SECRETABLE PAYLOAD REGULATION

Abstract
Described herein are chimeric proteins including membrane-cleavable chimeric proteins and degron-fusion chimeric proteins. Also described herein are nucleic acids, pharmaceutical compositions, methods, and methods of treatment directed to the same.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted via Patent Center and is hereby incorporated by reference in its entirety. Said XML copy, created on Aug. 25, 2023, is named STB-019WOC1.xml, and is 302,703 bytes in size.


BACKGROUND

Cancer ranks as a leading cause of death and an important barrier to increasing life expectancy in every country of the world. Worldwide, an estimated 19.3 million new cancer cases and almost 10.0 million cancer deaths occurred in 2020. Further, according to the World Health Organization (WHO) Global Health Estimates in 2019, cancer was the first or second leading cause of death before the age of 70 years in 112 of 183 countries analyzed, and ranked third or fourth in a further 23 countries analyzed. Subsequently, there is also a notable financial burden of cancer for both the individual with cancer and for society as a whole. In the U.S. alone, costs for cancer care were estimated to be $208.9 billion in 2020. There remains a need in the art for improved compositions and methods for treating cancer.


SUMMARY

Provided herein, in some embodiments, is a combinatorial cell-based immunotherapy for the targeted treatment of cancer, such as ovarian cancer, breast cancer, colon cancer, lung cancer, and pancreatic cancer. This immunotherapy relies on engineered cell circuits that enable multifactorial modulation within and/or near a tumor (a “tumor microenvironment (TME)”). Despite exciting advancements in immunotherapy, its efficacy against cancer has been limited.


The immunotherapy provided herein, however, is tumor-specific and effective yet limits systemic toxicity. This immunotherapy delivers to a tumor microenvironment proteins of interest, such as immunomodulatory effector molecules, in a regulated manner. The design of the delivery vehicle is optimized to improve overall function in cancer therapy, including, but not limited to, optimization of the promoters, linkers, signal peptides, delivery methods, combination, regulation, and order of the immunomodulatory effector molecules.


It has been increasingly recognized that tumors are a complex interplay between the tumor cells and the surrounding stroma, which includes the extracellular matrix, cancer-associated stromal cells (MSCs and fibroblasts), tumor vasculature, and the immune system. The TME suppresses anti-tumor immune responses through multiple mechanisms that target both the innate and adaptive immune system of the patient. For example, tumors can recruit and induce regulatory T cells that suppress the anti-tumor activity of conventional T cells by elaborating specific chemokines such as CCL22. Tumors can also express molecules that inhibit the activity of T cells and NK cells, such as immune checkpoints such as PD-L1. Thus, targeting a single pathway is likely insufficient for achieving robust efficacy against solid tumors.


Non-limiting examples of effector molecules encompassed by the present disclosure include cytokines, antibodies, chemokines, nucleotides, peptides, enzymes, and oncolytic viruses. For example, cells may be engineered to express and secrete in a regulated manner at least one, two, three or more of the following effector molecules: IL-12, IL-16, IFN-β, IFN-γ, IL-2, IL-15, IL-7, IL-36γ, IL-18, IL-1β, IL-21, OX40-ligand, CD40L, anti-PD-1 antibodies, anti-PD-L1 antibodies, anti-CTLA-4 antibodies, anti-TGFβ antibodies, anti-TNFR2, MIP1α (CCL3), MIP1 (CCL5), CCL21, CpG oligodeoxynucleotides, and anti-tumor peptides (e.g., anti-microbial peptides having anti-tumor activity, see, e.g., Gaspar, D. et al. Front Microbiol. 2013; 4: 294; Chu, H. et al. PLoS One. 2015; 10(5): e0126390, and website:aps.unmc.edu/AP/main.php).


Provided herein is an engineered nucleic acid comprising an expression cassette comprising a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D wherein S comprises a secretable effector molecule, C comprises a protease cleavage site, MT comprises a cell membrane tethering domain, and D comprises a degron, wherein the promoter is operably linked to the exogenous polynucleotide sequence, and wherein S-C-MT-D is configured to be expressed as a single polypeptide. In some aspects, the engineered nucleic acid is selected from the group consisting of: a DNA, cDNA, an RNA, an mRNA, and a naked plasmid.


In some aspects, the promoter is a constitutive promoter. In some aspects, the constitutive promoter is selected from the group consisting of: CAG, HLP, CMV, EFS, SFFV, SV40, MND, PGK, UbC, hEF1aV1, hCAGG, hEF1aV2, hACTb, heIF4A1, hGAPDH, hGRP78, hGRP94, hHSP70, hKINb, and hUBIb. In some aspects, the promoter is an inducible promoter. In some aspects, the inducible promoter is selected from the group consisting of: minP, NFkB response element, CREB response element, NFAT response element, SRF response element 1, SRF response element 2, AP1 response element, TCF-LEF response element promoter fusion, Hypoxia responsive element, SMAD binding element, STAT3 binding site, minCMV, YB_TATA, minTK, inducer molecule responsive promoters, and tandem repeats thereof. In some aspects, the promoter is a synthetic promoter. In some aspects, the promoter is a tissue-specific promoter.


In some aspects, the secretable effector molecule comprises a native signal peptide native to the secretable effector molecule. In some aspects, the secretable effector molecule comprises a non-native signal peptide non-native to the secretable effector molecule. In some aspects, the non-native signal peptide is selected from the group consisting of: IL12, IL2, optimized IL2, trypsiongen-2, Gaussia luciferase, CD5, human IgKVII, murine IgKVII, VSV-G, prolactin, serum albumin preprotein, azurocidin preprotein, osteonectin, CD33, IL6, IL8, CCL2, TIMP2, VEGFB, osteoprotegerin, serpin E1, GROalpha, CXCL12, IL21, CD8, and GMCSF.


In some aspects, the secretable effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a peptide, and an enzyme. In some aspects, the secretable effector molecule is a human-derived effector molecule. In some aspects, the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha. In some aspects, the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1. In some aspects, the homing molecule is selected from the group consisting of: anti-integrin alpha4,beta7; anti-MAdCAM; SDF1; and MMP-2. In some aspects, the growth factor is selected from the group consisting of: FLT3L and GM-CSF. In some aspects, the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40L. In some aspects, the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2. In some aspects, TGFbeta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereof. In some aspects, the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody. In some aspects, the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof.


In some aspects, the protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site. In some aspects, the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain. In some aspects, the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA. In some aspects, the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof. In some aspects, when expressed in a cell, the secretable effector molecule is tethered to a cell membrane of the cell. In some aspects, when expressed in a cell expressing a protease capable of cleaving the protease cleavage site, the secretable effector molecule is released from the cell membrane. In some aspects, the protease expressed on the cell membrane is endogenous to the cell. In some aspects, the protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease. In some aspects, the protease expressed on the cell membrane is heterologous to the cell. In some aspects, the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3). In some aspects, the protease cleavage site comprises an NS3 protease cleavage site. In some aspects, the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site. In some aspects, the protease can be repressed by a protease inhibitor. In some aspects, the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir. In some aspects, expression of the protease is capable of regulation.


In some aspects, the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule. In some aspects, the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN. In some aspects, the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences. In some aspects, the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189). In some aspects, the IMiD is an FDA-approved drug, wherein the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide. In some aspects, the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box. In some aspects, the degron is localized C-terminal of the cell membrane tethering domain.


Also provided for herein is an expression vector comprising any of the engineered nucleic acids described herein. In some aspects, the expression vector is a viral vector.


Also provided for herein is a membrane-cleavable chimeric protein encoded by any of the engineered nucleic acids described herein.


Also provided for herein is a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D wherein S comprises a secretable effector molecule, C comprises a protease cleavage site, MT comprises a cell membrane tethering domain, D comprises a degron, and wherein S-C-MT-D is configured to be expressed as a single polypeptide.


Also provided herein is an isolated cell comprising any of the engineered nucleic acids described herein, or any of the expression vectors described herein, or any of the membrane-cleavable chimeric proteins described herein. In some aspects, the cell is an autologous or allogeneic cell selected from the group consisting of: a T cell, a CD8+ T cell, a CD4+ T cell, a gamma-delta T cell, a cytotoxic T lymphocyte (CTL), a regulatory T cell, a viral-specific T cell, a Natural Killer T (NKT) cell, a Natural Killer (NK) cell, a B cell, a tumor-infiltrating lymphocyte (TIL), an innate lymphoid cell, a mast cell, an eosinophil, a basophil, a neutrophil, a myeloid cell, a macrophage, a monocyte, a dendritic cell, an erythrocyte, a platelet cell, a human embryonic stem cell (ESC), an ESC-derived cell, a pluripotent stem cell, a mesenchymal stromal cell (MSC), an induced pluripotent stem cell (iPSC), and an iPSC-derived cell.


Also provided herein is an isolated cell comprising an engineered nucleic acid, wherein the engineered nucleic acid comprises an expression cassette comprising a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D wherein S comprises a secretable effector molecule, C comprises a protease cleavage site, MT comprises a cell membrane tethering domain, and D comprises a degron, wherein the promoter is operably linked to the exogenous polynucleotide sequence, and wherein S-C-MT-D is configured to be expressed as a single polypeptide. In some aspects, the cell is an autologous or allogeneic cell selected from the group consisting of: a T cell, a CD8+ T cell, a CD4+ T cell, a gamma-delta T cell, a cytotoxic T lymphocyte (CTL), a regulatory T cell, a viral-specific T cell, a Natural Killer T (NKT) cell, a Natural Killer (NK) cell, a B cell, a tumor-infiltrating lymphocyte (TIL), an innate lymphoid cell, a mast cell, an eosinophil, a basophil, a neutrophil, a myeloid cell, a macrophage, a monocyte, a dendritic cell, an erythrocyte, a platelet cell, a human embryonic stem cell (ESC), an ESC-derived cell, a pluripotent stem cell, a mesenchymal stromal cell (MSC), an induced pluripotent stem cell (iPSC), and an iPSC-derived cell.


In some aspects, the cell further comprises an antigen recognizing receptor. In some aspects, the antigen recognizing receptor recognizes an antigen selected from the group consisting of: 5T4, ADAM9, AFP, AXL, B7-H3, B7-H4, B7-H6, C4.4, CA6, Cadherin 3, Cadherin 6, CCR4, CD123, CD133, CD138, CD142, CD166, CD25, CD30, CD352, CD37, CD38, CD44, CD56, CD66e, CD70, CD71, CD74, CD79b, CD80, CEA, CEACAM5, Claudin18.2, cMet, CSPG4, CTLA, DLK1, DLL3, DR5, EGFR, ENPP3, EpCAM, EphA2, Ephrin A4, ETBR, FGFR2, FGFR3, FRalpha, FRb, GCC, GD2, GFRa4, gpA33, GPC3, gpNBM, GPRC5, HER2, IL-13R, IL-13Ra, IL-13Ra2, IL-8, IL-15, IL1RAP, Integrin aV, KIT, L1CAM, LAMP1, Lewis Y, LeY, LIV-1, LRRC, LY6E, MCSP, Mesothelin, MUC1, MUC16, MUC1C, NaPi2B, Nectin 4, NKG2D, NOTCH3, NY ESO 1, Ovarin, P-cadherin, pan-Erb2, PSCA, PSMA, PTK7, ROR1, S Aures, SCT, SLAMF7, SLITRK6, SSTR2, STEAP1, Survivin, TDGF1, TIM1, TROP2, and WT1. In some aspects, the antigen recognizing receptor comprises an antigen-binding domain. In some aspects, the antigen-binding domain comprises an antibody, an antigen-binding fragment of an antibody, a F(ab) fragment, a F(ab′) fragment, a single chain variable fragment (scFv), or a single-domain antibody (sdAb). In some aspects, the scFv comprises a heavy chain variable domain (VH) and a light chain variable domain (VL). In some aspects, the VH and VL are separated by a peptide linker. In some aspects, the scFv comprises the structure VH-L-VL or VL-L-VH, wherein VH is the heavy chain variable domain, L is the peptide linker, and VL is the light chain variable domain. In some aspects, the antigen recognizing receptor is a chimeric antigen receptor (CAR). In some aspects, the CAR comprises one or more intracellular signaling domains, and the one or more intracellular signaling domains are selected from the group consisting of: a CD3zeta-chain intracellular signaling domain, a CD97 intracellular signaling domain, a CD11a-CD18 intracellular signaling domain, a CD2 intracellular signaling domain, an ICOS intracellular signaling domain, a CD27 intracellular signaling domain, a CD154 intracellular signaling domain, a CD8 intracellular signaling domain, an OX40 intracellular signaling domain, a 4-1BB intracellular signaling domain, a CD28 intracellular signaling domain, a ZAP40 intracellular signaling domain, a CD30 intracellular signaling domain, a GITR intracellular signaling domain, an HVEM intracellular signaling domain, a DAP10 intracellular signaling domain, a DAP12 intracellular signaling domain, and a MyD88 intracellular signaling domain. In some aspects, the CAR comprises a transmembrane domain, and the transmembrane domain is selected from the group consisting of: a CD8 transmembrane domain, a CD28 transmembrane domain a CD3zeta-chain transmembrane domain, a CD4 transmembrane domain, a 4-1BB transmembrane domain, an OX40 transmembrane domain, an ICOS transmembrane domain, a CTLA-4 transmembrane domain, a PD-1 transmembrane domain, a LAG-3 transmembrane domain, a 2B4 transmembrane domain, and a BTLA transmembrane domain. In some aspects, the CAR comprises a spacer region between the antigen-binding domain and the transmembrane domain.


Also provided herein is a composition comprising any of the engineered nucleic acids described herein, any of the expression vectors described herein, any of the membrane-cleavable chimeric proteins described herein, or any of the isolated cells described herein, and a pharmaceutically acceptable carrier, pharmaceutically acceptable excipient, or a combination thereof.


Also provided herein is a method of treating a subject in need thereof, the method comprising administering any of the engineered nucleic acids described herein, any of the expression vectors described herein, any of the membrane-cleavable chimeric proteins described herein, any of the isolated cells described herein, or any of the compositions described herein.


Provided for herein is an engineered nucleic acid comprising an expression cassette comprising a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D wherein S comprises a secretable effector molecule, C comprises a protease cleavage site, MT comprises a cell membrane tethering domain, and D comprises a degron, wherein the promoter is operably linked to the exogenous polynucleotide sequence, and wherein S-C-MT-D is configured to be expressed as a single polypeptide.


In some aspects, the promoter is a constitutive promoter. In some aspects, the constitutive promoter is selected from the group consisting of: CAG, HLP, CMV, EFS, SFFV, SV40, MND, PGK, UbC, hEF1aV1, hCAGG, hEF1aV2, hACTb, heIF4A1, hGAPDH, hGRP78, hGRP94, hHSP70, hKINb, and hUBIb. In some aspects, the promoter is an inducible promoter. In some aspects, the inducible promoter is selected from the group consisting of: minP, NFkB response element, CREB response element, NFAT response element, SRF response element 1, SRF response element 2, AP1 response element, TCF-LEF response element promoter fusion, Hypoxia responsive element, SMAD binding element, STAT3 binding site, minCMV, YB_TATA, minTK, inducer molecule responsive promoters, and tandem repeats thereof. In some aspects, the promoter is a synthetic promoter. In some aspects, the promoter is a tissue-specific promoter.


In some aspects, the secretable effector molecule comprises a signal peptide. In some aspects, the signal peptide comprises a native signal peptide native to the secretable effector molecule. In some aspects, the signal peptide comprises a non-native signal peptide non-native to the secretable effector molecule. In some aspects, the non-native signal peptide is selected from the group consisting of: IL12, IL2, optimized IL2, trypsiongen-2, Gaussia luciferase, CD5, human IgKVII, murine IgKVII, VSV-G, prolactin, serum albumin preprotein, azurocidin preprotein, osteonectin, CD33, IL6, IL8, CCL2, TIMP2, VEGFB, osteoprotegerin, serpin E1, GROalpha, CXCL12, IL21, CD8, and GMCSF.


In some aspects, the secretable effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a peptide, and an enzyme. In some aspects, the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha. In some aspects, the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1. In some aspects, the homing molecule is selected from the group consisting of: anti-integrin alpha4,beta7; anti-MAdCAM; SDF1; and MMP-2. In some aspects, the growth factor is selected from the group consisting of: FLT3L and GM-CSF. In some aspects, the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40L. In some aspects, the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2. In some aspects, the TGFbeta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereof. In some aspects, the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody. In some aspects, the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof. In some aspects, the secretable effector molecule is a human-derived effector molecule.


In some aspects, the protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site.


In some aspects, the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain. In some aspects, the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA. In some aspects, the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof.


In some aspects, when expressed in a cell, the secretable effector molecule is tethered to a cell membrane of the cell. In some aspects, when expressed in a cell expressing a protease capable of cleaving the protease cleavage site, the secretable effector molecule is released from the cell membrane. In some aspects, the protease expressed on the cell membrane is endogenous to the cell. In some aspects, the protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease. In some aspects, the protease expressed on the cell membrane is heterologous to the cell. In some aspects, the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3). In some aspects, the protease cleavage site comprises an NS3 protease cleavage site. In some aspects, the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site. In some aspects, the protease can be repressed by a protease inhibitor. In some aspects, the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir. In some aspects, expression of the protease is capable of regulation.


In some aspects, the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule. In some aspects, the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN. In some aspects, the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences. In some aspects, the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189). In some aspects, the IMiD is an FDA-approved drug. In some aspects, the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide.


In some aspects, the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box. In some aspects, the degron is localized C-terminal of the cell membrane tethering domain.


In some aspects, the engineered nucleic acid is selected from the group consisting of: a DNA, cDNA, an RNA, an mRNA, and a naked plasmid.


Also provided for herein is an expression vector comprising any of the engineered nucleic acids described herein. In some aspects, the expression vector is a viral vector. In some aspects, the viral vector is a lentiviral vector.


Also provided for herein, a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D wherein S comprises a secretable effector molecule, C comprises a protease cleavage site, MT comprises a cell membrane tethering domain, D comprises a degron, and wherein S-C-MT-D is configured to be expressed as a single polypeptide.


In some aspects, the secretable effector molecule comprises a signal peptide. In some aspects, the signal peptide comprises a native signal peptide native to the secretable effector molecule. In some aspects, the signal peptide comprises a non-native signal peptide non-native to the secretable effector molecule. In some aspects, the non-native signal peptide is selected from the group consisting of: IL12, IL2, optimized IL2, trypsiongen-2, Gaussia luciferase, CD5, human IgKVII, murine IgKVII, VSV-G, prolactin, serum albumin preprotein, azurocidin preprotein, osteonectin, CD33, IL6, IL8, CCL2, TIMP2, VEGFB, osteoprotegerin, serpin E1, GROalpha, CXCL12, IL21, CD8, and GMCSF.


In some aspects, the secretable effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a peptide, and an enzyme. In some aspects, the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha. In some aspects, the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1. In some aspects, the homing molecule is selected from the group consisting of: anti-integrin alpha4,beta7; anti-MAdCAM; SDF1; and MMP-2. In some aspects, the growth factor is selected from the group consisting of: FLT3L and GM-CSF. In some aspects, the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40L. In some aspects, the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2. In some aspects, the TGF-beta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereof. In some aspects, the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody. In some aspects, the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof. In some aspects, the secretable effector molecule is a human-derived effector molecule.


In some aspects, the protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site.


In some aspects, the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain. In some aspects, the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA. In some aspects, the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof. In some aspects, when expressed in a cell, the secretable effector molecule is tethered to a cell membrane of the cell.


In some aspects, when expressed in a cell expressing a protease capable of cleaving the protease cleavage site, the secretable effector molecule is released from the cell membrane. In some aspects, the protease expressed on the cell membrane is endogenous to the cell. In some aspects, the protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease. In some aspects, the protease expressed on the cell membrane is heterologous to the cell. In some aspects, the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3). In some aspects, the protease cleavage site comprises an NS3 protease cleavage site. In some aspects, the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site.


In some aspects, the protease can be repressed by a protease inhibitor. In some aspects, the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir.


In some aspects, the protease further comprises a degron. In some aspects, expression of the protease is capable of regulation.


In some aspects, the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule. In some aspects, the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN. In some aspects, the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences. In some aspects, the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189). In some aspects, the IMiD is an FDA-approved drug. In some aspects, the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide.


In some aspects, the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box. In some aspects, the degron is localized C-terminal of the cell membrane tethering domain.


Also provided for herein is a composition comprising any of the engineered nucleic acids of described herein, or any of the expression vectors described herein, or any of the membrane-cleavable chimeric proteins described herein, and a pharmaceutically acceptable carrier.


Also provided for herein is an isolated cell comprising any of the engineered nucleic acids of described herein, or any of the expression vectors described herein, or any of the membrane-cleavable chimeric proteins described herein.


Also provided for herein is an isolated cell comprising an engineered nucleic acid, wherein the engineered nucleic acid comprises an expression cassette comprising a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D wherein S comprises a secretable effector molecule, C comprises a protease cleavage site, MT comprises a cell membrane tethering domain, and D comprises a degron, wherein the promoter is operably linked to the exogenous polynucleotide sequence, and wherein S-C-MT-D is configured to be expressed as a single polypeptide.


In some aspects, the engineered nucleic acid is recombinantly expressed. In some aspects, the engineered nucleic acid is expressed from a vector or a selected locus from the genome of the cell.


Also provided for herein is an isolated cell comprising a membrane-cleavable chimeric protein, wherein the membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, has the formula: S-C-MT-D wherein S comprises a secretable effector molecule, C comprises a protease cleavage site, MT comprises a cell membrane tethering domain, D comprises a degron and and wherein S-C-MT-D is configured to be expressed as a single polypeptide.


In some aspects, the cell is selected from the group consisting of: a T cell, a CD8+ T cell, a CD4+ T cell, a gamma-delta T cell, a cytotoxic T lymphocyte (CTL), a regulatory T cell, a viral-specific T cell, a Natural Killer T (NKT) cell, a Natural Killer (NK) cell, a B cell, a tumor-infiltrating lymphocyte (TIL), an innate lymphoid cell, a mast cell, an eosinophil, a basophil, a neutrophil, a myeloid cell, a macrophage, a monocyte, a dendritic cell, an erythrocyte, a platelet cell, a human embryonic stem cell (ESC), an ESC-derived cell, a pluripotent stem cell, a mesenchymal stromal cell (MSC), an induced pluripotent stem cell (iPSC), and an iPSC-derived cell. In some aspects, the cell is autologous. In some aspects, the cell is allogeneic. In some aspects, the cell is a tumor cell selected from the group consisting of: a bladder tumor cell, a brain tumor cell, a breast tumor cell, a cervical tumor cell, a colorectal tumor cell, an esophageal tumor cell, a glioma cell, a kidney tumor cell, a liver tumor cell, a lung tumor cell, a melanoma cell, an ovarian tumor cell, a pancreatic tumor cell, a prostate tumor cell, a skin tumor cell, a thyroid tumor cell, and a uterine tumor cell.


In some aspects, the cell was engineered via transduction with an oncolytic virus. In some aspects, the oncolytic virus is selected from the group consisting of: an oncolytic herpes simplex virus, an oncolytic adenovirus, an oncolytic measles virus, an oncolytic influenza virus, an oncolytic Indiana vesiculovirus, an oncolytic Newcastle disease virus, an oncolytic vaccinia virus, an oncolytic poliovirus, an oncolytic myxoma virus, an oncolytic reovirus, an oncolytic mumps virus, an oncolytic Maraba virus, an oncolytic rabies virus, an oncolytic rotavirus, an oncolytic hepatitis virus, an oncolytic rubella virus, an oncolytic dengue virus, an oncolytic chikungunya virus, an oncolytic respiratory syncytial virus, an oncolytic lymphocytic choriomeningitis virus, an oncolytic morbillivirus, an oncolytic lentivirus, an oncolytic replicating retrovirus, an oncolytic rhabdovirus, an oncolytic Seneca Valley virus, an oncolytic sindbis virus, and any variant or derivative thereof. In some aspects, the oncolytic virus is a recombinant oncolytic virus comprising the first expression cassette and the second expression cassette.


In some aspects, the cell further comprises a protease capable of cleaving the protease cleavage site. In some aspects, the protease is an endogenous protease. In some aspects, the endogenous protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease. In some aspects, the protease is a heterologous protease. In some aspects, the heterologous protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3). In some aspects, the protease is expressed on the cell membrane of the cell. In some aspects, the protease is capable of cleaving the protease cleavage site. In some aspects, cleavage of the protease cleavage site releases the secretable effector molecule from the cell membrane of the cell.


In some aspects, the cell further comprises an antigen recognizing receptor. In some aspects, the antigen recognizing receptor recognizes an antigen selected from the group consisting of: 5T4, ADAM9, AFP, AXL, B7-H3, B7-H4, B7-H6, C4.4, CA6, Cadherin 3, Cadherin 6, CCR4, CD123, CD133, CD138, CD142, CD166, CD25, CD30, CD352, CD37, CD38, CD44, CD56, CD66e, CD70, CD71, CD74, CD79b, CD80, CEA, CEACAM5, Claudin18.2, cMet, CSPG4, CTLA, DLK1, DLL3, DR5, EGFR, ENPP3, EpCAM, EphA2, Ephrin A4, ETBR, FGFR2, FGFR3, FRalpha, FRb, GCC, GD2, GFRa4, gpA33, GPC3, gpNBM, GPRC5, HER2, IL-13R, IL-13Ra, IL-13Ra2, IL-8, IL-15, IL1RAP, Integrin aV, KIT, L1CAM, LAMP1, Lewis Y, LeY, LIV-1, LRRC, LY6E, MCSP, Mesothelin, MUC1, MUC16, MUC1C, NaPi2B, Nectin 4, NKG2D, NOTCH3, NY ESO 1, Ovarin, P-cadherin, pan-Erb2, PSCA, PSMA, PTK7, ROR1, S Aures, SCT, SLAMF7, SLITRK6, SSTR2, STEAP1, Survivin, TDGF1, TIM1, TROP2, and WT1. In some aspects, the antigen recognizing receptor comprises an antigen-binding domain. In some aspects, the antigen-binding domain comprises an antibody, an antigen-binding fragment of an antibody, a F(ab) fragment, a F(ab′) fragment, a single chain variable fragment (scFv), or a single-domain antibody (sdAb). In some aspects, the antigen-binding domain comprises a single chain variable fragment (scFv). In some aspects, the scFv comprises a heavy chain variable domain (VH) and a light chain variable domain (VL). In some aspects, the VH and VL are separated by a peptide linker. In some aspects, the scFv comprises the structure VH-L-VL or VL-L-VH, wherein VH is the heavy chain variable domain, L is the peptide linker, and VL is the light chain variable domain. In some aspects, the antigen recognizing receptor is a chimeric antigen receptor (CAR) or T cell receptor (TCR). In some aspects, the antigen recognizing receptor comprises an antigen-binding domain derived from PF-06263507, ASN004, GEN1044, MGA021, YW327.6S2, D9, E8, Mab173, obrindatamab, mAb378.96, XMT-1660, lupartumab amadotin, HKT288, mogamulizumab, CSL362, AC133, AC141, VIS832, CX-2009, daclizumab, basiliximab, brentuximab vedotin, SGN-CD352A, IMGN529, lilotomab, daratumumab, bivatuzumab, m900, m906, labetuzumab, R06958688, MFE-23, ARGX-110, vorsetuzumab mafodotin, INA01, milatuzumab, polatuzumab vedotin, galiximab, polatuzumab vedotin, galiximab, claudiximab, zolbetuximab, amivantamab, onartuzumab, emibetuzumab, ARGX-111, YYB-101, TP41.2, ipilimumab, AK104, rovalpituzumab tesirine, TRA-8, zapadicine-1, cetuximab, panitumumab, adecatumumab, DS-8895a, PF-06647263, DEDN6526A, BAY 1179470, bemarituzumab, MORAb-003, IMGN853, MLN0264, TAK-264, dinutuximab, MGD007, A33scFv-Fc, codrituzumab, ERY974, glembatumumab (CR011), taquetamab, trastuzumab, ASLAN004, ABX-IL8, BMS-986253, KD033, KTN0158, m3s193 BsAb, hu3S193, SGN-LIV1A, DLYE5953A, MMOT0530A, BMS-986148, anetumab ravtasine, pemtumomab, gatipotuzumab, oregovomab, abagovomab, Mab-AR-9.6, XMT-1592, upifitamab rilsodotin, lifatuzumab vedotin, efortumab vedotin, NNC0142-0002, PF-03732010, AGS-1C4D4, KM2777, KM2812, MT112, cofetuzumab pelidotin, zilovertamab vedotin, elotuzumab, ASG-15ME, tidutamab, vandortuzumab vedotin, AMG509, BIIB015, RMT1-10, Sacituzumab govitecan-hziy, or F7AK3.


In some aspects, the antigen recognizing receptor is a CAR. In some aspects, the CAR comprises one or more intracellular signaling domains, and the one or more intracellular signaling domains are selected from the group consisting of: a CD3zeta-chain intracellular signaling domain, a CD97 intracellular signaling domain, a CD11a-CD18 intracellular signaling domain, a CD2 intracellular signaling domain, an ICOS intracellular signaling domain, a CD27 intracellular signaling domain, a CD154 intracellular signaling domain, a CD8 intracellular signaling domain, an OX40 intracellular signaling domain, a 4-1BB intracellular signaling domain, a CD28 intracellular signaling domain, a ZAP40 intracellular signaling domain, a CD30 intracellular signaling domain, a GITR intracellular signaling domain, an HVEM intracellular signaling domain, a DAP10 intracellular signaling domain, a DAP12 intracellular signaling domain, and a MyD88 intracellular signaling domain. In some aspects, the CAR comprises a transmembrane domain, and the transmembrane domain is selected from the group consisting of: a CD8 transmembrane domain, a CD28 transmembrane domain a CD3zeta-chain transmembrane domain, a CD4 transmembrane domain, a 4-1BB transmembrane domain, an OX40 transmembrane domain, an ICOS transmembrane domain, a CTLA-4 transmembrane domain, a PD-1 transmembrane domain, a LAG-3 transmembrane domain, a 2B4 transmembrane domain, and a BTLA transmembrane domain. In some aspects, the CAR comprises a spacer region between the antigen-binding domain and the transmembrane domain.


Also provided for herein is a composition comprising any of the isolated cells described herein, and a pharmaceutically acceptable carrier.


Also provided for herein is a method of treating a subject in need thereof, the method comprising administering a therapeutically effective dose of any of the isolated cells described herein or any of the compositions described herein. In some aspects, the isolated cell is derived from the subject. In some aspects, the isolated cell is allogeneic with reference to the subject. In some aspects, the method further comprises administering a checkpoint inhibitor. In some aspects, the checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody. In some aspects, the method further comprises administering an anti-CD40 antibody.


Also provided for herein is a lipid-based structure comprising any of the engineered nucleic acids descrinbed herein, or any of the expression vectors described herein, or any of the membrane-cleavable chimeric proteins described herein. In some aspects, the lipid-based structure comprises a extracellular vesicle. In some aspects, the extracellular vesicle is selected from the group consisting of: a nanovesicle and an exosome. In some aspects, the lipid-based structure comprises a lipid nanoparticle or a micelle. In some aspects, the lipid-based structure comprises a liposome.


Also provided for herein is a composition comprising any of the lipid-based structures described herein, and a pharmaceutically acceptable carrier.


Also provided for herein is a method of treating a subject in need thereof, the method comprising administering a therapeutically effective dose of any of the lipid-based structures described herein or any of the compositions described herein. In some aspects, the administering comprises systemic administration. In some aspects, the lipid-based structure is capable of engineering a cell in the subject. In some aspects, the method further comprises administering a checkpoint inhibitor. In some aspects, the checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody. In some aspects, the method further comprises administering an anti-CD40 antibody.


Also provided for herein is a nanoparticle comprising any of the engineered nucleic acids described herein or any of the membrane-cleavable chimeric proteins described herein. In some aspects, the nanoparticle comprises an inorganic material.


Also provided for herein is a composition comprising any of the nanoparticles described herein.


Also provided for herein is a method of treating a subject in need thereof, the method comprising administering a therapeutically effective dose of any of the nanoparticles described herein, or any of the compositions described herein. In some aspects, the administering comprises systemic administration. In some aspects, the nanoparticle is capable of engineering a cell in the subject. In some aspects, the method further comprises administering a checkpoint inhibitor. In some aspects, the checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody. In some aspects, the method further comprises administering an anti-CD40 antibody.


Also provided for herein is a virus engineered to comprise any of the engineered nucleic acids described herein or any of the expression vectors described herein. In some aspects, the virus is selected from the group consisting of: a lentivirus, a retrovirus, an oncolytic virus, an adenovirus, an adeno-associated virus (AAV), and a virus-like particle (VLP).


Also provided for herein is a composition comprising any of the engineered viruses described herein, and a pharmaceutically acceptable carrier.


Also provided for herein is a method of treating a subject in need thereof, the method comprising administering a therapeutically effective dose of any of the engineered viruses described herein, or any of the compositions described herein. In some aspects, the administering comprises systemic administration. In some aspects, the engineered virus infects a cell in the subject and expresses the expression cassette. In some aspects, the method further comprises administering a checkpoint inhibitor. In some aspects, the checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody. In some aspects, the method further comprises administering an anti-CD40 antibody.


Also provided for herein is a method of inducing release of a membrane-tethered effector molecule, comprising: a) providing a cell, wherein the cell comprises a membrane-bound protease and a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D wherein S comprises an effector molecule, C comprises a cognate membrane-bound protease cleavage site, MT comprises a cell membrane tethering domain, D comprises a degron, and wherein S-C-MT-D is configured to be expressed as a single polypeptide; and b) culturing the cell under conditions suitable for expression of the membrane-bound protease and the membrane-cleavable chimeric protein, wherein upon expression, the membrane-cleavable chimeric protein is tethered to the cell membrane of the cell, and wherein, upon expression, the membrane-bound protease cleaves the cognate membrane-bound protease cleavage site of the membrane-cleavable chimeric protein, thereby releasing the effector molecule from the cell membrane. In some aspects, the degron is a drug-inducible degron. In some aspects, the method further comprises contacting the cell with a drug that induces the degron. In some aspects, induction of the degron degrades the membrane-cleavable chimeric protein.


Also provided for herein is a method of degrading a membrane-tethered effector molecule, comprising: a) providing a cell, wherein the cell comprises a membrane-bound protease and a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D wherein S comprises an effector molecule, C comprises a cognate membrane-bound protease cleavage site, MT comprises a cell membrane tethering domain, D comprises a drug-inducible degron, and wherein S-C-MT-D is configured to be expressed as a single polypeptide; b) culturing the cell under conditions suitable for expression of the membrane-bound protease and the membrane-cleavable chimeric protein, wherein upon expression, the membrane-cleavable chimeric protein is tethered to the cell membrane of the cell; and c) inducing the degron by contacting the cell with a drug, wherein degron induction of the degron degrades the membrane-cleavable chimeric protein. In some aspects, the effector molecule comprises a signal peptide.


In some aspects, the signal peptide comprises a native signal peptide native to the effector molecule. In some aspects, the signal peptide comprises a non-native signal peptide non-native to the effector molecule. In some aspects, the non-native signal peptide is selected from the group consisting of: IL12, IL2, optimized IL2, trypsiongen-2, Gaussia luciferase, CD5, human IgKVII, murine IgKVII, VSV-G, prolactin, serum albumin preprotein, azurocidin preprotein, osteonectin, CD33, IL6, IL8, CCL2, TIMP2, VEGFB, osteoprotegerin, serpin E1, GROalpha, CXCL12, IL21, CD8, and GMCSF.


In some aspects, the effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a polynucleotide, a peptide, and an enzyme. In some aspects, the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha. In some aspects, the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1. In some aspects, the homing molecule is selected from the group consisting of: anti-integrin alpha4,beta7; anti-MAdCAM; SDF1; and MMP-2. In some aspects, the growth factor is selected from the group consisting of: FLT3L and GM-CSF. In some aspects, the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40L. In some aspects, the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2. In some aspects, the TGFbeta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereof. In some aspects, the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody. In some aspects, the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof. In some aspects, the effector molecule is a human-derived effector molecule.


In some aspects, the cognate protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site.


In some aspects, the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain. In some aspects, the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA. In some aspects, the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof.


In some aspects, the protease is endogenous to the cell. In some aspects, the protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease. In some aspects, the protease is heterologous to the cell. In some aspects, the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3). In some aspects, the protease cleavage site comprises an NS3 protease cleavage site. In some aspects, the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site.


In some aspects, the protease can be repressed by a protease inhibitor. In some aspects, the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir.


In some aspects, protease further comprises a degron. In some aspects, expression of the protease is capable of regulation.


In some aspects, the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule. In some aspects, the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN. In some aspects, the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences. In some aspects, the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189). In some aspects, the IMiD is an FDA-approved drug. In some aspects, the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide.


In some aspects, the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box. In some aspects, the degron is localized C-terminal of the cell membrane tethering domain.


Also provided for herein is an adeno-associated virus (AAV) engineered to express a heterologous nucleic acid, wherein the heterologous nucleic acid encodes a degron-fusion chimeric protein, and wherein the degron-fusion chimeric protein comprises a degron fused to a protein of interest.


In some aspects, the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the protein of interest. In some aspects, the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN. In some aspects, the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences. In some aspects, the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189). In some aspects, the IMiD is an FDA-approved drug. In some aspects, the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide. In some aspects, the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box.


In some aspects, the AAV is selected from the group consisting of: AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV.Rh10, AAV11, and variants thereof.


In some aspects, the protein of interest is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a polynucleotide, a peptide, and an enzyme.


Also provided for herein is an adeno-associated viral (AAV) vector comprising a heterologous nucleic acid, wherein the heterologous nucleic acid encodes a degron-fusion chimeric protein, and wherein the degron-fusion chimeric protein comprises a degron fused to a protein of interest.


In some aspects, the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the protein of interest. In some aspects, the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN. In some aspects, the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences. In some aspects, the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189). In some aspects, the IMiD is an FDA-approved drug. In some aspects, the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide. In some aspects, the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box.


In some aspects, the AAV is selected from the group consisting of: AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV.Rh10, AAV11, and variants thereof.


In some aspects, the protein of interest is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a polynucleotide, a peptide, and an enzyme.


Also provided for herein is a pharmaceutical composition comprising any of the engineered AAV viruses described herein or any of the AAV vectors described herein, and a pharmaceutically acceptable carrier.


Also provided for herein is an isolated cell comprising any of the engineered AAV viruses described herein or any of the AAV vectors described herein.


Also provided for herein is a method for in vivo regulated protein expression, comprising administering to a subject in need thereof the engineered AAV viruses described herein or any of the AAV vectors described herein.


Also provided for herein is a method for regulating protein expression, comprising contacting a cell with any the engineered AAV viruses described herein or any of the AAV vectors described herein under conditions suitable for expression of the engineered AAV virus or AAV vector.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows SEAP enzymatic activity in supernatants from both HEK293Ft cells (left panel) and donor-derived T cells (right panel) following transduction with viral constructs that expressed either SEAP without a degron (“419”) or SEAP chimeric proteins with C-terminal degrons fused to SEAP using various linkers (“1118”, “1141”, “1143”, “1144”), or supernatant from untransduced cells (“no virus”). Shown are averaged replicates with either no drug added (right column for each construct, respectively) or 1 μM lenalidomide (left column for each construct, respectively).



FIG. 2 illustrates a representative Membrane-Cleavable Degron system in which a desired payload is expressed as a membrane-cleavable chimeric protein with a cleavable membrane-tethering domain (e.g., as shown here an ADAM10/17 cleavage site fused to PDGFRb transmembrane domain) inserted between the payload and degron domain.



FIG. 3 shows SEAP enzymatic activity in supernatants from both HEK293Ft cells (“HEK293T”) and donor-derived T cells (“T cells”) following transduction with viral constructs that expressed either SEAP without a degron (“419”) or SEAP chimeric proteins in Membrane-Cleavable Degron system formats (D-MT-C-S “1208”; S-C-MT-D “1209”), or supernatant from untransduced cells (“no virus”). Shown are averaged replicates with either no drug added (right column for each construct, respectively) or 1 μM lenalidomide (left column for each construct, respectively).



FIG. 4 shows SEAP enzymatic activity in supernatants from HEK293Ft cells (“HEK293T”) following transduction with viral constructs that expressed either SEAP without a degron (“419”) or SEAP chimeric proteins in Membrane-Cleavable Degron system formats (S-C-MT-D “1209”). Shown are averaged replicates with either no drug added (right column for each construct, respectively) or 1 μM lenalidomide (left column for each construct, respectively).



FIG. 5 shows SEAP enzymatic activity in supernatants from Jurkat T cells following transduction with viral constructs that expressed either SEAP without a degron (“419”), SEAP chimeric proteins with C-terminal degrons fused to SEAP using various linkers (“1118”, “1141”, “1143”, “1144”), or SEAP chimeric proteins in a Membrane-Cleavable Degron system format (S-C-MT-D “1209”), or supernatant following transduction with an irrelevant viral construct used as an infection control (“1321”). Shown are averaged replicates with either no drug added (left column for each construct, respectively) or 1 μM lenalidomide (right column for each construct, respectively).



FIG. 6 illustrates viral vector pL17d-based constructs engineered to express human IL-10 (hIL-10) either in its native form (“1584”) or as a membrane-cleavable chimeric protein fused to a membrane-cleavable degron (“1585”). The viral vectors are also engineered to express human IL-12 (hIL-12) from a separate promoter to serve as a normalization control for cell number, viability, and transduction efficiency.



FIG. 7 shows hIL-10 secretion in supernatants from donor-derived T cells, as normalized to IL-12 expression (hIL10/hIL12 ratio). The constructs tested either expressed native hIL-10 without a degron (“1584”), expressed a negative-control protein without a degron (“1043”), or expressed a hIL-10 membrane-cleavable chimeric protein in a S-C-MT-D orientation membrane-cleavable degron system (“1585”). Shown are averaged replicates with either no drug added (left column for each construct, respectively) or 1 μM lenalidomide (right column for each construct, respectively).



FIG. 8 shows luciferase activity in Hep3B cells transduced with either AAV2-based (left panel) or AAV8-based (right panel) constructs expressing a nanoluciferase degron-fusion chimeric protein containing a degron domain. Shown are averaged replicates with either no drug added (left column for each construct, respectively) or 1 μM lenalidomide (right column for each construct, respectively).



FIG. 9 shows luciferase activity in Hep3B cells transduced with an AAV2-based construct expressing a nanoluciferase degron-fusion chimeric protein containing a degron domain treated with two IMiDs, either 1-0.01 μM lenalidomide (left panel) or pomalidomide (left right panel).





DETAILED DESCRIPTION

Provided herein are immunotherapy compositions and methods directed to modulating different tumor-mediated immunosuppressive mechanisms within and/or near a tumor (a “tumor microenvironment (TME)”).


Chimeric proteins (or engineered nucleic acids encoding the chimeric proteins) are provided for herein having a degron domain.


In some aspects, membrane-cleavable chimeric proteins (or engineered nucleic acids encoding the chimeric proteins) are provided for herein having the formula S-C-MT-D, oriented from N-terminal to C-terminal and expressed as a single polypeptide. S refers to a secretable effector molecule. C refers to a protease cleavage site. MT refers to a cell membrane tethering domain. D refers to a degron. The membrane-cleavable chimeric protein is engineered such that secretion of the effector molecule can be regulated in a drug-dependent manner. Specifically, the membrane-cleavable chimeric protein is engineered such that secretion of the effector molecule can be regulated as part of a “Membrane-Cleavable Degron” system, where incorporation of a protease cleavage site (“C”), a cell membrane tethering domain (“MT”), and a degron (“D”) allow for regulated secretion of an effector molecule in a drug-dependent manner. Without wishing to be bound by theory, the components of the Membrane-Cleavable Degron system present in the membrane-cleavable chimeric protein generally regulate secretion through the below cellular processes:

    • MT: The cell membrane tethering domain contains a transmembrane domain (or a transmembrane-intracellular domain) that directs cellular-trafficking of the chimeric protein such that the protein is inserted into, or otherwise associated with, a cell membrane (“tethered”)
    • D: In the presence of an immunomodulatory drug (IMiD), the degron directs ubiquitin-mediated degradation of the chimeric protein such that secretion of the effector molecule is reduced or eliminated. In general, expression and localization of the chimeric protein into the cell membrane facilitates degron/ubiquitin-mediated degradation.
    • C: In the absence of an IMiD, degron/ubiquitin-mediated degradation of the chimeric protein does not occur. Following expression and localization of the chimeric protein into the cell membrane, the protease cleavage site directs cleavage of the chimeric protein such that the effector molecule is released (“secreted”) into the extracellular space


In some aspects, degron-fusion chimeric proteins (or engineered nucleic acids encoding the degron-fusion chimeric proteins) are provided for herein having a protein of interest (e.g., any of the effector molecules described herein) and a degron.


An “effector molecule,” refers to a molecule (e.g., a nucleic acid such as DNA or RNA, or a protein (polypeptide) or peptide) that binds to another molecule and modulates the biological activity of that molecule to which it binds. For example, an effector molecule may act as a ligand to increase or decrease enzymatic activity, gene expression, or cell signaling. Thus, in some embodiments, an effector molecule modulates (activates or inhibits) different immunomodulatory mechanisms. By directly binding to and modulating a molecule, an effector molecule may also indirectly modulate a second, downstream molecule.


In certain embodiments described herein (e.g., in general, for all membrane-cleavable chimeric proteins described herein), an effector molecule is a secretable effector molecule (e.g., referred to as “S” in the formula S-C-MT-D for membrane-cleavable chimeric proteins described herein). Non-limiting examples of effector molecules include cytokines, chemokines, enzymes that modulate metabolite levels, growth factors, co-activation molecules, tumor microenvironment modifiers, ligands, peptides, enzymes, antibodies, antibodies or decoy molecules that modulate cytokines, homing molecules, and/or integrins.


The term “modulate” encompasses maintenance of a biological activity, inhibition (partial or complete) of a biological activity, and stimulation/activation (partial or complete) of a biological activity. The term also encompasses decreasing or increasing (e.g., enhancing) a biological activity. Two different effector molecules are considered to “modulate different tumor-mediated immunosuppressive mechanisms” when one effector molecule modulates a tumor-mediated immunosuppressive mechanism (e.g., stimulates T cell signaling) that is different from the tumor-mediated immunosuppressive mechanism modulated by the other effector molecule (e.g., stimulates antigen presentation and/or processing).


Modulation by an effector molecule may be direct or indirect. Direct modulation occurs when an effector molecule binds to another molecule and modulates activity of that molecule. Indirect modulation occurs when an effector molecule binds to another molecule, modulates activity of that molecule, and as a result of that modulation, the activity of yet another molecule (to which the effector molecule is not bound) is modulated.


In some embodiments, modulation of a tumor-mediated immunosuppressive mechanism by at least one effector molecule results in an increase in an immunostimulatory and/or anti-tumor immune response (e.g., systemically or in the tumor microenvironment) by at least 10% (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or 200%). For example, modulation of a tumor-mediated immunosuppressive mechanism may result in an increase in an immunostimulatory and/or anti-tumor immune response by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%. In some embodiments, modulation of a tumor-mediated immunosuppressive mechanism results in an increase in an immunostimulatory and/or anti-tumor immune response 10-20%, 10-30%, 10-40%, 10-50%, 10-60%, 10-70%, 10-80%, 10-90%, 10-100%, 10-200%, 20-30%, 20-40%, 20-50%, 20-60%, 20-70%, 20-80%, 20-90%, 20-100%, 20-200%, 50-60%, 50-70%, 50-80%, 50-90%, 50-100%, or 50-200%. It should be understood that “an increase” in an immunostimulatory and/or anti-tumor immune response, for example, systemically or in a tumor microenvironment, is relative to the immunostimulatory and/or anti-tumor immune response that would otherwise occur, in the absence of the effector molecule(s).


In some embodiments, modulation of a tumor-mediated immunosuppressive mechanism by at least one effector molecule results in an increase in an immunostimulatory and/or anti-tumor immune response (e.g., systemically or in the tumor microenvironment) by at least 2 fold (e.g., 2, 3, 4, 5, 10, 25, 20, 25, 50, or 100 fold). For example, modulation of a tumor-mediated immunosuppressive mechanism may result in an increase in an immunostimulatory and/or anti-tumor immune response by at least 3 fold, at least 5 fold, at least 10 fold, at least 20 fold, at least 50 fold, or at least 100 fold. In some embodiments, modulation of a tumor-mediated immunosuppressive mechanism results in an increase in an immunostimulatory and/or anti-tumor immune response by 2-10, 2-20, 2-30, 2-40, 2-50, 2-60, 2-70, 2-80, 2-90, or 2-100 fold.


Non-limiting examples of immunostimulatory and/or anti-tumor immune mechanisms include T cell signaling, activity and/or recruitment, antigen presentation and/or processing, natural killer cell-mediated cytotoxic signaling, activity and/or recruitment, dendritic cell differentiation and/or maturation, immune cell recruitment, pro-inflammatory macrophage signaling, activity and/or recruitment, stroma degradation, immunostimulatory metabolite production, stimulator of interferon genes (STING) signaling (which increases the secretion of IFN and Th1 polarization, promoting an anti-tumor immune response), and/or Type I interferon signaling. An effector molecule may stimulate at least one (one or more) of the foregoing immunostimulatory mechanisms, thus resulting in an increase in an immunostimulatory response. Changes in the foregoing immunostimulatory and/or anti-tumor immune mechanisms may be assessed, for example, using in vitro assays for T cell proliferation or cytotoxicity, in vitro antigen presentation assays, expression assays (e.g., of particular markers), and/or cell secretion assays (e.g., of cytokines).


In some embodiments, modulation of a tumor-mediated immunosuppressive mechanism by at least one effector molecule results in a decrease in an immunosuppressive response (e.g., systemically or in the tumor microenvironment) by at least 10% (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or 200%). For example, modulation of a tumor-mediated immunosuppressive mechanism may result in a decrease in an immunosuppressive response by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%. In some embodiments, modulation of a tumor-mediated immunosuppressive mechanism results in a decrease in an immunosuppressive response 10-20%, 10-30%, 10-40%, 10-50%, 10-60%, 10-70%, 10-80%, 10-90%, 10-100%, 10-200%, 20-30%, 20-40%, 20-50%, 20-60%, 20-70%, 20-80%, 20-90%, 20-100%, 20-200%, 50-60%, 50-70%, 50-80%, 50-90%, 50-100%, or 50-200%. It should be understood that “a decrease” in an immunosuppressive response, for example, systemically or in a tumor microenvironment, is relative to the immunosuppressive response that would otherwise occur, in the absence of the effector molecule(s).


In some embodiments, modulation of a tumor-mediated immunosuppressive mechanism by at least one effector molecule results in a decrease in an immunosuppressive response (e.g., systemically or in the tumor microenvironment) by at least 2 fold (e.g., 2, 3, 4, 5, 10, 25, 20, 25, 50, or 100 fold). For example, modulation of a tumor-mediated immunosuppressive mechanism may result in a decrease in an immunosuppressive response by at least 3 fold, at least 5 fold, at least 10 fold, at least 20 fold, at least 50 fold, or at least 100 fold. In some embodiments, modulation of a tumor-mediated immunosuppressive mechanism results in a decrease in an immunosuppressive response by 2-10, 2-20, 2-30, 2-40, 2-50, 2-60, 2-70, 2-80, 2-90, or 2-100 fold.


Non-limiting examples of immunosuppressive mechanisms include negative costimulatory signaling, pro-apoptotic signaling of cytotoxic cells (e.g., T cells and/or NK cells), T regulatory (Treg) cell signaling, tumor checkpoint molecule production/maintenance, myeloid-derived suppressor cell signaling, activity and/or recruitment, immunosuppressive factor/metabolite production, and/or vascular endothelial growth factor signaling. An effector molecule may inhibit at least one (one or more) of the foregoing immunosuppressive mechanisms, thus resulting in a decrease in an immunosuppressive response. Changes in the foregoing immunosuppressive mechanisms may be assessed, for example, by assaying for an increase in T cell proliferation and/or an increase in IFNγ production (negative co-stimulatory signaling, Treg cell signaling and/or MDSC); Annexin V/PI flow staining (pro-apoptotic signaling); flow staining for expression, e.g., PDL1 expression (tumor checkpoint molecule production/maintenance); ELISA, LUMINEX®, RNA via qPCR, enzymatic assays, e.g., IDO tryptophan catabolism (immunosuppressive factor/metabolite production); and phosphorylation of PI3K, Akt, p38 (VEGF signaling).


In some embodiments, effector molecules function additively: the effect of two effector molecules, for example, may be equal to the sum of the effect of the two effector molecules functioning separately. In other embodiments, effector molecules function synergistically: the effect of two effector molecules, for example, may be greater than the combined function of the two effector molecules.


Effector molecules that modulate tumor-mediated immunosuppressive mechanisms and/or modify tumor microenvironments may be, for example, secreted factors (e.g., cytokines, chemokines, antibodies, and/or decoy receptors that modulate extracellular mechanisms involved in the immune system), inhibitors (e.g., antibodies, antibody fragments, ligand TRAP and/or small blocking peptides), intracellular factors that control cell state (e.g., microRNAs and/or transcription factors that modulate the state of cells to enhance pro-inflammatory properties), factors packaged into exosomes (e.g., microRNAs, cytosolic factors, and/or extracellular factors), surface displayed factors (e.g., checkpoint inhibitors, TRAIL), and and/or metabolic genes (e.g., enzymes that produce/modulate or degrade metabolites or amino acids).


In some embodiments, at least one of the effector molecules stimulates an immunostimulatory mechanism in the tumor microenvironment and/or inhibits an immunosuppressive mechanism in the tumor microenvironment.


In some embodiments, at least one of the effector molecules (a) stimulates T cell signaling, activity and/or recruitment, (b) stimulates antigen presentation and/or processing, (c) stimulates natural killer cell-mediated cytotoxic signaling, activity and/or recruitment, (d) stimulates dendritic cell differentiation and/or maturation, (e) stimulates immune cell recruitment, (f) stimulates pro-inflammatory macrophage signaling, activity and/or recruitment or inhibits anti-inflammatory macrophage signaling, activity and/or recruitment, (g) stimulates stroma degradation, (h) stimulates immunostimulatory metabolite production, (i) stimulates Type I interferon signaling, (j) inhibits negative costimulatory signaling, (k) inhibits pro-apoptotic signaling of anti-tumor immune cells, (l) inhibits T regulatory (Treg) cell signaling, activity and/or recruitment, (m) inhibits tumor checkpoint molecules, (n) stimulates stimulator of interferon genes (STING) signaling, (o) inhibits myeloid-derived suppressor cell signaling, activity and/or recruitment, (p) degrades immunosuppressive factors/metabolites, (q) inhibits vascular endothelial growth factor signaling, and/or (r) directly kills tumor cells.


In some embodiments, effector molecules may be selected from the following non-limiting classes of molecules: cytokines, antibodies, chemokines, nucleotides, peptides, and enzymes. Non-limiting examples of the foregoing classes of effector molecules are listed in Table 1 and specific sequences encoding exemplary effector molecules are listed in Table 2. Effector molecules can be human, such as those listed in Table 1 or Table 2 or human equivalents of murine effector molecules listed in Table 1 or Table 2. Effector molecules can be human-derived, such as the endogenous human effector molecule or an effector molecule modified and/or optimized for function, e.g., codon optimized to improve expression, modified to improve stability, or modified at its signal sequence (see below). Various programs and algorithms for optimizing function are known to those skilled in the art and can be selected based on the improvement desired, such as codon optimization for a specific species (e.g., human, mouse, bacteria, etc.).









TABLE 1







Exemplary Effector Molecules









Effector name
Category
Function





anti-CD40 or CD40
Agonist antibody
Stimulates B-cells and antigen


Ligand

presenting cells.


Flt3L
Ligand agonist
Stimulates myeloid cells and




antigen presenting cells


CXCL10-11 fusion
Chemokine
Attracts T-cells


TGFb blocking
Antagonist
Inhibit TGFb pathway, TME


peptides
peptides
modifier


Adenosine
TME modifier
Degradation of suppressive


deaminase (ADA)

adenosine in the TME


Kyneurinase
TME modifier
Degradation of kyneurine


HPGE2
TME modifier
Degradation of PGE2


CXCL13
Chemokine
Attracts B-cells


anti PD-1/PD-L1
Agonist antibody
Remove checkpoint


anti-CTLA-4
Agonist antibody
Remove checkpoint


anti-VEGF
Antagonist
Neutralizes an immuno-



antibody
suppressive/angiogenesis factor


anti-TNFa
Antagonist
Neutralizes cytokine/pro-tumor



antibody
factor


anti-IL-10
Antagonist
Neutralizes immunosuppressive



antibody
cytokine


anti-SDF1/CXCL12
Antagonist
Neutralizes pro-tumor chemokine



antibody



(TβRII)2 trap
Capture trap
Neutralizes an immuno-




suppressive cytokine


CCL21
Chemokine
Attracts leukocytes/NK


CCL1
Chemokine
Attracts leukocytes/NK


CCL17
Chemokine
Attracts leukocytes/NK


CCL19
Chemokine
Attracts leukocytes/NK


CCL21
Chemokine
Attracts leukocytes/NK


CCL20
Chemokine
Attracts leukocytes/NK


CCL21a
Chemokine
Attracts leukocytes/NK


MIP1b (CCL5)
Chemokine
Attracts leukocytes/NK


CXCL10
Chemokine
Attracts leukocytes/NK


CXCL11
Chemokine
Attracts leukocytes/NK


CCL2
Chemokine
Attracts monocytes


MIP-1 alpha (CCL3)
Chemokine
Attracts leukocytes/NK


XCL1
Chemokine
Attracts leukocytes/NK


IFNbeta
Cytokine
T cell response, tumor cell killing


IFNgamma
Cytokine
T cell response, tumor cell killing


IL-12
Cytokine
T cells, NK cells


IL-1beta
Cytokine
T cells, NK cells


IL-15
Cytokine
Stimulates T-cells and NK


IL-2
Cytokine
Stimulates T-cells and NK


IL-21
Cytokine
Stimulates T-cells


IL-24
Cytokine
Stimulates T-cells


IL36-gamma
Cytokine
Stimulates T-cells


IL-7
Cytokine
Stimulates T-cells


IL-22
Cytokine
Stimulates T-cells


IL-18
Cytokine
Stimulates T-cells


Granzymes/Perforin
Enzyme
Direct tumor cell killing


OX86 (anti-OX40)
Ligand
Stimulates T-cells


anti-TGFbeta
Neutralizing
Neutralizes an Immuno-



antibody
suppressive cytokine


TRAIL
Receptor/ligand
Direct tumor cell killing


FASL (CD49L)
Receptor/ligand
Direct tumor cell killing


OX40-L
Receptor/Ligand
Stimulates T-cells


cGAS
secreted molecule
Stimulates antigen-presenting




cells


41BBL
secreted molecule
Co-activation of T-cells


CD40L
secreted molecule
Stimulates T-cells


GM-CSF
secreted molecule
Growth factor for monocytes


STING
secreted molecule
Stimulates antigen-presenting




cells


HAC-V
Antagonist
inhibits checkpoint


‘microbody’_PD1
antibody



yCD
Pro-drug
Converts to cytotoxic molecule




upon activation


CpG/Nucleotides
Nucleotides
STING agonist
















TABLE 2





Sequences encoding exemplary


effector molecules

















IL12 (Human)



(SEQ ID NO: 56)



ATGTGCCATCAGCAGCTTGTCATATCTTGGTTTTCACTTG



TATTCCTGGCCAGCCCTTTGGTTGCGATCTGGGAGCTCAA



GAAGGATGTGTACGTTGTAGAGCTGGACTGGTACCCCGAT



GCTCCCGGTGAGATGGTCGTTTTGACATGTGACACTCCAG



AAGAGGACGGTATTACGTGGACTCTGGACCAGTCCTCCGA



AGTTCTTGGTTCTGGTAAGACTCTGACTATCCAGGTGAAA



GAATTTGGGGATGCGGGACAATACACATGCCACAAGGGAG



GCGAGGTGTTGTCTCATAGTTTGCTGCTTCTCCACAAGAA



AGAGGATGGAATCTGGAGCACCGACATACTCAAGGATCAA



AAGGAACCCAAAAATAAGACATTTCTGCGATGTGAGGCTA



AGAACTATAGTGGCCGCTTCACTTGTTGGTGGCTGACTAC



CATCAGCACAGATCTCACGTTTTCAGTAAAAAGTAGTAGA



GGTTCAAGTGATCCTCAAGGGGTAACGTGCGGTGCTGCAA



CACTGTCTGCTGAACGCGTAAGAGGAGATAATAAGGAGTA



CGAGTATTCCGTAGAATGCCAAGAGGACAGTGCTTGTCCT



GCGGCCGAGGAGTCTCTCCCAATAGAAGTGATGGTGGACG



CGGTGCATAAACTGAAATATGAGAACTACACAAGCAGTTT



TTTTATAAGAGATATCATCAAGCCCGATCCGCCGAAGAAT



TTGCAACTTAAACCGCTTAAAAACTCACGCCAGGTTGAAG



TATCCTGGGAGTATCCGGATACATGGTCAACACCACACAG



CTATTTTTCCCTTACCTTCTGTGTGCAGGTCCAAGGGAAG



AGCAAAAGGGAGAAGAAGGACAGGGTATTCACTGATAAAA



CTTCCGCGACGGTCATCTGCCGAAAAAACGCTAGTATATC



TGTACGGGCGCAGGATAGGTACTATAGTTCTTCTTGGTCT



GAGTGGGCCTCAGTTCCGTGCTCTGGGGGAGGAAGTGGAG



GAGGGTCCGGCGGTGGAAGCGGGGGAGGGAGTCGCAACTT



GCCAGTGGCTACACCAGATCCAGGCATGTTTCCATGTCTG



CATCATTCCCAGAATCTCCTGAGAGCGGTGTCAAATATGC



TCCAAAAAGCGAGACAAACACTGGAATTTTACCCGTGTAC



CAGTGAGGAGATTGATCACGAGGACATAACCAAGGACAAG



ACCTCAACTGTAGAAGCGTGTTTGCCGCTGGAGTTGACTA



AGAATGAGTCCTGCCTCAATTCCAGAGAAACTTCATTCAT



TACTAACGGCAGTTGTCTTGCATCCCGGAAAACGTCCTTT



ATGATGGCCCTTTGCCTTAGTTCAATTTACGAGGATCTTA



AAATGTATCAAGTGGAGTTTAAAACCATGAATGCTAAACT



TCTTATGGACCCCAAACGACAAATTTTTCTGGATCAGAAT



ATGCTTGCCGTGATAGACGAACTCATGCAGGCGCTTAATT



TTAACTCCGAAACAGTTCCACAAAAATCTAGCCTTGAAGA



ACCTGATTTTTATAAAACGAAGATTAAACTGTGTATCCTG



CTGCATGCCTTTCGCATCCGAGCTGTCACAATCGATAGGG



TTATGTCCTACCTTAACGCGAGCtaG







IL12p70 (Human; codon optimized;



bold denotes signal sequence)



(SEQ ID NO: 57)




ATGTGCCATCAGCAACTCGTCATCTCCTGGTTCTCCCTTG





TGTTCCTCGCTTCCCCTCTGGTCGCCATTTGGGAACTGAA




GAAGGACGTCTACGTGGTCGAGCTGGATTGGTACCCGGAC



GCCCCTGGAGAAATGGTCGTGCTGACTTGCGATACGCCAG



AAGAGGACGGCATAACCTGGACCCTGGATCAGAGCTCCGA



GGTGCTCGGAAGCGGAAAGACCCTGACCATTCAAGTCAAG



GAGTTCGGCGACGCGGGCCAGTACACTTGCCACAAGGGTG



GCGAAGTGCTGTCCCACTCCCTGCTGCTGCTGCACAAGAA



AGAGGATGGAATCTGGTCCACTGACATCCTCAAGGACCAA



AAAGAACCGAAGAACAAGACCTTCCTCCGCTGCGAAGCCA



AGAACTACAGCGGTCGGTTCACCTGTTGGTGGCTGACGAC



AATCTCCACCGACCTGACTTTCTCCGTGAAGTCGTCACGG



GGATCAAGCGATCCTCAGGGCGTGACCTGTGGAGCCGCCA



CTCTGTCCGCCGAGAGAGTCAGGGGAGACAACAAGGAATA



TGAGTACTCCGTGGAATGCCAGGAGGACAGCGCCTGCCCT



GCCGCGGAAGAGTCCCTGCCTATCGAGGTCATGGTCGATG



CCGTGCATAAGCTGAAATACGAGAACTACACTTCCTCCTT



CTTTATCCGCGACATCATCAAGCCTGACCCCCCCAAGAAC



TTGCAGCTGAAGCCACTCAAGAACTCCCGCCAAGTGGAAG



TGTCTTGGGAATATCCAGACACTTGGAGCACCCCGCACTC



ATACTTCTCGCTCACTTTCTGTGTGCAAGTGCAGGGAAAG



TCCAAACGGGAGAAGAAAGACCGGGTGTTCACCGACAAAA



CCTCCGCCACTGTGATTTGTCGGAAGAACGCGTCAATCAG



CGTCCGGGCGCAGGATAGATACTACTCGTCCTCCTGGAGC



GAATGGGCCAGCGTGCCTTGTTCCGGTGGCGGATCAGGCG



GAGGTTCAGGAGGAGGCTCCGGAGGAGGTTCCCGGAACCT



CCCTGTGGCAACCCCCGACCCTGGAATGTTCCCGTGCCTA



CACCACTCCCAAAACCTCCTGAGGGCTGTGTCGAACATGT



TGCAGAAGGCCCGCCAGACCCTTGAGTTCTACCCCTGCAC



CTCGGAAGAAATTGATCACGAGGACATCACCAAGGACAAG



ACCTCGACCGTGGAAGCCTGCCTGCCGCTGGAACTGACCA



AGAACGAATCGTGTCTGAACTCCCGCGAGACAAGCTTTAT



CACTAACGGCAGCTGCCTGGCGTCGAGAAAGACCTCATTC



ATGATGGCGCTCTGTCTTTCCTCGATCTACGAAGATCTGA



AGATGTATCAGGTCGAGTTCAAGACCATGAACGCCAAGCT



GCTCATGGACCCGAAGCGGCAGATCTTCCTGGACCAGAAT



ATGCTCGCCGTGATTGATGAACTGATGCAGGCCCTGAATT



TCAACTCCGAGACTGTGCCTCAAAAGTCCAGCCTGGAAGA



ACCGGACTTCTACAAGACCAAGATCAAGCTGTGCATCCTG



TTGCACGCTTTCCGCATTCGAGCCGTGACCATTGACCGCG



TGATGTCCTACCTGAACGCCAGT







IL12 (Mouse)



(SEQ ID NO: 58)



ATGTGTCCACAGAAGCTGACAATAAGTTGGTTTGCCATTG



TCCTCCTGGTGAGCCCACTCATGGCAATGTGGGAACTCGA



AAAGGATGTCTACGTGGTAGAAGTAGATTGGACTCCAGAC



GCGCCAGGGGAGACAGTGAATTTGACATGTGACACACCAG



AAGAAGATGACATTACATGGACATCTGACCAACGCCATGG



CGTAATAGGGAGTGGGAAAACACTCACGATCACAGTTAAA



GAGTTCTTGGATGCTGGTCAATATACTTGCCATAAAGGCG



GCGAGACACTCAGCCACTCACATTTGCTTTTGCATAAAAA



AGAGAATGGCATTTGGAGCACTGAAATACTTAAGAACTTT



AAGAACAAGACATTTCTCAAGTGTGAGGCCCCTAATTACA



GCGGCAGGTTCACGTGCTCATGGCTGGTCCAGCGCAACAT



GGACCTCAAGTTTAACATAAAATCTTCTTCCTCTTCACCT



GACTCCAGAGCTGTTACTTGCGGCATGGCTTCTCTGAGCG



CAGAAAAAGTAACGTTGGATCAAAGAGACTACGAAAAGTA



CTCTGTTTCTTGTCAAGAGGATGTTACGTGCCCGACGGCC



GAAGAAACGCTTCCAATTGAACTCGCGTTGGAAGCTCGCC



AACAAAACAAGTATGAAAACTACAGTACAAGCTTCTTTAT



ACGGGATATAATTAAACCCGATCCCCCCAAGAACTTGCAA



ATGAAACCACTTAAGAACAGCCAGGTGGAAGTTTCCTGGG



AGTATCCAGACTCATGGAGTACTCCTCACAGCTATTTTTC



TCTGAAATTCTTTGTAAGGATACAACGGAAGAAAGAGAAG



ATGAAAGAGACCGAGGAGGGTTGTAATCAGAAGGGAGCGT



TTCTCGTGGAGAAAACGTCTACCGAAGTCCAATGTAAAGG



TGGCAATGTGTGCGTCCAAGCTCAGGATAGATACTATAAT



TCAAGTTGCTCCAAGTGGGCCTGTGTTCCATGCCGCGTTC



GGAGCGGGGGAGGTAGCGGAGGAGGTAGTGGGGGTGGGTC



AGGAGGAGGGAGTCGAGTTATCCCGGTGTCAGGCCCCGCA



CGCTGCTTGAGCCAGAGTCGCAACCTCCTTAAGACAACAG



ATGACATGGTGAAAACAGCACGCGAAAAGCTTAAACACTA



CTCTTGTACGGCGGAGGATATTGATCACGAGGATATTACC



CGAGACCAAACTAGCACTTTGAAAACCTGTCTGCCCCTTG



AACTTCATAAAAATGAGAGCTGTCTGGCTACACGAGAGAC



GTCAAGTACGACTAGGGGCAGCTGTCTCCCGCCGCAAAAG



ACAAGCCTCATGATGACGCTCTGTTTGGGTTCCATTTACG



AGGACTTGAAAATGTATCAAACGGAGTTCCAGGCTATAAA



TGCGGCGTTGCAGAACCATAACCATCAACAAATTATACTT



GATAAAGGCATGTTGGTGGCGATTGATGAACTCATGCAGA



GTCTCAATCACAACGGGGAAACGTTGAGACAGAAACCCCC



AGTCGGTGAAGCGGACCCATATCGAGTAAAAATGAAGCTC



TGCATTCTGCTTCACGCATTCAGCACTAGAGTTGTTACCA



TCAACCGGGTAATGGGATATCTCTCCAGTGCGtaG







IL21 (Human; codon optimized;



bold denotes signal sequence)



(SEQ ID NO: 59)




ATGGAACGCATTGTGATCTGCCTGATGGTCATCTTCCTGG





GCACCTTAGTGCACAAGTCGAGCAGCCAGGGACAGGACAG




GCACATGATTAGAATGCGCCAGCTCATCGATATCGTGGAC



CAGTTGAAGAACTACGTGAACGACCTGGTGCCCGAGTTCC



TGCCGGCCCCCGAAGATGTGGAAACCAATTGCGAATGGTC



GGCATTTTCCTGCTTTCAAAAGGCACAGCTCAAGTCCGCT



AACACCGGGAACAACGAACGGATCATCAACGTGTCCATCA



AAAAGCTGAAGCGGAAGCCTCCCTCCACCAACGCCGGACG



GAGGCAGAAGCATAGGCTGACTTGCCCGTCATGCGACTCC



TACGAGAAGAAGCCGCCGAAGGAGTTCCTGGAGCGGTTCA



AGTCGCTCCTGCAAAAGATGATTCATCAGCACCTGTCCTC



CCGGACTCATGGGTCTGAGGATTCA







IL12p70_T2A_IL21 (Human; codon optimized;



bold denotes signal sequences)



(SEQ ID NO: 60)




ATGTGCCATCAGCAACTCGTCATCTCCTGGTTCTCCCTTG





TGTTCCTCGCTTCCCCTCTGGTCGCCATTTGGGAACTGAA




GAAGGACGTCTACGTGGTCGAGCTGGATTGGTACCCGGAC



GCCCCTGGAGAAATGGTCGTGCTGACTTGCGATACGCCAG



AAGAGGACGGCATAACCTGGACCCTGGATCAGAGCTCCGA



GGTGCTCGGAAGCGGAAAGACCCTGACCATTCAAGTCAAG



GAGTTCGGCGACGCGGGCCAGTACACTTGCCACAAGGGTG



GCGAAGTGCTGTCCCACTCCCTGCTGCTGCTGCACAAGAA



AGAGGATGGAATCTGGTCCACTGACATCCTCAAGGACCAA



AAAGAACCGAAGAACAAGACCTTCCTCCGCTGCGAAGCCA



AGAACTACAGCGGTCGGTTCACCTGTTGGTGGCTGACGAC



AATCTCCACCGACCTGACTTTCTCCGTGAAGTCGTCACGG



GGATCAAGCGATCCTCAGGGCGTGACCTGTGGAGCCGCCA



CTCTGTCCGCCGAGAGAGTCAGGGGAGACAACAAGGAATA



TGAGTACTCCGTGGAATGCCAGGAGGACAGCGCCTGCCCT



GCCGCGGAAGAGTCCCTGCCTATCGAGGTCATGGTCGATG



CCGTGCATAAGCTGAAATACGAGAACTACACTTCCTCCTT



CTTTATCCGCGACATCATCAAGCCTGACCCCCCCAAGAAC



TTGCAGCTGAAGCCACTCAAGAACTCCCGCCAAGTGGAAG



TGTCTTGGGAATATCCAGACACTTGGAGCACCCCGCACTC



ATACTTCTCGCTCACTTTCTGTGTGCAAGTGCAGGGAAAG



TCCAAACGGGAGAAGAAAGACCGGGTGTTCACCGACAAAA



CCTCCGCCACTGTGATTTGTCGGAAGAACGCGTCAATCAG



CGTCCGGGCGCAGGATAGATACTACTCGTCCTCCTGGAGC



GAATGGGCCAGCGTGCCTTGTTCCGGTGGCGGATCAGGCG



GAGGTTCAGGAGGAGGCTCCGGAGGAGGTTCCCGGAACCT



CCCTGTGGCAACCCCCGACCCTGGAATGTTCCCGTGCCTA



CACCACTCCCAAAACCTCCTGAGGGCTGTGTCGAACATGT



TGCAGAAGGCCCGCCAGACCCTTGAGTTCTACCCCTGCAC



CTCGGAAGAAATTGATCACGAGGACATCACCAAGGACAAG



ACCTCGACCGTGGAAGCCTGCCTGCCGCTGGAACTGACCA



AGAACGAATCGTGTCTGAACTCCCGCGAGACAAGCTTTAT



CACTAACGGCAGCTGCCTGGCGTCGAGAAAGACCTCATTC



ATGATGGCGCTCTGTCTTTCCTCGATCTACGAAGATCTGA



AGATGTATCAGGTCGAGTTCAAGACCATGAACGCCAAGCT



GCTCATGGACCCGAAGCGGCAGATCTTCCTGGACCAGAAT



ATGCTCGCCGTGATTGATGAACTGATGCAGGCCCTGAATT



TCAACTCCGAGACTGTGCCTCAAAAGTCCAGCCTGGAAGA



ACCGGACTTCTACAAGACCAAGATCAAGCTGTGCATCCTG



TTGCACGCTTTCCGCATTCGAGCCGTGACCATTGACCGCG



TGATGTCCTACCTGAACGCCAGTAGACGGAAACGCGGAAG



CGGAGAGGGCAGAGGCTCGCTGCTTACATGCGGGGACGTG



GAAGAGAACCCCGGTCCGATGGAACGCATTGTGATCTGCC




TGATGGTCATCTTCCTGGGCACCTTAGTGCACAAGTCGAG





CAGCCAGGGACAGGACAGGCACATGATTAGAATGCGCCAG




CTCATCGATATCGTGGACCAGTTGAAGAACTACGTGAACG



ACCTGGTGCCCGAGTTCCTGCCGGCCCCCGAAGATGTGGA



AACCAATTGCGAATGGTCGGCATTTTCCTGCTTTCAAAAG



GCACAGCTCAAGTCCGCTAACACCGGGAACAACGAACGGA



TCATCAACGTGTCCATCAAAAAGCTGAAGCGGAAGCCTCC



CTCCACCAACGCCGGACGGAGGCAGAAGCATAGGCTGACT



TGCCCGTCATGCGACTCCTACGAGAAGAAGCCGCCGAAGG



AGTTCCTGGAGCGGTTCAAGTCGCTCCTGCAAAAGATGAT



TCATCAGCACCTGTCCTCCCGGACTCATGGGTCTGAGGAT



TCA







IL-12_2A_CCL21a (Human)



(SEQ ID NO: 61)



ATGTGCCATCAGCAGCTTGTCATATCTTGGTTTTCACTTG



TATTCCTGGCCAGCCCTTTGGTTGCGATCTGGGAGCTCAA



GAAGGATGTGTACGTTGTAGAGCTGGACTGGTACCCCGAT



GCTCCCGGTGAGATGGTCGTTTTGACATGTGACACTCCAG



AAGAGGACGGTATTACGTGGACTCTGGACCAGTCCTCCGA



AGTTCTTGGTTCTGGTAAGACTCTGACTATCCAGGTGAAA



GAATTTGGGGATGCGGGACAATACACATGCCACAAGGGAG



GCGAGGTGTTGTCTCATAGTTTGCTGCTTCTCCACAAGAA



AGAGGATGGAATCTGGAGCACCGACATACTCAAGGATCAA



AAGGAACCCAAAAATAAGACATTTCTGCGATGTGAGGCTA



AGAACTATAGTGGCCGCTTCACTTGTTGGTGGCTGACTAC



CATCAGCACAGATCTCACGTTTTCAGTAAAAAGTAGTAGA



GGTTCAAGTGATCCTCAAGGGGTAACGTGCGGTGCTGCAA



CACTGTCTGCTGAACGCGTAAGAGGAGATAATAAGGAGTA



CGAGTATTCCGTAGAATGCCAAGAGGACAGTGCTTGTCCT



GCGGCCGAGGAGTCTCTCCCAATAGAAGTGATGGTGGACG



CGGTGCATAAACTGAAATATGAGAACTACACAAGCAGTTT



TTTTATAAGAGATATCATCAAGCCCGATCCGCCGAAGAAT



TTGCAACTTAAACCGCTTAAAAACTCACGCCAGGTTGAAG



TATCCTGGGAGTATCCGGATACATGGTCAACACCACACAG



CTATTTTTCCCTTACCTTCTGTGTGCAGGTCCAAGGGAAG



AGCAAAAGGGAGAAGAAGGACAGGGTATTCACTGATAAAA



CTTCCGCGACGGTCATCTGCCGAAAAAACGCTAGTATATC



TGTACGGGCGCAGGATAGGTACTATAGTTCTTCTTGGTCT



GAGTGGGCCTCAGTTCCGTGCTCTGGGGGAGGAAGTGGAG



GAGGGTCCGGCGGTGGAAGCGGGGGAGGGAGTCGCAACTT



GCCAGTGGCTACACCAGATCCAGGCATGTTTCCATGTCTG



CATCATTCCCAGAATCTCCTGAGAGCGGTGTCAAATATGC



TCCAAAAAGCGAGACAAACACTGGAATTTTACCCGTGTAC



CAGTGAGGAGATTGATCACGAGGACATAACCAAGGACAAG



ACCTCAACTGTAGAAGCGTGTTTGCCGCTGGAGTTGACTA



AGAATGAGTCCTGCCTCAATTCCAGAGAAACTTCATTCAT



TACTAACGGCAGTTGTCTTGCATCCCGGAAAACGTCCTTT



ATGATGGCCCTTTGCCTTAGTTCAATTTACGAGGATCTTA



AAATGTATCAAGTGGAGTTTAAAACCATGAATGCTAAACT



TCTTATGGACCCCAAACGACAAATTTTTCTGGATCAGAAT



ATGCTTGCCGTGATAGACGAACTCATGCAGGCGCTTAATT



TTAACTCCGAAACAGTTCCACAAAAATCTAGCCTTGAAGA



ACCTGATTTTTATAAAACGAAGATTAAACTGTGTATCCTG



CTGCATGCCTTTCGCATCCGAGCTGTCACAATCGATAGGG



TTATGTCCTACCTTAACGCGAGCCGGCGCAAGAGGGGTTC



CGGAGAGGGAAGGGGTAGTCTGCTCACCTGCGGCGATGTT



GAAGAAAATCCTGGTCCCATGGCGCAAAGTCTGGCTCTTT



CACTCCTGATCCTGGTCTTGGCCTTCGGGATTCCGAGGAC



CCAAGGAAGTGATGGTGGCGCCCAAGATTGTTGCCTTAAA



TACAGCCAGCGGAAAATACCCGCGAAAGTGGTCAGGAGTT



ATAGAAAACAGGAGCCTTCCCTGGGTTGTAGTATCCCCGC



CATACTTTTCCTCCCGAGAAAACGGAGCCAGGCCGAACTG



TGCGCTGACCCTAAGGAACTTTGGGTGCAACAACTTATGC



AACACCTGGATAAGACACCTTCTCCTCAAAAGCCAGCTCA



GGGCTGCCGAAAAGATAGAGGCGCCTCAAAAACCGGAAAA



AAGGGCAAAGGTTCTAAAGGATGTAAGCGGACTGAACGCT



CTCAAACGCCTAAAGGGCCGtaG







IL-12_2A_CCL21a (Mouse)



(SEQ ID NO: 62)



ATGTGTCCACAGAAGCTGACAATAAGTTGGTTTGCCATTG



TCCTCCTGGTGAGCCCACTCATGGCAATGTGGGAACTCGA



AAAGGATGTCTACGTGGTAGAAGTAGATTGGACTCCAGAC



GCGCCAGGGGAGACAGTGAATTTGACATGTGACACACCAG



AAGAAGATGACATTACATGGACATCTGACCAACGCCATGG



CGTAATAGGGAGTGGGAAAACACTCACGATCACAGTTAAA



GAGTTCTTGGATGCTGGTCAATATACTTGCCATAAAGGCG



GCGAGACACTCAGCCACTCACATTTGCTTTTGCATAAAAA



AGAGAATGGCATTTGGAGCACTGAAATACTTAAGAACTTT



AAGAACAAGACATTTCTCAAGTGTGAGGCCCCTAATTACA



GCGGCAGGTTCACGTGCTCATGGCTGGTCCAGCGCAACAT



GGACCTCAAGTTTAACATAAAATCTTCTTCCTCTTCACCT



GACTCCAGAGCTGTTACTTGCGGCATGGCTTCTCTGAGCG



CAGAAAAAGTAACGTTGGATCAAAGAGACTACGAAAAGTA



CTCTGTTTCTTGTCAAGAGGATGTTACGTGCCCGACGGCC



GAAGAAACGCTTCCAATTGAACTCGCGTTGGAAGCTCGCC



AACAAAACAAGTATGAAAACTACAGTACAAGCTTCTTTAT



ACGGGATATAATTAAACCCGATCCCCCCAAGAACTTGCAA



ATGAAACCACTTAAGAACAGCCAGGTGGAAGTTTCCTGGG



AGTATCCAGACTCATGGAGTACTCCTCACAGCTATTTTTC



TCTGAAATTCTTTGTAAGGATACAACGGAAGAAAGAGAAG



ATGAAAGAGACCGAGGAGGGTTGTAATCAGAAGGGAGCGT



TTCTCGTGGAGAAAACGTCTACCGAAGTCCAATGTAAAGG



TGGCAATGTGTGCGTCCAAGCTCAGGATAGATACTATAAT



TCAAGTTGCTCCAAGTGGGCCTGTGTTCCATGCCGCGTTC



GGAGCGGGGGAGGTAGCGGAGGAGGTAGTGGGGGTGGGTC



AGGAGGAGGGAGTCGAGTTATCCCGGTGTCAGGCCCCGCA



CGCTGCTTGAGCCAGAGTCGCAACCTCCTTAAGACAACAG



ATGACATGGTGAAAACAGCACGCGAAAAGCTTAAACACTA



CTCTTGTACGGCGGAGGATATTGATCACGAGGATATTACC



CGAGACCAAACTAGCACTTTGAAAACCTGTCTGCCCCTTG



AACTTCATAAAAATGAGAGCTGTCTGGCTACACGAGAGAC



GTCAAGTACGACTAGGGGCAGCTGTCTCCCGCCGCAAAAG



ACAAGCCTCATGATGACGCTCTGTTTGGGTTCCATTTACG



AGGACTTGAAAATGTATCAAACGGAGTTCCAGGCTATAAA



TGCGGCGTTGCAGAACCATAACCATCAACAAATTATACTT



GATAAAGGCATGTTGGTGGCGATTGATGAACTCATGCAGA



GTCTCAATCACAACGGGGAAACGTTGAGACAGAAACCCCC



AGTCGGTGAAGCGGACCCATATCGAGTAAAAATGAAGCTC



TGCATTCTGCTTCACGCATTCAGCACTAGAGTTGTTACCA



TCAACCGGGTAATGGGATATCTCTCCAGTGCGCGGCGCAA



GAGGGGTTCCGGAGAGGGAAGGGGTAGTCTGCTCACCTGC



GGCGATGTTGAAGAAAATCCTGGTCCCATGGCGCAAATGA



TGACCCTTTCCCTGCTGAGTCTTGTCCTCGCGCTCTGCAT



CCCGTGGACGCAGGGGTCTGATGGGGGGGGCCAAGACTGT



TGCCTGAAGTATTCACAAAAAAAGATACCGTACTCTATTG



TCAGAGGGTACAGGAAGCAAGAACCCTCCTTGGGTTGCCC



TATACCAGCAATTCTTTTCTCCCCACGCAAGCATTCCAAA



CCAGAACTGTGTGCGAACCCCGAGGAGGGTTGGGTACAGA



ACTTGATGCGAAGGCTTGACCAGCCCCCAGCCCCTGGCAA



GCAGTCACCTGGGTGCAGAAAAAACAGAGGTACTTCAAAG



AGCGGCAAGAAAGGCAAAGGGAGTAAAGGATGTAAAAGAA



CGGAGCAGACCCAGCCTTCACGAGGCtaG







CCL21a_2A_IL-12 (Mouse)



(SEQ ID NO: 63)



ATGGCGCAAATGATGACCCTTTCCCTGCTGAGTCTTGTCC



TCGCGCTCTGCATCCCGTGGACGCAGGGGTCTGATGGGGG



GGGCCAAGACTGTTGCCTGAAGTATTCACAAAAAAAGATA



CCGTACTCTATTGTCAGAGGGTACAGGAAGCAAGAACCCT



CCTTGGGTTGCCCTATACCAGCAATTCTTTTCTCCCCACG



CAAGCATTCCAAACCAGAACTGTGTGCGAACCCCGAGGAG



GGTTGGGTACAGAACTTGATGCGAAGGCTTGACCAGCCCC



CAGCCCCTGGCAAGCAGTCACCTGGGTGCAGAAAAAACAG



AGGTACTTCAAAGAGCGGCAAGAAAGGCAAAGGGAGTAAA



GGATGTAAAAGAACGGAGCAGACCCAGCCTTCACGAGGCC



GGCGCAAGAGGGGTTCCGGAGAGGGAAGGGGTAGTCTGCT



CACCTGCGGCGATGTTGAAGAAAATCCTGGTCCCATGTGT



CCACAGAAGCTGACAATAAGTTGGTTTGCCATTGTCCTCC



TGGTGAGCCCACTCATGGCAATGTGGGAACTCGAAAAGGA



TGTCTACGTGGTAGAAGTAGATTGGACTCCAGACGCGCCA



GGGGAGACAGTGAATTTGACATGTGACACACCAGAAGAAG



ATGACATTACATGGACATCTGACCAACGCCATGGCGTAAT



AGGGAGTGGGAAAACACTCACGATCACAGTTAAAGAGTTC



TTGGATGCTGGTCAATATACTTGCCATAAAGGCGGCGAGA



CACTCAGCCACTCACATTTGCTTTTGCATAAAAAAGAGAA



TGGCATTTGGAGCACTGAAATACTTAAGAACTTTAAGAAC



AAGACATTTCTCAAGTGTGAGGCCCCTAATTACAGCGGCA



GGTTCACGTGCTCATGGCTGGTCCAGCGCAACATGGACCT



CAAGTTTAACATAAAATCTTCTTCCTCTTCACCTGACTCC



AGAGCTGTTACTTGCGGCATGGCTTCTCTGAGCGCAGAAA



AAGTAACGTTGGATCAAAGAGACTACGAAAAGTACTCTGT



TTCTTGTCAAGAGGATGTTACGTGCCCGACGGCCGAAGAA



ACGCTTCCAATTGAACTCGCGTTGGAAGCTCGCCAACAAA



ACAAGTATGAAAACTACAGTACAAGCTTCTTTATACGGGA



TATAATTAAACCCGATCCCCCCAAGAACTTGCAAATGAAA



CCACTTAAGAACAGCCAGGTGGAAGTTTCCTGGGAGTATC



CAGACTCATGGAGTACTCCTCACAGCTATTTTTCTCTGAA



ATTCTTTGTAAGGATACAACGGAAGAAAGAGAAGATGAAA



GAGACCGAGGAGGGTTGTAATCAGAAGGGAGCGTTTCTCG



TGGAGAAAACGTCTACCGAAGTCCAATGTAAAGGTGGCAA



TGTGTGCGTCCAAGCTCAGGATAGATACTATAATTCAAGT



TGCTCCAAGTGGGCCTGTGTTCCATGCCGCGTTCGGAGCG



GGGGAGGTAGCGGAGGAGGTAGTGGGGGTGGGTCAGGAGG



AGGGAGTCGAGTTATCCCGGTGTCAGGCCCCGCACGCTGC



TTGAGCCAGAGTCGCAACCTCCTTAAGACAACAGATGACA



TGGTGAAAACAGCACGCGAAAAGCTTAAACACTACTCTTG



TACGGCGGAGGATATTGATCACGAGGATATTACCCGAGAC



CAAACTAGCACTTTGAAAACCTGTCTGCCCCTTGAACTTC



ATAAAAATGAGAGCTGTCTGGCTACACGAGAGACGTCAAG



TACGACTAGGGGCAGCTGTCTCCCGCCGCAAAAGACAAGC



CTCATGATGACGCTCTGTTTGGGTTCCATTTACGAGGACT



TGAAAATGTATCAAACGGAGTTCCAGGCTATAAATGCGGC



GTTGCAGAACCATAACCATCAACAAATTATACTTGATAAA



GGCATGTTGGTGGCGATTGATGAACTCATGCAGAGTCTCA



ATCACAACGGGGAAACGTTGAGACAGAAACCCCCAGTCGG



TGAAGCGGACCCATATCGAGTAAAAATGAAGCTCTGCATT



CTGCTTCACGCATTCAGCACTAGAGTTGTTACCATCAACC



GGGTAATGGGATATCTCTCCAGTGCGtaG







IL7 (Mouse)



(SEQ ID NO: 64)



ATGTTTCATGTGTCCTTCAGGTACATATTTGGTATCCCAC



CACTTATATTGGTGCTCTTGCCTGTAACCAGCTCTGAATG



TCATATAAAAGACAAGGAGGGCAAAGCATACGAGTCCGTA



TTGATGATCTCAATCGATGAACTTGACAAGATGACAGGGA



CCGATTCTAATTGTCCAAATAACGAGCCAAACTTCTTTCG



GAAACACGTGTGTGATGATACAAAAGAAGCTGCTTTTCTT



AACAGAGCTGCCAGAAAACTCAAGCAGTTCCTCAAGATGA



ATATATCCGAGGAATTTAACGTGCATCTCCTCACAGTATC



TCAGGGAACTCAAACCCTTGTAAACTGCACTTCTAAGGAG



GAGAAGAATGTCAAAGAGCAGAAGAAAAATGATGCATGTT



TTTTGAAACGGCTGTTGAGGGAGATCAAAACATGCTGGAA



TAAAATCCTCAAGGGCTCAATTtaG







IL15 (Human)



(SEQ ID NO: 65)



ATGGAAACAGACACATTGCTGCTTTGGGTATTGTTGCTCT



GGGTGCCTGGATCAACAGGAAACTGGGTAAACGTAATTTC



AGATCTGAAGAAGATCGAGGACCTTATTCAATCCATGCAC



ATCGATGCCACTCTCTACACCGAAAGCGACGTTCACCCAT



CTTGCAAGGTGACCGCTATGAAATGTGAATTGTTGGAACT



TCAGGTAATTTCTCTGGAGAGCGGCGATGCCTCAATACAT



GACACCGTTGAAAATCTTATCATCCTTGCTAATGATTCAC



TCTCTAGTAATGGGAACGTAACAGAGAGCGGGTGTAAGGA



GTGTGAAGAACTGGAGGAGAAAAACATTAAGGAATTTTTG



CAGTCATTCGTCCATATAGTGCAAATGTTCATAAACACTT



CCAGAAGAAAGCGAGGCTCTGGGGAGGGGCGAGGCTCTCT



GCTGACCTGTGGGGATGTAGAAGAGAATCCAGGTCCCATG



GACCGGCTGACCAGCTCATTCCTGCTTCTGATTGTGCCAG



CCTACGTGCTCTCCATCACATGTCCTCCCCCAATGAGCGT



CGAGCATGCTGACATCTGGGTGAAGTCATACTCCTTGTAC



AGCAGAGAGAGATACATTTGTAATTCCGGATTCAAGCGCA



AGGCCGGCACCTCCTCTCTGACAGAGTGCGTCCTTAACAA



AGCAACCAACGTAGCACATTGGACCACACCATCCTTGAAG



TGCATACGAGAACCTAAATCTTGCGATAAGACTCATACTT



GTCCACCTTGTCCAGCCCCAGAACTGCTTGGCGGACCCTC



AGTATTTTTGTTCCCACCAAAGCCAAAAGACACACTCATG



ATATCCAGAACTCCTGAGGTGACCTGTGTCGTTGTAGACG



TTTCCCACGAAGATCCTGAAGTAAAATTCAACTGGTACGT



GGATGGGGTCGAAGTCCATAACGCCAAGACTAAACCAAGG



GAGGAACAGTATAACTCTACTTACCGAGTAGTTTCTGTGT



TGACCGTGCTGCACCAGGACTGGTTGAACGGGAAGGAGTA



CAAATGCAAGGTGAGCAATAAAGCTCTGCCCGCACCAATC



GAAAAGACAATATCTAAGGCCAAGGGGCAGCCACGAGAGC



CCCAGGTATACACACTGCCACCCTCACGCGATGAATTGAC



TAAGAACCAGGTTTCCCTGACCTGTCTTGTAAAAGGTTTC



TACCCTTCCGACATAGCTGTTGAGTGGGAAAGTAACGGGC



AGCCAGAGAACAATTACAAGACAACTCCACCCGTTCTTGA



TAGCGATGGATCATTTTTTCTGTATTCCAAACTCACTGTC



GATAAAAGTCGCTGGCAGCAAGGCAATGTTTTTAGCTGCT



CAGTCATGCACGAAGCACTGCATAATCACTACACACAAAA



AAGTTTGTCCCTTAGCCCTGGTAAGtaG







IL15 (Human)



(SEQ ID NO: 66)



ATGTACTCAATGCAGTTGGCCTCCTGTGTAACATTGACCT



TGGTCCTCTTGGTCAACAGCAATTGGATCGATGTACGCTA



CGACTTGGAGAAGATTGAGTCCCTTATACAGAGTATACAC



ATAGATACAACCTTGTATACTGACAGTGACTTCCATCCCA



GCTGTAAAGTGACTGCAATGAACTGTTTTTTGTTGGAGTT



GCAAGTAATTCTGCATGAATACAGCAACATGACCCTCAAT



GAAACCGTTAGGAATGTCCTTTATCTCGCAAATTCTACTC



TGAGTAGCAATAAGAATGTTGCCGAAAGCGGCTGCAAGGA



GTGCGAAGAACTGGAGGAAAAAACTTTCACCGAGTTTCTC



CAGAGTTTCATCAGAATTGTCCAAATGTTCATTAATACAA



GTAGTGGTGGTGGGAGCGGGGGTGGAGGCAGTGGGGGAGG



TGGGAGCGGAGGTGGAGGGTCCGGAGGGGGGAGCCTTCAA



GGCACTACTTGTCCTCCACCCGTATCCATCGAGCACGCCG



ATATTCGAGTTAAAAATTATAGTGTTAATAGCAGAGAACG



ATACGTCTGCAACTCAGGGTTTAAGAGAAAGGCCGGAACT



TCAACTCTCATAGAATGCGTGATTAATAAGAATACTAACG



TCGCACATTGGACTACTCCCAGTCTCAAGTGCATACGCGA



TCCATCTCTCGCTCATTACTCACCAGTACCTACAGTGGTT



ACTCCTAAGGTGACCTCTCAGCCCGAATCACCATCTCCCA



GCGCAAAAGAGCCTGAGGCCTTTTCTCCTAAATCAGACAC



TGCTATGACTACAGAAACAGCCATAATGCCAGGAAGCCGG



CTGACACCATCTCAAACTACCAGCGCAGGCACAACTGGGA



CTGGCTCCCACAAAAGCTCACGCGCACCAAGTCTCGCCGC



AACAATGACATTGGAGCCTACAGCCAGCACATCTCTTAGA



ATCACAGAAATTTCTCCCCACAGTAGCAAGATGACCAAGG



TGGCAATTAGTACCAGCGTCCTTCTTGTAGGAGCTGGAGT



TGTGATGGCATTTTTGGCATGGTATATCAAAAGCAGGtaG







IL15 (Mouse)



(SEQ ID NO: 67)



ATGAAGATCCTCAAGCCATACATGCGAAACACTAGTATTA



GCTGTTACTTGTGTTTTCTGCTGAATAGTCATTTTTTGAC



TGAAGCAGGAATCCATGTATTTATACTCGGTTGTGTGTCT



GTAGGTCTGCCAAAGACTGAGGCTAATTGGATTGACGTGC



GCTATGATCTTGAAAAAATAGAGTCCTTGATTCAATCAAT



ACACATCGATACCACTCTCTACACCGACAGTGATTTCCAT



CCTTCCTGCAAGGTAACAGCTATGAATTGCTTCCTCCTGG



AGCTCCAAGTCATTCTCCATGAGTACTCCAACATGACTTT



GAACGAAACTGTAAGAAACGTATTGTATCTGGCTAATAGC



ACCTTGTCTAGTAACAAAAATGTGGCAGAGAGCGGCTGCA



AAGAATGTGAAGAATTGGAAGAGAAAACATTTACAGAGTT



CCTGCAATCCTTTATTCGCATCGTCCAAATGTTTATCAAT



ACCTCTtaG







IL15 (Mouse)



(SEQ ID NO: 68)



ATGTATTCCATGCAACTTGCCAGTTGTGTAACCCTTACTC



TCGTCCTGCTCGTTAATTCCGCTGGTGCTAACTGGATAGA



TGTTCGATACGATCTGGAAAAGATTGAGTCCCTTATCCAA



TCCATTCATATAGATACCACCCTTTATACTGACAGCGACT



TCCATCCTTCTTGCAAGGTGACCGCTATGAATTGTTTCCT



GCTGGAACTCCAAGTTATTCTGCATGAATACTCTAATATG



ACACTTAACGAGACCGTAAGAAATGTTCTCTATCTCGCTA



ATAGTACTTTGAGCTCAAATAAGAACGTGGCCGAGTCTGG



GTGTAAGGAATGCGAAGAGCTGGAAGAAAAGACATTCACC



GAGTTTCTCCAGTCTTTCATACGGATTGTGCAGATGTTTA



TCAACACATCAGATTACAAAGACGACGATGATAAGtaG







IL18 (Mouse)



(SEQ ID NO: 69)



ATGGCAGCCATGTCTGAGGACTCTTGTGTGAACTTTAAAG



AAATGATGTTCATAGACAATACACTCTACTTTATACCTGA



GGAGAATGGAGATTTGGAATCTGACAACTTTGGCAGGCTG



CATTGCACTACCGCAGTTATCCGAAACATCAACGATCAGG



TACTGTTTGTTGATAAAAGACAACCTGTATTCGAGGACAT



GACCGACATAGATCAGTCTGCCTCAGAGCCCCAGACTAGG



CTTATCATCTATATGTACAAGGACAGCGAAGTACGAGGCC



TGGCTGTTACACTCTCAGTCAAAGACTCTAAGATGAGCAC



CCTGTCATGCAAGAACAAAATTATCAGTTTTGAGGAGATG



GACCCACCTGAAAACATAGATGACATTCAGTCAGACCTCA



TTTTTTTTCAAAAGCGGGTACCAGGACACAACAAAATGGA



ATTTGAATCATCACTCTACGAAGGACATTTCCTTGCATGC



CAGAAAGAGGATGACGCATTCAAATTGATCCTGAAAAAAA



AGGACGAAAATGGTGATAAATCAGTCATGTTTACATTGAC



CAATCTTCACCAAAGTtaG







IL18 (Mouse)



(SEQ ID NO: 70)



ATGGCTGCAATGTCTGAAGATAGCTGTGTCAACTTTAAGG



AGATGATGTTCATTGATAATACTTTGTACTTTATACCTGA



AGAAAATGGAGACCTTGAGTCAGACAACTTCGGGAGACTG



CACTGCACAACTGCCGTTATCCGAAACATAAATGATCAAG



TATTGTTCGTGGACAAAAGACAACCAGTCTTTGAGGATAT



GACAGACATCGACCAATCCGCATCTGAACCTCAGACTAGG



CTGATCATCTATATGTACGCCGACTCCGAAGTAAGAGGCC



TTGCTGTGACACTTAGTGTTAAGGATAGTAAGATGAGCAC



ACTGTCCTGTAAGAATAAGATTATATCTTTTGAAGAGATG



GACCCTCCCGAGAACATAGATGACATCCAGAGCGACTTGA



TCTTCTTTCAGAAGCGAGTGCCAGGCCATAACAAGATGGA



ATTTGAATCATCTCTTTATGAAGGCCATTTCCTCGCATGT



CAAAAGGAGGACGATGCCTTCAAGCTCATTCTGAAAAAAA



AAGACGAGAACGGTGATAAGAGCGTGATGTTCACTCTGAC



AAATCTGCACCAGTCAtaG







IL18 (Human)



(SEQ ID NO: 71)



ATGTATCGCATGCAACTCCTGTCCTGCATTGCTCTGAGCT



TGGCTTTGGTAACCAACTCATACTTCGGGAAACTGGAGAG



TAAACTCTCCGTAATCAGGAATCTTAATGACCAAGTATTG



TTTATTGACCAGGGCAACCGCCCGTTGTTCGAGGATATGA



CTGATTCTGACTGTCGGGATAACGCTCCGAGAACTATCTT



TATCATTTCAATGTACAAGGACAGCCAACCGCGGGGTATG



GCTGTGACAATCAGTGTCAAATGTGAGAAGATTTCCACGC



TGTCCTGCGAAAACAAGATAATTTCTTTCAAAGAAATGAA



CCCCCCTGACAATATAAAGGATACAAAGAGTGATATCATC



TTCTTTCAGAGGTCCGTGCCCGGCCACGATAATAAGATGC



AATTTGAAAGTTCATCTTATGAGGGGTACTTTTTGGCATG



CGAGAAAGAAAGGGATCTCTTCAAGTTGATCCTGAAGAAG



GAGGACGAATTGGGCGACCGCTCCATCATGTTCACAGTCC



AGAACGAGGACtaG







IL18 (Human)



(SEQ ID NO: 72)



ATGTACCGCATGCAGCTCCTGAGTTGTATTGCCCTTTCCC



TCGCTCTCGTTACCAATTCTTACTTCGGTAAGCTTGCCTC



TAAACTCTCTGTTATTAGGAACTTGAACGACCAAGTCCTT



TTCATAGACCAAGGGAACAGACCACTGTTTGAAGATATGA



CGGATAGCGATTGCCGAGATAATGCCCCTAGGACGATTTT



TATCATTAGTATGTATGCGGACTCTCAACCGAGGGGGATG



GCCGTTACTATAAGTGTGAAATGCGAGAAAATATCAACGC



TCAGTTGTGAGAACAAAATCATAAGTTTCAAGGAGATGAA



TCCACCTGATAACATCAAAGACACTAAGTCTGATATTATA



TTTTTCCAACGAAGTGTTCCGGGACACGATAACAAAATGC



AATTTGAGAGCTCCTCATACGAGGGCTACTTCCTCGCGTG



TGAGAAAGAAAGGGATTTGTTTAAGCTTATCCTCAAGAAA



GAGGACGAGTTGGGGGATCGGAGCATAATGTTTACCGTAC



AGAATGAGGACtaG







IL21 (Mouse)



(SEQ ID NO: 73)



ATGGAGCGGACACTCGTGTGTCTTGTCGTAATTTTTCTCG



GGACAGTCGCACACAAGTCCTCACCCCAGGGTCCTGATCG



CCTTCTCATACGCCTCCGACATTTGATCGACATTGTAGAG



CAGCTCAAAATTTACGAGAATGACCTCGATCCCGAGCTTT



TGAGTGCTCCCCAAGACGTTAAGGGTCATTGCGAGCACGC



AGCTTTTGCTTGCTTCCAGAAGGCCAAGTTGAAACCAAGC



AACCCTGGTAATAATAAGACTTTCATCATCGACTTGGTCG



CCCAACTCCGAAGGAGGCTGCCTGCCCGGCGCGGAGGAAA



AAAACAAAAGCATATTGCAAAGTGTCCTTCATGTGATTCA



TACGAAAAGCGGACTCCCAAAGAGTTCTTGGAAAGGTTGA



AATGGCTTCTTCAGAAGATGATTCATCAACATTTGTCAta



G







IFN-beta (Human)



(SEQ ID NO: 74)



ATGACCAACAAATGCCTTTTGCAAATTGCCCTGCTTTTGT



GTTTTAGCACTACCGCATTGAGCATGTCATATAACCTCCT



CGGCTTCCTTCAGAGATCATCAAACTTTCAGTGTCAGAAA



CTGCTTTGGCAACTTAATGGCAGGCTCGAATATTGTCTGA



AAGATCGGATGAATTTCGACATTCCAGAAGAAATAAAACA



GCTTCAACAATTCCAGAAAGAGGACGCCGCCCTGACTATT



TACGAGATGCTCCAGAATATCTTCGCCATTTTCCGGCAGG



ACAGCTCATCCACGGGGTGGAATGAGACTATTGTAGAAAA



TCTTCTGGCTAATGTGTACCATCAAATTAATCACCTCAAA



ACGGTGCTTGAGGAAAAACTTGAAAAGGAAGATTTCACAC



GGGGCAAGTTGATGTCCTCCCTGCACCTTAAACGATACTA



CGGCAGGATTCTTCATTACTTGAAGGCTAAGGAGTATAGC



CATTGCGCGTGGACAATTGTACGGGTAGAAATACTGCGAA



ACTTTTATTTCATCAACCGGCTCACTGGATACCTTAGAAA



TtaG







IFN-beta (Mouse)



(SEQ ID NO: 75)



ATGAACAATCGGTGGATACTCCACGCCGCATTTCTCCTCT



GCTTTAGCACGACGGCCCTGTCCATC



AACTACAAACAGCTTCAGTTGCAGGAGCGGACTAACATAA



GGAAGTGCCAGGAACTGCTGGAACAGCTTAATGGTAAAAT



TAATCTTACATACCGAGCTGACTTCAAAATTCCTATGGAA



ATGACCGAGAAGATGCAGAAATCCTACACGGCATTCGCCA



TCCAGGAAATGCTCCAGAACGTATTTCTCGTGTTCCGCAA



TAATTTCTCTTCTACGGGTTGGAACGAAACCATTGTTGTT



AGACTGCTTGACGAACTGCATCAGCAAACCGTGTTCCTTA



AAACCGTGCTTGAGGAGAAGCAGGAGGAGCGCCTGACTTG



GGAGATGTCTAGTACCGCACTTCACTTGAAATCCTACTAC



TGGCGCGTTCAGCGGTATCTGAAGCTGATGAAGTATAACT



CATACGCCTGGATGGTAGTGCGCGCAGAGATCTTCAGAAA



CTTTCTTATCATCCGGCGACTGACCCGAAACTTTCAGAAT



taG







IFN-gamma (Human)



(SEQ ID NO: 76)



ATGAAGTACACTAGCTATATATTGGCCTTCCAGCTTTGCA



TCGTATTGGGTAGCCTCGGATGCTATTGCCAAGACCCGTA



TGTCAAAGAAGCCGAAAATCTCAAAAAGTATTTCAATGCC



GGACACTCAGACGTCGCGGATAACGGTACACTGTTTCTTG



GCATCCTGAAAAATTGGAAGGAAGAGAGTGACAGAAAAAT



AATGCAGTCACAAATAGTGTCCTTTTACTTTAAGCTGTTC



AAAAATTTCAAGGATGACCAAAGTATCCAGAAGAGTGTTG



AAACTATCAAAGAGGACATGAATGTGAAATTCTTTAACAG



TAATAAGAAGAAGCGCGATGACTTCGAGAAACTCACTAAT



TACAGCGTAACGGATCTTAACGTCCAACGCAAGGCAATCC



ACGAGCTTATACAGGTAATGGCTGAGCTTAGTCCCGCAGC



CAAGACAGGGAAGAGAAAAAGGTCTCAAATGCTTTTTCGG



GGCCGGCGAGCTTCACAAtaG







IFN-gamma (Mouse)



(SEQ ID NO: 77)



ATGAACGCTACGCATTGCATCCTCGCACTCCAATTGTTCC



TCATGGCTGTGTCAGGGTGTTACTGTCACGGTACTGTCAT



AGAAAGCCTCGAATCCCTGAATAACTATTTTAACAGTAGC



GGTATAGATGTAGAAGAAAAGTCTCTCTTTCTTGACATCT



GGAGGAATTGGCAAAAGGATGGAGACATGAAGATTCTCCA



ATCTCAGATTATATCATTTTACTTGAGGCTTTTTGAGGTT



CTGAAGGATAACCAGGCGATCAGCAATAATATCAGCGTAA



TTGAATCTCACCTTATTACAACATTTTTCTCAAATTCCAA



GGCAAAGAAAGATGCTTTCATGTCTATCGCGAAATTTGAG



GTGAACAATCCTCAGGTACAAAGGCAAGCCTTTAACGAGC



TGATTAGAGTTGTACATCAGTTGTTGCCCGAAAGTAGTCT



TAGAAAACGCAAACGGAGCCGATGCtaG







IFN-alpha (Mouse)



(SEQ ID NO: 78)



ATGGCAAGGTTGTGCGCTTTTCTCATGGTACTGGCTGTGC



TCTCCTATTGGCCTACTTGTTCTCTGGGATGCGACTTGCC



ACAGACCCACAATCTGCGGAATAAGAGGGCTCTGACTCTG



CTGGTGCAAATGAGACGGCTCTCTCCACTTAGCTGTTTGA



AAGATAGAAAGGATTTCGGGTTCCCCCAGGAGAAGGTGGA



TGCCCAGCAGATCAAGAAGGCACAGGCTATCCCCGTCCTT



TCCGAGCTGACCCAGCAAATTTTGAACATCTTTACAAGTA



AGGATAGTTCAGCTGCATGGAATACCACACTTTTGGATTC



TTTTTGTAACGATCTGCATCAGCAGCTGAACGATCTCCAG



GGATGCCTGATGCAGCAAGTCGGCGTGCAAGAATTTCCAC



TCACCCAGGAGGACGCTCTGCTCGCAGTGCGAAAGTATTT



TCACCGAATTACCGTGTACCTCCGGGAGAAAAAGCATTCA



CCCTGCGCTTGGGAAGTAGTCAGGGCCGAAGTATGGAGAG



CCCTTAGTAGCTCCGCTAATGTACTGGGCCGGTTGCGGGA



AGAGAAAtaG







CCL21 (Human)



(SEQ ID NO: 79)



ATGGCGCAAAGTCTGGCTCTTTCACTCCTGATCCTGGTCT



TGGCCTTCGGGATTCCGAGGACCCAAGGAAGTGATGGTGG



CGCCCAAGATTGTTGCCTTAAATACAGCCAGCGGAAAATA



CCCGCGAAAGTGGTCAGGAGTTATAGAAAACAGGAGCCTT



CCCTGGGTTGTAGTATCCCCGCCATACTTTTCCTCCCGAG



AAAACGGAGCCAGGCCGAACTGTGCGCTGACCCTAAGGAA



CTTTGGGTGCAACAACTTATGCAACACCTGGATAAGACAC



CTTCTCCTCAAAAGCCAGCTCAGGGCTGCCGAAAAGATAG



AGGCGCCTCAAAAACCGGAAAAAAGGGCAAAGGTTCTAAA



GGATGTAAGCGGACTGAACGCTCTCAAACGCCTAAAGGGC



CGtaG







CCL21a (Mouse)



(SEQ ID NO: 80)



ATGGCGCAAATGATGACCCTTTCCCTGCTGAGTCTTGTCC



TCGCGCTCTGCATCCCGTGGACGCAGGGGTCTGATGGGGG



GGGCCAAGACTGTTGCCTGAAGTATTCACAAAAAAAGATA



CCGTACTCTATTGTCAGAGGGTACAGGAAGCAAGAACCCT



CCTTGGGTTGCCCTATACCAGCAATTCTTTTCTCCCCACG



CAAGCATTCCAAACCAGAACTGTGTGCGAACCCCGAGGAG



GGTTGGGTACAGAACTTGATGCGAAGGCTTGACCAGCCCC



CAGCCCCTGGCAAGCAGTCACCTGGGTGCAGAAAAAACAG



AGGTACTTCAAAGAGCGGCAAGAAAGGCAAAGGGAGTAAA



GGATGTAAAAGAACGGAGCAGACCCAGCCTTCACGAGGCt



aG







Tail-less CCL21 (Human)



(SEQ ID NO: 81)



ATGGCGCAAAGTCTGGCTCTTTCACTCCTGATCCTGGTCT



TGGCCTTCGGGATTCCGAGGACCCAAGGAAGTGATGGTGG



CGCCCAAGATTGTTGCCTTAAATACAGCCAGCGGAAAATA



CCCGCGAAAGTGGTCAGGAGTTATAGAAAACAGGAGCCTT



CCCTGGGTTGTAGTATCCCCGCCATACTTTTCCTCCCGAG



AAAACGGAGCCAGGCCGAACTGTGCGCTGACCCTAAGGAA



CTTTGGGTGCAACAACTTATGCAACACCTGGATAAGACAC



CTTCTCCTCAAAAGCCAGCTCAGGGCtaG







Tail-less CCL21 (Mouse)



(SEQ ID NO: 82)



ATGGCGCAAATGATGACCCTTTCCCTGCTGAGTCTTGTCC



TCGCGCTCTGCATCCCGTGGACGCAGGGGTCTGATGGGGG



GGGCCAAGACTGTTGCCTGAAGTATTCACAAAAAAAGATA



CCGTACTCTATTGTCAGAGGGTACAGGAAGCAAGAACCCT



CCTTGGGTTGCCCTATACCAGCAATTCTTTTCTCCCCACG



CAAGCATTCCAAACCAGAACTGTGTGCGAACCCCGAGGAG



GGTTGGGTACAGAACTTGATGCGAAGGCTTGACCAGCCCC



CAGCCCCTGGCAAGCAGTCACCTGGGtaG







CCL19 (Mouse)



(SEQ ID NO: 83)



ATGGCACCCCGCGTCACACCCTTGCTTGCTTTTTCTCTGC



TTGTCCTCTGGACCTTCCCCGCTCCTACCCTTGGAGGAGC



CAATGATGCCGAGGATTGCTGCCTGAGTGTTACACAAAGG



CCAATACCAGGGAATATAGTGAAGGCATTCCGGTATCTGC



TCAATGAAGATGGGTGCAGAGTCCCCGCAGTTGTCTTTAC



AACATTGCGAGGTTACCAGCTTTGTGCTCCCCCAGACCAG



CCTTGGGTAGATCGCATTATTCGCCGGTTGAAGAAGAGCT



CAGCAAAGAATAAGGGCAATTCCACACGGAGAAGCCCCGT



CTCCtaG







CCL19 (Mouse)



(SEQ ID NO: 84)



ATGAAATCAGCAGTCCTTTTCTTGCTCGGGATTATTTTTC



TGGAACAATGTGGAGTGAGGGGAACACTCGTAATAAGAAA



CGCTCGGTGCTCATGCATATCAACATCACGGGGCACTATC



CACTACAAATCCCTGAAGGATCTGAAGCAGTTCGCCCCAA



GCCCTAACTGTAACAAGACCGAAATTATCGCAACTCTCAA



AAATGGAGATCAGACTTGTCTTGACCCAGATTCAGCAAAT



GTCAAGAAGCTGATGAAAGAGTGGGAAAAGAAGATTTCAC



AAAAAAAAAAGCAAAAACGCGGCAAGAAACATCAAAAGAA



CATGAAAAACAGGAAACCTAAGACTCCCCAGTCAAGGAGA



AGATCCCGCAAGACAACCtaG







CXCL11 (Mouse)



(SEQ ID NO: 85)



ATGAACAGAAAAGTTACCGCTATAGCACTTGCTGCCATAA



TATGGGCCACCGCAGCTCAAGGGTTCCTGATGTTCAAGCA



GGGCCGATGCCTCTGCATTGGCCCTGGAATGAAGGCCGTG



AAAATGGCCGAAATAGAAAAAGCTAGTGTCATATACCCCT



CTAACGGTTGCGATAAAGTCGAGGTTATAGTCACAATGAA



AGCTCATAAACGCCAACGCTGCCTCGACCCCCGGTCTAAG



CAGGCTAGGCTCATAATGCAAGCAATCGAGAAGAAAAACT



TTCTTAGACGGCAAAACATGtaG







CXCL10 (Mouse)



(SEQ ID NO: 86)



ATGAACCCATCTGCCGCCGTTATTTTCTGTCTGATACTCC



TTGGGCTGAGTGGCACACAAGGCATACCCCTCGCCCGCAC



AGTCCGGTGTAATTGTATACATATTGACGACGGCCCTGTT



AGAATGCGGGCCATCGGTAAGCTGGAGATTATACCAGCAA



GCCTTAGTTGTCCCAGGGTTGAAATCATAGCAACTATGAA



AAAAAACGACGAACAAAGATGTTTGAATCCCGAGAGCAAG



ACAATCAAAAACCTTATGAAAGCATTTAGTCAAAAACGCT



CTAAACGCGCTCCAtaG







CXCL10 (Human)



(SEQ ID NO: 87)



ATGAATCAGACGGCAATCCTTATATGCTGCCTTATATTCC



TTACTCTCTCAGGGATACAAGGGGTACCACTTTCTCGGAC



TGTTCGCTGCACTTGCATTTCAATATCTAACCAACCTGTA



AATCCGCGGAGCCTGGAAAAATTGGAGATTATACCTGCTT



CTCAATTCTGCCCTCGGGTGGAAATCATCGCCACTATGAA



GAAGAAGGGCGAGAAAAGGTGTCTGAATCCAGAGTCAAAG



GCAATCAAAAACCTGCTGAAAGCGGTGTCAAAGGAACGGT



CCAAGAGATCACCCtaG







CXCL11-CXCL10 (Mouse)



(SEQ ID NO: 88)



ATGAACAGGAAAGTAACAGCCATTGCATTGGCTGCCATCA



TCTGGGCCACCGCAGCACAGGGTTTTCTGATGTTTAAGCA



AGGGCGCTGTCTCTGTATAGGCCCAGGCATGAAGGCCGTG



AAGATGGCAGAGATTGAGAAGGCATCTGTGATTTATCCTT



CTAACGGGTGCGATAAAGTCGAAGTTATTGTGACAATGAA



GGCACACAAACGCCAACGGTGTTTGGACCCACGATCTAAA



CAGGCAAGATTGATTATGCAAGCCATCGAGAAAAAGAACT



TTCTCCGAAGGCAAAATATGATCCCTTTGGCTCGGACAGT



GCGGTGTAACTGTATTCACATCGACGATGGGCCAGTACGG



ATGAGAGCAATAGGAAAGCTCGAAATCATACCCGCCTCAT



TGTCTTGTCCCAGGGTGGAAATAATCGCCACTATGAAAAA



GAACGATGAACAGAGGTGTCTCAACCCAGAGAGTAAGACT



ATCAAGAACCTTATGAAGGCATTCAGTCAGAAGAGGTCAA



AGCGAGCACCAtaG







XCL1 (Human)



(SEQ ID NO: 89)



ATGAGACTTCTCATATTGGCGCTTCTCGGGATATGTTCTC



TTACGGCATACATAGTTGAGGGGGTGGGATCTGAGGTTAG



CGATAAACGAACTTGTGTTAGTCTTACAACACAGAGGCTT



CCAGTCTCCAGGATAAAAACATATACGATAACTGAGGGAT



CTCTCAGAGCGGTCATCTTCATAACGAAGAGGGGCCTGAA



GGTCTGTGCTGACCCACAAGCGACTTGGGTAAGGGACGTT



GTGCGGAGCATGGACAGGAAGAGCAATACTCGCAACAACA



TGATCCAAACCAAACCTACGGGCACCCAACAGTCAACCAA



TACTGCGGTAACATTGACGGGGtaG







XCL1 (Mouse)



(SEQ ID NO: 90)



ATGCGCCTCCTTCTGCTGACTTTTCTGGGTGTATGTTGCC



TGACACCCTGGGTCGTAGAAGGAGTAGGAACCGAGGTTCT



GGAAGAGTCCTCATGTGTAAACTTGCAGACACAACGACTC



CCCGTCCAAAAAATCAAGACCTATATAATCTGGGAGGGGG



CAATGCGGGCCGTCATTTTCGTGACTAAACGAGGTCTCAA



AATCTGCGCCGACCCCGAGGCTAAGTGGGTGAAGGCAGCC



ATTAAGACCGTGGATGGGAGAGCCAGCACCAGAAAGAACA



TGGCCGAAACAGTACCTACTGGCGCACAGCGGTCAACCTC



AACTGCTATAACCTTGACAGGAtaG







m_sCD40L #1



(SEQ ID NO: 91)



ATGGAGACTGACACTCTGCTTCTGTGGGTGTTGCTGCTGT



GGGTGCCTGGCAGTACAGGCGATATGCAACGAGGTGACGA



GGACCCTCAAATCGCCGCCCATGTAGTCTCTGAAGCTAAT



AGCAACGCTGCATCCGTCTTGCAGTGGGCAAAGAAAGGCT



ACTATACTATGAAGTCCAACTTGGTAATGCTTGAAAACGG



CAAGCAGTTGACTGTCAAGAGAGAGGGACTTTATTACGTC



TATACCCAAGTCACATTCTGTAGCAATCGAGAACCCTCCT



CACAGAGGCCTTTTATAGTGGGACTCTGGCTTAAACCAAG



TAGCGGCTCTGAGCGCATACTGTTGAAAGCCGCAAACACA



CACAGCTCTTCCCAACTCTGCGAGCAGCAATCCGTGCATC



TCGGTGGAGTATTTGAGCTTCAAGCCGGTGCCTCAGTGTT



TGTGAACGTCACTGAGGCCTCCCAGGTCATACATCGAGTT



GGGTTCAGCTCCTTCGGCTTGCTCAAGCTCtaG







m_sCD40L #2



(SEQ ID NO: 92)



ATGGAAACTGATACATTGCTGCTCTGGGTTTTGCTGCTCT



GGGTGCCTGGGAGTACAGGCGACATGAGGAGGCAGTTCGA



GGATCTCGTTAAGGATATTACCCTTAATAAGGAGGAGAAG



AAAGAAAACTCTTTTGAGATGCAACGAGGGGACGAAGATC



CTCAGATCGCTGCTCACGTGGTCTCTGAAGCTAACAGCAA



CGCCGCTTCTGTCCTCCAGTGGGCCAAGAAAGGTTATTAC



ACCATGAAATCAAACCTTGTAATGCTTGAAAACGGGAAAC



AGCTTACAGTGAAGAGGGAAGGTCTTTACTACGTCTATAC



CCAGGTAACCTTCTGCTCAAACAGAGAACCATCAAGCCAG



AGGCCATTCATAGTGGGGCTCTGGCTCAAACCTTCCAGTG



GCAGCGAGAGAATCTTGTTGAAAGCTGCTAATACACATAG



TAGTAGCCAGCTTTGCGAGCAACAGTCAGTCCACCTCGGG



GGGGTGTTTGAGTTGCAAGCAGGGGCCTCAGTATTCGTGA



ATGTCACTGAGGCTTCCCAGGTAATTCACAGGGTAGGCTT



TAGTTCATTCGGTTTGCTGAAGCTTtaG







m_sCD40L #3



(SEQ ID NO: 93)



ATGCGAAGAATGCAGCTTCTGCTCCTTATTGCTCTGAGTC



TCGCCCTTGTCACCAACTCCGGGGACAGAATGAAACAAAT



CGAGGACAAAATTGAAGAAATACTGAGTAAAATATATCAC



ATCGAAAACGAAATTGCACGCATTAAGAAATTGATTGGCG



AACGCACCAGTGGCGGCTCTGGTGGCACCGGAGGTTCAGG



CGGGACCGGGGGCTCTGACAAAGTCGAAGAGGAGGTTAAC



CTTCATGAGGACTTTGTGTTCATCAAGAAGCTGAAACGGT



GCAATAAAGGAGAAGGTTCTTTGAGCCTCCTTAATTGCGA



AGAGATGCGACGACAGTTCGAGGATCTGGTTAAGGACATT



ACACTTAATAAGGAAGAGAAAAAGGAGAACTCTTTCGAAA



TGCAGCGCGGCGATGAAGATCCCCAGATAGCCGCCCATGT



CGTCTCTGAGGCCAACTCTAACGCAGCATCCGTCCTCCAG



TGGGCTAAGAAAGGATATTATACTATGAAAAGCAATTTGG



TCATGCTCGAAAACGGTAAACAGCTCACTGTTAAGAGAGA



AGGCCTCTATTACGTATATACTCAAGTAACTTTCTGTTCT



AATAGGGAACCCTCCTCTCAAAGACCTTTTATCGTAGGAC



TCTGGTTGAAACCAAGTAGCGGTAGTGAAAGGATTCTGCT



CAAAGCAGCTAATACTCACTCCAGCAGTCAACTGTGCGAA



CAACAAAGCGTTCACCTCGGGGGCGTCTTTGAACTTCAGG



CAGGTGCCAGTGTTTTCGTCAACGTAACAGAAGCATCCCA



GGTAATTCATCGAGTAGGGTTTTCTAGCTTTGGTTTGCTG



AAGCTGtaG







anti-CD40_FGK4.5



(SEQ ID NO: 94)



ATGGAAACTGATCGCCTGTTGCTCTGGGTACTTCTTCTGT



GGGTGCCTGGGTCCACTGGTGACACTGTACTTACACAATC



ACCCGCTTTGGCCGTTTCTCCTGGTGAACGGGTCACAATT



AGTTGCCGAGCTTCCGATTCTGTATCTACTCTTATGCATT



GGTATCAACAAAAACCTGGTCAGCAGCCAAAATTGCTCAT



TTATCTTGCTAGTCACTTGGAGTCCGGCGTACCTGCTCGA



TTCAGCGGTAGTGGGTCTGGCACAGATTTCACTTTGACCA



TAGATCCCGTGGAGGCCGATGACACTGCAACCTACTATTG



CCAGCAATCCTGGAACGACCCTTGGACTTTCGGCGGCGGC



ACCAAGCTGGAACTCAAGCGAGCAGATGCTGCCCCAACCG



TTAGTATATTCCCACCCTCAACCGAACAACTCGCCACAGG



AGGCGCTAGTGTCGTGTGTCTTATGAACAATTTCTATCCA



CGAGACATTAGCGTCAAGTGGAAAATTGATGGGACAGAAA



GGCGAGATGGAGTTTTGGATTCAGTAACAGACCAGGATTC



AAAGGATTCTACCTATAGCATGAGCTCCACCTTGAGCCTG



ACCAAAGCTGATTATGAATCTCATAACCTGTATACTTGTG



AAGTGGTGCATAAGACTTCTAGCTCACCAGTGGTTAAATC



TTTTAACCGCAACGAATGTCGGCGCAAGAGGGGTTCCGGA



GAGGGAAGGGGTAGTCTGCTCACCTGCGGCGATGTTGAAG



AAAATCCTGGTCCCATGGACATTCGGCTCTCTTTGGTATT



CCTGGTACTTTTTATAAAGGGGGTGCAATGTGAAGTCCAG



CTCGTGGAAAGCGGTGGGGGCCTGGTTCAGCCCGGTCGCA



GCCTTAAACTTAGTTGCGCAGCATCCGGATTTACATTTTC



TGACTATAACATGGCCTGGGTTCGACAGGCACCCAAAAAA



GGGCTGGAGTGGGTCGCAACTATCATATACGATGGTTCCC



GGACATACTATAGAGATTCAGTGAAGGGGCGCTTTACAAT



AAGCAGGGACAATGCTAAGTCTACCTTGTATCTTCAGATG



GACTCCCTGAGGAGCGAAGATACAGCAACATATTATTGTG



CTACAAACCGCTGGTTGCTGCTTCATTATTTCGACTACTG



GGGTCAGGGCGTCATGGTAACTGTATCAAGCGCCGAGACC



ACAGCCCCTTCTGTATATCCATTGGCACCAGGTACTGCTC



TGAAATCCAACTCAATGGTAACCCTTGGATGTCTGGTTAA



GGGTTATTTTCCCGAGCCCGTCACAGTTACTTGGAACTCT



GGGGCCCTTTCTAGCGGAGTCCATACCTTTCCCGCCGTTT



TGCAGAGTGGTCTGTACACCCTTACCTCAAGCGTCACAGT



TCCATCTAGCACATGGAGCTCCCAGGCAGTAACTTGTAAT



GTGGCCCATCCAGCCTCCTCAACTAAGGTAGATAAAAAGA



TCGTTCCCAGAGAATGCAATCCATGTGGATGCACCGGGTC



TGAGGTCAGCAGTGTGTTCATTTTCCCACCCAAGACTAAA



GATGTATTGACTATTACTCTTACACCCAAAGTAACCTGCG



TGGTGGTTGATATTAGTCAAAATGATCCCGAGGTACGGTT



CTCTTGGTTTATCGACGACGTCGAAGTACATACAGCTCAG



ACACACGCTCCCGAGAAACAAAGCAATTCCACTCTTAGGA



GCGTGTCCGAGTTGCCAATCGTACATAGGGATTGGCTTAA



TGGCAAGACCTTTAAGTGTAAGGTCAATTCAGGGGCATTC



CCCGCACCAATAGAGAAGAGTATAAGCAAACCCGAGGGGA



CACCCAGAGGTCCACAGGTCTATACAATGGCTCCCCCCAA



GGAAGAGATGACCCAAAGTCAAGTCTCAATTACATGTATG



GTGAAGGGCTTTTATCCACCCGACATATACACTGAGTGGA



AGATGAATGGACAGCCCCAAGAGAATTATAAAAACACTCC



CCCTACCATGGACACCGACGGGTCCTATTTTCTTTATAGT



AAATTGAACGTGAAAAAGGAGACCTGGCAACAAGGCAACA



CTTTCACCTGCTCCGTTCTTCACGAGGGCCTGCATAATCA



TCATACCGAAAAGTCTCTCAGTCATTCTCCAGGTAAGtaG







CD40L_2 (Human)



(SEQ ID NO: 95)



ATGGAAACAGATACGTTGCTGTTGTGGGTACTTCTCCTTT



GGGTCCCTGGCAGCACAGGGGACGAGAATAGTTTCGAAAT



GCAGAAGGGCGACCAGAACCCACAGATCGCGGCTCACGTT



ATATCAGAAGCAAGTAGTAAGACCACTTCCGTACTTCAGT



GGGCTGAAAAAGGATATTACACCATGTCCAACAATCTCGT



GACACTGGAGAACGGTAAACAACTTACGGTGAAACGACAG



GGCCTCTATTACATCTACGCTCAGGTGACATTCTGCTCAA



ATAGGGAGGCTTCTAGTCAAGCGCCCTTCATCGCCAGCCT



GTGCCTCAAATCTCCCGGCCGGTTCGAACGAATCCTGTTG



CGAGCGGCCAATACCCATAGCTCAGCTAAACCTTGCGGCC



AGCAGAGTATTCATCTTGGTGGTGTGTTTGAACTTCAGCC



GGGAGCATCTGTGTTCGTCAACGTAACGGACCCTAGCCAA



GTGTCTCATGGGACAGGTTTTACATCCTTCGGACTCCTCA



AGTTGtaG







Flt3L (Human)



(SEQ ID NO: 96)



ATGACAGTTCTCGCGCCAGCTTGGAGTCCCACCACATACT



TGCTTTTGCTTCTGCTTCTGTCCTCTGGCCTGAGTGGGAC



CCAAGATTGTTCCTTTCAACATTCCCCAATTAGTTCTGAT



TTTGCAGTGAAGATTAGAGAGCTCTCAGACTATCTGCTGC



AAGATTATCCTGTCACAGTCGCTTCAAACCTGCAAGACGA



AGAGCTCTGCGGTGCCTTGTGGCGGTTGGTCTTGGCTCAA



AGATGGATGGAGAGACTGAAAACCGTAGCAGGCAGCAAGA



TGCAGGGTCTCCTGGAAAGGGTGAACACGGAAATCCATTT



TGTGACCAAGTGCGCGTTCCAGCCCCCACCGAGTTGTCTC



CGGTTTGTTCAAACGAATATATCCCGGTTGCTCCAGGAAA



CCTCAGAACAACTGGTGGCTTTGAAACCCTGGATCACAAG



ACAAAACTTTAGTCGGTGCCTCGAACTCCAGTGCCAACCA



GATTCTTCTACACTTCCCCCCCCGTGGTCCCCGCGCCCGT



TGGAAGCAACGGCCCCAtaG







TGFb TRAP (Human)



(SEQ ID NO: 97)



ATGGCCTGGAGTCCTCTGTTTCTGACTCTTATAACTCACT



GTGCCGGCAGTTGGGCTATACCCCCTCATGTACAGAAGTC



TGTAAACAACGACATGATTGTAACCGACAATAATGGCGCA



GTGAAATTCCCACAACTGTGTAAGTTCTGTGATGTACGGT



TTAGTACATGCGACAATCAAAAAAGCTGTATGTCTAACTG



CTCTATTACATCCATATGTGAAAAACCTCAGGAGGTGTGT



GTTGCCGTTTGGCGAAAAAATGATGAGAATATCACACTGG



AGACAGTATGTCATGACCCTAAACTGCCATACCATGATTT



CATACTGGAGGACGCCGCCAGTCCTAAGTGCATTATGAAA



GAGAAAAAGAAACCCGGTGAAACATTCTTTATGTGCTCTT



GTAGCTCTGACGAGTGTAACGACAACATTATATTCAGCGA



GGAGTACAATACAAGCAACCCCGATATACCACCTCACGTA



CAAAAAAGTGTCAACAACGATATGATTGTTACCGACAATA



ACGGAGCTGTTAAGTTTCCTCAGTTGTGCAAGTTCTGCGA



TGTACGATTCTCTACCTGCGACAACCAAAAGTCATGTATG



TCTAACTGTTCCATAACCTCCATCTGCGAGAAGCCCCAGG



AAGTCTGCGTCGCCGTGTGGCGGAAAAACGACGAGAATAT



CACTCTTGAAACCGTTTGTCATGATCCTAAACTGCCCTAT



CACGACTTTATTCTGGAAGATGCTGCTTCCCCTAAGTGTA



TCATGAAAGAAAAGAAGAAACCTGGGGAGACATTCTTTAT



GTGTTCATGCTCCTCCGATGAGTGTAACGACAATATCATC



TTCTCTGAGGAATACAACACTTCTAACCCTGATtaG







Fresolimumab (Human)



(SEQ ID NO: 98)



ATGGCCTGGTCCCCTCTTTTTCTGACCCTCATCACACACT



GTGCAGGCTCATGGGCTGAGACCGTCTTGACCCAGTCCCC



AGGAACTTTGTCTCTGTCTCCTGGTGAAAGAGCTACCCTT



AGTTGTCGAGCCTCTCAGTCCCTTGGTTCTAGCTATCTCG



CTTGGTACCAGCAAAAGCCAGGCCAGGCCCCACGACTGCT



GATCTACGGAGCATCTTCACGGGCTCCCGGCATTCCCGAT



CGATTTTCCGGATCTGGTAGTGGTACAGATTTCACACTGA



CCATATCTCGCCTGGAGCCCGAGGACTTTGCTGTTTATTA



TTGTCAGCAGTACGCCGATTCTCCTATCACTTTTGGACAG



GGAACCCGCCTGGAGATTAAGCGCACAGTAGCAGCTCCAT



CCGTCTTTATCTTTCCACCATCAGATGAACAGCTCAAGAG



TGGGACCGCAAGTGTAGTATGCCTGCTGAACAATTTTTAC



CCTAGAGAGGCCAAAGTGCAGTGGAAGGTGGATAACGCCC



TCCAGAGTGGCAATAGTCAAGAAAGTGTTACTGAGCAAGA



TAGTAAGGACTCTACATACTCTTTGAGTTCTACTTTGACC



CTGTCAAAAGCAGATTATGAAAAACATAAGGTGTATGCAT



GTGAAGTTACACACCAAGGGTTGTCCTCTCCAGTTACAAA



ATCTTTTAATAGAGGAGAGTGCCGCCGCAAACGCGGTAGT



GGAGAAGGTCGAGGCTCACTCTTGACCTGTGGCGACGTGG



AAGAAAATCCCGGTCCTATGGATTGGACTTGGAGGGTATT



TTGTCTTTTGGCAGTAACACCTGGAGCTCACCCCCAAGTA



CAGCTCGTCCAATCTGGTGCCGAGGTTAAAAAGCCTGGAA



GTTCAGTGAAGGTCTCTTGCAAGGCATCTGGATACACCTT



TTCATCTAACGTCATATCCTGGGTACGGCAAGCCCCAGGA



CAGGGACTTGAGTGGATGGGAGGGGTCATCCCCATCGTGG



ACATTGCTAATTACGCTCAGCGATTCAAAGGGCGGGTTAC



TATAACTGCCGACGAGTCTACCTCAACTACCTACATGGAG



TTGTCCTCTCTCCGCTCCGAGGACACTGCTGTATATTACT



GTGCCAGCACTCTCGGGTTGGTGTTGGATGCCATGGACTA



TTGGGGACAAGGAACCCTGGTGACAGTTAGCTCCGCAAGC



ACTAAAGGCCCTTCTGTTTTTCCCTTGGCACCTTGTAGTA



GGTCTACCTCTGAGTCTACAGCAGCACTTGGATGCTTGGT



TAAGGACTATTTTCCCGAGCCAGTTACAGTCTCTTGGAAC



AGTGGTGCCCTCACAAGTGGGGTTCATACCTTCCCCGCAG



TCCTCCAGAGTAGTGGCCTTTACAGCCTCTCATCAGTTGT



GACTGTTCCTAGTTCATCACTCGGTACTAAGACATATACA



TGTAACGTAGACCACAAGCCAAGCAACACAAAAGTAGACA



AACGAGTCGAATCTAAGTATGGACCCCCTTGTCCCTCCTG



TCCTGCTCCCGAGTTCCTTGGGGGCCCTTCCGTGTTCTTG



TTTCCTCCCAAGCCCAAGGATACCCTCATGATCTCACGAA



CCCCAGAGGTAACATGTGTGGTTGTTGACGTAAGTCAGGA



AGATCCCGAAGTGCAATTTAATTGGTACGTGGATGGCGTC



GAAGTCCATAACGCTAAAACAAAACCCCGAGAGGAACAAT



TCAATTCCACATATCGGGTGGTGAGTGTATTGACCGTTCT



TCACCAAGATTGGCTGAACGGCAAGGAGTATAAGTGTAAA



GTAAGCAACAAAGGTCTGCCAAGTAGCATAGAAAAAACAA



TATCTAAAGCTAAGGGCCAACCAAGGGAACCACAAGTATA



TACATTGCCCCCCTCTCAGGAAGAGATGACAAAGAATCAA



GTTAGCCTGACCTGTTTGGTAAAGGGGTTCTATCCCTCAG



ATATAGCAGTCGAGTGGGAATCTAACGGCCAGCCCGAGAA



TAATTATAAAACAACCCCCCCTGTGTTGGACTCAGACGGC



AGCTTCTTTCTCTATTCACGGCTCACTGTTGATAAGTCCC



GATGGCAGGAGGGGAATGTTTTCAGCTGTAGCGTGATGCA



CGAAGCTCTCCACAACCACTATACACAGAAAAGTTTGTCT



TTGTCCCTTGGAAAAtaG







TGFb neutralizing peptide



(Human)



(SEQ ID NO: 99)



ATGAGTACATCCTTTCCAGAGCTGGATCTGGAGAATTTTG



AGTATGACGACAGTGCCGAAGCCTGCTACCTCGGGGACAT



AGTCGCATTCGGGACAATCTTTTTGTCTGTATTTTACGCC



CTGGTGTTTACATTTGGCCTGGTTGGAAATCTGTTGGTCG



TACTCGCTCTCACCAATTCCCGAAAACCCAAAAGTATAAC



AGACATATACCTGTTGAATCTGGCACTGAGTGACCTTTTG



TTCGTCGCCACCCTTCCTTTTTGGACACACTACCTTATCA



GTCACGAGGGGCTTCATAATGCTATGTGCAAGCTCACTAC



TGCCTTCTTCTTTATCGGATTCTTCGGGGGTATCTTTTTT



ATCACAGTTATTAGCATTGACCGATACCTTGCCATAGTGC



TCGCAGCCAACTCAATGAACAACCGCACCGTGCAGCATGG



AGTGACTATTTCCTTGGGTGTGTGGGCCGCTGCTATACTT



GTCGCCAGCCCTCAATTCATGTTTACCAAAAGGAAAGACA



ATGAGTGCCTCGGAGATTACCCTGAGGTGTTGCAAGAAAT



GTGGCCTGTACTTCGAAATAGCGAAGTGAATATACTCGGC



TTTGCTCTTCCTCTGCTCATCATGTCATTCTGTTATTTTC



GAATAATCCAAACATTGTTCAGCTGTAAGAACCGAAAGAA



AGCCCGCGCCGTACGCCTGATTCTGCTCGTTGTGTTCGCC



TTTTTTCTGTTTTGGACTCCTTACAACATAATGATATTCC



TGGAGACTCTCAAATTCTATAACTTTTTTCCCTCCTGTGA



TATGAAAAGGGACCTTAGATTGGCTCTCAGTGTCACTGAA



ACAGTAGCCTTTAGCCATTGTTGTCTCAACCCTTTCATAT



ATGCATTTGCAGGGGAAAAGTTCCGGCGGTATCTCGGACA



TTTGTATCGGAAGTGCTTGGCCGTGTTGTGTGGTCATCCT



GTCCATACCGGATTCTCTCCTGAGAGTCAACGGAGCCGCC



AAGATTCAATCCTGTCCAGTTTCACTCACTATACTTCAGA



GGGGGATGGCAGCCTTCTGCTC







Kyneurinase #1



(SEQ ID NO: 100)



ATGGAGACCGACACTTTGTTGCTGTGGGTACTTTTGTTGT



GGGTCCCAGGATCTACCGGGGATATGGAACCCTCTCCTCT



TGAACTGCCAGTAGACGCCGTGCGCCGCATTGCAGCCGAG



TTGAATTGCGATCCAACAGATGAACGCGTTGCCCTGAGGC



TCGACGAAGAGGATAAATTGTCACATTTCAGGAACTGCTT



TTACATTCCAAAGATGAGGGATCTTCCATCCATAGATCTT



AGCCTCGTGTCCGAGGATGACGATGCCATATATTTTCTTG



GGAACAGTCTTGGGTTGCAGCCAAAAATGGTACGGACATA



TCTCGAAGAGGAGCTGGACAAATGGGCTAAAATGGGTGCT



TACGGCCACGACGTGGGAAAACGCCCCTGGATAGTTGGCG



ACGAATCTATCGTGAGTCTTATGAAAGATATAGTTGGAGC



ACATGAGAAAGAAATTGCACTGATGAATGCCCTTACTATC



AATCTGCATCTCCTCTTGCTTTCATTCTTTAAGCCCACTC



CTAAACGCCACAAAATACTTTTGGAAGCAAAAGCCTTTCC



AAGCGACCACTACGCTATTGAGTCACAAATACAACTCCAT



GGACTTGATGTGGAAAAGTCTATGCGGATGGTAAAACCAC



GCGAAGGCGAGGAGACCCTTCGAATGGAGGACATACTTGA



GGTCATCGAAGAAGAAGGAGATAGTATAGCAGTTATCCTT



TTCAGCGGGCTGCACTTCTACACAGGTCAACTCTTTAACA



TTCCAGCTATTACTAAGGCAGGCCACGCTAAAGGATGCTT



CGTGGGCTTTGACCTTGCACACGCAGTAGGAAACGTAGAG



CTCCGCTTGCACGATTGGGGCGTTGATTTCGCCTGCTGGT



GTTCATATAAGTATCTTAACTCAGGAGCTGGTGGGTTGGC



AGGCGCATTCGTACACGAGAAACACGCTCATACCGTAAAG



CCTGCACTGGTAGGGTGGTTCGGACACGATCTCTCTACCC



GCTTCAATATGGATAATAAACTCCAGCTTATACCTGGCGC



CAATGGATTCAGGATCTCAAATCCTCCTATTTTGCTCGTT



TGCAGTTTGCACGCATCTCTTGAGGTGTTCCAGCAGGCTA



CCATGACTGCACTCCGCCGGAAGTCAATCCTTTTGACCGG



ATACTTGGAGTATATGCTGAAACATTATCACTCAAAAGAT



AACACTGAGAATAAGGGCCCCATAGTAAACATTATCACTC



CATCTCGGGCTGAAGAGCGCGGCTGCCAACTCACATTGAC



TTTTTCCATTCCCAAGAAGTCAGTGTTCAAAGAGTTGGAG



AAACGGGGGGTTGTATGTGATAAGCGGGAGCCAGATGGAA



TCCGCGTTGCCCCAGTCCCCCTCTATAATTCTTTTCACGA



TGTATACAAGTTTATTAGACTGCTGACAAGTATCTTGGAC



TCATCTGAGCGATCTtaG







Kyneurinase #2



(SEQ ID NO: 101)



ATGGAACCCTCTCCTCTTGAACTGCCAGTAGACGCCGTGC



GCCGCATTGCAGCCGAGTTGAATTGCGATCCAACAGATGA



ACGCGTTGCCCTGAGGCTCGACGAAGAGGATAAATTGTCA



CATTTCAGGAACTGCTTTTACATTCCAAAGATGAGGGATC



TTCCATCCATAGATCTTAGCCTCGTGTCCGAGGATGACGA



TGCCATATATTTTCTTGGGAACAGTCTTGGGTTGCAGCCA



AAAATGGTACGGACATATCTCGAAGAGGAGCTGGACAAAT



GGGCTAAAATGGGTGCTTACGGCCACGACGTGGGAAAACG



CCCCTGGATAGTTGGCGACGAATCTATCGTGAGTCTTATG



AAAGATATAGTTGGAGCACATGAGAAAGAAATTGCACTGA



TGAATGCCCTTACTATCAATCTGCATCTCCTCTTGCTTTC



ATTCTTTAAGCCCACTCCTAAACGCCACAAAATACTTTTG



GAAGCAAAAGCCTTTCCAAGCGACCACTACGCTATTGAGT



CACAAATACAACTCCATGGACTTGATGTGGAAAAGTCTAT



GCGGATGGTAAAACCACGCGAAGGCGAGGAGACCCTTCGA



ATGGAGGACATACTTGAGGTCATCGAAGAAGAAGGAGATA



GTATAGCAGTTATCCTTTTCAGCGGGCTGCACTTCTACAC



AGGTCAACTCTTTAACATTCCAGCTATTACTAAGGCAGGC



CACGCTAAAGGATGCTTCGTGGGCTTTGACCTTGCACACG



CAGTAGGAAACGTAGAGCTCCGCTTGCACGATTGGGGCGT



TGATTTCGCCTGCTGGTGTTCATATAAGTATCTTAACTCA



GGAGCTGGTGGGTTGGCAGGCGCATTCGTACACGAGAAAC



ACGCTCATACCGTAAAGCCTGCACTGGTAGGGTGGTTCGG



ACACGATCTCTCTACCCGCTTCAATATGGATAATAAACTC



CAGCTTATACCTGGCGCCAATGGATTCAGGATCTCAAATC



CTCCTATTTTGCTCGTTTGCAGTTTGCACGCATCTCTTGA



GGTGTTCCAGCAGGCTACCATGACTGCACTCCGCCGGAAG



TCAATCCTTTTGACCGGATACTTGGAGTATATGCTGAAAC



ATTATCACTCAAAAGATAACACTGAGAATAAGGGCCCCAT



AGTAAACATTATCACTCCATCTCGGGCTGAAGAGCGCGGC



TGCCAACTCACATTGACTTTTTCCATTCCCAAGAAGTCAG



TGTTCAAAGAGTTGGAGAAACGGGGGGTTGTATGTGATAA



GCGGGAGCCAGATGGAATCCGCGTTGCCCCAGTCCCCCTC



TATAATTCTTTTCACGATGTATACAAGTTTATTAGACTGC



TGACAAGTATCTTGGACTCATCTGAGCGATCTtaG







VEGF



(SEQ ID NO: 102)



ATGAATTTCTTGCTGAGCTGGGTGCATTGGACACTCGCAT



TGTTGCTGTACTTGCACCATGCCAAGTGGTCCCAGGCTGC



ACCCACTACTGAGGGCGAGCAAAAGTCTCATGAGGTGATT



AAATTTATGGACGTTTACCAACGATCATACTGTCGGCCAA



TCGAAACCCTCGTAGATATATTCCAGGAGTACCCAGACGA



GATCGAATACATTTTCAAGCCCTCATGTGTCCCATTGATG



CGATGTGCTGGGTGCTGTAACGACGAAGCACTTGAATGTG



TCCCCACCTCCGAGAGTAACATCACAATGCAAATAATGAG



AATCAAGCCCCACCAATCCCAACATATCGGTGAAATGTCA



TTCCTTCAGCATTCCCGCTGCGAGTGCCGGCCTAAGAAGG



ACCGCACCAAACCAGAGAACCATTGTGAACCCTGTTCTGA



GAGACGGAAGCACTTGTTCGTACAGGACCCTCAAACATGC



AAGTGCAGCTGTAAGAATACCGACTCACGGTGTAAAGCTA



GGCAACTGGAGCTTAATGAAAGGACCTGCCGATGCGATAA



ACCCAGGAGGtaa







GM-CSF



(SEQ ID NO: 103)



ATGTGGTTGCAGAATTTGCTCTTCCTGGGGATTGTGGTCT



ACAGCCTCTCCGCACCTACCCGCTCTCCTATCACAGTTAC



AAGACCCTGGAAACATGTGGAGGCCATTAAAGAAGCATTG



AATTTGTTGGACGATATGCCCGTCACCCTGAATGAAGAAG



TAGAAGTTGTTTCTAATGAGTTCAGCTTTAAAAAATTGAC



CTGTGTGCAGACACGGCTTAAAATTTTTGAACAGGGACTT



AGAGGAAACTTTACTAAGCTGAAGGGGGCACTTAACATGA



CAGCTTCTTATTATCAGACCTATTGTCCTCCAACACCTGA



AACCGACTGTGAAACACAGGTAACCACTTACGCCGATTTT



ATTGATTCTTTGAAAACATTCCTCACCGATATACCATTTG



AGTGTAAGAAGCCAGGCCAAAAGtaG







Anti-PD1



(SEQ ID NO: 104)



ATGGAAACTGACACACTTCTTCTGTGGGTCTTGCTCCTGT



GGGTCCCAGGCTCTACTGGTGACAGTCCTGATAGGCCATG



GAACCCACCTACCTTTAGTCCAGCCTTGCTCGTCGTAACC



GAAGGGGACAACGCTACATTCACCTGCTCTTTTAGCAATA



CTTCTGAGAGTTTTCATGTAGTCTGGCATCGGGAGAGTCC



ATCCGGACAAACAGATACTTTGGCCGCTTTTCCAGAGGAT



AGGTCTCAACCTGGGCAAGACGCAAGGTTTCGAGTCACAC



AGCTTCCTAACGGGAGAGATTTTCACATGTCTGTAGTTCG



GGCACGCCGAAATGATTCTGGCACATATGTTTGCGGTGTG



ATCTCACTTGCTCCAAAGATTCAAATAAAGGAGAGCCTTC



GCGCCGAGTTGCGGGTGACTGAGCGGGAGCCCAAGTCCTG



CGACAAAACCCATACTTGTCCACCCTGTGGCGGCGGGTCA



TCCGGTGGCGGGTCTGGGGGGCAACCAAGAGAGCCACAGG



TATATACTCTTCCCCCCAGCAGAGAAGAAATGACAAAAAA



CCAAGTGTCCCTGACATGTCTGGTTAAAGGATTTTATCCC



AGTGACATTGCTGTAGAATGGGAATCCAATGGTCAACCCG



AGAATAACTACAAAACCACTCCTCCAGTATTGGACAGTGA



CGGTTCCTTCTTCCTCTATTCCAAACTTACAGTGGATAAA



TCCCGCTGGCAGCAAGGGAATGTATTCAGCTGTAGTGTCA



TGCACGAAGCTCTTCATAACCATTATACACAGAAATCTCT



TTCCCTGAGCCCAGGTAAAtaG







Adenosine Deaminase (ADA) #1



(Mouse)



(SEQ ID NO: 105)



ATGGAGACTGATACACTTTTGCTCTGGGTTTTGCTCTTGT



GGGTACCAGGGTCTACTGGAGATGCACAAACTCCTGCATT



CAACAAGCCTAAGGTAGAGCTTCATGTCCATTTGGACGGA



GCCATAAAACCTGAAACCATACTCTATTTCGGCAAGAAAC



GGGGTATAGCACTTCCCGCTGATACCGTGGAAGAGTTGAG



AAATATCATTGGCATGGACAAACCTCTTAGCCTGCCTGGC



TTTCTTGCAAAGTTCGACTACTATATGCCAGTTATAGCAG



GGTGTAGAGAAGCAATAAAGCGAATCGCCTATGAGTTCGT



TGAGATGAAGGCTAAAGAAGGAGTTGTTTACGTGGAAGTC



CGGTACTCACCTCATCTGCTTGCTAATAGCAAGGTGGACC



CAATGCCATGGAATCAAACTGAAGGTGATGTAACCCCTGA



CGATGTGGTCGATTTGGTCAATCAAGGTCTCCAAGAAGGC



GAGCAGGCTTTCGGCATTAAGGTAAGAAGTATATTGTGCT



GTATGCGACATCAACCTTCATGGTCCCTGGAGGTCCTCGA



ATTGTGCAAAAAGTACAATCAAAAAACAGTGGTCGCAATG



GATCTCGCTGGAGATGAGACCATAGAAGGTTCCTCTCTTT



TCCCCGGTCATGTCGAAGCATATGAAGGGGCTGTCAAAAA



TGGTATCCACCGCACCGTCCACGCAGGGGAAGTAGGGTCC



CCAGAAGTAGTCAGGGAAGCCGTTGACATTTTGAAAACAG



AAAGAGTCGGGCATGGCTACCATACAATAGAGGACGAAGC



CTTGTACAATCGACTTTTGAAAGAAAATATGCACTTCGAG



GTCTGTCCCTGGAGTTCATATCTCACCGGAGCATGGGACC



CCAAAACAACCCACGCCGTCGTACGCTTCAAGAATGATAA



GGCAAACTACAGTTTGAATACAGATGATCCACTGATATTC



AAGTCAACACTTGACACTGACTACCAGATGACAAAAAAAG



ATATGGGTTTCACCGAAGAAGAGTTCAAGAGATTGAACAT



TAACGCAGCAAAAAGCTCCTTCCTGCCAGAGGAAGAGAAA



AAAGAATTGCTTGAAAGGTTGTATCGAGAATACCAA







Adenosine Deaminase (ADA) #2



(Mouse)



(SEQ ID NO: 106)



ATGGCACAAACTCCAGCTTTTAATAAGCCCAAAGTGGAAC



TTCATGTTCATCTGGATGGGGCAATTAAGCCCGAAACTAT



ATTGTACTTTGGCAAAAAGAGGGGTATTGCCCTGCCAGCA



GATACCGTTGAGGAGCTTCGCAACATCATTGGGATGGACA



AGCCCCTCTCTCTGCCAGGTTTTCTCGCTAAATTCGATTA



TTATATGCCTGTTATTGCTGGTTGCCGGGAGGCCATCAAG



AGGATAGCCTACGAGTTTGTTGAGATGAAGGCCAAAGAGG



GCGTGGTGTACGTAGAGGTCAGATACAGCCCTCACCTGCT



TGCCAACAGCAAGGTGGACCCAATGCCCTGGAACCAAACC



GAGGGGGATGTCACTCCCGACGACGTTGTAGACCTCGTAA



ATCAGGGCCTTCAAGAGGGCGAGCAGGCATTTGGCATAAA



AGTCCGGTCTATACTCTGCTGTATGAGGCACCAACCCTCC



TGGTCTTTGGAGGTACTTGAGTTGTGTAAGAAATACAATC



AAAAGACTGTAGTCGCCATGGATCTTGCAGGCGATGAAAC



CATCGAGGGTAGCTCCTTGTTCCCTGGACATGTTGAAGCC



TACGAGGGGGCCGTAAAAAATGGGATACACAGGACTGTCC



ACGCTGGTGAAGTCGGAAGCCCAGAGGTGGTAAGGGAGGC



AGTTGACATACTCAAGACAGAGCGGGTTGGACACGGATAC



CACACAATTGAGGACGAGGCCCTGTATAACCGCCTCCTCA



AAGAGAACATGCATTTTGAGGTGTGTCCTTGGTCCAGCTA



CCTGACTGGTGCTTGGGACCCTAAAACAACTCACGCCGTG



GTCCGGTTCAAGAACGATAAAGCCAATTACTCTTTGAATA



CCGACGACCCCCTCATATTCAAATCAACATTGGATACCGA



CTACCAAATGACCAAAAAGGATATGGGGTTTACTGAAGAG



GAGTTCAAGAGGCTCAACATAAATGCCGCTAAATCCTCCT



TTCTCCCCGAGGAAGAAAAAAAAGAACTCCTTGAGCGGCT



GTATAGGGAGTATCAA







4-1BBL #1 (Mouse)



(SEQ ID NO: 107)



ATGGAAACAGATACACTCTTGCTCTGGGTACTGCTTCTGT



GGGTCCCCGGCTCTACTGGGGATGAAGATGATGTAACTAC



TACAGAAGAACTCGCTCCCGCTCTTGTCCCCCCACCCAAG



GGTACCTGCGCCGGTTGGATGGCTGGCATCCCAGGACATC



CAGGTCACAACGGTACCCCCGGAAGAGATGGTCGGGATGG



AACTCCCGGCGAGAAGGGCGAAAAAGGGGATGCAGGGCTT



CTGGGACCTAAAGGTGAAACAGGGGACGTTGGAATGACTG



GTGCAGAAGGGCCTCGCGGCTTTCCTGGCACCCCTGGGAG



GAAAGGAGAGCCCGGAGAGCTCCAGAGAACTGAACCTCGG



CCTGCACTCACTATAACTACTTCCCCTAATCTTGGGACCC



GCGAGAACAACGCCGATCAGGTTACACCTGTAAGCCATAT



CGGGTGCCCCAATACTACCCAGCAAGGGAGTCCCGTGTTC



GCAAAGCTTTTGGCTAAAAACCAAGCATCCCTGTGTAACA



CTACTCTTAATTGGCATTCACAAGACGGTGCTGGTAGCTC



TTATCTTTCTCAGGGGCTGCGGTACGAAGAAGATAAGAAG



GAATTGGTTGTGGATTCTCCAGGACTCTATTATGTCTTTC



TCGAATTGAAGCTCAGTCCCACCTTCACAAACACTGGACA



CAAAGTCCAGGGCTGGGTAAGTCTGGTACTCCAAGCAAAG



CCCCAGGTTGACGATTTCGACAATTTGGCACTCACCGTAG



AGCTTTTCCCATGCTCCATGGAAAATAAACTTGTTGATCG



GTCATGGTCACAGCTCTTGCTGCTTAAGGCAGGGCATCGC



CTCTCAGTGGGTCTGAGAGCTTATTTGCATGGTGCACAAG



ATGCTTACAGGGATTGGGAATTGTCCTACCCAAACACTAC



AAGTTTCGGGTTGTTCCTTGTCAAACCTGATAACCCATGG



GAGtaG







4-1BBL #2 (Mouse)



(SEQ ID NO: 108)



ATGGAAACTGATACACTCCTCCTGTGGGTCCTTCTTTTGT



GGGTGCCCGGATCAACCGGCGATGGCTGGATGGCAGGCAT



CCCAGGACACCCAGGACACAACGGTACTCCAGGTCGAGAC



GGTCGGGATGGGACTCCTGGGGAGAAAGGCGAGAAAGGGG



ACGCTGGTTTGCTCGGTCCTAAGGGGGAAACCGGGGATGT



AGGAATGACAGGGGCTGAAGGGCCTCGGGGATTTCCTGGG



ACACCAGGCAGGAAGGGTGAACCAGGGGAGGCCCTCCAGC



GCACCGAGCCACGGCCAGCTCTGACCATAACAACAAGTCC



AAACCTGGGCACACGCGAAAACAATGCTGACCAGGTGACT



CCTGTAAGTCACATCGGATGCCCTAACACTACACAACAGG



GCTCTCCTGTATTTGCAAAGCTTCTCGCAAAAAATCAAGC



ATCACTTTGTAATACAACCCTGAACTGGCATTCTCAGGAC



GGAGCAGGGTCCTCTTATTTGTCTCAAGGGCTCCGCTACG



AAGAAGATAAAAAGGAATTGGTTGTTGACAGTCCAGGTTT



GTATTATGTGTTTTTGGAACTTAAGCTGTCACCAACCTTC



ACTAACACCGGCCACAAGGTCCAAGGCTGGGTTAGTCTTG



TTTTGCAAGCCAAACCTCAAGTGGATGATTTTGACAATCT



GGCTTTGACTGTTGAGCTTTTTCCATGCAGTATGGAGAAT



AAACTGGTTGATCGGTCATGGTCACAGCTCCTTCTGCTCA



AGGCCGGACATAGGCTGAGTGTGGGACTTCGGGCCTACTT



GCACGGCGCCCAGGACGCATACCGAGACTGGGAACTCAGC



TACCCTAACACAACTTCTTTTGGGTTGTTCCTTGTCAAAC



CCGATAATCCTTGGGAAtaG







HPGE2 #1 (Mouse)



(SEQ ID NO: 109)



ATGGAGACTGATACTTTGCTCCTGTGGGTTCTTCTCCTGT



GGGTTCCTGGTTCCACAGGGGATATGCATGTCAATGGCAA



GGTAGCACTCGTGACTGGGGCTGCACAGGGTATCGGGAAA



GCTTTTGCCGAGGCCCTGTTGCTGCATGGCGCCAAGGTCG



CTTTGGTAGATTGGAACTTGGAGGCTGGAGTTAAATGCAA



AGCTGCACTCGACGAACAATTTGAGCCTCAAAAAACCCTC



TTTGTGCAGTGTGACGTTGCTGACCAAAAGCAACTCAGGG



ACACATTCAGGAAGGTCGTAGACCATTTCGGACGCCTCGA



TATACTCGTTAATAATGCCGGGGTAAACAACGAAAAGAAC



TGGGAACAAACATTGCAAATCAACCTGGTAAGTGTCATTA



GCGGAACTTATCTGGGTCTTGATTATATGAGCAAGCAGAA



CGGGGGCGAGGGCGGGATCATTATCAACATGTCAAGTCTT



GCCGGATTGATGCCAGTTGCTCAGCAGCCTGTTTACTGTG



CCAGCAAGCACGGTATTATTGGGTTTACCCGGAGTGCCGC



CATGGCCGCAAATCTTATGAAGAGTGGGGTAAGACTGAAT



GTTATCTGCCCAGGTTTCGTAGATACCCCAATCCTGGAGA



GCATCGAGAAGGAGGAAAATATGGGACAATACATTGAATA



TAAAGATCAAATCAAGGCTATGATGAAGTTCTACGGGGTT



CTGCATCCATCCACAATTGCCAACGGGCTCATTAATCTGA



TTGAGGACGACGCCTTGAACGGAGCTATAATGAAAATCAC



AGCTTCCAAAGGCATTCACTTCCAAGATTATGATATATCA



CCCTTGCTTGTCAAGGCTCCTCTGACAAGT







HPGE2 #2 (Mouse)



(SEQ ID NO: 110)



ATGCATGTCAATGGCAAGGTAGCACTCGTGACTGGGGCTG



CACAGGGTATCGGGAAAGCTTTTGCCGAGGCCCTGTTGCT



GCATGGCGCCAAGGTCGCTTTGGTAGATTGGAACTTGGAG



GCTGGAGTTAAATGCAAAGCTGCACTCGACGAACAATTTG



AGCCTCAAAAAACCCTCTTTGTGCAGTGTGACGTTGCTGA



CCAAAAGCAACTCAGGGACACATTCAGGAAGGTCGTAGAC



CATTTCGGACGCCTCGATATACTCGTTAATAATGCCGGGG



TAAACAACGAAAAGAACTGGGAACAAACATTGCAAATCAA



CCTGGTAAGTGTCATTAGCGGAACTTATCTGGGTCTTGAT



TATATGAGCAAGCAGAACGGGGGCGAGGGCGGGATCATTA



TCAACATGTCAAGTCTTGCCGGATTGATGCCAGTTGCTCA



GCAGCCTGTTTACTGTGCCAGCAAGCACGGTATTATTGGG



TTTACCCGGAGTGCCGCCATGGCCGCAAATCTTATGAAGA



GTGGGGTAAGACTGAATGTTATCTGCCCAGGTTTCGTAGA



TACCCCAATCCTGGAGAGCATCGAGAAGGAGGAAAATATG



GGACAATACATTGAATATAAAGATCAAATCAAGGCTATGA



TGAAGTTCTACGGGGTTCTGCATCCATCCACAATTGCCAA



CGGGCTCATTAATCTGATTGAGGACGACGCCTTGAACGGA



GCTATAATGAAAATCACAGCTTCCAAAGGCATTCACTTCC



AAGATTATGATATATCACCCTTGCTTGTCAAGGCTCCTCT



GACAAGT










Secretion Signals

The one or more effector molecules of the chimeric proteins provided for herein can be secretable effector molecules having a secretion signal peptide (also referred to as a signal peptide or signal sequence) at the effector molecule's N-terminus that direct newly synthesized proteins destined for secretion or membrane localization (also referred to as membrane insertion) to the proper protein processing pathways. In general, for all membrane-cleavable chimeric proteins described herein, the one or more effector molecules are secretable effector molecules (referred to as “S” in the formula S-C-MT-D). In embodiments with two or more effector molecules, each effector molecule can comprise a secretion signal. In embodiments with two or more effector molecules, each effector molecule can comprise a secretion signal such that each effector molecule is secreted from an engineered cell.


The secretion signal peptide operably associated with a effector molecule can be a native secretion signal peptide native secretion signal peptide (e.g., the secretion signal peptide generally endogenously associated with the given effector molecule). The secretion signal peptide operably associated with a effector molecule can be a non-native secretion signal peptide native secretion signal peptide. Non-native secretion signal peptides can promote improved expression and function, such as maintained secretion, in particular environments, such as tumor microenvironments. Non-limiting examples of non-native secretion signal peptide are shown in Table 3.









TABLE 3







Exemplary Signal Secretion Peptides










Name
Protein SEQUENCE
Source (Uniprot)
DNA SEQUENCE





IL-12
MCHQQLVISWFSL
P29460
ATGTGTCACCAGCAGCTCGTTATATC



VFLASPLVA (SEQ

CTGGTTTAGTTTGGTGTTTCTCGCTT



ID NO: 112)

CACCCCTGGTGGCA (SEQ ID NO: 31)





IL-12 (Codon
MCHQQLVISWFSL

ATGTGCCATCAGCAACTCGTCATCTC


Optimized)
VFLASPLVA (SEQ

CTGGTTCTCCCTTGTGTTCCTCGCTTC



ID NO: 112)

CCCTCTGGTCGCC (SEQ ID NO: 32)





IL-2 (Optimized)
MQLLSCIALILALV

ATGCAACTGCTGTCATGTATCGCACT



(SEQ ID NO: 113)

CATCCTGGCGCTGGTA (SEQ ID NO:





33)





IL-2 (Native)
MYRMQLLSCIALSL
P60568
ATGTATCGGATGCAACTTTTGAGCTG



ALVTNS (SEQ ID

CATCGCATTGTCTCTGGCGCTGGTGA



NO: 114)

CAAATTCC (SEQ ID NO: 34)





Trypsinogen-2
MNLLLILTFVAAAV
P07478
ATGAATCTCTTGCTCATACTTACGTT



A (SEQ ID NO: 115)

TGTCGCTGCTGCCGTTGCG (SEQ ID





NO: 35)






Gaussia

MGVKVLFALICIAV

ATGGGCGTGAAGGTCTTGTTTGCCCT



Luciferase

AEA (SEQ ID NO:

TATCTGCATAGCTGTTGCGGAGGCG



116)

(SEQ ID NO: 36)





CD5
MPMGSLQPLATLY
P06127
ATGCCGATGGGGAGCCTTCAACCTTT



LLGMLVASCLG

GGCAACGCTTTATCTTCTGGGGATGT



(SEQ ID NO: 117)

TGGTTGCTAGTTGCCTTGGG (SEQ ID





NO: 37)





IgKVII (mouse)
METDTLLLWVLLL

ATGGAAACTGACACGTTGTTGCTGT



WVPGSTGD (SEQ

GGGTATTGCTCTTGTGGGTCCCAGGA



ID NO: 118)

TCTACGGGCGAC (SEQ ID NO: 38)





IgKVII (human)
MDMRVPAQLLGLL
P01597
ATGGATATGAGGGTTCCCGCCCAGC



LLWLRGARC (SEQ

TTTTGGGGCTGCTTTTGTTGTGGCTT



ID NO: 119)

CGAGGGGCTCGGTGT (SEQ ID NO:





39)





VSV-G
MKCLLYLAFLFIGV

ATGAAGTGTCTGTTGTACCTGGCGTT



NC (SEQ ID NO:

TCTGTTCATTGGTGTAAACTGT (SEQ



120)

ID NO: 40)





Prolactin
MNIKGSPWKGSLL
P01236
ATGAATATCAAAGGAAGTCCGTGGA



LLLVSNLLLCQSVA

AGGGTAGTCTCCTGCTGCTCCTCGTA



P (SEQ ID NO: 121)

TCTAACCTTCTCCTTTGTCAATCCGT





GGCACCC (SEQ ID NO: 41)





Serum albumin
MKWVTFISLLFLFS
P02768
ATGAAATGGGTAACATTCATATCAC


preproprotein
SAYS (SEQ ID NO:

TTCTCTTTCTGTTCAGCTCTGCGTATT



122)

CT (SEQ ID NO: 42)





Azurocidin
MTRLTVLALLAGL
20160
ATGACAAGGCTTACTGTTTTGGCTCT


preproprotein
LASSRA (SEQ ID

CCTCGCTGGACTCTTGGCTTCCTCCC



NO: 123)

GAGCA (SEQ ID NO: 43)





Osteonectin
MRAWIFFLLCLAG
P09486
ATGAGGGCTTGGATTTTTTTTCTGCT


(BM40)
RALA (SEQ ID NO:

CTGCCTTGCCGGTCGAGCCCTGGCG



124)

(SEQ ID NO: 44)





CD33
MPLLLLLPLLWAG
P20138
ATGCCTCTTCTGCTTTTGCTTCCTCTT



ALA (SEQ ID NO:

TTGTGGGCAGGTGCCCTCGCA (SEQ



125)

ID NO: 45)





IL-6
MNSFSTSAFGPVAF
P05231
ATGAACTCTTTCTCAACCTCTGCGTT



SLGLLLVLPAAFPA

TGGTCCGGTCGCTTTCTCCCTTGGGC



P (SEQ ID NO: 126)

TCCTGCTTGTCTTGCCAGCAGCGTTT





CCTGCGCCA (SEQ ID NO: 46)





IL-8
MTSKLAVALLAAF
P10145
ATGACAAGTAAACTGGCGGTAGCCT



LISAALC (SEQ ID

TGCTCGCGGCCTTTTTGATTTCCGCA



NO: 127)

GCCCTTTGT (SEQ ID NO: 47)





CCL2
MKVSAALLCLLLIA
P13500
ATGAAGGTAAGTGCAGCGTTGCTTT



ATFIPQGLA (SEQ

GCCTTCTCCTCATTGCAGCGACCTTT



ID NO: 128)

ATTCCTCAAGGGCTGGCC (SEQ ID





NO: 48)





TIMP2
MGAAARTLRLALG
P16035
ATGGGAGCGGCAGCTAGAACACTTC



LLLLATLLRPADA

GACTTGCCCTTGGGCTCTTGCTCCTT



(SEQ ID NO: 129)

GCAACCCTCCTTAGACCTGCCGACG





CA (SEQ ID NO: 49)





VEGFB
MSPLLRRLLLAALL
P49765
ATGTCACCGTTGTTGCGGAGATTGCT



QLAPAQA (SEQ ID

GTTGGCCGCACTTTTGCAACTGGCTC



NO: 130)

CTGCTCAAGCC (SEQ ID NO: 50)





Osteoprotegerin
MNNLLCCALVFLDI
O00300
ATGAATAACCTGCTCTGTTGTGCGCT



SIKWTTQ (SEQ ID

CGTGTTCCTGGACATTTCTATAAAAT



NO: 131)

GGACAACGCAA (SEQ ID NO: 51)





Serpin El
MQMSPALTCLVLG
P05121
ATGCAAATGTCTCCTGCCCTTACCTG



LALVFGEGSA (SEQ

TCTCGTACTTGGTCTTGCGCTCGTAT



ID NO: 132)

TTGGAGAGGGATCAGCC (SEQ ID NO:





52)





GROalpha
MARAALSAAPSNP
P09341
ATGGCAAGGGCTGCACTCAGTGCTG



RLLRVALLLLLLVA

CCCCGTCTAATCCCAGATTGCTTCGA



AGRRAAG (SEQ ID

GTTGCATTGCTTCTTCTGTTGCTGGT



NO: 133)

TGCAGCTGGTAGGAGAGCAGCGGGT





(SEQ ID NO: 53)





CXCL12
MNAKVVVVLVLV
P48061
ATGAATGCAAAAGTCGTGGTCGTGC



LTALCLSDG (SEQ

TGGTTTTGGTTCTGACGGCGTTGTGT



ID NO: 134)

CTTAGTGATGGG (SEQ ID NO: 54)





IL-21 (Codon
MERIVICLMVIFLG
Q9HBE4
ATGGAACGCATTGTGATCTGCCTGAT


Optimized)
TL VHKSSS (SEQ ID

GGTCATCTTCCTGGGCACCTTAGTGC



NO: 135)

ACAAGTCGAGCAGC (SEQ ID NO: 55)





CD8
MALPVTALLLPLAL

ATGGCCTTACCAGTGACCGCCTTGCT



LLHAARP (SEQ ID

CCTGCCGCTGGCCTTGCTGCTCCACG



NO: 136)

CCGCCAGGCCG (SEQ ID NO: 139)





CD8 (Codon
MALPVTALLLPLAL

ATGGCGCTCCCGGTGACAGCACTTCT


Optimized)
LLHAARP (SEQ ID

CTTGCCTCTTGCCCTGCTGTTGCATG



NO: 137)

CCGCGCGCCCA (SEQ ID NO: 140)





GMCSF
MLLVTSLLLCELPH

ATGTTGCTCGTGACATCCCTCTTGCT



PAFLLIP (SEQ ID

TTGTGAGTTGCCTCATCCCGCATTCC



NO: 138)

TGCTCATCCCA (SEQ ID NO: 141)









Protease Cleavage Site

In certain embodiments, the chimeric proteins provided for herein (e.g., in general, for all membrane-cleavable chimeric proteins described herein) contain a protease cleavage site (e.g., referred to as “C” in the formula S-C-MT-D for membrane-cleavable chimeric proteins described herein). In general, the protease cleavage site can be any amino acid sequence motif capable of being cleaved by a protease. Examples of protease cleavage sites include, but are not limited to, a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, or an NS3 protease cleavage site. A specific example of a protease cleavage site is a hepatitis C virus (HCV) nonstructural protein 3 (NS3) protease cleavage site, including, but not limited to, a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B cleavage site. For a description of NS3 protease and representative sequences of its cleavage sites for various strains of HCV, see, e.g., Hepatitis C Viruses: Genomes and Molecular Biology (S. L. Tan ed., Taylor & Francis, 2006), Chapter 6, pp. 163-206; herein incorporated by reference in its entirety. For example, the sequences of HCV NS4A/4B protease cleavage site; HCV NS5A/5B protease cleavage site; C-terminal degron with NS4A/4B protease cleavage site; N-terminal degron with HCV NS5A/5B protease cleavage site are provided. Representative NS3 sequences are listed in the National Center for Biotechnology Information (NCBI) database. See, for example, NCBI entries: Accession Nos. YP_001491553, YP_001469631, YP_001469632, NP_803144, NP_671491, YP_001469634, YP_001469630, YP_001469633, ADA68311, ADA68307, AFP99000, AFP98987, ADA68322, AFP99033, ADA68330, AFP99056, AFP99041, CBF60982, CBF60817, AHH29575, AIZ00747, AIZ00744, AB136969, ABN05226, KF516075, KF516074, KF516056, AB826684, AB826683, JX171009, JX171008, JX171000, EU847455, EF154714, GU085487, JX171065, JX171063; all of which sequences (as entered by the date of filing of this application) are herein incorporated by reference.


The protease cleavage site can be C-terminal of the secretable effector molecule. The protease cleavage site can be N-terminal of the secretable effector molecule. In general, for all membrane-cleavable chimeric proteins described herein, the protease cleavage site is C-terminal of the secretable effector molecule and N-terminal of the cell membrane tethering domain (in other words, the protease cleavage site is in between the secretable effector molecule and the cell membrane tethering domain). The protease cleavage site can be connected to the secretable effector molecule by a polypeptide linker, i.e., a polypeptide sequence not generally considered to be part of the effector molecule or protease cleavage site. The protease cleavage site can be connected to the cell membrane tethering domain by a polypeptide linker, i.e., a polypeptide sequence not generally considered to be part of the cell membrane tethering domain or protease cleavage site. A polypeptide linker can be any amino acid sequence that connects a first polypeptide sequence and a second polypeptide sequence. A polypeptide linker can be a flexible linker (e.g., a Gly-Ser-Gly sequence). Examples of polypeptide linkers include, but are not limited to, GSG linkers (e.g., [GS]4GG [SEQ ID NO: 182]), A(EAAAK)3A (SEQ ID NO: 183), and Whitlow linkers (e.g., a “KEGS” (SEQ ID NO:195) linker such as the amino acid sequence KESGSVSSEQLAQFRSLD (SEQ ID NO: 184), a EGK linker such as the amino acid sequence EGKSSGSGSESKST (SEQ ID NO: 185), and linkers described in more detail in Issued U.S. Pat. No. 5,990,275 herein incorporated by reference). Other polypeptide linkers may be selected based on desired properties (e.g., length, flexibility, amino acid composition etc.) and are known to those skilled in the art.


In the Membrane-Cleavable Degron system, following expression and localization of the chimeric protein into the cell membrane, the protease cleavage site directs cleavage of the chimeric protein such that the effector molecule is released (“secreted”) into the extracellular space of a cell.


In general, a protease that cleaves the protease cleavage site is a protease specific for that specific protease cleavage site. For example, in the case of a disintegrin and metalloproteinase (“ADAM”) family protease, the protease that cleaves a specific ADAM protease cleavage site is generally limited to the ADAM protease(s) that specifically recognize the specific ADAM protease cleavage site motif. Proteases can be membrane-bound or membrane-associated. Proteases can be secreted, e.g., secreted in a specific cellular environment, such as a tumor microenvironment (“TME”).


A protease that cleaves the protease cleavage site of the chimeric protein can be expressed in the same cell that expresses the chimeric protein. A protease that cleaves the protease cleavage site of the chimeric protein can be endogenous to a cell expressing the chimeric protein. In other words, a cell engineered to express the chimeric protein can endogenously express the protease specific for the protease cleavage site present in the chimeric protein. Endogenous expression of the protease refers to both expression under generally homeostatic conditions (e.g., a cell generally considered to be healthy), and also to differential expression under non-homeostatic conditions (e.g., upregulated expression in a tumor cell). The protease cleavage site can be selected based on the known proteases endogenously expressed by a desired cell population. For example, a protease cleavage site can be selected for a specific tumor-associated protease known to be expressed in a particular tumor population of interest (e.g., in a specific tumor cell engineered to express the chimeric protein). Protease and/or expression databases can be used to select an appropriate protease cleavage site, such as selecting a protease cleavage site cleaved by a tumor-associated proteases through consulting Oncomine (www.oncomine.org), the European Bioinformatic Institute (www.ebi.ac.uk) in particular (www.ebi.ac.uk/gxa), PMAP (www.proteolysis.org), ExPASy Peptide Cutter (ca.expasy.org/tools/peptide cutter) and PMAP.Cut DB (cutdb.burnham.org), each of which is incorporated by reference for all purposes.


A protease that cleaves the protease cleavage site of the chimeric protein can be heterologous to a cell expressing the chimeric protein. For example, a cell engineered to express the chimeric protein can also be engineered to express a protease not generally expressed by the cell that is specific for the protease cleavage site present in the chimeric protein. A cell engineered to express both the chimeric protein and the protease can be engineered to express each from separate engineered nucleic acids or from a multicistronic systems (multicistronic and multi-promoter systems are described in greater detail in the Section herein titled “Multicistronic and Multiple Promoter Systems”). Heterologous proteases and their corresponding protease cleavage site can be selected as described above with reference to endogenous proteases.


A protease that cleaves the protease cleavage site of the chimeric protein can be expressed on a separate distinct cell than the cell that expresses the chimeric protein. For example, the protease can be generally expressed in a specific cellular environment, such as a tumor microenvironment. In such cases, in general, the cleavage of the protease cleavage site can be restricted to only those cellular environments of interest (e.g., a tumor microenvironment) due to the environment-restricted protease needing to come in contact with the protease cleavage site. In embodiments having membrane-cleavable chimeric proteins, in general, the secretion of the effector molecule can be restricted to only those cellular environments of interest (e.g., a tumor microenvironment) due to the environment-restricted protease needing to come in contact with the protease cleavage site. A protease that cleaves the protease cleavage site of the chimeric protein can be endogenous to the separate distinct cell. A protease that cleaves the protease cleavage site of the chimeric protein can be heterologous to the separate distinct cell. For example, the separate distinct cell can be engineered to express a protease not generally expressed by the separate distinct cell.


Proteases include, but are not limited to, a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease. A protease can be an NS3 protease.


Proteases can be tumor associated proteases, such as, a cathepsin, a cysteine protease, an aspartyl protease, a serine protease, or a metalloprotease. Specific examples of tumor associated proteases include Cathepsin B, Cathepsin L, Cathepsin S, Cathepsin D, Cathepsin E, Cathepsin A, Cathepsin G, Thrombin, Plasmin, Urokinase, Tissue Plasminogen Activator, Metalloproteinase 1 (MMP1), MMP2, MMP3, MMP4, MMP7, MMP8, MMP9, MMP1, MMP11, MMP12, MMP13, MMP14, MMP15, MMP16, MMP17, MMP20, MMP21, MMP23, MMP24, MMP25, MMP26, MMP28, ADAM, ADAMTS, CD10 (CALLA), or prostate specific antigen. Proteases can also include, but are not limited to, proteases listed in Table 4 below. Exemplary cognate protease cleavage sites for certain proteases are also listed in Table 4.









TABLE 4







Exemplary Proteases with Cognate Cleavage Sites and Inhibitors









Protease




(UniProt Accession No.)
Cognate cleavage site
Protease inhibitors





HCV NS4A/4B
DEMEECSQHL
Simeprevir,



(SEQ ID NO: 142)
Danoprevir,



EDVVPCSMG
Asunaprevir,



(SEQ ID NO: 143)
Ciluprevir,




Boceprevir,




Sovaprevir,




Paritaprevir,




Telaprevir,




Grazoprevir





HCV NS5A/5B
DEMEECSQHL
Simeprevir,



(SEQ ID NO: 142)
Danoprevir,



EDVVPCSMG
Asunaprevir,



(SEQ ID NO: 143)
Ciluprevir,




Boceprevir,




Sovaprevir,




Paritaprevir,




Telaprevir,




Grazoprevir





HCV NS3
DEMEECSQHL
Simeprevir,



(SEQ ID NO: 142)
Danoprevir,



EDVVPCSMG
Asunaprevir,



(SEQ ID NO: 143)
Ciluprevir,




Boceprevir,




Sovaprevir,




Paritaprevir,




Telaprevir,




Grazoprevir





HCV NS2-3
DEMEECSQHL
Simeprevir,



(SEQ ID NO: 142)
Danoprevir,



EDVVPCSMG
Asunaprevir,



(SEQ ID NO: 143)
Ciluprevir,




Boceprevir,




Sovaprevir,




Paritaprevir,




Telaprevir,




Grazoprevir





HIV-1 protease

Amprenavir,


(SEQ ID NO: 144)

Atazanavir,




Darunavir,




Fosamprenavir,




Indinavir, Lopinavir,




Nelfinavir, Ritonavir,




Saquinavir, Tipranavir





Signal peptidase (P67812,
preference of eukaryotic signal



P15367, P00804, P0803)
peptidase for cleavage after residue




20 (Xaa20↓) of pre(Δpro)apoA-II:




Ala, Cys > Gly > Ser, Thr > Pro >




Asn, Val, Ile, Leu, Tyr, His, Arg,




Asp.






proprotein convertases
(R/K)-X-(hydrophobic)-X↓, where



cleaving at hydrophobic
X is any amino acid



residues (e.g., Leu, Phe,




Val, or Met) (Q16549,




Q8NBP7, Q92824,




P29120, Q6UW60,




P29122, Q9QXV0)







proprotein convertases
(K/R)-(X)n-(K/R)↓, where n is 0, 2,



cleaving at small amino
4 or 6 and X is any amino acid



acid residues such as Ala




or Thr (Q16549,




Q8NBP7, Q92824,




P29120, Q6UW60,




P29122)







proopiomelanocortin
Cleavage at paired basic residues in



converting enzyme (PCE)
certain prohormones, either between



(Q9UO77615, O776133)
them, or on the carboxyl side






chromaffin granule
tends to cleave dipeptide bonds that



aspartic protease (CGAP)
have hydrophobic residues as well as




a beta-methylene group






prohormone thiol protease




(cathepsin L1) (P07154,




P07711, P06797, P25975,




Q28944)







carboxypeptidases (e.g.,
cleaves a peptide bond at the



carboxypeptidase E/H,
carboxy-terminal (C-terminal) end



carboxypeptidase D and
of a protein or peptide



carboxypeptidase Z)




(Q9M099, P15169,




Q04609, P08819, P08818,




O77564, P70627,




O35409, P07519,




Q8VZU3, P22792,




P15087, P16870,




Q9JHH6, Q96IY4,




Q7L8A9)







aminopeptidases (e.g.,
cleaves a peptide bond at the amino-



arginine aminopeptidase,
terminal (N-terminal) end of a



lysine aminopeptidase,
protein or peptide



aminopeptidase B)







prolyl endopeptidase
Hydrolysis of Pro-|-Xaa >> Ala-|-



(Q12884, P48147,
Xaa in oligopeptides.



P97321, Q4J6C6)
Release of an N-terminal dipeptide,




Xaa-Yaa-|-Zaa-, from a polypeptide,




preferentially when Yaa is Pro,




provided Zaa is neither Pro nor




hydroxyproline






aminopeptidase N
Release of an N-terminal amino



(P97449, P15144,
acid, Xaa-|-Yaa- from a peptide,



P15145, P15684)
amide or arylamide. Xaa is




preferably Ala, but may be most




amino acids including Pro (slow




action). When a terminal




hydrophobic residue is followed by a




prolyl residue, the two may be




released as an intact Xaa-Pro




dipeptide






insulin degrading enzyme
Degradation of insulin, glucagon and



(P14735, P35559,
other polypeptides. No action on



Q9JHR7, P22817,
proteins.



Q24K02)
Cleaves multiple short polypeptides




that vary considerably in sequence






Calpain (O08529,
No specific amino acid sequence is



P17655, Q07009,
uniquely recognized by calpains.



Q27971, P20807, P07384,
Amongst protein substrates, tertiary



O35350, 014815,
structure elements rather than



P04632, Q9Y6Q1,
primary amino acid sequences



O15484, Q9HC96,
appear to be responsible for



A6NHCO, Q9UMQ6)
directing cleavage to a specific




substrate. Amongst peptide and




small-molecule substrates, the most




consistently reported specificity is




for small, hydrophobic amino acids




(e.g., leucine, valine and isoleucine)




at the P2 position, and large




hydrophobic amino acids (e.g.,




phenylalanine and tyrosine) at the P1




position. One fluorogenic calpain




substrate is (EDANS)-Glu-Pro-Leu-




Phe═Ala-Glu-Arg-Lys-(DABCYL),




(EDANSEPLFAERKDABCYL




(SEQ ID NO: 145)) with cleavage




occurring at the Phe═Ala bond.






caspase 1 (P29466,
Strict requirement for an Asp residue



P29452)
at position P1 and has a preferred




cleavage sequence of Tyr-Val-Ala-




Asp-|- (YVAD; SEQ ID NO: 146).






caspase 2 (P42575,
Strict requirement for an Asp residue



P29594)
at P1, with 316-asp being essential




for proteolytic activity and has a




preferred cleavage sequence of Val-




Asp-Val-Ala-Asp-|- (VDVAD; SEQ




ID NO: 147).






caspase 3 (P42574,
Strict requirement for an Asp residue



P70677)
at positions P1 and P4. It has a




preferred cleavage sequence of Asp-




Xaa-Xaa-Asp-|- with a hydrophobic




amino-acid residue at P2 and a




hydrophilic amino-acid residue at




P3, although Val or Ala are also




accepted at this position.






caspase 4 (P70343,
Strict requirement for Asp at the P1



P49662)
position. It has a preferred cleavage




sequence of Tyr-Val-Ala-Asp-|-




(YVAD; SEQ ID NO: 146) but also




cleaves at Asp-Glu-Val-Asp-|-




(DEVD; SEQ ID NO: 148).






caspase 5 (P51878)
Strict requirement for Asp at the P1




position. It has a preferred cleavage




sequence of Tyr-Val-Ala-Asp-|-




(YVAD; SEQ ID NO: 146) but also




cleaves at Asp-Glu-Val-Asp-|- -|-




(DEVD; SEQ ID NO: 148).






caspase 6 (P55212)
Strict requirement for Asp at




position P1 and has a preferred




cleavage sequence of Val-Glu-His-




Asp-|-(VEHD; SEQ ID NO: 149).






caspase 7 (P97864,
Strict requirement for an Asp residue



P55210)
at position P1 and has a preferred




cleavage sequence of Asp-Glu-Val-




Asp-|- (DEVD; SEQ ID NO: 148).






caspase 8 (Q8IRY7,
Strict requirement for Asp at



089110, Q14790)
position P1 and has a preferred




cleavage sequence of




(Leu/Asp/Val)-Glu-Thr-Asp-|-




(Gly/Ser/Ala).






caspase 9 (P55211,
Strict requirement for an Asp residue



Q8C3Q9, Q5IS54)
at position P1 and with a marked




preference for His at position P2. It




has a preferred cleavage sequence of




Leu-Gly-His-Asp-|-Xaa (LGHD;




SEQ ID NO: 150).






caspase 10 (Q92851)
Strict requirement for Asp at




position P1 and has a preferred




cleavage sequence of Leu-Gln-Thr-




Asp-|-Gly (LQTDG; SEQ ID NO:




151).






puromycin sensitive
Release of an N-terminal amino



aminopeptidase (P55786,
acid, preferentially alanine, from a



Q11011)
wide range of peptides, amides and




arylamides.






angiotensin converting
Release of a C-terminal dipeptide,
Benazepril (Lotensin),


enzyme (ACE) (P12821,
oligopeptide-|-Xaa-Yaa, when Xaa is
Captopril, Enalapril


P09470, Q9BYF1)
not Pro, and Yaa is neither Asp nor
(Vasotec), Fosinopril,


SEQ ID NO: 156
Glu.
Lisinopril (Prinivil,




Zestril), Moexipril,




Perindopril (Aceon),




Quinapril (Accupril),




Ramipril (Altace),




Trandolapril (Mavik),




Zofenopril





pyroglutamyl peptidase II
Release of the N-terminal



(Q9NXJ5)
pyroglutamyl group from pGlu--His-




Xaa tripeptides and pGlu--His-Xaa-




Gly tetrapeptides






dipeptidyl peptidase IV
Release of an N-terminal dipeptide,



(P27487, P14740,
Xaa-Yaa-|-Zaa-, from a polypeptide,



P28843)
preferentially when Yaa is Pro,




provided Zaa is neither Pro nor




hydroxyproline






N-arginine dibasic
Hydrolysis of polypeptides,



convertase (O43847,
preferably at -Xaa-|-Arg-Lys-, and



Q8BHG1)
less commonly at -Arg-|-Arg-Xaa-,




in which Xaa is not Arg or Lys






endopeptidase 24.15
Preferential cleavage of bonds with



(thimet oligopeptidase)
hydrophobic residues at P1, P2 and



(P52888, P24155)
P3′ and a small residue at P1′ in




substrates of 5 to 15 residues






endopeptidase 24.16
Preferential cleavage in neurotensin:



(neurolysin) (Q9BYT8,
10-Pro-|-Tyr-11



Q91YP2)







amyloid precursor protein
Endopeptidase of broad specificity.



secretase alpha (P05067,




P12023, Q9Y5Z0,




P56817)







amyloid precursor protein
Broad endopeptidase specificity.



secretase beta (P05067,
Cleaves Glu-Val-Asn-Leu-|-Asp-



P12023, Q9Y5Z0,
Ala-Glu-Phe (EVNLDAEF; SEQ ID



P56817)
NO: 152) in the Swedish variant of




Alzheimer's amyloid precursor




protein






amyloid precursor protein
intramembrane cleavage of integral



secretase gamma (P05067,
membrane proteins



P12023, Q9Y5Z0,




P56817)







MMP 1 (P03956,
Cleavage of the triple helix of
SB-3CT


Q9EPL5uy)
collagen at about three-quarters of
p-OH SB-3CT



the length of the molecule from the
O-phosphate SB-3CT



N-terminus, at 775-Gly-|-Ile-776 in
RXP470.1



the alpha-1 (I) chain. Cleaves




synthetic substrates and alpha-




macroglobulins at bonds where P1′




is a hydrophobic residue.






MMP 2 (P08253, P33434)
Cleavage of gelatin type I and
SB-3CT



collagen types IV, V, VII, X.
p-OH SB-3CT



Cleaves the collagen-like sequence
O-phosphate SB-3CT



Pro-Gln-Gly-|-Ile-Ala-Gly-Gln
RXP470.1



(PQGIAGQ; SEQ ID NO: 153).






MMP 3 (P08254, P28862)
Preferential cleavage where P1′, P2′
SB-3CT



and P3′ are hydrophobic residues.
p-OH SB-3CT




O-phosphate SB-3CT




RXP470.1





MMP 7 (P09237,
Cleavage of 14-Ala-|-Leu-15 and 16-
SB-3CT


Q10738)
Tyr-|-Leu-17 in B chain of insulin.
p-OH SB-3CT



No action on collagen types I, II, IV,
O-phosphate SB-3CT



V. Cleaves gelatin chain alpha-2(I) >
RXP470.1



alpha-1(I).






MMP 8 (P22894,
Can degrade fibrillar type I, II, and
SB-3CT


O70138)
III collagens.
p-OH SB-3CT



Cleavage of interstitial collagens in
O-phosphate SB-3CT



the triple helical domain. Unlike EC
RXP470.1



3.4.24.7, this enzyme cleaves type




III collagen more slowly than type I.






MMP 9 (P14780, P41245)
Cleavage of gelatin types I and V
SB-3CT



and collagen types IV and V.
p-OH SB-3CT



Cleaves KiSS1 at a Gly-|-Leu bond.




Cleaves type IV and type V collagen
O-phosphate SB-3CT



into large C-terminal three quarter
RXP470.1



fragments and shorter N-terminal




one quarter fragments. Degrades




fibronectin but not laminin or Pz-




peptide.






MMP 10 (P09238,
Can degrade fibronectin, gelatins of
SB-3CT


055123)
type I, III, IV, and V; weakly
p-OH SB-3CT



collagens III, IV, and V.
O-phosphate SB-3CT




RXP470.1





MMP 11 (P24347,
A(A/Q)(N/A)↓(L/Y)(T/V/M/R)(R/K)
SB-3CT


Q02853)
G(G/A)EILR
p-OH SB-3CT



(SEQ ID NO: 191)
O-phosphate SB-3CT



↓ denotes the cleavage site
RXP470.1





MMP 12 (P39900,
Hydrolysis of soluble and insoluble
SB-3CT


P34960)
elastin. Specific cleavages are also
p-OH SB-3CT



produced at 14-Ala-|-Leu-15 and 16-
O-phosphate SB-3CT



Tyr-|-Leu-17 in the B chain of
RXP470.1



insulin




Has significant elastolytic activity.




Can accept large and small amino




acids at the P1′ site, but has a




preference for leucine. Aromatic or




hydrophobic residues are preferred




at the P1 site, with small




hydrophobic residues (preferably




alanine) occupying P3






MMP 13 (P45452,
Cleaves triple helical collagens,
SB-3CT


P33435)
including type I, type II and type III
p-OH SB-3CT



collagen, but has the highest activity
O-phosphate SB-3CT



with soluble type II collagen. Can
RXP470.1



also degrade collagen type IV, type




XIV and type X






MMP 14 (P50281,
Activates progelatinase A by
SB-3CT


P53690)
cleavage of the propeptide at 37-
p-OH SB-3CT



Asn-|-Leu-38. Other bonds
O-phosphate SB-3CT



hydrolyzed include 35-Gly-|-Ile-36
RXP470.1



in the propeptide of collagenase 3,




and 341-Asn-|-Phe-342, 441-Asp-|-




Leu-442 and 354-Gln-|-Thr-355 in




the aggrecan interglobular domain.






urokinase plasminogen
Specific cleavage of Arg-|-Val bond
Plasminogen activator


activator (uPA) (P00749,
in plasminogen to form plasmin.
inhibitors (PAI)


P06869)







tissue plasminogen
Specific cleavage of Arg-|-Val bond
Plasminogen activator


activator (tPA) (P00750,
in plasminogen to form plasmin.
inhibitors (PAI)


P11214)




tissue plasminogen
Specific cleavage of Arg-|-Val bond
Plasminogen activator


activator (tPA) (P00750,
in plasminogen to form plasmin.
inhibitors (PAI)


P11214)







Plasmin (P00747,
Preferential cleavage: Lys-|-Xaa >
α-2-antiplasmin (AP)


P20918)
Arg-|-Xaa, higher selectivity than




trypsin. Converts fibrin into soluble




products.






Thrombin (P00734,
Cleaves bonds after Arg and Lys



P19221)
Converts fibrinogen to fibrin and




activates factors V, VII, VIII, XIII,




and, in complex with




thrombomodulin, protein C.






BMP-1 (procollagen C-
Cleavage of the C-terminal



peptidase) (P13497,
propeptide at Ala-|-Asp in type I and



P98063)
II procollagens and at Arg-|-Asp in




type III.






ADAM (Q9POK1,

SB-3CT


Q9UKQ2, Q9JLN6,

p-OH SB-3CT


014672, Q13444,

O-phosphate SB-3CT


P78536, Q13443,

RXP470.1


O43184, P78325,




Q9UKF5, Q9BZ11,




Q9H2U9, Q99965,




O75077, Q9H013,




O43506)







granzyme A (P12544,
Preferential cleavage:- Arg-|-Xaa-,



P11032)
-Lys-|-Xaa- >> -Phe-|-Xaa- in small




molecule substrates.






granzyme B (P10144,
Preference for bulky and aromatic



P04187)
residues at the P1 position and acidic




residues at the P3′ and P4′ sites.






granzyme M (P51124,
Cleaves peptide substrates after



Q03238)
methionine, leucine, and norleucine.



tobacco Etch virus (TEV)
E-Xaa-Xaa-Y -Xaa-Q-(G/S), with






protease (P04517,
cleavage occurring between Q and



POCK09)
G/S. The most common sequence is




ENLYFQS (SEQ ID NO: 154)






chymotrypsin-like serine


-Thermobifida fusca



protease (P08217,

Thermopin


Q9UNI1, Q91X79,


-Pyrobaculum



P08861, P09093, P08218)


aerophilum Aeropin





-Thermococcus





kodakaraensis Tk-





serpin




-Alteromonas sp.




Marinostatin




-Streptomyces





misionensis SMTI





-Streptomyces sp.




chymostatin





alphavirus proteases




(P08411, P03317,




P13886, Q8JUX6,




Q86924, Q4QXJ8,




Q8QL53, P27282,




Q5XXP4)







chymotrypsin-like

-Thermobifidafusca


cysteine proteases

Thermopin


(Q86TL0, Q14790,

-Pyrobaculum


Q99538, 015553)


aerophilumAeropin





-Thermococcus





kodakaraensis Tk-





serpin




-Alteromonas sp.




Marinostatin




-Streptomyces





misionensis SMTI





-Streptomyces sp.




chymostatin





papain-like cysteine




proteases (P25774,




P53634, Q96K76)







picornavirus leader




proteases (P03305,




P03311, P13899)







HIV proteases (P04585,




P03367, P04584, P03369,




P12497, P03366, P04587)







Herpesvirus proteases




(P10220, Q2HRB6,




040922, Q69527)







adenovirus proteases




(P03252, P24937,




Q83906, P68985, P09569,




P11825, P10381)








Streptomyces griseus





protease A (SGPA)




(P00776)








Streptomyces griseus





protease B (SGPB)




(P00777)







alpha-lytic protease




(P85142, P00778)







serine proteases (P48740,




P98064, Q9UL52,




P05981, 060235)







cysteine proteases




(Q86TL0, Q14790,




Q8WYNO, Q96DT6,




P55211)







aspartic proteases




(Q9Y5Z0, P56817,




Q00663, Q53RT3,




POCY27)







threonine proteases




(Q9UI38, Q16512,




Q9H6P5, Q8IWU2)







Mast cell (MC) chymase
Abz-HPFHL(SEQ ID NO: 155)-
BAY 1142524


(CMA1) (NM_001836)
Lys(Dnp)-NH2 (SEQ ID NO: 194)
SUN13834





Rat mast cell protease-1, 
Abz-HPFHL(SEQ ID NO: 155)-
TY-51469


-2, -3, -4, -5 (NM_017145,
Lys(Dnp)-NH2 (SEQ ID NO: 194)



NM_172044,




NM_001170466,




NM_019321,




NM_013092)







Rat vascular chymase
Abz-HPFHL(SEQ ID NO: 155)-



(RVCH) (070500)
Lys(Dnp)-NH2 (SEQ ID NO: 194)






DENV NS3pro
A strong preference for basic amino
Anthraquinone


(NS2B/NS3)
acid residues (Arg/Lys) at the P1
BP13944


SEQ ID NOs: 157, 158,
positions was observed, whereas the
ZINC04321905


159, 160
preferences for the P2- 4 sites were
MB21



in the order of Arg > Thr >
Policresulen



Gln/Asn/Lys for P2, Lys > Arg >
SK-12



Asn for P3, and Nle > Leu > Lys >
NSC135618



Xaa for P4. The prime site substrate
Biliverdin



specificity was for small and polar




amino acids in P1 and P3.









A protease can be any of the following human proteases (MEROPS peptidase database number provided in parentheses; Rawlings N. D., Morton F. R., Kok, C. Y., Kong, J. & Barrett A. J. (2008) MEROPS: the peptidase database. Nucleic Acids Res. 36 Database issue, D320-325; herein incorporated by reference for all purposes): pepsin A (MER000885), gastricsin (MER000894), memapsin-2 (MER005870), renin (MER000917), cathepsin D (MER000911), cathepsin E (MER000944), memapsin-1 (MER005534), napsin A (MER004981), Mername-AA034 peptidase (MER014038), pepsin A4 (MER037290), pepsin A5 (Homo sapiens) (MER037291), hCG1733572 (Homo sapiens)-type putative peptidase (MER107386), napsin B pseudogene (MER004982), CYMP g.p. (Homo sapiens) (MER002929), subfamily A1A unassigned peptidases (MER181559), mouse mammary tumor virus retropepsin (MER048030), rabbit endogenous retrovirus endopeptidase (MER043650), S71-related human endogenous retropepsin (MER001812), RTVL-H-type putative peptidase (MER047117), RTVL-H-type putative peptidase (MER047133), RTVL-H-type putative peptidase (MER047160), RTVL-H-type putative peptidase (MER047206), RTVL-H-type putative peptidase (MER047253), RTVL-H-type putative peptidase (MER047260), RTVL-H-type putative peptidase (MER047291), RTVL-H-type putative peptidase (MER047418), RTVL-H-type putative peptidase (MER047440), RTVL-H-type putative peptidase (MER047479), RTVL-H-type putative peptidase (MER047559), RTVL-H-type putative peptidase (MER047583), RTVL-H-type putative peptidase (MER015446), human endogenous retrovirus retropepsin homologue 1 (MER015479), human endogenous retrovirus retropepsin homologue 2 (MER015481), endogenous retrovirus retropepsin pseudogene 1 (Homo sapiens chromosome 14) (MER029977), endogenous retrovirus retropepsin pseudogene 2 (Homo sapiens chromosome 8) (MER029665), endogenous retrovirus retropepsin pseudogene 3 (Homo sapiens chromosome 17) (MER002660), endogenous retrovirus retropepsin pseudogene 3 (Homo sapiens chromosome 17) (MER030286), endogenous retrovirus retropepsin pseudogene 3 (Homo sapiens chromosome 17) (MER047144), endogenous retrovirus retropepsin pseudogene 5 (Homo sapiens chromosome 12) (MER029664), endogenous retrovirus retropepsin pseudogene 6 (Homo sapiens chromosome 7) (MER002094), endogenous retrovirus retropepsin pseudogene 7 (Homo sapiens chromosome 6) (MER029776), endogenous retrovirus retropepsin pseudogene 8 (Homo sapiens chromosome Y) (MER030291), endogenous retrovirus retropepsin pseudogene 9 (Homo sapiens chromosome 19) (MER029680), endogenous retrovirus retropepsin pseudogene 10 (Homo sapiens chromosome 12) (MER002848), endogenous retrovirus retropepsin pseudogene 11 (Homo sapiens chromosome 17) (MER004378), endogenous retrovirus retropepsin pseudogene 12 (Homo sapiens chromosome 11) (MER003344), endogenous retrovirus retropepsin pseudogene 13 (Homo sapiens chromosome 2 and similar) (MER029779), endogenous retrovirus retropepsin pseudogene 14 (Homo sapiens chromosome 2) (MER029778), endogenous retrovirus retropepsin pseudogene 15 (Homo sapiens chromosome 4) (MER047158), endogenous retrovirus retropepsin pseudogene 15 (Homo sapiens chromosome 4) (MER047332), endogenous retrovirus retropepsin pseudogene 15 (Homo sapiens chromosome 4) (MER003182), endogenous retrovirus retropepsin pseudogene 16 (MER047165), endogenous retrovirus retropepsin pseudogene 16 (MER047178), endogenous retrovirus retropepsin pseudogene 16 (MER047200), endogenous retrovirus retropepsin pseudogene 16 (MER047315), endogenous retrovirus retropepsin pseudogene 16 (MER047405), endogenous retrovirus retropepsin pseudogene 16 (MER030292), endogenous retrovirus retropepsin pseudogene 17 (Homo sapiens chromosome 8) (MER005305), endogenous retrovirus retropepsin pseudogene 18 (Homo sapiens chromosome 4) (MER030288), endogenous retrovirus retropepsin pseudogene 19 (Homo sapiens chromosome 16) (MER001740), endogenous retrovirus retropepsin pseudogene 21 (Homo sapiens) (MER047222), endogenous retrovirus retropepsin pseudogene 21 (Homo sapiens) (MER047454), endogenous retrovirus retropepsin pseudogene 21 (Homo sapiens) (MER047477), endogenous retrovirus retropepsin pseudogene 21 (Homo sapiens) (MER004403), endogenous retrovirus retropepsin pseudogene 22 (Homo sapiens chromosome X) (MER030287), subfamily A2A non-peptidase homologues (MER047046), subfamily A2A non-peptidase homologues (MER047052), subfamily A2A non-peptidase homologues (MER047076), subfamily A2A non-peptidase homologues (MER047080), subfamily A2A non-peptidase homologues (MER047088), subfamily A2A non-peptidase homologues (MER047089), subfamily A2A non-peptidase homologues (MER047091), subfamily A2A non-peptidase homologues (MER047092), subfamily A2A non-peptidase homologues (MER047093), subfamily A2A non-peptidase homologues (MER047094), subfamily A2A non-peptidase homologues (MER047097), subfamily A2A non-peptidase homologues (MER047099), subfamily A2A non-peptidase homologues MER047101), subfamily A2A non-peptidase homologues (MER047102), subfamily A2A non-peptidase homologues (MER047107), subfamily A2A non-peptidase homologues (MER047108), subfamily A2A non-peptidase homologues (MER047109), subfamily A2A non-peptidase homologues (MER047110), subfamily A2A non-peptidase homologues MER047111), subfamily A2A non-peptidase homologues (MER047114), subfamily A2A non-peptidase homologues (MER047118), subfamily A2A non-peptidase homologues (MER047121), subfamily A2A non-peptidase homologues (MER047122), subfamily A2A non-peptidase homologues (MER047126), subfamily A2A non-peptidase homologues (MER047129), subfamily A2A non-peptidase homologues (MER047130), subfamily A2A non-peptidase homologues (MER047134), subfamily A2A non-peptidase homologues (MER047135), subfamily A2A non-peptidase homologues (MER047137), subfamily A2A non-peptidase homologues (MER047140), subfamily A2A non-peptidase homologues (MER047141), subfamily A2A non-peptidase homologues (MER047142), subfamily A2A non-peptidase homologues (MER047148), subfamily A2A non-peptidase homologues (MER047149), subfamily A2A non-peptidase homologues (MER047151), subfamily A2A non-peptidase homologues (MER047154), subfamily A2A non-peptidase homologues (MER047155), subfamily A2A non-peptidase homologues (MER047156), subfamily A2A non-peptidase homologues (MER047157), subfamily A2A non-peptidase homologues (MER047159), subfamily A2A non-peptidase homologues (MER047161), subfamily A2A non-peptidase homologues (MER047163), subfamily A2A non-peptidase homologues (MER047166), subfamily A2A non-peptidase homologues (MER047171), subfamily A2A non-peptidase homologues (MER047173), subfamily A2A non-peptidase homologues (MER047174), subfamily A2A non-peptidase homologues (MER047179), subfamily A2A non-peptidase homologues (MER047183), subfamily A2A non-peptidase homologues (MER047186), subfamily A2A non-peptidase homologues (MER047190), subfamily A2A non-peptidase homologues (MER047191), subfamily A2A non-peptidase homologues (MER047196), subfamily A2A non-peptidase homologues (MER047198), subfamily A2A non-peptidase homologues (MER047199), subfamily A2A non-peptidase homologues (MER047201), subfamily A2A non-peptidase homologues (MER047202), subfamily A2A non-peptidase homologues (MER047203), subfamily A2A non-peptidase homologues (MER047204), subfamily A2A non-peptidase homologues (MER047205), subfamily A2A non-peptidase homologues (MER047207), subfamily A2A non-peptidase homologues (MER047208), subfamily A2A non-peptidase homologues (MER047210), subfamily A2A non-peptidase homologues (MER047211), subfamily A2A non-peptidase homologues (MER047212), subfamily A2A non-peptidase homologues (MER047213), subfamily A2A non-peptidase homologues (MER047215), subfamily A2A non-peptidase homologues (MER047216), subfamily A2A non-peptidase homologues (MER047218), subfamily A2A non-peptidase homologues (MER047219), subfamily A2A non-peptidase homologues (MER047221), subfamily A2A non-peptidase homologues (MER047224), subfamily A2A non-peptidase homologues (MER047225), subfamily A2A non-peptidase homologues (MER047226), subfamily A2A non-peptidase homologues (MER047227), subfamily A2A non-peptidase homologues (MER047230), subfamily A2A non-peptidase homologues (MER047232), subfamily A2A non-peptidase homologues (MER047233), subfamily A2A non-peptidase homologues (MER047234), subfamily A2A non-peptidase homologues (MER047236), subfamily A2A non-peptidase homologues (MER047238), subfamily A2A non-peptidase homologues (MER047239), subfamily A2A non-peptidase homologues (MER047240), subfamily A2A non-peptidase homologues (MER047242), subfamily A2A non-peptidase homologues (MER047243), subfamily A2A non-peptidase homologues (MER047249), subfamily A2A non-peptidase homologues (MER047251), subfamily A2A non-peptidase homologues (MER047252), subfamily A2A non-peptidase homologues (MER047254), subfamily A2A non-peptidase homologues (MER047255), subfamily A2A non-peptidase homologues (MER047263), subfamily A2A non-peptidase homologues (MER047265), subfamily A2A non-peptidase homologues (MER047266), subfamily A2A non-peptidase homologues (MER047267), subfamily A2A non-peptidase homologues (MER047268), subfamily A2A non-peptidase homologues (MER047269), subfamily A2A non-peptidase homologues (MER047272), subfamily A2A non-peptidase homologues (MER047273), subfamily A2A non-peptidase homologues (MER047274), subfamily A2A non-peptidase homologues (MER047275), subfamily A2A non-peptidase homologues (MER047276), subfamily A2A non-peptidase homologues (MER047279), subfamily A2A non-peptidase homologues (MER047280), subfamily A2A non-peptidase homologues (MER047281), subfamily A2A non-peptidase homologues (MER047282), subfamily A2A non-peptidase homologues (MER047284), subfamily A2A non-peptidase homologues (MER047285), subfamily A2A non-peptidase homologues (MER047289), subfamily A2A non-peptidase homologues (MER047290), subfamily A2A non-peptidase homologues (MER047294), subfamily A2A non-peptidase homologues (MER047295), subfamily A2A non-peptidase homologues (MER047298), subfamily A2A non-peptidase homologues (MER047300), subfamily A2A non-peptidase homologues (MER047302), subfamily A2A non-peptidase homologues (MER047304), subfamily A2A non-peptidase homologues (MER047305), subfamily A2A non-peptidase homologues (MER047306), subfamily A2A non-peptidase homologues (MER047307), subfamily A2A non-peptidase homologues (MER047310), subfamily A2A non-peptidase homologues (MER047311), subfamily A2A non-peptidase homologues (MER047314), subfamily A2A non-peptidase homologues (MER047318), subfamily A2A non-peptidase homologues (MER047320), subfamily A2A non-peptidase homologues (MER047321), subfamily A2A non-peptidase homologues (MER047322), subfamily A2A non-peptidase homologues (MER047326), subfamily A2A non-peptidase homologues (MER047327), subfamily A2A non-peptidase homologues (MER047330), subfamily A2A non-peptidase homologues (MER047333), subfamily A2A non-peptidase homologues (MER047362), subfamily A2A non-peptidase homologues (MER047366), subfamily A2A non-peptidase homologues (MER047369), subfamily A2A non-peptidase homologues (MER047370), subfamily A2A non-peptidase homologues (MER047371), subfamily A2A non-peptidase homologues (MER047375), subfamily A2A non-peptidase homologues (MER047376), subfamily A2A non-peptidase homologues (MER047381), subfamily A2A non-peptidase homologues (MER047383), subfamily A2A non-peptidase homologues (MER047384), subfamily A2A non-peptidase homologues (MER047385), subfamily A2A non-peptidase homologues (MER047388), subfamily A2A non-peptidase homologues (MER047389), subfamily A2A non-peptidase homologues (MER047391), subfamily A2A non-peptidase homologues (MER047394), subfamily A2A non-peptidase homologues (MER047396), subfamily A2A non-peptidase homologues (MER047400), subfamily A2A non-peptidase homologues (MER047401), subfamily A2A non-peptidase homologues (MER047403), subfamily A2A non-peptidase homologues (MER047406), subfamily A2A non-peptidase homologues (MER047407), subfamily A2A non-peptidase homologues (MER047410), subfamily A2A non-peptidase homologues (MER047411), subfamily A2A non-peptidase homologues (MER047413), subfamily A2A non-peptidase homologues (MER047414), subfamily A2A non-peptidase homologues (MER047416), subfamily A2A non-peptidase homologues (MER047417), subfamily A2A non-peptidase homologues (MER047420), subfamily A2A non-peptidase homologues (MER047423), subfamily A2A non-peptidase homologues (MER047424), subfamily A2A non-peptidase homologues (MER047428), subfamily A2A non-peptidase homologues (MER047429), subfamily A2A non-peptidase homologues (MER047431), subfamily A2A non-peptidase homologues (MER047434), subfamily A2A non-peptidase homologues (MER047439), subfamily A2A non-peptidase homologues (MER047442), subfamily A2A non-peptidase homologues (MER047445), subfamily A2A non-peptidase homologues (MER047449), subfamily A2A non-peptidase homologues (MER047450), subfamily A2A non-peptidase homologues (MER047452), subfamily A2A non-peptidase homologues (MER047455), subfamily A2A non-peptidase homologues (MER047457), subfamily A2A non-peptidase homologues (MER047458), subfamily A2A non-peptidase homologues (MER047459), subfamily A2A non-peptidase homologues (MER047463), subfamily A2A non-peptidase homologues (MER047468), subfamily A2A non-peptidase homologues (MER047469), subfamily A2A non-peptidase homologues (MER047470), subfamily A2A non-peptidase homologues (MER047476), subfamily A2A non-peptidase homologues (MER047478), subfamily A2A non-peptidase homologues (MER047483), subfamily A2A non-peptidase homologues (MER047488), subfamily A2A non-peptidase homologues (MER047489), subfamily A2A non-peptidase homologues (MER047490), subfamily A2A non-peptidase homologues (MER047493), subfamily A2A non-peptidase homologues (MER047494), subfamily A2A non-peptidase homologues (MER047495), subfamily A2A non-peptidase homologues (MER047496), subfamily A2A non-peptidase homologues (MER047497), subfamily A2A non-peptidase homologues (MER047499), subfamily A2A non-peptidase homologues (MER047502), subfamily A2A non-peptidase homologues (MER047504), subfamily A2A non-peptidase homologues (MER047511), subfamily A2A non-peptidase homologues (MER047513), subfamily A2A non-peptidase homologues (MER047514), subfamily A2A non-peptidase homologues (MER047515), subfamily A2A non-peptidase homologues (MER047516), subfamily A2A non-peptidase homologues (MER047520), subfamily A2A non-peptidase homologues (MER047533), subfamily A2A non-peptidase homologues (MER047537), subfamily A2A non-peptidase homologues (MER047569), subfamily A2A non-peptidase homologues (MER047570), subfamily A2A non-peptidase homologues (MER047584), subfamily A2A non-peptidase homologues (MER047603), subfamily A2A non-peptidase homologues (MER047604), subfamily A2A non-peptidase homologues (MER047606), subfamily A2A non-peptidase homologues (MER047609), subfamily A2A non-peptidase homologues (MER047616), subfamily A2A non-peptidase homologues (MER047619), subfamily A2A non-peptidase homologues (MER047648), subfamily A2A non-peptidase homologues (MER047649), subfamily A2A non-peptidase homologues (MER047662), subfamily A2A non-peptidase homologues (MER048004), subfamily A2A non-peptidase homologues (MER048018), subfamily A2A non-peptidase homologues (MER048019), subfamily A2A non-peptidase homologues (MER048023), subfamily A2A non-peptidase homologues (MER048037), subfamily A2A unassigned peptidases (MER047164), subfamily A2A unassigned peptidases (MER047231), subfamily A2A unassigned peptidases (MER047386), skin aspartic protease (MER057097), presenilin 1 (MER005221), presenilin 2 (MER005223), impas 1 peptidase (MER019701), impas 1 peptidase (MER184722), impas 4 peptidase (MER019715), impas 2 peptidase (MER019708), impas 5 peptidase (MER019712), impas 3 peptidase (MER019711), possible family A22 pseudogene (Homo sapiens chromosome 18) (MER029974), possible family A22 pseudogene (Homo sapiens chromosome 11) (MER023159), cathepsin V (MER004437), cathepsin X (MER004508), cathepsin F (MER004980), cathepsin L (MER000622), cathepsin S (MER000633), cathepsin O (MER001690), cathepsin K (MER000644), cathepsin W (MER003756), cathepsin H (MER000629), cathepsin B (MER000686), dipeptidyl-peptidase I (MER001937), bleomycin hydrolase (animal) (MER002481), tubulointerstitial nephritis antigen (MER016137), tubulointerstitial nephritis antigen-related protein (MER021799), cathepsin L-like pseudogene 1 (Homo sapiens) (MER002789), cathepsin B-like pseudogene (chromosome 4, Homo sapiens) (MER029469), cathepsin B-like pseudogene (chromosome 1, Homo sapiens) (MER029457), CTSLL2 g.p. (Homo sapiens) (MER005210), CTSLL3 g.p. (Homo sapiens) (MER005209), calpain-1 (MER000770), calpain-2 (MER000964), calpain-3 (MER001446), calpain-9 (MER004042), calpain-8 (MER021474), calpain-15 (MER004745), calpain-5 (MER002939), calpain-11 (MER005844), calpain-12 (MER029889), calpain-10 (MER013510), calpain-13 (MER020139), calpain-14 (MER029744), Mername-AA253 peptidase (MER005537), calpamodulin (MER000718), hypothetical protein 940251 (MER003201), ubiquitinyl hydrolase-L1 (MER000832), ubiquitinyl hydrolase-L3 (MER000836), ubiquitinyl hydrolase-BAP1 (MER003989), ubiquitinyl hydrolase-UCH37 (MER005539), ubiquitin-specific peptidase 5 (MER002066), ubiquitin-specific peptidase 6 (MER000863), ubiquitin-specific peptidase 4 (MER001795), ubiquitin-specific peptidase 8 (MER001884), ubiquitin-specific peptidase 13 (MER002627), ubiquitin-specific peptidase 2 (MER004834), ubiquitin-specific peptidase 11 (MER002693), ubiquitin-specific peptidase 14 (MER002667), ubiquitin-specific peptidase 7 (MER002896), ubiquitin-specific peptidase 9X (MER005877), ubiquitin-specific peptidase 10 (MER004439), ubiquitin-specific peptidase 1 (MER004978), ubiquitin-specific peptidase 12 (MER005454), ubiquitin-specific peptidase 16 (MER005493), ubiquitin-specific peptidase 15 (MER005427), ubiquitin-specific peptidase 17 (MER002900), ubiquitin-specific peptidase 19 (MER005428), ubiquitin-specific peptidase 20 (MER005494), ubiquitin-specific peptidase 3 (MER005513), ubiquitin-specific peptidase 9Y (MER004314), ubiquitin-specific peptidase 18 (MER005641), ubiquitin-specific peptidase 21 (MER006258), ubiquitin-specific peptidase 22 (MER012130), ubiquitin-specific peptidase 33 (MER014335), ubiquitin-specific peptidase 29 (MER012093), ubiquitin-specific peptidase 25 (MER011115), ubiquitin-specific peptidase 36 (MER014033), ubiquitin-specific peptidase 32 (MER014290), ubiquitin-specific peptidase 26 (Homo sapiens-type) (MER014292), ubiquitin-specific peptidase 24 (MER005706), ubiquitin-specific peptidase 42 (MER011852), ubiquitin-specific peptidase 46 (MER014629), ubiquitin-specific peptidase 37 (MER014633), ubiquitin-specific peptidase 28 (MER014634), ubiquitin-specific peptidase 47 (MER014636), ubiquitin-specific peptidase 38 (MER014637), ubiquitin-specific peptidase 44 (MER014638), ubiquitin-specific peptidase 50 (MER030315), ubiquitin-specific peptidase 35 (MER014646), ubiquitin-specific peptidase 30 (MER014649), Mername-AA091 peptidase (MER014743), ubiquitin-specific peptidase 45 (MER030314), ubiquitin-specific peptidase 51 (MER014769), ubiquitin-specific peptidase 34 (MER014780), ubiquitin-specific peptidase 48 (MER064620), ubiquitin-specific peptidase 40 (MER015483), ubiquitin-specific peptidase 41 (MER045268), ubiquitin-specific peptidase 31 (MER015493), Mername-AA129 peptidase (MER016485), ubiquitin-specific peptidase 49 (MER016486), Mername-AA187 peptidase (MER052579), USP17-like peptidase (MER030192), ubiquitin-specific peptidase 54 (MER028714), ubiquitin-specific peptidase 53 (MER027329), ubiquitin-specific endopeptidase 39 [misleading] (MER064621), Mername-AA090 non-peptidase homologue (MER014739), ubiquitin-specific peptidase 43 [misleading] (MER030140), ubiquitin-specific peptidase 52 [misleading] (MER030317), NEK2 pseudogene (MER014736), C19 pseudogene (Homo sapiens: chromosome 5) (MER029972), Mername-AA088 peptidase (MER014750), autophagin-2 (MER013564), autophagin-1 (MER013561), autophagin-3 (MER014316), autophagin-4 (MER064622), Cezanne deubiquitinylating peptidase (MER029042), Cezanne-2 peptidase (MER029044), tumor necrosis factor alpha-induced protein 3 (MER029050), trabid peptidase (MER029052), VCIP135 deubiquitinating peptidase (MER152304), otubain-1 (MER029056), otubain-2 (MER029061), CylD protein (MER030104), UfSP1 peptidase (MER042724), UfSP2 peptidase (MER060306), DUBA deubiquitinylating enzyme (MER086098), KIAA0459 (Homo sapiens)-like protein (MER122467), Otud1 protein (MER125457), glycosyltransferase 28 domain containing 1, isoform CRA_c (Homo sapiens)-like (MER123606), hin1L g.p. (Homo sapiens) (MER139816), ataxin-3 (MER099998), ATXN3L putative peptidase (MER115261), Josephin domain containing 1 (Homo sapiens) (MER125334), Josephin domain containing 2 (Homo sapiens) (MER124068), YOD1 peptidase (MER116559), legumain (plant alpha form) (MER044591), legumain (MER001800), glycosylphosphatidylinositol:protein transamidase (MER002479), legumain pseudogene (Homo sapiens) (MER029741), family C13 unassigned peptidases (MER175813), caspase-1 (MER000850), caspase-3 (MER000853), caspase-7 (MER002705), caspase-6 (MER002708), caspase-2 (MER001644), caspase-4 (MER001938), caspase-5 (MER002240), caspase-8 (MER002849), caspase-9 (MER002707), caspase-10 (MER002579), caspase-14 (MER012083), paracaspase (MER019325), Mername-AA143 peptidase (MER021304), Mername-AA186 peptidase (MER020516), putative caspase (Homo sapiens) (MER021463), FLIP protein (MER003026), Mername-AA142 protein (MER021316), caspase-12 pseudogene (Homo sapiens) (MER019698), Mername-AA093 caspase pseudogene (MER014766), subfamily C14A non-peptidase homologues (MER185329), subfamily C14A non-peptidase homologues (MER179956), separase (Homo sapiens-type) (MER011775), separase-like pseudogene (MER014797), SENP1 peptidase (MER011012), SENP3 peptidase (MER011019), SENP6 peptidase (MER011109), SENP2 peptidase (MER012183), SENP5 peptidase (MER014032), SENP7 peptidase (MER014095), SENP8 peptidase (MER016161), SENP4 peptidase (MER005557), pyroglutamyl-peptidase I (chordate) (MER011032), Mername-AA073 peptidase (MER029978), Sonic hedgehog protein (MER002539), Indian hedgehog protein (MER002538), Desert hedgehog protein (MER012170), dipeptidyl-peptidase III (MER004252), Mername-AA164 protein (MER020410), LOC138971 g.p. (Homo sapiens) (MER020074), Atp23 peptidase (MER060642), prenyl peptidase 1 (MER004246), aminopeptidase N (MER000997), aminopeptidase A (MER001012), leukotriene A4 hydrolase (MER001013), pyroglutamyl-peptidase II (MER012221), cytosol alanyl aminopeptidase (MER002746), cystinyl aminopeptidase (MER002060), aminopeptidase B (MER001494), aminopeptidase PILS (MER005331), arginyl aminopeptidase-like 1 (MER012271), leukocyte-derived arginine aminopeptidase (MER002968), aminopeptidase Q (MER052595), aminopeptidase O (MER019730), Tata binding protein associated factor (MER026493), angiotensin-converting enzyme peptidase unit 1 (MER004967), angiotensin-converting enzyme peptidase unit 2 (MER001019), angiotensin-converting enzyme-2 (MER011061), Mername-AA153 protein (MER020514), thimet oligopeptidase (MER001737), neurolysin (MER010991), mitochondrial intermediate peptidase (MER003665), Mername-AA154 protein (MER021317), leishmanolysin-2 (MER014492), leishmanolysin-3 (MER180031), matrix metallopeptidase-1 (MER001063), matrix metallopeptidase-8 (MER001084), matrix metallopeptidase-2 (MER001080), matrix metallopeptidase-9 (MER001085), matrix metallopeptidase-3 (MER001068), matrix metallopeptidase-10 (Homo sapiens-type) (MER001072), matrix metallopeptidase-11 (MER001075), matrix metallopeptidase-7 (MER001092), matrix metallopeptidase-12 (MER001089), matrix metallopeptidase-13 (MER001411), membrane-type matrix metallopeptidase-1 (MER001077), membrane-type matrix metallopeptidase-2 (MER002383), membrane-type matrix metallopeptidase-3 (MER002384), membrane-type matrix metallopeptidase-4 (MER002595), matrix metallopeptidase-20 (MER003021), matrix metallopeptidase-19 (MER002076), matrix metallopeptidase-23B (MER004766), membrane-type matrix metallopeptidase-5 (MER005638), membrane-type matrix metallopeptidase-6 (MER012071), matrix metallopeptidase-21 (MER006101), matrix metallopeptidase-22 (MER014098), matrix metallopeptidase-26 (MER012072), matrix metallopeptidase-28 (MER013587), matrix metallopeptidase-23A (MER037217), macrophage elastase homologue (chromosome 8, Homo sapiens) (MER030035), Mername-AA156 protein (MER021309), matrix metallopeptidase-like 1 (MER045280), subfamily M10A non-peptidase homologues (MER175912), subfamily M10A non-peptidase homologues (MER187997), subfamily M10A non-peptidase homologues (MER187998), subfamily M10A non-peptidase homologues (MER180000), meprin alpha subunit (MER001111), meprin beta subunit (MER005213), procollagen C-peptidase (MER001113), mammalian tolloid-like 1 protein (MER005124), mammalian-type tolloid-like 2 protein (MER005866), ADAMTS9 peptidase (MER012092), ADAMTS14 peptidase (MER016700), ADAMTS15 peptidase (MER017029), ADAMTS16 peptidase (MER015689), ADAMTS17 peptidase (MER016302), ADAMTS18 peptidase (MER016090), ADAMTS19 peptidase (MER015663), ADAM8 peptidase (MER003902), ADAM9 peptidase (MER001140), ADAM10 peptidase (MER002382), ADAM12 peptidase (MER005107), ADAM19 peptidase (MER012241), ADAM15 peptidase (MER002386), ADAM17 peptidase (MER003094), ADAM20 peptidase (MER004725), ADAMDEC1 peptidase (MER000743), ADAMTS3 peptidase (MER005100), ADAMTS4 peptidase (MER005101), ADAMTS1 peptidase (MER005546), ADAM28 peptidase (Homo sapiens-type) (MER005495), ADAMTS5 peptidase (MER005548), ADAMTS8 peptidase (MER005545), ADAMTS6 peptidase (MER005893), ADAMTS7 peptidase (MER005894), ADAM30 peptidase (MER006268), ADAM21 peptidase (Homo sapiens-type) (MER004726), ADAMTS10 peptidase (MER014331), ADAMTS12 peptidase (MER014337), ADAMTS13 peptidase (MER015450), ADAM33 peptidase (MER015143), ovastacin (MER029996), ADAMTS20 peptidase (Homo sapiens-type) (MER026906), procollagen I N-peptidase (MER004985), ADAM2 protein (MER003090), ADAM6 protein (MER047044), ADAM7 protein (MER005109), ADAM18 protein (MER012230), ADAM32 protein (MER026938), non-peptidase homologue (Homo sapiens chromosome 4) (MER029973), family M12 non-peptidase homologue (Homo sapiens chromosome 16) (MER047654), family M12 non-peptidase homologue (Homo sapiens chromosome 15) (MER047250), ADAM3B protein (Homo sapiens-type) (MER005199), ADAM11 protein (MER001146), ADAM22 protein (MER005102), ADAM23 protein (MER005103), ADAM29 protein (MER006267), protein similar to ADAM21 peptidase preproprotein (Homo sapiens) (MER026944), Mername-AA225 peptidase homologue (Homo sapiens) (MER047474), putative ADAM pseudogene (chromosome 4, Homo sapiens) (MER029975), ADAM3A g.p. (Homo sapiens) (MER005200), ADAM1 g.p. (Homo sapiens) (MER003912), subfamily M12B non-peptidase homologues (MER188210), subfamily M12B non-peptidase homologues (MER188211), subfamily M12B non-peptidase homologues (MER188212), subfamily M12B non-peptidase homologues (MER188220), neprilysin (MER001050), endothelin-converting enzyme 1 (MER001057), endothelin-converting enzyme 2 (MER004776), DINE peptidase (MER005197), neprilysin-2 (MER013406), Kell blood-group protein (MER001054), PHEX peptidase (MER002062), i-AAA peptidase (MER001246), i-AAA peptidase (MER005755), paraplegin (MER004454), Afg3-like protein 2 (MER005496), Afg3-like protein 1A (MER014306), pappalysin-1 (MER002217), pappalysin-2 (MER014521), farnesylated-protein converting enzyme 1 (MER002646), metalloprotease-related protein-1 (MER030873), aminopeptidase AMZ2 (MER011907), aminopeptidase AMZ1 (MER058242), carboxypeptidase A1 (MER001190), carboxypeptidase A2 (MER001608), carboxypeptidase B (MER001194), carboxypeptidase N (MER001198), carboxypeptidase E (MER001199), carboxypeptidase M (MER001205), carboxypeptidase U (MER001193), carboxypeptidase A3 (MER001187), metallocarboxypeptidase D peptidase unit 1 (MER003781), metallocarboxypeptidase Z (MER003428), metallocarboxypeptidase D peptidase unit 2 (MER004963), carboxypeptidase A4 (MER013421), carboxypeptidase A6 (MER013456), carboxypeptidase A5 (MER017121), metallocarboxypeptidase O (MER016044), cytosolic carboxypeptidase-like protein 5 (MER033174), cytosolic carboxypeptidase 3 (MER033176), cytosolic carboxypeptidase 6 (MER033178), cytosolic carboxypeptidase 1 (MER033179), cytosolic carboxypeptidase 2 (MER037713), metallocarboxypeptidase D non-peptidase unit (MER004964), adipocyte-enhancer binding protein 1 (MER003889), carboxypeptidase-like protein X1 (MER013404), carboxypeptidase-like protein X2 (MER078764), cytosolic carboxypeptidase (MER026952), family M14 non-peptidase homologues (MER199530), insulysin (MER001214), mitochondrial processing peptidase beta-subunit (MER004497), nardilysin (MER003883), eupitrilysin (MER004877), mitochondrial processing peptidase non-peptidase alpha subunit (MER001413), ubiquinol-cytochrome c reductase core protein I (MER003543), ubiquinol-cytochrome c reductase core protein II (MER003544), ubiquinol-cytochrome c reductase core protein domain 2 (MER043998), insulysin unit 2 (MER046821), nardilysin unit 2 (MER046874), insulysin unit 3 (MER078753), mitochondrial processing peptidase subunit alpha unit 2 (MER124489), nardilysin unit 3 (MER142856), LOC133083 g.p. (Homo sapiens) (MER021876), subfamily M16B non-peptidase homologues (MER188757), leucyl aminopeptidase (animal) (MER003100), Mername-AA040 peptidase (MER003919), leucyl aminopeptidase-1 (Caenorhabditis-type) (MER013416), methionyl aminopeptidase 1 (MER001342), methionyl aminopeptidase 2 (MER001728), aminopeptidase P2 (MER004498), Xaa-Pro dipeptidase (eukaryote) (MER001248), aminopeptidase P1 (MER004321), mitochondrial intermediate cleaving peptidase 55 kDa (MER013463), mitochondrial methionyl aminopeptidase (MER014055), Mername-AA020 peptidase homologue (MER010972), proliferation-association protein 1 (MER005497), chromatin-specific transcription elongation factor 140 kDa subunit (MER026495), proliferation-associated protein 1-like (Homo sapiens chromosome X) (MER029983), Mername-AA226 peptidase homologue (Homo sapiens) (MER056262), Mername-AA227 peptidase homologue (Homo sapiens) (MER047299), subfamily M24A non-peptidase homologues (MER179893), aspartyl aminopeptidase (MER003373), Gly-Xaa carboxypeptidase (MER033182), carnosine dipeptidase II (MER014551), carnosine dipeptidase I (MER015142), Mername-AA161 protein (MER021873), aminoacylase (MER001271), glutamate carboxypeptidase II (MER002104), NAALADASE L peptidase (MER005239), glutamate carboxypeptidase III (MER005238), plasma glutamate carboxypeptidase (MER005244), Mername-AA103 peptidase (MER015091), Fxna peptidase (MER029965), transferrin receptor protein (MER002105), transferrin receptor 2 protein (MER005152), glutaminyl cyclise (MER015095), glutamate carboxypeptidase II (Homo sapiens)-type non-peptidase homologue (MER026971), nicalin (MER044627), membrane dipeptidase (MER001260), membrane-bound dipeptidase-2 (MER013499), membrane-bound dipeptidase-3 (MER013496), dihydro-orotase (MER005767), dihydropyrimidinase (MER033266), dihydropyrimidinase related protein-1 (MER030143), dihydropyrimidinase related protein-2 (MER030155), dihydropyrimidinase related protein-3 (MER030151), dihydropyrimidinase related protein-4 (MER030149), dihydropyrimidinase related protein-5 (MER030136), hypothetical protein like 5730457F1 IRIK (MER033184), 1300019j08rik protein (MER033186)), guanine aminohydrolase (MER037714), Kael putative peptidase (MER001577), OSGEPLI-like protein (MER013498), S2P peptidase (MER004458), subfamily M23B non-peptidase homologues (MER199845), subfamily M23B non-peptidase homologues (MER199846), subfamily M23B non-peptidase homologues (MER199847), subfamily M23B non-peptidase homologues (MER137320), subfamily M23B non-peptidase homologues (MER201557), subfamily M23B non-peptidase homologues (MER199417), subfamily M23B non-peptidase homologues (MER199418), subfamily M23B non-peptidase homologues (MER199419), subfamily M23B non-peptidase homologues (MER199420), subfamily M23B non-peptidase homologues (MER175932), subfamily M23B non-peptidase homologues (MER199665), Pohl peptidase (MER020382), Jabl/MPN domain metalloenzyme (MER022057), Mername-AA165 peptidase (MER021865), Brcc36 isopeptidase (MER021890), histone H2A deubiquitinase MYSM1 (MER021887), AMSH deubiquitinating peptidase (MER030146), putative peptidase (Homo sapiens chromosome 2) (MER029970), Mername-AA168 protein (MER021886), COP9 signalosome subunit 6 (MER030137), 26S proteasome non-ATPase regulatory subunit 7 (MER030134), eukaryotic translation initiation factor 3 subunit 5 (MER030133), IFP38 peptidase homologue (MER030132), subfamily M67A non-peptidase homologues (MER191181), subfamily M67A unassigned peptidases (MER191144), granzyme B (Homo sapiens-type) (MER000168), testisin (MER005212), tryptase beta (MER000136), kallikrein-related peptidase 5 (MER005544), corin (MER005881), kallikrein-related peptidase 12 (MER006038), DESC1 peptidase (MER006298), tryptase gamma 1 (MER011036), kallikrein-related peptidase 14 (MER011038), hyaluronan-binding peptidase (MER003612), transmembrane peptidase, serine 4 (MER011104), intestinal serine peptidase (rodent) (MER016130), adrenal secretory serine peptidase (MER003734), tryptase delta 1 (Homo sapiens) (MER005948), matriptase-3 (MER029902), marapsin (MER006119), tryptase-6 (MER006118), ovochymase-1 domain 1 (MER099182), transmembrane peptidase, serine 3 (MER005926), kallikrein-related peptidase 15 (MER000064), Mername-AA031 peptidase (MER014054), TMPRSS13 peptidase (MER014226), Mername-AA038 peptidase (MER062848), Mername-AA204 peptidase (MER029980), cationic trypsin (Homo sapiens-type) (MER000020), elastase-2 (MER000118), mannan-binding lectin-associated serine peptidase-3 (MER031968), cathepsin G (MER000082), myeloblastin (MER000170), granzyme A (MER001379), granzyme M (MER001541), chymase (Homo sapiens-type) (MER000123), tryptase alpha (MER000135), granzyme K (MER001936), granzyme H (MER000166), chymotrypsin B (MER000001), elastase-1 (MER003733), pancreatic endopeptidase E (MER000149), pancreatic elastase II (MER000146), enteropeptidase (MER002068), chymotrypsin C (MER000761), prostasin (MER002460), kallikrein 1 (MER000093), kallikrein-related peptidase 2 (MER000094), kallikrein-related peptidase 3 (MER000115), mesotrypsin (MER000022), complement component Clr-like peptidase (MER016352), complement factor D (MER000130), complement component activated Clr (MER000238), complement component activated CIs (MER000239), complement component C2a (MER000231), complement factor B (MER000229), mannan-binding lectin-associated serine peptidase 1 (MER000244), complement factor I (MER000228), pancreatic endopeptidase E form B (MER000150), pancreatic elastase IIB (MER000147), coagulation factor XIIa (MER000187), plasma kallikrein (MER000203) coagulation factor Xia (MER000210), coagulation factor IXa (MER000216), coagulation factor Vila (MER000215), coagulation factor Xa (MER000212), thrombin (MER000188), protein C (activated) (MER000222), acrosin (MER000078), hepsin (MER000156), hepatocyte growth factor activator (MER000186), mannan-binding lectin-associated serine peptidase 2 (MER002758), u-plasminogen activator (MER000195), t-plasminogen activator (MER000192), plasmin (MER000175), kallikrein-related peptidase 6 (MER002580), neurotrypsin (MER004171), kallikrein-related peptidase 8 (MER005400), kallikrein-related peptidase 10 (MER003645), epitheliasin (MER003736), kallikrein-related peptidase 4 (MER005266), prosemin (MER004214), chymopasin (MER001503), kallikrein-related peptidase 11 (MER004861), kallikrein-related peptidase 11 (MER216142), trypsin-2 type A (MER000021), HtrA1 peptidase (Homo sapiens-type) (MER002577), HtrA2 peptidase (MER208413), HtrA2 peptidase (MER004093), HtrA3 peptidase (MER014795), HtrA4 peptidase (MER016351), Tysndl peptidase (MER050461), TMPRSS12 peptidase (MER017085), HAT-like putative peptidase 2 (MER021884), trypsin C (MER021898), kallikrein-related peptidase 7 (MER002001), matriptase (MER003735), kallikrein-related peptidase 13 (MER005269), kallikrein-related peptidase 9 (MER005270), matriptase-2 (MER005278), umbelical vein peptidase (MER005421), LCLP peptidase (MER001900), spinesin (MER014385), marapsin-2 (MER021929), complement factor D-like putative peptidase (MER056164), ovochymase-2 (MER022410), HAT-like 4 peptidase (MER044589), ovochymase 1 domain 1 (MER022412), epidermis-specific SP-like putative peptidase (MER029900), testis serine peptidase 5 (MER029901), Mername-AA258 peptidase (MER000285), polyserase-IA unit 1 (MER030879), polyserase-IA unit 2 (MER030880), testis serine peptidase 2 (human-type) (MER033187), hypothetical acrosin-like peptidase (Homo sapiens) (MER033253), HAT-like 5 peptidase (MER028215), polyserase-3 unit 1 (MER061763), polyserase-3 unit 2 (MER061748), peptidase similar to tryptophan/serine protease (MER056263), polyserase-2 unit 1 (MER061777), Mername-AA123 peptidase (MER021930), HAT-like 2 peptidase (MER099184), hCG2041452-like protein (MER099172), hCG22067 (Homo sapiens) (MER099169), brain-rescue-factor-1 (Homo sapiens) (MER098873), hCG2041108 (Homo sapiens) (MER099173), polyserase-2 unit 2 (MER061760), polyserase-2 unit 3 (MER065694), Mername-AA201 (peptidase homologue) MER099175, secreted trypsin-like serine peptidase homologue (MER030000), polyserase-1A unit 3 (MER029880), azurocidin (MER000119), haptoglobin-1 (MER000233), haptoglobin-related protein (MER000235), macrophage-stimulating protein (MER001546), hepatocyte growth factor (MER000185), protein Z (MER000227), TESP1 protein (MER047214), LOC136242 protein (MER016132), plasma kallikrein-like protein 4 (MER016346), PRSS35 protein (MER016350), DKFZp586H2123-like protein (MER066474), apolipoprotein (MER000183), psi-KLK1 pseudogene (Homo sapiens) (MER033287), tryptase pseudogene I (MER015077), tryptase pseudogene II (MER015078), tryptase pseudogene III (MER015079), subfamily SlA unassigned peptidases (MER216982), subfamily SlA unassigned peptidases (MER216148), amidophosphoribosyltransferase precursor (MER003314), glutamine-fructose-6-phosphate transaminase 1 (MER003322), glutamine:fructose-6-phosphate amidotransferase (MER012158), Mername-AA144 protein (MER021319), asparagine synthetase (MER033254), family C44 non-peptidase homologues (MER159286), family C44 unassigned peptidases (MER185625) family C44 unassigned peptidases (MER185626), secernin 1 (MER045376), secernin 2 (MER064573), secernin 3 (MER064582), acid ceramidase precursor (MER100794), N-acylethanolamine acid amidase precursor (MER141667), proteasome catalytic subunit 1 (MER000556), proteasome catalytic subunit 2 (MER002625), proteasome catalytic subunit 3 (MER002149), proteasome catalytic subunit 1i (MER000552), proteasome catalytic subunit 2i (MER001515), proteasome catalytic subunit 3i (MER000555), proteasome catalytic subunit 5t (MER026203), protein serine kinase c17 (MER026497), proteasome subunit alpha 6 (MER000557), proteasome subunit alpha 2 (MER000550), proteasome subunit alpha 4 (MER000554), proteasome subunit alpha 7 (MER033250), proteasome subunit alpha 5 (MER000558), proteasome subunit alpha 1 (MER000549), proteasome subunit alpha 3 (MER000553), proteasome subunit XAPC7 (MER004372), proteasome subunit beta 3 (MER001710), proteasome subunit beta 2 (MER002676), proteasome subunit beta 1 (MER000551), proteasome subunit beta 4 (MER001711), Mername-AA230 peptidase homologue (Homo sapiens) (MER047329), Mername-AA231 pseudogene (Homo sapiens) (MER047172), Mername-AA232 pseudogene (Homo sapiens) (MER047316), glycosylasparaginase precursor (MER003299), isoaspartyl dipeptidase (threonine type) (MER031622), taspase-1 (MER016969), gamma-glutamyltransferase 5 (mammalian-type) (MER001977), gamma-glutamyltransferase 1 (mammalian-type) (MER001629), gamma-glutamyltransferase 2 (Homo sapiens) (MER001976), gamma-glutamyltransferase-like protein 4 (MER002721), gamma-glutamyltransferase-like protein 3 (MER016970), similar to gamma-glutamyltransferase 1 precursor (Homo sapiens) (MER026204), similar to gamma-glutamyltransferase 1 precursor (Homo sapiens) (MER026205), Mername-AA211 putative peptidase (MER026207), gamma-glutamyltransferase 6 (MER159283), gamma-glutamyl transpeptidase homologue (chromosome 2, Homo sapiens) (MER037241), polycystin-1 (MER126824), KIAA1879 protein (MER159329), polycystic kidney disease 1-like 3 (MER172554), gamma-glutamyl hydrolase (MER002963), guanine 5″-monophosphate synthetase (MER043387), carbamoyl-phosphate synthase (Homo sapiens-type) (MER078640), dihydro-orotase (N-terminal unit) (Homo sapiens-type) (MER060647), DJ-1 putative peptidase (MER003390), Mername-AA100 putative peptidase (MER014802), Mername-AA101 non-peptidase homologue (MER014803), KIAA0361 protein (Homo sapiens-type) (MER042827), F1134283 protein (Homo sapiens) (MER044553), non-peptidase homologue chromosome 21 open reading frame 33 (Homo sapiens) (MER160094), family C56 non-peptidase homologues (MER177016), family C56 non-peptidase homologues (MER176613), family C56 non-peptidase homologues (MER176918), EGF-like module containing mucin-like hormone receptor-like 2 (MER037230), CD97 antigen (human type) (MER037286), EGF-like module containing mucin-like hormone receptor-like 3 (MER037288), EGF-like module containing mucin-like hormone receptor-like 1 (MER037278), EGF-like module containing mucin-like hormone receptor-like 4 (MER037294), cadherin EGF LAG seven-pass G-type receptor 2 precursor (Homo sapiens) (MER045397), Gpr64 (Mus musculus)-type protein (MER123205), GPR56 (Homo sapiens)-type protein (MER122057), latrophilin 2 (MER122199), latrophilin-1 (MER126380), latrophilin 3 (MER124612), protocadherin Flamingo 2 (MER124239), ETL protein (MER126267), G protein-coupled receptor 112 (MER126114), seven transmembrane helix receptor (MER125448), Gpr114 protein (MER159320), GPR126 vascular inducible G protein-coupled receptor (MER140015), GPR125 (Homo sapiens)-type protein (MER159279), GPR116 (Homo sapiens)-type G-protein coupled receptor (MER159280), GPR128 (Homo sapiens)-type G-protein coupled receptor (MER162015), GPR133 (Homo sapiens)-type protein (MER159334), GPR110 G-protein coupled receptor (MER159277), GPR97 protein (MER159322), KPG_006 protein (MER161773), KPG_008 protein (MER161835), KPG_009 protein (MER159335), unassigned homologue (MER166269), GPR113 protein (MER159352), brain-specific angiogenesis inhibitor 2 (MER159746), PIDD auto-processing protein unit 1 (MER020001), PIDD auto-processing protein unit 2 (MER063690), MUC1 self-cleaving mucin (MER074260), dystroglycan (MER054741), proprotein convertase 9 (MER022416), site-1 peptidase (MER001948), furin (MER000375), proprotein convertase 1 (MER000376), proprotein convertase 2 (MER000377), proprotein convertase 4 (MER028255), PACE4 proprotein convertase (MER000383), proprotein convertase 5 (MER002578), proprotein convertase 7 (MER002984), tripeptidyl-peptidase II (MER000355), subfamily S8A non-peptidase homologues (MER201339), subfamily S8A non-peptidase homologues (MER191613), subfamily S8A unassigned peptidases (MER191611), subfamily S8A unassigned peptidases (MER191612), subfamily S8A unassigned peptidases (MER191614), tripeptidyl-peptidase I (MER003575), prolyl oligopeptidase (MER000393), dipeptidyl-peptidase IV (eukaryote) (MER000401), acylaminoacyl-peptidase (MER000408), fibroblast activation protein alpha subunit (MER000399), PREPL A protein (MER004227), dipeptidyl-peptidase 8 (MER013484), dipeptidyl-peptidase 9 (MER004923), FLJ1 putative peptidase (MER017240), Mername-AA194 putative peptidase (MER017353), Mername-AA195 putative peptidase (MER017367), Mername-AA196 putative peptidase (MER017368), Mername-AA197 putative peptidase (MER017371), C14orf29 protein (MER033244), hypothetical protein (MER033245), hypothetical esterase/lipase/thioesterase (MER047309), protein bat5 (MER037840), hypothetical protein flj40219 (MER033212), hypothetical protein flj37464 (MER033240), hypothetical protein flj33678 (MER033241), dipeptidylpeptidase homologue DPP6 (MER000403), dipeptidylpeptidase homologue DPP10 (MER005988), protein similar to Mus musculus chromosome 20 open reading frame 135 (MER037845), kynurenine formamidase (MER046020), thyroglobulin precursor (MER011604), acetylcholinesterase (MER033188), cholinesterase (MER033198), carboxylesterase D1 (MER033213), liver carboxylesterase (MER033220), carboxylesterase 3 (MER033224), carboxylesterase 2 (MER033226), bile salt-dependent lipase (MER033227), carboxylesterase-related protein (MER033231), neuroligin 3 (MER033232), neuroligin 4, X-linked (MER033235), neuroligin 4, Y-linked (MER033236), esterase D (MER043126), arylacetamide deacetylase (MER033237), KIAA1363-like protein (MER033242), hormone-sensitive lipase (MER033274), neuroligin 1 (MER033280), neuroligin 2 (MER033283), family S9 non-peptidase homologues (MER212939), family S9 non-peptidase homologues (MER211490), subfamily S9C unassigned peptidases (MER192341), family S9 unassigned peptidases (MER209181), family S9 unassigned peptidases (MER200434), family S9 unassigned peptidases (MER209507), family S9 unassigned peptidases (MER209142), serine carboxypeptidase A (MER000430), vitellogenic carboxypeptidase-like protein (MER005492), RISC peptidase (MER010960), family S15 unassigned peptidases (MER199442), family S15 unassigned peptidases (MER200437), family S15 unassigned peptidases (MER212825), lysosomal Pro-Xaa carboxypeptidase (MER000446), dipeptidyl-peptidase II (MER004952), thymus-specific serine peptidase (MER005538), epoxide hydrolase-like putative peptidase (MER031614), Loc328574-like protein (MER033246), abhydrolase domain-containing protein 4 (MER031616), epoxide hydrolase (MER000432), mesoderm specific transcript protein (MER199890), mesoderm specific transcript protein (MER017123), cytosolic epoxide hydrolase (MER029997), cytosolic epoxide hydrolase (MER213866), similar to hypothetical protein FLJ22408 (MER031608), CGI-58 putative peptidase (MER030163), Williams-Beuren syndrome critical region protein 21 epoxide hydrolase (MER031610), epoxide hydrolase (MER031612), hypothetical protein 922408 (epoxide hydrolase) (MER031617), monoglyceride lipase (MER033247), hypothetical protein (MER033249), valacyclovir hydrolase (MER033259), Ccg1-interacting factor b (MER210738), glycosylasparaginase precursor (MER003299), isoaspartyl dipeptidase (threonine type) (MER031622). taspase-1 (MER016969), gamma-glutamyltransferase 5 (mammalian-type) (MER001977), gamma-glutamyltransferase 1 (mammalian-type) (MER001629), gamma-glutamyltransferase 2 (Homo sapiens) (MER001976), gamma-glutamyltransferase-like protein 4 (MER002721). gamma-glutamyltransferase-like protein 3 (MER016970). similar to gamma-glutamyltransferase 1 precursor (Homo sapiens) (MER026204). similar to gamma-glutamyltransferase 1 precursor (Homo sapiens) (MER026205). Mername-AA211 putative peptidase (MER026207). gamma-glutamyltransferase 6 (MER159283). gamma-glutamyl transpeptidase homologue (chromosome 2, Homo sapiens) (MER037241). polycystin-1 (MER126824), KIAA1879 protein (MER159329). polycystic kidney disease 1-like 3 (MER172554). gamma-glutamyl hydrolase (MER002963). guanine 5″-monophosphate synthetase (MER043387). carbamoyl-phosphate synthase (Homo sapiens-type) (MER078640). dihydro-orotase (N-terminal unit) (Homo sapiens-type) (MER060647). DJ-1 putative peptidase (MER003390). Mername-AA100 putative peptidase (MER014802). Mername-AA101 non-peptidase homologue (MER014803). KIAA0361 protein (Homo sapiens-type) (MER042827). F1134283 protein (Homo sapiens) (MER044553). non-peptidase homologue chromosome 21 open reading frame 33 (Homo sapiens) (MER160094). family C56 non-peptidase homologues (MER177016), family C56 non-peptidase homologues (MER176613). family C56 non-peptidase homologues (MER176918). EGF-like module containing mucin-like hormone receptor-like 2 (MER037230). CD97 antigen (human type) (MER037286). EGF-like module containing mucin-like hormone receptor-like 3 (MER037288). EGF-like module containing mucin-like hormone receptor-like 1 (MER037278). EGF-like module containing mucin-like hormone receptor-like 4 (MER037294). cadherin EGF LAG seven-pass G-type receptor 2 precursor (Homo sapiens) (MER045397), Gpr64 (Mus musculus)-type protein (MER123205). GPR56 (Homo sapiens)-type protein (MER122057). latrophilin 2 (MER122199). latrophilin-1 (MER126380). latrophilin 3 (MER124612). protocadherin Flamingo 2 (MER124239). ETL protein (MER126267). G protein-coupled receptor 112 (MER126114). seven transmembrane helix receptor (MER125448). Gpr114 protein (MER159320). GPR126 vascular inducible G protein-coupled receptor (MER140015). GPR125 (Homo sapiens)-type protein (MER159279). GPR116 (Homo sapiens)-type G-protein coupled receptor (MER159280). GPR128 (Homo sapiens)-type G-protein coupled receptor (MER162015). GPR133 (Homo sapiens)-type protein (MER159334) GPR110 G-protein coupled receptor (MER159277), GPR97 protein (MER159322), KPG_006 protein (MER161773) KPG_008 protein (MER161835), KPG_009 protein (MER159335), unassigned homologue (MER166269), GPR113 protein (MER159352), brain-specific angiogenesis inhibitor 2 (MER159746), PIDD auto-processing protein unit 1 (MER020001), PIDD auto-processing protein unit 2 (MER063690), MUC1 self-cleaving mucin (MER074260), dystroglycan (MER054741), proprotein convertase 9 (MER022416), site-1 peptidase (MER001948), furin (MER000375), proprotein convertase 1 (MER000376), proprotein convertase 2 (MER000377), proprotein convertase 4 (MER028255), PACE4 proprotein convertase (MER000383), proprotein convertase 5 (MER002578), proprotein convertase 7 (MER002984), tripeptidyl-peptidase II (MER000355), subfamily S8A non-peptidase homologues (MER201339), subfamily S8A non-peptidase homologues (MER191613), subfamily S8A unassigned peptidases (MER191611), subfamily S8A unassigned peptidases (MER191612), subfamily S8A unassigned peptidases (MER191614), tripeptidyl-peptidase I (MER003575), prolyl oligopeptidase (MER000393), dipeptidyl-peptidase IV (eukaryote) (MER000401), acylaminoacyl-peptidase (MER000408), fibroblast activation protein alpha subunit (MER000399), PREPL A protein (MER004227), dipeptidyl-peptidase 8 (MER013484), dipeptidyl-peptidase 9 (MER004923), FLJ1 putative peptidase (MER017240), Mername-AA194 putative peptidase (MER017353), Mername-AA195 putative peptidase (MER017367), Mername-AA196 putative peptidase (MER017368), Mername-AA197 putative peptidase (MER017371), C14orf29 protein (MER033244), hypothetical protein (MER033245), hypothetical esterase/lipase/thioesterase (MER047309), protein bat5 (MER037840), hypothetical protein flj40219 (MER033212), hypothetical protein flj37464 (MER033240), hypothetical protein flj33678 (MER033241), dipeptidylpeptidase homologue DPP6 (MER000403), dipeptidylpeptidase homologue DPP10 (MER005988), protein similar to Mus musculus chromosome 20 open reading frame 135 (MER037845), kynurenine formamidase (MER046020), thyroglobulin precursor (MER011604), acetylcholinesterase (MER033188), cholinesterase (MER033198), carboxylesterase D1 (MER033213), liver carboxylesterase (MER033220), carboxylesterase 3 (MER033224), carboxylesterase 2 (MER033226), bile salt-dependent lipase (MER033227), carboxylesterase-related protein (MER033231), neuroligin 3 (MER033232), neuroligin 4, X-linked (MER033235), neuroligin 4, Y-linked (MER033236), esterase D (MER043126), arylacetamide deacetylase (MER033237), KIAA1363-like protein (MER033242), hormone-sensitive lipase (MER033274), neuroligin 1 (MER033280), neuroligin 2 (MER033283), family S9 non-peptidase homologues (MER212939), family S9 non-peptidase homologues (MER211490), subfamily S9C unassigned peptidases (MER192341), family S9 unassigned peptidases (MER209181), family S9 unassigned peptidases (MER200434), family S9 unassigned peptidases (MER209507), family S9 unassigned peptidases (MER209142), serine carboxypeptidase A (MER000430), vitellogenic carboxypeptidase-like protein (MER005492), RISC peptidase (MER010960), family S15 unassigned peptidases (MER199442), family S15 unassigned peptidases (MER200437), family S15 unassigned peptidases (MER212825), lysosomal Pro-Xaa carboxypeptidase (MER000446), dipeptidyl-peptidase II (MER004952), thymus-specific serine peptidase (MER005538), epoxide hydrolase-like putative peptidase (MER031614), Loc328574-like protein (MER033246), abhydrolase domain-containing protein 4 (MER031616), epoxide hydrolase (MER000432), mesoderm specific transcript protein (MER199890), mesoderm specific transcript protein (MER017123), cytosolic epoxide hydrolase (MER029997), cytosolic epoxide hydrolase (MER213866), similar to hypothetical protein FLJ22408 (MER031608), CGI-58 putative peptidase (MER030163), Williams-Beuren syndrome critical region protein 21 epoxide hydrolase (MER031610), epoxide hydrolase (MER031612), hypothetical protein flj22408 (epoxide hydrolase) (MER031617), monoglyceride lipase (MER033247), hypothetical protein (MER033249), valacyclovir hydrolase (MER033259), Ccg1-interacting factor b (MER210738).


Protease enzymatic activity can be regulated. For example, certain proteases are inhibited by specific inhibitors, such as specific small molecule inhibitors. Exemplary inhibitors for certain proteases are listed in Table 4. For example, an NS3 protease can be repressed by a protease inhibitor including, but not limited to, simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir. In another example, protease activity can be regulated through regulating expression of the protease itself, such as engineering a cell to express a protease using an inducible promoter system (e.g., Tet On/Off systems) or cell-specific promoters (promoters that can be used to express a heterologous protease are described in more detail in the Section herein titled “Promoters”). A protease can also contain a degron, such as any of the degrons described herein, and can be regulated using any of the degron systems described herein.


Cell Membrane Tethering Domain

The membrane-cleavable chimeric proteins provided for herein contain a cell-membrane tethering domain (referred to as “MT” in the formula S-C-MT-D). In general, the cell-membrane tethering domain can be any amino acid sequence motif capable of directing the chimeric protein to be localized to (e.g., inserted into), or otherwise associated with, the cell membrane of the cell expressing the chimeric protein. The cell-membrane tethering domain can be a transmembrane-intracellular domain. The cell-membrane tethering domain can be a transmembrane domain. The cell-membrane tethering domain can be an integral membrane protein domain (e.g., a transmembrane domain). Examples of cell-membrane tethering domains include, but are not limited to, a transmembrane-intracellular domain and/or transmembrane domain derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA. The cell membrane tethering domain can be a cell surface receptor or a cell membrane-bound portion thereof.


In general, for all membrane-cleavable chimeric proteins described herein, the cell membrane tethering domain is C-terminal of the protease cleavage site and N-terminal of the degron (in other words, the cell membrane tethering domain is in between the protease cleavage site and the degron). The cell membrane tethering domain can be connected to the protease cleavage site by a polypeptide linker, i.e., a polypeptide sequence not generally considered to be part of cell membrane tethering domain or protease cleavage site. The cell membrane tethering domain can be connected to the degron by a polypeptide linker, i.e., a polypeptide sequence not generally considered to be part of the cell membrane tethering domain or degron. A polypeptide linker can be any amino acid sequence that connects a first polypeptide sequence and a second polypeptide sequence. A polypeptide linker can be a flexible linker (e.g., a Gly-Ser-Gly sequence). Examples of polypeptide linkers include, but are not limited to, GSG linkers (e.g., [GS]4GG [SEQ ID NO: 182]), A(EAAAK)3A (SEQ ID NO: 183), and Whitlow linkers (e.g., a “KEGS (SEQ ID NO:195)” linker such as the amino acid sequence KESGSVSSEQLAQFRSLD (SEQ ID NO: 184), a EGK linker such as the amino acid sequence EGKSSGSGSESKST (SEQ ID NO: 185), and linkers described in more detail in Issued U.S. Pat. No. 5,990,275 herein incorporated by reference). Other polypeptide linkers may be selected based on desired properties (e.g., length, flexibility, amino acid composition etc.) and are known to those skilled in the art.


In general, the cell-membrane tethering domain is oriented such that the secreted effector molecule and the protease cleavage site are extracellularly exposed following insertion into, or association with, the cell membrane, such that the protease cleavage site is capable of being cleaved by its respective protease and releasing (“secreting”) the effector molecule into the extracellular space.


Degron

The chimeric proteins provided for herein contain a degron (e.g., referred to as “D” in the formula S-C-MT-D for membrane-cleavable chimeric proteins described herein). In general, the degron can be any amino acid sequence motif capable of directing regulated degradation of the chimeric protein, such as through regulated degradation through a ubiquitin-mediated pathway. The degron can be a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) including, but not limited to, IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN. The CRBN polypeptide substrate domain can be a chimeric fusion product of native CRBN polypeptide sequences, such as a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189). Degrons, and in particular CRBN degron systems, are described in more detail in International Application Pub. No. WO2019/089592A1, herein incorporated by reference for all purposes. Other examples of degrons include, but are not limited to HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα; SEQ ID NO: 161), GRR (residues 352-408 of human p105; SEQ ID NO: 162), DRR (residues 210-295 of yeast Cdc34; SEQ ID NO: 163), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B; e.g., SEQ ID NO: 164), RPB (four copies of residues 1688-1702 of yeast RPB; SEQ ID NO: 165), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein; SEQ ID NO: 166), NS2 (three copies of residues 79-93 of influenza A virus NS protein; SEQ ID NO: 167), ODC (residues 106-142 of ornithine decarboxylase; SEQ ID NO: 168), Nek2A, mouse ODC (residues 422-461; SEQ ID NO: 169), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, or a PCNA binding PIP box.


Regulated degradation can be drug-inducible. Drugs capable of mediating/regulating degradation can be small-molecule compounds. Drugs capable of mediating/regulating degradation can include an “immunomodulatory drug” (IMiD). In general, as used herein, IMiDs refer to a class of small-molecule immunomodulatory drugs containing an imide group. Cereblon (CRBN) is known target of IMiDs and binding of an IMiD to CRBN or a CRBN polypeptide substrate domain alters the substrate specificity of the CRBN E3 ubiquitin ligase complex leading to degradation of proteins having a CRBN polypeptide substrate domain (e.g., any of secretable effector molecules or other proteins of interest described herein). For degrons having a CRBN polypeptide substrate domain, examples of imide-containing IMiDs include, but are not limited to, a thalidomide, a lenalidomide, or a pomalidomide. The IMiD can be an FDA-approved drug.


In general, for all membrane-cleavable chimeric proteins described herein, the degron is C-terminal of the cell membrane tethering domain. The degron can be connected to the cell membrane tethering domain by a polypeptide linker, i.e., a polypeptide sequence not generally considered to be part of the cell membrane tethering domain or degron. A polypeptide linker can be any amino acid sequence that connects a first polypeptide sequence and a second polypeptide sequence. A polypeptide linker can be a flexible linker (e.g., a Gly-Ser-Gly sequence). Examples of polypeptide linkers include, but are not limited to, GSG linkers (e.g., [GS]4GG [SEQ ID NO: 182]), A(EAAAK)3A (SEQ ID NO: 183), and Whitlow linkers (e.g., a “KEGS” (SEQ ID NO:195) linker such as the amino acid sequence KESGSVSSEQLAQFRSLD (SEQ ID NO: 184), a EGK linker such as the amino acid sequence EGKSSGSGSESKST (SEQ ID NO: 185), and linkers described in more detail in Issued U.S. Pat. No. 5,990,275 herein incorporated by reference). Other polypeptide linkers may be selected based on desired properties (e.g., length, flexibility, amino acid composition etc.) and are known to those skilled in the art. In general, the degron is oriented in relation to the cell membrane tethering domain such that the degron is exposed to the cytosol following localization to the cell membrane such that the degron is capable of mediating degradation (e.g., exposure to the cytosol and cytosol) and is capable of mediating ubiquitin-mediated degradation.


For degron-fusion chimeric proteins, the degron can be N-terminal or C-terminal of the protein of interest, e.g., the effector molecule. The degron can be connected to the protein of interest by a polypeptide linker, i.e., a polypeptide sequence not generally considered to be part of the protein of interest or the degron. A polypeptide linker can be any amino acid sequence that connects a first polypeptide sequence and a second polypeptide sequence. A polypeptide linker can be a flexible linker (e.g., a Gly-Ser-Gly sequence). Examples of polypeptide linkers include, but are not limited to, GSG linkers (e.g., [GS]4GG [SEQ ID NO: 182]), A(EAAAK)3A (SEQ ID NO: 183), and Whitlow linkers (e.g., a “KEGS” (SEQ ID NO:195) linker such as the amino acid sequence KESGSVSSEQLAQFRSLD (SEQ ID NO: 184), a EGK linker such as the amino acid sequence EGKSSGSGSESKST (SEQ ID NO: 185), and linkers described in more detail in Issued U.S. Pat. No. 5,990,275 herein incorporated by reference). Other polypeptide linkers may be selected based on desired properties (e.g., length, flexibility, amino acid composition etc.) and are known to those skilled in the art. A polypeptide linker can be cleavable, e.g., any of the protease cleavage sites described herein.


Homing Molecules

A “tumor microenvironment” is the cellular environment in which a tumor exists, including surrounding blood vessels, immune cells, fibroblasts, bone marrow-derived inflammatory cells, lymphocytes, signaling molecules and the extracellular matrix (ECM) (see, e.g., Pattabiraman, D. R. & Weinberg, R. A. Nature Reviews Drug Discovery 13, 497-512 (2014); Balkwill, F. R. et al. J Cell Sci 125, 5591-5596, 2012; and Li, H. et al. J Cell Biochem 101(4), 805-15, 2007).


In some embodiments, engineered nucleic acids are configured to produce at least one homing molecule. For example, in degron-fusion chimeric proteins described herein containing a protein of interest fused to degron domain (e.g., see fusions proteins related to engineered AAV vectors and viruses described elsewhere herein), the protein of interest can be a homing molecule. “Homing,” refers to active navigation (migration) of a cell to a target site (e.g., a cell, tissue (e.g., tumor), or organ). A “homing molecule” refers to a molecule that directs cells to a target site. In some embodiments, a homing molecule functions to recognize and/or initiate interaction of an engineered cell to a target site. Non-limiting examples of homing molecules include CXCR1, CCR9, CXCR2, CXCR3, CXCR4, CCR2, CCR4, FPR2, VEGFR, IL6R, CXCR1, CSCR7, and PDGFR.


In some embodiments, a homing molecule is a chemokine receptor (cell surface molecule that binds to a chemokine). Chemokines are small cytokines or signaling proteins secreted by cells that can induce directed chemotaxis in cells. Chemokines can be classified into four main subfamilies: CXC, CC, CX3C and XC, all of which exert biological effects by binding selectively to chemokine receptors located on the surface of target cells. In some embodiments, engineered nucleic acids are configured to produce CXCR4, a chemokine receptor which allows engineered cells to home along a chemokine gradient towards a stromal cell-derived factor 1 (also known as SDF1, C-X-C motif chemokine 12, and CXCL12)-expressing cell, tissue, or tumor. Non-limiting examples of chemokine receptors that may be encoded by the engineered nucleic acids of the present disclosure include: CXC chemokine receptors (e.g., CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, and CXCR7), CC chemokine receptors (CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, and CCR11), CX3C chemokine receptors (e.g., CX3CR1, which binds to CX3CL1), and XC chemokine receptors (e.g., XCR1). In some embodiments, a chemokine receptor is a G protein-linked transmembrane receptor, or a member of the tumor necrosis factor (TNF) receptor superfamily (including but not limited to TNFRSF1A, TNFRSF1B). In some embodiments, engineered nucleic acids are configured to produce CXCL8, CXCL9, and/or CXCL10 (promote T-cell recruitment), CCL3 and/or CXCL5, CCL21 (Th1 recruitment and polarization).


In some embodiments, engineered nucleic acids are configured to produce G-protein coupled receptors (GPCRs) that detect N-formylated-containing oligopeptides (including but not limited to FPR2 and FPRL1).


In some embodiments, engineered nucleic acids are configured to produce receptors that detect interleukins (including but not limited to IL6R).


In some embodiments, engineered nucleic acids are configured to produce receptors that detect growth factors secreted from other cells, tissues, or tumors (including but not limited to FGFR, PDGFR, EGFR, and receptors of the VEGF family, including but not limited to VEGF-C and VEGF-D).


In some embodiments, a homing molecule is an integrin. Integrins are transmembrane receptors that facilitate cell-extracellular matrix (ECM) adhesion. Integrins are obligate heterodimers having two subunits: u (alpha) and R (beta). The a subunit of an integrin may be, without limitation: ITGA1, ITGA2, ITGA3, ITGA4, ITGA5, ITGA6, IGTA7, ITGA8, ITGA9, IGTA10, IGTA11, ITGAD, ITGAE, ITGAL, ITGAM, ITGAV, ITGA2B, ITGAX. The β subunit of an integrin may be, without limitation: ITGB1, ITGB2, ITGB3, ITGB4, ITGB5, ITGB6, ITGB7, and ITGB8. Engineered nucleic acids can be configured to produce any combination of the integrin a and R subunits.


In some embodiments, a homing molecule is a matrix metalloproteinase (MMP). MMPs are enzymes that cleave components of the basement membrane underlying the endothelial cell wall. Non-limiting examples of MMPs include MMP-2, MMP-9, and MMP. In some embodiments, engineered nucleic acids are configured to produce an inhibitor of a molecule (e.g., protein) that inhibits MMPs. For example, engineered nucleic acids can be configured to express an inhibitor (e.g., an RNAi molecule) of membrane type 1 MMP (MT1-MMP) or TIMP metallopeptidase inhibitor 1 (TIMP-1).


In some embodiments, a homing molecule is a ligand that binds to selectin (e.g., hematopoietic cell E-/L-selectin ligand (HCELL), Dykstra et al., Stem Cells. 2016 October; 34(10):2501-2511) on the endothelium of a target tissue, for example.


The term “homing molecule” also encompasses transcription factors that regulate the production of molecules that improve/enhance homing of cells.


Engineered Nucleic Acids


Provided herein are engineered nucleic acids encoding at least one chimeric protein of the present disclosure, such as such as the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein. Provided herein are engineered nucleic acids encoding two or more chimeric proteins.


In certain embodiments described herein, the engineered nucleic acids encode an expression cassette containing a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-D. S refers to a secretable effector molecule. C refers to a protease cleavage site. MT refers to a cell membrane tethering domain. D refers to a degron. The promoter is operably linked to the exogenous polynucleotide sequence and S-C-MT-D is configured to be expressed as a single polypeptide.


In certain embodiments described herein, the engineered nucleic acids encode an expression cassette containing a promoter and an exogenous polynucleotide sequence encoding a degron-fusion chimeric protein having a protein of interest (e.g., any of the effector molecules described herein) and a degron. The promoter is operably linked to the exogenous polynucleotide sequence and the degron-fusion chimeric protein is configured to be expressed as a single polypeptide.


An “engineered nucleic acid” is a nucleic acid that does not occur in nature. It should be understood, however, that while an engineered nucleic acid as a whole is not naturally-occurring, it may include nucleotide sequences that occur in nature. In some embodiments, an engineered nucleic acid comprises nucleotide sequences from different organisms (e.g., from different species). For example, in some embodiments, an engineered nucleic acid includes a murine nucleotide sequence, a bacterial nucleotide sequence, a human nucleotide sequence, and/or a viral nucleotide sequence. The term “engineered nucleic acids” includes recombinant nucleic acids and synthetic nucleic acids. A “recombinant nucleic acid” refers to a molecule that is constructed by joining nucleic acid molecules and, in some embodiments, can replicate in a live cell. A “synthetic nucleic acid” refers to a molecule that is amplified or chemically, or by other means, synthesized. Synthetic nucleic acids include those that are chemically modified, or otherwise modified, but can base pair with naturally-occurring nucleic acid molecules. Modifications include, but are not limited to, one or more modified internucleotide linkages and non-natural nucleic acids. Modifications are described in further detail in U.S. Pat. No. 6,673,611 and U.S. Application Publication 2004/0019001 and, each of which is incorporated by reference in their entirety. Modified internucleotide linkages can be a phosphorodithioate or phosphorothioate linkage. Non-natural nucleic acids can be a locked nucleic acid (LNA), a peptide nucleic acid (PNA), glycol nucleic acid (GNA), a phosphorodiamidate morpholino oligomer (PMO or “morpholino”), and threose nucleic acid (TNA). Non-natural nucleic acids are described in further detail in International Application WO 1998/039352, U.S. Application Pub. No. 2013/0156849, and U.S. Pat. Nos. 6,670,461; 5,539,082; 5,185,444, each herein incorporated by reference in their entirety. Recombinant nucleic acids and synthetic nucleic acids also include those molecules that result from the replication of either of the foregoing. Engineered nucleic acid of the present disclosure may be encoded by a single molecule (e.g., included in the same plasmid or other vector) or by multiple different molecules (e.g., multiple different independently-replicating molecules). Engineered nucleic acids can be an isolated nucleic acid. Isolated nucleic acids include, but are not limited to a cDNA polynucleotide, an RNA polynucleotide, an RNAi oligonucleotide (e.g., siRNAs, miRNAs, antisense oligonucleotides, shRNAs, etc.), an mRNA polynucleotide, a circular plasmid, a linear DNA fragment, a vector, a minicircle, a ssDNA, and an oligonucleotide.


Engineered nucleic acid of the present disclosure may be produced using standard molecular biology methods (see, e.g., Green and Sambrook, Molecular Cloning, A Laboratory Manual, 2012, Cold Spring Harbor Press). In some embodiments, engineered nucleic acid constructs are produced using GIBSON ASSEMBLY® Cloning (see, e.g., Gibson, D. G. et al. Nature Methods, 343-345, 2009; and Gibson, D. G. et al. Nature Methods, 901-903, 2010, each of which is incorporated by reference herein). GIBSON ASSEMBLY® typically uses three enzymatic activities in a single-tube reaction: 5′ exonuclease, the ′Y extension activity of a DNA polymerase and DNA ligase activity. The 5′ exonuclease activity chews back the 5′end sequences and exposes the complementary sequence for annealing. The polymerase activity then fills in the gaps on the annealed regions. A DNA ligase then seals the nick and covalently links the DNA fragments together. The overlapping sequence of adjoining fragments is much longer than those used in Golden Gate Assembly, and therefore results in a higher percentage of correct assemblies. In some embodiments, engineered nucleic acid constructs are produced using IN-FUSION® cloning (Clontech).


Promoters

In general, in all embodiments described herein, the engineered nucleic acids encoding the one or more chimeric proteins encode an expression cassette containing a promoter. In some embodiments, an engineered nucleic acid (e.g., an engineered nucleic acid comprising an expression cassette) comprises a promoter operably linked to a nucleotide sequence (e.g., an exogenous polynucleotide sequence) encoding at least 2 chimeric proteins. For example, the engineered nucleic acid may comprise a promoter operably linked to a nucleotide sequence encoding at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 8, at least 9, or at least 10 chimeric proteins. In some embodiments, an engineered nucleic acid comprises a promoter operably linked to a nucleotide sequence encoding 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more chimeric proteins.


A “promoter” refers to a control region of a nucleic acid sequence at which initiation and rate of transcription of the remainder of a nucleic acid sequence are controlled. A promoter may also contain sub-regions at which regulatory proteins and molecules may bind, such as RNA polymerase and other transcription factors. Promoters may be constitutive, inducible, repressible, tissue-specific or any combination thereof. A promoter drives expression or drives transcription of the nucleic acid sequence that it regulates. Herein, a promoter is considered to be “operably linked” when it is in a correct functional location and orientation in relation to a nucleic acid sequence it regulates to control (“drive”) transcriptional initiation and/or expression of that sequence.


A promoter may be one naturally associated with a gene or sequence, as may be obtained by isolating the 5′ non-coding sequences located upstream of the coding segment of a given gene or sequence. Such a promoter can be referred to as “endogenous.” In some embodiments, a coding nucleic acid sequence may be positioned under the control of a recombinant or heterologous promoter, which refers to a promoter that is not normally associated with the encoded sequence in its natural environment. Such promoters may include promoters of other genes; promoters isolated from any other cell; and synthetic promoters or enhancers that are not “naturally occurring” such as, for example, those that contain different elements of different transcriptional regulatory regions and/or mutations that alter expression through methods of genetic engineering that are known in the art. In addition to producing nucleic acid sequences of promoters and enhancers synthetically, sequences may be produced using recombinant cloning and/or nucleic acid amplification technology, including polymerase chain reaction (PCR) (see, e.g., U.S. Pat. Nos. 4,683,202 and 5,928,906).


Promoters of an engineered nucleic acid may be “inducible promoters,” which refer to promoters that are characterized by regulating (e.g., initiating or activating) transcriptional activity when in the presence of, influenced by or contacted by a signal. The signal may be endogenous or a normally exogenous condition (e.g., light), compound (e.g., chemical or non-chemical compound) or protein (e.g., cytokine) that contacts an inducible promoter in such a way as to be active in regulating transcriptional activity from the inducible promoter. Activation of transcription may involve directly acting on a promoter to drive transcription or indirectly acting on a promoter by inactivation a repressor that is preventing the promoter from driving transcription. Conversely, deactivation of transcription may involve directly acting on a promoter to prevent transcription or indirectly acting on a promoter by activating a repressor that then acts on the promoter.


A promoter is “responsive to” or “modulated by” a local tumor state (e.g., inflammation or hypoxia) or signal if in the presence of that state or signal, transcription from the promoter is activated, deactivated, increased, or decreased. In some embodiments, the promoter comprises a response element. A “response element” is a short sequence of DNA within a promoter region that binds specific molecules (e.g., transcription factors) that modulate (regulate) gene expression from the promoter. Response elements that may be used in accordance with the present disclosure include, without limitation, a phloretin-adjustable control element (PEACE), a zinc-finger DNA-binding domain (DBD), an interferon-gamma-activated sequence (GAS) (Decker, T. et al. J Interferon Cytokine Res. 1997 March; 17(3):121-34, incorporated herein by reference), an interferon-stimulated response element (ISRE) (Han, K. J. et al. J Biol Chem. 2004 Apr. 9; 279(15):15652-61, incorporated herein by reference), a NF-kappaB response element (Wang, V. et al. Cell Reports. 2012; 2(4): 824-839, incorporated herein by reference), and a STAT3 response element (Zhang, D. et al. J of Biol Chem. 1996; 271: 9503-9509, incorporated herein by reference). Other response elements are encompassed herein. Response elements can also contain tandem repeats (e.g., consecutive repeats of the same nucleotide sequence encoding the response element) to generally increase sensitivity of the response element to its cognate binding molecule. Tandem repeats can be labeled 2×, 3×, 4×, 5×, etc. to denote the number of repeats present.


Non-limiting examples of responsive promoters (also referred to as “inducible promoters”) (e.g., TGF-beta responsive promoters) are listed in Table 5A, which shows the design of the promoter and transcription factor, as well as the effect of the inducer molecule towards the transcription factor (TF) and transgene transcription (T) is shown (B, binding; D, dissociation; n.d., not determined) (A, activation; DA, deactivation; DR, derepression) (see Horner, M. & Weber, W. FEBS Letters 586 (2012) 20784-2096m, and references cited therein). Other non-limiting examples of inducible promoters include those presented in Table 5B.









TABLE 5A







Examples of Responsive Promoters.















Response to



Promoter and
Transcription

inducer












System
operator
factor (TF)
Inducer molecule
TF
T










Transcriptional activator-responsive promoters












AIR
PAIR (OalcA-
AlcR
Acetaldehyde
n.d.
A



PhCMVmin)






ART
PART (OARG-
ArgR-VP16
l-Arginine
B
A



PhCMVmin)






BIT
PBIT3 (OBirA3-
BIT (BirA-
Biotin
B
A



PhCMVmin)
VP16)





Cumate-activator
PCR5 (OCuO6-
cTA (CymR-
Cumate
D
DA



PhCMVmin)
VP16)





Cumate-reverse
PCR5 (OCuO6-
rcTA (rCymR-
Cumate
B
A


activator
PhCMVmin)
VP16)





E-OFF
PETR (OETR-
ET (E-VP16)
Erythromycin
D
DA



PhCMVmin)






NICE-OFF
PNIC (ONIC-
NT (HdnoR-
6-Hydroxy-nicotine
D
DA



PhCMVmin)
VP16)





PEACE
PTtgR1 (OTtgR-
TtgA1 (TtgR-
Phloretin
D
DA



PhCMVmin)
VP16)





PIP-OFF
PPIR (OPIR-
PIT (PIP-
Pristinamycin I
D
DA



Phsp70min)
VP16)





QuoRex
PSCA (OscbR-
SCA (ScbR-
SCB1
D
DA



PhCMVmin)PSPA
VP16)






(OpapRI-







PhCMVmin)






Redox
PROP (OROP-
REDOX
NADH
D
DA



PhCMVmin)
(REX-VP16)





TET-OFF
PhCMV*-1
tTA (TetR-
Tetracycline
D
DA



(OtetO7-
VP16)






PhCMVmin)






TET-ON
PhCMV*-1
rtTA (rTetR-
Doxycycline
B
A



(OtetO7-
VP16)






PhCMVmin)






TIGR
PCTA (OrheO-
CTA (RheA-
Heat
D
DA



PhCMVmin)
VP16)





TraR
O7x(tra box)-
p65-TraR
3-Oxo-C8-HSL
B
A



PhCMVmin






VAC-OFF
P1VanO2
VanA1 (VanR-
Vanillic acid
D
DA



(OVanO2-
VP16)






PhCMVmin)











Transcriptional repressor-responsive promoters












Cumate-repressor
PCuO (PCMV5-
CymR
Cumate
D
DR



OCuO)






E-ON
PETRON8
E-KRAB
Erythromycin
D
DR



(PSV40-OETR8)






NICE-ON
PNIC (PSV40-
NS (HdnoR-
6-Hydroxy-nicotine
D
DR



ONIC8)
KRAB)





PIP-ON
PPIRON (PSV40-
PIT3 (PIP-
Pristinamycin I
D
DR



OPIR3)
KRAB)





Q-ON
PSCAON8
SCS (ScbR-
SCB1
D
DR



(PSV40-OscbR8)
KRAB)





TET-ON
OtetO-PHPRT
tTS-H4 (TetR-
Doxycycline
D
DR


repressor-based

HDAC4)





T-REX
PTetO (PhCMV-
TetR
Tetracycline
D
DR



OtetO2)






UREX
PUREX8 (PSV40-
mUTS
Uric acid
D
DR



OhucO8)
(KRAB-HucR)





VAC-ON
PVanON8
VanA4 (VanR-
Vanillic acid
D
DR



(PhCMV-
KRAB)






OVanO8)















Hybrid promoters
















QuoRexPIP-
OscbR8-OPIR3-
SCAPIT3
SCB 1Pristinamycin I
DD
DADR


ON(NOT IF gate)
PhCMVmin






QuoRexE-
OscbR-OETR8-
SCAE-KRAB
SCB1Erythromycin
DD
DADR


ON(NOT IF gate)
PhCMVmin






TET-OFFE-
OtetO7-OETR8-
tTAE-KRAB
TetracyclineErythromycin
DD
DADR


ON(NOT IF gate)
PhCMVmin






TET-OFFPIP-
OtetO7-OPIR3-
tTAPIT3E-
TetracyclinePristinamycin
DDD
DADRDR


ONE-ON
OETR8-
KRAB
IErythromycin





PhCMVmin
















TABLE 5B







Exemplary Inducible Promoters








Name
DNA SEQUENCE





minimal promoter; minP
AGAGGGTATATAATGGAAGCTCGACTTCCAG (SEQ ID NO: 1)


NFkB response element
GGGAATTTCCGGGGACTTTCCGGGAATTTCCGGGGACTTTCCGGGAAT


protein promoter; 5x
TTCC (SEQ ID NO: 2)


NFkB-RE






CREB response element
CACCAGACAGTGACGTCAGCTGCCAGATCCCATGGCCGTCATACTGTG


protein promoter; 4x CRE
ACGTCTTTCAGACACCCCATTGACGTCAATGGGAGAA (SEQ ID NO: 3)





NFAT response element
GGAGGAAAAACTGTTTCATACAGAAGGCGTGGAGGAAAAACTGTTTC


protein promoter; 3x NFAT
ATACAGAAGGCGTGGAGGAAAAACTGTTTCATACAGAAGGCGT (SEQ


binding sites
ID NO: 4)





SRF response element
AGGATGTCCATATTAGGACATCTAGGATGTCCATATTAGGACATCTAG


protein promoter; 5x SRE
GATGTCCATATTAGGACATCTAGGATGTCCATATTAGGACATCTAGGA



TGTCCATATTAGGACATCT (SEQ ID NO: 5)





SRF response element
AGTATGTCCATATTAGGACATCTACCATGTCCATATTAGGACATCTAC


protein promoter 2; 5x
TATGTCCATATTAGGACATCTTGTATGTCCATATTAGGACATCTAAAA


SRF-RE
TGTCCATATTAGGACATCT (SEQ ID NO: 6)





AP1 response element
TGAGTCAGTGACTCAGTGAGTCAGTGACTCAGTGAGTCAGTGACTCA


protein promoter; 6x AP1-
G (SEQ ID NO: 7)


RE






TCF-LEF response element
AGATCAAAGGGTTTAAGATCAAAGGGCTTAAGATCAAAGGGTATAAG


promoter; 8x TCF-LEF-RE
ATCAAAGGGCCTAAGATCAAAGGGACTAAGATCAAAGGGTTTAAGAT



CAAAGGGCTTAAGATCAAAGGGCCTA (SEQ ID NO: 8)





SBEx4
GTCTAGACGTCTAGACGTCTAGACGTCTAGAC (SEQ ID NO: 9)





SMAD2/3-CAGACA x4
CAGACACAGACACAGACACAGACA (SEQ ID NO: 10)





STAT3 binding site
Ggatccggtactcgagatctgcgatctaagtaagcttggcattccg



gtactgttggtaaagccac (SEQ ID NO: 11)





minCMV
taggcgtgtacggtgggaggcctatataagcagagctcgtttagtg



aaccgtcagatcgcctgga (SEQ ID NO: 170)





YB_TATA
TCTAGAGGGTATATAATGGGGGCCA (SEQ ID NO: 171)





minTK
Ttcgcatattaaggtgacgcgtgtggcctcgaacaccgagcgaccct



gcagcgacccgcttaa (SEQ ID NO: 172)









Other non-limiting examples of promoters include the cytomegalovirus (CMV) promoter, the elongation factor 1-alpha (EFla) promoter, the elongation factor (EFS) promoter, the MND promoter (a synthetic promoter that contains the U3 region of a modified MoMuLV LTR with myeloproliferative sarcoma virus enhancer), the phosphoglycerate kinase (PGK) promoter, the spleen focus-forming virus (SFFV) promoter, the simian virus 40 (SV40) promoter, and the ubiquitin C (UbC) promoter (see Table 5C).









TABLE 5C







Exemplary Constitutive Promoters








Name
DNA SEQUENCE





CMV
GTTGACATTGATTATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATTA



GTTCATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCC



GCCTGGCTGACCGCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTAT



GTTCCCATAGTAACGCCAATAGGGACTTTCCATTGACGTCAATGGGTGGAGT



ATTTACGGTAAACTGCCCACTTGGCAGTACATCAAGTGTATCATATGCCAAG



TACGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCATTATGCC



CAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGT



CATCGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGGGCGTGGA



TAGCGGTTTGACTCACGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGG



GAGTTTGTTTTGGCACCAAAATCAACGGGACTTTCCAAAATGTCGTAACAAC



TCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGTACGGTGGGAGGTCTATA



TAAGCAGAGCTC (SEQ ID NO: 12)





EF1a
GGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCACAGTCCCCGAGA



AGTTGGGGGGAGGGGTCGGCAATTGAACCGGTGCCTAGAGAAGGTGGCGCG



GGGTAAACTGGGAAAGTGATGCCGTGTACTGGCTCCGCCTTTTTCCCGAGGG



TGGGGGAGAACCGTATATAAGTGCAGTAGTCGCCGTGAACGTTCTTTTTCGC



AACGGGTTTGCCGCCAGAACACAGGTAAGTGCCGTGTGTGGTTCCCGCGGGC



CTGGCCTCTTTACGGGTTATGGCCCTTGCGTGCCTTGAATTACTTCCACCTGG



CTGCAGTACGTGATTCTTGATCCCGAGCTTCGGGTTGGAAGTGGGTGGGAGA



GTTCGAGGCCTTGCGCTTAAGGAGCCCCTTCGCCTCGTGCTTGAGTTGAGGCC



TGGCCTGGGCGCTGGGGCCGCCGCGTGCGAATCTGGTGGCACCTTCGCGCCT



GTCTCGCTGCTTTCGATAAGTCTCTAGCCATTTAAAATTTTTGATGACCTGCT



GCGACGCTTTTTTTCTGGCAAGATAGTCTTGTAAATGCGGGCCAAGATCTGCA



CACTGGTATTTCGGTTTTTGGGGCCGCGGGCGGCGACGGGGCCCGTGCGTCC



CAGCGCACATGTTCGGCGAGGCGGGGCCTGCGAGCGCGACCACCGAGAATC



GGACGGGGGTAGTCTCAAGCTGGCCGGCCTGCTCTGGTGCCTGTCCTCGCGC



CGCCGTGTATCGCCCCGCCCCGGGCGGCAAGGCTGGCCCGGTCGGCACCAGT



TGCGTGAGCGGAAAGATGGCCGCTTCCCGGTCCTGCTGCAGGGAGCTCAAAA



TGGAGGACGCGGCGCTCGGGAGAGCGGGCGGGTGAGTCACCCACACAAAGG



AAAAGGGCCTTTCCGTCCTCAGCCGTCGCTTCATGTGACTCCACGGAGTACC



GGGCGCCGTCCAGGCACCTCGATTAGTTCTCGAGCTTTTGGAGTACGTCGTCT



TTAGGTTGGGGGGAGGGGTTTTATGCGATGGAGTTTCCCCACACTGAGTGGG



TGGAGACTGAAGTTAGGCCAGCTTGGCACTTGATGTAATTCTCCTTGGAATTT



GCCCTTTTTGAGTTTGGATCTTGGTTCATTCTCAAGCCTCAGACAGTGGTTCA



AAGTTTTTTTCTTCCATTTCAGGTGTCGTGA (SEQ ID NO: 13)





EFS
GGATCTGCGATCGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCAC



AGTCCCCGAGAAGTTGGGGGGAGGGGTCGGCAATTGAACCGGTGCCTAGAG



AAGGTGGCGCGGGGTAAACTGGGAAAGTGATGTCGTGTACTGGCTCCGCCTT



TTTCCCGAGGGTGGGGGAGAACCGTATATAAGTGCAGTAGTCGCCGTGAACG



TTCTTTTTCGCAACGGGTTTGCCGCCAGAACACAGCTGAAGCTTCGAGGGGC



TCGCATCTCTCCTTCACGCGCCCGCCGCCCTACCTGAGGCCGCCATCCACGCC



GGTTGAGTCGCGTTCTGCCGCCTCCCGCCTGTGGTGCCTCCTGAACTGCGTCC



GCCGTCTAGGTAAGTTTAAAGCTCAGGTCGAGACCGGGCCTTTGTCCGGCGC



TCCCTTGGAGCCTACCTAGACTCAGCCGGCTCTCCACGCTTTGCCTGACCCTG



CTTGCTCAACTCTACGTCTTTGTTTCGTTTTCTGTTCTGCGCCGTTACAGATCC



AAGCTGTGACCGGCGCCTAC (SEQ ID NO: 14)





MND
TTTATTTAGTCTCCAGAAAAAGGGGGGAATGAAAGACCCCACCTGTAGGTTT



GGCAAGCTAGGATCAAGGTTAGGAACAGAGAGACAGCAGAATATGGGCCAA



ACAGGATATCTGTGGTAAGCAGTTCCTGCCCCGGCTCAGGGCCAAGAACAGT



TGGAACAGCAGAATATGGGCCAAACAGGATATCTGTGGTAAGCAGTTCCTGC



CCCGGCTCAGGGCCAAGAACAGATGGTCCCCAGATGCGGTCCCGCCCTCAGC



AGTTTCTAGAGAACCATCAGATGTTTCCAGGGTGCCCCAAGGACCTGAAATG



ACCCTGTGCCTTATTTGAACTAACCAATCAGTTCGCTTCTCGCTTCTGTTCGC



GCGCTTCTGCTCCCCGAGCTCAATAAAAGAGCCCA (SEQ ID NO: 15)





PGK
GGGGTTGGGGTTGCGCCTTTTCCAAGGCAGCCCTGGGTTTGCGCAGGGACGC



GGCTGCTCTGGGCGTGGTTCCGGGAAACGCAGCGGCGCCGACCCTGGGTCTC



GCACATTCTTCACGTCCGTTCGCAGCGTCACCCGGATCTTCGCCGCTACCCTT



GTGGGCCCCCCGGCGACGCTTCCTGCTCCGCCCCTAAGTCGGGAAGGTTCCT



TGCGGTTCGCGGCGTGCCGGACGTGACAAACGGAAGCCGCACGTCTCACTAG



TACCCTCGCAGACGGACAGCGCCAGGGAGCAATGGCAGCGCGCCGACCGCG



ATGGGCTGTGGCCAATAGCGGCTGCTCAGCGGGGCGCGCCGAGAGCAGCGG



CCGGGAAGGGGCGGTGCGGGAGGCGGGGTGTGGGGCGGTAGTGTGGGCCCT



GTTCCTGCCCGCGCGGTGTTCCGCATTCTGCAAGCCTCCGGAGCGCACGTCG



GCAGTCGGCTCCCTCGTTGACCGAATCACCGACCTCTCTCCCCAG (SEQ ID



NO: 16)





SFFV
GTAACGCCATTTTGCAAGGCATGGAAAAATACCAAACCAAGAATAGAGAAG



TTCAGATCAAGGGCGGGTACATGAAAATAGCTAACGTTGGGCCAAACAGGA



TATCTGCGGTGAGCAGTTTCGGCCCCGGCCCGGGGCCAAGAACAGATGGTCA



CCGCAGTTTCGGCCCCGGCCCGAGGCCAAGAACAGATGGTCCCCAGATATGG



CCCAACCCTCAGCAGTTTCTTAAGACCCATCAGATGTTTCCAGGCTCCCCCAA



GGACCTGAAATGACCCTGCGCCTTATTTGAATTAACCAATCAGCCTGCTTCTC



GCTTCTGTTCGCGCGCTTCTGCTTCCCGAGCTCTATAAAAGAGCTCACAACCC



CTCACTCGGCGCGCCAGTCCTCCGACAGACTGAGTCGCCCGGG (SEQ ID



NO: 17)





SV40
CTGTGGAATGTGTGTCAGTTAGGGTGTGGAAAGTCCCCAGGCTCCCCAGCAG



GCAGAAGTATGCAAAGCATGCATCTCAATTAGTCAGCAACCAGGTGTGGAAA



GTCCCCAGGCTCCCCAGCAGGCAGAAGTATGCAAAGCATGCATCTCAATTAG



TCAGCAACCATAGTCCCGCCCCTAACTCCGCCCATCCCGCCCCTAACTCCGCC



CAGTTCCGCCCATTCTCCGCCCCATGGCTGACTAATTTTTTTTATTTATGCAGA



GGCCGAGGCCGCCTCTGCCTCTGAGCTATTCCAGAAGTAGTGAGGAGGCTTT



TTTGGAGGCCTAGGCTTTTGCAAAAAGCT (SEQ ID NO: 18)





UbC
GCGCCGGGTTTTGGCGCCTCCCGCGGGCGCCCCCCTCCTCACGGCGAGCGCT



GCCACGTCAGACGAAGGGCGCAGGAGCGTTCCTGATCCTTCCGCCCGGACGC



TCAGGACAGCGGCCCGCTGCTCATAAGACTCGGCCTTAGAACCCCAGTATCA



GCAGAAGGACATTTTAGGACGGGACTTGGGTGACTCTAGGGCACTGGTTTTC



TTTCCAGAGAGCGGAACAGGCGAGGAAAAGTAGTCCCTTCTCGGCGATTCTG



CGGAGGGATCTCCGTGGGGCGGTGAACGCCGATGATTATATAAGGACGCGCC



GGGTGTGGCACAGCTAGTTCCGTCGCAGCCGGGATTTGGGTCGCGGTTCTTG



TTTGTGGATCGCTGTGATCGTCACTTGGTGAGTTGCGGGCTGCTGGGCTGGCC



GGGGCTTTCGTGGCCGCCGGGCCGCTCGGTGGGACGGAAGCGTGTGGAGAG



ACCGCCAAGGGCTGTAGTCTGGGTCCGCGAGCAAGGTTGCCCTGAACTGGGG



GTTGGGGGGAGCGCACAAAATGGCGGCTGTTCCCGAGTCTTGAATGGAAGAC



GCTTGTAAGGCGGGCTGTGAGGTCGTTGAAACAAGGTGGGGGGCATGGTGG



GCGGCAAGAACCCAAGGTCTTGAGGCCTTCGCTAATGCGGGAAAGCTCTTAT



TCGGGTGAGATGGGCTGGGGCACCATCTGGGGACCCTGACGTGAAGTTTGTC



ACTGACTGGAGAACTCGGGTTTGTCGTCTGGTTGCGGGGGCGGCAGTTATGC



GGTGCCGTTGGGCAGTGCACCCGTACCTTTGGGAGCGCGCGCCTCGTCGTGT



CGTGACGTCACCCGTTCTGTTGGCTTATAATGCAGGGTGGGGCCACCTGCCG



GTAGGTGTGCGGTAGGCTTTTCTCCGTCGCAGGACGCAGGGTTCGGGCCTAG



GGTAGGCTCTCCTGAATCGACAGGCGCCGGACCTCTGGTGAGGGGAGGGATA



AGTGAGGCGTCAGTTTCTTTGGTCGGTTTTATGTACCTATCTTCTTAAGTAGC



TGAAGCTCCGGTTTTGAACTATGCGCTCGGGGTTGGCGAGTGTGTTTTGTGAA



GTTTTTTAGGCACCTTTTGAAATGTAATCATTTGGGTCAATATGTAATTTTCA



GTGTTAGACTAGTAAAGCTTCTGCAGGTCGACTCTAGAAAATTGTCCGCTAA



ATTCTGGCCGTTTTTGGCTTTTTTGTTAGAC (SEQ ID NO: 19)





hEF1aV1
GGCTCCGGTGCCCGTCAGTGGGCAGAGCGCACATCGCCCACAGTCCCCGAGA



AGTTGGGGGGAGGGGTCGGCAATTGAACCGGTGCCTAGAGAAGGTGGCGCG



GGGTAAACTGGGAAAGTGATGTCGTGTACTGGCTCCGCCTTTTTCCCGAGGG



TGGGGGAGAACCGTATATAAGTGCAGTAGTCGCCGTGAACGTTCTTTTTCGC



AACGGGTTTGCCGCCAGAACACAGGTAAGTGCCGTGTGTGGTTCCCGCGGGC



CTGGCCTCTTTACGGGTTATGGCCCTTGCGTGCCTTGAATTACTTCCACCTGG



CTGCAGTACGTGATTCTTGATCCCGAGCTTCGGGTTGGAAGTGGGTGGGAGA



GTTCGAGGCCTTGCGCTTAAGGAGCCCCTTCGCCTCGTGCTTGAGTTGAGGCC



TGGCCTGGGCGCTGGGGCCGCCGCGTGCGAATCTGGTGGCACCTTCGCGCCT



GTCTCGCTGCTTTCGATAAGTCTCTAGCCATTTAAAATTTTTGATGACCTGCT



GCGACGCTTTTTTTCTGGCAAGATAGTCTTGTAAATGCGGGCCAAGATCTGCA



CACTGGTATTTCGGTTTTTGGGGCCGCGGGCGGCGACGGGGCCCGTGCGTCC



CAGCGCACATGTTCGGCGAGGCGGGGCCTGCGAGCGCGGCCACCGAGAATC



GGACGGGGGTAGTCTCAAGCTGGCCGGCCTGCTCTGGTGCCTGGTCTCGCGC



CGCCGTGTATCGCCCCGCCCTGGGCGGCAAGGCTGGCCCGGTCGGCACCAGT



TGCGTGAGCGGAAAGATGGCCGCTTCCCGGCCCTGCTGCAGGGAGCTCAAAA



TGGAGGACGCGGCGCTCGGGAGAGCGGGCGGGTGAGTCACCCACACAAAGG



AAAAGGGCCTTTCCGTCCTCAGCCGTCGCTTCATGTGACTCCACGGAGTACC



GGGCGCCGTCCAGGCACCTCGATTAGTTCTCGAGCTTTTGGAGTACGTCGTCT



TTAGGTTGGGGGGAGGGGTTTTATGCGATGGAGTTTCCCCACACTGAGTGGG



TGGAGACTGAAGTTAGGCCAGCTTGGCACTTGATGTAATTCTCCTTGGAATTT



GCCCTTTTTGAGTTTGGATCTTGGTTCATTCTCAAGCCTCAGACAGTGGTTCA



AAGTTTTTTTCTTCCATTTCAGGTGTCGTGA (SEQ ID NO: 20)





hCAGG
ACTAGTTATTAATAGTAATCAATTACGGGGTCATTAGTTCATAGCCCATATAT



GGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGACCGCCC



AACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGC



CAATAGGGACTTTCCATTGACGTCAATGGGTGGAGTATTTACGGTAAACTGC



CCACTTGGCAGTACATCAAGTGTATCATATGCCAAGTACGCCCCCTATTGAC



GTCAATGACGGTAAATGGCCCGCCTGGCATTATGCCCAGTACATGACCTTAT



GGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCATCGCTATTACCATG



GTCGAGGTGAGCCCCACGTTCTGCTTCACTCTCCCCATCTCCCCCCCCTCCCC



ACCCCCAATTTTGTATTTATTTATTTTTTAATTATTTTGTGCAGCGATGGGGGC



GGGGGGGGGGGGGGGGCGCGCGCCAGGCGGGGCGGGGGGGGCGAGGGGC



GGGGCGGGGCGAGGCGGAGAGGTGCGGCGGCAGCCAATCAGAGCGGCGCG



CTCCGAAAGTTTCCTTTTATGGCGAGGCGGCGGCGGCGGCGGCCCTATAAAA



AGCGAAGCGCGCGGCGGGCGGGGAGTCGCTGCGACGCTGCCTTCGCCCCGT



GCCCCGCTCCGCCGCCGCCTCGCGCCGCCCGCCCCGGCTCTGACTGACCGCG



TTACTCCCACAGGTGAGCGGGCGGGACGGCCCTTCTCCTCCGGGCTGTAATT



AGCGCTTGGTTTAATGACGGCTTGTTTCTTTTCTGTGGCTGCGTGAAAGCCTT



GAGGGGCTCCGGGAGGGCCCTTTGTGCGGGGGGAGCGGCTCGGGGGGTGCG



TGCGTGTGTGTGTGCGTGGGGAGCGCCGCGTGCGGCTCCGCGCTGCCCGGCG



GCTGTGAGCGCTGCGGGCGCGGCGCGGGGCTTTGTGCGCTCCGCAGTGTGCG



CGAGGGGAGCGCGGCCGGGGGCGGTGCCCCGCGGTGCGGGGGGGGCTGCGA



GGGGAACAAAGGCTGCGTGCGGGGTGTGTGCGTGGGGGGGTGAGCAGGGGG



TGTGGGCGCGTCGGTCGGGCTGCAACCCCCCCTGCACCCCCCTCCCCGAGTT



GCTGAGCACGGCCCGGCTTCGGGTGCGGGGCTCCGTACGGGGCGTGGCGCG



GGGCTCGCCGTGCCGGGCGGGGGGTGGCGGCAGGTGGGGGTGCCGGGCGGG



GCGGGGCCGCCTCGGGCCGGGGAGGGCTCGGGGGAGGGGCGCGGCGGCCCC



CGGAGCGCCGGCGGCTGTCGAGGCGCGGCGAGCCGCAGCCATTGCCTTTTAT



GGTAATCGTGCGAGAGGGCGCAGGGACTTCCTTTGTCCCAAATCTGTGCGGA



GCCGAAATCTGGGAGGCGCCGCCGCACCCCCTCTAGCGGGCGCGGGGCGAA



GCGGTGCGGCGCCGGCAGGAAGGAAATGGGCGGGGAGGGCCTTCGTGCGTC



GCCGCGCCGCCGTCCCCTTCTCCCTCTCCAGCCTCGGGGCTGTCCGCGGGGG



GACGGCTGCCTTCGGGGGGGACGGGGCAGGGCGGGGTTCGGCTTCTGGCGTG



TGACCGGCGGCTCTAGAGCCTCTGCTAACCATGTTCATGCCTTCTTCTTTTTCC



TACAGCTCCTGGGCAACGTGCTGGTTATTGTGCTGTCTCATCATTTTGGCAAA



GAATTC (SEQ ID NO: 21)





hEF1aV2
Gggcagagcgcacatcgcccacagtccccgagaagttggggggaggggtcgg



caattgaaccggtgcctagagaaggtggcgcggggtaaactgggaaagtgat



gtcgtgtactggctccgcctttttcccgaggggggggagaaccgtatataag



tgcagtagtcgccgtgaacgttctttttcgcaacgggtttgccgccagaaca



cag (SEQ ID NO: 22)





hACTb
CCACTAGTTCCATGTCCTTATATGGACTCATCTTTGCCTATTGCGACACACAC



TCAATGAACACCTACTACGCGCTGCAAAGAGCCCCGCAGGCCTGAGGTGCCC



CCACCTCACCACTCTTCCTATTTTTGTGTAAAAATCCAGCTTCTTGTCACCACC



TCCAAGGAGGGGGAGGAGGAGGAAGGCAGGTTCCTCTAGGCTGAGCCGAAT



GCCCCTCTGTGGTCCCACGCCACTGATCGCTGCATGCCCACCACCTGGGTAC



ACACAGTCTGTGATTCCCGGAGCAGAACGGACCCTGCCCACCCGGTCTTGTG



TGCTACTCAGTGGACAGACCCAAGGCAAGAAAGGGTGACAAGGACAGGGTC



TTCCCAGGCTGGCTTTGAGTTCCTAGCACCGCCCCGCCCCCAATCCTCTGTGG



CACATGGAGTCTTGGTCCCCAGAGTCCCCCAGCGGCCTCCAGATGGTCTGGG



AGGGCAGTTCAGCTGTGGCTGCGCATAGCAGACATACAACGGACGGTGGGC



CCAGACCCAGGCTGTGTAGACCCAGCCCCCCCGCCCCGCAGTGCCTAGGTCA



CCCACTAACGCCCCAGGCCTGGTCTTGGCTGGGCGTGACTGTTACCCTCAAA



AGCAGGCAGCTCCAGGGTAAAAGGTGCCCTGCCCTGTAGAGCCCACCTTCCT



TCCCAGGGCTGCGGCTGGGTAGGTTTGTAGCCTTCATCACGGGCCACCTCCA



GCCACTGGACCGCTGGCCCCTGCCCTGTCCTGGGGAGTGTGGTCCTGCGACT



TCTAAGTGGCCGCAAGCCACCTGACTCCCCCAACACCACACTCTACCTCTCA



AGCCCAGGTCTCTCCCTAGTGACCCACCCAGCACATTTAGCTAGCTGAGCCC



CACAGCCAGAGGTCCTCAGGCCCTGCTTTCAGGGCAGTTGCTCTGAAGTCGG



CAAGGGGGAGTGACTGCCTGGCCACTCCATGCCCTCCAAGAGCTCCTTCTGC



AGGAGCGTACAGAACCCAGGGCCCTGGCACCCGTGCAGACCCTGGCCCACC



CCACCTGGGCGCTCAGTGCCCAAGAGATGTCCACACCTAGGATGTCCCGCGG



TGGGTGGGGGGCCCGAGAGACGGGCAGGCCGGGGGCAGGCCTGGCCATGCG



GGGCCGAACCGGGCACTGCCCAGCGTGGGGCGCGGGGGCCACGGCGCGCGC



CCCCAGCCCCCGGGCCCAGCACCCCAAGGCGGCCAACGCCAAAACTCTCCCT



CCTCCTCTTCCTCAATCTCGCTCTCGCTCTTTTTTTTTTTCGCAAAAGGAGGGG



AGAGGGGGTAAAAAAATGCTGCACTGTGCGGCGAAGCCGGTGAGTGAGCGG



CGCGGGGCCAATCAGCGTGCGCCGTTCCGAAAGTTGCCTTTTATGGCTCGAG



CGGCCGCGGCGGCGCCCTATAAAACCCAGCGGCGCGACGCGCCACCACCGC



CGAGACCGCGTCCGCCCCGCGAGCACAGAGCCTCGCCTTTGCCGATCCGCCG



CCCGTCCACACCCGCCGCCAGgtaagcccggccagccgaccggggcagggg



ctcacggcccggccgcaggcggccgcggccccttcgcccgtgcagagccgc



cgtctgggccgcagcggggggcgcatggggggggaaccggaccgccgtggg



gggcgcgggagaagcccctgggcctccggagatgggggacaccccacgcca



gttcggaggcgcgaggccgcgctcgggaggcgcgctccgggggtgccgctc



tcgggggggggcaaccggcggggtctttgtctgagccgggctcttgccaat



ggggatcgcagggtgggcgcggcggagcccccgccaggcccggtgggggct



ggggcgccattgcgcgtgcgcgctggtcctttgggcgctaactgcgtgcgc



gctgggaattggcgctaattgcgcgtgcgcgctgggactcaaggcgctaac



tgcgcgtgcgttctggggcccggggtgccgcggcctgggctggggcgaagg



cgggctcggccggaaggggggggtcgccgcggctcccgggcgcttgcgcgc



acttcctgcccgagccgctggccgcccgagggtgtggccgctgcgtgcgcg



cgcgccgacccggcgctgtttgaaccgggcggaggcggggctggcgcccgg



ttgggagggggttggggcctggcttcctgccgcgcgccgcggggacgcctc



cgaccagtgtttgccttttatggtaataacgcggccggcccggcttccttt



gtccccaatctgggcgcgcgccggcgccccctggcggcctaaggactcggc



gcgccggaagtggccagggcgggggcgacctcggctcacagcgcgcccggc



tat (SEQ ID NO: 23)





heIF4A1
GTTGATTTCCTTCATCCCTGGCACACGTCCAGGCAGTGTCGAATCCATCTCTG



CTACAGGGGAAAACAAATAACATTTGAGTCCAGTGGAGACCGGGAGCAGAA



GTAAAGGGAAGTGATAACCCCCAGAGCCCGGAAGCCTCTGGAGGCTGAGAC



CTCGCCCCCCTTGCGTGATAGGGCCTACGGAGCCACATGACCAAGGCACTGT



CGCCTCCGCACGTGTGAGAGTGCAGGGCCCCAAGATGGCTGCCAGGCCTCGA



GGCCTGACTCTTCTATGTCACTTCCGTACCGGCGAGAAAGGCGGGCCCTCCA



GCCAATGAGGCTGCGGGGCGGGCCTTCACCTTGATAGGCACTCGAGTTATCC



AATGGTGCCTGCGGGCCGGAGCGACTAGGAACTAACGTCATGCCGAGTTGCT



GAGCGCCGGCAGGCGGGGCCGGGGCGGCCAAACCAATGCGATGGCCGGGGC



GGAGTCGGGCGCTCTATAAGTTGTCGATAGGCGGGCACTCCGCCCTAGTTTC



TAAGGACCATG (SEQ ID NO: 24)





hGAPDH
AGTTCCCCAACTTTCCCGCCTCTCAGCCTTTGAAAGAAAGAAAGGGGGGGG



GCAGGCCGCGTGCAGTCGCGAGCGGTGCTGGGCTCCGGCTCCAATTCCCCAT



CTCAGTCGCTCCCAAAGTCCTTCTGTTTCATCCAAGCGTGTAAGGGTCCCCGT



CCTTGACTCCCTAGTGTCCTGCTGCCCACAGTCCAGTCCTGGGAACCAGCACC



GATCACCTCCCATCGGGCCAATCTCAGTCCCTTCCCCCCTACGTCGGGGCCCA



CACGCTCGGTGCGTGCCCAGTTGAACCAGGCGGCTGCGGAAAAAAAAAAGC



GGGGAGAAAGTAGGGCCCGGCTACTAGCGGTTTTACGGGCGCACGTAGCTCA



GGCCTCAAGACCTTGGGCTGGGACTGGCTGAGCCTGGCGGGAGGCGGGGTCC



GAGTCACCGCCTGCCGCCGCGCCCCCGGTTTCTATAAATTGAGCCCGCAGCC



TCCCGCTTCGCTCTCTGCTCCTCCTGTTCGACAGTCAGCCGCATCTTCTTTTGC



GTCGCCAGgtgaagacgggcggagagaaacccgggaggctagggacggcct



gaaggcggcaggggcgggcgcaggccggatgtgttcgcgccgctgcggggt



gggcccgggcggcctccgcattgcaggggcgggcggaggacgtgatgcggc



gcgggctgggcatggaggcctggtgggggaggggaggggaggcgtgggtgt



cggccggggccactaggcgctcactgttctctccctccgcgcagCCGAGCC



ACATCGCTGAGACAC (SEQ ID NO: 25)





hGRP78
AGTGCGGTTACCAGCGGAAATGCCTCGGGGTCAGAAGTCGCAGGAGAGATA



GACAGCTGCTGAACCAATGGGACCAGCGGATGGGGCGGATGTTATCTACCAT



TGGTGAACGTTAGAAACGAATAGCAGCCAATGAATCAGCTGGGGGGGCGGA



GCAGTGACGTTTATTGCGGAGGGGGCCGCTTCGAATCGGCGGCGGCCAGCTT



GGTGGCCTGGGCCAATGAACGGCCTCCAACGAGCAGGGCCTTCACCAATCGG



CGGCCTCCACGACGGGGCTGGGGGAGGGTATATAAGCCGAGTAGGCGACGG



TGAGGTCGACGCCGGCCAAGACAGCACAGACAGATTGACCTATTGGGGTGTT



TCGCGAGTGTGAGAGGGAAGCGCCGCGGCCTGTATTTCTAGACCTGCCCTTC



GCCTGGTTCGTGGCGCCTTGTGACCCCGGGCCCCTGCCGCCTGCAAGTCGGA



AATTGCGCTGTGCTCCTGTGCTACGGCCTGTGGCTGGACTGCCTGCTGCTGCC



CAACTGGCTGGCAC (SEQ ID NO: 26)





hGRP94
TAGTTTCATCACCACCGCCACCCCCCCGCCCCCCCGCCATCTGAAAGGGTTCT



AGGGGATTTGCAACCTCTCTCGTGTGTTTCTTCTTTCCGAGAAGCGCCGCCAC



ACGAGAAAGCTGGCCGCGAAAGTCGTGCTGGAATCACTTCCAACGAAACCC



CAGGCATAGATGGGAAAGGGTGAAGAACACGTTGCCATGGCTACCGTTTCCC



CGGTCACGGAATAAACGCTCTCTAGGATCCGGAAGTAGTTCCGCCGCGACCT



CTCTAAAAGGATGGATGTGTTCTCTGCTTACATTCATTGGACGTTTTCCCTTA



GAGGCCAAGGCCGCCCAGGCAAAGGGGCGGTCCCACGCGTGAGGGGCCCGC



GGAGCCATTTGATTGGAGAAAAGCTGCAAACCCTGACCAATCGGAAGGAGC



CACGCTTCGGGCATCGGTCACCGCACCTGGACAGCTCCGATTGGTGGACTTC



CGCCCCCCCTCACGAATCCTCATTGGGTGCCGTGGGTGCGTGGTGCGGCGCG



ATTGGTGGGTTCATGTTTCCCGTCCCCCGCCCGCGAGAAGTGGGGGTGAAAA



GCGGCCCGACCTGCTTGGGGTGTAGTGGGCGGACCGCGCGGCTGGAGGTGTG



AGGATCCGAACCCAGGGGTGGGGGGTGGAGGCGGCTCCTGCGATCGAAGGG



GACTTGAGACTCACCGGCCGCACGTC (SEQ ID NO: 27)





hHSP70
GGGCCGCCCACTCCCCCTTCCTCTCAGGGTCCCTGTCCCCTCCAGTGAATCCC



AGAAGACTCTGGAGAGTTCTGAGCAGGGGGCGGCACTCTGGCCTCTGATTGG



TCCAAGGAAGGCTGGGGGGCAGGACGGGAGGCGAAAACCCTGGAATATTCC



CGACCTGGCAGCCTCATCGAGCTCGGTGATTGGCTCAGAAGGGAAAAGGCG



GGTCTCCGTGACGACTTATAAAAGCCCAGGGGCAAGCGGTCCGGATAACGGC



TAGCCTGAGGAGCTGCTGCGACAGTCCACTACCTTTTTCGAGAGTGACTCCC



GTTGTCCCAAGGCTTCCCAGAGCGAACCTGTGCGGCTGCAGGCACCGGCGCG



TCGAGTTTCCGGCGTCCGGAAGGACCGAGCTCTTCTCGCGGATCCAGTGTTCC



GTTTCCAGCCCCCAATCTCAGAGCGGAGCCGACAGAGAGCAGGGAACCC



(SEQ ID NO: 28)





hKINb
GCCCCACCCCCGTCCGCGTTACAACCGGGAGGCCCGCTGGGTCCTGCACCGT



CACCCTCCTCCCTGTGACCGCCCACCTGATACCCAAACAACTTTCTCGCCCCT



CCAGTCCCCAGCTCGCCGAGCGCTTGCGGGGAGCCACCCAGCCTCAGTTTCC



CCAGCCCCGGGCGGGGCGAGGGGCGATGACGTCATGCCGGCGCGCGGCATT



GTGGGGCGGGGCGAGGCGGGGCGCCGGGGGGAGCAACACTGAGACGCCATT



TTCGGCGGCGGGAGCGGCGCAGGCGGCCGAGCGGGACTGGCTGGGTCGGCT



GGGCTGCTGGTGCGAGGAGCCGCGGGGCTGTGCTCGGCGGCCAAGGGGACA



GCGCGTGGGTGGCCGAGGATGCTGCGGGGCGGTAGCTCCGGCGCCCCTCGCT



GGTGACTGCTGCGCCGTGCCTCACACAGCCGAGGCGGGCTCGGCGCACAGTC



GCTGCTCCGCGCTCGCGCCCGGCGGCGCTCCAGGTGCTGACAGCGCGAGAGA



GCGCGGCCTCAGGAGCAACAC (SEQ ID NO: 29)





hUBIb
TTCCAGAGCTTTCGAGGAAGGTTTCTTCAACTCAAATTCATCCGCCTGATAAT



TTTCTTATATTTTCCTAAAGAAGGAAGAGAAGCGCATAGAGGAGAAGGGAA



ATAATTTTTTAGGAGCCTTTCTTACGGCTATGAGGAATTTGGGGCTCAGTTGA



AAAGCCTAAACTGCCTCTCGGGAGGTTGGGCGCGGCGAACTACTTTCAGCGG



CGCACGGAGACGGCGTCTACGTGAGGGGTGATAAGTGACGCAACACTCGTTG



CATAAATTTGCGCTCCGCCAGCCCGGAGCATTTAGGGGCGGTTGGCTTTGTTG



GGTGAGCTTGTTTGTGTCCCTGTGGGTGGACGTGGTTGGTGATTGGCAGGATC



CTGGTATCCGCTAACAGgtactggcccacagccgtaaagacctgcgggggc



gtgagaggggggaatgggtgaggtcaagctggaggcttcttggggttgggg



ggccgctgaggggaggggagggcgaggtgacgcgacacccggcctttctgg



gagagtgggccttgttgacctaaggggggcgagggcagttggcacgcgcac



gcgccgacagaaactaacagacattaaccaacagcgattccgtcgcgttta



cttgggaggaaggcggaaaagaggtagtttgtgtggcttctggaaacccta



aatttggaatcccagtatgagaatggtgtcccttcttgtgtttcaatggga



tttttacttcgcgagtcttgtgggtttggttttgttttcagtttgcctaac



accgtgcttaggtttgaggcagattggagttcggtcgggggagtttgaata



tccggaacagttagtggggaaagctgtggacgcttggtaagagagcgctct



ggattttccgctgttgacgttgaaaccttgaatgacgaatttcgtattaag



tgacttagccttgtaaaattgaggggaggcttgcggaatattaacgtattt



aaggcattttgaaggaatagttgctaattttgaagaatattaggtgtaaaa



gcaagaaatacaatgatcctgaggtgacacgcttatgttttacttttaaac



tagGTCACC (SEQ ID NO: 30)





CAG
gacattgattattgactagttattaatagtaatcaattacggggtcattag



ttcatagcccatatatggagttccgcgttacataacttacggtaaatggcc



cgcctggctgaccgcccaacgacccccgcccattgacgtcaataatgacgt



atgttcccatagtaacgccaatagggactttccattgacgtcaatgggtgg



agtatttacggtaaactgcccacttggcagtacatcaagtgtatcatatgc



caagtacgccccctattgacgtcaatgacggtaaatggcccgcctggcatt



atgcccagtacatgaccttatgggactttcctacttggcagtacatctacg



tattagtcatcgctattaccatggtcgaggtgagccccacgttctgcttca



ctctccccatctcccccccctccccacccccaattttgtatttatttattt



tttaattattttgtgcagcgatgggggcggggggggggggggggcgcgcgc



caggcggggggggcggggcgaggggggggcggggcgaggcggagaggtgcg



gcggcagccaatcagagcggcgcgctccgaaagtttccttttatggcgagg



cggggcggcggcggccctataaaaagcgaagcgcgcgggggcg (SEQ ID



NO: 173)





HLP
Tgtttgctgcttgcaatgtttgcccattttagggtggacacaggacgctgt



ggtttctgagccagggggcgactcagatcccagccagtggacttagcccct



gtttgctcctccgataactggggtgaccttggttaatattcaccagcagcc



tcccccgttgcccctctggatccactgcttaaatacggacgaggacagggc



cctgtctcctcagcttcaggcaccaccactgacctgggacagtgaat 



(SEQ ID NO: 174)









The promoter can be a tissue-specific promoter. In general, a tissue-specific promoter directs transcription of a nucleic acid, (e.g., the engineered nucleic acids encoding the chimeric proteins, such as membrane-cleavable chimeric proteins having the formula S-C-MT-D) such that expression is limited to a specific cell type, organelle, or tissue. Tissue specific promoters include, but are not limited to, albumin (liver specific, Pinkert et al., (1987)), lymphoid specific promoters (Calame and Eaton, 1988), particular promoters of T-cell receptors (Winoto and Baltimore, (1989)) and immunoglobulins; Banerji et al., (1983); Queen and Baltimore, 1983), neuron specific promoters (e.g. the neurofilament promoter; Byrne and Ruddle, 1989), pancreas specific promoters (Edlund et al., (1985)) or mammary gland specific promoters (milk whey promoter, U.S. Pat. No. 4,873,316 and European Application Publication No. 264,166) as well as developmentally regulated promoters such as the murine hox promoters (Kessel and Gruss, Science 249:374-379 (1990)) or the α-fetoprotein promoter (Campes and Tilghman, Genes Dev. 3:537-546 (1989)), the contents of each of which are fully incorporated by reference herein. The promoter can be constitutive in the respective specific cell type, organelle, or tissue. Tissue-specific promoters and/or regulatory elements can also include promoters from the liver fatty acid binding (FAB) protein gene, specific for colon epithelial cells; the insulin gene, specific for pancreatic cells; the transphyretin, .alpha.1-antitrypsin, plasminogen activator inhibitor type 1 (PAI-I), apolipoprotein A1 and LDL receptor genes, specific for liver cells; the myelin basic protein (MBP) gene, specific for oligodendrocytes; the glial fibrillary acidic protein (GFAP) gene, specific for glial cells; OPSIN, specific for targeting to the eye; and the neural-specific enolase (NSE) promoter that is specific for nerve cells. Examples of tissue-specific promoters include, but are not limited to, the promoter for creatine kinase, which has been used to direct expression in muscle and cardiac tissue and immunoglobulin heavy or light chain promoters for expression in B cells. Other tissue specific promoters include the human smooth muscle alpha-actin promoter. Exemplary tissue-specific expression elements for the liver include but are not limited to HMG-COA reductase promoter, sterol regulatory element 1, phosphoenol pyruvate carboxy kinase (PEPCK) promoter, human C-reactive protein (CRP) promoter, human glucokinase promoter, cholesterol L 7-alpha hydroylase (CYP-7) promoter, beta-galactosidase alpha-2,6 sialylkansferase promoter, insulin-like growth factor binding protein (IGFBP-I) promoter, aldolase B promoter, human transferrin promoter, and collagen type I promoter. Exemplary tissue-specific expression elements for the prostate include but are not limited to the prostatic acid phosphatase (PAP) promoter, prostatic secretory protein of 94 (PSP 94) promoter, prostate specific antigen complex promoter, and human glandular kallikrein gene promoter (hgt-1). Exemplary tissue-specific expression elements for gastric tissue include but are not limited to the human H+/K+-ATPase alpha subunit promoter. Exemplary tissue-specific expression elements for the pancreas include but are not limited to pancreatitis associated protein promoter (PAP), elastase 1 transcriptional enhancer, pancreas specific amylase and elastase enhancer promoter, and pancreatic cholesterol esterase gene promoter. Exemplary tissue-specific expression elements for the endometrium include, but are not limited to, the uteroglobin promoter. Exemplary tissue-specific expression elements for adrenal cells include, but are not limited to, cholesterol side-chain cleavage (SCC) promoter. Exemplary tissue-specific expression elements for the general nervous system include, but are not limited to, gamma-gamma enolase (neuron-specific enolase, NSE) promoter. Exemplary tissue-specific expression elements for the brain include, but are not limited to, the neurofilament heavy chain (NF-H) promoter. Exemplary tissue-specific expression elements for lymphocytes include, but are not limited to, the human CGL-1/granzyme B promoter, the terminal deoxy transferase (TdT), lambda 5, VpreB, and lck (lymphocyte specific tyrosine protein kinase p561ck) promoter, the humans CD2 promoter and its 3′transcriptional enhancer, and the human NK and T cell specific activation (NKG5) promoter. Exemplary tissue-specific expression elements for the colon include, but are not limited to, pp60c-src tyrosine kinase promoter, organ-specific neoantigens (OSNs) promoter, and colon specific antigen-P promoter. Tissue-specific expression elements for breast cells are for example, but are not limited to, the human alpha-lactalbumin promoter. Exemplary tissue-specific expression elements for the lung include, but are not limited to, the cystic fibrosis transmembrane conductance regulator (CFTR) gene promoter.


In some embodiments, a promoter of the present disclosure is modulated by signals within a tumor microenvironment. A tumor microenvironment is considered to modulate a promoter if, in the presence of the tumor microenvironment, the activity of the promoter is increased or decreased by at least 10%, relative to activity of the promoter in the absence of the tumor microenvironment. In some embodiments, the activity of the promoter is increased or decreased by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, relative to activity of the promoter in the absence of the tumor microenvironment. For example, the activity of the promoter is increased or decreased by 10-20%, 10-30%, 10-40%, 10-50%, 10-60%, 10-70%, 10-80%, 10-90%, 10-100%, 10-200%, 20-30%, 20-40%, 20-50%, 20-60%, 20-70%, 20-80%, 20-90%, 20-100%, 20-200%, 50-60%, 50-70%, 50-80%, 50-90%, 50-100%, or 50-200%, relative to activity of the promoter in the absence of the tumor microenvironment.


In some embodiments, the activity of the promoter is increased or decreased by at least 2 fold (e.g., 2, 3, 4, 5, 10, 25, 20, 25, 50, or 100 fold), relative to activity of the promoter in the absence of the tumor microenvironment. For example, the activity of the promoter is increased or decreased by at least 3 fold, at least 5 fold, at least 10 fold, at least 20 fold, at least 50 fold, or at least 100 fold, relative to activity of the promoter in the absence of the tumor microenvironment. In some embodiments, the activity of the promoter is increased or decreased by 2-10, 2-20, 2-30, 2-40, 2-50, 2-60, 2-70, 2-80, 2-90, or 2-100 fold, relative to activity of the promoter in the absence of the tumor microenvironment.


In some embodiments, a promoter of the present disclosure is activated under a hypoxic condition. A “hypoxic condition” is a condition where the body or a region of the body is deprived of adequate oxygen supply at the tissue level. Hypoxic conditions can cause inflammation (e.g., the level of inflammatory cytokines increase under hypoxic conditions). In some embodiments, the promoter that is activated under hypoxic condition is operably linked to a nucleotide encoding a chimeric proteins that decreases the expression of activity of inflammatory cytokines, thus reducing the inflammation caused by the hypoxic condition. In some embodiments, the promoter that is activated under hypoxic conditions comprises a hypoxia responsive element (HRE). A “hypoxia responsive element (HRE)” is a response element that responds to hypoxia-inducible factor (HIF). The HRE, in some embodiments, comprises a consensus motif NCGTG (where N is either A or G).


Multicistronic and Multiple Promoter Systems

In some embodiments, engineered nucleic acids (e.g., an engineered nucleic acid comprising an expression cassette) are configured to produce multiple chimeric proteins. For example, nucleic acids may be configured to produce 2-20 different chimeric proteins. In some embodiments, nucleic acids are configured to produce 2-20, 2-19, 2-18, 2-17, 2-16, 2-15, 2-14, 2-13, 2-12, 2-11, 2-10, 2-9, 2-8, 2-7, 2-6, 2-5, 2-4, 2-3, 3-20, 3-19, 3-18, 3-17, 3-16, 3-15, 3-14, 3-13, 3-12, 3-11, 3-10, 3-9, 3-8, 3-7, 3-6, 3-5, 3-4, 4-20, 4-19, 4-18, 4-17, 4-16, 4-15, 4-14, 4-13, 4-12, 4-11, 4-10, 4-9, 4-8, 4-7, 4-6, 4-5, 5-20, 5-19, 5-18, 5-17, 5-16, 5-15, 5-14, 5-13, 5-12, 5-11, 5-10, 5-9, 5-8, 5-7, 5-6, 6-20, 6-19, 6-18, 6-17, 6-16, 6-15, 6-14, 6-13, 6-12, 6-11, 6-10, 6-9, 6-8, 6-7, 7-20, 7-19, 7-18, 7-17, 7-16, 7-15, 7-14, 7-13, 7-12, 7-11, 7-10, 7-9, 7-8, 8-20, 8-19, 8-18, 8-17, 8-16, 8-15, 8-14, 8-13, 8-12, 8-11, 8-10, 8-9, 9-20, 9-19, 9-18, 9-17, 9-16, 9-15, 9-14, 9-13, 9-12, 9-11, 9-10, 10-20, 10-19, 10-18, 10-17, 10-16, 10-15, 10-14, 10-13, 10-12, 10-11, 11-20, 11-19, 11-18, 11-17, 11-16, 11-15, 11-14, 11-13, 11-12, 12-20, 12-19, 12-18, 12-17, 12-16, 12-15, 12-14, 12-13, 13-20, 13-19, 13-18, 13-17, 13-16, 13-15, 13-14, 14-20, 14-19, 14-18, 14-17, 14-16, 14-15, 15-20, 15-19, 15-18, 15-17, 15-16, 16-20, 16-19, 16-18, 16-17, 17-20, 17-19, 17-18, 18-20, 18-19, or 19-20 chimeric proteins. In some embodiments, nucleic acids are configured to produce 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 chimeric proteins.


In some embodiments, engineered nucleic acids can be multicistronic, i.e., more than one separate polypeptide (e.g., multiple chimeric proteins) can be produced from a single mRNA transcript. Engineered nucleic acids can be multicistronic through the use of various linkers, e.g., a polynucleotide sequence encoding a first chimeric proteins can be linked to a nucleotide sequence encoding a second chimeric protein, such as in a first gene:linker:second gene 5′ to 3′ orientation. A linker can encode a 2A ribosome skipping element, such as T2A. Other 2A ribosome skipping elements include, but are not limited to, E2A, P2A, and F2A. 2A ribosome skipping elements allow production of separate polypeptides encoded by the first and second genes are produced during translation. A linker can encode a cleavable linker polypeptide sequence, such as a Furin cleavage site or a TEV cleavage site, wherein following expression the cleavable linker polypeptide is cleaved such that separate polypeptides encoded by the first and second genes are produced. A cleavable linker can include a polypeptide sequence, such as such a flexible linker (e.g., a Gly-Ser-Gly sequence), that further promotes cleavage.


A linker can encode an Internal Ribosome Entry Site (IRES), such that separate polypeptides encoded by the first and second genes are produced during translation. A linker can encode a splice acceptor, such as a viral splice acceptor.


A linker can be a combination of linkers, such as a Furin-2A linker that can produce separate polypeptides through 2A ribosome skipping followed by further cleavage of the Furin site to allow for complete removal of 2A residues. In some embodiments, a combination of linkers can include a Furin sequence, a flexible linker, and 2A linker. Accordingly, in some embodiments, the linker is a Furin-Gly-Ser-Gly-2A fusion polypeptide. In some embodiments, a linker of the present disclosure is a Furin-Gly-Ser-Gly-T2A fusion polypeptide.


In general, a multicistronic system can use any number or combination of linkers, to express any number of genes or portions thereof (e.g., an engineered nucleic acid can encode a first, a second, and a third chimeric protein, each separated by linkers such that separate polypeptides encoded by the first, second, and third chimeric proteins are produced).


Engineered nucleic acids can use multiple promoters to express genes from multiple ORFs, i.e., more than one separate mRNA transcript can be produced from a single engineered nucleic acid. For example, a first promoter can be operably linked to a polynucleotide sequence encoding a first chimeric protein, and a second promoter can be operably linked to a polynucleotide sequence encoding a second chimeric protein. In general, any number of promoters can be used to express any number of chimeric proteins. In some embodiments, at least one of the ORFs expressed from the multiple promoters can be multicistronic.


“Linkers,” as used herein can refer to polypeptides that link a first polypeptide sequence and a second polypeptide sequence, the multicistronic linkers described above, or the additional promoters that are operably linked to additional ORFs described above.


Engineered Cells


Provided herein are engineered cells, and methods of producing the engineered cells, that produce chimeric proteins that modulate different tumor-mediated immunosuppressive mechanisms. In general, engineered cells of the present disclosure may be engineered to express the chimeric proteins provided for herein, such as the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein. These cells are referred to herein as “engineered cells.” These cells, which typically contain engineered nucleic acid, do not occur in nature. In some embodiments, the cells are engineered to include a nucleic acid comprising a promoter operably linked to a nucleotide sequence encoding a chimeric protein, for example, one that stimulates an immune response. An engineered cell can comprise an engineered nucleic acid integrated into the cell's genome. An engineered cell can comprise an engineered nucleic acid capable of expression without integrating into the cell's genome, for example, engineered with a transient expression system such as a plasmid or mRNA.


The present disclosure also encompasses additivity and synergy between a chimeric protein(s) and the engineered cell from which they are produced. In some embodiments, cells are engineered to produce at least two (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) chimeric proteins, each of which modulates a different tumor-mediated immunosuppressive mechanism. In other embodiments, cells are engineered to produce at least one chimeric proteins having an effector molecule that is not natively produced by the cells. Such an effector molecule may, for example, complement the function of effector molecules natively produced by the cells.


In some embodiments, cells are engineered to express membrane-tethered anti-CD3 and/or anti-CD28 agonist extracellular domains.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce multiple chimeric proteins. For example, cells may be engineered to produce 2-20 different chimeric proteins. In some embodiments, cells engineered to produce 2-20, 2-19, 2-18, 2-17, 2-16, 2-15, 2-14, 2-13, 2-12, 2-11, 2-10, 2-9, 2-8, 2-7, 2-6, 2-5, 2-4, 2-3, 3-20, 3-19, 3-18, 3-17, 3-16, 3-15, 3-14, 3-13, 3-12, 3-11, 3-10, 3-9, 3-8, 3-7, 3-6, 3-5, 3-4,4-20,4-19, 4-18, 4-17, 4-16, 4-15, 4-14, 4-13, 4-12, 4-11, 4-10, 4-9, 4-8, 4-7, 4-6, 4-5, 5-20, 5-19,5-18,5-17, 5-16, 5-15, 5-14, 5-13, 5-12, 5-11, 5-10, 5-9, 5-8, 5-7, 5-6, 6-20, 6-19, 6-18, 6-17, 6-16, 6-15,6-14, 6-13,6-12,6-11,6-10, 6-9, 6-8, 6-7,7-20,7-19,7-18,7-17,7-16,7-15,7-14,7-13, 7-12, 7-11, 7-10, 7-9, 7-8, 8-20, 8-19, 8-18, 8-17, 8-16, 8-15, 8-14, 8-13, 8-12, 8-11, 8-10, 8-9, 9-20, 9-19, 9-18, 9-17, 9-16, 9-15, 9-14, 9-13, 9-12, 9-11, 9-10, 10-20, 10-19, 10-18, 10-17, 10-16, 10-15, 10-14, 10-13, 10-12, 10-11, 11-20, 11-19, 11-18, 11-17, 11-16, 11-15, 11-14, 11-13, 11-12, 12-20, 12-19, 12-18, 12-17, 12-16, 12-15, 12-14, 12-13, 13-20, 13-19, 13-18, 13-17, 13-16, 13-15, 13-14, 14-20, 14-19, 14-18, 14-17, 14-16, 14-15, 15-20, 15-19, 15-18, 15-17, 15-16, 16-20, 16-19, 16-18, 16-17, 17-20, 17-19, 17-18, 18-20, 18-19, or 19-20 chimeric proteins. In some embodiments, cells are engineered to produce 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 chimeric proteins.


In some embodiments, engineered cells comprise one or more engineered nucleic acids encoding a promoter operably linked to a nucleotide sequence encoding a chimeric protein. In some embodiments, cells are engineered to include a plurality of engineered nucleic acids, e.g., at least two engineered nucleic acids, each encoding a promoter operably linked to a nucleotide sequence encoding at least one (e.g., 1, 2 or 3) chimeric protein. For example, cells may be engineered to comprise at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 8, at least 9, or at least 10, engineered nucleic acids, each encoding a promoter operably linked to a nucleotide sequence encoding at least one (e.g., 1, 2 or 3) chimeric protein. In some embodiments, the cells are engineered to comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, or more engineered nucleic acids, each encoding a promoter operably linked to a nucleotide sequence encoding at least one (e.g., 1, 2 or 3) chimeric protein. Engineered cells can comprise an engineered nucleic acid encoding at least one of the linkers described above, such as polypeptides that link a first polypeptide sequence and a second polypeptide sequence, one or more multicistronic linker described above, one or more additional promoters operably linked to additional ORFs, or a combination thereof.


Engineered cells of the present disclosure (e.g., an immune cell or a stem cell) typically produce multiple chimeric proteins, at least two of which modulate different tumor-mediated immunosuppressive mechanisms. In some embodiments, at least one of the chimeric proteins stimulates an inflammatory pathway in the tumor microenvironment, and at least one of the chimeric proteins inhibits a negative regulator of inflammation in the tumor microenvironment.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce at least one homing molecule. “Homing,” refers to active navigation (migration) of a cell to a target site (e.g., a cell, tissue (e.g., tumor), or organ). A “homing molecule” refers to a molecule that directs cells to a target site. In some embodiments, a homing molecule functions to recognize and/or initiate interaction of an engineered cell to a target site. Non-limiting examples of homing molecules include CXCR1, CCR9, CXCR2, CXCR3, CXCR4, CCR2, CCR4, FPR2, VEGFR, IL6R, CXCR1, CSCR7, and PDGFR.


In some embodiments, a homing molecule is a chemokine receptor (cell surface molecule that binds to a chemokine). Non-limiting examples of chemokine receptors that may be produced by the engineered cells of the present disclosure include: CXC chemokine receptors (e.g., CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, and CXCR7), CC chemokine receptors (CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, and CCR11), CX3C chemokine receptors (e.g., CX3CR1, which binds to CX3CL1), and XC chemokine receptors (e.g., XCR1). In some embodiments, a chemokine receptor is a G protein-linked transmembrane receptor, or a member of the tumor necrosis factor (TNF) receptor superfamily (including but not limited to TNFRSF1A, TNFRSF1B). In some embodiments, cells are engineered to produce CXCL8, CXCL9, and/or CXCL10 (promote T-cell recruitment), CCL3 and/or CXCL5, CCL21 (Th1 recruitment and polarization). In some embodiments, cells are engineered to produce CXCR4.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce G-protein coupled receptors (GPCRs) that detect N-formylated-containing oligopeptides (including but not limited to FPR2 and FPRL1).


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce receptors that detect interleukins (including but not limited to IL6R).


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce receptors that detect growth factors secreted from other cells, tissues, or tumors (including but not limited to FGFR, PDGFR, EGFR, and receptors of the VEGF family, including but not limited to VEGF-C and VEGF-D).


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce one or more integrins. Cells of the present disclosure may be engineered to produce any combination of integrin a and R subunits. The a subunit of an integrin may be, without limitation: ITGA1, ITGA2, ITGA3, ITGA4, ITGA5, ITGA6, IGTA7, ITGA8, ITGA9, IGTA10, IGTA11, ITGAD, ITGAE, ITGAL, ITGAM, ITGAV, ITGA2B, ITGAX. The R subunit of an integrin may be, without limitation: ITGB1, ITGB2, ITGB3, ITGB4, ITGB5, ITGB6, ITGB7, and ITGB8.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce one or more matrix metalloproteinases (MMP). Non-limiting examples of MMPs include MMP-2, MMP-9, and MMP. In some embodiments, cells are engineered to produce an inhibitor of a molecule (e.g., protein) that inhibits MMPs. For example, cells may be engineered to express an inhibitor (e.g., an RNAi molecule) of membrane type 1 MMP (MT1-MMP) or TIMP metallopeptidase inhibitor 1 (TIMP-1).


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce a ligand that binds to selectin (e.g., hematopoietic cell E-/L-selectin ligand (HCELL), Dykstra et al., Stem Cells. 2016 October; 34(10):2501-2511) on the endothelium of a target tissue, for example.


The term “homing molecule” also encompasses transcription factors that regulate the production of molecules that improve/enhance homing of cells.


Also provided herein are engineered cells (e.g., tumor cells, erythrocytes, platelet cells, or bacterial cells) that are engineered to produce multiple chimeric proteins, at least two of which modulate different tumor-mediated immunosuppressive mechanisms. In some embodiments, at least one (e.g., 1, 2, 3, 4, 5, or more) chimeric protein stimulates at least one immunostimulatory mechanism in the tumor microenvironment, or inhibits at least one immunosuppressive mechanism in the tumor microenvironment. In some embodiments, at least one (e.g., 1, 2, 3, 4, 5, or more) chimeric protein inhibits at least one immunosuppressive mechanism in the tumor microenvironment, and at least one chimeric protein (e.g., 1, 2, 3, 4, 5, or more) inhibits at least one immunosuppressive mechanism in the tumor microenvironment. In yet other embodiments, at least two (e.g., 2, 3, 4, 5, or more) chimeric proteins stimulate at least one immunostimulatory mechanism in the tumor microenvironment. In still other embodiments, at least two (e.g., 1, 2, 3, 4, 5, or more) chimeric proteins inhibit at least one immunosuppressive mechanism in the tumor microenvironment.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce at least one chimeric protein that stimulates T cell signaling, activity and/or recruitment. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates antigen presentation and/or processing. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates natural killer cell-mediated cytotoxic signaling, activity and/or recruitment. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates dendritic cell differentiation and/or maturation. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates immune cell recruitment. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates M1 macrophage signaling, activity and/or recruitment. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates Th1 polarization. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates stroma degradation. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates immunostimulatory metabolite production. In some embodiments, a cell is engineered to produce at least one chimeric protein that stimulates Type I interferon signaling. In some embodiments, a cell is engineered to produce at least one chimeric protein that inhibits negative costimulatory signaling. In some embodiments, a cell is engineered to produce at least one chimeric protein that inhibits pro-apoptotic signaling (e.g., via TRAIL) of anti-tumor immune cells. In some embodiments, a cell is engineered to produce at least one chimeric protein that inhibits T regulatory (Treg) cell signaling, activity and/or recruitment. In some embodiments, a cell is engineered to produce at least one chimeric protein that inhibits tumor checkpoint molecules. In some embodiments, a cell is engineered to produce at least one chimeric protein that activates stimulator of interferon genes (STING) signaling. In some embodiments, a cell is engineered to produce at least one chimeric protein that inhibits myeloid-derived suppressor cell signaling, activity and/or recruitment. In some embodiments, a cell is engineered to produce at least one chimeric protein that degrades immunosuppressive factors/metabolites. In some embodiments, a cell is engineered to produce at least one chimeric protein that inhibits vascular endothelial growth factor signaling. In some embodiments, a cell is engineered to produce at least one chimeric protein that directly kills tumor cells (e.g., granzyme, perforin, oncolytic viruses, cytolytic peptides and enzymes, anti-tumor antibodies, e.g., that trigger ADCC).


In some embodiments, at least one chimeric protein: stimulates T cell signaling, activity and/or recruitment, stimulates antigen presentation and/or processing, stimulates natural killer cell-mediated cytotoxic signaling, activity and/or recruitment, stimulates dendritic cell differentiation and/or maturation, stimulates immune cell recruitment, stimulates macrophage signaling, stimulates stroma degradation, stimulates immunostimulatory metabolite production, or stimulates Type I interferon signaling; and at least one chimeric protein inhibits negative costimulatory signaling, inhibits pro-apoptotic signaling of anti-tumor immune cells, inhibits T regulatory (Treg) cell signaling, activity and/or recruitment, inhibits tumor checkpoint molecules, activates stimulator of interferon genes (STING) signaling, inhibits myeloid-derived suppressor cell signaling, activity and/or recruitment, degrades immunosuppressive factors/metabolites, inhibits vascular endothelial growth factor signaling, or directly kills tumor cells.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce at least one chimeric protein selected from IL-12, IFN-β, IFN-γ, IL-2, IL-15, IL-7, IL-36γ, IL-18, IL-1β, OX40-ligand, and CD40L; and/or at least one chimeric protein selected from a checkpoint inhibitor. Illustrative immune checkpoint molecules that can be targeted for blocking or inhibition include, but are not limited to, CTLA-4, 4-1BB (CD137), 4-1BBL (CD137L), PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, TIM3, B7H3, B7H4, VISTA, KIR, 2B4 (belongs to the CD2 family of molecules and is expressed on all NK, γδ T, and memory CD8+(αβ) T cells), CD160 (also referred to as BY55), and CGEN-15049. Immune checkpoint inhibitors include antibodies, or antigen binding fragments thereof, or other binding proteins, that bind to and block or inhibit the activity of one or more of CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, TIM3, B7H3, B7H4, VISTA, KIR, 2B4, CD160, and CGEN-15049. Exemplary checkpoint inhibitors include, but are not limited to, anti-PD-1 antibodies, anti-PD-L1 antibodies, anti-PD-L2 antibodies, anti-CTLA-4 antibodies, anti-LAG-3 antibodies, anti-TIM-3 antibodies, anti-TIGIT antibodies, anti-VISTA antibodies, anti-KIR antibodies, anti-B7-H3 antibodies, anti-B7-H4 antibodies, anti-HVEM antibodies, anti-BTLA antibodies, anti-GAL9 antibodies, anti-A2AR antibodies, anti-phosphatidylserine antibodies, anti-CD27 antibodies, anti-TNFa antibodies, anti-TREM1 antibodies, and anti-TREM2 antibodies. Illustrative immune checkpoint inhibitors include pembrolizumab (anti-PD-1; MK-3475/Keytruda®—Merck), nivolumamb (anti-PD-1; Opdivo®—BMS), pidilizumab (anti-PD-1 antibody; CT-011—Teva/CureTech), AMP224 (anti-PD-1; NCI), avelumab (anti-PD-L1; Bavencio®—Pfizer), durvalumab (anti-PD-L1; MEDI4736/Imfinzi®—Medimmune/AstraZeneca), atezolizumab (anti-PD-L1; Tecentriq®—Roche/Genentech), BMS-936559 (anti-PD-L1—BMS), tremelimumab (anti-CTLA-4; Medimmune/AstraZeneca), ipilimumab (anti-CTLA-4; Yervoy®—BMS), lirilumab (anti-KIR; BMS), monalizumab (anti-NKG2A; Innate Pharma/AstraZeneca).


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce at least one chimeric protein selected from IL-12, IFN-β, IFN-γ, IL-2, IL-15, IL-7, IL-36γ, IL-18, IL-1β, OX40-ligand, and CD40L; and/or at least one chimeric protein selected from anti-PD-1 antibodies, anti-PD-L1 antibodies, anti-CTLA-4 antibodies, and anti-IL-35 antibodies; and/or at least one chimeric protein selected from MIP1α (CCL3), MIP1 (CCL5), and CCL21; and/or at least one chimeric protein selected from CpG oligodeoxynucleotides; and/or at least one chimeric protein selected from microbial peptides.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-β and at least one chimeric protein selected from cytokines, antibodies, chemokines, nucleotides, peptides, enzymes, and stimulators of interferon genes (STINGs). In some embodiments, a cell is engineered to produce IFN-β and at least one cytokine or receptor/ligand (e.g., IL-12, IFN-γ, IL-2, IL-15, IL-7, IL-36γ, IL-18, IL-1β, OX40-ligand, and/or CD40L).


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-β and at least one cytokine or receptor/ligand (e.g., IL-12, IFN-γ, IL-2, IL-15, IL-7, IL-36γ, IL-18, IL-1, OX40-ligand, and/or CD40L).


In some embodiments the cytokine is produced as an engineered fusion protein with an antibody, antibody-fragment, or receptor that self-binds to the cytokine to induce cell-specific targeted binding such as with IL-2 fused to an antibody fragment preventing it from binding to Treg cells and preferentially binding to CD8 and NK cells. In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-β and at least one antibody (e.g., anti-PD-1 antibody, anti-PD-L1 antibody, anti-CTLA-4 antibody, anti-VEGF, anti-TGF-β, anti-IL-10, anti-TNF-α, and/or anti-IL-35 antibody). In some embodiments, a cell is engineered to produce IFN-β and at least one chemokine (MIP1α (CCL3), MIPiI (CCL5), and/or CCL21). In some embodiments, a cell is engineered to produce IFN-β and at least one nucleotide (e.g., a CpG oligodeoxynucleotide). In some embodiments, a cell is engineered to produce IFN-β and at least one peptide (e.g., an anti-tumor peptide). In some embodiments, a cell is engineered to produce IFN-β and at least one enzyme. In some embodiments, a cell is engineered to produce IFN-β and at least one STING activator. In some embodiments, a cell is engineered to produce IFN-β and at least one effector with direct anti-tumor activity (e.g., oncolytic virus).


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-α and MIP1-α. In some embodiments, a cell is engineered to produce IFN-α and MIP1-β. In some embodiments, a cell is engineered to produce IFN-α and CXCL9. In some embodiments, a cell is engineered to produce IFN-α and CXCL10. In some embodiments, a cell is engineered to produce IFN-α and CXCL11. In some embodiments, a cell is engineered to produce IFN-α and CCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL36-γ, IL-18, CD40L and/or 41BB-L. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-β and MIP1-α. In some embodiments, a cell is engineered to produce IFN-β and MIP1-β. In some embodiments, a cell is engineered to produce IFN-β and CXCL9. In some embodiments, a cell is engineered to produce IFN-β and CXCL10. In some embodiments, a cell is engineered to produce IFN-β and CXCL11. In some embodiments, a cell is engineered to produce IFN-β and CCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL36-γ, IL-18, CD40L and/or 41BB-L. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IL-12 and MIP1-α. In some embodiments, a cell is engineered to produce IL-12 and MIP1-β. In some embodiments, a cell is engineered to produce IL-12 and CXCL9. In some embodiments, a cell is engineered to produce IL-12 and CXCL10. In some embodiments, a cell is engineered to produce IL-12 and CXCL11. In some embodiments, a cell is engineered to produce IL-12 and CCL21. In some embodiments, the cell is engineered to further produce IFN-β, IFN-γ, IL-2, IL-7, IL-15, IL36-γ, IL-18, CD40L and/or 41BB-L. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce TNF-related apoptosis-inducing ligand (TRAIL) and MIP1-α. In some embodiments, a cell is engineered to produce TRAIL and MIP1-β. In some embodiments, a cell is engineered to produce TRAIL and CXCL9. In some embodiments, a cell is engineered to produce TRAIL and CXCL10. In some embodiments, a cell is engineered to produce TRAIL and CXCL11. In some embodiments, a cell is engineered to produce TRAIL and CCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL36-γ, IL-18, CD40L and/or 41BB-L. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce a stimulator of interferon gene (STING) and MIP1-α. In some embodiments, a cell is engineered to produce STING and MIP1-β. In some embodiments, a cell is engineered to produce STING and CXCL9. In some embodiments, a cell is engineered to produce STING and CXCL10. In some embodiments, a cell is engineered to produce STING and CXCL11. In some embodiments, a cell is engineered to produce STING and CCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL36-γ, IL-18, CD40L and/or 41BB-L. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CD40L and MIP1-α. In some embodiments, a cell is engineered to produce CD40L and MIP1-β. In some embodiments, a cell is engineered to produce CD40L and CXCL9. In some embodiments, a cell is engineered to produce CD40L and CXCL10. In some embodiments, a cell is engineered to produce CD40L and CXCL11. In some embodiments, a cell is engineered to produce CD40L and CCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL36-γ, IL-18, and/or 41BB-L. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce cytosine deaminase and MIP1-α. In some embodiments, a cell is engineered to produce cytosine deaminase and MIP1-β. In some embodiments, a cell is engineered to produce cytosine deaminase and CXCL9. In some embodiments, a cell is engineered to produce cytosine deaminase and CXCL10. In some embodiments, a cell is engineered to produce cytosine deaminase and CXCL11. In some embodiments, a cell is engineered to produce cytosine deaminase and CCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL36-γ, IL-18, CD40L, and/or 41BB-L. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-α and IL-12. In some embodiments, a cell is engineered to produce IFN-α and IFN-γ. In some embodiments, a cell is engineered to produce IFN-α and IL-2. In some embodiments, a cell is engineered to produce IFN-α and IL-7. In some embodiments, a cell is engineered to produce IFN-α and IL-15. In some embodiments, a cell is engineered to produce IFN-α and IL-36γ. In some embodiments, a cell is engineered to produce IFN-α and IL-18. In some embodiments, a cell is engineered to produce IFN-α and CD40L. In some embodiments, a cell is engineered to produce IFN-α and 41BB-L. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-β and IL-12. In some embodiments, a cell is engineered to produce IFN-β and IFN-γ. In some embodiments, a cell is engineered to produce IFN-β and IL-2. In some embodiments, a cell is engineered to produce IFN-β and IL-7. In some embodiments, a cell is engineered to produce IFN-β and IL-15. In some embodiments, a cell is engineered to produce IFN-β and IL-36γ. In some embodiments, a cell is engineered to produce IFN-β and IL-18. In some embodiments, a cell is engineered to produce IFN-β and CD40L. In some embodiments, a cell is engineered to produce IFN-β and 41BB-L. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce TNF-related apoptosis-inducing ligand (TRAIL) and IL-12. In some embodiments, a cell is engineered to produce TRAIL and IFN-γ. In some embodiments, a cell is engineered to produce TRAIL and IL-2. In some embodiments, a cell is engineered to produce TRAIL and IL-7. In some embodiments, a cell is engineered to produce TRAIL and IL-15. In some embodiments, a cell is engineered to produce TRAIL and IL-36γ. In some embodiments, a cell is engineered to produce TRAIL and IL-18. In some embodiments, a cell is engineered to produce TRAIL and CD40L. In some embodiments, a cell is engineered to produce TRAIL and 41BB-L. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce a stimulator of interferon gene (STING) and IL-12. In some embodiments, a cell is engineered to produce STING and IFN-γ. In some embodiments, a cell is engineered to produce STING and IL-2. In some embodiments, a cell is engineered to produce STING and IL-7. In some embodiments, a cell is engineered to produce STING and IL-15. In some embodiments, a cell is engineered to produce STING and IL-36γ. In some embodiments, a cell is engineered to produce STING and IL-18. In some embodiments, a cell is engineered to produce STING and CD40L. In some embodiments, a cell is engineered to produce STING and 41BB-L. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-$, CXCL9, CXCL10, CXCL11, and/or CCL21. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CD40L and IL-12. In some embodiments, a cell is engineered to produce CD40L and IFN-γ. In some embodiments, a cell is engineered to produce CD40L and IL-2. In some embodiments, a cell is engineered to produce CD40L and IL-7. In some embodiments, a cell is engineered to produce CD40L and IL-15. In some embodiments, a cell is engineered to produce CD40L and IL-36γ. In some embodiments, a cell is engineered to produce CD40L and IL-18. In some embodiments, a cell is engineered to produce CD40L and 41BB-L. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce cytosine deaminase and IL-12. In some embodiments, a cell is engineered to produce cytosine deaminase and IFN-γ. In some embodiments, a cell is engineered to produce cytosine deaminase and IL-2. In some embodiments, a cell is engineered to produce cytosine deaminase and IL-7. In some embodiments, a cell is engineered to produce cytosine deaminase and IL-15. In some embodiments, a cell is engineered to produce cytosine deaminase and IL-36γ. In some embodiments, a cell is engineered to produce cytosine deaminase and IL-18. In some embodiments, a cell is engineered to produce cytosine deaminase and CD40L. In some embodiments, a cell is engineered to produce cytosine deaminase and 41BB-L. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce MIP1-α and IL-12. In some embodiments, a cell is engineered to produce MIP1-α and MIP1-γ. In some embodiments, a cell is engineered to produce MIP1-α and IL-2. In some embodiments, a cell is engineered to produce MIP1-α and IL-7. In some embodiments, a cell is engineered to produce MIP1-α and IL-15. In some embodiments, a cell is engineered to produce MIP1-α and IL-36γ. In some embodiments, a cell is engineered to produce MIP1-α and IL-18. In some embodiments, a cell is engineered to produce MIP1-α and CD40L. In some embodiments, a cell is engineered to produce MIP1-α and 41BB-L. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce MIP1-β and IL-12. In some embodiments, a cell is engineered to produce MIP1-3 and MIP1-γ. In some embodiments, a cell is engineered to produce MIP1-β and IL-2. In some embodiments, a cell is engineered to produce MIP1-β and IL-7. In some embodiments, a cell is engineered to produce MIP1-β and IL-15. In some embodiments, a cell is engineered to produce MIP1-β and IL-36γ. In some embodiments, a cell is engineered to produce MIP1-3 and IL-18. In some embodiments, a cell is engineered to produce MIP1-β and CD40L. In some embodiments, a cell is engineered to produce MIP1-β and 41BB-L. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CXCL9 and IL-12. In some embodiments, a cell is engineered to produce CXCL9 and IFN-γ. In some embodiments, a cell is engineered to produce CXCL9 and IL-2. In some embodiments, a cell is engineered to produce CXCL9 and IL-7. In some embodiments, a cell is engineered to produce CXCL9 and IL-15. In some embodiments, a cell is engineered to produce CXCL9 and IL-36γ. In some embodiments, a cell is engineered to produce CXCL9 and IL-18. In some embodiments, a cell is engineered to produce CXCL9 and CD40L. In some embodiments, a cell is engineered to produce CXCL9 and 41BB-L. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce a CXCL10 and IL-12. In some embodiments, a cell is engineered to produce CXCL10 and IFN-γ. In some embodiments, a cell is engineered to produce CXCL10 and IL-2. In some embodiments, a cell is engineered to produce CXCL10 and IL-7. In some embodiments, a cell is engineered to produce CXCL10 and IL-15. In some embodiments, a cell is engineered to produce CXCL10 and IL-36γ. In some embodiments, a cell is engineered to produce CXCL10 and IL-18. In some embodiments, a cell is engineered to produce CXCL10 and CD40L. In some embodiments, a cell is engineered to produce CXCL10 and 41BB-L. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CXCL11 and IL-12. In some embodiments, a cell is engineered to produce CXCL11 and IFN-γ. In some embodiments, a cell is engineered to produce CXCL11 and IL-2. In some embodiments, a cell is engineered to produce CXCL11 and IL-7. In some embodiments, a cell is engineered to produce CXCL11 and IL-15. In some embodiments, a cell is engineered to produce CXCL11 and IL-36γ. In some embodiments, a cell is engineered to produce CXCL11 and IL-18. In some embodiments, a cell is engineered to produce CXCL11 and 41BB-L. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CCL21 and IL-12. In some embodiments, a cell is engineered to produce CCL21 and IFN-γ. In some embodiments, a cell is engineered to produce CCL21 and IL-2. In some embodiments, a cell is engineered to produce CCL21 and IL-7. In some embodiments, a cell is engineered to produce CCL21 and IL-15. In some embodiments, a cell is engineered to produce CCL21 and IL-36γ. In some embodiments, a cell is engineered to produce CCL21 and IL-18. In some embodiments, a cell is engineered to produce CCL21 and CD40L. In some embodiments, a cell is engineered to produce CCL21 and 41BB-L. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce anti-CD40 antibody, anti-CTLA4 antibody, anti-PD-L1 antibody, and/or OX40L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-α and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IFN-α and OX40L. In some embodiments, a cell is engineered to produce IFN-α and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IFN-α and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CXCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-β and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IFN-β and OX40L. In some embodiments, a cell is engineered to produce IFN-β and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IFN-β and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CXCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce TRAIL and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce TRAIL and OX40L. In some embodiments, a cell is engineered to produce TRAIL and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce TRAIL and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CXCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce STING and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce STING and OX40L. In some embodiments, a cell is engineered to produce STING and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce STING and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CXCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CD40L and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce CD40L and OX40L. In some embodiments, a cell is engineered to produce CD40L and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce CD40L and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CXCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce cytosine deaminase and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce cytosine deaminase and OX40L. In some embodiments, a cell is engineered to produce cytosine deaminase and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce cytosine deaminase and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CXCL21. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce MIP1-α and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce MIP1-α and OX40L. In some embodiments, a cell is engineered to produce MIP1-α and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce MIP1-α and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce MIP1-β and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce MIP1-β and OX40L. In some embodiments, a cell is engineered to produce MIP1-3 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce MIP1-β and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CXCL9 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce CXCL9 and OX40L. In some embodiments, a cell is engineered to produce CXCL9 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce CXCL9 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CXCL10 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce CXCL10 and OX40L. In some embodiments, a cell is engineered to produce CXCL10 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce CXCL10 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CXCL11 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce CXCL11 and OX40L. In some embodiments, a cell is engineered to produce CXCL11 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce CXCL11 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CCL21 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce CCL21 and OX40L. In some embodiments, a cell is engineered to produce CCL21 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce CCL21 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce IL-12, IFN-γ, IL-2, IL-7, IL-15, IL-36γ, IL-18, CD40L, and/or 41BB-L.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IL-12 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IL-12 and OX40L. In some embodiments, a cell is engineered to produce IL-12 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IL-12 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IFN-γ and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IFN-γ and OX40L. In some embodiments, a cell is engineered to produce IFN-γ and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IFN-γ and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IL-2 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IL-2 and OX40L. In some embodiments, a cell is engineered to produce IL-2 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IL-2 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IL-7 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IL-7 and OX40L. In some embodiments, a cell is engineered to produce IL-7 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IL-7 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IL-15 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IL-15 and OX40L. In some embodiments, a cell is engineered to produce IL-15 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IL-15 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IL-36-γ and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IL-36-γ and OX40L. In some embodiments, a cell is engineered to produce IL-36-γ and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IL-36-γ and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce IL-18 and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce IL-18 and OX40L. In some embodiments, a cell is engineered to produce IL-18 and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce IL-18 and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce CD40L and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce CD40L and OX40L. In some embodiments, a cell is engineered to produce CD40L and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce CD40L and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


In some embodiments, a cell (e.g., an immune cell or a stem cell) is engineered to produce 41BB-L and anti-PD-L1 antibody. In some embodiments, a cell is engineered to produce 41BB-L and OX40L. In some embodiments, a cell is engineered to produce 41BB-L and anti-CTLA4 antibody. In some embodiments, a cell is engineered to produce 41BB-L and anti-CD47 antibody. In some embodiments, the cell is engineered to further produce IFN-α, IFN-β, TRAIL, STING, CD40L, and/or cytosine deaminase. In some embodiments, the cell is engineered to further produce MIP1-α, MIP1-β, CXCL9, CXCL10, CXCL11, and/or CCL21.


A cell can also be further engineered to express additional proteins in addition to the chimeric proteins (e.g., the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein), proteins of interest, or effector molecules described herein. A cell can be further engineered to express antigen recognizing receptor. Examples of antigen recognizing receptors include, but are not limited to, 5T4, ADAM9, AFP, AXL, B7-H3, B7-H4, B7-H6, C4.4, CA6, Cadherin 3, Cadherin 6, CCR4, CD123, CD133, CD138, CD142, CD166, CD25, CD30, CD352, CD37, CD38, CD44, CD56, CD66e, CD70, CD71, CD74, CD79b, CD80, CEA, CEACAM5, Claudin18.2, cMet, CSPG4, CTLA, DLK1, DLL3, DR5, EGFR, ENPP3, EpCAM, EphA2, Ephrin A4, ETBR, FGFR2, FGFR3, FRalpha, FRb, GCC, GD2, GFRa4, gpA33, GPC3, gpNBM, GPRC5, HER2, IL-13R, IL-13Ra, IL-13Ra2, IL-8, IL-15, IL1RAP, Integrin aV, KIT, L1CAM, LAMP1, Lewis Y, LeY, LIV-1, LRRC, LY6E, MCSP, Mesothelin, MUC1, MUC16, MUCIC, NaPi2B, Nectin 4, NKG2D, NOTCH3, NY ESO 1, Ovarin, P-cadherin, pan-Erb2, PSCA, PSMA, PTK7, ROR1, S Aures, SCT, SLAMF7, SLITRK6, SSTR2, STEAP1, Survivin, TDGF1, TIM1, TROP2, and WT1.


An antigen recognizing receptor can include an antigen-binding domain, such as an antibody, an antigen-binding fragment of an antibody, a F(ab) fragment, a F(ab′) fragment, a single chain variable fragment (scFv), or a single-domain antibody (sdAb). An antigen recognizing receptors can include an scFv. An scFv can include a heavy chain variable domain (VH) and a light chain variable domain (VL), which can be separated by a peptide linker. For example, an scFv can include the structure VH-L-VL or VL-L-VH, wherein VH is the heavy chain variable domain, L is the peptide linker, and VL is the light chain variable domain.


An antigen recognizing receptor can be a chimeric antigen receptor (CAR). A CAR can have one or more intracellular signaling domains, such as a CD3zeta-chain intracellular signaling domain, a CD97 intracellular signaling domain, a CD11a-CD18 intracellular signaling domain, a CD2 intracellular signaling domain, an ICOS intracellular signaling domain, a CD27 intracellular signaling domain, a CD154 intracellular signaling domain, a CD8 intracellular signaling domain, an OX40 intracellular signaling domain, a 4-1BB intracellular signaling domain, a CD28 intracellular signaling domain, a ZAP40 intracellular signaling domain, a CD30 intracellular signaling domain, a GITR intracellular signaling domain, an HVEM intracellular signaling domain, a DAP10 intracellular signaling domain, a DAP12 intracellular signaling domain, a MyD88 intracellular signaling domain, fragments thereof, combinations thereof, or combinations of fragments thereof. A CAR can have a transmembrane domain, such as a CD8 transmembrane domain, a CD28 transmembrane domain a CD3zeta-chain transmembrane domain, a CD4 transmembrane domain, a 4-1BB transmembrane domain, an OX40 transmembrane domain, an ICOS transmembrane domain, a CTLA-4 transmembrane domain, a PD-1 transmembrane domain, a LAG-3 transmembrane domain, a 2B4 transmembrane domain, a BTLA transmembrane domain, fragments thereof, combinations thereof, or combinations of fragments thereof. A CAR can have a spacer region between the antigen-binding domain and the transmembrane domain.


An antigen recognizing receptor can be a T cell receptor (TCR).


Engineered Cell Types

Also provided herein are engineered cells. Cells can be engineered to comprise any of the engineered nucleic acids described herein (e.g., any of the engineered nucleic acids encoding the chimeric protein described herein). Cells can be engineered to possess any of the features of any of the engineered cells described herein. In a particular aspect, provided herein are cells engineered to produce one or more chimeric proteins, where the one or more chimeric proteins are the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein. In a particular aspect, provided herein are cells engineered to produce two or more chimeric proteins. In a particular aspect, provided herein are cells engineered to produce two or more of the chimeric proteins described herein, where each chimeric protein is a different protein of interest or effector molecule. In a particular aspect, provided herein are cells engineered to produce any of the chimeric proteins described herein and engineered to separately produce a different effector molecule or protein of interest (e.g., a homing molecule, antigen receptor, etc.).


The engineered cells can be an immune cell, including but not limited to, a T cell, a CD8+ T cell, a CD4+ T cell, a gamma-delta (γδ) T cell, a cytotoxic T lymphocyte (CTL), a regulatory T cell, a viral-specific T cell, a Natural Killer T (NKT) cell, a Natural Killer (NK) cell, a B cell, a tumor-infiltrating lymphocyte (TIL), an innate lymphoid cell, a mast cell, an eosinophil, a basophil, a neutrophil, a myeloid cell, a macrophage, a monocyte, or a dendritic cell.


The engineered cells can be a stem cell, including but not limited to, a human embryonic stem cell (ESC), an ESC-derived cell, a pluripotent stem cell, a mesenchymal stromal cell (MSC), an induced pluripotent stem cell (iPSC), or an iPSC-derived cell.


The engineered cells can be tumor-derived cells. Examples of tumor cells include, but are not limited to, a bladder tumor cell, a brain tumor cell, a breast tumor cell, a cervical tumor cell, a colorectal tumor cell, an esophageal tumor cell, a glioma cell, a kidney tumor cell, a liver tumor cell, a lung tumor cell, a melanoma cell, an ovarian tumor cell, a pancreatic tumor cell, a prostate tumor cell, a skin tumor cell, a thyroid tumor cell, and a uterine tumor cell.


A cell can be engineered to produce the chimeric proteins using methods known to those skilled in the art. For example, cells can be transduced to engineer the tumor. In an embodiment, the cell is transduced using a virus.


In a particular embodiment, the cell is transduced using an oncolytic virus. Examples of oncolytic viruses include, but are not limited to, an oncolytic herpes simplex virus, an oncolytic adenovirus, an oncolytic measles virus, an oncolytic influenza virus, an oncolytic Indiana vesiculovirus, an oncolytic Newcastle disease virus, an oncolytic vaccinia virus, an oncolytic poliovirus, an oncolytic myxoma virus, an oncolytic reovirus, an oncolytic mumps virus, an oncolytic Maraba virus, an oncolytic rabies virus, an oncolytic rotavirus, an oncolytic hepatitis virus, an oncolytic rubella virus, an oncolytic dengue virus, an oncolytic chikungunya virus, an oncolytic respiratory syncytial virus, an oncolytic lymphocytic choriomeningitis virus, an oncolytic morbillivirus, an oncolytic lentivirus, an oncolytic replicating retrovirus, an oncolytic rhabdovirus, an oncolytic Seneca Valley virus, an oncolytic sindbis virus, and any variant or derivative thereof.


The virus, including any of the oncolytic viruses described herein, can be a recombinant virus that encodes one more transgenes encoding one or more chimeric proteins, such as any of the engineered nucleic acids described herein. The virus, including any of the oncolytic viruses described herein, can be a recombinant virus that encodes one more transgenes encoding one or more of the two or more chimeric proteins, such as any of the engineered nucleic acids described herein.


Also provided herein are engineered erythrocytes. Erythrocytes can be engineered to comprise any of the engineered nucleic acids described herein. Erythrocytes can be engineered to possess any of the features of any of the engineered cells described herein. In a particular aspect, provided herein are erythrocytes engineered to produce one or more of the chimeric proteins described herein. In a particular aspect, provided herein are erythrocytes engineered to produce two or more of the chimeric proteins described herein.


Also provided herein are engineered platelet cells. Platelet cells can be engineered to comprise any of the engineered nucleic acids described herein. Platelet cells can be engineered to possess any of the features of any of the engineered cells described herein. In a particular aspect, provided herein are platelet cells engineered to produce one or more of the chimeric proteins described herein. In a particular aspect, provided herein are platelet cells engineered to produce two or more of the chimeric proteins described herein.


Also provided herein are engineered bacterial cells. Bacterial cells can be engineered to comprise any of the engineered nucleic acids described herein. Bacterial cells can be engineered to possess any of the features of any of the engineered cells described herein. In a particular aspect, provided herein are bacterial cells engineered to produce two or more of the chimeric proteins described herein. Bacterial cells can be engineered to produce one or more mammalian-derived chimeric proteins. Bacterial cells can be engineered to produce two or more mammalian-derived chimeric proteins. Examples of bacterial cells include, but are not limited to, Clostridium beijerinckii, Clostridium sporogenes, Clostridium novyi, Escherichia coli, Pseudomonas aeruginosa, Listeria monocytogenes, Salmonella typhimurium, and Salmonella choleraesuis.


An engineered cell can be a human cell. An engineered cell can be a human primary cell. An engineered primary cell can be a tumor infiltrating primary cell. An engineered primary cell can be a primary T cell. An engineered primary cell can be a hematopoietic stem cell (HSC). An engineered primary cell can be a natural killer cell. An engineered primary cell can be any somatic cell. An engineered primary cell can be a MSC. Human cells (e.g., immune cells) can be engineered to comprise any of the engineered nucleic acids described herein. Human cells (e.g., immune cells) can be engineered to possess any of the features of any of the engineered cells described herein. In a particular aspect, provided herein are human cells (e.g., immune cells) engineered to produce one or more of the chimeric proteins described herein. In a particular aspect, provided herein are human cells (e.g., immune cells) engineered to produce two or more of the chimeric proteins described herein.


An engineered cell can be isolated from a subject (autologous), such as a subject known or suspected to have cancer. Cell isolation methods are known to those skilled in the art and include, but are not limited to, sorting techniques based on cell-surface marker expression, such as FACS sorting, positive isolation techniques, and negative isolation, magnetic isolation, and combinations thereof. An engineered cell can be allogenic with reference to the subject being administered a treatment. Allogenic modified cells can be HLA-matched to the subject being administered a treatment. An engineered cell can be a cultured cell, such as an ex vivo cultured cell. An engineered cell can be an ex vivo cultured cell, such as a primary cell isolated from a subject. Cultured cell can be cultured with one or more cytokines.


Also provided herein are methods that include culturing the engineered cells of the present disclosure. Methods of culturing the engineered cells described herein are known. One skilled in the art will recognize that culturing conditions will depend on the particular engineered cell of interest. One skilled in the art will recognize that culturing conditions will depend on the specific downstream use of the engineered cell, for example, specific culturing conditions for subsequent administration of the engineered cell to a subject.


Methods of Engineering Cells


Also provided herein are compositions and methods for engineering cells to produce one or more proteins of interest or effector molecules (e.g., the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein).


In general, cells are engineered to produce proteins of interest or effector molecules through introduction (i.e., delivery) of polynucleotides encoding the one or more proteins of interest or effector molecules, e.g., the chimeric proteins described herein having the protein of interest or effector molecule, into the cell's cytosol and/or nucleus. For example, the polynucleotides encoding the one or more chimeric proteins can be any of the engineered nucleic acids encoding the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein. Delivery methods include, but are not limited to, viral-mediated delivery, lipid-mediated transfection, nanoparticle delivery, electroporation, sonication, and cell membrane deformation by physical means. One skilled in the art will appreciate the choice of delivery method can depend on the specific cell type to be engineered.


Viral-Mediated Delivery

Viral vector-based delivery platforms can be used to engineer cells. In general, a viral vector-based delivery platform engineers a cell through introducing (i.e., delivering) into a host cell. For example, a viral vector-based delivery platform can engineer a cell through introducing any of the engineered nucleic acids described herein (e.g., any of the exogenous polynucleotide sequences encoding the chimeric proteins described herein, such as the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein, and/or any of the expression cassettes described herein containing a promoter and an exogenous polynucleotide sequence encoding the chimeric proteins, oriented from N-terminal to C-terminal). A viral vector-based delivery platform can be a nucleic acid, and as such, an engineered nucleic acid can also encompass an engineered virally-derived nucleic acid. Such engineered virally-derived nucleic acids can also be referred to as recombinant viruses or engineered viruses.


A viral vector-based delivery platform can encode more than one engineered nucleic acid, gene, or transgene within the same nucleic acid. For example, an engineered virally-derived nucleic acid, e.g., a recombinant virus or an engineered virus, can encode one or more transgenes, including, but not limited to, any of the engineered nucleic acids described herein that encode one or more of the chimeric proteins described herein. The one or more transgenes encoding the one or more chimeric proteins can be configured to express the one or more chimeric proteins and/or other protein of interest. A viral vector-based delivery platform can encode one or more genes in addition to the one or more transgenes (e.g., transgenes encoding the one or more chimeric proteins and/or other protein of interest), such as viral genes needed for viral infectivity and/or viral production (e.g., capsid proteins, envelope proteins, viral polymerases, viral transcriptases, etc.), referred to as cis-acting elements or genes.


A viral vector-based delivery platform can comprise more than one viral vector, such as separate viral vectors encoding the engineered nucleic acids, genes, or transgenes described herein, and referred to as trans-acting elements or genes. For example, a helper-dependent viral vector-based delivery platform can provide additional genes needed for viral infectivity and/or viral production on one or more additional separate vectors in addition to the vector encoding the one or more chimeric proteins and/or other protein of interest. One viral vector can deliver more than one engineered nucleic acids, such as one vector that delivers engineered nucleic acids that are configured to produce two or more chimeric proteins and/or other protein of interest. More than one viral vector can deliver more than one engineered nucleic acids, such as more than one vector that delivers one or more engineered nucleic acid configured to produce one or more chimeric proteins and/or other protein of interest. The number of viral vectors used can depend on the packaging capacity of the above mentioned viral vector-based vaccine platforms, and one skilled in the art can select the appropriate number of viral vectors.


In general, any of the viral vector-based systems can be used for the in vitro production of molecules, such as the chimeric proteins, effector molecules, and/or other protein of interest described herein, or used in vivo and ex vivo gene therapy procedures, e.g., for in vivo delivery of the engineered nucleic acids encoding one or more chimeric proteins and/or other protein of interest. The selection of an appropriate viral vector-based system will depend on a variety of factors, such as cargo/payload size, immunogenicity of the viral system, target cell of interest, gene expression strength and timing, and other factors appreciated by one skilled in the art.


Viral vector-based delivery platforms can be RNA-based viruses or DNA-based viruses. Exemplary viral vector-based delivery platforms include, but are not limited to, a herpes simplex virus, a adenovirus, a measles virus, an influenza virus, a Indiana vesiculovirus, a Newcastle disease virus, a vaccinia virus, a poliovirus, a myxoma virus, a reovirus, a mumps virus, a Maraba virus, a rabies virus, a rotavirus, a hepatitis virus, a rubella virus, a dengue virus, a chikungunya virus, a respiratory syncytial virus, a lymphocytic choriomeningitis virus, a morbillivirus, a lentivirus, a replicating retrovirus, a rhabdovirus, a Seneca Valley virus, a sindbis virus, and any variant or derivative thereof. Other exemplary viral vector-based delivery platforms are described in the art, such as vaccinia, fowlpox, self-replicating alphavirus, marabavirus, adenovirus (See, e.g., Tatsis et al., Adenoviruses, Molecular Therapy (2004) 10, 616-629), or lentivirus, including but not limited to second, third or hybrid second/third generation lentivirus and recombinant lentivirus of any generation designed to target specific cell types or receptors (See, e.g., Hu et al., Immunization Delivered by Lentiviral Vectors for Cancer and Infectious Diseases, Immunol Rev. (2011) 239(1): 45-61, Sakuma et al., Lentiviral vectors: basic to translational, Biochem J. (2012) 443(3):603-18, Cooper et al., Rescue of splicing-mediated intron loss maximizes expression in lentiviral vectors containing the human ubiquitin C promoter, Nucl. Acids Res. (2015) 43 (1): 682-690, Zufferey et al., Self-Inactivating Lentivirus Vector for Safe and Efficient In vivo Gene Delivery, J. Virol. (1998) 72 (12): 9873-9880).


The sequences may be preceded with one or more sequences targeting a subcellular compartment. Upon introduction (i.e. delivery) into a host cell, infected cells (i.e., an engineered cell) can express the chimeric proteins and/or other protein of interest. Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848. Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described in Stover et al. (Nature 351:456-460 (1991)). A wide variety of other vectors useful for the introduction (i.e., delivery) of engineered nucleic acids, e.g., Salmonella typhi vectors, and the like will be apparent to those skilled in the art from the description herein.


The viral vector-based delivery platforms can be a virus that targets a cell, herein referred to as an oncolytic virus. Examples of oncolytic viruses include, but are not limited to, an oncolytic herpes simplex virus, an oncolytic adenovirus, an oncolytic measles virus, an oncolytic influenza virus, an oncolytic Indiana vesiculovirus, an oncolytic Newcastle disease virus, an oncolytic vaccinia virus, an oncolytic poliovirus, an oncolytic myxoma virus, an oncolytic reovirus, an oncolytic mumps virus, an oncolytic Maraba virus, an oncolytic rabies virus, an oncolytic rotavirus, an oncolytic hepatitis virus, an oncolytic rubella virus, an oncolytic dengue virus, an oncolytic chikungunya virus, an oncolytic respiratory syncytial virus, an oncolytic lymphocytic choriomeningitis virus, an oncolytic morbillivirus, an oncolytic lentivirus, an oncolytic replicating retrovirus, an oncolytic rhabdovirus, an oncolytic Seneca Valley virus, an oncolytic sindbis virus, and any variant or derivative thereof. Any of the oncolytic viruses described herein can be a recombinant oncolytic virus comprising one more transgenes (e.g., an engineered nucleic acid) encoding one or more chimeric proteins and/or other protein of interest. The transgenes encoding the one or more chimeric proteins and/or other protein of interest can be configured to express the chimeric proteins and/or other protein of interest.


The viral vector-based delivery platform can be retrovirus-based. In general, retroviral vectors are comprised of cis-acting long terminal repeats with packaging capacity for up to 6-10 kb of foreign sequence. The minimum cis-acting LTRs are sufficient for replication and packaging of the vectors, which are then used to integrate the one or more engineered nucleic acids (e.g., transgenes encoding the one or more chimeric proteins and/or other protein of interest) into the target cell to provide permanent transgene expression. Retroviral-based delivery systems include, but are not limited to, those based upon murine leukemia, virus (MuLV), gibbon ape leukemia virus (GaLV), Simian Immuno deficiency vims (SIV), human immuno deficiency vims (HIV), and combinations thereof (see, e.g., Buchscher et al., J. Virol. 66:2731-2739 (1992); Johann et ah, J. Virol. 66:1635-1640 (1992); Sommnerfelt et al., Virol. 176:58-59 (1990); Wilson et ah, J. Virol. 63:2374-2378 (1989); Miller et al, J, Virol. 65:2220-2224 (1991); PCT/US94/05700). Other retroviral systems include the Phoenix retrovirus system.


The viral vector-based delivery platform can be lentivirus-based. In general, lentiviral vectors are retroviral vectors that are able to transduce or infect non-dividing cells and typically produce high viral titers. Lentiviral-based delivery platforms can be HIV-based, such as ViraPower systems (ThermoFisher) or pLenti systems (Cell Biolabs). Lentiviral-based delivery platforms can be SIV, or FIV-based. Other exemplary lentivirus-based delivery platforms are described in more detail in U.S. Pat. Nos. 7,311,907; 7,262,049; 7,250,299; 7,226,780; 7,220,578; 7,211,247; 7,160,721; 7,078,031; 7,070,993; 7,056,699; 6,955,919, each herein incorporated by reference for all purposes.


The viral vector-based delivery platform can be adenovirus-based. In general, adenoviral based vectors are capable of very high transduction efficiency in many cell types, do not require cell division, achieve high titer and levels of expression, and can be produced in large quantities in a relatively simple system. In general, adenoviruses can be used for transient expression of a transgene within an infected cell since adenoviruses do not typically integrate into a host's genome. Adenovirus-based delivery platforms are described in more detail in Li et al., Invest Opthalmol Vis Sci 35:2543 2549, 1994; Borras et al., Gene Ther 6:515 524, 1999; Li and Davidson, PNAS 92:7700 7704, 1995; Sakamoto et al., H Gene Ther 5:1088 1097, 1999; WO 94/12649, WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655, each herein incorporated by reference for all purposes. Other exemplary adenovirus-based delivery platforms are described in more detail in U.S. Pat. Nos. 5,585,362; 6,083,716, 7,371,570; 7,348,178; 7,323,177; 7,319,033; 7,318,919; and 7,306,793 and International Patent Application WO96/13597, each herein incorporated by reference for all purposes.


The viral vector-based delivery platform can be adeno-associated virus (AAV)-based. Adeno-associated virus (“AAV”) vectors may be used to transduce cells with engineered nucleic acids (e.g., any of the engineered nucleic acids described herein). AAV systems can be used for the in vitro production of proteins of interest, such as the chimeric proteins described herein and/or effector molecules, or used in vivo and ex vivo gene therapy procedures, e.g., for in vivo delivery of the engineered nucleic acids encoding one or more chimeric proteins and/or other protein of interest (see, e.g., West et al., Virology 160:38-47 (1987); U.S. Pat. Nos. 4,797,368; 5,436,146; 6,632,670; 6,642,051; 7,078,387; 7,314,912; 6,498,244; 7,906,111; US patent publications US 2003-0138772, US 2007/0036760, and US 2009/0197338; Gao, et al., J. Virol, 78(12):6381-6388 (June 2004); Gao, et al, Proc Natl Acad Sci USA, 100(10):6081-6086 (May 13, 2003); and International Patent applications WO 2010/138263 and WO 93/24641; Kotin, Human Gene Therapy 5:793-801 (1994); Muzyczka, J. Clin. Invest. 94:1351 (1994), each herein incorporated by reference for all purposes). Exemplary methods for constructing recombinant AAV vectors are described in more detail in U.S. Pat. No. 5,173,414; Tratschin et ah, Mol. Cell. Biol. 5:3251-3260 (1985); Tratschin, et ah, Mol. Cell, Biol. 4:2072-2081 (1984); Hermonat & amp; Muzyczka, PNAS 81:64666470 (1984); and Samuiski et ah, J. Virol. 63:03822-3828 (1989), each herein incorporated by reference for all purposes. In general, an AAV-based vector comprises a capsid protein having an amino acid sequence corresponding to any one of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV.Rh10, AAV11 and variants thereof. In particular examples, an AAV-based vector has a capsid protein having an amino acid sequence corresponding to AAV2. In particular examples, an AAV-based vector has a capsid protein having an amino acid sequence corresponding to AAV8.


AAV vectors can be engineered to have any of the exogenous polynucleotide sequences encoding the membrane-cleavable chimeric proteins described herein having the formula: S-C-MT-D.


AAV vectors can be engineered to express a heterologous nucleic acid, wherein the heterologous nucleic acid encodes any of the degron-fusion chimeric proteins described herein. The protein of interest in the degron-fusion chimeric protein can be secretable. The protein of interest in the degron-fusion chimeric protein can be non-secretable. The degron domain can be any of the degron domains described elsewhere in more detail herein, such as a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the protein of interest. CRBN polypeptide substrate domains can be any of the CRBN polypeptide substrate domains described elsewhere in more detail herein, such as IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN. The CRBN polypeptide substrate domain can be a chimeric fusion product of native CRBN polypeptide sequences, such as a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189). Regulated degradation can be drug-inducible. Drugs capable of mediating/regulating degradation can be small-molecule inhibitors. Drugs capable of mediating/regulating degradation can include an IMiD, as described in greater detail elsewhere herein. For degrons having a CRBN polypeptide substrate domain, examples of imide-containing IMiDs include, but are not limited to, a thalidomide, a lenalidomide, or a pomalidomide. The IMiD can be an FDA-approved drug. Other examples of degrons in the fusion protein include, but are not limited to, a HCV NS4 degron, PEST (two copies of residues 277-307 of human IxBU), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box. Examples of proteins of interest in the fusion protein can be proteins selected from therapeutic classes including, but not limited to, a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a polynucleotide, a peptide, or an enzyme.


Described herein are AAVs engineered to express a heterologous nucleic acid, wherein the heterologous nucleic acid encodes a degron-fusion chimeric protein, and wherein the degron-fusion chimeric protein contains a degron fused to a protein of interest. For example, AAVs engineered to express a degron-fusion chimeric protein (e.g., any of the degron-fusion chimeric described herein) can be produced using any of the AAV vectors described herein. AAV vectors and/or engineered AAV viruses can be combined with a pharmaceutically acceptable carrier.


The viral vector-based delivery platform can be a virus-like particle (VLP) platform. In general, VLPs are constructed by producing viral structural proteins and purifying resulting viral particles. Then, following purification, a cargo/payload (e.g., any of the engineered nucleic acids described herein) is encapsulated within the purified particle ex vivo. Accordingly, production of VLPs maintains separation of the nucleic acids encoding viral structural proteins and the nucleic acids encoding the cargo/payload. The viral structural proteins used in VLP production can be produced in a variety of expression systems, including mammalian, yeast, insect, bacterial, or in vivo translation expression systems. The purified viral particles can be denatured and reformed in the presence of the desired cargo to produce VLPs using methods known to those skilled in the art. Production of VLPs are described in more detail in Seow et al. (Mol Ther. 2009 May; 17(5): 767-777), herein incorporated by reference for all purposes.


The viral vector-based delivery platform can be engineered to target (i.e., infect) a range of cells, target a narrow subset of cells, or target a specific cell. In general, the envelope protein chosen for the viral vector-based delivery platform will determine the viral tropism. The virus used in the viral vector-based delivery platform can be pseudotyped to target a specific cell of interest. The viral vector-based delivery platform can be pantropic and infect a range of cells. For example, pantropic viral vector-based delivery platforms can include the VSV-G envelope. The viral vector-based delivery platform can be amphotropic and infect mammalian cells. Accordingly, one skilled in the art can select the appropriate tropism, pseudotype, and/or envelope protein for targeting a desired cell type.


Lipid Structure Delivery Systems

Engineered nucleic acids (e.g., any of the engineered nucleic acids described herein) can be introduced into a cell using a lipid-mediated delivery system. In general, a lipid-mediated delivery system uses a structure composed of an outer lipid membrane enveloping an internal compartment. Examples of lipid-based structures include, but are not limited to, a lipid-based nanoparticle, a liposome, a micelle, an exosome, a vesicle, an extracellular vesicle, a cell, or a tissue. Lipid structure delivery systems can deliver a cargo/payload (e.g., any of the engineered nucleic acids described herein) in vitro, in vivo, or ex vivo.


A lipid-based nanoparticle can include, but is not limited to, a unilamellar liposome, a multilamellar liposome, and a lipid preparation. As used herein, a “liposome” is a generic term encompassing in vitro preparations of lipid vehicles formed by enclosing a desired cargo, e.g., an engineered nucleic acid, such as any of the engineered nucleic acids described herein, within a lipid shell or a lipid aggregate. Liposomes may be characterized as having vesicular structures with a bilayer membrane, generally comprising a phospholipid, and an inner medium that generally comprises an aqueous composition. Liposomes include, but are not limited to, emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. Liposomes can be unilamellar liposomes. Liposomes can be multilamellar liposomes. Liposomes can be multivesicular liposomes. Liposomes can be positively charged, negatively charged, or neutrally charged. In certain embodiments, the liposomes are neutral in charge. Liposomes can be formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of a desired purpose, e.g., criteria for in vivo delivery, such as liposome size, acid lability and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al., Ann. Rev. Biophys. Bioeng. 9; 467 (1980), U.S. Pat. Nos. 4,235,871, 4,501,728, 4,501,728, 4,837,028, and 5,019,369, each herein incorporated by reference for all purposes.


A multilamellar liposome is generated spontaneously when lipids comprising phospholipids are suspended in an excess of aqueous solution such that multiple lipid layers are separated by an aqueous medium. Water and dissolved solutes are entrapped in closed structures between the lipid bilayers following the lipid components undergoing self-rearrangement. A desired cargo (e.g., a polypeptide, a nucleic acid, a small molecule drug, an engineered nucleic acid, such as any of the engineered nucleic acids described herein, a viral vector, a viral-based delivery system, etc.) can be encapsulated in the aqueous interior of a liposome, attached to a liposome via a linking molecule that is associated with both the liposome and the polypeptide/nucleic acid, interspersed within the lipid bilayer of a liposome, entrapped in a liposome, complexed with a liposome, or otherwise associated with the liposome such that it can be delivered to a target entity. Lipophilic molecules or molecules with lipophilic regions may also dissolve in or associate with the lipid bilayer.


A liposome used according to the present embodiments can be made by different methods, as would be known to one of ordinary skill in the art. Preparations of liposomes are described in further detail in WO 2016/201323, International Applications PCT/US85/01161 and PCT/US89/05040, and U.S. Pat. Nos. 4,728,578, 4,728,575, 4,737,323, 4,533,254, 4,162,282, 4,310,505, and 4,921,706; each herein incorporated by reference for all purposes.


Liposomes can be cationic liposomes. Examples of cationic liposomes are described in more detail in U.S. Pat. Nos. 5,962,016; 5,030,453; 6,680,068, U.S. Application 2004/0208921, and International Patent Applications WO03/015757A1, WO04029213A2, and WO02/100435A1, each hereby incorporated by reference in their entirety.


Lipid-mediated gene delivery methods are described, for instance, in WO 96/18372; WO 93/24640; Mannino & Gould-Fogerite, BioTechniques 6(7): 682-691 (1988); U.S. Pat. No. 5,279,833 Rose U.S. Pat. No. 5,279,833; WO91/06309; and Felgner et al., Proc. Natl. Acad. Sci. USA 84: 7413-7414 (1987), each herein incorporated by reference for all purposes.


Exosomes are small membrane vesicles of endocytic origin that are released into the extracellular environment following fusion of multivesicular bodies with the plasma membrane. The size of exosomes ranges between 30 and 100 nm in diameter. Their surface consists of a lipid bilayer from the donor cell's cell membrane, and they contain cytosol from the cell that produced the exosome, and exhibit membrane proteins from the parental cell on the surface. Exosomes useful for the delivery of nucleic acids are known to those skilled in the art, e.g., the exosomes described in more detail in U.S. Pat. No. 9,889,210, herein incorporated by reference for all purposes.


As used herein, the term “extracellular vesicle” or “EV” refers to a cell-derived vesicle comprising a membrane that encloses an internal space. In general, extracellular vesicles comprise all membrane-bound vesicles that have a smaller diameter than the cell from which they are derived. Generally extracellular vesicles range in diameter from 20 nm to 1000 nm, and can comprise various macromolecular cargo either within the internal space, displayed on the external surface of the extracellular vesicle, and/or spanning the membrane. The cargo can comprise nucleic acids (e.g., any of the engineered nucleic acids described herein), proteins, carbohydrates, lipids, small molecules, and/or combinations thereof. By way of example and without limitation, extracellular vesicles include apoptotic bodies, fragments of cells, vesicles derived from cells by direct or indirect manipulation (e.g., by serial extrusion or treatment with alkaline solutions), vesiculated organelles, and vesicles produced by living cells (e.g., by direct plasma membrane budding or fusion of the late endosome with the plasma membrane). Extracellular vesicles can be derived from a living or dead organism, explanted tissues or organs, and/or cultured cells.


As used herein the term “exosome” refers to a cell-derived small (between 20-300 nm in diameter, more preferably 40-200 nm in diameter) vesicle comprising a membrane that encloses an internal space, and which is generated from the cell by direct plasma membrane budding or by fusion of the late endosome with the plasma membrane. The exosome comprises lipid or fatty acid and polypeptide and optionally comprises a payload (e.g., a therapeutic agent), a receiver (e.g., a targeting moiety), a polynucleotide (e.g., a nucleic acid, RNA, or DNA, such as any of the engineered nucleic acids described herein), a sugar (e.g., a simple sugar, polysaccharide, or glycan) or other molecules. The exosome can be derived from a producer cell, and isolated from the producer cell based on its size, density, biochemical parameters, or a combination thereof. An exosome is a species of extracellular vesicle. Generally, exosome production/biogenesis does not result in the destruction of the producer cell. Exosomes and preparation of exosomes are described in further detail in WO 2016/201323, which is hereby incorporated by reference in its entirety.


As used herein, the term “nanovesicle” (also referred to as a “microvesicle”) refers to a cell-derived small (between 20-250 nm in diameter, more preferably 30-150 nm in diameter) vesicle comprising a membrane that encloses an internal space, and which is generated from the cell by direct or indirect manipulation such that said nanovesicle would not be produced by said producer cell without said manipulation. In general, a nanovesicle is a sub-species of an extracellular vesicle. Appropriate manipulations of the producer cell include but are not limited to serial extrusion, treatment with alkaline solutions, sonication, or combinations thereof. The production of nanovesicles may, in some instances, result in the destruction of said producer cell. Preferably, populations of nanovesicles are substantially free of vesicles that are derived from producer cells by way of direct budding from the plasma membrane or fusion of the late endosome with the plasma membrane. The nanovesicle comprises lipid or fatty acid and polypeptide, and optionally comprises a payload (e.g., a therapeutic agent), a receiver (e.g., a targeting moiety), a polynucleotide (e.g., a nucleic acid, RNA, or DNA, such as any of the engineered nucleic acids described herein), a sugar (e.g., a simple sugar, polysaccharide, or glycan) or other molecules. The nanovesicle, once it is derived from a producer cell according to said manipulation, may be isolated from the producer cell based on its size, density, biochemical parameters, or a combination thereof.


Lipid nanoparticles (LNPs), in general, are synthetic lipid structures that rely on the amphiphilic nature of lipids to form membranes and vesicle like structures (Riley 2017). In general, these vesicles deliver cargo/payloads, such as any of the engineered nucleic acids or viral systems described herein, by absorbing into the membrane of target cells and releasing the cargo into the cytosol. Lipids used in LNP formation can be cationic, anionic, or neutral. The lipids can be synthetic or naturally derived, and in some instances biodegradable. Lipids can include fats, cholesterol, phospholipids, lipid conjugates including, but not limited to, polyethyleneglycol (PEG) conjugates (PEGylated lipids), waxes, oils, glycerides, and fat soluble vitamins. Lipid compositions generally include defined mixtures of materials, such as the cationic, neutral, anionic, and amphipathic lipids. In some instances, specific lipids are included to prevent LNP aggregation, prevent lipid oxidation, or provide functional chemical groups that facilitate attachment of additional moieties. Lipid composition can influence overall LNP size and stability. In an example, the lipid composition comprises dilinoleylmethyl-4-dimethylaminobutyrate (MC3) or MC3-like molecules. MC3 and MC3-like lipid compositions can be formulated to include one or more other lipids, such as a PEG or PEG-conjugated lipid, a sterol, or neutral lipids. In addition, LNPs can be further engineered or functionalized to facilitate targeting of specific cell types. Another consideration in LNP design is the balance between targeting efficiency and cytotoxicity.


Micelles, in general, are spherical synthetic lipid structures that are formed using single-chain lipids, where the single-chain lipid's hydrophilic head forms an outer layer or membrane and the single-chain lipid's hydrophobic tails form the micelle center. Micelles typically refer to lipid structures only containing a lipid mono-layer. Micelles are described in more detail in Quader et al. (Mol Ther. 2017 Jul. 5; 25(7): 1501-1513), herein incorporated by reference for all purposes.


Nucleic-acid vectors, such as expression vectors, exposed directly to serum can have several undesirable consequences, including degradation of the nucleic acid by serum nucleases or off-target stimulation of the immune system by the free nucleic acids. Similarly, viral delivery systems exposed directly to serum can trigger an undesired immune response and/or neutralization of the viral delivery system. Therefore, encapsulation of an engineered nucleic acid and/or viral delivery system can be used to avoid degradation, while also avoiding potential off-target affects. In certain examples, an engineered nucleic acid and/or viral delivery system is fully encapsulated within the delivery vehicle, such as within the aqueous interior of an LNP. Encapsulation of an engineered nucleic acid and/or viral delivery system within an LNP can be carried out by techniques well-known to those skilled in the art, such as microfluidic mixing and droplet generation carried out on a microfluidic droplet generating device. Such devices include, but are not limited to, standard T-junction devices or flow-focusing devices. In an example, the desired lipid formulation, such as MC3 or MC3-like containing compositions, is provided to the droplet generating device in parallel with an engineered nucleic acid or viral delivery system and any other desired agents, such that the delivery vector and desired agents are fully encapsulated within the interior of the MC3 or MC3-like based LNP. In an example, the droplet generating device can control the size range and size distribution of the LNPs produced. For example, the LNP can have a size ranging from 1 to 1000 nanometers in diameter, e.g., 1, 10, 50, 100, 500, or 1000 nanometers. Following droplet generation, the delivery vehicles encapsulating the cargo/payload (e.g., an engineered nucleic acid and/or viral delivery system) can be further treated or engineered to prepare them for administration.


Nanoparticle Delivery

Nanomaterials can be used to deliver engineered nucleic acids (e.g., any of the engineered nucleic acids described herein). Nanomaterial vehicles, importantly, can be made of non-immunogenic materials and generally avoid eliciting immunity to the delivery vector itself. These materials can include, but are not limited to, lipids (as previously described), inorganic nanomaterials, and other polymeric materials. Nanomaterial particles are described in more detail in Riley et al. (Recent Advances in Nanomaterials for Gene Delivery-A Review. Nanomaterials 2017, 7(5), 94), herein incorporated by reference for all purposes.


Genomic Editing Systems

A genomic editing systems can be used to engineer a host genome to encode an engineered nucleic acid, such as an engineered nucleic acid encoding one or more of the chimeric proteins (e.g., any of the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein). In general, a “genomic editing system” refers to any system for integrating an exogenous gene into a host cell's genome. Genomic editing systems include, but are not limited to, a transposon system, a nuclease genomic editing system, and a viral vector-based delivery platform.


A transposon system can be used to integrate an engineered nucleic acid, such as an engineered nucleic acid encoding one or more of the chimeric proteins (e.g., any of the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein), into a host genome. Transposons generally comprise terminal inverted repeats (TIR) that flank a cargo/payload nucleic acid and a transposase. The transposon system can provide the transposon in cis or in trans with the TIR-flanked cargo. A transposon system can be a retrotransposon system or a DNA transposon system. In general, transposon systems integrate a cargo/payload (e.g., an engineered nucleic acid) randomly into a host genome. Examples of transposon systems include systems using a transposon of the Tc1/mariner transposon superfamily, such as a Sleeping Beauty transposon system, described in more detail in Hudecek et al. (Crit Rev Biochem Mol Biol. 2017 August; 52(4):355-380), and U.S. Pat. Nos. 6,489,458, 6,613,752 and 7,985,739, each of which is herein incorporated by reference for all purposes. Another example of a transposon system includes a PiggyBac transposon system, described in more detail in U.S. Pat. Nos. 6,218,185 and 6,962,810, each of which is herein incorporated by reference for all purposes.


A nuclease genomic editing system can be used to engineer a host genome to encode an engineered nucleic acid, such as an engineered nucleic acid encoding one or more of the chimeric proteins (e.g., any of the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein). Without wishing to be bound by theory, in general, the nuclease-mediated gene editing systems used to introduce an exogenous gene take advantage of a cell's natural DNA repair mechanisms, particularly homologous recombination (HR) repair pathways. Briefly, following an insult to genomic DNA (typically a double-stranded break), a cell can resolve the insult by using another DNA source that has identical, or substantially identical, sequences at both its 5′ and 3′ ends as a template during DNA synthesis to repair the lesion. In a natural context, HDR can use the other chromosome present in a cell as a template. In gene editing systems, exogenous polynucleotides are introduced into the cell to be used as a homologous recombination template (HRT or HR template). In general, any additional exogenous sequence not originally found in the chromosome with the lesion that is included between the 5′ and 3′ complimentary ends within the HRT (e.g., a gene or a portion of a gene) can be incorporated (i.e., “integrated”) into the given genomic locus during templated HDR. Thus, a typical HR template for a given genomic locus has a nucleotide sequence identical to a first region of an endogenous genomic target locus, a nucleotide sequence identical to a second region of the endogenous genomic target locus, and a nucleotide sequence encoding a cargo/payload nucleic acid (e.g., any of the engineered nucleic acids described herein, such as any of the engineered nucleic acids encoding one or more chimeric proteins (e.g., any of the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein)).


In some examples, a HR template can be linear. Examples of linear HR templates include, but are not limited to, a linearized plasmid vector, a ssDNA, a synthesized DNA, and a PCR amplified DNA. In particular examples, a HR template can be circular, such as a plasmid. A circular template can include a supercoiled template.


The identical, or substantially identical, sequences found at the 5′ and 3′ ends of the HR template, with respect to the exogenous sequence to be introduced, are generally referred to as arms (HR arms). HR arms can be identical to regions of the endogenous genomic target locus (i.e., 100% identical). HR arms in some examples can be substantially identical to regions of the endogenous genomic target locus. While substantially identical HR arms can be used, it can be advantageous for HR arms to be identical as the efficiency of the HDR pathway may be impacted by HR arms having less than 100% identity.


Each HR arm, i.e., the 5′ and 3′ HR arms, can be the same size or different sizes. Each HR arm can each be greater than or equal to 50, 100, 200, 300, 400, or 500 bases in length. Although HR arms can, in general, be of any length, practical considerations, such as the impact of HR arm length and overall template size on overall editing efficiency, can also be taken into account. An HR arms can be identical, or substantially identical to, regions of an endogenous genomic target locus immediately adjacent to a cleavage site. Each HR arms can be identical to, or substantially identical to, regions of an endogenous genomic target locus immediately adjacent to a cleavage site. Each HR arms can be identical, or substantially identical to, regions of an endogenous genomic target locus within a certain distance of a cleavage site, such as 1 base-pair, less than or equal to 10 base-pairs, less than or equal to 50 base-pairs, or less than or equal to 100 base-pairs of each other.


A nuclease genomic editing system can use a variety of nucleases to cut a target genomic locus, including, but not limited to, a Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) family nuclease or derivative thereof, a Transcription activator-like effector nuclease (TALEN) or derivative thereof, a zinc-finger nuclease (ZFN) or derivative thereof, and a homing endonuclease (HE) or derivative thereof.


A CRISPR-mediated gene editing system can be used to engineer a host genome to encode an engineered nucleic acid, such as an engineered nucleic acid encoding one or more of the chimeric proteins (e.g., any of the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein). CRISPR systems are described in more detail in M. Adli (“The CRISPR tool kit for genome editing and beyond” Nature Communications; volume 9 (2018), Article number: 1911), herein incorporated by reference for all that it teaches. In general, a CRISPR-mediated gene editing system comprises a CRISPR-associated (Cas) nuclease and a RNA(s) that directs cleavage to a particular target sequence. An exemplary CRISPR-mediated gene editing system is the CRISPR/Cas9 systems comprised of a Cas9 nuclease and a RNA(s) that has a CRISPR RNA (crRNA) domain and a trans-activating CRISPR (tracrRNA) domain. The crRNA typically has two RNA domains: a guide RNA sequence (gRNA) that directs specificity through base-pair hybridization to a target sequence (“a defined nucleotide sequence”), e.g., a genomic sequence; and an RNA domain that hybridizes to a tracrRNA. A tracrRNA can interact with and thereby promote recruitment of a nuclease (e.g., Cas9) to a genomic locus. The crRNA and tracrRNA polynucleotides can be separate polynucleotides. The crRNA and tracrRNA polynucleotides can be a single polynucleotide, also referred to as a single guide RNA (sgRNA). While the Cas9 system is illustrated here, other CRISPR systems can be used, such as the Cpf1 system. Nucleases can include derivatives thereof, such as Cas9 functional mutants, e.g., a Cas9 “nickase” mutant that in general mediates cleavage of only a single strand of a defined nucleotide sequence as opposed to a complete double-stranded break typically produced by Cas9 enzymes.


In general, the components of a CRISPR system interact with each other to form a Ribonucleoprotein (RNP) complex to mediate sequence specific cleavage. In some CRISPR systems, each component can be separately produced and used to form the RNP complex. In some CRISPR systems, each component can be separately produced in vitro and contacted (i.e., “complexed”) with each other in vitro to form the RNP complex. The in vitro produced RNP can then be introduced (i.e., “delivered”) into a cell's cytosol and/or nucleus, e.g., a T cell's cytosol and/or nucleus. The in vitro produced RNP complexes can be delivered to a cell by a variety of means including, but not limited to, electroporation, lipid-mediated transfection, cell membrane deformation by physical means, lipid nanoparticles (LNP), virus like particles (VLP), and sonication. In a particular example, in vitro produced RNP complexes can be delivered to a cell using a Nucleofactor/Nucleofection® electroporation-based delivery system (Lonza®). Other electroporation systems include, but are not limited to, MaxCyte electroporation systems, Miltenyi CliniMACS electroporation systems, Neon electroporation systems, and BTX electroporation systems. CRISPR nucleases, e.g., Cas9, can be produced in vitro (i.e., synthesized and purified) using a variety of protein production techniques known to those skilled in the art. CRISPR system RNAs, e.g., an sgRNA, can be produced in vitro (i.e., synthesized and purified) using a variety of RNA production techniques known to those skilled in the art, such as in vitro transcription or chemical synthesis.


An in vitro produced RNP complex can be complexed at different ratios of nuclease to gRNA. An in vitro produced RNP complex can be also be used at different amounts in a CRISPR-mediated editing system. For example, depending on the number of cells desired to be edited, the total RNP amount added can be adjusted, such as a reduction in the amount of RNP complex added when editing a large number of cells in a reaction.


In some CRISPR systems, each component (e.g., Cas9 and an sgRNA) can be separately encoded by a polynucleotide with each polynucleotide introduced into a cell together or separately. In some CRISPR systems, each component can be encoded by a single polynucleotide (i.e., a multi-promoter or multicistronic vector, see description of exemplary multicistronic systems below) and introduced into a cell. Following expression of each polynucleotide encoded CRISPR component within a cell (e.g., translation of a nuclease and transcription of CRISPR RNAs), an RNP complex can form within the cell and can then direct site-specific cleavage.


Some RNPs can be engineered to have moieties that promote delivery of the RNP into the nucleus. For example, a Cas9 nuclease can have a nuclear localization signal (NLS) domain such that if a Cas9 RNP complex is delivered into a cell's cytosol or following translation of Cas9 and subsequent RNP formation, the NLS can promote further trafficking of a Cas9 RNP into the nucleus.


The engineered cells described herein can be engineered using non-viral methods, e.g., the nuclease and/or CRISPR mediated gene editing systems described herein can be delivered to a cell using non-viral methods. The engineered cells described herein can be engineered using viral methods, e.g., the nuclease and/or CRISPR mediated gene editing systems described herein can be delivered to a cell using viral methods such as adenoviral, retroviral, lentiviral, or any of the other viral-based delivery methods described herein.


In some CRISPR systems, more than one CRISPR composition can be provided such that each separately target the same gene or general genomic locus at more than target nucleotide sequence. For example, two separate CRISPR compositions can be provided to direct cleavage at two different target nucleotide sequences within a certain distance of each other. In some CRISPR systems, more than one CRISPR composition can be provided such that each separately target opposite strands of the same gene or general genomic locus. For example, two separate CRISPR “nickase” compositions can be provided to direct cleavage at the same gene or general genomic locus at opposite strands.


In general, the features of a CRISPR-mediated editing system described herein can apply to other nuclease-based genomic editing systems. TALEN is an engineered site-specific nuclease, which is composed of the DNA-binding domain of TALE (transcription activator-like effectors) and the catalytic domain of restriction endonuclease Fokl. By changing the amino acids present in the highly variable residue region of the monomers of the DNA binding domain, different artificial TALENs can be created to target various nucleotides sequences. The DNA binding domain subsequently directs the nuclease to the target sequences and creates a double-stranded break. TALEN-based systems are described in more detail in U.S. Ser. No. 12/965,590; U.S. Pat. Nos. 8,450,471; 8,440,431; 8,440,432; 10,172,880; and U.S. Ser. No. 13/738,381, all of which are incorporated by reference herein in their entirety. ZFN-based editing systems are described in more detail in U.S. Pat. Nos. 6,453,242; 6,534,261; 6,599,692; 6,503,717; 6,689,558; 7,030,215; 6,794,136; 7,067,317; 7,262,054; 7,070,934; 7,361,635; 7,253,273; and U.S. Patent Publication Nos. 2005/0064474; 2007/0218528; 2005/0267061, all incorporated herein by reference in their entireties for all purposes.


Other Engineering Delivery Systems

Various additional means to introduce engineered nucleic acids (e.g., any of the engineered nucleic acids described herein) into a cell or other target recipient entity, such as any of the lipid structures described herein.


Electroporation can used to deliver polynucleotides to recipient entities. Electroporation is a method of internalizing a cargo/payload into a target cell or entity's interior compartment through applying an electrical field to transiently permeabilize the outer membrane or shell of the target cell or entity. In general, the method involves placing cells or target entities between two electrodes in a solution containing a cargo of interest (e.g., any of the engineered nucleic acids described herein). The lipid membrane of the cells is then disrupted, i.e., permeabilized, by applying a transient set voltage that allows the cargo to enter the interior of the entity, such as the cytoplasm of the cell. In the example of cells, at least some, if not a majority, of the cells remain viable. Cells and other entities can be electroporated in vitro, in vivo, or ex vivo. Electroporation conditions (e.g., number of cells, concentration of cargo, recovery conditions, voltage, time, capacitance, pulse type, pulse length, volume, cuvette length, electroporation solution composition, etc.) vary depending on several factors including, but not limited to, the type of cell or other recipient entity, the cargo to be delivered, the efficiency of internalization desired, and the viability desired. Optimization of such criteria are within the scope of those skilled in the art. A variety devices and protocols can be used for electroporation. Examples include, but are not limited to, Neon® Transfection System, MaxCyte® Flow Electroporation™, Lonza® Nucleofector™ systems, and Bio-Rad© electroporation systems.


Other means for introducing engineered nucleic acids (e.g., any of the engineered nucleic acids described herein) into a cell or other target recipient entity include, but are not limited to, sonication, gene gun, hydrodynamic injection, and cell membrane deformation by physical means.


Compositions and methods for delivering engineered mRNAs in vivo, such as naked plasmids or mRNA, are described in detail in Kowalski et al. (Mol Ther. 2019 Apr. 10; 27(4): 710-728) and Kaczmarek et al. (Genome Med. 2017; 9: 60.), each herein incorporated by reference for all purposes.


Delivery Vehicles


Also provided herein are compositions for delivering a cargo/payload (a “delivery vehicle”).


The cargo can comprise nucleic acids (e.g., any of the engineered nucleic acids described herein, such as any of the engineered nucleic acids described herein encoding the chimeric proteins including the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein)), as described above. The cargo can comprise proteins, carbohydrates, lipids, small molecules, and/or combinations thereof. The cargo can be any of the chimeric proteins provided for herein (e.g., any of the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein). The cargo can be a combination of the chimeric proteins described herein, e.g., two or more of the chimeric proteins described herein. The cargo can be a combination of the chimeric protein described herein and another cargo of interest, such as another protein, carbohydrate, lipid, small molecule, and/or combination thereof.


The delivery vehicle can comprise any composition suitable for delivering a cargo. The delivery vehicle can comprise any composition suitable for delivering a protein (e.g., any of the chimeric proteins described herein). The delivery vehicle can be any of the lipid structure delivery systems described herein. For example, a delivery vehicle can be a lipid-based structure including, but not limited to, a lipid-based nanoparticle, a liposome, a micelle, an exosome, a vesicle, an extracellular vesicle, a cell, or a tissue. The delivery vehicle can be any of the nanoparticles described herein, such as nanoparticles comprising lipids (as previously described), inorganic nanomaterials, and other polymeric materials.


The delivery vehicle can be capable of delivering the cargo to a cell, such as delivering any of the chimeric proteins described herein to a cell. The delivery vehicle can be capable of delivering the cargo to a cell, such as delivering any of the chimeric proteins described herein to a cell. The delivery vehicle can be configured to target a specific cell, such as configured with a re-directing antibody to target a specific cell. The delivery vehicle can be capable of delivering the cargo to a cell in vivo.


The delivery vehicle can be capable of delivering the cargo to a tissue or tissue environment (e.g., a tumor microenvironment), such as delivering any of the chimeric proteins described herein to a tissue or tissue environment in vivo. Delivering a cargo can include secreting the cargo, such as secreting any of the chimeric proteins described herein. Accordingly, the delivery vehicle can be capable of secreting the cargo, such as secreting any of the chimeric proteins described herein. The delivery vehicle can be capable of secreting the cargo to a tissue or tissue environment (e.g., a tumor microenvironment), such as secreting any of the chimeric proteins described herein into a tissue or tissue environment. The delivery vehicle can be configured to target a specific tissue or tissue environment (e.g., a tumor microenvironment), such as configured with a re-directing antibody to target a specific tissue or tissue environment.


Methods of Treatment


Further provided herein are methods that include delivering, or administering, to a subject (e.g., a human subject) engineered cells as provided herein to produce in vivo at least one protein of interest produced by the engineered cells (e.g., any of the chimeric proteins provided for herein, such as the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein, or the secreted effector molecules provided for herein following protease cleavage of the chimeric protein). Further provided herein are methods that include delivering, or administering, to a subject (e.g., a human subject) engineered cells as provided herein to produce in vivo at least two proteins of interest, e.g., at least two of the chimeric proteins provided for herein, such as the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein, produced by the engineered cells.


Further provided herein are methods that include delivering, or administering, to a subject (e.g., a human subject) any of the delivery vehicles described herein, such as any of the delivery vehicles described herein comprising any of the proteins of interest described herein, e.g., any of the chimeric proteins provided for herein, such as the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein. Further provided herein are methods that include delivering, or administering, to a subject (e.g., a human subject) any of the delivery vehicles described herein, such as any of the delivery vehicles described herein comprising two or more proteins of, e.g., at least two of the chimeric proteins provided for herein, such as the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein.


In some embodiments, the engineered cells or delivery vehicles are administered via intravenous, intraperitoneal, intratracheal, subcutaneous, intratumoral, oral, anal, intranasal (e.g., packed in a delivery particle), or arterial (e.g., internal carotid artery) routes. Thus, the engineered cells or delivery vehicles may be administered systemically or locally (e.g., to a TME or via intratumoral administration). An engineered cell can be isolated from a subject, such as a subject known or suspected to have cancer. An engineered cell can be allogenic with reference to the subject being administered a treatment. Allogenic modified cells can be HLA-matched to the subject being administered a treatment. Delivery vehicles can be any of the lipid structure delivery systems described herein. Delivery vehicles can be any of the nanoparticles described herein.


Engineered cells or delivery vehicles can be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated. For example, engineered cells or delivery vehicles can be administered in combination with one or more IMiDs described herein. FDA-approved IMiDs can be administered in their approved fashion. In another example, engineered cells or delivery vehicles can be administered in combination with a checkpoint inhibitor therapy. Exemplary checkpoint inhibitors include, but are not limited to, anti-PD-1 antibodies, anti-PD-L1 antibodies, anti-PD-L2 antibodies. anti-CTLA-4 antibodies, anti-LAG-3 antibodies, anti-TIM-3 antibodies, anti-TIGIT antibodies, anti-VISTA antibodies, anti-KIR antibodies, anti-B7-H3 antibodies, anti-B74-14 antibodies, anti-HVEM anibodies, anti-BTLA antibodies, anti-GAL9 antibodies, anti-A2AR antibodies, anti-CD27 antibodies. anti-TNFa antibodies, anti-TREM1 antibodies, and anti-TREM2 antibodies. Illustrative immune checkpoint inhibitors include pembrolizumab (anti-PD-1; MK-3475/Keytruda®—Merck), nivolumamb (anti-PD-1; Opdivo®—BMS), pidilizumab (anti-PD-1 antibody; CT-011—Teva/CureTech), AMP224 (anti-PD-1; NCI), avelumab (anti-PD-L1; Bavencio®—Pfizer), durvalumab (anti-PD-L1; MEDI4736/Imfinzi®—Medimmune/AstraZeneca), atezolizumab (anti-PD-L1; Tecentriq®—Roche/Genentech), BMS-936559 (anti-PD-L1—BMS), tremelimumab (anti-CTLA-4; Medimmune/AstraZeneca), ipilimumab (anti-CTLA-4; Yervoy®—BMS), lirilumab (anti-KIR; BMS), monalizumab (anti-NKG2A; Innate Pharma/AstraZeneca). In other examples, engineered cells or delivery vehicles can be administered in combination with TGFbeta inhibitors, VEGF inhibitors, or HPGE2. In another example, engineered cells or delivery vehicles can be administered in combination with an anti-CD40 antibody.


Some methods comprise selecting a subject (or patient population) having a tumor (or cancer) and treating that subject with engineered cells or delivery vehicles that modulate tumor-mediated immunosuppressive mechanisms.


The engineered cells or delivery vehicles of the present disclosure may be used, in some instances, to treat cancer, such as ovarian cancer. Other cancers are described herein. For example, the engineered cells may be used to treat bladder tumors, brain tumors, breast tumors, cervical tumors, colorectal tumors, esophageal tumors, gliomas, kidney tumors, liver tumors, lung tumors, melanomas, ovarian tumors, pancreatic tumors, prostate tumors, skin tumors, thyroid tumors, and/or uterine tumors. The engineered cells or delivery vehicles of the present disclosure can be used to treat cancers with tumors located in the peritoneal space of a subject.


The methods provided herein also include delivering a preparation of engineered cells or delivery vehicles. A preparation, in some embodiments, is a substantially pure preparation, containing, for example, less than 5% (e.g., less than 4%, 3%, 2%, or 1%) of cells other than engineered cells. A preparation may comprise 1×105 cells/kg to 1×107 cells/kg cells. Preparation of engineered cells or delivery vehicles can include pharmaceutical compositions having one or more pharmaceutically acceptable carriers. For example, preparations of engineered cells or delivery vehicles can include any of the engineered viruses, such as an engineered AAV virus, or any of the engineered viral vectors, such as AAV vector, described herein.


In Vivo Expression

The methods provided herein also include delivering a composition in vivo capable of producing the engineered cells described herein, e.g., capable of delivering any of the engineered nucleic acids described herein to a cell in vivo. Such compositions include any of the viral-mediated delivery platforms, any of the lipid structure delivery systems, any of the nanoparticle delivery systems, any of the genomic editing systems, or any of the other engineering delivery systems described herein capable of engineering a cell in vivo.


The methods provided herein also include delivering a composition in vivo capable of producing any of the proteins of interest described herein, e.g., any of the chimeric proteins provided for herein, such as the degron-fusion chimeric proteins described herein or the membrane-cleavable chimeric proteins having the formula S-C-MT-D described herein. The methods provided herein also include delivering a composition in vivo capable of producing two or more of the proteins of interest described herein. Compositions capable of in vivo production of proteins of interest include, but are not limited to, any of the engineered nucleic acids described herein. Compositions capable of in vivo production proteins of interest can be a naked mRNA or a naked plasmid.


ADDITIONAL EMBODIMENTS

The paragraphs below provide additional enumerated embodiments.

    • 1. An engineered nucleic acid comprising an expression cassette comprising a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula:





S-C-MT-D

      • wherein
        • S comprises a secretable effector molecule,
        • C comprises a protease cleavage site,
        • MT comprises a cell membrane tethering domain, and
        • D comprises a degron,
      • wherein the promoter is operably linked to the exogenous polynucleotide sequence, and wherein S-C-MT-D is configured to be expressed as a single polypeptide.
    • 2. The engineered nucleic acid of paragraph 1, wherein the promoter is a constitutive promoter.
    • 3. The engineered nucleic acid of paragraph 2, wherein the constitutive promoter is selected from the group consisting of: CAG, HLP, CMV, EFS, SFFV, SV40, MND, PGK, UbC, hEF1aV1, hCAGG, hEF1aV2, hACTb, heIF4A1, hGAPDH, hGRP78, hGRP94, hHSP70, hKINb, and hUBIb.
    • 4. The engineered nucleic acid of any one of paragraphs 1-3, wherein the promoter is an inducible promoter.
    • 5. The engineered nucleic acid of paragraph 4, wherein the inducible promoter is selected from the group consisting of: minP, NFkB response element, CREB response element, NFAT response element, SRF response element 1, SRF response element 2, AP1 response element, TCF-LEF response element promoter fusion, Hypoxia responsive element, SMAD binding element, STAT3 binding site, minCMV, YB_TATA, minTK, inducer molecule responsive promoters, and tandem repeats thereof.
    • 6. The engineered nucleic acid of any one of paragraphs 1-3, wherein the promoter is a synthetic promoter.
    • 7. The engineered nucleic acid of any one of paragraphs 1-3, wherein the promoter is a tissue-specific promoter.
    • 8. The engineered nucleic acid of any one of paragraphs 1-7, wherein the secretable effector molecule comprises a signal peptide.
    • 9. The engineered nucleic acid of paragraph 8, wherein the signal peptide comprises a native signal peptide native to the secretable effector molecule.
    • 10. The engineered nucleic acid of paragraph 8, wherein the signal peptide comprises a non-native signal peptide non-native to the secretable effector molecule.
    • 11. The engineered nucleic acid of paragraph 10, wherein the non-native signal peptide is selected from the group consisting of: IL12, IL2, optimized IL2, trypsiongen-2, Gaussia luciferase, CD5, human IgKVII, murine IgKVII, VSV-G, prolactin, serum albumin preprotein, azurocidin preprotein, osteonectin, CD33, IL6, IL8, CCL2, TIMP2, VEGFB, osteoprotegerin, serpin E1, GROalpha, CXCL12, IL21, CD8, and GMCSF.
    • 12. The engineered nucleic acid of any one of paragraphs 1-11, wherein the secretable effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a peptide, and an enzyme.
    • 13. The engineered nucleic acid of paragraph 12, wherein the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha.
    • 14. The engineered nucleic acid of paragraph 12, wherein the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1.
    • 15. The engineered nucleic acid of paragraph 12, wherein the homing molecule is selected from the group consisting of: anti-integrin alpha4,beta7; anti-MAdCAM; SDF1; and MMP-2.
    • 16. The engineered nucleic acid of paragraph 12, wherein the growth factor is selected from the group consisting of: FLT3L and GM-CSF.
    • 17. The engineered nucleic acid of paragraph 12, wherein the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40L.
    • 18. The engineered nucleic acid of paragraph 12, wherein the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2.
    • 19. The engineered nucleic acid of paragraph 18, wherein the TGFbeta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereof.
    • 20. The engineered nucleic acid of paragraph 18, wherein the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anwd-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody.
    • 21. The engineered nucleic acid of paragraph 18, wherein the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof.
    • 22. The engineered nucleic acid of any one of paragraphs 1-18, wherein the secretable effector molecule is a human-derived effector molecule.
    • 23. The engineered nucleic acid of any one of paragraphs 1-22, wherein the protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site.
    • 24. The engineered nucleic acid of any one of paragraphs 1-23, wherein the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain.
    • 25. The engineered nucleic acid of paragraph 24, wherein the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA.
    • 26. The engineered nucleic acid of any one of paragraphs 1-23, wherein the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof.
    • 27. The engineered nucleic acid of any one of paragraphs 1-26, wherein when expressed in a cell, the secretable effector molecule is tethered to a cell membrane of the cell.
    • 28. The engineered nucleic acid of paragraph 27, wherein when expressed in a cell expressing a protease capable of cleaving the protease cleavage site, the secretable effector molecule is released from the cell membrane.
    • 29. The engineered nucleic acid of paragraph 28, wherein the protease expressed on the cell membrane is endogenous to the cell.
    • 30. The engineered nucleic acid of paragraph 29, wherein the protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease.
    • 31. The engineered nucleic acid of paragraph 28, wherein the protease expressed on the cell membrane is heterologous to the cell.
    • 32. The engineered nucleic acid of paragraph 31, wherein the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3).
    • 33. The engineered nucleic acid of paragraph 32, wherein the protease cleavage site comprises an NS3 protease cleavage site.
    • 34. The engineered nucleic acid of paragraph 33, wherein the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site.
    • 35. The engineered nucleic acid of any one of paragraphs 28-34, wherein the protease can be repressed by a protease inhibitor.
    • 36. The engineered nucleic acid of paragraph 35, wherein the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir.
    • 37. The engineered nucleic acid of any one of paragraphs 28-36, wherein expression of the protease is capable of regulation.
    • 38. The engineered nucleic acid of any one of paragraphs 1-37, wherein the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule.
    • 39. The engineered nucleic acid of paragraph 38, wherein the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN.
    • 40. The engineered nucleic acid of paragraph 38, wherein the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences.
    • 41. The engineered nucleic acid of paragraph 38, wherein the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189).
    • 42. The engineered nucleic acid of any one of paragraphs 38-41, wherein the IMiD is an FDA-approved drug.
    • 43. The engineered nucleic acid of any one of paragraphs 38-42, wherein the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide.
    • 44. The engineered nucleic acid of any one of paragraphs 1-36, wherein the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box.
    • 45. The engineered nucleic acid of any one of paragraphs 1-44, wherein the degron is localized C-terminal of the cell membrane tethering domain.
    • 46. The engineered nucleic acid of any one of paragraphs 1-45, wherein the engineered nucleic acid is selected from the group consisting of: a DNA, cDNA, an RNA, an mRNA, and a naked plasmid.
    • 47. An expression vector comprising the engineered nucleic acid of any one of paragraphs 1-46.
    • 48. The expression vector of paragraph 47, wherein the expression vector is a viral vector.
    • 49. The expression vector of paragraph 48, wherein the viral vector is a lentiviral vector.
    • 50. A membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula:





S-C-MT-D

      • wherein
        • S comprises a secretable effector molecule,
        • C comprises a protease cleavage site,
        • MT comprises a cell membrane tethering domain,
        • D comprises a degron, and
        • wherein S-C-MT-D is configured to be expressed as a single polypeptide.
    • 51. The chimeric protein of paragraph 50, wherein the secretable effector molecule comprises a signal peptide.
    • 52. The chimeric protein of paragraph 51, wherein the signal peptide comprises a native signal peptide native to the secretable effector molecule.
    • 53. The chimeric protein of paragraph 51, wherein the signal peptide comprises a non-native signal peptide non-native to the secretable effector molecule.
    • 54. The chimeric protein of paragraph 53, wherein the non-native signal peptide is selected from the group consisting of: IL12, IL2, optimized IL2, trypsiongen-2, Gaussia luciferase, CD5, human IgKVII, murine IgKVII, VSV-G, prolactin, serum albumin preprotein, azurocidin preprotein, osteonectin, CD33, IL6, IL8, CCL2, TIMP2, VEGFB, osteoprotegerin, serpin E1, GROalpha, CXCL12, IL21, CD8, and GMCSF.
    • 55. The chimeric protein of any one of paragraphs 50-54, wherein the secretable effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a peptide, and an enzyme.
    • 56. The chimeric protein of paragraph 55, wherein the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha.
    • 57. The chimeric protein of paragraph 55, wherein the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1.
    • 58. The chimeric protein of paragraph 55, wherein the homing molecule is selected from the group consisting of: anti-integrin alpha4,beta7; anti-MAdCAM; SDF1; and MMP-2.
    • 59. The chimeric protein of paragraph 55, wherein the growth factor is selected from the group consisting of: FLT3L and GM-CSF.
    • 60. The chimeric protein of paragraph 55, wherein the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40L.
    • 61. The chimeric protein of paragraph 55, wherein the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2.
    • 62. The chimeric protein of paragraph 61, wherein the TGF-beta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereof.
    • 63. The chimeric protein of paragraph 61, wherein the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody.
    • 64. The chimeric protein of paragraph 61, wherein the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof.
    • 65. The chimeric protein of any one of paragraphs 50-61, wherein the secretable effector molecule is a human-derived effector molecule.
    • 66. The chimeric protein of any one of paragraphs 50-65, wherein the protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site.
    • 67. The chimeric protein of any one of paragraphs 50-66, wherein the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain.
    • 68. The chimeric protein of paragraph 67, wherein the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA.
    • 69. The chimeric protein of any one of paragraphs 50-66, wherein the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof.
    • 70. The chimeric protein of any one of paragraphs 50-69, wherein when expressed in a cell, the secretable effector molecule is tethered to a cell membrane of the cell.
    • 71. The chimeric protein of paragraph 70, wherein when expressed in a cell expressing a protease capable of cleaving the protease cleavage site, the secretable effector molecule is released from the cell membrane.
    • 72. The chimeric protein of paragraph 71, wherein the protease expressed on the cell membrane is endogenous to the cell.
    • 73. The chimeric protein of paragraph 72, wherein the protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease.
    • 74. The chimeric protein of paragraph 71, wherein the protease expressed on the cell membrane is heterologous to the cell.
    • 75. The chimeric protein of paragraph 74, wherein the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3).
    • 76. The chimeric protein of paragraph 75, wherein the protease cleavage site comprises an NS3 protease cleavage site.
    • 77. The chimeric protein of paragraph 76, wherein the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site.
    • 78. The chimeric protein of any one of paragraphs 71-77, wherein the protease can be repressed by a protease inhibitor.
    • 79. The chimeric protein of paragraph 78, wherein the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir.
    • 80. The chimeric protein of any one of paragraphs 71-79, wherein the protease further comprises a degron.
    • 81. The chimeric protein of any one of paragraphs 71-80, wherein expression of the protease is capable of regulation.
    • 82. The chimeric protein of any one of paragraphs 50-81, wherein the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule.
    • 83. The chimeric protein of paragraph 82, wherein the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN.
    • 84. The chimeric protein of paragraph 82, wherein the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences.
    • 85. The chimeric protein of paragraph 82, wherein the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189).
    • 86. The chimeric protein of any one of paragraphs 82-85, wherein the IMiD is an FDA-approved drug.
    • 87. The chimeric protein of any one of paragraphs 82-86, wherein the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide.
    • 88. The chimeric protein of any one of paragraphs 50-80, wherein the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box.
    • 89. The chimeric protein of any one of paragraphs 50-88, wherein the degron is localized C-terminal of the cell membrane tethering domain.
    • 90. A composition comprising the engineered nucleic acid of any one of paragraphs 1-46, or the expression vector of any one of paragraphs 47-49, or the membrane-cleavable chimeric protein of any one of paragraphs 50-89, and a pharmaceutically acceptable carrier.
    • 91. An isolated cell comprising the engineered nucleic acid of any one of paragraphs 1-46, or the expression vector of any one of paragraphs 47-49, or the membrane-cleavable chimeric protein of any one of paragraphs 50-89.
    • 92. An isolated cell comprising an engineered nucleic acid, wherein the engineered nucleic acid comprises an expression cassette comprising a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula:





S-C-MT-D

      • wherein
        • S comprises a secretable effector molecule,
        • C comprises a protease cleavage site,
        • MT comprises a cell membrane tethering domain, and
        • D comprises a degron,
      • wherein the promoter is operably linked to the exogenous polynucleotide sequence, and wherein S-C-MT-D is configured to be expressed as a single polypeptide.
    • 93. The isolated cell of paragraph 91 or paragraph 92, wherein the engineered nucleic acid is recombinantly expressed.
    • 94. The isolated cell of any one of paragraphs 91-93, wherein the engineered nucleic acid is expressed from a vector or a selected locus from the genome of the cell.
    • 95. An isolated cell comprising a membrane-cleavable chimeric protein, wherein the membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, has the formula:





S-C-MT-D

      • wherein
        • S comprises a secretable effector molecule,
        • C comprises a protease cleavage site,
        • MT comprises a cell membrane tethering domain,
        • D comprises a degron and
        • and wherein S-C-MT-D is configured to be expressed as a single polypeptide.
    • 96. The isolated cell of any one of paragraphs 91-95, wherein the cell is selected from the group consisting of: a T cell, a CD8+ T cell, a CD4+ T cell, a gamma-delta T cell, a cytotoxic T lymphocyte (CTL), a regulatory T cell, a viral-specific T cell, a Natural Killer T (NKT) cell, a Natural Killer (NK) cell, a B cell, a tumor-infiltrating lymphocyte (TIL), an innate lymphoid cell, a mast cell, an eosinophil, a basophil, a neutrophil, a myeloid cell, a macrophage, a monocyte, a dendritic cell, an erythrocyte, a platelet cell, a human embryonic stem cell (ESC), an ESC-derived cell, a pluripotent stem cell, a mesenchymal stromal cell (MSC), an induced pluripotent stem cell (iPSC), and an iPSC-derived cell.
    • 97. The isolated cell of any one of paragraphs 91-96, wherein the cell is autologous.
    • 98. The isolated cell of any one of paragraphs 91-96, wherein the cell is allogeneic.
    • 99. The isolated cell of any one of paragraphs 91-95, wherein the cell is a tumor cell selected from the group consisting of: a bladder tumor cell, a brain tumor cell, a breast tumor cell, a cervical tumor cell, a colorectal tumor cell, an esophageal tumor cell, a glioma cell, a kidney tumor cell, a liver tumor cell, a lung tumor cell, a melanoma cell, an ovarian tumor cell, a pancreatic tumor cell, a prostate tumor cell, a skin tumor cell, a thyroid tumor cell, and a uterine tumor cell.
    • 100. The isolated cell of paragraph 99, wherein the cell was engineered via transduction with an oncolytic virus.
    • 101. The isolated cell of paragraph 100, wherein the oncolytic virus is selected from the group consisting of: an oncolytic herpes simplex virus, an oncolytic adenovirus, an oncolytic measles virus, an oncolytic influenza virus, an oncolytic Indiana vesiculovirus, an oncolytic Newcastle disease virus, an oncolytic vaccinia virus, an oncolytic poliovirus, an oncolytic myxoma virus, an oncolytic reovirus, an oncolytic mumps virus, an oncolytic Maraba virus, an oncolytic rabies virus, an oncolytic rotavirus, an oncolytic hepatitis virus, an oncolytic rubella virus, an oncolytic dengue virus, an oncolytic chikungunya virus, an oncolytic respiratory syncytial virus, an oncolytic lymphocytic choriomeningitis virus, an oncolytic morbillivirus, an oncolytic lentivirus, an oncolytic replicating retrovirus, an oncolytic rhabdovirus, an oncolytic Seneca Valley virus, an oncolytic sindbis virus, and any variant or derivative thereof.
    • 102. The isolated cell of paragraph 100 or paragraph 101 wherein the oncolytic virus is a recombinant oncolytic virus comprising the first expression cassette and the second expression cassette.
    • 103. The isolated cell of any one of paragraphs 91-102, wherein the cell further comprises a protease capable of cleaving the protease cleavage site.
    • 104. The isolated cell of paragraph 103, wherein the protease is an endogenous protease.
    • 105. The isolated cell of paragraph 104, wherein the endogenous protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease.
    • 106. The isolated cell of paragraph 103, wherein the protease is a heterologous protease.
    • 107. The isolated cell of paragraph 106, wherein the heterologous protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3).
    • 108. The isolated cell of any one of paragraphs 103-107, wherein the protease is expressed on the cell membrane of the cell.
    • 109. The isolated cell of paragraph 108, wherein the protease is capable of cleaving the protease cleavage site.
    • 110. The isolated cell of paragraph 109, wherein cleavage of the protease cleavage site releases the secretable effector molecule from the cell membrane of the cell.
    • 111. The isolated cell of any one of paragraphs 91-110, wherein the cell further comprises an antigen recognizing receptor.
    • 112. The isolated cell of paragraph 111, wherein the antigen recognizing receptor recognizes an antigen selected from the group consisting of: 5T4, ADAM9, AFP, AXL, B7-H3, B7-H4, B7-H6, C4.4, CA6, Cadherin 3, Cadherin 6, CCR4, CD123, CD133, CD138, CD142, CD166, CD25, CD30, CD352, CD37, CD38, CD44, CD56, CD66e, CD70, CD71, CD74, CD79b, CD80, CEA, CEACAM5, Claudin18.2, cMet, CSPG4, CTLA, DLK1, DLL3, DR5, EGFR, ENPP3, EpCAM, EphA2, Ephrin A4, ETBR, FGFR2, FGFR3, FRalpha, FRb, GCC, GD2, GFRa4, gpA33, GPC3, gpNBM, GPRC5, HER2, IL-13R, IL-13Ra, IL-13Ra2, IL-8, IL-15, IL1RAP, Integrin aV, KIT, L1CAM, LAMP1, Lewis Y, LeY, LIV-1, LRRC, LY6E, MCSP, Mesothelin, MUC1, MUC16, MUC1C, NaPi2B, Nectin 4, NKG2D, NOTCH3, NY ESO 1, Ovarin, P-cadherin, pan-Erb2, PSCA, PSMA, PTK7, ROR1, S Aures, SCT, SLAMF7, SLITRK6, SSTR2, STEAP1, Survivin, TDGF1, TIM1, TROP2, and WT1.
    • 113. The isolated cell of paragraph 111 or paragraph 112, wherein the antigen recognizing receptor comprises an antigen-binding domain.
    • 114. The isolated cell of paragraph 113, wherein the antigen-binding domain comprises an antibody, an antigen-binding fragment of an antibody, a F(ab) fragment, a F(ab′) fragment, a single chain variable fragment (scFv), or a single-domain antibody (sdAb).
    • 115. The isolated cell of paragraph 113, wherein the antigen-binding domain comprises a single chain variable fragment (scFv).
    • 116. The isolated cell of paragraph 115, wherein the scFv comprises a heavy chain variable domain (VH) and a light chain variable domain (VL).
    • 117. The isolated cell of paragraph 116, wherein the VH and VL are separated by a peptide linker.
    • 118. The isolated cell of paragraph 117, wherein the scFv comprises the structure VH-L-VL or VL-L-VH, wherein VH is the heavy chain variable domain, L is the peptide linker, and VL is the light chain variable domain.
    • 119. The isolated cell of any one of paragraphs 111-118, wherein the antigen recognizing receptor is a chimeric antigen receptor (CAR) or T cell receptor (TCR).
    • 120. The isolated cell of any one of paragraphs 111-118, wherein the antigen recognizing receptor is a CAR.
    • 121. The isolated cell of paragraph 120, wherein the CAR comprises one or more intracellular signaling domains, and the one or more intracellular signaling domains are selected from the group consisting of: a CD3zeta-chain intracellular signaling domain, a CD97 intracellular signaling domain, a CD11a-CD18 intracellular signaling domain, a CD2 intracellular signaling domain, an ICOS intracellular signaling domain, a CD27 intracellular signaling domain, a CD154 intracellular signaling domain, a CD8 intracellular signaling domain, an OX40 intracellular signaling domain, a 4-1BB intracellular signaling domain, a CD28 intracellular signaling domain, a ZAP40 intracellular signaling domain, a CD30 intracellular signaling domain, a GJTR intracellular signaling domain, an HVEM intracellular signaling domain, a DAP10 intracellular signaling domain, a DAP12 intracellular signaling domain, and a MyD88 intracellular signaling domain.
    • 122. The isolated cell of paragraph 120 or paragraph 121, wherein the CAR comprises a transmembrane domain, and the transmembrane domain is selected from the group consisting of: a CD8 transmembrane domain, a CD28 transmembrane domain a CD3zeta-chain transmembrane domain, a CD4 transmembrane domain, a 4-1BB transmembrane domain, an OX40 transmembrane domain, an ICOS transmembrane domain, a CTLA-4 transmembrane domain, a PD-1 transmembrane domain, a LAG-3 transmembrane domain, a 2B4 transmembrane domain, and a BTLA transmembrane domain.
    • 123. The isolated cell of any one of paragraphs 120-122, wherein the CAR comprises a spacer region between the antigen-binding domain and the transmembrane domain.
    • 124. A composition comprising the isolated cell of any one of paragraphs 91-123, and a pharmaceutically acceptable carrier.
    • 125. A method of treating a subject in need thereof, the method comprising administering a therapeutically effective dose of any of the isolated cells of any one of paragraphs 91-123 or the composition of paragraph 124.
    • 126. The method of paragraph 125, wherein the isolated cell is derived from the subject.
    • 127. The method of paragraph 125 or paragraph 126, wherein the isolated cell is allogeneic with reference to the subject.
    • 128. The method of any one of paragraphs 125-127, wherein the method further comprises administering a checkpoint inhibitor.
    • 129. The method of paragraph 128, wherein the checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody.
    • 130. The method of any one of paragraphs 125-129, wherein the method further comprises administering an anti-CD40 antibody.
    • 131. A lipid-based structure comprising the engineered nucleic acid of any one of paragraphs 1-46, or the expression vector of any one of paragraphs 47-49, or the membrane-cleavable chimeric protein of any one of paragraphs 50-89.
    • 132. The lipid-based structure of paragraph 131, wherein the lipid-based structure comprises a extracellular vesicle.
    • 133. The lipid-based structure of paragraph 132, wherein the extracellular vesicle is selected from the group consisting of: a nanovesicle and an exosome.
    • 134. The lipid-based structure of paragraph 131, wherein the lipid-based structure comprises a lipid nanoparticle or a micelle.
    • 135. The lipid-based structure of paragraph 131, wherein the lipid-based structure comprises a liposome.
    • 136. A composition comprising the lipid-based structure of any one of paragraphs 131-135, and a pharmaceutically acceptable carrier.
    • 137. A method of treating a subject in need thereof, the method comprising administering a therapeutically effective dose of any of the lipid-based structures of any one of paragraphs 131-135 or the composition of paragraph 136.
    • 138. The method of paragraph 137, wherein the administering comprises systemic administration.
    • 139. The method of paragraph 137 or paragraph 138, wherein the lipid-based structure is capable of engineering a cell in the subject.
    • 140. The method of any one of paragraphs 137-139, wherein the method further comprises administering a checkpoint inhibitor.
    • 141. The method of paragraph 140, wherein the checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody.
    • 142. The method of any one of paragraphs 137-141, wherein the method further comprises administering an anti-CD40 antibody.
    • 143. A nanoparticle comprising the engineered nucleic acid of any one of paragraphs 1-46 or the membrane-cleavable chimeric protein of any one of paragraphs 50-89.
    • 144. The nanoparticle of paragraph 143, wherein the nanoparticle comprises an inorganic material.
    • 145. A composition comprising the nanoparticle of paragraph 143 or paragraph 144.
    • 146. A method of treating a subject in need thereof, the method comprising administering a therapeutically effective dose of any of the nanoparticles of paragraph 143 or paragraph 144, or the composition of paragraph 145.
    • 147. The method of paragraph 146, wherein the administering comprises systemic administration.
    • 148. The method of paragraph 146 or paragraph 147, wherein the nanoparticle is capable of engineering a cell in the subject.
    • 149. The method of any one of paragraphs 146-148, wherein the method further comprises administering a checkpoint inhibitor.
    • 150. The method of paragraph 149, wherein the checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody.
    • 151. The method of any one of paragraphs 146-150, wherein the method further comprises administering an anti-CD40 antibody.
    • 152. A virus engineered to comprise the engineered nucleic acid of any one of paragraphs 1-46 or the expression vector of any one of paragraphs 47-49.
    • 153. The engineered virus of paragraph 152, wherein the virus is selected from the group consisting of: a lentivirus, a retrovirus, an oncolytic virus, an adenovirus, an adeno-associated virus (AAV), and a virus-like particle (VLP).
    • 154. A composition comprising the engineered virus of paragraph 152 or paragraph 153, and a pharmaceutically acceptable carrier.
    • 155. A method of treating a subject in need thereof, the method comprising administering a therapeutically effective dose of the engineered virus of paragraph 152 or paragraph 153, or the composition of paragraph 154.
    • 156. The method of paragraph 155, wherein the administering comprises systemic administration.
    • 157. The method of paragraph 155 or paragraph 156, wherein the engineered virus infects a cell in the subject and expresses the expression cassette.
    • 158. The method of any one of paragraphs 155-157, wherein the method further comprises administering a checkpoint inhibitor.
    • 159. The method of paragraph 158, wherein the checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody.
    • 160. The method of any one of paragraphs 155-159, wherein the method further comprises administering an anti-CD40 antibody.
    • 161. A method of inducing release of a membrane-tethered effector molecule, comprising:
      • a) providing a cell, wherein the cell comprises a membrane-bound protease and a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula:





S-C-MT-D

      • wherein
        • S comprises an effector molecule,
        • C comprises a cognate membrane-bound protease cleavage site,
        • MT comprises a cell membrane tethering domain,
        • D comprises a degron, and
        • wherein S-C-MT-D is configured to be expressed as a single polypeptide; and
      • b) culturing the cell under conditions suitable for expression of the membrane-bound protease and the membrane-cleavable chimeric protein, wherein upon expression, the membrane-cleavable chimeric protein is tethered to the cell membrane of the cell, and
        • wherein, upon expression, the membrane-bound protease cleaves the cognate membrane-bound protease cleavage site of the membrane-cleavable chimeric protein, thereby releasing the effector molecule from the cell membrane.
    • 162. The method of paragraph 161, wherein the degron is a drug-inducible degron.
    • 163. The method of paragraph 162, further comprising contacting the cell with a drug that induces the degron.
    • 164. The method of paragraph 163, wherein induction of the degron degrades the membrane-cleavable chimeric protein.
    • 165. A method of degrading a membrane-tethered effector molecule, comprising:
      • a) providing a cell, wherein the cell comprises a membrane-bound protease and a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula:





S-C-MT-D

      • wherein
        • S comprises an effector molecule,
        • C comprises a cognate membrane-bound protease cleavage site,
        • MT comprises a cell membrane tethering domain,
        • D comprises a drug-inducible degron, and
        • wherein S-C-MT-D is configured to be expressed as a single polypeptide;
      • b) culturing the cell under conditions suitable for expression of the membrane-bound protease and the membrane-cleavable chimeric protein, wherein upon expression, the membrane-cleavable chimeric protein is tethered to the cell membrane of the cell; and
      • c) inducing the degron by contacting the cell with a drug, wherein degron induction of the degron degrades the membrane-cleavable chimeric protein.
    • 166. The method of any one of paragraphs 161-165, wherein the effector molecule comprises a signal peptide.
    • 167. The method of paragraph 166, wherein the signal peptide comprises a native signal peptide native to the effector molecule.
    • 168. The method of paragraph 166, wherein the signal peptide comprises a non-native signal peptide non-native to the effector molecule.
    • 169. The method of paragraph 168, wherein the non-native signal peptide is selected from the group consisting of: IL12, IL2, optimized IL2, trypsiongen-2, Gaussia luciferase, CD5, human IgKVII, murine IgKVII, VSV-G, prolactin, serum albumin preprotein, azurocidin preprotein, osteonectin, CD33, IL6, IL8, CCL2, TIMP2, VEGFB, osteoprotegerin, serpin E1, GROalpha, CXCL12, IL21, CD8, and GMCSF.
    • 170. The method of any one of paragraphs 161-169, wherein the effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a polynucleotide, a peptide, and an enzyme.
    • 171. The method of paragraph 170, wherein the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha.
    • 172. The method of paragraph 170, wherein the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1.
    • 173. The method of paragraph 170, wherein the homing molecule is selected from the group consisting of: anti-integrin alpha4,beta7; anti-MAdCAM; SDF1; and MMP-2.
    • 174. The method of paragraph 170, wherein the growth factor is selected from the group consisting of: FLT3L and GM-CSF.
    • 175. The method of paragraph 170, wherein the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40L.
    • 176. The method of paragraph 170, wherein the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2.
    • 177. The method of paragraph 176, wherein the TGFbeta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereof.
    • 178. The method of paragraph 176, wherein the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody.
    • 179. The method of paragraph 176, wherein the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof.
    • 180. The method of any one of paragraphs 161-175, wherein the effector molecule is a human-derived effector molecule.
    • 181. The method of any one of paragraphs 161-180, wherein the cognate protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site.
    • 182. The method of any one of paragraphs 161-181, wherein the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain.
    • 183. The method of paragraph 182, wherein the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA.
    • 184. The method of any one of paragraphs 161-181, wherein the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof.
    • 185. The method of any one of paragraphs 161-184, wherein the protease is endogenous to the cell.
    • 186. The method of paragraph 185, wherein the protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease.
    • 187. The method of any one of paragraphs 161-184, wherein the protease is heterologous to the cell.
    • 188. The method of paragraph 187, wherein the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3).
    • 189. The method of paragraph 188, wherein the protease cleavage site comprises an NS3 protease cleavage site.
    • 190. The method of paragraph 189, wherein the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site.
    • 191. The method of any one of paragraphs 161-190, wherein the protease can be repressed by a protease inhibitor.
    • 192. The method of paragraph 191, wherein the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir.
    • 193. The method of any one of paragraphs 161-192, wherein the protease further comprises a degron.
    • 194. The method of any one of paragraphs 161-193, wherein expression of the protease is capable of regulation.
    • 195. The method of any one of paragraphs 161-194, wherein the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule.
    • 196. The method of paragraph 195, wherein the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN.
    • 197. The method of paragraph 195, wherein the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences.
    • 198. The method of paragraph 195, wherein the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189).
    • 199. The method of any one of paragraphs 195-198, wherein the IMiD is an FDA-approved drug.
    • 200. The method of any one of paragraphs 195-199, wherein the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide.
    • 201. The method of any one of paragraphs 161-193, wherein the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box.
    • 202. The method of any one of paragraphs 161-201, wherein the degron is localized C-terminal of the cell membrane tethering domain.
    • 203. An adeno-associated virus (AAV) engineered to express a heterologous nucleic acid, wherein the heterologous nucleic acid encodes a degron-fusion chimeric protein, and wherein the degron-fusion chimeric protein comprises a degron fused to a protein of interest.
    • 204. The engineered virus of paragraph 203, wherein the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the protein of interest.
    • 205. The engineered virus of paragraph 204, wherein the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN.
    • 206. The engineered virus of paragraph 204, wherein the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences.
    • 207. The engineered virus of paragraph 204, wherein the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189).
    • 208. The engineered virus of any one of paragraphs 204-207, wherein the IMiD is an FDA-approved drug.
    • 209. The engineered virus of any one of paragraphs 204-208, wherein the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide.
    • 210. The engineered virus of paragraph 203, wherein the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box.
    • 211. The engineered virus of any one of paragraphs 203-210, wherein the AAV is selected from the group consisting of: AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV.Rh10, AAV11, and variants thereof.
    • 212. The engineered virus of any one of paragraphs 203-211, wherein the protein of interest is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a polynucleotide, a peptide, and an enzyme.
    • 213. An adeno-associated viral (AAV) vector comprising a heterologous nucleic acid, wherein the heterologous nucleic acid encodes a degron-fusion chimeric protein, and wherein the degron-fusion chimeric protein comprises a degron fused to a protein of interest.
    • 214. The AAV vector of paragraph 213, wherein the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the protein of interest.
    • 215. The AAV vector of paragraph 214, wherein the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN.
    • 216. The AAV vector of paragraph 214, wherein the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences.
    • 217. The AAV vector of paragraph 214, wherein the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189).
    • 218. The AAV vector of any one of paragraphs 213-217, wherein the IMiD is an FDA-approved drug.
    • 219. The AAV vector of any one of paragraphs 213-218, wherein the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide.
    • 220. The AAV vector of paragraph 213, wherein the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box.
    • 221. The AAV vector of any one of paragraphs 213-220, wherein the AAV is selected from the group consisting of: AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV.Rh10, AAV11, and variants thereof.
    • 222. The AAV vector of any one of paragraphs 213-221, wherein the protein of interest is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a polynucleotide, a peptide, and an enzyme.
    • 223. A pharmaceutical composition comprising the engineered virus of any one of paragraphs 203-212 or the AAV vector of any one of paragraphs 213-222 and a pharmaceutically acceptable carrier.
    • 224. An isolated cell comprising the engineered virus of any one of paragraphs 203-212 or the AAV vector of any one of paragraphs 213-222.
    • 225. A method for in vivo regulated protein expression, comprising administering to a subject in need thereof the engineered virus of any one of paragraphs 203-212 or the AAV vector of any one of paragraphs 213-222.
    • 226. A method for regulating protein expression, comprising contacting a cell with the engineered virus of any one of paragraphs 203-212 or the AAV vector of any one of paragraphs 213-222 under conditions suitable for expression of the engineered virus or AAV vector.


EXAMPLES

Below are examples of specific embodiments for carrying out the present invention. The examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way. For example, the experiments described and performed below demonstrate the general utility of engineering cells to secrete payloads (e.g., effector molecules) and delivering those cells to induce an immunogenic response against tumors. Specifically, the examples described below demonstrate the ability to induce an immunogenic response against tumors through delivery of engineered mesenchymal stem cells (MSCs) secreting a variety of the effector molecules described herein. The use of MSCs is not intended to limit the scope of the present invention directed to engineering the cells described herein.


Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperatures, etc.), but some experimental error and deviation should, of course, be allowed for.


Example 1—Degron Domain Alone Does Not Regulate Secretion

The ability to regulate secretion of a molecule through attachment of a degron domain was assessed.


Methods

A series of pL17d-based viral vector constructs were generated using Secreted Alkaline Phosphatase (SEAP) fused to a degron (d913 super degron) using various linkers, as described in Table 6A with sequences listed in Table 6B. The d913 super degron is described in more detail in International Application Pub. No. WO2019/089592A1, herein incorporated by reference for all purposes.


SEAP assays were performed using the QuantiBlue kit (Invivogen; rep-qb1). For experiments performed in HEK293Ft cells, the following protocol was followed:

    • Cells were plated at 2e5 cells/well in a 12 well plate on Day 0
    • On Day 1, cells were transduced with 300K GS units of virus
    • On Day 2, cells were split into 2 wells in 6 well plates+/−1 μM lenalidomide
    • On Day 4, cells were passaged and lenalidomide treatment was continued
    • On Day 7, 20 uL supernatant from each well was harvested and incubated with 180 uL QuantiBlue (Invivogen) for 1 h at 37 deg, SEAP activity was read out on a plate reader at 655 nm (Cells are adherent so do not need to be pelleted from the media)


      For experiments performed in donor-derived T cells, the following protocol was followed:
    • T cells were enriched from leukapheresis material using anti-CD4 and anti-CD8 magnetic beads at a targeted 1:1 ratio on the Miltenyi Biotec CliniMACS Prodigy instrument. The CliniMACS Prodigy utilizes a closed-system, single-use disposable kits for many different cell types. The automated program performs platelet washes, CD4+& CD8+ bead labeling, and positive selection over a magnet resulting in cell suspensions with high purity. Positively selected T cells were then sampled, analyzed for cell surface phenotype via flow cytometry and cryopreserved via a controlled rate freezer.
    • Cells were then thawed and activated on Day 0 (1e6 cells/mL in a 24 well plate were activated 3:1 with CD3/CD28 Dynabeads)
    • On Day 1, 100K GS units of virus were added to cells
    • On Day 2, fresh media was added to T cells
    • On Day 4, each well was split into 2 wells of 6 well plate with +/−1 μM lenalidomide
    • On Day 7, cells were pelleted and 20 uL supernatant from each well was harvested and incubated with 180 uL QuantiBlue (Invivogen) for 1 h at 37 deg, SEAP activity was read out on a plate reader at 655 nm









TABLE 6A







SEAP/Degron Construct Configuration










Construct
Description







SB01118
SEAP-(GS)4 GG (SEQ ID NO: 182)-d913 super




degron



SB01141
SEAP-A(EAAAK)3A (SEQ ID NO: 183)-d913




super degron



SB01143
SEAP-KEGS (SEQ ID NO: 195) Whitlow linker-




d913 super degron



SB01144
SEAP-EGK Whitlow linker-d913 super degron



SB00419
SEAP

















TABLE 6B





SEAP/Degron Construct Sequences


Sequences















SB01118 (SEQ ID NO: 175)


ATGTGGGGTGCCTGTCTGCTATTGCTGGGCTTAAGTCTTCAAGTTTGCCCCAGTGTCATTCC


TGTGGAGGAGGAGAATCCTGCTTTTTGGAATAGGAAGGCAGCTGAAGCCTTGGATGCAGC


CAAGAAGCTCAAGCCCATTCAGACATCTGCAAAGAATCTTGTCATCCTCATGGGTGATGGA


ATGGGTGTCTCCACTGTAACAGCCACCAGGATTCTGAAGGGCCAGCAACAAGGTCATCTAG


GCCCAGAGACCCAGTTGGCAATGGACAGGTTCCCTCACATGGCCCTTTCCAAGACTTACAA


CACTGACAAGCAGATTCCTGACTCTGCTGGGACAGGCACAGCATTCTTGTGTGGAGTAAAA


ACCAACATGAAAGTCATTGGTCTTTCAGCTGCTGCCAGATTCAACCAGTGCAACACCACAT


GGGGCAATGAAGTGGTCTCTGTAATGCACAGGGCCAAAAAAGCTGGGAAAAGTGTGGGTG


TGGTGACAACCACCTCTGTCCAGCATGCCTCTCCTGCTGGAACTTATGCCCACACAGTGAA


CAGAGGTTGGTACTCTGATGCTCAGATGCCTGCCTCAGCTTTACAAGATGGCTGCAAGGAC


ATCAGCACCCAGCTCATCTCAAACATGGACATAGATGTCATCTTAGGGGGTGGGAGAAAG


TTCATGTTCCCAAAGGGGACTCCTGACCAGGAGTACCCCACAGACACAAAGCAGGCTGGC


ACAAGATTAGATGGTAGGAACCTTGTGCAAGAGTGGCTTGCCAAGCATCAGGGAGCAAGG


TATGTCTGGAACAGGAGTGAGCTAATCCAGGCCTCTTTGAACAGGTCTGTCACTCACCTAA


TGGGGTTATTTGAGCCCAATGACATGAAGTATGAGATACACAGGGACCCTGCCCAGGACC


CCTCCTTAGCAGAAATGACTGAAGTTGCTGTGAGGATGTTGTCCAGAAATCCAAAAGGGTT


CTACCTCTTTGTTGAGGGGGGAAGGATTGATCATGGTCACCATGAGACAGTTGCTTACAGA


GCCTTAACTGAGGCTGTGATGTTTGATTCTGCTGTGGACAAGGCTGACAAACTGACCTCTG


AGCAGGACACAATGATTCTAGTGACTGCTGACCACAGTCATGTTTTCTCCTTTGGGGGCTA


CACCCAGAGGGGTGCTTCAATCTTTGGCCTGGCCCCTTTCAAGGCAGAAGATGGGAAGAGT


TTCACCTCCATCCTCTATGGGAATGGTCCTGGGTACAAGCTGCACAATGGGGCCAGAGCTG


ATGTGACAGAAGAGGAGAGCTCCAACCCAACCTACCAGCAGCAAGCAGCAGTCCCTCTTT


CTTCAGAAACCCACTCTGGGGAAGATGTGGCCATATTTGCCAGAGGCCCCCAAGCCCACTT


GGTGCATGGAGTTCAGGAGCAGAATTACATAGCTCATGTAATGGCTTTTGCTGCTTGCTTG


GAGCCCTACACAGACTGTGGCCTAGCCAGCCCAGCAGGCCAGTCCTCTGCAGTAAGCCCA


GGCGGCAGTGGCTCGGGCTCGGGGTCCGGCGGATTCAATGTCTTAATGGTTCATAAGCGAA


GCCATACTGGTGAACGCCCATTGCAGTGCGAAATATGCGGCTTTACCTGCCGCCAGAAAGG


TAACCTCCTCCGCCACATTAAACTGCACACAGGGGAAAAACCTTTTAAGTGTCACCTCTGC


AACTATGCATGCCAAAGAAGAGATGCGCTCTAG





SB01141 (SEQ ID NO: 176)


ATGTGGGGTGCCTGTCTGCTATTGCTGGGCTTAAGTCTTCAAGTTTGCCCCAGTGTCATTCC


TGTGGAGGAGGAGAATCCTGCTTTTTGGAATAGGAAGGCAGCTGAAGCCTTGGATGCAGC


CAAGAAGCTCAAGCCCATTCAGACATCTGCAAAGAATCTTGTCATCCTCATGGGTGATGGA


ATGGGTGTCTCCACTGTAACAGCCACCAGGATTCTGAAGGGCCAGCAACAAGGTCATCTAG


GCCCAGAGACCCAGTTGGCAATGGACAGGTTCCCTCACATGGCCCTTTCCAAGACTTACAA


CACTGACAAGCAGATTCCTGACTCTGCTGGGACAGGCACAGCATTCTTGTGTGGAGTAAAA


ACCAACATGAAAGTCATTGGTCTTTCAGCTGCTGCCAGATTCAACCAGTGCAACACCACAT


GGGGCAATGAAGTGGTCTCTGTAATGCACAGGGCCAAAAAAGCTGGGAAAAGTGTGGGTG


TGGTGACAACCACCTCTGTCCAGCATGCCTCTCCTGCTGGAACTTATGCCCACACAGTGAA


CAGAGGTTGGTACTCTGATGCTCAGATGCCTGCCTCAGCTTTACAAGATGGCTGCAAGGAC


ATCAGCACCCAGCTCATCTCAAACATGGACATAGATGTCATCTTAGGGGGTGGGAGAAAG


TTCATGTTCCCAAAGGGGACTCCTGACCAGGAGTACCCCACAGACACAAAGCAGGCTGGC


ACAAGATTAGATGGTAGGAACCTTGTGCAAGAGTGGCTTGCCAAGCATCAGGGAGCAAGG


TATGTCTGGAACAGGAGTGAGCTAATCCAGGCCTCTTTGAACAGGTCTGTCACTCACCTAA


TGGGGTTATTTGAGCCCAATGACATGAAGTATGAGATACACAGGGACCCTGCCCAGGACC


CCTCCTTAGCAGAAATGACTGAAGTTGCTGTGAGGATGTTGTCCAGAAATCCAAAAGGGTT


CTACCTCTTTGTTGAGGGGGGAAGGATTGATCATGGTCACCATGAGACAGTTGCTTACAGA


GCCTTAACTGAGGCTGTGATGTTTGATTCTGCTGTGGACAAGGCTGACAAACTGACCTCTG


AGCAGGACACAATGATTCTAGTGACTGCTGACCACAGTCATGTTTTCTCCTTTGGGGGCTA


CACCCAGAGGGGTGCTTCAATCTTTGGCCTGGCCCCTTTCAAGGCAGAAGATGGGAAGAGT


TTCACCTCCATCCTCTATGGGAATGGTCCTGGGTACAAGCTGCACAATGGGGCCAGAGCTG


ATGTGACAGAAGAGGAGAGCTCCAACCCAACCTACCAGCAGCAAGCAGCAGTCCCTCTTT


CTTCAGAAACCCACTCTGGGGAAGATGTGGCCATATTTGCCAGAGGCCCCCAAGCCCACTT


GGTGCATGGAGTTCAGGAGCAGAATTACATAGCTCATGTAATGGCTTTTGCTGCTTGCTTG


GAGCCCTACACAGACTGTGGCCTAGCCAGCCCAGCAGGCCAGTCCTCTGCAGTAAGCCCA


GGCGCCGAGGCGGCAGCAAAGGAAGCAGCAGCGAAAGAGGCGGCTGCCAAAGCGTTCAA


TGTCTTAATGGTTCATAAGCGAAGCCATACTGGTGAACGCCCATTGCAGTGCGAAATATGC


GGCTTTACCTGCCGCCAGAAAGGTAACCTCCTCCGCCACATTAAACTGCACACAGGGGAAA


AACCTTTTAAGTGTCACCTCTGCAACTATGCATGCCAAAGAAGAGATGCGCTCTAA





SB01143 (SEQ ID NO: 177)


ATGTGGGGTGCCTGTCTGCTATTGCTGGGCTTAAGTCTTCAAGTTTGCCCCAGTGTCATTCC


TGTGGAGGAGGAGAATCCTGCTTTTTGGAATAGGAAGGCAGCTGAAGCCTTGGATGCAGC


CAAGAAGCTCAAGCCCATTCAGACATCTGCAAAGAATCTTGTCATCCTCATGGGTGATGGA


ATGGGTGTCTCCACTGTAACAGCCACCAGGATTCTGAAGGGCCAGCAACAAGGTCATCTAG


GCCCAGAGACCCAGTTGGCAATGGACAGGTTCCCTCACATGGCCCTTTCCAAGACTTACAA


CACTGACAAGCAGATTCCTGACTCTGCTGGGACAGGCACAGCATTCTTGTGTGGAGTAAAA


ACCAACATGAAAGTCATTGGTCTTTCAGCTGCTGCCAGATTCAACCAGTGCAACACCACAT


GGGGCAATGAAGTGGTCTCTGTAATGCACAGGGCCAAAAAAGCTGGGAAAAGTGTGGGTG


TGGTGACAACCACCTCTGTCCAGCATGCCTCTCCTGCTGGAACTTATGCCCACACAGTGAA


CAGAGGTTGGTACTCTGATGCTCAGATGCCTGCCTCAGCTTTACAAGATGGCTGCAAGGAC


ATCAGCACCCAGCTCATCTCAAACATGGACATAGATGTCATCTTAGGGGGTGGGAGAAAG


TTCATGTTCCCAAAGGGGACTCCTGACCAGGAGTACCCCACAGACACAAAGCAGGCTGGC


ACAAGATTAGATGGTAGGAACCTTGTGCAAGAGTGGCTTGCCAAGCATCAGGGAGCAAGG


TATGTCTGGAACAGGAGTGAGCTAATCCAGGCCTCTTTGAACAGGTCTGTCACTCACCTAA


TGGGGTTATTTGAGCCCAATGACATGAAGTATGAGATACACAGGGACCCTGCCCAGGACC


CCTCCTTAGCAGAAATGACTGAAGTTGCTGTGAGGATGTTGTCCAGAAATCCAAAAGGGTT


CTACCTCTTTGTTGAGGGGGGAAGGATTGATCATGGTCACCATGAGACAGTTGCTTACAGA


GCCTTAACTGAGGCTGTGATGTTTGATTCTGCTGTGGACAAGGCTGACAAACTGACCTCTG


AGCAGGACACAATGATTCTAGTGACTGCTGACCACAGTCATGTTTTCTCCTTTGGGGGCTA


CACCCAGAGGGGTGCTTCAATCTTTGGCCTGGCCCCTTTCAAGGCAGAAGATGGGAAGAGT


TTCACCTCCATCCTCTATGGGAATGGTCCTGGGTACAAGCTGCACAATGGGGCCAGAGCTG


ATGTGACAGAAGAGGAGAGCTCCAACCCAACCTACCAGCAGCAAGCAGCAGTCCCTCTTT


CTTCAGAAACCCACTCTGGGGAAGATGTGGCCATATTTGCCAGAGGCCCCCAAGCCCACTT


GGTGCATGGAGTTCAGGAGCAGAATTACATAGCTCATGTAATGGCTTTTGCTGCTTGCTTG


GAGCCCTACACAGACTGTGGCCTAGCCAGCCCAGCAGGCCAGTCCTCTGCAGTAAGCCCA


GGCAAGGAAAGCGGGTCCGTAAGTTCAGAGCAGTTGGCTCAGTTTCGGTCACTTGACTTCA


ATGTCTTAATGGTTCATAAGCGAAGCCATACTGGTGAACGCCCATTGCAGTGCGAAATATG


CGGCTTTACCTGCCGCCAGAAAGGTAACCTCCTCCGCCACATTAAACTGCACACAGGGGAA


AAACCTTTTAAGTGTCACCTCTGCAACTATGCATGCCAAAGAAGAGATGCGCTCTAA





SB01144 (SEQ ID NO: 178)


ATGTGGGGTGCCTGTCTGCTATTGCTGGGCTTAAGTCTTCAAGTTTGCCCCAGTGTCATTCC


TGTGGAGGAGGAGAATCCTGCTTTTTGGAATAGGAAGGCAGCTGAAGCCTTGGATGCAGC


CAAGAAGCTCAAGCCCATTCAGACATCTGCAAAGAATCTTGTCATCCTCATGGGTGATGGA


ATGGGTGTCTCCACTGTAACAGCCACCAGGATTCTGAAGGGCCAGCAACAAGGTCATCTAG


GCCCAGAGACCCAGTTGGCAATGGACAGGTTCCCTCACATGGCCCTTTCCAAGACTTACAA


CACTGACAAGCAGATTCCTGACTCTGCTGGGACAGGCACAGCATTCTTGTGTGGAGTAAAA


ACCAACATGAAAGTCATTGGTCTTTCAGCTGCTGCCAGATTCAACCAGTGCAACACCACAT


GGGGCAATGAAGTGGTCTCTGTAATGCACAGGGCCAAAAAAGCTGGGAAAAGTGTGGGTG


TGGTGACAACCACCTCTGTCCAGCATGCCTCTCCTGCTGGAACTTATGCCCACACAGTGAA


CAGAGGTTGGTACTCTGATGCTCAGATGCCTGCCTCAGCTTTACAAGATGGCTGCAAGGAC


ATCAGCACCCAGCTCATCTCAAACATGGACATAGATGTCATCTTAGGGGGTGGGAGAAAG


TTCATGTTCCCAAAGGGGACTCCTGACCAGGAGTACCCCACAGACACAAAGCAGGCTGGC


ACAAGATTAGATGGTAGGAACCTTGTGCAAGAGTGGCTTGCCAAGCATCAGGGAGCAAGG


TATGTCTGGAACAGGAGTGAGCTAATCCAGGCCTCTTTGAACAGGTCTGTCACTCACCTAA


TGGGGTTATTTGAGCCCAATGACATGAAGTATGAGATACACAGGGACCCTGCCCAGGACC


CCTCCTTAGCAGAAATGACTGAAGTTGCTGTGAGGATGTTGTCCAGAAATCCAAAAGGGTT


CTACCTCTTTGTTGAGGGGGGAAGGATTGATCATGGTCACCATGAGACAGTTGCTTACAGA


GCCTTAACTGAGGCTGTGATGTTTGATTCTGCTGTGGACAAGGCTGACAAACTGACCTCTG


AGCAGGACACAATGATTCTAGTGACTGCTGACCACAGTCATGTTTTCTCCTTTGGGGGCTA


CACCCAGAGGGGTGCTTCAATCTTTGGCCTGGCCCCTTTCAAGGCAGAAGATGGGAAGAGT


TTCACCTCCATCCTCTATGGGAATGGTCCTGGGTACAAGCTGCACAATGGGGCCAGAGCTG


ATGTGACAGAAGAGGAGAGCTCCAACCCAACCTACCAGCAGCAAGCAGCAGTCCCTCTTT


CTTCAGAAACCCACTCTGGGGAAGATGTGGCCATATTTGCCAGAGGCCCCCAAGCCCACTT


GGTGCATGGAGTTCAGGAGCAGAATTACATAGCTCATGTAATGGCTTTTGCTGCTTGCTTG


GAGCCCTACACAGACTGTGGCCTAGCCAGCCCAGCAGGCCAGTCCTCTGCAGTAAGCCCA


GGCGAGGGGAAAAGCAGTGGATCAGGCAGCGAAAGCAAGTCTACTTTCAATGTCTTAATG


GTTCATAAGCGAAGCCATACTGGTGAACGCCCATTGCAGTGCGAAATATGCGGCTTTACCT


GCCGCCAGAAAGGTAACCTCCTCCGCCACATTAAACTGCACACAGGGGAAAAACCTTTTA


AGTGTCACCTCTGCAACTATGCATGCCAAAGAAGAGATGCGCTCTAA





SB00419 (SEQ ID NO: 179)


ATGTGGGGTGCCTGTCTGCTATTGCTGGGCTTAAGTCTTCAAGTTTGCCCCAGTGTCATTCC


TGTGGAGGAGGAGAATCCTGCTTTTTGGAATAGGAAGGCAGCTGAAGCCTTGGATGCAGC


CAAGAAGCTCAAGCCCATTCAGACATCTGCAAAGAATCTTGTCATCCTCATGGGTGATGGA


ATGGGTGTCTCCACTGTAACAGCCACCAGGATTCTGAAGGGCCAGCAACAAGGTCATCTAG


GCCCAGAGACCCAGTTGGCAATGGACAGGTTCCCTCACATGGCCCTTTCCAAGACTTACAA


CACTGACAAGCAGATTCCTGACTCTGCTGGGACAGGCACAGCATTCTTGTGTGGAGTAAAA


ACCAACATGAAAGTCATTGGTCTTTCAGCTGCTGCCAGATTCAACCAGTGCAACACCACAT


GGGGCAATGAAGTGGTCTCTGTAATGCACAGGGCCAAAAAAGCTGGGAAAAGTGTGGGTG


TGGTGACAACCACCTCTGTCCAGCATGCCTCTCCTGCTGGAACTTATGCCCACACAGTGAA


CAGAGGTTGGTACTCTGATGCTCAGATGCCTGCCTCAGCTTTACAAGATGGCTGCAAGGAC


ATCAGCACCCAGCTCATCTCAAACATGGACATAGATGTCATCTTAGGGGGTGGGAGAAAG


TTCATGTTCCCAAAGGGGACTCCTGACCAGGAGTACCCCACAGACACAAAGCAGGCTGGC


ACAAGATTAGATGGTAGGAACCTTGTGCAAGAGTGGCTTGCCAAGCATCAGGGAGCAAGG


TATGTCTGGAACAGGAGTGAGCTAATCCAGGCCTCTTTGAACAGGTCTGTCACTCACCTAA


TGGGGTTATTTGAGCCCAATGACATGAAGTATGAGATACACAGGGACCCTGCCCAGGACC


CCTCCTTAGCAGAAATGACTGAAGTTGCTGTGAGGATGTTGTCCAGAAATCCAAAAGGGTT


CTACCTCTTTGTTGAGGGGGGAAGGATTGATCATGGTCACCATGAGACAGTTGCTTACAGA


GCCTTAACTGAGGCTGTGATGTTTGATTCTGCTGTGGACAAGGCTGACAAACTGACCTCTG


AGCAGGACACAATGATTCTAGTGACTGCTGACCACAGTCATGTTTTCTCCTTTGGGGGCTA


CACCCAGAGGGGTGCTTCAATCTTTGGCCTGGCCCCTTTCAAGGCAGAAGATGGGAAGAGT


TTCACCTCCATCCTCTATGGGAATGGTCCTGGGTACAAGCTGCACAATGGGGCCAGAGCTG


ATGTGACAGAAGAGGAGAGCTCCAACCCAACCTACCAGCAGCAAGCAGCAGTCCCTCTTT


CTTCAGAAACCCACTCTGGGGAAGATGTGGCCATATTTGCCAGAGGCCCCCAAGCCCACTT


GGTGCATGGAGTTCAGGAGCAGAATTACATAGCTCATGTAATGGCTTTTGCTGCTTGCTTG


GAGCCCTACACAGACTGTGGCCTAGCCAGCCCAGCAGGCCAGTCCTCTGCAGTAAGCCCA


GGCtaG









Results

A SEAP assay was designed to test the efficacy of a degron domain's ability to regulate secretion of molecules through assessing secreted SEAP activity in conditioned media (free of cells). A CRBN-based degron system was used in which lenalidomide-dependent degradation has been shown in regulated degradation of membrane-associated proteins (WO2019/089592A1). As shown in FIG. 1, SEAP enzymatic activity was determined in supernatants from both HEK293Ft cells (left panel) and donor-derived T cells (right panel). SEAP enzymatic activity was quantified as a percentage of “Off” (+lenalidomide) relative to “On” (no lenalidomide) and is presented in Table 7A (HEK) and Table 7B (T Cells). Additionally, quantification of SEAP enzymatic activity was also normalized to cell number by dividing SEAP absorbance by cell count per well (from Countess), see Table 7A and Table 7B. Notably, the addition of lenalidomide did not result in reduced secretion of SEAP in any of the constructs tested. The results indicate adding a degron domain alone to a secreted molecule does not allow for drug-dependent degradation/regulation.









TABLE 7A







SEAP/Degron QuantiBlue Assay (HEK Cells)











% SEAP OFF vs. ON



% SEAP OFF vs. ON
(Len treated/untreated *100)


Construct
(Len treated/untreated *100)
Normalized to cell count












1118
134
153


1141
161 (p = .011)
109


1143
57.6 (p < .01)  
187


1144
308 (p < .01) 
473 (p < .01)


419
103
133


No virus
100
133
















TABLE 7B







SEAP/Degron QuantiBlue Assay (T Cells)











% SEAP OFF vs. ON



% SEAP OFF vs. ON
(Len treated/untreated *100)


Construct
(Len treated/untreated *100)
Normalized to cell count












1118
96.1
33 (p < .01)


1141
82
142


1143
68
109


1144
100
90


419
127
169


No virus
44
ND









Example 2—Membrane-Cleavable Degron System

The ability to regulate secretion of a molecule through attachment of a degron domain in conjunction with a cleavable membrane-tethering domain was assessed.


Methods

A series of pL17d-based viral vector constructs were generated using Secreted Alkaline Phosphatase (SEAP) fused to a degron (d913 super degron) using various linkers, including transmembrane domain containing linkers, as described in Table 8A with sequences listed in Table 8B.


SEAP assays were performed using the QuantiBlue kit (Invivogen; rep-qb1). For experiments performed in HEK293Ft cells, the following protocol was followed:

    • Cells were plated at 1e5 cells/well in a 24 well plate on Day 0
    • On Day 1, cells were transduced with 100K GS units of virus
    • On Day 2, cells were split into 2 wells in 12 well plates+/−1 μM lenalidomide
    • On Day 4, cells were passaged in triplicate into 24 well plates and lenalidomide treatment was continued
    • On Day 7, 20 uL supernatant from each well was harvested and incubated with 180 uL QuantiBlue (Invivogen) for 1 h at 37 deg, SEAP activity was read out on a plate reader at 655 nm (Cells are adherent so do not need to be pelleted from the media)


      For experiments performed in donor-derived T cells, the following protocol was followed:
    • T cells were enriched from leukapheresis material using anti-CD4 and anti-CD8 magnetic beads at a targeted 1:1 ratio on the Miltenyi Biotec CliniMACS Prodigy instrument. The CliniMACS Prodigy utilizes a closed-system, single-use disposable kits for many different cell types. The automated program performs platelet washes, CD4+& CD8+ bead labeling, and positive selection over a magnet resulting in cell suspensions with high purity. Positively selected T cells were then sampled, analyzed for cell surface phenotype via flow cytometry and cryopreserved via a controlled rate freezer.
    • Cells were then thawed and activated on Day 0 (1e6 cells/mL in a 24 well plate were activated 3:1 with CD3/CD28 Dynabeads)
    • On Day 1, 100K GS units of virus were added to cells
    • On Day 2, fresh media was added to T cells
    • On Day 4, each well was split into 6 wells of 48 well plate with +/−1 μM lenalidomide (3 wells with drug, 3 wells without)
    • On Day 7, cells were pelleted and 20 uL supernatant from each well was harvested and incubated with 180 uL QuantiBlue (Invivogen) for 1 h at 37 deg, SEAP activity was read out on a plate reader at 655 nm









TABLE 8A







SEAP/Degron/Membrane-Cleavable Construct Configurations








Construct
Description





SB01208
d913 degron-A(EAAK)3A (SEQ ID NO: 193) linker-hFasL



min type2 TM domain-ADAM10/17 cs-SEAP


SB01209
SEAP-ADAM10/17 cleavage site-PDGFRb TM-



A(EAAAK)3A (SEQ ID NO: 183)-d913 degron


SB00419
SEAP
















TABLE 8B





SEAP/Degron/Membrane-Cleavable Construct Sequences


Sequences















SB01208 (SEQ ID NO: 180)


ATGTTCAATGTCTTAATGGTTCATAAGCGAAGCCATACTGGTGAACGCCCATTGCAGTGCG


AAATATGCGGCTTTACCTGCCGCCAGAAAGGTAACCTCCTCCGCCACATTAAACTGCACAC


AGGGGAAAAACCTTTTAAGTGTCACCTCTGCAACTATGCATGCCAAAGAAGAGATGCGCTC


GCCGAGGCGGCAGCAAAGGAAGCAGCAGCGAAAGAGGCGGCTGCCAAAGCGATGCTGAA


GAAACGAGGTAACCATAGCACCGGCTTGTGTCTGTTGGTCATGTTTTTTATGGTCCTTGTAG


CTCTCGTAGGTCTTGGGCTGGGTATGTTTCAACTGTTCCACCTCCAAAAGGAAACAGGCCC


CCGAGCCGAGGCCCTGAAGGGTGGGGTCATTCCTGTGGAGGAGGAGAATCCTGCTTTTTGG


AATAGGAAGGCAGCTGAAGCCTTGGATGCAGCCAAGAAGCTCAAGCCCATTCAGACATCT


GCAAAGAATCTTGTCATCCTCATGGGTGATGGAATGGGTGTCTCCACTGTAACAGCCACCA


GGATTCTGAAGGGCCAGCAACAAGGTCATCTAGGCCCAGAGACCCAGTTGGCAATGGACA


GGTTCCCTCACATGGCCCTTTCCAAGACTTACAACACTGACAAGCAGATTCCTGACTCTGCT


GGGACAGGCACAGCATTCTTGTGTGGAGTAAAAACCAACATGAAAGTCATTGGTCTTTCAG


CTGCTGCCAGATTCAACCAGTGCAACACCACATGGGGCAATGAAGTGGTCTCTGTAATGCA


CAGGGCCAAAAAAGCTGGGAAAAGTGTGGGTGTGGTGACAACCACCTCTGTCCAGCATGC


CTCTCCTGCTGGAACTTATGCCCACACAGTGAACAGAGGTTGGTACTCTGATGCTCAGATG


CCTGCCTCAGCTTTACAAGATGGCTGCAAGGACATCAGCACCCAGCTCATCTCAAACATGG


ACATAGATGTCATCTTAGGGGGTGGGAGAAAGTTCATGTTCCCAAAGGGGACTCCTGACCA


GGAGTACCCCACAGACACAAAGCAGGCTGGCACAAGATTAGATGGTAGGAACCTTGTGCA


AGAGTGGCTTGCCAAGCATCAGGGAGCAAGGTATGTCTGGAACAGGAGTGAGCTAATCCA


GGCCTCTTTGAACAGGTCTGTCACTCACCTAATGGGGTTATTTGAGCCCAATGACATGAAG


TATGAGATACACAGGGACCCTGCCCAGGACCCCTCCTTAGCAGAAATGACTGAAGTTGCTG


TGAGGATGTTGTCCAGAAATCCAAAAGGGTTCTACCTCTTTGTTGAGGGGGGAAGGATTGA


TCATGGTCACCATGAGACAGTTGCTTACAGAGCCTTAACTGAGGCTGTGATGTTTGATTCTG


CTGTGGACAAGGCTGACAAACTGACCTCTGAGCAGGACACAATGATTCTAGTGACTGCTGA


CCACAGTCATGTTTTCTCCTTTGGGGGCTACACCCAGAGGGGTGCTTCAATCTTTGGCCTGG


CCCCTTTCAAGGCAGAAGATGGGAAGAGTTTCACCTCCATCCTCTATGGGAATGGTCCTGG


GTACAAGCTGCACAATGGGGCCAGAGCTGATGTGACAGAAGAGGAGAGCTCCAACCCAAC


CTACCAGCAGCAAGCAGCAGTCCCTCTTTCTTCAGAAACCCACTCTGGGGAAGATGTGGCC


ATATTTGCCAGAGGCCCCCAAGCCCACTTGGTGCATGGAGTTCAGGAGCAGAATTACATAG


CTCATGTAATGGCTTTTGCTGCTTGCTTGGAGCCCTACACAGACTGTGGCCTAGCCAGCCCA


GCAGGCCAGTCCTCTGCAGTAAGCCCAGGCTAA





SB01209 (SEQ ID NO: 181)


ATGTGGGGTGCCTGTCTGCTATTGCTGGGCTTAAGTCTTCAAGTTTGCCCCAGTGTCATTCC


TGTGGAGGAGGAGAATCCTGCTTTTTGGAATAGGAAGGCAGCTGAAGCCTTGGATGCAGC


CAAGAAGCTCAAGCCCATTCAGACATCTGCAAAGAATCTTGTCATCCTCATGGGTGATGGA


ATGGGTGTCTCCACTGTAACAGCCACCAGGATTCTGAAGGGCCAGCAACAAGGTCATCTAG


GCCCAGAGACCCAGTTGGCAATGGACAGGTTCCCTCACATGGCCCTTTCCAAGACTTACAA


CACTGACAAGCAGATTCCTGACTCTGCTGGGACAGGCACAGCATTCTTGTGTGGAGTAAAA


ACCAACATGAAAGTCATTGGTCTTTCAGCTGCTGCCAGATTCAACCAGTGCAACACCACAT


GGGGCAATGAAGTGGTCTCTGTAATGCACAGGGCCAAAAAAGCTGGGAAAAGTGTGGGTG


TGGTGACAACCACCTCTGTCCAGCATGCCTCTCCTGCTGGAACTTATGCCCACACAGTGAA


CAGAGGTTGGTACTCTGATGCTCAGATGCCTGCCTCAGCTTTACAAGATGGCTGCAAGGAC


ATCAGCACCCAGCTCATCTCAAACATGGACATAGATGTCATCTTAGGGGGTGGGAGAAAG


TTCATGTTCCCAAAGGGGACTCCTGACCAGGAGTACCCCACAGACACAAAGCAGGCTGGC


ACAAGATTAGATGGTAGGAACCTTGTGCAAGAGTGGCTTGCCAAGCATCAGGGAGCAAGG


TATGTCTGGAACAGGAGTGAGCTAATCCAGGCCTCTTTGAACAGGTCTGTCACTCACCTAA


TGGGGTTATTTGAGCCCAATGACATGAAGTATGAGATACACAGGGACCCTGCCCAGGACC


CCTCCTTAGCAGAAATGACTGAAGTTGCTGTGAGGATGTTGTCCAGAAATCCAAAAGGGTT


CTACCTCTTTGTTGAGGGGGGAAGGATTGATCATGGTCACCATGAGACAGTTGCTTACAGA


GCCTTAACTGAGGCTGTGATGTTTGATTCTGCTGTGGACAAGGCTGACAAACTGACCTCTG


AGCAGGACACAATGATTCTAGTGACTGCTGACCACAGTCATGTTTTCTCCTTTGGGGGCTA


CACCCAGAGGGGTGCTTCAATCTTTGGCCTGGCCCCTTTCAAGGCAGAAGATGGGAAGAGT


TTCACCTCCATCCTCTATGGGAATGGTCCTGGGTACAAGCTGCACAATGGGGCCAGAGCTG


ATGTGACAGAAGAGGAGAGCTCCAACCCAACCTACCAGCAGCAAGCAGCAGTCCCTCTTT


CTTCAGAAACCCACTCTGGGGAAGATGTGGCCATATTTGCCAGAGGCCCCCAAGCCCACTT


GGTGCATGGAGTTCAGGAGCAGAATTACATAGCTCATGTAATGGCTTTTGCTGCTTGCTTG


GAGCCCTACACAGACTGTGGCCTAGCCAGCCCAGCAGGCCAGTCCTCTGCAGTAAGCCCA


GGCCCCCGAGCCGAGGCCCTGAAGGGTGGGGCTGTGGGCCAGGACACGCAGGAGGTCATC


GTGGTGCCACACTCCTTGCCCTTTAAGGTGGTGGTGATCTCAGCCATCCTGGCCCTGGTGGT


GCTCACCATCATCTCCCTTATCATCCTCATCATGCTTTGGCAGAAGAAGCCACGTGCCGAG


GCGGCAGCAAAGGAAGCAGCAGCGAAAGAGGCGGCTGCCAAAGCGTTCAATGTCTTAATG


GTTCATAAGCGAAGCCATACTGGTGAACGCCCATTGCAGTGCGAAATATGCGGCTTTACCT


GCCGCCAGAAAGGTAACCTCCTCCGCCACATTAAACTGCACACAGGGGAAAAACCTTTTA


AGTGTCACCTCTGCAACTATGCATGCCAAAGAAGAGATGCGCTCTAA









Results

Combining a degron domain and a cleavable membrane-tethering domain to regulate secretion of molecules was assessed. In general, the system (referred to as the “Membrane-Cleavable Degron” system), uses a chimeric protein having the formula S-C-MT-D (oriented from N-terminal to C-terminal), in which S refers to a secretable payload (e.g., an effector molecule), C refers to a protease cleavage site, MT refers to a cell membrane tethering domain, and D refers to a degron. FIG. 2 illustrates a representative Membrane-Cleavable Degron system in which a desired payload is expressed as a chimeric protein in which a cleavable membrane-tethering domain (e.g., an ADAM10/17 cleavage site fused to PDGFRb transmembrane domain) is inserted between the payload and degron domain.


A SEAP assay was designed to test the efficacy of the Membrane-Cleavable Degron system through assessing secreted SEAP activity in conditioned media (free of cells). The domains for the constructs tested are outlined below:


SB01208 (D-MT-C-S Orientation):





    • S is SEAP

    • C is an ADAM10/17 cleavage site

    • MT is a type II transmembrane domain (human FasL)

    • D is a CRBN-based degron (d913)





SB01209 (S-C-MT-D Orientation):





    • S is SEAP

    • C is an ADAM10/17 cleavage site

    • MT is a PDGFRb transmembrane domain

    • D is a CRBN-based degron (d913)





As shown in FIG. 3, SEAP enzymatic activity was determined in supernatants from both HEK293Ft cells (“HEK293T”) and donor-derived T cells (“T cells”). SEAP enzymatic activity was quantified as a percentage of “Off” (+lenalidomide) relative to “On” (no lenalidomide) and is presented in Table 9A (HEK) and Table 9B (T Cells). SEAP secretion was reduced upon addition of lenalidomide in HEK293Ft cells and donor-derived T cells for the Membrane-Cleavable Degron construct having the S-C-MT-D orientation (“1209”). As shown in FIG. 4 and quantified in Table 9A, SEAP enzymatic activity was also normalized to cell number in HEK293Ft cells. Normalization was performed by dividing SEAP absorbance by cell count per well (from Countess). SEAP secretion was reduced in a lenalidomide-dependent manner in HEK293Ft cells for the S-C-MT-D orientated construct (“1209”) relative to SEAP activity for the construct without a degron (“419”). The results suggest degradation of the membrane-tethered SEAP occurred in the presence of the immunomodulatory drug (IMiD) lenalidomide. The Membrane-Cleavable Degron construct having the orientation D-MT-C-S (“1208”) did not result in functional secretion of SEAP. The results indicate a Membrane-Cleavable Degron system using a chimeric protein in the S-C-MT-D orientation allows for drug-dependent degradation/regulation of secreted molecules.









TABLE 9A







SEAP/Degron/Membrane-Cleavable QuantiBlue Assay (HEK Cells)











% SEAP OFF vs. ON



% SEAP OFF vs. ON
(Len treated/untreated *100)


Construct
(Len treated/untreated *100)
Normalized to cell count












419
70
104.3


1208
325
ND


1209
18 (p < .001)
15.7 (p = .017)


no virus
314
ND
















TABLE 9B







SEAP/Degron/Membrane-Cleavable QuantiBlue Assay (T Cells)











% SEAP OFF vs. ON



Construct
(Len treated/untreated *100)














419
129



1208
182



1209
32 (p = 0.007)



no virus
189










Example 3—Membrane-Cleavable Degron System in Jurkats

The Membrane-Cleavable Degron system was further assessed in a Jurkat T cell line.


Methods

The viral vector constructs were generated, as described above, using Secreted Alkaline Phosphatase (SEAP) fused to a degron (d913 super degron) with various linkers, including transmembrane domain containing linkers. The constructs examined are described in Table 10.


SEAP assays were performed using the QuantiBlue kit (Invivogen; rep-qb1). For experiments performed in Jurkat cells, the following protocol was followed:

    • On Day 1, cells were plated at 1e6 cells in 1 mL in a 24 well plate and 300 k GS units of virus was added
    • On Day 3, cells were split into 6 wells of a 48 well plate (1e5 cells in 0.5 mL) and drug was added to 3 of these wells (1 μM lenalidomide)
    • On Day 6, cells were pelleted and 20 uL supernatant from each well was harvested and incubated with 180 uL QuantiBlue (Invivogen) for 1 h at 37 deg, SEAP activity was read out on a plate reader at 655 nm









TABLE 10







SEAP/Degron and SEAP/Degron/Membrane-Cleavable


Construct Configurations











Construct
Description
SEQ ID NO







SB01118
SEAP-(GS)4 GG (SEQ ID
175




NO: 182)-d913 super degron




SB01141
SEAP-A(EAAAK)3A (SEQ ID
176




NO: 183)-d913 super degron




SB01143
SEAP-KEGS (SEQ ID NO: 195)
177




Whitlow linker-d913 super





degron




SB01144
SEAP-EGK Whitlow linker-
178




d913 super degron




SB01209
SEAP-ADAM10/17 cleavage site-
181




PDGFRb TM-A(EAAAK)3A





(SEQ ID NO: 183)-d913 degron




SB00419
SEAP
179



SB01321
mCherry Expression Vector





(viral transfection control)











Results

The Membrane-Cleavable Degron system was further assessed in a Jurkat T cell line using the SEAP activity assay. The constructs tested either had no degron (“419”), a C-terminal degron only with various linkers (“1118”, “1141”, “1143”, “1144”), an irrelevant viral construct used as an infection control (“1321”), or was the Membrane-Cleavable Degron system below:


SB01209 (S-C-MT-D orientation):

    • S is SEAP
    • C is an ADAM10/17 cleavage site
    • MT is a PDGFRb transmembrane domain
    • D is a CRBN-based degron (d913)


As shown in FIG. 5, SEAP enzymatic activity was determined in supernatants from Jurkat T cells. SEAP enzymatic activity was quantified as a percentage of “Off” (+lenalidomide) relative to “On” (no lenalidomide) and is presented in Table 11. SEAP secretion was significantly reduced upon addition of lenalidomide in Jurkat T cells for the Membrane-Cleavable Degron construct having the S-C-MT-D orientation (“1209”). SEAP secretion was also reduced in a lenalidomide-dependent manner for the S-C-MT-D orientated construct (“1209”) relative to SEAP activity for the construct without a degron (“419”). The results suggest degradation of the membrane-tethered SEAP occurred in the presence of the immunomodulatory drug (IMiD) lenalidomide indicating a Membrane-Cleavable Degron system using a chimeric protein in the S-C-MT-D orientation allows for drug-dependent degradation/regulation of secreted molecules.









TABLE 11







SEAP/Degron/Membrane-Cleavable QuantiBlue Assay (Jurkat Cells)










Construct
OFF % of ON







SB01118
108.2 (ns)



SB01141
105.0 (ns)



SB01143
105.4 (ns)



SB01144
 82.3 (ns)



SB01209
5.94 (p < .001)



SB00419
105.3 (ns)



SB01321
N/A no Len




treatment










Example 4—Regulated Cytokine Secretion Using Membrane-Cleavable Degron System

The Membrane-Cleavable Degron system was further assessed for the ability to regulate the secretion of cytokines.


Methods

As illustrated in FIG. 6, pL17d viral vector constructs were generated that expressed human IL-10 (hIL-10) either in its native form (“1584”) or as a chimeric protein fused to a membrane-cleavable degron (“1585”). The viral vectors also expressed human IL-12 (hIL-12) from a separate promoter as a normalization control for cell number, viability, and transduction efficiency. The constructs examined are described in Table 12A with relevant sequences listed in Table 12B.


Cytokine secretion was assessed following the general protocol below:

    • Day 0: Donor T cells (enriched as previously described above) were thawed and activated with CD3/CD28 Dynabeads
    • Day 1: T cells were transduced with viral constructs (100 k GoStix values per 1e6 cells)
    • Day 2: Media was changed
    • Day 4: Cells were passaged
    • Day 7: Cells were spun down, washed 3× with media, then resuspended in media+/−1 μM Lenalidomide
    • Day 9: Supernatants were harvested and were evaluated for hIL10 and hIL12 concentration by Luminex assay









TABLE 12A







IL-10/Degron/Membrane-Cleavable Construct Configurations








Construct
Description





1584
SFFV promoter-hIL-10; minTK-hIL-12 (opposite direction)


1585
SFFV promoter-hIL-10-ADAM10/17 cleavage site-PDGFRb



TM-A(EAAAK)3A (SEQ ID NO: 183)-d913 degron; minTK-



hIL-12(opposite direction)


1043
SFFV promoter-control protein; minTK-hIL-12 (opposite



direction)
















TABLE 12B





IL-10/Degron/Membrane-Cleavable Construct Sequences


Sequences















minTK-hIL-12 (opposite direction) (SEQ ID NO: 192)


ttaACTGGCGTTCAGGTAGGACATCACGCGGTCAATGGTCACGGCTCGAATGCGGAA


AGCGTGCAACAGGATGCACAGCTTGATCTTGGTCTTGTAGAAGTCCGGTTCTTCCA


GGCTGGACTTTTGAGGCACAGTCTCGGAGTTGAAATTCAGGGCCTGCATCAGTTCA


TCAATCACGGCGAGCATATTCTGGTCCAGGAAGATCTGCCGCTTCGGGTCCATGAG


CAGCTTGGCGTTCATGGTCTTGAACTCGACCTGATACATCTTCAGATCTTCGTAGAT


CGAGGAAAGACAGAGCGCCATCATGAATGAGGTCTTTCTCGACGCCAGGCAGCTG


CCGTTAGTGATAAAGCTTGTCTCGCGGGAGTTCAGACACGATTCGTTCTTGGTCAG


TTCCAGCGGCAGGCAGGCTTCCACGGTCGAGGTCTTGTCCTTGGTGATGTCCTCGT


GATCAATTTCTTCCGAGGTGCAGGGGTAGAACTCAAGGGTCTGGCGGGCCTTCTGC


AACATGTTCGACACAGCCCTCAGGAGGTTTTGGGAGTGGTGTAGGCACGGGAACA


TTCCAGGGTCGGGGGTTGCCACAGGGAGGTTCCGGGAACCTCCTCCGGAGCCTCCT


CCTGAACCTCCGCCTGATCCGCCACCGGAACAAGGCACGCTGGCCCATTCGCTCCA


GGAGGACGAGTAGTATCTATCCTGCGCCCGGACGCTGATTGACGCGTTCTTCCGAC


AAATCACAGTGGCGGAGGTTTTGTCGGTGAACACCCGGTCTTTCTTCTCCCGTTTGG


ACTTTCCCTGCACTTGCACACAGAAAGTGAGCGAGAAGTATGAGTGCGGGGTGCTC


CAAGTGTCTGGATATTCCCAAGACACTTCCACTTGGCGGGAGTTCTTGAGTGGCTT


CAGCTGCAAGTTCTTGGGGGGGTCAGGCTTGATGATGTCGCGGATAAAGAAGGAG


GAAGTGTAGTTCTCGTATTTCAGCTTATGCACGGCATCGACCATGACCTCGATAGG


CAGGGACTCTTCCGCGGCAGGGCAGGCGCTGTCCTCCTGGCATTCCACGGAGTACT


CATATTCCTTGTTGTCTCCCCTGACTCTCTCGGCGGACAGAGTGGCGGCTCCACAG


GTCACGCCCTGAGGATCGCTTGATCCCCGTGACGACTTCACGGAGAAAGTCAGGTC


GGTGGAGATTGTCGTCAGCCACCAACAGGTGAACCGACCGCTGTAGTTCTTGGCTT


CGCAGCGGAGGAAGGTCTTGTTCTTCGGTTCTTTTTGGTCCTTGAGGATGTCAGTGG


ACCAGATTCCATCCTCTTTCTTGTGCAGCAGCAGCAGGGAGTGGGACAGCACTTCG


CCACCCTTGTGGCAAGTGTACTGGCCCGCGTCGCCGAACTCCTTGACTTGAATGGT


CAGGGTCTTTCCGCTTCCGAGCACCTCGGAGCTCTGATCCAGGGTCCAGGTTATGC


CGTCCTCTTCTGGCGTATCGCAAGTCAGCACGACCATTTCTCCAGGGGCGTCCGGG


TACCAATCCAGCTCGACCACGTAGACGTCCTTCTTCAGTTCCCAAATGGCGACCAG


AGGGGAAGCGAGGAACACAAGGGAGAACCAGGAGATGACGAGTTGCTGATGGCA


CATGGTGGCGGCttaagcgggtcgctgcagggtcgctcggtgttcgaggccacacg


cgtcaccttaatatgcgaa





1584 (SEQ ID NO: 186) SFFV promoter-hIL-10


ccgataaaataaaagattttatttagtctccagaaaaaggggggaatgaaagaccc


cacctgtaggtttggcaagctagctgcagtaacgccattttgcaaggcatggaaaa


ataccaaaccaagaatagagaagttcagatcaaggggggtacatgaaaatagctaa


cgttgggccaaacaggatatctgcggtgagcagtttcggccccggcccggggccaa


gaacagatggtcaccgcagtttcggccccggcccgaggccaagaacagatggtccc


cagatatggcccaaccctcagcagtttcttaagacccatcagatgtttccaggctc


ccccaaggacctgaaatgaccctgcgccttatttgaattaaccaatcagcctgctt


ctcgcttctgttcgcgcgcttctgcttcccgagctctataaaagagctcacaaccc


ctcactcggcgcgccagtcctccgacagactgagtcgcccgggggggatctggagc


tctcgagaattcATGCACTCATCTGCTCTCCTTTGCTGTCTCGTGCTCCTGACTGG


AGTACGCGCGTCACCCGGTCAAGGGACACAAAGCGAAAACTCCTGTACGCATTTTC


CGGGCAACCTCCCTAATATGCTTCGCGACTTGCGAGATGCATTTTCCAGGGTGAAG


ACCTTCTTCCAGATGAAGGACCAGTTGGATAATCTTCTCCTCAAGGAGTCTCTTTT


GGAGGACTTCAAGGGCTACCTGGGATGTCAAGCCCTTAGCGAAATGATTCAGTTTT


ATCTTGAAGAGGTCATGCCGCAAGCTGAGAACCAGGACCCCGACATAAAAGCCCAT


GTGAACTCCCTCGGCGAGAATTTGAAGACGCTGAGACTTAGATTGAGAAGATGTCA


TAGATTTCTCCCATGTGAGAATAAGTCCAAGGCTGTCGAGCAGGTTAAGAATGCTT


TCAATAAGTTGCAGGAAAAAGGAATCTATAAGGCAATGAGCGAGTTTGACATCTTT


ATTAACTACATTGAGGCTTATATGACTATGAAAATTAGAAACTAA





1585 (SEQ ID NO: 187) SFFV promoter-hIL-10-ADAM10/17 


cleavage site-PDGFRb TM-A(EAAAK)3A (SEQ ID NO: 183)-


d913 degron:


ccgataaaataaaagattttatttagtctccagaaaaaggggggaatgaaagaccc


cacctgtaggtttggcaagctagctgcagtaacgccattttgcaaggcatggaaaa


ataccaaaccaagaatagagaagttcagatcaagggcgggtacatgaaaatagcta


acgttgggccaaacaggatatctgcggtgagcagtttcggccccggcccggggcca


agaacagatggtcaccgcagtttcggccccggcccgaggccaagaacagatggtcc


ccagatatggcccaaccctcagcagtttcttaagacccatcagatgtttccaggct


cccccaaggacctgaaatgaccctgcgccttatttgaattaaccaatcagcctgct


tctcgcttctgttcgcgcgcttctgcttcccgagctctataaaagagctcacaacc


cctcactcggcgcgccagtcctccgacagactgagtcgcccgggggggatctggag


ctctcgagaattcATGCACTCATCTGCTCTCCTTTGCTGTCTCGTGCTCCTGACTG


GAGTACGCGCGTCACCCGGTCAAGGGACACAAAGCGAAAACTCCTGTACGCATTTT


CCGGGCAACCTCCCTAATATGCTTCGCGACTTGCGAGATGCATTTTCCAGGGTGAA


GACCTTCTTCCAGATGAAGGACCAGTTGGATAATCTTCTCCTCAAGGAGTCTCTTT


TGGAGGACTTCAAGGGCTACCTGGGATGTCAAGCCCTTAGCGAAATGATTCAGTTT


TATCTTGAAGAGGTCATGCCGCAAGCTGAGAACCAGGACCCCGACATAAAAGCCCA


TGTGAACTCCCTCGGCGAGAATTTGAAGACGCTGAGACTTAGATTGAGAAGATGTC


ATAGATTTCTCCCATGTGAGAATAAGTCCAAGGCTGTCGAGCAGGTTAAGAATGCT


TTCAATAAGTTGCAGGAAAAAGGAATCTATAAGGCAATGAGCGAGTTTGACATCTT


TATTAACTACATTGAGGCTTATATGA









Results

The Membrane-Cleavable Degron system was further assessed for the ability to regulate secretion of cytokines. The constructs tested either expressed native hIL-10 without a degron (“1584”), expressed a negative-control protein without a degron (“1043”), or expressed a hIL-10 chimeric protein in a membrane-cleavable degron system as described below:


1585 (S-C-MT-D Orientation):





    • S is hIL-12

    • C is an ADAM10/17 cleavage site

    • MT is a PDGFRb transmembrane domain

    • D is a CRBN-based degron (d913)





As shown in FIG. 7, hIL-10 secretion was determined in supernatants from donor-derived T cells, as normalized to IL-12 expression, and is presented in Table 13. No hIL-10 was detected for T cells transduced with the construct expressing an irrelevant protein (“1043”). The addition of lenalidomide did not result in reduced secretion of in the viral construct expressing the native hIL-10 (“1584”). In contrast, addition of lenalidomide resulted in an ˜50% decrease in hIL-10 detected in the supernatant, indicating the membrane cleavable degron format enabled regulation of hIL-10 secretion from primary T cells.













TABLE 13






Mean
Mean
% 1 uM
p value



IL10/IL12 ratio
IL10/IL12 ratio
Len of
(no drug vs.


Construct
(no drug)
(1 uM Len)
no drug
1 uM Len)



















1584
3.11
4.09
131%
0.01


1585
2.97
1.65
 56%
0.002


1043
0.113
0.110
 97%
0.53


No virus
0
0
n/a
n/a









Example 5—AAV Degron Systems

The degron-mediated regulation was assessed using an AAV-based delivery system.


Methods

AAV2 and AAV8 viral vector constructs were generated that expressed nanoluciferase as a chimeric protein fused to a C-terminal degron using an A(EAAK)3A linker (SEQ ID NO: 193). Constructs were generated using packaging vectors with either AAV2 or AAV8 capsid protein. The sequence of the construct nanoluciferase/degron fusion is listed in Table 14.


Luciferase activity was assessed following the general protocol below:

    • Day 0: Hep3b cells were plated at 50,000 cells per well in a 96 well plate
    • Day 1: 1e6 vg/cell AAV2 or 3e6 vg/cell AAV8 was added to cells in presence or absence of 1 μM Lenalidomide (or 1 μM-0.01 μM Lenalidomide or Pomalidomide)
    • Day 3: Cells were lysed and luciferase activity was measured with Promega Nanoglo kit









TABLE 14





Nanoluciferase/Degron Construct Sequence


Sequences















Nanoluciferase/Degron AAV Vector (SEQ ID NO: 188)


CCTGCAGGCAGCTGCGCGCTCGCTCGCTCACTGAGGCCGCCCGGGCGTCGGGCGAC


CTTTGGTCGCCCGGCCTCAGTGAGCGAGCGAGCGCGCAGAGAGGGAGTGGCCAAC


TCCATCACTAGGGGTTCCTGCGGCCGCATCGATTGAATTCtgtttgctgcttgca


atgtttgcccattttagggtggacacaggacgctgtggtttctgagccagggggcg


actcagatcccagccagtggacttagcccctgtttgctcctccgataactggggtg


accttggttaatattcaccagcagcctcccccgttgcccctctggatccactgc


ttaaatacggacgaggacagggccctgtctcctcagcttcaggcaccacca


ctgacctgggacagtgaatGCGGATTCGAATCCCGGCCGGGAACGGTGC


ATTGGAACGCGGATTCCCCGTGCCAAGAGTGACGTAAGTACCGCCTATAGAGTCTA


TAGGCCCACAAAAAATGCTTTCTTCTTTTAATATACTTTTTTGTTTATCTTATTTCTA


ATACTTTCCCTAATCTCTTTCTTTCAGGGCAATAATGATACAATGTATCATGCCTCT


TTGCACCATTCTAAAGAATAACAGTGATAATTTCTGGGTTAAGGCAATAGCAATAT


TTCTGCATATAAATATTTCTGCATATAAATTGTAACTGATGTAAGAGGTTTCATATT


GCTAATAGCAGCTACAATCCAGCTACCATTCTGCTTTTATTTTATGGTTGGGATAAG


GCTGGATTATTCTGAGTCCAAGCTAGGCCCTTTTGCTAATCATGTTCATACCTCTTA


TCTTCCTCCCACAGCTCCTGGGCAACGTGCTGGTCTGTGTGCTGGCCCATCACTTTG


GCAAAGAATTGGGATTCGAACATGGTCTTCACACTCGAAGATTTCGTTGGGGACTG


GCGACAGACAGCCGGCTACAACCTGGACCAAGTCCTTGAACAGGGAGGTGTGTCC


AGTTTGTTTCAGAATCTCGGGGTGTCCGTAACTCCGATCCAAAGGATTGTCCTGAG


CGGTGAAAATGGGCTGAAGATCGACATCCATGTCATCATCCCGTATGAAGGTCTGA


GCGGCGACCAAATGGGCCAGATCGAAAAAATTTTTAAGGTGGTGTACCCTGTGGA


TGATCATCACTTTAAGGTGATCCTGCACTATGGCACACTGGTAATCGACGGGGTTA


CGCCGAACATGATCGACTATTTCGGACGGCCGTATGAAGGCATCGCCGTGTTCGAC


GGCAAAAAGATCACTGTAACAGGGACCCTGTGGAACGGCAACAAAATTATCGACG


AGCGCCTGATCAACCCCGACGGCTCCCTGCTGTTCCGAGTAACCATCAACGGAGTG


ACCGGCTGGCGGCTGTGCGAACGCATTCTGGCGGGCAGTGGCTCGGGCTCGGGGT


CCGGCGGATTCAATGTCTTAATGGTTCATAAGCGAAGCCATACTGGTGAACGCCCA


TTGCAGTGCGAAATATGCGGCTTTACCTGCCGCCAGAAAGGTAACCTCCTCCGCCA


CATTAAACTGCACACAGGGGAAAAACCTTTTAAGTGTCACCTCTGCAACTATGCAT


GCCAAAGAAGAGATGCGCTCTAGTAAGTCGACCTGCAGAAGCTTGCCTCGAGCAG


CGCTGCTCGAGAGATCTACGGGTGGCATCCCTGTGACCCCTCCCCAGTGCCTCTCC


TGGCCCTGGAAGTTGCCACTCCAGTGCCCACCAGCCTTGTCCTAATAAAATTAAGT


TGCATCATTTTGTCTGACTAGGTGTCCTTCTATAATATTATGGGGTGGAGGGGGGT


GGTATGGAGCAAGGGGCAAGTTGGGAAGACAACCTGTAGGGCCTGCGGGGTCTAT


TGGGAACCAAGCTGGAGTGCAGTGGCACAATCTTGGCTCACTGCAATCTCCGCCTC


CTGGGTTCAAGCGATTCTCCTGCCTCAGCCTCCCGAGTTGTTGGGATTCCAGGCAT


GCATGACCAGGCTCAGCTAATTTTTGTTTTTTTGGTAGAGACGGGGTTTCACCATAT


TGGCCAGGCTGGTCTCCAACTCCTAATCTCAGGTGATCTACCCACCTTGGCCTCCCA


AATTGCTGGGATTACAGGCGTGAACCACTGCTCCCTTCCCTGTCCTTCTGATTTTGT


AGGTAACCACGTGCGGACCGAGCGGCCGCAGGAACCCCTAGTGATGGAGTTGGCC


ACTCCCTCTCTGCGCGCTCGCTCGCTCACTGAGGCCGGGCGACCAAAGGTCGCCCG


ACGCCCGGGCTTTGCCCGGGCGGCCTCAGTGAGCGAGCGAGCGCGCAGCTGCCTG


CAGGGGCGCCTGATGCGGTATTTTCTCCTTACGCATCTGTGCGGTATTTCACACCGC


ATACGTCAAAGCAACCATAGTACGCGCCCTGTAGCGGCGCATTAAGCGCGGCGGG


TGTGGTGGTTACGCGCAGCGTGACCGCTACACTTGCCAGCGCCCTAGCGCCCGCTC


CTTTCGCTTTCTTCCCTTCCTTTCTCGCCACGTTCGCCGGCTTTCCCCGTCAAGCTCT


AAATCGGGGGCTCCCTTTAGGGTTCCGATTTAGTGCTTTACGGCACCTCGACCCCA


AAAAACTTGATTTGGGTGATGGTTCACGTAGTGGGCCATCGCCCTGATAGACGGTT


TTTCGCCCTTTGACGTTGGAGTCCACGTTCTTTAATAGTGGACTCTTGTTCCAAACT


GGAACAACACTCAACCCTATCTCGGGCTATTCTTTTGATTTATAAGGGATTTTGCCG


ATTTCGGCCTATTGGTTAAAAAATGAGCTGATTTAACAAAAATTTAACGCGAATTT


TAACAAAATATTAACGTTTACAATTTTATGGTGCACTCTCAGTACAATCTGCTCTGA


TGCCGCATAGTTAAGCCAGCCCCGACACCCGCCAACACCCGCTGACGCGCCCTGAC


GGGCTTGTCTGCTCCCGGCATCCGCTTACAGACAAGCTGTGACCGTCTCCGGGAGC


TGCATGTGTCAGAGGTTTTCACCGTCATCACCGAAACGCGCGAGACGAAAGGGCCT


CGTGATACGCCTATTTTTATAGGTTAATGTCATGATAATAATGGTTTCTTAGACGTC


AGGTGGCACTTTTCGGGGAAATGTGCGCGGAACCCCTATTTGTTTATTTTTCTAAAT


ACATTCAAATATGTATCCGCTCATGAGACAATAACCCTGATAAATGCTTCAATAAT


ATTGAAAAAGGAAGAGTATGAGTATTCAACATTTCCGTGTCGCCCTTATTCCCTTTT


TTGCGGCATTTTGCCTTCCTGTTTTTGCTCACCCAGAAACGCTGGTGAAAGTAAAA


GATGCTGAAGATCAGTTGGGTGCACGAGTGGGTTACATCGAACTGGATCTCAACA


GCGGTAAGATCCTTGAGAGTTTTCGCCCCGAAGAACGTTTTCCAATGATGAGCACT


TTTAAAGTTCTGCTATGTGGCGCGGTATTATCCCGTATTGACGCCGGGCAAGAGCA


ACTCGGTCGCCGCATACACTATTCTCAGAATGACTTGGTTGAGTACTCACCAGTCA


CAGAAAAGCATCTTACGGATGGCATGACAGTAAGAGAATTATGCAGTGCTGCCAT


AACCATGAGTGATAACACTGCGGCCAACTTACTTCTGACAACGATCGGAGGACCG


AAGGAGCTAACCGCTTTTTTGCACAACATGGGGGATCATGTAACTCGCCTTGATCG


TTGGGAACCGGAGCTGAATGAAGCCATACCAAACGACGAGCGTGACACCACGATG


CCTGTAGCAATGGCAACAACGTTGCGCAAACTATTAACTGGCGAACTACTTACTCT


AGCTTCCCGGCAACAATTAATAGACTGGATGGAGGCGGATAAAGTTGCAGGACCA


CTTCTGCGCTCGGCCCTTCCGGCTGGCTGGTTTATTGCTGATAAATCTGGAGCCGGT


GAGCGTGGGTCTCGCGGTATCATTGCAGCACTGGGGCCAGATGGTAAGCCCTCCCG


TATCGTAGTTATCTACACGACGGGGAGTCAGGCAACTATGGATGAACGAAATAGA


CAGATCGCTGAGATAGGTGCCTCACTGATTAAGCATTGGTAACTGTCAGACCAAGT


TTACTCATATATACTTTAGATTGATTTAAAACTTCATTTTTAATTTAAAAGGATCTA


GGTGAAGATCCTTTTTGATAATCTCATGACCAAAATCCCTTAACGTGAGTTTTCGTT


CCACTGAGCGTCAGACCCCGTAGAAAAGATCAAAGGATCTTCTTGAGATCCTTTTT


TTCTGCGCGTAATCTGCTGCTTGCAAACAAAAAAACCACCGCTACCAGCGGTGGTT


TGTTTGCCGGATCAAGAGCTACCAACTCTTTTTCCGAAGGTAACTGGCTTCAGCAG


AGCGCAGATACCAAATACTGTCCTTCTAGTGTAGCCGTAGTTAGGCCACCACTTCA


AGAACTCTGTAGCACCGCCTACATACCTCGCTCTGCTAATCCTGTTACCAGTGGCT


GCTGCCAGTGGCGATAAGTCGTGTCTTACCGGGTTGGACTCAAGACGATAGTTACC


GGATAAGGCGCAGCGGTCGGGCTGAACGGGGGGTTCGTGCACACAGCCCAGCTTG


GAGCGAACGACCTACACCGAACTGAGATACCTACAGCGTGAGCTATGAGAAAGCG


CCACGCTTCCCGAAGGGAGAAAGGCGGACAGGTATCCGGTAAGCGGCAGGGTCGG


AACAGGAGAGCGCACGAGGGAGCTTCCAGGGGGAAACGCCTGGTATCTTTATAGT


CCTGTCGGGTTTCGCCACCTCTGACTTGAGCGTCGATTTTTGTGATGCTCGTCAGGG


GGGCGGAGCCTATGGAAAAACGCCAGCAACGCGGCCTTTTTACGGTTCCTGGCCTT


TTGCTGGCCTTTTGCTCACATGT









Results

The AAV-based delivery system was assessed for degron-mediated regulation of protein expression. The constructs tested expressed a nanoluciferase chimeric protein containing a degron domain. As shown in FIG. 8, Hep3B cells transduced with either an AAV2-based (left panel) or AAV8-based (right panel) construct and treated with lenalidomide resulted in a significant decrease (˜40-80 fold) in luciferase activity relative to cells not treated with lenalidomide, indicating the degron system was functional in AAV-based formats enabling regulation of protein expression.


Drug responsiveness of the degron system was further assessed in AAV-based formats. Using the AAV2-based construct, two IMiDs (lenalidomide and pomalidomide) were titrated to determine the minimal effective dose in vitro. As shown in FIG. 9 and quantified in Table 15, Hep3B cells transduced with the AAV2 construct and treated with either 1-0.01 μM lenalidomide (left panel) or pomalidomide (left right panel) resulted in a significant decreases in luciferase activity at all concentrations tested relative to cells not treated with lenalidomide. Notably, nanoluciferase signal increased from 0.05 to 0.01 μM of either drug, demonstrating efficacy at physiological concentrations of drug (based on PK studies in mice) and indicating the 0.01 μM concentration may be approaching the minimal effective dose.









TABLE 15







Nanoluciferase/Degron Chimera Response to IMiD












Fold change relative



Drug
Dose (uM)
to no drug
p-value













Lenalidomide
1
60
.01


Lenalidomide
0.5
35
.01


Lenalidomide
0.1
25
.01


Lenalidomide
0.05
18
.01


Lenalidomide
0.01
6
.01


Pomalidomide
1
19
.02


Pomalidomide
0.5
18
.02


Pomalidomide
0.1
19
.02


Pomalidomide
0.05
12
.02


Pomalidomide
0.01
4
.02

















Other Sequences



HIV-1 protease (SEQ ID NO: 144):


PQVTLWQRPLVTIKIGGQLKEALLDTGADDTVLEEMSLPGRWKPKMIGGIGGFIKVR





QYDQILIEICGHKAIGTVLVGPTPVNIIGRNLLTQIGCTLNF





Angiotensin converting enzyme (ACE) (SEQ ID NO: 156):


MGAASGRRGPGLLLPLPLLLLLPPQPALALDPGLQPGNFSADEAGAQLFAQSYNSSA





EQVLFQSVAASWAHDTNITAENARRQEEAALLSQEFAEAWGQKAKELYEPIWQNFT





DPQLRRIIGAVRTLGSANLPLAKRQQYNALLSNMSRIYSTAKVCLPNKTATCWSLDP





DLTNILASSRSYAMLLFAWEGWHNAAGIPLKPLYEDFTALSNEAYKQDGFTDTGAY





WRSWYNSPTFEDDLEHLYQQLEPLYLNLHAFVRRALHRRYGDRYINLRGPIPAHLLG





DMWAQSWENIYDMVVPFPDKPNLDVTSTMLQQGWNATHMFRVAEEFFTSLELSPM





PPEFWEGSMLEKPADGREVVCHASAWDFYNRKDFRIKQCTRVTMDQLSTVHHEMG





HIQYYLQYKDLPVSLRRGANPGFHEAIGDVLALSVSTPEHLHKIGLLDRVTNDTESDI





NYLLKMALEKIAFLPFGYLVDQWRWGVFSGRTPPSRYNFDWWYLRTKYQGICPPVT





RNETHFDAGAKFHVPNVTPYIRYFVSFVLQFQFHEALCKEAGYEGPLHQCDIYRSTK





AGAKLRKVLQAGSSRPWQEVLKDMVGLDALDAQPLLKYFQPVTQWLQEQNQQNG





EVLGWPEYQWHPPLPDNYPEGIDLVTDEAEASKFVEEYDRTSQVVWNEYAEANWN





YNTNITTETSKILLQKNMQIANHTLKYGTQARKFDVNQLQNTTIKRIIKKVQDLERAA





LPAQELEEYNKILLDMETTYSVATVCHPNGSCLQLEPDLTNVMATSRKYEDLLWAW





EGWRDKAGRAILQFYPKYVELINQAARLNGYVDAGDSWRSMYETPSLEQDLERLFQ





ELQPLYLNLHAYVRRALHRHYGAQHINLEGPIPAHLLGNMWAQTWSNIYDLVVPFP





SAPSMDTTEAMLKQGWTPRRMFKEADDFFTSLGLLPVPPEFWNKSMLEKPTDGREV





VCHASAWDFYNGKDFRIKQCTTVNLEDLVVAHHEMGHIQYFMQYKDLPVALREGA





NPGFHEAIGDVLALSVSTPKHLHSLNLLSSEGGSDEHDINFLMKMALDKIAFIPFSYL





VDQWRWRVFDGSITKENYNQEWWSLRLKYQGLCPPVPRTQGDFDPGAKFHIPSSVP





YIRYFVSFIIQFQFHEALCQAAGHTGPLHKCDIYQSKEAGQRLATAMKLGFSRPWPE





AMQLITGQPNMSASAMLSYFKPLLDWLRTENELHGEKLGWPQYNWTPNSARSEGPL





PDSGRVSFLGLDLDAQQARVGQWLLLFLGIALLVATLGLSQRLFSIRHRSLHRHSHG





PQFGSEVELRHS





DENV NS3pro (NS2B/NS3) >sp|P33478|1475-2093 (SEQ ID NO: 157):


SGVLWDTPSPPEVERAVLDDGIYRIMQRGLLGRSQVGVGVFQDGVFHTMWHVTRG





AVLMYQGKRLEPSWASVKKDLISYGGGWRFQGSWNTGEEVQVIAVEPGKNPKNVQ





TAPGTFKTPEGEVGAIALDFKPGTSGSPIVNREGKIVGLYGNGVVTTSGTYVSAIAQA





KASQEGPLPEIEDEVFRKRNLTIMDLHPGSGKTRRYLPAIVREAIRRNVRTLILAPTRV





VASEMAEALKGMPIRYQTTAVKSEHTGKEIVDLMCHATFTMRLLSPVRVPNYNMII





MDEAHFTDPASIARRGYISTRVGMGEAAAIFMTATPPGSVEAFPQSNAVIQDEERDIP





ERSWNSGYEWITDFPGKTVWFVPSIKSGNDIANCLRKNGKRVIQLSRKTFDTEYQKT





KNNDWDYVVTTDISEMGANFRADRVIDPRRCLKPVILKDGPERVILAGPMPVTVAS





AAQRRGRIGRNQNKEGDQYVYMGQPLNNDEDHAHWTEAKMLLDNINTPEGIIPALF





EPEREKSAAIDGEYRLRGEARKTFVELMRRGDLPVWLSYKVASEGFQYSDRRWCFD





GERNNQVLEENMDVEMWTKEGERKKLRPRWLDARTYSDPLALREFKEFAAGRR





DENV NS3pro (NS2B/NS3) >sp|P14340|1476-2093 (SEQ ID NO: 158):


AGVLWDVPSPPPVGKAELEDGAYRIKQKGILGYSQIGAGVYKEGTFHTMWHVTRGA





VLMHKGKRIEPSWADVKKDLISYGGGWKLEGEWKEGEEVQVLALEPGKNPRAVQT





KPGLFKTNAGTIGAVSLDFSPGTSGSPIIDKKGKVVGLYGNGVVTRSGAYVSAIAQTE





KSIEDNPEIEDDIFRKRKLTIMDLHPGAGKTKRYLPAIVREAIKRGLRTLILAPTRVVA





AEMEEALRGLPIRYQTPAIRAEHTGREIVDLMCHATFTMRLLSPVRVPNYNLIIMDEA





HFTDPASIAARGYISTRVEMGEAAGIFMTATPPGSRDPFPQSNAPIMDEEREIPERSWS





SGHEWVTDFKGKTVWFVPSIKAGNDIAACLRKNGKKVIQLSRKTFDSEYVKTRTND





WDFVVTTDISEMGANFKAERVIDPRRCMKPVILTDGEERVILAGPMPVTHSSAAQRR





GRIGRNPKNENDQYIYMGEPLENDEDCAHWKEAKMLLDNINTPEGIIPSMFEPEREK





VDAIDGEYRLRGEARKTFVDLMRRGDLPVWLAYRVAAEGINYADRRWCFDGIKNN





QILEENVEVEIWTKEGERKKLKPRWLDAKIYSDPLALKEFKEFAAGRK





DENV NS3pro (NS2B/NS3) >sp|Q99D35|1474-2092 (SEQ ID NO: 159):


SGVLWDVPSPPETQKAELEEGVYRIKQQGIFGKTQVGVGVQKEGVFHTMWHVTRG





AVLTHNGKRLEPNWASVKKDLISYGGGWRLSAQWQKGEEVQVIAVEPGKNPKNFQ





TMPGIFQTTTGEIGAIALDFKPGTSGSPIINREGKVVGLYGNGVVTKNGGYVSGIAQT





NAEPDGPTPELEEEMFKKRNLTIMDLHPGSGKTRKYLPAIVREAIKRRLRTLILAPTR





VVAAEMEEALKGLPIRYQTTATKSEHTGREIVDLMCHATFTMRLLSPVRVPNYNLII





MDEAHFTDPASIAARGYISTRVGMGEAAAIFMTATPPGTADAFPQSNAPIQDEERDIP





ERSWNSGNEWITDFVGKTVWFVPSIKAGNDIANCLRKNGKKVIQLSRKTFDTEYQKT





KLNDWDFVVTTDISEMGANFKADRVIDPRRCLKPVILTDGPERVILAGPMPVTVASA





AQRRGRVGRNPQKENDQYIFMGQPLNKDEDHAHWTEAKMLLDNINTPEGIIPALFEP





EREKSAAIDGEYRLKGESRKTFVELMRRGDLPVWLAHKVASEGIKYTDRKWCFDGE





RNNQILEENMDVEIWTKEGEKKKLRPRWLDARTYSDPLALKEFKDFAAGRK





DENV NS3pro (NS2B/NS3) >sp|Q5UCB8|1475-2092 (SEQ ID NO: 160):


SGALWDVPSPAATQKAALSEGVYRIMQRGLFGKTQVGVGIHIEGVFHTMWHVTRGS





VICHETGRLEPSWADVRNDMISYGGGWRLGDKWDKEEDVQVLAIEPGKNPKHVQT





KPGLFKTLTGEIGAVTLDFKPGTSGSPIINRKGKVIGLYGNGVVTKSGDYVSAITQAE





RIGEPDYEVDEDIFRKKRLTIMDLHPGAGKTKRILPSIVREALKRRLRTLILAPTRVVA





AEMEEALRGLPIRYQTPAVKSEHTGREIVDLMCHATFTTRLLSSTRVPNYNLIVMDE





AHFTDPSSVAARGYISTRVEMGEAAAIFMTATPPGTTDPFPQSNSPIEDIEREIPERSW





NTGFDWITDYQGKTVWFVPSIKAGNDIANCLRKSGKKVIQLSRKTFDTEYPKTKLTD





WDFVVTTDISEMGANFRAGRVIDPRRCLKPVILPDGPERVILAGPIPVTPASAAQRRG





RIGRNPAQEDDQYVFSGDPLKNDEDHAHWTEAKMLLDNIYTPEGIIPTLFGPEREKT





QAIDGEFRLRGEQRKTFVELMRRGDLPVWLSYKVASAGISYKDREWCFTGERNNQI





LEENMEVEIWTREGEKKKLRPKWLDARVYADPMALKDFKEFASGRK





PEST (two copies of residues 277-307 of human IκBα; SEQ ID NO: 161):


LQMLPESEDEESYDTESEFTEFTEDELPYDDGSLQMLPESEDEESYDTESEFTEFTEDE





LPYDD





GRR (residues 352-408 of human p105; SEQ ID NO: 162):


EIKDKEEVQRKRQKLMPNFSDSFGGGSGAGAGGGGMFGSGGGGGGTGSTGPGYSFP





H





DRR (residues 210-295 of yeast Cdc34; SEQ ID NO: 163):


IDDENGSVILQDDDYDDGNNHIPFEDDDVYNYNDNDDDDERIEFEDDDDDDDDSID





NDSVMDRKQPHKAEDESEDVEDVERVSKKD





SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or


influenza B; e.g., SEQ ID NO: 164):


PESMREEYRKEGSKRIKCPDCEPFCNKRGSPESMREEYRKE





RPB (four copies of residues 1688-1702 of yeast RPB; SEQ ID NO: 165):


RSYSPTSPNYSPTSPSGSYSPTSPNYSPTSPSGGSRSYSPTSPNYSPTSPSGSYSPTSPNY





SPTSPSG





SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza


A virus M2 protein; SEQ ID NO: 166):


PESMREEYRKEGSSLLTEVETPGSPESMREEYRKEGSSLLTEVETPGSPESMREEYRK





E





NS2 (three copies of residues 79-93 of influenza A virus NS protein;


SEQ ID NO: 167):


LIEEVRHRLKTTENSGSLIEEVRHRLKTTENSGSLIEEVRHRLKTTENSGS





ODC (residues 106-142 of ornithine decarboxylase; SEQ ID NO: 168):


FPPEVEEQDDGTLPMSCAQESGMDRHPAACASARINV





mouse ODC (residues 422-461; SEQ ID NO: 169):


SHGFPPEVEEQAAGTLPMSCAQESGMDRHPAACASARINV






Interpretations

All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.


The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.”


It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.


In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of” and “consisting essentially of” shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.

Claims
  • 1-18. (canceled)
  • 19. An engineered nucleic acid comprising an expression cassette comprising a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-Dwherein S comprises a secretable effector molecule,C comprises a protease cleavage site,MT comprises a cell membrane tethering domain, andD comprises a degron,wherein the promoter is operably linked to the exogenous polynucleotide sequence, and wherein S-C-MT-D is configured to be expressed as a single polypeptide,optionally wherein the engineered nucleic acid is selected from the group consisting of: a DNA, cDNA, an RNA, an mRNA, and a naked plasmid.
  • 20. The engineered nucleic acid of claim 19, wherein: a) the promoter is a constitutive promoter,optionally wherein the constitutive promoter is selected from the group consisting of: CAG, HLP, CMV, EFS, SFFV, SV40, MND, PGK, UbC, hEF1aV1, hCAGG, hEF1aV2, hACTb, heIF4A1, hGAPDH, hGRP78, hGRP94, hHSP70, hKINb, and hUBIb; and/orb) the promoter is an inducible promoter,optionally wherein the inducible promoter is selected from the group consisting of: minP, NFkB response element, CREB response element, NFAT response element, SRF response element 1, SRF response element 2, AP1 response element, TCF-LEF response element promoter fusion, Hypoxia responsive element, SMAD binding element, STAT3 binding site, minCMV, YB_TATA, minTK, inducer molecule responsive promoters, and tandem repeats thereof; and/orc) the promoter is a synthetic promoter; and/ord) the promoter is a tissue-specific promoter.
  • 21. The engineered nucleic acid of claim 19, wherein: a) the secretable effector molecule comprises a native signal peptide native to the secretable effector molecule; orb) the secretable effector molecule comprises a non-native signal peptide non-native to the secretable effector molecule,optionally wherein the non-native signal peptide is selected from the group consisting of: IL12, IL2, optimized IL2, trypsinogen-2, Gaussia luciferase, CD5, human IgKVII, murine IgKVII, VSV-G, prolactin, serum albumin preprotein, azurocidin preprotein, osteonectin, CD33, IL6, IL8, CCL2, TIMP2, VEGFB, osteoprotegerin, serpin E1, GROalpha, CXCL12, IL21, CD8, and GMCSF.
  • 22. The engineered nucleic acid of claim 19, wherein the secretable effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a peptide, and an enzyme,optionally wherein the secretable effector molecule is a human-derived effector molecule,optionally wherein the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha,optionally wherein the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1 optionally wherein the homing molecule is selected from the group consisting of: anti-integrin alpha4,beta7; anti-MAdCAM; SDF1; and MMP-2optionally wherein the growth factor is selected from the group consisting of: FLT3L and GM-CSFoptionally wherein the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40Loptionally wherein the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2,optionally wherein the TGFbeta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereofoptionally wherein the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-L2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TNFa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody, andoptionally wherein the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof.
  • 23. The engineered nucleic acid of claim 19, wherein: a) the protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site; and/orb) the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain; and/orc) the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA; and/ord) the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof; and/ore) when expressed in a cell, the secretable effector molecule is tethered to a cell membrane of the cell; and/orf) when expressed in a cell expressing a protease capable of cleaving the protease cleavage site, the secretable effector molecule is released from the cell membrane; and/org) the protease expressed on the cell membrane is endogenous to the cell; and/orh) the protease is selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease; and/ori) the protease expressed on the cell membrane is heterologous to the cell;j) the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3); and/ork) the protease cleavage site comprises an NS3 protease cleavage site; and/orl) the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site; and/orm) the protease can be repressed by a protease inhibitor,optionally wherein the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir; and/orn) expression of the protease is capable of regulation.
  • 24. The engineered nucleic acid of claim 19, wherein the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule, optionally wherein the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN,optionally wherein the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences,optionally wherein the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189),optionally wherein the IMiD is an FDA-approved drug, wherein the IMiD is selected from the group consisting of: thalidomide, lenalidomide, and pomalidomide,optionally wherein the degron is selected from the group consisting of HCV NS4 degron, PEST (two copies of residues 277-307 of human IκBα), GRR (residues 352-408 of human p105), DRR (residues 210-295 of yeast Cdc34), SNS (tandem repeat of SP2 and NB (SP2-NB-SP2 of influenza A or influenza B), RPB (four copies of residues 1688-1702 of yeast RPB), SPmix (tandem repeat of SP1 and SP2 (SP2-SP1-SP2-SP1-SP2 of influenza A virus M2 protein), NS2 (three copies of residues 79-93 of influenza A virus NS protein), ODC (residues 106-142 of ornithine decarboxylase), Nek2A, mouse ODC (residues 422-461), mouse ODC_DA (residues 422-461 of mODC including D433A and D434A point mutations), an APC/C degron, a COP1 E3 ligase binding degron motif, a CRL4-Cdt2 binding PIP degron, an actinfilin-binding degron, a KEAP1 binding degron, a KLHL2 and KLHL3 binding degron, an MDM2 binding motif, an N-degron, a hydroxyproline modification in hypoxia signaling, a phytohormone-dependent SCF-LRR-binding degron, an SCF ubiquitin ligase binding phosphodegron, a phytohormone-dependent SCF-LRR-binding degron, a DSGxxS (SEQ ID NO: 190) phospho-dependent degron, an Siah binding motif, an SPOP SBC docking motif, and a PCNA binding PIP box, oroptionally wherein the degron is localized C-terminal of the cell membrane tethering domain.
  • 25. An expression vector comprising the engineered nucleic acid claim 19, optionally wherein the expression vector is a viral vector.
  • 26. A membrane-cleavable chimeric protein encoded by the engineered nucleic acid of claim 19.
  • 27. A membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-Dwherein S comprises a secretable effector molecule,C comprises a protease cleavage site,MT comprises a cell membrane tethering domain,D comprises a degron, andwherein S-C-MT-D is configured to be expressed as a single polypeptide.
  • 28. An isolated cell comprising the membrane-cleavable chimeric protein of claim 27, optionally wherein the cell is an autologous or allogeneic cell selected from the group consisting of: a T cell, a CD8+ T cell, a CD4+ T cell, a gamma-delta T cell, a cytotoxic T lymphocyte (CTL), a regulatory T cell, a viral-specific T cell, a Natural Killer T (NKT) cell, a Natural Killer (NK) cell, a B cell, a tumor-infiltrating lymphocyte (TIL), an innate lymphoid cell, a mast cell, an eosinophil, a basophil, a neutrophil, a myeloid cell, a macrophage, a monocyte, a dendritic cell, an erythrocyte, a platelet cell, a human embryonic stem cell (ESC), an ESC-derived cell, a pluripotent stem cell, a mesenchymal stromal cell (MSC), an induced pluripotent stem cell (iPSC), and an iPSC-derived cell.
  • 29. An isolated cell comprising an engineered nucleic acid, wherein the engineered nucleic acid comprises an expression cassette comprising a promoter and an exogenous polynucleotide sequence encoding a membrane-cleavable chimeric protein, oriented from N-terminal to C-terminal, having the formula: S-C-MT-Dwherein S comprises a secretable effector molecule,C comprises a protease cleavage site,MT comprises a cell membrane tethering domain, andD comprises a degron,wherein the promoter is operably linked to the exogenous polynucleotide sequence, and wherein S-C-MT-D is configured to be expressed as a single polypeptide,optionally wherein the cell is an autologous or allogeneic cell selected from the group consisting of: a T cell, a CD8+ T cell, a CD4+ T cell, a gamma-delta T cell, a cytotoxic T lymphocyte (CTL), a regulatory T cell, a viral-specific T cell, a Natural Killer T (NKT) cell, a Natural Killer (NK) cell, a B cell, a tumor-infiltrating lymphocyte (TIL), an innate lymphoid cell, a mast cell, an eosinophil, a basophil, a neutrophil, a myeloid cell, a macrophage, a monocyte, a dendritic cell, an erythrocyte, a platelet cell, a human embryonic stem cell (ESC), an ESC-derived cell, a pluripotent stem cell, a mesenchymal stromal cell (MSC), an induced pluripotent stem cell (iPSC), and an iPSC-derived cell.
  • 30. The isolated cell of claim 29, wherein the cell further comprises an antigen recognizing receptor, optionally wherein the antigen recognizing receptor recognizes an antigen selected from the group consisting of: 5T4, ADAM9, AFP, AXL, B7-H3, B7-H4, B7-H6, C4.4, CA6, Cadherin 3, Cadherin 6, CCR4, CD123, CD133, CD138, CD142, CD166, CD25, CD30, CD352, CD37, CD38, CD44, CD56, CD66e, CD70, CD71, CD74, CD79b, CD80, CEA, CEACAM5, Claudin18.2, cMet, CSPG4, CTLA, DLK1, DLL3, DR5, EGFR, ENPP3, EpCAM, EphA2, Ephrin A4, ETBR, FGFR2, FGFR3, FRalpha, FRb, GCC, GD2, GFRa4, gpA33, GPC3, gpNBM, GPRC5, HER2, IL-13R, IL-13Ra, IL-13Ra2, IL-8, IL-15, IL1RAP, Integrin aV, KIT, L1CAM, LAMP1, Lewis Y, LeY, LIV-1, LRRC, LY6E, MCSP, Mesothelin, MUC1, MUC16, MUC1C, NaPi2B, Nectin 4, NKG2D, NOTCH3, NY ESO 1, Ovarin, P-cadherin, pan-Erb2, PSCA, PSMA, PTK7, ROR1, S Aures, SCT, SLAMF7, SLITRK6, SSTR2, STEAP1, Survivin, TDGF1, TIM1, TROP2, and WT1,optionally wherein the antigen recognizing receptor comprises an antigen-binding domain,optionally wherein the antigen-binding domain comprises an antibody, an antigen-binding fragment of an antibody, a F(ab) fragment, a F(ab′) fragment, a single chain variable fragment (scFv), or a single-domain antibody (sdAb),optionally wherein the scFv comprises a heavy chain variable domain (VH) and a light chain variable domain (VL)optionally wherein the VH and VL are separated by a peptide linkeroptionally wherein the scFv comprises the structure VH-L-VL or VL-L-VH, wherein VH is the heavy chain variable domain, L is the peptide linker, and VL is the light chain variable domainoptionally wherein the antigen recognizing receptor is a chimeric antigen receptor (CAR)optionally wherein the CAR comprises one or more intracellular signaling domains, and the one or more intracellular signaling domains are selected from the group consisting of: a CD3zeta-chain intracellular signaling domain, a CD97 intracellular signaling domain, a CD11a-CD18 intracellular signaling domain, a CD2 intracellular signaling domain, an ICOS intracellular signaling domain, a CD27 intracellular signaling domain, a CD154 intracellular signaling domain, a CD8 intracellular signaling domain, an OX40 intracellular signaling domain, a 4-1BB intracellular signaling domain, a CD28 intracellular signaling domain, a ZAP40 intracellular signaling domain, a CD30 intracellular signaling domain, a GITR intracellular signaling domain, an HVEM intracellular signaling domain, a DAP10 intracellular signaling domain, a DAP12 intracellular signaling domain, and a MyD88 intracellular signaling domainoptionally wherein the CAR comprises a transmembrane domain, and the transmembrane domain is selected from the group consisting of: a CD8 transmembrane domain, a CD28 transmembrane domain a CD3zeta-chain transmembrane domain, a CD4 transmembrane domain, a 4-1BB transmembrane domain, an OX40 transmembrane domain, an ICOS transmembrane domain, a CTLA-4 transmembrane domain, a PD-1 transmembrane domain, a LAG-3 transmembrane domain, a 2B4 transmembrane domain, and a BTLA transmembrane domain, and/oroptionally wherein the CAR comprises a spacer region between the antigen-binding domain and the transmembrane domain.
  • 31. A composition comprising the isolated cell of claim 29, and a pharmaceutically acceptable carrier, pharmaceutically acceptable excipient, or a combination thereof.
  • 32. A method of treating a subject in need thereof, the method comprising administering the isolated cell of claim 29.
  • 33. A method of treating a subject in need thereof, the method comprising administering the composition of claim 31.
  • 34. A method of inducing release of a membrane-tethered effector molecule, comprising: a) providing the isolated cell of claim 29; andb) culturing the isolated cell under conditions suitable for expression of the membrane-bound protease and the membrane-cleavable chimeric protein,wherein upon expression, the membrane-cleavable chimeric protein is tethered to the cell membrane of the isolated cell, andwherein upon expression, the membrane-bound protease cleaves the cognate membrane-bound protease cleavage site of the membrane-cleavable chimeric protein, thereby releasing the effector molecule from the cell membrane.
  • 35. The method of claim 34, further comprising contacting the isolated cell with a drug that induces the degron, optionally wherein induction of the degron degrades the membrane-cleavable chimeric protein.
  • 36. The method of claim 34, wherein the secretable effector molecule is selected from a therapeutic class, wherein the therapeutic class is selected from the group consisting of: a cytokine, a chemokine, a homing molecule, a growth factor, a co-activation molecule, a tumor microenvironment modifier, a ligand, an antibody, a peptide, and an enzyme,optionally wherein the secretable effector molecule is a human-derived effector molecule,optionally wherein the cytokine is selected from the group consisting of: IL1-beta, IL2, IL4, IL6, IL7, IL10, IL12, an IL12p70 fusion protein, IL15, IL17A, IL18, IL21, IL22, Type I interferons, Interferon-gamma, and TNF-alpha,optionally wherein the chemokine is selected from the group consisting of: CCL21a, CXCL10, CXCL11, CXCL13, a CXCL10-CXCL11 fusion protein, CCL19, CXCL9, and XCL1optionally wherein the homing molecule is selected from the group consisting of: anti-integrin alpha4, beta7; anti-MAdCAM; SDF1; and MMP-2optionally wherein the growth factor is selected from the group consisting of: FLT3L and GM-CSFoptionally wherein the co-activation molecule is selected from the group consisting of: 4-1BBL and CD40Loptionally wherein the tumor microenvironment modifier is selected from the group consisting of: adenosine deaminase, a TGFbeta inhibitor, an immune checkpoint inhibitor, a VEGF inhibitor, and HPGE2,optionally wherein the TGFbeta inhibitor is selected from the group consisting of: an anti-TGFbeta peptide, an anti-TGFbeta antibody, a TGFb-TRAP, and a combination thereofoptionally wherein the immune checkpoint inhibitor is selected from the group consisting of: an anti-PD-1 antibody, an anti-PD-L1 antibody, an anti-PD-=2 antibody, an anti-CTLA-4 antibody, an anti-LAG-3 antibody, an anti-TIM-3 antibody, an anti-TIGIT antibody, an anti-VISTA antibody, an anti-KIR antibody, an anti-B7-H3 antibody, an anti-B7-H4 antibody, an anti-HVEM antibody, an anti-BTLA antibody, an anti-GAL9 antibody, an anti-A2AR antibody, an anti-phosphatidylserine antibody, an anti-CD27 antibody, an anti-TN Fa antibody, an anti-TREM1 antibody, and an anti-TREM2 antibody, andoptionally wherein the VEGF inhibitor comprises an anti-VEGF antibody, an anti-VEGF peptide, or a combination thereof.
  • 37. The method of claim 34, wherein: a) the cognate protease cleavage site is cleavable by a protease selected from the group consisting of: a Type 1 transmembrane protease cleavage site, a Type II transmembrane protease cleavage site, a GPI anchored protease cleavage site, an ADAM8 protease cleavage site, an ADAM9 protease cleavage site, an ADAM10 protease cleavage site, an ADAM12 protease cleavage site, an ADAM15 protease cleavage site, an ADAM17 protease cleavage site, an ADAM19 protease cleavage site, an ADAM20 protease cleavage site, an ADAM21 protease cleavage site, an ADAM28 protease cleavage site, an ADAM30 protease cleavage site, an ADAM33 protease cleavage site, a BACE1 protease cleavage site, a BACE2 protease cleavage site, a SIP protease cleavage site, an MT1-MMP protease cleavage site, an MT3-MMP protease cleavage site, an MT5-MMP protease cleavage site, a furin protease cleavage site, a PCSK7 protease cleavage site, a matriptase protease cleavage site, a matriptase-2 protease cleavage site, an MMP9 protease cleavage site, and an NS3 protease cleavage site; and/orb) the cell membrane tethering domain comprises a transmembrane-intracellular domain or a transmembrane domain,optionally wherein the transmembrane-intracellular domain and/or transmembrane domain is derived from PDGFR-beta, CD8, CD28, CD3zeta-chain, CD4, 4-1BB, OX40, ICOS, CTLA-4, PD-1, LAG-3, 2B4, LNGFR, EpoR, or BTLA; and/orc) the cell membrane tethering domain comprises a cell surface receptor, or a cell membrane-bound portion thereof; and/ord) the protease is endogenous to the cell and selected from the group consisting of: a Type 1 transmembrane protease, a Type II transmembrane protease, a GPI anchored protease, an ADAM8 protease, an ADAM9 protease, an ADAM10 protease, an ADAM12 protease, an ADAM15 protease, an ADAM17 protease, an ADAM19 protease, an ADAM20 protease, an ADAM21 protease, an ADAM28 protease, an ADAM30 protease, an ADAM33 protease, a BACE1 protease, a BACE2 protease, a SIP protease, an MT1-MMP protease, an MT3-MMP protease, an MT5-MMP protease, a furin protease, a PCSK7 protease, a matriptase protease, a matriptase-2 protease, and an MMP9 protease; and/ore) the protease is heterologous to the cell, optionally wherein the protease is hepatitis C virus (HCV) nonstructural protein 3 (NS3); and/orf) the protease cleavage site comprises an NS3 protease cleavage site, optionally wherein the NS3 protease cleavage site comprises a NS3/NS4A, a NS4A/NS4B, a NS4B/NS5A, or a NS5A/NS5B junction cleavage site; and/org) the protease can be repressed by a protease inhibitor, optionally wherein the protease inhibitor is selected from the group consisting of: simeprevir, danoprevir, asunaprevir, ciluprevir, boceprevir, sovaprevir, paritaprevir, telaprevir, grazoprevir, glecaprevir, and voxiloprevir; and/orh) the protease further comprises a degron, optionally wherein expression of the protease is capable of regulation,optionally wherein the degron comprises a cereblon (CRBN) polypeptide substrate domain capable of binding CRBN in response to an immunomodulatory drug (IMiD) thereby promoting ubiquitin pathway-mediated degradation of the secretable effector molecule,optionally wherein the CRBN polypeptide substrate domain is selected from the group consisting of: IKZF1, IKZF3, CK1a, ZFP91, GSPT1, MEIS2, GSS E4F1, ZN276, ZN517, ZN582, ZN653, ZN654, ZN692, ZN787, and ZN827, or a fragment thereof that is capable of drug-inducible binding of CRBN,optionally wherein the CRBN polypeptide substrate domain is a chimeric fusion product of native CRBN polypeptide sequences, optionally wherein the CRBN polypeptide substrate domain is a IKZF3/ZFP91/IKZF3 chimeric fusion product having the amino acid sequence of FNVLM VHKRS HTGER PLQCE ICGFT CRQKG NLLRH IKLHT GEKPF KCHLC NYACQ RRDAL (SEQ ID NO: 189), and
RELATED APPLICATIONS

This application is a continuation of International Application No. PCT/US2022/012570, filed Jan. 14, 2022, which claims the benefit of and priority to U.S. Provisional Patent Application No. 63/137,419, filed Jan. 14, 2021, each of which are hereby incorporated by reference in their entireties for all purposes.

Provisional Applications (1)
Number Date Country
63137419 Jan 2021 US
Continuations (1)
Number Date Country
Parent PCT/US2022/012570 Jan 2022 US
Child 18352079 US