will be determined in light of factors related to the subject requiring treatment. Dosage and administration are adjusted to provide sufficient levels of the cell composition or to maintain the desired effect. Factors that may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combination(s), reaction sensitivities, and response to therapy. Appropriate dosages may be ascertained through use of appropriate dose-response data. A number of biomarkers or physiological markers for therapeutic effect can be monitored including T cell activation or proliferation, cytokine synthesis or production (e.g., production of TNF-α, IFN-γ, IL-2), induction of various activation markers (e.g., CD25, IL-2 receptor), inflammation, joint swelling or tenderness, serum level of C-reactive protein, anti-collagen antibody production, and/or T cell-dependent antibody response(s).


A variety of means are known for determining if administration of a therapeutic composition of the invention sufficiently modulates immunological activity by eliminating, sequestering, or inactivating immune cells mediating or capable of mediating an undesired immune response; inducing, generating, or turning on immune cells that mediate or are capable of mediating a protective immune response; changing the physical or functional properties of immune cells; or a combination of these effects. Examples of measurements of the modulation of immunological activity include, but are not limited to, examination of the presence or absence of immune cell populations (using flow cytometry, immunohistochemistry, histology, electron microscopy, polymerase chain reaction (PCR)); measurement of the functional capacity of immune cells including ability or resistance to proliferate or divide in response to a signal (such as using T cell proliferation assays and pepscan analysis based on 3H-thymidine incorporation following stimulation with anti-CD3 antibody, anti-T cell receptor antibody, anti-CD28 antibody, calcium ionophores, PMA, antigen presenting cells loaded with a peptide or protein antigen; B cell proliferation assays); measurement of the ability to kill or lyse other cells (such as cytotoxic T cell assays); measurements of the cytokines, chemokines, cell surface molecules, antibodies and other products of the cells (e.g., by flow cytometry, enzyme-linked immunosorbent assays, Western blot analysis, protein microarray analysis, immunoprecipitation analysis); measurement of biochemical markers of activation of immune cells or signaling pathways within immune cells (e.g., Western blot and immunoprecipitation analysis of tyrosine, serine or threonine phosphorylation, polypeptide cleavage, and formation or dissociation of protein complexes; protein array analysis; DNA transcriptional, profiling using DNA arrays or subtractive hybridization); measurements of cell death by apoptosis, necrosis, or other mechanisms (e.g., annexin V staining, TUNEL assays, gel electrophoresis to measure DNA laddering, histology; fluorogenic caspase assays, Western blot analysis of caspase substrates); measurement of the genes, proteins, and other molecules produced by immune cells (e.g., Northern blot analysis, polymerase chain reaction, DNA microarrays, protein microarrays, 2-dimensional gel electrophoresis, Western blot analysis, enzyme linked immunosorbent assays, flow cytometry); and measurement of clinical symptoms or outcomes such as improvement of autoimmune, neurodegenerative, and other diseases involving self proteins or self polypeptides (clinical scores, requirements for use of additional therapies, functional status, imaging studies) for example, by measuring relapse rate or disease severity (using clinical scores known to the ordinarily skilled artisan) in the case of multiple sclerosis, measuring blood glucose in the case of type I diabetes, or joint inflammation in the case of rheumatoid arthritis.


IX. Exemplary Embodiments

By way of example, among the provided embodiments are:


1. An immunomodulatory protein comprising at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein:

    • the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain;
    • the immunomodulatory protein does not comprise a transmembrane domain; and
    • the immunomodulatory protein is not conjugated to a half-life extending moiety.


2. The immunomodulatory protein of embodiment 1, wherein the half-life extending moiety is a multimerization domain.


3. The immunomodulatory protein of embodiment 1 or 2, wherein the half-life extending moiety is an Fc domain.


4. The immunomodulatory protein of any one of embodiments 1-3, wherein the immunomodulatory protein further comprises a signal peptide.


5. The immunomodulatory protein of embodiment 4, wherein the signal peptide is a native signal peptide from the corresponding wild-type IgSF member.


6. The immunomodulatory protein of embodiment 4, wherein the signal peptide is a non-native signal peptide.


7. The immunomodulatory protein of embodiment 4 or 6, wherein the signal peptide is an IgG-kappa signal peptide, an IL-2 signal peptide, or a CD33 signal peptide.


8. The immunomodulatory protein of any one of embodiments 1-7, wherein the at least one cell surface cognate binding partner is expressed on a mammalian cell.


9. The immunomodulatory protein of embodiment 8, wherein the mammalian cell is an antigen presenting cell (APC), a tumor cell, or a lymphocyte.


10. The immunomodulatory protein of embodiment 8 or 9, wherein the mammalian cell is a T-cell.


11. The immunomodulatory protein of any of embodiments 8-10, wherein the mammalian cell is a mouse, rat, cynomolgus monkey, or human cell.


12. The immunomodulatory protein of any of embodiments 1-11, wherein the at least one affinity modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the wild-type IgSF domain.


13. The immunomodulatory protein of any of embodiments 8-12, wherein specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain modulates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


14. The immunomodulatory protein of any of embodiments 8-13, wherein specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain increases immunological activity of the mammalian cell compared to the wild-type IgSF domain.


15. The immunomodulatory protein of any of embodiments 8-13, wherein specific binding of the immunomodulatory protein attenuates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


16. The immunomodulatory protein of any of embodiments 1-15, wherein the wild-type IgSF domain is from an IgSF family member of a family selected from the group consisting of Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family, and Killer-cell immunoglobulin-like receptors (KIR) family.


17. The immunomodulatory protein of any of embodiments 1-16, wherein the wild-type IgSF domain is from an IgSF member selected from the group consisting of CD80, CD86, PD-L1, PD-L2, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAG3, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R, NKp30, VISTA, VSIG3, and VSIG8.


18. The immunomodulatory protein of any of embodiments 1-17, wherein the wild-type IgSF domain is a human IgSF domain.


19. The immunomodulatory protein of any of embodiments 1-18, wherein the at least one affinity modified IgSF domain has at least 90% sequence identity to a wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27 and 408.


20. The immunomodulatory protein of any of embodiments 1-19, wherein the immunomodulatory protein has at least 90% sequence identity to the amino acid sequence selected from any of SEQ ID NOS:28-54 and 410 or to a specific binding fragment thereof containing an IgSF domain.


21. The immunomodulatory protein of any of embodiments 1-20, wherein the wild-type IgSF domain is a member of the B7 family.


22. The immunomodulatory protein of any of embodiments 1-21, wherein the wild-type IgSF domain is a domain of CD80, CD86 or ICOSL.


23. The immunomodulatory protein of any of embodiments 1-22, wherein the at least one cell surface cognate binding partner is a stimulatory receptor expressed on a T-cell, and the at least one affinity-modified IgSF domain has increased binding affinity to the stimulatory receptor compared to the binding affinity of the wild-type IgSF domain to the stimulatory receptor.


24. The immunomodulatory protein of embodiment 23, wherein binding of the affinity-modified IgSF domain to the stimulatory receptor increases immunological activity of the T-cell.


25. The immunomodulatory protein of embodiment 23 or 24, wherein the stimulatory receptor is CD28, ICOS, or CD226.


26. The immunomodulatory protein of any one of embodiments 23-25, wherein the at least one affinity-modified IgSF domain is an affinity-modified ICOSL IgSF domain that has increased binding affinity to at least one of: ICOS and CD28.


27. The immunomodulatory protein of any one of embodiments 23-26, wherein the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is ICOS.


28. The immunomodulatory protein of any one of embodiments 23-26, wherein the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is CD28.


29. The immunomodulatory protein of any one of embodiments 23-25, wherein the at least one affinity-modified IgSF domain is an affinity modified CD80 IgSF domain and the stimulatory receptor is CD28.


30. The immunomodulatory protein of any one of embodiments 23-29, wherein the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4 or exhibits decreased binding affinity to CTLA-4 compared to the binding affinity of wild-type IgSF domain to CTLA-4.


31. The immunomodulatory protein of any of embodiments 1-30, wherein the at least one affinity-modified IgSF domain specifically binds to no more than one cell surface cognate binding partner.


32. The immunomodulatory protein of any of embodiments 1-31, wherein the immunomodulatory protein specifically binds to no more than one cell surface cognate binding partner.


33. The immunomodulatory protein of any of embodiments 1-30, wherein the at least one affinity-modified domain specifically binds to at least two cell surface cognate binding partners.


34. The immunomodulatory protein of embodiment 33, wherein:


the first cell surface cognate binding partner is a stimulatory receptor expressed on a T cell; and


the second cell surface cognate binding partner is an inhibitory ligand of an inhibitory receptor, wherein the inhibitory receptor is expressed on a T-cell.


35. The immunomodulatory protein of embodiment 34, wherein binding of the affinity-modified domain to the inhibitory ligand competitively inhibits binding of the inhibitory ligand to the inhibitory receptor.


36. The immunomodulatory protein of embodiment 34 or embodiment 35, wherein:


the inhibitory receptor is PD-1, CTLA-4, LAG-3, TIGIT, CD96, CD112R, BTLA, CD160, TIM-3, VSIG3, or VSIG8; or


the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, HVEM, MHC class II, PVR, CEACAM-1, GAL9, or VISTA.


37. The immunomodulatory protein of any one of embodiments 34-36, wherein the affinity modified IgSF domain is an affinity modified CD80 domain and the stimulatory receptor is CD28.


38. The immunomodulatory protein of embodiment 37, wherein the inhibitory ligand is PD-L1 and the inhibitory receptor is PD-1.


39. The immunomodulatory protein of embodiment 37 or embodiment 38, wherein the affinity-modified IgSF domain exhibits decreased binding affinity to CTLA-4 compared to the wild-type IgSF domain.


40. The immunomodulatory protein of any one of embodiments 37-39, wherein the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4.


41. The immunomodulatory protein of any of embodiments 1-20, wherein the affinity modified IgSF domain is an affinity modified CD155 IgSF domain or an affinity modified CD112 IgSF domain and the at least one cell surface cognate binding partner is CD226, TIGIT or CD112R.


42. The immunomodulatory protein of embodiment 41, wherein the affinity-modified IgSF domain exhibits decreased binding affinity to CD226 compared to the binding affinity of the wild-type IgSF domain to CD226 and, optionally, retains or exhibits increased binding to TIGIT (T-cell immunoreceptor with Ig and ITIM domains) or CD112R compared to the binding affinity of the wild-type IgSF domain.


43. The immunomodulatory protein of any of embodiments 1-20, wherein the at least one affinity-modified IgSF domain specifically binds to a cell surface cognate binding partner that is a tumor specific antigen.


44. The immunomodulatory protein of embodiment 43, wherein the tumor specific antigen is B7-H6.


45. The immunomodulatory protein of embodiment 43 or 44, wherein the affinity-modified IgSF domain is an affinity modified NKp30 IgSF domain.


46. The immunomodulatory protein of any one of embodiments 1-45, wherein the at least one affinity-modified IgSF domain comprises a first affinity-modified IgSF domain and a second affinity-modified IgSF domain.


47. The immunomodulatory protein of embodiment 46, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain are different.


48. The immunomodulatory protein of embodiment 46 or 47, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more different amino acid substitutions in the same wild-type IgSF domain.


49. The immunomodulatory protein of embodiment 46 or 47, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more amino acid substitutions in a different wild-type IgSF domain.


50. The immunomodulatory protein of any of embodiments 1-20, wherein the wild-type IgSF domain is from an IgSF member that is a ligand of an inhibitory receptor, the inhibitory receptor comprising an ITIM signaling domain.


51. The immunomodulatory protein of embodiment 50, wherein:


the inhibitory receptor is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, or BTLA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8, respectively; or


the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA, respectively.


52. The immunomodulatory protein of embodiment 50 or 51, wherein the inhibitory receptor is PD-1 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of PD-1.


53. The immunomodulatory protein of any of embodiments 50-52, wherein the affinity-modified IgSF domain has increased binding affinity for a trans surface cognate binding partner compared to the wildtype IgSF domain, whereby the increased binding affinity competitively inhibits binding of the trans surface cognate binding partner to the inhibitory receptor.


54. The immunomodulatory protein of any of embodiments 1-53, wherein the affinity modified IgSF domain differs by no more than ten amino acid substitutions from the wildtype IgSF domain.


55. The immunomodulatory protein of any of embodiments 1-54, wherein the affinity modified IgSF domain differs by no more than five amino acid substitutions from the wildtype IgSF domain.


56. The immunomodulatory protein of any of embodiments 1-55, wherein the one or more affinity-modified IgSF domain is or comprises an affinity modified IgV domain, affinity modified IgC1 domain, or an affinity modified IgC2 domain, or is a specific binding fragment thereof comprising the one or more amino acid substitutions.


57. The immunomodulatory protein of any of embodiments 1-56, wherein the immunomodulatory protein further comprises one or more non-affinity modified IgSF domains.


58. The immunomodulatory protein of any one of embodiments 1-56, wherein the immunomodulatory protein has been secreted from an engineered cell.


59. The immunomodulatory protein of embodiment 58, wherein the engineered cell is an immune cell.


60. The immunomodulatory protein of embodiment 58 or 59, wherein the engineered cell is a primary cell.


61. A recombinant nucleic acid encoding the immunomodulatory protein of any of embodiments 1-60.


62. The recombinant nucleic acid of embodiment 61, wherein the nucleic acid molecule further comprises at least one promoter operably linked to control expression of the immunomodulatory protein.


63. The recombinant nucleic acid of embodiment 62, wherein the promoter is a constitutively active promoter.


64. The recombinant nucleic acid of embodiment 62, wherein the promoter is an inducible promoter.


65. The recombinant nucleic acid of embodiment 64, wherein the promoter is responsive to an element responsive to T-cell activation signaling.


66. The recombinant nucleic acid of embodiment 64, or 65, wherein the promoter comprises a binding site for NFAT or a binding site for NF-κB.


67. A recombinant expression vector comprising the nucleic acid of any of embodiments 61-66.


68. A recombinant expression vector comprising a nucleic acid encoding an immunomodulatory protein under the operable control of a signal sequence for secretion, wherein:

    • the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; and
    • the encoded immunomodulatory protein is secreted when expressed from a cell.


69. The expression vector of embodiment 68, wherein the immunomodulatory protein does not comprise a transmembrane domain.


70. The expression vector of embodiment 68 or 69, wherein the immunomodulatory protein is not conjugated to a half-life extending moiety.


71. The expression vector of any one of embodiments 68-70, wherein the half-life extending moiety is a multimerization domain.


72. The expression vector of any one of embodiments 68-71, wherein the half-life extending moiety is an Fc domain.


73. The expression vector of any one of embodiments 68-72, wherein the signal sequence for secretion encodes a secretory signal peptide.


74. The expression vector of embodiment 73, wherein the signal peptide is a native signal peptide from the corresponding wild-type IgSF member.


75. The expression vector of embodiment 73, wherein the signal peptide is a non-native signal peptide.


76. The expression vector of embodiment 73 or 75, wherein the signal peptide is an IgG-kappa signal peptide, an IL-2 signal peptide, or a CD33 signal peptide.


77. The expression vector of any of embodiments 68-76, wherein the nucleic acid molecule further comprises at least one promoter operably linked to control expression of the immunomodulatory protein.


78. The expression vector of embodiment 77, wherein the promoter is a constitutively active promoter.


79. The expression vector of embodiment 77, wherein the promoter is an inducible promoter.


80. The expression vector of embodiment 79 wherein the promoter is responsive to an element responsive to T-cell activation signaling.


81. The expression vector of embodiment 79 or 80, wherein the promoter comprises a binding site for NFAT or a binding site for NF-κB.


82. The expression vector of any of embodiments 68-81, wherein the vector is a viral vector.


83. The expression vector of embodiment 82, wherein the viral vector is a retroviral vector.


84. The expression vector of embodiment 82 or 83, wherein the viral vector is a lentiviral vector or a gammaretroviral vector.


85. The expression vector of any one of embodiments 68-84, wherein the at least one affinity-modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the wild-type IgSF domain.


86. The expression vector of any one of embodiments 68-85, wherein the at least one cell surface cognate binding partner is expressed on a mammalian cell.


87. The expression vector of embodiment 86, wherein the mammalian cell is an antigen presenting cell (APC), a tumor cell, or a lymphocyte.


88. The expression vector of embodiment 86 or 87, wherein the mammalian cell is a T-cell.


89. The expression vector of any one of embodiments 86-88, wherein the mammalian cell is a mouse, rat, cynomolgus monkey, or human cell.


90. The expression vector of any one of embodiments 86-89, wherein specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain modulates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


91. The expression vector of any one of embodiments 86-90, wherein specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain increases immunological activity of the mammalian cell compared to the wild-type IgSF domain.


92. The expression vector of any one of embodiments 86-910, wherein specific binding of the immunomodulatory protein attenuates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


93. The expression vector of any one of embodiments 68-92, wherein the wild-type IgSF domain is from an IgSF family member of a family selected from the group consisting of Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family, and Killer-cell immunoglobulin-like receptors (KIR) family.


94. The expression vector of any one of embodiments 68-93, wherein the wild-type IgSF domain is from an IgSF member selected from the group consisting of CD80, CD86, PD-L1, PD-L2, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAG3, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R, NKp30, VISTA, VSIG3, and VSIG8.


95. The expression vector of any one of embodiments 68-94, wherein the wild-type IgSF domain is a human IgSF domain.


96. The expression vector of any one of embodiments 68-95, wherein the at least one affinity modified IgSF domain has at least 90% sequence identity to a wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27 and 408.


97. The expression vector of any one of embodiments 68-96, wherein the immunomodulatory protein has at least 90% sequence identity to the amino acid sequence selected from any of SEQ ID NOS: 28-54 and 410.


98. The expression vector of any of embodiments 68-97, wherein the wild-type IgSF domain is a member of the B7 family.


99. The expression vector of any of embodiments 68-98, wherein the wild-type IgSF domain is a domain of CD80, CD86 or ICOSL.


100. The expression vector of any one of embodiments 68-99, wherein the at least one cell surface cognate binding partner is a stimulatory receptor expressed on a T-cell and the at least one affinity-modified IgSF domain has increased binding affinity to the stimulatory receptor compared to the binding affinity of the wild-type IgSF domain to the stimulatory receptor.


101. The expression vector of embodiment 100, wherein binding of the affinity-modified IgSF domain to the stimulatory receptor increases immunological activity of the T-cell.


102. The expression vector of embodiment 100 or 101, wherein the stimulatory receptor is CD28, ICOS, or CD226.


103. The expression vector of any one of embodiments 100-102, wherein the at least one affinity-modified IgSF domain is an affinity-modified ICOSL IgSF domain that has increased binding affinity to at least one of: ICOS and CD28.


104. The expression vector of any one of embodiments 100-103, wherein the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is ICOS.


105. The expression vector of any one of embodiments 100-103, wherein the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is CD28.


106. The expression vector of any one of embodiments 100-102, wherein the at least one affinity-modified IgSF domain is an affinity modified CD80 IgSF domain and the stimulatory receptor is CD28.


107. The expression vector of any one of embodiments 100-106, wherein the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4 or exhibits decreased binding affinity to CTLA-4 compared to the binding affinity of wild-type IgSF domain to CTLA-4.


108. The expression vector of any one of embodiments 68-107, wherein the at least one affinity-modified IgSF domain specifically binds to no more than one cell surface cognate binding partner.


109. The expression vector of any one of embodiments 68-108, wherein the immunomodulatory protein specifically binds to no more than one cell surface cognate binding partner.


110. The expression vector of any one of embodiments 68-107, wherein the at least one affinity-modified domain specifically binds to at least two cell surface cognate binding partners.


111. The expression vector of embodiment 110, wherein:


the first cell surface cognate binding partner is a stimulatory receptor expressed on a T cell; and


the second cell surface cognate binding partner is an inhibitory ligand of an inhibitory receptor, wherein the inhibitory receptor is expressed on a T-cell.


112. The expression vector of embodiment 111, wherein binding of the affinity-modified IgSF domain to the inhibitory ligand competitively inhibits binding of the inhibitory ligand to the inhibitory receptor.


113. The expression vector of embodiment 111 or 112, wherein:


the inhibitory receptor is PD-1, CTLA-4, LAG-3, TIGIT, CD96, CD112R, BTLA, CD160, TIM-3, VSIG3, or VSIG8; or


the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, HVEM, MHC class II, PVR, CEACAM-1, GAL9 or VISTA.


114. The expression vector of any one of embodiments 111-113, wherein the affinity modified IgSF domain is an affinity modified CD80 domain and the stimulatory receptor is CD28.


115. The expression vector of embodiment 114, wherein the inhibitory ligand is PD-L1 and the inhibitory receptor is PD-1.


116. The expression vector of embodiment 114 or 115, wherein the affinity-modified IgSF domain exhibits decreased binding affinity to CTLA-4 compared to the wild-type IgSF domain.


117. The expression vector of any one of embodiments 114-116, wherein the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4.


118. The expression vector of any of embodiments 68-97, wherein the affinity-modified IgSF domain is an affinity modified CD155 IgSF domain or an affinity modified CD112 IgSF domain and the at least one cell surface cognate binding partner is CD226, TIGIT or CD112R.


119. The expression vector of embodiment 118, wherein the affinity-modified IgSF domain exhibits decreased binding affinity to CD226 compared to the binding affinity of the wild-type IgSF domain to CD226 and, optionally, retains or exhibits increased binding to TIGIT (T-cell immunoreceptor with Ig and ITIM domains) or CD112R compared to the binding affinity of the wild-type IgSF domain.


120. The expression vector of any of embodiments 68-97, wherein the at least one affinity-modified IgSF domain specifically binds to a cell surface cognate binding partner that is a tumor specific antigen.


121. The expression vector of embodiment 120, wherein the tumor specific antigen is B7-H6.


122. The expression vector of embodiment 120 or 121, wherein the affinity-modified IgSF domain is an affinity modified NKp30 IgSF domain.


123. The expression vector of any one of embodiments 68-122, wherein the at least one affinity-modified IgSF domain comprises a first affinity-modified IgSF domain and a second affinity-modified IgSF domain.


124. The expression vector of embodiment 123, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain are different.


125. The expression vector of embodiment 123 or 124, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more different amino acid substitutions in the same wild-type IgSF domain.


126. The expression vector of embodiment 123 or 124, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more amino acid substitutions in a different wild-type IgSF domain.


127. The expression vector of any of embodiments 68-97, wherein the wild-type IgSF domain is from an IgSF member that is a ligand of an inhibitory receptor, the inhibitory receptor comprising an ITIM signaling domain.


128. The expression vector of embodiment 127, wherein:


the inhibitory receptor is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, or BTLA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8, respectively; or


the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA, respectively.


129. The expression vector of embodiment 127 or 128, wherein the inhibitory receptor is PD-1 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of PD-1.


130. The expression vector of any of embodiments 127-129, wherein the affinity-modified IgSF domain has increased binding affinity for a trans surface cognate binding partner compared to the wildtype IgSF domain, whereby the increased binding affinity competitively inhibits binding of the trans surface cognate binding partner to the inhibitory receptor.


131. The expression vector of any of embodiments 68-130, wherein the affinity modified IgSF domain differs by no more than ten amino acid substitutions from the wildtype IgSF domain.


132. The expression vector of any of embodiments 68-131, wherein the affinity modified IgSF domain differs by no more than five amino acid substitutions from the wildtype IgSF domain.


133. The expression vector of any of embodiments 68-132, wherein the one or more affinity-modified IgSF domain is or comprises an affinity modified IgV domain, affinity modified IgC1 domain, or an affinity modified IgC2 domain, or is a specific binding fragment thereof comprising the one or more amino acid substitutions.


134. The expression vector of any of embodiments 68-133, wherein the immunomodulatory protein further comprises one or more non-affinity modified IgSF domains.


135. An engineered cell comprising the nucleic acid of any one of embodiments 61-66 or the expression vector of any one of embodiments 67-134.


136. An engineered cell comprising the immunomodulatory protein of any one of embodiments 1-60.


137. An engineered cell that secretes the immunomodulatory protein of any one of embodiments 1-60.


138. The engineered cell of any of embodiments 135-137, wherein the cell is an immune cell.


139. An engineered immune cell comprising a nucleic acid molecule that encodes an immunomodulatory protein, wherein:

    • the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; and
    • the engineered cell expresses and secretes the immunomodulatory protein.


140. The engineered immune cell of embodiment 139, wherein the immunomodulatory protein does not comprise a transmembrane domain.


141. The engineered immune cell of embodiment 139 or 140, wherein the immunomodulatory protein is not conjugated to a half-life extending moiety.


142. The engineered immune cell of embodiment 141, wherein the half-life extending moiety is a multimerization domain.


143. The engineered immune cell of embodiment 141 or 142, wherein the half-life extending moiety is an Fc domain.


144. The engineered immune cell of any one of embodiments 139-143, wherein the nucleic acid molecule comprises a sequence encoding a secretory signal peptide operably linked to the sequence encoding the immunomodulatory protein.


145. The engineered immune cell of embodiment 144, wherein the signal peptide is the native signal peptide from the corresponding wild-type IgSF member.


146. The engineered immune cell of embodiment 144, wherein the signal peptide is a non-native signal sequence.


147. The engineered immune cell of embodiment 144 or 146, wherein the signal peptide is an IgG-kappa signal peptide, an IL-2 signal peptide, or a CD33 signal peptide.


148. The engineered immune cell of any one of embodiments 139-148, wherein the nucleic acid molecule further comprises at least one promoter operably linked to control expression of the immunomodulatory protein.


149. The engineered immune cell of embodiment 148, wherein the promoter is a constitutively active promoter.


150. The engineered immune cell of embodiment 148, wherein the promoter is an inducible promoter.


151. The engineered immune cell of embodiment 148 or 150, wherein the promoter is responsive to an element responsive to T-cell activation signaling.


152. The engineered immune cell of any one of embodiments 148, 150, and 151, wherein the promoter comprises a binding site for NFAT or a binding site for NF-κB.


153. The engineered cell of embodiments 135-152, wherein the immunomodulatory protein is expressed and secreted by the engineered cell after the engineered cell is contacted with an inducing agent or after induction of T cell activation signaling, which optionally is induced upon binding of an antigen to a chimeric antigen receptor (CAR) or engineered T-cell receptor (TCR) expressed by the engineered cell.


154. The engineered cell of any one of embodiments 135-153, wherein the cell is a lymphocyte.


155. The engineered cell of embodiment 154, wherein the lymphocyte is a T cell, a B cell or an NK cell.


156. The engineered cell of any one of embodiments 135-155, wherein the cell is a T cell.


157. The engineered cell of embodiment 156, wherein the T cells is CD4+ or CD8+.


158. The engineered cell of any one of embodiments 135-154, wherein the cell is an antigen presenting cell.


159. The engineered cell of any one of embodiments 135-158, wherein the cell is a primary cell obtained from a subject.


160. The engineered cell of embodiment 159, wherein the subject is a human subject.


161. The engineered cell of any one of embodiments 135-160, wherein the at least one affinity modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the wild-type IgSF domain.


162. The engineered cell of any one of embodiments 135-161, wherein the at least one cell surface cognate binding partner is expressed on a mammalian cell.


163. The engineered cell of embodiment 162, wherein the mammalian cell is an antigen presenting cell (APC), a tumor cell, or a lymphocyte.


164. The engineered cell of embodiment 162 or 163, wherein the mammalian cell is a T-cell.


165. The engineered cell of any one of embodiments 162-164, wherein the mammalian cell is a mouse, rat, cynomolgus monkey, or human cell.


166. The engineered cell of any one of embodiments 162-165, wherein specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain modulates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


167. The engineered cell of any one of embodiments 162-166, wherein specific binding of the immunomodulatory protein comprising the at least one affinity-modified IgSF domain increases immunological activity of the mammalian cell compared to the wild-type IgSF domain.


168. The engineered cell of any one of embodiments 162-166, wherein specific binding of the immunomodulatory protein attenuates immunological activity of the mammalian cell compared to the wild-type IgSF domain.


169. The engineered cell of any one of embodiments 135-168, wherein the wild-type IgSF domain is from an IgSF family member of a family selected from the group consisting of Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family, and Killer-cell immunoglobulin-like receptors (KIR) family.


170. The engineered cell of any one of embodiments 135-169, wherein the wild-type IgSF domain is from an IgSF member selected from the group consisting of CD80, CD86, PD-L1, PD-L2, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAG3, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R, NKp30, VISTA, VSIG3, and VSIG8.


171. The engineered cell of any one of embodiments 135-170, wherein the wild-type IgSF domain is a human IgSF domain.


172. The engineered cell of any one of embodiments 135-171, wherein the at least one affinity modified IgSF domain has at least 90% sequence identity to a wild-type IgSF domain or a specific binding fragment thereof contained in the sequence of amino acids set forth in any of SEQ ID NOS: 1-27 and 408.


173. The engineered cell of any one of embodiments 135-172, wherein the immunomodulatory protein has at least 90% sequence identity to the amino acid sequence selected from any of SEQ ID NOS: 28-54 and 410.


174. The engineered cell of any one of embodiments 135-173, wherein the at least one cell surface cognate binding partner is a stimulatory receptor expressed on a T-cell and the at least one affinity-modified IgSF domain has increased binding affinity to the stimulatory receptor compared to the binding affinity of the wild-type IgSF domain to the stimulatory receptor.


175. The engineered cell of embodiment 174, wherein binding of the affinity-modified IgSF domain to the stimulatory receptor increases immunological activity of the T-cell.


176. The engineered cell of embodiment 174 or 175, wherein the stimulatory receptor is CD28, ICOS, or CD226.


177. The engineered cell of any one of embodiments 174-176, wherein the at least one affinity-modified IgSF domain is an affinity-modified ICOSL IgSF domain that has increased binding affinity to at least one of: ICOS and CD28.


178. The engineered cell of any one of embodiments 174-177, wherein the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is ICOS.


179. The engineered cell of any one of embodiments 174-177, wherein the at least one affinity-modified IgSF domain is an affinity modified ICOSL IgSF domain and the stimulatory receptor is CD28.


180. The engineered cell of any one of embodiments 174-176, wherein the at least one affinity-modified IgSF domain is an affinity modified CD80 IgSF domain and the stimulatory receptor is CD28.


181. The engineered cell of any one of embodiments 174-180, wherein the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4 or exhibits decreased binding affinity to CTLA-4 compared to the binding affinity of wild-type IgSF domain to CTLA-4.


182. The engineered cell of any one of embodiments 135-181, wherein the at least one affinity-modified IgSF domain specifically binds to no more than one cell surface cognate binding partner.


183. The engineered cell of any one of embodiments 135-182, wherein the immunomodulatory protein specifically binds to no more than one cell surface cognate binding partner.


184. The engineered cell of any one of embodiments 135-181, wherein the at least one affinity-modified domain specifically binds to at least two cell surface cognate binding partners.


185. The engineered cell of embodiment 184, wherein:


the first cell surface cognate binding partner is a stimulatory receptor expressed on a T cell; and


the second cell surface cognate binding partner is an inhibitory ligand of an inhibitory receptor, wherein the inhibitory receptor is expressed on a T-cell.


186. The engineered cell of embodiment 185, wherein binding of the affinity-modified domain to the inhibitory ligand competitively inhibits binding of the inhibitory ligand to the inhibitory receptor.


187. The engineered cell of embodiment 185 or 186, wherein:


the inhibitory receptor is PD-1, CTLA-4, LAG-3, TIGIT, CD96, CD112R, BTLA, CD160, TIM-3, VSIG3, or VSIG8; or


the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, HVEM, MHC class II, PVR, CEACAM-1, GAL9 or VISTA.


188. The engineered cell of any one of embodiments 185-187, wherein the affinity modified IgSF domain is an affinity modified CD80 domain and the stimulatory receptor is CD28.


189. The engineered cell of embodiment 188, wherein the inhibitory ligand is PD-L1 and the inhibitory receptor is PD-1.


190. The engineered cell of embodiment 188 or 189, wherein the affinity-modified IgSF domain exhibits decreased binding affinity to CTLA-4 compared to the wild-type IgSF domain.


191. The engineered cell of any one of embodiments 188-190, wherein the affinity-modified IgSF domain does not substantially specifically bind to CTLA-4.


192. The engineered cell of any of embodiments 135-173, wherein the affinity modified IgSF domain is an affinity modified CD155 IgSF domain or an affinity modified CD112 IgSF domain and the at least one cell surface cognate binding partner is CD226, TIGIT or CD112R.


193. The engineered cell of embodiment 192, wherein the affinity-modified IgSF domain exhibits decreased binding affinity to CD226 compared to the binding affinity of the wild-type IgSF domain to CD226 and, optionally, retains or exhibits increased binding to TIGIT (T-cell immunoreceptor with Ig and ITIM domains) or CD112R compared to the binding affinity of the wild-type IgSF domain.


194. The engineered cell of any of embodiments 135-173, wherein the at least one affinity-modified IgSF domain specifically binds to a cell surface cognate binding partner that is a tumor specific antigen.


195. The engineered cell of embodiment 194, wherein the tumor specific antigen is B7-H6.


196. The engineered cell of embodiment 194 or 195, wherein the affinity-modified IgSF domain is an affinity modified NKp30 IgSF domain.


197. The engineered cell of any one of embodiments 135-173, wherein the at least one affinity-modified IgSF domain comprises a first affinity-modified IgSF domain and a second affinity-modified IgSF domain.


198. The engineered cell of embodiment 197, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain are different.


199. The engineered cell of embodiment 197 or 198, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprises one or more different amino acid substitutions in the same wild-type IgSF domain.


200. The engineered cell of embodiment 197 or 198, wherein the first affinity-modified IgSF domain and the second affinity-modified IgSF domain each comprise one or more amino acid substitutions in a different wild-type IgSF domain.


201. The engineered cell of any of embodiments 135-173, wherein the wild-type IgSF domain is from an IgSF member that is a ligand of an inhibitory receptor, the inhibitory receptor comprising an ITIM signaling domain.


202. The engineered cell of embodiment 201, wherein:


the inhibitory receptor is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, or BTLA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8, respectively; or


the ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-L1, PD-L2, B7-1, B7-2, MHC class II, PVR, CEACAM-1, GAL9 or VISTA, respectively.


203. The engineered cell of embodiment 201 or 202, wherein the inhibitory receptor is PD-1 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of PD-1.


204. The engineered cell of any of embodiments 201-203, wherein the affinity-modified IgSF domain has increased binding affinity for a trans surface cognate binding partner compared to the wildtype IgSF domain, whereby the increased binding affinity competitively inhibits binding of the trans surface cognate binding partner to the inhibitory receptor.


205. The engineered cell of any of embodiments 135-204, wherein the affinity modified IgSF domain differs by no more than ten amino acid substitutions from the wildtype IgSF domain.


206. The engineered cell of any of embodiments 135-205, wherein the affinity modified IgSF domain differs by no more than five amino acid substitutions from the wildtype IgSF domain.


207. The engineered cell of any of embodiments 135-206, wherein the one or more affinity-modified IgSF domain is or comprises an affinity modified IgV domain, affinity modified IgC1 domain, or an affinity modified IgC2 domain, or is a specific binding fragment thereof comprising the one or more amino acid substitutions.


208. The engineered cell of any of embodiments 135-207, wherein the immunomodulatory protein further comprises one or more non-affinity modified IgSF domains.


209. The engineered cell of any of embodiments 135-208, further comprising a chimeric antigen receptor (CAR) or an engineered T-cell receptor (TCR).


210. A pharmaceutical composition comprising the cell of any of embodiments 135-209 or the infectious agent of any of embodiments 230-252 and a pharmaceutically acceptable carrier.


211. The pharmaceutical composition of embodiment 210 that is sterile.


212. A method of introducing an immunomodulatory protein into a subject, comprising administering an engineered cell of any one of embodiments 135-209, the infectious agent of any of embodiments 230-252 or a pharmaceutical composition of embodiment 210 or 211 to the subject.


213. A method of modulating an immune response in a subject, comprising administering the cell of any one of embodiments 135-209, an infectious agent of any of embodiments 230-252 or a pharmaceutical composition of embodiment 210 or 211 to the subject.


214. The method of embodiment 213, wherein modulating the immune response treats a disease or disorder in the subject.


215. The method of embodiment 213 or 214, wherein the modulated immune response is increased.


216. The method of embodiment 214 or 215, wherein the disease or disorder is a tumor.


217. The method of any one of embodiments 214-216, wherein the disease or disorder is a cancer.


218. The method of any one of embodiments 214-217, wherein the disease or disorder is melanoma, lung cancer, bladder cancer, or a hematological malignancy.


219. The method of embodiment 213 or 214, wherein the modulated immune response is decreased.


220. The method of embodiment 214 or 219, wherein the disease or disorder is an inflammatory disease or condition.


221. The method of any one of embodiments 214, 219, and 220, wherein the disease or condition is Crohn's disease, ulcerative colitis, multiple sclerosis, asthma, rheumatoid arthritis, or psoriasis.


222. The method of any one of embodiments 212-221, wherein the subject is human.


223. The method of any of embodiments 212-222, wherein the cell is autologous to the subject.


224. The method of any of embodiments 212-222, wherein the cell is allogenic to the subject.


225. The method of any one of embodiments 212-224 wherein the engineered cell expresses and secretes the immunomodulatory protein.


226. The method of any one of embodiments 212-225, wherein the immunomodulatory protein is constitutively expressed by the engineered cell.


227. The method of any one of embodiments 212-225, wherein the immunomodulatory protein is expressed and secreted by the engineered cell after the engineered cell is contacted with an inducing agent.


228. The method of any one of embodiments 212-227, wherein the immunomodulatory protein is expressed and secreted by the engineered cell upon T cell activation signaling.


229. The method of embodiment 228, wherein the engineered cell expresses a chimeric antigen receptor (CAR) or an engineered T-cell receptor (TCR) and T cell activation signaling is induced upon binding of an antigen by the CAR or TCR.


230. An infectious agent, comprising a nucleic acid molecule encoding the immunomodulatory protein of any one of embodiments 1-60, a nucleic acid molecule of any of embodiments 61-66 or the expression vector of any one of embodiments 67-134.


231. An infectious agent, comprising a nucleic acid molecule encoding a transmembrane immunomodulatory protein (TIP) comprising:

    • (i) an ectodomain comprising at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitution(s) in a wild-type IgSF domain, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; and
    • (ii) a transmembrane domain.


232. The infectious agent of embodiment 231, wherein the at least one affinity modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the reference wild-type IgSF domain.


233. The infectious agent of embodiment 231 or embodiment 232, wherein the wild-type IgSF domain is from an IgSF family member of a family selected from Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, B7 family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family or Killer-cell immunoglobulin-like receptors (KIR) family.


234. The infectious agent of any of embodiments 231-233, wherein the wild-type IgSF domain is from an IgSF member selected from CD80, CD86, PD-L1, PD-L2, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAGS, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R or NKp30.


235. The infectious agent of any of embodiments 231-234, wherein the wild-type IgSF domain is a human IgSF member.


236. The infectious agent of any of embodiments 231-235, wherein the transmembrane immunomodulatory protein has at least 90% sequence identity to the amino acid sequence selected from any of SEQ ID NOS: 381-407 and 409 or to a contiguous portion thereof containing the affinity-modified IgSF domain and a transmembrane domain.


237. The infectious agent of any of embodiments 231-236, wherein the transmembrane immunomodulatory protein is a chimeric receptor, wherein the endodomain is not the endodomain from the wild-type IgSF member comprising the wild-type IgSF domain.


238. The infectious agent of embodiment 237, wherein the endodomain comprises at least one ITAM (immunoreceptor tyrosine-based activation motif)-containing signaling domain.


239. The infectious agent of embodiment 238, wherein the endodomain comprises a CD3-zeta signaling domain.


240. The infectious agent of embodiment 238 or embodiment 239, wherein the endodomain further comprises at least one of: a CD28 costimulatory domain, an ICOS signaling domain, an OX40 signaling domain, and a 41BB signaling domain.


241. The infectious agent of any of embodiments 231-240, wherein the affinity modified IgSF domain differs by no more than ten amino acid substitutions or no more than five amino acid substitutions from the wildtype IgSF domain.


242. The infectious agent of any of embodiments 231-241, wherein the affinity-modified IgSF domain is or comprises an affinity modified IgV domain, affinity modified IgC1 domain or an affinity modified IgC2 domain or is a specific binding fragment thereof comprising the one or more amino acid substitutions.


243. The infectious agent of any of embodiments 231-242, wherein the transmembrane domain is the native transmembrane domain from the corresponding wild-type IgSF member.


244. The infectious agent of any of embodiments 231-243, wherein the transmembrane domain is not the native transmembrane domain from the corresponding wild-type IgSF member.


245. The infectious agent of embodiment 244, wherein the transmembrane protein is a transmembrane protein derived from CD8.


246. The infectious agent of any of embodiments 230-245, wherein the infectious agent is a bacteria or a virus.


247. The infectious agent of embodiment 246, wherein the virus is an oncolytic virus.


248. The infectious agent of embodiment 247, wherein the oncolytic virus is an adenovirus, adeno-associated virus, herpes virus, Herpes Simplex Virus, Vesticular Stomatic virus, Reovirus, Newcastle Disease virus, parvovirus, measles virus, vesticular stomatitis virus (VSV), Coxsackie virus or a Vaccinia virus.


249. The infectious agent of embodiment 246, wherein the virus specifically targets dendritic cells (DCs) and/or is dendritic cell-tropic.


250. The infectious agent of embodiment 249, wherein the virus is a lentiviral vector that is pseudotyped with a modified Sindbis virus envelope product.


251. The infectious agent of any of embodiments 230-250, further comprising a nucleic acid molecule encoding a further gene product that results in death of a target cell or that can augment or boost an immune response.


252. The infectious agent of embodiment 251, wherein the further gene product is selected from an anticancer agent, anti-metastatic agent, an antiangiogenic agent, an immunomodulatory molecule, an immune checkpoint inhibitor, an antibody, a cytokine, a growth factor, an antigen, a cytotoxic gene product, a pro-apoptotic gene product, an anti-apoptotic gene product, a cell matrix degradative gene, genes for tissue regeneration or a reprogramming human somatic cells to pluripotency.


X. Examples

The following examples are included for illustrative purposes only and are not intended to limit the scope of the invention.


Examples 1-12 describe the design, creation, and screening of exemplary affinity modified CD80 (B7-1), CD86 (B7-2), ICOSL, and NKp30 immunomodulatory proteins, which are components of the immune synapse (IS) that have a demonstrated dual role in both immune activation and inhibition. These examples demonstrate that affinity modification of IgSF domains yields proteins that can act to both increase and decrease immunological activity. This work also describes the various combinations of those domains fused in pairs (i.e., stacked) to form a Type II immunomodulatory protein to achieve immunomodulatory activity. The Examples also describe the generation of exemplary secretable immunomodulatory proteins containing such affinity-modified immunomodulatory proteins.


Example 1
Generation of Mutant DNA Constructs of IgSF Domains

Example 1 describes the generation of mutant DNA constructs of human CD80, CD86, ICOSL and NKp30 IgSF domains for translation and expression on the surface of yeast as yeast display libraries. The below examples exemplify binding and activity of affinity-modified domains of the exemplary IgSF proteins in an Fc-fusion format; such affinity-modified domains are contemplated in connection with a secretable immunomodulatory protein or transmembrane immunomodulatory protein as described.


A. Degenerate Libraries


For libraries that target specific residues of target protein for complete or partial randomization with degenerate codons, such as specific mixed base sets to code for various amino acid substitutions, the coding DNAs for the extracellular domains (ECD) of human CD80 (SEQ ID NO:28), ICOSL (SEQ ID NO:32), and NKp30 (SEQ ID NO:54) were ordered from Integrated DNA Technologies (Coralville, Iowa) as a set of overlapping oligonucleotides of up to 80 base pairs (bp) in length. To generate a library of diverse variants of each ECD, the oligonucleotides contained desired degenerate codons at desired amino acid positions. Degenerate codons were generated using an algorithm at the URL: rosettadesign.med.unc.edu/SwiftLib/.


In general, positions to mutate and degenerate codons were chosen from crystal structures (CD80, NKp30) or homology models (ICOSL) of the target-ligand pairs of interest were used to identify ligand contact residues as well as residues that are at the protein interaction interface. This analysis was performed using a structure viewer available at the URL:spdbv.vital-it.ch). For example, a crystal structure for CD80 bound to CTLA4 is publicly available at the URL:www.rcsb.org/pdb/explore/explore.do?structureId=1I8L and a targeted library was designed based on the CD80::CTLA4 interface for selection of improved binders to CTLA4. However, there are no CD80 structures available with ligands CD28 and PD-L1, so the same library was also used to select for binders of CD28 (binds the same region on CD80 as CTLA4) and PD-L1 (not known if PD-L1 binds the same site as CTLA4). The next step in library design was the alignment of human, mouse, rat and monkey CD80, ICOSL or NKp30 sequences to identify conserved residues. Based on this analysis, conserved target residues were mutated with degenerate codons that only specified conservative amino acid changes plus the wild-type residue. Residues that were not conserved were mutated more aggressively, but also included the wild-type residue. Degenerate codons that also encoded the wild-type residue were deployed to avoid excessive mutagenesis of target protein. For the same reason, only up to 20 positions were targeted for mutagenesis at a time. These residues were a combination of contact residues and non-contact interface residues.


The oligonucleotides were dissolved in sterile water, mixed in equimolar ratios, heated to 95° C. for five minutes and slowly cooled to room temperature for annealing. ECD-specific oligonucleotide primers that anneal to the start and end of the ECDs, respectively, were then used to generate PCR product. The ECD-specific oligonucleotides which overlap by 40-50 bp with a modified version of pBYDS03 cloning vector (Life Technologies USA), beyond and including the BamHI and Kpnl cloning sites, were then used to amplify 100 ng of PCR product from the prior step to generate a total of at least 12 μg of DNA for every electroporation. Both PCR's were by polymerase chain reaction (PCR) using OneTaq 2× PCR master mix (New England Biolabs, USA). The second PCR products were purified using a PCR purification kit (Qiagen, Germany) and resuspended in sterile deionized water. To prepare for library insertion, a modified yeast display version of vector pBYDS03 was digested with BamHI and Kpnl restriction enzymes (New England Biolabs, USA) and the large vector fragment was gel-purified and dissolved in sterile, deionized water. Electroporation-ready DNA for the next step was generated by mixing 12 μg of library DNA for every electroporation with 4 μg of linearized vector in a total volume of 50 μL deionized and sterile water. An alternative way to generate targeted libraries was to carry out site-directed mutagenesis (Multisite kit, Agilent, USA) of target ECDs with oligonucleotides containing degenerate codons. This approach was used to generate sublibraries that only target specific stretches of the DNA for mutagenesis. In these cases, sublibraries were mixed before proceeding to the selection steps. In general, library sizes were in the range of 10×107 to 10×108 clones, except that sublibraries were only in the range of 10×104 to 10×105. Large libraries and sublibraries were generated for CD80, ICOSL, CD86 and NKp30. Sublibraries were generated for CD80, ICOSL and NKp30.


B. Random Libraries


Random libraries were also constructed to identify variants of the ECD of CD80 (SEQ ID NO:28), CD86 (SEQ ID NO: 29), ICOSL (SEQ ID NO:32) and NKp30 (SEQ ID NO:54. DNA encoding wild-type ECDs was cloned between the BamHI and Kpnl restriction sites of modified yeast display vector pBYDS03 and in some cases, the DNA was released using the same restriction enzymes. The released DNA or undigested plasmid was then mutagenized with the Genemorph II Kit (Agilent, USA) so as to generate an average of three to five amino acid changes per library variant. Mutagenized DNA was then amplified by the two-step PCR and further processed as described above for targeted libraries.


Example 2
Introduction of DNA Libraries into Yeast

Example 2 describes the introduction of CD80, CD86, ICOSL and NKp30 DNA libraries into yeast.


To introduce degenerate and random library DNA into yeast, electroporation-competent cells of yeast strain BJ5464 (ATCC.org; ATCC number 208288) were prepared and electroporated on a Gene Pulser II (Biorad, USA) with the electroporation-ready DNA from the step above essentially as described (Colby, D. W. et al. 2004 Methods Enzymology 388, 348-358). The only exception is that transformed cells were grown in non-inducing minimal selective SCD-Leu medium to accommodate the LEU2 selective marker carried by modified plasmid pBYDS03.


Library size was determined by plating serial dilutions of freshly recovered cells on SCD-Leu agar plates and then extrapolating library size from the number of single colonies from plating that generated at least 50 colonies per plate. The remainder of the electroporated culture was grown to saturation and cells from this culture were subcultured into the same medium once more to minimize the fraction of untransformed cells. To maintain library diversity, this subculturing step was carried out using an inoculum that contained at least 10× more cells than the calculated library size. Cells from the second saturated culture were resuspended in fresh medium containing sterile 25% (weight/volume) glycerol to a density of 10×1010 per mL and frozen and stored at −80° C. (frozen library stock).


One liter of SCD-Leu media consists of 14.7 grams of sodium citrate dihydrate, 4.29 grams of citric acid monohydrate, 20 grams of dextrose, 6.7 grams of Difco brand yeast nitrogen base, and 1.6 grams yeast synthetic drop-out media supplement without leucine. Media was filtered sterilized before use using a 0.2 μM vacuum filter device.


Library size was determined by plating dilutions of freshly recovered cells on SCD-Leu agar plates and then extrapolating library size from the number of single colonies from a plating that generate at least 50 colonies per plate.


To segregate plasmid from cells that contain two or more different library clones, a number of cells corresponding to 10 times the library size, were taken from the overnight SCD-Leu culture and subcultured 1/100 into fresh SCD-Leu medium and grown overnight. Cells from this overnight culture were resuspended in sterile 25% (weight/volume) glycerol to a density of 10E10/mL and frozen and stored at −80° C. (frozen library stock).


Example 3
Yeast Selection

Example 3 describes the selection of yeast expressing affinity modified variants of CD80, CD86, ICOSL and NKp30.


A number of cells equal to at least 10 times the library size were thawed from individual library stocks, suspended to 0.1×10E6 cells/mL in non-inducing SCD-Leu medium, and grown overnight. The next day, a number of cells equal to 10 times the library size were centrifuged at 2000 RPM for two minutes and resuspended to 0.5×10E6 cells/mL in inducing SCDG-Leu media. One liter of the SCDG-Leu induction media consists of 5.4 grams Na2HPO4, 8.56 grams of NaH2PO4*H2O, 20 grams galactose, 2.0 grams dextrose, 6.7 grams Difco yeast nitrogen base, and 1.6 grams of yeast synthetic drop out media supplement without leucine dissolved in water and sterilized through a 0.22 μm membrane filter device. The culture was grown for two days at 20° C. to induce expression of library proteins on the yeast cell surface.


Cells were processed with magnetic beads to reduce non-binders and enrich for all CD80, CD86, ICOSL or NKp30 variants with the ability to bind their exogenous recombinant counter-structure proteins (cognate binding partners). For example, yeast displayed targeted or random CD80 libraries were selected against each of CD28, CTLA-4, PD-L1. ICOSL libraries were selected against ICOS and CD28 and NKp30 libraries were selected against B7-H6. This was then followed by two to three rounds of fluorescence activated cell sorting (FACS) using exogenous cognate binding partner protein staining to enrich the fraction of yeast cells that displays improved binders. Magnetic bead enrichment and selections by flow cytometry are essentially as described in Keith D. Miller, 1 Noah B. Pefaur, 2 and Cheryl L. Bairdl Current Protocols in Cytometry 4.7.1-4.7.30, July 2008.


With CD80, CD86, ICOSL, and NKp30 libraries, target ligand proteins were sourced from R&D Systems (USA) as follows: human rCD28.Fc (i.e., recombinant CD28-Fc fusion protein), rPDL1.Fc, rCTLA4.Fc, rICOS.Fc, and rB7H6.Fc. Magnetic streptavidin beads were obtained from New England Biolabs, USA. For biotinylation of cognate binding partner protein, biotinylation kit cat #21955, Life Technologies, USA, was used. For two-color, flow cytometric sorting, a Becton Dickinson FACS Aria II sorter was used. CD80, CD86, ICOSL, or NKp30 display levels were monitored with an anti-hemagglutinin tag antibody labeled with Alexafluor 488 (Life Technologies, USA). Ligand binding Fc fusion proteins rCD28.Fc, rCTLA4.Fc, rPDL1.Fc, rICOS.Fc, or rB7-H6.Fc were detected with PE conjugated human Ig specific goat Fab (Jackson ImmunoResearch, USA). Doublet yeast were gated out using forward scatter (FSC)/side scatter (SSC) parameters, and sort gates were based upon higher ligand binding detected in FL2 that possessed more limited HA tag expression binding in FL1.


Yeast outputs from the flow cytometric sorts were assayed for higher specific binding affinity. Sort output yeast were expanded and re-induced to express the particular IgSF affinity modified domain variants they encode. This population then can be compared to the parental, wild-type yeast strain, or any other selected outputs, such as the bead output yeast population, by flow cytometry.


For ICOSL, the second sort outputs (F2) were compared to parental ICOSL yeast for binding of each rICOS.Fc, rCD28.Fc, and rCTLA4.Fc by double staining each population with anti-HA (hemagglutinin) tag expression and the anti-human Fc secondary to detect ligand binding.


In the case of ICOSL yeast variants selected for binding to ICOS, the F2 sort outputs gave Mean Fluorescence Intensity (MFI) values of 997, when stained with 5.6 nM rICOS.Fc, whereas the parental ICOSL strain MFI was measured at 397 when stained with the same concentration of rICOS.Fc. This represents a roughly three-fold improvement of the average binding in this F2 selected pool of clones, and it is predicted that individual clones from that pool will have much better improved MFI/affinity when individually tested.


In the case of ICOSL yeast variants selected for binding to CD28, the F2 sort outputs gave MFI values of 640 when stained with 100 nM rCD28.Fc, whereas the parental ICOSL strain MFI was measured at 29 when stained with the same concentration of rCD28.Fc (22-fold improvement). In the case of ICOSL yeast variants selected for binding to CTLA4, the F2 sort outputs gave MFI values of 949 when stained with 100 nM rCTLA4.Fc, whereas the parental ICOSL strain MFI was measured at 29 when stained with the same concentration of rCTLA4.Fc (32-fold improvement).


In the case of NKp30 yeast variants selected for binding to B7-H6, the F2 sort outputs gave MFI values of 533 when stained with 16.6 nM rB7H6.Fc, whereas the parental NKp30 strain MFI was measured at 90 when stained with the same concentration of rB7H6.Fc (6-fold improvement).


Among the NKp30 variants that were identified, was a variant that contained mutations L30V/A60V/S64P/S86G with reference to positions in the NKp30 extracellular domain corresponding to positions set forth in SEQ ID NO:54. Among the CD86 variants that were identified, was a variant that contained mutations Q35H/H90L/Q102H with reference to positions in the CD86 extracellular domain corresponding to positions set forth in SEQ ID NO:29.


Importantly, the MFIs of all F2 outputs described above when measured with the anti-HA tag antibody on FL1 did not increase and sometimes decreased compared to wild-type strains, indicating that increased binding was not a function of increased expression of the selected variants on the surface of yeast, and validated gating strategies of only selecting mid to low expressors with high ligand binding.


Example 4
Reformatting Selection Outputs as Fc-Fusions and in Various Immunomodulatory Protein Types

Example 4 describes reformatting of selection outputs as immunomodulatory proteins containing an affinity modified (variant) extracellular domain (ECD) of CD80 or ICOSL fused to an Fc molecule (variant ECD-Fc fusion molecules).


Output cells from final flow cytometric CD80 and ICOSL sorts were grown to terminal density in SCD-Leu medium. Plasmid DNA from each output was isolated using a yeast plasmid DNA isolation kit (Zymo Research, USA). For Fc fusions, PCR primers with added restriction sites suitable for cloning into the Fc fusion vector of choice were used to batch-amplify from the plasmid DNA preps the coding DNA for the mutant target's ECD. After restriction digestion, the PCR products were ligated into an appropriate Fc fusion vector followed by chemical transformation into strain E. coli strain XL1 Blue E. coli (Agilent, USA) or NEB5alpha (New England Biolabs, USA) as directed by supplier. Exemplary of an Fc fusion vector is pFUSE-hIgG1-Fc2 (InvivoGen, USA).


Dilutions of transformation reactions were plated on LB-agar containing 100 μg/mL carbenicillin (Teknova, USA) to generate single colonies. Up to 96 colonies from each transformation were then grown in 96 well plates to saturation overnight at 37° C. in LB-broth (Teknova cat # L8112) and a small aliquot from each well was submitted for DNA sequencing of the ECD insert in order to identify mutation(s) in all clones. Sample preparation for DNA sequencing was carried out using protocols provided by the service provider (Genewiz; South Plainfield, N.J.). After removal of sample for DNA sequencing, glycerol was then added to the remaining cultures for a final glycerol content of 25% and plates were stored at −20° C. for future use as master plates (see below). Alternatively, samples for DNA sequencing were generated by replica plating from grown liquid cultures to solid agar plates using a disposable 96 well replicator (VWR, USA). These plates were incubated overnight to generate growth patches and the plates were submitted to Genewiz as specified by Genewiz.


After identification of clones of interest from analysis of Genewiz-generated DNA sequencing data, clones of interest were recovered from master plates and individually grown to density in 5 mL liquid LB-broth containing 100 μg/mL carbenicillin (Teknova, USA) and 2 mL of each culture were then used for preparation of approximately 10 μg of miniprep plasmid DNA of each clone using a standard kit such as the Pureyield kit (Promega). Identification of clones of interest generally involved the following steps. First, DNA sequence data files were downloaded from the Genewiz website. All sequences were then manually curated so that they start at the beginning of the ECD coding region. The curated sequences were then batch-translated using a suitable program available at the URL: www.ebi.ac.uk/Tools/st/emboss_transeq/. The translated sequences were then aligned using a suitable program available at the URL:multalin.toulouse.inra.fr/multalin/multalin.html. Alternatively, Genewiz sequenced were processed to generate alignments using Ugene software (http://ugene.net).


Clones of interest were then identified using the following criteria: 1.) identical clone occurs at least two times in the alignment and 2.) a mutation occurs at least two times in the alignment and preferably in distinct clones. Clones that meet at least one of these criteria were clones that have been enriched by our sorting process most likely due to improved binding.


The methods generated immunomodulatory proteins containing an ECD of CD80 or ICOSL with at least one affinity-modified domain in which the encoding DNA was generated to encode a protein designed as follows: signal peptide followed by variant (mutant) ECD followed by a linker of three alanines (AAA) followed by a human IgG1 Fc set forth in SEQ ID NO:2084 containing the mutation N297G (N82G with reference to wild-type human IgG1 Fc set forth in SEQ ID NO: 226). The human IgG1 Fc also contained the mutations R292C and V302C (corresponding to R77C and V87C with reference to wild-type human IgG1 Fc set forth in SEQ ID NO: 226). Since the construct does not include any antibody light chains that can form a covalent bond with a cysteine, the human IgG1 Fc also contained replacement of the cysteine residues to a serine residue at position 220 (C220S) by EU numbering (corresponding to position 5 (C5S) with reference 5 (C5S) compared to the wild-type or unmodified Fc set forth in SEQ ID NO: 226.


In addition, Example 8 below describes further immunomodulatory proteins that were generated as stack constructs containing at least two different affinity modified domains from identified variant CD80, CD86, ICOSL, and NKp30 molecules linked together and fused to an Fc.


Example 5
Expression and Purification of Fc-Fusions

Example 5 describes the high throughput expression and purification of Fc-fusion proteins containing variant ECD CD80, CD86, ICOSL, and NKp30 as described in the above Examples.


Recombinant variant Fc fusion proteins were produced from suspension-adapted human embryonic kidney (HEK) 293 cells using the Expi293 expression system (Invitrogen, USA). 4 μg of each plasmid DNA from the previous step was added to 200 μL Opti-MEM (Invitrogen, USA) at the same time as 10.8 μL ExpiFectamine was separately added to another 200 μL Opti-MEM. After 5 minutes, the 200 μL of plasmid DNA was mixed with the 200 μL of ExpiFectamine and was further incubated for an additional 20 minutes before adding this mixture to cells. Ten million Expi293 cells were dispensed into separate wells of a sterile 10 mL, conical bottom, deep 24 well growth plate (Thomson Instrument Company, USA) in a volume 3.4 mL Expi293 media (Invitrogen, USA). Plates were shaken for 5 days at 120 RPM in a mammalian cell culture incubator set to 95% humidity and 8% CO2. Following a 5 day incubation, cells were pelleted and culture supernatants were retained.


Proteins were purified from supernatants using a high throughput 96 well Protein A purification kit using the manufacturer's protocol (Catalog number 45202, Life Technologies, USA). Resulting elution fractions were buffer exchanged into PBS using Zeba 96 well spin desalting plate (Catalog number 89807, Life Technologies, USA) using the manufacturer's protocol. Purified protein was quantitated using 280 nm absorbance measured by Nanodrop instrument (Thermo Fisher Scientific, USA), and protein purity was assessed by loading 5 μg of protein on NUPAGE pre-cast, polyacrylamide gels (Life Technologies, USA) under denaturing and reducing conditions and subsequent gel electrophoresis. Proteins were visualized in gel using standard Coomassie staining.


Example 6
Assessment of Binding and Activity of Affinity-Matured IgSF Domain-Containing Molecules

A. Binding to Cell Surface-Expressed Cognate Binding Partners


This Example describes Fc-fusion binding studies of purified proteins from the above Examples to assess specificity and affinity of CD80 and ICOSL domain variant immunomodulatory proteins for cognate binding partners.


To produce cells expressing cognate binding partners, full-length mammalian surface expression constructs for each of human CD28, CTLA4, PD-L1, ICOS and B7-H6 were designed in pcDNA3.1 expression vector (Life Technologies) and sourced from Genscript, USA. Binding studies were carried out using the Expi293F transient transfection system (Life Technologies, USA) described above. The number of cells needed for the experiment was determined, and the appropriate 30 mL scale of transfection was performed using the manufacturer's suggested protocol. For each CD28, CTLA-4, PD-L1, ICOS, B7-H6, or mock 30 mL transfection, 75 million Expi293F cells were incubated with 30 μg expression construct DNA and 1.5 mL diluted ExpiFectamine 293 reagent for 48 hours, at which point cells were harvested for staining.


For staining by flow cytometry, 200,000 cells of appropriate transient transfection or negative control were plated in 96 well round bottom plates. Cells were spun down and resuspended in staining buffer (PBS (phosphate buffered saline), 1% BSA (bovine serum albumin), and 0.1% sodium azide) for 20 minutes to block non-specific binding. Afterwards, cells were centrifuged again and resuspended in staining buffer containing 100 nM to 1 nM variant immunomodulatory protein, depending on the experiment of each candidate CD80 variant Fc, ICOSL variant Fc, or stacked IgSF variant Fc fusion protein in 50 μl. Primary staining was performed on ice for 45 minutes, before washing cells in staining buffer twice. PE-conjugated anti-human Fc (Jackson ImmunoResearch, USA) was diluted 1:150 in 50 μl staining buffer and added to cells and incubated another 30 minutes on ice. Secondary antibody was washed out twice, cells were fixed in 4% formaldehyde/PBS, and samples were analyzed on FACScan flow cytometer (Becton Dickinson, USA).


Mean Fluorescence Intensity (MFI) was calculated for each transfectant and negative parental line with Cell Quest Pro software (Becton Dickinson, USA).


B. Bioactivity Characterization


This Example further describes Fc-fusion variant protein bioactivity characterization in human primary T cell in vitro assays.


I. Mixed Lymphocyte Reaction (MLR)


Soluble rICOSL.Fc or rCD80.Fc bioactivity was tested in a human Mixed Lymphocyte Reaction (MLR). Human primary dendritic cells (DC) were generated by culturing monocytes isolated from PBMC (BenTech Bio, USA) in vitro for 7 days with 500U/mL rIL-4 (R&D Systems, USA) and 250 U/mL rGM-CSF (R&D Systems, USA) in Ex-Vivo 15 media (Lonza, Switzerland). 10,000 matured DC and 100,000 purified allogeneic CD4+ T cells (BenTech Bio, USA) were co-cultured with ICOSL variant fusion protein, CD80 variant Fc fusion protein, or controls in 96 well round bottom plates in 200 μl final volume of Ex-Vivo 15 media. On day 5, IFN-gamma secretion in culture supernatants was analyzed using the Human IFN-gamma Duoset ELISA kit (R&D Systems, USA). Optical density was measured by VMax ELISA Microplate Reader (Molecular Devices, USA) and quantitated against titrated rIFN-gamma standard included in the IFN-gamma Duo-set kit (R&D Systems, USA).


2 Anti-CD3 Coimmobilization Assay


Costimulatory bioactivity of ICOSL fusion variants and CD80 Fc fusion variants was determined in anti-CD3 coimmobilization assays. 1 nM or 4 nM mouse anti-human CD3 (OKT3, Biolegends, USA) was diluted in PBS with 1 nM to80 nM rICOSL.Fc or rCD80.Fc variant proteins. This mixture was added to tissue culture treated flat bottom 96 well plates (Corning, USA) overnight to facilitate adherence of the stimulatory proteins to the wells of the plate. The next day, unbound protein was washed off the plates and 100,000 purified human pan T cells (BenTech Bio, US) or human T cell clone BC3 (Astarte Biologics, USA) were added to each well in a final volume of 200 μL of Ex-Vivo 15 media (Lonza, Switzerland). Cells were cultured 3 days before harvesting culture supernatants and measuring human IFN-gamma levels with Duoset ELISA kit (R&D Systems, USA) as mentioned above.


C. Results


Results for the binding and activity studies for exemplary tested variants are shown in Tables 12-15. In particular, Table 12 indicates exemplary IgSF domain amino acid substitutions (replacements) in the ECD of CD80 selected in the screen for affinity-maturation against the respective cognate structure CD28. Table 13 indicates exemplary IgSF domain amino acid substitutions (replacements) in the ECD of CD80 selected in the screen for affinity-maturation against the respective cognate structure PD-L1. Table 14 indicates exemplary IgSF domain amino acid substitutions (replacements) in the ECD of ICOSL selected in the screen for affinity-maturation against the respective cognate structures ICOS and CD28. For each Table, the exemplary amino acid substitutions are designated by amino acid position number corresponding to the respective reference unmodified ECD sequence as follows. For example, the reference unmodified ECD sequence in Tables 12 and 13 is the unmodified CD80 ECD sequence set forth in SEQ ID NO:28 and the reference unmodified ECD sequence in Table 14 is the unmodified ICOSL ECD sequence (SEQ ID NO:32). The amino acid position is indicated in the middle, with the corresponding unmodified (e.g. wild-type) amino acid listed before the number and the identified variant amino acid substitution listed after the number. Column 2 sets forth the SEQ ID NO identifier for the variant ECD for each variant ECD-Fc fusion molecule.


Also shown is the binding activity as measured by the Mean Fluorescence Intensity (MFI) value for binding of each variant Fc-fusion molecule to cells engineered to express the cognate binding partner ligand and the ratio of the MFI compared to the binding of the corresponding unmodified ECD-Fc fusion molecule not containing the amino acid substitution(s) to the same cell-expressed cognate binding partner ligand. The functional activity of the variant Fc-fusion molecules to modulate the activity of T cells also is shown based on the calculated levels of IFN-gamma in culture supernatants (pg/mL) generated either i) with the indicated variant ECD-Fc fusion molecule coimmoblized with anti-CD3 or ii) with the indicated variant ECD-Fc fusion molecule in an MLR assay. The Tables also depict the ratio of IFN-gamma produced by each variant ECD-Fc compared to the corresponding unmodified ECD-Fc in both functional assays.


As shown, the selections resulted in the identification of a number of CD80 or ICOSL IgSF domain variants that were affinity-modified to exhibit increased binding for at least one, and in some cases more than one, cognate binding partner ligand. In addition, the results showed that affinity modification of the variant molecules also exhibited improved activities to both increase and decrease immunological activity depending on the format of the molecule. For example, coimmobilization of the ligand likely provides a multivalent interaction with the cell to cluster or increase the avidity to favor agonist activity and increase T cell activation compared to the unmodified (e.g. wildtype) ECD-Fc molecule not containing the amino acid replacement(s). However, when the molecule is provided as a bivalent Fc molecule in solution, the same IgSF domain variants exhibited an antagonist activity to decrease T cell activation compared to the unmodified (e.g. wildtype) ECD-Fv molecule not containing the amino acid replacement(s).









TABLE 12







CD80 variants selected against CD28. Molecule sequences, binding data, and


costimulatory bioactivity data.


















Coimmobilization
MLR













Binding
with anti-CD3
IFN-gamma














SEQ
CD28
CTLA-4
PD-L1
IFN-gamma
levels



ID
MFI
MFI
MFI
pg/mL
pg/mL



NO
(parental
(parental
(parental
(parental
(parental


CD80 mutation(s)
(ECD)
ratio)
ratio)
ratio)
ratio)
ratio)
















L70Q/A91G
55
125
283
6
93
716




(1.31)
(1.36)
(0.08)
(1.12)
(0.83)


L70Q/A91G/T130A
56
96
234
7
99
752




(1.01)
(1.13)
(0.10)
(1.19)
(0.87)


L70Q/A91G/I118A/
57
123
226
7
86
741


T120S/T130A

(1.29)
(1.09)
(0.10)
(1.03)
(0.86)


V4M/L70Q/A91G/
58
89
263
6
139
991


T120S/T130A

(0.94)
(1.26)
(0.09)
(1.67)
(1.14)


L70Q/A91G/T120S/
59
106
263
6
104
741


T130A

(1.12)
(1.26)
(0.09)
(1.25)
(0.86)


V20L/L70Q/A91S/
60
105
200
9
195
710


T120S/T130A

(1.11)
(0.96)
(0.13)
(2.34)
(0.82)


S44P/L70Q/A91G/
61
88
134
5
142
854


T130A

(0.92)
(0.64)
(0.07)
(1.71)
(0.99)


L70Q/A91G/E117G/
62
120
193
6
98
736


T120S/T130A

(1.27)
(0.93)
(0.08)
(1.05)
(0.85)


A91G/T120S/
63
84
231
44
276
714


T130A

(0.89)
(1.11)
(0.62)
(3.33)
(0.82)


L70R/A91G/T120S/
64
125
227
6
105
702


T130A

(1.32)
(1.09)
(0.09)
(1.26)
(0.81)


L70Q/E81A/A91G/
65
140
185
18
98
772


T120S/I127T/T130A

(1.48)
(0.89)
(0.25)
(1.18)
(0.89)


L70Q/Y87N/A91G/
66
108
181
6
136
769


T130A

(1.13)
(0.87)
(0.08)
(1.63)
(0.89)


T28S/L70Q/A91G/
67
32
65
6
120
834


E95K/T120S/T130A

(0.34)
(0.31)
(0.08)
(1.44)
(0.96)


N63S/L70Q/A91G/
68
124
165
6
116
705


T120S/T130A

(1.30)
(0.79)
(0.08)
(1.39)
(0.81)


K36E/I67T/L70Q/
69
8
21
5
53
852


A91G/T120S/

(0.09)
(0.10)
(0.08)
(0.63)
(0.98)


T130A/N152T
70
113
245
6
94
874


E52G/L70Q/A91G/

(1.19)
(1.18)
(0.08)
(1.13)
(1.01)


T120S/T130A








K37E/F59S/L70Q/
71
20
74
6
109
863


A91G/T120S/T130A

(0.21)
(0.36)
(0.08)
(1.31)
(1.00)


A91G/S103P
72
39
56
9
124
670




(0.41)
(0.27)
(0.13)
(1.49)
(0.77)


K89E/T130A
73
90
148
75
204
761




(0.95)
(0.71)
(1.07)
(2.45)
(0.88)


A91G
74
96
200
85
220
877




(1.01)
(0.96)
(1.21)
(2.65)
(1.01)


D60V/A91G/T120S/
75
111
222
12
120
744


T130A

(1.17)
(1.07)
(0.18)
(1.44)
(0.86)


K54M/A91G/T120S
76
68
131
5
152
685




(0.71)
(0.63)
(0.08)
(1.83)
(0.79)


M38T/L70Q/E77G/
77
61
102
5
119
796


A91G/T120S/

(0.64)
(0.49)
(0.07)
(1.43)
(0.92)


T130A/N152T








R29H/E52G/L70R/
78
100
119
5
200
740


E88G/A91G/T130A

(1.05)
(0.57)
(0.08)
(2.41)
(0.85)


Y31H/T41G/L70Q/
79
85
85
6
288
782


A91G/T120S/T130A

(0.89)
(0.41)
(0.08)
(3.47)
(0.90)


V68A/T110A
80
103
233
48
163
861




(1.08)
(1.12)
(0.68)
(1,96)
(0.99)


S66H/D90G/T110A/
81
33
121
11
129
758


F116L

(0.35)
(0.58)
(0.15)
(1.55)
(0.88)


R29H/E52G/T120S/
82
66
141
11
124
800


T130A

(0.69)
(0.68)
(0.15)
(1.49)
(0.92)


A91G/L102S
83
6
6
5
75
698




(0.06)
(0.03)
(0.08)
(0.90)
(0.81)


I67T/L70Q/A91G/
84
98
160
5
1751
794


T120S

(1.03)
(0.77)
(0.08)
(21.1)
(0.92)


L70Q/A91G/T110A/
85
8
14
5
77
656


T120S/T130A

(0.09)
(0.07)
(0.07)
(0.93)
(0.76)


M38V/T41D/M43I/
86
5
8
8
82
671


W50G/D76G/V83A/

(0.06)
(0.04)
(0.11)
(0.99)
(0.78)


K89E/T120S/T130A








V22A/L70Q/S121P
87
5
7
5
105
976




(0.06)
(0.04)
(0.07)
(1.27)
(1.13)


A12V/S15F/Y31H/
88
6
6
5
104
711


T41G/T130A/P137L/

(0.06)
(0.03)
(0.08)
(1.25)
(0.82)


N152T








I67F/L70R/E88G/
89
5
6
6
62
1003


A91G/T120S/T130A

(0.05)
(0.03)
(0.08)
(0.74)
(1.16)


E24G/L25P/L70Q/
90
26
38
8
101
969


T120S

(0.27)
(0.18)
(0.11)
(1.21)
(1.12)


A91G/F92L/F108L/
91
50
128
16
59
665


T120S

(0.53)
(0.61)
(0.11)
(0.71)
(0.77)


WT CD80
28
95
208
70
83
866




(1.00)
(1.00)
(1.00)
(1.00)
(1.00)
















TABLE 13







CD80 variants selected against PD-L1. Molecule sequences, binding data, and


costimulatory bioactivity data.


















Coimmobilization
MLR













Binding
with anti-CD3
IFN-gamma














SEQ
CD28
CTLA-4
PD-L1
IFN-gamma
levels



ID
MFI
MFI
MFI
pg/mL
pg/mL



NO
(parental
(parental
(parental
(parental
(parental


CD80 mutation(s)
(ECD)
ratio)
ratio)
ratio)
ratio)
ratio)
















R29D/Y31L/Q33H/
92
1071
1089
37245
387
5028


K36G/M38I/T41A/

(0.08)
(0.02)
(2.09)
(0.76)
(0.26)


M43R/M47T/E81V/








L85R/K89N/A91T/








F92P/K93V/R94L/








I118T/N149S








R29D/Y31L/Q33H/
93
1065
956
30713
400
7943


K36G/M381/T41A/

(0.08)
(0.02)
(1.72)
(0.79)
(0.41)


M43R/M47T/E81V/








L85R/K89N/A91T/








F92P/K93V/R94L/








N144S/N149S








R29D/Y31L/Q33H/
94
926
954
47072
464
17387


K36G/M38I/T41A/

(0.07)
(0.02)
(2.64)
(0.91)
(0.91)


M42T/M43R/M47T/








E81V/L85R/K89N/








A91T/F92P/K93V/








R94L/L148S/N149S








E24G/R29D/Y31L/
95
1074
1022
1121
406
13146


Q33H/K36G/M38I/

(0.08)
(0.02)
(0.06)
(0.80)
(0.69)


T41A/M43R/M47T/








F59L/E81V/L85R/








K89N/A91T/F92P/








K93V/R94L/H96R








R29D/Y31L/Q33H/
96
1018
974
25434
405
24029


K36G/M38I/T41A/

(0.08)
(0.02)
(1.43)
(0.80)
(1.25)


M43R/M47T/E81V/








L85R/K89N/A91T/








F92P/K93V/R94L/








N149S








R29V/M43Q/E81R/
97
1029
996
1575
342
11695


L85I/K89R/D90L/

(0.08)
(0.02)
(0.09)
(0.67)
(0.61)


A91E/F92N/K93Q/








R94G








T41I/A91G
98
17890
50624
12562
433
26052




(1.35)
(1.01)
(0.70)
(0.85)
(1.36)


K89R/D90K/A91G/
99
41687
49429
20140
773
6345


F92Y/K93R/N122S/

(3.15)
(0.99)
(1.13)
(1.52)
(0.33)


N178S








K89R/D90K/A91G/
100
51663
72214
26405
1125
9356


F92Y/K93R

(3.91)
(1.44)
(1.48)
(2.21)
(0.49)


K36G/K37Q/M38I/
101
1298
1271
3126
507
3095


F59L/E81V/L85R/

(0.10)
(0.03)
(0.18)
(1.00)
(0.16)


K89N/A91T/F92P/








K93V/R94L/E99G/








T130A/N149S








AE88D/K89R/D90K/
102
31535
50868
29077
944
5922


A91G/F92Y/K93R

(2.38)
(1.02)
(1.63)
(1.85)
(0.31)


K36G/K37Q/M38I/
103
1170
1405
959
427
811


L40M

(0.09)
(0.03)
(0.05)
(0.84)
(0.04)


K36G
104
29766
58889
20143
699
30558




(2.25)
(1.18)
(1.13)
(1.37)
(1.59)


WTCD80
28
13224
50101
17846
509
19211




(1.00)
(1.00)
(1.00)
(1.00)
(1.00)
















TABLE 14







ICOSL variants selected against CD28 or ICOS. Molecule sequences, binding data, and


costimulatory bioactivity data.














Coimmobilization
MLR





with anti-CD3
IFN-gamma



SEQ
Binding
IFN-gamma
levels pg/mL













ID NO
ICOS OD
CD28 MFI
pg/mL
(parental


ICOSL mutation(s)
(ECD)
(parental ratio)
(parental ratio)
(parental ratio)
ratio)















N52S
109
1.33
162
1334    
300   




(1.55)
(9.00)
(1.93)
(0.44)


N52H
110
1.30
368
1268    
39   




(1.51)
(20.44)
(1.83)
(0.06)


N52D
111
1.59
130
1943    
190   




(1.85)
(7.22)
(2.80)
(0.28)


N52Y/N57Y/
112
1.02
398
510*   
18   


F138L/L203P

(1.19)
(22.11)
 (1.47*)
(0.03)


N52H/N57Y/Q100P
113
1.57
447
2199    
25   




(1.83)
(24.83)
(3.18)
(0.04)


N52S/Y146C/
114
1.26
39
1647    
152   


Y152C

(1.47)
(2.17)
(2.38)
(0.22)


N52H/C198R
115
1.16
363
744*   
ND




(1.35)
(20.17)
 (2.15*)
(ND)


N52H/C140del/
1563
ND
154
522*   
ND


T225A

(ND)
(8.56)
 (1.51*)
(ND)


N52H/C198R/
117
1.41
344
778*   
0  


T225A

(1.64)
(19.11)
 (2.25*)
(0)  


N52H/K92R
118
1.48
347
288*   
89   




(1.72)
(19.28)
 (0.83*)
(0.13)


N52H/S99G
119
0.09
29
184*   
421   




(0.10)
(1.61)
 (0.53*)
(0.61)


N52Y
120
0.08
18
184*   
568   




(0.09)
(1.00)
 (0.53*)
(0.83)


N57Y
121
1.40
101
580*   
176   




(1.63)
(5.61)
 (1.68*)
(0.26)


N57Y/Q100P
122
0.62
285
301*   
177   




(0.72)
(15.83)
 (0.87*)
(0.26)


N52S/S130G/
123
0.16
24
266*   
1617    


Y152C

(0.19)
(1.33)
 (0.77*)
(2.35)


N52S/Y152C
124
0.18
29
238*   
363   




(0.21)
(1.61)
 (0.69*)
(0.53)


N52S/C198R
125
1.80
82
1427    
201   




(2.09)
(4.56)
(2.06)
(0.29)


N52Y/N57Y/Y152C
126
0.08
56
377*   
439   




(0.09)
(3.11)
 (1.09*)
(0.64)


N52Y/N57Y/
127
ND
449
1192   
ND


H129P/C198R

(ND)
(24.94)
(1.72)
(ND)


N52H/L161P/
128
0.18
343
643*   
447   


C198R

(0.21)
(19.05)
 (1.86*)
(0.65)


N52S/T113E
129
1.51
54
451*   
345   




(1.76)
(3.00)
 (1.30*)
(0.50)


S54A
130
1.62
48
386*   
771   




(1.88)
(2.67)
 (1.12*)
(1.12)


N52S/S54P
1564
1.50
38
476*   
227   




(1.74)
(2.11)
 (1.38*)
(0.33)


N52K/L208P
132
1.91
291
1509    
137   




(2.22)
(16.17)
(2.18)
(0.20)


N52S/Y152H
133
0.85
68
2158    
221   




(0.99)
(3.78)
(3.12)
(0.32)


N52D/V151A
134
0.90
19
341*   
450   




(1.05)
(1.06)
 (0.99*)
(0.66)


N52H/I143T
135
1.83
350
2216    
112   




(2.13)
(19.44)
(3.20)
(0.16)


N52S/L80P
136
0.09
22
192*   
340   




(0.10)
(1.22)
 (0.55*)
(0.49)


F120S/Y152H/
137
0.63
16
351*   
712   


N201S

(0.73)
(0.89)
 (1.01*)
(1.04)


N52S/R75Q/L203P
138
1.71
12
1996    
136   




(1.99)
(0.67)
(2.88)
(0.20)


N52S/D158G
139
1.33
39
325*   
277   




(1.55)
(2.17)
 (0.94*)
(0.40)


N52D/Q133H
140
1.53
104
365*   
178   




(1.78)
(5.78)
 (1.05*)
(0.26)


WT ICOSL
32
0.86
18
692/346*
687   




(1.00)
(1.00)
(1.00)
(1.00)





*Parental ratio calculated using 346 pg/mL IFN-gamma for WT ICOSL






Example 7
Ligand Binding Competition Assay

As shown in Example 6, several CD80 variant molecules exhibited improved binding to one or both of CD28 and PD-L1. To further assess the binding activity of CD80 to ligands CD28 and PD-L1, this Example describes a ligand competition assay assessing the non-competitive nature of exemplary CD80 variants to bind both CD28 and PD-L1.


An ELISA based binding assay was set up incorporating plate-bound CD80 variant A91G ECD-Fc to assess the ability of CD80 to simultaneously bind CD28 and PD-L1. Maxisorp 96 well ELISA plates (Nunc, USA) were coated overnight with 100 nM human recombinant CD80 variant A91G ECD-Fc fusion protein in PBS. The following day unbound protein was washed out, and the plate was blocked with 1% bovine serum albumin (Millipore, USA)/PBS for 1 hour at room temperature. This blocking reagent was washed out three times with PBS/0.05% Tween, which included a two minute incubation on a platform shaker for each wash.


In one arm of the competition assay, CD80 was incubated with CD28, and then CD28-bound CD80 was then assessed for competitive binding in the presence of either the other known CD80 cognate binding partner PD-L1 or CTLA-4 or negative control ligand PD-L2. Specifically, biotinylated recombinant human CD28 Fc fusion protein (rCD28.Fc; R&D Systems) was titrated into the wells starting at 10 nM, diluting out for eight points with 1:2 dilutions in 25 μL volume. Immediately after adding the biotinylated rCD28.Fc, unlabeled competitive binders, recombinant human PD-L1 monomeric his-tagged protein, recombinant human CTLA-4 monomeric his-tagged protein, or a negative control human recombinant PD-L2 Fc fusion protein (R&D Systems) were added to wells at 2000/1000/500 nM respectively in 25 μL volume for a final volume of 50 μL. These proteins were incubated together for one hour before repeating the three wash steps as described above.


After washing, 2.5 ng per well of HRP-conjugated streptavidin (Jackson Immunoresearch, USA) was diluted in 1% BSA/PBS and added to wells to detect bound biotinylated rCD28.Fc. After one hour incubation, wells were washed again three times as described above. To detect signal, 50 μL of TMB substrate (Pierce, USA) was added to wells following wash and incubated for 7 minutes, before adding 50 ul 2M sulfuric acid stop solution. Optical density was determined on an Emax Plus microplate reader (Molecular Devices, USA). Optical density values were graphed in Prism (Graphpad, USA).


The results are set forth in FIG. 1A. The results showed decreased binding of biotinylated rCD28.Fc to the CD80 variant A91G ECD-Fc fusion protein with titration of the rCD28.Fc. When rCD28.Fc binding was performed in the presence of non-competitive control protein, rPDL2, there was no decrease in CD28 binding for CD80 (solid triangle). In contrast, a competitive control protein, rCTLA-4, when incubated with the CD28.Fc, did result in decreased CD28 binding for CD80 as expected (x line). When recombinant PD-L1 was incubated with CD28.Fc, no decrease in CD28 binding to CD80 was observed, which demonstrated that the epitopes of CD28 and PD-L1 for CD80 are non-competitive. Binding of the recombinant PD-L1 protein used in the CD28 competition assay to CD80 was confirmed by incubating the biotinylated PD-L1 in the presence of non-biotinylated rCD28.Fc (square).


The reverse competition also was set up in which CD80 was incubated with PD-L1, and then PD-L1-bound CD80 was then assessed for competitive binding in the presence of either the other known CD80 cognate binding partners CD28 or CTLA-4 or negative control ligand PD-L2. Specifically, the assay was performed by titrating biotinylated recombinant human PD-L1-his monomeric protein into wells containing the recombinant CD80 variant. Because binding is weaker with this ligand, titrations started at 5000 nM with similar 1:2 dilutions over eight points in 25 μL. When the rPD-L1-his was used to detect binding, the competitive ligands human rCD28.Fc, human rCTLA-4.Fc, or human rPD-L2.Fc control were added at 2.5 nM final concentration in 25 μL for a total volume of 50 μL. The subsequent washes, detection, and OD measurements were the same as described above.


The results are set forth in FIG. 1B. Titrated PD-L1-his binding alone confirmed that PD-L1 bound to the CD80 variant A91G ECD-Fc fusion molecule immobilized on the plate (square). When PD-L1-his binding was performed in the presence of non-competitive control protein, rPDL2, there was no decrease in PD-L1 binding for CD80 (triangle). The CD28-competitive control protein, rCTLA-4, when incubated with the PD-L1-his, did not result in decreased PD-L1 binding for CD80 (x line), even though CTLA-4 is competitive for CD28. This result further demonstrated that lack of competition between CD28 and PD-L1 for CD80 binding. Finally, when PD-L1-his was incubated with CD28.Fc, no decrease in PD-L1 binding to CD80 was observed, which demonstrated that the epitopes of CD28 and PD-L1 for CD80 are non-competitive.


Thus, the results showed that CTLA-4, but not PD-L1 or the negative control PD-L2, competed for binding of CD28 to CD80 (FIG. 1A) and that CD28, CTLA-4, and PD-L2 did not compete for binding of PD-L1 to CD80 (FIG. 1B). Thus, these results demonstrated that CD28 and PD-L1 are non-competitive binders of CD80, and that this non-competitive binding can be demonstrated independently of which ligand is being detected in the ELISA.


Example 8
Generation and Assessment of Stacked Molecules Containing Different Affinity-Modified Domains

Selected variant molecules described above that were affinity-modified for one or more cognate binding partners were used to generate “stack” molecule (i.e., Type II immunomodulatory protein) containing two or more affinity-modified IgSF domains. Stack constructs were obtained as geneblocks (Integrated DNA Technologies, Coralville, Iowa) that encode the stack in a format that enables its fusion to Fc by standard Gibson assembly using a Gibson assembly kit (New England Biolabs). As above, this example exemplifies binding and activity of a molecule containing the affinity-modified domains in an Fc-fusion format; such stack molecules containing two or more affinity-modified domains are contemplated in connection with a secretable immunomodulatory protein or transmembrane immunomodulatory protein as described.


The encoding nucleic acid molecule of all stacks was generated to encode a protein designed as follows: Signal peptide, followed by the first variant IgV of interest, followed by a 15 amino acid linker which is composed of three GGGGS(G4S) motifs (SEQ ID NO:228), followed by the second IgV of interest, followed by two GGGGS linkers (SEQ ID NO: 229) followed by three alanines (AAA), followed by a human IgG1 Fc as described above. To maximize the chance for correct folding of the IgV domains in each stack, the first IgV was preceded by all residues that normally occur in the wild-type protein between this IgV and the signal peptide (leading sequence). Similarly, the first IgV was followed by all residues that normally connect it in the wild-type protein to either the next Ig domain (typically an IgC domain) or if such a second IgV domain is absent, the residues that connect it to the transmembrane domain (trailing sequence). The same design principle was applied to the second IgV domain except that when both IgV domains were derived from same parental protein (e.g. a CD80 IgV stacked with another CD80 IgV), the linker between both was not duplicated.


Table 15 sets forth the design for exemplary stacked constructs. The exemplary stack molecules shown in Table 15 contain the IgV domains as indicated and additionally leading or trailing sequences as described above. In the Table, the following components are present in order: signal peptide (SP; SEQ ID NO:225), IgV domain 1 (IgV1), trailing sequence 1 (TS1), linker 1 (LR1; SEQ ID NO:228), IgV domain 2(IgV2), trailing sequence 2 (TS2), linker 2 (LR2; SEQ ID NO:230) and Fc domain (SEQ ID NO:226 containing C5S/N82G amino acid substitution). In some cases, a leading sequence 1 (LS1) is present between the signal peptide and IgV1 and in some cases a leading sequence 2 (LS2) is present between the linker and IgV2.









TABLE 15







Amino acid sequence (SEQ ID NO) of components of exemplary stacked constructs















First domain

Second domain




















SP
LS1
IgV1
TS1
LR1
LS2
IgV2
TS2
LR2
Fc




















NKp30 WT
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


ICOSL WT


NO: 214
NO: 235


NO: 196
NO: 233




NKp30
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


L30V/A60V/S64P/


NO: 215
NO: 235


NO: 212
NO: 233




S86G












ICOSL












N525/N57Y/H94D/












L96F/L98F/Q100R












NKp30
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


L30V/A60V/S64P/


NO: 215
NO: 235


NO: 199
NO: 233




S86G)












ICOSL N52D












NKp30
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


L30V/A60V/S64P/


NO: 215
NO: 235


NO: 201
NO: 233




S86G












ICOSL












N52H/N57Y/Q100P












ICOSL WT
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


Nkp30 WT


NO: 196
NO: 233


NO: 214
NO: 235




ICOSL N52D
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


NKp30


NO: 199
NO: 233


NO: 215
NO: 235




L30V/A60V/S64P/












S86G












ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52H/N57Y/Q100P


NO: 201
NO: 233


NO: 215
NO: 235




NKp30












L30V/A60V/S64P/












S86G












Domain 1: NKp30
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


WT


NO: 214
NO: 235


NO: 152
NO: 231




Domain 2: CD80












WT












Domain 1: NKp30
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


WT


NO: 214
NO: 235

NO: 236
NO: 220
NO: 237




Domain 2: CD86












WT












Domain 1: NKp30
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


L30V/A60V/S64P/


NO: 215
NO: 235


NO: 192
NO: 231




S86G












Domain 2: CD80












R29H/Y31H/T41G/












Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 1: NKp30
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


L30V/A60V/S64P/


NO: 215
NO: 235


NO: 175
NO: 231




S86G












Domain 2: CD80












I67T/L70Q/A91G/












T120S












Domain 1: NKp30
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


L30V/A60V/S64P/


NO: 215
NO: 235

NO: 236
NO: 221
NO: 237




S86G












Domain 2: CD86












Q35H/H90L/Q102H












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


WT


NO: 152
NO: 231


NO: 214
NO: 235




Domain 2: Nkp30












WT












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


WT

NO: 236
NO: 220
NO: 237


NO: 214
NO: 235




Domain 2: Nkp30












WT












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


R29H/Y31H/T41G/


NO: 192
NO: 231


NO: 215
NO: 235




Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 2: NKp30












L30V/A60V/S64P/












S86G












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


I67T/L70Q/A91G/


NO: 175
NO: 231


NO: 215
NO: 235




T120S












Domain 2: NKp30












L30V/A60V/S64P/












S86G












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


Q35H/H9OL/Q102H

NO: 236
NO: 221
NO: 237


NO: 215
NO: 235




Domain 2: NKp30












L30V/A60V/S64P/












S86G












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


WT


NO: 152
NO: 231


NO: 196
NO: 233




Domain 2: ICOSL












WT












Domain 1: CD80
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


WT


NO: 152
NO: 231

NO: 236
NO: 220
NO: 237




Domain 2: CD86












WT












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


WT


NO: 152
NO: 231


NO: 152
NO: 231




Domain 2: CD80












WT












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


E88D/K89R/D90K/


NO: 189
NO: 231


NO: 192
NO: 231




A91G/F92Y/K93R












Domain 2: CD80












R29H/Y31H/T41G/












Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


A12T/H18L/M43V/


NO: 193
NO: 231


NO: 192
NO: 231




F59L/E77K/P109S/












I118T












Domain 2: CD80












R29H/Y31H/T41G/












Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


A12T/H18L/M43V/


NO: 193
NO: 231


NO: 175
NO: 231




F59L/E77K/P109S/












I118T












Domain 2: CD80












I67T/L70Q/A91G/












T120S












Domain 1: CD80
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


E88D/K89R/D90K/


NO: 189
NO: 231

NO: 236
NO: 221
NO: 237




A91G/F92Y/K93R












Domain 2: CD86












Q35H/H90L/Q102H












Domain 1: CD80
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


A12T/H18L/M43V/


NO: 193
NO: 231

NO: 236
NO: 221
NO: 237




F59L/E77K/P109S/












I118T












Domain 2: CD86












Q35H/H90L/Q102H












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


E88D/K89R/D90K/


NO: 189
NO: 231


NO: 213
NO: 233




A91G/F92Y/K93R












Domain 2: ICOSL












N525/N57Y/H94D/












L96F/L98F/Q100R/












G103E/F120S












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


A12T/H18L/M43V/


NO: 193
NO: 231


NO: 213
NO: 233




F59L/E77K/P109S/












I118T












Domain 2: ICOSL












N525/N57Y/H94D/












L96F/L98F/Q100R/












G103E/F120S












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


A12T/H18L/M43V/


NO: 193
NO: 231


NO: 199
NO: 233




F59L/E77K/P109S/












I118T












Domain 2: ICOSL












N52D












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


E88D/K89R/D90K/


NO: 189
NO: 231


NO: 201
NO: 233




A91G/F92Y/K93R












Domain 2: ICOSL












N52H/N57Y/Q100P












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


A12T/H18L/M43V/


NO: 193
NO: 231


NO: 201
NO: 233




F59L/E77K/P109S/












I118T












Domain 2: ICOSL












N52H/N57Y/Q100P












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


WT


NO: 196
NO: 233


NO: 152
NO: 231




Domain 2: CD80












WT












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


WT

NO: 236
NO: 220
NO: 237


NO: 152
NO: 231




Domain 2: CD80












WT












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


R29H/Y31H/T41G/


NO: 192
NO: 231


NO: 189
NO: 231




Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 2: CD80












E88D/K89R/D90K/












A91G/F92Y/K93R












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


R29H/Y31H/T41G/


NO: 192
NO: 231


NO: 193
NO: 231




Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 2: CD80












A12T/H18L/M43V/












F59L/E77K/P109S/












I118T












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


I67T/L70Q/A91G/


NO: 175
NO: 231


NO: 189
NO: 231




T120S












Domain 2: CD80












E88D/K89R/D90K/












A91G/F92Y/K93R












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


I67T/L70Q/A91G/


NO: 175
NO: 231


NO: 193
NO: 231




T120S












Domain 2: CD80












A12T/H18L/M43V/












F59L/E77K/P109S/












I118T












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


Q35H/H90L/Q102H

NO: 236
NO: 221
NO: 237


NO: 189
NO: 231




Domain 2: CD80












E88D/K89R/D90K/












A91G/F92Y/K93R












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


Q35H/H90L/Q102H

NO: 236
NO: 221
NO: 237


NO: 193
NO: 231




Domain 2: CD80












A12T/H18L/M43V/












F59L/E77K/P109S/












I118T












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52S/N57Y/H94D/


NO: 213
NO: 233


NO: 189
NO: 231




L96F/L98F/Q100R/












G103E/












F120S












Domain 2: CD80












E88D/K89R/D90K/












A91G/F92Y/K93R












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52S/N57Y/H94D/


NO: 213
NO: 233


NO: 193
NO: 231




L96F/L98F/Q100R/












G103E/F120S












Domain 2: CD80












A12T/H18L/M43V/












F59L/E77K/P109S/












I118T












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52D


NO: 199
NO: 233


NO: 189
NO: 231




Domain 2: CD80












E88D/K89R/D90K/












A91G/F92Y/K93R












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52D


NO: 199
NO: 233


NO: 193
NO: 231




Domain: 2 CD80












A12T/H18L/M43V/












F59L/E77K/P109S/












I118T












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52H/N57Y/Q100P


NO: 201
NO: 233


NO: 189
NO: 231




Domain 2: CD80












E88D/K89R/D90K/












A91G/F92Y/K93R












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52H/N57Y/Q100P


NO: 201
NO: 233


NO: 193
NO: 231




Domain 2: CD80












A12T/H18L/M43V/












F59L/E77K/P109S/












I118T












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


V68M/L70P/L72P/


NO: 195
NO: 231


NO: 189
NO: 231




K86E












Domain 2: CD80












E88D/K89R/D90K/












A91G/F92Y/K93R












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


R29V/Y31F/K36G/


NO: 194
NO: 231


NO: 189
NO: 231




M38L/M43Q/E81R












N83I/L85I/K89R/












D90L/A91E/F92N/












K93Q/R94G












Domain 2: CD80












E88D/K89R/D90K/












A91G/F92Y/K93R












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


V68M/L70P/L72P/


NO: 195
NO: 231


NO: 193
NO: 231




K86E












Domain 2: CD80












A12T/H18L/M43V/












F59L/E77K/P109S/












I118T












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


R29V/Y31F/K36G/


NO: 194
NO: 231


NO: 193
NO: 231




M38L/M43Q/E81R












N83I/L85I/K89R/












D90L/A91E/F92N/












K93Q/R94G












Domain 2: CD80












A12T/H18L/M43V/












F59L/E77K/P109S/












I118T












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


E88D/K89R/D90K/


NO: 189
NO: 231


NO: 195
NO: 231




A91G/F92Y/K93R












Domain 2: CD80












V68M/L70P/L72P/












K86E












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


E88D/K89R/D90K/


NO: 189
NO: 231


NO: 194
NO: 231




A91G/F92Y/K93R












Domain 2: CD80












R29V/Y31F/K36G/












M38L/M43Q/E81R












N83I/L85I/K89R/












D90L/A91E/F92N/












K93Q/R94G












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


A12T/H18L/M43V/


NO: 193
NO: 231


NO: 195
NO: 231




F59L/E77K/P109S/












I118T












Domain 2: CD80












V68M/L70P/L72P/












K86E












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


A12T/H18L/M43V/


NO: 193
NO: 231


NO: 194
NO: 231




F59L/E77K/P109S/












I118T












Domain 2: CD80












R29V/Y31F/K36G/












M38L/M43Q/E81R/












V83I/L85I/K89R/












D90L/A91E/F92N/












K93Q/R94G












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


WT

NO: 236
NO: 220
NO: 237


NO: 196
NO: 233




Domain 2: ICOSL












WT












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


R29H/Y31H/T41G/


NO: 192
NO: 231


NO: 213
NO: 233




Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 2: ICOSL












N525/N57Y/H94D/












L96F/L98F/Q100R/












G103E/F120S












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


I67T/L70Q/A91G/


NO: 175
NO: 231


NO: 213
NO: 233




T120S












Domain 2: ICOSL












N52S/N57Y/H94D/












L96F/L98F/Q100R/












G103E/F120S












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


R29H/Y31H/T41G/


NO: 192
NO: 231


NO: 199
NO: 233




Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 2: ICOSL












N52D












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


I67T/L70Q/A91G/


NO: 175
NO: 231


NO: 199
NO: 233




T120S












Domain 2: ICOSL












N52D












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


R29H/Y31H/T41G/


NO: 192
NO: 231


NO: 201
NO: 233




Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 2: ICOSL












N52H/N57Y/Q100P












Domain 1: CD80
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


I67T/L70Q/A91G/


NO: 175
NO: 231


NO: 201
NO: 233




T120S












Domain 2: ICOSL












N52H/N57Y/Q100P












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID


SEQ ID
SEQ ID




Q35H/H90L/Q102H

NO: 236
NO: 221
NO: 237
+

NO: 213
NO: 233
+
+


Domain 2: ICOSL












N52S/N57Y/H94D/












L96F/L98F/Q100R/












G103E/F120S












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


Q35H/H90L/Q102H

NO: 236
NO: 221
NO: 237


NO: 199
NO: 233




Domain 2: ICOSL












N52D












Domain 1: CD86
+
SEQ ID
SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


Q35H/H90L/Q102H

NO: 236
NO: 221
NO: 237


NO: 201
NO: 233




Domain 2: ICOSL












N52H/N57Y/Q100P












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


WT


NO: 196
NO: 233

NO: 236
NO: 220
NO: 237




Domain 2: CD86












WT












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52S/N57Y/H94D/


NO: 213
NO: 233


NO: 192
NO: 231




L96F/L98F/Q100R/












G103E/F120S












Domain 2: CD80












R29H/Y31H/T41G/












Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52S/N57Y/H94D/


NO: 213
NO: 233


NO: 175
NO: 231




L96F/L98F/Q100R/












G103E/F120S












Domain 2: CD80












I67T/L70Q/A91G/












T120S












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52D


NO: 199
NO: 233


NO: 192
NO: 231




Domain 2: CD80












R29H/Y31H/T41G/












Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52D


NO: 199
NO: 233


NO: 175
NO: 231




Domain 2: CD80












I67T/L70Q/A91G/












T120S












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+

SEQ ID
SEQ ID
+
+


N52H/N57Y/Q100P


NO: 201
NO: 233


NO: 192
NO: 231




Domain 2: CD80












R29H/Y31H/T41G/












Y87N/E88G/K89E/












D90N/A91G/P109S












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


N52S/N57Y/H94D/


NO: 213
NO: 233

NO: 236
NO: 221
NO: 237




L96F/L98F/Q100R/












G103E/F120S












Domain 2: CD86












Q35H/H90L/Q102H












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


N52D


NO: 199
NO: 233

NO: 236
NO: 221
NO: 237




Domain 2: CD86












Q35H/H90L/Q102H












Domain 1: ICOSL
+

SEQ ID
SEQ ID
+
SEQ ID
SEQ ID
SEQ ID
+
+


N52H/N57Y/Q100P


NO: 201
NO: 233

NO: 236
NO: 221
NO: 237




Domain 2: CD86












Q35H/H90L/Q102H









High throughput expression and purification of the variant IgV-stacked-Fc fusion molecules containing various combinations of variant IgV domains from CD80, CD86, ICOSL or NKp30 containing at least one affinity-modified IgV domain were generated as described in Example 5. Binding of the variant IgV-stacked-Fc fusion molecules to respective cognate binding partners and functional activity by anti-CD3 coimmobilization assay also were assessed as described in Example 6. For example, costimulatory bioactivy of the stacked IgSF Fc fusion proteins was determined in a similar immobilized anti-CD3 assay as above. In this case, 4 nM of anti-CD3 (OKT3, Biolegend, USA) was coimmobilized with 4 nM to 120 nM of human rB7-H6.Fc (R&D Systems, USA) or human rPD-L1.Fc (R&D Systems, USA) overnight on tissue-culture treated 96 well plates (Corning, USA). The following day unbound protein was washed off with PBS and 100,000 purified pan T cells were added to each well in 100u1 Ex-Vivo 15 media (Lonza, Switzerland). The stacked IgSF domains were subsequently added at concentrations ranging from 8 nM to 40 nM in a volume of 100u1 for 200u1 volume total. Cells were cultured 3 days before harvesting culture supernatants and measuring human IFN-gamma levels with Duoset ELISA kit (R&D Systems, USA) as mentioned above.


The results are set forth in Tables 16-20. Specifically, Table 16 sets forth binding and functional activity results for variant IgV-stacked-Fc fusion molecules containing an NKp30 IgV domain and an ICOSL IgV domain. Table 17 sets forth binding and functional activity results for variant IgV-stacked-Fv fusion molecules containing an NKp30 IgV domain and a CD80 or CD86 IgV domain. Table 18 sets forth binding and functional activity results for variant IgV-stacked-Fc fusion molecules containing a variant CD80 IgV domain and a CD80, CD86 or ICOSL IgV domain. Table 19 sets forth binding and functional activity results for variant IgV-stacked-Fc fusion molecules containing two variant CD80 IgV domains. Table 20 sets forth results for variant IgV-stacked Fc fusion molecules containing a variant CD80 or CD86 IgV domain and a variant ICOSL IgV domain.


For each of Tables 16-20, Column 1 indicates the structural organization and orientation of the stacked, affinity modified or wild-type (WT) domains beginning with the amino terminal (N terminal) domain, followed by the middle WT or affinity modified domain located before the C terminal human IgG1 Fc domains. Column 2 sets forth the SEQ ID NO identifier for the sequence of each IgV domain contained in a respective “stack” molecule. Column 3 shows the binding partners which the indicated affinity modified stacked domains from column 1 were selected against.


Also shown is the binding activity as measured by the Mean Fluorescence Intensity (MFI) value for binding of each stack molecule to cells engineered to express various cognate binding partners and the ratio of the MFI compared to the binding of the corresponding stack molecule containing unmodified IgV domains not containing the amino acid substitution(s) to the same cell-expressed cognate binding partner. The functional activity of the variant stack molecules to modulate the activity of T cells also is shown based on the calculated levels of IFN-gamma in culture supernatants (pg/mL) generated with the indicated variant stack molecule in solution and the appropriate ligand coimmoblized with anti-CD3 as described in Example 6. The Tables also depict the ratio of IFN-gamma produced by each variant stack molecule compared to the corresponding unmodified stack molecule in the coimmobilization assay.


As shown, the results showed that it was possible to generate stack molecules containing at least one variant IgSF domains that exhibited affinity-modified activity of increased binding for at least one cognate binding partner compared to a corresponding stack molecule containing the respective unmodified (e.g. wild-type) IgV domain. In some cases, the stack molecule, either from one or a combination of both variant IgSF domains in the molecule, exhibited increased binding for more than one cognate binding partner. The results also showed that the order of the IgV domains in the stacked molecules could, in some cases, alter the degree of improved binding activity. In some cases, functional T cell activity also was altered when assessed in the targeted coimmobilization assay.









TABLE 16







Stacked variant IgV Fc fusion proteins containing an NKp30 IgV domain and an ICOSL IgV domain



















Anti-CD3








coimmobilization













cognate
Binding Activity
assay














SEQ
binding
B7H6 MFI
ICOS MFI
CD28 MFI
pg/mL


Domain Structure
ID
partner
(WT
(WT
(WT
IFN-gamma


N terminal to C terminal:
NO
selected
parental
parental
parental
(WT parental


domain 1/domain 2/Fc
(IgV)
against
MFI ratio)
MFI ratio)
MFI ratio)
IFN-gamma ratio)
















Domain 1: NKp30 WT
214

64538
26235
6337
235


Domain 2: ICOSL WT
196

(1.00)
(1.00)
(1.00)
(1.00)


Domain 1: NKp30 (L30V
215
B7-H6
59684
12762
9775
214


A60V S64P S86G)


(0.92)
(0.49)
(1.54)
(0.91)


Domain 2: ICOSL (N52S
212
ICOS-






N57Y H94D L96F L98F

CD28






Q100R)








Domain 1: NKp30 (L30V
215
B7-H6
65470
30272
9505
219


A60V S64P S86G)


(1.01)
(1.15)
(1.50)
(0.93)


Domain 2: ICOSL (N52D)
199
ICOS-








CD28






Domain 1: NKp30 (L30V
215
B7-H6
38153
27903
11300
189


A60V S64P S86G)/


(0.59)
(1.06)
(1.78)
(0.80)


Domain 2: ICOSL (N52H
201
ICOS-






N57Y Q100P)

CD28






Domain 1: ICOSL WT
196

117853
70320
7916
231


Domain 2: Nkp30 WT
214

(1.0)
(1.0)
(1.0)
(1.0)


Domain 1: ICOSL (N52D)
199
ICOS-
100396
83912
20778
228




CD28
(0.85)
(1.19)
(2.62)
(0.98)


Domain 2: NKp30 (L30V
215
B7-H6






A60V S64P S86G)








Domain 1: ICOSL (N52H
201
ICOS-
82792
68874
72269
561


N57Y Q100P)

CD28
(0.70)
(0.98)
(9.12)
(2.43)


Domain 2: NKp30 (L30V
215
B7-H6






A60V S64P S86G)






















TABLE 17







Stacked variant IgV Fc fusion proteins containing an NKp30 IgV domain and a CD80 or CD86 IgV


domain













Anti-CD3





coimmobilization



cognate

assay



binding
Binding Activity
pg/mL IFN-












Domain Structure
SEQ ID
partner
B7H6 MFI
CD28 MFI
gamma


N terminal to C terminal:
NO
selected
(WT parental
(WT parental
(WT parental IFN-


domain 1/domain 2/Fc
(IgV)
against
MFI ratio)
MFI ratio)
gamma ratio)















Domain 1: NKp30 WT
214

88823
7022
68


Domain 2: CD80 WT
152

(1.00)
(1.00)
(1.00)


Domain 1: NKp30 WT
214

14052
1690
92


Domain 2: CD86 WT
220

(1.00)
(1.00)
(1.00)


Domain 1: NKp30 (L30V
215
B7-H6
53279
9027
94


A60V S64P S86G)


(0.60)
(1.29)
(1.38)


Domain 2: CD80
192
CD28


R29H/Y31H/T41G/Y87N/E88G/


K89E/D90N/A91G/P109S


Domain 1: NKp30 (L30V
215
B7-H6
41370
11240
60


(A60V/S64P/S86G)


(0.47)
(1.60)
(0.88)


Domain 2: CD80
175
CD28


(I67T/L70Q/A91G/T120S)


Domain 1: NKp30 (L30V
215
B7-H6
68480
9115
110


(A60V/S64P/S86G)


(4.87)
(5.39)
(1.19)


Domain 2: CD86
221
CD28


(Q35H/H90L/Q102H)


Domain 1: CD80 WT
152

110461
13654
288





(1.00)
(1.00)
(1.00)


Domain 2: Nkp30 WT
214


Domain 1: CD86 WT
220
CD28
128899
26467
213





(1.00)
(1.00)
(1.00)


Domain 2: Nkp30 WT
214
B7-H6


Domain 1: CD80
192
CD28
55727
4342
100


(R29H/Y31H/T41G/Y87N/E88G/


(0.50)
(0.32)
(0.35)


K89E/D90N/A91G/P109S)


Domain 2: NKp30
215
B7-H6


(L30V/A60V/S64P.S86G)


Domain 1: CD80
175
CD28
40412
7094
84


(I67T/L70Q/A91G/T120S)


(0.37)
(0.52)
(0.29)


Domain 2: NKp30
215
B7-H6


(L30V/A60V/S64P/S86G)


Domain 1: CD86
221
CD28
220836
11590
113


(Q35H/H90L/Q102H)


(1.71)
(0.44)
(0.53)


Domain 2: NKp30
215
B7-H6


(L30V/A60V/S64P/S86G)
















TABLE 18







Stacked variant IgV Fc fusion proteins containing a CD80 IgV domain and a CD80, CD86, or


ICOSL IgV domain















Anti-CD3





Binding Activity
coimmobilization















cognate

PD-L1

assay



SEQ
binding
CD28 MFI
MFI
ICOS MFI
pg/mL


Domain Structure
ID
partner
(WT
(WT
(WT
IFN-gamma


N terminal to C terminal:
NO
selected
parental
parental
parental
(WT parental


domain 1/domain 2/Fc
(IgV)
against
MFI ratio)
MFI ratio)
MFI ratio)
IFN-gamma ratio)
















Domain 1: CD80 WT
152

1230
2657
11122
69


Domain 2: ICOSL WT
196

(1.00)
(1.00)
(1.00)
(1.00)


Domain 1: CD80 WT
152

60278
2085

59


Domain 2: CD86 WT
220

(1.00)
(1.00)

(1.00)


Domain 1: CD80 WT
152

1634
6297

98


Domain 2: CD80 WT
152

(1.00)
(1.00)

(1.00)


Domain 1: CD80
189
PD-L1
4308
4234

214


E88D/K89R/D90K/A91G/


(2.64)
(0.67)

(2.18)


F92Y/K93R








Domain 2: CD80
192
CD28






R29H/Y31H/T41G/Y87N/








E88G/K89E/D90N/A91G/








P109S








Domain 1: CD80
193
PD-L1
7613
2030

137


A12T/H18L/M43V/F59L/


(4.66)
(0.32)

(1.40)


E77K/P109S/I118T








Domain 2: CD80
192
CD28






R29H/Y31H/T41G/Y87N/








E88G/K89E/D90N/A91G/








P109S








Domain 1: CD80
193
PD-L1
3851
3657

81


A12T/H18L/M43V/F59L/


(2.36)
(0.58)

(0.83)


E77K/P109S/I118T








Domain 2: CD80
175
CD28






I67T/L70Q/A91G/T120S








Domain 1: CD80
189
PD-L1
4117
2914

96


E88D/K89R/D90K/A91G/


(0.07)
(1.40)

(1.63)


F92Y/K93R








Domain 2: CD86
221
CD28






Q35H/H90L/Q102H








Domain 1: CD80
193
PD-L1
2868
3611

94


A12T/H18L/M43V/F59L/


(0.05)
(1.73)

(1.60)


E77K/P109S/I118T








Domain 2: CD86
221
CD28






Q35H/H90L/Q102H








Domain 1: CD80
189
PD-L1
3383
4515
5158
90


E88D/K89R/D90K/A91G/


(2.75)
(1.70)
(0.46)
(1.30)


F92Y/K93R








Domain 2: ICOSL
213
ICOS/






N52S/N57Y/H94D/L96F/

CD28






L98F/Q100R/G103E/








F120S








Domain 1: CD80
193
PD-L1
2230
2148
3860
112


A12T/H18L/M43V/F59L/


(1.81)
(0.81)
(0.35)
(1.62)


E77K/P109S/I118T








Domain 2: ICOSL
213
ICOS/






N52S/N57Y/H94D/L96F/

CD28






L98F/Q100R/G103E/








F120S








Domain 1: CD80
193
PD-L1
5665
6446
15730
126


A12T/H18L/M43V/F59L/

ICOS/
(4.61)
(2.43)
(1.41)
(1.83)


E77K/P109S/I118T

CD28






Domain 2: ICOSL
199







N52D








Domain 1: CD80
189
PD-L1
6260
4543
11995
269


E88D/K89R/D90K/A91G/


(5.09)
(1.71)
(1.08)
(3.90)


F92Y/K93R








Domain 2: ICOSL
201
ICOS/






N52H/N57Y/Q100P

CD28






Domain 1: CD80
193
PD-L1
3359
3874
8541
97


A12T/H18L/M43V/F59L/


(2.73)
(1.46)
(0.77)
(1.41)


E77K/P109S/I118T








Domain 2: ICOSL
201
ICOS/






N52H/N57Y/Q100P

CD28






Domain 1: ICOSL WT
196

3000
2966
14366
101


Domain 2: CD80 WT
152

(1.00)
(1.00)
(1.00)
(1.00)


Domain 1: CD86 WT
220

4946
1517

125


Domain 2: CD80 WT
152

(1.00)
(1.00)

(1.00)


Domain 1: CD80
192
CD28
2832
3672

114


R29H/Y31H/T41G/Y87N/


(1.73)
(0.58)

(1.16)


E88G/K89E/D90N/A91G/








P109S








Domain 2: CD80
189
PD-L1






E88D/K89R/D90K/A91G/








F92Y/K93R








Domain 1: CD80
192
CD28
4542
2878

142


R29H/Y31H/T41G/Y87N/


(2.78)
(0.45)

(1.45)


E88G/K89E/D90N/A91G/








P109S








Domain 2: CD80
193
PD-L1






A12T/H18L/M43V/F59L/








E77K/P109S/I118T








Domain 1: CD80
175
CD28
938
995

102


I67T/L70Q/A91G/T120S


(0.57)
(0.16)

(1.04)


Domain 2: CD80
189
PD-L1






E88D/K89R/D90K/A91G/








F92Y/K93R








Domain 1: CD80
175
CD28
4153
2827

108


I67T/L70Q/A91G/T120S


(2.54)
(0.45)

(1.10)


Domain 2: CD80
193
PD-L1






A12T/H18L/M43V/F59L/








E77K/P109S/I118T








Domain 1: CD86
221
CD28
14608
2535

257


Q35H/H90L/Q102H


(2.95)
(1.67)

(2.06)


Domain 2: CD80
189
PD-L1






E88D/K89R/D90K/A91G/








F92Y/K93R








Domain 1: CD86
221
CD28
2088
2110

101


Q35H/H90L/Q102H


(0.42)
(1.39)

(0.81)


Domain 2: CD80
193
PD-L1






A12T/H18L/M43V/F59L/








E77K/P109S/I118T








Domain 1: ICOSL
213
ICOS/
3634
4893
6403
123


N52S/N57Y/H94D/L96F/

CD28
(1.21)
(1.65)
(0.45)
(1.22)


L98F/Q100R/G103E/








F120S








Domain 2: CD80
189
PD-L1
1095
5929
7923
127


E88D/K89R/D90K/A91G/


(0.37)
(2.0)
(0.55)
(1.26)


F92Y/K93R








Domain 1: ICOSL
213
ICOS/






N52S/N57Y/H94D/L96F/

CD28






L98F/Q100R/G103E/








F120S








Domain 2: CD80
193
PD-L1






A12T/H18L/M43V/F59L/








E77K/P109S/I118T








Domain 1: ICOSL
199
ICOSL/
2023
5093
16987
125


N52D

CD28
(0.67)
(1.72)
(1.18)
(1.24)


Domain 2: CD80
189
PD-L1






E88D/K89R/D90K/A91G/








F92Y/K93R








Domain 1: ICOSL
199
ICOS/
3441
3414
20889
165


N52D

CD28
(1.15)
(1.15)
(1.45)
(1.63)


Domain 2: CD80
193
PD-L1






A12T/H18L/M43V/F59L/








E77K/P109S/I118T








Domain 1: ICOSL
201
ICOS/
7835
6634
20779
95


N52H/N57Y/Q100P

CD28
(2.61)
(2.24)
(1.45)
(0.94)


Domain 2: CD80
189
PD-L1






E88D/K89R/D90K/A91G/








F92Y/K93R








Domain 1: ICOSL
201
ICOS/
8472
3789
13974
106


N52H/N57Y/Q100P

CD28
(2.82)
(1.28)
(0.97)
(1.05)


Domain 2: CD80
193
PD-L1






A12T/H18L/M43V/F59L/








E77K/P109S/I118T
















TABLE 19







Stacked variant IgV Fc fusion proteins containing two CD80 IgV domains











Cognate





binding
Binding Activity
Functional












Domain Structure
SEQ ID
partner
PD-L1 MFI
CTLA-4 MFI
Activity


N terminal to C terminal:
NO
selected
(WT parental
(WT parental
MLR IFN-gamma


domain 1/domain 2/Fc
(IgV)
against
MFI ratio)
MFI ratio)
pg/mL















Domain 1: CD80 WT
152

6297
4698
35166





(1.00)
(1.00)
(1.00)


Domain 2: CD80 WT
152


Domain 1: CD80
195
CTLA-4
2464
4955
5705


V68M/L70P/L72P/K86E


(0.39)
(1.05)
(0.16)


Domain 2: CD80
189
PD-L1


E88D/K89R/D90K/A91G/F92Y/


K93R


Domain 1: CD80
194
CTLA-4
1928
1992
1560


R29V/Y31F/K36G/M38L/M43Q/


(0.31)
(0.42)
(0.04)


E81R/V83I/L85I/K89R/


D90L/A91E/F92N/K93Q/R94G


Domain 2: CD80
189
PD-L1


E88D/K89R/D90K/A91G/F92Y/


K93R


Domain 1: CD80
195
CTLA-4
1215
1382
2171


V68M/L70P/L72P/K86E


(0.19)
(0.29)
(0.06)


Domain 2: CD80
193
PD-L1


A12T/H18L/M43V/F59L/E77K/


P109S/I118T


Domain 1: CD80
194
CTLA-4
1592
1962
1512


R29V/Y31F/K36G/M38L/M43Q/


(0.25)
(0.42)
(0.04)


E81R/V83I/L851/K89R/


D90L/A91E/F92N/K93Q/R94G


Domain 2: CD80
193
PD-L1


A12T/H18L/M43V/F59L/E77K/


P109S/I118T


Domain 1: CD80
189
PD-L1
1747
2057
9739


E88D/K89R/D90K/A91G/F92Y/


(0.28)
(0.44)
(0.28)


K93R


Domain 2: CD80
195
CTLA-4


V68M/L70P/L72P/K86E


Domain 1: CD80
189
PD-L1
1752
1772
5412


E88D/K89R/D90K/A91G/F92Y/


(0.28)
(0.38)
(0.15)


K93R


Domain 2: CD80
194
CTLA-4


R29V/Y31F/K36G/M38L/M43Q/


E81R/V83I/L85I/K89R/


D90L/A91E/F92N/K93Q/R94G


Domain 1: CD80
193
PD-L1
1636
1887
7608


A12T/H18L/M43V/F59L/E77K/


(0.26)
(0.40)
(0.22)


P109S/I118T


Domain 2: CD80
195
CTLA-4


V68M/L70P/L72P/K86E


Domain 1: CD80
193
PD-L1
2037
4822
11158


A12T/H18L/M43V/F59L/E77K/


(0.32)
(1.03)
(0.32)


P109S/I118T


Domain 2: CD80
194
CTLA-4


R29V/Y31F/K36G/M38L/M43Q/


E81R/V83I/


L85I/K89R/D90L/A91E/F92N/


K93Q/R94G
















TABLE 20







Stacked variant IgV Fc fusion proteins containing a CD80 or CD86 IgV domain and an ICOSL IgV


domain











Cognate





binding
Binding Activity
Functional












Domain Structure
SEQ ID
partner
PD-L1 MFI
CTLA-4 MFI
Activity


N terminal to C terminal:
NO
selected
(WT parental
(WT parental
MLR IFN-gamma


domain 1/domain 2/Fc
(IgV)
against
MFI ratio)
MFI ratio)
pg/mL















Domain 1: CD80 WT
152

1230
11122
1756





(1.00)
(1.00)
(1.00)


Domain 2: ICOSL WT
196


Domain 1: CD86 WT
220

29343
55193
6305


Domain 2: ICOSL WT
196

(1.00)
(1.00)
(1.00)


Domain 1: CD80
192
CD28
2280
3181
2281


R29H/Y31H/T41G/Y87N/E88G/


(1.85)
(0.29)
(1.30)


K89E/D90N/A91G/P109S


Domain 2: ICOSL
213
ICOS/CD28


N52S/N57Y/H94D/L96F/L98F/


Q100R/G103E/


F120S


Domain 1: CD80
175
CD28
2309
26982
1561


I67T/L70Q/A91G/T120S


(1.88)
(2.43)
(0.89)


Domain 2: ICOSL
213
ICOS/CD28


N52S/N57Y/H94D/L96F/L98F/


Q100R/G103E/


F120S


Domain 1: CD80
192
CD28
4285
22744
1612


R29H/Y31H/T41G/Y87N/E88G/


(3.48)
(2.04)
(0.92)


K89E/D90N/A91G/P109S


Domain 2: ICOSL
199
ICOS/CD28


N52D


Domain 1: CD80
175
CD28
3024
16916
3857


I67T/L70Q/A91G/T120S


(2.46)
(1.52)
(2.20)


Domain 2: ICOSL
199
ICOS/CD28


N52D


Domain 1: CD80
192
CD28
6503
7240
6886


R29H/Y31H/T41G/Y87N/E88G/


(5.29)
(0.65)
(3.92)


K89E/D90N/A91G/P109S


Domain 2: ICOSL
201
ICOS/CD28


N52H/N57Y/Q100P


Domain 1: CD80
175
CD28
3110
4848
3393


I67T/L70Q/A91G/T120S


(2.53)
(0.44)
(1.93)


Domain 2: ICOSL
201
ICOS/CD28


N52H/N57Y/Q100P


Domain 1: CD86
221
CD28
11662
21165
880


Q35H/H9OL/Q102H


(0.40)
(0.38)
(0.14)


Domain 2: ICOSL
213
ICOS/CD28


N52S/N57Y/H94D/L96F/L98F/


Q100R/G103E/


F120S


Domain 1: CD86
221
CD28
24230
73287
1110


Q35H/H9OL/Q102H


(0.83)
(1.33)
(0.18)


Domain 2: ICOSL
199
ICOS/CD28


N52D


Domain 1: CD86
221
CD28
1962
1630
587


Q35H/H90L/Q102H


(0.07)
(0.03)
(0.09)


Domain 2: ICOSL
201
ICOS/CD28


N52H/N57Y/Q100P


Domain 1: ICOSL WT
196

3000
14366
4113


Domain 2: CD80 WT
152

(1.00)
(1.00)
(1.00)


Domain 1: ICOSL WT
196

18005
53602
18393


Domain 2: CD86 WT
220

(1.00)
(1.00)
(1.00)


Domain 1: ICOSL
213
ICOSL/CD28
10426
51286
18680


N52S/N57Y/H94D/L96F/L98F/


(3.48)
(3.57)
(4.54)


Q100R/G103E/


F120S


Domain 2: CD80
192
CD28


R29H/Y31H/T41G/Y87N/E88G/


K89E/D90N/A91G/P109S


Domain 1: ICOSL
213
ICOS/CD28
17751
29790
10637


N52S/N57Y/H94D/L96F/L98F/


(5.92)
(2.07)
(2.59)


Q100R/G103E/


F120S


Domain 2: CD80
175
CD28


I67T/L70Q/A91G/T120S


Domain 1: ICOSL
199
ICOS/CD28
2788
25870
6205


N52D


(0.93)
(1.80)
(1.51)


Domain 2: CD80
192
CD28


R29H/Y31H/T41G/Y87N/E88G/


K89E/D90N/A91G/P109S


Domain 1: ICOSL
199
ICOS/CD28
2522
13569
5447


N52D


(0.84)
(0.94)
(1.32)


Domain 2: CD80
175
CD28


I67T/L70Q/A91G/T120S


Domain 1: ICOSL
201
ICOS/CD28
9701
9187
5690


N52H/N57Y/Q100P


(3.23)
(0.64)
(1.38)


Domain 2: CD80
192
CD28


R29H/Y31H/T41G/Y87N/E88G/


K89E/D90N/A91G/P109S


Domain 1: ICOSL
213
ICOS/CD28
27050
21257
8131


N52S/N57Y/H94D/L96F/L98F/


(1.50)
(0.40)
(0.44)


Q100R/G103E/


F120S


Domain 2: CD86
221
CD28


Q35H/H90L/Q102H


Domain 1: ICOSL
199
ICOS/CD28
34803
80210
6747


N52D


(1.93)
(1.50)
(0.37)


Domain 2: CD86
221
CD28


Q35H/H90L/Q102H


Domain 1: ICOSL
201
ICOS/CD28
5948
4268
26219


N52H/N57Y/Q100P


(0.33)
(0.08)
(1.43)


Domain 2: CD86
221
CD28


Q35H/H90L/Q102H









Example 9
Generation, Selection and Screening of Affinity-Modified IgSF Domain Variants of CD155

Affinity-modified IgSF domain variants of CD155 were generated substantially as described in Examples 1-6 above with some slight modifications. For example, for the generation of CD155 variants, only the IgV domain, and not the other two domains of the ECD, was included in the generated proteins. The example exemplifies binding and activity of the exemplary affinity-modified domains in an Fc-fusion format; such affinity-modified domains are contemplated in connection with a secretable immunomodulatory protein or transmembrane immunomodulatory protein as described.


To generate a library targeting specific residues of CD155 by complete or partial randomization with degenerate codons, the coding DNA for the immunoglobulin-like V-type (IgV) domain of human CD155 (SEQ ID NO:241) was ordered from Integrated DNA Technologies (Coralville, Iowa) as a set of overlapping oligonucleotides of up to 80 base pairs (bp) in length. In general, to generate a library of diverse variants of the IgV domain, the oligonucleotides contained desired degenerate codons at desired amino acid positions. Degenerate codons were generated using an algorithm at the URL: rosettadesign.med.unc.edu/SwiftLib/. In general, positions to mutate and degenerate codons were chosen from crystal structure information (PDB: 3UDW) or homology models built from this structure containing the target-ligand pairs of interest to identify ligand contact residues as well as residues that are at the protein interaction interface. For example, a crystal structure of CD155 bound to TIGIT is publicly available at the URL: www.rcsb.org/pdb/explore/explore.do?structureId=3UDW for Protein Data Base code 3UDW. This analysis was performed using a structure viewer available at the URL: spdbv.vital-it.ch).


The oligonucleotides were used for PCR amplification to generate library DNA inserts for insertion into the modified yeast display version of vector pBYDS03 substantially as described in Example 1. Alternatively, Ultramers (Integrated DNA Technologies) of up to 200 bp in length were used in conjunction with megaprimer PCR (URL: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC146891/pdf/253371.pdf) to generate larger stretches of degenerate codons that could not be as easily incorporated using multiple small overlapping primers. Following the generation of full length product using megaprimer PCR, the mutant IgV domain library was PCR amplified again using DNA primers containing 40 bp overlap region with the modified pBYDS03 cloning variant for homologous recombination into yeast. The library of DNA inserts were prepared for library insertion substantially as descried in Example 1 and electroporation-ready DNA was prepared.


As alternative approaches, either sublibraries generated by site-directed mutagenesis to target specific residues of the IgV domain of CD155 or random libraries of the IgV domain of CD155 were generated to further identify variants of the IgV domain of CD155 substantially as described in Example 1.


The degenerate or random library DNA was inserted into yeast substantially as described in Example 2. Yeast expressing affinity modified variants of CD155 were selected using the method substantially described in Example 3. For the selection, the following target ligand proteins were employed: human rTIGIT.Fc (i.e., recombinant TIGIT-Fc fusion protein) and rCD226.Fc. Magnetic Protein A beads were obtained from New England Biolabs, USA. For two-color, flow cytometric sorting, a Bio-Rad S3e sorter was used. CD155 display levels were monitored with an anti-hemagglutinin antibody labeled with Alexafluor 488 (Life Technologies, USA). Ligand binding of Fc fusion proteins, rTIGIT.Fc or rCD226.Fc, were detected with PE conjugated human Ig specific goat Fab (Jackson ImmunoResearch, USA). Doublet yeast were gated out using forward scatter (FSC)/side scatter (SSC) parameters, and sort gates were based upon higher ligand binding detected in FL2 that possessed more limited tag expression binding in FL1.


Second sort outputs (F2) were obtained substantially as described in Example 3 by expanding and re-inducing expression of sort outputs that had been assayed for higher specific binding affinity and were used to assess binding compared to the parental, wild-type yeast strain. For CD155, the second FACS outputs (F2) were compared to parental CD155 yeast for binding rTIGIT.Fc or rCD226.Fc by double staining each population with anti-HA (hemagglutinin) tag expression and the anti-human Fc secondary to detect ligand binding.


Selected outputs were reformatted as immunomodulatory proteins containing an affinity modified (variant) immunoglobulin-like V-type (IgV) domain of CD155 fused to an Fc molecule (variant IgV domain-Fc fusion molecules) substantially as described in Example 4, except including only the IgV domain and not the full ECD domain. In some alternative methods for the CD155 outputs, DNA from the outputs were PCR amplified with primers containing 40 bp overlap regions on either end with an Fc fusion vector to carry out in vitro recombination using Gibson Assembly Mastermix (New England Biolabs), which was subsequently used in heat shock transformation into E. Coli strain NEB5alpha. Exemplary of an Fc fusion vector is pFUSE-hIgG1-Fc2 (Invivogen, USA).


After transformation, samples were prepared for DNA sequencing substantially as described in Example 4. The sequences were then manually curated as described in Example 4, except that they were manually curated so that they start at the beginning of the IgV coding region. The curated sequences were batch-translated and aligned as described in Example 4. Clones of interest were identified using the criteria as described in Example 4. Table 21 sets forth the identified variant CD155 affinity-modified molecules, including the amino acid substitutions contained in each variant.


The methods generated immunomodulatory proteins containing an affinity-modified IgV domain of CD155 in which the encoding DNA was generated to encode a protein as follows: signal peptide followed by variant (mutant) IgV domain followed by a linker of three alanines (AAA) followed by a human IgG1 Fc containing the mutation N297G (N82G with reference to wild-type human IgG1 Fc set forth in SEQ ID NO: 226). The human IgG1 Fc also contained the mutations R292C and V302C by EU numbering (corresponding to R77C and V87C with reference to wild-type human IgG1 Fc set forth in SEQ ID NO: 226). Since the construct does not include any antibody light chains that can form a covalent bond with a cysteine, the human IgG1 Fc also contained replacement of the cysteine residues to a serine residue at position 220 (C220S) by EU numbering (corresponding to position 5 (C5S) with reference to the wild-type or unmodified Fc set forth in SEQ ID NO: 226.


Recombinant variant CD155 Fc fusion proteins were expressed and purified substantially as described in Example 5. Binding and activity of the affinity-modified variant CD155 Fc fusion proteins was assessed substantially as described in Example 6, except that cells expressing full length human CD226 and TIGIT cognate binding partners were generated. Cells were stained by flow cytometry with CD155 Fc variant and mean fluorescence intensity (MFI) was determined as described in Example 5. For bioactivity characterization, recombinant CD155 Fc variants were assessed in an anti-CD3 coimmobilization assay substantially as described in Example 5.


The results for the binding and activity studies are set forth in Table 21. The Table indicates exemplary IgSF domain amino acid substitutions (replacements) in the IgV domain of CD155 selected in the screen for affinity-maturation against the respective cognate structures TIGIT and CD226. The exemplary amino acid substitutions are designated by amino acid position number corresponding to position of the unmodified sequence set forth in SEQ ID NO: 241 (IgV). The amino acid position is indicated in the middle, with the corresponding unmodified (e.g. wild-type) amino acid listed before the number and the identified variant amino acid substitution listed after the number. Column 2 sets forth the SEQ ID NO identifier for the variant for each variant ECD-Fc fusion molecule.


Also shown is the binding activity as measured by the Mean Fluorescence Intensity (MFI) value for binding of each variant Fc-fusion molecule to cells engineered to express the cognate binding partner as the ratio of the MFI compared to the binding of the corresponding unmodified ECD-Fc fusion molecule not containing the amino acid substitution(s) to the same cell-expressed cognate binding partner. The functional activity of the variant Fc-fusion molecules to modulate the activity of T cells also is shown based on the calculated levels of IFN-gamma in culture supernatants (pg/mL) generated with the indicated variant Fc fusion molecule coimmoblized with anti-CD3 as a ratio of IFN-gamma produced by each variant CD155 IgV-Fc compared to the corresponding unmodified CD155 IgV-Fc in both functional assays.









TABLE 21







CD155 variants selected against cognate binding partners. Molecule sequences,


binding data, and costimulatory bioactivity data.
















TIGIT

Mock
Anti-CD3




CD226
tfxn
CD96
Expi293
IFN-gamma




tfxn MFI
MFI
MFI
MFI
(pg/mL)




(CD226
(TIGIT
(CD96
(Mock
(Anti-CD3



SEQ
MFI
MFI
MFI
MFI
IFN-gamma



ID NO
parental
parental
parental
parental
parental


CD155 mutations
(IgV)
ratio)
ratio)
ratio)
ratio)
ratio)
















P18S, P64S, F91S
653
497825
247219
140065
3528
270.1




(133.7)
(91.1)
(45.4)
(1.2)
(0.7)


P18S, F91S, L104P
654
26210
75176
10867
2130
364.2




(7.0)
(27.7)
(3.5)
(0.7)
(0.9)


L44P
655
581289
261931
152252
3414
277.6




(156.1)
(96.5)
(49.4)
(1.2)
(0.7)


A56V
656
455297
280265
161162
2601
548.2




(122.3)
(103.2)
(52.2)
(0.9)
(1.4)


P18L, L79V, F91S
657
5135
4073
3279
2719
1241.5




(1.4)
(1.5)
(1.1)
(0.9)
(3.2)


P18S, F91S
658
408623
284190
147463
3348
760.6




(109.8)
(104.7)
(47.8)
(1.1)
(2.0)


P18T, F91S
659
401283
223985
157644
3065
814.7




(107.8)
(82.5)
(51.1)
(1.1)
(2.1)


P18T, S42P, F91S
660
554105
223887
135395
3796
539.7




(148.8)
(82.5)
(43.9)
(1.3)
(1.4)


G7E, Pl8T, Y30C, F91S
661
12903
12984
7906
2671
275.9




(3.5)
(4.8)
(2.6)
(0.9)
(0.7)


P18T, F91S, G111D
662
438327
287315
167583
4012
307.2




(117.7)
(105.8)
(54.3)
(1.4)
(0.8)


P18S, F91P
663
4154
3220
2678
2816
365.7




(1.1)
(1.2)
(0.9)
(1.0)
(0.9)


P18T, F91S, F108L
664
394546
298680
193122
2926
775.4




(106.0)
(110.0)
(62.6)
(1.0)
(2.0)


P18T, T45A, F91S
665
435847
222044
191026
2948
1546.8




(117.1)
(81.8)
(61.9)
(1.0)
(4.0)


P18T, F91S, R94H
666
3589
2942
2509
2390
1273.2




(1.0)
(1.1)
(0.8)
(0.8)
(3.3)


P18S, Y30C, F91S
667
382352
276358
56934
3540
426.5




(102.7)
(101.8)
(18.5)
(1.2)
(1.1)


A81V, L83P
668
4169
2912
2616
2993
339.7




(1.1)
(1.1)
(0.8)
(1.0)
(0.9)


L88P
669
65120
74845
35280
2140
969.2




(17.5)
(27.6)
(11.4)
(0.7)
(2.5)


Wild type
652
3723
2715
3085
2913
389.6




(1.0)
(1.0)
(1.0)
(1.0)
(1.0)


R94H
670
18905
104013
11727
1663
372.6




(5.1)
(38.3)
(3.8)
(0.6)
(1.0)


A13E, P18S, A56V,
671
357808
179060
118570
2844
349.2


F91S

(96.1)
(66.0)
(38.4)
(1.0)
(0.9)


P18T, F91S, V115A
672
38487
46313
22718
2070
1574.5




(10.3)
(17.1)
(7.4)
(0.7)
(4.0)


P18T, Q60K
673
238266
173730
154448
4778
427.2




(64.0)
(64.0)
(50.1)
(1.6)
(1.1)









Example 10
Generation, Selection and Screening of Affinity-Modified IgSF Domain Variants of CD112

Affinity-modified IgSF domain variants of CD112 were generated substantially as described in Examples 1-6 above with some slight modifications. For example, for the generation of CD112 variants, only the IgV domain, and not the other two domains of the ECD, was included in the generated proteins. The example exemplifies binding and activity of the exemplary affinity-modified domains in an Fc-fusion format; such affinity-modified domains are contemplated in connection with a secretable immunomodulatory protein or transmembrane immunomodulatory protein as described.


To generate a library targeting specific residues of CD155 by complete or partial randomization with degenerate codons, the coding DNA for the immunoglobulin-like V-type (IgV) domain of human CD112 (SEQ ID NO:286) was ordered from Integrated DNA Technologies (Coralville, Iowa) as a set of overlapping oligonucleotides of up to 80 base pairs (bp) in length. In general, to generate a library of diverse variants of the IgV domain, the oligonucleotides contained desired degenerate codons at desired amino acid positions. Degenerate codons were generated using an algorithm at the URL: rosettadesign.med.unc.edu/SwiftLib/. In general, positions to mutate and degenerate codons were chosen from crystal structure information (PDB: 3UDW) or homology models built from this structure containing the target-ligand pairs of interest to identify ligand contact residues as well as residues that are at the protein interaction interface.


The oligonucleotides were used for PCR amplification to generate library DNA inserts for insertion into the modified yeast display version of vector pBYDS03 substantially as described in Example 1. Alternatively, Ultramers (Integrated DNA Technologies) of up to 200 bp in length were used in conjunction with megaprimer PCR (URL: http://www.ncbi.nlm.nih.gov/pmc/articles/PMC146891/pdf/253371.pdf) to generate larger stretches of degenerate codons that could not be as easily incorporated using multiple small overlapping primers. Following the generation of full length product using megaprimer PCR, the mutant IgV domain library was PCR amplified again using DNA primers containing 40 bp overlap region with the modified pBYDS03 cloning variant for homologous recombination into yeast. The library of DNA inserts were prepared for library insertion substantially as descried in Example 1 and electroporation-ready DNA was prepared.


As alternative approaches, either sublibraries generated by site-directed mutagenesis to target specific residues of the IgV domain of CD112 or random libraries of the IgV domain of CD112 were generated to further identify variants of the IgV domain of CD112 substantially as described in Example 1.


The degenerate or random library DNA was inserted into yeast substantially as described in Example 2. Yeast expressing affinity modified variants of CD112 were selected using the method substantially described in Example 3. For the selection, the following target ligand proteins were employed: human rTIGIT.Fc (i.e., recombinant TIGIT-Fc fusion protein), rCD226.Fc and rCD112R.Fc. Magnetic Protein A beads were obtained from New England Biolabs, USA. For two-color, flow cytometric sorting, a Bio-Rad S3e sorter was used. CD112 display levels were monitored with an anti-hemagglutinin antibody labeled with Alexafluor 488 (Life Technologies, USA). Ligand binding of Fc fusion proteins, rTIGIT.Fc, rCD226.Fc, or rCD112R.Fc, were detected with PE conjugated human Ig specific goat Fab (Jackson ImmunoResearch, USA). Doublet yeast were gated out using forward scatter (FSC)/side scatter (SSC) parameters, and sort gates were based upon higher ligand binding detected in FL2 that possessed more limited tag expression binding in FL1.


Second sort outputs (F2) were obtained substantially as described in Example 3 by expanding and re-inducing expression of sort outputs that had been assayed for higher specific binding affinity and were used to assess binding compared to the parental, wild-type yeast strain. For CD112, the second FACS outputs (F2) were compared to parental CD112 yeast for binding of each rTIGIT.Fc, rCD226.Fc, and rCD112R by double staining each population with anti-HA (hemagglutinin) tag expression and the anti-human Fc secondary to detect ligand binding.


Selected outputs were reformatted as immunomodulatory proteins containing an affinity modified (variant) immunoglobulin-like V-type (IgV) domain of CD112 fused to an Fc molecule (variant IgV domain-Fc fusion molecules) substantially as described in Example 4, except including only the IgV domain and not the full ECD domain. In some alternative methods for the CD112 outputs, DNA from the outputs were PCR amplified with primers containing 40 bp overlap regions on either end with an Fc fusion vector to carry out in vitro recombination using Gibson Assembly Mastermix (New England Biolabs), which was subsequently used in heat shock transformation into E. Coli strain NEB5alpha. Exemplary of an Fc fusion vector is pFUSE-hIgG1-Fc2 (Invivogen, USA).


After transformation, samples were prepared for DNA sequencing substantially as described in Example 4. The sequences were then manually curated as described in Example 4, except that they were manually curated so that they start at the beginning of the IgV coding region. The curated sequences were batch-translated and aligned as described in Example 4. Clones of interest were identified using the criteria as described in Example 4. Table 22 sets forth the identified variant CD112 affinity-modified molecules, including the amino acid substitutions contained in each variant.


The methods generated immunomodulatory proteins containing an affinity-modified IgV domain of CD112 in which the encoding DNA was generated to encode a protein as follows: signal peptide followed by variant (mutant) IgV domain followed by a linker of three alanines (AAA) followed by a human IgG1 Fc containing the mutation N297G (N82G with reference to wild-type human IgG1 Fc set forth in SEQ ID NO: 226). The human IgG1 Fc also contained the mutations R292C and V302C (corresponding to R77C and V87C with reference to wild-type human IgG1 Fc set forth in SEQ ID NO: 226). Since the construct does not include any antibody light chains that can form a covalent bond with a cysteine, the human IgG1 Fc also contained replacement of the cysteine residues to a serine residue at position 5 (C5S) compared to the wild-type or unmodified Fc set forth in SEQ ID NO: 226.


Recombinant variant CD112 Fc fusion proteins were expressed and purified substantially as described in Example 5. Binding and activity of the affinity-modified variant CD112 Fc fusion proteins was assessed substantially as described in Example 6, except that cells expressing full length human CD226, TIGIT and CD112R cognate binding partners were generated. Cells were stained by flow cytometry with CD112 Fc variant and mean fluorescence intensity (MFI) was determined as described in Example 5. For bioactivity characterization, recombinant CD112 Fc variants were assessed in an anti-CD3 coimmobilization assay substantially as described in Example 5.


The results for the binding and activity studies are set forth in Table 22. The Table indicates exemplary IgSF domain amino acid substitutions (replacements) in the IgV domain of CD112 selected in the screen for affinity-maturation against the respective cognate structures TIGIT, CD226 and CD112R. The exemplary amino acid substitutions are designated by amino acid position number corresponding to position of the unmodified sequence set forth in SEQ ID NO: 286. The amino acid position is indicated in the middle, with the corresponding unmodified (e.g. wild-type) amino acid listed before the number and the identified variant amino acid substitution listed after the number. Column 2 sets forth the SEQ ID NO identifier for the variant ECD for each variant IgV-Fc fusion molecule.


Also shown is the binding activity as measured by the Mean Fluorescence Intensity (MFI) value for binding of each variant Fc-fusion molecule to cells engineered to express the cognate binding partner as the ratio of the MFI compared to the binding of the corresponding unmodified IgV-Fc fusion molecule not containing the amino acid substitution(s) to the same cell-expressed cognate binding partner. The functional activity of the variant Fc-fusion molecules to modulate the activity of T cells also is shown based on the calculated levels of IFN-gamma in culture supernatants (pg/ml) generated with the indicated variant Fc fusion molecule coimmoblized with anti-CD3 as a ratio of IFN-gamma produced by each variant CD112 IgV-Fc compared to the corresponding unmodified CD112 IgV-Fc in both functional assays.









TABLE 22







CD112 variants selected against cognate binding partners. Molecule sequences,


binding data, and costimulatory bioactivity data.


















Mock
Anti-CD3




TIGIT
CD112R
CD226
Expi293
IFN-gamma




tfxn MFI
tfxn MFI
MFI
MFI
(pg/mL)




(TIGIT
(CD112R
(CD226
(Mock
(Anti-CD3



SEQ
MFI
MFI
MFI
MFI
IFN-gamma



ID NO
parental
parental
parental
parental
parental


CD112 mutations
(IgV)
ratio)
ratio)
ratio)
ratio)
ratio)
















WTCD112
965
210829
1452
265392
1112
676.6




(1.00)
(1.00)
(1.00)
(1.00)
(1.00)


Y33H, A112V, G117D
966
12948
1552
1368
1241
164.8




(0.06)
(1.07)
(0.01)
(1.12)
(0.24)


V19A, Y33H, S64G, S80G,
967
48356
1709
2831
1098



G98S, N106Y, A112V

(0.23)
(1.18)
(0.01)
(0.99)



L32P, A112V
968
191432
1557
11095
1259
390.4




(0.91)
(1.07)
(0.04)
(1.13)
(0.58)


A95V, A112I
969
238418
1706
51944
1215
282.5




(1.13)
(1.17)
(0.20)
(1.09)
(0.42)


P28S, A112V
970
251116
1985
153382
1189
503.4




(1.19)
(1.37)
(0.58)
(1.07)
(0.74)


P27A, T38N, V101A,
971
255803
2138
222822
1399
240.7


A112V

(1.21)
(1.47)
(0.84)
(1.26)
(0.36)


S118F
972
11356
5857
6938
1270
271.7




(0.05)
(4.03)
(0.03)
(1.14)
(0.40)


R12W, H48Y, F54S, S118F
973
10940
3474
5161
1069





(0.05)
(2.39)
(0.02)
(0.96)



R12W, Q79R, S118F
974
2339
7370
1880
1338
447.4




(0.01)
(5.08)
(0.01)
(1.20)
(0.66)


T113S, S118Y
975
6212
6823
1554
1214
225.1




(0.03)
(4.70)
(0.01)
(1.09)
(0.33)


S118Y
976
2921
6535
2003
1463
190.4




(0.01)
(4.50)
(0.01)
(1.32)
(0.28)


N106I, S118Y
977
2750
7729
1815
1222
265.8




(0.01)
(5.32)
(0.01)
(1.10)
(0.39)


N106I, S118F
978
1841
9944
1529
1308
437.9




(0.01)
(6.85)
(0.01)
(1.18)
(0.65)


A95T, L96P, S118Y
979
2352
4493
1412
1329
292.4




(0.01)
(3.09)
(0.01)
(1.19)
(0.43)


Y33H, P67S, N106Y,
980
225015
3259
204434
1296
618.8


A112V

(1.07)
(2.24)
(0.77)
(1.17)
(0.91)


N106Y, A112V
981
6036
1974
15334
1108
409.9




(0.03)
(1.36)
(0.06)
(1.00)
(0.61)


T18S, Y33H, A112V
982
252647
1347
183181
1412
601.8




(1.20)
(0.93)
(0.69)
(1.27)
(0.89)


P9S, Y33H, N47S, A112V
983
240467
1418
203608
1361
449.1




(1.14)
(0.98)
(0.77)
(1.22)
(0.66)


P42S, P67H, A112V
984
204484
1610
188647
1174
530.6




(0.97)
(1.11)
(0.71)
(1.06)
(0.78)


P27L, L32P, P42S, A112V
985
219883
1963
84319
1900
251.6




(1.04)
(1.35)
(0.32)
(1.71)
(0.37)


G98D, A112V
986
4879
2369
6100
1729
387.0




(0.02)
(1.63)
(0.02)
(1.55)
(0.57)


Y33H, S35P, N106Y,
987
250724
1715
94373
1495
516.2


A112V

(1.19)
(1.18)
(0.36)
(1.34)
(0.76)


L32P, P42S, T100A,
988
242675
1742
202567
1748
435.3


A112V

(1.15)
(1.20)
(0.76)
(1.57)
(0.64)


P27S, P45S, N106I, A112V
989
223557
1799
84836
1574
277.5




(1.06)
(1.24)
(0.32)
(1.42)
(0.41)


Y33H, N47K, A112V
990
251339
1525
199601
1325
483.2




(1.19)
(1.05)
(0.75)
(1.19)
(0.71)


Y33H, N106Y, A112V
991
297169
1782
258315
1440
485.4




(1.41)
(1.23)
(0.97)
(1.30)
(0.72)


K78R, D84G, A112V,
992
236662
1638
24850
1345
142.5


F114S

(1.12)
(1.13)
(0.09)
(1.21)
(0.21)


Y33H, N47K, F54L, A112V
993
14483
1617
2371
1353
352.8




(0.07)
(1.11)
(0.01)
(1.22)
(0.52)


Y33H, A112V
994
98954
1216
1726
1298





(0.47)
(0.84)
(0.01)
(1.17)



A95V, A112V
995
168521
2021
200789
1459
412.9




(0.80)
(1.39)
(0.76)
(1.31)
(0.61)


R12W, A112V
996
135635
1582
23378
1412
165.8




(0.64)
(1.09)
(0.09)
(1.27)
(0.24)


A112V
1002
213576
1986
151900
1409
211.4




(1.01)
(1.37)
(0.57)
(1.27)
(0.31)


Y33H, A112V
994
250667
1628
230578
1216
612.7




(1.19)
(1.12)
(0.87)
(1.09)
(0.91)


R12W, P27S, A112V
997
3653
1308
9105
1051





(0.02)
(0.90)
(0.03)
(0.94)



Y33H, V51M, A112V
998
218698
1384
195450
1170
709.4




(1.04)
(0.95)
(0.74)
(1.05)
(1.05)


Y33H, A112V, S118T
999
219384
1566
192645
1313
396.3




(1.04)
(1.08)
(0.73)
(1.18)
(0.59)


Y33H, V101A, A112V,
1000
5605
1582
5079
1197



P115S

(0.03)
(1.09)
(0.02)
(1.08)



H24R, T38N, D43G,
1001
227095
1537
229311
1336
858.6


A112V

(1.08)
(1.06)
(0.86)
(1.20)
(1.27)


A112V
1002
4056
1356
10365
986





(0.02)
(0.93)
(0.04)
(0.89)



P27A, A112V
1003
193537
1531
230708
3084
355.1




(0.92)
(1.05)
(0.87)
(2.77)
(0.52)


A112V, S118T
1004
233173
1659
121817
845
533.3




(1.11)
(1.14)
(0.46)
(0.76)
(0.79)


R12W, A112V, M122I
1005
235935
1463
217748
1350
528.0




(1.12)
(1.01)
(0.82)
(1.21)
(0.78)


Q83K, N106Y, A112V
1006
205948
2042
234958
1551
481.4




(0.98)
(1.41)
(0.89)
(1.39)
(0.71)


R12W, P27S, A112V,
1007
11985
2667
12756
1257
334.4


S118T

(0.06)
(1.84)
(0.05)
(1.13)
(0.49)


P28S, Y33H, A112V
1008
4711
1412
3968
955





(0.02)
(0.97)
(0.01)
(0.86)



P27S, Q90R, A112V
1009
3295
1338
6755
1048





(0.02)
(0.92)
(0.03)
(0.94)



L15V, P27A, A112V,
1010
209888
1489
84224
1251
512.3


S118T

(1.00)
(1.03)
(0.32)
(1.13)
0.76)









Y33H, N106Y, T108I,
1011
Not tested













A112V















Y33H, P56L, V75M,
1012
Not tested













V101M, A112V















Example 11
Additional Affinity Modified IgSF Domains

This examples describe the design, creation, and screening of additional affinity modified CD80, CD155, CD112, PD-L1, PD-L2, CD86 (B7-2) immunomodulatory proteins, which are other components of the immune synapse (IS) that have a demonstrated dual role in both immune activation and inhibition. Also described are generation and screening of Nkp30 variants. These examples demonstrate that affinity modification of IgSF domains yields proteins that can act to both increase and decrease immunological activity. Various combinations of those domains fused in pairs (i.e., stacked) with a variant affinity modified CD80 to form a Type II immunomodulatory protein to achieve immunomodulatory activity. The example exemplifies binding and activity of the exemplary affinity-modified domains in an Fc-fusion format; such affinity-modified domains are contemplated in connection with a secretable immunomodulatory protein or transmembrane immunomodulatory protein as described.


Mutant DNA constructs of encoding a variant of the IgV domain of human CD80, CD155, CD112, PD-L1, PD-L2, CD86 (B7-2), and NKp30 for translation and expression as yeast display libraries were generated substantially as described in Example 1. For target libraries that target specific residues for complete or partial randomization with degenerate codons and/or random libraries were constructed to identify variants of the IgV of CD80 (SEQ ID NO:578), IgV of CD112 (SEQ ID NO: 965), CD155(SEQ ID NO: 652), PD-L1 (SEQ ID NO:1332), and variants of the IgV of PD-L2 (SEQ ID NO:1393) substantially as described in Example 1. Similar methods also were used to generate libraries of the IgC-like domain of NKp30 (SEQ ID NO:1566).


The degenerate or random library DNA was introduced into yeast substantially as described in Example 2 to generate yeast libraries. The libraries were used to select yeast expressing affinity modified variants of CD80, CD155, CD112, PD-L1, PD-L2, CD86 (B7-2), and NKp30 substantially as described in Example 3. Cells were processed to reduce non-binders and to enrich for CD80, CD155, CD112, PD-L1 or PD-L2 variants with the ability to bind their exogenous recombinant counter-structure proteins substantially as described in Example 3.


With CD80, CD86 and NKp30 libraries, target ligand proteins were sourced from R&D Systems (USA) as follows: human rCD28.Fc (i.e., recombinant CD28-Fc fusion protein), rPDL1.Fc, rCTLA4.Fc, and rB7H6.Fc. Two-color flow cytometry was performed substantially as described in Example 3. Yeast outputs from the flow cytometric sorts were assayed for higher specific binding affinity. Sort output yeast were expanded and re-induced to express the particular IgSF affinity modified domain variants they encode. This population then can be compared to the parental, wild-type yeast strain, or any other selected outputs, such as the bead output yeast population, by flow cytometry.


In the case of NKp30 yeast variants selected for binding to B7-H6, the F2 sort outputs gave MFI values of 533 when stained with 16.6 nM rB7H6.Fc, whereas the parental NKp30 strain MFI was measured at 90 when stained with the same concentration of rB7H6.Fc (6-fold improvement).


Among the NKp30 variants that were identified, was a variant that contained mutations L30V/A60V/S64P/S86G with reference to positions in the NKp30 extracellular domain corresponding to positions set forth in SEQ ID NO:54.


For CD155 variants provided in Table 23A-E, selection involved two positive selections with the desired counter structures TIGIT and CD96 followed by one negative selection with the counter structure CD226 to select away from CD226 and improve binding specificity of the variant CD155. Selection was performed essentially as described in Example 3 above except the concentrations of the counter structures (TIGIT/CD96) and selection stringency of the positive sorts were varied to optimize lead identification. The concentration of CD226 for the negative selection was kept at 100 nM.


For additional CD112 variants provided in Table 24A-B, selection involved two positive selections with the desired counter structures TIGIT and CD112R followed by one negative selection with the counter structure CD226 to select away from CD226 and improve binding specificity of the variant CD112. Selection was performed essentially as described in Example 3 above except the concentrations of the counter structures (TIGIT/CD112R) and selection stringency of the positive sorts were varied to optimize lead identification. The concentration of CD226 for the negative selection was kept at 100 nM.


For PD-L1 and PD-L2 variants provided in Table 25 and 26A-B, yeast display targeted or random PD-L1 or PD-L2 libraries were selected against PD-1. This was then followed by two to three rounds of flow cytometry sorting using exogenous counter-structure protein staining to enrich the fraction of yeast cells that displays improved binders. Alternatively, for PD-L1, selections were performed with human rCD80.Fc (i.e., human recombinant CD80 Fc fusion protein from R&D Systems, USA). Selections were carried out largely as described for PD-1 above. Magnetic bead enrichment and selections by flow cytometry are essentially as described in Miller K. D. et al., Current Protocols in Cytometry 4.7.1-4.7.30, July 2008.


For CD80 variants provided in Tables 27A-B, CD80 libraries consisted of positive selection with the desired counter structure CTLA4 and negative selection with the counter structure CD28.


Exemplary selection outputs were reformatted as immunomodulatory proteins containing an affinity modified (variant) IgV of CD80, variant IgV of CD155, variant IgV of CD112, variant IgV of PD-L1, variant IgV of PD-L2, each fused to an Fc molecule (variant IgV-Fc fusion molecules) substantially as described in Example 4 and the Fc-fusion protein was expressed and purified substantially as described in Example 5. In some cases, Fc-fusion proteins were generated containing: affinity-modified IgSF domain followed by a GSGGGGS linker followed by a human IgG1 Fc containing the mutations L234A, L235E, G237A, E356D and M358L by EU numbering (SEQ ID NO:2153).


Binding of exemplary IgSF domain variants to cell-expressed counter structures (cognate binding partners) was then assessed substantially as described in Example 6. Cells expressing cognate binding partners were produced and binding studies and flow cytometry were carried out substantially as described in Example 6. In addition, the bioactivity of the Fc-fusion variant protein was characterized by either mixed lymphocyte reaction (MLR) or anti-CD3 coimmobilization assay substantially as described in Example 6.


As above, for each Table, the exemplary amino acid substitutions are designated by amino acid position number corresponding to the respective reference unmodified ECD s (see Table 1). The amino acid position is indicated in the middle, with the corresponding unmodified (e.g. wild-type) amino acid listed before the number and the identified variant amino acid substitution listed (or inserted designated by a) after the number.


Also shown is the binding activity as measured by the Mean Fluorescence Intensity (MFI) value for binding of each variant Fc-fusion molecule to cells engineered to express the cognate counter structure ligand and the ratio of the MFI compared to the binding of the corresponding unmodified Fc fusion molecule not containing the amino acid substitution(s) to the same cell-expressed counter structure ligand. The functional activity of the PD-L2 variant Fc-fusion molecules to modulate the activity of T cells also is shown based on the calculated levels of IFN-gamma in culture supernatants (pg/mL) generated with the indicated variant Fc fusion molecule in an MLR assay. Table 26B also depicts the ratio of IFN-gamma produced by each variant IgV-Fc compared to the corresponding unmodified IgV-Fc in an MLR assay.


As shown, the selections resulted in the identification of a number of CD80, CD155, CD112, PD-L1, and PD-L2 IgSF domain variants that were affinity-modified to exhibit increased binding for at least one, and in some cases more than one, cognate counter structure ligand. In addition, the results showed that affinity modification of the variant molecules also exhibited improved activities to both increase and decrease immunological activity.









TABLE 23A







Additional CD155 Variants and Binding Data.














TIGIT
CD226
CD112R
CD96



















Fold

Fold

Fold

Fold



SEQ

↑ to

↑ to

↑ to

↑ to



ID NO
MFI at
WT
MFI at
WT
MFI at
WT
MFI at
WT


CD155 Mutation(s)
(IgV)
100 nM
ECD
100 nM
ECD
100 nM
ECD
100 nM
ECD



















S52M
868
1865.3
0.00
1901.0
0.01
1553.4
0.87
1609.8
0.02


T45Q, S52L, L104E,
869
2287.0
0.01
2390.4
0.01
1735.1
0.97
1575.1
0.02


G111R











S42G
879
4837.5
0.01
2448.1
0.01
1815.4
1.02
1699.6
0.02


Q62F
871
2209.5
0.01
2572.1
0.01
2706.5
1.52
2760.7
0.03


S52Q
872
2288.1
0.01
2022.3
0.01
1790.1
1.00
1822.3
0.02


S42A, L104Q, G111R
873
1923.7
0.00
1901.7
0.01
1815.1
1.02
1703.8
0.02


S42A, S52Q, L104Q,
874
1807.5
0.00
2157.2
0.01
1894.4
1.06
1644.0
0.02


G111R











S52W, L104E
875
1938.2
0.00
1905.6
0.01
2070.6
1.16
1629.5
0.02


S42C
876
1914.0
0.00
2096.1
0.01
1685.0
0.95
1592.4
0.02


S52W
877
1991.6
0.00
2037.3
0.01
1612.8
0.90
1712.9
0.02


S52M, L104Q
878
2666.6
0.01
2252.2
0.01
1706.0
0.96
1633.1
0.02


S42L, S52L, Q62F,
879
2021.4
0.00
2643.8
0.02
1730.1
0.97
2318.7
0.02


L104Q











S42W
880
2434.5
0.01
2133.4
0.01
2325.7
1.30
2555.4
0.03


S42Q
881
2073.5
0.00
2225.9
0.01
1905.1
1.07
2143.1
0.02


S52L
882
2224.8
0.01
2676.3
0.02
2038.6
1.14
2043.2
0.02


S52R
883
4395.4
0.01
3964.4
0.02
2741.7
1.54
4846.9
0.05


L104E
884
3135.4
0.01
2264.2
0.01
1803.5
1.01
1556.7
0.02


G111R
885
2082.7
0.00
2791.3
0.02
2470.9
1.39
3317.1
0.03


S52E
886
2655.4
0.01
2599.8
0.02
1904.9
1.07
1799.0
0.02


Q62Y
887
2528.6
0.01
2621.4
0.02
1918.4
1.08
1827.5
0.02


T45Q, S52M, L104E
888
79498.2
0.19
143238.5
0.83
2600.6
1.46
6310.4
0.06


S42N, L104Q, G111R
889
2432.1
0.01
2311.3
0.01
1847.4
1.04
1958.3
0.02


S52M, V57L
890
1760.7
0.00
2431.6
0.01
2006.9
1.13
1858.7
0.02


S42N, S52Q, Q62F
891
2402.7
0.01
2152.0
0.01
1855.0
1.04
1737.6
0.02


S42A, S52L, L104E,
892
2262.7
0.01
1889.4
0.01
1783.2
1.00
1606.2
0.02


G111R











S42W, S52Q, V57L,
893
1961.4
0.00
2138.3
0.01
1844.9
1.03
1699.6
0.02


Q62Y











L104Q
894
10314.4
0.02
3791.4
0.02
2119.9
1.19
1542.6
0.02


S42L, S52Q, L104E
895
1946.9
0.00
6474.3
0.04
1749.0
0.98
1702.2
0.02


S42C, S52L
896
1762.5
0.00
2147.3
0.01
1663.4
0.93
1484.7
0.01


S42W, S52R, Q62Y,
897
1918.8
0.00
2300.1
0.01
1824.6
1.02
1756.0
0.02


L104Q











T45Q, S52R, L104E
898
121636.9
0.29
142381.2
0.82
2617.9
1.47
3748.2
0.04


S52R, Q62F, L104Q,
899
2969.2
0.01
3171.6
0.02
1725.4
0.97
2362.3
0.02


G111R











T45Q, S52L, V57L,
900
2857.7
0.01
5943.5
0.03
1496.8
0.84
1533.3
0.02


L104E











S52M, Q62Y
901
1926.6
0.00
2000.3
0.01
1771.6
0.99
1651.1
0.02


Q62F, L104E, G111R
902
1966.4
0.00
2043.5
0.01
1701.9
0.95
1524.8
0.02


T45Q, S52Q
903
4812.8
0.01
5787.5
0.03
1765.6
0.99
2451.3
0.02


S52L, L104E
904
4317.8
0.01
2213.9
0.01
1756.9
0.99
1829.3
0.02


S42V, S52E
905
2055.0
0.00
2272.6
0.01
1808.0
1.01
2530.2
0.03


T45Q, S52R, G111R
906
4092.3
0.01
2075.2
0.01
1793.6
1.01
2336.6
0.02


S42G, S52Q, L104E,
907
2010.1
0.00
2019.2
0.01
1706.4
0.96
1707.6
0.02


G111R











S42N, S52E, V57L,
908
1784.2
0.00
1743.6
0.01
1690.1
0.95
1538.7
0.02


L104E











Wildtype
652
1964.7
0.00
2317.1
0.01
2169.6
1.22
1893.4
0.02


S42C, S52M, Q62F
909
1861.0
0.00
2084.2
0.01
1592.3
0.89
1481.3
0.01


S42L
910
1930.4
0.00
2187.2
0.01
1743.2
0.98
1618.4
0.02


Wildtype
652
2182.6
0.01
2374.5
0.01
1743.1
0.98
1680.4
0.02


S42A
911
1929.2
0.00
2188.6
0.01
1733.7
0.97
1623.6
0.02


S42G, S52L, Q62F,
912
1924.3
0.00
2157.6
0.01
1661.3
0.93
1642.1
0.02


L104Q











S42N
913
1817.4
0.00
1910.9
0.01
1699.7
0.95
1691.5
0.02


CD155 IgV Fc
652
4690
0.01
4690
0.03
2941
1.65
3272
0.03



(IgV)










Wildtype CD155 ECD-
 47
423797
1.00
172839
1.00
1783
1.00
99037
1.00


Fc
(ECD)










Anti-human Fc PE

1506.3
0.00
3774
0.02
1587
0.89
1618
0.02
















TABLE 23B







Additional CD155 Variants and Binding Data.













TIGIT
CD226
CD96















SEQ

Fold

Fold

Fold



ID

Increase

Increase

Increase



NO
MFI at
to WT
MFI at
to WT
MFI at
to WT


CD155 Mutation(s)
(IgV)
100 nM
ECD
100 nM
ECD
100nM
ECD





P18T, S65A, S67V, F91S
914
297843
1.99
351195
3.22
128180
1.68









P18F, T39A, T45Q, T61R,
915
Little to no protein produced


S65N, S67L, E73G, R78G
















P18T, T45Q, T61R, S65N, S67L
916
224682
1.50
270175
2.48
 22820
0.30


P18F, S65A, S67V, F91S
917
534106
3.57
350410
3.21
144069
1.89









P18F, T45Q, T61R, S65N,
918
Little to no protein produced


S67L, F91S, L104P
















P18S, L79P, L104M
919
342549
2.29
320823
2.94
107532
1.41


P18S, L104M
920
449066
3.00
295126
2.70
121266
1.59


L79P, L104M
921
 3210
0.02
 8323
0.08
 2894
0.04


P18T, T45Q, L79P
922
542878
3.63
371498
3.40
193719
2.55


P18T, T45Q, T61R, S65H, S67H
923
312337
2.09
225439
2.07
152903
2.01









P18T, A81E
924
Little to no protein produced


P18S, D23Y, E37P, S52G,
925
Little to no protein produced


Q62M, G80S, A81P, G99Y,




S112N
















A13R, D23Y, E37P, S42P, Q62Y,
926
 4161
0.03
 11673
0.11
 5762
0.08


A81E
















A13R, D23Y, E37P, G99Y,
927
Little to no protein produced














S112N
















A13R, D23Y, E37P, Q62M,
928
Little to no protein produced














A77V, G805, A81P, G99Y









P18L, E37S, Q62M, G80S, A81P,
929
 5900
0.04
 14642
0.13
 3345
0.04


G99Y, S112N









P18S, L104T
930
321741
2.15
367470
3.37
108569
1.43


P18S, Q62H, L79Q, F91S
931
283357
1.89
324877
2.98
125541
1.65


P18S, F91S
658
222780
1.49
300049
2.75
 48542
0.64









T45Q, S52K, Q62F, L104Q,
932
Little to no protein produced


G111R




T45Q, 552Q, Q62Y, L104Q,
933
Little to no protein produced


G111R




T45Q, 552Q, Q62Y, L104E,
934
Little to no protein produced


G111R




V57A, T61M, S65W, 567A,
935
Little to no protein produced


E96D, L104T
















P18L, V57T, T61S, S65Y, S67A,
936
278178
1.86
276870
2.54
121499
1.60


L104T









P18T, T45Q
937
326769
2.18
357515
3.28
 92389
1.21









Pl8L, V57A, T61M, S65W,
938
Little to no protein produced














567A, L104T









T61M, S65W, S67A, L104T
939
360915
2.41
417897
3.83
148954
1.96


P18S, V41A, S42G, T45G,
940
 3821
0.03
 11449
0.10
 3087
0.04


L104N









P18H, S42G, T451, S52T, G53R,
941
 5066
0.03
177351
1.63
 3700
0.05


S54H, V57L, H59E, T61S, S65D,









E68G, L104N









P18S, S42G, T45V, F58L, S67W,
942
 14137
0.09
 15175
0.14
 15324
0.20


L104N









P18S, T45I, L104N
943
141745
0.95
298011
2.73
 97246
1.28


P18S, S42G, T45G, L104N,
944
29387
0.20
117965
1.08
 15884
0.21


V106A









P18H, H40R, S42G, T45I,
945
12335
0.08
 14657
0.13
 15779
0.21


S52T, G53R, S54H, V57L,









H59E, T61S, S65D, E68G,









L104Y, V106L, F108H
















E37V, 542G, T45G, L104N
946
Little to no protein produced














P18S, T45Q, L79P, L104T
947
206674
1.38
285512
2.62
 87790
1.15


P18L, Q62R
948
66939
0.45
 25063
0.23
 10928
0.14









Al3R, D23Y, E37P, 542L,
949
Little to no protein produced














552G, Q62Y, A81E









P18L, H49R, L104T, D116N
950
167980
1.12
214677
1.97
 62451
0.82









A13R, D23Y, E37P, Q62M,
951
Little to no protein produced














G80S, A81P, L104T









S65T, L104T
952
205942
1.38
187147
1.71
 65207
0.86









A13R, D23Y, E37P, S52G,
953
Little to no protein produced


V57A, Q62M, K70E, L104T
















P18L, A47V, Q62Y, E73D,
954
146142
0.98
248926
2.28
 73956
0.97


L104T
















H40T, V41M, A47V, S52Q,
955
Little to no protein produced














Q62L, S65T, E73R, D97G,









E98S, L104T, D116N









P18L, S42P, T45Q, T61G, S65H,
956
153536
1.03
402503
3.69
 53044
0.70


S67E, L104T, D116N
















P18S, H40T, V41M, A47V,
957
Little to no protein produced














S52Q, Q62L, S65T, E73R,









L104M, V106A
















H40T, V41M, A47V, S52Q,
958
Little to no protein produced














Q62L, S65T, E68G, E73R,









D97G, E98S, L104T
















T45Q, S52E, L104E
959
Little to no protein produced














T45Q, S52E, Q62F, L104E
960
132850
0.89
276434
2.53
 14558
0.19


Wildtype CD155 ECD-Fc
47
149692
1.00
109137
1.00
 76083
1.00



(ECD)








Anti-human Fc PE

 2287
0.02
 4799
0.04
 2061
0.03
















TABLE 23C







Additional CD155 Variants and Binding Data.













TIGIT
CD226
CD96

















Fold

Fold

Fold





Increase

Increase

Increase



SEQ ID
MFI at
to WT
MFI at
to WT
MFI at
to WT


CD155 Mutations
NO (IgV)
100nM
IgV
100 nM
IgV
100 nM
IgV





P18F, T26M, L44V, Q62K,
961
117327
1.2
 1613
0.1
 1629
0.1


L79P, F91S, L104M,









G111D









P18S, T45S, T61K, S65W,
962
124936
1.3
 2114
0.1
 2223
0.1


S67A, F91S, G111R









P18S, L79P, L104M,
963
110512
1.1
18337
0.9
22793
1.3


T107M









P18S, S65W, S67A, M90V,
964
101726
1.0
 1605
0.1
 2571
0.1


V95A, L104Q, G111R









Wildtype CD155-ECD
47 (ECD)
 98935
1.0
20029
1.0
17410
1.0
















TABLE 23D







Additional CD155 Variants and Binding Data.













TIGIT
CD226
CD96

















Fold

Fold

Fold





Change

Change

Change





from

from

from



SEQ ID
MFI at
CD155-
MFI at
CD155-
MFI at
CD155


CD155 Mutations
NO (IgV)
11.1 nM
ECD
11.1 nM
ECD
11.1 nM
ECD

















P18S, A47G, L79P, F91S,
1678
56,409
1.19
1,191
0.08
25,362
1.49


L104M, T107A, R113W









P18T, D23G, S24A, N35D,
1679
128,536
2.72
987
0.06
3,497
0.20


H49L, L79P, F91S,









L104M, G111R









V9L, P18S, Q60R, V75L,
1680
125,329
2.65
986
0.06
959
0.06


L79P, R89K, F91S,









L104E, G111R
















P18S, H49R, E73D, L79P,
1681
Little to no protein produced














N85D, F91S, V95A,









L104M, G111R









V11A, P18S, L79P, F91S,
1682
48,246
1.02
974
0.06
923
0.05


L104M, G111R









V11A, P18S, S54R, Q60P,
1683
190,392
4.02
1,019
0.07
1,129
0.07


Q62K, L79P, N85D, F91S,









T107M









P18T, S52P, S65A, S67V,
1684
121,611
2.57
986
0.06
16,507
0.97


L79P, F91S, L104M,









G111R









P18T, M36T, L79P, F91S,
1685
150,015
3.17
1,029
0.07
2,514
0.15


G111R









D8G, P18S, M36I, V38A,
1686
79,333
1.68
1,026
0.07
2,313
0.14


H49Q, A76E, F91S,









L104M, T107A, R113W









P18S, S52P, S65A, S67V,
1687
23,766
0.50
1,004
0.07
1,080
0.06


L79P, F91S, L104M,









T107S, R113W









T151, P18T, L79P, F91S,
1688
55,498
1.17
1,516
0.10
1,030
0.06


L104M, G111R









P18F, T26M, L44V,
1689
213,640
4.51
991
0.06
1,276
0.07


Q62K, L79P, E82D, F91S,









L104M, G111D









P18T, E37G, G53R, Q62K,
1690
251,288
5.31
2,001
0.13
45,878
2.69


L79P, F91S, E98D,









L104M, T107M









P18L, K70E, L79P, F91S,
1691
62,608
1.32
1,117
0.07
973
0.06


V95A, G111R









V9I, Q12K, P18F, S65A,
1692
81,932
1.73
803
0.05
68,295
4.00


S67V, L79P, L104T,









G111R, S1121









P18F, S65A, S67V, F91S,
1693
30,661
0.65
901
0.06
3,193
0.19


L104M, G111R









V9I, V101, P18S, F20S,
1694
151,489
3.20
973
0.06
974
0.06


T45A, L79P, F91S,









L104M, F108Y, G111R,









S112V









V9L, P18L, L79P, M90I,
1695
155,279
3.28
910
0.06
10,568
0.62


F91S, T102S, L104M,









G111R









P18C, T26M, L44V, M55I,
1696
137,521
2.91
973
0.06
111,085
6.51


Q62K, L79P, F91S,









L104M, T107M









V9I, P18T, D23G, L79P,
1697
151,426
3.20
897
0.06
2,725
0.16


F91S, G111R









P18F, L79P, M90L, F91S,
1698
125,639
2.66
917
0.06
3,939
0.23


V95A, L104M, G111R









P18F, L79P, M9OL, F91S,
1698
115,156
2.43
1,073
0.07
2,464
0.14


V95A, L104M, G111R









P18T, M36T, 565A, 567E,
1699
10,616
0.22
1,130
0.07
963
0.06


L79Q, A81T, F91S,









G111R









V9L, P18T, Q62R, L79P,
1700
195,111
4.12
835
0.05
1,497
0.09


F91S, L104M, G111R









CD155-ECD-Fe
47 (ECD)
47,319
1.00
15,421
1.00
17,067
1.00


Fe Control
-
2,298
0.05
1,133
0.07
996
0.06
















TABLE 23E







Additional CD155 Variants and Binding Data.














TIGIT
CD226
CD112R
CD96



















Fold

Fold

Fold

Fold





Change

Change

Change

Change



SEQ

from

from

from

from



ID NO
MFI at
CD155-
MFI at
CD155-
MFI at
CD155-
MFI at
CD155-


CD155 Mutations
(IgV)
25nM
ECD
25 nM
ECD
25nM
ECD
25nM
ECD



















P18T, G19D, M36T, S54N,
1819
905
0.02
748
0.02
1276
1.56
726
0.01


L79P, L83Q, F91S, T107M,











F108Y











V9L, P18L, M55V, S69L,
1820
58656
1.34
11166
0.29
920
1.13
67364
1.39


L79P, A81E, F91S, T107M











P18F, H40Q, T61K, Q62K,
1821
108441
2.48
853
0.02
918
1.13
8035
0.17


L79P, F91S, L104M,











T107V











P18S, Q32R, Q62K, R78G,
1822
5772
0.13
701
0.02
843
1.03
831
0.02


L79P, F91S, T107A,











R113W











Q12H, P18T, L21S, G22S,
1823
1084
0.02
687
0.02
876
1.07
818
0.02


V57A, Q62R, L79P, F91S,











T107M











V9I, P18S, S24P, H49Q,
1824
69926
1.60
1089
0.03
1026
1.26
43856
0.90


F58Y, Q60R, Q62K, L79P,











F91S, T107M











P18T, W46C, H49R, S65A,
1825
918
0.02
640
0.02
803
0.98
717
0.01


S67V, A76T, L79P, S87T,











L104M











P18S, S42T, E51G, L79P,
1826
12630
0.29
707
0.02
857
1.05
1050
0.02


F91S, G92W, T107M











P18S, S42T, E51G, L79P,
1826
7476
0.17
851
0.02
935
1.15
924
0.02


F91S, G92W, T107M











V10F, T15S, Pl8L, R48Q,
1827
1168
0.03
792
0.02
901
1.10
998
0.02


L79P, F91S, T107M,











V115M











P18S, L21M, Y30F, N35D,
1828
1377
0.03
743
0.02
946
1.16
1033
0.02


R84W, F91S, T107M,











D116G











P18F, E51V, S54G, Q60R,
1829
46090
1.05
15701
0.41
1012
1.24
61814
1.27


L79Q, E82G, S87T, M90I,











F91S, G92R, T107M


















Q16H, P18F, F91S, T107M
1830
Little to no protein produced
















P18T, D23G, Q60R, S67L,
1831
64091
1.47
30931
0.81
874
1.07
108875
2.24


L79P, F91S, T107M,











V115A











D8G, V9I, V11A, P18T,
1832
52508
1.20
9483
0.25
817
1.00
97770
2.01


T26M, S52P, L79P, F91S,











G92A, T107L, V115A











V9I, P18F, A47E, G50S,
1833
55167
1.26
54341
1.43
752
0.92
102115
2.10


E68G, L79P, F91S, T107M


















P18S, M55I, Q62K, S69P,
1834
Little to no protein produced
















L79P, F91S, T107M











P18T, T39S, S52P, S54R,
1835
45927
1.05
744
0.02
1038
1.27
1225
0.03


L79P, F91S, T107M


















P18S, D23N, L79P, F91S,
1836
Little to no protein produced
















T107M, S114N











P18S, P34S, E51V, L79P,
1837
7917
0.18
769
0.02
853
1.04
892
0.02


F91S, G111R











P18S, H59N, V75A, L79P,
1838
800
0.02
676
0.02
915
1.12
759
0.02


A81T, F91S, L104M,











T107M











P18S, W46R, E68D, L79P,
1839
1359
0.03
717
0.02
798
0.98
737
0.02


F91S, T107M, R113G











V9L, P18F, T45A, S65A,
1840
130274
2.98
153569
4.04
812
1.00
85605
1.76


S67V, R78K, L79V, F91S,











T107M, S114T











P18T, M55L, T61R, L79P,
1841
133399
3.05
1906
0.05
827
1.01
57927
1.19


F91S, V1061, T107M











T151, P18S, V33M, N35F,
1842
7550
0.17
1015
0.03
789
0.97
2709
0.06


T39S, M55L, R78S, L79P,











F91S, T107M











P18S, Q62K, K70E, L79P,
1843
11173
0.26
691
0.02
735
0.90
1951
0.04


F91S, G92E, R113W











P18F, F20I, T26M, A47V,
1844
136088
3.11
54026
1.42
1401
1.72
96629
1.99


E51K, L79P, F91S











P18T, D23A, Q60H, L79P,
1845
43795
1.00
98241
2.58
888
1.09
70891
1.46


M90V, F91S, T107M











P18S, D23G, C29R, N35D,
1846
1599
0.04
1030
0.03
1115
1.37
1944
0.04


E37G, M55I, Q62K, S65A,











S67G, R78G, L79P, F91S,











L104M, T107M, Q110R


















A13E, P18S, M36R, Q62K,
1847
Little to no protein produced
















S67T, L79P, N85D, F91S,











T107M











V9I, P18T, H49R, L79P,
1848
46375
1.06
76851
2.02
794
0.97
80210
1.65


N85D, F91S, L104T,











T107M











V9A, P18F, T61S, Q62L,
1849
26109
0.60
891
0.02
825
1.01
2633
0.05


L79P, F91S, G111R


















D8E, P18T, T61A, L79P,
1850
Little to no protein produced
















F91S, T107M











P18S, V41A, H49R, S54C,
1851
1098
0.03
830
0.02
876
1.07
1678
0.03


L79S, N85Y, L88P, F91S,











L104M, T107M











V11E, P18H, F20Y, V25E,
1852
979
0.02
846
0.02
844
1.03
928
0.02


N35S, H49R, L79P, F91S,











T107M, G111R











V11A, P18F, D23A, L79P,
1853
45249
1.04
913
0.02
830
1.02
33883
0.70


G80D, V95A, T107M











P18S, K70R, L79P, F91S,
1854
16180
0.37
793
0.02
854
1.05
1182
0.02


G111R











P18T, D23A, Q60H, L79P,
1845
175673
4.02
161958
4.26
879
1.08
50981
1.05


M90V, F91S, T107M











V9L, V11M, P18S, N35S,
1855
2999
0.07
2315
0.06
893
1.09
925
0.02


S54G, Q62K, L79P,











L104M, T107M, V115M











V9L, P18Y, V25A, V38G,
1856
138011
3.16
26015
0.68
919
1.13
17970
0.37


M55V, A77T, L79P, M90I,











F91S, L104M











VlOG, P18T, L72Q, L79P,
1857
4253
0.10
1584
0.04
863
1.06
3643
0.07


F91S, T107M











P18S, H59R, A76G, R78S,
1858
130622
2.99
79435
2.09
1009
1.24
44493
0.91


L79P











V9A, P18S, M36T, S65G,
1859
92503
2.12
989
0.03
886
1.09
7850
0.16


L79P, F91S, L104T,











G111R, S1121











P18T, S52A, V57A, Q60R,
1860
187338
4.29
10579
0.28
908
1.11
3791
0.08


Q62K, S65C, L79P, F91T,











N100Y, T107M


















V11A, P18F, N35D, A47E,
1861
Little to no protein produced
















Q62K, L79P, F91S, G99D,











T107M, S114N











V11A, P18T, N35S, L79P,
1862
218660
5.00
273825
7.20
1269
1.56
69871
1.44


S87T, F91S











V9D, V11M, Q12L, P18S,
1863
8693
0.20
790
0.02
852
1.04
1991
0.04


E37V, M55I, Q60R, K70Q,











L79P, F91S, L104M,











T107M











T15S, P18S, Y30H, Q32L,
1864
16213
0.37
2092
0.06
1056
1.29
6994
0.14


Q62R, L79P, F91S, T107M











CD155-ECD-Fc
47
43704
1.00
38032
1.00
816
1.00
48638
1.00



(ECD)










CD112-IgV
965
1289

824

17819

1172
0.02
















TABLE 24A







Additional CD112 Variants and Binding Data.














TIGIT
CD226
CD112R
CD96



















Fold

Fold

Fold

Fold



SEQ

Increase

Increase

Increase

Increase


CD112
ID NO
MFI
to WT
MFI at
to WT
MFI at
to WT
MFI at
to WT


Mutation(s)
(IgV)
100 nM
IgV
100 nM
IgV
100 nM
IgV
100 nM
IgV



















S118F
972
1763
0.02
1645
0.08
2974
0.61
1659
0.19


N47K, Q79R,
1095
1738
0.02
1689
0.09
2637
0.54
1647
0.19


S118F











Q40R, P60T,
1096
4980
0.06
1608
0.08
2399
0.50
2724
0.32


A112V, S118T











F114Y, S118F
1097
110506
1.34
7325
0.37
1502
0.31
1553
0.18


N106I, S118Y
977
1981
0.02
1700
0.09
2394
0.49
1582
0.19


S118Y
976
101296
1.23
9990
0.50
1429
0.30
1551
0.18


Y33H, K78R,
1098
2276
0.03
2115
0.11
3429
0.71
2082
0.24


S118Y











N1061, S118F
978
1875
0.02
1675
0.08
2365
0.49
1662
0.19


R12W, A46T,
1099
3357
0.04
1808
0.09
1664
0.34
4057
0.48


K66M, Q79R,











N106I, T113A,











S118F











Y33H, A112V,
1100
3376
0.04
2886
0.15
3574
0.74
3685
0.43


S118F











R12W, Y33H,
1101
100624
1.22
24513
1.24
1490
0.31
2060
0.24


N106I, S118F











L15V, Q90R,
1102
5791
0.07
4169
0.21
2752
0.57
4458
0.52


S118F











N47K, D84G,
1103
3334
0.04
2819
0.14
2528
0.52
3498
0.41


N106I, S118Y











L32P, S118F
1104
3881
0.05
2506
0.13
2659
0.55
2518
0.29


Y33H, Q79R,
1105










A112V, S118Y











T18A, N106I,
1106
84035
1.02
10208
0.52
1585
0.33
1590
0.19


S118T











L15V, Y33H,
1107










N106Y, A112V,











S118F











V37M, S118F
1108
96986
1.18
2523
0.13
1985
0.41
1849
0.22


N47K, A112V,
1109
1980
0.02
1859
0.09
2733
0.56
1825
0.21


S118Y











A46T, A112V
1110
4224
0.05
4685
0.24
3288
0.68
4273
0.50


P28S, Y33H,
1111
6094
0.07
2181
0.11
1891
0.39
3021
0.35


N106I, S118Y











P3OS, Y33H,
1112
2247
0.03
2044
0.10
1796
0.37
2658
0.31


N47K, V75M,











Q79R, N106I,











S118Y











V19A, N47K,
1113
2504
0.03
2395
0.12
2174
0.45
2852
0.33


N106Y, K116E,











S118Y











Q79R, T85A,
1114
2192
0.03
1741
0.09
2367
0.49
1620
0.19


A112V, S118Y











Y33H, A112V
994
20646
0.25
1465
0.07
1794
0.37
2589
0.30


V101M, N106I,
1115
55274
0.67
6625
0.33
1357
0.28
1494
0.17


S118Y











Y33H, Q79R,
1116
6095
0.07
1760
0.09
2393
0.49
3033
0.36


N1061, A112V,











S118T











Q79R, A112V
1117
1571
0.02
1490
0.08
2284
0.47
1326
0.16


Y33H, A46T,
1118
90813
1.10
15626
0.79
1298
0.27
3571
0.42


Q79R, N106I,











S118F











A112V, G121S
1119
95674
1.16
19992
1.01
1252
0.26
4005
0.47


Y33H, Q79R,
1120
36246
0.44
2118
0.11
1970
0.41
3250
0.38


N106I, S118Y











Y33H, N106I,
1121
47352
0.57
4217
0.21
2641
0.55
1488
0.17


A112V











Y33H, A46T,
1122
14413
0.17
1596
0.08
2335
0.48
1441
0.17


V101M,











A112V, S118T











L32P, L99M,
1123
3056
0.04
1791
0.09
2210
0.46
2000
0.23


N106I, S118F











L32P, T108A,
1124
104685
1.27
4531
0.23
2308
0.48
1518
0.18


S118F











A112V
1002
4937
0.06
1903
0.10
1646
0.34
3011
0.35


R12W, Q79R,
1125
55539
0.67
6918
0.35
1386
0.29
1740
0.20


A112V











Y33H, N106Y,
1126
2786
0.03
2517
0.13
1787
0.37
2023
0.24


E110G, A112V











Y33H, N106I,
1127
1967
0.02
1579
0.08
2601
0.54
1517
0.18


S118Y











Q79R, S118F
1128
82055
1.00
7582
0.38
1298
0.27
1970
0.23


Y33H, Q79R,
1129
21940
0.27
1632
0.08
1141
0.24
18423
2.16


G98D, V101M,











A112V











N47K, T81S,
1130
6889
0.08
1311
0.07
1303
0.27
1145
0.13


V101M,











A112V, S118F











G82S, S118Y
1131
4267
0.05
1938
0.10
2140
0.44
2812
0.33


Y33H, All2V,
1132
14450
0.18
1532
0.08
2353
0.49
3004
0.35


S118Y











Y33H, N47K,
1133
70440
0.85
3557
0.18
1447
0.30
1679
0.20


Q79R, N106Y,











A112V











Y33H, S118T
1134
113896
1.38
17724
0.89
1252
0.26
5001
0.59


R12W, Y33H,
1135
3376
0.04
2727
0.14
2047
0.42
2339
0.27


Q79R, V101M,











A112V











S118F
972
2685
0.03
1864
0.09
2520
0.52
1566
0.18


Wildtype
965
82414
1.00
19803
1.00
4842
1.00
8541
1.00


CD112-IgV Fc
(IgV)










CD112 ECD-Fc
48
29157
0.35
8755
0.44
1107
0.23
1103
0.13



(ECD)










Anti-hFc PE

1383
0.02
1461
0.07
1358
0.28
1468
0.17
















TABLE 24B







Additional CD112 Variants and Binding Data.














TIGIT
CD226
CD112R
CD96



















Fold

Fold

Fold

Fold



SEQ

Increase

Increase

Increase

Increase


CD112
ID NO
MFI
to WT
MFI at
to WT
MFI at
to WT
MFI at
to WT


Mutation(s)
(IgV)
20 nM
IgV
20 nM
IgV
20 nM
IgV
20 nM
IgV



















N1061, S118Y
977
1288
0.04
1334
0.12
6920
4.16
1102
0.44


Y33H, Q83K,
1631
115690
3.31
10046
0.93
1128
0.68
2053
0.82


A112V, S118T











R12W, Q79R,
974
1436
0.04
1296
0.12
6546
3.93
1046
0.42


S118F











V29M, Y33H,
1632
Not tested









N106I, S118F











Y33H, A46T,
1633
111256
3.18
14974
1.39
1148
0.69
3333
1.34


A112V











Y33H, Q79R,
1634
1483
0.04
1326
0.12
7425
4.46
1138
0.46


S118F











Y33H, N47K,
1635
1338
0.04
1159
0.11
1516
0.91
1140
0.46


F74L, S118F











R12W, V101M,
1636
1378
0.04
1249
0.12
5980
3.59
1182
0.47


N106I, S118Y











A46T, V101A,
1637
1359
0.04
1199
0.11
6729
4.04
1173
0.47


N106I, S118Y











Y33H, N106Y,
991
113580
3.25
17771
1.65
1207
0.72
2476
0.99


A112V


















N106Y, A112V,
1638
Not tested
















S118T


















S76P, T81I,
1639
Not tested
















V101M,











N106Y, A112V,











S118F











N106Y, A112V
981
29015
0.83
2760
0.26
1159
0.70
1639
0.66


P9R, L21V,
1640
1920
0.05
1218
0.11
1107
0.66
1074
0.43


P22L, I34M,











S69F, F74L,











A87V, A112V,











L125A











Y33H, V101M,
1641
126266
3.61
24408
2.27
1150
0.69
4535
1.82


A112V











N1061, S118F
978
1776
0.05
1385
0.13
9058
5.44
1370
0.55


V29A, L32P,
1642
1265
0.04
1148
0.11
5057
3.04
1194
0.48


S118F











A112V
1002
69673
1.99
6387
0.59
1140
0.68
1214
0.49


Y33H, V101M,
1641
133815
3.83
24992
2.32
1184
0.71
6338
2.54


A112V











P28S, Y33H,
1111
2745
0.08
1689
0.16
6625
3.98
1978
0.79


N106I, S118Y











Y33H, V101M,
1643
118654
3.40
21828
2.03
1253
0.75
3871
1.55


N106I, A112V











R12W, Y33H,
1644
171390
4.91
5077
0.47
1124
0.68
2636
1.06


N47K, Q79R,











S118Y











A112V, S118T
1004
103203
2.95
15076
1.40
1155
0.69
1426
0.57


Y33H, A46T,
1645
141859
4.06
29436
2.74
1184
0.71
5760
2.31


A112V, S118T











Y33H, All2V,
1646
5161
0.15
1734
0.16
1184
0.71
1249
0.50


F114L, S118T











A112V
1002
78902
2.26
6224
0.58
1114
0.67
1181
0.47


Y33H, T38A,
1647
111293
3.19
25702
2.39
1192
0.72
99015
39.69


A46T, V101M,











A112V











Q79R, A112V
1117
96674
2.77
7264
0.67
1130
0.68
1216
0.49


Y33H, N1061,
1127
5720
0.16
1453
0.14
6543
3.93
1248
0.50


S118Y











P28S, Y33H,
1648
22393
0.64
1378
0.13
1550
0.93
19174
7.68


S69P, N106I,











A112V, S118Y











Y33H, P42L,
1649
214116
6.13
13878
1.29
1315
0.79
4753
1.91


N47K, V101M,











A112V











Y33H, N47K,
1650
6719
0.19
1319
0.12
1305
0.78
1278
0.51


F74S, Q83K,











N106I, F111L,











A112V, S118T











Y33H, A112V,
1651
184794
5.29
10204
0.95
1269
0.76
4321
1.73


S118T, V119A











Y33H, N106I,
1652
6872
0.20
1591
0.15
2308
1.39
2796
1.12


A112V, S118F











Y33H, K66M,
1653
1724
0.05
1259
0.12
6782
4.07
1197
0.48


S118F, W124L











S118F
972
1325
0.04
1213
0.11
7029
4.22
1135
0.46


N1061, A112V
1654
111342
3.19
4241
0.39
1546
0.93
1178
0.47


Y33H, A112V
994
177926
5.09
13761
1.28
1152
0.69
3117
1.25


WT CD112 IgV
965
34932
1.00
10762
1.00
1665
1.00
2495
1.00


WT CD112-Fc
48
28277
0.81
8023
0.75
1253
0.75
1064
0.43


ECD
(ECD)










Anti-huFc PE

1138
0.03
1006
0.09
1010
0.61
1062
0.43
















TABLE 25A







Selected PD-L1 variants and binding data.









Binding to Jurkat/PD-1 Cells













Fold increase over



SEQ ID NO

wildtype PD-L1


PD-L1 Mutation(s)
(IgV)
MFI at 50 nM
IgV-Fc













K28N, M41V, N45T, H51N, K57E
1267
12585
2.4


I20L, I36T, N45D, I47T
1268
3119
0.6


I20L, M41K, K44E
1269
9206
1.8


P6S, N45T, N78I, I83T
1270
419
0.1


N78I
1271
2249
0.4









M41K, N78I
1272
Little or no protein produced


I20L, I36T, N45D
1277
Little or no protein produced


N17D, N45T, V50A, D72G
1278
Little or no protein produced


I20L, F49S
1279
Little or no protein produced










N45T, V50A
1280
23887
4.6


I20L, N45T, N78I
1281
29104
5.6


N45T, N78I
1273
24865
4.7


I20L, N45T
1274
24279
4.6


I20L, N45T, V50A
1282
34158
6.5


N45T
1275
6687
1.3


M41K
1276
5079
1.0









M41V, N45T
1283
Little or no protein produced


M41K, N45T
1284
Little or no protein produced










A33D, S75P, D85E
1285
685
0.1


M18I, M41K, D43G, H51R, N78I
1286
20731
4.0


V11E, I20L, I36T, N45D, H60R, S75P
1287
3313
0.6









A33D, V50A
1288
Little or no protein produced


S16G, A33D, K71E, S75P
1289
Little or no protein produced










E27G, N45T, M97I
1290
881
0.2


E27G, N45T, K57R
1291
5022
1.0


A33D, E53V
1292
650
0.1


D43G, N45D, V58A
1293
63960
12.2


E40G, D43V, N45T, V50A
1294
809
0.2


Y14S, K28E, N45T
1295
16232
3.1


A33D, N78S
1296
1725
0.3


A33D, N78I
1297
8482
1.6


A33D, N45T
1298
17220
3.3


A33D, N45T, N78I
1299


E27G, N45T, V50A
1300
25267
4.8


N45T, V50A, N78S
1301
28572
5.4


N45T, V50A
1280
18717
3.6


I20L, N45T, V110M
1302
464
0.1


I20L, I36T, N45T, V50A
1303
7658
1.5


N45T, L74P, S75P
1304
5251
1.0


N45T, S75P
1305
12200
2.3


S75P, K106R
1306
388
0.1


S75P
1307
1230
0.2


A33D, S75P
1308
306
0.1


A33D, S75P, D104G
1309
251
0.0


A33D, S75P
1310
1786
0.3


I20L, E27G, N45T, V50A
1311
29843
5.7


I20L, E27G, D43G, N45D, V58A,
1312
69486
13.3


N78I


I20L, D43G, N45D, V58A, N781
1313
72738
13.9


I20L, A33D, D43G, N45D, V58A,
1314
80205
15.3


N78I


I20L, D43G, N45D, N78I
1315
67018
12.8


E27G, N45T, V50A, N78I
1316
30677
5.9


N45T, V50A, N78I
1317
32165
6.1


V11A, I20L, E27G, D43G, N45D,
1318
73727
14.1


H51Y, S99G


I20L, E27G, D43G, N45T, V50A
1319
36739
7.0


I20L, K28E, D43G, N45D, V58A,
1320
80549
15.4


Q89R, G101G-ins


I20L, I36T, N45D
1321
16870
3.2


I20L, K28E, D43G, N45D, E53G,
1322
139
0.0


V58A, N78I


A33D, D43G, N45D, V58A, S75P
1323
58484
11.2


K23R, D43G, N45D
1324
67559
12.9


I20L, D43G, N45D, V58A, N78I,
1325
259
0.0


D90G, G101D


D43G, N45D, L56Q, V58A, G101G-
1326
88277
16.8


ins (G101GG)


I20L, K23E, D43G, N45D, V58A,
1327
89608
17.1


N78I


I20L, K23E, D43G, N45D, V50A,
1328
88829
16.9


N78I


T19I, E27G, N45I, V50A, N78I, M97K
1329
25496
4.9


I20L, M41K, D43G, N45D
1330
599
0.1


K23R, N45T, N78I
1331
84980
16.2


Full length PD-L1 Fc

18465
3.5


Wild type PD-L1 IgV
1332
5243
1.0


Anti-PD-1 monoclonal antibody

79787
15.2


(nivolumab)


Human IgG

198
0.0
















TABLE 25B







Flow Binding to Cells Expressing PD-1 or CD80










PD-1
CD80















Fold

Fold





Change

Change



SEQ ID

Compared

Compared



NO
MFI at
to WT PD-
MFI at
to WT


PD-L1 Mutation(s)
(ECD)
20 nM
L1
20 nM
PD-L1















K57R, S99G
1875
2953
0.9
16253
121.3


K57R, S99G, F189L
1876
1930
0.6
12906
96.3


M18V, M97L, F193S, R195G,
1877
69
0.0
241
1.8


E200K, H202Q


I36S, M41K, M97L, K144Q,
1878
3498
1.1
68715
512.8


R195G, E200K, H202Q, L206F









C22R, Q65L, L124S, K144Q,
1879
Little or no protein produced


R195G, E200N, H202Q, T221L












M18V, I98L, L124S, P198T,
1880
2187
0.7
143
1.1


L206F









S99G, N117S, I148V, K171R,
1881
Little or no protein produced


R180S












I36T, M97L, A103V, Q155H
1882
120
0.0
128
1.0


K28I, S99G
1883
830
0.3
693
5.2


R195S
1884
3191
1.0
138
1.0


A79T, S99G, T185A, R195G,
1885
1963
0.6
643
4.8


E200K, H202Q, L206F


K57R, S99G, L124S, K144Q
1886
2081
0.7
14106
105.3


K57R, S99G, R195G
1887
2479
0.8
10955
81.8


D55V, M97L, S99G
1888
11907
3.8
71242
531.7


E27G, I36T, D55N, M97L, K111E
1889
1904
0.6
88724
662.1


E54G, M97L, S99G
1890
8414
2.7
51905
387.4


G15A, I36T, M97L, K111E,
1891
112
0.0
13530
101.0


H202Q


G15A, I36T, V129D
1892
114
0.0
136
1.0


G15A, I36T, V129D, R195G
1893
125
0.0
134
1.0


G15A, V129D
1894
2075
0.7
128
1.0


I36S, M97L
1895
3459
1.1
44551
332.5


I36T, D55N, M97L, K111E,
1896
265
0.1
62697
467.9


A204T


I36T, D55N, M97L, K111E,
1897
393
0.1
72641
542.1


V129A, F173L


I36T, D55S, M97L, K111E, I148V,
1898
94
0.0
30704
229.1


R180S


I36T, G52R, M97L, V112A,
1899
81
0.0
149
1.1


K144E, V175A, P198T


I36T, I46V, D55G, M97L, K106E,
1900
69
0.0
190
1.4


K144E, T185A, R195G


I36T, I83T, M97L, K144E, P198T
1901
62
0.0
6216
46.4









I36T, M97L, K111E
1902
Little or no protein produced












I36T, M97L, K144E, P198T
1903
197
0.1
40989
305.9


I36T, M97L, Q155H, F193S,
1904
69
0.0
1251
9.3


N201Y


I36T, M97L, V129D
1905
523
0.2
50905
379.9


L35P, I36S, M97L, K111E
1906
190
0.1
155
1.2


M18I, I36T, E53G, M97L, K144E,
1907
104
0.0
47358
353.4


E199G, V207A


M18T, I36T, D55N, M97L, K111E
1908
138
0.0
71440
533.1


M18V, M97L, T176N, R195G
1909
1301
0.4
45300
338.1


M97L, S99G
1910
12906
4.1
81630
609.2


N17D, M97L, S99G
1911
10079
3.2
73249
546.6


S99G, T185A, R195G, P198T
1912
2606
0.8
22062
164.6


V129D, H202Q
1913
2001
0.6
219
1.6


V129D, P198T
1914
3245
1.0
152
1.1


V129D, T150A
1915
1941
0.6
142
1.1


V93E, V129D
1916
1221
0.4
150
1.1


Y10F, M18V, S99G, Q138R,
1917
70
0.0
412
3.1


T203A


WT PD-L1 (IgV + IgC) Fc

3121
1.0
134
1.0


CTLA4-Fc

59
N/A
199670
N/A


Anti-PD1 mAb

31482
N/A
134
N/A


Fc Control

59
N/A
132
N/A
















TABLE 25C







Additional Affinity-Matured IgSF Domain-Containing Molecules











SEQ ID




NO



PD-L1 Mutation(s)
(ECD)







N45D
1918



K160M, R195G
1919



N45D, K144E
1920



N45D, P198S
1921



N45D, P198T
1922



N45D, R195G
1923



N45D, R195S
1924



N45D, S131F
1925



N45D, V58D
1926



V129D, R195S
1927



I98T, F173Y, L196S
1928



N45D, E134G, L213P
1929



N45D, F173I, S177C
1930



N45D, I148V, R195G
1931



N45D, K111T, R195G
1932



N45D, N113Y, R195S
1933



N45D, N165Y, E170G
1934



N45D, Q89R, I98V
1935



N45D, S131F, P198S
1936



N45D, S75P, P198S
1937



N45D, V50A, R195T
1938



E27D, N45D, T183A, I188V
1939



F173Y, T183I, L196S, T203A
1940



K23N, N45D, S75P, N120S
1941



N45D, G102D, R194W, R195G
1943



N45D, G52V, Q121L, P198S
1943



N45D, I148V, R195G, N201D
1944



N45D, K111T, T183A, I188V
1945



N45D, Q89R, F189S, P198S
1946



N45D, S99G, C137R, V207A
1947



N45D, T163I, K167R, R195G
1948



N45D, T183A, T192S, R194G
1949



N45D, V50A, I119T, K144E
1950



T19A, N45D, K144E, R195G
1951



V11E, N45D, T130A, P198T
1952



V26A, N45D, T163I, T185A
1953



K23N, N45D, L124S, K167T, R195G
1954



K23N, N45D, Q73R, T163I
1955



K28E, N45D, W149R, S158G, P198T
1956



K28R, N45D, K57E, I98V, R195S
1957



K28R, N45D, V129D, T163N, R195T
1958



M41K, D43G, N45D, R64S, R195G
1959



M41K, D43G, N45D, R64S, S99G
1960



N45D, R68L, F173L, D197G, P198S
1961



N45D, V50A, I148V, R195G, N201D
1962



M41K, D43G, K44E, N45D, R195G,
1963



N201D



N45D, V50A, L124S, K144E, L179P,
1964



R195G

















TABLE 26A







Variant PD-L2 selected against PD-1. Molecule sequence and binding data.










Binding to Jurkat/PD-1
Fortebio



Cells
binding to












SEQ

Fold increase
PD-1-Fc



ID NO
MFI at
over wildtype
Response


PD-L2 mutation(s)
(IgV)
50 nM
PD-L2 IgV-Fc
Units














H15Q
1487
15998
1.63
0.007


N24D
1488
1414
0.14
−0.039


E44D
1489
2928
0.3
−0.006


V89D
1490
3361
0.34
0.005


Q82R, V89D
1491
44977
4.57
1.111


E59G, Q82R
1492
12667
1.29
−0.028


S39I, V89D
1493
26130
2.65
0.26


S67L, V89D
1494
15991
1.62
0.608


S67L, I85F
1495
529
0.05
−0.005


S67L, I86T
1496
6833
0.69
0.141


H15Q, K65R
1497
13497
1.37
−0.001


H15Q, Q72H, V89D
1498
12629
1.28
0.718


H15Q, S67L, R76G
1499
47201
4.8
0.418


H15Q, R76G, I85F
1500
2941
0.3
−0.038


H15Q, T47A, Q82R
1501
65174
6.62
0.194


H15Q, Q82R, V89D
1502
49652
5.04
1.198


H15Q, C23S, I86T
1503
830
0.08
−0.026


H15Q, S39I, I86T
1504
1027
0.1
0.309


H15Q, R76G, I85F
1505
1894
0.19
−0.006


E44D, V89D, W91R
1506
614
0.06
−0.048


I13V, S67L, V89D
1507
26200
2.66
1.42


H15Q, S67L, I86T
1508
15952
1.62
0.988


I13V, H15Q, S67L, I86T
1509
21570
2.19
1.391


I13V, H15Q, E44D, V89D
1510
23958
2.43
1.399


I13V, S39I, E44D, Q82R, V89D
1511
71423
7.26
0.697


I13V, E44D, Q82R, V89D
1512
45191
4.59
1.283


I13V, Q72H, R76G, I86T
1513
10429
1.06
0.733


I13V, H15Q, R76G, I85F
1514
4736
0.48
−0.04


H15Q, S67L, R76G, I85F
1516
2869
0.29
0.025









H15Q, S39I, R76G, V89D
1515
Little or no protein produced











H15Q, T47A, Q72H, R76G, I86T
1517
32103
3.26
0.512


H15Q, T47A, Q72H, R76G
1518
16500
1.68
0.327


I13V, H15Q, T47A, Q72H, R76G
1519
73412
7.46
0.896


H15Q, E44D, R76G, I85F
1520
2885
0.29
−0.013


H15Q, S39I, S67L, V89D
1521
45502
4.62
1.174


H15Q, N32D, S67L, V89D
1522
25880
2.63
1.407


N32D, S67L, V89D
1523
31753
3.23
1.155


H15Q, S67L, Q72H, R76G, V89D
1524
40180
4.08
1.464


H15Q, Q72H, Q74R, R76G, I86T
1525
4049
0.41
0.093


G28V, Q72H, R76G, I86T
1526
5563
0.57
0.003


I13V, H15Q, S39I, E44D, S67L
1527
63508
6.45
0.889


E44D, S67L, Q72H, Q82R, V89D
1528
51467
5.23
1.061


H15Q, V89D
1529
17672
1.8
0.31


H15Q, T47A
1530
26578
2.7
0.016


I13V, H15Q, Q82R
1531
76146
7.74
0.655


I13V, H15Q, V89D
1532
28745
2.92
1.331


I13V, S67L, Q82R, V89D
1533
58992
5.99
1.391


I13V, H15Q, Q82R, V89D
1534
49523
5.03
1.419


H15Q, V31M, S67L, Q82R, V89D
1535
67401
6.85
1.37


I13V, H15Q, T47A, Q82R
1536
89126
9.05
0.652


I13V, H15Q, V31A, N45S, Q82R, V89D
1537
68016
6.91
1.327









I13V, T20A, T47A, K65X, Q82R, V89D
1538
Not tested











H15Q, T47A, H69L, Q82R, V89D
1539
65598
6.66
1.44


I13V, H15Q, T47A, H69L, R76G, V89D
1540
54340
5.52
1.719


I12V, I13V, H15Q, T47A, Q82R, V89D
1541
61207
6.22
1.453


I13V, H15Q, R76G, D77N, Q82R, V89D
1542
33079
3.36
0.065


I13V, H15Q, T47A, R76G, V89D
1543
53668
5.45
1.596


I13V, H15Q, T47A, Q82R, V89D
1544
63320
6.43
1.418


I13V, H15Q, T47A, Q82R, V89D
1545
60980
6.2
1.448


I13V, H15Q, I36V, T47A, S67L, V89D
1546
52835
5.37
1.627


H15Q, T47A, K65R, S67L, Q82R, V89D
1547
79692
8.1
1.453


H15Q, L33P, T47A, S67L, P71S, V89D
1548
45726
4.65
1.467


I13V, H15Q, Q72H, R76G, I86T
1549
24450
2.48
1.355


H15Q, T47A, S67L, Q82R, V89D
1550
67962
6.9
1.479


F2L, H15Q, D46E, T47A, Q72H, R76G, Q82R, V89D
1551
23039
2.34
1.045


I13V, H15Q, L33F, T47A, Q82R, V89D
1552
62254
6.32
1.379









I13V, H15Q, T47A, E58G, S67L, Q82R, V89D
1543
Not tested











H15Q, N24S, T47A, Q72H, R76G, V89D
1554
32077
3.26
0.4


I13V, H15Q, E44V, T47A, Q82R, V89D
1555
61005
6.2
1.329


H15Q, N18D, T47A, Q72H, V73A, R76G, I86T, V89D
1556
48317
4.91
0.475


I13V, H15Q, T37A, E44D, S48C, S67L, Q82R, V89D
1557
47605
4.84
1.255


H15Q, L33H, S67L, R76G, Q82R, V89D
1558
62326
6.33
1.507


I13V, H15Q, T47A, Q72H, R76G, I86T
1559
49016
4.98
1.477


H15Q, S39I, E44D, Q72H, V75G, R76G, Q82R, V89D
1560
43713
4.44
0.646


H15Q, T47A, S67L, R76G, Q82R, V89D
1561
71897
7.3
1.539


I13V, H15Q, T47A, S67L, Q72H, R76G, Q82R, V89D
1562
71755
7.29
1.536


Wild Type PD-L2 IgV
1393
9843
1
−0.024


Full length ECD of PD-L2
31
2145
0.22
0.071



(ECD)


Full length ECD of PD-L1 (R&D Systems)

23769
2.41
1.263


Anti-PD-1 monoclonal antibody (nivolumab)

87002
8.84
0.899
















TABLE 26B







Bioactivity Data of PD-L2 variants selected against PD-1 in MLR.













Fold





increase




IFN
over



SEQ ID
gamma
wildtype



NO
levels
PD-L2


PD-L2 mutation(s)
(IgV)
pg/mL
IgV-Fc













H15Q
1487
1817.1
1.32


N24D
1488
1976.3
1.44


E44D
1489
1499.4
1.09


V89D
1490
1168.1
0.85


Q82R, V89D
1491
1617
1.17


E59G, Q82R
1492
1511.3
1.1


S39I, V89D
1493
1314.5
0.95


S67L, V89D
1494
1230.1
0.89


S67L, I85F
1495
1281.9
0.93


S67L, I86T
1496
1020.4
0.74


H15Q, K65R
1497
1510.8
1.1


H15Q, Q72H, V89D
1498
1272.2
0.92


H15Q, S67L, R76G
1499
1426.2
1.04


H15Q, R76G, I85F
1500
1725.7
1.25


H15Q, T47A, Q82R
1501
1317.9
0.96


H15Q, Q82R, V89D
1502
1081.2
0.79


H15Q, C23S, I86T
1503
1847.2
1.34


H15Q, S39I, I86T
1504
1415.2
1.03


H15Q, R76G, I85F
1505
1437.8
1.04


E44D, V89D, W91R
1506
1560.1
1.13


I13V, S67L, V89D
1507
867.5
0.63


H15Q, S67L, I86T
1508
1034.2
0.75


I13V, H15Q, S67L, I86T
1509
1014.4
0.74


I13V, H15Q, E44D, V89D
1510
1384.2
1.01


I13V, S39I, E44D, Q82R, V89D
1511
935.6
0.68


I13V, E44D, Q82R, V89D
1512
1009.5
0.73


I13V, Q72H, R76G, I86T
1513
1953
1.42


I13V, H15Q, R76G, I85F
1514
1528.5
1.11


H15Q, S67L, R76G, I85F
1516
1318.7
0.96


H15Q, T47A, Q72H, R76G, I86T
1517
1599.6
1.16


H15Q, T47A, Q72H, R76G
1518
1462.5
1.06


I13V, H15Q, T47A, Q72H, R76G
1519
1469.8
1.07


H15Q, E44D, R76G, I85F
1520
1391.6
1.01


H15Q, S39I, S67L, V89D
1521
1227
0.89


H15Q, N32D, S67L, V89D
1522
1285.7
0.93


N32D, S67L, V89D
1523
1194
0.87


H15Q, S67L, Q72H, R76G, V89D
1524
1061.2
0.77


H15Q, Q72H, Q74R, R76G, I86T
1525
933.8
0.68


G28V, Q72H, R76G, I86T
1526
1781.6
1.29


I13V, H15Q, S39I, E44D, S67L
1527
1256.9
0.91


E44D, S67L, Q72H, Q82R, V89D
1528
1281.4
0.93


H15Q, V89D
1529
1495.4
1.09


H15Q, T47A
1530
1637.2
1.19


I13V, H15Q, Q82R
1531
1432.9
1.04


I13V, H15Q, V89D
1532
1123
0.82


I13V, S67L, Q82R, V89D
1533
1372.8
1


I13V, H15Q, Q82R, V89D
1534
1596.6
1.16


H15Q, V31M, S67L, Q82R, V89D
1535
1206.5
0.88


I13V, H15Q, T47A, Q82R
1536
1703.3
1.24


I13V, H15Q, V31A, N45S, Q82R, V89D
1537
1723.1
1.25


H15Q, T47A, H69L, Q82R, V89D
1539
1732.5
1.26


I13V, H15Q, T47A, H69L, R76G, V89D
1540
1075.5
0.78


I12V, I13V, H15Q, T47A, Q82R, V89D
1541
1533.2
1.11


I13V, H15Q, R76G, D77N, Q82R, V89D
1542
1187.9
0.86


I13V, H15Q, T47A, R76G, V89D
1543
1253.7
0.91


I13V, H15Q, T47A, Q82R, V89D
1544
1445.5
1.05


I13V, H15Q, T47A, Q82R, V89D
1545
1737
1.26


I13V, H15Q, I36V, T47A, S67L, V89D
1546
1357.4
0.99


H15Q, T47A, K65R, S67L, Q82R, V89D
1547
1335.3
0.97


H15Q, L33P, T47A, S67L, P71S, V89D
1548
1289.1
0.94


I13V, H15Q, Q72H, R76G, I86T
1549
1221
0.89


H15Q, T47A, S67L, Q82R, V89D
1550
1197.1
0.87


F2L, H15Q, D46E, T47A, Q72H, R76G, Q82R, V89D
1551
1170.7
0.85


I13V, H15Q, L33F, T47A, Q82R, V89D
1552
1468.4
1.07


I13V, H15Q, T47A, E58G, S67L, Q82R, V89D
1543
836.1
0.61


H15Q, N24S, T47A, Q72H, R76G, V89D
1554
1091.8
0.79


I13V, H15Q, E44V, T47A, Q82R, V89D
1555
1270.5
0.92


H15Q, N18D, T47A, Q72H, V73A, R76G, I86T, V89D
1556
1065.8
0.77


I13V, H15Q, T37A, E44D, S48C, S67L, Q82R, V89D
1557
1751.7
1.27


H15Q, L33H, S67L, R76G, Q82R, V89D
1558
1502
1.09


I13V, H15Q, T47A, Q72H, R76G, I86T
1559
1088.1
0.79


H15Q, S39I, E44D, Q72H, V75G, R76G, Q82R, V89D
1560
940.9
0.68


H15Q, T47A, S67L, R76G, Q82R, V89D
1561
1097.8
0.8


I13V, H15Q, T47A, S67L, Q72H, R76G, Q82R, V89D
1562
1559.6
1.13


Wild Type PD-L2 IgV
1393
1376.8
1


Full length ECD of PD-L2
31
1173.2
0.85



(ECD)


Full length ECD of PD-L1
2118
2190.9
1.59



(ECD)


Nivolumab (anti-PD-1)

418.9
0.3
















TABLE 27A







Variant CD80 Binding to HEK293 Cells Transfected with CTLA4, CD28 or PD-L1














CTLA4
CD28
PD-L1

















SEQ ID
MFI at
Fold
MFI
Fold
MFI at
Fold
Ratio of



NO
66.6
change
at 66.6
change
22.2
change
CTLA4:


CD80 mutation(s)
(IgV)
nM
to WT
nM
to WT
nM
to WT
CD28












L70P
579
Not tested


130F/L70P
580
Not tested















Q27H/T41S/A71D
581
368176
2.3
25051
1.01
24181
N/A
14.7


130T/L70R
582
2234
0.0
2596
0.10
5163
N/A
0.9


T13R/C16R/L70Q/A71D
583
197357
1.2
16082
0.65
9516
N/A
12.3


T571
584
393810
2.4
23569
0.95
3375
N/A
16.7


M431/C82R
585
3638
0.0
3078
0.12
7405
N/A
1.2


V22L/M38V/M47T/A71D/
586
175235
1.1
3027
0.12
6144
N/A
57.9


L85M










130V/T571/L70P/A71D/
587
116085
0.7
10129
0.41
5886
N/A
11.5


A91T










V221/L70M/A71D
588
163825
1.0
22843
0.92
33404
N/A
7.2


N55D/L70P/E77G
589
Not tested








T57A/I69T
590
Not tested








N55D/K86M
591
3539
0.0
3119
0.13
5091
N/A
1.1


L72P/T791
592
50176
0.3
3397
0.14
6023
N/A
14.8


L70P/F92S
593
4035
0.0
2948
0.12
6173
N/A
1.4


T79P
594
2005
0.0
2665
0.11
4412
N/A
0.8


E35D/M471/L65P/D90N
595
4411
0.0
2526
0.10
4034
N/A
1.7


L25S/E35D/M471/D90N
596
61265
0.4
4845
0.20
20902
N/A
12.6


Q27X*/S44P/I67T/P74S/
597
195637
1.2
17524
0.71
17509
N/A
11.2


E81G/E95D










A71D
598
220090
1.4
16785
0.68
29642
N/A
13.1


T13A/Q27X*/I61N/A71D
599
195061
1.2
17519
0.71
21717
N/A
11.1


E81K/A91S
600
98467
0.6
3309
0.13
44557
N/A
29.8


A12V/M47V/L70M
601
81616
0.5
7400
0.30
31077
N/A
11.0


K34E/T41A/L72V
602
88982
0.6
3755
0.15
35293
N/A
23.7


T41S/A71D/V84A
603
103010
0.6
5573
0.22
83541
N/A
18.5


E35D/A71D
604
106069
0.7
18206
0.73
40151
N/A
5.8


E35D/M47I
605
353590
2.2
14350
0.58
149916
N/A
24.6


K36R/G78A
606
11937
0.1
2611
0.11
5715
N/A
4.6


Q33E/T41A
607
8292
0.1
2442
0.10
3958
N/A
3.4


M47V/N48H
608
207012
1.3
14623
0.59
145529
N/A
14.2


M47LN68A
609
74238
0.5
13259
0.53
11223
N/A
5.6


S44P/A71D
610
8839
0.1
2744
0.11
6309
N/A
3.2


Q27H/M43I/A71D/R73S
611
136251
0.8
12391
0.50
8242
N/A
11.0


E35D/T57I/L70Q/A71D
613
121901
0.8
21284
0.86
2419
N/A
5.7


M47I/E88D
614
105192
0.7
7337
0.30
97695
N/A
14.3


M42I/I61V/A71D
615
54478
0.3
6074
0.24
4226
N/A
9.0


P51A/A71D
616
67256
0.4
4262
0.17
5532
N/A
15.8


H18Y/M47I/T57I/A71G
617
136455
0.8
20081
0.81
13749
N/A
6.8


V20I/M47V/T57I/V84I
618
183516
1.1
26922
1.08
3583
N/A
6.8


WT
578
161423
1.0
24836
1.00
Not
N/A
6.5








tested







*Stop codon at indicated position













TABLE 27B







Variant CD80 Binding to HEK293 Cells Transfected with CTLA4, CD28 or PD-L1














CTLA4
CD28
PD-L1

















SEQ ID
MFI at
Fold
MFI
Fold
MFI at
Fold
Ratio of



NO
66.6
change
at 66.6
change
22.2
change
CTLA4:


CD80 mutation(s)
(IgV)
nM
to WT
nM
to WT
nM
to WT
CD28


















V20I/M47V/A71D
619
149937
7.23
15090
9.33
9710
5.48
9.9


A71D/L72V/E95K
620
140306
6.77
6314
3.90
8417
4.75
22.2


V22L/E35G/A71D/L72P
621
152588
7.36
8150
5.04
1403
0.79
18.7


E35D/A71D
622
150330
7.25
14982
9.26
13781
7.77
10.0


E35D/I67L/A71D
623
146087
7.04
11175
6.91
9354
5.28
13.1


T13R/M42V/M47I/A71D
625
108900
5.25
16713
10.33
1869
1.05
6.5


E35D
626
116494
5.62
3453
2.13
25492
14.38
33.7


E35D/M47I/L70M
627
116531
5.62
14395
8.90
49131
27.71
8.1


E35D/A71/L72V
628
134252
6.47
11634
7.19
13125
7.40
11.5


E35D/M43L/L70M
629
102499
4.94
3112
1.92
40632
22.92
32.9


A26P/E35D/M43I/L85Q/
630
83139
4.01
5406
3.34
9506
5.36
15.4


E88D










E35D/D46V/L85Q
631
85989
4.15
7510
4.64
38133
21.51
11.4


Q27L/E35D/M47I/T57I/
632
59793
2.88
14011
8.66
1050
0.59
4.3


L70Q/E88D










Q27H/E35G/A71D/L72P/
624
85117
4.10
10317
6.38
1452
0.82
8.3


T791










M47V/169F/A71D/V831
633
76944
3.71
15906
9.83
3399
1.92
4.8


E35D/T57A/A71D/L85Q
634
85724
4.13
3383
2.09
1764
0.99
25.3


H18Y/A26T/E35D/A71D/
635
70878
3.42
6487
4.01
8026
4.53
10.9


L85Q










E35D/M47L
636
82410
3.97
11508
7.11
58645
33.08
7.2


E23D/M42V/M43I/158V/
637
37331
1.80
10910
6.74
2251
1.27
3.4


L70R










V68M/L70M/A71D/E95K
638
56479
2.72
10541
6.51
38182
21.53
5.4


N55I/T57I/I69F
639
2855
0.14
1901
1.17
14759
8.32
1.5


E35D/M43I/A71D
640
63789
3.08
6369
3.94
27290
15.39
10.0


T41S/T57I/L70R
641
59844
2.89
4902
3.03
19527
11.01
12.2


H18Y/A71D/L72P/E88V
642
68391
3.30
8862
5.48
1085
0.61
7.7


V20I/A71D
643
60323
2.91
10500
6.49
3551
2.00
5.7


E23G/A26S/E35D/T62N/
644
59025
2.85
5484
3.39
10662
6.01
10.8


A71D/L72V/L85M










Al2T/E24D/E35D/D46V/
645
63738
3.07
7411
4.58
1221
0.69
8.6


I61V/L72P/E95V










V22L/E35D/M43L/A71G/
646
2970
0.14
1498
0.93
1851
1.04
2.0


D76H










E35G/K54E/A71D/L72P
647
71899
3.47
3697
2.29
1575
0.89
19.4


L70Q/A71D
648
45012
2.17
18615
11.50
1692
0.95
2.4


A26E/E35D/M47L/L85Q
649
40325
1.94
2266
1.40
55548
31.33
17.8


D46E/A71D
650
69674
3.36
16770
10.36
22777
12.85
4.2


Y31H/E35D/T41SN68L/
651
3379
0.16
2446
1.51
18863
10.64
1.4


K93R/R94W










CD80 IgV Fc
578
20739
1.00
1618
1.00
1773
1.00
12.8



(IgV)









CD80 ECD Fc
 28
72506
3.50
3072
1.90
4418
2.49
23.6



(ECD)
















Example 12
Generation of Secreted Immunomodulatory Protein

To generate a PD-L2 secreted immunomodulatory protein (SIP), DNA encoding exemplary SIPs was obtained as gene blocks from Integrated DNA Technologies (Coralville, USA) and then cloned by Gibson assembly (New England Biolabs Gibson assembly kit) into a modified version of pRRL vector (Dull et al., (1998) J Virol, 72(11): 8463-8471) between restriction sites downstream of MND promoter to remove GFP. Exemplary SIP constructs were generated to encode a protein set forth in SEQ ID NO: 1571-1573, including the signal peptide. In this exemplary Example, the constructs were generated to additionally include a tag moiety. The gene blocks had the following structure in order: 39 base pair overlap with pRRL prior to first restriction site-first restriction site-GCCGCCACC (Kozak); complete ORF encoding PD-L2 IgV wildtype amino acid sequence set forth in SEQ ID NO:1393 or variant PD-L2 IgV set forth in SEQ ID NO:1535 (H15Q, V31M, S67L, Q82R, V89D), SEQ ID NO:1547 (H15Q, T47A, K65R, S67L, Q82R, V89D) or SEQ ID NO: 1561 (H15Q, T47A, S67L, R76G, Q82R, V89D), also including in all cases the PD-L2 signal peptide MIFLLLMLSLELQLHQIAA as set forth in SEQ ID NO: 1567; DNA encoding Avitag as set forth in SEQ ID NO:1568 (GLNDIFEAQKIEWHE); DNA encoding His tag as set forth in SEQ ID NO: 1569 (HHHHHH); TAA stop codon; second restriction site-41 base pair overlap with pRRL beyond second restriction site.


To prepare lentiviral vectors, 3×106 HEK293 cells were plated per 100 mm dish. On the next day, 4.5 μg of P-Mix (3 μg of PAX2 and 1.5 μg of pMD2G) was added to 6 μg of DNA encoding the SIPs constructs in a 5 mL polypropylene tube. Diluent buffer (10 mM HEPES/150 mM NaCl pH7.05/1 L TC grade H20) was added to the tube to bring up the total volume of 500 μL. To the diluent DNA(PEI:total DNA 4:1), 42 μL of PEI (1 μg/μL) was added and mixed by vortexing. The mixture was incubated at room temperature for 10 minutes and cells were prepared by aspirating medium from the dish gently without disturbing the adherent cells, then replaced with 6 mL of Opti-MEM (1×). DNA/PEI mixture was then added to the dish and incubated at 37° C. for 24 hours. After 24 hours, media was aspirated from the dishes and replaced with 10 mL of fresh DMEM media and then incubated at 37° C. Viral supernatant was collected after 48 hours using a syringe attached to a 0.45 μm filter PES to remove cells and debris from the culture (Thermo Scientific Nalgene Syringe Filter). A separate lentiviral vector stock also was prepared encoding an anti-CD19 CAR (containing an anti-CD19 scFv, a hinge and transmembrane domain derived from CD8, and a CD3zeta signaling domain) substantially as described. The exemplary anti-CD19 CAR used is set forth in SEQ ID NO: 2160 (encoded by the sequence in set forth in SEQ ID NO: 2161) containing the scFv set forth in SEQ ID NO:1576, the CD8-derived hinge and transmembrane domain set forth in SEQ ID NO: 1574, and the CD3zeta set forth in SEQ ID NO:1575.


Pan T-cells were transduced with the viral vectors encoding the PD-L2 SIPs. T-cells were thawed and activated with anti-CD3/anti-CD28 beads (Dynal) at a 1:1 ratio. The T-cells (1×106 cells) were mixed with 1 mL total lentiviral vector supernatant containing equal volume (0.5 mL each) of the lentiviral vector supernatant encoding the indicated PD-L2 SIPs and a lentiviral vector supernatant encoding the anti-CD19 CAR. As a control, cells were transduced only with the lentiviral vector encoding the anti-CD19 CAR or were transduced with mock vector control. Transduction was performed in the presence of 10 μg/mL polybrene and 50 IU/mL IL-2. Cells were spun down at 2500 rpm for 60 min at 30° C. After 24 hours, 3 mL of Xvivol5 plus media and IL2 was added to each well. The cells were fed every two days with fresh media and cytokines.


Transduction also was carried out on HEK-293 cells, which were resuspended at 2×105 cells with 1 mL of the lentiviral supernatant encoding the indicated PD-L2 SIPs. To the cells, 3 mL of DMEM media was added and cells were fed every two days with fresh media.


To assess the amount of secreted SIP, a cell-based assay was performed to assess binding of the secretable variant PD-L2 to PD-1. Approximately, 100,000 PD-1+Jurkat cells were plated per well in the presence of 50 μL of culture supernatant containing PD-L2 SIP obtained from transduced cells above and incubated at 4° C. for 30 minutes. To generate a standard curve, 50 μL of the respective variant PD-L2 protein was added to PD-1+Jurkat cells at 10 μg/mL, 3 μg/mL, 1 μg/mL, 0.3 μg/mL, 0.1 μg/mL, and 0 μg/mL and also incubated at 4° C. for 30 minutes. Cells were washed and 50 μL of anti-his-APC were added (1:50) and this was incubated at 4° C. for 30 minutes. Surface bound PD-L2 protein was detected by flow cytometry and the concentration of SIP in the supernatant sample was determined by comparison to the standard curve. As shown in FIGS. 2A and 2B, SIP proteins were detected in the supernatant of transduced T cells and transduced HEK293 cell, but were not detected from supernatant samples from mock transduced or cells transduced without SIPs.


Example 13
Assessment of Proliferation and Bioactivity of Pan T Cells Transduced with PD-L2 SIP

Pan T-cells were transduced essentially as described in Example 12 with the viral vectors encoding the PD-L2 SIPs. T-cells were thawed and activated with anti-CD3/anti-CD28 beads (Dynal) at a 1:1 ratio. The T-cells (1×106 cells) were mixed with 1 mL total lentiviral vector supernatant containing equal volume (0.5 mL each) of the lentiviral vector supernatant encoding the indicated PD-L2 SIPs and a lentiviral vector supernatant encoding the anti-CD19 CAR. As a control, cells were transduced only with the lentiviral vector encoding the anti-CD19 CAR or were transduced with mock vector control. Transduction was performed in the presence of 10 μg/mL polybrene and 50 IU/mL IL-2. Cells were spun down at 2500 rpm for 60 min at 30° C. After 24 hours, 3 mL of Xvivol5 plus media and IL2 was added to each well. The cells were fed every two days with fresh media and cytokines.


At 14 days after activation, cells were re-stimulated with Nalm6 cells that had been transduced with a lenti-viral vector to provide expression of PD-L1 (Nalm6 PDL1+). Transduced T cells were labeled with Cell Trace Far Red and proliferation was measured at day 5 by determining the fraction of the cells that showed dilution of the dye. Results for the proliferation studies for T cells transduced with exemplary tested variant PD-L2 SIP are shown in FIG. 3A.


Levels of IFN-gamma released into the supernatant were measured by ELISA on day 5 after re-stimulation. Results for the bioactivity studies for T cells transduced with exemplary tested variant PD-L2 SIP are shown in FIG. 3B. The T cells transduced with PD-L2 variant SIP are identified with reference to the amino acid substitutions in the IgV of PD-L2 with reference to positions corresponding to positions of the unmodified (wildtype) PD-L2 ECD sequence set forth in SEQ ID NO:31. As shown in FIGS. 3A and 3B, proliferation and improved activities to increase immunological activity was observed.


A similar study was carried out except that T cells were co-transduced with the anti-CD19 CAR and a lentiviral vector encoding a SIP, either a variant PD-L2 IgV or wild-type (WT) PD-L2 IgV. Following stimulation of transduced T cells with Nalm6 PDL1+ cells as described above, the proliferation of T cells was measured at day 3 by determining the fraction of the cells that showed dilution of the dye. As shown in FIG. 3C, cells engineered with the variant PD-L2 SIP improved proliferation compared to proliferation of T cells only expressing the CAR, and the improved proliferation was also greater than the proliferation of T cells expressing the wild-type PD-L2 SIP.


Example 14
Generation of Secreted Immunomodulatory Protein and Assessment of Proliferation of Pan T Cells Transduced with PD-L1 SIP

To generate a PD-L1 secreted immunomodulatory protein (SIP), DNA encoding exemplary SIPs was obtained as gene blocks from Integrated DNA Technologies (Coralville, USA) and then cloned by Gibson assembly (New England Biolabs Gibson assembly kit) into a modified version of pRRL vector (Dull et al., (1998) J Virol, 72(11): 8463-8471) between restriction sites downstream of MND promoter to remove GFP. Exemplary PD-L1 SIP constructs were generated to encode a protein set forth in SEQ ID NO: 2155-2156, including the signal peptide. In this exemplary Example, the constructs were generated to additionally include a tag moiety. The gene blocks had the following structure in order: 39 base pair overlap with pRRL prior to first restriction site-first restriction site-GCCGCCACC (Kozak); complete ORF encoding PD-L1 IgV wildtype amino acid sequence set forth in SEQ ID NO: 1332 or variant PD-L1 IgV set forth in SEQ ID NO: 1326 (D43G/N45D/L56Q/V58A/G101G-ins (G101GG), also including in all cases the signal peptide MGSTAILALLLAVLQGVSA as set forth in SEQ ID NO: 2157; DNA encoding Flag-tag as set forth in SEQ ID NO:2154 (DYKDDDDK); DNA encoding His tag as set forth in SEQ ID NO: 1569 (HHHHHH); TAA stop codon; second restriction site-41 base pair overlap with pRRL beyond second restriction site. For comparison, a SIP encoding wild-type PD-L1 also was assessed.


To prepare lentiviral vectors, 3×106 HEK293 cells were plated per 100 mm dish. On the next day, 4.5 μg of P-Mix (3 μg of PAX2 and 1.5 μg of pMD2G) was added to 6 μg of DNA encoding the SIPs constructs in a 5 mL polypropylene tube. Diluent buffer (10 mM HEPES/150 mM NaCl pH7.05/1 L TC grade H20) was added to the tube to bring up the total volume of 500 μL. To the diluent DNA(PEI:total DNA 4:1), 42 μL of PEI (1 μg/μL) was added and mixed by vortexing. The mixture was incubated at room temperature for 10 minutes and cells were prepared by aspirating medium from the dish gently without disturbing the adherent cells, then replaced with 6 mL of Opti-MEM (1×). DNA/PEI mixture was then added to the dish and incubated at 37° C. for 24 hours. After 24 hours, media was aspirated from the dishes and replaced with 10 mL of fresh DMEM media and then incubated at 37° C. Viral supernatant was collected after 48 hours using a syringe attached to a 0.45 μm filter PES to remove cells and debris from the culture (Thermo Scientific Nalgene Syringe Filter). A separate lentiviral vector stock also was prepared encoding an anti-CD19 CAR (containing an anti-CD19 scFv, a hinge and transmembrane domain derived from CD8, and a CD3zeta signaling domain) substantially as described. The exemplary anti-CD19 CAR used is set forth in SEQ ID NO: 2160 (encoded by the sequence in set forth in SEQ ID NO: 2161) containing the scFv set forth in SEQ ID NO:1576, the CD8-derived hinge and transmembrane domain set forth in SEQ ID NO: 1574, and the CD3zeta set forth in SEQ ID NO:1575.


T-cells were thawed and activated with anti-CD3/anti-CD28 beads (Dynal) at a 1:1 ratio. The T-cells (1×106 cells) were mixed with 1 mL total lentiviral vector supernatant containing equal volume (0.5 mL each) of the lentiviral vector supernatant encoding the indicated PD-L1 SIP (D43G/N45D/L56Q/V58A/G101GG or wildtype) and a lentiviral vector supernatant encoding the anti-CD19 CAR. As a control, cells were transduced only with the lentiviral vector encoding the anti-CD19 CAR or were transduced with mock vector control. Transduction was performed in the presence of 10 μg/mL polybrene and 50 IU/mL IL-2. Cells were spun down at 2500 rpm for 60 min at 30° C. After 24 hours, 3 mL of Xvivol5 plus media and IL2 was added to each well. The cells were fed every two days with fresh media and cytokines.


At 14 days after activation, cells were re-stimulated with Nalm6 cells that had been transduced with a lenti-viral vector to provide expression of PD-L1 (Nalm6 PDL1+). Transduced T cells were labeled with Cell Trace Far Red and proliferation was measured at day 3 by determining the fraction of the cells that showed dilution of the dye. Results for the proliferation studies for T cells transduced with exemplary tested variant PD-L1 SIP are shown in FIG. 5.


Example 15
Detection of Secreted Immunomodulatory Protein in Supernatant of Transduced Cells

A cell-based assay was employed to detect the presence of SIPs in culture supernatant. HEK-293 cells were transduced with a lentiviral vector encoding exemplary SIPs, variant PD-L1 IgV (D43G/N45D/L56Q/V58A/G101G-ins(G101GG) set forth in SEQ ID NO:1326), wild-type PD-L1 IgV (set forth in SEQ ID NO:1332), variant PD-L2 IgV (H15Q/T47A/K65R/S67L/Q82R/V89D set forth in SEQ ID NO:1547) or wild-type PD-L2 IgV (set forth in SEQ ID NO:1393), as described in Examples 12 and 14. Four days after transduction, supernatant was collected. Approximately 50 μL of supernatant, diluted 1:2, 1:4, 1:8, 1:16, 1:32, 1:64, 1:128 or neat, was added to 1×105 K562 cells transduced to express PD-1 (K562 PD-1+) in a 96-well round bottom plate, and incubated at 4° C. for 30 minutes. To generate a standard curve, PD-L2 his tag protein, diluted to 10,000 pg/mL, 1,000 pg/mL, 100 pg/mL, 10 pg/mL, 1 pg/mL and 0.1 pg/mL and also incubated with K562 PD-1+ cells at 4° C. for 30 minutes. Cells were washed and 50 μL of anti-his-APC were added (1:50) and this was incubated at 4° C. for 30 minutes. Surface bound PD-L2 protein was detected by flow cytometry and the concentration of SIP in the supernatant sample was determined by comparison to the standard curve. As shown in FIG. 6, the variant PD-L1 and variant PD-L2 SIPs, but not the wild-type proteins, were detected in the supernatant of cells.


The cell-based assay described above was used to assess the presence of PD-L2 SIP in supernatant of T cells following culture with antigen-expressing target cells. T cells were activated and transduced with a lentiviral vector encoding an anti-CD19 CAR and a lentiviral vector encoding either variant PD-L2 H15Q/T47A/K65R/S67L/Q82R/V89D set forth in SEQ ID NO:1547) or wild-type PD-L2 IgV (set forth in SEQ ID NO:1393), as described in Example 9. At 14 days after activation, transduced T cells were cultured with CD19-expressing Nalm6 PD-L1+ cells or Raji cells. At days 3, 6, 9 and 12 after initiation of culture of T cells, supernatant was collected and the presence of PD-L2 was detected as described above. The variant PD-L2 SIP, but not the wild-type PD-L2 SIP, was detected in supernatant following stimulation with target antigen-expressing cells using this assay. This result may be due to the wild-type PD-L2 SIP not having a high enough affinity to bind to the K562/PD-1+ cells. The variant PD-L2 SIP was detected at substantially higher levels in supernatant of T cells stimulated with Raji cells compared to Nalm6 PD-L1 cells, and the level of variant PD-L2 SIP in the supernatant following stimulation with Raji cells was sustained throughout the time course of this study.


While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims
  • 1. An engineered immune cell comprising a nucleic acid molecule that encodes an immunomodulatory protein, wherein: the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain of an IgSF family member, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; andthe engineered immune cell expresses and secretes the immunomodulatory protein.
  • 2. The engineered immune cell of claim 1, wherein the immunomodulatory protein does not comprise a transmembrane domain.
  • 3. The engineered immune cell of claim 1 or claim 2, wherein the nucleic acid molecule comprises a sequence encoding a secretory signal peptide operably linked to the sequence encoding the immunomodulatory protein.
  • 4. The engineered immune cell of claim 3, wherein the signal peptide is the native signal peptide from the corresponding wild-type IgSF member.
  • 5. The engineered immune cell of claim 3, wherein the signal peptide is a non-native signal sequence.
  • 6. The engineered immune cell of claim 3 or claim 5, wherein the signal peptide is an IgG-kappa signal peptide, an IL-2 signal peptide, or a CD33 signal peptide.
  • 7. The engineered immune cell of any of claims 1-6, wherein the nucleic acid molecule further comprises at least one promoter operably linked to control expression of the immunomodulatory protein.
  • 8. The engineered immune cell of claim 7, wherein the promoter is a constitutively active promoter.
  • 9. The engineered immune cell of claim 7, wherein the promoter is an inducible promoter.
  • 10. The engineered immune cell of claim 7 or claim 9, wherein the promoter is responsive to an element responsive to T-cell activation signaling.
  • 11. The engineered immune cell of any of claims 7, 9, and 10, wherein the promoter comprises a binding site for NFAT or a binding site for NF-κB.
  • 12. The engineered immune cell of any one of claims 1-11, wherein the immune cell is a lymphocyte.
  • 13. The engineered immune cell of claim 12, wherein the lymphocyte is a T cell, a B cell or an NK cell.
  • 14. The engineered immune cell of any of claims 1-13, wherein the immune cell is a T cell.
  • 15. The engineered immune cell of claim 14, wherein the T cell is CD4+ or CD8+.
  • 16. The engineered immune cell of any of claims 1-15, wherein the immune cell is an antigen presenting cell.
  • 17. The engineered immune cell of any of claims 1-16, wherein the immune cell is a primary cell obtained from a subject.
  • 18. The engineered immune cell of claim 17, wherein the subject is a human subject.
  • 19. The engineered immune cell of any of claims 1-18, wherein the at least one affinity-modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the binding affinity of the wild-type IgSF domain for the at least one cell surface cognate binding partner.
  • 20. The engineered immune cell of any of claims 1-19, wherein the wild-type IgSF domain is from an IgSF family member of a family selected from B7 family, Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family, or Killer-cell immunoglobulin-like receptors (KIR) family.
  • 21. The engineered immune cell of any of claims 1-20, wherein the wild-type IgSF domain is from an IgSF member selected from the group consisting of PD-L1, PD-L2, CD80, CD86, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAG3, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R, NKp30, VISTA, VSIG3, and VSIG8.
  • 22. The engineered immune cell of any of claims 1-21, wherein the wild-type IgSF domain is a human IgSF domain.
  • 23. The engineered immune cell of any of claims 1-22, wherein the wild-type IgSF domain is from an IgSF member that is a ligand of an stimulatory receptor, wherein the stimulatory receptor comprises a costimulatory signaling domain.
  • 24. The engineered immune cell of any of claims 1-23, wherein the at least one cell surface cognate binding partner is a stimulatory receptor expressed on a T-cell and the at least one affinity-modified IgSF domain has increased binding affinity to the stimulatory receptor compared to the binding affinity of the wild-type IgSF domain to the stimulatory receptor.
  • 25. The engineered immune cell of claim 23 or claim 24, wherein the stimulatory receptor is CD28, ICOS, or CD226.
  • 26. The engineered immune cell of any of claims 1-22, wherein the wild-type IgSF domain is from an IgSF member that is a ligand of an inhibitory receptor, wherein the inhibitory receptor comprising an ITIM signaling domain.
  • 27. The engineered immune cell of any one of claims 1-22 and 26, wherein the at least one cell surface cognate binding partner is an inhibitory receptor expressed on a T-cell and the at least one affinity-modified IgSF domain has increased binding affinity to the inhibitory receptor compared to the binding affinity of the wild-type IgSF domain to the inhibitory receptor.
  • 28. The engineered immune cell of claim 25 or claim 26, wherein: the inhibitory receptor is PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of a ligand of PD-1, CTLA-4, LAG3, TIGIT, TIM-3, BTLA, VSIG3, or VSIG8, respectively; orthe ligand of the inhibitory receptor is PD-L1, PD-L2, B7-1, B7-2, MHC class II, CD155, CD112, CEACAM-1, GAL9 or VISTA and the at least one affinity-modified IgSF domain is an affinity-modified IgSF domain of PD-L1, PD-L2, B7-1, B7-2, MHC class II, CD155, CD112, CEACAM-1, GAL9 or VISTA, respectively.
  • 29. The engineered immune cell of any of claims 26-28, wherein the inhibitory receptor is PD-1 and the at least one affinity-modified IgSF domain is an affinity-modified IgSF of PD-L1 or is an affinity-modified IgSF of PD-L2.
  • 30. The engineered immune cell of any of claims 1-28, wherein the affinity modified IgSF domain is an affinity modified CD155 IgSF domain or an affinity modified CD112 IgSF domain and the at least one cell surface cognate binding partner is CD226, TIGIT or CD112R.
  • 31. The engineered immune cell of any of claims 1-30, wherein the affinity modified IgSF domain differs by no more than ten amino acid substitutions from the wildtype IgSF domain.
  • 32. The engineered immune cell of any of claims 1-30, wherein the affinity-modified IgSF domain differs by no more than five amino acid substitutions from the wildtype IgSF domain.
  • 33. The engineered immune cell of any of claims 1-32, wherein the one or more affinity-modified IgSF domain is or comprises an affinity modified IgV domain, an affinity modified IgC1 domain, or an affinity modified IgC2 domain, or is a specific binding fragment thereof comprising the one or more amino acid substitutions.
  • 34. The engineered immune cell of any of claims 1-33, wherein the immunomodulatory protein further comprises one or more non-affinity modified IgSF domains.
  • 35. The engineered cell of any of claims 1-34, wherein the engineered immune cell further comprises a chimeric antigen receptor (CAR) or an engineered T-cell receptor (TCR).
  • 36. An infectious agent, comprising a nucleic acid molecule that encodes an immunomodulatory protein, wherein: the immunomodulatory protein comprises at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitutions in a wild-type IgSF domain of an IgSF family member, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; andthe infectious agent expresses and secretes the immunomodulatory protein.
  • 37. The infectious agent of claim 36, wherein the immunomodulatory protein does not comprise a transmembrane domain.
  • 38. An infectious agent, comprising a nucleic acid molecule encoding a transmembrane immunomodulatory protein (TIP), wherein the TIP comprises: (i) an ectodomain comprising at least one non-immunoglobulin affinity-modified immunoglobulin superfamily (IgSF) domain comprising one or more amino acid substitution(s) in a wild-type IgSF domain of an IgSF family member, wherein the at least one affinity-modified IgSF domain specifically binds at least one cell surface cognate binding partner of the wild-type IgSF domain; and(ii) a transmembrane domain.
  • 39. The infectious agent of claim 38, wherein the transmembrane domain is the native transmembrane domain from the corresponding wild-type IgSF member.
  • 40. The infectious agent of claim 38 or claim 39, wherein the transmembrane domain is not the native transmembrane domain from the corresponding wild-type IgSF member.
  • 41. The infectious agent of claim 40, wherein the transmembrane domain is a transmembrane domain derived from CD8.
  • 42. The infectious agent of any of claims 36-41, wherein the at least one affinity-modified IgSF domain has increased binding affinity to the at least one cell surface cognate binding partner compared with the binding affinity of the wild-type IgSF domain for the at least one cell surface cognate binding partner.
  • 43. The infectious agent of any of claims 36-42, wherein the wild-type IgSF domain is from an IgSF family member of a family selected from B7 family, Signal-Regulatory Protein (SIRP) Family, Triggering Receptor Expressed On Myeloid Cells Like (TREML) Family, Carcinoembryonic Antigen-related Cell Adhesion Molecule (CEACAM) Family, Sialic Acid Binding Ig-Like Lectin (SIGLEC) Family, Butyrophilin Family, CD28 family, V-set and Immunoglobulin Domain Containing (VSIG) family, V-set transmembrane Domain (VSTM) family, Major Histocompatibility Complex (MHC) family, Signaling lymphocytic activation molecule (SLAM) family, Leukocyte immunoglobulin-like receptor (LIR), Nectin (Nec) family, Nectin-like (NECL) family, Poliovirus receptor related (PVR) family, Natural cytotoxicity triggering receptor (NCR) family, T cell immunoglobulin and mucin (TIM) family or Killer-cell immunoglobulin-like receptors (KIR) family.
  • 44. The infectious agent of any of claims 36-43, wherein the wild-type IgSF domain is from an IgSF member selected from PD-L1, PD-L2, CD80, CD86, ICOS Ligand, B7-H3, B7-H4, CD28, CTLA4, PD-1, ICOS, BTLA, CD4, CD8-alpha, CD8-beta, LAGS, TIM-3, CEACAM1, TIGIT, PVR, PVRL2, CD226, CD2, CD160, CD200, CD200R or NKp30.
  • 45. The infectious agent of any of claims 36-44, wherein the wild-type IgSF domain is a human IgSF member.
  • 46. The infectious agent of any of claims 36-45, wherein the affinity-modified IgSF domain differs by no more than ten amino acid substitutions or no more than five amino acid substitutions from the wildtype IgSF domain.
  • 47. The infectious agent of any of claims 36-46, wherein the affinity-modified IgSF domain is or comprises an affinity-modified IgV domain, an affinity-modified IgC1 domain or an affinity-modified IgC2 domain or is a specific binding fragment thereof comprising the one or more amino acid substitutions.
  • 48. The infectious agent of any of claims 36-47, wherein the infectious agent is a bacteria or a virus.
  • 49. The infectious agent of claim 48, wherein infectious agent is a virus and the virus is an oncolytic virus.
  • 50. The infectious agent of claim 49, wherein the oncolytic virus is an adenovirus, adeno-associated virus, herpes virus, Herpes Simplex Virus, Vesticular Stomatic virus, Reovirus, Newcastle Disease virus, parvovirus, measles virus, vesticular stomatitis virus (VSV), Coxsackie virus or a Vaccinia virus.
  • 51. The infectious agent of claim 48, wherein the infectious agent is a virus and the virus specifically targets dendritic cells (DCs) and/or is dendritic cell-tropic.
  • 52. The infectious agent of claim 48 or claim 51, wherein the virus is a lentiviral vector that is pseudotyped with a modified Sindbis virus envelope product.
  • 53. The infectious agent of any of claims 36-52, further comprising a nucleic acid molecule encoding a further gene product that results in death of a target cell or that augments or boosts an immune response.
  • 54. The infectious agent of claim 53, wherein the further gene product is selected from an anticancer agent, anti-metastatic agent, an antiangiogenic agent, an immunomodulatory molecule, an immune checkpoint inhibitor, an antibody, a cytokine, a growth factor, an antigen, a cytotoxic gene product, a pro-apoptotic gene product, an anti-apoptotic gene product, a cell matrix degradative gene, genes for tissue regeneration or a reprogramming human somatic cells to pluripotency.
  • 55. A pharmaceutical composition comprising the engineered immune cell of any of claims 1-35 or the infectious agent of any of claims 36-54 and a pharmaceutically acceptable carrier.
  • 56. The pharmaceutical composition of claim 55 that is sterile.
  • 57. A method of introducing an immunomodulatory protein into a subject, comprising administering the engineered cell of any one of claims 1-35, the infectious agent of any of claims 36-54 and a pharmaceutical composition of claim 55 or claim 56 to the subject.
  • 58. A method of modulating an immune response in a subject, comprising administering the engineered immune cell of any one of claims 1-35, an infectious agent of any of claims 36-54 or a pharmaceutical composition of claim 55 or claim 56 to the subject.
  • 59. The method of claim 58, wherein modulating the immune response treats a disease or disorder in the subject.
  • 60. The method of claim 58 or claim 59, wherein the modulating the immune response comprises increasing the immune response.
  • 61. The method of any of claim 59 or claim 60, wherein the disease or disorder is a cancer.
  • 62. The method of claim 58 or claim 59, wherein the modulating the immune response comprises decreasing the immune response.
  • 63. The method of claim 59 or claim 62, wherein the disease or disorder is an inflammatory disease or condition.
  • 64. The method of any of claims 58-63, wherein the subject is human.
  • 65. A composition for use in the treatment of a disease or disorder, wherein the composition comprises the engineered immune cell of any of claims 1-35 or the infectious agent of any of claims 36-54 and a pharmaceutically acceptable carrier.
  • 66. Use of a composition for the manufacture of a medicament for the treatment of a disease or disorder, wherein the composition comprises the engineered immune cell of any of claims 1-35 or the infectious agent of any of claims 36-54 and a pharmaceutically acceptable carrier.
  • 67. The composition of claim 65 or the use of claim 66, wherein the composition modulates an immune response.
  • 68. The composition or use of claim 67, wherein the modulating the immune response comprises increasing the immune response.
  • 69. The composition or use of any of claims 65-68, wherein the disease or disorder is a cancer.
  • 70. The composition or use of claim 67, wherein the modulating the immune response comprises decreasing the immune response.
  • 71. The composition or use of any of claims 65-68 and 70, wherein the disease or disorder is an inflammatory disease or condition.
  • 72. The composition or use of any of claims 65-71, wherein the subject is human.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority from U.S. provisional application No. 62/410,827, filed Oct. 20, 2016, entitled “Secretable Variant Immunomodulatory Proteins and Engineered Cell Therapy,” U.S. provisional application No. 62/475,210 filed Mar. 22, 2017, entitled “Secretable Variant Immunomodulatory Proteins and Engineered Cell Therapy,” and U.S. provisional application No. 62/537,921 filed Jul. 27, 2017, entitled “Secretable Variant Immunomodulatory Proteins and Engineered Cell Therapy,” the contents of each of which are incorporated by reference in their entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2017/057743 10/20/2017 WO 00
Provisional Applications (3)
Number Date Country
62537921 Jul 2017 US
62475210 Mar 2017 US
62410827 Oct 2016 US