Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
A METHOD TO SPECIFICALLY STIMULATE SURVIVAL AND EXPANSION OF GENETICALLY-MODIFIED IMMUNE CELLS
Document Type and Number:
WIPO Patent Application WO/2020/044239
Kind Code:
A1
Abstract:
The present invention relates to a vector encoding a wildtype or truncated form of erythropoietin receptor (EpoR) to promote T cell survival and proliferation. Specifically, it exemplifies a bicist ronic vector that expresses a truncated EpoR with an anti-CD19-41 ΒΒ-003ζ chimeric antigen receptor (CAR) that has a greater ex vivo expansion than CAR- T cells and demonstrates a significantly higher anti-leukemic activity in vivo. It also describes the method of producing cells expressing said vector and the use of these cells to kill CD19+ tumour cells.

Inventors:
VINANICA NATASHA (SG)
YONG ARTHUR (SG)
CAMPANA DARIO (SG)
IMAMURA MASARU (JP)
Application Number:
PCT/IB2019/057217
Publication Date:
March 05, 2020
Filing Date:
August 27, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NAT UNIV SINGAPORE (SG)
International Classes:
C07K14/715; A61K35/17; A61P35/00; C07K14/725; C12N5/071
Domestic Patent References:
WO2013043196A12013-03-28
WO2016118857A12016-07-28
WO2005000890A12005-01-06
Other References:
ROYER Y. ET AL.: "High-throughput gateway bicistronic retroviral vectors for stable expression in mammalian cells: exploring the biologic effects of STAT5 overexpression", DNA CELL BIOL., vol. 23, no. 6, June 2004 (2004-06-01), pages 355 - 365, XP055695462, [retrieved on 20191105]
MINAMOTO S. ET AL.: "Acquired Erythropoietin Responsiveness of Interleukin- 2-dependent T lymphocytes Retrovirally Transduced with Genes Encoding Chimeric Erythropoietin/lnterleukin-2 Receptors", BLOOD, vol. 86, no. 6, 15 September 1995 (1995-09-15), pages 2281 - 2287, XP055777976
MOHAMMED S. ET AL.: "Improving Chimeric Antigen Receptor-Modified T Cell Function by Reversing the Immunosuppressive Tumor Microenvironment of Pancreatic Cancer", MOL THER., vol. 25, no. 1, 4 January 2017 (2017-01-04), pages 249 - 258, XP055695458, [retrieved on 20191105]
WILKIE S. ET AL.: "Selective Expansion of Chimeric Antigen Receptor-targeted T-cells with Potent Effector Function using Interleukin-4", J BIOL CHEM., vol. 285, no. 33, 18 June 2010 (2010-06-18), pages 25538 - 25544, XP002762491, [retrieved on 20191105]
DE LA CHAPELLE A. ET AL.: "Truncated erythropoietin receptor causes dominantly inherited benign human erythrocytosis", PROC NATL ACAD SCI USA., vol. 90, no. 1 0, 15 May 1993 (1993-05-15), pages 4495 - 4499, XP055695455, [retrieved on 20191105]
See also references of EP 3844186A4
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A vector comprising a nucleic acid encoding:

a) an erythropoietin (Epo) receptor;

b) a self-cleaving peptide or internal ribosome entry site; and

c) a cell surface receptor.

2. The vector of Claim 1, wherein the Epo receptor has at least 90% sequence identity to SEQ ID NO: 2.

3. The vector of Claim 1, wherein the Epo receptor is a mutant Epo receptor.

4. The vector of Claim 3, wherein the nucleic acid has a mutation that encodes a stop codon within exon 8 of the Epo receptor.

5. The vector of Claim 3, wherein the Epo receptor has at least 90% sequence identity to SEQ ID NO: 6.

6. The vector of Claim 1, wherein the nucleic acid further encodes a Flag tag

(DYKDDDDK (SEQ ID NO: 23)) that is C terminal to the Epo receptor.

7. The vector of Claim 1, wherein the nucleic acid comprises a self-cleaving peptide.

8. The vector of Claim 7, wherein the self-cleaving peptide is a 2A peptide.

9. The vector of Claim 1, wherein the cell surface receptor comprises an extracellular receptor domain that binds a target cell antigen.

10. The vector of any one of Claims 1 through 8, wherein the cell surface receptor is a chimeric antigen receptor comprising:

i) a signal peptide;

ii) an extracellular receptor domain that binds a target cell antigen;

iii) a hinge and transmembrane domain that anchors the extracellular receptor domain on the surface of a cell; and

iv) an effector domain.

11. The vector of Claim 10, wherein the extracellular receptor domain comprises a variable immunoglobulin light chain domain and a variable immunoglobulin heavy chain domain joined by a linker domain.

12. The vector of Claim 11, wherein the linker domain is (G4S)x (SEQ ID NO: 24), wherein x is an integer from 1 to 100.

13. The vector of Claim 10, wherein the extracellular receptor domain is a single-chain variable fragment (scFv).

14. The vector of any one of Claims 1 through 8, wherein the cell surface receptor is a T cell receptor.

15. The vector of Claim 10, wherein the extracellular receptor domain is an anti-CD 19 single-chain variable fragment (scFv).

16. The vector of Claim 10, wherein the chimeric antigen receptor is anti-CD 19-41BB- Oϋ3z.

17. A method of making a transgenic mammalian host cell, the method comprising

introducing into a mammalian host cell the vector of any of Claims 1 through 16.

18. The method of Claim 17, wherein the mammalian host cell is an immune cell.

19. The method of Claim 18, wherein the immune cell is a natural killer (NK) cell, a monocyte/macrophage cell, or a dendritic cell.

20. The method of Claim 18, wherein the immune cell is a T cell.

21. The method of Claim 20, wherein the T cell further expresses a T-cell receptor (TCR) that binds a tumor antigen or a viral antigen.

22. The method of Claim 21, wherein the TCR is endogenous.

23. The method of Claim 22, wherein T cell is a tumor-infiltrating lymphocyte (TIL), and wherein the method further comprises extracting the tumor-infiltrating lymphocyte from a tumor and expanding the TIL ex vivo.

24. A mammalian immune cell comprising the vector of any one Claims 1 through 16.

25. The mammalian immune cell of Claim 24, wherein the mammalian immune cell is a natural killer (NK) cell, a monocyte/macrophage cell, or a dendritic cell.

26. The mammalian immune cell of Claim 24, wherein the mammalian immune cell is a T cell.

27. The mammalian immune cell of Claim 26, wherein the T cell is a human T cell.

28. The mammalian immune cell of Claim 26, wherein the T cell is a human peripheral blood T lymphocyte.

29. A method of reducing the number of CD 19+ cells in a mammal, the method

comprising introducing mammalian T cells into the subject, wherein the mammalian T cells comprise the vector of any of Claims 1 through 16.

Description:
A Method to Specifically Stimulate Survival and Expansion of Genetically-Modified Immune

Cells

RELATED APPLICATION

[0001] This application claims the benefit of LT.S. Provisional Application No.

62/724,488, filed on August 29, 2018. The entire teachings of the above application is incorporated herein by reference.

INCORPORATION BY REFERENCE OF MATERIAL IN ASCII TEXT FILE

[0002] This application incorporates by reference the Sequence Listing contained in the following ASCII text file being submitted concurrently herewith:

a) File name: 4459_l l5l-00l_PCT_SL.txt; created August 26, 2019, 54,419 bytes in size.

BACKGROUND

[0003] For patients with cancer, adoptive transfer of immune cells is a promising and increasingly available treatment option, one that can lead to clinical responses even when all standard treatment has failed. Clinical studies with tumor-infiltrating lymphocytes, and T lymphocytes redirected towards tumor-associated molecules with T-cell receptors (TCRs) or chimeric antigen receptors (CARs) have provided compelling evidence of the potential of these approaches in patients with leukemia and solid tumors. 1 For example, treatment of B- cell leukemia and lymphoma with anti-CD 19 CAR-T cells has led to durable remissions in patients with disease resistant to conventional therapy. 2 10

[0004] Several factors collectively influence proliferation and life-span of infused T lymphocytes, including the intensity of lymphodepleting therapy prior to infusion, and the proliferative potential and exhaustion propensity of the T cell infused. 1,9 In the case of CAR- engineered T cells, the quality of the CAR is an important feature and the type of co stimulation that the CAR can deliver appears to play an important role. 11 13

[0005] Interleukin-2 (IL-2) promotes expansion and persistence of T cells in vivo and it is used in some cell therapy trials for this purpose. 14,15 However, administration of IL-2 can have considerable toxi cities. 16,17 Moreover, it lacks specificity as it reacts with all T cells expressing IL-2 receptors, regardless of their anti-tumor capacity. To this end, IL-2 stimulates regulatory T cells, which dampen immune responses. 18

SUMMARY

[0006] Described herein are vectors, nucleic acids, and transgenic cells that can be used to improve the clinical efficacy of adoptive cell therapy by improving expansion and/or persistence of the infused cells.

[0007] Described herein is a vector that includes a nucleic acid. The nucleic acid encodes an erythropoietin (Epo) receptor; a self-cleaving peptide or internal ribosome entry site; and a cell surface protein.

[0008] The Epo receptor can have at least 90% sequence identity to any of SEQ ID NOS: 2, 4, 6, and 8. In some embodiments, the Epo receptor can have at least 90% sequence identity to SEQ ID NOS: 2. In some embodiments, the Epo receptor can have at least 90% sequence identity to SEQ ID NO: 4. In some embodiments, the Epo receptor can have at least 90% sequence identity to SEQ ID NO: 6. In some embodiments, the Epo receptor can have at least 90% sequence identity to SEQ ID NO: 8. In some embodiments, the Epo receptor is a mutant Epo receptor. In some embodiments, the nucleic acid has a mutation that encodes a stop codon within exon 8 of the Epo receptor.

[0009] The nucleotide can further encode a Flag tag (DYKDDDDK (SEQ ID NO: 23)) that is C terminal to the Epo receptor. The nucleic acid can include a self-cleaving peptide, such as a 2A peptide (e.g., T2A, P2A, E2A, F2A). In some instances, the 2A peptide can be a T2A peptide.

[0010] The signal peptide can be a signal peptide of a surface protein, such as CD8a signal peptide.

[0011] The cell surface receptor can include an extracellular receptor domain that binds a target cell antigen.

[0012] The cell surface receptor can be a chimeric antigen receptor (CAR). The CAR can include a signal peptide; an extracellular receptor domain that binds a target cell antigen; a hinge and transmembrane domain that anchors the extracellular receptor domain on the surface of a cell; and an effector domain. Where the cell surface receptor is a chimeric antigen receptor, the extracellular domain is typically a single-chain variable fragment (scFv).

[0013] The extracellular receptor domain can include a variable immunoglobulin light chain domain and a variable immunoglobulin heavy chain domain joined by a linker domain. The linker domain can be (G4S)x (SEQ ID NO: 24), wherein x is an integer from 1 to 100. The linker domain can be (G4S)3 (SEQ ID NO: 25).

[0014] The cell surface receptor can be a T-cell receptor.

[0015] The extracellular domain can include a monoclonal antibody, a recombinant antibody, a human antibody, a humanized antibody, a Fab, a Fab', a F(ab')2, an Fv, a single- chain variable fragment (scFv), a minibody, a diabody, a single-domain antibody, or a functional derivative or variant or fragment thereof.

[0016] The extracellular receptor domain can include an immunoglobulin Fc receptor, such as CD 16, CD32 or CD64. The extracellular receptor domain can include a cytokine, such as IL-13, IL-4, IL-7, or IL-3.

[0017] The cell surface receptor can activate immune cells. For example, the cell surface receptor can include NKG2D, NKG2C, NCR1, NCR2, NCR3, CD137, CD28, or ICOS. The cell surface receptor can include a fragment of NKG2D, NKG2C, NCR1, NCR2, NCR3, CD137, CD28, or ICOS. The cell surface receptor can include a ligand of NKG2D, NKG2C, NCR1, NCR2, NCR3, CD137, CD28, or ICOS.

[0018] The cell surface receptor can inhibit immune cells. For example, the cell surface receptor can include NKG2A, PD-l, or CTLA-4. The cell surface receptor can include a fragment of NKG2A, PD-l, or CTLA-4. The cell surface receptor can include a ligand of NKG2A, PD-l, or CTLA-4.

[0019] The cell surface receptor can be a receptor for a cytokine. For example, the cell surface receptor can be a receptor for IL-6, IL-l, or TNF alpha.

[0020] The target cell antigen can be a tumor associated antigen or a tumor specific antigen. The target cell antigen can be a viral, bacterial, fungal, or parasite associated antigen.

[0021] The target cell antigen is CD19, CD20, CD22, CD123, CD33, B-cell maturation antigen (BCMA), mesothelin, human epidermal growth factor receptor 2 (Her2), prostate- specific membrane antigen (PSMA), or disialoganglioside (GD)-2.

[0022] The target cell antigen can be CD 19.

[0023] The extracellular domain can be an anti-CD 19 single-chain variable fragment (scFv). The hinge and transmembrane domain can be a CD8a hinge and transmembrane domain. The hinge can include a plurality of amino acid residues. The transmembrane domain can be a transmembrane domain from CD4, CD8P, CD 16, CD28, CD32, CD34, CD64, CD 137, FceRIy, 0X40, Oϋ3z, CD3e, CD3y, CD35, TCRa, VEGFR2, FAS, or FGFR2B. [0024] The effector domain can include 4-1BB and CD3z. The CAR can be anti-CDl9- 41BB-Oϋ3z.

[0025] The vector can be a retrovirus, such as a murine stem cell virus (MSCV) retroviral vector. The vector can further encode a fluorescent protein. The vector can encode an internal ribosomal entry site (IRES). The vector can further encode at least one regulatory element for expression of the nucleic acid.

[0026] Described herein is a method of making a transgenic mammalian host cell. The method can include introducing into a mammalian host cell any of the vectors described herein. The mammalian host cell can be an immune cell, such as a natural killer (NK) cell, a monocyte/macrophage cell, a dendritic cell, or a T cell. The T cell can be a human peripheral blood T lymphocyte. The T cell can be a CD4+ T cell. The T cell can be a CD8+ T cell.

The T cell can further expresses a T-cell receptor (TCR) that binds a tumor antigen or a viral antigen. The TCR is endogenous. For example, the T cell can be a tumor-infiltrating lymphocyte (TIL), and the method can further include extracting the tumor-infiltrating lymphocyte from a tumor and expanding the TIL ex vivo. The TCR can be exogenous. For example, the method can further include introducing into the T cell a vector that expresses the exogenous TCR.

[0027] Described herein is a mammalian immune cell comprising any of the vectors described herein. The mammalian immune cell can be a natural killer (NK) cell, a monocyte/macrophage cell, a dendritic cell, or a T cell. The T cell can be a human T cell.

The T cell can be a human peripheral blood T lymphocyte. The T cell can be as described in the preceding paragraph or as otherwise described herein.

[0028] Described herein is a method of reducing the number of CD 19+ cells in a mammal. The method can include introducing mammalian T cells into the subject. The mammalian T cells can include any of the vectors described herein. The mammal can be a human. The mammalian T cells can be autologous cells isolated from the mammal. The mammalian T cells can be allogenic cells isolated from a donor. The method can further include administering Epo to the subject. The method can further include administering IL-2 to the subject. Reducing the number of CD 19+ cells in the mammal can treat acute lymphoblastic leukemia (ALL).

[0029] Described herein is use of any of the vectors described herein in the manufacture of a medicament for treating or preventing cancer, a viral infection, a bacterial infection, a fungal infection, or a parasite in a mammal in need thereof. [0030] Described herein is use of any of the mammalian immune cells described herein for reducing the number of CD19+ cells in a mammal.

[0031] Described herein is a vector for use in a method for reducing the number of CD19+ cells in a mammal. The vector can be any of the vectors described herein.

[0032] Described herein is a mammalian immune cell for use in a method for reducing the number of CD19+ cells in a mammal. The mammalian immune cell can be any of the mammalian immune cells described herein.

[0033] Described herein is a vector that includes a nucleic acid encoding a mutant erythropoietin (Epo) receptor. For example, the mutant Epo receptor can have at least 90% sequence identity to SEQ ID NO: 6. Also described herein is a method of making a transgenic mammalian host cell by introducing into a mammalian host cell the vector that includes a nucleic acid encoding a mutant erythropoietin (Epo) receptor. Also described herein is a mammalian immune cell that includes the vector that includes a nucleic acid encoding a mutant erythropoietin (Epo) receptor. The mammalian immune cell can be a T cell, natural killer (NK) cell, a monocyte/macrophage cell, or a dendritic cell.

[0034] Epo receptor can be expressed in T cells and delivers signals.

[0035] EpoRm is expressed at higher levels than EpoR.

[0036] EpoRm induces stronger and more durable signals.

[0037] Epo can support the proliferation of EpoRm-CAR-T cells.

[0038] Expression of EpoRm and exposure to Epo do not interfere with cytotoxicity of CAR-T cells.

[0039] EpoRm-CAR-T cells can expand and exert cytotoxicity in vivo.

BRIEF DESCRIPTION OF THE DRAWINGS

[0040] The foregoing will be apparent from the following more particular description of example embodiments, as illustrated in the accompanying drawings in which like reference characters refer to the same parts throughout the different views. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating embodiments.

[0041] FIGs. 1 A-E show expression of EpoR in human peripheral blood T cells confers survival signal. FIG. 1 A is flow cytometric dot-plots that illustrate surface EpoR expression in Jurkat cells, as detected by a PE-conjugated anti-EpoR antibody (R&D Systems); cells transduced with GFP only are shown as control. Percentage of cells in each quadrant is shown. FIG. 1B is flow cytometric histograms that illustrate Epo binding to Jurkat cells. Cells were labelled with biotin-conjugated Epo (R&D Systems) and streptavidin-PE (Jackson ImmunoResearch). FIG. 1C is flow cytometric dot-plots that illustrate surface EpoR expression in T lymphocytes transduced with GFP only or GFP plus EpoR. FIG. 1D is representative flow cytometric dot-plots that illustrate phosphorylation of STAT5 Y647, detected with an AF647-conjugated antibody (BD Biosciences), after stimulation with 10 IU/mL Epo for 15 minutes; results with cells treated with 10 mM ruxolitinib for 1 hour prior to Epo stimulation are also shown. FIG. 1E is graphs showing survival of EpoR-transduced T cells and of T cells transduced with GFP only cultured in absence of exogenous cytokines (no Epo) or in presence of Epo (4 IU/mL). Symbols indicate percentage of cell recovery relative to the number of input cells.

[0042] FIGs. 2A-F show that EpoRm has higher and more stable expression than EpoR. FIG. 2A is a western blot analysis of EpoR expression in 293T cells. Cell lysates of 293T cells transduced with EpoR, EpoRm or GFP only were separated on a 10% polyacrylamide gel under reducing condition. The blotted membrane was probed with mouse anti-Flag antibody (9 A3; Cell Signaling Technology) followed by goat anti-mouse IgG conjugated to horseradish peroxidase (HRP, R&D Systems); rabbit anti-human GAPDH (EPR16891;

Abeam) followed by goat anti-rabbit IgG conjugated to HRP (Abeam) was used to detect GAPDH (loading control). Antibody binding was revealed by Clarity Western ECL substrate (Bio-Rad) and visualized by ChemiDoc Touch Imager (Bio-Rad). FIG. 2B is flow cytometric analysis of T lymphocytes transduced with EpoR, EpoRm or GFP only. Flow cytometric dot- plots illustrate EpoR expression as detected by a PE-conjugated anti-EpoR antibody (R&D Systems). Percentage of cells in each quadrant is shown. FIG. 2C is charts showing the percentage of GFP+ T cells expressing EpoR (left) or mean fluorescent intensity (MFI) of EpoR (right) of T cells transduced with EpoR or EpoRm. ****, p < 0.0001. FIG. 2D is charts showing the percentage of CD4+ (left) or CD8+ (right) T cells from 6 donors expressing EpoR. ****, P < 0.0001. FIG. 2E is graphs showing the relation between MFI of GFP and MFI of EpoR in T cells from 3 donors transduced with either EpoR or EpoRm. Each panel shows results with T cells of 1 donor. ****, p < 0.0001. FIG. 2F is graph showing T lymphocytes from 3 donors that were transduced with EpoR or EpoRm and then stimulated with 10 IU/mL Epo. Expression of surface EpoR was assessed by flow cytometry at the indicated time points after stimulation. Each panel shows results with T cells of 1 donor.

[0043] FIGs. 3 A-E show that EpoRm enhances Epo signaling. FIG. 3 A is representative flow cytometric contour plots that illustrate phosphorylation of STAT5 in T cells transduced with EpoR, EpoRm or GFP only after stimulation with Epo (10 IU/mL) for 15 minutes. FIG. 3B is graphs showing the relation between MFI of GFP and MFI of pSTAT5 in T cells from 3 donors transduced with either EpoR or EpoRm. Each panel shows results with T cells of 1 donor. ****, p < 0.0001. FIG. 3C is plots showing percentage of GFP+ cells expressing pSTAT5 in T cells transduced with EpoR, EpoRm or GFP only after stimulation with 10 IU/mL Epo or treated with 10 mM ruxolitinib prior to Epo stimulation. Each symbol represents results of one experiment. ****, p < 0.0001; **, P < 0.01. FIG. 3D is a graph showing percentage of GFP+ cells expressing pSTAT5 following stimulation with different concentration of Epo for 15 minutes. Each symbol indicates the average value of 2 experiments. FIG. 3E is a graph showing T cells transduced with EpoR, EpoRm or GFP only stimulated with 10 IU/mL Epo, and pSTAT5 was assessed by flow cytometry at the indicated time points. Each symbol indicates the average value of 2 experiments.

[0044] FIGs. 4A-D show proliferation and survival signals induced by EpoRm in T cells. FIG. 4A are flow cytometry dot-plots illustrate cell cycle analysis of T lymphocytes transduced with EpoR, EpoRm or GFP only, unstimulated (top row) or stimulated with 10 IU/mL Epo (bottom row) after 3 days of culture in cytokine-free medium. DNA content, detected by FxCycle staining, is shown in the x axes; DNA synthesis, shown by Edu incorporation, is shown on the y axes. Edu+ cells are shown in box, with their percentage. FIG. 4B is graphs showing survival of T lymphocytes transduced with EpoR, EpoRm or GFP-only cultured in absence of exogenous cytokines (no Epo) or in presence of Epo (10 IU/mL) for 3 weeks. Symbols indicate mean (± SD) percentage of cell recovery relative to the number of input cells in triplicate measurements. **** P <0.0001; ** P <0.01. FIG. 4C is graphs showing percentage of T cell recovery relative to input cells after 6-8 days culture with or without 100 IU/mL IL-2 and/or 10 IU/mL Epo. Each symbol indicates measurements with T cells of 1 of 4 donors (mean of 3 measurements for 3 donors, and 1 measurement for 1 donor). * P = 0.02. FIG. 4D is graphs showing survival of T lymphocytes transduced with EpoR, EpoRm or GFP only cultured with 100 IU/mL IL-2 in the absence or presence of Epo (10 IU/mL). Percentage of T cell recovery relative to input cells at the indicated days is shown.

[0045] FIGs. 5A-F show expression and function of EpoRm-CAR T cells. FIG. 5 A is a schematic representation of the EpoRm-CAR construct. FIG. 5B is flow cytometric dot-plots illustrate surface expression of CAR and EpoR in T cells transduced with EpoRm-CAR; cells transduced with GFP only were used as control. Percentage of cells in each quadrant is shown. Cells were stained with EpoR-PE antibody to detect EpoR expression; CAR expression was detected with goat anti-mouse F(ab’)2 antibody and streptavidin-APC

(Jackson ImmunoResearch). FIG. 5C are plots showing percentage of GFP+ cells expressing CAR (left) and EpoR (right) among T lymphocytes transduced with CAR, EpoRm-CAR or GFP only. Each symbol corresponds to a measurement for one transduction. Horizontal bars indicate median value. FIG. 5D are plots showing percentage of GFP+ cells expressing pSTAT5 in T lymphocytes transduced with CAR, EpoRm-CAR or GFP only after 15 minutes stimulation with 10 IU/mL Epo, or 1 hour pre-treatment with 10 mM ruxolitinib prior to Epo stimulation. Each symbol corresponds to a measurement for one transduction. Horizontal bars indicate median value. FIG. 5E is charts showing cytotoxicity of T lymphocytes transduced with CAR, EpoRm-CAR or GFP only against the CD19+ cell lines OP-l, RS4;l 1 and Nalm6. Bars represent mean (± SD) of triplicate experiments in a 4-hour cytotoxicity assay at a 1 :1 E:T ratio. FIG. 5F is graphs showing long-term cytotoxicity of T lymphocytes transduced with CAR, EpoRm-CAR or GFP only to OP-l mCherry cells at the indicated E:T ratios in presence of 10 IU/mL Epo. Symbols represent the mean (± SD) percentage cytotoxicity measured at the indicated time points. * P = 0.03; ** P < 0.01.

[0046] FIGs. 6A-F show that Epo supports the proliferation of EpoRm-CAR T cells. FIG. 6A is charts that show survival and expansion of T lymphocytes from 3 donors transduced with CAR, EpoRm-CAR or GFP only co-cultured with Streck-treated or irradiated OP-l cells at 1 : 1 ratio, in the absence or presence of 10 IU/mL Epo. Each symbol indicates percentage of cell recovery as compared with the number of input cells. Mean (± SD) of triplicate cultures is shown, except for the day 14 measurement for 1 donor, where only the mean of 2 measurements is shown. FIG. 6B is a plot showing recovery of EpoRm-CAR-transduced T cells relative to input cells after 7 days of culture with or without 1 mM ruxolitinib in the presence of 10 IU/mL Epo. *** P < 0.00l. FIG. 6C is charts showing percentage of T lymphocytes transduced with CAR, EpoRm-CAR or GFP only recovered 7 days after co culture with irradiated CD 19+ OP-l at 1 : 1 ratio in the presence of 10 IU/mL Epo with 10 IU/mL or 100 IU/mL of IL-2. Each symbol indicates the mean of triplicate cultures. FIG. 6D is plots showing MFI of pSTAT5 in EpoRm-CAR-transduced T cells after stimulation with 10 IU/mL Epo and/or 100 IU/mL IL-2 at the indicated time points. FIG. 6E is plots showing percentage of GFP+ EpoRm-CAR-transduced T cells expressing pSTAT5 stimulated with the indicated concentrations of Epo and IL-2. * P = 0.045; ** P <0.01. FIG. 6F is charts showing EpoRm-CAR-transduced T cells were treated with either tofacitinib (n = 2) or ruxolitinib (n = 1) for 1 hour at the indicated doses prior to stimulation with either 10 IU/mL Epo or 100 IU/mL IL-2. Bars represent mean of two measurements for tofacitinib and a single measurement for ruxolitinib.

[0047] FIGs. 7A-E show in vivo tumor killing and expansion of EpoRm-CAR T cells in immunodeficient mice. FIG. 7A are representative images of NSG mice infused i.v. with 5 x 10 5 Nalm6-luciferase cells. Two days later, 4 mice were injected i.v. with 2 x 10 7 EpoRm- CAR T cells; 2 of them received i.p. injections of 100 IU Epo 3 times a week for 2 weeks; 1 mouse received no T cells. Bioluminescence ventral images on day 2 are shown with enhanced sensitivity to document Nalm6 engraftment. FIG. 7B is plots showing leukemia cell growth expressed as photons per second in mice shown in Panel 7A. Symbols corresponds to total bioluminescence by ventral and dorsal imaging. Mice were euthanized when the aggregate ventral and dorsal bioluminescence signal reached 1 x 10 10 photons per second.

FIG. 7C is a plot showing absolute number of human CD45+ cells in peripheral blood of mice injected i.v. with T cells transduced with CAR, EpoRm-CAR, or GFP only; some mice received 100 IU Epo i.p. 3 times a week for 2 weeks, as indicated. Blood was obtained via cheek bleed 13 days after T-cell infusion. * P = 0.02; ** P<0.0l. FIG. 7D is representative flow cytometric contour plots that illustrate the presence of human CD45+ GFP+ (top) and human CD45+ (bottom) T cells in mouse peripheral blood. FIG. 7E is a chart showing pSTAT5 MFI in T cells transduced with CAR, EpoRm-CAR, or GFP only following stimulation with 80 ng/mL of either mouse or human Epo for 15 minutes.

[0048] FIGs. 8A-C show degranulation and cytokine release profile of EpoRm-CAR T cells. FIG. 8A is a chart showing percentage of GFP+ cells expressing CDl07a in T cells transduced with CAR, EpoRm-CAR, or GFP only, after co-culture with OP-l cells at 1 : 1 E:T ratio for 4 hours. Bars represent mean (± SD) triplicate measurements with cells from 3 donors. FIG. 8B is graphs showing percentage of GFP+ cells expressing IFNy (left) and TNFa (right) in T cells transduced as in FIG. 8 A after 6 hours of co-culture with OP-l cells at 1 : 1 E:T ratio. Mean (± SD) of triplicate measurement with cells from 3 donors for IFNy and 1 donor for TNFa are shown. *** P <0 .001. FIG. 8C is plots of Luminex analysis of cytokines production by EpoRm-CAR-transduced T cells after co-culture with OP-l cells at 1 :4 E:T ratio for 24 hours in absence or presence of 10 IU/mL Epo.

[0049] FIGs. 9A-F show anti -leukemic activity of EpoRm-CAR-T cells in xenograft models. FIG. 9A is images of NOD/scid IL2RGnull mice injected i.v. with 1 x 10 7 T cells transduced with CAR, EpoRm-CAR or GFP only; one group of mice received no T cells. Two weeks later, mice were injected i.v. with 2.5 x 10 5 Nalm6 cells expressing luciferase. Ventral images illustrate Nalm6 cell engraftment as measured by luminescence after i.p. injection of aqueous D-luciferin potassium salt (150 pg/g body weight). FIG. 9B is charts showing luminescence measurements in the groups of mice shown in FIG. 9A. Shown is the number of mice in each group with signals below l0 8 photons/second on day 59. FIG. 9C is a chart showing persistence of infused T cells in the mice shown in FIG. 9A. Mouse blood was collected by cheek prick and cells were stained with APC (2D1; Biolegend) or PE/Cy7- conjugated anti-human CD45 (HI30; BD Pharmingen), and PE-conjugated anti-mouse CD45 (30-F11; BD Pharmingen). Percentage of hCD45+ GFP+ cells among all CD45+ (human and mouse) lymphoid cells is shown. FIG. 9D is images where Nalm6 cells transduced with luciferase were injected i.v. in NOD/scid IL2RGnull mice (5 x 10 5 cells per mouse). Four days later, tumor engraftment was assessed and 1-2 x 10 7 T cells were injected i.v.. Ventral images illustrate Nalm6 cell engraftment as measured by luminescence after i.p. injection of aqueous D-luciferin potassium salt (150 pg/g body weight). FIG. 9E is charts showing luminescence measurements in the groups of mice shown in FIG. 9D. The number of mice in each group with signals below l0 8 photons/second on day 55 is shown. FIG. 9F is a chart showing aggregate long-term survival of mice included in the two sets of experiments.

[0050] FIG. 10 is charts showing growth curves of Jurkat cells transduced with Epo receptors or GFP only. Each symbol represents the mean of three measurements.

DETAILED DESCRIPTION

[0051] A description of example embodiments follows.

[0052] Described herein is experiments pertinent to whether ectopic expression of the wild-type erythropoietin (Epo) receptor and of a naturally occurring truncated form associated with erythrocytosis could confer Epo responsiveness to human peripheral blood lymphocytes. ETsing T cells transduced with a single construct encoding for the receptor and a CAR, the potential of Epo to specifically expand CAR-T cells in vitro and in vivo was assessed.

[0053] Epo receptors can be expressed in immune cells (e.g., T cells) and are functional. Compared to normal Epo receptor, a mutant Epo receptor exhibited higher and more durable expression, higher signal intensity, and greater stimulation of T cell activity. Expression and function of Epo receptor in T cells are unexpected, as is the superiority of the mutant EpoR. Acute Lymphoblastic Leukemia and CD 19

[0054] Acute lymphoblastic leukemia (ALL) is a cancer of lymphoid blood cells. ALL progresses rapidly and is fatal if untreated. Standard treatment includes chemotherapy and hematopoietic stem cell transplant. CD 19 is a B-cell-specific antigen that is expressed on all leukemic cells in the majority of cases of ALL.

[0055] The vectors described herein can be used to generate modified T cells, which, in turn, can be used for targeted treatment of ALL. The processes described herein can be used to create transgenic T cells that can target CD 19+ B-cells for destruction, thereby decreasing the risk and/or severity of ALL.

[0056] While the particular examples described herein target CD 19+ B-cells as a paradigm, the approach is applicable to the targeting other antigens that are markers of cells in the pathogenesis of cancer and other diseases.

Nucleic Acids

[0057] As used herein, the term“nucleic acid” refers to a polymer comprising multiple nucleotide monomers ( e.g ., ribonucleotide monomers or deoxyribonucleotide monomers). “Nucleic acid” includes, for example, DNA (e.g., genomic DNA and cDNA), RNA, and DNA-RNA hybrid molecules. Nucleic acid molecules can be naturally occurring, recombinant, or synthetic. In addition, nucleic acid molecules can be single-stranded, double-stranded or triple-stranded. In certain embodiments, nucleic acid molecules can be modified. In the case of a double-stranded polymer,“nucleic acid” can refer to either or both strands of the molecule.

[0058] The terms“nucleotide” and“nucleotide monomer” refer to naturally occurring ribonucleotide or deoxyribonucleotide monomers, as well as non-naturally occurring derivatives and analogs thereof. Accordingly, nucleotides can include, for example, nucleotides comprising naturally occurring bases (e.g, adenosine, thymidine, guanosine, cytidine, uridine, inosine, deoxyadenosine, deoxythymidine, deoxyguanosine, or

deoxycytidine) and nucleotides comprising modified bases known in the art.

[0059] As used herein, the term“sequence identity,” refers to the extent to which two nucleotide sequences, or two amino acid sequences, have the same residues at the same positions when the sequences are aligned to achieve a maximal level of identity, expressed as a percentage. For sequence alignment and comparison, typically one sequence is designated as a reference sequence, to which a test sequences are compared. The sequence identity between reference and test sequences is expressed as the percentage of positions across the entire length of the reference sequence where the reference and test sequences share the same nucleotide or amino acid upon alignment of the reference and test sequences to achieve a maximal level of identity. As an example, two sequences are considered to have 70% sequence identity when, upon alignment to achieve a maximal level of identity, the test sequence has the same nucleotide or amino acid residue at 70% of the same positions over the entire length of the reference sequence.

[0060] Alignment of sequences for comparison to achieve maximal levels of identity can be readily performed by a person of ordinary skill in the art using an appropriate alignment method or algorithm. In some instances, the alignment can include introduced gaps to provide for the maximal level of identity. Examples include the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol. 48:443 (1970), the search for similarity method of Pearson & Lipman, Proc. Natl. Acad. Sci. USA 85:2444 (1988), computerized

implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the

Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr.,

Madison, Wis.), and visual inspection (see generally Ausubel et al, Current Protocols in Molecular Biology).

[0061] When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequent coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters. A commonly used tool for determining percent sequence identity is Protein Basic Local Alignment Search Tool (BLASTP) available through National Center for Biotechnology Information, National Library of Medicine, of the LTnited States National Institutes of Health. (Altschul et al., JMol Biol. 215(3):403-10 (1990)).

[0062] In various embodiments, two nucleotide sequences, or two amino acid sequences, can have at least, e.g, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more, sequence identity. When ascertaining percent sequence identity to one or more sequences described herein, the sequences described herein are the reference sequences. Vectors

[0063] The terms“vector”,“vector construct” and“expression vector” mean the vehicle by which a DNA or RNA sequence (e.g. a foreign gene) can be introduced into a host cell, so as to transform the host and promote expression (e.g. transcription and translation) of the introduced sequence. Vectors typically comprise the DNA of a transmissible agent, into which foreign DNA encoding a protein is inserted by restriction enzyme technology. A common type of vector is a“plasmid”, which generally is a self-contained molecule of double-stranded DNA that can readily accept additional (foreign) DNA and which can readily introduced into a suitable host cell. A large number of vectors, including plasmid and fungal vectors, have been described for replication and/or expression in a variety of eukaryotic and prokaryotic hosts.

The terms“express” and“expression” mean allowing or causing the information in a gene or DNA sequence to become manifest, for example producing a protein by activating the cellular functions involved in transcription and translation of a corresponding gene or DNA sequence. A DNA sequence is expressed in or by a cell to form an“expression product” such as a protein. The expression product itself, e.g. the resulting protein, may also be said to be “expressed” by the cell. A polynucleotide or polypeptide is expressed recombinantly, for example, when it is expressed or produced in a foreign host cell under the control of a foreign or native promoter, or in a native host cell under the control of a foreign promoter. Gene delivery vectors generally include a transgene (e.g., nucleic acid encoding an enzyme) operably linked to a promoter and other nucleic acid elements required for expression of the transgene in the host cells into which the vector is introduced. Suitable promoters for gene expression and delivery constructs are known in the art. Recombinant plasmids can also comprise inducible, or regulatable, promoters for expression of an enzyme in cells.

[0064] Various gene delivery vehicles are known in the art and include both viral and non-viral (e.g., naked DNA, plasmid) vectors. Viral vectors suitable for gene delivery are known to those skilled in the art. Such viral vectors include, e.g., vector derived from the herpes virus, baculovirus vector, lentiviral vector, retroviral vector, adenoviral vector, adeno- associated viral vector (AAV), and murine stem cell virus (MSCV). The viral vector can be replicating or non-replicating. Such vectors may be introduced into many appropriate host cells, using methods disclosed or cited herein or otherwise known to those skilled in the relevant art. [0065] Non-viral vectors for gene delivery include naked DNA, plasmids, transposons, and mRNA, among others. Non-limiting examples include pKK plasmids (Clonetech), pUC plasmids, pET plasmids (Novagen, Inc., Madison, Wis.), pRSET or pREP plasmids

(Invitrogen, San Diego, Calif.), pMAL plasmids (New England Biolabs, Beverly, Mass.). Such vectors may be introduced into many appropriate host cells, using methods disclosed or cited herein or otherwise known to those skilled in the relevant art.

[0066] In certain embodiments, the vector comprises an internal ribosome entry site (IRES). In some embodiments, the vector includes a selection marker, such as an ampicillin resistance gene (Amp). In some embodiments, the nucleic acid encodes a fluorescent protein, such as green fluorescent protein (GFP) or mCherry. In some embodiments, the nucleic acid is suitable for subcloning into pMSCV-IRES-GFP between EcoRI and Xhol. In some embodiments, the vector contains a multiple cloning site (MCS) for the insertion of the desired gene.

[0067] Although the genetic code is degenerate in that most amino acids are represented by multiple codons (called“synonyms” or“synonymous” codons), it is understood in the art that codon usage by particular organisms is nonrandom and biased towards particular codon triplets. Accordingly, in some embodiments, the vector includes a nucleotide sequence that has been optimized for expression in a particular type of host cell ( e.g ., through codon optimization). Codon optimization refers to a process in which a polynucleotide encoding a protein of interest is modified to replace particular codons in that polynucleotide with codons that encode the same amino acid(s), but are more commonly used/recognized in the host cell in which the nucleic acid is being expressed. In some aspects, the polynucleotides described herein are codon optimized for expression in T cells.

Erythropoietin (Epo) receptors and mutants thereof

[0068] As used herein, the term“Epo receptor” refers to a protein that binds

erythropoietin, which is a glycoprotein cytokine. Particular Epo receptors and mutants thereof are described in the Exemplification.

[0069] Examples of mutant Epo receptors include truncated Epo receptors, which can be formed by several different types of mutations, including frameshifts, insertions, and deletions. Those lacking the C-terminal negative regulatory domain exhibit hypersensitivity to Epo stimulation in red cells. One example is represented by a nucleic acid encoding an Epo receptor that has nonsense mutations within exon 8 of the Epo receptor gene that encode premature stop codons. Such mutants can produce a truncated form of EpoR with augmented Epo signaling in erythrocyte progenitors. One particular example of an EpoR mutant has a mutation at nucleotide 6002 so that codon 439 encodes a stop codon (TAG) instead of tryptophan (TGG).

Chimeric antigen receptor (CAR) Constructs

[0070] FIG. 5 A illustrates a particular construct that is an anti-CD 19 single-chain variable fragment (anti-CD 19 scFv) coupled to a hinge and transmembrane domain. In the particular example of FIG. 5 A, the hinge and transmembrane domain are a CD8a hinge and

transmembrane domain. The anti-CD 19 scFv includes an anti-CD 19 variable light chain domain, an anti-CD 19 variable heavy chain domain, and a linker domain joining the variable light chain domain and the variable heavy chain domain. The relative positions of the variable light and variable heavy chain domain can be reversed, but they are both N’ terminal to a transmembrane domain, illustrated in FIG. 5A as a CD8a hinge and transmembrane domain. The construct can also include an N-terminal signal peptide, such as a CD8a signal peptide (see SEQ ID NOS: 21 and 22). Signal peptides of surface proteins are generally suitable.

[0071] A variety of linker domains are suitable. In some embodiments, the linker domain can be (G4S)x (SEQ ID NO: 24), wherein x is an integer from 1 to 100; preferably, x is an integer from 1 to 10; even more preferably, x is an integer from 2 to 5. In some

embodiments, the linker domain can be (G4S)3 (SEQ ID NO: 25). In other embodiments, the linker domain can be one or more glycine residues (e.g., (G)y (SEQ ID NO: 26), where y is an integer from 2 to 100. In other embodiments, the linker domain can be (EAAAK) 3 (SEQ ID NO: 27). (G4S) X (SEQ ID NO: 24), (G4S) 3 (SEQ ID NO: 25), and (G) y (SEQ ID NO: 26) are examples of flexible linkers, while (EAAAK) 3 (SEQ ID NO: 27) is an example of a more rigid linker.

[0072] A variety of hinge and transmembrane domains are suitable. In some

embodiments, the hinge domain can be a CD8a hinge domain. In some embodiments, the transmembrane domain can be a CD8a transmembrane domain. In some embodiments, the hinge and transmembrane domain can be a CD8a hinge and transmembrane domain. In some embodiments, the hinge can be a plurality of amino acid residues. In some embodiments, the transmembrane domain can be a transmembrane domain from CD4, CD8P, CD 16, CD28, CD32, CD34, CD64, CD137, FceRIy, 0X40, Oϋ3z, CD3e, CD3y, CD35, TCRa, VEGFR2, FAS, or FGFR2B.

[0073] While the embodiment of FIG. 5 A is an anti-CD 19 construct, a similar approach can be applied to generate constructs for other target antigens, such as CD20, CD22, CD 123, CD33, B-cell maturation antigen (BCMA), mesothelin, human epidermal growth factor receptor 2 (Her2), prostate-specific membrane antigen (PSMA), or disialoganglioside (GD)- 2. For example, based on the schema in FIG. 5A, the anti-CDl9 scFv portion can be replaced with a different scFv that specifically binds to a different target antigen.

[0074] The constructs also encode an Epo receptor. In the construct of FIG. 5 A, the Epo receptor is a mutant (EpoRm) that was generated using site-directed mutagenesis polymerase chain reaction (PCR) to alter the codon for amino acid 439 from TGG (Trp) to TAG (stop). Other mutants of Epo receptor are known in the art, and other Epo receptors variants not existing in nature can be created. One example is an Epo receptor lacking the

immunoreceptor tyrosine-based inhibition motif (ITIM) present in the intracellular portion of the receptor, which may have enhanced signaling properties and are likely to be suitable. Another example is an Epo receptor with multiple JAK2 -binding domains in the intracellular portion.

[0075] In the construct, the Epo receptor is joined with the chimeric antigen receptor (CAR) by a 2A peptide, which is a self-cleaving peptide. By joining the Epo receptor with the CAR, coexpression of both proteins can be achieved from a single vector. Examples of 2A peptides are P2A (SEQ ID NOS: 13 and 14), T2A (SEQ ID NOS: 15 and 16), E2A (SEQ ID NOS: 17 and 18), and F2A (SEQ ID NOS: 19 and 20), though other 2A peptides are known in the art.

Methods of Making Transgenic Host Cells

[0076] Described herein are methods of making a transgenic host cell, such as transgenic T cells. The transgenic host cells can be made, for example, by introducing one or more of the vector embodiments described herein into the host cell.

[0077] In one embodiment, the method comprises introducing into a host cell a vector that includes a nucleic acid that encodes an Epo receptor and a chimeric antigen receptor (CAR), such as an anti-CD19-41BB-CD3z. In some embodiments, a nucleic acid, such as a bicistronic vector, expresses Epo receptor and the CAR. In some embodiments, two separate vectors can be used to create a transgenic cell, such as a transgenic T cell, that expresses Epo receptor and the CAR.

[0078] In some embodiments, one or more of the nucleic acids are integrated into the genome of the host cell. In some embodiments, the nucleic acids to be integrated into a host genome can be introduced into the host cell using any of a variety of suitable methodologies known in the art, including, for example, homologous recombination, CRISPR-based systems (e g., CRISPR/Cas9; CRISPR/Cpfl) and TALEN systems.

[0079] A variety of host cells are suitable, most typically immune cells. In addition to T cells (T lymphocytes), expression of EpoR is expected to activate natural killer (NK) cells, monocytes/macrophages, dendritic cells, and other immune cells. In some instances, the T cell can be a human peripheral blood T lymphocyte. In some instances, the T cell can be a CD4+ T cell. In some instances, the T cell can be a CD8+ T cell.

[0080] In some instances, the T cell can also expresses a T-cell receptor (TCR) that binds a tumor antigen or a viral antigen. In some instances, the TCR can be endogenous. For example, the T cell can be a tumor-infiltrating lymphocyte (TIL) that is extracted from a tumor and expanded ex vivo. In some instances, the TCR can be exogenous. For example, the TCR can be expressed in the T cell by viral transduction or other means. The TCR can be specific for a viral peptide, such as a peptide derived from hepatitis B virus, hepatitis C virus, Epstein-Barr virus, cytomegalovirus, or from a tumor cell, such as melanoma-associated antigen (MAGE), NY-ESO-l, telomerase reverse transcriptase (TERT).

Values and Ranges

[0081] ETnless otherwise indicated or otherwise evident from the context and

understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or subrange within the stated ranges in various embodiments, unless the context clearly dictates otherwise.“About” in reference to a numerical value generally refers to a range of values that fall within ±8%, in some embodiments ±6%, in some embodiments ±4%, in some embodiments ±2%, in some embodiments ±1%, in some embodiments ±0.5% of the value unless otherwise stated or otherwise evident from the context. EXEMPLIFICATION

Materials and Methods

Cells

[0082] The leukemia cell lines Jurkat, Nalm6, and RS4; 11 were obtained from the American Type Culture Collection (ATCC; Rockville, MD). The CD 19+ B-lineage ALL cell line OP-l was developed in our laboratory. 19 A murine stem cell virus (MSCV) retroviral vector, containing green fluorescent protein (GFP) or mCherry and an internal ribosomal entry site (IRES) was used to express the firefly luciferase gene in Nalm6, and mCherry in OP-l, respectively. Cell lines were maintained in RPMI-1640 (Thermo Fisher Scientific, Waltham, MA) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin- streptomycin. Human embryonic kidney fibroblast 293T (HEK 293T) cells were cultured in DMEM (HyClone, GE Life Sciences, Logan, Utah) supplemented with 10% FBS and 1% penicillin-streptomycin.

[0083] Peripheral blood samples were obtained from discarded anonymized by-products of platelet donations from healthy adult donors at the National University Hospital Blood Bank or the Health Science Authority Blood Bank, Singapore. Mononucleated cells were separated by centrifugation on a Lymphoprep density step (Nycomed, Oslo, Norway) and washed twice in RPMI-1640. T cells were enriched with Dynabeads Human T-Activator CD3/CD28 (Invitrogen, Carlsbad, CA) and cultured in RPMI-1640, 10% FBS, 1% penicillin- streptomycin, and interleukin-2 (IL-2; 120 IU/mL; Proleukin, Novartis, Basel, Switzerland).

Gene cloning and retroviral transduction

[0084] The Epo receptor (EpoR) cDNA was obtained from GeneCopoeia (Rockville, MD). The mutant EpoR (EpoRm) was generated using site-directed mutagenesis polymerase chain reaction (PCR) to alter the codon for amino acid 439 from TGG (Trp) to TAG (stop). 20 In some experiments, a Flag tag (DYKDDDDK (SEQ ID NO: 23)) was added to C-terminal of EpoR and EpoRm. The anti-CD19-41BB-CD3z CAR was previously made in our laboratory. 21 The EpoRm-2A-CAR was generated by fusion PCR, combining EpoRm and anti-CD19-41BB-CD3z through 2A peptide sequence. 22 The constructs and expression cassette were subcloned into EcoRI and Xhol sites of the pMSCV-IRES-GFP vector.

[0085] Preparation of retroviral supernatant and transduction were performed as previously described. 23 Briefly, pMSCY retroviral vector-conditioned medium was added to RetroNectin (Takara, Otsu, Japan)-coated polypropylene tubes; after centrifugation and removal of the supernatant, T cells (5 x 10 5 ) were added to the tubes and left at 37°C for 12 hours; fresh viral supernatant was added on two other successive days. T lymphocytes were then maintained in RPMI-1640 with FBS, antibiotics and 200 IU/mL IL-2 until the time of the experiments, 7-21 days after transduction.

Detection of EpoR and CAR expression

[0086] Surface expression of EpoR was detected with phycoerythrin (PE)-conjugated anti-human EpoR antibody (38409; R&D Systems, Minneapolis, MN). In some experiments, surface staining of EpoR was done on cells that had been cultured in cytokine-free media for 2 hours followed by incubation with 10 IU/mL of recombinant human Epo (Thermo Fisher Scientific) at 37°C for 15-60 minutes. Expression of CAR was detected using a biotin- conjugated goat anti-mouse F(ab’)2 antibody (Jackson ImmunoResearch, West Grove, PA) followed by streptavidin conjugated to allophycocyanin (APC; Jackson ImmunoResearch). PE/Cy7-conjugated anti-CD4 (SK3) antibody was from BD Biosciences (San Jose, CA); APC-conjugated anti-CD8 (BW135/80) antibody was from Miltenyl Biotec (Bergisch Gladbach, Germany). In all tests, non-reactive isotype-matched antibodies were used as controls. Cell staining was analysed using Accuri C6 or Fortessa flow cytometers (BD Bioscience), with Diva (BD Biosciences) or FlowJo software (FlowJo, Ashland, OR).

[0087] Western blot analysis of EpoR expression in 293T cells was performed as previously described. 24 Briefly, cell lysates were extracted using CelLytic M cell lysis reagent (Sigma-Aldrich, Saint Louis, MO) prior to protein quantification with Pierce BCA protein assay kit (ThermoFisher Scientific). Cell lysates were diluted with 4x Laemmli sample buffer (Bio-rad, Hercules, CA) prior to separation on 10% polyacrylamide gel by electrophoresis under reducing condition. Blotted membrane was probed with mouse anti- Flag (9 A3; Cell Signaling Technology, Danvers, MA) followed by goat anti-mouse IgG conjugated to horseradish peroxidase (HRP) (R&D Systems); rabbit anti-human

glyceraldehyde 3-phosphate dehydrogenase (GAPDH) (EPR16891; Abeam, Cambridge, UK) followed by HRP-conjugated goat anti-rabbit IgG antibody (Abeam) was used as a loading control. Antibody binding was revealed by Clarity Western ECL substrate (Bio-Rad) and visualized by ChemiDoc Touch Imager (Bio-Rad). Detection of Epo binding and signaling

[0088] To determine binding of Epo to EpoR, cells were incubated with biotinylated Epo (R&D Systems) for 2 hours at room temperature. Biotinylated Epo was visualized with streptavidin-PE (Jackson ImmunoRe search).

[0089] To detect Epo signaling, cells were incubated in cytokine-free media for 2 hours before stimulated with Epo (0.01 - 10 IU/mL) at 37°C for 15 minutes to 24 hours. In some experiments, cells were treated with 0.1 - 10 mM of ruxolitinib (Selleckchem) or 0.5 - 5 nM tofacitinib (Santa Cruz Biotechnology, Santa Cruz, CA) for 1 hour prior to Epo stimulation. Cells were then fixed with lx lyse/fix buffer (BD Biosciences), permeabilized with Perm Buffer III (BD Biosciences), and stained with anti-STAT5 (pY694) conjugated to Alexa Fluor 647 (AF647) (47; BD Biosciences). In some experiments, the effects of human Epo were compared to those of murine Epo (Biolegend, San Diego, CA).

Cell proliferation assay

[0090] To determine the effect of Epo on cell cycle, cells were cultured in cytokine-free media for 3 days followed by stimulation with 10 IU/mL of Epo for 1 day. DNA synthesis was measured by Click-iT EdU AF647 Flow Cytometry Assay Kit (Thermo Fisher

Scientific), and DNA content was measured with FxCycle Violet Stain (Thermo Fisher Scientific).

[0091] To assess cell survival, T cells were cultured in absence of exogenous cytokines, with or without Epo (4 - 10 IU/mL) in a flat bottom 96-well or 24-well plate (Cellstar). For cell proliferation, T cells were co-cultured with target cells (OP-l cells) at 1 : 1 effector-to- target (E:T) ratio in a flat bottom 96-well plate; Epo (10 IU/mL) was added every two days. Target cells, irradiated (100 Gy) or treated with Streck cell preservative (Streck Laboratories, Omaha, NE) to inhibit growth, were added at the beginning of the cultures, and every 7 days thereafter. In some experiments, low dose (10 IU/mL) or high dose (100 IU/mL) IL-2 was added to the culture as well. The number of GFP+ T cells was measured by flow cytometry.

Cytotoxicity and cytokine production

[0092] To test cytotoxicity, CD19+ target cells (OP-l, RS4; 11, and Nalm6) were labelled with calcein red-orange AM (Thermo Fisher Scientific) and placed into a 96-well round bottom plate (Coming Costar, Coming, NY). T cells were added at E:T ratio of 1 : 1 and co cultured with target cells for 4 hours at 37°C and 5% C02 incubator. Viable target cells were counted by flow cytometry. 25 For long-term cytotoxicity, OP-l mCherry cells were placed into a 96-well flat bottom plate, T cells were added at different E:T ratios and cultured for 3 days with 10 IU/mL of Epo. Plates were placed in IncuCyte Zoom System (Essen

BioScience) set to collect data collection (whole-well imaging) every 4 hours.

[0093] To measure exocytosis of lytic granules, T cells were co-cultured with OP-l cells at 1 : 1 E:T ratio for 4 hours in a 96-well round bottom plate. PE-conjugated anti-human CD 107a antibody (H4A3; BD Biosciences) was added at the beginning of the cultures and monensin (BD Golgi Stop) 1 hour later.

[0094] To measure interferon-g (IFN-g) and tumor necrosis factor-a (TNF-a) production, target and effector cells at a 1 : 1 E:T ratio were plated as above. After 1 hour, brefeldin A (BD GolgiPlug) was added to the culture and incubated for another 5 hours. Subsequently, intracellular staining with PE-conjugated anti-IFN-g (clone 25723.11; BD Biosciences) or anti-TNF-a (clone 6401.1111; BD Biosciences) was done prior to analysis by flow cytometry. To assess cytokine profile, target and effector cells were co-cultured at 1 :4 E:T ratio in absence or presence of 10 IU/mL Epo for 24 hours. Culture supernatant was collected to be analysed by Luminex Multiplex Assay (Bio-Rad).

Xenograft experiments

[0095] To determine survival of T cells in vivo, NOD.Cg-Prkdcscid åL2rgtmlWjl/SzJ (NOD/scid IL2RGnull) mice (The Jackson Laboratory, Bar Harbor, ME) were injected intravenously (i.v.) with 1 x 10 7 T cells transduced with GFP alone, CAR, or EpoRm-CAR.

In some mice, 100 IU of Epo were injected intraperitoneally (i.p) every two days for two weeks. On day 13, blood cells were counted with a cell counter (Beckman Coulter, Miami, FL). After treatment with red blood cell lysis solution (Sigma-Aldrich), cells were stained with APC-conjugated anti-human CD45 (2D1; Biolegend) and PE-conjugated anti-mouse CD45 (30-F11; BD Pharmingen).

[0096] To assess anti -leukemic activity, NOD-scid-IL2RGnull mice were injected with lxlO 7 T cells i.v., followed 2 weeks later by 2.5xl0 5 Nalm6 cells expressing luciferase i.v.. ALL cell engraftment was determined by measuring luminescence signal with the Xenogen IVIS-200 System (Perkin Elmer, Waltham, MA), after i.p. injection of aqueous D-luciferin potassium salt (150 pg/g body weight; Perkin Elmer); signals were analyzed with Living Image 3.0 software. In another model, 5xl0 5 Nalm6-luciferase cells were injected i.v.

followed 4 days later by 1- 2xl0 7 T cells i.v.. Mice were euthanized when the luminescence reached 1 x 10 10 photons per second, or earlier if there were physical signs warranting euthanasia.

[0097] In another model to test anti-leukemic activity of T cells, Nalm6 cells transduced with luciferase were injected i.v. (5 x 10 5 cells per mouse), followed 2 days later by T cells expressing EpoRm-CAR (2 x 10 7 cells per mouse, i.v.), while control mice received no T cells; some mice received Epo (100 IU) 3 times per week i.p.. Tumor cell load was determined with the Xenogen IVIS-200 System (Perkin Elmer, Waltham, MA) after injecting aqueous D-luciferin potassium salt (150 pg/g body weight; Perkin Elmer) i.p.. Luminescence was analyzed with Living Image 3.0 software. Mice were euthanized when the luminescence reached 1 x 10 10 photons per second, or earlier if there were physical signs warranting euthanasia.

Jurkat cell growth curves

[0098] To determine the effect of EpoR and EpoRm expression on cell growth, numbers of Jurkat cells transduced with GFP only, EpoR and EpoRm and maintained under the same culture conditions were counted periodically.

RESULTS

EpoR can be expressed in T cells and mediates Epo survival signals

[0099] Retroviral transduction of Jurkat cells with the EpoR gene resulted in high expression of the receptor (FIG. 1 A). Cells expressing EpoR could bind Epo, while there was no detectable binding in cells transduced with GFP alone (FIG. 1B). Next, we determined whether EpoR could also be expressed in peripheral blood T lymphocytes. For this purpose, we stimulated peripheral blood mononucleated cells with anti-CD3 and CD28 antibodies and IL-2, and transduced them with the EpoR gene or GFP alone. In 5 experiments with T cells from 4 donors, the percentage of transduced T lymphocytes (GFP+) expressing EpoR was 74.9% ± 4.8%; EpoR was undetectable in cells transduced with GFP only (FIG. 1C).

[00100] To test whether EpoR was functional, we exposed EpoR-transduced T

lymphocytes to Epo (10 IU/mL) for 15 minutes, and measured phosphorylation of STAT5 Y647, one of the downstream activation signals triggered by EpoR ligation in erythroid cells. 26,27 In experiments with T cells from 3 donors, percentage of pSTAT5-positive cells among GFP+ EpoR-transduced T lymphocytes increased from 1.0% ± 0.9% to 85.9% ±

5.1% after Epo exposure; it remained essentially unchanged in T lymphocytes transduced with GFP only (1.3% ± 0.4% to 1.5% ± 0.7%) (Fig. 1D). After EpoR ligation in erythroid cells, JAK2 kinase phosphorylates STAT5. 28 After exposing EpoR T lymphocytes to the JAK1/2 inhibitor ruxolitinib, 29 STAT5 phosphorylation induced by Epo was abrogated (FIG. 1D). Finally, addition of Epo (4 IU/mL) to EpoR-transduced T cells supported their survival in culture, in the absence of exogenous IL-2 (FIG. 1E). Together, these results show that EpoR can be expressed in T lymphocytes, and that exposure to Epo can transduce survival signals in EpoR-expressing T lymphocytes.

An EpoR mutant enhances EpoR expression in T cells

[00101] It has been previously reported that nonsense mutations in the EpoR exon 8 produced a truncated form of EpoR with augmented Epo signaling in erythrocyte progenitors, resulting in an increased erythrocyte output. 20,30 We generated a cDNA encoding an EpoR mutant in which a mutation of the codon 439 (TGG) encoding for tryptophan was inserted to change it to the stop codon TAG (“EpoRm”). After expressing the cDNA in 293T cells, we found that the encoded protein had the predicted size of 54 kDa by western blotting, as compared to 62 kDa for EpoR (FIG. 2A). We expressed EpoR and EpoRm in activated peripheral blood T lymphocytes, and observed that levels of expression of EpoRm were consistently higher than those measured for the wild-type EpoR construct in the same T cells (FIG. 2B). Thus, in 18 experiments with T lymphocytes from 7 donors, the percentage of GFP+ T cells expressing EpoR was 71.9% ± 14.1% as compared to 93.1% ± 6.1% for EpoRm (P <0.0001 by paired t test); mean fluorescence intensity (MFI) was 3,620 ± 2,449 and 8,773 ± 5,851, respectively (P <0.0001) (Fig. 2C). EpoRm expression was higher regardless of whether cells were CD4+ or CD8+ (Fig. 2D).

[00102] Overall GFP expression in T cells transduced with either EpoR or EpoRm was not significantly different: percentage of GFP+ T cells was 75.2% ± 8.4% with EpoR and 80.4% ± 7.5% with EpoRm; MFI was 7,003 ± 2,820 and 8,331 ± 3,343, respectively (P >0.05 for either comparison). Therefore, it is unlikely that differences in expression between the 2 receptors was simply due to different transduction efficiency. Nevertheless, we addressed this possibility and performed a detailed analysis of the levels of receptor expression in relation to a given level of GFP expression in 3 experiments. By these measurements, EpoRm expression was higher than that of EpoR (FIG. 2E; P <0.0001 for all 3 comparisons).

[00103] Higher expression of EpoRm was associated with a longer persistence after exposure to Epo. In experiments with T cells from 3 donors, reduction in percentage of EpoR-positive cells was clearly higher in cells transduced with EpoR than in those transduced with EpoRm (FIG. 2F).

Signaling properties of EpoRm

[00104] In line with the higher and more sustained expression of EpoRm, exposure of T cells to Epo resulted in more vigorous activation if these cells expressed EpoRm. EpoRm T cells had higher pSTAT5 Y647 phosphorylation than EpoR T cells, which was suppressed by exposure to the JAK1/2 inhibitor ruxolitinib (10 mM) (FIG. 3A, B, C). The higher STAT5 phosphorylation caused by EpoRm was corroborated by an analysis of pSTAT5 Y647 MFI according to levels of GFP. At any given level of retroviral transduction measured by GFP, pSTAT5 MFI was higher with EpoRm, which approached a plateau in pSTAT5 signaling at lower levels of transduction (FIG. 3B). STAT5 phosphorylation triggered by Epo was dose- dependent, and EpoRm required lower Epo levels to induce STAT5 phosphorylation. Thus, in 2 experiments, the average pSTAT5 Y647 with a dose of 0.1 IU/mL was 8.9% for EpoR versus 29.1% with EpoRm; it was 66.1% versus 88.6% with 1 IU/mL of Epo (Fig. 3D). STAT5 phosphorylation was also more durable in cells expressing EpoRm than in those expressing EpoR (FIG. 3E).

[00105] Exposure to Epo for 24 hours elicited DNA synthesis in T cells bearing Epo receptors, and stimulation was higher in those expressing EpoRm (FIG. 4A). In the absence of IL-2, T cells rapidly died regardless of Epo receptor expression. However, when Epo was added to the culture, those expressing Epo receptors persisted for at least 2 weeks, with cultures of T cells expressing EpoRm yielding higher cell numbers (FIG. 4B).

[00106] We determined whether Epo stimulation would further improve T cell recovery if cells were cultured in the presence of 100 IU/mL of IL-2. In experiments with cells from 4 donors, recovery of T cells transduced with EpoRm was significantly higher after 7 days of culture when Epo (10 IU/mL) was added to IL-2 (100 IU/mL) (P = 0.020); under these conditions, cell recovery was better than with cell transduced with EpoR (P = 0.019) (FIG. 4C). In separate cultures prolonged for 14 days, T cells expressing Epo receptors show higher and more durable expansion than those transduced with GFP only; again, EpoRm cells performed better than EpoR cells (FIG. 4D). Functional activity of simultaneously expressed EpoRm and CAR

[00107] The previous experiments indicated that EpoRm had higher expression, and produced stronger and more durable signals than the wild-type Epo receptor in T cells.

Therefore, we incorporated the gene encoding EpoRm in a bicistronic vector also containing the gene encoding an anti-CD 19-41BB-Oϋ3z CAR developed in our laboratory (FIG. 5 A). 21 Transduction of this construct in T lymphocytes resulted in expression of both EpoRm and CAR (FIG. 5B), and either gene was expressed at levels which were similar to those of cells transduced with a single-gene vector (FIG. 5C). The functionality of EpoRm was maintained regardless of whether it was expressed alone or in combination with the CAR (FIG. 5D).

[00108] CAR-T cell function was also retained in cells expressing EpoRm. Thus, there were no differences in exocytosis of cytotoxic granules, as measured by CD 107a expression after 4 hours of co-culture with the CD19+ ALL cell line OP-l cells, regardless of whether CAR-T cells expressed EpoRm or whether there was Epo in the cultures (FIG. 8A).

Secretion of IFNy triggered by CAR stimulation under the same culture conditions was also not affected by Epo signaling, but we observed that EpoRm-CAR-T cells secreted more TNFoc in the presence of Epo than CAR-T cells lacking EpoRm (P <0.001) (FIG. 8B). The cytokine profile of EpoRm-CAR T cells co-cultured with OP-l cells with and without Epo, was generally similar, although we noted higher levels of IL-6 and IL-4 and lower levels of IL-2 in cells cultured with Epo (FIG. 8C).

[00109] We determined the capacity of EpoRm-CAR T cells to kill CD 19+ target cells. In experiments with 3 ALL cell lines ( OP-l, RS4;l 1 and Nalm6), 4-hours cytotoxicity of these T cells at 1 : 1 E:T was indistinguishable from that of T cells transduced with CAR alone, regardless of whether Epo (10 IU/mL) was added to the cultures (FIG. 5E). However, when cytotoxicity was tested over 72-hours in the presence of Epo there was a significantly higher killing by EpoRm-CAR T cells over that of CAR-T cells at low (1 :4, 1 :8) E:T ratios (FIG.

5F).

Proliferative signals of Epo and IL-2 in EpoRm-CAR T cells

[00110] The higher killing exerted by EpoRm-CAR T cells in long-term cultures might be explained by a higher rate of proliferation of these cells, thereby creating a higher E:T ratio. To test this notion, we co-cultured T cells, transduced with EpoRm-CAR, CAR alone or GFP, with Streck-treated or irradiated OP-l cells, and monitored T cell growth over 2 weeks. We added Epo (10 IU/mL) to the cultures but no exogenous IL-2. As shown in FIG. 6A, the expansion of T cells from 3 donors was higher when CAR-T cells expressed EpoRm, indicating that the CAR-driven cell proliferation is enhanced by EpoRm signaling. EpoRm- CAR T cell proliferation was abrogated by ruxolotinib (1 mM) (FIG. 6B). To determine whether Epo stimulation could add to the stimulation provided by exogenous IL-2, we assessed T cell recovery in co-culture with irradiated OP-l after 7 days, in the presence of either 10 IU/mL (4 donors) or 100 IU/mL of IL-2 (3 donors). CAR-stimulated EpoRm-CAR T cells cultured with both IL-2 and Epo had considerably higher expansion than those cultured with IL-2 alone (FIG. 6C).

[00111] To further determine the relation between IL-2- and Epo-triggered signaling, we measured STAT5 Y647 phosphorylation. As shown in FIG. 6D, cells exposed to Epo and IL- 2 had higher pSTAT5 MFI than those exposed to either growth factor. In side-to-side comparisons, the pSTAT5 levels induced by 1 IU/mL of Epo were significantly higher than those induced by 50 IU/mL IL-2 (P = 0.0019) and surpassed those induced by 100 IU/mL of IL-2) (FIG. 6E). Finally, we determined the relative sensitivity to JAK inhibitors of EpoRm- CAR T cells exposed to either Epo (10 IU/mL) or IL-2 (100 IU/mL). We used tofacitinib, which predominantly inhibits JAK3, and ruxolitinib, which predominantly inhibits JAK1 and JAK2 signaling. 29 31 At concentration that completely abrogated STAT5 Y647

phosphorylation caused by IL-2, the inhibitors had no effect of Epo signaling (FIG. 6F). Hence, EpoRm signaling in CAR-T cells triggers a signaling cascade that is distinct from that of IL-2, and enhances the proliferative stimulus of IL-2.

Xenograft models

[00112] To begin to assess the anti-leukemic activity of EpoRm-CAR- T cells in vivo, we infused 5 NSG mice with luciferase-labelled CD19+ Nalm6 ALL cells i.v. After confirming engraftment 2 days later, 4 mice received EpoRm-CAR-T cells i.v. and 1 was left untreated. As shown in FIG. 7A and B, ALL cells rapidly expanded in the untreated mice, while the treated mice were in remission 2 months after leukemic cell infusion. Of note, injection of 100 IU Epo i.p. in 2 of these mice 3 times a week for 2 weeks did not affect the anti-leukemic activity of the EpoR-CAR-T cells (FIG. 7A).

[00113] In a second set of experiments, T lymphocytes transduced with CAR, or EpoRm- CAR of GFP alone were injected i.v. into NSG mice. On day 13 after injection, peripheral blood was examined for the presence of cells expressing GFP and human CD45. As shown in FIG. 7C and D, levels of GFP+/hCD45+ cells in mice injected with EpoRm-CAR-T cells were higher than those measured in mice injected with cells transduced with GFP only, or with CAR lacking EpoRm; injection of 100 IU Epo i.p. 3 times a week for 2 weeks in the latter group did not significantly increase cell numbers. Levels of GFP+/hCD45+ cells were highest in mice injected with EpoRm-CAR-T cells who received Epo i.p. injections.

Notably, however, a significant increase in GFP+/hCD45+ cells was evident in those who had not received Epo injections, although levels were lower than with Epo supplementation. This result suggested that endogenous murine Epo might stimulate cells expressing human EpoRm. Indeed, there is considerable homology between murine and human Epo genes. 32 To determine whether murine Epo could induce signals through human EpoRm in T cells, we examined pSTAT5 Y647 following murine and human Epo stimulation. As shown in FIG.

7E, either produced similar increases in pSTAT5, providing an explanation for the relative increase in GFP+/hCD45+ cells observed when EpoRm-CAR-T cells were infused without human Epo supplementation.

[00114] To assess whether increased numbers of EpoRm-CAR-T cells in vivo would provide superior protection against ALL engraftment, we injected T cells i.v. in NOD-scid- IL2RGnull mice followed 14 days later by i.v. injection of Nalm6 cells. Engraftment of ALL cells was only delayed by CAR-T cells but completely abrogated by EpoRm-CAR-T cells (FIGs. 9A and 9B). Two months after ALL cell infusion, there was ALL engraftment in all 5 mice injected with CAR-T cells, while none of the 5 mice injected with EpoRm-CAR-T cells had ALL (P=0.0079 by Fisher’s exact test). On day 13, 1 day prior to ALL cell inoculation and with no exogenous Epo, percentage of CAR-T cells among peripheral blood lymphoid cells was l8.8%±6.7% versus 48.9%±l7.0% for EpoRm-CAR-T cells (P=0.0062) (FIG. 9C). Levels of EpoRm-CAR-T cells remained higher than those of CAR-T cells at subsequent time points.

[00115] In another model, Nalm6 cells were first engrafted in NOD-scid-IL2RGnull mice by i.v. injection. On day 4, mice were distributed in 4 groups with similar tumor load; three groups received either CAR-T cells, EpoRm-CAR-T cells, or T cells transduced with GFP only by i.v. injection; a 4 th group received only tissue culture medium. ALL cells rapidly expanded in the untreated mice, and in mice that received control T cells without CAR. Both CAR- and EpoRm-CAR-T cells markedly reduced leukemic signals (FIGs. 9D and 9E). On day 55 post-infusion, however, 5 of the 10 mice treated with CAR-T cells had relapsed versus none of the 9 who had received EpoRm-CAR-T cells (P=0.0325 by Fisher’s exact test). FIG. 9F shows the combined long-term survival of the 2 cohorts; EpoRm-CAR-T cells produced a significantly higher overall anti-leukemic effect than CAR-T cells (P=0.0076 by log rank test).

Jurkat cell growth curves

[00116] Expression of either EpoR or EpoRm does not adversely affect long-term cell growth of Jurkat cells (FIG. 10).

EMBODIMENTS

[00117] Embodiment 1. A vector comprising a nucleic acid encoding: a) an erythropoietin (Epo) receptor; b) a self-cleaving peptide or internal ribosome entry site; and c) a cell surface receptor.

[00118] Embodiment 2. The vector of Embodiment 1, wherein the Epo receptor has at least 90% sequence identity to SEQ ID NO: 2.

[00119] Embodiment 3. The vector of Embodiment 1, wherein the Epo receptor is a mutant Epo receptor.

[00120] Embodiment 4. The vector of Embodiment 3, wherein the nucleic acid has a mutation that encodes a stop codon within exon 8 of the Epo receptor.

[00121] Embodiment 5. The vector of Embodiment 3, wherein the Epo receptor has at least 90% sequence identity to SEQ ID NO: 6.

[00122] Embodiment 6. The vector of any of Embodiments 1 through 5, wherein the nucleic acid further encodes a Flag tag (DYKDDDDK (SEQ ID NO: 23)) that is C terminal to the Epo receptor.

[00123] Embodiment 7. The vector of any of Embodiments 1 through 6, wherein the nucleic acid comprises a self-cleaving peptide.

[00124] Embodiment 8. The vector of Embodiment 7, wherein the self-cleaving peptide is a 2A peptide.

[00125] Embodiment 9. The vector of Embodiment 8, wherein the 2A peptide is a T2A peptide.

[00126] Embodiment 10. The vector of any one of Embodiments 1 through 9, wherein the signal peptide is a CD8a signal peptide.

[00127] Embodiment 11. The vector of Embodiment 10, wherein the cell surface receptor comprises an extracellular receptor domain that binds a target cell antigen. [00128] Embodiment 12. The vector of any one of Embodiments 1 through 10, wherein the cell surface receptor is a chimeric antigen receptor comprising: i) a signal peptide; ii) an extracellular receptor domain that binds a target cell antigen; iii) a hinge and transmembrane domain that anchors the extracellular receptor domain on the surface of a cell; and iv) an effector domain.

[00129] Embodiment 13. The vector of Embodiment 11 or 12, wherein the extracellular receptor domain comprises a variable immunoglobulin light chain domain and a variable immunoglobulin heavy chain domain joined by a linker domain.

[00130] Embodiment 14. The vector of Embodiment 13, wherein the linker domain is (G4S)x (SEQ ID NO: 24), wherein x is an integer from 1 to 100.

[00131] Embodiment 15. The vector of Embodiment 13, wherein the linker domain is (G4S)3 (SEQ ID NO: 25).

[00132] Embodiment 16. The vector of Embodiment 12, wherein the extracellular receptor domain is a single-chain variable fragment (scFv).

[00133] Embodiment 17. The vector of any one of Embodiments 1 through 10, wherein the cell surface receptor is a T cell receptor.

[00134] Embodiment 18. The vector of Embodiment 11, wherein the extracellular receptor domain comprises a monoclonal antibody, a recombinant antibody, a human antibody, a humanized antibody, a Fab, a Fab', a F(ab')2, an Fv, a single-chain variable fragment (scFv), a minibody, a diabody, a single-domain antibody, or a functional derivative or variant or fragment thereof.

[00135] Embodiment 19. The vector of Embodiment 11, wherein the extracellular receptor domain comprises an immunoglobulin Fc receptor.

[00136] Embodiment 20. The vector of Embodiment 19, wherein the immunoglobulin Fc receptor is CD 16, CD32 or CD64.

[00137] Embodiment 21. The vector of Embodiment 11, wherein the extracellular receptor domain comprises a cytokine.

[00138] Embodiment 22. The vector of Embodiment 21, wherein the cytokine is IL-13, IL- 4, IL-7, or IL-3.

[00139] Embodiment 23. The vector of Embodiment 11, wherein the cell surface receptor activates immune cells. [00140] Embodiment 24. The vector of Embodiment 23, wherein the cell surface receptor comprises NKG2D, NKG2C, NCR1, NCR2, NCR3, CD137, CD28, or ICOS, or a fragment or a ligand thereof.

[00141] Embodiment 25. The vector of Embodiment 11, wherein the cell surface receptor inhibits immune cells.

[00142] Embodiment 26. The vector of Embodiment 25, wherein the cell surface receptor comprises NKG2A, PD-l, or CTLA-4, or a fragment or ligand thereof.

[00143] Embodiment 27. The vector of Embodiment 11, wherein the cell surface receptor is a receptor for a cytokine.

[00144] Embodiment 28. The vector of Embodiment 27, wherein the cell surface receptor is a receptor for IL-6, IL-l, or TNF alpha.

[00145] Embodiment 29. The vector of any one of Embodiments 1 through 28, wherein the target cell antigen is a tumor associated antigen or a tumor specific antigen.

[00146] Embodiment 30. The vector of any one of Embodiments 1 through 28, wherein the target cell antigen is a viral, bacterial, fungal, or parasite associated antigen or a viral, bacterial, fungal, or parasite specific antigen.

[00147] Embodiment 31. The vector of any one of Embodiments 1 through 28, wherein the target cell antigen is CD19, CD20, CD22, CD123, CD33, B-cell maturation antigen (BCMA), mesothelin, human epidermal growth factor receptor 2 (Her2), prostate-specific membrane antigen (PSMA), or disialoganglioside (GD2).

[00148] Embodiment 32. The vector of any one of Embodiments 1 through 28, wherein the target cell antigen is CD 19.

[00149] Embodiment 33. The vector of any one of Embodiments 1 through 32, wherein the extracellular domain is an anti-CD 19 single-chain variable fragment (scFv).

[00150] Embodiment 34. The vector of any one of Embodiments 1 through 33, wherein the hinge and transmembrane domain is a CD8a hinge and transmembrane domain.

[00151] Embodiment 35. The vector of any one of Embodiments 1 through 34, wherein the hinge comprises a plurality of amino acid residues.

[00152] Embodiment 36. The vector of any one of Embodiments 1 through 35, wherein the transmembrane domain is a transmembrane domain from CD4, CD8P, CD 16, CD28, CD32, CD34, CD64, CD137, FceRIy, 0X40, Oϋ3z, CD3e, CD3y, CD35, TCRa, VEGFR2, FAS, or FGFR2B. [00153] Embodiment 37. The vector of any one of Embodiments 1 through 36, wherein the effector domain comprises 4-1BB and O03z.

[00154] Embodiment 38. The vector of any one of Embodiments 1 through 37, wherein the CAR is anti-CD 19-41 BB -CD3 z.

[00155] Embodiment 39. A vector comprising a nucleic acid encoding a mutant erythropoietin (Epo) receptor.

[00156] Embodiment 40. The vector of Embodiment 39, wherein the nucleic acid has a mutation that encodes a stop codon within exon 8 of the Epo receptor.

[00157] Embodiment 41. The vector of Embodiment 39, wherein the mutant Epo receptor has at least 90% sequence identity to SEQ ID NO: 6.

[00158] Embodiment 42. The vector of any one of Embodiments 1 through 41, wherein the vector is a retrovirus.

[00159] Embodiment 43. The vector of any one of Embodiments 1 through 42, wherein the vector is a murine stem cell virus (MSCV) retroviral vector.

[00160] Embodiment 44. The vector of any one of Embodiments 1 through 43, wherein the vector further encodes a fluorescent protein.

[00161] Embodiment 45. The vector of any one of Embodiments 1 through 44, wherein the vector encodes an internal ribosomal entry site (IRES).

[00162] Embodiment 46. The vector of any one of Embodiments 1 through 45, wherein the vector further encodes at least one regulatory element for expression of the nucleic acid.

[00163] Embodiment 47. A method of making a transgenic mammalian host cell, the method comprising introducing into a mammalian host cell the vector of any of Embodiments 1 through 46.

[00164] Embodiment 48. The method of Embodiment 47, wherein the mammalian host cell is an immune cell.

[00165] Embodiment 49. The method of Embodiment 48, wherein the immune cell is a natural killer (NK) cell, a monocyte/macrophage cell, or a dendritic cell.

[00166] Embodiment 50. The method of Embodiment 48, wherein the immune cell is a T cell.

[00167] Embodiment 51. The method of Embodiment 50, wherein the T cell is a human peripheral blood T lymphocyte.

[00168] Embodiment 52. The method of Embodiment 50, wherein the T cell is a CD4+ T cell. [00169] Embodiment 53. The method of Embodiment 50, wherein the T cell is a CD8+ T cell.

[00170] Embodiment 54. The method of Embodiment 50, wherein the T cell further expresses a T-cell receptor (TCR) that binds a tumor antigen or a viral antigen.

[00171] Embodiment 55. The method of Embodiment 54, wherein the TCR is endogenous.

[00172] Embodiment 56. The method of Embodiment 55, wherein T cell is a tumor- infiltrating lymphocyte (TIL), and wherein the method further comprises extracting the tumor-infiltrating lymphocyte from a tumor and expanding the TIL ex vivo.

[00173] Embodiment 57. The method of Embodiment 54, wherein the TCR is exogenous.

[00174] Embodiment 58. The method of Embodiment 57, wherein the method further comprises introducing into the T cell a vector that expresses the exogenous TCR.

[00175] Embodiment 59. A mammalian immune cell comprising the vector of any one Embodiments 1 through 46.

[00176] Embodiment 60. The mammalian immune cell of Embodiment 59, wherein the mammalian immune cell is a natural killer (NK) cell, a monocyte/macrophage cell, or a dendritic cell.

[00177] Embodiment 61. The mammalian immune cell of Embodiment 59, wherein the mammalian immune cell is a T cell.

[00178] Embodiment 62. The mammalian immune cell of Embodiment 61, wherein the T cell is a human T cell.

[00179] Embodiment 63. The mammalian immune cell of Embodiment 61, wherein the T cell is a human peripheral blood T lymphocyte.

[00180] Embodiment 64. A method of reducing the number of CD 19+ cells in a mammal, the method comprising introducing mammalian T cells into the subject, wherein the mammalian T cells comprise the vector of any of Embodiments 1 through 46.

[00181] Embodiment 65. The method of Embodiment 64, wherein the mammal is a human.

[00182] Embodiment 66. The method of any of Embodiments 64 through 65, wherein the mammalian T cells are autologous cells isolated from the mammal.

[00183] Embodiment 67. The method of any of Embodiments 64 through 65, wherein the mammalian T cells are allogenic cells isolated from a donor.

[00184] Embodiment 68. The method of any of Embodiments 64 through 67, further comprising administering Epo to the subject. [00185] Embodiment 69. The method of any of Embodiments 64 through 68, further comprising administering IL-2 to the subject.

[00186] Embodiment 70. The method of any of Embodiments 64 through 69, wherein reducing the number of CD 19+ cells in the mammal treats acute lymphoblastic leukemia (ALL).

[00187] Embodiment 71. Lise of the vector according to any one of Embodiments 1 through 46 in the manufacture of a medicament for treating or preventing cancer, a viral infection, a bacterial infection, a fungal infection, or a parasite in a mammal in need thereof.

[00188] Embodiment 72. ETse of a mammalian immune cell according to any one of Embodiments 59 through 63 for reducing the number of CD19+ cells in a mammal.

[00189] Embodiment 73. A vector according to any one of Embodiments 1 through 46 for use in a method for reducing the number of CD19+ cells in a mammal.

[00190] Embodiment 74. A mammalian immune cell according to any one of

Embodiments 59 through 63 for use in a method for reducing the number of CD 19+ cells in a mammal.

[00191] Embodiment 75. A method of making a transgenic mammalian host cell, the method comprising introducing into a mammalian host cell the vector of any of Embodiments 39 through 46.

[00192] Embodiment 76. The method of Embodiment 75, wherein the mammalian host cell is an immune cell.

[00193] Embodiment 77. The method of Embodiment 76, wherein the immune cell is a T cell.

[00194] Embodiment 78. The method of Embodiment 77, wherein the T cell is a tumor- infiltrating lymphocyte (TIL).

[00195] Embodiment 79. The method of Embodiment 76, wherein the immune cell is a natural killer cell.

[00196] Embodiment 80. A mammalian immune cell comprising the vector of any one Embodiments 39 through 46.

[00197] Embodiment 81. The mammalian immune cell of Embodiment 80, wherein the mammalian immune cell is a T cell, natural killer (NK) cell, a monocytic/macrophage cell, or a dendritic cell. REFERENCES

1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science. 20l5;348(6230):62-68.

2. Kochenderfer IN, Wilson WH, Janik JE, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010; 116(20):4099-4102.

3. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor- modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725-733.

4. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 20l4;37l(l6): 1507-1517.

5. Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of l9-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med.

20l4;6(224):224ra225.

6. Kochenderfer JN, Dudley ME, Kassim SH, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CDl9 chimeric antigen receptor. J Clin Oncol. 2015; 33(6):540-549.

7. Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015; 385(9967):5l7-528.

8. Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123-2138.

9. Sadelain M, Riviere I, Riddell S. Therapeutic T cell engineering. Nature.

20l7;545(7655):423-43 l. 10. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl JMed. 20l8;378(5):439-448.

11. Campana D, Schwarz H, Imai C. 4-1BB chimeric antigen receptors. Cancer J.

20l4;20(2): 134-140.

12. Long AH, Haso WM, Shern JF, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med.

20l5;2l(6):58l-590.

13. Zhao Z, Condomines M, van der Stegen SJ, et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell. 2015;28(4):415-428.

14. Besser MJ, Shapira-Frommer R, Schachter J. Tumor-infiltrating lymphocytes: clinical experience. Cancer J. 20l5;2l(6):465-469.

15. Lu YC, Parker LL, Lu T, et al. Treatment of patients with metastatic cancer using a major histocompatibility complex class II-restricted T-cell receptor targeting the cancer germline antigen MAGE- A3. J Clin Oncol. 2017; 35(29): 3322-3329.

16. Atkins MB. Interleukin-2: clinical applications. Semin Oncol. 2002;29(3 Suppl 7): 12-

17.

17. Li Y, Strick-Marchand H, Lim Al, et al. Regulatory T cells control toxicity in a humanized model of IL-2 therapy. Nat Commun. 20l7;8(l): 1762.

18. Matsuoka K, Koreth J, Kim HT, et al. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med. 20l3;5(l79): l79ral43.

19. Manabe A, Coustan-Smith E, Kumagai M, et al. Interleukin-4 induces programmed cell death (apoptosis) in cases of high-risk acute lymphoblastic leukemia. Blood.

l994;83(7): 1731-1737. 20. de la Chapelle A, Traskelin AL, Juvonen E. Truncated erythropoietin receptor causes dominantly inherited benign human erythrocytosis. Proc Natl Acad Sci USA.

1993;90(10):4495-4499.

21. Imai C, Mihara K, Andreansky M, Nicholson IC, Pui CH, Campana D. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004;18:676-684.

22. Szymczak-Workman AL, Vignali KM, Vignali DA. Design and construction of 2A peptide-linked multi cistronic vectors. Cold Spring Harb Protoc. 20l2;20l2(2): 199-204.

23. Png YT, Vinanica N, Kamiya T, Shimasaki N, Coustan-Smith E, Campana D.

Blockade of CD7 expression in T cells for effective chimeric antigen receptor targeting of T- cell malignancies. Blood Advances, 1(25), 2348-2360. . Blood Advances. 20l7;l(25):2348- 2360.

24. Kudo K, Imai C, Lorenzini P, et al. T lymphocytes expressing a CD16 signaling receptor exert antibody-dependent cancer cell killing. Cancer Res. 20l4;74(l):93-l03.

25. Chang YH, Connolly J, Shimasaki N, Mimura K, Kono K, Campana D. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 20l3;73(6): 1777-1786.

26. Gouilleux F, Pallard C, Dusanter-Fourt I, et al. Prolactin, growth hormone, erythropoietin and granulocyte-macrophage colony stimulating factor induce MGF-Stat5 DNA binding activity. EMBO J. 1995;14(9):2005-2013.

27. Wakao H, Harada N, Kitamura T, Mui AL, Miyajima A. Interleukin 2 and erythropoietin activate STAT5/MGF via distinct pathways. EMBO J. 1995; 14(11):2527-

2535. 28. Witthuhn BA, Quelle FW, Silvennoinen O, et al. JAK2 associates with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietin. Cell. l993;74(2):227-236.

29. Kiladjian JJ, Winton EF, Talpaz M, Verstovsek S. Ruxolitinib for the treatment of patients with polycythemia vera. Expert Rev Hematol . 20l5;8(4):39l-40l.

30. Juvonen E, Ikkala E, Fyhrquist F, Ruutu T. Autosomal dominant erythrocytosis caused by increased sensitivity to erythropoietin. Blood. 1991 ;78(l l):3066-3069.

31. Meyer SC, Levine RL. Molecular pathways: molecular basis for sensitivity and resistance to JAK kinase inhibitors. Clin Cancer Res. 20l4;20(8):2051-2059.

32. Galson DL, Tan CC, Ratcliffe PJ, Bunn HF. Comparison of the hu006Dan and mouse erythropoietin genes shows extensive homology in the flanking regions. Blood. l993;82(l l):3321-3326.

SEQUENCES

[00198] SEQ ID NO: 1 : Epo receptor (EpoR) cDNA:

CCGGAATTCGCCACCATGGACCACCTCGGGGCGTCCCTCTGGCCCCAGGTCGGCT

CCCTTTGTCTCCTGCTCGCTGGGGCCGCCTGGGCGCCCCCGCCTAACCTCCCGGA

CCCCAAGTTCGAGAGCAAAGCGGCCTTGCTGGCGGCCCGGGGGCCCGAAGAGCT

TCTGTGCTTCACCGAGCGGTTGGAGGACTTGGTGTGTTTCTGGGAGGAAGCGGCG

AGCGCTGGGGTGGGCCCGGGCAACTACAGCTTCTCCTACCAGTTAGAAGATGAG

CCATGGAAGCTGTGTCGCCTGCACCAGGCTCCCACGGCTCGTGGTGCGGTGCGCT

TCTGGTGTTCGCTGCCTACAGCCGACACGTCGAGCTTCGTGCCCCTAGAGTTGCG

CGTCACAGCAGCCTCCGGCGCTCCGCGATATCACCGTGTCATCCACATCAATGAA

GTAGTGCTCCTAGACGCCCCCGTGGGGCTGGTGGCGCGGTTGGCTGACGAGAGC

GGCCACGTAGTGTTGCGCTGGCTCCCGCCGCCTGAGACACCCATGACGTCTCACA

TCCGCTACGAGGTGGACGTCTCGGCCGGCAACGGCGCAGGGAGCGTACAGAGGG

TGGAGATCCTGGAGGGCCGCACCGAGTGTGTGCTGAGCAACCTGCGGGGCCGGA

CGCGCTACACCTTCGCCGTCCGCGCGCGTATGGCTGAGCCGAGCTTCGGCGGCTT

CTGGAGCGCCTGGTCGGAGCCTGTGTCGCTGCTGACGCCTAGCGACCTGGACCCC CTCATCCTGACGCTCTCCCTCATCCTCGTGGTCATCCTGGTGCTGCTGACCGTGCT

CGCGCTGCTCTCCCACCGCCGGGCTCTGAAGCAGAAGATCTGGCCTGGCATCCCG

AGCCCAGAGAGCGAGTTTGAAGGCCTCTTCACCACCCACAAGGGTAACTTCCAG

CTGTGGCTGTACCAGAATGATGGCTGCCTGTGGTGGAGCCCCTGCACCCCCTTCA

CGGAGGACCCACCTGCTTCCCTGGAAGTCCTCTCAGAGCGCTGCTGGGGGACGA

TGCAGGCAGTGGAGCCGGGGACAGATGATGAGGGCCCCCTGCTGGAGCCAGTGG

GCAGTGAGCATGCCCAGGATACCTATCTGGTGCTGGACAAATGGTTGCTGCCCCG

GAACCCGCCCAGTGAGGACCTCCCAGGGCCTGGTGGCAGTGTGGACATAGTGGC

CATGGATGAAGGCTCAGAAGCATCCTCCTGCTCATCTGCTTTGGCCTCGAAGCCC

AGCCCAGAGGGAGCCTCTGCTGCCAGCTTTGAGTACACTATCCTGGACCCCAGCT

CCCAGCTCTTGCGTCCATGGACACTGTGCCCTGAGCTGCCCCCTACCCCACCCCA

CCTAAAGTACCTGTACCTTGTGGTATCTGACTCTGGCATCTCAACTGACTACAGC

TCAGGGGACTCCCAGGGAGCCCAAGGGGGCTTATCCGATGGCCCCTACTCCAAC

CCTTATGAGAACAGCCTTATCCCAGCCGCTGAGCCTCTGCCCCCCAGCTATGTGG

CTTGCTCTTAGCTCGAGCGG

[00199] SEQ ID NO: 2: Epo receptor (EpoR) amino acid:

MDHLGASLWPQVGSLCLLLAGAAWAPPPNLPDPKFESKAALLAARGPEELLCFTER LEDL VCF WEE AAS AGVGPGNY SF S Y QLEDEPWKLCRLHQ APT ARGA VRF W C SLPT A DTSSFVPLELRVTAASGAPRYHRVIHINEVVLLDAPVGLVARLADESGHVVLRWLPP PETPMTSHIRYEVDVSAGNGAGSVQRVEILEGRTECVLSNLRGRTRYTFAVRARMAE P SF GGF W SAW SEP V SLLTP SDLDPLILTL SLIL VVIL VLLTVL ALL SHRRALKQKIWPGI PSPESEFEGLFTTHKGNF QLWL Y QNDGCLWW SPCTPFTEDPP ASLEVLSERCW GTM QAVEPGTDDEGPLLEPVGSEHAQDTYLVLDKWLLPRNPPSEDLPGPGGSVDIVAMD EGSE AS SC S S AL ASKP SPEGAS AASFE YTILDP S SQLLRPWTLCPELPPTPPHLK YL YL VVSDSGISTDYSSGDSQGAQGGLSDGPYSNPYENSLIPAAEPLPPSYVACS

[00200] SEQ ID NO: 3 : EpoR with Flag tag cDNA:

ATATATGAATTCGCCACCATGGACCACCTCGGGGCGTCCCTCTGGCCCCAGGTCG

GCTCCCTTTGTCTCCTGCTCGCTGGGGCCGCCTGGGCGCCCCCGCCTAACCTCCC

GGACCCCAAGTTCGAGAGCAAAGCGGCCTTGCTGGCGGCCCGGGGGCCCGAAGA

GCTTCTGTGCTTCACCGAGCGGTTGGAGGACTTGGTGTGTTTCTGGGAGGAAGCG

GCGAGCGCTGGGGTGGGCCCGGGCAACTACAGCTTCTCCTACCAGTTAGAAGAT

GAGCCATGGAAGCTGTGTCGCCTGCACCAGGCTCCCACGGCTCGTGGTGCGGTG

CGCTTCTGGTGTTCGCTGCCTACAGCCGACACGTCGAGCTTCGTGCCCCTAGAGT TGCGCGTCACAGCAGCCTCCGGCGCTCCGCGATATCACCGTGTCATCCACATCAA

TGAAGTAGTGCTCCTAGACGCCCCCGTGGGGCTGGTGGCGCGGTTGGCTGACGA

GAGCGGCCACGTAGTGTTGCGCTGGCTCCCGCCGCCTGAGACACCCATGACGTCT

CACATCCGCTACGAGGTGGACGTCTCGGCCGGCAACGGCGCAGGGAGCGTACAG

AGGGTGGAGATCCTGGAGGGCCGCACCGAGTGTGTGCTGAGCAACCTGCGGGGC

CGGACGCGCTACACCTTCGCCGTCCGCGCGCGTATGGCTGAGCCGAGCTTCGGCG

GCTTCTGGAGCGCCTGGTCGGAGCCTGTGTCGCTGCTGACGCCTAGCGACCTGGA

CCCCCTCATCCTGACGCTCTCCCTCATCCTCGTGGTCATCCTGGTGCTGCTGACCG

TGCTCGCGCTGCTCTCCCACCGCCGGGCTCTGAAGCAGAAGATCTGGCCTGGCAT

CCCGAGCCCAGAGAGCGAGTTTGAAGGCCTCTTCACCACCCACAAGGGTAACTT

CCAGCTGTGGCTGTACCAGAATGATGGCTGCCTGTGGTGGAGCCCCTGCACCCCC

TTCACGGAGGACCCACCTGCTTCCCTGGAAGTCCTCTCAGAGCGCTGCTGGGGGA

CGAT GC AGGC AGT GGAGCCGGGGAC AGAT GAT GAGGGCCCCCTGCTGGAGCC AG

TGGGCAGTGAGCATGCCCAGGATACCTATCTGGTGCTGGACAAATGGTTGCTGCC

CCGGAACCCGCCCAGTGAGGACCTCCCAGGGCCTGGTGGCAGTGTGGACATAGT

GGCCATGGATGAAGGCTCAGAAGCATCCTCCTGCTCATCTGCTTTGGCCTCGAAG

CCCAGCCCAGAGGGAGCCTCTGCTGCCAGCTTTGAGTACACTATCCTGGACCCCA

GCTCCCAGCTCTTGCGTCCATGGACACTGTGCCCTGAGCTGCCCCCTACCCCACC

CCACCTAAAGTACCTGTACCTTGTGGTATCTGACTCTGGCATCTCAACTGACTAC

AGCTCAGGGGACTCCCAGGGAGCCCAAGGGGGCTTATCCGATGGCCCCTACTCC

AACCCTTATGAGAACAGCCTTATCCCAGCCGCTGAGCCTCTGCCCCCCAGCTATG

TGGCTTGCTCTGACTACAAAGACGATGACGACAAGTAGCTCGAGTATATA

[00201] SEQ ID NO: 4: EpoR with Flag tag amino acid:

MDHLGASLWPQVGSLCLLLAGAAWAPPPNLPDPKFESKAALLAARGPEELLCFTER LEDL VCF WEE AAS AGVGPGNY SF S Y QLEDEPWKLCRLHQ APT ARGA VRF W C SLPT A DTSSFVPLELRVTAASGAPRYHRVIHINEVVLLDAPVGLVARLADESGHVVLRWLPP PETPMTSHIRYEVDVSAGNGAGSVQRVEILEGRTECVLSNLRGRTRYTFAVRARMAE P SF GGF W SAW SEP V SLLTP SDLDPLILTL SLIL VVIL VLLTVL ALL SHRRALKQKIWPGI PSPESEFEGLFTTHKGNF QLWL Y QNDGCLWW SPCTPFTEDPP ASLEVLSERCW GTM QAVEPGTDDEGPLLEPVGSEHAQDTYLVLDKWLLPRNPPSEDLPGPGGSVDIVAMD EGSE AS SC S S AL ASKP SPEGAS AASFE YTILDP S SQLLRPWTLCPELPPTPPHLK YL YL VV SD SGISTD Y S SGD SQGAQGGLSDGP Y SNP YEN SLIP AAEPLPP S YVAC SD YKDDDD K [00202] SEQ ID NO: 5 : mutant EpoR (EpoRm) cDNA:

CCGGAATTCGCCACCATGGACCACCTCGGGGCGTCCCTCTGGCCCCAGGTCGGCT

CCCTTTGTCTCCTGCTCGCTGGGGCCGCCTGGGCGCCCCCGCCTAACCTCCCGGA

CCCCAAGTTCGAGAGCAAAGCGGCCTTGCTGGCGGCCCGGGGGCCCGAAGAGCT

TCTGTGCTTCACCGAGCGGTTGGAGGACTTGGTGTGTTTCTGGGAGGAAGCGGCG

AGCGCTGGGGTGGGCCCGGGCAACTACAGCTTCTCCTACCAGTTAGAAGATGAG

CCATGGAAGCTGTGTCGCCTGCACCAGGCTCCCACGGCTCGTGGTGCGGTGCGCT

TCTGGTGTTCGCTGCCTACAGCCGACACGTCGAGCTTCGTGCCCCTAGAGTTGCG

CGTCACAGCAGCCTCCGGCGCTCCGCGATATCACCGTGTCATCCACATCAATGAA

GTAGTGCTCCTAGACGCCCCCGTGGGGCTGGTGGCGCGGTTGGCTGACGAGAGC

GGCCACGTAGTGTTGCGCTGGCTCCCGCCGCCTGAGACACCCATGACGTCTCACA

TCCGCTACGAGGTGGACGTCTCGGCCGGCAACGGCGCAGGGAGCGTACAGAGGG

TGGAGATCCTGGAGGGCCGCACCGAGTGTGTGCTGAGCAACCTGCGGGGCCGGA

CGCGCTACACCTTCGCCGTCCGCGCGCGTATGGCTGAGCCGAGCTTCGGCGGCTT

CTGGAGCGCCTGGTCGGAGCCTGTGTCGCTGCTGACGCCTAGCGACCTGGACCCC

CTCATCCTGACGCTCTCCCTCATCCTCGTGGTCATCCTGGTGCTGCTGACCGTGCT

CGCGCTGCTCTCCCACCGCCGGGCTCTGAAGCAGAAGATCTGGCCTGGCATCCCG

AGCCCAGAGAGCGAGTTTGAAGGCCTCTTCACCACCCACAAGGGTAACTTCCAG

CTGTGGCTGTACCAGAATGATGGCTGCCTGTGGTGGAGCCCCTGCACCCCCTTCA

CGGAGGACCCACCTGCTTCCCTGGAAGTCCTCTCAGAGCGCTGCTGGGGGACGA

TGCAGGCAGTGGAGCCGGGGACAGATGATGAGGGCCCCCTGCTGGAGCCAGTGG

GCAGTGAGCATGCCCAGGATACCTATCTGGTGCTGGACAAATGGTTGCTGCCCCG

GAACCCGCCCAGTGAGGACCTCCCAGGGCCTGGTGGCAGTGTGGACATAGTGGC

CATGGATGAAGGCTCAGAAGCATCCTCCTGCTCATCTGCTTTGGCCTCGAAGCCC

AGCCCAGAGGGAGCCTCTGCTGCCAGCTTTGAGTACACTATCCTGGACCCCAGCT

CCCAGCTCTTGCGTCCATAGACACTGTGCCCTGAGCTGCCCCCTACCCCACCCCA

CCTAAAGTACCTGTACCTTGTGGTATCTGACTCTGGCATCTCAACTGACTACAGC

TCAGGGGACTCCCAGGGAGCCCAAGGGGGCTTATCCGATGGCCCCTACTCCAAC

CCTTATGAGAACAGCCTTATCCCAGCCGCTGAGCCTCTGCCCCCCAGCTATGTGG

CTTGCTCTTAGCTCGAGCGG

[00203] SEQ ID NO: 6: mutant EpoR (EpoRm) amino acid:

MDHLGASLWPQVGSLCLLLAGAAWAPPPNLPDPKFESKAALLAARGPEELLCFTER LEDL VCF WEE AAS AGVGPGNY SF S Y QLEDEPWKLCRLHQ APT ARGA VRF W C SLPT A DTSSFVPLELRVTAASGAPRYHRVIHINEVVLLDAPVGLVARLADESGHVVLRWLPP PETPMTSHIRYEVDVSAGNGAGSVQRVEILEGRTECVLSNLRGRTRYTFAVRARMAE P SF GGF W SAW SEP V SLLTP SDLDPLILTL SLIL VVIL VLLTVL ALL SHRRALKQKIWPGI PSPESEFEGLFTTHKGNF QLWL Y QNDGCLWW SPCTPFTEDPP ASLEVLSERCW GTM QAVEPGTDDEGPLLEPVGSEHAQDTYLVLDKWLLPRNPPSEDLPGPGGSVDIVAMD EGSE AS SC S S AL ASKPSPEGAS AASFE YTILDP S SQLLRP

[00204] SEQ ID NO: 7: EpoRm with Flag tag cDNA:

ATATATGAATTCGCCACCATGGACCACCTCGGGGCGTCCCTCTGGCCCCAGGTCG

GCTCCCTTTGTCTCCTGCTCGCTGGGGCCGCCTGGGCGCCCCCGCCTAACCTCCC

GGACCCCAAGTTCGAGAGCAAAGCGGCCTTGCTGGCGGCCCGGGGGCCCGAAGA

GCTTCTGTGCTTCACCGAGCGGTTGGAGGACTTGGTGTGTTTCTGGGAGGAAGCG

GCGAGCGCTGGGGTGGGCCCGGGCAACTACAGCTTCTCCTACCAGTTAGAAGAT

GAGCCATGGAAGCTGTGTCGCCTGCACCAGGCTCCCACGGCTCGTGGTGCGGTG

CGCTTCTGGTGTTCGCTGCCTACAGCCGACACGTCGAGCTTCGTGCCCCTAGAGT

TGCGCGTCACAGCAGCCTCCGGCGCTCCGCGATATCACCGTGTCATCCACATCAA

TGAAGTAGTGCTCCTAGACGCCCCCGTGGGGCTGGTGGCGCGGTTGGCTGACGA

GAGCGGCCACGTAGTGTTGCGCTGGCTCCCGCCGCCTGAGACACCCATGACGTCT

CACATCCGCTACGAGGTGGACGTCTCGGCCGGCAACGGCGCAGGGAGCGTACAG

AGGGTGGAGATCCTGGAGGGCCGCACCGAGTGTGTGCTGAGCAACCTGCGGGGC

CGGACGCGCTACACCTTCGCCGTCCGCGCGCGTATGGCTGAGCCGAGCTTCGGCG

GCTTCTGGAGCGCCTGGTCGGAGCCTGTGTCGCTGCTGACGCCTAGCGACCTGGA

CCCCCTCATCCTGACGCTCTCCCTCATCCTCGTGGTCATCCTGGTGCTGCTGACCG

TGCTCGCGCTGCTCTCCCACCGCCGGGCTCTGAAGCAGAAGATCTGGCCTGGCAT

CCCGAGCCCAGAGAGCGAGTTTGAAGGCCTCTTCACCACCCACAAGGGTAACTT

CCAGCTGTGGCTGTACCAGAATGATGGCTGCCTGTGGTGGAGCCCCTGCACCCCC

TTCACGGAGGACCCACCTGCTTCCCTGGAAGTCCTCTCAGAGCGCTGCTGGGGGA

CGAT GC AGGC AGT GGAGCCGGGGAC AGAT GAT GAGGGCCCCCTGCTGGAGCC AG

TGGGCAGTGAGCATGCCCAGGATACCTATCTGGTGCTGGACAAATGGTTGCTGCC

CCGGAACCCGCCCAGTGAGGACCTCCCAGGGCCTGGTGGCAGTGTGGACATAGT

GGCCATGGATGAAGGCTCAGAAGCATCCTCCTGCTCATCTGCTTTGGCCTCGAAG

CCCAGCCCAGAGGGAGCCTCTGCTGCCAGCTTTGAGTACACTATCCTGGACCCCA

GCTCCCAGCTCTTGCGTCCAGACTACAAAGACGATGACGACAAGTAGCTCGAGT

ATATA [00205] SEQ ID NO: 8: EpoRm with Flag tag amino acid:

MDHLGASLWPQVGSLCLLLAGAAWAPPPNLPDPKFESKAALLAARGPEELLCFTER LEDL VCF WEE AAS AGVGPGNY SF S Y QLEDEPWKLCRLHQ APT ARGA VRF W C SLPT A DTSSFVPLELRVTAASGAPRYHRVIHINEVVLLDAPVGLVARLADESGHVVLRWLPP PETPMTSHIRYEVDVSAGNGAGSVQRVEILEGRTECVLSNLRGRTRYTFAVRARMAE P SF GGF W SAW SEP V SLLTP SDLDPLILTL SLIL VVIL VLLTVL ALL SHRRALKQKIWPGI PSPESEFEGLFTTHKGNF QLWL Y QNDGCLWW SPCTPFTEDPP ASLEVLSERCW GTM QAVEPGTDDEGPLLEPVGSEHAQDTYLVLDKWLLPRNPPSEDLPGPGGSVDIVAMD EGSE AS SC S S AL ASKP SPEGAS AASFE YTILDP S SQLLRPD YKDDDDK

[00206] SEQ ID NO: 9: anti-CDl9-4lBB-CD3C CAR cDNA:

GAATTCGGCTTCCACCATGGCCTTACCAGTGACCGCCTTGCTCCTGCCGCTGGCC

TTGCTGCTCCACGCCGCCAGGCCGGACATCCAGATGACACAGACTACATCCTCCC

TGTCTGCCTCTCTGGGAGACAGAGTCACCATCAGTTGCAGGGCAAGTCAGGACA

TTAGTAAATATTTAAATTGGTATCAGCAGAAACCAGATGGAACTGTTAAACTCCT

GATCTACCATACATCAAGATTACACTCAGGAGTCCCATCAAGGTTCAGTGGCAGT

GGGTCTGGAACAGATTATTCTCTCACCATTAGCAACCTGGAGCAAGAAGATATTG

CCACTTACTTTTGCCAACAGGGTAATACGCTTCCGTACACGTTCGGAGGGGGGAC

CAAGCTGGAGATCACAGGTGGCGGTGGCTCGGGCGGTGGTGGGTCGGGTGGCGG

CGGATCTGAGGTGAAACTGCAGGAGTCAGGACCTGGCCTGGTGGCGCCCTCACA

GAGCCTGTCCGTCACATGCACTGTCTCAGGGGTCTCATTACCCGACTATGGTGTA

AGCTGGATTCGCCAGCCTCCACGAAAGGGTCTGGAGTGGCTGGGAGTAATATGG

GGTAGTGAAACCACATACTATAATTCAGCTCTCAAATCCAGACTGACCATCATCA

AGGACAACTCCAAGAGCCAAGTTTTCTTAAAAATGAACAGTCTGCAAACTGATG

ACACAGCCATTTACTACTGTGCCAAACATTATTACTACGGTGGTAGCTATGCTAT

GGACTACTGGGGCCAAGGAACCTCAGTCACCGTCTCCTCAACCACGACGCCAGC

GCCGCGACCACCAACACCGGCGCCCACCATCGCGTCGCAGCCCCTGTCCCTGCGC

CCAGAGGCGTGCCGGCCAGCGGCGGGGGGCGCAGTGCACACGAGGGGGCTGGA

CTTCGCCTGTGATATCTACATCTGGGCGCCCTTGGCCGGGACTTGTGGGGTCCTT

CTCCTGTCACTGGTTATCACCCTTTACTGCAAACGGGGCAGAAAGAAACTCCTGT

ATATATTCAAACAACCATTTATGAGACCAGTACAAACTACTCAAGAGGAAGATG

GCTGT AGCTGCCGATTTCC AGAAGAAGAAGAAGGAGGAT GT GAACTGAGAGT GA

AGTTCAGCAGGAGCGCAGACGCCCCCGCGTACCAGCAGGGCCAGAACCAGCTCT

AT A ACGAGCTC A ATCT AGGAC GA AGAGAGGAGT AC GAT GTTTT GGAC A AGAGAC GTGGCCGGGACCCTGAGATGGGGGGAAAGCCGAGAAGGAAGAACCCTCAGGAA

GGCCTGTACAATGAACTGCAGAAAGATAAGATGGCGGAGGCCTACAGTGAGATT

GGGATGAAAGGCGAGCGCCGGAGGGGCAAGGGGCACGATGGCCTTTACCAGGG

TCTCAGTACAGCCACCAAGGACACCTACGACGCCCTTCACATGCAGGCCCTGCCC

CCTCGCTAACAGCCAGGGGATTTCACCACTCAAAGGCCAGACCTGCAGACGCCC

AGATTATGAGACACACTCGAGCC

[00207] SEQ ID NO: 10: anti-CD 19-41BB-Oϋ3z CAR amino acid:

MALPVTALLLPLALLLHAARPDIQMTQTTSSLSASLGDRVTISCRASQDISKYLNWY

QQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGNT LP

YTFGGGTKLEITGGGGSGGGGSGGGGSEVKLQESGPGLVAPSQSLSVTCTVSGVSLP

D Y GV SWIRQPPRKGLEWLGVIWGSETT YYN S ALKSRLTIIKDN SKSQVFLKMN SLQT

DDT AIYY C AKHYYY GGS YAMD YW GQGT S VT V S STTTP APRPPTP APTI ASQPL SLRP

E ACRP A AGGA VHTRGLDF ACDI YIW APL AGTCGVLLL SL VITL Y CKRGRKKLL YIFK

QPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNL

GRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERR

RGKGHDGLYQGLSTATKDTYDALHMQALPPR

[00208] SEQ ID NO: 11 : EpoRm-2A-CAR cDNA:

GCCACCATGGACCACCTCGGGGCGTCCCTCTGGCCCCAGGTCGGCTCCCTTTGTC

TCCTGCTCGCTGGGGCCGCCTGGGCGCCCCCGCCTAACCTCCCGGACCCCAAGTT

CGAGAGCAAAGCGGCCTTGCTGGCGGCCCGGGGGCCCGAAGAGCTTCTGTGCTT

CACCGAGCGGTTGGAGGACTTGGTGTGTTTCTGGGAGGAAGCGGCGAGCGCTGG

GGTGGGCCCGGGCAACTACAGCTTCTCCTACCAGTTAGAAGATGAGCCATGGAA

GCTGTGTCGCCTGCACCAGGCTCCCACGGCTCGTGGTGCGGTGCGCTTCTGGTGT

TCGCTGCCTACAGCCGACACGTCGAGCTTCGTGCCCCTAGAGTTGCGCGTCACAG

CAGCCTCCGGCGCTCCGCGATATCACCGTGTCATCCACATCAATGAAGTAGTGCT

CCTAGACGCCCCCGTGGGGCTGGTGGCGCGGTTGGCTGACGAGAGCGGCCACGT

AGTGTTGCGCTGGCTCCCGCCGCCTGAGACACCCATGACGTCTCACATCCGCTAC

GAGGTGGACGTCTCGGCCGGCAACGGCGCAGGGAGCGTACAGAGGGTGGAGAT

CCTGGAGGGCCGCACCGAGTGTGTGCTGAGCAACCTGCGGGGCCGGACGCGCTA

CACCTTCGCCGTCCGCGCGCGTATGGCTGAGCCGAGCTTCGGCGGCTTCTGGAGC

GCCTGGTCGGAGCCTGTGTCGCTGCTGACGCCTAGCGACCTGGACCCCCTCATCC

TGACGCTCTCCCTCATCCTCGTGGTCATCCTGGTGCTGCTGACCGTGCTCGCGCTG

CTCTCCCACCGCCGGGCTCTGAAGCAGAAGATCTGGCCTGGCATCCCGAGCCCA GAGAGCGAGTTTGAAGGCCTCTTCACCACCCACAAGGGTAACTTCCAGCTGTGG

CTGTACCAGAATGATGGCTGCCTGTGGTGGAGCCCCTGCACCCCCTTCACGGAGG

ACCCACCTGCTTCCCTGGAAGTCCTCTCAGAGCGCTGCTGGGGGACGATGCAGGC

AGTGGAGCCGGGGACAGATGATGAGGGCCCCCTGCTGGAGCCAGTGGGCAGTGA

GCATGCCCAGGATACCTATCTGGTGCTGGACAAATGGTTGCTGCCCCGGAACCCG

CCCAGTGAGGACCTCCCAGGGCCTGGTGGCAGTGTGGACATAGTGGCCATGGAT

GAAGGCTCAGAAGCATCCTCCTGCTCATCTGCTTTGGCCTCGAAGCCCAGCCCAG

AGGGAGCCTCTGCTGCCAGCTTTGAGTACACTATCCTGGACCCCAGCTCCCAGCT

CTTGCGTCCAGGATCAGGCGAGGGCAGAGGAAGTCTGCTAACATGCGGTGACGT

CGAGGAGAATCCTGGCCCAATGGCCTTACCAGTGACCGCCTTGCTCCTGCCGCTG

GCCTTGCTGCTCCACGCCGCCAGGCCGGACATCCAGATGACACAGACTACATCCT

CCCTGTCTGCCTCTCTGGGAGACAGAGTCACCATCAGTTGCAGGGCAAGTCAGG

AC ATT AGT AAAT ATTT AA ATT GGT AT C AGC AGAAACC AGAT GGAACTGTT AAACT

CCTGATCTACCATACATCAAGATTACACTCAGGAGTCCCATCAAGGTTCAGTGGC

AGTGGGTCTGGAACAGATTATTCTCTCACCATTAGCAACCTGGAGCAAGAAGAT

ATTGCCACTTACTTTTGCCAACAGGGTAATACGCTTCCGTACACGTTCGGAGGGG

GG AC C A AGC T GG AG AT C AC AGGT GGC GGT GGC T C GGGC GGT GGT GGGT C GGGT G

GCGGCGGATCTGAGGTGAAACTGCAGGAGTCAGGACCTGGCCTGGTGGCGCCCT

CACAGAGCCTGTCCGTCACATGCACTGTCTCAGGGGTCTCATTACCCGACTATGG

TGTAAGCTGGATTCGCCAGCCTCCACGAAAGGGTCTGGAGTGGCTGGGAGTAAT

ATGGGGTAGTGAAACCACATACTATAATTCAGCTCTCAAATCCAGACTGACCATC

ATCAAGGACAACTCCAAGAGCCAAGTTTTCTTAAAAATGAACAGTCTGC AAACT

GATGACACAGCCATTTACTACTGTGCCAAACATTATTACTACGGTGGTAGCTATG

CTATGGACTACTGGGGCCAAGGAACCTCAGTCACCGTCTCCTCAACCACGACGCC

AGCGCCGCGACCACCAACACCGGCGCCCACCATCGCGTCGCAGCCCCTGTCCCT

GCGCCCAGAGGCGTGCCGGCCAGCGGCGGGGGGCGCAGTGCACACGAGGGGGC

TGGACTTCGCCTGTGATATCTACATCTGGGCGCCCTTGGCCGGGACTTGTGGGGT

CCTTCTCCTGTCACTGGTTATCACCCTTTACTGCAAACGGGGCAGAAAGAAACTC

CTGTATATATTCAAACAACCATTTATGAGACCAGTACAAACTACTCAAGAGGAA

GAT GGC T GT AGC T GC C G ATT T C C AG A AG A AG A AG A AGG AGG AT GT G A AC T GAGA

GTGAAGTTCAGCAGGAGCGCAGACGCCCCCGCGTACCAGCAGGGCCAGAACCAG

CTCT AT AACGAGCTC AATCT AGGACGAAGAGAGGAGT ACGATGTTTTGGAC AAG

AGACGTGGCCGGGACCCTGAGATGGGGGGAAAGCCGAGAAGGAAGAACCCTCA GG A AGGC C T GT AC A AT G A AC T GC AG A A AG AT A AG AT GGC GG AGGC C T AC AGT G AGATTGGGATGAAAGGCGAGCGCCGGAGGGGCAAGGGGCACGATGGCCTTTAC CAGGGTCTCAGTACAGCCACCAAGGACACCTACGACGCCCTTCACATGCAGGCC CTGCCCCCTCGCTAA

[00209] SEQ ID NO: 12: EpoRm-2A-CAR amino acid:

MDHLGASLWPQVGSLCLLLAGAAWAPPPNLPDPKFESKAALLAARGPEELLCFTER

LEDL VCF WEE AAS AGVGPGNY SF S Y QLEDEPWKLCRLHQ APT ARGA VRF W C SLPT A

DTSSFVPLELRVTAASGAPRYHRVIHINEVVLLDAPVGLVARLADESGHVVLRWLPP

PETPMTSHIRYEVDVSAGNGAGSVQRVEILEGRTECVLSNLRGRTRYTFAVRARMAE

P SF GGF W SAW SEP V SLLTP SDLDPLILTL SLIL VVIL VLLTVL ALL SHRRALKQKIWPGI

PSPESEFEGLFTTHKGNF QLWL Y QNDGCLWW SPCTPFTEDPP ASLEVLSERCW GTM

QAVEPGTDDEGPLLEPVGSEHAQDTYLVLDKWLLPRNPPSEDLPGPGGSVDIVAMD

EGSEASSCSSALASKPSPEGASAASFEYTILDPSSQLLRPGSGEGRGSLLTCGDVEE NP

GPMALPVTALLLPLALLLHAARPDIQMTQTTSSLSASLGDRVTISCRASQDISKYLN W

YQQKPDGTVKLLIYHTSRLHSGVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQGN T

LP YTF GGGTKLEIT GGGGS GGGGS GGGGSEVKLQE S GPGL V AP S Q SL S VTCT V SGV S

LPD Y GV SWIRQPPRKGLEWLGVIWGSETT YYN S ALKSRLTIIKDN SKSQVFLKMN SL

QTDDTAIYYCAKHYYYGGSYAMDYWGQGTSVTVSSTTTPAPRPPTPAPTIASQPLSL

RPE ACRP A AGGA VHTRGLDF ACDI YIW APL AGTCGVLLL SL VITL Y CKRGRKKLL YI

FKQPFMRP VQTTQEEDGC SCRFPEEEEGGCELRVKF SRS ADAP AYQQGQNQLYNEL

NLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGE

RRRGKGHDGLYQGLSTATKDTYDALHMQALPPR

[00210] SEQ ID NO: 13 : P2A cDNA:

GGAAGCGGAGCTACTAACTTCAGCCTGCTGAAGCAGGCTGGAGACGTGGAGGAG

AACCCTGGACCT

[00211] SEQ ID NO: 14: P2A amino acid: GSGATNF SLLKQ AGD VEENPGP

[00212] SEQ ID NO: 15 : T2A cDNA:

GGA AGC GGAGAGGGC AGAGGA AGTCTGCT A AC AT GCGGT GAC GT C GAGGAGA A TCCTGGACCT

[00213] SEQ ID NO: 16: T2A amino acid: GSGEGRGSLLTCGD VEENPGP

[00214] SEQ ID NO: 17: E2A cDNA:

GGAAGCGGACAGTGTACTAATTATGCTCTCTTGAAATTGGCTGGAGATGTTGAGA GC AACCCTGGACCT [00215] SEQ ID NO: 18: E2A amino acid: GSGQCTNYALLKLAGDVESNPGP

[00216] SEQ ID NO: 19: F2A cDNA:

GGAAGCGGAGTGAAACAGACTTTGAATTTTGACCTTCTCAAGTTGGCGGGAGAC

GTGGAGTCCAACCCTGGACCT

[00217] SEQ ID NO: 20: F2A amino acid: GSGVKQTLNFDLLKLAGDVESNPGP

[00218] SEQ ID NO: 21 : CD8a signal peptide cDNA:

ATGGCCTTACCAGTGACCGCCTTGCTCCTGCCGCTGGCCTTGCTGCTCCACGCCG

CCAGGCCG

[00219] SEQ ID NO: 22: CD8a signal peptide amino acid:

M ALP VT ALLLPL ALLLH AARP

INCORPORATION BY REFERENCE; EQUIVALENTS

[00220] The teachings of all patents, published applications and references cited herein are incorporated by reference in their entirety.

[00221] While example embodiments have been particularly shown and described, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the embodiments encompassed by the appended claims.