US20230331808A1 - Chimeric molecules providing targeted costimulation for adoptive cell therapy - Google Patents

Chimeric molecules providing targeted costimulation for adoptive cell therapy Download PDF

Info

Publication number
US20230331808A1
US20230331808A1 US18/316,548 US202318316548A US2023331808A1 US 20230331808 A1 US20230331808 A1 US 20230331808A1 US 202318316548 A US202318316548 A US 202318316548A US 2023331808 A1 US2023331808 A1 US 2023331808A1
Authority
US
United States
Prior art keywords
seq
domain
cells
sequence
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/316,548
Inventor
John Bridgeman
Robert Hawkins
Ruben Rodriguez
Sujita Sukumaran
Xingliang ZHOU
Eric Gschweng
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Instil Bio UK Ltd
Original Assignee
Instil Bio UK Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Instil Bio UK Ltd filed Critical Instil Bio UK Ltd
Priority to US18/316,548 priority Critical patent/US20230331808A1/en
Assigned to INSTIL BIO INC. reassignment INSTIL BIO INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: RODRIGUEZ, RUBEN
Assigned to INSTIL BIO, INC. reassignment INSTIL BIO, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GSCHWENG, Eric, SUKUMARAN, Sujita, ZHOU, Xingliang
Assigned to INSTIL BIO (UK) LIMITED reassignment INSTIL BIO (UK) LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HAWKINS, ROBERT, BRIDGEMAN, JOHN
Assigned to INSTIL BIO (UK) LIMITED reassignment INSTIL BIO (UK) LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: INSTIL BIO INC.
Publication of US20230331808A1 publication Critical patent/US20230331808A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70517CD8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/50Colon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/54Pancreas
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/55Lung
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/57Skin; melanoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/59Reproductive system, e.g. uterus, ovaries, cervix or testes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies

Definitions

  • the present invention relates to a chimeric molecule useful in adoptive cell therapy (ACT), and cells comprising the same.
  • the chimeric molecule can act as a modulator of cellular activity enhancing responses when an endogenous T-cell receptor (TCR) is engaged with its cognate antigen.
  • TCR T-cell receptor
  • the present invention also provides proteins, nucleic acids encoding the chimeric molecule and therapeutic uses thereof.
  • T-cells Adoptive cell therapy (ACT) using autologous T-cells to mediate cancer regression has shown much promise in early clinical trials.
  • TILs tumor reactive or tumor infiltrating lymphocytes
  • T-cells may be genetically modified to retarget them towards defined tumor antigens. This can be done via the gene transfer of peptide (p)-major histocompatibility complex (MHC) specific T-cell Receptors (TCRs) or synthetic fusions between tumor specific single chain antibody fragment (scFv) and T-cell signaling domains (e.g. CD3), the latter being termed chimeric antigen receptors (CARs).
  • MHC peptide
  • TCRs tumor specific T-cell Receptors
  • scFv tumor specific single chain antibody fragment
  • CD3 T-cell signaling domains
  • TIL and TCR transfer has proven particularly good when targeting melanoma (Rosenberg et al. 2011; Morgan 2006), whereas CAR therapy has shown much promise in the treatment of certain B-cell malignancies (Grupp et al. 2013).
  • Costimulatory signals are useful to achieve robust CAR T cell expansion, function, persistence and antitumor activity.
  • CAR therapy in leukemia has been partly attributed to the incorporation of costimulatory domains (e.g. CD28 or CD137) into the CAR construct, signals from which synergize with the signal provided by CD3 ⁇ to enhance anti-tumor activity.
  • costimulatory domains e.g. CD28 or CD137
  • signal 1 provided by the TCR complex
  • costimulatory receptors such as CD28, CD137 or CD134
  • costimulatory receptors such as CD28, CD137 or CD134
  • AICD activation induced cell death
  • Targeted costimulation will have beneficial effects for non-CAR-based T-cell therapies.
  • incorporating costimulatory domains into a chimeric TCR has been shown to enhance responses of T-cells towards pMHC (Govers 2014).
  • tumor infiltrating lymphocytes TILs
  • TILs utilize their endogenous TCRs to mediate tumor recognition, it has not been possible to engineer the endogenous TCR.
  • TIL are subject to substantial limitations as tumor cells express very few costimulatory ligands.
  • the ability to induce targeted costimulation of TIL, or indeed any other adoptive T-cell therapy product, would be beneficial.
  • chimeric molecules in particular chimeric proteins, designed to provide costimulation when the endogenous TCR is engaged with its cognate antigen.
  • the proposed constructs may be incorporated in the endogenous TCR complex.
  • the TCR receptor complex aggregates, forcing the clustering of these chimeric constructs. This clustering results in the activation of their signaling domains, causing an increase in costimulation.
  • This costimulation manifests itself in a measurable improvement in the effector function of the recipient T cell: increased in activation markers, increase cytokine secretion (IL-2 in particular) and increased proliferation.
  • a chimeric molecule advantageously a chimeric protein, that provides costimulation to the T cell when the endogenous T cell receptor is engaged.
  • This molecule may comprise a TCR clustering domain and a signaling domain that may contain a CD40 intracellular domain or signaling fragment thereof.
  • the TCR clustering domain may be one or more of the proteins typically found in the TCR complex, such as but not limited to, CD3D, CD3E, CD3G, CD3Z, CD3-eta and the constant chains of pre-TCR alpha (PTCRA) TCR alpha, TCR beta, TCR gamma or TCR delta.
  • proteins typically found in the TCR complex such as but not limited to, CD3D, CD3E, CD3G, CD3Z, CD3-eta and the constant chains of pre-TCR alpha (PTCRA) TCR alpha, TCR beta, TCR gamma or TCR delta.
  • PTCRA pre-TCR alpha
  • the signaling domain may also comprise, an additional full length costimulatory domain, including but not limited to CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6.
  • TCR alpha containing constructs are advantageously co-expressed with TCR beta and vice versa; and TCR gamma containing constructs should be co-expressed with TCR delta and vice versa.
  • the preferred configuration includes TCR gamma-delta; and in gamma-delta T cells, the preferred configuration includes TCR alpha-beta to minimize interference/disruption with the endogenous TCR machinery and the TCR pairing.
  • the transmembrane and extracellular portions are advantageously utilized.
  • the present invention also contemplates portions or the totality of their intracellular components, which could potentially minimize the disruption of the endogenous TCR complex signaling or help to further amplify the endogenous TCR signaling.
  • the invention provides a chimeric protein comprising a clustering domain and a signaling domain that may contain a CD40 intracellular domain or signaling fragment thereof.
  • the clustering domain is capable of oligomerization and/or self assembly.
  • clustering comprises formation of a homodimer or homotrimer.
  • clustering comprises oligomerization with a different protein to form a heterodimer or heterotrimer.
  • the chimeric protein is constitutive as signaling, for example independent of receptor engagement by an extracellular ligand or independent of receptor engagement by an extracellular ligand attached to a different cell.
  • the clustering domain comprises a transmembrane domain.
  • the clustering domain comprises a transmembrane domain and further comprises activating mutations that promote dimerization or oligomerization.
  • the clustering domain comprises an extracellular domain, such as but not limited to an extracellular domain of a receptor.
  • the clustering domain comprises an extracellular domain of a receptor and further comprises activating mutations in the extracellular domain that promote dimerization or oligomerization.
  • the clustering domain comprises a leucine zipper.
  • the leucine zipper comprises or constitutes a transmembrane domain.
  • the leucine zipper comprises or constitutes a soluble domain.
  • Non-limiting examples of clustering domains include clustering domains of the thrombopoietin receptor (TpoR), erythropoietin receptor (EpoR), growth hormone receptor (GHR), glycophorin A (GPA) transmembrane domain, and activating mutants thereof.
  • clustering may be modulated by a small molecule. In some embodiments, clustering may be modulated by post-translational modifications.
  • the invention provides a chimeric protein which comprises an extracellular ligand binding domain linked to an intracellular signaling domain by a transmembrane domain.
  • the extracellular ligand binding domain is selected or engineered to bind to an extracellular ligand that maintains two or more copies of the chimeric protein in proximity to one another such that the signaling domain is activated.
  • the extracellular ligand binding domain is considered one part of a specific binding pair (sbp) and the extracellular ligand is the second part of the specific binding pair.
  • one member of the sbp comprises a protein or receptor or extracellular portion thereof and the second sbp comprises a binding protein specific for the first member of the sbp.
  • the extracellular sbp is bivalent. In some embodiments, the extracellular sbp is trivalent.
  • extracellular ligands include antibodies and bivalent antigen binding fragments thereof.
  • Non-limiting examples of extracellular ligand binding domains of chimeric proteins of the invention include, without limitation, NKG2A, CD27, CD137, GITR, PD-1, PD-L1, FasL, OX40, CTLA4, ICOS, CD40, EGFR, HER2 and extracellular portions thereof.
  • Complementary sbp members include, without limitation, pembrolizumab for PD1, trastuzumab for HER2, cetuximab for EGFR, tremelimumab for CTLA4, varlilumab for CD27, and urelumab for CD137.
  • the intracellular signaling domain comprises a CD40 intracellular domain or signaling fragment thereof.
  • the CD40 signaling domain comprises SEQ ID NO:154, SEQ ID NO:155, or SEQ ID NO:156.
  • the CD40 signaling fragment comprises, consists, or consists essentially of an SH3 motif (KPTNKAPH, PTNKAPHP or PTNKAPH), TRAF2 motif (PKQE, PKQET, PVQE, PVQET, SVQE, SVQET), TRAF6 motif (QEPQEINFP or QEPQEINFP), PKA motif (KKPTNKA, SRISVQE, or a combination thereof, or is a full length CD40 intracellular domain.
  • SH3 motif KPTNKAPH, PTNKAPHP or PTNKAPH
  • TRAF2 motif PKQE, PKQET, PVQE, PVQET, SVQE, SVQET
  • TRAF6 motif QEPQEINFP or QEPQEINFP
  • PKA motif KKPTNKA, SRISVQE, or a combination thereof
  • one or more of the SH3, TRAF2, TRAF6, or PKA motifs of the CD40 signaling domain is mutated. In some embodiments, one or more of the SH3, TRAF2, TRAF6, or PKA motifs of the CD40 signaling domain is present in multiple copies.
  • the engineered protein has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 166, wherein the sequence is not SEQ ID NO: 123.
  • the engineered protein has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 167, wherein the sequence is not SEQ ID NO: 123.
  • the engineered protein further comprises a binding domain, CD28 domain, and CD40 domain.
  • the engineered protein further comprises a signal peptide sequence.
  • the signal peptide sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 157.
  • the binding domain comprises a VL sequence, a VH sequence, and an at least one linker.
  • the at least one linker has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 159 or 161.
  • the binding domain comprises two linker sequences.
  • the two linker sequences have at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to amino acid sequences SEQ ID NO: 159 and SEQ ID NO: 161, respectively.
  • the VL sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 158. In some embodiments, the VH sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 160. In some embodiments, the CD40 domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 165.
  • the CD28 domain comprises a CD28 transmembrane domain. In some embodiments, the CD28 transmembrane domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 163. In some embodiments, the CD28 domain comprises a CD28 extracellular domain. In some embodiments, the CD28 extracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 162. In some embodiments, the CD28 domain comprises a CD28 intracellular domain.
  • the CD28 intracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 164.
  • the protein further comprises 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDSAVYAYALNI (SEQ ID NO: 173).
  • the binding domain and CD28 domain are connected by an at least one linker.
  • an engineered protein is provided. It can comprise an amino acid sequence that is at least 80% identical to the amino acid sequence of SEQ ID NO: 166 or 167, wherein the amino acid sequence does not include at least one of:
  • a CoStAR is provided. It can comprise: a) an optional signal peptide; b) a binding domain, wherein the binding domain binds to an anti-pembrolizumab antibody or binding fragment thereof; c) a CD28 domain; and d) a CD40 domain. Wherein a) is optionally linked to b), wherein b) is linked to c), wherein c) is linked to d), and wherein the CoStAR comprises an amino acid sequence that: i) lacks at least one of:
  • a fusion protein comprises a) a means for binding to an antibody that binds to pembrolizumab; b) a CD28 domain; and c) CD40 domain. Wherein a) is linked to b), wherein b) is linked to c), and wherein the fusion protein comprises an amino acid sequence that: i) lacks at least one of:
  • a fusion protein that comprises the amino acid sequence of SEQ ID NO: 166.
  • a fusion protein that comprises the amino acid sequence of SEQ ID NO: 167.
  • nucleic acid which encodes the protein of any one of the preceding claims.
  • nucleic acid which encodes a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a vector which comprises a nucleic acid of any one of the embodiments of the present application.
  • Also disclosed herein is a cell which expresses a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a cell which expresses at least two proteins of any one of the embodiments of the present application.
  • Also disclosed herein is a method of making the cell of any one of the embodiments of the present application which comprises the step of transducing or transfecting a cell with a vector of any one of the embodiments of the present application.
  • Also disclosed herein is a method for preparing a population of cells that express a protein of any one of the embodiments of the present application, comprising detecting expression of the protein on the surface of cells transfected or transduced with a vector according to any one of the embodiments of the present application and selecting cells which are identified as expressing the protein.
  • Also disclosed herein is a cell population produced by the method of any one of the methods disclosed in the present application.
  • Also disclosed herein is a cell population which is enriched for cell expression a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a method for treating a disease in a subject in need thereof, which comprises the step of administering the cell of any one of the embodiments of the present application, or the cell population of any one of the embodiments of the present application, to the subject.
  • FIG. 1 A- 1 C Schematic models for universal costimulatory proteins.
  • TCR incorporated antigen agnostic receptor (TIAAR) comprises modifying components of the TCR complex and associated signaling adaptors.
  • FIG. 1 B A constitutive costimulatory receptor comprising transmembrane domains (TMDs) and features that enable inducible or constitutive activation.
  • FIG. 1 C An inducible costimulatory receptor capable of induction and activation by extracellular ligand binding.
  • FIG. 2 Cytokine production by TCR incorporated antigen agnostic receptor (TIAAR) transduced cells.
  • Cytokine production (Bcl-xL, IL2, IFNg and TNFa) from genetically modified and non-transduced T cells (NTD) of two donors was determined after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only).
  • FIGS. 3 A- 3 B Provides and activation marker expression by TIAAR transduced cells.
  • Proliferation T cell counts
  • activation marker expression 41BB and CD69
  • NTD genetically modified and non-transduced T cells
  • FIG. 4 Cytokine production in leucine zipper based universal CoStAR (LZ) transduced cells. Cytokine production (Bcl-xL, IL2, IFNg and TNFa) from genetically modified and non-transduced T cells (NTD) of two donors was determined after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only).
  • FIGS. 5 A- 5 B Provided and activation marker expression by LZ-CoStAR transduced cells. Proliferation (T cell counts) and activation marker expression (41BB and CD69) was determined for genetically modified and non-transduced T cells (NTD) from donor 1 ( FIG. 5 A ) and donor 2 ( FIG. 5 B ) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only).
  • NTD genetically modified and non-transduced T cells
  • FIG. 6 Cytokine production in inducible universal CoStAR transduced cells. Cytokine production (Bcl-xL, IL2, IFNg and TNFa) from genetically modified and non-transduced T cells (NTD) of two donors was determined after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only). The universal CoStAR is inducible by pembrolizumab.
  • FIGS. 7 A- 7 B Provides and activation marker expression by inducible universal CoStAR transduced cells. Proliferation (T cell counts) and activation marker expression (41BB and CD69) was determined for genetically modified and non-transduced T cells (NTD) from donor 1 ( FIG. 7 A ) and donor 2 ( FIG. 7 B ) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only).
  • the universal CoStAR is inducible by pembrolizumab.
  • FIG. 8 A is a schematic of a protein of some embodiments provided herein. It is a protein comprising, Universal CoStAR sequence, comprising an optional section, a binding domain, a CD28 domain, and a CD40 domain.
  • FIG. 8 B is a schematic of some embodiments provided herein.
  • FIG. 8 C outlines a set of sequences of some embodiments provided herein.
  • FIG. 9 depicts a sequence of an anti-pembrolizumab CoStAR Sequence (“Universal CoStAR”) (SEQ ID NO: 166), containing an optional signal domain.
  • Universal CoStAR Anti-pembrolizumab CoStAR Sequence
  • FIG. 10 depicts a sequence of an anti-pembrolizumab CoStAR Sequence (“Universal CoStAR”) (SEQ ID NO: 166), without the optional signal domain.
  • Universal CoStAR Anti-pembrolizumab CoStAR Sequence
  • FIG. 11 depicts a sequence alignment between the anti-pembrolizumab CoStAR Sequence (“Universal CoStAR”) (SEQ ID NO: 166) and the vector clone pIB1102 (SEQ ID NO: 123).
  • FIG. 12 depicts the transduction efficiency of constructs into TILs after 21 days. Round 1 (left panel) and Round 2 (right panel) of transduction was performed with constructs CTP386.1 and CTP387.1 across a variety of TIL organ types (x-axis).
  • FIG. 13 depicts the transduction efficiency of constructs into TILs after 21 days. Round 1 (left panel) and Round 2 (right panel) of transduction was performed with constructs 322 and 1324 across a variety of TIL organ types (x-axis).
  • FIG. 14 depicts the transduction efficiency of constructs into TILs after 21 days. Round 1 (left panel) and Round 2 (right panel) of transduction was performed with construct CTP205 across a variety of TIL organ types (x-axis).
  • FIGS. 15 A- 15 E depict the fold-expansion of TILs following a serial stimulation assay.
  • Anti-CEA or anti-FOLR modified TILs from CRC 9823 were treated every 7 days with the target K562 OKT3 CEACAM5 or OKT3 FOLR, respectively. The readout was measured using the cell count over time. Shown in panels are the fold-expansion for anti-CEA TILs with K562 OKT3 CEACAM5 exposure ( FIG. 15 A ), anti-FOLR TILs with OKT3 FOLR exposure ( FIG. 15 B ), a universal CoStAR with K562 OKT3 CEACAM5 exposure ( FIG.
  • FIG. 15 C a universal CoStAR with K562 OKT3 CEACAM5 exposure and 5 ug/mL pembro
  • FIG. 15 E a universal CoStAR with K562 OKT3 CEACAM5 exposure and 250 ug/mL pembdro
  • FIGS. 16 A- 16 E depict the fold-expansion of TILs following a serial stimulation assay.
  • Anti-CEA or anti-FOLR modified TILs from Mel 11909 were treated every 7 days with the target K562 OKT3 CEACAM5 or OKT3 FOLR, respectively. The readout was measured using the cell count over time. Shown in panels are the fold-expansion for anti-CEA TILs with K562 OKT3 CEACAM5 exposure ( FIG. 16 A ), anti-FOLR TILs with OKT3 FOLR exposure ( FIG. 16 B ), a universal CoStAR with K562 OKT3 CEACAM5 exposure ( FIG.
  • FIG. 16 C a universal CoStAR with K562 OKT3 CEACAM5 exposure and 5 ug/mL pembro
  • FIG. 16 E a universal CoStAR with K562 OKT3 CEACAM5 exposure and 250 ug/mL pembdro
  • FIGS. 17 A- 17 B depict the increase in IL2 production (pg/mL) in anti-CEA, anti-FOLR, and Universal CoStAR modified TLS from CRC983 ( FIG. 17 A ) or Mel 11909 ( FIG. 17 B ).
  • FIG. 19 depicts an example timeline for transducing CoStAR constructs into cells using the protocol outlined in Example 3.
  • FIG. 20 depicts the transduction efficiency of constructs into TILs after 1 day. Round 1 (left panel) and Round 2 (right panel) of transduction was performed with across a variety of TIL organ types (x-axis). Plotted on the y-axis, is the percent of cells with positive expression of FOLR1 following transduction.
  • FIG. 21 depicts the transduction efficiency of constructs into TILs after 1 day. Round 1 (left panel) and Round 2 (right panel) of transduction was performed across a variety of TIL organ types (x-axis). Plotted on the y-axis, is the percent of cells with positive expression of CEA following transduction.
  • FIG. 22 depicts the transduction efficiency of constructs into TILs after 1 day. Round 1 (left panel) and Round 2 (right panel) of transduction was performed across a variety of TIL organ types (x-axis). Plotted on the y-axis, is the percent of cells with positive expression of both FOLR1 and CEA following transduction.
  • FIG. 23 depicts the round 1 (left panel) and round 2 (right panel) of Anti-CEA (386.1) cells that were percent positive for CEA expression after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIG. 24 depicts the round 1 (left panel) and round 2 (right panel) of Anti-CEA (387.1) cells that were percent positive for CEA expression after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIG. 25 depicts the round 1 (left panel) and round 2 (right panel) of Anti-FOLR cells that were percent positive for FOLR expression after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIG. 26 depicts the round 1 (left panel) and round 2 (right panel) of Universal CoStAR (pIB1322) cells that were percent positive for both FOLR and CEA after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIG. 27 depicts the round 1 (left panel) and round 2 (right panel) of Universal CoStAR (pIB1324) cells that were percent positive for both FOLR and CEA after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIGS. 28 A- 28 D depict the ratio of CD4 to CD8 positive TIL cells of those enriched for FOLR and/or CEA expression after 21 days.
  • FIG. 28 A depicts the ratio in CRC cells
  • FIG. 28 B depicts the ratio in NSCLC cells
  • FIG. 28 C depicts the ratio in ovarian cells
  • FIG. 28 D depicts the ratio in melanoma cells.
  • FIG. 29 depicts an example timeline for sorting TIL cells and testing for function, using the protocol outlined in Example 4.
  • FIG. 30 A- 30 H depicts the increase in IFNg production (pg/mL) in TIL and K562 cells lines following co-culturing, across the cell types CRC-11974 ( FIG. 30 A ), CRC-11959 ( FIG. 30 B ), NSCLC-9332 ( FIG. 30 C ), NSCLC-9596 ( FIG. 30 D ), Ovarian cells ( FIG. 30 E ), Melanoma-CC60 ( FIG. 30 F ), Melanoma-11909 ( FIG. 30 G ), and Melanoma-17614 ( FIG. 30 H ).
  • FIG. 31 A- 31 H depicts the increase in IL2 production (pg/mL) in TIL and K562 cells lines following co-culturing, across the cell types CRC-11974 ( FIG. 31 A ), CRC-11959 ( FIG. 31 B ), NSCLC-9332 ( FIG. 31 C ), NSCLC-9596 ( FIG. 31 D ), Ovarian cells ( FIG. 31 E ), Melanoma-CC60 ( FIG. 31 F ), Melanoma-11909 ( FIG. 31 G ), and Melanoma-17614 ( FIG. 31 H ).
  • FIG. 32 A- 32 H depicts the increase in TNFa production (pg/mL) in TIL and K562 cells lines following co-culturing, across the cell types CRC-11974 ( FIG. 32 A ), CRC-11959 ( FIG. 32 B ), NSCLC-9332 ( FIG. 32 C ), NSCLC-9596 ( FIG. 32 D ), Ovarian cells ( FIG. 32 E ), Melanoma-CC60 ( FIG. 32 F ), Melanoma-11909 ( FIG. 32 G ), and Melanoma-17614 ( FIG. 32 H ).
  • the engineered protein that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 166, and wherein the sequence is not SEQ ID NO: 123.
  • the engineered protein has at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 70 and 100%, identity to SEQ ID NO: 166.
  • the engineered protein has an at least 80% identity to SEQ ID NO:166.
  • the engineered protein has an at least 90% identity to SEQ ID NO:166. In some embodiments, the engineered protein is SEQ ID NO:166 ( FIG. 9 ). In some embodiments, the sequence is at least 80% identical and is not the sequence of SEQ ID NO: 123.
  • the engineered protein that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 167, and wherein the sequence is not SEQ ID NO: 123.
  • the engineered protein has an at least 80% identity to SEQ ID NO:167.
  • the engineered protein has an at least 90% identity to SEQ ID NO:167.
  • the engineered protein is SEQ ID NO:167 ( FIG. 10 ).
  • the sequence is at least 80% identical, and is not the sequence of SEQ ID NO: 123.
  • an engineered protein has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 166, and wherein the sequence is not SEQ ID NO: 123.
  • an engineered protein has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 167, and wherein the sequence is not SEQ ID NO: 123.
  • the engineered protein, CoStAR or fusion protein has a general structure as depicted in FIG. 8 A .
  • the engineered protein CoStAR or fusion protein has a general structure as depicted in FIG. 8 B .
  • the engineered protein CoStAR or fusion protein comprises at least one sequence depicted in FIG. 8 C .
  • the arrangement in FIG. 8 A is an embodiment separate from the embodiments in FIGS. 8 B and/or 8 C .
  • the arrangement in FIG. 8 B is an embodiment separate from the embodiments in FIGS. 8 C and/or 8 A .
  • the arrangement in FIG. 8 C is an embodiment separate from the embodiments in FIGS. 8 A and/or 8 B (thus, the sequence itself is envisioned, in some embodiments, as the entirety of the engineered protein).
  • FIG. 8 B depicts some embodiments that are a subset of FIG. 8 C .
  • FIG. 8 C depicts some embodiments that are a subset of FIG. 8 A and FIG. 8 B .
  • Exemplary CDRs are underlined in FIG. 8 C (SEQ ID NO: 158 and 160).
  • the engineered protein, CoStAR or fusion protein is as depicted in FIG. 9 or 10 . Exemplary CDRs are underlined in FIG. 9 and FIG. 10 . In some embodiments, the engineered protein, CoStAR or fusion protein is different from other fusion proteins, as shown in FIG. 11 . In some embodiments, the engineered protein, CoStAR or fusion protein can lack a tag component and/or a section of CD28.
  • the engineered protein comprises a binding domain. In some embodiments, the engineered protein comprises a CD28 domain. In some embodiments, the engineered protein comprises a CD40 domain. In some embodiments, the engineered protein comprises 1, 2, or all 3 of a binding domain, a CD28 domain, and/or a CD40 domain.
  • the engineered protein comprises a signal peptide sequence.
  • signal peptide is given its usual scientific meaning, and thus refers a short peptide that functions in translocating the rest of the attached protein. “Signal peptide” thus may also be called a signal sequence, targeting signal, localization signal, localization sequence, transit peptide, leader sequence or leader peptide. It will be understood that the signal peptide may be any peptide with the function of signaling for the attached peptide to be translocated to the plasma membrane of a cell.
  • the signal peptide sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 157.
  • the binding domain comprises 1, 2, or all 3 of a VL sequence, a VH sequence, and/or an at least one linker.
  • the at least one linker has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 159 or 161.
  • the binding domain comprises two linker sequences.
  • the two linker sequences have at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to amino acid sequences SEQ ID NO: 159 and SEQ ID NO: 161, respectively.
  • the VL sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 158.
  • the VH sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 160.
  • all 3 of the heavy chain CDRs and/or all three of the light chain CDRs within the VH and VL are identical to the heavy and/or light chain CDRs contained within SEQ ID NOs: 158 and 160.
  • 1, 2, 3, 4, 5 or 6 of the CDRs have 1, 2, 3, 4 or more point mutations.
  • 1, 2, 3, 4, 5, or 6 CDRs are 70, 80, 85, 90, 95, 96, 97, 98, 99 or 100% identical to the corresponding CDRs within SEQ ID NOs: 158 and/or 160.
  • the protein comprises 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDSAVYAYALNI (SEQ ID NO: 173).
  • the binding domain and CD28 domain are connected by an at least one linker.
  • 1, 2, 3, 4, 5 or 6 of the CDRs have 1, 2, 3, 4, or more point mutations.
  • 1, 2, 3, 4, 5, or 6 CDRs are 70, 80, 85, 90, 95, 96, 97, 98, 99 or 100% identical to the CDRs.
  • an engineered protein comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to the amino acid sequence of SEQ ID NO: 166 or 167.
  • an engineered protein comprises an amino acid sequence that is at least 80% identical to the amino acid sequence of SEQ ID NO: 166 or 167, wherein the amino acid sequence does not include at least one of:
  • the CD40 domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 165.
  • the CD28 domain comprises a CD28 transmembrane domain.
  • the CD28 transmembrane domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 163.
  • the CD28 domain comprises a CD28 extracellular domain.
  • the CD28 extracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 162.
  • the CD28 domain comprises a CD28 intracellular domain.
  • the CD28 intracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 164.
  • the amino acid sequence and/or fusion protein and/or engineered protein does not include at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQQLQV YSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175).
  • the engineered protein lacks both of QKLISEEDLE (SEQ ID NO: 174) and
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 157.
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 158.
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 159.
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 160.
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 161.
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 162.
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 163.
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 164.
  • the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 165.
  • the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) that have at least 80% identity to SEQ ID NOS: 157-165, respectively. In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) that have at least 90% identity to SEQ ID NOS: 157-165, respectively. In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) that have at least 95% identity to SEQ ID NOS: 157-165, respectively. In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) that have at least 98% identity to SEQ ID NOS: 157-165, respectively. In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) selected from the group consisting of: SEQ ID NOS: 157-165.
  • the CoStAR comprises an optional signal peptide, a binding domain, wherein the binding domain binds to an anti-pembrolizumab antibody or binding fragment thereof, a CD28 domain, and a CD40 domain, wherein the signal peptide is optionally linked to the binding domain, wherein the binding domain is linked to the CD28 domain, wherein the CD28 domain is linked to the CD40 domain, and wherein the CoStAR comprises an amino acid sequence that: i) lacks at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQ QLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175); ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167;
  • the fusion protein comprises a means for binding to an antibody that binds to pembrolizumab, a CD28 domain, and a CD40 domain, wherein the means for binding to an antibody is linked to a CD28 domain, wherein the CD28 domain is linked to the CD40 domain, and wherein the fusion protein comprises an amino acid sequence that: i) lacks at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQQLQV YSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175); ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167; iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is
  • the binding domain or the means for binding to an antibody that binds to pembrolizumab comprises: 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDSAVYAYALNI (SEQ ID NO: 173).
  • 1, 2, 3, 4, 5 or 6 of the CDRs have 1, 2, 3, 4, or more point mutations.
  • 1, 2, 3, 4, 5, or 6 CDRs are 70, 80, 85, 90, 95, 96, 97, 98, 99 or 100% identical to the CDRs.
  • the CD28 domain comprises: SEQ ID Nos: 162, 163, and 164, or a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical thereto.
  • the CD40 domain comprises: SEQ ID No: 165, or a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical thereto.
  • a fusion protein comprising the amino acid sequence of SEQ ID NO: 166.
  • the fusion protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 166.
  • a fusion protein comprising the amino acid sequence of SEQ ID NO: 167.
  • the fusion protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 167.
  • nucleic acid which encodes a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a vector which comprises a nucleic acid of any one of the embodiments of the present application.
  • Also disclosed herein is a cell which expresses a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a cell which expresses at least two proteins of any one of the embodiments of the present application.
  • Also disclosed herein is a method of making the cell of any one of the embodiments of the present application which comprises the step of transducing or transfecting a cell with a vector of any one of the embodiments of the present application.
  • Also disclosed herein is a method for preparing a population of cells that express a protein of any one of the embodiments of the present application, comprising detecting expression of the protein on the surface of cells transfected or transduced with a vector according to any one of the embodiments of the present application and selecting cells which are identified as expressing the protein.
  • Also disclosed herein is a cell population produced by the method of any one of the methods disclosed in the present application.
  • Also disclosed herein is a cell population which is enriched for cell expression a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a method for treating a disease in a subject in need thereof, which comprises the step of administering the cell of any one of the embodiments of the present application, or the cell population of any one of the embodiments of the present application, to the subject.
  • full length protein or “full length receptor” refers to a receptor protein, such as, for example, a CD40 receptor protein.
  • full length encompasses receptor proteins lacking up to about 5 or up to 10 amino acids, for example 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids, at the N-terminal of the mature receptor protein once its signal peptide has been cleaved. For instance, while a specific cleavage site of a receptors N-terminal signal peptide may be defined, variability in exact point of cleavage has been observed.
  • the term “full length” does not imply presence or absence of amino acids of the receptors N-terminal signal peptide. In one embodiment, the term “full length” (e.g.
  • a full length CD28 or a full length CD40 intracellular domain encompasses mature receptor proteins (e.g. CD28 according to certain aspects of the invention) lacking the N terminal signal peptide lacking up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein once its signal peptide has been cleaved.
  • a “full length” CD28 receptor or other receptor or TCR clustering domain does not include the signal peptide and may lack up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein (e.g. N terminal residues N, K, I, L and/or V).
  • SEQ ID Nos. 4-12 This is shown in the exemplary fusions, e.g. SEQ ID Nos. 4-12 (note that these may lack up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein as shown in the boxed region).
  • the chimeric protein of the present invention may comprise a TCR clustering domain as well as a signaling domain that advantageously may comprise a CD40 intracellular domain.
  • T cell receptor refers to a heterodimeric receptor composed of ⁇ or ⁇ chains that pair on the surface of a T cell.
  • Each ⁇ , ⁇ , ⁇ , and ⁇ chain is composed of two Ig-like domains: a variable domain (V) that confers antigen recognition through the complementarity determining regions (CDR), followed by a constant domain I that is anchored to cell membrane by a connecting peptide and a Transmembral TM) region.
  • V variable domain
  • CDR complementarity determining regions
  • TM Transmembral Transmembral region
  • the TM region associates with the invariant subunits of the CD3 signaling apparatus.
  • Each of the V domains has three CDRs.
  • CDRs interact with a complex between an antigenic peptide bound to a protein encoded by the major histocompatibility complex (pMHC) (Davis and Bjorkman (1988) Nature, 334, 395-402; Davis et al. (1998) Annu Rev Immunol, 16, 523-544; Murphy (2012), xix, 868 p.).
  • pMHC major histocompatibility complex
  • Costimulatory receptor proteins useful in the chimeric proteins of the invention include, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6, which in their natural form comprise extracellular ligand binding domains and intracellular signal transducing domains.
  • CD2 is characterized as a cell adhesion molecule found on the surface of T cells and is capable of initiating intracellular signals necessary for T cell activation.
  • CD27 is characterized as a type II transmembrane glycoprotein belonging to the TNFR superfamily (TNFRSF) whose expression on B cells is induced by antigen-receptor activation in B cells.
  • CD28 is one of the proteins on T cells and is the receptor for CD80 (B7.1) and CD86 (B7.2) ligands on antigen-presenting cells.
  • CD137 (4-1BB) ligand is found on most leukocytes and on some non-immune cells.
  • OX40 ligand is expressed on many antigen-presenting cells such as DC2s (dendritic cells), macrophages, and B lymphocytes.
  • the costimulatory receptor protein is full length CD28 as defined herein.
  • CD40 is a member of the tumor necrosis factor receptor (TNFR) superfamily and several isoforms are generated by alternative splicing. Its ligand, CD154 (also called CD40L) is a protein that is primarily expressed on activated T cells.
  • CD40 isoform 1 protein sequence is set forth in GenBank accession No. NP_001241.1, including signal peptide (amino acids 1-20), transmembrane domain (amino acids 194-215), and cytoplasmic domain (amino acids 216-277) (SEQ ID NO:22).
  • CD40 receptor signaling involves adaptor proteins including but not limited to TNF receptor-associated factors (TRAF), and the cytoplasmic domain comprises signaling components, including but not limited to an SH3 motif (KPTNKAPH), TRAF2 motif (PKQE, PVQE, SVQE), TRAF6 motif (QEPQEINFP) and PKA motif (KKPTNKA, SRISVQE).
  • Further motifs for binding to TRAF1, TRAF2, TRAF3, and TRAF5 comprise the major consensus sequence (P/S/A/T)X(Q/E)E or minor consensus sequence PXQXXD and can be identified in or obtained from, without limitation, TNFR family members such as CD30, Ox40, 4-1BB, and the EBV oncoprotein LMP1.
  • Examples disclosed herein demonstrate operation of CD40 as a signaling domain and further that cytokine and chemokine expression profiles are altered by signaling domain selection.
  • the CD40 signaling domains of the invention provide distinct and overlapping responses induced by the different factor binding sites.
  • Ahonen, C L et al. The CD 40- TRAF 6 axis controls affinity maturation and the generation of long - lived plasma cells . Nat Immunol. 2002; 3: 451-456; Mackey M F et al., Distinct contributions of different CD 40 TRAF binding sites to CD 154- induced dendritic cell maturation and IL -12 secretion . Eur J Immunol.
  • TNF receptor - associated factor 6 is an essential mediator of CD 40- activated proinflammatory pathways in monocytes and macrophages . J Immunol. 2005; 174: 1081-1090.
  • a chimeric protein of the invention comprises substantially all of a CD40 costimulatory domain. In some embodiments, a chimeric protein of the invention comprises two or more CD40 costimulatory domains. In some embodiments, a chimeric protein of the invention comprises a CD40 costimulatory domain signaling component or motif, including but not limited to an SH3 motif (KPTNKAPH), TRAF2 motif (PKQE, PVQE, SVQE), TRAF3 motif, TRAF6 motif (QEPQEINFP) or PKA motif (KKPTNKA, SRISVQE) as well as two or more, or three or more, or four or more such components of motifs, which can be in multiple copies and arranged in any order. In some embodiments, a chimeric protein of the invention comprises a CD40 costimulatory domain and a CD40 costimulatory domain signaling component or motif.
  • selection of one or more costimulatory domain signaling component or motif is guided by the cell in which the chimeric protein is to be expressed and/or a desired costimulatory activity more closely identified with a signaling component or motif, or avoidance of a costimulatory activity more closely identified with a signaling component or motif.
  • a chimeric protein signaling domain comprises, in addition to a CD40 costimulatory domain or signaling component or motif thereof, or two or more such domains or components or motifs or combinations thereof, an additional full length costimulatory domain or signaling component thereof from, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6,
  • the human CD28 protein sequence is set forth in GenBank accession No. NP_006130.1, including signal peptide (amino acids 1-18), extracellular domain (amino acids 19-152), transmembrane domain (amino acids 153-179) and cytoplasmic domain (amino acids 180-200).
  • the extracellular domain includes an immunoglobulin type domain (amino acids 21-136) which contains amino acids with compose the antigen binding site and amino acids that form the homodimer interface.
  • the extracellular domain includes several asparagine residues which may be glycosylated, and the intracellular domain comprises serine and tyrosine residues, which may be phosphorylated.
  • the human CD8 alpha chain protein sequence is set forth by GenBank accession No. NP_001139345.1, including signal peptide (amino acids 1-21), extracellular domain (amino acids 22-182), transmembrane domain (amino acids 183-203), and cytoplasmic domain (amino acids 204-235).
  • the extracellular domain includes an immunoglobulin type domain (amino acids 28-128) which contains amino acids with compose the antigen binding site and amino acids that form the homodimer interface.
  • the extracellular domain includes several asparagine residues which may be glycosylated, and the intracellular domain comprises serine and tyrosine residues, which may be phosphorylated.
  • the human IgG4 constant region sequence is set forth in UniProtKB/Swiss-Prot: accession No. P01861.1, including CH1 (amino acids 1-98), hinge (amino acids 99-110), CH2 (amino acids 111-220), CH3 (amino acids 221-327).
  • the CH2 region includes asparagine at amino acid 177, which is the glycosylated and associated with Fc receptor and antibody-dependent cell-mediated cytotoxicity (ADCC).
  • the protein sequence of human CD137 (41BB), another TNFR superfamily member, is set forth by GenBank accession No. NP_001552.2, including signal peptide (amino acids 1-23), extracellular domain (amino acids 24-186), transmembrane domain (amino acids 187-213), and cytoplasmic domain (amino acids 214-255).
  • the human CD134 (OX40) protein sequence is set forth by GenBank accession No. NP_003318.1, including signal peptide (amino acids 1-28), extracellular domain (amino acids 29-214), transmembrane domain (amino acids 215-235), and cytoplasmic domain (amino acids 236-277).
  • This receptor has been shown to activate NF-kappaB through its interaction with adaptor proteins TRAF2 and TRAF5 and studies suggest that this receptor promotes expression of apoptosis inhibitors BCL2 and BCL21L1/BCL2-XL.
  • the human T-cell surface antigen CD2 has at least two isoforms.
  • the human CD2 isoform1 protein sequence is set forth by NP_001315538.1, including signal peptide (amino acids 1-24), extracellular domain (amino acids 25-235), transmembrane domain (amino acids 236-261), and cytoplasmic domain (amino acids 262-377).
  • the human CD2 isoform2 protein sequence is set forth by NP_001758.2
  • the human CD357 (GITR) isoform-1 protein sequence is set forth by GenBank accession No. NP_004186.1, including signal peptide (amino acids 1-25), extracellular domain (amino acids 26-162), transmembrane domain (amino acids 163-183), and cytoplasmic domain (amino acids 184-241).
  • the human CD29 (beta1 integrin) protein sequence is set forth by GenBank accession No. NP_596867, including signal peptide (amino acids 1-20), extracellular domain (amino acids 21-728), transmembrane domain (amino acids 729-751), and cytoplasmic domain (amino acids 752-798).
  • human CD150 (SLAM) protein sequence has at several isoforms.
  • mCD150 transmembrane form of CD150
  • sCD150 secreted form of CD150
  • human SLAM isoform b is set forth by GenBank accession No. NP_003028.1, including signal peptide (amino acids 1-20), extracellular domain (amino acids 21-237), transmembrane domain (amino acids 238-258), and cytoplasmic domain (amino acids 259-335).
  • Human SLAM isoform a is set forth by GenBank accession No. NP_001317683.1.
  • a chimeric protein may be expressed alone under the control of a promoter in a therapeutic population of cells that have therapeutic activity, for example, Tumor Infiltrating Lymphocytes (TILs).
  • TILs Tumor Infiltrating Lymphocytes
  • the chimeric protein may be expressed along with a therapeutic transgene such as a chimeric antigen receptor (CAR) and/or T-cell Receptor (TCR).
  • CAR chimeric antigen receptor
  • TCR T-cell Receptor
  • Suitable TCRs and CARs are well known in the literature, for example HLA-A*02-NYESO-1 specific TCRs (Rapoport et al. Nat Med 2015) or anti-CD19scFv.CD3 ⁇ fusion CARs (Kochenderfer et al.
  • the chimeric proteins described herein may be expressed with any known CAR or TCR thus providing the cell with a regulatable growth switch to allow cell expansion in-vitro or in-vivo, and a conventional activation mechanism in the form of the TCR or CAR for anti-cancer activity.
  • the invention provides a cell for use in adoptive cell therapy comprising a chimeric protein as described herein and a TCR and/or CAR that specifically binds to a tumor associated antigen.
  • An exemplary chimeric protein comprising CD28 includes an extracellular antigen binding domain and an extracellular, transmembrane and intracellular signaling domain.
  • a chimeric protein of the invention optionally comprises a spacer region between the TCR clustering domain and the costimulatory receptor.
  • spacer refers to the extracellular structural region of a chimeric protein that separates the TCR clustering domain from the signaling domain of the chimeric protein.
  • long spacers are employed, for example to target membrane-proximal epitopes or glycosylated antigens (see Guest R. D. et al. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J. Immunother. 2005; 28:203-211; Wilkie S.
  • chimeric proteins bear short spacers, for example to target membrane distal epitopes (see Hudecek M. et al., Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 2013; 19:3153-3164; Hudecek M. et al 27acarbazine27nalling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol. Res. 2015; 3:125-135).
  • the spacer comprises all or part of or is derived from an IgG hinge, including but not limited to IgG1, IgG2, or IgG4.
  • a spacer can comprise all or part of one or more antibody constant domains, such as but not limited to CH2 and/or CH3 domains.
  • the CH2 domain in a spacer comprising all or part of a CH2 domain, is modified so as not to bind to an Fc receptor. For example, Fc receptor binding in myeloid cells has been found to impair CAR T cell functionality.
  • the spacer comprises all or part of an Ig-like hinge from CD28, CD8, or other protein comprising a hinge region. In some embodiments of the invention that comprise a spacer, the spacer is from 1 and 50 amino acids in length.
  • the chimeric protein extracellular domain comprises a linker.
  • Linkers comprise short runs of amino acids used to connect domains, for example a binding domain with a spacer or transmembrane domain.
  • a ligand binding domain will usually be connected to a spacer or a transmembrane domain by flexible linker comprising from about 5 to 25 amino acids, such as, for example, AAAGSGGSG or GGGGSGGGGSGGGGS.
  • a chimeric protein comprises a TCR clustering domain joined directly to a signaling domain by a linker, and without a spacer.
  • a chimeric protein comprises a binding domain joined directly to a transmembrane by a spacer and without a linker.
  • a chimeric protein comprises a full length primary costimulatory receptor which can comprise an extracellular ligand binding and intracellular signaling portion of, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6.
  • the chimeric protein for instance may comprise an extracellular ligand binding domain of one of the aforementioned proteins and an intracellular signaling domain of another of the aforementioned proteins.
  • the signaling portion of the chimeric protein comprises a single signaling domain.
  • the signaling portion of the chimeric protein comprises a second intracellular signaling domain such as but not limited to: CD2, CD27, CD28, CD40, CD134 (OX40), CD137 (4-1BB), CD150 (SLAM).
  • the first and second intracellular signaling domains are the same.
  • the first and second intracellular signaling domains are different.
  • the costimulatory receptor is capable of dimerization. Without being bound by theory, it is thought that chimeric proteins dimerize or associate with other accessory molecules for signal initiation.
  • chimeric proteins dimerize or associate with accessory molecules through transmembrane domain interactions. In some embodiments, dimerization or association with accessory molecules is assisted by costimulatory receptor interactions in the intracellular portion, and/or the extracellular portion of the costimulatory receptor.
  • the transmembrane domain influences chimeric protein function.
  • the transmembrane domain is comprised by the full length primary costimulatory receptor domain.
  • the transmembrane domain can be that of the extracellular domain or the intracellular domain.
  • the transmembrane domain is from CD4, CD8a, CD28, or ICOS. Gueden et al.
  • the transmembrane domain comprises a hydrophobic ⁇ helix that spans the cell membrane.
  • amino acid sequence variants of the TCR clustering domain or other moieties provided herein are contemplated.
  • Amino acid sequence variants of an antibody moiety may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the clustering moiety, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody moiety. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • TCR clustering domain moieties comprising one or more amino acid substitutions, deletions, or insertions are provided.
  • Amino acid substitutions may be introduced into a binding domain of interest and the products screened for a desired activity, e.g., retained/improved clustering or decreased immunogenicity.
  • amino acid substitutions may be introduced into one or more of the primary co-stimulatory receptor domain (extracellular or intracellular), secondary costimulatory receptor domain, or extracellular co-receptor domain. Accordingly, the invention encompasses chimeric proteins and component parts particularly disclosed herein as well as variants thereof, i.e.
  • chimeric proteins and component parts having at least 75%, at least 80%, at least 85%, at least 87%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity to the amino acid sequences particularly disclosed herein.
  • the terms “percent similarity,” “percent identity,” and “percent homology” when referring to a particular sequence are used as set forth in the University of Wisconsin GCG software program BestFit. Other algorithms may be used, e.g. BLAST, psiBLAST or TBLASTN (which use the method of Altschul et al. (1990) J. Mol. Biol. 215: 405-410), FASTA (which uses the method of Pearson and Lipman (1988) PNAS USA 85: 2444-2448).
  • Particular amino acid sequence variants may differ from a reference sequence by insertion, addition, substitution or deletion of 1 amino acid, 2, 3, 4, 5-10, 10-20 or 20-30 amino acids.
  • a variant sequence may comprise the reference sequence with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more residues inserted, deleted or substituted. For example, 5, 10, 15, up to 20, up to 30 or up to 40 residues may be inserted, deleted or substituted.
  • a variant may differ from a reference sequence by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more conservative substitutions.
  • Conservative substitutions involve the replacement of an amino acid with a different amino acid having similar properties. For example, an aliphatic residue may be replaced by another aliphatic residue, a non-polar residue may be replaced by another non-polar residue, an acidic residue may be replaced by another acidic residue, a basic residue may be replaced by another basic residue, a polar residue may be replaced by another polar residue or an aromatic residue may be replaced by another aromatic residue.
  • Conservative substitutions may, for example, be between amino acids within the following groups:
  • Amino acids may be grouped into different classes according to common side-chain properties: a. hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile; b. neutral hydrophilic: Cys, Ser, Thr, Asn, Gln; c. acidic: Asp, Glu; d. basic: His, Lys, Arg; e. residues that influence chain orientation: Gly, Pro; aromatic: Trp, Tyr, Phe. Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • the cells used in the present invention may be any lymphocyte that is useful in adoptive cell therapy, such as a T-cell or a natural killer (NK) cell, an NKT cell, a gamma/delta T-cell or T regulatory cell.
  • the cells may be allogeneic or autologous to the patient.
  • T cells or T lymphocytes are a type of lymphocyte that have a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells), by the presence of a T-cell receptor (TCR) on the cell surface.
  • TCR T-cell receptor
  • TC cells Cytotoxic T cells
  • CTLs destroy virally infected cells and tumor cells, and are also implicated in transplant rejection.
  • CTLs express the CD8 molecule at their surface.
  • Memory T cells are a subset of antigen-specific T cells that persist long-term after an infection has resolved. They quickly expand to large numbers of effector T cells upon re-exposure to their cognate antigen, thus providing the imm“ne sys”em with “memory” against past infections.
  • Memory T cells comprise three subtypes: central memory T cells (TCM cells) and two types of effector memory T cells (TEM cells and TEMRA cells). Memory cells may be either CD4+ or CD8+.
  • Memory T cells typically express the cell surface protein CD45RO.
  • Regulatory T cells (Treg cells), formerly known as suppressor T cells, are crucial for the maintenance of immunological tolerance. Their major role is to shut down T cell-mediated immunity toward the end of an immune reaction and to suppress auto-reactive T cells that escaped the process of negative selection in the thymus.
  • Treg cells Two major classes of CD4 + Treg cells have been described—naturally occurring Treg cells and adaptive Treg cells.
  • Naturally occurring Treg cells also known as CD4 + CD25 + FoxP3 + Treg cells
  • Naturally occurring Treg cells can be distinguished from other T cells by the presence of an intracellular molecule called FoxP3.
  • Adaptive Treg cells also known as Tr1 cells or Th3 cells may originate during a normal immune response.
  • Natural Killer Cells are a type of cytolytic cell which form part of the innate immune system. NK cells provide rapid responses to innate signals from virally infected cells in an MHC independent manner. NK cells (belonging to the group of innate lymphoid cells) are defined as large granular lymphocytes (LGL) and constitute the third kind of cells differentiated from the common lymphoid progenitor generating B and T lymphocytes.
  • LGL large granular lymphocytes
  • therapeutic cells of the invention comprise autologous cells engineered to express a chimeric protein.
  • therapeutic cells of the invention comprise allogeneic cells engineered to express a chimeric protein.
  • Autologous cells expressing chimeric proteins may be advantageous in avoiding graft-versus-host disease (GVHD) due to TCR-mediated recognition of recipient alloantigens.
  • GVHD graft-versus-host disease
  • An aspect of the invention provides a nucleic acid sequence of the invention, encoding any of the chimeric proteins, polypeptides, or proteins described herein (including functional portions and functional variants thereof).
  • polynucleotide “nucleotide”, and “nucleic acid” are intended to be synonymous with each other. It will be understood by a skilled person that numerous different polynucleotides and nucleic acids can encode the same polypeptide as a result of the degeneracy of the genetic code.
  • Nucleic acids according to the invention may comprise DNA or RNA. They may be single stranded or double-stranded. They may also be polynucleotides which include within them synthetic or modified nucleotides. A number of different types of modification to oligonucleotides are known in the art.
  • polynucleotides may be modified by any method available in the art. Such modifications may be carried out in order to enhance the in vivo activity or life span of polynucleotides of interest.
  • variant in relation to a nucleotide sequence include any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) nucleic acid from or to the sequence.
  • the invention also provides a nucleic acid sequence which comprises a nucleic acid sequence encoding a chimeric protein and a further nucleic acid sequence encoding a T-cell receptor (TCR) and/or chimeric antigen receptor (CAR).
  • TCR T-cell receptor
  • CAR chimeric antigen receptor
  • the nucleic acid sequences may be joined by a sequence allowing co-expression of the two or more nucleic acid sequences.
  • the construct may comprise an internal promoter, an internal ribosome entry sequence (IRES) sequence or a sequence encoding a cleavage site.
  • the cleavage site may be self-cleaving, such that when the polypeptide is produced, it is immediately cleaved into the discrete proteins without the need for any external cleavage activity.
  • Various self-cleaving sites are known, including the Foot- and Mouth disease virus (FMDV) and the 2A self-cleaving peptide.
  • the co-expressing sequence may be an internal ribosome entry sequence (IRES).
  • the co-expressing sequence may be an internal promoter.
  • the present invention provides a vector which comprises a nucleic acid sequence or nucleic acid construct of the invention.
  • Such a vector may be used to introduce the nucleic acid sequence(s) or nucleic acid construct(s) into a host cell so that it expresses one or more chimeric protein(s) according to the first aspect of the invention and, optionally, one or more other proteins of interest (POI), for example a TCR or a CAR.
  • POI proteins of interest
  • the vector may, for example, be a plasmid or a viral vector, such as a retroviral vector or a lentiviral vector, or a transposon-based vector or synthetic mRNA.
  • nucleic acids of the present invention may also be used for nucleic acid immunization and gene therapy, using standard gene delivery protocols. Methods for gene delivery are known in the art. See, e.g., U.S. Pat. Nos. 5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties.
  • promoter elements e.g., enhancers
  • promoters regulate the frequency of transcriptional initiation.
  • these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well.
  • the spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another.
  • tk thymidine kinase
  • the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline.
  • a suitable promoter is the immediate early cytomegalovirus (CMV) promoter sequence.
  • CMV immediate early cytomegalovirus
  • This promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operatively linked thereto.
  • Another example of a suitable promoter is Elongation Growth Factor-1 ⁇ (EF-1 ⁇ ).
  • constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, MSCV promoter, MND promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter.
  • SV40 simian virus 40
  • MMTV mouse mammary tumor virus
  • HSV human immunodeficiency virus
  • LTR long terminal repeat
  • MoMuLV promoter MoMuLV promoter
  • MSCV MND promoter
  • MND promoter an avian leukemia virus promoter
  • the vectors can be suitable for replication and integration in eukaryotic cells.
  • Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence.
  • Viral vector technology is well known in the art and is described, for example, in Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York), and in other virology and molecular biology manuals, see also, WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193).
  • the constructs expressed are as shown in SEQ ID NOS:32-65 and 67-79.
  • the nucleic acids are multi-cistronic constructs that permit the expression of multiple transgenes (e.g., chimeric protein and a TCR and/or CAR etc.) under the control of a single promoter.
  • the transgenes e.g., chimeric protein and a TCR and/or CAR etc.
  • the transgenes are separated by a self-cleaving 2A peptide.
  • 2A peptides useful in the nucleic acid constructs of the invention include F2A, P2A, T2A and E2A.
  • a source of cells e.g., immune effector cells, e.g., T cells or NK cells
  • T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors.
  • T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation through a PERCOLLTM gradient or by counterflow centrifugal elutriation.
  • Tumor infiltrating cells are isolated and/or expanded from a tumor, for example by a fragmented, dissected, or enzyme digested tumor biopsy or mass.
  • T cells can be further isolated by positive or negative selection techniques.
  • T cells are isolated by incubation with anti-CD3/anti-CD28-conjugated beads, such as DYNABEADS® M-450 CD3/CD28 T, for a time period sufficient for positive selection of the desired T cells.
  • the time period is about 30 minutes.
  • the time period ranges from 30 minutes to 36 hours or longer and all integer values there between.
  • the time period is at least 1, 2, 3, 4, 5, or 6 hours.
  • the time period is 10 to 24 hours.
  • the incubation time period is 24 hours. Longer incubation times may be used to isolate T cells in any situation where there are few T cells as compared to other cell types, such in isolating tumor infiltrating lymphocytes (TIL) from tumor tissue or from immunocompromised individuals. Further, use of longer incubation times can increase the efficiency of capture of CD8+ T cells. Thus, by simply shortening or lengthening the time T cells are allowed to bind to the CD3/CD28 beads and/or by increasing or decreasing the ratio of beads to T cells (as described further herein), subpopulations of T cells can be preferentially selected for or against at culture initiation or at other time points during the process.
  • TIL tumor infiltrating lymphocytes
  • subpopulations of T cells can be preferentially selected for or against at culture initiation or at other desired time points.
  • multiple rounds of selection can also be used in the context of this invention.
  • Enrichment of a T cell population by negative selection can be accomplished with a combination of antibodies directed to surface markers unique to the negatively selected cells.
  • One method is cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected.
  • a monoclonal antibody cocktail typically includes antibodies to CD14, CD20, CD16, HLA-DR, and CD8.
  • T regulatory cells are depleted by anti-CD25 conjugated beads or other similar method of selection.
  • a specific subpopulation of chimeric protein effector cells that specifically bind to a target antigen can be enriched for by positive selection techniques.
  • effector cells are enriched for by incubation with target antigen-conjugated beads for a time period sufficient for positive selection of the desired abTCR effector cells.
  • the time period is about 30 minutes.
  • the time period ranges from 30 minutes to 36 hours or longer (including all ranges between these values).
  • the time period is at least one, 2, 3, 4, 5, or 6 hours.
  • the time period is 10 to 24 hours.
  • the incubation time period is 24 hours.
  • T cells for stimulation can also be frozen after a washing step. After the washing step that removes plasma and platelets, the cells may be suspended in a freezing solution. While many freezing solutions and parameters are known in the art and will be useful in this context, one method involves using PBS containing 20% DMSO and 8% human serum albumin, or culture media containing 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin and 7.5% DMSO, or 31.25% Plasmalyte-A, 31.25% Dextrose 5%, 0.45% NaCl, 10% Dextran 40 and 5% Dextrose, 20% Human Serum Albumin, and 7.5% DMSO or other suitable cell freezing media containing for example, Hespan and PlasmaLyte A, the cells then are frozen to ⁇ 80° C. at a rate of 1° per minute and stored in the vapor phase of a liquid nitrogen storage tank. Other methods of controlled freezing may be used as well as uncontrolled freezing immediately at ⁇ 20° C. or in liquid nitrogen.
  • the immune effector cell can be an allogeneic immune effector cell, e.g., T cell or NK cell.
  • a T cell described herein can be, e.g., engineered such that it does not express a functional HLA on its surface.
  • a T cell described herein can be engineered such that cell surface expression HLA, e.g., HLA class 1 and/or HLA class II, is downregulated.
  • HLA e.g., HLA class 1 and/or HLA class II
  • downregulation of HLA may be accomplished by reducing or eliminating expression of beta-2 microglobulin (B2M).
  • the T cell can lack a functional TCR and a functional HLA, e.g., HLA class I and/or HLA class II.
  • Modified T cells that lack expression of a functional TCR and/or HLA can be obtained by any suitable means, including a knock out or knock down of one or more subunit of TCR or HLA.
  • the T cell can include a knock down of TCR and/or HLA using siRNA, shRNA, clustered regularly interspaced short palindromic repeats (CRISPR) transcription-activator like effector nuclease (TALEN), or zinc finger endonuclease (ZFN).
  • siRNA siRNA
  • shRNA clustered regularly interspaced short palindromic repeats
  • TALEN clustered regularly interspaced short palindromic repeats
  • ZFN zinc finger endonuclease
  • the allogeneic cell can be a cell which does not expresses or expresses at low levels an inhibitory molecule, e.g. a cell engineered by any method described herein.
  • the cell can be a cell that does not express or expresses at low levels an inhibitory molecule, e.g., that can decrease the ability of a chimeric protein-expressing cell to mount an immune effector response.
  • inhibitory molecules examples include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAGS, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, Gal9, adenosine, and TGFR beta. Inhibition of an inhibitory molecule, e.g., by inhibition at the DNA, RNA or protein level, can optimize a CAR-expressing cell performance.
  • an inhibitory nucleic acid e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., an siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used.
  • an inhibitory nucleic acid e.g., a dsRNA, e.g., an siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used.
  • CRISPR clustered regularly interspaced short palindromic repeats
  • TALEN transcription-activator like effector nu
  • T cells may be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 20060121005.
  • the T cells of the invention may be expanded by contact with a surface having attached thereto an agent that stimulates a CD3/TCR complex associated signal and a ligand that stimulates a costimulatory molecule on the surface of the T cells.
  • T cell populations may be stimulated as described herein, such as by contact with an anti-CD3 antibody, or antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore.
  • a ligand that binds the accessory molecule is used for co-stimulation of an accessory molecule on the surface of the T cells.
  • a population of T cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T cells.
  • an anti-CD3 antibody and an anti-CD28 antibody can be used.
  • an anti-CD28 antibody include 9.3, B-T3, XR-CD28 (Diaclone, Besancon, France) can be used as can other methods commonly known in the art (Berg et al., Transplant Proc. 30(8):3975-3977, 1998; Haanen et al., J. Exp. Med. 190(9):13191328, 1999; Garland et al., J. Immunol Meth. 227(1-2):53-63, 1999).
  • the expansion can be performed using non-specific T-cell receptor stimulation in the presence of interleukin-2 (IL-2) or interleukin-15 (IL-15).
  • the non-specific T-cell receptor stimulus can include, for example, an anti-CD3 antibody, such as about 30 ng/ml of OKT3, a mouse monoclonal anti-CD3 antibody (commercially available from Ortho-McNeil, Raritan, N.J. or Miltenyi Biotech, Auburn, Calif.) or UHCT-1 (commercially available from BioLegend, San Diego, Calif., USA).
  • Chimeric protein cells can be expanded in vitro by including one or more antigens, including antigenic portions thereof, such as epitope(s), of a cancer, which can be optionally expressed from a vector, such as a human leukocyte antigen A2 (HLA-A2) binding peptide, e.g., 0.3 .mu.M MART-1:26-35 (27L) or gp100:209-217 (210M), optionally in the presence of a T-cell growth factor, such as 300 IU/mL IL-2 or IL-15.
  • HLA-A2 human leukocyte antigen A2
  • T-cell growth factor such as 300 IU/mL IL-2 or IL-15.
  • Chimeric protein cells may also be rapidly expanded by re-stimulation with the same antigen(s) of the cancer pulsed onto HLA-A2-expressing antigen-presenting cells.
  • the chimeric protein cells can be further stimulated with, e.g., example, irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2.
  • the stimulation occurs as part of the expansion.
  • the expansion occurs in the presence of irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2.
  • the cell culture medium comprises IL-2. In some embodiments, the cell culture medium comprises about 1000 IU/mL, about 1500 IU/mL, about 2000 IU/mL, about 2500 IU/mL, about 3000 IU/mL, about 3500 IU/mL, about 4000 IU/mL, about 4500 IU/mL, about 5000 IU/mL, about 5500 IU/mL, about 6000 IU/mL, about 6500 IU/mL, about 7000 IU/mL, about 7500 IU/mL, or about 8000 IU/mL, or between 1000 and 2000 IU/mL, between 2000 and 3000 IU/mL, between 3000 and 4000 IU/mL, between 4000 and 5000 IU/mL, between 5000 and 6000 IU/mL, between 6000 and 7000 IU/mL, between 7000 and 8000 IU/mL, or between 8000 IU/mL of IL-2.
  • the cell culture medium comprises OKT3 antibody. In some embodiments, the cell culture medium comprises about 0.1 ng/mL, about 0.5 ng/mL, about 1 ng/mL, about 2.5 ng/mL, about 5 ng/mL, about 7.5 ng/mL, about 10 ng/mL, about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 50 ng/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 200 ng/mL, about 500 ng/mL, about 1 ⁇ g/mL or between 0.1 ng/mL and 1 ng/mL, between 1 ng/mL and 5 ng/mL, between 5 ng/mL and 10 ng/mL, between 10 ng/mL,
  • a combination of IL-2, IL-7, IL-15, and/or IL-21 are employed as a combination during the expansion.
  • IL-2, IL-7, IL-15, and/or IL-21 as well as any combinations thereof can be included during the expansion.
  • a combination of IL-2, IL-15, and IL-21 are employed as a combination during the expansion.
  • IL-2, IL-15, and IL-21 as well as any combinations thereof can be included.
  • the expansion can be conducted in a supplemented cell culture medium comprising IL-2, OKT-3, and antigen-presenting feeder cells.
  • the expansion culture media comprises about 500 IU/mL of IL-15, about 400 IU/mL of IL-15, about 300 IU/mL of IL-15, about 200 IU/mL of IL-15, about 180 IU/mL of IL-15, about 160 IU/mL of IL-15, about 140 IU/mL of IL-15, about 120 IU/mL of IL-15, or about 100 IU/mL of IL-15, or about 500 IU/mL of IL-15 to about 100 IU/mL of IL-15, or about 400 IU/mL of IL-15 to about 100 IU/mL of IL-15 or about 300 IU/mL of IL-15 to about 100 IU/mL of IL-15 or about 200 IU/mL of IL-15, or about 180 IU/mL of IL-15.
  • the expansion culture media comprises about 20 IU/mL of IL-21, about 15 IU/mL of IL-21, about 12 IU/mL of IL-21, about 10 IU/mL of IL-21, about 5 IU/mL of IL-21, about 4 IU/mL of IL-21, about 3 IU/mL of IL-21, about 2 IU/mL of IL-21, about 1 IU/mL of IL-21, or about 0.5 IU/mL of IL-21, or about 20 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 15 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 12 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 10 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 5 IU/mL of IL-21 to about 1 IU/m
  • the antigen-presenting feeder cells are PBMCs.
  • the ratio of chimeric protein cells to PBMCs and/or antigen-presenting cells in the expansion is about 1 to 25, about 1 to 50, about 1 to 100, about 1 to 125, about 1 to 150, about 1 to 175, about 1 to 200, about 1 to 225, about 1 to 250, about 1 to 275, about 1 to 300, about 1 to 325, about 1 to 350, about 1 to 375, about 1 to 400, or about 1 to 500, or between 1 to 50 and 1 to 300, or between 1 to 100 and 1 to 200.
  • the primary stimulatory signal and the costimulatory signal for the T cell may be provided by different protocols.
  • the agents providing each signal may be in solution or coupled to a surface.
  • the agents may be coupled to the same su“fac” (i.e., in “cis” formation) or to separate sur“aces “i.e., in “trans” formation).
  • one agent may be coupled to a surface and the other agent in solution.
  • the agent providing the costimulatory signal is bound to a cell surface and the agent providing the primary activation signal is in solution or coupled to a surface. In certain aspects, both agents can be in solution.
  • the agents may be in soluble form, and then cross-linked to a surface, such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents.
  • a surface such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents.
  • aAPCs artificial antigen presenting cells
  • the two agents are immobilized on beads, either on the same bead, i.e., “cis,” or to separate beads, i.e., “trans.”
  • the agent providing the primary activation signal is an anti-CD3 antibody or an antigen-binding fragment thereof and the agent providing the costimulatory signal is an anti-CD28 antibody or antigen-binding fragment thereof; and both agents are co-immobilized to the same bead in equivalent molecular amounts.
  • a 1:1 ratio of each antibody bound to the beads for CD4+ T cell expansion and T cell growth is used.
  • a ratio of anti CD3:CD28 antibodies bound to the beads is used such that an increase in T cell expansion is observed as compared to the expansion observed using a ratio of 1:1. In one particular aspect an increase of from about 1 to about 3 fold is observed as compared to the expansion observed using a ratio of 1:1.
  • the ratio of CD3:CD28 antibody bound to the beads ranges from 100:1 to 1:100 and all integer values there between. In one aspect of the present invention, more anti-CD28 antibody is bound to the particles than anti-CD3 antibody, i.e., the ratio of CD3:CD28 is less than one. In certain aspects of the invention, the ratio of anti CD28 antibody to anti CD3 antibody bound to the beads is greater than 2:1.
  • a 1:100 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:75 CD3:CD28 ratio of antibody bound to beads is used. In a further aspect, a 1:50 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:30 CD3:CD28 ratio of antibody bound to beads is used. In one preferred aspect, a 1:10 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:3 CD3:CD28 ratio of antibody bound to the beads is used. In yet one aspect, a 3:1 CD3:CD28 ratio of antibody bound to the beads is used.
  • Ratios of particles to cells from 1:500 to 500:1 and any integer values in between may be used to stimulate T cells or other target cells.
  • the ratio of particles to cells may depend on particle size relative to the target cell. For example, small sized beads could only bind a few cells, while larger beads could bind many.
  • the ratio of cells to particles ranges from 1:100 to 100:1 and any integer values in-between and in further aspects the ratio comprises 1:9 to 9:1 and any integer values in between, can also be used to stimulate T cells.
  • the ratio of anti-CD3- and anti-CD28-coupled particles to T cells that result in T cell stimulation can vary as noted above, however certain preferred values include 1:100, 1:50, 1:40, 1:30, 1:20, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, and 15:1 with one preferred ratio being at least 1:1 particles per T cell.
  • a ratio of particles to cells of 1:1 or less is used.
  • a preferred particle:cell ratio is 1:5.
  • the ratio of particles to cells can be varied depending on the day of stimulation.
  • the ratio of particles to cells is from 1:1 to 10:1 on the first day and additional particles are added to the cells every day or every other day thereafter for up to 10 days, at final ratios of from 1:1 to 1:10 (based on cell counts on the day of addition).
  • the ratio of particles to cells is 1:1 on the first day of stimulation and adjusted to 1:5 on the third and fifth days of stimulation.
  • particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:5 on the third and fifth days of stimulation.
  • the ratio of particles to cells is 2:1 on the first day of stimulation and adjusted to 1:10 on the third and fifth days of stimulation.
  • particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:10 on the third and fifth days of stimulation.
  • ratios will vary depending on particle size and on cell size and type.
  • the most typical ratios for use are in the neighborhood of 1:1, 2:1 and 3:1 on the first day.
  • the cells such as T cells
  • the cells are combined with agent-coated beads, the beads and the cells are subsequently separated, and then the cells are cultured.
  • the agent-coated beads and cells prior to culture, are not separated but are cultured together.
  • the beads and cells are first concentrated by application of a force, such as a magnetic force, resulting in increased ligation of cell surface markers, thereby inducing cell stimulation.
  • Retrovirus-based gene delivery is a mature, well-characterized technology, which has been used to permanently integrate CARs into the host cell genome (Scholler J., e.g. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci. Transl. Med. 2012; 4:132ra53; Rosenberg S. A. et al., Gene transfer into humans—immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N. Engl. J. Med. 1990; 323:570-578)
  • Non-viral DNA transfection methods can also be used.
  • Singh et al describes use of the Sleeping Beauty (SB) transposon system developed to engineer CAR T cells (Singh H., et al., Redirecting specificity of T-cell populations for CD19 using the Sleeping Beauty system. Cancer Res. 2008; 68:2961-2971) and is being used in clinical trials (see e.g., ClinicalTrials.gov: NCT00968760 and NCT01653717).
  • SB Sleeping Beauty
  • SB100X hyperactive transposase
  • MMs L. et al. Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat. Genet. 2009; 41:753-761
  • multiple transgenes can be delivered from multicistronic single plasmids (e.g., Thokala R.
  • Morita et al describes the piggyBac transposon system to integrate larger transgenes (Morita D. et al., Enhanced expression of anti-CD19 chimeric antigen receptor in piggyBac transposon-engineered T cells. Mol. Ther. Methods Clin. Dev. 2017; 8:131-140)
  • Nakazawa et al. describes use of the system to generate EBV-specific cytotoxic T-cells expressing HER2-specific chimeric antigen receptor (Nakazawa Y et al, PiggyBac-mediated cancer immunotherapy using EBV-specific cytotoxic T-cells expressing HER2-specific chimeric antigen receptor. Mol. Ther. 2011; 19:2133-2143).
  • Manuri et al used the system to generate CD-19 specific T cells (Manuri P. V. R. et al., piggyBac transposon/transposase system to generate CD19-specific T cells for the treatment of B-lineage malignancies. Hum. Gene Ther. 2010; 21:427-437).
  • Transposon technology is easy and economical.
  • One potential drawback is the longer expansion protocols currently employed may result in T cell differentiation, impaired activity and poor persistence of the infused cells.
  • Monjezi et al describe development minicircle vectors that minimize these difficulties through higher efficiency integrations (Monjezi R. et al., Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors. Leukemia. 2017; 31:186-194). These transposon technologies can be used for chimeric proteins of the invention.
  • the present invention also relates to a pharmaceutical composition containing a vector or a chimeric protein expressing cell of the invention together with a pharmaceutically acceptable carrier, diluent or excipient, and optionally one or more further pharmaceutically active polypeptides and/or compounds.
  • a pharmaceutical composition comprising a chimeric protein described above and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising a nucleic acid encoding a chimeric protein according to any of the embodiments described above and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition is provided comprising an effector cell expressing a chimeric protein described above and a pharmaceutically acceptable carrier.
  • Such a formulation may, for example, be in a form suitable for intravenous infusion.
  • pharmaceutically acceptable or “pharmacologically compatible” is meant a material that is not biologically or otherwise undesirable, e.g., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained.
  • Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration.
  • An aspect of the invention provides a population of modified T cells expressing a recombinant chimeric protein.
  • a suitable population may be produced by a method described above.
  • the population of modified T cells may be for use as a medicament.
  • a population of modified T cells as described herein may be used in cancer immunotherapy therapy, for example adoptive T cell therapy.
  • aspects of the invention provide the use of a population of modified T cells as described herein for the manufacture of a medicament for the treatment of cancer, a population of modified T cells as described herein for the treatment of cancer, and a method of treatment of cancer may comprise administering a population of modified T cells as described herein to an individual in need thereof.
  • the population of modified T cells may be autologous i.e. the modified T cells were originally obtained from the same individual to whom they are subsequently administered (i.e. the donor and recipient individual are the same).
  • a suitable population of modified T cells for administration to the individual may be produced by a method comprising providing an initial population of T cells obtained from the individual, modifying the T cells to express a cAMP PDE or fragment thereof and an antigen receptor which binds specifically to cancer cells in the individual, and culturing the modified T cells.
  • the population of modified T cells may be allogeneic i.e. the modified T cells were originally obtained from a different individual to the individual to whom they are subsequently administered (i.e. the donor and recipient individual are different).
  • the donor and recipient individuals may be HLA matched to avoid GVHD and other undesirable immune effects.
  • a suitable population of modified T cells for administration to a recipient individual may be produced by a method comprising providing an initial population of T cells obtained from a donor individual, modifying the T cells to express a chimeric protein which binds specifically to cancer cells in the recipient individual, and culturing the modified T cells.
  • the recipient individual may exhibit a T cell mediated immune response against cancer cells in the recipient individual. This may have a beneficial effect on the cancer condition in the individual.
  • Cancer conditions may be characterized by the abnormal proliferation of malignant cancer cells and may include leukaemias, such as AML, CML, ALL and CLL, lymphomas, such as Hodgkin lymphoma, non-Hodgkin lymphoma and multiple myeloma, and solid cancers such as sarcomas, skin cancer, melanoma, bladder cancer, brain cancer, breast cancer, uterus cancer, ovary cancer, prostate cancer, lung cancer, colorectal cancer, cervical cancer, liver cancer, head and neck cancer, oesophageal cancer, pancreas cancer, renal cancer, adrenal cancer, stomach cancer, testicular cancer, cancer of the gall bladder and biliary tracts, thyroid cancer, thymus cancer, cancer of bone, and cerebral cancer, as well as cancer of unknown primary (CUP).
  • leukaemias such as AML, CML, ALL and CLL
  • lymphomas such as Hodgkin lymphoma, non-Hodgkin lymphoma and multiple mye
  • Cancer cells within an individual may be immunologically distinct from normal somatic cells in the individual (i.e. the cancerous tumor may be immunogenic).
  • the cancer cells may be capable of eliciting a systemic immune response in the individual against one or more antigens expressed by the cancer cells.
  • the tumor antigens that elicit the immune response may be specific to cancer cells or may be shared by one or more normal cells in the individual.
  • An individual suitable for treatment as described above may be a mammal, such as a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon), an ape (e.g. gorilla, chimpanzee, orangutan, gibbon), or a human.
  • a rodent e.g. a guinea pig, a hamster, a rat, a mouse
  • murine e.g. a mouse
  • canine e.g. a dog
  • feline e.g. a cat
  • equine e.g. a horse
  • the individual is a human.
  • non-human mammals especially mammals that are conventionally used as models for demonstrating therapeutic efficacy in humans (e.g. murine, primate, porcine, canine, or rabbit animals) may be employed.
  • therapeutically effective amount refers to an amount of a chimeric protein or composition comprising a chimeric protein as disclosed herei”, eff”ctive to “treat” a disease or disorder in an individual.
  • the therapeutically effective amount of a chimeric protein or composition comprising a chimeric protein as disclosed herein can reduce the number of cancer cells; reduce the tumor size or weight; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • a chimeric protein or composition comprising a chimeric protein as disclosed herein can prevent growth and/or kill existing cancer cells, it can be cytostatic and/or cytotoxic.
  • the therapeutically effective amount is a growth inhibitory amount. In some embodiments, the therapeutically effective amount is an amount that improves progression free survival of a patient.
  • the therapeutically effective amount of a chimeric protein or composition comprising a chimeric protein as disclosed herein can reduce the number of cells infected by the pathogen; reduce the production or release of pathogen-derived antigens; inhibit (i.e., slow to some extent and preferably stop) spread of the pathogen to uninfected cells; and/or relieve to some extent one or more symptoms associated with the infection.
  • the therapeutically effective amount is an amount that extends the survival of a patient.
  • T and NK cells expressing chimeric proteins for use in the methods of the present may either be created ex vivo eithe' from a patient's own peripheral blood (autologous), or in the setting of a haematopoietic stem cell transplant from donor peripheral blood (allogenic), or peripheral blood from an unconnected donor (allogenic).
  • T-cells or NK cells may be derived from ex-vivo differentiation of inducible progenitor cells or embryonic progenitor cells to T-cells or NK cells.
  • T-cells expressing a chimeric protein and, optionally, a CAR and/or TCR are generated by introducing DNA or RNA coding for the chimeric protein and, optionally, a CAR and/or TCR, by one of many means including transduction with a viral vector, transfection with DNA or RNA.
  • T or NK cells expressing a chimeric protein of the present invention and, optionally, expressing a TCR and/or CAR may be used for the treatment of haematological cancers or solid tumors.
  • a method for the treatment of disease relates to the therapeutic use of a vector or cell, including a T or NK cell, of the invention.
  • the vector, or T or NK cell may be administered to a subject having an existing disease or condition in order to lessen, reduce or improve at least one symptom associated with the disease and/or to slow down, reduce or block the progression of the disease.
  • the method of the invention may cause or promote T-cell mediated killing of cancer cells.
  • the vector, or T or NK cell according to the present invention may be administered to a patient with one or more additional therapeutic agents.
  • the one or more additional therapeutic agents can be co-administered to the patient.
  • co-administering is meant administering one or more additional therapeutic agents and the vector, or T or NK cell of the present invention sufficiently close in time such that the vector, or T or NK cell can enhance the effect of one or more additional therapeutic agents, or vice versa.
  • the vectors or cells can be administered first and the one or more additional therapeutic agents can be administered second, or vice versa.
  • the vectors or cells and the one or more additional therapeutic agents can be administered simultaneously.
  • One co-administered therapeutic agent that may be useful is IL-2, as this is currently used in existing cell therapies to boost the activity of administered cells.
  • IL-2 treatment is associated with toxicity and tolerability issues.
  • the addition of the engineered protein to a subject induces cytokine secretion. In some embodiments, the addition of CoStAR to a subject induces cytokine secretion. In some embodiments, the cytokine secretion lowers cytokine levels in the subject, including but not limited to IL-2. In some embodiments, the cytokine secretion following CoStAR exposure results in no detectable IL-2 in the subject. In some embodiments, the addition of the engineered protein to a subject reduces or eliminates the need for administration of exogenous IL-2. In some embodiments, the addition of the CoStAR to a subject reduces or eliminates the need for administration of exogenous IL-2.
  • tumor cell killing potency is characterized by flow cytometry to enumerate T cells and target cells and plate-based fluorescence or luminescence to measure percent killing.
  • cytokine secretion potency is characterized at the single cell level by flow cytometry and ELISA/MSD to characterize the population.
  • proliferation potency is determined by flow cytometry to characterize the population.
  • TIL potency may be determined by additional analytes, memory phenotype, cytotoxicity using cell lines, cytotoxicity using a patient specific tumor, a cytokine panel, cell proliferation and/or cellular composition.
  • the chimeric protein effector cells can be allogeneic or autologous to the patient.
  • allogeneic cells are further genetically modified, for example by gene editing, so as to minimize or prevent GVHD and/or a patient's immune response against the chimeric protein cells.
  • the chimeric protein effector cells are used to treat cancers and neoplastic diseases associated with a target antigen.
  • Cancers and neoplastic diseases that may be treated using any of the methods described herein include tumors that are not vascularized, or not yet substantially vascularized, as well as vascularized tumors.
  • the cancers may comprise non-solid tumors (such as hematological tumors, for example, leukemias and lymphomas) or may comprise solid tumors.
  • Types of cancers to be treated with the chimeric protein effector cells of the invention include, but are not limited to, carcinoma, blastoma, and sarcoma, and certain leukemia or lymphoid malignancies, benign and malignant tumors, and malignancies e.g., sarcomas, carcinomas, and melanomas.
  • carcinoma blastoma
  • sarcoma certain leukemia or lymphoid malignancies
  • benign and malignant tumors e.g., sarcomas, carcinomas, and melanomas.
  • malignancies e.g., sarcomas, carcinomas, and melanomas.
  • adult tumors/cancers and pediatric tumors/cancers are also included.
  • Hematologic cancers are cancers of the blood or bone marrow.
  • hematological (or hematogenous) cancers include leukemias, including acute leukemias (such as acute lymphocytic leukemia, acute myelocytic leukemia, acute myelogenous leukemia and myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia), chronic leukemias (such as chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, l'mphoma, Hodgkin's dise'se, non-Hodgkin's lymphoma (indolent and high grade forms), multiple myeloma, plasmacyt'ma, Waldenstrom's macroglobulinemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia and
  • Solid tumors are abnormal masses of tissue that usually do not contain cysts or liquid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors, such as sarcomas and carcinomas, include adrenocortical carcinoma, cholangiocarcinoma, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, and other sarcomas, synovioma, mes'thelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, stomach cancer, lymphoid malignancy, pancreatic cancer, breast cancer, lung cancers, ovarian cancer, prostate cancer, hepatocellular carcinoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma,
  • an immunologically effective amount When “an immunologically effective amount,” “an anti-tumor effective amount,” “a tumor-inhibiting effective amount,” or “therapeutic amount” is indicated, the 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 the T cells described herein may be administered at a dosage of 10 4 to 10 9 cells/kg body weight, in some instances 10 5 to 10 6 cells/kg body weight, including all integer values within those ranges. 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).
  • a chimeric protein-expressing cell described herein may be used in combination with other known agents and therapie.
  • “Administered” “in combination”, as used herein, means that two (or more) different treatments are delivered to the subject during the cours' of the subject's affliction with the disorder, e.g., the two or more treatments are delivered after the subject has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons.
  • the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referr“d to herein”s “s“multaneous” or “con”urrent delivery”.
  • the delivery of one treatment ends before the delivery of the other treatment begins.
  • the treatment is more effective because of combined administration.
  • the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment.
  • delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other.
  • the effect of the two treatments can be partially additive, wholly additive, or greater than additive.
  • the delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
  • a chimeric protein-expressing cell described herein and the at least one additional therapeutic agent can be administered simultaneously, in the same or in separate compositions, or sequentially.
  • the CAR-expressing cell described herein can be administered first, and the additional agent can be administered second, or the order of administration can be reversed.
  • the chimeric protein therapy and/or other therapeutic agents, procedures or modalities can be administered during periods of active disorder, or during a period of remission or less active disease.
  • the chimeric protein therapy can be administered before the other treatment, concurrently with the treatment, post-treatment, or during remission of the disorder.
  • the therapy and the additional agent can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the administered amount or dosage of the chimeric protein therapy, the additional agent (e.g., second or third agent), or all is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy.
  • the amount or dosage of the chimeric protein therapy, the additional agent (e.g., second or third agent), or all, that results in a desired effect is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower) than the amount or dosage of each agent used individually, e.g., as a monotherapy, required to achieve the same therapeutic effect.
  • a chimeric protein-expressing cell described herein may be used in a treatment regimen in combination with surgery, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, cytokines, and irradiation, peptide vaccine, such as that described in Izumoto et al. 2008 J Neurosurg 108:963-971.
  • immunosuppressive agents such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies
  • immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506,
  • compounds of the present invention are combined with other therapeutic agents, such as other anti-cancer agents, anti-allergic agents, anti-nausea agents (or anti-emetics), pain relievers, cytoprotective agents, and combinations thereof.
  • other therapeutic agents such as other anti-cancer agents, anti-allergic agents, anti-nausea agents (or anti-emetics), pain relievers, cytoprotective agents, and combinations thereof.
  • a chimeric protein-expressing cell described herein can be used in combination with a chemotherapeutic agent.
  • chemotherapeutic agents include an anthracycline (e.g., doxorubicin (e.g., liposomal doxorubicin)), a vinca alkaloid (e.g., vinblastine, vincristine, vindesine, vinorelbine), an alkylating agent (e.g., cy53acarbazineide, decarbazine, melphalan, ifosfamide, temozolomide), an immune cell antibody (e.g., alemtuzamab, gemtuzumab, rituximab, ofatumumab, tositumomab, brentuximab), an antimetabolite (including, e.g., folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deamina
  • chemotherapeutic agents considered for use in combination therapies include busulfan (Myleran®), busulfan injection (Busulfex®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), daunorubicin hydrochloride (Cerubidine®), daunorubicin citrate liposome injection (DaunoXome®), dexamethasone, doxorubicin hydrochloride (Adriamycin®, Rubex®), etoposide (Vepesid®), fludarabine phosphate (Fludara®), hydroxyurea (Hydrea®), Idarubicin (Idamycin®), mitoxantrone (Novantrone®), Gemtuzumab Ozogamicin
  • general chemotherapeutic agents considered for use in combination therapies include anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4-pentoxycarbonyl-5-deoxy-5-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), dacarbazine (DTIC-Dome®), d
  • Treatments can be evaluated, for example, by tumor regression, tumor weight or size shrinkage, time to progression, duration of survival, progression free survival, overall response rate, duration of response, quality of life, protein expression and/or activity.
  • Approaches to determining efficacy of the therapy can be employed, including for example, measurement of response through radiological imaging.
  • TCR Incorporated Antigen Agnostic Receptors TIAARs
  • Table 2 provides exemplary, non-limiting examples of components of TCR incorporated antigen agnostic receptors (TIAARs) of the invention.
  • Table 3 shows exemplary arrangements of the components.
  • TCR incorporated antigen agnostic receptor (TIAAR) components Code Signal peptide Tag ECD_TMD ICD Costim pIB1026 CD3D Myc CD3D N/A CD28-CD40 pIB1027 CD3E FLAG CD3E N/A CD28-CD40 pIB1028 CD3G Myc CD3G N/A CD28-CD40 pIB1029 CD3Z Myc IC CD3Z N/A CD28-CD40 pIB1030 CD8A Myc hTRDC N/A CD28-CD40 pIB1031 CD8A FLAG hTRGC1 N/A CD28-CD40 pIB1032 CD8A Myc mTRAC N/A CD28-CD40 pIB1033 CD8A FLAG mTRBC1 N/A CD28-CD40 pIB1046 CD8A ⁇ 2 Myc and hTRDC_hTRGC1 N/A CD28-CD40 FLAG
  • TCR incorporated antigen agnostic receptors Code Description pIB1026 CD3D_CD3D_CD28CD40 pIB1027 CD3E_CD3E_CD28CD40 pIB1028 CD3G_CD3G_CD28CD40 pIB1029 CD3Z_CD3Z_CD28CD40_Myc pIB1030 CD8A_hTRDC_CD28CD40 pIB1031 CD8A_hTRGC1_CD28CD40 pIB1032 CD8A_mTRAC_CD28CD40 pIB1033 CD8A_mTRBC1_CD28CD40 pIB1046 CD8A_hTRDC_CD28CD40-T2A-CD8a_hTRGC1_CD28CD40 pIB1047 CD8A_mTRAC_CD28CD40-T2A-CD8A_mTRBC1_CD28CD40 pIB1048 CD3D_
  • Table 4 provides exemplary, non-limiting examples of components of constitutive costimulatory proteins of the invention.
  • Table 5 shows the exemplary arrangements of the components.
  • Table 6 provides exemplary, non-limiting examples of inducible costimulatory receptors of the invention.
  • Table 7 shows exemplary arrangements of the components.
  • Components may include a signal peptide (SP), a TCR clustering domain (CD) and/or a signaling domain (SD).
  • SP signal peptide
  • CD TCR clustering domain
  • SD signaling domain
  • any one or more of the arrangements below are contemplated:
  • T cells from 2 healthy donors were either modified to express the constructs tested or left non-transduced (NTD) at MOI 10.
  • NTD non-transduced
  • effector T cells were thawed and resuspended at 1 ⁇ 10 6 cells/mL in T cell media (TCM) without IL2 and incubated overnight at 37° C. with 5% CO2.
  • TCM T cell media
  • effectors and Ba/F3-OKT3 targets were collected and counted using a ViCELL BLU as per manufacturer's instructions.
  • T cells were then cocultured with B a/F3 OKT3 targets at the 10:1, 1:1 and 1:10 E:T (effector:target) ratios overnight.
  • the invention includes modifying components of the TCR complex and associated signaling adaptors (such as, for example, in a TCR incorporated antigen agnostic receptor “TIAAR”), identifying transmembrane domains (TMDs) and modifications that enable constitutive activation of receptors (“constitutive”) and utilizing antibodies to induce activation of the receptor (“inducible”).
  • TCR complex and associated signaling adaptors such as, for example, in a TCR incorporated antigen agnostic receptor “TIAAR”
  • TMDs transmembrane domains
  • the scFV targeting co-stimulatory or inhibitor receptors and ligands.
  • the scFV are derived from antibodies targeting co-stimulatory or inhibitory molecules expressed on immune cells.
  • TIAAR TCR incorporated list of constructs Signal ECD_ GOI Code Concept peptide Tag TMD ICD Costim description pIB1026 TIAAR CD3D Myc CD3D N/A CD28- CD3D_CD3D_ CD40 CD28CD40 pIB1027 TIAAR CD3E FLAG CD3E N/A CD28- CD3E_CD3E_ CD40 CD28CD40 pIB1028 TIAAR CD3G Myc CD3G N/A CD28- CD3G_CD3G_ CD40 CD28CD40 pIB1029 TIAAR CD3Z Myc IC CD3Z N/A CD28- CD3Z_CD3Z_ CD40 CD28CD40_Myc pIB1030 TIAAR CD8A Myc hTRDC N/A CD28- CD8A_hTRDC_ CD40 CD28CD40 pIB1031 TIAAR CD8A FLAG hTRGC1 N/
  • Cytokine production (Bcl-xL, IL2, IFNgamma and TNFalpha) from genetically modified and non-transduced T cells (NTD) after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only) ( FIG. 2 ).
  • Bcl-xL, IL2, IFNgamma and TNFalpha production from genetically modified T cells as compared to NTD cells in Donor 1.
  • IFN ⁇ production and comparable or lower levels of Bcl-xL, IL2, IFNgamma and TNFalpha production in genetically modified as compared to NTD cells in Donor 2.
  • T cell counts from CD45+ (TIARR)) and activation marker expression (41BB from CD45+ and CD69 from CD45+) from genetically modified and non-transduced T cells (NTD) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only) FIGS. 3 A- 3 B ).
  • T cell counts from CD45+ (TIARR) T cell counts from CD45+
  • activation marker expression 41BB from CD45+ and CD69 from CD45+
  • NTD non-transduced T cells
  • Cytokine production (Bcl-xL, IL2, IFNgamma and TNFalpha) from genetically modified and non-transduced T cells (NTD) after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only) ( FIG. 4 ).
  • Bcl-xL, IL2, and TNFalpha production were increased or comparable Bcl-xL, IL2, and TNFalpha production and comparable or lower levels of IFNgamma from genetically modified T cells as compared to NTD cells in Donor 1.
  • IFNgamma and IL2 production and comparable or lower levels of Bcl-xL and TNFalpha production in genetically modified as compared to NTD cells in Donor 2.
  • T cell counts from CD45+(LZ) and activation marker expression (41BB from CD45+ and CD69 from CD45+) from genetically modified and non-transduced T cells (NTD) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only) FIGS. 5 A- 5 B ).
  • T cell counts in genetically modified as compared to NTD cells in Donor 1 There was increased 41BB and CD69 expression in genetically modified T cells as compared to NTD cells in Donor 1.
  • T cell counts as compared to NTD cells in Donor 2 an increase in 41BB expression in genetically modified T cells as compared to NTD cells, and similar or decreased expression of CD169 in genetically modified T cells as compared to NTD cells in Donor 2.
  • Cytokine production (Bcl-xL, IL2, IFNg and TNFa) from genetically modified and non-transduced T cells (NTD) after overnight stimulation with either Ba/F3 OKT3 targets or Ba/F3 OKT3 targets with 10 ug/mL pembrolizumab or left unstimulated (i.e., T cells only) ( FIG. 6 ).
  • Bcl-xL, IL2, IFNgamma and TNFalpha production from genetically modified T cells in the presence of Ba/F3 OKT3 and pembrolizumab as compared to conditions with Ba/F3 OKT3 stimulation alone and NTD cells in both Donor 1 and Donor 2.
  • An engineered protein having the sequence of SEQ ID NO: 166 will be transfected into a Tumor Infiltrating Lymphocyte (TIL) cell using standard procedures by incorporating vectors. This cell will then be used to generate a population of TIL cells expressing those proteins. The population will be derived through detecting expression of the protein on the surface of the cells transfected to express the two proteins, and selecting cells which are identified as expressing those proteins. Through this process, the population of cells will be enriched for those expressing the protein. Following enrichment, the TIL cells will be administered at a therapeutic amount to a patient as a therapeutic treatment for cancer.
  • TIL Tumor Infiltrating Lymphocyte
  • CoStAR constructs were transduced into TIL cells from tumor digests.
  • the efficiency of CEA or FOLR expression on tumor digests at Day 1 is as shown in FIGS. 20 - 22 , and in Table 13.
  • the efficiency of CEA or FOLR expression by Day 21 is as shown in FIGS. 12 - 14 .
  • the amount of CEA and FOLR expression was significantly higher by 21 days of treatment compared to 1 day of treatment.
  • the transduction was performed using the following materials and methodology:
  • the tumor digest was thawed (no stimulation) in media 1.
  • 0.5-1e6 cells were seeded per condition (6 conditions). Phenotype was recorded were applicable.
  • TCM base media GMP/TCM media+25 mM HEPES+25 ⁇ M 2-Mercaptoethanol
  • TIL cells expressing CoStAR constructs then underwent a screen to assess function.
  • the results of this screen are as shown in FIGS. 17 A- 17 B .
  • a population of cells were generated that were enriched for CEA and/or FOLR expression.
  • the fraction of anti-CEA, anti-FOLR, and universal CoStAR cells with positive expression are as shown in FIGS. 23 - 27 .
  • the CD4/8 ratio was as shown in FIGS. 28 A- 28 D .
  • the methodology for the assay was as follows:
  • TIL cells were thawed.
  • CoStAR-modified TILS were sorted, then ran on fortessa.
  • the media change was completed, and the cells were stained and ran on fortessa.
  • the TCM media was changed to exclude IL2.
  • the co-culture and serial stimulation assay was set up. As can be seen in FIGS. 30 A- 30 H, 31 A- 31 H, and 32 A- 32 H , the co-culture with autologous digest is inconclusive probably due to variability introduced in tumor reactivity of the TILs between different conditions as a result of sorting for CoStAR.
  • TIL cells expressing CoStAR constructs then underwent a serial stimulation assay.
  • the results of the stimulation assay are as shown in FIGS. 15 A- 15 E and 16 A- 16 E .
  • the methodology for the stimulation assay was as follows:

Abstract

The present invention relates to a chimeric molecule useful in adoptive cell therapy (ACT), and cells comprising the same. The chimeric molecule can act as a modulator of cellular activity enhancing responses when an endogenous T-cell receptor (TCR) is engaged with its cognate antigen. The present invention also provides proteins, nucleic acids encoding the chimeric molecule and therapeutic uses thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS FOR PRIORITY
  • This application is a continuation of U.S. patent application Ser. No. 17/822,251, filed Aug. 25, 2022, which is a continuation in part of PCT Patent Application Serial No. PCT/US2022/073741, filed Jul. 14, 2022, which claims priority to U.S. Provisional Application Ser. No. 63/222,916, filed Jul. 16, 2021, both of which are hereby expressly incorporated by reference in its entirety.
  • RELATED APPLICATIONS AND INCORPORATION BY REFERENCE
  • Reference is made to U.S. Provisional Patent Application No. 63/222,916, filed Jul. 16, 2021, U.S. Provisional Patent Application Ser. No. 63/053,498 filed Jul. 17, 2020, U.S. Provisional Patent Application Ser. No. 63/222,916, filed Jul. 16, 2021, PCT Patent Application Serial No. PCT/US2021/042079 filed Jul. 16, 2021, U.S. Provisional Patent Application Ser. No. 63/345,821, filed May 25, 2022, the contents of which are incorporated herein by reference in their entireties.
  • Reference is made to GB patent application Serial No. 1900858.0, filed 22 Jan. 2019, U.S. patent application Ser. No. 62/951,770, filed 20 Dec. 2019, International application PCT/GB2020/050120, filed 20 Jan. 2020, and U.S. provisional patent application 63/053,494, filed Jul. 17, 2020.
  • The foregoing applications, and all documents cited therein or during their prosecution (“appln cited documents”) and all documents cited or referenced in the appln cited documents, and all documents cited or referenced herein (“herein cited documents”), and all documents cited or referenced in herein cited documents, together with any manufacturer's instructions, descriptions, product specifications, and product sheets for any products mentioned herein or in any document incorporated by reference herein, are hereby incorporated herein by reference, and may be employed in the practice of the invention. More specifically, all referenced documents are incorporated by reference to the same extent as if each individual document was specifically and individually indicated to be incorporated by reference.
  • REFERENCE TO 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 INSTB.006C3.xml, created on May 3, 2023, which is 266,974 bytes in size. The information in the electronic format of the Sequence Listing is incorporated herein by reference in its entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to a chimeric molecule useful in adoptive cell therapy (ACT), and cells comprising the same. The chimeric molecule can act as a modulator of cellular activity enhancing responses when an endogenous T-cell receptor (TCR) is engaged with its cognate antigen. The present invention also provides proteins, nucleic acids encoding the chimeric molecule and therapeutic uses thereof.
  • BACKGROUND OF THE INVENTION
  • Adoptive cell therapy (ACT) using autologous T-cells to mediate cancer regression has shown much promise in early clinical trials. Several general approaches have been taken such as the use of naturally occurring tumor reactive or tumor infiltrating lymphocytes (TILs) expanded ex vivo. Additionally, T-cells may be genetically modified to retarget them towards defined tumor antigens. This can be done via the gene transfer of peptide (p)-major histocompatibility complex (MHC) specific T-cell Receptors (TCRs) or synthetic fusions between tumor specific single chain antibody fragment (scFv) and T-cell signaling domains (e.g. CD3), the latter being termed chimeric antigen receptors (CARs).
  • TIL and TCR transfer has proven particularly good when targeting melanoma (Rosenberg et al. 2011; Morgan 2006), whereas CAR therapy has shown much promise in the treatment of certain B-cell malignancies (Grupp et al. 2013).
  • Costimulatory signals are useful to achieve robust CAR T cell expansion, function, persistence and antitumor activity. The success of CAR therapy in leukemia has been partly attributed to the incorporation of costimulatory domains (e.g. CD28 or CD137) into the CAR construct, signals from which synergize with the signal provided by CD3ζ to enhance anti-tumor activity. The basis of this observation relates to the classical signal 1/signal 2 paradigm of T-cell activation. Here signal 1, provided by the TCR complex, synergizes with signal 2 provided by costimulatory receptors such as CD28, CD137 or CD134 to permit the cells to undergo clonal expansion, IL2 production and long term survival without the activation induced cell death (AICD) associated with signal 1 alone. Furthermore the involvement of signal 2 enhances the signal generated through signal 1 allowing the cells to respond better to low avidity interactions such as those encountered during anti-tumor responses.
  • Targeted costimulation will have beneficial effects for non-CAR-based T-cell therapies. For example, incorporating costimulatory domains into a chimeric TCR has been shown to enhance responses of T-cells towards pMHC (Govers 2014). While tumor infiltrating lymphocytes (TILs) utilize their endogenous TCRs to mediate tumor recognition, it has not been possible to engineer the endogenous TCR. Thus TIL are subject to substantial limitations as tumor cells express very few costimulatory ligands. The ability to induce targeted costimulation of TIL, or indeed any other adoptive T-cell therapy product, would be beneficial.
  • Citation or identification of any document in this application is not an admission that such document is available as prior art to the present invention.
  • SUMMARY OF THE INVENTION
  • Provided herein are chimeric molecules, in particular chimeric proteins, designed to provide costimulation when the endogenous TCR is engaged with its cognate antigen. Mechanistically, the proposed constructs may be incorporated in the endogenous TCR complex. When the endogenous TCR complex machinery is engaged with their cognate antigen, the TCR receptor complex aggregates, forcing the clustering of these chimeric constructs. This clustering results in the activation of their signaling domains, causing an increase in costimulation. This costimulation manifests itself in a measurable improvement in the effector function of the recipient T cell: increased in activation markers, increase cytokine secretion (IL-2 in particular) and increased proliferation.
  • Some embodiments herein relate to a chimeric molecule, advantageously a chimeric protein, that provides costimulation to the T cell when the endogenous T cell receptor is engaged. This molecule may comprise a TCR clustering domain and a signaling domain that may contain a CD40 intracellular domain or signaling fragment thereof.
  • The TCR clustering domain may be one or more of the proteins typically found in the TCR complex, such as but not limited to, CD3D, CD3E, CD3G, CD3Z, CD3-eta and the constant chains of pre-TCR alpha (PTCRA) TCR alpha, TCR beta, TCR gamma or TCR delta.
  • The signaling domain may also comprise, an additional full length costimulatory domain, including but not limited to CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6.
  • While CD3D, CD3E, CD3G, CD3Z work alone; the constructs containing TCR constant chains (either alpha/beta or gamma/delta) are preferably co-expressed with their respective partner in bicistronic configuration: TCR alpha with TCR beta and TCR gamma with TCR delta. Therefore, TCR alpha containing constructs are advantageously co-expressed with TCR beta and vice versa; and TCR gamma containing constructs should be co-expressed with TCR delta and vice versa. In the context of TILs and any other alpha-beta T cells; the preferred configuration includes TCR gamma-delta; and in gamma-delta T cells, the preferred configuration includes TCR alpha-beta to minimize interference/disruption with the endogenous TCR machinery and the TCR pairing.
  • For CD3D, CD3E, CD3G and CD3Z, the transmembrane and extracellular portions are advantageously utilized. However, the present invention also contemplates portions or the totality of their intracellular components, which could potentially minimize the disruption of the endogenous TCR complex signaling or help to further amplify the endogenous TCR signaling.
  • In another aspect, the invention provides a chimeric protein comprising a clustering domain and a signaling domain that may contain a CD40 intracellular domain or signaling fragment thereof. In some embodiments, the clustering domain is capable of oligomerization and/or self assembly. In some embodiments, clustering comprises formation of a homodimer or homotrimer. In some embodiments, clustering comprises oligomerization with a different protein to form a heterodimer or heterotrimer. In some embodiments, the chimeric protein is constitutive as signaling, for example independent of receptor engagement by an extracellular ligand or independent of receptor engagement by an extracellular ligand attached to a different cell. In some embodiments, the clustering domain comprises a transmembrane domain. In some embodiments, the clustering domain comprises a transmembrane domain and further comprises activating mutations that promote dimerization or oligomerization. In some embodiments, the clustering domain comprises an extracellular domain, such as but not limited to an extracellular domain of a receptor. In some embodiments, the clustering domain comprises an extracellular domain of a receptor and further comprises activating mutations in the extracellular domain that promote dimerization or oligomerization. In some embodiments, the clustering domain comprises a leucine zipper. In some embodiments, the leucine zipper comprises or constitutes a transmembrane domain. In some embodiments, the leucine zipper comprises or constitutes a soluble domain. Non-limiting examples of clustering domains include clustering domains of the thrombopoietin receptor (TpoR), erythropoietin receptor (EpoR), growth hormone receptor (GHR), glycophorin A (GPA) transmembrane domain, and activating mutants thereof. In some embodiments, clustering may be modulated by a small molecule. In some embodiments, clustering may be modulated by post-translational modifications.
  • In another aspect, the invention provides a chimeric protein which comprises an extracellular ligand binding domain linked to an intracellular signaling domain by a transmembrane domain. In some embodiments, the extracellular ligand binding domain is selected or engineered to bind to an extracellular ligand that maintains two or more copies of the chimeric protein in proximity to one another such that the signaling domain is activated. The extracellular ligand binding domain is considered one part of a specific binding pair (sbp) and the extracellular ligand is the second part of the specific binding pair. In some embodiments, one member of the sbp comprises a protein or receptor or extracellular portion thereof and the second sbp comprises a binding protein specific for the first member of the sbp. In some embodiments, the extracellular sbp is bivalent. In some embodiments, the extracellular sbp is trivalent. Nonlimiting examples of extracellular ligands include antibodies and bivalent antigen binding fragments thereof. Non-limiting examples of extracellular ligand binding domains of chimeric proteins of the invention (i.e., sbp members) include, without limitation, NKG2A, CD27, CD137, GITR, PD-1, PD-L1, FasL, OX40, CTLA4, ICOS, CD40, EGFR, HER2 and extracellular portions thereof. Complementary sbp members include, without limitation, pembrolizumab for PD1, trastuzumab for HER2, cetuximab for EGFR, tremelimumab for CTLA4, varlilumab for CD27, and urelumab for CD137. In some embodiments, the intracellular signaling domain comprises a CD40 intracellular domain or signaling fragment thereof.
  • In some embodiments, the CD40 signaling domain comprises SEQ ID NO:154, SEQ ID NO:155, or SEQ ID NO:156. In some embodiments, the CD40 signaling fragment comprises, consists, or consists essentially of an SH3 motif (KPTNKAPH, PTNKAPHP or PTNKAPH), TRAF2 motif (PKQE, PKQET, PVQE, PVQET, SVQE, SVQET), TRAF6 motif (QEPQEINFP or QEPQEINFP), PKA motif (KKPTNKA, SRISVQE, or a combination thereof, or is a full length CD40 intracellular domain. In some embodiments, one or more of the SH3, TRAF2, TRAF6, or PKA motifs of the CD40 signaling domain is mutated. In some embodiments, one or more of the SH3, TRAF2, TRAF6, or PKA motifs of the CD40 signaling domain is present in multiple copies.
  • Disclosed in this application is an engineered protein. In some embodiments, the engineered protein has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 166, wherein the sequence is not SEQ ID NO: 123.
  • In some embodiments, the engineered protein has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 167, wherein the sequence is not SEQ ID NO: 123.
  • In some embodiments, the engineered protein further comprises a binding domain, CD28 domain, and CD40 domain. In some embodiments, the engineered protein further comprises a signal peptide sequence. In some embodiments, the signal peptide sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 157. In some embodiments, the binding domain comprises a VL sequence, a VH sequence, and an at least one linker. In some embodiments, the at least one linker has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 159 or 161. In some embodiments, the binding domain comprises two linker sequences. In some embodiments, the two linker sequences have at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to amino acid sequences SEQ ID NO: 159 and SEQ ID NO: 161, respectively. In some embodiments, the VL sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 158. In some embodiments, the VH sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 160. In some embodiments, the CD40 domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 165. In some embodiments, the CD28 domain comprises a CD28 transmembrane domain. In some embodiments, the CD28 transmembrane domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 163. In some embodiments, the CD28 domain comprises a CD28 extracellular domain. In some embodiments, the CD28 extracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 162. In some embodiments, the CD28 domain comprises a CD28 intracellular domain. In some embodiments, the CD28 intracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 164. In some embodiments, the protein further comprises 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDSAVYAYALNI (SEQ ID NO: 173). In some embodiments, the binding domain and CD28 domain are connected by an at least one linker.
  • In some embodiments, an engineered protein is provided. It can comprise an amino acid sequence that is at least 80% identical to the amino acid sequence of SEQ ID NO: 166 or 167, wherein the amino acid sequence does not include at least one of:
  • (SEQ ID NO: 174)
    QKLISEEDLE
    or
    (SEQ ID NO: 175)
    LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGN
    YSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI.
  • In some embodiments, a CoStAR is provided. It can comprise: a) an optional signal peptide; b) a binding domain, wherein the binding domain binds to an anti-pembrolizumab antibody or binding fragment thereof; c) a CD28 domain; and d) a CD40 domain. Wherein a) is optionally linked to b), wherein b) is linked to c), wherein c) is linked to d), and wherein the CoStAR comprises an amino acid sequence that: i) lacks at least one of:
  • (SEQ ID NO: 174)
    QKLISEEDLE
    or
    (SEQ ID NO: 175)
    LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGN
    YSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI;

    ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167; iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is not SEQ ID NO: 123; or iv) any combination of i-iv.
  • In some embodiments, a fusion protein is provided. The fusion protein comprises a) a means for binding to an antibody that binds to pembrolizumab; b) a CD28 domain; and c) CD40 domain. Wherein a) is linked to b), wherein b) is linked to c), and wherein the fusion protein comprises an amino acid sequence that: i) lacks at least one of:
  • (SEQ ID NO: 174)
    QKLISEEDLE
    or
    (SEQ ID NO: 175)
    LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGN
    YSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI;

    ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167; iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is not SEQ ID NO: 123; or iv) any combination of i-iv.
  • In some embodiments, a fusion protein is provided that comprises the amino acid sequence of SEQ ID NO: 166.
  • In some embodiments, a fusion protein is provided that comprises the amino acid sequence of SEQ ID NO: 167.
  • In some embodiments, a nucleic acid which encodes the protein of any one of the preceding claims.
  • Also disclosed herein is a nucleic acid which encodes a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a vector which comprises a nucleic acid of any one of the embodiments of the present application.
  • Also disclosed herein is a cell which expresses a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a cell which expresses at least two proteins of any one of the embodiments of the present application.
  • Also disclosed herein is a method of making the cell of any one of the embodiments of the present application which comprises the step of transducing or transfecting a cell with a vector of any one of the embodiments of the present application.
  • Also disclosed herein is a method for preparing a population of cells that express a protein of any one of the embodiments of the present application, comprising detecting expression of the protein on the surface of cells transfected or transduced with a vector according to any one of the embodiments of the present application and selecting cells which are identified as expressing the protein.
  • Also disclosed herein is a cell population produced by the method of any one of the methods disclosed in the present application.
  • Also disclosed herein is a cell population which is enriched for cell expression a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a method for treating a disease in a subject in need thereof, which comprises the step of administering the cell of any one of the embodiments of the present application, or the cell population of any one of the embodiments of the present application, to the subject.
  • Accordingly, it is an object of the invention not to encompass within the invention any previously known product, process of making the product, or method of using the product such that Applicants reserve the right and hereby disclose a disclaimer of any previously known product, process, or method. It is further noted that the invention does not intend to encompass within the scope of the invention any product, process, or making of the product or method of using the product, which does not meet the written description and enablement requirements of the USPTO (35 U.S.C. § 112, first paragraph) or the EPO (Article 83 of the EPC), such that Applicants reserve the right and hereby disclose a disclaimer of any previously described product, process of making the product, or method of using the product. It may be advantageous in the practice of the invention to be in compliance with Art. 53(c) EPC and Rule 28(b) and (c) EPC. All rights to explicitly disclaim any embodiments that are the subject of any granted patent(s) of applicant in the lineage of this application or in any other lineage or in any prior filed application of any third party is explicitly reserved. Nothing herein is to be construed as a promise.
  • It is noted that in this disclosure and particularly in the claims and/or paragraphs, terms such as “comprises”, “comprised”, “comprising” and the like can have the meaning attributed to it in U.S. patent law; e.g., they can mean “includes”, “included”, “including”, and the like; and that terms such as “consisting essentially of” and “consists essentially of” have the meaning ascribed to them in U.S. patent law, e.g., they allow for elements not explicitly recited, but exclude elements that are found in the prior art or that affect a basic or novel characteristic of the invention.
  • These and other embodiments are disclosed or are obvious from and encompassed by, the following Detailed Description.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The following detailed description, given by way of example, but not intended to limit the invention solely to the specific embodiments described, may best be understood in conjunction with the accompanying drawings.
  • FIG. 1A-1C—Schematic models for universal costimulatory proteins. (FIG. 1A) TCR incorporated antigen agnostic receptor (TIAAR) comprises modifying components of the TCR complex and associated signaling adaptors. (FIG. 1B) A constitutive costimulatory receptor comprising transmembrane domains (TMDs) and features that enable inducible or constitutive activation. (FIG. 1C) An inducible costimulatory receptor capable of induction and activation by extracellular ligand binding.
  • FIG. 2 —Cytokine production by TCR incorporated antigen agnostic receptor (TIAAR) transduced cells. Cytokine production (Bcl-xL, IL2, IFNg and TNFa) from genetically modified and non-transduced T cells (NTD) of two donors was determined after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only).
  • FIGS. 3A-3B—Proliferation and activation marker expression by TIAAR transduced cells. Proliferation (T cell counts) and activation marker expression (41BB and CD69) was determined for genetically modified and non-transduced T cells (NTD) from donor 1 (FIG. 3A) and donor 2 (FIG. 3B) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only).
  • FIG. 4 —Cytokine production in leucine zipper based universal CoStAR (LZ) transduced cells. Cytokine production (Bcl-xL, IL2, IFNg and TNFa) from genetically modified and non-transduced T cells (NTD) of two donors was determined after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only).
  • FIGS. 5A-5B—Proliferation and activation marker expression by LZ-CoStAR transduced cells. Proliferation (T cell counts) and activation marker expression (41BB and CD69) was determined for genetically modified and non-transduced T cells (NTD) from donor 1 (FIG. 5A) and donor 2 (FIG. 5B) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only).
  • FIG. 6 —Cytokine production in inducible universal CoStAR transduced cells. Cytokine production (Bcl-xL, IL2, IFNg and TNFa) from genetically modified and non-transduced T cells (NTD) of two donors was determined after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only). The universal CoStAR is inducible by pembrolizumab.
  • FIGS. 7A-7B—Proliferation and activation marker expression by inducible universal CoStAR transduced cells. Proliferation (T cell counts) and activation marker expression (41BB and CD69) was determined for genetically modified and non-transduced T cells (NTD) from donor 1 (FIG. 7A) and donor 2 (FIG. 7B) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only). The universal CoStAR is inducible by pembrolizumab.
  • FIG. 8A—is a schematic of a protein of some embodiments provided herein. It is a protein comprising, Universal CoStAR sequence, comprising an optional section, a binding domain, a CD28 domain, and a CD40 domain.
  • FIG. 8B—is a schematic of some embodiments provided herein.
  • FIG. 8C—outlines a set of sequences of some embodiments provided herein.
  • FIG. 9 —depicts a sequence of an anti-pembrolizumab CoStAR Sequence (“Universal CoStAR”) (SEQ ID NO: 166), containing an optional signal domain.
  • FIG. 10 —depicts a sequence of an anti-pembrolizumab CoStAR Sequence (“Universal CoStAR”) (SEQ ID NO: 166), without the optional signal domain.
  • FIG. 11 —depicts a sequence alignment between the anti-pembrolizumab CoStAR Sequence (“Universal CoStAR”) (SEQ ID NO: 166) and the vector clone pIB1102 (SEQ ID NO: 123).
  • FIG. 12 —depicts the transduction efficiency of constructs into TILs after 21 days. Round 1 (left panel) and Round 2 (right panel) of transduction was performed with constructs CTP386.1 and CTP387.1 across a variety of TIL organ types (x-axis).
  • FIG. 13 —depicts the transduction efficiency of constructs into TILs after 21 days. Round 1 (left panel) and Round 2 (right panel) of transduction was performed with constructs 322 and 1324 across a variety of TIL organ types (x-axis).
  • FIG. 14 —depicts the transduction efficiency of constructs into TILs after 21 days. Round 1 (left panel) and Round 2 (right panel) of transduction was performed with construct CTP205 across a variety of TIL organ types (x-axis).
  • FIGS. 15A-15E—depict the fold-expansion of TILs following a serial stimulation assay. Anti-CEA or anti-FOLR modified TILs from CRC 9823 were treated every 7 days with the target K562 OKT3 CEACAM5 or OKT3 FOLR, respectively. The readout was measured using the cell count over time. Shown in panels are the fold-expansion for anti-CEA TILs with K562 OKT3 CEACAM5 exposure (FIG. 15A), anti-FOLR TILs with OKT3 FOLR exposure (FIG. 15B), a universal CoStAR with K562 OKT3 CEACAM5 exposure (FIG. 15C), a universal CoStAR with K562 OKT3 CEACAM5 exposure and 5 ug/mL pembro (FIG. 15D), and a universal CoStAR with K562 OKT3 CEACAM5 exposure and 250 ug/mL pembdro (FIG. 15E).
  • FIGS. 16A-16E—depict the fold-expansion of TILs following a serial stimulation assay. Anti-CEA or anti-FOLR modified TILs from Mel 11909 were treated every 7 days with the target K562 OKT3 CEACAM5 or OKT3 FOLR, respectively. The readout was measured using the cell count over time. Shown in panels are the fold-expansion for anti-CEA TILs with K562 OKT3 CEACAM5 exposure (FIG. 16A), anti-FOLR TILs with OKT3 FOLR exposure (FIG. 16B), a universal CoStAR with K562 OKT3 CEACAM5 exposure (FIG. 16C), a universal CoStAR with K562 OKT3 CEACAM5 exposure and 5 ug/mL pembro (FIG. 16D), and a universal CoStAR with K562 OKT3 CEACAM5 exposure and 250 ug/mL pembdro (FIG. 16E).
  • FIGS. 17A-17B—depict the increase in IL2 production (pg/mL) in anti-CEA, anti-FOLR, and Universal CoStAR modified TLS from CRC983 (FIG. 17A) or Mel 11909 (FIG. 17B).
  • FIG. 18 —depicts a pie chart of the tumor types (n=15) used as digests to generate TIL cells in Example 3.
  • FIG. 19 —depicts an example timeline for transducing CoStAR constructs into cells using the protocol outlined in Example 3.
  • FIG. 20 —depicts the transduction efficiency of constructs into TILs after 1 day. Round 1 (left panel) and Round 2 (right panel) of transduction was performed with across a variety of TIL organ types (x-axis). Plotted on the y-axis, is the percent of cells with positive expression of FOLR1 following transduction.
  • FIG. 21 —depicts the transduction efficiency of constructs into TILs after 1 day. Round 1 (left panel) and Round 2 (right panel) of transduction was performed across a variety of TIL organ types (x-axis). Plotted on the y-axis, is the percent of cells with positive expression of CEA following transduction.
  • FIG. 22 —depicts the transduction efficiency of constructs into TILs after 1 day. Round 1 (left panel) and Round 2 (right panel) of transduction was performed across a variety of TIL organ types (x-axis). Plotted on the y-axis, is the percent of cells with positive expression of both FOLR1 and CEA following transduction.
  • FIG. 23 —depicts the round 1 (left panel) and round 2 (right panel) of Anti-CEA (386.1) cells that were percent positive for CEA expression after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIG. 24 —depicts the round 1 (left panel) and round 2 (right panel) of Anti-CEA (387.1) cells that were percent positive for CEA expression after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIG. 25 —depicts the round 1 (left panel) and round 2 (right panel) of Anti-FOLR cells that were percent positive for FOLR expression after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIG. 26 —depicts the round 1 (left panel) and round 2 (right panel) of Universal CoStAR (pIB1322) cells that were percent positive for both FOLR and CEA after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIG. 27 —depicts the round 1 (left panel) and round 2 (right panel) of Universal CoStAR (pIB1324) cells that were percent positive for both FOLR and CEA after pre-sort, post-sort, day 2, and day 4 as outlined in Example 4.
  • FIGS. 28A-28D—depict the ratio of CD4 to CD8 positive TIL cells of those enriched for FOLR and/or CEA expression after 21 days. FIG. 28A depicts the ratio in CRC cells, FIG. 28B depicts the ratio in NSCLC cells, FIG. 28C depicts the ratio in ovarian cells, and FIG. 28D depicts the ratio in melanoma cells.
  • FIG. 29 —depicts an example timeline for sorting TIL cells and testing for function, using the protocol outlined in Example 4.
  • FIG. 30A-30H—depicts the increase in IFNg production (pg/mL) in TIL and K562 cells lines following co-culturing, across the cell types CRC-11974 (FIG. 30A), CRC-11959 (FIG. 30B), NSCLC-9332 (FIG. 30C), NSCLC-9596 (FIG. 30D), Ovarian cells (FIG. 30E), Melanoma-CC60 (FIG. 30F), Melanoma-11909 (FIG. 30G), and Melanoma-17614 (FIG. 30H).
  • FIG. 31A-31H—depicts the increase in IL2 production (pg/mL) in TIL and K562 cells lines following co-culturing, across the cell types CRC-11974 (FIG. 31A), CRC-11959 (FIG. 31B), NSCLC-9332 (FIG. 31C), NSCLC-9596 (FIG. 31D), Ovarian cells (FIG. 31E), Melanoma-CC60 (FIG. 31F), Melanoma-11909 (FIG. 31G), and Melanoma-17614 (FIG. 31H).
  • FIG. 32A-32H—depicts the increase in TNFa production (pg/mL) in TIL and K562 cells lines following co-culturing, across the cell types CRC-11974 (FIG. 32A), CRC-11959 (FIG. 32B), NSCLC-9332 (FIG. 32C), NSCLC-9596 (FIG. 32D), Ovarian cells (FIG. 32E), Melanoma-CC60 (FIG. 32F), Melanoma-11909 (FIG. 32G), and Melanoma-17614 (FIG. 32H).
  • DETAILED DESCRIPTION OF THE INVENTION
  • Disclosed herein are a variety of engineered proteins. In some embodiments, the engineered protein that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 166, and wherein the sequence is not SEQ ID NO: 123. In some embodiments, the engineered protein has at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 70 and 100%, identity to SEQ ID NO: 166. In some embodiments, the engineered protein has an at least 80% identity to SEQ ID NO:166. In some embodiments, the engineered protein has an at least 90% identity to SEQ ID NO:166. In some embodiments, the engineered protein is SEQ ID NO:166 (FIG. 9 ). In some embodiments, the sequence is at least 80% identical and is not the sequence of SEQ ID NO: 123.
  • In some embodiments, the engineered protein that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 167, and wherein the sequence is not SEQ ID NO: 123. In some embodiments, the engineered protein that has at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 70 and 100%, identity to SEQ ID NO: 167. In some embodiments, the engineered protein has an at least 80% identity to SEQ ID NO:167. In some embodiments, the engineered protein has an at least 90% identity to SEQ ID NO:167. In some embodiments, the engineered protein is SEQ ID NO:167 (FIG. 10 ). In some embodiments, the sequence is at least 80% identical, and is not the sequence of SEQ ID NO: 123.
  • In some embodiments, an engineered protein is provided that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 166, and wherein the sequence is not SEQ ID NO: 123.
  • In some embodiments, an engineered protein is provided that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 167, and wherein the sequence is not SEQ ID NO: 123.
  • In some embodiments, the engineered protein, CoStAR or fusion protein has a general structure as depicted in FIG. 8A.
  • In some embodiments, the engineered protein CoStAR or fusion protein has a general structure as depicted in FIG. 8B.
  • In some embodiments, the engineered protein CoStAR or fusion protein comprises at least one sequence depicted in FIG. 8C.
  • In some embodiments, the arrangement in FIG. 8A is an embodiment separate from the embodiments in FIGS. 8B and/or 8C. In some embodiments, the arrangement in FIG. 8B is an embodiment separate from the embodiments in FIGS. 8C and/or 8A. In some embodiments, the arrangement in FIG. 8C is an embodiment separate from the embodiments in FIGS. 8A and/or 8B (thus, the sequence itself is envisioned, in some embodiments, as the entirety of the engineered protein). In some embodiments, FIG. 8B depicts some embodiments that are a subset of FIG. 8C. In some embodiments, FIG. 8C depicts some embodiments that are a subset of FIG. 8A and FIG. 8B. Exemplary CDRs are underlined in FIG. 8C (SEQ ID NO: 158 and 160).
  • In some embodiments, the engineered protein, CoStAR or fusion protein is as depicted in FIG. 9 or 10 . Exemplary CDRs are underlined in FIG. 9 and FIG. 10 . In some embodiments, the engineered protein, CoStAR or fusion protein is different from other fusion proteins, as shown in FIG. 11 . In some embodiments, the engineered protein, CoStAR or fusion protein can lack a tag component and/or a section of CD28.
  • In some embodiments, the engineered protein comprises a binding domain. In some embodiments, the engineered protein comprises a CD28 domain. In some embodiments, the engineered protein comprises a CD40 domain. In some embodiments, the engineered protein comprises 1, 2, or all 3 of a binding domain, a CD28 domain, and/or a CD40 domain.
  • In some embodiments, the engineered protein comprises a signal peptide sequence. The term “signal peptide” is given its usual scientific meaning, and thus refers a short peptide that functions in translocating the rest of the attached protein. “Signal peptide” thus may also be called a signal sequence, targeting signal, localization signal, localization sequence, transit peptide, leader sequence or leader peptide. It will be understood that the signal peptide may be any peptide with the function of signaling for the attached peptide to be translocated to the plasma membrane of a cell. In some embodiments, the signal peptide sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 157.
  • In some embodiments, the binding domain comprises 1, 2, or all 3 of a VL sequence, a VH sequence, and/or an at least one linker. In some embodiments, the at least one linker has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 159 or 161. In some embodiments, the binding domain comprises two linker sequences. In some embodiments, the two linker sequences have at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to amino acid sequences SEQ ID NO: 159 and SEQ ID NO: 161, respectively.
  • In some embodiments, the VL sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 158. In some embodiments, the VH sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 160. In some embodiments, all 3 of the heavy chain CDRs and/or all three of the light chain CDRs within the VH and VL are identical to the heavy and/or light chain CDRs contained within SEQ ID NOs: 158 and 160. In some embodiments, 1, 2, 3, 4, 5 or 6 of the CDRs have 1, 2, 3, 4 or more point mutations. In some embodiments, 1, 2, 3, 4, 5, or 6 CDRs are 70, 80, 85, 90, 95, 96, 97, 98, 99 or 100% identical to the corresponding CDRs within SEQ ID NOs: 158 and/or 160.
  • In some embodiments, the protein comprises 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDSAVYAYALNI (SEQ ID NO: 173). In some embodiments, the binding domain and CD28 domain are connected by an at least one linker. In some embodiments, 1, 2, 3, 4, 5 or 6 of the CDRs have 1, 2, 3, 4, or more point mutations. In some embodiments, 1, 2, 3, 4, 5, or 6 CDRs are 70, 80, 85, 90, 95, 96, 97, 98, 99 or 100% identical to the CDRs.
  • Also disclosed herein is an engineered protein comprising an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to the amino acid sequence of SEQ ID NO: 166 or 167.
  • In some embodiments, an engineered protein is provided that comprises an amino acid sequence that is at least 80% identical to the amino acid sequence of SEQ ID NO: 166 or 167, wherein the amino acid sequence does not include at least one of:
  • (SEQ ID NO: 174)
    QKLISEEDLE
    or
    (SEQ ID NO: 175)
    LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGN
    YSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI.
  • In some embodiments, the CD40 domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 165. In some embodiments, the CD28 domain comprises a CD28 transmembrane domain. In some embodiments, the CD28 transmembrane domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 163. In some embodiments, the CD28 domain comprises a CD28 extracellular domain. In some embodiments, the CD28 extracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 162. In some embodiments, the CD28 domain comprises a CD28 intracellular domain. In some embodiments, the CD28 intracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 164.
  • In some embodiments, the amino acid sequence and/or fusion protein and/or engineered protein does not include at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQQLQV YSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175). In some embodiments, the engineered protein lacks both of QKLISEEDLE (SEQ ID NO: 174) and
  • (SEQ ID NO: 175)
    LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGN
    YSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 157.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 158.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 159.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 160.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 161.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 162.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 163.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 164.
  • In some embodiments, the engineered protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 165.
  • In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) that have at least 80% identity to SEQ ID NOS: 157-165, respectively. In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) that have at least 90% identity to SEQ ID NOS: 157-165, respectively. In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) that have at least 95% identity to SEQ ID NOS: 157-165, respectively. In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) that have at least 98% identity to SEQ ID NOS: 157-165, respectively. In some embodiments, the engineered protein comprises 1, 2, 3, 4, 5, 6, 7, 8, and/or 9 sequence(s) selected from the group consisting of: SEQ ID NOS: 157-165.
  • Also disclosed herein is a CoStAR. In some embodiments, the CoStAR comprises an optional signal peptide, a binding domain, wherein the binding domain binds to an anti-pembrolizumab antibody or binding fragment thereof, a CD28 domain, and a CD40 domain, wherein the signal peptide is optionally linked to the binding domain, wherein the binding domain is linked to the CD28 domain, wherein the CD28 domain is linked to the CD40 domain, and wherein the CoStAR comprises an amino acid sequence that: i) lacks at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQ QLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175); ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167; iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is not SEQ ID NO: 123; or iv) any combination of i-iv.
  • Also disclosed herein is a fusion protein. In some embodiments, the fusion protein comprises a means for binding to an antibody that binds to pembrolizumab, a CD28 domain, and a CD40 domain, wherein the means for binding to an antibody is linked to a CD28 domain, wherein the CD28 domain is linked to the CD40 domain, and wherein the fusion protein comprises an amino acid sequence that: i) lacks at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQQLQV YSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175); ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167; iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is not SEQ ID NO: 123; or iv) any combination of i-iv. In some embodiments, the means for binding to pembrolizumab is an anti-pembrolizumab antibody. In some embodiments, the anti-pembrolizumab antibody is
  • In some embodiments, the binding domain or the means for binding to an antibody that binds to pembrolizumab comprises: 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDSAVYAYALNI (SEQ ID NO: 173). In some embodiments, 1, 2, 3, 4, 5 or 6 of the CDRs have 1, 2, 3, 4, or more point mutations. In some embodiments, 1, 2, 3, 4, 5, or 6 CDRs are 70, 80, 85, 90, 95, 96, 97, 98, 99 or 100% identical to the CDRs.
  • In some embodiments, the CD28 domain comprises: SEQ ID Nos: 162, 163, and 164, or a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical thereto. In some embodiments, the CD40 domain comprises: SEQ ID No: 165, or a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical thereto.
  • Also disclosed herein is a fusion protein comprising the amino acid sequence of SEQ ID NO: 166. In some embodiments, the fusion protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 166. Also disclosed herein is a fusion protein comprising the amino acid sequence of SEQ ID NO: 167. In some embodiments, the fusion protein comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identical to SEQ ID NO: 167.
  • Also disclosed herein is a nucleic acid which encodes a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a vector which comprises a nucleic acid of any one of the embodiments of the present application.
  • Also disclosed herein is a cell which expresses a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a cell which expresses at least two proteins of any one of the embodiments of the present application.
  • Also disclosed herein is a method of making the cell of any one of the embodiments of the present application which comprises the step of transducing or transfecting a cell with a vector of any one of the embodiments of the present application.
  • Also disclosed herein is a method for preparing a population of cells that express a protein of any one of the embodiments of the present application, comprising detecting expression of the protein on the surface of cells transfected or transduced with a vector according to any one of the embodiments of the present application and selecting cells which are identified as expressing the protein.
  • Also disclosed herein is a cell population produced by the method of any one of the methods disclosed in the present application.
  • Also disclosed herein is a cell population which is enriched for cell expression a protein of any one of the embodiments of the present application.
  • Also disclosed herein is a method for treating a disease in a subject in need thereof, which comprises the step of administering the cell of any one of the embodiments of the present application, or the cell population of any one of the embodiments of the present application, to the subject.
  • As used herein, “full length protein” or “full length receptor” refers to a receptor protein, such as, for example, a CD40 receptor protein. The term “full length” encompasses receptor proteins lacking up to about 5 or up to 10 amino acids, for example 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids, at the N-terminal of the mature receptor protein once its signal peptide has been cleaved. For instance, while a specific cleavage site of a receptors N-terminal signal peptide may be defined, variability in exact point of cleavage has been observed. The term “full length” does not imply presence or absence of amino acids of the receptors N-terminal signal peptide. In one embodiment, the term “full length” (e.g. a full length CD28 or a full length CD40 intracellular domain, according to certain aspects of the invention) encompasses mature receptor proteins (e.g. CD28 according to certain aspects of the invention) lacking the N terminal signal peptide lacking up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein once its signal peptide has been cleaved. As mentioned above, a “full length” CD28 receptor or other receptor or TCR clustering domain according to the various aspects of the invention does not include the signal peptide and may lack up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein (e.g. N terminal residues N, K, I, L and/or V). This is shown in the exemplary fusions, e.g. SEQ ID Nos. 4-12 (note that these may lack up to about 5, for example 1, 2, 3, 4, 5, or up to 10 amino acids at the N-terminal of the mature receptor protein as shown in the boxed region).
  • The chimeric protein of the present invention may comprise a TCR clustering domain as well as a signaling domain that advantageously may comprise a CD40 intracellular domain.
  • The term “T cell receptor,” or “TCR,” refers to a heterodimeric receptor composed of αβ or γδ chains that pair on the surface of a T cell. Each α, β, γ, and δ chain is composed of two Ig-like domains: a variable domain (V) that confers antigen recognition through the complementarity determining regions (CDR), followed by a constant domain I that is anchored to cell membrane by a connecting peptide and a Transmembral TM) region. The TM region associates with the invariant subunits of the CD3 signaling apparatus. Each of the V domains has three CDRs. These CDRs interact with a complex between an antigenic peptide bound to a protein encoded by the major histocompatibility complex (pMHC) (Davis and Bjorkman (1988) Nature, 334, 395-402; Davis et al. (1998) Annu Rev Immunol, 16, 523-544; Murphy (2012), xix, 868 p.).
  • Costimulatory receptor proteins useful in the chimeric proteins of the invention include, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6, which in their natural form comprise extracellular ligand binding domains and intracellular signal transducing domains. For example, CD2 is characterized as a cell adhesion molecule found on the surface of T cells and is capable of initiating intracellular signals necessary for T cell activation. CD27 is characterized as a type II transmembrane glycoprotein belonging to the TNFR superfamily (TNFRSF) whose expression on B cells is induced by antigen-receptor activation in B cells. CD28 is one of the proteins on T cells and is the receptor for CD80 (B7.1) and CD86 (B7.2) ligands on antigen-presenting cells. CD137 (4-1BB) ligand is found on most leukocytes and on some non-immune cells. OX40 ligand is expressed on many antigen-presenting cells such as DC2s (dendritic cells), macrophages, and B lymphocytes. In one embodiment, the costimulatory receptor protein is full length CD28 as defined herein.
  • CD40 is a member of the tumor necrosis factor receptor (TNFR) superfamily and several isoforms are generated by alternative splicing. Its ligand, CD154 (also called CD40L) is a protein that is primarily expressed on activated T cells. For reference, the human CD40 isoform 1 protein sequence is set forth in GenBank accession No. NP_001241.1, including signal peptide (amino acids 1-20), transmembrane domain (amino acids 194-215), and cytoplasmic domain (amino acids 216-277) (SEQ ID NO:22). CD40 receptor signaling involves adaptor proteins including but not limited to TNF receptor-associated factors (TRAF), and the cytoplasmic domain comprises signaling components, including but not limited to an SH3 motif (KPTNKAPH), TRAF2 motif (PKQE, PVQE, SVQE), TRAF6 motif (QEPQEINFP) and PKA motif (KKPTNKA, SRISVQE). Further motifs for binding to TRAF1, TRAF2, TRAF3, and TRAF5 comprise the major consensus sequence (P/S/A/T)X(Q/E)E or minor consensus sequence PXQXXD and can be identified in or obtained from, without limitation, TNFR family members such as CD30, Ox40, 4-1BB, and the EBV oncoprotein LMP1. (See, e.g., Ye, H et al., The Structural Basis for the Recognition of Diverse Receptor Sequences by TRAF2. Molecular Cell, 1999; 4(3):321-30. doi: 10.1016/S1097-2765(00)80334-2; Park H H, Structure of TRAF Family: Current Understanding of Receptor Recognition. Front. Immunol. 2018; 9:1999. doi: 10.3389/fimmu.2018.01999).
  • Examples disclosed herein demonstrate operation of CD40 as a signaling domain and further that cytokine and chemokine expression profiles are altered by signaling domain selection. In this regard, the CD40 signaling domains of the invention provide distinct and overlapping responses induced by the different factor binding sites. (See, e.g., Ahonen, C L et al., The CD40-TRAF6 axis controls affinity maturation and the generation of long-lived plasma cells. Nat Immunol. 2002; 3: 451-456; Mackey M F et al., Distinct contributions of different CD40 TRAF binding sites to CD154-induced dendritic cell maturation and IL-12 secretion. Eur J Immunol. 2003; 33: 779-789; Mukundan L et al., TNF receptor-associated factor 6 is an essential mediator of CD40-activated proinflammatory pathways in monocytes and macrophages. J Immunol. 2005; 174: 1081-1090.
  • In some embodiments, a chimeric protein of the invention comprises substantially all of a CD40 costimulatory domain. In some embodiments, a chimeric protein of the invention comprises two or more CD40 costimulatory domains. In some embodiments, a chimeric protein of the invention comprises a CD40 costimulatory domain signaling component or motif, including but not limited to an SH3 motif (KPTNKAPH), TRAF2 motif (PKQE, PVQE, SVQE), TRAF3 motif, TRAF6 motif (QEPQEINFP) or PKA motif (KKPTNKA, SRISVQE) as well as two or more, or three or more, or four or more such components of motifs, which can be in multiple copies and arranged in any order. In some embodiments, a chimeric protein of the invention comprises a CD40 costimulatory domain and a CD40 costimulatory domain signaling component or motif.
  • In some embodiments, selection of one or more costimulatory domain signaling component or motif is guided by the cell in which the chimeric protein is to be expressed and/or a desired costimulatory activity more closely identified with a signaling component or motif, or avoidance of a costimulatory activity more closely identified with a signaling component or motif.
  • In some embodiments, a chimeric protein signaling domain comprises, in addition to a CD40 costimulatory domain or signaling component or motif thereof, or two or more such domains or components or motifs or combinations thereof, an additional full length costimulatory domain or signaling component thereof from, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6,
  • For reference, the human CD28 protein sequence is set forth in GenBank accession No. NP_006130.1, including signal peptide (amino acids 1-18), extracellular domain (amino acids 19-152), transmembrane domain (amino acids 153-179) and cytoplasmic domain (amino acids 180-200). The extracellular domain includes an immunoglobulin type domain (amino acids 21-136) which contains amino acids with compose the antigen binding site and amino acids that form the homodimer interface. The extracellular domain includes several asparagine residues which may be glycosylated, and the intracellular domain comprises serine and tyrosine residues, which may be phosphorylated.
  • For reference, the human CD8 alpha chain protein sequence is set forth by GenBank accession No. NP_001139345.1, including signal peptide (amino acids 1-21), extracellular domain (amino acids 22-182), transmembrane domain (amino acids 183-203), and cytoplasmic domain (amino acids 204-235). The extracellular domain includes an immunoglobulin type domain (amino acids 28-128) which contains amino acids with compose the antigen binding site and amino acids that form the homodimer interface. The extracellular domain includes several asparagine residues which may be glycosylated, and the intracellular domain comprises serine and tyrosine residues, which may be phosphorylated.
  • For reference, the human IgG4 constant region sequence is set forth in UniProtKB/Swiss-Prot: accession No. P01861.1, including CH1 (amino acids 1-98), hinge (amino acids 99-110), CH2 (amino acids 111-220), CH3 (amino acids 221-327). The CH2 region includes asparagine at amino acid 177, which is the glycosylated and associated with Fc receptor and antibody-dependent cell-mediated cytotoxicity (ADCC).
  • For reference, the protein sequence of human CD137 (41BB), another TNFR superfamily member, is set forth by GenBank accession No. NP_001552.2, including signal peptide (amino acids 1-23), extracellular domain (amino acids 24-186), transmembrane domain (amino acids 187-213), and cytoplasmic domain (amino acids 214-255).
  • For reference, the human CD134 (OX40) protein sequence is set forth by GenBank accession No. NP_003318.1, including signal peptide (amino acids 1-28), extracellular domain (amino acids 29-214), transmembrane domain (amino acids 215-235), and cytoplasmic domain (amino acids 236-277). This receptor has been shown to activate NF-kappaB through its interaction with adaptor proteins TRAF2 and TRAF5 and studies suggest that this receptor promotes expression of apoptosis inhibitors BCL2 and BCL21L1/BCL2-XL.
  • The human T-cell surface antigen CD2 has at least two isoforms. For reference, the human CD2 isoform1 protein sequence is set forth by NP_001315538.1, including signal peptide (amino acids 1-24), extracellular domain (amino acids 25-235), transmembrane domain (amino acids 236-261), and cytoplasmic domain (amino acids 262-377). The human CD2 isoform2 protein sequence is set forth by NP_001758.2
  • For reference, the human CD357 (GITR) isoform-1 protein sequence is set forth by GenBank accession No. NP_004186.1, including signal peptide (amino acids 1-25), extracellular domain (amino acids 26-162), transmembrane domain (amino acids 163-183), and cytoplasmic domain (amino acids 184-241).
  • For reference, the human CD29 (beta1 integrin) protein sequence is set forth by GenBank accession No. NP_596867, including signal peptide (amino acids 1-20), extracellular domain (amino acids 21-728), transmembrane domain (amino acids 729-751), and cytoplasmic domain (amino acids 752-798).
  • The human CD150 (SLAM) protein sequence has at several isoforms. In addition to the transmembrane form of CD150 (mCD150), cells of hematopoietic lineage express mRNA encoding the secreted form of CD150 (sCD150), which lacks the entire transmembrane region of 30 amino acids. For reference, human SLAM isoform b is set forth by GenBank accession No. NP_003028.1, including signal peptide (amino acids 1-20), extracellular domain (amino acids 21-237), transmembrane domain (amino acids 238-258), and cytoplasmic domain (amino acids 259-335). Human SLAM isoform a is set forth by GenBank accession No. NP_001317683.1.
  • In embodiments of the invention, a chimeric protein may be expressed alone under the control of a promoter in a therapeutic population of cells that have therapeutic activity, for example, Tumor Infiltrating Lymphocytes (TILs). Alternatively, the chimeric protein may be expressed along with a therapeutic transgene such as a chimeric antigen receptor (CAR) and/or T-cell Receptor (TCR). Suitable TCRs and CARs are well known in the literature, for example HLA-A*02-NYESO-1 specific TCRs (Rapoport et al. Nat Med 2015) or anti-CD19scFv.CD3ζ fusion CARs (Kochenderfer et al. J Clin Oncol 2015) which have been successfully used to treat Myeloma or B-cell malignancies respectively. The chimeric proteins described herein may be expressed with any known CAR or TCR thus providing the cell with a regulatable growth switch to allow cell expansion in-vitro or in-vivo, and a conventional activation mechanism in the form of the TCR or CAR for anti-cancer activity. Thus the invention provides a cell for use in adoptive cell therapy comprising a chimeric protein as described herein and a TCR and/or CAR that specifically binds to a tumor associated antigen. An exemplary chimeric protein comprising CD28 includes an extracellular antigen binding domain and an extracellular, transmembrane and intracellular signaling domain.
  • A chimeric protein of the invention optionally comprises a spacer region between the TCR clustering domain and the costimulatory receptor. As used herein, the term “spacer” refers to the extracellular structural region of a chimeric protein that separates the TCR clustering domain from the signaling domain of the chimeric protein. In some embodiments long spacers are employed, for example to target membrane-proximal epitopes or glycosylated antigens (see Guest R. D. et al. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J. Immunother. 2005; 28:203-211; Wilkie S. et al., Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor. J. Immunol. 2008; 180:4901-4909). In other embodiments, chimeric proteins bear short spacers, for example to target membrane distal epitopes (see Hudecek M. et al., Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 2013; 19:3153-3164; Hudecek M. et al 27acarbazine27nalling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol. Res. 2015; 3:125-135). In some embodiments, the spacer comprises all or part of or is derived from an IgG hinge, including but not limited to IgG1, IgG2, or IgG4. By “derived from an Ig hinge” is meant a spacer comprising insertions, deletions, or mutations in an IgG hinge. In some embodiments, a spacer can comprise all or part of one or more antibody constant domains, such as but not limited to CH2 and/or CH3 domains. In some embodiments, in a spacer comprising all or part of a CH2 domain, the CH2 domain is modified so as not to bind to an Fc receptor. For example, Fc receptor binding in myeloid cells has been found to impair CAR T cell functionality. In some embodiments, the spacer comprises all or part of an Ig-like hinge from CD28, CD8, or other protein comprising a hinge region. In some embodiments of the invention that comprise a spacer, the spacer is from 1 and 50 amino acids in length.
  • In some embodiments, the chimeric protein extracellular domain comprises a linker. Linkers comprise short runs of amino acids used to connect domains, for example a binding domain with a spacer or transmembrane domain. In order for there to be flexibility to bind ligand, a ligand binding domain will usually be connected to a spacer or a transmembrane domain by flexible linker comprising from about 5 to 25 amino acids, such as, for example, AAAGSGGSG or GGGGSGGGGSGGGGS. In some embodiments, a chimeric protein comprises a TCR clustering domain joined directly to a signaling domain by a linker, and without a spacer. In some embodiments, a chimeric protein comprises a binding domain joined directly to a transmembrane by a spacer and without a linker.
  • As discussed above, in some embodiments, a chimeric protein comprises a full length primary costimulatory receptor which can comprise an extracellular ligand binding and intracellular signaling portion of, without limitation, CD2, CD9, CD26, CD27, CD28, CD29, CD38, CD40, CD43, CD46, CD49d, CD55, CD73, CD81, CD82, CD99, CD100, CD134 (OX40), CD137 (41BB), CD150 (SLAM), CD270 (HVEM), CD278 (ICOS), CD357 (GITR), or EphB6. In other embodiments, the chimeric protein, for instance may comprise an extracellular ligand binding domain of one of the aforementioned proteins and an intracellular signaling domain of another of the aforementioned proteins. In some embodiments, the signaling portion of the chimeric protein comprises a single signaling domain. In other embodiments, the signaling portion of the chimeric protein comprises a second intracellular signaling domain such as but not limited to: CD2, CD27, CD28, CD40, CD134 (OX40), CD137 (4-1BB), CD150 (SLAM). In some embodiments, the first and second intracellular signaling domains are the same. In other embodiments, the first and second intracellular signaling domains are different. In some embodiments, the costimulatory receptor is capable of dimerization. Without being bound by theory, it is thought that chimeric proteins dimerize or associate with other accessory molecules for signal initiation. In some embodiments, chimeric proteins dimerize or associate with accessory molecules through transmembrane domain interactions. In some embodiments, dimerization or association with accessory molecules is assisted by costimulatory receptor interactions in the intracellular portion, and/or the extracellular portion of the costimulatory receptor.
  • Although the main function of the transmembrane is to anchor the chimeric protein in the T cell membrane, in some embodiments, the transmembrane domain influences chimeric protein function. In some embodiments, the transmembrane domain is comprised by the full length primary costimulatory receptor domain. In embodiments of the invention wherein the chimeric protein construct comprises an extracellular domain of one receptor and an intracellular signaling domain of a second receptor, the transmembrane domain can be that of the extracellular domain or the intracellular domain. In some embodiments, the transmembrane domain is from CD4, CD8a, CD28, or ICOS. Gueden et al. associated use of the ICOS transmembrane domain with increased CART cell persistence and overall anti-tumor efficacy (Guedan S. et al., Enhancing CART cell persistence through ICOS and 4-1BB costimulation. JCI Insight. 2018; 3:96976). In an embodiment, the transmembrane domain comprises a hydrophobic α helix that spans the cell membrane.
  • In some embodiments, amino acid sequence variants of the TCR clustering domain or other moieties provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the moiety. Amino acid sequence variants of an antibody moiety may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the clustering moiety, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody moiety. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • In some embodiments, TCR clustering domain moieties comprising one or more amino acid substitutions, deletions, or insertions are provided. Amino acid substitutions may be introduced into a binding domain of interest and the products screened for a desired activity, e.g., retained/improved clustering or decreased immunogenicity. In some embodiments, amino acid substitutions may be introduced into one or more of the primary co-stimulatory receptor domain (extracellular or intracellular), secondary costimulatory receptor domain, or extracellular co-receptor domain. Accordingly, the invention encompasses chimeric proteins and component parts particularly disclosed herein as well as variants thereof, i.e. chimeric proteins and component parts having at least 75%, at least 80%, at least 85%, at least 87%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% sequence identity to the amino acid sequences particularly disclosed herein. The terms “percent similarity,” “percent identity,” and “percent homology” when referring to a particular sequence are used as set forth in the University of Wisconsin GCG software program BestFit. Other algorithms may be used, e.g. BLAST, psiBLAST or TBLASTN (which use the method of Altschul et al. (1990) J. Mol. Biol. 215: 405-410), FASTA (which uses the method of Pearson and Lipman (1988) PNAS USA 85: 2444-2448).
  • Particular amino acid sequence variants may differ from a reference sequence by insertion, addition, substitution or deletion of 1 amino acid, 2, 3, 4, 5-10, 10-20 or 20-30 amino acids. In some embodiments, a variant sequence may comprise the reference sequence with 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more residues inserted, deleted or substituted. For example, 5, 10, 15, up to 20, up to 30 or up to 40 residues may be inserted, deleted or substituted.
  • In some preferred embodiments, a variant may differ from a reference sequence by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more conservative substitutions. Conservative substitutions involve the replacement of an amino acid with a different amino acid having similar properties. For example, an aliphatic residue may be replaced by another aliphatic residue, a non-polar residue may be replaced by another non-polar residue, an acidic residue may be replaced by another acidic residue, a basic residue may be replaced by another basic residue, a polar residue may be replaced by another polar residue or an aromatic residue may be replaced by another aromatic residue. Conservative substitutions may, for example, be between amino acids within the following groups:
  • Conservative substitutions are shown in Table 1 below.
  • TABLE 1
    Original Exemplary Preferred
    Residue Substitutions Substitutions
    IArg (R) Lys; Gln; Asn Lys
    Asn (N) Gln; His; Asp; Lys; Arg Gln
    Asp (D) Glu; AsnIu
    Cys (C) Ser; Ala Ser
    Gln (Q) Asn; I Asn
    Glu (E) Asp; Gln Asp
    Gly (G) Ala Ala
    His (H) Asn; Gln; Lys; Arg Arg
    Ile (I) Leu; Val; Met; Ala; Phe; Norleucine Leu
    Leu (L) Norleucinne; Ile; Val; Met; Ala; Phe Ile
    Lys (K) Arg; Gln; Asn Arg
    Met (M) Leu; Phe; Ile Leu
    Phe (F) Trp; Leu; Val; Ile; Ala; Tyr Tyr
    Pro (P) Ala Ala
    Ser (S) Thr Thr
    Thr (T) Val; Ser Ser
    Trp (W) Tyr; Phe Tyr
    Tyr (Y) Trp; Phe; Thr; Ser Phe
    Val (V) Il e; Le u; Met; Phe; Ala; Norleucine Leu
  • Amino acids may be grouped into different classes according to common side-chain properties: a. hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile; b. neutral hydrophilic: Cys, Ser, Thr, Asn, Gln; c. acidic: Asp, Glu; d. basic: His, Lys, Arg; e. residues that influence chain orientation: Gly, Pro; aromatic: Trp, Tyr, Phe. Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • The cells used in the present invention may be any lymphocyte that is useful in adoptive cell therapy, such as a T-cell or a natural killer (NK) cell, an NKT cell, a gamma/delta T-cell or T regulatory cell. The cells may be allogeneic or autologous to the patient.
  • T cells or T lymphocytes are a type of lymphocyte that have a central role in cell-mediated immunity. They can be distinguished from other lymphocytes, such as B cells and natural killer cells (NK cells), by the presence of a T-cell receptor (TCR) on the cell surface. There are various types of T cell, as summarized below. Cytotoxic T cells (TC cells, or CTLs) destroy virally infected cells and tumor cells, and are also implicated in transplant rejection. CTLs express the CD8 molecule at their surface.
  • These cells recognize their targets by binding to antigen associated with MHC class I, which is present on the surface of all nucleated cells. Through IL-10, adenosine and other molecules secreted by regulatory T cells, the CD8+ cells can be inactivated to an anergic state, which prevent autoimmune diseases such as experimental autoimmune encephalomyelitis.
  • Memory T cells are a subset of antigen-specific T cells that persist long-term after an infection has resolved. They quickly expand to large numbers of effector T cells upon re-exposure to their cognate antigen, thus providing the imm“ne sys”em with “memory” against past infections. Memory T cells comprise three subtypes: central memory T cells (TCM cells) and two types of effector memory T cells (TEM cells and TEMRA cells). Memory cells may be either CD4+ or CD8+. Memory T cells typically express the cell surface protein CD45RO. Regulatory T cells (Treg cells), formerly known as suppressor T cells, are crucial for the maintenance of immunological tolerance. Their major role is to shut down T cell-mediated immunity toward the end of an immune reaction and to suppress auto-reactive T cells that escaped the process of negative selection in the thymus.
  • Two major classes of CD4+ Treg cells have been described—naturally occurring Treg cells and adaptive Treg cells. Naturally occurring Treg cells (also known as CD4+CD25+FoxP3+ Treg cells) arise in the thymus and have been linked to interactions between developing T cells with both myeloid (CD11c+) and plasmacytoid (CD123+) dendritic cells that have been activated with TSLP. Naturally occurring Treg cells can be distinguished from other T cells by the presence of an intracellular molecule called FoxP3. Adaptive Treg cells (also known as Tr1 cells or Th3 cells) may originate during a normal immune response.
  • Natural Killer Cells (or NK cells) are a type of cytolytic cell which form part of the innate immune system. NK cells provide rapid responses to innate signals from virally infected cells in an MHC independent manner. NK cells (belonging to the group of innate lymphoid cells) are defined as large granular lymphocytes (LGL) and constitute the third kind of cells differentiated from the common lymphoid progenitor generating B and T lymphocytes.
  • In some embodiments, therapeutic cells of the invention comprise autologous cells engineered to express a chimeric protein. In some embodiments, therapeutic cells of the invention comprise allogeneic cells engineered to express a chimeric protein. Autologous cells expressing chimeric proteins may be advantageous in avoiding graft-versus-host disease (GVHD) due to TCR-mediated recognition of recipient alloantigens.
  • An aspect of the invention provides a nucleic acid sequence of the invention, encoding any of the chimeric proteins, polypeptides, or proteins described herein (including functional portions and functional variants thereof). As used herein, the terms “polynucleotide”, “nucleotide”, and “nucleic acid” are intended to be synonymous with each other. It will be understood by a skilled person that numerous different polynucleotides and nucleic acids can encode the same polypeptide as a result of the degeneracy of the genetic code. In addition, it is to be understood that skilled persons may, using routine techniques, make nucleotide substitutions that do not affect the polypeptide sequence encoded by the polynucleotides described here to reflect the codon usage of any particular host organism in which the polypeptides are to be expressed, e.g. codon optimization. Nucleic acids according to the invention may comprise DNA or RNA. They may be single stranded or double-stranded. They may also be polynucleotides which include within them synthetic or modified nucleotides. A number of different types of modification to oligonucleotides are known in the art. These include methylphosphonate and phosphorothioate backbones, addition of acridine or polylysine ‘chains at’ the 3′ and/or 5′ ends of the molecule. For the purposes of the present invention, it is to be understood that the polynucleotides may be modified by any method available in the art. Such modifications may be carried out in order to enhance the in vivo activity or life span of polynucleotides of interest.
  • The terms “variant”, “homologue” or “derivative” in relation to a nucleotide sequence include any substitution of, variation of, modification of, replacement of, deletion of or addition of one (or more) nucleic acid from or to the sequence.
  • The nucleic acid sequence may encode the protein sequence shown as SEQ ID NO:2 or a variant thereof. The nucleotide sequence may comprise a codon optimized nucleic acid sequence shown engineered for expression in human cells.
  • The invention also provides a nucleic acid sequence which comprises a nucleic acid sequence encoding a chimeric protein and a further nucleic acid sequence encoding a T-cell receptor (TCR) and/or chimeric antigen receptor (CAR).
  • The nucleic acid sequences may be joined by a sequence allowing co-expression of the two or more nucleic acid sequences. For example, the construct may comprise an internal promoter, an internal ribosome entry sequence (IRES) sequence or a sequence encoding a cleavage site. The cleavage site may be self-cleaving, such that when the polypeptide is produced, it is immediately cleaved into the discrete proteins without the need for any external cleavage activity. Various self-cleaving sites are known, including the Foot- and Mouth disease virus (FMDV) and the 2A self-cleaving peptide. The co-expressing sequence may be an internal ribosome entry sequence (IRES). The co-expressing sequence may be an internal promoter.
  • In an aspect, the present invention provides a vector which comprises a nucleic acid sequence or nucleic acid construct of the invention.
  • Such a vector may be used to introduce the nucleic acid sequence(s) or nucleic acid construct(s) into a host cell so that it expresses one or more chimeric protein(s) according to the first aspect of the invention and, optionally, one or more other proteins of interest (POI), for example a TCR or a CAR. The vector may, for example, be a plasmid or a viral vector, such as a retroviral vector or a lentiviral vector, or a transposon-based vector or synthetic mRNA.
  • The nucleic acids of the present invention may also be used for nucleic acid immunization and gene therapy, using standard gene delivery protocols. Methods for gene delivery are known in the art. See, e.g., U.S. Pat. Nos. 5,399,346, 5,580,859, 5,589,466, incorporated by reference herein in their entireties.
  • Vectors derived from retroviruses, such as the lentivirus, are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene or transgenes and its propagation in daughter cells. The vector may be capable of transfecting or transducing a lymphocyte including a T cell or an NK cell. The present invention also provides vectors in which a nucleic acid of the present invention is inserted. The expression of natural or synthetic nucleic acids encoding a chimeric protein, and optionally a TCR or CAR is typically achieved by operably linking a nucleic acid encoding the chimeric protein and TCR/CAR polypeptide or portions thereof to one or more promoters, and incorporating the construct into an expression vector.
  • Additional promoter elements, e.g., enhancers, regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline.
  • One example of a suitable promoter is the immediate early cytomegalovirus (CMV) promoter sequence. This promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence operatively linked thereto. Another example of a suitable promoter is Elongation Growth Factor-1α (EF-1α). However, other constitutive promoter sequences may also be used, including, but not limited to the simian virus 40 (SV40) early promoter, mouse mammary tumor virus (MMTV), human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, MoMuLV promoter, MSCV promoter, MND promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter.
  • The vectors can be suitable for replication and integration in eukaryotic cells. Typical cloning vectors contain transcription and translation terminators, initiation sequences, and promoters useful for regulation of the expression of the desired nucleic acid sequence. Viral vector technology is well known in the art and is described, for example, in Sambrook et al. (2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York), and in other virology and molecular biology manuals, see also, WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193). In some embodiments, the constructs expressed are as shown in SEQ ID NOS:32-65 and 67-79. In some embodiments the nucleic acids are multi-cistronic constructs that permit the expression of multiple transgenes (e.g., chimeric protein and a TCR and/or CAR etc.) under the control of a single promoter. In some embodiments, the transgenes (e.g., chimeric protein and a TCR and/or CAR etc.) are separated by a self-cleaving 2A peptide. Examples of 2A peptides useful in the nucleic acid constructs of the invention include F2A, P2A, T2A and E2A. In other embodiments of the invention, the nucleic acid construct of the invention is a multi-cistronic construct comprising two promoters; one promoter driving the expression of chimeric protein and the other promoter driving the expression of the TCR or CAR. In some embodiments, the dual promoter constructs of the invention are uni-directional. In other embodiments, the dual promoter constructs of the invention are bi-directional. In order to assess the expression of the chimeric protein polypeptide or portions thereof, the expression vector to be introduced into a cell can also contain either a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from the population of cells sought to be transfected or transduced through viral vectors.
  • Prior to expansion and genetic modification, a source of cells (e.g., immune effector cells, e.g., T cells or NK cells) is obtained from a subject. The term “subject” is intended to include living organisms in which an immune response can be elicited (e.g., mammals). Examples of subjects include humans, dogs, cats, mice, rats, and transgenic species thereof. T cells can be obtained from a number of sources, including peripheral blood mononuclear cells, bone marrow, lymph node tissue, cord blood, thymus tissue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, and tumors.
  • In one aspect, T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation through a PERCOLL™ gradient or by counterflow centrifugal elutriation.
  • In another aspect, Tumor infiltrating cells (TILs) are isolated and/or expanded from a tumor, for example by a fragmented, dissected, or enzyme digested tumor biopsy or mass.
  • A specific subpopulation of T cells, such as CD3+, CD28+, CD4+, CD8+, CD45RA+, and CD45RO+T cells, can be further isolated by positive or negative selection techniques. For example, in one aspect, T cells are isolated by incubation with anti-CD3/anti-CD28-conjugated beads, such as DYNABEADS® M-450 CD3/CD28 T, for a time period sufficient for positive selection of the desired T cells. In one aspect, the time period is about 30 minutes. In a further aspect, the time period ranges from 30 minutes to 36 hours or longer and all integer values there between. In a further aspect, the time period is at least 1, 2, 3, 4, 5, or 6 hours. In yet another preferred aspect, the time period is 10 to 24 hours. In one aspect, the incubation time period is 24 hours. Longer incubation times may be used to isolate T cells in any situation where there are few T cells as compared to other cell types, such in isolating tumor infiltrating lymphocytes (TIL) from tumor tissue or from immunocompromised individuals. Further, use of longer incubation times can increase the efficiency of capture of CD8+ T cells. Thus, by simply shortening or lengthening the time T cells are allowed to bind to the CD3/CD28 beads and/or by increasing or decreasing the ratio of beads to T cells (as described further herein), subpopulations of T cells can be preferentially selected for or against at culture initiation or at other time points during the process. Additionally, by increasing or decreasing the ratio of anti-CD3 and/or anti-CD28 antibodies on the beads or other surface, subpopulations of T cells can be preferentially selected for or against at culture initiation or at other desired time points. The skilled artisan would recognize that multiple rounds of selection can also be used in the context of this invention. In certain aspects, it may be desirable to perform the selection procedure and use“the “unselected” cells in the activation and exp“nsion proc”ss. “Unselected” cells can also be subjected to further rounds of selection.
  • Enrichment of a T cell population by negative selection can be accomplished with a combination of antibodies directed to surface markers unique to the negatively selected cells. One method is cell sorting and/or selection via negative magnetic immunoadherence or flow cytometry that uses a cocktail of monoclonal antibodies directed to cell surface markers present on the cells negatively selected. For example, to enrich for CD4+ cells by negative selection, a monoclonal antibody cocktail typically includes antibodies to CD14, CD20, CD16, HLA-DR, and CD8. In certain aspects, it may be desirable to enrich for or positively select for regulatory T cells which typically express CD4+, CD25+, CD62Lhi, GITR+, CD137, PD1, TIM3, LAG-3, CD150 and FoxP3+. Alternatively, in certain aspects, T regulatory cells are depleted by anti-CD25 conjugated beads or other similar method of selection.
  • The methods described herein can include, e.g., selection of a specific subpopulation of immune effector cells, e.g., T cells, that are a T regulatory cell-depleted population, CD25+ depleted cells, using, e.g., a negative selection technique, e.g., described herein. Preferably, the population of T regulatory depleted cells contains less than 30%, 25%, 20%, 15%, 10%, 5%, 4%, 3%, 2%, 1% of CD25+ cells.
  • A specific subpopulation of chimeric protein effector cells that specifically bind to a target antigen can be enriched for by positive selection techniques. For example, in some embodiments, effector cells are enriched for by incubation with target antigen-conjugated beads for a time period sufficient for positive selection of the desired abTCR effector cells. In some embodiments, the time period is about 30 minutes. In some embodiments, the time period ranges from 30 minutes to 36 hours or longer (including all ranges between these values). In some embodiments, the time period is at least one, 2, 3, 4, 5, or 6 hours. In some embodiments, the time period is 10 to 24 hours. In some embodiments, the incubation time period is 24 hours. For isolation of effector cells present at low levels in the heterogeneous cell population, use of longer incubation times, such as 24 hours, can increase cell yield. Longer incubation times may be used to isolate effector cells in any situation where there are few effector cells as compared to other cell types. The skilled artisan would recognize that multiple rounds of selection can also be used in the context of this invention.
  • T cells for stimulation can also be frozen after a washing step. After the washing step that removes plasma and platelets, the cells may be suspended in a freezing solution. While many freezing solutions and parameters are known in the art and will be useful in this context, one method involves using PBS containing 20% DMSO and 8% human serum albumin, or culture media containing 10 % Dextran 40 and 5% Dextrose, 20% Human Serum Albumin and 7.5% DMSO, or 31.25% Plasmalyte-A, 31.25% Dextrose 5%, 0.45% NaCl, 10 % Dextran 40 and 5% Dextrose, 20% Human Serum Albumin, and 7.5% DMSO or other suitable cell freezing media containing for example, Hespan and PlasmaLyte A, the cells then are frozen to −80° C. at a rate of 1° per minute and stored in the vapor phase of a liquid nitrogen storage tank. Other methods of controlled freezing may be used as well as uncontrolled freezing immediately at −20° C. or in liquid nitrogen.
  • In embodiments described herein, the immune effector cell can be an allogeneic immune effector cell, e.g., T cell or NK cell.
  • A T cell described herein can be, e.g., engineered such that it does not express a functional HLA on its surface. For example, a T cell described herein, can be engineered such that cell surface expression HLA, e.g., HLA class 1 and/or HLA class II, is downregulated. In some aspects, downregulation of HLA may be accomplished by reducing or eliminating expression of beta-2 microglobulin (B2M).
  • In some embodiments, the T cell can lack a functional TCR and a functional HLA, e.g., HLA class I and/or HLA class II. Modified T cells that lack expression of a functional TCR and/or HLA can be obtained by any suitable means, including a knock out or knock down of one or more subunit of TCR or HLA. For example, the T cell can include a knock down of TCR and/or HLA using siRNA, shRNA, clustered regularly interspaced short palindromic repeats (CRISPR) transcription-activator like effector nuclease (TALEN), or zinc finger endonuclease (ZFN).
  • In some embodiments, the allogeneic cell can be a cell which does not expresses or expresses at low levels an inhibitory molecule, e.g. a cell engineered by any method described herein. For example, the cell can be a cell that does not express or expresses at low levels an inhibitory molecule, e.g., that can decrease the ability of a chimeric protein-expressing cell to mount an immune effector response. Examples of inhibitory molecules include PD1, PD-L1, PD-L2, CTLA4, TIM3, CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5), LAGS, VISTA, BTLA, TIGIT, LAIR1, CD160, 2B4, CD80, CD86, B7-H3 (CD276), B7-H4 (VTCN1), HVEM (TNFRSF14 or CD270), KIR, A2aR, MHC class I, MHC class II, Gal9, adenosine, and TGFR beta. Inhibition of an inhibitory molecule, e.g., by inhibition at the DNA, RNA or protein level, can optimize a CAR-expressing cell performance. In embodiments, an inhibitory nucleic acid, e.g., an inhibitory nucleic acid, e.g., a dsRNA, e.g., an siRNA or shRNA, a clustered regularly interspaced short palindromic repeats (CRISPR), a transcription-activator like effector nuclease (TALEN), or a zinc finger endonuclease (ZFN), e.g., as described herein, can be used.
  • T cells may be activated and expanded generally using methods as described, for example, in U.S. Pat. Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 20060121005.
  • Generally, the T cells of the invention may be expanded by contact with a surface having attached thereto an agent that stimulates a CD3/TCR complex associated signal and a ligand that stimulates a costimulatory molecule on the surface of the T cells. In particular, T cell populations may be stimulated as described herein, such as by contact with an anti-CD3 antibody, or antigen-binding fragment thereof, or an anti-CD2 antibody immobilized on a surface, or by contact with a protein kinase C activator (e.g., bryostatin) in conjunction with a calcium ionophore. For co-stimulation of an accessory molecule on the surface of the T cells, a ligand that binds the accessory molecule is used. For example, a population of T cells can be contacted with an anti-CD3 antibody and an anti-CD28 antibody, under conditions appropriate for stimulating proliferation of the T cells. To stimulate proliferation of either CD4+ T cells or CD8+ T cells, an anti-CD3 antibody and an anti-CD28 antibody can be used. Examples of an anti-CD28 antibody include 9.3, B-T3, XR-CD28 (Diaclone, Besancon, France) can be used as can other methods commonly known in the art (Berg et al., Transplant Proc. 30(8):3975-3977, 1998; Haanen et al., J. Exp. Med. 190(9):13191328, 1999; Garland et al., J. Immunol Meth. 227(1-2):53-63, 1999).
  • In some embodiments, expansion can be performed using flasks or containers, or gas-permeable containers known by those of skill in the art and can proceed for 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days, about 7 days to about 14 days, about 8 days to about 14 days, about 9 days to about 14 days, about 10 days to about 14 days, about 11 days to about 14 days, about 12 days to about 14 days, or about 13 days to about 14 days. In some embodiments, the second TIL expansion can proceed for about 14 days.
  • In some embodiments, the expansion can be performed using non-specific T-cell receptor stimulation in the presence of interleukin-2 (IL-2) or interleukin-15 (IL-15). The non-specific T-cell receptor stimulus can include, for example, an anti-CD3 antibody, such as about 30 ng/ml of OKT3, a mouse monoclonal anti-CD3 antibody (commercially available from Ortho-McNeil, Raritan, N.J. or Miltenyi Biotech, Auburn, Calif.) or UHCT-1 (commercially available from BioLegend, San Diego, Calif., USA). Chimeric protein cells can be expanded in vitro by including one or more antigens, including antigenic portions thereof, such as epitope(s), of a cancer, which can be optionally expressed from a vector, such as a human leukocyte antigen A2 (HLA-A2) binding peptide, e.g., 0.3 .mu.M MART-1:26-35 (27L) or gp100:209-217 (210M), optionally in the presence of a T-cell growth factor, such as 300 IU/mL IL-2 or IL-15. Other suitable antigens may include, e.g., NY-ESO-1, TRP-1, TRP-2, tyrosinase cancer antigen, MAGE-A3, SSX-2, and VEGFR2, or antigenic portions thereof. Chimeric protein cells may also be rapidly expanded by re-stimulation with the same antigen(s) of the cancer pulsed onto HLA-A2-expressing antigen-presenting cells. Alternatively, the chimeric protein cells can be further stimulated with, e.g., example, irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2. In some embodiments, the stimulation occurs as part of the expansion. In some embodiments, the expansion occurs in the presence of irradiated, autologous lymphocytes or with irradiated HLA-A2+ allogeneic lymphocytes and IL-2.
  • In some embodiments, the cell culture medium comprises IL-2. In some embodiments, the cell culture medium comprises about 1000 IU/mL, about 1500 IU/mL, about 2000 IU/mL, about 2500 IU/mL, about 3000 IU/mL, about 3500 IU/mL, about 4000 IU/mL, about 4500 IU/mL, about 5000 IU/mL, about 5500 IU/mL, about 6000 IU/mL, about 6500 IU/mL, about 7000 IU/mL, about 7500 IU/mL, or about 8000 IU/mL, or between 1000 and 2000 IU/mL, between 2000 and 3000 IU/mL, between 3000 and 4000 IU/mL, between 4000 and 5000 IU/mL, between 5000 and 6000 IU/mL, between 6000 and 7000 IU/mL, between 7000 and 8000 IU/mL, or between 8000 IU/mL of IL-2.
  • In some embodiments, the cell culture medium comprises OKT3 antibody. In some embodiments, the cell culture medium comprises about 0.1 ng/mL, about 0.5 ng/mL, about 1 ng/mL, about 2.5 ng/mL, about 5 ng/mL, about 7.5 ng/mL, about 10 ng/mL, about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 50 ng/mL, about 60 ng/mL, about 70 ng/mL, about 80 ng/mL, about 90 ng/mL, about 100 ng/mL, about 200 ng/mL, about 500 ng/mL, about 1 μg/mL or between 0.1 ng/mL and 1 ng/mL, between 1 ng/mL and 5 ng/mL, between 5 ng/mL and 10 ng/mL, between 10 ng/mL and 20 ng/mL, between 20 ng/mL and 30 ng/mL, between 30 ng/mL and 40 ng/mL, between 40 ng/mL and 50 ng/mL, or between 50 ng/mL and 100 ng/mL of OKT3 antibody.
  • In some embodiments, a combination of IL-2, IL-7, IL-15, and/or IL-21 are employed as a combination during the expansion. In some embodiments, IL-2, IL-7, IL-15, and/or IL-21 as well as any combinations thereof can be included during the expansion. In some embodiments, a combination of IL-2, IL-15, and IL-21 are employed as a combination during the expansion. In some embodiments, IL-2, IL-15, and IL-21 as well as any combinations thereof can be included.
  • In some embodiments, the expansion can be conducted in a supplemented cell culture medium comprising IL-2, OKT-3, and antigen-presenting feeder cells.
  • In some embodiments, the expansion culture media comprises about 500 IU/mL of IL-15, about 400 IU/mL of IL-15, about 300 IU/mL of IL-15, about 200 IU/mL of IL-15, about 180 IU/mL of IL-15, about 160 IU/mL of IL-15, about 140 IU/mL of IL-15, about 120 IU/mL of IL-15, or about 100 IU/mL of IL-15, or about 500 IU/mL of IL-15 to about 100 IU/mL of IL-15, or about 400 IU/mL of IL-15 to about 100 IU/mL of IL-15 or about 300 IU/mL of IL-15 to about 100 IU/mL of IL-15 or about 200 IU/mL of IL-15, or about 180 IU/mL of IL-15.
  • In some embodiments, the expansion culture media comprises about 20 IU/mL of IL-21, about 15 IU/mL of IL-21, about 12 IU/mL of IL-21, about 10 IU/mL of IL-21, about 5 IU/mL of IL-21, about 4 IU/mL of IL-21, about 3 IU/mL of IL-21, about 2 IU/mL of IL-21, about 1 IU/mL of IL-21, or about 0.5 IU/mL of IL-21, or about 20 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 15 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 12 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 10 IU/mL of IL-21 to about 0.5 IU/mL of IL-21, or about 5 IU/mL of IL-21 to about 1 IU/mL of IL-21, or about 2 IU/mL of IL-21. In some embodiments, the cell culture medium comprises about 1 IU/mL of IL-21, or about 0.5 IU/mL of IL-21.
  • In some embodiments the antigen-presenting feeder cells (APCs) are PBMCs. In an embodiment, the ratio of chimeric protein cells to PBMCs and/or antigen-presenting cells in the expansion is about 1 to 25, about 1 to 50, about 1 to 100, about 1 to 125, about 1 to 150, about 1 to 175, about 1 to 200, about 1 to 225, about 1 to 250, about 1 to 275, about 1 to 300, about 1 to 325, about 1 to 350, about 1 to 375, about 1 to 400, or about 1 to 500, or between 1 to 50 and 1 to 300, or between 1 to 100 and 1 to 200.
  • In certain aspects, the primary stimulatory signal and the costimulatory signal for the T cell may be provided by different protocols. For example, the agents providing each signal may be in solution or coupled to a surface. When coupled to a surface, the agents may be coupled to the same su“fac” (i.e., in “cis” formation) or to separate sur“aces “i.e., in “trans” formation). Alternatively, one agent may be coupled to a surface and the other agent in solution. In one aspect, the agent providing the costimulatory signal is bound to a cell surface and the agent providing the primary activation signal is in solution or coupled to a surface. In certain aspects, both agents can be in solution. In one aspect, the agents may be in soluble form, and then cross-linked to a surface, such as a cell expressing Fc receptors or an antibody or other binding agent which will bind to the agents. In this regard, see for example, U.S. Patent Application Publication Nos. 20040101519 and 20060034810 for artificial antigen presenting cells (aAPCs) that are contemplated for use in activating and expanding T cells in the present invention.
  • In one aspect, the two agents are immobilized on beads, either on the same bead, i.e., “cis,” or to separate beads, i.e., “trans.” By way of example, the agent providing the primary activation signal is an anti-CD3 antibody or an antigen-binding fragment thereof and the agent providing the costimulatory signal is an anti-CD28 antibody or antigen-binding fragment thereof; and both agents are co-immobilized to the same bead in equivalent molecular amounts. In one aspect, a 1:1 ratio of each antibody bound to the beads for CD4+ T cell expansion and T cell growth is used. In certain aspects of the present invention, a ratio of anti CD3:CD28 antibodies bound to the beads is used such that an increase in T cell expansion is observed as compared to the expansion observed using a ratio of 1:1. In one particular aspect an increase of from about 1 to about 3 fold is observed as compared to the expansion observed using a ratio of 1:1. In one aspect, the ratio of CD3:CD28 antibody bound to the beads ranges from 100:1 to 1:100 and all integer values there between. In one aspect of the present invention, more anti-CD28 antibody is bound to the particles than anti-CD3 antibody, i.e., the ratio of CD3:CD28 is less than one. In certain aspects of the invention, the ratio of anti CD28 antibody to anti CD3 antibody bound to the beads is greater than 2:1. In one particular aspect, a 1:100 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:75 CD3:CD28 ratio of antibody bound to beads is used. In a further aspect, a 1:50 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:30 CD3:CD28 ratio of antibody bound to beads is used. In one preferred aspect, a 1:10 CD3:CD28 ratio of antibody bound to beads is used. In one aspect, a 1:3 CD3:CD28 ratio of antibody bound to the beads is used. In yet one aspect, a 3:1 CD3:CD28 ratio of antibody bound to the beads is used.
  • Ratios of particles to cells from 1:500 to 500:1 and any integer values in between may be used to stimulate T cells or other target cells. As those of ordinary skill in the art can readily appreciate, the ratio of particles to cells may depend on particle size relative to the target cell. For example, small sized beads could only bind a few cells, while larger beads could bind many. In certain aspects the ratio of cells to particles ranges from 1:100 to 100:1 and any integer values in-between and in further aspects the ratio comprises 1:9 to 9:1 and any integer values in between, can also be used to stimulate T cells. The ratio of anti-CD3- and anti-CD28-coupled particles to T cells that result in T cell stimulation can vary as noted above, however certain preferred values include 1:100, 1:50, 1:40, 1:30, 1:20, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 10:1, and 15:1 with one preferred ratio being at least 1:1 particles per T cell. In one aspect, a ratio of particles to cells of 1:1 or less is used. In one particular aspect, a preferred particle:cell ratio is 1:5. In further aspects, the ratio of particles to cells can be varied depending on the day of stimulation. For example, in one aspect, the ratio of particles to cells is from 1:1 to 10:1 on the first day and additional particles are added to the cells every day or every other day thereafter for up to 10 days, at final ratios of from 1:1 to 1:10 (based on cell counts on the day of addition). In one particular aspect, the ratio of particles to cells is 1:1 on the first day of stimulation and adjusted to 1:5 on the third and fifth days of stimulation. In one aspect, particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:5 on the third and fifth days of stimulation. In one aspect, the ratio of particles to cells is 2:1 on the first day of stimulation and adjusted to 1:10 on the third and fifth days of stimulation. In one aspect, particles are added on a daily or every other day basis to a final ratio of 1:1 on the first day, and 1:10 on the third and fifth days of stimulation. One of skill in the art will appreciate that a variety of other ratios may be suitable for use in the present invention. In particular, ratios will vary depending on particle size and on cell size and type. In one aspect, the most typical ratios for use are in the neighborhood of 1:1, 2:1 and 3:1 on the first day.
  • In further aspects of the present invention, the cells, such as T cells, are combined with agent-coated beads, the beads and the cells are subsequently separated, and then the cells are cultured. In an alternative aspect, prior to culture, the agent-coated beads and cells are not separated but are cultured together. In a further aspect, the beads and cells are first concentrated by application of a force, such as a magnetic force, resulting in increased ligation of cell surface markers, thereby inducing cell stimulation.
  • Viral- and non-viral-based genetic engineering tools can be used to generate chimeric protein cells, including without limitation T cells, NK cells resulting in permanent or transient expression of therapeutic genes. Retrovirus-based gene delivery is a mature, well-characterized technology, which has been used to permanently integrate CARs into the host cell genome (Scholler J., e.g. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci. Transl. Med. 2012; 4:132ra53; Rosenberg S. A. et al., Gene transfer into humans—immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N. Engl. J. Med. 1990; 323:570-578)
  • Non-viral DNA transfection methods can also be used. For example, Singh et al describes use of the Sleeping Beauty (SB) transposon system developed to engineer CAR T cells (Singh H., et al., Redirecting specificity of T-cell populations for CD19 using the Sleeping Beauty system. Cancer Res. 2008; 68:2961-2971) and is being used in clinical trials (see e.g., ClinicalTrials.gov: NCT00968760 and NCT01653717). The same technology is applicable to engineer chimeric protein cells.
  • Multiple SB enzymes have been used to deliver transgenes. Mates describes a hyperactive transposase (SB100X) with approximately 100-fold enhancement in efficiency when compared to the first-generation transposase. SB100X supported 35-50% stable gene transfer in human CD34(+) cells enriched in hematopoietic stem or progenitor cells. (Mates L. et al., Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat. Genet. 2009; 41:753-761) and multiple transgenes can be delivered from multicistronic single plasmids (e.g., Thokala R. et al., Redirecting specificity of T cells using the Sleeping Beauty system to express chimeric antigen receptors by mix-and-matching of VL and VH domains targeting cD123+ tumors. PLoS ONE. 2016; 11:e0159477) or multiple plasmids (e.g., Hurton L. V. et al., Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc. Natl. Acad. Sci. USA. 2016; 113:E7788-E7797). Such systems are used with chimeric proteins of the invention.
  • Morita et al, describes the piggyBac transposon system to integrate larger transgenes (Morita D. et al., Enhanced expression of anti-CD19 chimeric antigen receptor in piggyBac transposon-engineered T cells. Mol. Ther. Methods Clin. Dev. 2017; 8:131-140) Nakazawa et al. describes use of the system to generate EBV-specific cytotoxic T-cells expressing HER2-specific chimeric antigen receptor (Nakazawa Y et al, PiggyBac-mediated cancer immunotherapy using EBV-specific cytotoxic T-cells expressing HER2-specific chimeric antigen receptor. Mol. Ther. 2011; 19:2133-2143). Manuri et al used the system to generate CD-19 specific T cells (Manuri P. V. R. et al., piggyBac transposon/transposase system to generate CD19-specific T cells for the treatment of B-lineage malignancies. Hum. Gene Ther. 2010; 21:427-437).
  • Transposon technology is easy and economical. One potential drawback is the longer expansion protocols currently employed may result in T cell differentiation, impaired activity and poor persistence of the infused cells. Monjezi et al describe development minicircle vectors that minimize these difficulties through higher efficiency integrations (Monjezi R. et al., Enhanced CAR T-cell engineering using non-viral Sleeping Beauty transposition from minicircle vectors. Leukemia. 2017; 31:186-194). These transposon technologies can be used for chimeric proteins of the invention.
  • The present invention also relates to a pharmaceutical composition containing a vector or a chimeric protein expressing cell of the invention together with a pharmaceutically acceptable carrier, diluent or excipient, and optionally one or more further pharmaceutically active polypeptides and/or compounds.
  • In some embodiments, a pharmaceutical composition is provided comprising a chimeric protein described above and a pharmaceutically acceptable carrier. In some embodiments, a pharmaceutical composition is provided comprising a nucleic acid encoding a chimeric protein according to any of the embodiments described above and a pharmaceutically acceptable carrier. In some embodiments, a pharmaceutical composition is provided comprising an effector cell expressing a chimeric protein described above and a pharmaceutically acceptable carrier. Such a formulation may, for example, be in a form suitable for intravenous infusion.
  • As used herein, by “pharmaceutically acceptable” or “pharmacologically compatible” is meant a material that is not biologically or otherwise undesirable, e.g., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained. Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration.
  • An aspect of the invention provides a population of modified T cells expressing a recombinant chimeric protein. A suitable population may be produced by a method described above.
  • The population of modified T cells may be for use as a medicament. For example, a population of modified T cells as described herein may be used in cancer immunotherapy therapy, for example adoptive T cell therapy.
  • Other aspects of the invention provide the use of a population of modified T cells as described herein for the manufacture of a medicament for the treatment of cancer, a population of modified T cells as described herein for the treatment of cancer, and a method of treatment of cancer may comprise administering a population of modified T cells as described herein to an individual in need thereof.
  • The population of modified T cells may be autologous i.e. the modified T cells were originally obtained from the same individual to whom they are subsequently administered (i.e. the donor and recipient individual are the same). A suitable population of modified T cells for administration to the individual may be produced by a method comprising providing an initial population of T cells obtained from the individual, modifying the T cells to express a cAMP PDE or fragment thereof and an antigen receptor which binds specifically to cancer cells in the individual, and culturing the modified T cells.
  • The population of modified T cells may be allogeneic i.e. the modified T cells were originally obtained from a different individual to the individual to whom they are subsequently administered (i.e. the donor and recipient individual are different). The donor and recipient individuals may be HLA matched to avoid GVHD and other undesirable immune effects. A suitable population of modified T cells for administration to a recipient individual may be produced by a method comprising providing an initial population of T cells obtained from a donor individual, modifying the T cells to express a chimeric protein which binds specifically to cancer cells in the recipient individual, and culturing the modified T cells.
  • Following administration of the modified T cells, the recipient individual may exhibit a T cell mediated immune response against cancer cells in the recipient individual. This may have a beneficial effect on the cancer condition in the individual.
  • Cancer conditions may be characterized by the abnormal proliferation of malignant cancer cells and may include leukaemias, such as AML, CML, ALL and CLL, lymphomas, such as Hodgkin lymphoma, non-Hodgkin lymphoma and multiple myeloma, and solid cancers such as sarcomas, skin cancer, melanoma, bladder cancer, brain cancer, breast cancer, uterus cancer, ovary cancer, prostate cancer, lung cancer, colorectal cancer, cervical cancer, liver cancer, head and neck cancer, oesophageal cancer, pancreas cancer, renal cancer, adrenal cancer, stomach cancer, testicular cancer, cancer of the gall bladder and biliary tracts, thyroid cancer, thymus cancer, cancer of bone, and cerebral cancer, as well as cancer of unknown primary (CUP).
  • Cancer cells within an individual may be immunologically distinct from normal somatic cells in the individual (i.e. the cancerous tumor may be immunogenic). For example, the cancer cells may be capable of eliciting a systemic immune response in the individual against one or more antigens expressed by the cancer cells. The tumor antigens that elicit the immune response may be specific to cancer cells or may be shared by one or more normal cells in the individual.
  • An individual suitable for treatment as described above may be a mammal, such as a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon), an ape (e.g. gorilla, chimpanzee, orangutan, gibbon), or a human.
  • In preferred embodiments, the individual is a human. In other preferred embodiments, non-human mammals, especially mammals that are conventionally used as models for demonstrating therapeutic efficacy in humans (e.g. murine, primate, porcine, canine, or rabbit animals) may be employed.
  • The term “therapeutically effective amount” refers to an amount of a chimeric protein or composition comprising a chimeric protein as disclosed herei“, eff”ctive to “treat” a disease or disorder in an individual. In the case of cancer, the therapeutically effective amount of a chimeric protein or composition comprising a chimeric protein as disclosed herein can reduce the number of cancer cells; reduce the tumor size or weight; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent a chimeric protein or composition comprising a chimeric protein as disclosed herein can prevent growth and/or kill existing cancer cells, it can be cytostatic and/or cytotoxic. In some embodiments, the therapeutically effective amount is a growth inhibitory amount. In some embodiments, the therapeutically effective amount is an amount that improves progression free survival of a patient. In the case of infectious disease, such as viral infection, the therapeutically effective amount of a chimeric protein or composition comprising a chimeric protein as disclosed herein can reduce the number of cells infected by the pathogen; reduce the production or release of pathogen-derived antigens; inhibit (i.e., slow to some extent and preferably stop) spread of the pathogen to uninfected cells; and/or relieve to some extent one or more symptoms associated with the infection. In some embodiments, the therapeutically effective amount is an amount that extends the survival of a patient.
  • Cells, including T and NK cells, expressing chimeric proteins for use in the methods of the present may either be created ex vivo eithe' from a patient's own peripheral blood (autologous), or in the setting of a haematopoietic stem cell transplant from donor peripheral blood (allogenic), or peripheral blood from an unconnected donor (allogenic). Alternatively, T-cells or NK cells may be derived from ex-vivo differentiation of inducible progenitor cells or embryonic progenitor cells to T-cells or NK cells. In these instances, T-cells expressing a chimeric protein and, optionally, a CAR and/or TCR, are generated by introducing DNA or RNA coding for the chimeric protein and, optionally, a CAR and/or TCR, by one of many means including transduction with a viral vector, transfection with DNA or RNA.
  • T or NK cells expressing a chimeric protein of the present invention and, optionally, expressing a TCR and/or CAR may be used for the treatment of haematological cancers or solid tumors.
  • A method for the treatment of disease relates to the therapeutic use of a vector or cell, including a T or NK cell, of the invention. In this respect, the vector, or T or NK cell may be administered to a subject having an existing disease or condition in order to lessen, reduce or improve at least one symptom associated with the disease and/or to slow down, reduce or block the progression of the disease. The method of the invention may cause or promote T-cell mediated killing of cancer cells. The vector, or T or NK cell according to the present invention may be administered to a patient with one or more additional therapeutic agents. The one or more additional therapeutic agents can be co-administered to the patient. By “co-administering” is meant administering one or more additional therapeutic agents and the vector, or T or NK cell of the present invention sufficiently close in time such that the vector, or T or NK cell can enhance the effect of one or more additional therapeutic agents, or vice versa. In this regard, the vectors or cells can be administered first and the one or more additional therapeutic agents can be administered second, or vice versa. Alternatively, the vectors or cells and the one or more additional therapeutic agents can be administered simultaneously. One co-administered therapeutic agent that may be useful is IL-2, as this is currently used in existing cell therapies to boost the activity of administered cells. However, IL-2 treatment is associated with toxicity and tolerability issues.
  • In some embodiments, the addition of the engineered protein to a subject induces cytokine secretion. In some embodiments, the addition of CoStAR to a subject induces cytokine secretion. In some embodiments, the cytokine secretion lowers cytokine levels in the subject, including but not limited to IL-2. In some embodiments, the cytokine secretion following CoStAR exposure results in no detectable IL-2 in the subject. In some embodiments, the addition of the engineered protein to a subject reduces or eliminates the need for administration of exogenous IL-2. In some embodiments, the addition of the CoStAR to a subject reduces or eliminates the need for administration of exogenous IL-2.
  • In some embodiments, other mechanisms of action are involved in the killing of tumor cells apart from the direct effect of CoStAR. In some embodiments, secretion of cytokines and/or proliferation are evaluated. In some embodiments, tumor cell killing potency is characterized by flow cytometry to enumerate T cells and target cells and plate-based fluorescence or luminescence to measure percent killing. In some embodiments, cytokine secretion potency is characterized at the single cell level by flow cytometry and ELISA/MSD to characterize the population. In some embodiments, proliferation potency is determined by flow cytometry to characterize the population. In some embodiments, TIL potency may be determined by additional analytes, memory phenotype, cytotoxicity using cell lines, cytotoxicity using a patient specific tumor, a cytokine panel, cell proliferation and/or cellular composition.
  • As mentioned, for administration to a patient, the chimeric protein effector cells can be allogeneic or autologous to the patient. In some embodiments, allogeneic cells are further genetically modified, for example by gene editing, so as to minimize or prevent GVHD and/or a patient's immune response against the chimeric protein cells.
  • The chimeric protein effector cells are used to treat cancers and neoplastic diseases associated with a target antigen. Cancers and neoplastic diseases that may be treated using any of the methods described herein include tumors that are not vascularized, or not yet substantially vascularized, as well as vascularized tumors. The cancers may comprise non-solid tumors (such as hematological tumors, for example, leukemias and lymphomas) or may comprise solid tumors. Types of cancers to be treated with the chimeric protein effector cells of the invention include, but are not limited to, carcinoma, blastoma, and sarcoma, and certain leukemia or lymphoid malignancies, benign and malignant tumors, and malignancies e.g., sarcomas, carcinomas, and melanomas. Adult tumors/cancers and pediatric tumors/cancers are also included.
  • Hematologic cancers are cancers of the blood or bone marrow. Examples of hematological (or hematogenous) cancers include leukemias, including acute leukemias (such as acute lymphocytic leukemia, acute myelocytic leukemia, acute myelogenous leukemia and myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia), chronic leukemias (such as chronic myelocytic (granulocytic) leukemia, chronic myelogenous leukemia, and chronic lymphocytic leukemia), polycythemia vera, l'mphoma, Hodgkin's dise'se, non-Hodgkin's lymphoma (indolent and high grade forms), multiple myeloma, plasmacyt'ma, Waldenstrom's macroglobulinemia, heavy chain disease, myelodysplastic syndrome, hairy cell leukemia and myelodysplasia.
  • Solid tumors are abnormal masses of tissue that usually do not contain cysts or liquid areas. Solid tumors can be benign or malignant. Different types of solid tumors are named for the type of cells that form them (such as sarcomas, carcinomas, and lymphomas). Examples of solid tumors, such as sarcomas and carcinomas, include adrenocortical carcinoma, cholangiocarcinoma, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteosarcoma, and other sarcomas, synovioma, mes'thelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, stomach cancer, lymphoid malignancy, pancreatic cancer, breast cancer, lung cancers, ovarian cancer, prostate cancer, hepatocellular carcinoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, thyroid cancer (e.g., medullary thyroid carcinoma and papillary thyroid carcinoma), pheochromocytomas sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, chorio'arcinoma, Wilms' tumor, cervical cancer (e.g., cervical carcinoma and pre-invasive cervical dysplasia), colorectal cancer, cancer of the anus, anal canal, or anorectum, vaginal cancer, cancer of the vulva (e.g., squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, and fibrosarcoma), penile cancer, oropharyngeal cancer, esophageal cancer, head cancers (e.g., squamous cell carcinoma), neck cancers (e.g., squamous cell carcinoma), testicular cancer (e.g., seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, Leydig cell tumor, fibroma, fibroadenoma, adenomatoid tumors, and lipoma), bladder carcinoma, kidney cancer, melanoma, cancer of the uterus (e.g., endometrial carcinoma), urothelial cancers (e.g., squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma, ureter cancer, and urinary bladder cancer), and CNS tumors (such as a glioma (such as brainstem glioma and mixed gliomas), glioblastoma (also known as glioblastoma multiforme) astrocytoma, CNS lymphoma, germinoma, medulloblastoma, Schwannoma craniopharyogioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, neuroblastoma, retinoblastoma and brain metastases).
  • When “an immunologically effective amount,” “an anti-tumor effective amount,” “a tumor-inhibiting effective amount,” or “therapeutic amount” is indicated, the 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 the T cells described herein may be administered at a dosage of 104 to 109 cells/kg body weight, in some instances 105 to 106 cells/kg body weight, including all integer values within those ranges. 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).
  • A chimeric protein-expressing cell described herein may be used in combination with other known agents and therapie. “Administered” “in combination”, as used herein, means that two (or more) different treatments are delivered to the subject during the cours' of the subject's affliction with the disorder, e.g., the two or more treatments are delivered after the subject has been diagnosed with the disorder and before the disorder has been cured or eliminated or treatment has ceased for other reasons. In some embodiments, the delivery of one treatment is still occurring when the delivery of the second begins, so that there is overlap in terms of administration. This is sometimes referr“d to herein”s “s“multaneous” or “con”urrent delivery”. In other embodiments, the delivery of one treatment ends before the delivery of the other treatment begins. In some embodiments of either case, the treatment is more effective because of combined administration. For example, the second treatment is more effective, e.g., an equivalent effect is seen with less of the second treatment, or the second treatment reduces symptoms to a greater extent, than would be seen if the second treatment were administered in the absence of the first treatment, or the analogous situation is seen with the first treatment. In some embodiments, delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one treatment delivered in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive. The delivery can be such that an effect of the first treatment delivered is still detectable when the second is delivered.
  • A chimeric protein-expressing cell described herein and the at least one additional therapeutic agent can be administered simultaneously, in the same or in separate compositions, or sequentially. For sequential administration, the CAR-expressing cell described herein can be administered first, and the additional agent can be administered second, or the order of administration can be reversed.
  • The chimeric protein therapy and/or other therapeutic agents, procedures or modalities can be administered during periods of active disorder, or during a period of remission or less active disease. The chimeric protein therapy can be administered before the other treatment, concurrently with the treatment, post-treatment, or during remission of the disorder.
  • When administered in combination, the therapy and the additional agent (e.g., second or third agent), or all, can be administered in an amount or dose that is higher, lower or the same than the amount or dosage of each agent used individually, e.g., as a monotherapy. In some embodiments, the administered amount or dosage of the chimeric protein therapy, the additional agent (e.g., second or third agent), or all, is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of each agent used individually, e.g., as a monotherapy. In other embodiments, the amount or dosage of the chimeric protein therapy, the additional agent (e.g., second or third agent), or all, that results in a desired effect (e.g., treatment of cancer) is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower) than the amount or dosage of each agent used individually, e.g., as a monotherapy, required to achieve the same therapeutic effect.
  • In further aspects, a chimeric protein-expressing cell described herein may be used in a treatment regimen in combination with surgery, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies or other antibody therapies, cytoxin, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, cytokines, and irradiation, peptide vaccine, such as that described in Izumoto et al. 2008 J Neurosurg 108:963-971.
  • In certain instances, compounds of the present invention are combined with other therapeutic agents, such as other anti-cancer agents, anti-allergic agents, anti-nausea agents (or anti-emetics), pain relievers, cytoprotective agents, and combinations thereof.
  • In one embodiment, a chimeric protein-expressing cell described herein can be used in combination with a chemotherapeutic agent. Exemplary chemotherapeutic agents include an anthracycline (e.g., doxorubicin (e.g., liposomal doxorubicin)), a vinca alkaloid (e.g., vinblastine, vincristine, vindesine, vinorelbine), an alkylating agent (e.g., cy53acarbazineide, decarbazine, melphalan, ifosfamide, temozolomide), an immune cell antibody (e.g., alemtuzamab, gemtuzumab, rituximab, ofatumumab, tositumomab, brentuximab), an antimetabolite (including, e.g., folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors (e.g., fludarabine)), an mTOR inhibitor, a TNFR glucocorticoid induced TNFR related protein (GITR) agonist, a proteasome inhibitor (e.g., aclacinomycin A, gliotoxin or bortezomib), an immunomodulator such as thalidomide or a thalidomide derivative (e.g., lenalidomide).
  • General Chemotherapeutic agents considered for use in combination therapies include busulfan (Myleran®), busulfan injection (Busulfex®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), daunorubicin hydrochloride (Cerubidine®), daunorubicin citrate liposome injection (DaunoXome®), dexamethasone, doxorubicin hydrochloride (Adriamycin®, Rubex®), etoposide (Vepesid®), fludarabine phosphate (Fludara®), hydroxyurea (Hydrea®), Idarubicin (Idamycin®), mitoxantrone (Novantrone®), Gemtuzumab Ozogamicin (Mylotarg®).
  • In embodiments, general chemotherapeutic agents considered for use in combination therapies include anastrozole (Arimidex®), bicalutamide (Casodex®), bleomycin sulfate (Blenoxane®), busulfan (Myleran®), busulfan injection (Busulfex®), capecitabine (Xeloda®), N4-pentoxycarbonyl-5-deoxy-5-fluorocytidine, carboplatin (Paraplatin®), carmustine (BiCNU®), chlorambucil (Leukeran®), cisplatin (Platinol®), cladribine (Leustatin®), cyclophosphamide (Cytoxan® or Neosar®), cytarabine, cytosine arabinoside (Cytosar-U®), cytarabine liposome injection (DepoCyt®), dacarbazine (DTIC-Dome®), dactinomycin (Actinomycin D, Cosmegan), daunorubicin hydrochloride (Cerubidine®), daunorubicin citrate liposome injection (DaunoXome®), dexamethasone, docetaxel (Taxotere®), doxorubicin hydrochloride (Adriamycin®, Rubex®), etoposide (Vepesid®), fludarabine phosphate (Fludara®), 5-fluorouracil (Adrucil®, Efudex®), flutamide (Eulexin®), tezacitibine, Gemcitabine (difluorodeoxycitidine), hydroxyurea (Hydrea®), Idarubicin (Idamycin®), ifosfamide (IFEX®), irinotecan (Camptosar®), L-asparaginase (ELSPAR®), leucovorin calcium, melphalan (Alkeran®), 6-mercaptopurine (Purinethol®), methotrexate (Folex®), mitoxantrone (Novantrone®), mylotarg, paclitaxel (Taxol®), phoenix (Yttrium90/MX-DTPA), pentostatin, polifeprosan 20 with carmustine implant (Gliadel®), tamoxifen citrate (Nolvadex®), teniposide (Vumon®), 6-thioguanine, thiotepa, tirapazamine (Tirazone®), topotecan hydrochloride for injection (Hycamptin®), vinblastine (Velban®), vincristine (Oncovin®), and vinorelbine (Navelbine®).
  • Treatments can be evaluated, for example, by tumor regression, tumor weight or size shrinkage, time to progression, duration of survival, progression free survival, overall response rate, duration of response, quality of life, protein expression and/or activity. Approaches to determining efficacy of the therapy can be employed, including for example, measurement of response through radiological imaging.
  • TCR Incorporated Antigen Agnostic Receptors (TIAARs)
  • Table 2 provides exemplary, non-limiting examples of components of TCR incorporated antigen agnostic receptors (TIAARs) of the invention. Table 3 shows exemplary arrangements of the components.
  • TABLE 2
    TCR incorporated antigen agnostic receptor (TIAAR) components
    Code Signal peptide Tag ECD_TMD ICD Costim
    pIB1026 CD3D Myc CD3D N/A CD28-CD40
    pIB1027 CD3E FLAG CD3E N/A CD28-CD40
    pIB1028 CD3G Myc CD3G N/A CD28-CD40
    pIB1029 CD3Z Myc IC CD3Z N/A CD28-CD40
    pIB1030 CD8A Myc hTRDC N/A CD28-CD40
    pIB1031 CD8A FLAG hTRGC1 N/A CD28-CD40
    pIB1032 CD8A Myc mTRAC N/A CD28-CD40
    pIB1033 CD8A FLAG mTRBC1 N/A CD28-CD40
    pIB1046 CD8A ×2 Myc and hTRDC_hTRGC1 N/A CD28-CD40
    FLAG
    pIB1047 CD8A ×2 Myc and mTRAC_mTRBC1 N/A CD28-CD40
    FLAG
    pIB1048 CD3D and Myc and CD3D_CD3E N/A CD28-CD40
    CD3E FLAG
    pIB1049 CD3G and Myc and CD3G_CD3E N/A CD28-CD40
    CD3E FLAG
    pIB1050 CD3D and Myc and CD3D_CD3E CD3D_CD3E CD28-CD40
    CD3E FLAG
    pIB1051 CD3D and Myc and CD3D_CD3E CD3D_CD3E N/A
    CD3E FLAG
    pIB1052 CD3D and Myc and CD3D_CD3E N/A N/A
    CD3E FLAG
    pIB1053 CD3G and Myc and CD3G_CD3E CD3G_CD3E CD28-CD40
    CD3E FLAG
    pIB1054 CD3G and Myc and CD3G_CD3E CD3G_CD3E N/A
    CD3E FLAG
    pIB1055 CD3G and Myc and CD3G_CD3E N/A N/A
    CD3E FLAG
    pIB1056 CD3Z Myc IC CD3Z CD3Z CD28-CD40
    pIB1057 CD3Z Myc IC CD3Z CD3Z N/A
    pIB1058 CD3Z Myc IC CD3Z N/A N/A
    pIB1059 CD3Z Myc IC CD3Z CD3Z (×2) CD28-CD40
    pIB1060 CD3Z Myc IC CD3Z CD3Z (×2) N/A
    pIB1061 CD3Z Myc IC CD3Z CD3Z (×2) CD28-CD40
    (swaped)
    pIB1062 CD3Z Myc IC CD3Z CD3Z CD28-CD40
    (swaped)
    pIB1063 CD80 Myc CD80 CD80 N/A
    pIB1064 no signal Myc IC N/A Lck N/A
    peptide
    pIB1065 no signal Myc IC N/A Lck (Y505F) N/A
    peptide
    pIB1066 CD80 Myc CD80 Lck N/A
    pIB1067 CD80 Myc CD80 Lck CD28-CD40
    pIB1068 CD80 Myc CD80 CD80_Lck N/A
    (Y505F)
    pIB1069 CD80 Myc CD80 CD80_Lck CD28-CD40
    (Y505F)
    pIB1070 CD8A Myc LAT LAT N/A
    pIB1071 CD8A Myc LAT LAT CD28-CD40
    pIB1072 CD4 Myc CD4 CD4 CD28-CD40
    pIB1073 CD4 Myc CD4 CD4 CD28-CD40
    pIB1074 CD8A and Myc and CD8A and CD8A and N/A
    CD8B FLAG CD8B CD8B
    pIB1075 CD8A and Myc and CD8A and CD8A and CD28-CD40
    CD8B FLAG CD8B CD8B
  • TABLE 3
    TCR incorporated antigen agnostic receptors (TIAARs)
    Code Description
    pIB1026 CD3D_CD3D_CD28CD40
    pIB1027 CD3E_CD3E_CD28CD40
    pIB1028 CD3G_CD3G_CD28CD40
    pIB1029 CD3Z_CD3Z_CD28CD40_Myc
    pIB1030 CD8A_hTRDC_CD28CD40
    pIB1031 CD8A_hTRGC1_CD28CD40
    pIB1032 CD8A_mTRAC_CD28CD40
    pIB1033 CD8A_mTRBC1_CD28CD40
    pIB1046 CD8A_hTRDC_CD28CD40-T2A-CD8a_hTRGC1_CD28CD40
    pIB1047 CD8A_mTRAC_CD28CD40-T2A-CD8A_mTRBC1_CD28CD40
    pIB1048 CD3D_CD3D_CD28CD40-T2A-CD3E_CD3E_CD28CD40
    pIB1049 CD3G_CD3G_CD28CD40-T2A-CD3E_CD3E_CD28CD40
    pIB1050 CD3D_CD3D_CD3D (ICD)_CD28CD40-T2A-CD3E_CD3E_CD3E (ICD)_CD28CD40
    pIB1051 CD3D_CD3D_CD3D ICD -T2A-CD3E_CD3E_CD3E ICD
    pIB1052 CD3D_CD3D (control)-T2A-CD3E_CD3E (control)
    pIB1053 CD3G_CD3G_CD3G (ICD)_CD28CD40-T2A-CD3E_CD3E_CD3E (ICD)_CD28CD40
    pIB1054 CD3G_CD3G_CD3G ICD -T2A-CD3E_CD3E_CD3E ICD
    pIB1055 CD3G_CD3G (control)-T2A-CD3E_CD3E (control)
    pIB1056 CD3z_CD3z_CD3z ICD_CD28CD40_Myc
    pIB1057 CD3z_CD3z_CD3z ICD_Myc
    pIB1058 CD3z_CD3z (control)
    pIB1059 CD3Z_CD3Z_CD3Z ICD (duplicating CD3z endodomain-6 ITAMs)_CD28CD40_Myc
    pIB1060 CD3Z_CD3Z_CD3Z ICD (duplicating CD3z endodomain-6 ITAMs)_Myc
    pIB1061 CD3Z_CD3Z_CD28CD40_CD3Z (6 ITAMs)_Myc
    pIB1062 CD3Z_CD3Z_CD28CD40_CD3Z ICD_Myc
    pIB1063 CD80 (control)
    pIB1064 Lck (control)
    pIB1065 Lck (Y505F) (control)
    pIB1066 CD80_Lck
    pIB1067 CD80_Lck_CD28CD40
    pIB1068 CD80_Lck (Y505F)
    pIB1069 CD80_Lck (Y505F)_CD28CD40
    pIB1070 LAT (control)
    pIB1071 LAT_C28CD40
    pIB1072 CD4 control
    pIB1073 CD4_CD28_CD40
    pIB1074 CD8 control
    pIB1075 CD8_CD28_CD40
  • Constitutive Costimulatory Proteins
  • Table 4 provides exemplary, non-limiting examples of components of constitutive costimulatory proteins of the invention. Table 5 shows the exemplary arrangements of the components.
  • TABLE 4
    Constitutively stimulating antigen agnostic receptor (C-SAAR) components
    Code Signal peptide Tag ECD_TMD Costim
    pIB1076 CD8A Myc LZ (cFos)_EGFR CD28-CD40
    pIB1077 CD8A Myc LZ (cFos)_CD28 CD28-CD40
    pIB1078 CD8A Myc LZ (cJun)_EGFR CD28-CD40
    pIB1079 CD8A Myc LZ (cJun)_CD28 CD28-CD40
    pIB1080 CD8A Myc LZ (c/EBP)_EGFR CD28-CD40
    pIB1081 CD8A Myc LZ (c/EBP)_CD28 CD28-CD40
    pIB1103 GpA Myc GpA ECD_TMD CD28-CD40
    pIB1104 GpA Myc GpA TMD CD28-CD40
    pIB1105 EPOR Myc EPOR ECD_TMD CD28-CD40
    pIB1106 EPOR Myc EPOR TMD CD28-CD40
    pIB1107 TPOR Myc TPOR ECD_TMD CD28-CD40
    pIB1108 TPOR Myc TPOR TMD CD28-CD40
    pIB1109 TPOR Myc TPOR ECD_TMD (S505N) CD28-CD40
    pIB1110 TPOR Myc TPOR TMD (S505N) CD28-CD40
    pIB1111 TPOR Myc TPOR ECD_TMD (W515K) CD28-CD40
    pIB1112 TPOR Myc TPOR TMD (W515K) CD28-CD40
    pIB1113 TPOR Myc TPOR ECD_TMD (H499L) CD28-CD40
    pIB1114 TPOR Myc TPOR TMD (H499L) CD28-CD40
    pIB1115 TPOR Myc TPOR ECD_TMD (S505N-W515K) CD28-CD40
    pIB1116 TPOR Myc TPOR TMD (S505N-W515K) CD28-CD40
    pIB1117 TPOR Myc TPOR ECD_TMD (H499Y-S505N) CD28-CD40
    pIB1118 TPOR Myc TPOR TMD (H499Y-S505N) CD28-CD40
    pIB1119 TPOR Myc TPOR ECD_TMD (L498W-H499C) CD28-CD40
    pIB1120 TPOR Myc TPOR TMD (L498W-H499C) CD28-CD40
    pIB1025 CD8a Myc CD28 CD28-CD40
    pIB1179 CD8a N/A IgG1 + CD28TM CD28-CD40
    pIB1180 CD8a N/A IgG1mut + CD28TM CD28-CD40
    pIB1181 CD8a N/A IgG2 + CD28TM CD28-CD40
    pIB1182 CD8a N/A IgG3 + CD28TM CD28-CD40
    pIB1183 CD8a N/A IgG4 + CD28TM CD28-CD40
    pIB1184 CD8a N/A IgG4mut +CD28TM CD28-CD40
    pIB1185 CD8a N/A IgG1 + CD28 stalk/TM CD28-CD40
    pIB1186 CD8a N/A IgG1mut + CD28 stalk/TM CD28-CD40
    pIB1187 CD8a N/A IgG2 + CD28 stalk/TM CD28-CD40
    pIB1188 CD8a N/A IgG3 + CD28 stalk/TM CD28-CD40
    pIB1189 CD8a N/A IgG4 + CD28 stalk/TM CD28-CD40
    pIB1190 CD8a N/A IgG4mut + CD28 stalk/TM CD28-CD40
  • TABLE 5
    C-SAAR Proteins
    Code Description
    pIB1076 LZ (cFos)- EGFRTM/JMD- CD28-CD40
    pIB1077 LZ (cFos)- CD28TM- CD28-CD40
    pIB1078 LZ (cJun)- EGFRTM/JMD- CD28-CD40
    pIB1079 LZ (cJun)- CD28TM- CD28-CD40
    pIB1080 LZ (c/EBP)- EGFRTM/JMD- CD28-CD40
    pIB1081 LZ (c/EBP)- CD28TM- CD28-CD40
    pIB1103 GpA ECD-TMD-CD28-CD40
    pIB1104 GpA TMD-CD28-CD40
    pIB1105 EpoR ECD-TMD-CD28-CD40
    pIB1106 EpoR TMD-CD28-CD40
    pIB1107 TPO ECD- TPO (WT) TMD -CD28-CD40
    pIB1108 TPO (WT) TMD -CD28-CD40
    pIB1109 TPO ECD- TPO (S505N) TMD -CD28-CD40
    pIB1110 TPO (S505N) TMD -CD28-CD40
    pIB1111 TPO ECD-TPO (W515K) TMD -CD28-CD40
    pIB1112 TPO (W515K) TMD -CD28-CD40
    pIB1113 TPO ECD- TPO (H499L) TMD -CD28-CD40
    pIB1114 TPO (H499L) TMD -CD28-CD40
    pIB1115 TPO ECD- TPO (S505N-W515K) TMD -CD28-CD40
    pIB1116 TPO (S505N-W515K) TMD -CD28-CD40
    pIB1117 TPO ECD- TPO (H499Y-S505N) TMD -CD28-CD40
    pIB1118 TPO (H499Y-S505N) TMD -CD28-CD40
    pIB1119 TPO ECD- TPO (L498W-H499C) TMD -CD28-CD40
    pIB1120 TPO (L498W-H499C) TMD -CD28-CD40
    pIB1025 CD28 TM_CD28_CD40
    pIB1179 IICH2CH3)-CD28(TM)-CD28(CoStim)-CD40(CoStim)
    pIB1180 IgG1ICH3, mutant)-CD28(TM)-CD28(CoStim)-CD40(CoStim)
    pIB1182 IgG3(CH2CH3)-CD28(TM)-CD28(CoStim)-CD40(CoI)
    pIB1184 IgG4(CH2CH3, mutant)-CD28(TM)-CD28(CoStim)-CD40(CoStim)
    pIB1185 IgG1(CH2CH3)-CD28(Stalk + TM)-CD28(CoStim)-CD40(CoStim)
    pIB1186 IgG1(CH2CH3, mutant)-CD28(Stalk + TM)-CD28(CoStim)-CD40(CoStim)
    pIB1187 IgG2(CH2CH3)-CD28(Stalk + TM)-CD28(CoStim)-CD40(CoStim)
    pIB1188 IgG3(CH2CH3)-CD28(Stalk + TM)-CD28(CoStim)-CD40(CoStim)
    pIB1189 IgG4(CH2CH3)-CD28(Stalk + TM)-CD28(CoStim)-CD40(CoStim)
    pIB1190 IgG4(CH2CH3, mutant)-CD28(Stalk + TM)-CD28(CoStim)-CD40(CoStim)
  • Inducible Costimulatory Receptors
  • Table 6 provides exemplary, non-limiting examples of inducible costimulatory receptors of the invention. Table 7 shows exemplary arrangements of the components.
  • TABLE 6
    Inducible costimulatory protein components
    Code Signal peptide Tag ECD_TMD ICD Costim
    pIB1082 EGFR Myc EGFR N/A CD28-CD40
    pIB1083 EGFR Myc EGFR (domain IV) N/A CD28-CD40
    pIB1084 EGFR Myc EGFR (623-668) N/A CD28-CD40
    pIB1085 Her2 Myc Her2 N/A CD28-CD40
    pIB1086 Her2 Myc Her2 (V659E) N/A CD28-CD40
    pIB1087 Her2 Myc Her2 (V660D) N/A CD28-CD40
    pIB1088 Her2 Myc Her2 (V660R) N/A CD28-CD40
    pIB1089 Her2 Myc Her2 domain IV_TMD N/A CD28-CD40
    pIB1090 Her2 Myc Her2 domain IV_TMD (V659E) N/A CD28-CD40
    pIB1091 Her2 Myc Her2 domain IV_TMD (G660D) N/A CD28-CD40
    pIB1092 Her2 Myc Her2 domain IV_TMD (G660R) N/A CD28-CD40
    pIB1093 Her2 Myc Her2 TMD N/A CD28-CD40
    pIB1094 Her2 Myc Her2 TMD (V659E) N/A CD28-CD40
    pIB1095 Her2 Myc Her2 TMD (G660D) N/A CD28-CD40
    pIB1096 Her2 Myc Her2 TMD (G660R) N/A CD28-CD40
    pIB1097 CD8A Myc A30514 VH_VL N/A CD28-CD40
    pIB1098 CD8A Myc A30514 VL_VH N/A CD28-CD40
    pIB1099 CD8A Myc A30523 VH_VL N/A CD28-CD40
    pIB1100 CD8A Myc A30523 VL_VH N/A CD28-CD40
    pIB1101 CD8A Myc A30633 VH_VL N/A CD28-CD40
    pIB1102 CD8A Myc A30633 VL_VH N/A CD28-CD40
  • TABLE 7
    Inducible costimulatory proteins
    Code GOI description
    pIB1082 WT EGFR ECD- EGFRTM/JMD- CD28-CD40
    pIB1083 domain IV - EGFRTM/JMD- CD28-CD40
    pIB1084 EGFRTM/JMD- CD28-CD40 (control)
    pIB1085 Her 2 (Domain 1 to IV)-TMD/JMD-CD28-CD40
    pIB1086 Her 2 (Domain 1 to IV)-TMD (V659E)/JMD-CD28-CD40
    pIB1087 Her 2 (Domain 1 to IV)-TMD (G660D)/JMD-CD28-CD40
    pIB1088 Her 2 (Domain 1 to IV)-TMD (G660R)/JMD-CD28-CD40
    pIB1089 Her 2 (Domain IV)-TMD/JMD-CD28-CD40
    pIB1090 Her 2 (Domain IV)-TMD (V659E)/JMD-CD28-CD40
    pIB1091 Her 2 (Domain IV)-TMD (G660D)/JMD-CD28-CD40
    pIB1092 Her 2 (Domain IV)-TMD (G660R)/JMD-CD28-CD40
    pIB1093 Her 2 TMD/JMD-CD28-CD40
    pIB1094 Her 2 TMD (V659E)/JMD-CD28-CD40
    pIB1095 Her 2 TMD (G660D)/JMD-CD28-CD40
    pIB1096 Her 2 TMD (G660R)/JMD-CD28-CD40
    pIB1097 Anti-ID1 VH-VL (A30514-pembrolizumab)-
    CD28TMD CD28-CD40
    pIB1098 Anti-ID1 VL-VH (A30514-pembrolizumab)-
    CD28TMD CD28-CD40
    pIB1099 Anti-ID2 Vh-VL (A30523-pembrolizumab)-
    CD28TMD CD28-CD40
    pIB1100 Anti-ID2 VL-Vh (A30523-pembrolizumab)-
    CD28TMD CD28-CD40
    pIB1101 Anti-ID3 Vh-VL (A30633-pembrolizumab)-
    CD28TMD CD28-CD40
    pIB1102 Anti-ID3 VL-VH (A30633-pembrolizumab)-
    CD28TMD CD28-CD40
  • The following sequences in the below table include complete components and are non-limiting. Components may include a signal peptide (SP), a TCR clustering domain (CD) and/or a signaling domain (SD). It will be understood that whereas certain proteins may comprise N-terminal signal peptides when expressed, those signal peptides are cleaved and may be imprecisely cleaved when the proteins are expressed, and that the resulting proteins from which signal peptides are removed comprise binding domains having variation of up to about five amino acids in the location of the N-terminal amino acid.
  • TABLE 8
    TCR costimulation construct examples
    SEQ ID NO: 1
    Component: SP CD3D_
    Sequence: MEHSTFLSGL VLATLLSQVS P
    SEQ ID NO: 2
    Component: CD CD3D_
    Sequence: FKIPIEELED RVFVNCNTSI TWVEGTVGTL LSDITRLDLG KRILDPRGIY
    RCNGTDIYKD KESTVQVHYR MCQSCVELDP ATVAGIIVTD VIATLLLALG
    VFCFA
    SEQ ID NO: 3
    Component: SD CD28CD40
    Sequence: RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN
    KAPHPKQEPQ EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ
    ERQ
    SEQ ID NO: 4
    Full length: CD3D_CD3D_CD28CD40
    Sequence: MEHSTFLSGL VLATLLSQVS PFKIPIEELE DRVFVNCNTS ITWVEGTVGT
    LLSDITRLDL GKRILDPRGI YRCNGTDIYK DKESTVQVHY RMCQSCVELD
    PATVAGIIVT DVIATLLLAL GVFCFARSKR SRLLHSDYMN MTPRRPGPTR
    KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH PKQEPQEINF PDDLPGSNTA
    APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 5
    Component: SP CD3E_
    Sequence: MQSGTHWRVL GLCLLSVGVW GQ
    SEQ ID NO: 6
    Component: CD CD3E_
    Sequence: DGNEEMGGIT QTPYKVSISG TTVILTCPQY PGSEILWQHN DKNIGGDEDD
    KNIGSDEDHL SLKEFSELEQ SGYYVCYPRG SKPEDANFYL YLRARVCENC
    MEMDVMSVAT IVIVDICITG GLLLLVYYWS
    SEQ ID NO: 7
    Component: SD CD28CD40
    Sequence: RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN
    KAPHPKQEPQ EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ
    ERQ
    SEQ ID NO: 8
    Full length: CD3E_CD3E_CD28CD40
    Sequence: MQSGTHWRVL GLCLLSVGVW GQDGNEEMGG ITQTPYKVSI SGTTVILTCPQ
    YPGSEILWQH NDKNIGGDED DKNIGSDEDH LSLKEFSELE QSGYYVCYPRG
    SKPEDANFYL YLRARVCENC MEMDVMSVAT IVIVDICITG GLLLLVYYWSR
    SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS KKVAKKPTNKA
    PHPKQEPQEI NFPDDLPGSN TAAPVQETLH GCQPVTQEDG KESRISVQERQ
    SEQ ID NO: 9
    Component: SP CD3G_
    Sequence: MEQGKGLAVL ILAIILLQGT LA
    SEQ ID NO: 10
    Component: CD CD3G_
    Sequence: QSIKGNHLVK VYDYQEDGSV LLTCDAEAKN ITWFKDGKMI GFLTEDKKKW
    NLGSNAKDPR GMYQCKGSQN KSKPLQVYYR MCQNCIELNA ATISGFLFAE
    IVSIFVLAVG VYFIA
    SEQ ID NO: 11
    Component: SD CD28CD40
    Sequence: RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN
    KAPHPKQEPQ EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ
    ERQ
    SEQ ID NO: 12
    Full length: CD3G_CD3G_CD28CD40
    Sequence: MEQGKGLAVL ILAIILLQGT LAQSIKGNHL VKVYDYQEDG SVLLTCDAEA
    KNITWFKDGK MIGFLTEDKK KWNLGSNAKD PRGMYQCKGS QNKSKPLQVY
    YRMCQNCIEL NAATISGFLF AEIVSIFVLA VGVYFIARSK RSRLLHSDYM
    NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN
    FPDDLPGSNT AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 13
    Component: SP CD3Z_
    Sequence: MKWKALFTAA ILQAQLPITE A
    SEQ ID NO: 14
    Component: CD CD3Z_
    Sequence: QSFGLLDPKL CYLLDGILFI YGVILTALFL
    SEQ ID NO: 15
    Component: SD CD28CD40
    Sequence: RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN
    KAPHPKQEPQ EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ
    ERQ
    SEQ ID NO: 16
    Full length: CD3Z_CD3Z_CD28CD40
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LRSKRSRLLH SDYMNMTPRR PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT
    NKAPHPKQEP QEINFPDDLP GSNTAAPVQE TLHGCQPVTQ EDGKESRISV
    QERQ
    SEQ ID NO: 17
    Component: SP CD8A_
    Sequence: MALPVTALLL PLALLLHAAR P
    SEQ ID NO: 18
    Component: CD TRDC_
    Sequence: SQPHTKPSVF VMKNGTNVAC LVKEFYPKDI RINLVSSKKI TEFDPAIVIS
    PSGKYNAVKL GKYEDSNSVT CSVQHDNKTV HSTDFEVKTD STDHVKPKET
    ENTKQPSKSC HKPKAIVHTE KVNMMSLTVL GLRMLFAKTV AVNFLLTAKL
    FFL
    SEQ ID NO: 19
    Component: SD CD28CD40
    Sequence: RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN
    KAPHPKQEPQ EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ
    ERQ
    SEQ ID NO: 20
    Full length: CD8A_TRDC_CD28CD40
    Sequence: MALPVTALLL PLALLLHAAR PSQPHTKPSV FVMKNGTNVA CLVKEFYPKD
    IRINLVSSKK ITEFDPAIVI SPSGKYNAVK LGKYEDSNSV TCSVQHDNKT
    VHSTDFEVKT DSTDHVKPKE TENTKQPSKS CHKPKAIVHT EKVNMMSLTV
    LGLRMLFAKT VAVNFLLTAK LFFLRSKRSR LLHSDYMNMT PRRPGPTRKH
    YQPYAPPRDF AAYRSKKVAK KPTNKAPHPK QEPQEINFPD DLPGSNTAAP
    VQETLHGCQP VTQEDGKESR ISVQERQ
    SEQ ID NO: 21
    Component: SP CD8A_
    Sequence: MALPVTALLL PLALLLHAAR P
    SEQ ID NO: 22
    Component: CD TRGC1_
    Sequence: DKQLDADVSP KPTIFLPSIA ETKLQKAGTY LCLLEKFFPD VIKIHWQEKK
    SNTILGSQEG NTMKTNDTYM KFSWLTVPEK SLDKEHRCIV RHENNKNGVD
    QEIIFPPIKT DVITMDPKDN CSKDANDTLL LQLTNTSAYY MYLLLLLKSV
    VYFAIITCCL L
    SEQ ID NO: 23
    Component: SD CD28CD40
    Sequence: RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN
    KAPHPKQEPQ EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ
    ERQ
    SEQ ID NO: 24
    Full length: CD8A_TRGC1_CD28CD40
    Sequence: MALPVTALLL PLALLLHAAR PDKQLDADVS PKPTIFLPSI AETKLQKAGT
    YLCLLEKFFP DVIKIHWQEK KSNTILGSQE GNTMKTNDTY MKFSWLTVPE
    KSLDKEHRCI VRHENNKNGV DQEIIFPPIK TDVITMDPKD NCSKDANDTL
    LLQLTNTSAY YMYLLLLLKS VVYFAIITCC LLRSKRSRLL HSDYMNMTPR
    RPGPTRKHYQ PYAPPRDFAA YRSKKVAKKP TNKAPHPKQE PQEINFPDDL
    PGSNTAAPVQ ETLHGCQPVT QEDGKESRIS VQERQ
    SEQ ID NO: 25
    Component: SP CD8A_
    Sequence: MALPVTALLL PLALLLHAAR P
    SEQ ID NO: 26
    Component: CD TRAC_
    Sequence: IQNPEPAVYQ LKDPRSQDST LCLFTDFDSQ INVPKTMESG TFITDKCVLD
    MKAMDSKSNG AIAWSNQTSF TCQDIFKETN ATYPSSDVPC DATLTEKSFE
    TDMNLNFQNL LVIVLRILLL KVAGFNLLMT LRLWSS
    SEQ ID NO: 27
    Component: SD CD28CD40
    Sequence: RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN
    KAPH PKQEP QEINFPDDLP GSNTAAPVQE TLHGCQPVTQ EDGKESRISV
    QERQ
    SEQ ID NO: 28
    Full length: CD8A_TRAC_CD28CD40
    Sequence: MALPVTALLL PLALLLHAAR PIQNPEPAVY QLKDPRSQDS TLCLFTDFDS
    QINVPKTMES GTFITDKCVL DMKAMDSKSN GAIAWSNQTS FTCQDIFKET
    NATYPSSDVP CDATLTEKSF ETDMNLNFQN LLVIVLRILL LKVAGFNLLM
    TLRLWSSRSK RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK
    VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT AAPVQETLHG CQPVTQEDGK
    ESRISVQERQ
    SEQ ID NO: 29
    Component: SP CD8A_
    Sequence: MALPVTALLL PLALLLHAAR P
    SEQ ID NO: 30
    Component: CD TRBC1_
    Sequence: VLTPPKVSLF EPSKAEIANK QKATLVCLAR GFFPDHVELS WWVNGKEVHS
    GVCTDPQAYK ESNYSYCLSS RLRVSATFWH NPRNHFRCQV QFHGLSEEDK
    WPEGSPKPVT QNISAEAWGR ADCGITSASY QQGVLSATIL YEILLGKATL
    YAVLVSTLVV MAMVKRKNS
    SEQ ID NO: 31
    Component: SD CD28CD40
    Sequence: RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN
    KAPHPKQEPQ EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ
    ERQ
    SEQ ID NO: 32
    Full length: CD8A_TRBC1_CD28CD40
    Sequence: MALPVTALLL PLALLLHAAR PVLTPPKVSL FEPSKAEIAN KQKATLVCLA
    RGFFPDHVEL SWWVNGKEVH SGVCTDPQAY KESNYSYCLS SRLRVSATFW
    HNPRNHFRCQ VQFHGLSEED KWPEGSPKPV TQNISAEAWG RADCGITSAS
    YQQGVLSATI LYEILLGKAT LYAVLVSTLV VMAMVKRKNS RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 33
    vector clone: pIB1001
    Sequence MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLVQSGA EVKKPGASVK
    VSCKASGYTF TSYWMNWVRQ APGQGLEWMG RIDPYDSETH
    YAQKLQGRVT
    MTTDTSTSTA YMELRSLRSD DTAVYYCARG GYDFDVGTLY WFFDVWGQGT
    TVTVSSGGGG SGGGGSGGGG SDIQMTQSPS SLSASVGDRV TITCRASENI
    YSYLAWYQQK PGKAPKLLIY NAKTLAEGVP SRFSGSGSGT DFTLTISSLQ
    PEDFATYYCQ HHYGTPRTFG GGTKVEIKAA AGSGGSGILV KQSPMLVAYD
    NAVNLSCKYS YNLFSREFRA SLHKGLDSAV EVCVVYGNYS QQLQVYSKTG
    FNCDGKLGNE SVTFYLQNLY VNQTDIYFCK IEVMYPPPYL DNEKSNGTII
    HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR
    SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH
    PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 34
    vector clone: pIB1002
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LDIQMTQSPS SLSASVGDRV
    TITCRASENI YSYLAWYQQK PGKAPKLLIY NAKTLAEGVP SRFSGSGSGT
    DFTLTISSLQ PEDFATYYCQ HHYGTPRTFG GGTKVEIKGG GGSGGGGSGG
    GGSQVQLVQS GAEVKKPGAS VKVSCKASGY TFTSYWMNWV
    RQAPGQGLEW
    MGRIDPYDSE THYAQKLQGR VTMTTDTSTS TAYMELRSLR SDDTAVYYCA
    RGGYDFDVGT LYWFFDVWGQ GTTVTVSSAA AGSGGSGILV KQSPMLVAYD
    NAVNLSCKYS YNLFSREFRA SLHKGLDSAV EVCVVYGNYS QQLQVYSKTG
    FNCDGKLGNE SVTFYLQNLY VNQTDIYFCK IEVMYPPPYL DNEKSNGTII
    HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR
    SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH
    PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 35
    vector clone: pIB1003
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLVESGG GVVQPGRSLR
    LSCAASGFTF SSYDMHWVRQ APGKGLEWVA VIWYDGSNKY YADSVKGRFT
    ISRDNSKNTL YLQMNSLRAE DTAVYYCARG SGNWGFFDYW GQGTLVTVSS
    GGGGSGGGGS GGGGSDIQMT QSPSSLSASV GDRVTITCRA SQGISRWLAW
    YQQKPEKAPK SLIYAASSLQ SGVPSRFSGS GSGTDFTLTI SSLQPEDFAT
    YYCQQYNTYP RTFGQGTKVE IKAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 36
    vector clone: pIB1004
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LDIQMTQSPS SLSASVGDRV
    TITCRASQGI SRWLAWYQQK PEKAPKSLIY AASSLQSGVP SRFSGSGSGT
    DFTLTISSLQ PEDFATYYCQ QYNTYPRTFG QGTKVEIKGG GGSGGGGSGG
    GGSQVQLVES GGGVVQPGRS LRLSCAASGF TFSSYDMHWV RQAPGKGLEW
    VAVIWYDGSN KYYADSVKGR FTISRDNSKN TLYLQMNSLR AEDTAVYYCA
    RGSGNWGFFD YWGQGTLVTV SSAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 37
    vector clone: pIB1005
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLQQWGA GLLKPSETLS
    LTCAVYGGSF SGYYWSWIRQ SPEGLEWIGE INHGGYVTYN PSLESRVTIS
    VDTSKNQFSL KLSSVTAADT AVYYCARDYG PGNYDWYFDL WGRGTLVTVS
    SGGGGSGGGG SGGGGSEIVL TQSPATLSLS PGERATLSCR ASQSVSSYLA
    WYQQKPGQAP RLLIYDASNR ATGIPARFSG SGSGTDFTLT ISSLEPEDFA
    VYYCQQRSNW PPALTFGGGT KVEIKRAAAG SGGSGILVKQ SPMLVAYDNA
    VNLSCKYSYN LFSREFRASL HKGLDSAVEV CVVYGNYSQQ LQVYSKTGFN
    CDGKLGNESV TFYLQNLYVN QTDIYFCKIE VMYPPPYLDN EKSNGTIIHV
    KGKHLCPSPL FPGPSKPFWV LVVVGGVLAC YSLLVTVAFI IFWVRSKRSR
    LLHSDYMNMT PRRPGPTRKH YQPYAPPRDF AAYRSKKVAK KPTNKAPHPK
    QEPQEINFPD DLPGSNTAAP VQETLHGCQP VTQEDGKESR ISVQERQ
    SEQ ID NO: 38
    vector clone : pIB1006
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LEIVLTQSPA TLSLSPGERA
    TLSCRASQSV SSYLAWYQQK PGQAPRLLIY DASNRATGIP ARFSGSGSGT
    DFTLTISSLE PEDFAVYYCQ QRSNWPPALT FGGGTKVEIK RGGGGSGGGG
    SGGGGSQVQL QQWGAGLLKP SETLSLTCAV YGGSFSGYYW SWIRQSPEGL
    EWIGEINHGG YVTYNPSLES RVTISVDTSK NQFSLKLSSV TAADTAVYYC
    ARDYGPGNYD WYFDLWGRGT LVTVSSAAAG SGGSGILVKQ SPMLVAYDNA
    VNLSCKYSYN LFSREFRASL HKGLDSAVEV CVVYGNYSQQ LQVYSKTGFN
    CDGKLGNESV TFYLQNLYVN QTDIYFCKIE VMYPPPYLDN EKSNGTIIHV
    KGKHLCPSPL FPGPSKPFWV LVVVGGVLAC YSLLVTVAFI IFWVRSKRSR
    LLHSDYMNMT PRRPGPTRKH YQPYAPPRDF AAYRSKKVAK KPTNKAPHPK
    QEPQEINFPD DLPGSNTAAP VQETLHGCQP VTQEDGKESR ISVQERQ
    SEQ ID NO: 39
    vector clone: pIB1007
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVTLRESGP ALVKPTQTLT
    LTCTFSGFSL STSGMGVGWI RQPPGKALEW LAHIWWDDDK YYNPSLKSRL
    TISKDTSKNQ VVLTMTNMDP VDTATYYCAR TRRYFPFAYW GQGTLVTVSS
    GGGGSGGGGS GGGGSEIVMT QSPATLSVSP GERATLSCKA SQNVGTNVAW
    YQQKPGQAPR LLIYSASYRY SGIPARFSGS GSGTEFTLTI SSLQSEDFAV
    YYCQQYNTDP LTFGGGTKVE IKAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 40
    vector clone: pIB1008
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LEIVMTQSPA TLSVSPGERA
    TLSCKASQNV GTNVAWYQQK PGQAPRLLIY SASYRYSGIP ARFSGSGSGT
    EFTLTISSLQ SEDFAVYYCQ QYNTDPLTFG GGTKVEIKGG GGSGGGGSGG
    GGSQVTLRES GPALVKPTQT LTLTCTFSGF SLSTSGMGVG WIRQPPGKAL
    EWLAHIWWDD DKYYNPSLKS RLTISKDTSK NQVVLTMTNM DPVDTATYYC
    ARTRRYFPFA YWGQGTLVTV SSAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 41
    vector clone: pIB1009
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLVQSGV EVKKPGASVK
    VSCKASGYTF TNYYMYWVRQ APGQGLEWMG GINPSNGGTN FNEKFKNRVT
    LTTDSSTTTA YMELKSLQFD DTAVYYCARR DYRFDMGFDY WGQGTTVTVS
    SGGGGSGGGG SGGGGSEIVL TQSPATLSLS PGERATLSCR ASKGVSTSGY
    SYLHWYQQKP GQAPRLLIYL ASYLESGVPA RFSGSGSGTD FTLTISSLEP
    EDFAVYYCQH SRDLPLTFGG GTKVEIKRAA AGSGGSGILV KQSPMLVAYD
    NAVNLSCKYS YNLFSREFRA SLHKGLDSAV EVCVVYGNYS QQLQVYSKTG
    FNCDGKLGNE SVTFYLQNLY VNQTDIYFCK IEVMYPPPYL DNEKSNGTII
    HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR
    SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH
    PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 42
    vector clone: pIB1010
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LEIVLTQSPA TLSLSPGERA
    TLSCRASKGV STSGYSYLHW YQQKPGQAPR LLIYLASYLE SGVPARFSGS
    GSGTDFTLTI SSLEPEDFAV YYCQHSRDLP LTFGGGTKVE IKRGGGGSGG
    GGSGGGGSQV QLVQSGVEVK KPGASVKVSC KASGYTFTNY
    YMYWVRQAPG
    QGLEWMGGIN PSNGGTNFNE KFKNRVTLTT DSSTTTAYME LKSLQFDDTA
    VYYCARRDYR FDMGFDYWGQ GTTVTVSSAA AGSGGSGILV KQSPMLVAYD
    NAVNLSCKYS YNLFSREFRA SLHKGLDSAV EVCVVYGNYS QQLQVYSKTG
    FNCDGKLGNE SVTFYLQNLY VNQTDIYFCK IEVMYPPPYL DNEKSNGTII
    HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR
    SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH
    PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 43
    vector clone: pIB1011
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LEVQLVESGG GLVQPGGSLR
    LSCAASGFTF SDSWIHWVRQ APGKGLEWVA WISPYGGSTY YADSVKGRFT
    ISADTSKNTA YLQMNSLRAE DTAVYYCARR HWPGGFDYWG QGTLVTVSSG
    GGGSGGGGSG GGGSDIQMTQ SPSSLSASVG DRVTITCRAS QDVSTAVAWY
    QQKPGKAPKL LIYSASFLYS GVPSRFSGSG SGTDFTLTIS SLQPEDFATY
    YCQQYLYHPA TFGQGTKVEI KRAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 44
    vector clone: pIB1012
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LDIQMTQSPS SLSASVGDRV
    TITCRASQDV STAVAWYQQK PGKAPKLLIY SASFLYSGVP SRFSGSGSGT
    DFTLTISSLQ PEDFATYYCQ QYLYHPATFG QGTKVEIKRG GGGSGGGGSG
    GGGSEVQLVE SGGGLVQPGG SLRLSCAASG FTFSDSWIHW VRQAPGKGLE
    WVAWISPYGG STYYADSVKG RFTISADTSK NTAYLQMNSL RAEDTAVYYC
    ARRHWPGGFD YWGQGTLVTV SSAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 45
    vector clone: pIB1013
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLVQSGA EVKKPGASVK
    VSCKASGYTF TNYWIGWVKQ APGQGLEWIG YLYPGGLYTN YNEKFKGKAT
    MTADTSTNTA YMELSSLRSE DTAVYYCARY RDYDYAMDYW GQGTLVTVSS
    GGGGSGGGGS GGGGSDVVMT QTPLSLPVTL GQPASISCKS TKSLLNSDGF
    TYLGWCLQKP GQSPQLLIYL VSNRFSGVPD RFSGSGSGTD FTLKISRVEA
    EDVGVYYCFQ SNYLPLTFGQ GTKLEIKRAA AGSGGSGILV KQSPMLVAYD
    NAVNLSCKYS YNLFSREFRA SLHKGLDSAV EVCVVYGNYS QQLQVYSKTG
    FNCDGKLGNE SVTFYLQNLY VNQTDIYFCK IEVMYPPPYL DNEKSNGTII
    HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR
    SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH
    PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 46
    vector clone: pIB1014
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LDVVMTQTPL SLPVTLGQPA
    SISCKSTKSL LNSDGFTYLG WCLQKPGQSP QLLIYLVSNR FSGVPDRFSG
    SGSGTDFTLK ISRVEAEDVG VYYCFQSNYL PLTFGQGTKL EIKRGGGGSG
    GGGSGGGGSQ VQLVQSGAEV KKPGASVKVS CKASGYTFTN YWIGWVKQAP
    GQGLEWIGYL YPGGLYTNYN EKFKGKATMT ADTSTNTAYM ELSSLRSEDT
    AVYYCARYRD YDYAMDYWGQ GTLVTVSSAA AGSGGSGILV
    KQSPMLVAYD
    NAVNLSCKYS YNLFSREFRA SLHKGLDSAV EVCVVYGNYS QQLQVYSKTG
    FNCDGKLGNE SVTFYLQNLY VNQTDIYFCK IEVMYPPPYL DNEKSNGTII
    HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR
    SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH
    PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 47
    vector clone: pIB1015
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLQESGP GLVKPSQTLS
    LTCAVYGGSF SSGYWNWIRK HPGKGLEYIG YISYNGITYH NPSLKSRITI
    NRDTSKNQYS LQLNSVTPED TAVYYCARYK YDYDGGHAMD
    YWGQGTLVTV
    SSGGGGSGGG GSGGGGSDIQ MTQSPSSLSA SVGDRVTITC RASQDISNYL
    NWYQQKPGKA PKLLIYYTSK LHSGVPSRFS GSGSGTDYTL TISSLQPEDF
    ATYYCQQGSA LPWTFGQGTK VEIKAAAGSG GSGILVKQSP MLVAYDNAVN
    LSCKYSYNLF SREFRASLHK GLDSAVEVCV VYGNYSQQLQ VYSKTGFNCD
    GKLGNESVTF YLQNLYVNQT DIYFCKIEVM YPPPYLDNEK SNGTIIHVKG
    KHLCPSPLFP GPSKPFWVLV VVGGVLACYS LLVTVAFIIF WVRSKRSRLL
    HSDYMNMTPR RPGPTRKHYQ PYAPPRDFAA YRSKKVAKKP TNKAPHPKQE
    PQEINFPDDL PGSNTAAPVQ ETLHGCQPVT QEDGKESRIS VQERQ
    SEQ ID NO: 48
    vector clone: pIB1016
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LDIQMTQSPS SLSASVGDRV
    TITCRASQDI SNYLNWYQQK PGKAPKLLIY YTSKLHSGVP SRFSGSGSGT
    DYTLTISSLQ PEDFATYYCQ QGSALPWTFG QGTKVEIKGG GGSGGGGSGG
    GGSQVQLQES GPGLVKPSQT LSLTCAVYGG SFSSGYWNWI RKHPGKGLEY
    IGYISYNGIT YHNPSLKSRI TINRDTSKNQ YSLQLNSVTP EDTAVYYCAR
    YKYDYDGGHA MDYWGQGTLV TVSSAAAGSG GSGILVKQSP
    MLVAYDNAVN
    LSCKYSYNLF SREFRASLHK GLDSAVEVCV VYGNYSQQLQ VYSKTGFNCD
    GKLGNESVTF YLQNLYVNQT DIYFCKIEVM YPPPYLDNEK SNGTIIHVKG
    KHLCPSPLFP GPSKPFWVLV VVGGVLACYS LLVTVAFIIF WVRSKRSRLL
    HSDYMNMTPR RPGPTRKHYQ PYAPPRDFAA YRSKKVAKKP TNKAPHPKQE
    PQEINFPDDL PGSNTAAPVQ ETLHGCQPVT QEDGKESRIS VQERQ
    SEQ ID NO: 49
    vector clone: pIB1017
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLVESGG GVVQPGRSLR
    LSCAASGFTF SSYTMHWVRQ APGKGLEWVT FISYDGNNKY YADSVKGRFT
    ISRDNSKNTL YLQMNSLRAE DTAIYYCART GWLGPFDYWG QGTLVTVSSG
    GGGSGGGGSG GGGSEIVLTQ SPGTLSLSPG ERATLSCRAS QSVGSSYLAW
    YQQKPGQAPR LLIYGAFSRA TGIPDRFSGS GSGTDFTLTI SRLEPEDFAV
    YYCQQYGSSP WTFGQGTKVE IKAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 50
    vector clone: pIB1018
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LEIVLTQSPG TLSLSPGERA
    TLSCRASQSV GSSYLAWYQQ KPGQAPRLLI YGAFSRATGI PDRFSGSGSG
    TDFTLTISRL EPEDFAVYYC QQYGSSPWTF GQGTKVEIKG GGGSGGGGSG
    GGGSQVQLVE SGGGVVQPGR SLRLSCAASG FTFSSYTMHW VRQAPGKGLE
    WVTFISYDGN NKYYADSVKG RFTISRDNSK NTLYLQMNSL RAEDTAIYYC
    ARTGWLGPFD YWGQGTLVTV SSAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 51
    vector clone: pIB1019
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LEVQLVESGG GVVRPGGSLR
    LSCVASGVTF DDYGMSWVRQ APGKGLEWVS GINWNGGDTD YSDSVKGRFT
    ISRDNAKNSL YLQMNSLRAE DTALYYCARD FYGSGSYYHV PFDYWGQGIL
    VTVSSGGGGS GGGGSGGGGS EIVLTQSPGT LSLSPGERAT LSCRASQSVS
    RSYLAWYQQK RGQAPRLLIY GASSRATGIP DRFSGDGSGT DFTLSISRLE
    PEDFAVYYCH QYDMSPFTFG PGTKVDIKAA AGSGGSGILV KQSPMLVAYD
    NAVNLSCKYS YNLFSREFRA SLHKGLDSAV EVCVVYGNYS QQLQVYSKTG
    FNCDGKLGNE SVTFYLQNLY VNQTDIYFCK IEVMYPPPYL DNEKSNGTII
    HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR
    SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH
    PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 52
    vector clone: pIB1020
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LEIVLTQSPG TLSLSPGERA
    TLSCRASQSV SRSYLAWYQQ KRGQAPRLLI YGASSRATGI PDRFSGDGSG
    TDFTLSISRL EPEDFAVYYC HQYDMSPFTF GPGTKVDIKG GGGSGGGGSG
    GGGSEVQLVE SGGGVVRPGG SLRLSCVASG VTFDDYGMSW VRQAPGKGLE
    WVSGINWNGG DTDYSDSVKG RFTISRDNAK NSLYLQMNSL RAEDTALYYC
    ARDFYGSGSY YHVPFDYWGQ GILVTVSSAA AGSGGSGILV KQSPMLVAYD
    NAVNLSCKYS YNLFSREFRA SLHKGLDSAV EVCVVYGNYS QQLQVYSKTG
    FNCDGKLGNE SVTFYLQNLY VNQTDIYFCK IEVMYPPPYL DNEKSNGTII
    HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR
    SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH
    PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 53
    vector clone: pIB1021
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLVESGG GVVQPGRSLR
    LSCAASGFSF SSTYVCWVRQ APGKGLEWIA CIYTGDGTNY SASWAKGRFT
    ISKDSSKNTV YLQMNSLRAE DTAVYFCARP DITYGFAINF WGPGTLVTVS
    SGGGGSGGGG SGGGGSDIQM TQSPSSLSAS VGDRVTIKCQ ASQSISSRLA
    WYQQKPGKPP KLLIYRASTL ASGVPSRFSG SGSGTDFTLT ISSLQPEDVA
    TYYCQCTGYG ISWPIGGGTK VEIKAAAGSG GSGILVKQSP MLVAYDNAVN
    LSCKYSYNLF SREFRASLHK GLDSAVEVCV VYGNYSQQLQ VYSKTGFNCD
    GKLGNESVTF YLQNLYVNQT DIYFCKIEVM YPPPYLDNEK SNGTIIHVKG
    KHLCPSPLFP GPSKPFWVLV VVGGVLACYS LLVTVAFIIF WVRSKRSRLL
    HSDYMNMTPR RPGPTRKHYQ PYAPPRDFAA YRSKKVAKKP TNKAPHPKQE
    PQEINFPDDL PGSNTAAPVQ ETLHGCQPVT QEDGKESRIS VQERQ
    SEQ ID NO: 54
    vector clone: pIB1022
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LDIQMTQSPS SLSASVGDRV
    TIKCQASQSI SSRLAWYQQK PGKPPKLLIY RASTLASGVP SRFSGSGSGT
    DFTLTISSLQ PEDVATYYCQ CTGYGISWPI GGGTKVEIKG GGGSGGGGSG
    GGGSQVQLVE SGGGVVQPGR SLRLSCAASG FSFSSTYVCW VRQAPGKGLE
    WIACIYTGDG TNYSASWAKG RFTISKDSSK NTVYLQMNSL RAEDTAVYFC
    ARPDITYGFA INFWGPGTLV TVSSAAAGSG GSGILVKQSP MLVAYDNAVN
    LSCKYSYNLF SREFRASLHK GLDSAVEVCV VYGNYSQQLQ VYSKTGFNCD
    GKLGNESVTF YLQNLYVNQT DIYFCKIEVM YPPPYLDNEK SNGTIIHVKG
    KHLCPSPLFP GPSKPFWVLV VVGGVLACYS LLVTVAFIIF WVRSKRSRLL
    HSDYMNMTPR RPGPTRKHYQ PYAPPRDFAA YRSKKVAKKP TNKAPHPKQE
    PQEINFPDDL PGSNTAAPVQ ETLHGCQPVT QEDGKESRIS VQERQ
    SEQ ID NO: 55
    vector clone: pIB1023
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQVQLVQSGA EVKKPGASVK
    VSCKASGYTF TGYYMHWVRQ APGQGLEWMG WINPDSGGTN
    YAQKFQGRVT
    MTRDTSISTA YMELNRLRSD DTAVYYCARD QPLGYCTNGV CSYFDYWGQG
    TLVTVSSGGG GSGGGGSGGG GSDIQMTQSP SSVSASVGDR VTITCRASQG
    IYSWLAWYQQ KPGKAPNLLI YTASTLQSGV PSRFSGSGSG TDFTLTISSL
    QPEDFATYYC QQANIFPLTF GGGTKVEIKA AAGSGGSGIL VKQSPMLVAY
    DNAVNLSCKY SYNLFSREFR ASLHKGLDSA VEVCVVYGNY SQQLQVYSKT
    GFNCDGKLGN ESVTFYLQNL YVNQTDIYFC KIEVMYPPPY LDNEKSNGTI
    IHVKGKHLCP SPLFPGPSKP FWVLVVVGGV LACYSLLVTV AFIIFWVRSK
    RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP
    HPKQEPQEIN FPDDLPGSNT AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 56
    vector clone: pIB1024
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LDIQMTQSPS SVSASVGDRV
    TITCRASQGI YSWLAWYQQK PGKAPNLLIY TASTLQSGVP SRFSGSGSGT
    DFTLTISSLQ PEDFATYYCQ QANIFPLTFG GGTKVEIKGG GGSGGGGSGG
    GGSQVQLVQS GAEVKKPGAS VKVSCKASGY TFTGYYMHWV
    RQAPGQGLEW
    MGWINPDSGG TNYAQKFQGR VTMTRDTSIS TAYMELNRLR SDDTAVYYCA
    RDQPLGYCTN GVCSYFDYWG QGTLVTVSSA AAGSGGSGIL VKQSPMLVAY
    DNAVNLSCKY SYNLFSREFR ASLHKGLDSA VEVCVVYGNY SQQLQVYSKT
    GFNCDGKLGN ESVTFYLQNL YVNQTDIYFC KIEVMYPPPY LDNEKSNGTI
    IHVKGKHLCP SPLFPGPSKP FWVLVVVGGV LACYSLLVTV AFIIFWVRSK
    RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP
    HPKQEPQEIN FPDDLPGSNT AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 57
    vector clone: pIB1025
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 58
    vector clone: pIB1026
    Sequence: MEHSTFLSGL VLATLLSQVS PEQKLISEED LFKIPIEELE DRVFVNCNTS
    ITWVEGTVGT LLSDITRLDL GKRILDPRGI YRCNGTDIYK DKESTVQVHY
    RMCQSCVELD PATVAGIIVT DVIATLLLAL GVFCFARSKR SRLLHSDYMN
    MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH PKQEPQEINF
    PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 59
    vector clone: pIB1027
    Sequence: MQSGTHWRVL GLCLLSVGVW GQDYKDDDDK DGNEEMGGIT QTPYKVSISG
    TTVILTCPQY PGSEILWQHN DKNIGGDEDD KNIGSDEDHL SLKEFSELEQ
    SGYYVCYPRG SKPEDANFYL YLRARVCENC MEMDVMSVAT IVIVDICITG
    GLLLLVYYWS RSKRSRLLHS DYMNMTPRRP GPTRKHYQPY APPRDFAAYR
    SKKVAKKPTN KAPHPKQEPQ EINFPDDLPG SNTAAPVQET LHGCQPVTQE
    DGKESRISVQ ERQ
    SEQ ID NO: 60
    vector clone: pIB1028
    Sequence: MEQGKGLAVL ILAIILLQGT LAEQKLISEE DLQSIKGNHL VKVYDYQEDG
    SVLLTCDAEA KNITWFKDGK MIGFLTEDKK KWNLGSNAKD PRGMYQCKGS
    QNKSKPLQVY YRMCQNCIEL NAATISGFLF AEIVSIFVLA VGVYFIARSK
    RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP
    HPKQEPQEIN FPDDLPGSNT AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 61
    vector clone: pIB1029
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LRSKRSRLLH SDYMNMTPRR PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT
    NKAPHPKQEP QEINFPDDLP GSNTAAPVQE TLHGCQPVTQ EDGKESRISV
    QERQEQKLIS EEDL
    SEQ ID NO: 62
    vector clone: pIB1030
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LSQPHTKPSV FVMKNGTNVA
    CLVKEFYPKD IRINLVSSKK ITEFDPAIVI SPSGKYNAVK LGKYEDSNSV
    TCSVQHDNKT VHSTDFEVKT DSTDHVKPKE TENTKQPSKS CHKPKAIVHT
    EKVNMMSLTV LGLRMLFAKT VAVNFLLTAK LFFLRSKRSR LLHSDYMNMT
    PRRPGPTRKH YQPYAPPRDF AAYRSKKVAK KPTNKAPHPK QEPQEINFPD
    DLPGSNTAAP VQETLHGCQP VTQEDGKESR ISVQERQ
    SEQ ID NO: 63
    vector clone: pIB1031
    Sequence: MALPVTALLL PLALLLHAAR PDYKDDDDKD KQLDADVSPK PTIFLPSIAE
    TKLQKAGTYL CLLEKFFPDV IKIHWQEKKS NTILGSQEGN TMKTNDTYMK
    FSWLTVPEKS LDKEHRCIVR HENNKNGVDQ EIIFPPIKTD VITMDPKDNC
    SKDANDTLLL QLTNTSAYYM YLLLLLKSVV YFAIITCCLL RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 64
    vector clone: pIB1032
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LIQNPEPAVY QLKDPRSQDS
    TLCLFTDFDS QINVPKTMES GTFITDKCVL DMKAMDSKSN GAIAWSNQTS
    FTCQDIFKET NATYPSSDVP CDATLTEKSF ETDMNLNFQN LLVIVLRILL
    LKVAGFNLLM TLRLWSSRSK RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP
    RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT AAPVQETLHG
    CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 65
    vector clone: pIB1033
    Sequence: MALPVTALLL PLALLLHAAR PDYKDDDDKV LTPPKVSLFE PSKAEIANKQ
    KATLVCLARG FFPDHVELSW WVNGKEVHSG VCTDPQAYKE SNYSYCLSSR
    LRVSATFWHN PRNHFRCQVQ FHGLSEEDKW PEGSPKPVTQ NISAEAWGRA
    DCGITSASYQ QGVLSATILY EILLGKATLY AVLVSTLVVM AMVKRKNSRS
    KRSRLLHSDY MNMTPRRPGP TRKHYQPYAP PRDFAAYRSK KVAKKPTNKA
    PHPKQEPQEI NFPDDLPGSN TAAPVQETLH GCQPVTQEDG KESRISVQER
    Q
    SEQ ID NO: 66
    vector clone: pIB1046
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LSQPHTKPSV FVMKNGTNVA
    CLVKEFYPKD IRINLVSSKK ITEFDPAIVI SPSGKYNAVK LGKYEDSNSV
    TCSVQHDNKT VHSTDFEVKT DSTDHVKPKE TENTKQPSKS CHKPKAIVHT
    EKVNMMSLTV LGLRMLFAKT VAVNFLLTAK LFFLRSKRSR LLHSDYMNMT
    PRRPGPTRKH YQPYAPPRDF AAYRSKKVAK KPTNKAPHPK QEPQEINFPD
    DLPGSNTAAP VQETLHGCQP VTQEDGKESR ISVQERQRAK RGSGEGRGSL
    LTCGDVEENP GPMALPVTAL LLPLALLLHA ARPDYKDDDD KDKQLDADVS
    PKPTIFLPSI AETKLQKAGT YLCLLEKFFP DVIKIHWQEK KSNTILGSQE
    GNTMKTNDTY MKFSWLTVPE KSLDKEHRCI VRHENNKNGV DQEIIFPPIK
    TDVITMDPKD NCSKDANDTL LLQLTNTSAY YMYLLLLLKS VVYFAIITCC
    LLRSKRSRLL HSDYMNMTPR RPGPTRKHYQ PYAPPRDFAA YRSKKVAKKP
    TNKAPHPKQE PQEINFPDDL PGSNTAAPVQ ETLHGCQPVT QEDGKESRIS
    VQERQ
    SEQ ID NO: 67
    vector clone: pIB1047
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LIQNPEPAVY QLKDPRSQDS
    TLCLFTDFDS QINVPKTMES GTFITDKCVL DMKAMDSKSN GAIAWSNQTS
    FTCQDIFKET NATYPSSDVP CDATLTEKSF ETDMNLNFQN LLVIVLRILL
    LKVAGFNLLM TLRLWSSRSK RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP
    RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT AAPVQETLHG
    CQPVTQEDGK ESRISVQERQ RAKRGSGEGR GSLLTCGDVE ENPGPMALPV
    TALLLPLALL LHAARPDYKD DDDKVLTPPK VSLFEPSKAE IANKQKATLV
    CLARGFFPDH VELSWWVNGK EVHSGVCTDP QAYKESNYSY CLSSRLRVSA
    TFWHNPRNHF RCQVQFHGLS EEDKWPEGSP KPVTQNISAE AWGRADCGIT
    SASYQQGVLS ATILYEILLG KATLYAVLVS TLVVMAMVKR KNSRSKRSRL
    LHSDYMNMTP RRPGPTRKHY QPYAPPRDFA AYRSKKVAKK PTNKAPHPKQ
    EPQEINFPDD LPGSNTAAPV QETLHGCQPV TQEDGKESRI SVQERQ
    SEQ ID NO: 68
    vector clone: pIB1048
    Sequence: MEHSTFLSGL VLATLLSQVS PEQKLISEED LFKIPIEELE DRVFVNCNTS
    ITWVEGTVGT LLSDITRLDL GKRILDPRGI YRCNGTDIYK DKESTVQVHY
    RMCQSCVELD PATVAGIIVT DVIATLLLAL GVFCFARSKR SRLLHSDYMN
    MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH PKQEPQEINF
    PDDLPGSNTA APVQETLHGC QPVTQEDGKE SRISVQERQR AKRGSGEGRG
    SLLTCGDVEE NPGPMQSGTH WRVLGLCLLS VGVWGQDYKD
    DDDKDGNEEM
    GGITQTPYKV SISGTTVILT CPQYPGSEIL WQHNDKNIGG DEDDKNIGSD
    EDHLSLKEFS ELEQSGYYVC YPRGSKPEDA NFYLYLRARV CENCMEMDVM
    SVATIVIVDI CITGGLLLLV YYWSRSKRSR LLHSDYMNMT PRRPGPTRKH
    YQPYAPPRDF AAYRSKKVAK KPTNKAPHPK QEPQEINFPD DLPGSNTAAP
    VQETLHGCQP VTQEDGKESR ISVQERQ
    SEQ ID NO: 69
    vector clone: pIB1049
    Sequence: MEQGKGLAVL ILAIILLQGT LAEQKLISEE DLQSIKGNHL VKVYDYQEDG
    SVLLTCDAEA KNITWFKDGK MIGFLTEDKK KWNLGSNAKD PRGMYQCKGS
    QNKSKPLQVY YRMCQNCIEL NAATISGFLF AEIVSIFVLA VGVYFIARSK
    RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP
    HPKQEPQEIN FPDDLPGSNT AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    RAKRGSGMQS GTHWRVLGLC LLSVGVWGQD YKDDDDKDGN
    EEMGGITQTP
    YKVSISGTTV ILTCPQYPGS EILWQHNDKN IGGDEDDKNI GSDEDHLSLK
    EFSELEQSGY YVCYPRGSKP EDANFYLYLR ARVCENCMEM DVMSVATIVI
    VDICITGGLL LLVYYWSRSK RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP
    RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT AAPVQETLHG
    CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 70
    vector clone: pIB1050
    Sequence: MEHSTFLSGL VLATLLSQVS PEQKLISEED LFKIPIEELE DRVFVNCNTS
    ITWVEGTVGT LLSDITRLDL GKRILDPRGI YRCNGTDIYK DKESTVQVHY
    RMCQSCVELD PATVAGIIVT DVIATLLLAL GVFCFAGHET GRLSGAADTQ
    ALLRNDQVYQ PLRDRDDAQY SHLGGNWARN KRSKRSRLLH SDYMNMTPRR
    PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT NKAPHPKQEP QEINFPDDLP
    GSNTAAPVQE TLHGCQPVTQ EDGKESRISV QERQRAKRGS GEGRGSLLTC
    GDVEENPGPM QSGTHWRVLG LCLLSVGVWG QDYKDDDDKD
    GNEEMGGITQ
    TPYKVSISGT TVILTCPQYP GSEILWQHND KNIGGDEDDK NIGSDEDHLS
    LKEFSELEQS GYYVCYPRGS KPEDANFYLY LRARVCENCM EMDVMSVATI
    VIVDICITGG LLLLVYYWSK NRKAKAKPVT RGAGAGGRQR GQNKERPPPV
    PNPDYEPIRK GQRDLYSGLN QRRIRSKRSR LLHSDYMNMT PRRPGPTRKH
    YQPYAPPRDF AAYRSKKVAK KPTNKAPHPK QEPQEINFPD DLPGSNTAAP
    VQETLHGCQP VTQEDGKESR ISVQERQ
    SEQ ID NO: 71
    vector clone: pIB1051
    Sequence: MEHSTFLSGL VLATLLSQVS PEQKLISEED LFKIPIEELE DRVFVNCNTS
    ITWVEGTVGT LLSDITRLDL GKRILDPRGI YRCNGTDIYK DKESTVQVHY
    RMCQSCVELD PATVAGIIVT DVIATLLLAL GVFCFAGHET GRLSGAADTQ
    ALLRNDQVYQ PLRDRDDAQY SHLGGNWARN KRAKRGSGEG RGSLLTCGDV
    EENPGPMQSG THWRVLGLCL LSVGVWGQDY KDDDDKDGNE EMGGITQTPY
    KVSISGTTVI LTCPQYPGSE ILWQHNDKNI GGDEDDKNIG SDEDHLSLKE
    FSELEQSGYY VCYPRGSKPE DANFYLYLRA RVCENCMEMD VMSVATIVIV
    DICITGGLLL LVYYWSKNRK AKAKPVTRGA GAGGRQRGQN KERPPPVPNP
    DYEPIRKGQR DLYSGLNQRR I
    SEQ ID NO: 72
    vector clone: pIB1052
    Sequence: MEHSTFLSGL VLATLLSQVS PEQKLISEED LFKIPIEELE DRVFVNCNTS
    ITWVEGTVGT LLSDITRLDL GKRILDPRGI YRCNGTDIYK DKESTVQVHY
    RMCQSCVELD PATVAGIIVT DVIATLLLAL GVFCFARAKR GSGEGRGSLL
    TCGDVEENPG PMQSGTHWRV LGLCLLSVGV WGQDYKDDDD
    KDGNEEMGGI
    TQTPYKVSIS GTTVILTCPQ YPGSEILWQH NDKNIGGDED DKNIGSDEDH
    LSLKEFSELE QSGYYVCYPR GSKPEDANFY LYLRARVCEN CMEMDVMSVA
    TIVIVDICIT GGLLLLVYYW S
    SEQ ID NO: 73
    vector clone: pIB1053
    Sequence: MEQGKGLAVL ILAIILLQGT LAEQKLISEE DLQSIKGNHL VKVYDYQEDG
    SVLLTCDAEA KNITWFKDGK MIGFLTEDKK KWNLGSNAKD PRGMYQCKGS
    QNKSKPLQVY YRMCQNCIEL NAATISGFLF AEIVSIFVLA VGVYFIAGQD
    GVRQSRASDK QTLLPNDQLY QPLKDREDDQ YSHLQGNQLR RNRSKRSRLL
    HSDYMNMTPR RPGPTRKHYQ PYAPPRDFAA YRSKKVAKKP TNKAPHPKQE
    PQEINFPDDL PGSNTAAPVQ ETLHGCQPVT QEDGKESRIS VQERQRAKRG
    SGEGRGSLLT CGDVEENPGP MQSGTHWRVL GLCLLSVGVW GQDYKDDDDK
    DGNEEMGGIT QTPYKVSISG TTVILTCPQY PGSEILWQHN DKNIGGDEDD
    KNIGSDEDHL SLKEFSELEQ SGYYVCYPRG SKPEDANFYL YLRARVCENC
    MEMDVMSVAT IVIVDICITG GLLLLVYYWS KNRKAKAKPV TRGAGAGGRQ
    RGQNKERPPP VPNPDYEPIR KGQRDLYSGL NQRRIRSKRS RLLHSDYMNM
    TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP
    DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 74
    vector clone: pIB1054
    Sequence: MEQGKGLAVL ILAIILLQGT LAEQKLISEE DLQSIKGNHL VKVYDYQEDG
    SVLLTCDAEA KNITWFKDGK MIGFLTEDKK KWNLGSNAKD PRGMYQCKGS
    QNKSKPLQVY YRMCQNCIEL NAATISGFLF AEIVSIFVLA VGVYFIAGQD
    GVRQSRASDK QTLLPNDQLY QPLKDREDDQ YSHLQGNQLR RNRAKRGSGE
    GRGSLLTCGD VEENPGPMQS GTHWRVLGLC LLSVGVWGQD
    YKDDDDKDGN
    EEMGGITQTP YKVSISGTTV ILTCPQYPGS EILWQHNDKN IGGDEDDKNI
    GSDEDHLSLK EFSELEQSGY YVCYPRGSKP EDANFYLYLR ARVCENCMEM
    DVMSVATIVI VDICITGGLL LLVYYWSKNR KAKAKPVTRG AGAGGRQRGQ
    NKERPPPVPN PDYEPIRKGQ RDLYSGLNQR RI
    SEQ ID NO: 75
    vector clone: pIB1055
    Sequence: MEQGKGLAVL ILAIILLQGT LAEQKLISEE DLQSIKGNHL VKVYDYQEDG
    SVLLTCDAEA KNITWFKDGK MIGFLTEDKK KWNLGSNAKD PRGMYQCKGS
    QNKSKPLQVY YRMCQNCIEL NAATISGFLF AEIVSIFVLA VGVYFIARAK
    RGSGEGRGSL LTCGDVEENP GPMQSGTHWR VLGLCLLSVG VWGQDYKDDD
    DKDGNEEMGG ITQTPYKVSI SGTTVILTCP QYPGSEILWQ HNDKNIGGDE
    DDKNIGSDED HLSLKEFSEL EQSGYYVCYP RGSKPEDANF YLYLRARVCE
    NCMEMDVMSV ATIVIVDICI TGGLLLLVYY WS
    SEQ ID NO: 76
    vector clone: pIB1056
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LRVKFSRSAD APAYQQGQNQ LYNELNLGRR EEYDVLDKRR GRDPEMGGKP
    QRRKNPQEGL YNELQKDKMA EAYSEIGMKG ERRRGKGHDG LYQGLSTATK
    DTYDALHMQA LPPRRSKRSR LLHSDYMNMT PRRPGPTRKH YQPYAPPRDF
    AAYRSKKVAK KPTNKAPHPK QEPQEINFPD DLPGSNTAAP VQETLHGCQP
    VTQEDGKESR ISVQERQEQK LISEEDL
    SEQ ID NO: 78
    vector clone: pIB1057
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LRVKFSRSAD APAYQQGQNQ LYNELNLGRR EEYDVLDKRR GRDPEMGGKP
    QRRKNPQEGL YNELQKDKMA EAYSEIGMKG ERRRGKGHDG LYQGLSTATK
    DTYDALHMQA LPPREQKLIS EEDL
    SEQ ID NO: 79
    vector clone: pIB1058
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LEQKLISEED L
    SEQ ID NO: 80
    vector clone: pIB1059
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LRVKFSRSAD APAYQQGQNQ LYNELNLGRR EEYDVLDKRR GRDPEMGGKP
    QRRKNPQEGL YNELQKDKMA EAYSEIGMKG ERRRGKGHDG LYQGLSTATK
    DTYDALHMQA LPPRRVKFSR SADAPAYQQG QNQLYNELNL GRREEYDVLD
    KRRGRDPEMG GKPQRRKNPQ EGLYNELQKD KMAEAYSEIG MKGERRRGKG
    HDGLYQGLST ATKDTYDALH MQALPPRRSK RSRLLHSDYM NMTPRRPGPT
    RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT
    AAPVQETLHG CQPVTQEDGK ESRISVQERQ EQKLISEEDL
    SEQ ID NO: 81
    vector clone: pIB1060
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LRVKFSRSAD APAYQQGQNQ LYNELNLGRR EEYDVLDKRR GRDPEMGGKP
    QRRKNPQEGL YNELQKDKMA EAYSEIGMKG ERRRGKGHDG LYQGLSTATK
    DTYDALHMQA LPPRRVKFSR SADAPAYQQG QNQLYNELNL GRREEYDVLD
    KRRGRDPEMG GKPQRRKNPQ EGLYNELQKD KMAEAYSEIG MKGERRRGKG
    HDGLYQGLST ATKDTYDALH MQALPPREQK LISEEDL
    SEQ ID NO: 82
    vector clone: pIB1061
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LRSKRSRLLH SDYMNMTPRR PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT
    NKAPHPKQEP QEINFPDDLP GSNTAAPVQE TLHGCQPVTQ EDGKESRISV
    QERQRVKFSR SADAPAYQQG QNQLYNELNL GRREEYDVLD KRRGRDPEMG
    GKPQRRKNPQ EGLYNELQKD KMAEAYSEIG MKGERRRGKG HDGLYQGLST
    ATKDTYDALH MQALPPRRVK FSRSADAPAY QQGQNQLYNE LNLGRREEYD
    VLDKRRGRDP EMGGKPQRRK NPQEGLYNEL QKDKMAEAYS EIGMKGERRR
    GKGHDGLYQG LSTATKDTYD ALHMQALPPR EQKLISEEDL
    SEQ ID NO: 83
    vector clone: pIB1062
    Sequence: MKWKALFTAA ILQAQLPITE AQSFGLLDPK LCYLLDGILF IYGVILTALF
    LRSKRSRLLH SDYMNMTPRR PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT
    NKAPHPKQEP QEINFPDDLP GSNTAAPVQE TLHGCQPVTQ EDGKESRISV
    QERQRVKFSR SADAPAYQQG QNQLYNELNL GRREEYDVLD KRRGRDPEMG
    GKPQRRKNPQ EGLYNELQKD KMAEAYSEIG MKGERRRGKG HDGLYQGLST
    ATKDTYDALH MQALPPREQK LISEEDL
    SEQ ID NO: 84
    vector clone: pIB1063
    Sequence: MGHTRRQGTS PSKCPYLNFF QLLVLAGLSH FCSGEQKLIS EEDLVIHVTK
    EVKEVATLSC GHNVSVEELA QTRIYWQKEK KMVLTMMSGD MNIWPEYKNR
    TIFDITNNLS IVILALRPSD EGTYECVVLK YEKDAFKREH LAEVTLSVKA
    DFPTPSISDF EIPTSNIRRI ICSTSGGFPE PHLSWLENGE ELNAINTTVS
    QDPETELYAV SSKLDFNMTT NHSFMCLIKY GHLRVNQTFN WNTTKQEHFP
    DNLLPSWAIT LISVNGIFVI CCL
    SEQ ID NO: 85
    vector clone: pIB1064
    Sequence: MGCGCSSHPE DDWMENIDVC ENCHYPIVPL DGKGTLLIRN GSEVRDPLVT
    YEGSNPPASP LQDNLVIALH SYEPSHDGDL GFEKGEQLRI LEQSGEWWKA
    QSLTTGQEGF IPFNFVAKAN SLEPEPWFFK NLSRKDAERQ LLAPGNTHGS
    FLIRESESTA GSFSLSVRDF DQNQGEVVKH YKIRNLDNGG FYISPRITFP
    GLHELVRHYT NASDGLCTRL SRPCQTQKPQ KPWWEDEWEV PRETLKLVER
    LGAGQFGEVW MGYYNGHTKV AVKSLKQGSM SPDAFLAEAN
    LMKQLQHQRL
    VRLYAVVTQE PIYIITEYME NGSLVDFLKT PSGIKLTINK LLDMAAQIAE
    GMAFIEERNY IHRDLRAANI LVSDTLSCKI ADFGLARLIE DNEYTAREGA
    KFPIKWTAPE AINYGTFTIK SDVWSFGILL TEIVTHGRIP YPGMTNPEVI
    QNLERGYRMV RPDNCPEELY QLMRLCWKER PEDRPTFDYL RSVLEDFFTA
    TEGQYQPQPE QKLISEEDL
    SEQ ID NO: 86
    vector clone: pIB1065
    Sequence: MGCGCSSHPE DDWMENIDVC ENCHYPIVPL DGKGTLLIRN GSEVRDPLVT
    YEGSNPPASP LQDNLVIALH SYEPSHDGDL GFEKGEQLRI LEQSGEWWKA
    QSLTTGQEGF IPFNFVAKAN SLEPEPWFFK NLSRKDAERQ LLAPGNTHGS
    FLIRESESTA GSFSLSVRDF DQNQGEVVKH YKIRNLDNGG FYISPRITFP
    GLHELVRHYT NASDGLCTRL SRPCQTQKPQ KPWWEDEWEV PRETLKLVER
    LGAGQFGEVW MGYYNGHTKV AVKSLKQGSM SPDAFLAEAN
    LMKQLQHQRL
    VRLYAVVTQE PIYIITEYME NGSLVDFLKT PSGIKLTINK LLDMAAQIAE
    GMAFIEERNY IHRDLRAANI LVSDTLSCKI ADFGLARLIE DNEYTAREGA
    KFPIKWTAPE AINYGTFTIK SDVWSFGILL TEIVTHGRIP YPGMTNPEVI
    QNLERGYRMV RPDNCPEELY QLMRLCWKER PEDRPTFDYL RSVLEDFFTA
    TEGQFQPQPE QKLISEEDL
    SEQ ID NO: 87
    vector clone: pIB1066
    Sequence: MGHTRRQGTS PSKCPYLNFF QLLVLAGLSH FCSGEQKLIS EEDLVIHVTK
    EVKEVATLSC GHNVSVEELA QTRIYWQKEK KMVLTMMSGD MNIWPEYKNR
    TIFDITNNLS IVILALRPSD EGTYECVVLK YEKDAFKREH LAEVTLSVKA
    DFPTPSISDF EIPTSNIRRI ICSTSGGFPE PHLSWLENGE ELNAINTTVS
    QDPETELYAV SSKLDFNMTT NHSFMCLIKY GHLRVNQTFN WNTTKQEHFP
    DNLLPSWAIT LISVNGIFVI CCLGCGCSSH PEDDWMENID VCENCHYPIV
    PLDGKGTLLI RNGSEVRDPL VTYEGSNPPA SPLQDNLVIA LHSYEPSHDG
    DLGFEKGEQL RILEQSGEWW KAQSLTTGQE GFIPFNFVAK ANSLEPEPWF
    FKNLSRKDAE RQLLAPGNTH GSFLIRESES TAGSFSLSVR DFDQNQGEVV
    KHYKIRNLDN GGFYISPRIT FPGLHELVRH YTNASDGLCT RLSRPCQTQK
    PQKPWWEDEW EVPRETLKLV ERLGAGQFGE VWMGYYNGHT
    KVAVKSLKQG
    SMSPDAFLAE ANLMKQLQHQ RLVRLYAVVT QEPIYIITEY MENGSLVDFL
    KTPSGIKLTI NKLLDMAAQI AEGMAFIEER NYIHRDLRAA NILVSDTLSC
    KIADFGLARL IEDNEYTARE GAKFPIKWTA PEAINYGTFT IKSDVWSFGI
    LLTEIVTHGR IPYPGMTNPE VIQNLERGYR MVRPDNCPEE LYQLMRLCWK
    ERPEDRPTFD YLRSVLEDFF TATEGQYQPQ P
    SEQ ID NO: 88
    vector clone: pIB1067
    Sequence: MGHTRRQGTS PSKCPYLNFF QLLVLAGLSH FCSGEQKLIS EEDLVIHVTK
    EVKEVATLSC GHNVSVEELA QTRIYWQKEK KMVLTMMSGD MNIWPEYKNR
    TIFDITNNLS IVILALRPSD EGTYECVVLK YEKDAFKREH LAEVTLSVKA
    DFPTPSISDF EIPTSNIRRI ICSTSGGFPE PHLSWLENGE ELNAINTTVS
    QDPETELYAV SSKLDFNMTT NHSFMCLIKY GHLRVNQTFN WNTTKQEHFP
    DNLLPSWAIT LISVNGIFVI CCLGCGCSSH PEDDWMENID VCENCHYPIV
    PLDGKGTLLI RNGSEVRDPL VTYEGSNPPA SPLQDNLVIA LHSYEPSHDG
    DLGFEKGEQL RILEQSGEWW KAQSLTTGQE GFIPFNFVAK ANSLEPEPWF
    FKNLSRKDAE RQLLAPGNTH GSFLIRESES TAGSFSLSVR DFDQNQGEVV
    KHYKIRNLDN GGFYISPRIT FPGLHELVRH YTNASDGLCT RLSRPCQTQK
    PQKPWWEDEW EVPRETLKLV ERLGAGQFGE VWMGYYNGHT
    KVAVKSLKQG
    SMSPDAFLAE ANLMKQLQHQ RLVRLYAVVT QEPIYIITEY MENGSLVDFL
    KTPSGIKLTI NKLLDMAAQI AEGMAFIEER NYIHRDLRAA NILVSDTLSC
    KIADFGLARL IEDNEYTARE GAKFPIKWTA PEAINYGTFT IKSDVWSFGI
    LLTEIVTHGR IPYPGMTNPE VIQNLERGYR MVRPDNCPEE LYQLMRLCWK
    ERPEDRPTFD YLRSVLEDFF TATEGQYQPQ PRSKRSRLLH SDYMNMTPRR
    PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT NKAPHPKQEP QEINFPDDLP
    GSNTAAPVQE TLHGCQPVTQ EDGKESRISV QERQ
    SEQ ID NO: 89
    vector clone: pIB1068
    Sequence: MGHTRRQGTS PSKCPYLNFF QLLVLAGLSH FCSGEQKLIS EEDLVIHVTK
    EVKEVATLSC GHNVSVEELA QTRIYWQKEK KMVLTMMSGD MNIWPEYKNR
    TIFDITNNLS IVILALRPSD EGTYECVVLK YEKDAFKREH LAEVTLSVKA
    DFPTPSISDF EIPTSNIRRI ICSTSGGFPE PHLSWLENGE ELNAINTTVS
    QDPETELYAV SSKLDFNMTT NHSFMCLIKY GHLRVNQTFN WNTTKQEHFP
    DNLLPSWAIT LISVNGIFVI CCLGCGCSSH PEDDWMENID VCENCHYPIV
    PLDGKGTLLI RNGSEVRDPL VTYEGSNPPA SPLQDNLVIA LHSYEPSHDG
    DLGFEKGEQL RILEQSGEWW KAQSLTTGQE GFIPFNFVAK ANSLEPEPWF
    FKNLSRKDAE RQLLAPGNTH GSFLIRESES TAGSFSLSVR DFDQNQGEVV
    KHYKIRNLDN GGFYISPRIT FPGLHELVRH YTNASDGLCT RLSRPCQTQK
    PQKPWWEDEW EVPRETLKLV ERLGAGQFGE VWMGYYNGHT
    KVAVKSLKQG
    SMSPDAFLAE ANLMKQLQHQ RLVRLYAVVT QEPIYIITEY MENGSLVDFL
    KTPSGIKLTI NKLLDMAAQI AEGMAFIEER NYIHRDLRAA NILVSDTLSC
    KIADFGLARL IEDNEYTARE GAKFPIKWTA PEAINYGTFT IKSDVWSFGI
    LLTEIVTHGR IPYPGMTNPE VIQNLERGYR MVRPDNCPEE LYQLMRLCWK
    ERPEDRPTFD YLRSVLEDFF TATEGQFQPQ P
    SEQ ID NO: 90
    vector clone: pIB1069
    Sequence: MGHTRRQGTS PSKCPYLNFF QLLVLAGLSH FCSGEQKLIS EEDLVIHVTK
    EVKEVATLSC GHNVSVEELA QTRIYWQKEK KMVLTMMSGD MNIWPEYKNR
    TIFDITNNLS IVILALRPSD EGTYECVVLK YEKDAFKREH LAEVTLSVKA
    DFPTPSISDF EIPTSNIRRI ICSTSGGFPE PHLSWLENGE ELNAINTTVS
    QDPETELYAV SSKLDFNMTT NHSFMCLIKY GHLRVNQTFN WNTTKQEHFP
    DNLLPSWAIT LISVNGIFVI CCLGCGCSSH PEDDWMENID VCENCHYPIV
    PLDGKGTLLI RNGSEVRDPL VTYEGSNPPA SPLQDNLVIA LHSYEPSHDG
    DLGFEKGEQL RILEQSGEWW KAQSLTTGQE GFIPFNFVAK ANSLEPEPWF
    FKNLSRKDAE RQLLAPGNTH GSFLIRESES TAGSFSLSVR DFDQNQGEVV
    KHYKIRNLDN GGFYISPRIT FPGLHELVRH YTNASDGLCT RLSRPCQTQK
    PQKPWWEDEW EVPRETLKLV ERLGAGQFGE VWMGYYNGHT
    KVAVKSLKQG
    SMSPDAFLAE ANLMKQLQHQ RLVRLYAVVT QEPIYIITEY MENGSLVDFL
    KTPSGIKLTI NKLLDMAAQI AEGMAFIEER NYIHRDLRAA NILVSDTLSC
    KIADFGLARL IEDNEYTARE GAKFPIKWTA PEAINYGTFT IKSDVWSFGI
    LLTEIVTHGR IPYPGMTNPE VIQNLERGYR MVRPDNCPEE LYQLMRLCWK
    ERPEDRPTFD YLRSVLEDFF TATEGQFQPQ PRSKRSRLLH SDYMNMTPRR
    PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT NKAPHPKQEP QEINFPDDLP
    GSNTAAPVQE TLHGCQPVTQ EDGKESRISV QERQ
    SEQ ID NO: 91
    vector clone: pIB1070
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LMEEAILVPC VLGLLLLPIL
    AMLMALCVHC HRLPGSYDST SSDSLYPRGI QFKRPHTVAP WPPAYPPVTS
    YPPLSQPDLL PIPRSPQPLG GSHRTPSSRR DSDGANSVAS YENEGASGIR
    GAQAGWGVWG PSWTRLTPVS LPPEPACEDA DEDEDDYHNP GYLVVLPDST
    PATSTAAPSA PALSTPGIRD SAFSMESIDD YVNVPESGES AEASLDGSRE
    YVNVSQELHP GAAKTEPAAL SSQEAEEVEE EGAPDYENLQ ELN
    SEQ ID NO: 92
    vector clone: pIB1071
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LMEEAILVPC VLGLLLLPIL
    AMLMALCVHC HRLPGSYDST SSDSLYPRGI QFKRPHTVAP WPPAYPPVTS
    YPPLSQPDLL PIPRSPQPLG GSHRTPSSRR DSDGANSVAS YENEGASGIR
    GAQAGWGVWG PSWTRLTPVS LPPEPACEDA DEDEDDYHNP GYLVVLPDST
    PATSTAAPSA PALSTPGIRD SAFSMESIDD YVNVPESGES AEASLDGSRE
    YVNVSQELHP GAAKTEPAAL SSQEAEEVEE EGAPDYENLQ ELNRSKRSRL
    LHSDYMNMTP RRPGPTRKHY QPYAPPRDFA AYRSKKVAKK PTNKAPHPKQ
    EPQEINFPDD LPGSNTAAPV QETLHGCQPV TQEDGKESRI SVQERQ
    SEQ ID NO: 93
    vector clone: pIB1072
    Sequence: MNRGVPFRHL LLVLQLALLP AATQGEQKLI SEEDLKKVVL GKKGDTVELT
    CTASQKKSIQ FHWKNSNQIK ILGNQGSFLT KGPSKLNDRA DSRRSLWDQG
    NFPLIIKNLK IEDSDTYICE VEDQKEEVQL LVFGLTANSD THLLQGQSLT
    LTLESPPGSS PSVQCRSPRG KNIQGGKTLS VSQLELQDSG TWTCTVLQNQ
    KKVEFKIDIV VLAFQKASSI VYKKEGEQVE FSFPLAFTVE KLTGSGELWW
    QAERASSSKS WITFDLKNKE VSVKRVTQDP KLQMGKKLPL HLTLPQALPQ
    YAGSGNLTLA LEAKTGKLHQ EVNLVVMRAT QLQKNLTCEV WGPTSPKLML
    SLKLENKEAK VSKREKAVWV LNPEAGMWQC LLSDSGQVLL ESNIKVLPTW
    STPVQPMALI VLGGVAGLLL FIGLGIFFCV RCRHRRRQAE RMSQIKRLLS
    EKKTCQCPHR FQKTCSPI
    SEQ ID NO: 94
    vector clone: pIB1073
    Sequence: MNRGVPFRHL LLVLQLALLP AATQGEQKLI SEEDLKKVVL GKKGDTVELT
    CTASQKKSIQ FHWKNSNQIK ILGNQGSFLT KGPSKLNDRA DSRRSLWDQG
    NFPLIIKNLK IEDSDTYICE VEDQKEEVQL LVFGLTANSD THLLQGQSLT
    LTLESPPGSS PSVQCRSPRG KNIQGGKTLS VSQLELQDSG TWTCTVLQNQ
    KKVEFKIDIV VLAFQKASSI VYKKEGEQVE FSFPLAFTVE KLTGSGELWW
    QAERASSSKS WITFDLKNKE VSVKRVTQDP KLQMGKKLPL HLTLPQALPQ
    YAGSGNLTLA LEAKTGKLHQ EVNLVVMRAT QLQKNLTCEV WGPTSPKLML
    SLKLENKEAK VSKREKAVWV LNPEAGMWQC LLSDSGQVLL ESNIKVLPTW
    STPVQPMALI VLGGVAGLLL FIGLGIFFCV RCRHRRRQAE RMSQIKRLLS
    EKKTCQCPHR FQKTCSPIRS KRSRLLHSDY MNMTPRRPGP TRKHYQPYAP
    PRDFAAYRSK KVAKKPTNKA PHPKQEPQEI NFPDDLPGSN TAAPVQETLH
    GCQPVTQEDG KESRISVQER Q
    SEQ ID NO: 95
    vector clone: pIB1074
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LSQFRVSPLD RTWNLGETVE
    LKCQVLLSNP TSGCSWLFQP RGAAASPTFL LYLSQNKPKA AEGLDTQRFS
    GKRLGDTFVL TLSDFRRENE GYYFCSALSN SIMYFSHFVP VFLPAKPTTT
    PAPRPPTPAP TIASQPLSLR PEACRPAAGG AVHTRGLDFA CDIYIWAPLA
    GTCGVLLLSL VITLYCNHRN RRRVCKCPRP VVKSGDKPSL SARYVRAKRG
    SGEGRGSLLT CGDVEENPGP MRPRLWLLLA AQLTVLHGNS VDYKDDDDKL
    QQTPAYIKVQ TNKMVMLSCE AKISLSNMRI YWLRQRQAPS SDSHHEFLAL
    WDSAKGTIHG EEVEQEKIAV FRDASRFILN LTSVKPEDSG IYFCMIVGSP
    ELTFGKGTQL SVVDFLPTTA QPTKKSTLKK RVCRLPRPET QKGPLCSPIT
    LGLLVAGVLV LLVSLGVAIH LCCRRRRARL RFMKQFYK
    SEQ ID NO: 96
    vector clone: pIB1075
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LSQFRVSPLD RTWNLGETVE
    LKCQVLLSNP TSGCSWLFQP RGAAASPTFL LYLSQNKPKA AEGLDTQRFS
    GKRLGDTFVL TLSDFRRENE GYYFCSALSN SIMYFSHFVP VFLPAKPTTT
    PAPRPPTPAP TIASQPLSLR PEACRPAAGG AVHTRGLDFA CDIYIWAPLA
    GTCGVLLLSL VITLYCNHRN RRRVCKCPRP VVKSGDKPSL SARYVRSKRS
    RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP
    KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQRA
    KRGSGEGRGS LLTCGDVEEN PGPMRPRLWL LLAAQLTVLH GNSVDYKDDD
    DKLQQTPAYI KVQTNKMVML SCEAKISLSN MRIYWLRQRQ APSSDSHHEF
    LALWDSAKGT IHGEEVEQEK IAVFRDASRF ILNLTSVKPE DSGIYFCMIV
    GSPELTFGKG TQLSVVDFLP TTAQPTKKST LKKRVCRLPR PETQKGPLCS
    PITLGLLVAG VLVLLVSLGV AIHLCCRRRR ARLRFMKQFY KRSKRSRLLH
    SDYMNMTPRR PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT NKAPHPKQEP
    QEINFPDDLP GSNTAAPVQE TLHGCQPVTQ EDGKESRISV QERQ
    SEQ ID NO: 97
    vector clone: pIB1076
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LELTDTLQAE TDQLEDEKSA
    LQTEIANLLK EKEKLEFILA AHNCTYGCTG PGLEGCPTNG PKIPSIATGM
    VGALLLLLVV ALGIGLFMRS KRSRLLHSDY MNMTPRRPGP TRKHYQPYAP
    PRDFAAYRSK KVAKKPTNKA PHPKQEPQEI NFPDDLPGSN TAAPVQETLH
    GCQPVTQEDG KESRISVQER Q
    SEQ ID NO: 98
    vector clone: pIB1077
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LELTDTLQAE TDQLEDEKSA
    LQTEIANLLK EKEKLEFILA AHFWVLVVVG GVLACYSLLV TVAFIIFWVR
    SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS KKVAKKPTNK
    APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED GKESRISVQE
    RQ
    SEQ ID NO: 99
    vector clone: pIB1078
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LLEEKVKTLK AQNSELASTA
    NMLREQVAQL NCTYGCTGPG LEGCPTNGPK IPSIATGMVG ALLLLLVVAL
    GIGLFMRSKR SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV
    AKKPTNKAPH PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE
    SRISVQERQ
    SEQ ID NO: 100
    vector clone: pIB1079
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LLEEKVKTLK AQNSELASTA
    NMLREQVAQL FWVLVVVGGV LACYSLLVTV AFIIFWVRSK RSRLLHSDYM
    NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN
    FPDDLPGSNT AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 101
    vector clone: pIB1080
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LETQHKVLEL TAENERLQKK
    VEQLSRELST NCTYGCTGPG LEGCPTNGPK IPSIATGMVG ALLLLLVVAL
    GIGLFMRSKR SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR DFAAYRSKKV
    AKKPTNKAPH PKQEPQEINF PDDLPGSNTA APVQETLHGC QPVTQEDGKE
    SRISVQERQ
    SEQ ID NO: 102
    vector clone: pIB1081
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LETQHKVLEL TAENERLQKK
    VEQLSRELST FWVLVVVGGV LACYSLLVTV AFIIFWVRSK RSRLLHSDYM
    NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN
    FPDDLPGSNT AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 103
    vector clone: pIB1082
    Sequence: MRPSGTAGAA LLALLAALCP ASRAEQKLIS EEDLLEEKKV CQGTSNKLTQ
    LGTFEDHFLS LQRMFNNCEV VLGNLEITYV QRNYDLSFLK TIQEVAGYVL
    IALNTVERIP LENLQIIRGN MYYENSYALA VLSNYDANKT GLKELPMRNL
    QEILHGAVRF SNNPALCNVE SIQWRDIVSS DFLSNMSMDF QNHLGSCQKC
    DPSCPNGSCW GAGEENCQKL TKIICAQQCS GRCRGKSPSD CCHNQCAAGC
    TGPRESDCLV CRKFRDEATC KDTCPPLMLY NPTTYQMDVN PEGKYSFGAT
    CVKKCPRNYV VTDHGSCVRA CGADSYEMEE DGVRKCKKCE GPCRKVCNGI
    GIGEFKDSLS INATNIKHFK NCTSISGDLH ILPVAFRGDS FTHTPPLDPQ
    ELDILKTVKE ITGFLLIQAW PENRTDLHAF ENLEIIRGRT KQHGQFSLAV
    VSLNITSLGL RSLKEISDGD VIISGNKNLC YANTINWKKL FGTSGQKTKI
    ISNRGENSCK ATGQVCHALC SPEGCWGPEP RDCVSCRNVS RGRECVDKCN
    LLEGEPREFV ENSECIQCHP ECLPQAMNIT CTGRGPDNCI QCAHYIDGPH
    CVKTCPAGVM GENNTLVWKY ADAGHVCHLC HPNCTYGCTG PGLEGCPTNG
    PKIPSIATGM VGALLLLLVV ALGIGLFMRS KRSRLLHSDY MNMTPRRPGP
    TRKHYQPYAP PRDFAAYRSK KVAKKPTNKA PHPKQEPQEI NFPDDLPGSN
    TAAPVQETLH GCQPVTQEDG KESRISVQER Q
    SEQ ID NO: 104
    vector clone: pIB1083
    Sequence: MRPSGTAGAA LLALLAALCP ASRAEQKLIS EEDLGTSGQK TKIISNRGEN
    SCKATGQVCH ALCSPEGCWG PEPRDCVSCR NVSRGRECVD KCNLLEGEPR
    EFVENSECIQ CHPECLPQAM NITCTGRGPD NCIQCAHYID GPHCVKTCPA
    GVMGENNTLV WKYADAGHVC HLCHPNCTYG CTGPGLEGCP TNGPKIPSIA
    TGMVGALLLL LVVALGIGLF MRSKRSRLLH SDYMNMTPRR PGPTRKHYQP
    YAPPRDFAAY RSKKVAKKPT NKAPHPKQEP QEINFPDDLP GSNTAAPVQE
    TLHGCQPVTQ EDGKESRISV QERQ
    SEQ ID NO: 105
    vector clone: pIB1084
    Sequence: MRPSGTAGAA LLALLAALCP ASRAEQKLIS EEDLNCTYGC TGPGLEGCPT
    NGPKIPSIAT GMVGALLLLL VVALGIGLFM RSKRSRLLHS DYMNMTPRRP
    GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ EINFPDDLPG
    SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 106
    vector clone: pIB1085
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLTQVCTGTD MKLRLPASPE
    THLDMLRHLY QGCQVVQGNL ELTYLPTNAS LSFLQDIQEV QGYVLIAHNQ
    VRQVPLQRLR IVRGTQLFED NYALAVLDNG DPLNNTTPVT GASPGGLREL
    QLRSLTEILK GGVLIQRNPQ LCYQDTILWK DIFHKNNQLA LTLIDTNRSR
    ACHPCSPMCK GSRCWGESSE DCQSLTRTVC AGGCARCKGP LPTDCCHEQC
    AAGCTGPKHS DCLACLHFNH SGICELHCPA LVTYNTDTFE SMPNPEGRYT
    FGASCVTACP YNYLSTDVGS CTLVCPLHNQ EVTAEDGTQR CEKCSKPCAR
    VCYGLGMEHL REVRAVTSAN IQEFAGCKKI FGSLAFLPES FDGDPASNTA
    PLQPEQLQVF ETLEEITGYL YISAWPDSLP DLSVFQNLQV IRGRILHNGA
    YSLTLQGLGI SWLGLRSLRE LGSGLALIHH NTHLCFVHTV PWDQLFRNPH
    QALLHTANRP EDECVGEGLA CHQLCARGHC WGPGPTQCVN CSQFLRGQEC
    VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE ADQCVACAHY
    KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC THSCVDLDDK
    GCPAEQRASP LTSIISAVVG ILLVVVLGVV FGILIRSKRS RLLHSDYMNM
    TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP
    DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 107
    vector clone: pIB1086
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLTQVCTGTD MKLRLPASPE
    THLDMLRHLY QGCQVVQGNL ELTYLPTNAS LSFLQDIQEV QGYVLIAHNQ
    VRQVPLQRLR IVRGTQLFED NYALAVLDNG DPLNNTTPVT GASPGGLREL
    QLRSLTEILK GGVLIQRNPQ LCYQDTILWK DIFHKNNQLA LTLIDTNRSR
    ACHPCSPMCK GSRCWGESSE DCQSLTRTVC AGGCARCKGP LPTDCCHEQC
    AAGCTGPKHS DCLACLHFNH SGICELHCPA LVTYNTDTFE SMPNPEGRYT
    FGASCVTACP YNYLSTDVGS CTLVCPLHNQ EVTAEDGTQR CEKCSKPCAR
    VCYGLGMEHL REVRAVTSAN IQEFAGCKKI FGSLAFLPES FDGDPASNTA
    PLQPEQLQVF ETLEEITGYL YISAWPDSLP DLSVFQNLQV IRGRILHNGA
    YSLTLQGLGI SWLGLRSLRE LGSGLALIHH NTHLCFVHTV PWDQLFRNPH
    QALLHTANRP EDECVGEGLA CHQLCARGHC WGPGPTQCVN CSQFLRGQEC
    VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE ADQCVACAHY
    KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC THSCVDLDDK
    GCPAEQRASP LTSIISAVEG ILLVVVLGVV FGILIRSKRS RLLHSDYMNM
    TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP
    DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 108
    vector clone: pIB1087
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLTQVCTGTD MKLRLPASPE
    THLDMLRHLY QGCQVVQGNL ELTYLPTNAS LSFLQDIQEV QGYVLIAHNQ
    VRQVPLQRLR IVRGTQLFED NYALAVLDNG DPLNNTTPVT GASPGGLREL
    QLRSLTEILK GGVLIQRNPQ LCYQDTILWK DIFHKNNQLA LTLIDTNRSR
    ACHPCSPMCK GSRCWGESSE DCQSLTRTVC AGGCARCKGP LPTDCCHEQC
    AAGCTGPKHS DCLACLHFNH SGICELHCPA LVTYNTDTFE SMPNPEGRYT
    FGASCVTACP YNYLSTDVGS CTLVCPLHNQ EVTAEDGTQR CEKCSKPCAR
    VCYGLGMEHL REVRAVTSAN IQEFAGCKKI FGSLAFLPES FDGDPASNTA
    PLQPEQLQVF ETLEEITGYL YISAWPDSLP DLSVFQNLQV IRGRILHNGA
    YSLTLQGLGI SWLGLRSLRE LGSGLALIHH NTHLCFVHTV PWDQLFRNPH
    QALLHTANRP EDECVGEGLA CHQLCARGHC WGPGPTQCVN CSQFLRGQEC
    VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE ADQCVACAHY
    KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC THSCVDLDDK
    GCPAEQRASP LTSIISAVVD ILLVVVLGVV FGILIRSKRS RLLHSDYMNM
    TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP
    DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 109
    vector clone: pIB1088
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLTQVCTGTD MKLRLPASPE
    THLDMLRHLY QGCQVVQGNL ELTYLPTNAS LSFLQDIQEV QGYVLIAHNQ
    VRQVPLQRLR IVRGTQLFED NYALAVLDNG DPLNNTTPVT GASPGGLREL
    QLRSLTEILK GGVLIQRNPQ LCYQDTILWK DIFHKNNQLA LTLIDTNRSR
    ACHPCSPMCK GSRCWGESSE DCQSLTRTVC AGGCARCKGP LPTDCCHEQC
    AAGCTGPKHS DCLACLHFNH SGICELHCPA LVTYNTDTFE SMPNPEGRYT
    FGASCVTACP YNYLSTDVGS CTLVCPLHNQ EVTAEDGTQR CEKCSKPCAR
    VCYGLGMEHL REVRAVTSAN IQEFAGCKKI FGSLAFLPES FDGDPASNTA
    PLQPEQLQVF ETLEEITGYL YISAWPDSLP DLSVFQNLQV IRGRILHNGA
    YSLTLQGLGI SWLGLRSLRE LGSGLALIHH NTHLCFVHTV PWDQLFRNPH
    QALLHTANRP EDECVGEGLA CHQLCARGHC WGPGPTQCVN CSQFLRGQEC
    VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE ADQCVACAHY
    KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC THSCVDLDDK
    GCPAEQRASP LTSIISAVVR ILLVVVLGVV FGILIRSKRS RLLHSDYMNM
    TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP
    DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 110
    vector clone: pIB1089
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLQLCARGHC WGPGPTQCVN
    CSQFLRGQEC VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE
    ADQCVACAHY KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC
    THSCVDLDDK GCPAEQRASP LTSIISAVVG ILLVVVLGVV FGILIRSKRS
    RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP
    KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 111
    vector clone: pIB1090
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLQLCARGHC WGPGPTQCVN
    CSQFLRGQEC VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE
    ADQCVACAHY KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC
    THSCVDLDDK GCPAEQRASP LTSIISAVEG ILLVVVLGVV FGILIRSKRS
    RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP
    KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 112
    vector clone: pIB1091
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLQLCARGHC WGPGPTQCVN
    CSQFLRGQEC VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE
    ADQCVACAHY KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC
    THSCVDLDDK GCPAEQRASP LTSIISAVVD ILLVVVLGVV FGILIRSKRS
    RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP
    KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 113
    vector clone: pIB1092
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLQLCARGHC WGPGPTQCVN
    CSQFLRGQEC VEECRVLQGL PREYVNARHC LPCHPECQPQ NGSVTCFGPE
    ADQCVACAHY KDPPFCVARC PSGVKPDLSY MPIWKFPDEE GACQPCPINC
    THSCVDLDDK GCPAEQRASP LTSIISAVVR ILLVVVLGVV FGILIRSKRS
    RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP
    KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 114
    vector clone: pIB1093
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLSIISAVVG ILLVVVLGVV
    FGILIRSKRS RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA
    KKPTNKAPHP KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES
    RISVQERQ
    SEQ ID NO: 115
    vector clone: pIB1094
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLSIISAVEG ILLVVVLGVV
    FGILIRSKRS RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA
    KKPTNKAPHP KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES
    RISVQERQ
    SEQ ID NO: 116
    vector clone: pIB1095
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLSIISAVVD ILLVVVLGVV
    FGILIRSKRS RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA
    KKPTNKAPHP KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES
    RISVQERQ
    SEQ ID NO: 117
    vector clone: pIB1096
    Sequence: MELAALCRWG LLLALLPPGA ASEQKLISEE DLSIISAVVR ILLVVVLGVV
    FGILIRSKRS RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA
    KKPTNKAPHP KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES
    RISVQERQ
    SEQ ID NO: 118
    vector clone: pIB1097
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQEQLVESGG RLVTPGTPLT
    LTCTASGFSL GSDFMSWVRQ APGKGLEWIG YIDPRSDIPY YASWAKGRFT
    ISKTSTTVDL KITSPTTEDT ATYFCARDLN AGYFNGIFYI WGPGTLVTVS
    SGGGGSGGGG SGGGGSELVM TQTPSSVSAA VGDTVTINCQ ASETVATLLA
    WYQQKPGQPP KLLIYGASNL ESGVPSRFRG SGSGTEFTLT ISGMKAEDAA
    TYYCQYGYIS TGSNTFGAGT NVEIKAAAGS GGSGILVKQS PMLVAYDNAV
    NLSCKYSYNL FSREFRASLH KGLDSAVEVC VVYGNYSQQL QVYSKTGFNC
    DGKLGNESVT FYLQNLYVNQ TDIYFCKIEV MYPPPYLDNE KSNGTIIHVK
    GKHLCPSPLF PGPSKPFWVL VVVGGVLACY SLLVTVAFII FWVRSKRSRL
    LHSDYMNMTP RRPGPTRKHY QPYAPPRDFA AYRSKKVAKK PTNKAPHPKQ
    EPQEINFPDD LPGSNTAAPV QETLHGCQPV TQEDGKESRI SVQERQ
    SEQ ID NO: 119
    vector clone: pIB1098
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LELVMTQTPS SVSAAVGDTV
    TINCQASETV ATLLAWYQQK PGQPPKLLIY GASNLESGVP SRFRGSGSGT
    EFTLTISGMK AEDAATYYCQ YGYISTGSNT FGAGTNVEIK GGGGSGGGGS
    GGGGSQEQLV ESGGRLVTPG TPLTLTCTAS GFSLGSDFMS WVRQAPGKGL
    EWIGYIDPRS DIPYYASWAK GRFTISKTST TVDLKITSPT TEDTATYFCA
    RDLNAGYFNG IFYIWGPGTL VTVSSAAAGS GGSGILVKQS PMLVAYDNAV
    NLSCKYSYNL FSREFRASLH KGLDSAVEVC VVYGNYSQQL QVYSKTGFNC
    DGKLGNESVT FYLQNLYVNQ TDIYFCKIEV MYPPPYLDNE KSNGTIIHVK
    GKHLCPSPLF PGPSKPFWVL VVVGGVLACY SLLVTVAFII FWVRSKRSRL
    LHSDYMNMTP RRPGPTRKHY QPYAPPRDFA AYRSKKVAKK PTNKAPHPKQ
    EPQEINFPDD LPGSNTAAPV QETLHGCQPV TQEDGKESRI SVQERQ
    SEQ ID NO: 120
    vector clone: pIB1099
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQEQLVESGG RLVTPGTPLT
    LTCTASGFSL GSDFMSWVRQ APGKGLEWIG YIDPRSDIPY YASWAKGRFT
    ISKTSTTVDL KITSPTTEDT ATYFCARDLN AGYFNGIFYI WGPGTLVTVS
    SGGGGSGGGG SGGGGSELDM TQTPSSTSEP VGGTVTINCQ ASQTISSYLS
    WYQQKPGHPP KLLIYDASDL ASGVPSRFSG SRSGTQFTLT ISGVQCDDAA
    TYYCLGVYDY RSDDGAAFGG GTELEILAAA GSGGSGILVK QSPMLVAYDN
    AVNLSCKYSY NLFSREFRAS LHKGLDSAVE VCVVYGNYSQ QLQVYSKTGF
    NCDGKLGNES VTFYLQNLYV NQTDIYFCKI EVMYPPPYLD NEKSNGTIIH
    VKGKHLCPSP LFPGPSKPFW VLVVVGGVLA CYSLLVTVAF IIFWVRSKRS
    RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP
    KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 121
    vector clone: pIB1100
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LELDMTQTPS STSEPVGGTV
    TINCQASQTI SSYLSWYQQK PGHPPKLLIY DASDLASGVP SRFSGSRSGT
    QFTLTISGVQ CDDAATYYCL GVYDYRSDDG AAFGGGTELE ILGGGGSGGG
    GSGGGGSQEQ LVESGGRLVT PGTPLTLTCT ASGFSLGSDF MSWVRQAPGK
    GLEWIGYIDP RSDIPYYASW AKGRFTISKT STTVDLKITS PTTEDTATYF
    CARDLNAGYF NGIFYIWGPG TLVTVSSAAA GSGGSGILVK QSPMLVAYDN
    AVNLSCKYSY NLFSREFRAS LHKGLDSAVE VCVVYGNYSQ QLQVYSKTGF
    NCDGKLGNES VTFYLQNLYV NQTDIYFCKI EVMYPPPYLD NEKSNGTIIH
    VKGKHLCPSP LFPGPSKPFW VLVVVGGVLA CYSLLVTVAF IIFWVRSKRS
    RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP
    KQEPQEINFP DDLPGSNTAA PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 122
    vector clone: pIB1101
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LQSLEESGGR LVTPGTPLTL
    TCTVSGFSLS TNDMNWVRQA PGKGLEWIGV IYSDDTPDYA TWAKGRFTIS
    RTSTTVDLKI TSPTTEDTAT YFCARGHYDS AVYAYALNIW GPGTLVTVSS
    GGGGSGGGGS GGGGSELVMT QTPSSVSAAV GGTVTITCQA SQSLSNLLAW
    YQQKPGQPPK LLIYGASNLE SGVPSRFRGS GSGTDFTLTI SGMKAEDAAT
    YYCQGGHYSG LTFGNGTNVE IKAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 123
    vector clone: pIB1102
    Sequence: MALPVTALLL PLALLLHAAR PEQKLISEED LELVMTQTPS SVSAAVGGTV
    TITCQASQSL SNLLAWYQQK PGQPPKLLIY GASNLESGVP SRFRGSGSGT
    DFTLTISGMK AEDAATYYCQ GGHYSGLTFG NGTNVEIKGG GGSGGGGSGG
    GGSQSLEESG GRLVTPGTPL TLTCTVSGFS LSTNDMNWVR QAPGKGLEWI
    GVIYSDDTPD YATWAKGRFT ISRTSTTVDL KITSPTTEDT ATYFCARGHY
    DSAVYAYALN IWGPGTLVTV SSAAAGSGGS GILVKQSPML VAYDNAVNLS
    CKYSYNLFSR EFRASLHKGL DSAVEVCVVY GNYSQQLQVY SKTGFNCDGK
    LGNESVTFYL QNLYVNQTDI YFCKIEVMYP PPYLDNEKSN GTIIHVKGKH
    LCPSPLFPGP SKPFWVLVVV GGVLACYSLL VTVAFIIFWV RSKRSRLLHS
    DYMNMTPRRP GPTRKHYQPY APPRDFAAYR SKKVAKKPTN KAPHPKQEPQ
    EINFPDDLPG SNTAAPVQET LHGCQPVTQE DGKESRISVQ ERQ
    SEQ ID NO: 124
    vector clone: pIB1103
    Sequence: MYGKIIFVLL LSEIVSISAE QKLISEEDLS STTGVAMHTS TSSSVTKSYI
    SSQTNDTHKR DTYAATPRAH EVSEISVRTV YPPEEETGER VQLAHHFSEP
    EITLIIFGVM AGVIGTILLI SYGRSKRSRL LHSDYMNMTP RRPGPTRKHY
    QPYAPPRDFA AYRSKKVAKK PTNKAPHPKQ EPQEINFPDD LPGSNTAAPV
    QETLHGCQPV TQEDGKESRI SVQERQ
    SEQ ID NO: 125
    vector clone: pIB1104
    Sequence: MYGKIIFVLL LSEIVSISAE QKLISEEDLI TLIIFGVMAG VIGTILLISY
    GRSKRSRLLH SDYMNMTPRR PGPTRKHYQP YAPPRDFAAY RSKKVAKKPT
    NKAPHPKQEP QEINFPDDLP GSNTAAPVQE TLHGCQPVTQ EDGKESRISV
    QERQ
    SEQ ID NO: 126
    vector clone: pIB1105
    Sequence: MDHLGASLWP QVGSLCLLLA GAAWEQKLIS EEDLAPPPNL PDPKFESKAA
    LLAARGPEEL LCFTERLEDL VCFWEEAASA GVGPGNYSFS YQLEDEPWKL
    CRLHQAPTAR GAVRFWCSLP TADTSSFVPL ELRVTAASGA PRYHRVIHIN
    EVVLLDAPVG LVARLADESG HVVLRWLPPP ETPMTSHIRY EVDVSAGNGA
    GSVQRVEILE GRTECVLSNL RGRTRYTFAV RARMAEPSFG GFWSAWSEPV
    SLLTPSDLDP LILTLSLILV VILVLLTVLA LLSRSKRSRL LHSDYMNMTP
    RRPGPTRKHY QPYAPPRDFA AYRSKKVAKK PTNKAPHPKQ EPQEINFPDD
    LPGSNTAAPV QETLHGCQPV TQEDGKESRI SVQERQ
    SEQ ID NO: 127
    vector clone: pIB1106
    Sequence: MDHLGASLWP QVGSLCLLLA GAAWEQKLIS EEDLLILTLS LILVVILVLL
    TVLALLSRSK RSRLLHSDYM NMTPRRPGPT RKHYQPYAPP RDFAAYRSKK
    VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT AAPVQETLHG CQPVTQEDGK
    ESRISVQERQ
    SEQ ID NO: 128
    vector clone: pIB1107
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLQDVSL LASDSEPLKC
    FSRTFEDLTC FWDEEEAAPS GTYQLLYAYP REKPRACPLS SQSMPHFGTR
    YVCQFPDQEE VRLFFPLHLW VKNVFLNQTR TQRVLFVDSV GLPAPPSIIK
    AMGGSQPGEL QISWEEPAPE ISDFLRYELR YGPRDPKNST GPTVIQLIAT
    ETCCPALQRP HSASALDQSP CAQPTMPWQD GPKQTSPSRE ASALTAEGGS
    CLISGLQPGN SYWLQLRSEP DGISLGGSWG SWSLPVTVDL PGDAVALGLQ
    CFTLDLKNVT CQWQQQDHAS SQGFFYHSRA RCCPRDRYPI WENCEEEEKT
    NPGLQTPQFS RCHFKSRNDS IIHILVEVTT APGTVHSYLG SPFWIHQAVR
    LPTPNLHWRE ISSGHLELEW QHPSSWAAQE TCYQLRYTGE GHQDWKVLEP
    PLGARGGTLE LRPRSRYRLQ LRARLNGPTY QGPWSSWSDP TRVETATETA
    WISLVTALHL VLGLSAVLGL LLLRWRSKRS RLLHSDYMNM TPRRPGPTRK
    HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP DDLPGSNTAA
    PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 129
    vector clone: pIB1108
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLISLVT ALHLVLGLSA
    VLGLLLLRWR SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS
    KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
    SEQ ID NO: 130
    vector clone: pIB1109
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLQDVSL LASDSEPLKC
    FSRTFEDLTC FWDEEEAAPS GTYQLLYAYP REKPRACPLS SQSMPHFGTR
    YVCQFPDQEE VRLFFPLHLW VKNVFLNQTR TQRVLFVDSV GLPAPPSIIK
    AMGGSQPGEL QISWEEPAPE ISDFLRYELR YGPRDPKNST GPTVIQLIAT
    ETCCPALQRP HSASALDQSP CAQPTMPWQD GPKQTSPSRE ASALTAEGGS
    CLISGLQPGN SYWLQLRSEP DGISLGGSWG SWSLPVTVDL PGDAVALGLQ
    CFTLDLKNVT CQWQQQDHAS SQGFFYHSRA RCCPRDRYPI WENCEEEEKT
    NPGLQTPQFS RCHFKSRNDS IIHILVEVTT APGTVHSYLG SPFWIHQAVR
    LPTPNLHWRE ISSGHLELEW QHPSSWAAQE TCYQLRYTGE GHQDWKVLEP
    PLGARGGTLE LRPRSRYRLQ LRARLNGPTY QGPWSSWSDP TRVETATETA
    WISLVTALHL VLGLNAVLGL LLLRWRSKRS RLLHSDYMNM TPRRPGPTRK
    HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP DDLPGSNTAA
    PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 131
    vector clone: pIB1110
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLISLVT ALHLVLGLNA
    VLGLLLLRWR SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS
    KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
    SEQ ID NO: 132
    vector clone: pIB1111
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLQDVSL LASDSEPLKC
    FSRTFEDLTC FWDEEEAAPS GTYQLLYAYP REKPRACPLS SQSMPHFGTR
    YVCQFPDQEE VRLFFPLHLW VKNVFLNQTR TQRVLFVDSV GLPAPPSIIK
    AMGGSQPGEL QISWEEPAPE ISDFLRYELR YGPRDPKNST GPTVIQLIAT
    ETCCPALQRP HSASALDQSP CAQPTMPWQD GPKQTSPSRE ASALTAEGGS
    CLISGLQPGN SYWLQLRSEP DGISLGGSWG SWSLPVTVDL PGDAVALGLQ
    CFTLDLKNVT CQWQQQDHAS SQGFFYHSRA RCCPRDRYPI WENCEEEEKT
    NPGLQTPQFS RCHFKSRNDS IIHILVEVTT APGTVHSYLG SPFWIHQAVR
    LPTPNLHWRE ISSGHLELEW QHPSSWAAQE TCYQLRYTGE GHQDWKVLEP
    PLGARGGTLE LRPRSRYRLQ LRARLNGPTY QGPWSSWSDP TRVETATETA
    WISLVTALHL VLGLSAVLGL LLLRKRSKRS RLLHSDYMNM TPRRPGPTRK
    HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP DDLPGSNTAA
    PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 133
    vector clone: pIB1112
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLISLVT ALHLVLGLSA
    VLGLLLLRKR SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS
    KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
    SEQ ID NO: 134
    vector clone: pIB1113
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLQDVSL LASDSEPLKC
    FSRTFEDLTC FWDEEEAAPS GTYQLLYAYP REKPRACPLS SQSMPHFGTR
    YVCQFPDQEE VRLFFPLHLW VKNVFLNQTR TQRVLFVDSV GLPAPPSIIK
    AMGGSQPGEL QISWEEPAPE ISDFLRYELR YGPRDPKNST GPTVIQLIAT
    ETCCPALQRP HSASALDQSP CAQPTMPWQD GPKQTSPSRE ASALTAEGGS
    CLISGLQPGN SYWLQLRSEP DGISLGGSWG SWSLPVTVDL PGDAVALGLQ
    CFTLDLKNVT CQWQQQDHAS SQGFFYHSRA RCCPRDRYPI WENCEEEEKT
    NPGLQTPQFS RCHFKSRNDS IIHILVEVTT APGTVHSYLG SPFWIHQAVR
    LPTPNLHWRE ISSGHLELEW QHPSSWAAQE TCYQLRYTGE GHQDWKVLEP
    PLGARGGTLE LRPRSRYRLQ LRARLNGPTY QGPWSSWSDP TRVETATETA
    WISLVTALLL VLGLSAVLGL LLLRWRSKRS RLLHSDYMNM TPRRPGPTRK
    HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP DDLPGSNTAA
    PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 135
    vector clone: pIB1114
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLISLVT ALLLVLGLSA
    VLGLLLLRWR SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS
    KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
    SEQ ID NO: 136
    vector clone: pIB1115
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLQDVSL LASDSEPLKC
    FSRTFEDLTC FWDEEEAAPS GTYQLLYAYP REKPRACPLS SQSMPHFGTR
    YVCQFPDQEE VRLFFPLHLW VKNVFLNQTR TQRVLFVDSV GLPAPPSIIK
    AMGGSQPGEL QISWEEPAPE ISDFLRYELR YGPRDPKNST GPTVIQLIAT
    ETCCPALQRP HSASALDQSP CAQPTMPWQD GPKQTSPSRE ASALTAEGGS
    CLISGLQPGN SYWLQLRSEP DGISLGGSWG SWSLPVTVDL PGDAVALGLQ
    CFTLDLKNVT CQWQQQDHAS SQGFFYHSRA RCCPRDRYPI WENCEEEEKT
    NPGLQTPQFS RCHFKSRNDS IIHILVEVTT APGTVHSYLG SPFWIHQAVR
    LPTPNLHWRE ISSGHLELEW QHPSSWAAQE TCYQLRYTGE GHQDWKVLEP
    PLGARGGTLE LRPRSRYRLQ LRARLNGPTY QGPWSSWSDP TRVETATETA
    WISLVTALHL VLGLNAVLGL LLLRKRSKRS RLLHSDYMNM TPRRPGPTRK
    HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP DDLPGSNTAA
    PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 137
    vector clone: pIB1116
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLISLVT ALHLVLGLNA
    VLGLLLLRKR SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS
    KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
    SEQ ID NO: 138
    vector clone: pIB1117
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLQDVSL LASDSEPLKC
    FSRTFEDLTC FWDEEEAAPS GTYQLLYAYP REKPRACPLS SQSMPHFGTR
    YVCQFPDQEE VRLFFPLHLW VKNVFLNQTR TQRVLFVDSV GLPAPPSIIK
    AMGGSQPGEL QISWEEPAPE ISDFLRYELR YGPRDPKNST GPTVIQLIAT
    ETCCPALQRP HSASALDQSP CAQPTMPWQD GPKQTSPSRE ASALTAEGGS
    CLISGLQPGN SYWLQLRSEP DGISLGGSWG SWSLPVTVDL PGDAVALGLQ
    CFTLDLKNVT CQWQQQDHAS SQGFFYHSRA RCCPRDRYPI WENCEEEEKT
    NPGLQTPQFS RCHFKSRNDS IIHILVEVTT APGTVHSYLG SPFWIHQAVR
    LPTPNLHWRE ISSGHLELEW QHPSSWAAQE TCYQLRYTGE GHQDWKVLEP
    PLGARGGTLE LRPRSRYRLQ LRARLNGPTY QGPWSSWSDP TRVETATETA
    WISLVTALYL VLGLNAVLGL LLLRWRSKRS RLLHSDYMNM TPRRPGPTRK
    HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP DDLPGSNTAA
    PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 139
    vector clone: pIB1118
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLISLVT ALYLVLGLNA
    VLGLLLLRWR SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS
    KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
    SEQ ID NO: 140
    vector clone: pIB1119
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLQDVSL LASDSEPLKC
    FSRTFEDLTC FWDEEEAAPS GTYQLLYAYP REKPRACPLS SQSMPHFGTR
    YVCQFPDQEE VRLFFPLHLW VKNVFLNQTR TQRVLFVDSV GLPAPPSIIK
    AMGGSQPGEL QISWEEPAPE ISDFLRYELR YGPRDPKNST GPTVIQLIAT
    ETCCPALQRP HSASALDQSP CAQPTMPWQD GPKQTSPSRE ASALTAEGGS
    CLISGLQPGN SYWLQLRSEP DGISLGGSWG SWSLPVTVDL PGDAVALGLQ
    CFTLDLKNVT CQWQQQDHAS SQGFFYHSRA RCCPRDRYPI WENCEEEEKT
    NPGLQTPQFS RCHFKSRNDS IIHILVEVTT APGTVHSYLG SPFWIHQAVR
    LPTPNLHWRE ISSGHLELEW QHPSSWAAQE TCYQLRYTGE GHQDWKVLEP
    PLGARGGTLE LRPRSRYRLQ LRARLNGPTY QGPWSSWSDP TRVETATETA
    WISLVTAWCL VLGLSAVLGL LLLRWRSKRS RLLHSDYMNM TPRRPGPTRK
    HYQPYAPPRD FAAYRSKKVA KKPTNKAPHP KQEPQEINFP DDLPGSNTAA
    PVQETLHGCQ PVTQEDGKES RISVQERQ
    SEQ ID NO: 141
    vector clone: pIB1120
    Sequence: MPSWALFMVT SCLLLAPQNL AQVSSEQKLI SEEDLISLVT AWCLVLGLSA
    VLGLLLLRWR SKRSRLLHSD YMNMTPRRPG PTRKHYQPYA PPRDFAAYRS
    KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
    SEQ ID NO: 142
    vector clone: pIB1179
    Sequence: MALPVTALLL PLALLLHAAR PEPKSCDKTH TCPPCPAPEL LGGPSVFLFP
    PKPKDTLMIS RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE
    QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR
    EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT
    PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS
    PGKAAAFWVL VVVGGVLACY SLLVTVAFII FWVRSKRSRL LHSDYMNMTP
    RRPGPTRKHY QPYAPPRDFA AYRSKKVAKK PTNKAPHPKQ EPQEINFPDD
    LPGSNTAAPV QETLHGCQPV TQEDGKESRI SVQERQ
    SEQ ID NO: 143
    vector clone: pIB1180
    Sequence: MALPVTALLL PLALLLHAAR PAEPKSPDKT HTCPPCPAPP VAGPSVFLFP
    PKPKDTLMIA RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE
    QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR
    EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT
    PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS
    PGKKDPKFWV LVVVGGVLAC YSLLVTVAFIIFWVRSKRSR LLHSDYMNMT
    PRRPGPTRKH YQPYAPPRDF AAYRSKKVAK KPTNKAPHPK QEPQEINFPD
    DLPGSNTAAP VQETLHGCQP VTQEDGKESR ISVQERQ
    SEQ ID NO: 144
    vector clone: pIB1181
    Sequence: MALPVTALLL PLALLLHAAR PERKCCVECP PCPAPPVAGP SVFLFPPKPK
    DTLMISRTPE VTCVVVDVSH EDPEVQFNWY VDGVEVHNAK TKPREEQFNS
    TFRVVSVLTV VHQDWLNGKE YKCKVSNKGL PAPIEKTISK TKGQPREPQV
    YTLPPSREEM TKNQVSLTCL VKGFYPSDIS VEWESNGQPE NNYKTTPPML
    DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVM HEALHNHYTQ KSLSLSPGKF
    WVLVVVGGVL ACYSLLVTVA FIIFWVRSKR SRLLHSDYMN MTPRRPGPTR
    KHYQPYAPPR DFAAYRSKKV AKKPTNKAPH PKQEPQEINF PDDLPGSNTA
    APVQETLHGC QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 145
    vector clone: pIB1182
    Sequence: MALPVTALLL PLALLLHAAR PELKTPLGDT THTCPRCPEP KSCDTPPPCP
    RCPEPKSCDT PPPCPRCPEP KSCDTPPPCP RCPAPELLGG PSVFLFPPKP
    KDTLMISRTP EVTCVVVDVS HEDPEVQFKW YVDGVEVHNA KTKPREEQYN
    STFRVVSVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS KTKGQPREPQ
    VYTLPPSREE MTKNQVSLTC LVKGFYPSDI AVEWESSGQP ENNYNTTPPM
    LDSDGSFFLY SKLTVDKSRW QQGNIFSCSV MHEALHNRFT QKSLSLSPGK
    FWVLVVVGGV LACYSLLVTV AFIIFWVRSK RSRLLHSDYM NMTPRRPGPT
    RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT
    AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 146
    vector clone: pIB1183
    Sequence: MALPVTALLL PLALLLHAAR PESKYGPPCP SCPAPEFLGG PSVFLFPPKP
    KDTLMISRTP EVTCVVVDVS QEDPEVQFNW YVDGVEVHNA KTKPREEQFN
    STYRVVSVLT VLHQDWLNGK EYKCKVSNKG LPSSIEKTIS KAKGQPREPQ
    VYTLPPSQEE MTKNQVSLTC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV
    LDSDGSFFLY SRLTVDKSRW QEGNVFSCSV MHEALHNHYT QKSLSLSLGK
    FWVLVVVGGV LACYSLLVTV AFIIFWVRSK RSRLLHSDYM NMTPRRPGPT
    RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT
    AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 147
    vector clone: pIB1184
    Sequence: MALPVTALLL PLALLLHAAR PESKYGPPCP PCPAPEFEGG PSVFLFPPKP
    KDTLMISRTP EVTCVVVDVS QEDPEVQFNW YVDGVEVHNA KTKPREEQFQ
    STYRVVSVLT VLHQDWLNGK EYKCKVSNKG LPSSIEKTIS KAKGQPREPQ
    VYTLPPSQEE MTKNQVSLTC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV
    LDSDGSFFLY SRLTVDKSRW QEGNVFSCSV MHEALHNHYT QKSLSLSLGK
    FWVLVVVGGV LACYSLLVTV AFIIFWVRSK RSRLLHSDYM NMTPRRPGPT
    RKHYQPYAPP RDFAAYRSKK VAKKPTNKAP HPKQEPQEIN FPDDLPGSNT
    AAPVQETLHG CQPVTQEDGK ESRISVQERQ
    SEQ ID NO: 148
    vector clone: pIB1185
    Sequence: MALPVTALLL PLALLLHAAR PEPKSCDKTH TCPPCPAPEL LGGPSVFLFP
    PKPKDTLMIS RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE
    QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR
    EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT
    PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS
    PGKAAAIEVM YPPPYLDNEK SNGTIIHVKG KHLCPSPLFP GPSKPFWVLV
    VVGGVLACYS LLVTVAFIIF WVRSKRSRLL HSDYMNMTPR RPGPTRKHYQ
    PYAPPRDFAA YRSKKVAKKP TNKAPHPKQE PQEINFPDDL PGSNTAAPVQ
    ETLHGCQPVT QEDGKESRIS VQERQ
    SEQ ID NO: 149
    vector clone: pIB1186
    Sequence: MALPVTALLL PLALLLHAAR PAEPKSPDKT HTCPPCPAPP VAGPSVFLFP
    PKPKDTLMIA RTPEVTCVVV DVSHEDPEVK FNWYVDGVEV HNAKTKPREE
    QYNSTYRVVS VLTVLHQDWL NGKEYKCKVS NKALPAPIEK TISKAKGQPR
    EPQVYTLPPS RDELTKNQVS LTCLVKGFYP SDIAVEWESN GQPENNYKTT
    PPVLDSDGSF FLYSKLTVDK SRWQQGNVFS CSVMHEALHN HYTQKSLSLS
    PGKKDPKIEV MYPPPYLDNE KSNGTIIHVK GKHLCPSPLF PGPSKPFWVL
    VVVGGVLACY SLLVTVAFII FWVRSKRSRL LHSDYMNMTP RRPGPTRKHY
    QPYAPPRDFA AYRSKKVAKK PTNKAPHPKQ EPQEINFPDD LPGSNTAAPV
    QETLHGCQPV TQEDGKESRI SVQERQ
    SEQ ID NO: 150
    vector clone: pIB1187
    Sequence: MALPVTALLL PLALLLHAAR PERKCCVECP PCPAPPVAGP SVFLFPPKPK
    DTLMISRTPE VTCVVVDVSH EDPEVQFNWY VDGVEVHNAK TKPREEQFNS
    TFRVVSVLTV VHQDWLNGKE YKCKVSNKGL PAPIEKTISK TKGQPREPQV
    YTLPPSREEM TKNQVSLTCL VKGFYPSDIS VEWESNGQPE NNYKTTPPML
    DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVM HEALHNHYTQ KSLSLSPGKI
    EVMYPPPYLD NEKSNGTIIH VKGKHLCPSP LFPGPSKPFW VLVVVGGVLA
    CYSLLVTVAF IIFWVRSKRS RLLHSDYMNM TPRRPGPTRK HYQPYAPPRD
    FAAYRSKKVA KKPTNKAPHP KQEPQEINFP DDLPGSNTAA PVQETLHGCQ
    PVTQEDGKES RISVQERQ
    SEQ ID NO: 151
    vector clone: pIB1188
    Sequence: MALPVTALLL PLALLLHAAR PELKTPLGDT THTCPRCPEP KSCDTPPPCP
    RCPEPKSCDT PPPCPRCPEP KSCDTPPPCP RCPAPELLGG PSVFLFPPKP
    KDTLMISRTP EVTCVVVDVS HEDPEVQFKW YVDGVEVHNA KTKPREEQYN
    STFRVVSVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS KTKGQPREPQ
    VYTLPPSREE MTKNQVSLTC LVKGFYPSDI AVEWESSGQP ENNYNTTPPM
    LDSDGSFFLY SKLTVDKSRW QQGNIFSCSV MHEALHNRFT QKSLSLSPGK
    IEVMYPPPYL DNEKSNGTII HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL
    ACYSLLVTVA FIIFWVRSKR SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR
    DFAAYRSKKV AKKPTNKAPH PKQEPQEINF PDDLPGSNTA APVQETLHGC
    QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 152
    vector clone: pIB1189
    Sequence: MALPVTALLL PLALLLHAAR PESKYGPPCP SCPAPEFLGG PSVFLFPPKP
    KDTLMISRTP EVTCVVVDVS QEDPEVQFNW YVDGVEVHNA KTKPREEQFN
    STYRVVSVLT VLHQDWLNGK EYKCKVSNKG LPSSIEKTIS KAKGQPREPQ
    VYTLPPSQEE MTKNQVSLTC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV
    LDSDGSFFLY SRLTVDKSRW QEGNVFSCSV MHEALHNHYT QKSLSLSLGK
    IEVMYPPPYL DNEKSNGTII HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL
    ACYSLLVTVA FIIFWVRSKR SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR
    DFAAYRSKKV AKKPTNKAPH PKQEPQEINF PDDLPGSNTA APVQETLHGC
    QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 153
    vector clone: pIB1190
    Sequence: MALPVTALLL PLALLLHAAR PESKYGPPCP PCPAPEFEGG PSVFLFPPKP
    KDTLMISRTP EVTCVVVDVS QEDPEVQFNW YVDGVEVHNA KTKPREEQFQ
    STYRVVSVLT VLHQDWLNGK EYKCKVSNKG LPSSIEKTIS KAKGQPREPQ
    VYTLPPSQEE MTKNQVSLTC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV
    LDSDGSFFLY SRLTVDKSRW QEGNVFSCSV MHEALHNHYT QKSLSLSLGK
    IEVMYPPPYL DNEKSNGTII HVKGKHLCPS PLFPGPSKPF WVLVVVGGVL
    ACYSLLVTVA FIIFWVRSKR SRLLHSDYMN MTPRRPGPTR KHYQPYAPPR
    DFAAYRSKKV AKKPTNKAPH PKQEPQEINF PDDLPGSNTA APVQETLHGC
    QPVTQEDGKE SRISVQERQ
    SEQ ID NO: 154
    designation CD40
    Sequence KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
    SEQ ID NO: 155
    designation CD40_tandem
    Sequence KKVAKKPTNK APHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQKKVAKKPT NKAPHPKQEP QEINFPDDLP GSNTAAPVQE
    TLHGCQPVTQ EDGKESRISV QERQ
    SEQ ID NO: 156
    designation CD40_P227A
    Sequence KKVAKKPTNK AAHPKQEPQE INFPDDLPGS NTAAPVQETL HGCQPVTQED
    GKESRISVQE RQ
  • In some embodiments, any one or more of the arrangements below are contemplated:
      • 1. An engineered protein that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 166, and wherein the sequence is not SEQ ID NO: 123.
      • 2. An engineered protein that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 167, and wherein the sequence is not SEQ ID NO: 123.
      • 3. The engineered protein of any one of arrangements 1 or 2, further comprising a binding domain, CD28 domain, and CD40 domain.
      • 4. The engineered protein of any one of arrangements 2-3, further comprising a signal peptide sequence.
      • 5. The engineered protein of arrangement 4, wherein the signal peptide sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 157.
      • 6. The engineered protein of any one of arrangements 3-5, wherein the binding domain comprises a VL sequence, a VH sequence, and an at least one linker.
      • 7. The engineered protein of arrangement 6, wherein the at least one linker has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 159 or 161.
      • 8. The engineered protein of any one of arrangements 6-7, wherein the binding domain comprises two linker sequences.
      • 9. The engineered protein of arrangement 8, wherein the two linker sequences have at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to amino acid sequences SEQ ID NO: 159 and SEQ ID NO: 161, respectively.
      • 10. The engineered protein of any one of arrangements 6-9, wherein the VL sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 158.
      • 11. The engineered protein of any one of arrangements 6-10, wherein the VH sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 160.
      • 12. The engineered protein of any one of arrangements 3-11, wherein the CD40 domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 165.
      • 13. The engineered protein of any one of arrangements 3-12, wherein the CD28 domain comprises a CD28 transmembrane domain.
      • 14. The engineered protein of arrangement 13, wherein the CD28 transmembrane domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 163.
      • 15. The engineered protein of any one of arrangements 3-14, wherein the CD28 domain comprises a CD28 extracellular domain.
      • 16. The engineered protein of arrangement 15, wherein the CD28 extracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 162.
      • 17. The engineered protein of any one of arrangements 3-16, wherein the CD28 domain comprises a CD28 intracellular domain.
      • 18. The engineered protein of arrangement 17, wherein the CD28 intracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 164.
      • 19. The engineered protein of any one of arrangements 1-18, wherein the protein further comprises 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of:
  • (SEQ ID NO: 168)
    QASQSLSNLLA,  
    (SEQ ID NO: 169)
    GASNLES,
    (SEQ ID NO: 170)
    QGGHYSGL,
    (SEQ ID NO: 171)
    TNDMN,
    (SEQ ID NO: 172)
    VIYSDDTPDYATWAKG,
    and/or
    (SEQ ID NO: 173)
    GHYDSAVYAYALNI.
      • 20. An engineered protein comprising an amino acid sequence that is at least 80% identical to the amino acid sequence of SEQ ID NO: 166 or 167, wherein the amino acid sequence does not include at least one of: QKLISEEDLE (SEQ ID NO: 174) or
  • (SEQ ID NO: 175)
    LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYG
    NYSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI.
      • 21. The engineered protein of arrangement 20, wherein the engineered protein lacks both of QKLISEEDLE (SEQ ID NO: 174) and
  • (SEQ ID NO: 175)
    LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYG
    NYSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI.
      • 22. The engineered protein of either of arrangements 20 or 21, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 158.
      • 23. The engineered protein of either of arrangements 20 or 21, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 160.
      • 24. The engineered protein of either of arrangements 23, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 158.
      • 25. The engineered protein of any one of arrangements 20-24, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 162.
      • 26. The engineered protein of any one of arrangements 20-25, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 164.
      • 27. The engineered protein of any one of arrangements 20-26, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 165.
      • 28. The engineered protein of any one of arrangements 20-27, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 163.
      • 29. The engineered protein of any one of arrangements 20-28, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 161.
      • 30. The engineered protein of any one of arrangements 20-29, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 159.
      • 31. The engineered protein of any one of arrangements 20-30, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 157.
      • 32. The engineered protein of any one of arrangements 20-31, wherein SEQ ID NO: 166 or 167 comprises a sequence that is at least 80, 90, 95, 96, 97, 98, 99 or 100% identical to SEQ ID NO: 162.
      • 33. The engineered protein of any one of arrangements 20-32, wherein SEQ ID NO: 166 or 167 comprises 1, 2, 3, 4, 5, 6, 7, and/or all 8 sequence(s) that is/are at least 95% identical to a sequence selected from the group consisting of SEQ ID NOs: 158, 160, 162, 163, 164, and 165.
      • 34. The engineered protein of any one of arrangements 20-33, wherein SEQ ID NO: 166 or 167 comprises a sequence that is more than 98% identical to each of SEQ ID NOs: 158, 160, 162, 163, 164, and 165.
      • 35. A CoStAR comprising:
      • (a) an optional signal peptide;
      • (b) a binding domain, wherein the binding domain binds to an anti-pembrolizumab antibody or binding fragment thereof;
      • (c) a CD28 domain;
      • (d) a CD40 domain;
      • wherein a) is optionally linked to b), wherein b) is linked to c), wherein c) is linked to d), and wherein the CoStAR comprises an amino acid sequence that:
        • i) lacks at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQQLQV YSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175);
        • ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167;
        • iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is not SEQ ID NO: 123; or
        • iv) any combination of i-iv.
      • 36. A fusion protein comprising:
      • (a) a means for binding to an antibody that binds to pembrolizumab;
      • (b) a CD28 domain;
      • (c) a CD40 domain;
      • wherein a) is linked to b), wherein b) is linked to c), and wherein the fusion protein comprises an amino acid sequence that:
        • i) lacks at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQQLQV YSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175);
        • ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167;
        • iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is not SEQ ID NO: 123; or
        • iv) any combination of i-iv.
      • 37. The CoStAR or fusion protein of arrangements 35 or 36, wherein the binding domain or the means for binding to an antibody that binds to pembrolizumab comprises: 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDSAVYAYALNI (SEQ ID NO: 173).
      • 38. The CoStAR or fusion protein of one of arrangements 35-37, wherein the CD28 domain comprises: SEQ ID Nos: 162, 163, and 164, or a sequence that is at least 80% or 95, or 98% identical thereto.
      • 39. The CoStAR or fusion protein of one of arrangements 35-38, wherein the CD40 domain comprises: SEQ ID No: 165, or a sequence that is at least 80% or 95, or 98% identical thereto.
      • 40. A fusion protein comprising the amino acid sequence of SEQ ID NO: 166.
      • 41. A fusion protein comprising the amino acid sequence of SEQ ID NO: 167.
      • 42. A nucleic acid which encodes the protein of any one of the preceding arrangements.
      • 43. A vector which comprises the nucleic acid of any one of the preceding arrangements.
      • 44. A cell which expresses the protein of any one of the preceding arrangements.
      • 45. A cell which expresses at least two proteins of any one of the preceding arrangements.
      • 46. A method of making the cell of any one of arrangement 44 or 45, which comprises the step of transducing or transfecting a cell with a vector of arrangement 43.
      • 47. A method for preparing a population of cells that express a protein of any one of arrangements 1 to 41, comprising detecting expression of the protein on the surface of cells transfected or transduced with a vector according to arrangement 43 and selecting cells which are identified as expressing the protein.
      • 48. A cell population produced by the method of arrangement 47.
      • 49. A cell population which is enriched for cell expression a protein of any one of arrangements 1 to 41.
      • 50. A method for treating a disease in a subject in need thereof, which comprises the step of administering the cell of any one of arrangements 44-45 or the cell population of arrangement 48 or 49 to the subject.
      • 51. The engineered protein of any one of arrangements 1-41, wherein the binding domain and CD28 domain are connected by at least one linker.
  • Although the present invention and its advantages have been described in detail, it should be understood that various changes, substitutions and alterations can be made herein without departing from the spirit and scope of the invention as defined in the appended claims.
  • EXAMPLES
  • The present invention will be further illustrated in the following Examples which are given for illustration purposes only and are not intended to limit the invention in any way.
  • Examples 1
  • Coculture assay set up. T cells from 2 healthy donors were either modified to express the constructs tested or left non-transduced (NTD) at MOI 10. One day prior to coculture set up, effector T cells were thawed and resuspended at 1×106 cells/mL in T cell media (TCM) without IL2 and incubated overnight at 37° C. with 5% CO2. On the day of coculture, T cells (effectors) and Ba/F3-OKT3 targets were collected and counted using a ViCELL BLU as per manufacturer's instructions. T cells were then cocultured with B a/F3 OKT3 targets at the 10:1, 1:1 and 1:10 E:T (effector:target) ratios overnight. For the inducible costimulatory protein constructs tested (i.e., pIB1097 to pIB1102), an additional set of wells were setup to which 10 μg/mL pembrolizumab was added in addition to Ba/F3 OKT3 targets. Each condition was performed in duplicates. Unstimulated T cells served as negative controls. Two sets of each E:T ratio as well as the T cell only control plates were set up. Brefeldin A was added at 1:1000 dilution to one set of plates to assess cytokine production by intracellular cytokine staining (ICS) using a flow cytometer after overnight co-culture. The second set was incubated for 5 days following which T cell counts and activation marker expression (i.e., 41BB and CD69) was assessed by flow cytometry.
  • The invention includes modifying components of the TCR complex and associated signaling adaptors (such as, for example, in a TCR incorporated antigen agnostic receptor “TIAAR”), identifying transmembrane domains (TMDs) and modifications that enable constitutive activation of receptors (“constitutive”) and utilizing antibodies to induce activation of the receptor (“inducible”).
  • scFV targeting co-stimulatory or inhibitor receptors and ligands. The scFV are derived from antibodies targeting co-stimulatory or inhibitory molecules expressed on immune cells.
  • TABLE 9
    Signal peptide Tag ECD_TMD ICD Costim GOI description
    CD8a Myc hZ270_HL NA CD28-CD40 anti-NKG2A blocking
    CD8a Myc hZ270_LH NA CD28-CD40 anti-NKG2A blocking
    CD8a Myc Varlilumab_HL NA CD28-CD40 anti-CD27 agonist
    CD8a Myc Varlilumab_LH NA CD28-CD40 anti-CD27 agonist
    CD8a Myc Urelumab_HL NA CD28-CD40 anti-CD137 agonist
    CD8a Myc Urelumab_LH NA CD28-CD40 anti-CD137 agonist
    CD8a Myc TRX518_HL NA CD28-CD40 anti-GITR agonist
    CD8a Myc TRX518 LH NA CD28-CD40 anti-GITR agonist
    CD8a Myc Pembroluzimab_HL NA CD28-CD40 anti-PD1 blocking
    CD8a Myc Pembroluzimab_LH NA CD28-CD40 anti-PD1 blocking
    CD8a Myc Atezolizumab_HL NA CD28-CD40 anti-PDL1 blocking
    CD8a Myc Atezolizumab_LH NA CD28-CD40 anti-PDL1 blocking
    CD8a Myc RONK203_HL NA CD28-CD40 anti-FasL blocking
    CD8a Myc RONK203_LH NA CD28-CD40 anti-FasL blocking
    CD8a Myc Tavolimab_HL NA CD28-CD40 anti-OX40 agonist
    CD8a Myc Tavolimab_LH NA CD28-CD40 anti-OX40 agonist
    CD8a Myc Ipilimumab_HL NA CD28-CD40 anti-CTLA4 blocking
    CD8a Myc Ipilimumab_LH NA CD28-CD40 anti-CTLA4 blocking
    CD8a Myc KY1044_HL NA CD28-CD40 anti-ICOS agonist
    CD8a Myc KY1044_LH NA CD28-CD40 anti-ICOS agonist
    CD8a Myc APX005_HL NA CD28-CD40 anti-CD40 agonist
    CD8a Myc APX005_LH NA CD28-CD40 anti-CD40 agonist
    CD8a Myc Selicrelumab_HL NA CD28-CD40 anti-CD40 agonist
    CD8a Myc Selicrelumab_LH NA CD28-CD40 anti-CD40 agonist
  • TABLE 10
    TIAAR (TCR incorporated) list of constructs
    Signal ECD_ GOI
    Code Concept peptide Tag TMD ICD Costim description
    pIB1026 TIAAR CD3D Myc CD3D N/A CD28- CD3D_CD3D_
    CD40 CD28CD40
    pIB1027 TIAAR CD3E FLAG CD3E N/A CD28- CD3E_CD3E_
    CD40 CD28CD40
    pIB1028 TIAAR CD3G Myc CD3G N/A CD28- CD3G_CD3G_
    CD40 CD28CD40
    pIB1029 TIAAR CD3Z Myc IC CD3Z N/A CD28- CD3Z_CD3Z_
    CD40 CD28CD40_Myc
    pIB1030 TIAAR CD8A Myc hTRDC N/A CD28- CD8A_hTRDC_
    CD40 CD28CD40
    pIB1031 TIAAR CD8A FLAG hTRGC1 N/A CD28- CD8A_hTRGC1_
    CD40 CD28CD40
    pIB1032 TIAAR CD8A Myc mTRAC N/A CD28- CD8A_mTRAC_
    CD40 CD28CD40
    pIB1033 TIAAR CD8A FLAG mTRBC1 N/A CD28- CD8A_mTRBC1_
    CD40 CD28CD40
    pIB1046 TIAAR CD8A x2 Myc hTRDC_ N/A CD28- CD8A_hTRDC_
    and hTRGC1 CD40 CD28CD40-T2A-
    FLAG CD8a_hTRGC1_
    CD28CD40
    pIB1047 TIAAR CD8A x2 Myc mTRAC_ N/A CD28- CD8A_mTRAC_
    and mTRBC1 CD40 CD28CD40-T2A-
    FLAG CD8A_mTRBC1_
    CD28CD40
    pIB1048 TIAAR CD3D Myc CD3D_ N/A CD28- CD3D_CD3D_
    and and CD3E CD40 CD28CD40-T2A-
    CD3E FLAG CD3E_CD3E_
    CD28CD40
    pIB1049 TIAAR CD3G Myc CD3G_ N/A CD28- CD3G_CD3G_
    and and CD3E CD40 CD28CD40-T2A-
    CD3E FLAG CD3E_CD3E_
    CD28CD40
    pIB1050 TIAAR CD3D Myc CD3D_ CD3D CD28- CD3D_CD3D_
    and and CD3E CD3E CD40 CD3D
    CD3E FLAG (ICD)_
    CD28CD40-T2A-
    CD3E_CD3E_CD3E
    (ICD)_CD28CD40
    pIB1051 TIAAR CD3D Myc CD3D_ CD3D N/A CD3D_CD3D_
    and and CD3E CD3E CD3D ICD-
    CD3E FLAG T2A-CD3E_CD3E_
    CD3EICD
    pIB1052 TIAAR CD3D Myc CD3D_ N/A N/A CD3D_CD3D
    and and CD3E (control)-
    CD3E FLAG T2A-CD3E_CD3E
    (control)
    pIB1053 TIAAR CD3G Myc CD3G_ CD3G_ CD28- CD3G_CD3G_
    and and CD3E CD3E CD40 CD3G (ICD)_
    CD3E FLAG CD28CD40-T2A-
    CD3E_CD3E_
    CD3E (ICD)_
    CD28CD40
    pIB1054 TIAAR CD3G Myc CD3G_ CD3G_ N/A CD3G_CD3G_
    and and CD3E CD3E CD3G ICD-
    CD3E FLAG T2A-CD3E_CD3E_
    CD3E ICD
    pIB1055 TIAAR CD3G Myc CD3G_ N/A N/A CD3G_CD3G
    and and CD3E (control)-
    CD3E FLAG T2A-CD3E_
    CD3E (control)
    pIB1056 TIAAR CD3Z Myc IC CD3Z CD3Z CD28- CD3z_CD3z_
    CD40 CD3z ICD_
    CD28CD40_Myc
    pIB1057 TIAAR CD3Z Myc IC CD3Z CD3Z N/A CD3z_CD3z_
    CD3z ICD_Myc
    pIB1058 TIAAR CD3Z Myc IC CD3Z N/A N/A CD3z_CD3z
    (control)
    pIB1059 TIAAR CD3Z Myc IC CD3Z CD3Z CD28- CD3Z_CD3Z_CD3Z
    (x2) CD40 ICD (duplicating
    CD3z endodomain-
    6 ITAMs)_
    CD28CD40_Myc
    pIB1060 TIAAR CD3Z Myc IC CD3Z CD3Z N/A CD3Z_CD3Z_
    (x2) CD3Z ICD
    (duplicating CD3z
    endodomain-6
    ITAMs)_Myc
    pIB1061 TIAAR CD3Z Myc IC CD3Z CD3Z CD28- CD3Z_CD3Z_
    (x2) CD40 CD28CD40_
    (swaped) CD3Z (6 ITAMs)_
    Myc
    pIB1062 TIAAR CD3Z Myc IC CD3Z CD3Z CD28- CD3Z_CD3Z_
    CD40 CD28CD40
    (swaped) CD3Z ICD_Myc
    pIB1063 TIAAR CD80 Myc CD80 CD80 N/A CD80 (control)
    pIB1064 TIAAR no Myc IC N/A Lck N/A Lck (control)
    signal
    peptide
    pIB1065 TIAAR no Myc IC N/A Lck N/A Lck (Y505F)
    signal (Y505F) (control)
    peptide
    pIB1066 TIAAR CD80 Myc CD80 Lck N/A CD80_Lck
    pIB1067 TIAAR CD80 Myc CD80 Lck CD28- CD80_Lck_
    CD40 CD28CD40
    pIB1068 TIAAR CD80 Myc CD80 CD80_ N/A CD80_Lck
    Lck (Y505F)
    (Y505F)
    pIB1069 TIAAR CD80 Myc CD80 CD80_ CD28- CD80_Lck
    Lck CD40 (Y505F)_
    (Y505F) CD28CD40
    pIB1070 TIAAR CD8A Myc LAT LAT N/A LAT (control)
    pIB1071 TIAAR CD8A Myc LAT LAT CD28- LAT_
    CD40 C28CD40
    pIB1072 TIAAR CD4 Myc CD4 CD4 CD28- CD4 control
    CD40
    pIB1073 TIAAR CD4 Myc CD4 CD4 CD28- CD4_CD28_
    CD40 CD40
    pIB1074 TIAAR CD8A Myc CD8A CD8A N/A CD8 control
    and and and and
    CD8B FLAG CD8B CD8B
    pIB1075 TIAAR CD8A Myc CD8A CD8A CD28- CD8_CD28_
    and and and and CD40 CD40
    CD8B FLAG CD8B CD8B
  • Cytokine production (Bcl-xL, IL2, IFNgamma and TNFalpha) from genetically modified and non-transduced T cells (NTD) after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only) (FIG. 2 ). There was increased Bcl-xL, IL2, IFNgamma and TNFalpha production from genetically modified T cells as compared to NTD cells in Donor 1. There was an increase in IFNγ production and comparable or lower levels of Bcl-xL, IL2, IFNgamma and TNFalpha production in genetically modified as compared to NTD cells in Donor 2.
  • Proliferation (T cell counts from CD45+ (TIARR)) and activation marker expression (41BB from CD45+ and CD69 from CD45+) from genetically modified and non-transduced T cells (NTD) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only) (FIGS. 3A-3B). There was decreased T cell counts in genetically modified as compared to NTD cells in Donor 1. There was increased or similar 41BB and CD69 expression in genetically modified as compared to NTD cells in Donor 1. There was increased or similar 41BB and CD69 expression in genetically modified T cells as compared to NTD cells in Donor 1. There was increased T cell counts for 2 modifications tested, and a similar or decreased T cell counts for the remaining genetically modified T cells as compared to NTD cells in Donor 2. There was increased or comparable 41BB and CD169 expression in genetically modified as compared to NTD cells in Donor 2.
  • TABLE 11
    List of constitutive constructs
    Signal
    Code Concept peptide Tag ECD_TMD ICD Costim GOI description
    pIB1076 C-SAAR CD8A Myc LZ (cFos)_ N/A CD28- LZ (cFos)-EGFRTM/JMD-
    EGFR CD40 CD28-CD40
    pIB1077 C-SAAR CD8A Myc LZ (cFos)_ N/A CD28- LZ (cFos)-CD28TM-CD28-
    CD28 CD40 CD40
    pIB1078 C-SAAR CD8A Myc LZ (cJun)_ N/A CD28- LZ (cJun)-EGFRTM/JMD-
    EGFR CD40 CD28-CD40
    pIB1079 C-SAAR CD8A Myc LZ (cJun)_ N/A CD28- LZ (cJun)-CD28TM-CD28-
    CD28 CD40 CD40
    pIB1080 C-SAAR CD8A Myc LZ (c/EBP)_ N/A CD28- LZ (c/EBP)-EGFRTM/JMD-
    EGFR CD40 CD28-CD40
    pIB1081 C-SAAR CD8A Myc LZ (c/EBP)_ N/A CD28- LZ (c/EBP)-CD28TM-
    CD28 CD40 CD28-CD40
    pIB1103 C-SAAR GpA Myc GpA ECD_ N/A CD28- GpA ECD-TMD-CD28-CD40
    TMD CD40
    pIB1104 C-SAAR GpA Myc GpA TMD N/A CD28- GpA TMD-CD28-CD40
    CD40
    pIB1105 C-SAAR EPOR Myc EPOR ECD_ N/A CD28- EpoR ECD-TMD-CD28-
    TMD CD40 CD40
    pIB1106 C-SAAR EPOR Myc EPOR TMD N/A CD28- EpoR TMD-CD28-CD40
    CD40
    pIB1107 C-SAAR TPOR Myc TPOR ECD_ N/A CD28- TPO ECD-TPO (WT) TMD -
    TMD CD40 CD28-CD40
    pIB1108 C-SAAR TPOR Myc TPOR TMD N/A CD28- TPO (WT) TMD-CD28-
    CD40 CD40
    pIB1109 C-SAAR TPOR Myc TPOR ECD_ N/A CD28- TPO ECD-TPO (S505N)
    TMD (S505N) CD40 TMD -CD28-CD40
    pIB1110 C-SAAR TPOR Myc TPOR TMD N/A CD28- TPO (S505N) TMD -CD28-
    (S505N) CD40 CD40
    pIB1111 C-SAAR TPOR Myc TPOR ECD_ N/A CD28- TPO ECD-TPO (W515K)
    TMD(W515K) CD40 TMD-CD28-CD40
    pIB1112 C-SAAR TPOR Myc TPOR TMD N/A CD28- TPO (W515K) TMD -CD28-
    (W515K) CD40 CD40
    pIB1113 C-SAAR TPOR Myc TPOR ECD_ N/A CD28- TPO ECD-TP (H499L)
    TMD (H499L) CD40 TMD-CD28-CD40
    pIB1114 C-SAAR TPOR Myc TPOR TMD N/A CD28- TPO (H499L) TMD-CD28-
    (H499L) CD40 CD40
    pIB1115 C-SAAR TPOR Myc TPOR ECD_ N/A CD28- TPO ECD-TPO (S505N-
    TMD (S505N- CD40 W515K) TMD -CD28-CD40
    W515K)
    pIB1116 C-SAAR TPOR Myc TPOR TMD N/A CD28- TPO (S505N-W515K) TMD -
    (S505N- CD40 CD28-CD40
    W515K)
    pIB1117 C-SAAR TPOR Myc TPOR ECD_ N/A CD28- TPO ECD-TPO (H499Y-
    TMD CD40 S505N) TMD-CD28-CD40
    (H499Y-
    S505N)
    pIB1118 C-SAAR TPOR Myc TPOR TMD N/A CD28- TPO (H499Y-S505N) TMD -
    (H499Y- CD40 CD28-CD40
    S505N)
    pIB1119 C-SAAR TPOR Myc TPOR ECD_ N/A CD28- TPO ECD-TPO (L498W-
    TMD CD40 H499C) TMD -CD28-CD40
    (L498W-
    H499C)
    pIB1120 C-SAAR TPOR Myc TPOR TMD N/A CD28- TPO (L498W-H499C) TMD -
    (L498W- CD40 CD28-CD40
    H499C)
    pIB1025 C-SAAR CD8a Myc CD28 N/A CD28- CD28 TM_CD28_CD40
    CD40
    pIB1179 C-SAAR CD8a N/A IgGI28TM N/A CD28- IgG1(CH2CH3)-CD28(TM)-
    CD40 CD28(CoStim)-
    CD40(CoStim)
    pIB1180 C-SAAR CD8a N/A IgG1mut + N/A CD28- IgG1(CH2CH3,mutant)-
    CIM CD40 CD28(TM)-CD28(CoStim)-
    CD40(CoStim)
    pIB1181 C-SAAR CD8al N/A IgG2 + N/A CD28- IgG2(CH2CH3)-CD28(TM)-
    CD28TM CD40 CD28(CoStim)-
    CD40(CoStim)
    pIB1182 C-SAAR I N /A IgG3 + N/A CD28- IgG3(CH2CH3)-CD28(TM)-
    CD28TM CD40 CD28(CoStim)-
    CD40(CoStim)
    pIB1183 C-SICD8a CD8a N/A IgG4 + N/A CD28- IgG4(CH2CH3)-CD28(TM)-
    CD28TM CD40 CD28(CoStim)-
    CD40(CoStim)
    pIB1184 C-SAAR CDI/A N/A IgG4mut + N/A CD28- IgG4(CH2CH3,mutant)-
    CD28TM CD40 CD28I-CD28(CoStim)-
    CD40(CoStim)
    pIB1185 C-SAAR CD8a N/A IgG1 + CD28 N/A CD28- IgG1(CH2CH3)-
    stalk/TM CD40 CD28(Stalk + TM)-
    CD28(CoStim)-
    CD40(CoStim)
    pIB1186 C-SAAR CD8a N/A IgG1mut + N/A CD28- IgG1(CH2CH3,mutant)-
    CD28 stalk/ CD40 CD28(Stalk + TM)-
    TM CD28(CoStim)-
    CD40(CoStim)
    pIB1187 C-SAAR CD8a N/A IgG2 + CD28 N/A CD28- IgG2(CH2CH3)-
    stalk/TM CD40 CD28(Stalk + TM)-
    CD28(CoStim)-
    CD40(CoStim)
    pIB1188 C-SAAR CD8a N/A IgG3 + CD28 N/A CD28- IgG3(CH2CH3)-
    stalk/TM CD40 CD28(Stalk + TM)-
    CD28(CoStim)-
    CD40(CoStim)
    pIB1189 C-SAAR CD8a N/A IgG4 + CD28 N/A CD28- IgG4(CH2CH3)-
    stalk/TM CD40 CD28(Stalk + TM)-
    CD28(CoStim)-
    CD40(CoStim)
    pIB1190 C-SAAR CD8a N/A IgG4mut + N/A CD28- IgG4(CH2CH3,mutant)-
    CD28 stalk/ CD40 CD28(Stalk + TM)-
    TM CD28(CoStim)-
    CD40(CoStim)
  • Cytokine production (Bcl-xL, IL2, IFNgamma and TNFalpha) from genetically modified and non-transduced T cells (NTD) after overnight stimulation with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only) (FIG. 4 ). There was increased or comparable Bcl-xL, IL2, and TNFalpha production and comparable or lower levels of IFNgamma from genetically modified T cells as compared to NTD cells in Donor 1. There was an increase in IFNgamma and IL2 production and comparable or lower levels of Bcl-xL and TNFalpha production in genetically modified as compared to NTD cells in Donor 2.
  • Proliferation (T cell counts from CD45+(LZ)) and activation marker expression (41BB from CD45+ and CD69 from CD45+) from genetically modified and non-transduced T cells (NTD) after 5-day co-culture with either Ba/F3 OKT3 targets or left unstimulated (i.e., T cells only) (FIGS. 5A-5B). There was decreased T cell counts in genetically modified as compared to NTD cells in Donor 1. There was increased 41BB and CD69 expression in genetically modified T cells as compared to NTD cells in Donor 1. There was increased T cell counts as compared to NTD cells in Donor 2, an increase in 41BB expression in genetically modified T cells as compared to NTD cells, and similar or decreased expression of CD169 in genetically modified T cells as compared to NTD cells in Donor 2.
  • TABLE 12
    List of inducible constructs
    Signal
    Code Concept peptide Tag ECD_TMD ICD Costim GOI description
    pIB1082 Inducible EGFR Myc EGFR N/A CD28- WT EGFR ECD
    CD40 EGFRTM/JMD-CD28-CD40
    pIB1083 Inducible EGFR Myc EGFR N/A CD28- domain IV - EGFRTM/JMD-
    (domain IV) CD40 CD28-CD40
    pIB1084 Inducible EGFR Myc EGFR (623- N/A CD28- EGFRTM/JMD-CD28-CD40
    668) CD40 (control)
    pIB1085 Inducible Her2 Myc Her2 N/A CD28- Her 2 (Domain 1 to IV)-
    CD40 TMD/JMD-CD28-CD40
    pIB1086 Inducible Her2 Myc Her2 N/A CD28- Her 2 (Domain 1 to IV)-TMD
    (V659E) CD40 (V659E)/JMD-CD28-CD40
    pIB1087 Inducible Her2 Myc Her2 N/A CD28- Her 2 (Domain 1 to IV)-TMD
    (V660D) CD40 (G660D)/JMD-CD28-CD40
    pIB1088 Inducible Her2 Myc Her2 N/A CD28- Her 2 (Domain 1 to IV)-TMD
    (V660R) CD40 (G660R)/JMD-CD28-CD40
    pIB1089 Inducible Her2 Myc Her2 domain N/A CD28- Her 2 (Domain IV)-TMD/JMD-
    IV_TMD CD40 CD28-CD40
    pIB1090 Inducible Her2 Myc Her2 domain N/A CD28- Her 2 (Domain IV)-TMD
    IV_TMD CD40 (V659E)/JMD-CD28-CD40
    (V659E)
    pIB1091 Inducible Her2 Myc Her2 domain N/A CD28- Her 2 (Domain IV)-TMD
    IV_TMD CD40 (G660D)/JMD-CD28-CD40
    (G660D)
    pIB1092 Inducible Her2 Myc IV_TMD N/A CD28- Her 2 (Domain IV)-TMD
    Her2 domain CD40 (G660R)/JMD-CD28-CD40
    (G660R)
    pIB1093 Inducible Her2 Myc Her2 TMD N/A CD28- Her 2 TMD/JMD-CD28-CD40
    CD40
    pIB1094 Inducible Her2 Myc Her2 TMD N/A CD28- Her 2 TMD (V659E)/JMD-
    (V659E) CD40 CD28-CD40
    pIB1095 Inducible Her2 Myc Her2 TMD N/A CD28- Her 2 TMD (G660D)/JMD-
    (G660D) CD40 CD28-CD40
    pIB1096 Inducible Her2 Myc Her2 TMD N/A CD28- Her 2 TMD (G660R)/JMD-
    (G660R) CD40 CD28-CD40
    pIB1097 Inducible CD8A Myc A30514 N/A CD28- Anti-ID1 VH-VL (A30514-
    VH_VL CD40 pembrolizumab)-CD28TMD
    CD28-CD40
    pIB1098 Inducible CD8A Myc A30514 N/A CD28- Anti-ID1 VL-VH (A30514-
    VL_VH CD40 pembrolizumab)-CD28TMD
    CD28-CD40
    pIB1099 Inducible CD8A Myc A30523 N/A CD28- Anti-ID2 Vh-VL (A30523-
    VH_VL CD40 pembrolizumab)-CD28TMD
    CD28-CD40
    pIB1100 Inducible CD8A Myc A30523 N/A CD28- Anti-ID2 VL-Vh (A30523-
    VL_VH CD40 pembrolizumab)-CD28TMD
    CD28-CD40
    pIB1101 Inducible CD8A Myc A30633 N/A CD28- Anti-ID3 Vh-VL (A30633-
    VH_VL CD40 pembrolizumab)-CD28TMD
    CD28-CD40
    pIB1102 Inducible CD8A Myc A30633 N/A CD28- Anti-ID3 VL-VH (A30633-
    VL_VH CD40 pembrolizumab)-CD28TMD
    CD28-CD40
  • Cytokine production (Bcl-xL, IL2, IFNg and TNFa) from genetically modified and non-transduced T cells (NTD) after overnight stimulation with either Ba/F3 OKT3 targets or Ba/F3 OKT3 targets with 10 ug/mL pembrolizumab or left unstimulated (i.e., T cells only) (FIG. 6 ). There was increased or comparable Bcl-xL, IL2, IFNgamma and TNFalpha production from genetically modified T cells in the presence of Ba/F3 OKT3 and pembrolizumab as compared to conditions with Ba/F3 OKT3 stimulation alone and NTD cells in both Donor 1 and Donor 2.
  • Proliferation (T cell counts from CD45+ (Inducible)) and activation marker expression (41BB from CD45+ and CD69 from CD45+) from genetically modified and non-transduced T cells (NTD) after 5-day co-culture with either Ba/F3 OKT3 targets or Ba/F3 OKT3 targets with 10 ug/mL pembrolizumab or left unstimulated (i.e., T cells only) (FIGS. 7A-7B). There was increased, comparable and decreased T cell counts in genetically modified T cells as compared to NTD cells in Donor 1. There was increased or similar 41BB and CD169 expression in genetically modified T cells as compared to NTD cells in Donor 1. There was increased or comparable T cell counts in genetically modified T cells as compared to NTD cells in Donor 2. There was increased, similar and decreased 41BB and CD169 expression in genetically modified cells as compared to NTD cells in Donor 2. Increased T cell counts and activation marker expression in both donors were observed when the genetically modified T cells were stimulated with Ba/F3 OKT3 in the presence of pembrolizumab compared to conditions with Ba/F3 OKT3 stimulation alone as well as NTD cells.
  • Example 2 Use of Sequences in a Cell System for the Treatment of a Patient
  • An engineered protein having the sequence of SEQ ID NO: 166 will be transfected into a Tumor Infiltrating Lymphocyte (TIL) cell using standard procedures by incorporating vectors. This cell will then be used to generate a population of TIL cells expressing those proteins. The population will be derived through detecting expression of the protein on the surface of the cells transfected to express the two proteins, and selecting cells which are identified as expressing those proteins. Through this process, the population of cells will be enriched for those expressing the protein. Following enrichment, the TIL cells will be administered at a therapeutic amount to a patient as a therapeutic treatment for cancer.
  • Example 3
  • Transduction of CoStAR Constructs into TIL Cells
  • CoStAR constructs were transduced into TIL cells from tumor digests. The efficiency of CEA or FOLR expression on tumor digests at Day 1 is as shown in FIGS. 20-22 , and in Table 13.
  • TABLE 13
    % CEA from % FOLR1 % CEA+ %
    CD45- expression FOLR+
    Construct (FSC-H (FSC-H (CEA
    Tumor type Name vs CEA) vs FOLR1) vs FOLR1)
    CRC CRC 11959 71.7 4.53 2.65
    (metastatic)
    CRC CRC 11974 54.6 9.9 5.31
    (metastatic)
    NSCLC NSCLC- 3.88 15.3 1.56
    9332
    NSCLC NSCLC- 15.1 34.8 14.7
    9596
    Ovarian OV-9662 2.03 41.3 0.93
    Melanoma MEL-CC50 1.37 0.77 0.061
    Melanoma MEL- 1.26 4.53 0.25
    11909
    Melanoma MEL- 4.4 1.41 0.18
    17614
  • The efficiency of CEA or FOLR expression by Day 21 is as shown in FIGS. 12-14 . As can be shown in FIGS. 12-14 and 20-22 , the amount of CEA and FOLR expression was significantly higher by 21 days of treatment compared to 1 day of treatment. The transduction was performed using the following materials and methodology:
  • Methodology: Experimental Outline
  • On day 1, the tumor digest was thawed (no stimulation) in media 1. There were a total of 15 tumor digests, and comprised pancreatic, CRC, NSCLC, ovarian, melanoma, and cervical tumors (FIG. 18 ). 0.5-1e6 cells were seeded per condition (6 conditions). Phenotype was recorded were applicable. On days 3 and 4, cells were transduced in media 1 at MOI 10. On day 8, cells were given outgrowth feed, through the addition of 1 mL of media 2 with gentamycin/amphotericin and fresh IL2 (6000 U/mL). On day 10, static REP (G-REX 6) was performed; cells were stimulated with OKT3 (30 ng/mL)+IL2 (3000 U/mL)+irradiated feeders (1:200). On day 15, dynamic REP was performed; cells were transferred to G-REX 6M with 100 mL media 3. On day 18, cells were counted, split as needed, and/or removed 65 mL media and replenished with fresh media 3. On day 21, the REP was ended. This timeline is also schematically depicted in FIG. 19 .
  • Methodology: Media Preparation
  • TCM base media: GMP/TCM media+25 mM HEPES+25 μM 2-Mercaptoethanol
  • Media 1: TCM base media+10% FBS, 1× Gentamycin/Amphotericin (500× stock)+50 ug/mL vancomycin+3000 IU/mL IL2
  • Media 2: TCM base media+10% FBS, 2× Gentamicin/Amphotericin B (500× stock)+100 ug/mL vancomycin+6000 IU/mL IL2
  • Media 3: TCM base media+8% human AB serum, 3000 IU/mL IL2
  • Methodology: Detailed Protocol by Day
  • Day 1: Thaw frozen tumor digest
      • 1. Prepare media 1 as per table in excel sheet. Add 200 uL IL2 to 200 mL of media 1
      • 2. Label a 15 ML conical tube for each tumor digest vial
      • 3. Remove the tumor digest vials from LN, and immediately thaw in 37-degree water bath until small chunks remain.
      • 4. Sterilize the exterior and bring the vials to BSC.
      • 5. Add in 1 ml of pre-warmed media 1 very gently, drop by drop, to each vial.
      • 6. Transfer the total of 2 ml cell suspension from the vial using a 5 ml serological pipette, and add very gently, drop by drop, to the match labeled 15 ml conical tube which contains 2 ml of media 1.
      • 7. Add in 9 ml of pre-warmed media 1 very gently to the cell suspension in the 15 ml conical tube.
      • 8. Centrifuge at 400 g for 10 min.
      • 9. Aspirate off the supernatant carefully without touching the cell pellet.
      • 10. Based on the frozen cell number, resuspend the cells in 5 ml of media 1 gently
      • 11. Count using Vicell and record in excel sheet (1:10 dilution; 30 uL cell suspension in 270 uL media 1)
      • 12. Add 1 mL media and split into 6 conditions
      • 13. Resuspend cells in either 0.5 M/mL
      • 14. Seed 0.5 M/mL in a 24 well plate or 1 M/2 mL in 6 well plate
      • 15. Seed 6 wells for each tumor digest (pIB1322, pIB1324, CTP386.1, CTP387.1, CTP025 and NTD T cells)
      • 16. Add 10 uL/mL amphotericin B only for CRC samples
      • 17. Place plates in incubator
      • 18. Collect cells if available for checking for CEA and FOLR expression on tumor
  • Day 3: TIL Transduction During Outgrowth
      • 1. Prepare required media 1 and add 1 uL/mL IL2 (25 uL IL2 to 25 mL media 1)
      • 2. Dilute the needed volume of LVV in 0.1 mL for 24 well plate or 0.5 ml for 6 well plate with Media 1 in a separate vial for each sample, gently pipette to mix well and add into cell culture.
      • 3. Add equivalent volume of media 1 alone to the nontransduced well.
      • 4. Mix gently and place plate in the incubator.
  • Day 4: TIL Second Transduction
      • 1. Prepare required media 1 and add 1 uL/mL IL2 (35 uL IL2 to 35 mL media 1)
      • 2. Use the TVC and LVV volume calculation from day 3, dilute the needed volume of LVV in 0.1 ml with Media 1 in a separate vial for each sample
      • 3. Mix gently and place plate in the incubator.
  • Day 8: Outgrowth Feed
      • 1. Prepare media 2 as per table in the excel sheet. Add 200 uL IL2 to 100 mL media 2.
      • 2. Mix each well
      • 3. Add 1 ml of fresh Media 2 to each well for 6 well plate and 500 uL for 24 well plate
      • 4. Continue the culture to day 10
  • Day 10: Static REP
      • 1. Prepare media 3 as per table in the excel sheet
      • 2. Collect all conditions
      • 3. Count using Vicell and record in excel sheet
      • 4. Calculate required number of irPBMC feeders needed
        • a. Assuming 0.1e6 cells per condition based on cell counts below
        • b. At 1:200—will need 12e9 irradiated feeders total
        • c. Thaw feeders in media 3 and centrifuge at 300 g×5 mins
        • d. Count feeders using Vicell
      • 5. Prepare master mix of media 3+OKT3+IL2 as needed, using the ratios outline in Table 14 below.
  • TABLE 14
    Stock Per mL of Volume
    Reagent concentration media 4 needed
    Media 3 NA 1 mL 30 mL
    OKT3 (30 ng/mL 100 ug/mL NA 30 uL (100 ng/ml)
    or 0.3 uL/mL)
    IL2 3000 U/uL 1 uL 30 uL 
      • 6. Add required volumes in the following order to each well of a 6-well G-Rex
        • a. Add 30 mL media 4+OKT3+IL2 first to each well
        • b. Add irradiated feeders to the bottom of the well
        • c. Add TILs gently to the bottom of well
      • 7. Plate an extra well of irradiated feeders alone to monitor and phenotype (seeded in Grex as well)
  • Day 15: Dynamic REP
      • 1. Collect all cell culture, resuspend wells
      • 2. Count using Vicell and record in excel sheet
      • 3. Transfer 30 mL cell suspension to 6M G-REX and top to 100 ml (ie, add 60 mL media 3+10 mL to rinse the well)
      • 4. Add fresh IL2 (1 uL/mL for 3000 U/mL IL2). Add 100 uL IL2 for each condition
  • Day 18: Dynamic REP Maintenance
      • 1. Take 6M G-REX from incubator
      • 2. Resuspend cells in the wells
      • 3. Count using Vicell and record in excel sheet
      • 4. Either do a media change for cell counts<1e6 cells/mL or split conditions with cell counts>1e6 cells/mL
      • Notes:
        • a. Let G-Rex sit in Biosafety cabinet for 30 minutes if cells are in suspension or disturbed prior to removing media.
        • b. For media change, remove 60 mL media and replenish with 60 mL media 4+IL2
        • c. For splitting conditions, transfer 50 mL cell suspension to a second G-REX 6M and make up volume to 100 mL for all conditions.
      • 5. Top wells with media 4 to 100 ml and place in incubator.
  • Day 21: Harvest
      • 1. Harvest all cells.
      • 2. Centrifuge at 400 g×10 mins
      • 3. Resuspend in 50 mL media 3 and pass through 100 um cell strainer into a fresh 50 mL conical tube
      • 4. Count using Vicell and record in excel sheet
      • 5. Spin down cells at 400 g for 10 min and aspirate off the supernatant.
      • 6. Cryopreserve the cells with CryoStor CS10 and freeze in cryovials.
    Example 4 TIL Functional Screening
  • TIL cells expressing CoStAR constructs then underwent a screen to assess function. The results of this screen are as shown in FIGS. 17A-17B. From the screening, a population of cells were generated that were enriched for CEA and/or FOLR expression. The fraction of anti-CEA, anti-FOLR, and universal CoStAR cells with positive expression are as shown in FIGS. 23-27 . Of these TILS, the CD4/8 ratio was as shown in FIGS. 28A-28D. The methodology for the assay was as follows:
  • Methodology: Experiment Outline
  • On day 1, TIL cells were thawed. On day 2, CoStAR-modified TILS were sorted, then ran on fortessa. On day 4, the media change was completed, and the cells were stained and ran on fortessa. On day 6, the TCM media was changed to exclude IL2. On day 7, the co-culture and serial stimulation assay was set up. As can be seen in FIGS. 30A-30H, 31A-31H, and 32A-32H, the co-culture with autologous digest is inconclusive probably due to variability introduced in tumor reactivity of the TILs between different conditions as a result of sorting for CoStAR. Additionally, a higher level of IL2 production observed from both anti-CEA and Universal CoStAR modified TILs in the presence of signal 1+2 in 24-hour co-culture assay. Similar trends observed for IFNg and TNFa. On day 8, the supernatant was collected, and MSD was performed. Cells were then stained to assess the expression of activation markers. An outline of this time is as shown in FIG. 29 .
  • Methodology: TCM Media with IL2
  • To a bottle of 500 mL RPMI 1640, add: 50 mL FBS, 5 mL pen/strep, 5 mL HEPES, 500 uL 2-mercaptoethanol, and 1 uL/mL of IL-2 stock (3e6 U/mL). The final concentration of the media should be 3000 U/mL.
  • Methodology: Detailed Protocol by Day
  • Day 1: Thaw TILS
      • 1) Place vials in water bath
      • 2) Transfer to hood and add cell suspension to 9 mL TCM media
      • 3) Centrifuge at 400 g×5 mins
      • 4) Aspirate
      • 5) Add 5 mL media
      • 6) Count using Vicell
      • 7) Rest cells overnight at 1e6 cells/mL
  • Day 2: Sort CoStAR-modified TILS
      • 1. Collect cells
      • 2. Centrifuge at 400 g×5 mins
      • 3. Aspirate and resuspend in 5 mL media
      • 4. Count using Vicell
      • 5. Prepare buffer (10% FBS+2 mM EDTA), add following to 500 mL of RPMI 1640
        • 50 mL of fetal bovine serum
        • 2 mL of 0.5M EDTA
      • 6. Determine cell number, collect cells for LS columns (see “Pre- and Post-sort counts”).
      • 7. Centrifuge@400×g for 5 minutes, aspirate supernatant. Break pellet.
      • 8. Transfer to 15 mL conical tubes
      • 9. Incubate cells with antibodies as per table
        • For CTP205, add 1 uL sol FOLR Fc APC antibody per 1e6 cells
        • For pIB1322 and pIB1324, add 10 uL donkey anti-rabbit PE antibody per 1e6 cells
        • For CTP386.1 and 387.1, add 2.5 uL CEACAM5-Fc FITC antibody per 1e6 cells
      • 10. Mix well, incubate in the dark@4 C for 30 minutes.
      • 11. Wash cells by adding 1-2 mL buffer per 10e6 cells
      • 12. Centrifuge@400×g for 5 minutes. Aspirate supernatant.
      • 13. Add 5 mL buffer to wash.
      • 14. Centrifuge@400×g for 5 minutes. Aspirate supernatant.
      • 15. Resuspend in 80 uL of buffer 10e6 cells (see table)
      • 16. Add 20 uL of microbeads 10e6 cells (see table)
      • 17. Mix well, incubate for 15 minutes@4 C
      • 18. Wash cells by adding 1-2 mL buffer per 10e6 cells
      • 19. Centrifuge@400×g for 5 minutes. Aspirate supernatant.
      • 20. Resuspend up to 100e6 cells in 500 uL of buffer.
      • 21. Magnetic separation using LS column
        • a) Place LS column on MACS separator.
        • b) Rinse with 2 mL buffer. Use column immediately. Rinse column right before adding cell suspension.
        • c) Apply cell suspension to column
        • d) Wash column 3× with 2 mL of buffer. Collect unlabelled cells that pass through the column
        • e) Remove column form separator and place on appropriate tube.
        • f) Pipette 5 mL buffer onto column.
        • g) Immediately flush by firmly pushing plunger into column.
        • h) Centrifuge at 400 g×5 mins
        • i) Resuspend in 1 mL (or 5 mL, depending on pellet) TCM media+3000 U/mL IL2
        • j) Count on Vicell. (see Pre- and post-sort counts)
        • k) Add media to maintain cells at 1e6/mL and incubate flasks
  • Day 4: Complete Media Change
      • 1. Collect cells in tubes
      • 2. Centrifuge at 400 g×5 mins
      • 3. Aspirate
      • 4. Add TCM media with IL2
      • 5. Count using Vicell
      • 6. Add media to maintain cells at 1e6 cells/mL
      • 7. Collect 0.05 to 0.1e6 cells to run on fortessa-stain with L/D Near Far IR
  • Day 6: Change to Media without IL2
      • 8. Collect cells in tubes
      • 9. Centrifuge at 400 g×5 mins
      • 10. Aspirate
      • 11. Add TCM media WITHOUT IL2
      • 12. Count using Vicell
      • 13. Add TCM media WITHOUT IL2 media to maintain cells at 1e6 cells/mL
  • Day 7: Set Up Co-Culture
      • 1. Collect sorted TILs
      • 2. Centrifuge at 400 g×5 mins
      • 3. Aspirate and add 5 mL TCM media without IL2
      • 4. Count using Vicell
      • 5. Collect targets—K562 WT, OKT3, CEACAM5 and OKT3 CEACAM5 and OKT3 FOLR
      • 6. Thaw autologous digest in TCM media without IL2
      • 7. Centrifuge at 400 g×5 mins
      • 8. Aspirate and resuspend in 5 ml TCM media without IL2
      • 9. Count effectors, target cell lines and autologous digest using Vicell
      • 10. Collect 2e6 cells and resuspend in 2 mLs final volume with TCM media without IL2
      • 11. Collect 1e6 targets and resuspend in 10 mLs final volume with TCM media without IL2
      • 12. Collect either 2e6 or 10e6 cells for autologous digest and resuspend in 2 mL TCM media without IL2 (this will be either 1:1 or 1:5 ratio)
      • 13. Plate 50 uL TILs as per plate map
      • 14. Plate 50 uL targets as per plate map.
      • 15. Resuspend targets according to TILs to adjust E:T ratio to 10:1 (this is only for cell lines)
      • 16. Prepare pembro from stock (5 mg/mL) as per table
      • 17. Add 100 uL pembro as per plate map
      • 18. Add 100 uL media to plate 1
      • 19. Make up volume to 200 uL final in all wells. Note that effectors only and target only wells will have 150 uL; add 50 uL media or appropriate pembro concentration depending on plates
      • 20. Add TILs for FMOs
      • 21. Add PBS to empty wells
      • 22. Plate plates in the incubator
    Example 5 Serial Stimulation Assay of CoStAR-TIL Cells
  • TIL cells expressing CoStAR constructs then underwent a serial stimulation assay. The results of the stimulation assay are as shown in FIGS. 15A-15E and 16A-16E. As can be seen in those figures, the TILs modified with anti-CEA CoStAR but not Universal CoStAR expanded in a serial stimulation assay comparable to anti-FOLR CoStAR modified TILs. The methodology for the stimulation assay was as follows:
  • Day 1:
      • 1. Collect effector cells and count using Vicell
      • 2. Thaw targets (irradiated K562 OKT3 CEACAM5 and K562 OKT3 FOLR1) and count using Vicell
      • 3. Record counts in excel sheet and make up volume to 0.2M/ml for each sample
      • 4. Plate 50 uL of effectors (ie, 50K cells) and targets (ie, 10K cells) as per plate map. Note: ADD K562 OKT3 FOLR to control plate
      • 5. Add 100 ul of media into appropriate wells as per plate map
      • 6. Dilute 10 uL of stock pembro in 4.99 ml of fresh media to make 2× concentration for plate 2 (10 ug/ml) and add 100 ul into appropriate wells as per plate map
      • 7. Dilute 0.5 mL of stock pembro in 4.5 ml of fresh media to make 2× concentration for plate 3 (500 ug/ml) and add 100 ul into appropriate wells as per plate map
      • 8. Add 200 uL PBS to wells at the edge of the plates
      • 9. Place the plates into incubator
  • Day 6, 13, 20, 27, and 34: Collect cells for cell count
      • 1. Prepare cell counting by adding 180 ul of media into the Vi-cell 96 well counting plates
      • 2. Resuspend cells in the coculture plates
      • 3. Take 20 ul of resuspended cells and add into the Vi-cell 96 well plates. Refer to “plate map for counting” tab in the excel sheet.
      • 4. Put the coculture plates back into the incubator
      • 5. Count the Vi-cell 96 well plates (1:10 dilution)
      • 6. Put plates back in the incubator
  • Day 7, 14, 21, 28, and 35: re-stimulation of cells with targets
      • 1. Take the cell count files from vi-cell and plug into the excel sheet for calculation
      • 2. Collect 50 uL sample for staining
      • 3. Seal plates with parafilm and spin down the coculture plates at 500 g×5 min
      • 4. Decant the plates inside the biosafety cabinet
      • 5. Resuspend the cells in 50 ul/well of fresh media
      • 6. Prepare pembro and add 100 ul of pembro based on concentration calculation
      • 7. Dilute 12 uL of stock pembro in 6 ml of fresh media to make 2×concentration (10 ug/ml) and add 100 ul into appropriate wells as per plate map
      • 8. Dilute 0.6 mL of stock pembro in 5.4 ml of fresh media to make 2×concentration (500 ug/ml) and add 100 ul into appropriate wells as per plate map
      • 9. Add 100 ul of media into appropriate wells as per plate map
      • 10. Add additional media based on calculation in table
      • 11. Thaw irradiated K562 OKT3 CEACAM5 and K562 OKT3 FOLR targets
      • 12. resuspend target cells at 1M/ml and add target cells based on calculation in table
      • 13. Mix wells and place in incubator
      • 14. Add PBS to empty wells along the edges
  • Having thus described in detail preferred embodiments of the present invention, it is to be understood that the invention defined by the above paragraphs is not to be limited to particular details set forth in the above description as many apparent variations thereof are possible without departing from the spirit or scope of the present invention.

Claims (31)

What is claimed is:
1. An engineered protein that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 166, and wherein the sequence is not SEQ ID NO: 123.
2. An engineered protein that has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 167, and wherein the sequence is not SEQ ID NO: 123.
3. The engineered protein of any one of claim 1 or 2, further comprising a binding domain, CD28 domain, and CD40 domain.
4. The engineered protein of any one of claims 2-3, further comprising a signal peptide sequence.
5. The engineered protein of claim 4, wherein the signal peptide sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 157.
6. The engineered protein of any one of claims 3-5, wherein the binding domain comprises a VL sequence, a VH sequence, and an at least one linker.
7. The engineered protein of claim 6, wherein the at least one linker has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 159 or 161.
8. The engineered protein of any one of claims 6-7, wherein the VL sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 158.
9. The engineered protein of any one of claims 6-8, wherein the VH sequence has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 160.
10. The engineered protein of any one of claims 3-9, wherein the CD40 domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to the amino acid sequence of SEQ ID NO: 165.
11. The engineered protein of any one of claims 3-10, wherein the CD28 domain comprises a CD28 transmembrane domain.
12. The engineered protein of claim 11, wherein the CD28 transmembrane domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 163.
13. The engineered protein of any one of claims 3-12, wherein the CD28 domain comprises a CD28 extracellular domain.
14. The engineered protein of claim 13, wherein the CD28 extracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 162.
15. The engineered protein of any one of claims 3-14, wherein the CD28 domain comprises a CD28 intracellular domain.
16. The engineered protein of claim 15, wherein the CD28 intracellular domain has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 100%, or any integer that is between 80 and 100%, identity to SEQ ID NO: 164.
17. The engineered protein of any one of claims 1-16, wherein the protein further comprises 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDASVYAYALNI (SEQ ID NO: 173).
18. An engineered protein comprising an amino acid sequence that is at least 80% identical to the amino acid sequence of SEQ ID NO: 166 or 167, wherein the amino acid sequence does not include at least one of: QKLISEEDLE (SEQ ID NO: 174) or
(SEQ ID NO: 175) LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGN YSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI.
19. A CoStAR comprising:
(a) an optional signal peptide;
(b) a binding domain, wherein the binding domain binds to an anti-pembrolizumab antibody or binding fragment thereof;
(c) a CD28 domain;
(d) a CD40 domain;
wherein a) is optionally linked to b), wherein b) is linked to c), wherein c) is linked to d), and
wherein the CoStAR comprises an amino acid sequence that:
i) lacks at least one of: QKLISEEDLE (SEQ ID NO: 174) or
(SEQ ID NO: 175) LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGN YSQQLQVYSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI;
ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167;
iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is not SEQ ID NO: 123; or
iv) any combination of i-iv.
20. A fusion protein comprising:
(a) a means for binding to an antibody that binds to pembrolizumab;
(b) a CD28 domain;
(c) a CD40 domain;
wherein a) is linked to b), wherein b) is linked to c), and wherein the fusion protein comprises an amino acid sequence that:
i) lacks at least one of: QKLISEEDLE (SEQ ID NO: 174) or LVKQSPMLVAYDNAVNLSCKYSYNLFSREFRASLHKGLDSAVEVCVVYGNYSQQLQV YSKTGFNCDGKLGNESVTFYLQNLYVNQTDIYFCKI (SEQ ID NO: 175);
ii) has an amino acid sequence that is greater than 95% identical to SEQ ID NO: 166 or 167;
iii) has an amino acid sequence that is greater than 80% identical to SEQ ID NO: 166 or 167 and is not SEQ ID NO: 123; or
iv) any combination of i-iv.
21. The CoStAR or fusion protein of claim 19 or 20, wherein the binding domain or the means for binding to an antibody that binds to pembrolizumab comprises: 1, 2, 3, 4, 5, or all 6 CDR sequence(s) selected from the group consisting of: QASQSLSNLLA (SEQ ID NO: 168), GASNLES (SEQ ID NO: 169), QGGHYSGL (SEQ ID NO: 170), TNDMN (SEQ ID NO: 171), VIYSDDTPDYATWAKG (SEQ ID NO: 172), and/or GHYDASVYAYALNI (SEQ ID NO: 173).
22. A fusion protein comprising the amino acid sequence of SEQ ID NO: 166.
23. A fusion protein comprising the amino acid sequence of SEQ ID NO: 167.
24. A nucleic acid which encodes the protein of any one of the preceding claims.
25. A vector which comprises the nucleic acid of any one of the preceding claims.
26. A cell which expresses the protein of any one of the preceding claims.
27. A cell which expresses at least two proteins of any one of the preceding claims.
28. A method of making the cell of any one of claim 26 or 27, which comprises the step of transducing or transfecting a cell with a vector of claim 25.
29. A method for preparing a population of cells that express a protein of any one of claims 1 to 23, comprising detecting expression of the protein on the surface of cells transfected or transduced with a vector according to claim 25 and selecting cells which are identified as expressing the protein.
30. A cell population which is enriched for cell expression a protein of any one of claims 1 to 23.
31. A method for treating a disease in a subject in need thereof, which comprises the step of administering the cell of any one of claims 26-27 or the cell population of claim 30 to the subject.
US18/316,548 2021-07-16 2023-05-12 Chimeric molecules providing targeted costimulation for adoptive cell therapy Pending US20230331808A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/316,548 US20230331808A1 (en) 2021-07-16 2023-05-12 Chimeric molecules providing targeted costimulation for adoptive cell therapy

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202163222916P 2021-07-16 2021-07-16
PCT/US2022/073741 WO2023288278A1 (en) 2021-07-16 2022-07-14 Chimeric molecules providing targeted costimulation for adoptive cell therapy
US17/822,251 US11697677B2 (en) 2021-07-16 2022-08-25 Chimeric molecules providing targeted costimulation for adoptive cell therapy
US18/316,548 US20230331808A1 (en) 2021-07-16 2023-05-12 Chimeric molecules providing targeted costimulation for adoptive cell therapy

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US17/822,251 Continuation US11697677B2 (en) 2021-07-16 2022-08-25 Chimeric molecules providing targeted costimulation for adoptive cell therapy

Publications (1)

Publication Number Publication Date
US20230331808A1 true US20230331808A1 (en) 2023-10-19

Family

ID=84919708

Family Applications (2)

Application Number Title Priority Date Filing Date
US17/822,251 Active US11697677B2 (en) 2021-07-16 2022-08-25 Chimeric molecules providing targeted costimulation for adoptive cell therapy
US18/316,548 Pending US20230331808A1 (en) 2021-07-16 2023-05-12 Chimeric molecules providing targeted costimulation for adoptive cell therapy

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US17/822,251 Active US11697677B2 (en) 2021-07-16 2022-08-25 Chimeric molecules providing targeted costimulation for adoptive cell therapy

Country Status (4)

Country Link
US (2) US11697677B2 (en)
AR (1) AR126463A1 (en)
TW (1) TW202321285A (en)
WO (1) WO2023288278A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW202306997A (en) 2021-06-16 2023-02-16 英商英斯特生物科技有限公司 Receptors providing targeted costimulation for adoptive cell therapy

Family Cites Families (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5399346A (en) 1989-06-14 1995-03-21 The United States Of America As Represented By The Department Of Health And Human Services Gene therapy
IL104570A0 (en) 1992-03-18 1993-05-13 Yeda Res & Dev Chimeric genes and cells transformed therewith
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
EP1235842A4 (en) 1999-10-15 2003-04-23 Univ Massachusetts Rna interference pathway genes as tools for targeted genetic interference
US6326193B1 (en) 1999-11-05 2001-12-04 Cambria Biosciences, Llc Insect control agent
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US7572631B2 (en) 2000-02-24 2009-08-11 Invitrogen Corporation Activation and expansion of T cells
MXPA02008265A (en) 2000-02-24 2004-09-10 Xcyte Therapies Inc Simultaneous stimulation and concentration of cells.
WO2001096584A2 (en) 2000-06-12 2001-12-20 Akkadix Corporation Materials and methods for the control of nematodes
US7745140B2 (en) 2002-01-03 2010-06-29 The Trustees Of The University Of Pennsylvania Activation and expansion of T-cells using an engineered multivalent signaling platform as a research tool
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
US7232888B2 (en) 2002-07-01 2007-06-19 Massachusetts Institute Of Technology Antibodies against tumor surface antigens
EP1765988B1 (en) 2004-05-27 2017-09-20 The Trustees of The University of Pennsylvania Novel artificial antigen presenting cells and uses therefor
US9402865B2 (en) 2011-01-18 2016-08-02 The Trustees Of The University Of Pennsylvania Compositions and methods for treating cancer
EP3915588A1 (en) 2011-07-29 2021-12-01 The Trustees of the University of Pennsylvania Switch costimulatory receptors
ES2743738T3 (en) 2012-10-02 2020-02-20 Memorial Sloan Kettering Cancer Center Compositions and methods for immunotherapy
US9598489B2 (en) 2012-10-05 2017-03-21 The Trustees Of The Univeristy Of Pennsylvania Human alpha-folate receptor chimeric antigen receptor
WO2014055657A1 (en) 2012-10-05 2014-04-10 The Trustees Of The University Of Pennsylvania Use of a trans-signaling approach in chimeric antigen receptors
KR102417657B1 (en) 2013-02-06 2022-07-07 안트로제네시스 코포레이션 Modified t lymphocytes having improved specificity
KR20150131218A (en) * 2013-03-14 2015-11-24 벨리쿰 파마슈티컬스, 인크. Methods for controlling t cell proliferation
US10640569B2 (en) 2013-12-19 2020-05-05 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
WO2015112626A1 (en) 2014-01-21 2015-07-30 June Carl H Enhanced antigen presenting ability of car t cells by co-introduction of costimulatory molecules
GB201506423D0 (en) 2015-04-15 2015-05-27 Tc Biopharm Ltd Gamma delta T cells and uses thereof
EP3174546B1 (en) 2014-07-31 2019-10-30 Novartis AG Subset-optimized chimeric antigen receptor-containing t-cells
MA41538A (en) 2014-10-17 2017-12-26 Baylor College Medicine BIPARTITE AND TRIPARTITE IMMUNE CELLS OF SIGNALING
JP6917896B2 (en) 2015-03-05 2021-08-18 フレッド ハッチンソン キャンサー リサーチ センター Immunomodulatory fusion proteins and their use
GB201507368D0 (en) 2015-04-30 2015-06-17 Ucl Business Plc Cell
CA2992551A1 (en) * 2015-07-21 2017-01-26 Novartis Ag Methods for improving the efficacy and expansion of immune cells
US9995753B2 (en) * 2015-09-25 2018-06-12 Merck Sharp & Dohme Corp. Anti-pembrolizumab antibodies
MA44314A (en) 2015-11-05 2018-09-12 Juno Therapeutics Inc CHEMERICAL RECEPTORS CONTAINING TRAF-INDUCING DOMAINS, AND ASSOCIATED COMPOSITIONS AND METHODS
CN109069537B (en) 2016-04-04 2022-04-15 亘喜生物科技(上海)有限公司 Car with repeated binding motions in co-stimulation domain
WO2018156711A1 (en) 2017-02-22 2018-08-30 H. Lee Moffitt Cancer Center And Research Institute, Inc. Il13ra2-binding chimeric antigen receptors
SG10202109731YA (en) 2017-03-13 2021-10-28 Poseida Therapeutics Inc Compositions and methods for selective elimination and replacement of hematopoietic stem cells
WO2018169922A2 (en) * 2017-03-13 2018-09-20 Kite Pharma, Inc. Chimeric antigen receptors for melanoma and uses thereof
WO2018170475A1 (en) 2017-03-17 2018-09-20 Fred Hutchinson Cancer Research Center Immunomodulatory fusion proteins and uses thereof
KR20200014294A (en) 2017-04-26 2020-02-10 유레카 쎄라퓨틱스, 인코포레이티드 Chimeric Antibody / T-Cell Receptor Constructs and Uses thereof
WO2018208849A1 (en) 2017-05-09 2018-11-15 Bellicum Pharmaceuticals, Inc. Methods to augment or alter signal transduction
AU2019304174A1 (en) 2018-07-09 2021-01-28 Multitude Inc. Antibodies specific to folate receptor alpha
GB201900858D0 (en) * 2019-01-22 2019-03-13 Price Nicola Kaye Receptors providing targeted costimulation for adoptive cell therapy
EP4028526A4 (en) 2019-09-11 2023-08-30 Migal Galilee Research Institute Ltd. Chimeric antigen receptor comprising cd40 cytoplasmic domain and uses thereof
US20210277149A1 (en) * 2020-03-06 2021-09-09 Mustang Bio, Inc. Anti-idiotype antibodies and methods of using the same
WO2022016112A1 (en) 2020-07-17 2022-01-20 Instil Bio (Uk) Limited Receptors providing targeted costimulation for adoptive cell therapy
TW202306997A (en) 2021-06-16 2023-02-16 英商英斯特生物科技有限公司 Receptors providing targeted costimulation for adoptive cell therapy

Also Published As

Publication number Publication date
WO2023288278A1 (en) 2023-01-19
US20230059511A1 (en) 2023-02-23
TW202321285A (en) 2023-06-01
AR126463A1 (en) 2023-10-11
US11697677B2 (en) 2023-07-11

Similar Documents

Publication Publication Date Title
US20220160760A1 (en) Receptors providing targeted costimulation for adoptive cell therapy
RU2732925C2 (en) Mat-directed chimeric antigen receptor systems for sorting/depleting the engineered immune cells
WO2018144535A1 (en) Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
US20220348631A1 (en) Receptors providing targeted costimulation for adoptive cell therapy
US20230277670A1 (en) Chimeric molecules providing targeted costimulation for adoptive cell therapy
JP2021532742A (en) T cells containing NEF and their production methods
US20230055694A1 (en) Receptors providing targeted costimulation for adoptive cell therapy
US20230331808A1 (en) Chimeric molecules providing targeted costimulation for adoptive cell therapy
US20230227576A1 (en) Receptors providing targeted costimulation for adoptive cell therapy
WO2022037562A1 (en) Engineered immunoresponsive cells and uses thereof
US20230226181A1 (en) GENETIC ENGINEERING OF gamma delta T CELLS FOR IMMUNOTHERAPY
WO2021244654A1 (en) Activation induced cytokine production in immune cells
TW202122574A (en) Nef-containing t cells and methods of producing thereof
US20240058447A1 (en) Use of fusion constructs for il-2 independent t cell therapy
TW202216752A (en) Chimeric molecules providing targeted costimulation for adoptive cell therapy
WO2022007784A1 (en) Methods of reducing graft rejection of allogeneic cell therapy
US20230338422A1 (en) Engineering gamma delta t cells with interleukin-36 for immunotherapy
WO2023141530A2 (en) Receptors providing targeted costimulation for adoptive cell therapy
TW202216751A (en) Receptors providing targeted costimulation for adoptive cell therapy

Legal Events

Date Code Title Description
AS Assignment

Owner name: INSTIL BIO INC., TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:RODRIGUEZ, RUBEN;REEL/FRAME:063635/0155

Effective date: 20221003

Owner name: INSTIL BIO (UK) LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRIDGEMAN, JOHN;HAWKINS, ROBERT;SIGNING DATES FROM 20221004 TO 20221006;REEL/FRAME:063628/0266

Owner name: INSTIL BIO (UK) LIMITED, UNITED KINGDOM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INSTIL BIO INC.;REEL/FRAME:063628/0241

Effective date: 20230324

Owner name: INSTIL BIO, INC., TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SUKUMARAN, SUJITA;ZHOU, XINGLIANG;GSCHWENG, ERIC;SIGNING DATES FROM 20230222 TO 20230323;REEL/FRAME:063631/0196

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION