BIFUNCTIONAL SUPERKINES AND USES THEREOF

Information

  • Patent Application
  • 20230365642
  • Publication Number
    20230365642
  • Date Filed
    June 24, 2021
    2 years ago
  • Date Published
    November 16, 2023
    5 months ago
Abstract
Bispecific human interleukin-2 (IL-2) cytokine fusions are provided. In particular, provided are bispecific IL-2 cytokine fusions that have an increased binding capacity for IL-2Rβ receptor as compared to wild-type IL-2 for use in monotherapeutic applications as well as in combination therapies with anti-PD-1 antibodies for the treatment of cancer. Also provided are pharmaceutical compositions that include such bispecific IL-2 cytokine fusions.
Description
DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY

The contents of the text file submitted electronically herewith are incorporated herein by reference in their entirety: A computer readable format copy of the Sequence Listing (Filename: “117802-5013-US_Sequence_Listing.txt”; Date created: Dec. 21, 2022; File size: 873 KB).


BACKGROUND

Interleukin 2 (IL-2) is a pluripotent cytokine produced primarily by activated CD4+ T cells, which plays a crucial role in producing a normal immune response. IL-2 promotes proliferation and expansion of activated T lymphocytes, potentiates B cell growth, and activates monocytes and natural killer cells. It was by virtue of these activities that IL-2 was tested and is used as an approved treatment of cancer (aldesleukin, Proleukin®). In eukaryotic cells, human IL-2 is synthesized as a precursor polypeptide of 153 amino acids, from which 20 amino acids are removed to generate mature secreted IL-2 (Taniguchi 1983). Recombinant human IL-2 has been produced in E. coli (Rosenberg 1984), in insect cells (Smith 1985) and in mammalian COS cells (Taniguchi 1983).


Interleukin-2 (IL-2) is a four α-helical bundle type I cytokine first identified as a T cell growth factor (Morgan et al., Science 193: 1007 (1976)) but subsequently shown to have broad actions. IL-2 promotes T helper differentiation (Zhu et al., Annual review of immunology 28: 445 (2010); Liao et al., Nat Immunol 9: 1288 (2008); and Liao et al., Nat Immunol 12: 551 (2011)) and the development of regulatory T (Treg) cells (Cheng et al., Immunol Rev 241: 63 (2011)), induces natural killer and lymphokine activated killer activity (Liao et al., Immunity 38: 13 (2013)), and mediates activation-induced cell death (AICD) (Lenardo et al., Nature 353: 858 (1991)).


IL-2 works by interacting with three different receptors: the interleukin 2 receptor alpha (IL-2Rα; CD25), the interleukin 2 receptor beta (IL-2Rβ;CD122), and the interleukin 2 receptor gamma (IL-2Rγ;CD132; common gamma chain). The first receptor to be identified was the IL-2Rα, which is a 55 kD polypeptide (p55) that appears upon T cell activation and was originally called Tac (for T activation) antigen. The IL-2Rα binds IL-2 with a Kd of approximately 10−8 M, and is also known as the “low affinity” IL-2 receptor. Binding of IL-2 to cells expressing only the IL-2Rα does not lead to any detectable biologic response. In most circumstances, IL-2 works through three different receptors: the IL-2Rα, the IL-2Rβ, and the IL-2Rγ. Most cells, such as resting T cells, are not responsive to IL-2 since they only express the IL-2Rβ, and the IL-2Rγ, which have low affinity for IL-2. Upon stimulation, resting T cells express the relatively high affinity IL-2 receptor IL-2Rα. Binding of IL-2 to the IL-2Rα causes this receptor to sequentially engage the IL-2Rβ, and the IL-2Rγ, bringing about T cell activation. IL-2 “superkines” with augmented action due to enhanced binding affinity for IL-2Rβ were previously developed (Levin et al., Nature 484: 529 (2012)).


Despite the wealth of knowledge around IL-2, including IL-2 superagonists, there remains a need in the art for bispecific IL-2 cytokine fusions. The present invention meets this need, providing IL-2 superagonists or agonists as bispecific fusions with a second cytokine. In some embodiments, the IL-2 muteins portions of the bispecific fusions comprise substitutions L80F, R81D, L85V, 186V and I92F, numbered in accordance with wild-type IL-2.


BRIEF SUMMARY

IL-2 exerts a wide spectrum of effects on the immune system, and it plays crucial roles in regulating both immune activation and homeostasis. As an immune system stimulator, bispecific IL-2 cytokine fusions are described and also find use in monotherapies as well as in combination with anti-PD-1 antibodies for the treatment of cancer.


In some embodiments, the present invention provides a bispecific IL-2 cytokine fusion, comprising an IL-2 mutein as described herein fused to a second cytokine.


In some embodiments of the bispecific IL-2 cytokine fusion, the IL-2 mutein comprises the following amino acid substitutions: L80F, R81D, L85V, 186V, and I92F, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


In some embodiments of the bispecific IL-2 cytokine fusion, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


In some embodiments, the bispecific IL-2 cytokine fusion of claim 1, wherein the IL-2 mutein further comprises K43N substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


In some embodiments of the bispecific IL-2 cytokine fusion, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


In some embodiments of the bispecific IL-2 cytokine fusion, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


In some embodiments of the bispecific IL-2 cytokine fusion, the IL-2 mutein further comprises an E62A substitution and an F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


In some embodiments of the bispecific IL-2 cytokine fusion, the IL-2 mutein is an IL-2 mutein as described in Tables 2, 4, 5, and/or 6.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion comprises a sequence as described in Tables 2, 4, 5, 6, 10, and/or 12 and/or FIG. 54.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion comprises SEQ ID NO:146, SEQ ID NO:147, SEQ ID NO: 148, SEQ ID NO: 149, or SEQ ID NO:150.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion comprises MDNA413-Fc-MDNA109 or MDNA109FEAA-Fc-MDNA413.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion further comprises an Fc antibody fragment.


In some embodiments of the bispecific IL-2 cytokine fusion, the Fc antibody fragment is a human Fc antibody fragment.


In some embodiments of the bispecific IL-2 cytokine fusion, the Fc antibody fragment comprises a N297A substitution.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion further comprises albumin.


In some embodiments of the bispecific IL-2 cytokine fusion, the second cytokine is selected from the group consisting of IL-4, IL-13, IL-10, IL-12, IL15, and IL-18.


In some embodiments of the bispecific IL-2 cytokine fusion, the second cytokine is IL-4 or IL-13.


In some embodiments of the bispecific IL-2 cytokine fusion, the second cytokine is as described in Table 7 and/or Table 8 and/or Table 12 and/or Table 28 and/or FIG. 54.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion exhibits increased binding capacity for IL-2Rβ as compared to wild-type human IL-2.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion exhibits a greater binding affinity for IL-2Rβ as compared to wild-type human IL-2.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion exhibits abrogated IL2Ru binding (i.e., does not significantly bind to IL2Ru).


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion exhibits decreased binding affinity for CD25 as compared to wild-type human IL-2.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion comprises SEQ ID NO:146, SEQ ID NO:147, SEQ ID NOs:148 and 213, SEQ ID NOs:149 and 213, SEQ ID NO:150, SEQ ID NOs:151 and 214, SEQ ID NO:152, SEQ ID NO:153, SEQ ID NO:154, SEQ ID NO:155, SEQ ID NO:156, SEQ ID NOs:157 and 213, SEQ ID NO:158, SEQ ID NO:159, SEQ ID NO:160, SEQ ID NOs:161 and 215, SEQ ID NOs:162 and 216, SEQ ID NO:163, SEQ ID NO:164, SEQ ID NOs:165 and 213, SEQ ID NOs:166 and 213, or SEQ ID NOs:167 and 217, or a sequence from Table 12 or Table 28 or FIG. 54.


In some embodiments of the bispecific IL-2 cytokine fusion, the bispecific IL-2 cytokine fusion comprises MDNA413-Fc-MDNA109, MDNA109FEAA-Fc-MDNA413, Fc-MDNA132 (1:1 KiH), Fc-A11 (1:2), Fc-A11 (1:2), Fc-MDNA413 (1:2), Fc-MDNA413 (1:2), Fc4-MDNA413 (1:2), MDNA413-Fc (1:1 KIH), MDNA109-Fc (2:1), MDNA-109FEAA-Fc (2:1), Fc-MDNA109 (1:2), MDNA109FEAA-Fc-MDNA132 (2:1:1 KiH), MDNA413-Fc-MDNA132 (2:1:1 KiH), MDNA109FEAA-Fc-MDNA413 (2:1:2)—version 1, MDNA109FEAA-Fc-MDNA413 (2:1:2)-version 2, MDNA132-Fc-MDNA109 (1:1:1 KIH), MDNA413-Fc-MDNA109 (1:1:1 KIH), MDNA132-Fc-MDNA109FEAA (1:1:1 KIH), or MDNA109-Fc-MDNA413.


The present invention also provides a method of treating cancer comprising administering a bispecific IL-2 cytokine fusion as described herein.


In some embodiments, the cancer is selected from the group consisting of prostate cancer, ovarian cancer, breast cancer, endometrial cancer, multiple myeloma, melanoma, lymphomas, lung cancers including small cell lung cancer, kidney cancer, liver cancer, colon cancer, colorectal cancer, pancreatic cancer, gastric cancer, and brain cancer. In some embodiments, the cancer is colon cancer.


The present invention also provides a method of treating cancer comprising administering a combination treatment comprising:

    • (i) an anti-PD-1 antibody or inhibitor or an anti-PD-L1 antibody or inhibitor and
    • (ii) a bispecific IL-2 cytokine fusion as described herein.


In some embodiments, the anti-PD-1 antibody or inhibitor is selected from the group consisting of nivolumab, BMS-936558, MDX-1106, ONO-4538, AMP224, CT-011, and MK-3475(pembrolizumab), cemiplimab (REGN2810), SHR-1210 (CTR20160175 and CTR20170090), SHR-1210 (CTR20170299 and CTR20170322), JS-001 (CTR20160274), IBI308 (CTR20160735), BGB-A317 (CTR20160872) and a PD-1 antibody as recited in U.S. Patent Publication No. 2017/0081409 or in Table 38.


In some embodiments, the anti-PD-L1 antibody or inhibitor is selected from the group consisting of atezolizumab, avelumab, and Durvalumab.


In some embodiments, the cancer is selected from the group consisting of prostate cancer, ovarian cancer, breast cancer, endometrial cancer, multiple myeloma, melanoma, lymphomas, lung cancers including small cell lung cancer, kidney cancer, liver cancer, colon cancer, colorectal cancer, pancreatic cancer, gastric cancer, and brain cancer. In some embodiments, the cancer is colon cancer.


The present invention also provides pharmaceutical compositions comprising a bispecific IL-2 cytokine fusion as described herein, and a pharmaceutically acceptable carrier.


The present invention also provides a pharmaceutical composition comprising an anti-PD-1 antibody or inhibitor, a bispecific IL-2 cytokine fusion as described herein, and a pharmaceutically acceptable carrier.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1: Provides examples of IgG1, IgG2, IgG3, and IgG4 sequences.



FIG. 2: Provides exemplary H9-Fc fusion sequences.



FIG. 3: Exemplary oncolytic viruses.



FIG. 4: SDS-PAGE and HPLC analysis of MDNA132-Fc-MDNA109 (KIH) [Lot: T1848A12] post purification.



FIG. 5: SDS-PAGE and HPLC analysis of MDNA413-Fc-MDNA109 (KIH) [Lot: T1903A08] post purification.



FIG. 6: SDS-PAGE and HPLC analysis of MDNA109FEAA-Fc-MDNA413 (2:1:2) [Lot: T1914B08] post purification.



FIG. 7: Gating strategy. PBMCs were stimulated with IL-2 (25 nM) for 15 minutes prior to staining and analysis by flow-cytometry. Single lymphocytes were identified from which NK cells (CD3CD56+), CD8 T cells (CD3+CD8+), Tregs (CD3+CD4+CD25+FOXP3+) and non-Treg CD4 T cells (CD3+CD25CD4+FOXP3) were gated. Frequency of pSTAT5 positive cells was determined for each immune subset.



FIG. 8: Fluorescence minus one (FMO) controls. A) Following stimulation and gating strategy described in FIG. 4, all antibodies used to identify the different immune subsets were included except for pSTAT5 to provide an FMO control that could be used to create a pSTAT5+ gate for each population. B) FMOs for FOXP3 and CD25 were used to determine Treg population.



FIG. 9: Representative dose-response of IL-2, IL2-Fc, MDNA109 variants, and bi-specific superkines on pSTAT5 PBMCs. Graphs and EC50 values were calculated using non-linear regression. Each data point is a singlet.



FIG. 10: EC50 ratios of naïve CD8T cells (left) and NK cells (right) to Tregs.



FIG. 11: Survival BALB/c mice receiving different doses of MDNA132-Fc-MDNA109 (left) and MDNA413-Fc-MDNA109 (right).



FIG. 12: Survival C57Bl/6 mice receiving different doses of MDNA132-Fc-MDNA109 (left) and MDNA413-Fc-MDNA109 (right).



FIG. 13: MDNA132-Fc-MDNA109 (KIH) inhibits the growth of CT26 colon tumors. (a) Average tumor size in vehicle control and MDNA132-Fc-MDNA109 treatment groups. (b) growth of individual tumors in each groups. Each line represents a single tumor. CR=complete regression.



FIG. 14: CT26 tumor re-challenge. (a) Naïve untreated mice or mice previously cured of their CT26 tumors by MDNA132-Fc-MDNA109 were re-challenged on the opposite flank with CT26 tumor cells. (b) Overall survival to date.



FIG. 15: MDNA413-Fc-MDNA109 inhibits the growth of B16 F10 melanomas. Average tumor size of each treatment group (left) Individual tumor growth (right).



FIG. 16A-16B: Treatment of B16F10 tumor bearing mice with Vehicle (PBS) or MDNA109FEAA-Fc-MDNA413.



FIG. 17: Efficacy study in 4T1 model with MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 monotherapy and in combination with anti-PD1.



FIG. 18: B16F10 intra-tumoral injection with PBS or MDNA132-Fc-MDNA109 (KIH). Injections were performed on Day 14, 17, and 20 post-implant.



FIG. 19: CT26 intra-tumoral injection with MDNA109FEAA-Fc-MDNA413. Injections were performed on Day 11, 14, and 17.



FIG. 20: Activation of STAT6 signaling pathway in HEK Blue™ IL-4/IL-13 cells in the presence of human IL-4 (hIL4), human IL-13 (hIL13) or mouse IL-13 (mIL13).



FIG. 21: Inhibition of STAT6 signaling pathway in HEK Blue™ IL-4/IL-13 cells when cells were incubated with hIL13 together with MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, or Fc-MDNA413.



FIG. 22: Inhibition of STAT6 signaling pathway in HEK Blue™ IL-4/IL-13 cells when cells were incubated with mIL13 together with MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, or Fc-MDNA413.



FIG. 23: Inhibition of STAT6 signaling pathway in HEK Blue™ IL-4/IL-13 cells when cells were incubated with hIL4 together with MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, or Fc-MDNA413.



FIG. 24: Proliferation of CTLL2 cells when the cell culture was supplemented with the indicated proteins.



FIG. 25: Schematic and binding information for MDNA11.



FIG. 26: MDNA11 exhibits enhanced potency on naïve CD8+ T and NK cells & diminished activity on Tregs.



FIG. 27: MDNA11 inhibits tumor growth & induces a strong memory response as monotherapy & in combination with αCTLA4 in CT26 tumor model. CT26 tumor (˜60 mm3) bearing Balb/c mice were treated with MDNA11 (5 mg/kg Ix/week, 2 weeks) or Anti-CTLA4 (200 μg 2×/week, 2 weeks) by IP injection. Re-challenge experiment performed by implanting 2×106 CT26 cells in opposite flank (Day 49, Day 116 and Day 165), without further treatment. Control mice showed robust tumor growth at each re-challenge experiment (representative data for control shown).



FIG. 28: Development of long-term (gp70) specific CD8 T cells protected mice against CT26 tumors. Mice challenged with CT26 cells 5 days prior to euthanasia; control mice were age matched. Analysis by flow cytometry using APC anti-mouse CD3ε, anti-CD8 FITC clone KT15 and H-2Ld MuLV gp70 Tetramer-PE.



FIG. 29: MDNA11 delays the growth of B16F10 melanomas as monotherapy & in combination with TA99 MAb by promoting Tumor Infiltrating Lymphocytes (TILs). For efficacy study: 1) Treatment initiated 9-days post implant; average tumor size ˜15 mm3 and 2) MDNA11 (5 mg/kg, IP, Q.W); TA99 MAb (150 μg, IP, B.I.W.) for 3 cycle. For TILs analysis: 1) Average tumor size ˜50 mm3 when mice received a single dosing with MDNA11 (5 mg/kg; IP) and 2) TILs analyzed by flow cytometry. For TILs analyzed by flow cytometry: 1) CD8 T cells (CD45+CD8+), 2) NK cells (CD45+NK1.1), and 3) Tregs (CD4+CD25+FoxP3+).



FIG. 30: MDNA11 displays extended half-life in mice and NHP.



FIG. 31: MDNA11 induces proliferation & expansion of CD4+ T, CD8+ T and NK cells but not Tregs in NHP.



FIG. 32: Targeting immunologic “cold tumors” by modulation of TME with IL-2/IL-13 Bi-specific Superkines. “Cold” tumors are not responsive to check-point inhibitors because of a pro-tumoral TME: 1) Low CD8 & NK cell counts; high Treg counts and 2) High number of immune-suppressive myeloid cells (i.e. TAM & MDSC).



FIG. 33: Bi-specific MDNA109FEAA-Fc-MDNA413 Superkine induces Th1 and reduces Th2 immune responses.



FIG. 34: Enhanced potency on CD8+ T and NK cells; diminished activity on Tregs-Strong Th1 response-MDNA109FEAA-Fc (MDNA11) and MDNA109FEAA-Fc-MDNA413 inhibits hIL-4/hIL-13 induced signaling in HEK-Blue reporter cells-Suppression of Th2 response.



FIG. 35: ELISA assay measuring binding of Fc-hIL13 to various IL13 and IL4 receptors, including human IL13Rα2, mouse IL13Rα2, human IL13Rα1, mouse IL13Rα1, human IL4R and mouse IL4R.



FIG. 36A: ELISA assay measuring binding of MDNA132-Fc-MDNA109 binding to human IL13Rα2. FIG. 36B: ELISA assay measuring binding of MDNA132-Fc-MDNA109 binding to mouse IL13Rα2.



FIG. 37A: ELISA assay measuring binding of various fusion protein comprising MDNA413 to human IL13Rα1. FIG. 37B: ELISA assay measuring binding of various fusion protein comprising MDNA413 to mouse IL13Rα1.



FIG. 38A-38B: Sensograms of the biacore analysis for Fc-MDNA132 (1:1 KIH) as ligand and receptors, IL13Rα1 and IL13Rα2 as antigens.



FIG. 39A-39B: Sensograms of the biacore analysis for Fc-MDNA413 (1:2) as ligand and receptors, IL13Rα1 as antigens.



FIG. 40A-40B: Sensograms of the biacore analysis for Fc-MDNA413 (1:2) as ligand and receptors, IL13Rα2 as antigens.



FIG. 41A-41D: Binding affinity to IL13Rα1 by SPR (MCK, multi-cycle kinetics).



FIG. 42A-42B: pSTAT5 Signaling Assay in Human PBMC.



FIG. 43A-43C: Measurement of pSTAT6 signaling.



FIG. 44A-44B: Macrophage polarization assay.



FIG. 45A-45E: Binding affinity to IL13Rα1 and IL13Rα2 by SPR (MCK, multi-cycle kinetics).



FIG. 46A-46C: Inhibition of tumor growth in a CT26 colon cancer model by bispecific construct MDNA132-Fc-MDNA109.



FIG. 47: HEK Blue IL2 Assay.



FIG. 48: CTTL-2 Assay.



FIG. 49A-49B: HEK Blue IL4 Assay.



FIG. 50A-50B: HEK Blue IL13 Assay.



FIG. 51A-51D: TF-1 Assay.



FIG. 52A-52B: pSTAT5 Assay in human PBMCs.



FIG. 53A-53D: SPR Study.



FIG. 54A-54J: Bispecific sequence information for various construct embodiments.



FIG. 55: Directed evolution combined with yeast display results in tunable superkines.



FIG. 56: MDNA109 is an engineered version of human IL-2 showing enhanced agonist activity.



FIG. 57: Introduction to long-acting IL-2 superkines.



FIG. 58: MDNA11 is a ‘beta-only’ superkine with uniquely enhanced affinity for CD122.



FIG. 59: MDNA11 preferentially stimulates immune effector cells.



FIG. 60: MDNA11 shows monotherapy anti-tumor efficacy and combination effect with anti-PD1 in MC38 tumor model.



FIG. 61: MDNA11+ anti-CTLA4 induces tumor clearance, protects against, re-challenges, and promotes antigen-specific CD8 T-cells.



FIG. 62: MDNA11 induces durable and sustained proliferation and expansion of immune effector cells but not Tregs in NHP.



FIG. 63: Superkine targeted with antibody (STAb) enhances accumulation in tumors.



FIG. 64: STAb overcomes checkpoint resistance and ‘cold’ tumors.



FIG. 65: Role of IL-4 and IL-13 receptors in cancer.



FIG. 66: MDNA132 is an engineered version of human IL-13 targeting tumor specific antigen.



FIG. 67: MDNA132 plays a role in localizing T-cell engager and checkpoint inhibitor to tumors.



FIG. 68: MDNA413 is an engineered version of human IL-13 showing antagonist activity.



FIG. 69: Fc-MDNA413 inhibits IL-4 and IL-13 induced signaling and function.



FIG. 70: Mechanism of action of dual specific cytokine (DUCK Cancer) MDNA109FEAA-Fc-MDNA413.



FIG. 71: TF-1 Assay.



FIG. 72: SPR Study.



FIG. 73A-73B: SPR Study.



FIG. 74A-74B: SPR Study.



FIG. 75A-75D: SPR Study.



FIG. 76A-76E: Inhibition of IL-4 induced M2 polarization.



FIG. 77A-77E: Inhibition of IL-13 induced M2 polarization.





DETAILED DESCRIPTION

In order for the present disclosure to be more readily understood, certain terms and phrases are defined below as well as throughout the specification.


Definitions

All references cited herein are incorporated by reference in their entirety as though fully set forth. Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Singleton et al., Dictionary of Microbiology and Molecular Biology 3rd ed., J. Wiley & Sons (New York, NY 2001); March, Advanced Organic Chemistry Reactions, Mechanisms and Structure 5th ed., J. Wiley & Sons (New York, NY 2001); and Sambrook and Russell, Molecular Cloning: A Laboratory Manual 3rd ed., Cold Spring harbor Laboratory Press (Cold Spring Harbor, NY 2001), provide one skilled in the art with a general guide to many terms used in the present disclosure. As appropriate, procedures involving the use of commercially available kits and reagents are generally carried out in accordance with manufacturer defined protocols and/or parameters unless otherwise noted.


As used herein, “IL-2” means wild-type IL-2, whether native or recombinant. Mature human IL-2 occurs as a 133 amino acid sequence (less the signal peptide, consisting of an additional 20 N-terminal amino acids), as described in Fujita, et. al., PNAS USA, 80, 7437-7441 (1983). The amino acid sequence of human IL-2 (SEQ ID NO:1; full length) is found in Genbank under accession locator NP_000577.2. The amino acid sequence of mature human IL-2 is depicted in SEQ ID NO:2 (human wild-type mature; position numbering of the substitutions is based on this sequence). The murine (Mus musculus) IL-2 amino acid sequence is found in Genbank under accession locator (SEQ ID NO:3). The amino acid sequence of mature murine IL-2 is depicted in SEQ ID NO:4.









SEQ ID NO: 1


MYRMQLLSCIALSLALVTNSAPTSSSTKKTQLQLEHLLLDLQMILNGIN





NYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNLAQSKNF





HLRPRDLISNINVIVLELKGSETTFMCEYADETATIVEFLNRWITFCQS





IISTLT





SEQ ID NO: 2


APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKK





ATELKHLQCLEEELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKG





SETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 3


MYSMQLASCVTLTLVLLVNSAPTSSSTSSSTAEAQQQQQQQQQQQQHLE





QLLMDLQELLSRMENYRNLKLPRMLTFKFYLPKQATELKDLQCLEDELG





PLRHVLDLTQSKSFQLEDAENFISNIRVTVVKLKGSDNTFECQFDDESA





TVVDFLRRWIAFCQSIISTSPQ





SEQ ID NO: 4


APTSSSTSSSTAEAQQQQQQQQQQQQHLEQLLMDLQELLSRMENYRNLK





LPRMLTFKFYLPKQATELKDLQCLEDELGPLRHVLDLTQSKSFQLEDAE





NFISNIRVTVVKLKGSDNTFECQFDDESATVVDFLRRWIAFCQSIISTS





PQ






As used herein, “IL-2 mutein” means an IL-2 polypeptide wherein specific substitutions to the interleukin-2 protein have been made. The IL-2 muteins are characterized by amino acid insertions, deletions, substitutions and modifications at one or more sites in or at the other residues of the native IL-2 polypeptide chain. In accordance with this disclosure, any such insertions, deletions, substitutions and modifications result in an IL-2 mutein that retains the IL-2RD binding activity. Exemplary muteins can include substitutions of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids.


Muteins also include conservative modifications and substitutions at other positions of IL-2 (i.e., those that have a minimal effect on the secondary or tertiary structure of the mutein). Such conservative substitutions include those described by Dayhoff in The Atlas of Protein Sequence and Structure 5 (1978), and by Argos in EMBO J., 8:779-785 (1989). For example, amino acids belonging to one of the following groups represent conservative changes: Group I: ala, pro, gly, gln, asn, ser, thr; Group II: cys, ser, tyr, thr; Group III: val, ile, leu, met, ala, phe; Group IV: lys, arg, his; Group V: phe, tyr, trp, his; and Group VI: asp, glu.


“Numbered in accordance with IL-2” means identifying a chosen amino acid with reference to the position at which that amino acid normally occurs in the mature sequence of wild type IL-2, for example R81 refers to the eighty-first amino acid, arginine, that occurs in SEQ ID NO:2. L80 refers to the eightieth amino acid, leucine, that occurs in SEQ ID NO:2. L85 refers to the eighty-fifth amino acid, leucine, that occurs in SEQ ID NO:2. 186 refers to the eighty-sixth amino acid, isoleucine, that occurs in SEQ ID NO:2. 192 refers to the ninety-second amino acid, isoleucine, that occurs in SEQ ID NO:2. F42 refers to the forty-second amino acid, phenylalanine, that occurs in SEQ ID NO:2. K43 refers to the forty-third amino acid, lysine, that occurs in SEQ ID NO:2.


As used herein, the abbreviations for the genetically encoded L-enantiomeric amino acids used in the disclosure methods are conventional and are as follows in Table 1.









TABLE 1







Amino acid abbreviations












One-Letter
Common



Amino Acid
Symbol
Abbreviation







Alanine
A
Ala



Arginine
R
Arg



Asparagine
N
Asn



Aspartic acid
D
Asp



Cysteine
C
Cys



Glutamine
Q
Gln



Glutamic acid
E
Glu



Glycine
G
Gly



Histidine
H
His



Isoleucine
I
Ile



Leucine
L
Leu



Lysine
K
Lys



Methionine
M
Met



Phenylalanine
F
Phe



Proline
P
Pro



Serine
S
Ser



Threonine
T
Thr



Tryptophan
W
Trp



Tyrosine
Y
Tyr



Valine
V
Val










“Hydrophilic Amino Acid” refers to an amino acid exhibiting a hydrophobicity of less than zero according to the normalized consensus hydrophobicity scale of Eisenberg et al., 1984, J. Mol. Biol. 179: 125-142. Genetically encoded hydrophilic amino acids include Thr (T), Ser (S), His (H), Glu (E), Asn (N), Gln (Q), Asp (D), Lys (K) and Arg (R).


The term “cell types having the IL-2Rαγ receptor” means the cells known to have this receptor type, i.e., T cells, activated T cells, B cells, activated monocytes, and activated NK cells. The term “cell types having the IL-2Rβγ receptor” means the cells known to have that receptor type, i.e., B cells, resting monocytes, and resting NK cells.


The term “identity,” as used herein in reference to polypeptide or DNA sequences, refers to the subunit sequence identity between two molecules. When a subunit position in both of the molecules is occupied by the same monomeric subunit (i.e., the same amino acid residue or nucleotide), then the molecules are identical at that position. The similarity between two amino acid or two nucleotide sequences is a direct function of the number of identical positions. In general, the sequences are aligned so that the highest order match is obtained. If necessary, identity can be calculated using published techniques and widely available computer programs, such as the GCS program package (Devereux et al., Nucleic Acids Res. 12:387, 1984), BLASTP, BLASTN, FASTA (Atschul et al., J. Molecular Biol. 215:403, 1990). Sequence identity can be measured using sequence analysis software such as the Sequence Analysis Software Package of the Genetics Computer Group at the University of Wisconsin Biotechnology Center (1710 University Avenue, Madison, Wis. 53705), with the default parameters thereof.


The terms “polypeptide,” “protein” or “peptide” refer to any chain of amino acid residues, regardless of its length or post-translational modification (e.g., glycosylation or phosphorylation).


In the event the mutant IL-2 polypeptides of the disclosure are “substantially pure,” they can be at least about 60% by weight (dry weight) the polypeptide of interest, for example, a polypeptide containing the mutant IL-2 amino acid sequence. For example, the polypeptide can be at least about 75%, about 80%, about 85%, about 90%, about 95% or about 99%, by weight, the polypeptide of interest. Purity can be measured by any appropriate standard method, for example, column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis.


An “agonist” is a compound that interacts with a target to cause or promote an increase in the activation of the target.


A “partial agonist” is a compound that interacts with the same target as an agonist but does not produce as great a magnitude of a biochemical and/or physiological effect as the agonist, even by increasing the dosage of the partial agonist.


A “superagonist” (also referred to as a “superkine”) is a type of agonist that is capable of producing a maximal response greater than the endogenous agonist for the target receptor, and thus has an efficacy of more than 100%.


“Operably linked” is intended to mean that the nucleotide sequence of interest (i.e., a sequence encoding an IL-2 mutein) is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). “Regulatory sequences” include promoters, enhancers, and other expression control elements (e.g., polyadenylation signals). See, for example, Goeddel (1990) in Gene Expression Technology: Methods in Enzymology 185 (Academic Press, San Diego, Calif). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cells and those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, and the like. The expression constructs of the invention can be introduced into host cells to thereby produce the human IL-2 muteins disclosed herein or to produce biologically active variants thereof.


The terms “host cell” and “recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell but are still included within the scope of the term as used herein.


As used herein, the terms “transformation” and “transfection” refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, particle gun, or electroporation.


As used herein, the term “pharmaceutically acceptable carrier” includes, but is not limited to, saline, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Supplementary active compounds (e.g., antibiotics) can also be incorporated into the compositions.


As used herein, the term “anti-PD-1 antibody” refers to any antibody that binds to PD-1, including inhibitory antibodies. An “anti-PD-1 inhibitor” refers to an inhibitor that binds to and inhibits PD-1. Such anti-PD-1 antibodies and/or inhibitors include but are not limited to nivolumab, BMS-936558, MDX-1106, ONO-4538, AMP224, CT-011, and MK-3475, among others.


As used herein, the terms “cancer” (or “cancerous”), “hyperproliferative,” and “neoplastic” to refer to cells having the capacity for autonomous growth (i.e., an abnormal state or condition characterized by rapidly proliferating cell growth). Hyperproliferative and neoplastic disease states may be categorized as pathologic (i.e., characterizing or constituting a disease state), or they may be categorized as non-pathologic (i.e., as a deviation from normal but not associated with a disease state). The terms are meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. “Pathologic hyperproliferative” cells occur in disease states characterized by malignant tumor growth. Examples of non-pathologic hyperproliferative cells include proliferation of cells associated with wound repair. The terms “cancer” or “neoplasm” are used to refer to malignancies of the various organ systems, including those affecting the lung, breast, thyroid, lymph glands and lymphoid tissue, reproductive systems, gastrointestinal organs, and the genitourinary tract, as well as to adenocarcinomas which are generally considered to include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumors, non-small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus. Cancers generally can include prostate cancer, ovarian cancer, breast cancer, endometrial cancer, multiple myeloma, melanoma, lymphomas, lung cancers including small cell lung cancer, kidney cancer, colorectal cancer, pancreatic cancer, gastric cancer, and brain cancer.


The term “carcinoma” is art-recognized and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas. An “adenocarcinoma” refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.


As used herein, the term “hematopoietic neoplastic disorders” refers to diseases involving hyperplastic/neoplastic cells of hematopoietic origin, e.g., arising from myeloid, lymphoid or erythroid lineages, or precursor cells thereof. Preferably, the diseases arise from poorly differentiated acute leukemias (e.g., erythroblastic leukemia and acute megakaryoblastic leukemia). Additional exemplary myeloid disorders include, but are not limited to, acute promyeloid leukemia (APML), acute myelogenous leukemia (AML) and chronic myelogenous leukemia (CML) (reviewed in Vaickus, L. (1991) Crit Rev. in Oncol./Hemotol. 11:267-97); lymphoid malignancies include, but are not limited to acute lymphoblastic leukemia (ALL) which includes B-lineage ALL and T-lineage ALL, chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), hairy cell leukemia (HLL) and Waldenstrom's macroglobulinemia (WM). Additional forms of malignant lymphomas include, but are not limited to non-Hodgkin lymphoma and variants thereof, peripheral T cell lymphomas, adult T cell leukemia/lymphoma (ATL), cutaneous T cell lymphoma (CTCL), large granular lymphocytic leukemia (LGF), Hodgkin's disease and Reed-Stemberg disease.


As used herein, the terms “treatment,” “treating,” and the like, refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease. “Treatment,” as used herein, covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject predisposed to the disease or at risk of acquiring the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease. A therapeutically effective amount can be an amount that reduces tumor number, tumor size, and/or increases survival.


The terms “individual,” “subject,” and “patient” are used interchangeably herein, and refer to a mammal, including, but not limited to, human and non-human primates, including simians and humans; mammalian sport animals (e.g., horses); mammalian farm animals (e.g., sheep, goats, etc.); mammalian pets (dogs, cats, etc.); and rodents (e.g., mice, rats, etc.).


The terms “pharmaceutically acceptable” and “physiologically acceptable” mean a biologically acceptable formulation, gaseous, liquid or solid, or mixture thereof, suitable for one or more routes of administration, in vivo delivery or contact. A “pharmaceutically acceptable” or “physiologically acceptable” composition is a material that is not biologically or otherwise undesirable, e.g., the material may be administered to a subject without causing substantial undesirable biological effects. Thus, such a pharmaceutical composition may be used, for example in administering an IL-2 mutein to a subject. In particular, an IL-2 mutein comprising the substitutions L80F, R81D, L85V, I86V, and I92F is administered in combination with anti-PD-1 to a subject with cancer. In some embodiments, the IL-2 mutein administered further comprises a substitution at position F42A. In some embodiments, the IL-2 administered mutein further comprises a substitution at position K43N.


The phrase a “unit dosage form” as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity optionally in association with a pharmaceutical carrier (excipient, diluent, vehicle or filling agent) which, when administered in one or more doses, produces a desired effect (e.g., prophylactic or therapeutic effect). In some embodiments, the therapeutic effect is to reduce tumor number. In some embodiments, the therapeutic effect is to reduce tumor size. In some embodiments, the therapeutic effect is to increase survival.


In some embodiments, unit dosage forms may be within, for example, ampules and vials, including a liquid composition, or a composition in a freeze-dried or lyophilized state; a sterile liquid carrier, for example, can be added prior to administration or delivery in vivo. Individual unit dosage forms can be included in multi-dose kits or containers. IL-2 muteins in combination with anti-PD-1 antibodies, and pharmaceutical compositions thereof can be packaged in a single or multiple unit dosage form for ease of administration and uniformity of dosage.


A “therapeutically effective amount” will fall in a relatively broad range determinable through experimentation and/or clinical trials. For example, for in vivo injection, e.g., injection directly into the tissue or vasculature of a subject (for example, liver tissue or veins). Other effective dosages can be readily established by one of ordinary skill in the art through routine trials establishing dose response curves.


An “effective amount” or “sufficient amount” refers to an amount providing, in single or multiple doses, alone or in combination, with one or more other compositions (therapeutic agents such as a drug), treatments, protocols, or therapeutic regimens agents (including, for example, vaccine regimens), a detectable response of any duration of time (long or short term), an expected or desired outcome in or a benefit to a subject of any measurable or detectable degree or for any duration of time (e.g., for minutes, hours, days, months, years, or cured).


The doses of an “effective amount” or “sufficient amount” for treatment (e.g., to ameliorate or to provide a therapeutic benefit or improvement) typically are effective to provide a response to one, multiple or all adverse symptoms, consequences or complications of the disease, one or more adverse symptoms, disorders, illnesses, pathologies, or complications, for example, caused by or associated with the disease, to a measurable extent, although decreasing, reducing, inhibiting, suppressing, limiting or controlling progression or worsening of the disease is also a satisfactory outcome. In some embodiments, the effective amount is an amount sufficient to reduce tumor number. In some embodiments, the effective amount is an amount sufficient to reduce tumor size. In some embodiments, the effective amount is an amount sufficient to increase survival.


“Prophylaxis” and grammatical variations thereof mean a method in which contact, administration or in vivo delivery to a subject is prior to disease. Administration or in vivo delivery to a subject can be performed prior to development of an adverse symptom, condition, complication, etc. caused by or associated with the disease. For example, a screen (e.g., genetic) can be used to identify such subjects as candidates for the described methods and uses, but the subject may not manifest the disease. Such subjects therefore include those screened positive for an insufficient amount or a deficiency in a functional gene product (protein), or producing an aberrant, partially functional or non-functional gene product (protein), leading to disease; and subjects screening positive for an aberrant, or defective (mutant) gene prod]uct (protein) leading to disease, even though such subjects do not manifest symptoms of the disease.


I. DETAILED DESCRIPTION

Described herein are bispecific IL-2 cytokine fusions, which comprise an IL-2 mutein fused to a second cytokine. Also described herein IL-2 muteins comprising the substitutions L80F, R81D, L85V, I86V, and I92F, which have an increased binding capacity for IL-2RD receptor, which can be included in the bispecific IL-2 cytokine fusions. Also described herein are uses of bispecific IL-2 cytokine fusions for use in monotherapies as well as in combination treatments with anti-PD-1 antibodies. In some embodiments, the IL-2 mutein comprising L80F, R81D, L85V, I86V and I92F, numbered in accordance with wild-type human IL-2 (SEQ ID NO:2; wild-type hIL-2) is referred to as H9. Such IL-2 muteins find use, for example, when combined with anti-PD-1 antibodies for the treatment of cancer. Also provided are nucleic acids encoding such IL-2 muteins, methods of making such IL-2 muteins, pharmaceutical compositions that include such IL-2 muteins and methods of treatment using such IL-2 muteins.


A. IL-2 Muteins for Use in Bispecific IL-2 Cytokine Fusions


The substituted amino acid residue(s) can be, but are not necessarily, conservative substitutions, which typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. These mutations can be at amino acid residues that contact the IL-2Rβ and/or the IL-2Rγ.


More specifically, a mutation (whether conservative or non-conservative, by way of addition(s) or deletion(s)) can be made at one or more of positions. For example, the mutation can be: I24V, P65H, Q74R, Q74H, Q74N, Q74S, L80F, L80V, R81I, R81T, R81D, L85V, I86V, I89V, I92F, V93I. The sequences of exemplary IL-2 muteins are as follows: 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO: 9; B11 SEQ ID NO: 10; C5 SEQ ID NO:11; D10 SEQ ID NO: 12; E10 SEQ ID NO:13; G8 SEQ ID NO: 14; H4 SEQ ID NO: 15; and H9 SEQ ID NO: 16.


In some embodiments, the substitutions in the IL-2 mutein comprise L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the substitutions in the IL-2 mutein comprise F42A, L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the substitutions in the IL-2 mutein comprise F42A, Y45A, L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the substitutions in the IL-2 mutein comprise F42A, E62A, L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the substitutions in the IL-2 mutein comprise F42A, Y45A, E62A, L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the substitutions in the IL-2 mutein comprise E62A, L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the substitutions in the IL-2 mutein comprise Y45A, E62A, L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the substitutions in the IL-2 mutein comprise Y45A and E62A, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2.


In some embodiments, the substitutions in the IL-2 mutein that lead to increased and/or enhanced IL-2Rβ binding include L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, an IL-2 mutein for use in the invention comprises L80F, R81D, L85V, I86V, and I92F and exhibits increased IL-2Rβ binding. In some embodiments, an IL-2 mutein for use in the invention further comprises a substitution at position F42A. In some embodiments, the IL-2 mutein for use in the invention further comprises a substitution at position K43N. In some embodiments, the mutein comprises substitutions L80F, R81D, L85V, I86V, and I92F, and one or more substitutions selected from the group consisting of F42A, Y45A, and E62A, all as compared to wild-type human IL-2 (SEQ ID NO:2).


In some embodiments, the amino acid substitutions increasing IL-2Rβ binding affinity include: L80F, R81D, L85V, I86V, and I92F. In some embodiments, the amino acid substitutions that increase IL-2Rβ binding affinity include: L80F, R81D, L85V, 186V, and I92F.


In some embodiments, the subject IL-2 mutein having a greater binding affinity for IL-2Rβ as compared to wild-type human IL-2, includes the amino acid substitutions L80F, R81D, L85V, I86V, and I92F. In some embodiments, the IL-2 mutein has the amino acid sequence: APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEE ELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWI TFCQSIISTLT (SEQ ID NO:5; H9 as used in Example 1).


In some embodiments, the IL-2 mutein has increased capabilities to stimulate one or more signaling pathways that are dependent on IL-2Rβ/IL-2Rγc heterodimerization. In some embodiments, the subject IL-2 mutein has an enhanced capability to stimulate STAT5 phosphorylation in an IL-2Rβ+ cell as compared to wild-type human IL-2. In some embodiments, the IL-2 mutein stimulates STAT5 phosphorylation in an IL-2Rβ+ cell at a level that is 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more of the level that wild-type IL-2 stimulates STAT5 phosphorylation in the same cell. In some embodiments, the IL-2 mutein stimulates STAT5 phosphorylation in an IL-2Rβ+ cell at a level that is 105%, 110%, 115%, 120%, 125%, 130%, 135%, 140%, 145%, 150%, 155%, 160%, 165%, 170%, 175%, 180%, 185%, 190%, 195% or more as compared to the level that wild-type IL-2 stimulates STAT5 phosphorylation in the same cell. In some embodiments, the IL-2Rβ+ cell is a T cell. In particular embodiments, the T cell is a CD8+ T cell. In some embodiments, the CD8+ T cell is a freshly isolated CD8+ T cell. In other embodiments, the CD8+ T cell T cell is an activated CD8+ T cell. In other embodiments, the IL-2Rβ+ cell is a natural killer (NK) cell. In some embodiments, the IL-2 mutein comprises substitutions L80F, R81D, L85V, 186V, and I92F, as compared to wild-type human IL-2 (SEQ ID NO:2).


In some embodiments, the mutein has an enhanced capability to stimulate ERK1/ERK2 signaling in an IL-2Rβ+ cell as compared to wild-type human IL-2. In some embodiments, the IL-2 mutein stimulates pERK1/ERK2 signaling in an IL-2Rβ+ cell at a level that is 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or more of the level that wild-type IL-2 stimulates pERK1/ERK2 signaling in the same cell. In some embodiments, the IL-2 mutein stimulates pERK1/ERK2 phosphorylation in an IL-2Rβ+ cell at a level that is 105%, 110%, 115%, 120%, 125%, 130%, 135%, 140%, 145%, 150%, 155%, 160%, 165%, 170%, 175%, 180%, 185%, 190%, 195% or more as compared to the level that wild-type IL-2 stimulates pERK1/ERK2 phosphorylation in the same cell. In some embodiments, the IL-2Rβ+ cell is a T cell. In particular embodiments, the T cell is a CD8+ T cell.


In some embodiments, the CD8+ T cell is a freshly isolated CD8+ T cell. In other embodiments, the CD8+ T cell T cell is an activated CD8+ T cell. In other embodiments, the IL-2Rβ+ cell is a natural killer (NK) cell. In some embodiments, the IL-2 mutein comprises substitutions L80F, R81D, L85V, I86V, and I92F, as compared to wild-type human IL-2 (SEQ ID NO:2). STAT5 and ERK1/2 signaling can be measured, for example, by phosphorylation of STAT5 and ERK1/2 using any suitable method known in the art. For example, STAT5 and ERK1/2 phosphorylation can be measured using antibodies specific for the phosphorylated version of these molecules in combination with flow cytometry analysis as described herein. In some embodiments, the mutein has an enhanced capability to stimulate PI 3-kinase signaling in a IL-2Rβ+ cell as compared to wild-type human IL-2. In some embodiments, the IL-2 mutein stimulates PI 3-kinase signaling in an IL-2Rβ+ cell at a level that is 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or less of the level that wild-type IL-2 stimulates PI 3-kinase signaling in the same cell. In some embodiments, the IL-2 mutein stimulates PI 3-kinase signaling in an IL-2Rβ+ cell at a level that is 105%, 110%, 115%, 120%, 125%, 130%, 135%, 140%, 145%, 150%, 155%, 160%, 165%, 170%, 175%, 180%, 185%, 190%, 195% or more as compared to the level that wild-type IL-2 stimulates PI 3-kinase signaling phosphorylation in the same cell. In some embodiments, the IL-2Rβ+ cell is a T cell. In particular embodiments, the T cell is a CD8+ T cell. In some embodiments, the CD8+ T cell T cell is an activated CD8+ T cell. In other embodiments, the IL-2Rβ+ cell is a natural killer (NK) cell. In some embodiments, the IL-2 mutein comprises substitutions L80F, R81D, L85V, 186V, and I92F, as compared to wild-type human IL-2 (SEQ ID NO:2). PI3-kinase signaling can be measured using any suitable method known in the art. For example, PI 3-kinase signaling can be measured using antibodies that are specific for phospho-S6 ribosomal protein in conjunction with flow cytometry analysis as described herein.


In some embodiments the IL-2 mutein is a stimulator of IL-2 and/or IL-15 STAT5 phosphorylation in CD8+ T cells. In some embodiments, the mutein is a promoter of IL-2 and/or IL-15 induced proliferation of CD8+ T cells. In some embodiments, the mutein is a stimulator of IL-2 dependent, TCR-induced cell proliferation. In some embodiments, the IL-2 mutein comprises substitutions L80F, R81D, L85V, I86V, and I92F, as compared to wild-type human IL-2 (SEQ ID NO:2).


IL-2 promotes Th1, Th9, and Treg T cell differentiation and inhibits Th17 differentiation. Therefore, without being bound by any particular theory of operation, it is believed that IL-2 muteins that function as IL-2 superagonists are capable of promoting Th1, Th9, and/or Treg cell differentiation or inhibiting Th17 cell differentiation. In some embodiments, the IL-2 mutein is a promoter of IL-2 dependent Th1, Th9 and/or Treg differentiation. In some embodiments, the mutein is an inhibitor of Th17 differentiation. In some embodiments, the IL-2 mutein comprises substitutions L80F, R81D, L85V, I86V, and I92F, as compared to wild-type human IL-2 (SEQ ID NO:2).


In some embodiments, the IL-2 mutein signals less and/or independently of CD25 (for example, has reduced or ablated CD25 binding) as compared to wild-type human IL-2. In some embodiments the reduced and/or independent signaling with regard to CD25 allows for preferential activation of effector T-cells while limiting the stimulation of Tregs. In some embodiments the reduced and/or independent signaling with regard to CD25 allows for reduced toxicity. In some embodiments, the mutein comprises substitutions L80F, R81D, L85V, I86V, and I92F, and one or more substitutions selected from the group consisting of F42A, Y45A, and E62A, all as compared to wild-type human IL-2 (SEQ ID NO:2).


In some embodiments, the IL-2 mutein is capable of increasing and/or restoring responsiveness to anergic NK cells. In some embodiments, the IL-2 mutein is capable of increasing and/or restoring responsiveness to anergic NK cells in the tumor microenvironment. In some embodiments, the IL-2 mutein comprises substitutions L80F, R81D, L85V, 186V, and I92F, as compared to wild-type human IL-2 (SEQ ID NO:2).


In some embodiments the mutein is an inhibitor an inhibitor of IL-2 dependent activation of natural killer (NK) cells. IL-2 activation of NK cells can be measured by any suitable method known in the art, for example, by measuring IL-2 induced CD69 expression and/or cytotoxicity, as described herein.


In some embodiments, an increase in IL-2Rβ binding affinity is any binding affinity for IL-2Rβ that is greater than the wild-type human IL-2 binding affinity for IL-2Rβ. In some embodiments, the binding affinity is a 2-fold, 5-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 120-fold, 150-fold, 170-fold, 190-fold, 200-fold, 220-fold, 240-fold or more increase in binding affinity for IL-2Rβ as compared to the wild-type human IL-2 binding affinity for IL-2Rβ.


In some embodiments, an increase in binding capacity for IL-2Rβ is any binding capacity for IL-2Rβ that is greater than the wild-type human IL-2 binding capacity for IL-2Rβ. In some embodiments, the binding capacity is a 2-fold, 5-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 120-fold, 150-fold, 170-fold, 190-fold, 200-fold, 220-fold, 240-fold or more increase in binding capacity for IL-2Rβ as compared to the wild-type human IL-2 binding capacity for IL-2Rβ.


In some embodiments, the subject IL-2 mutein having a greater binding affinity for IL-2Rβ as compared to wild-type human IL-2 also exhibits reduced binding to CD25 and includes the amino acid substitutions F42A, L80F, R81D, L85V, I86V, and I92F. In some embodiments, the reduce binding affinity is about 220-fold, i.e., from about Kd of 6.6 nM for wild-type human IL-2 to about 1.4 μM for the mutein comprising F42A, L80F, R81D, L85V, I86V, and I92F. In some embodiments, the IL-2 mutein has the amino acid sequence:









(SEQ ID NO: 6; also referred to as H9-F42A)


APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKK


ATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG


SETTFMCEYADETATIVEFLNRWITFCQSIISTLT.






In some embodiments, the subject IL-2 mutein having a greater binding affinity for IL-2Rβ as compared to wild-type human IL-2 also exhibits reduced binding to CD25 and includes the amino acid substitutions K43N, L80F, R81D, L85V, I86V, and I92F. In some embodiments, the reduce binding affinity is due to allowing for glycosylation at position 43 with the K43N substitution. By substituting lysine for asparagine (K43N), CD25 binding is reduced and/or ablated in the IL-2 mutein comprising the amino acid substitutions K43N, L80F, R81D, L85V, 186V, and I92F. In some embodiments, the IL-2 mutein has the amino acid sequence:









(SEQ ID NO: 7; also referred to as H9-K43N)


APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFNFYMPKK


ATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG


SETTFMCEYADETATIVEFLNRWITFCQSIISTLT.






In some embodiments, a reduction in binding affinity for CD25 is any binding affinity for CD25 that is less than the wild-type human IL-2 binding affinity. In some embodiments, the binding affinity is a 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 60-fold, 70-fold, 80-fold, 90-fold, 100-fold, 120-fold, 150-fold, 170-fold, 190-fold, 200-fold, 220-fold, 240-fold or more decrease in binding affinity for CD25 as compared to the wild-type human IL-2 binding affinity for CD25.


In some embodiments, the subject IL-2 mutein having a greater binding affinity for IL-2Rβ and a reduced binding affinity for CD25 as compared to wild-type human IL-2 includes the amino acid substitutions F42A, Y45A L80F, R81D, L85V, I86V, and I92F. In some embodiments, the IL-2 mutein has the amino acid sequence:









(SEQ ID NO: 8; H9-F42A/Y45A; H9-FYAA)


APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFAMPKK


ATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG


SETTFMCEYADETATIVEFLNRWITFCQSIISTLT.






In some embodiments, the subject IL-2 mutein having a greater binding affinity for IL-2Rβ and a reduced binding affinity for CD25 as compared to wild-type human IL-2 includes the amino acid substitutions F42A, E62A L80F, R81D, L85V, I86V, and I92F. In some embodiments, the IL-2 mutein has the amino acid sequence:









(SEQ ID NO: 9; H9-F42A/E62A; H9-FEAA)


APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKK


ATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG


SETTFMCEYADETATIVEFLNRWITFCQSIISTLT.






In some embodiments, the subject IL-2 mutein having a greater binding affinity for IL-2Rβ and a reduced binding affinity for CD25 as compared to wild-type human IL-2 includes the amino acid substitutions F42A, Y45A, E62A, L80F, R8TD, L85V, 186V, and I92F. In some embodiments, the IL-2 mutein has the amino acid sequence:









(SEQ ID NO: 10; H9-F42A/Y45A/E62A; H9-FYEAAA)


APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFAMPKK


ATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG


SETTFMCEYADETATIVEFLNRWITFCQSIISTLT.






In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO: 10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein sequence is 95% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO: 10. In some embodiments, the IL-2 mutein sequence is 98% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO: 10. In some embodiments, the IL-2 mutein sequence is 99% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO: 10.


Further exemplary IL-2 sequences are provided in the table below.









TABLE 2







List of Exemplary IL-2 Muteins








Amino Acid Sequences



SEQ ID NO:



(Information)
Amino acid sequence





SEQ ID NO: 6
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKL


(also referred to as H9-F42A)
TRMLTAKFYMPKKATELKHLQCLEEELKPLEEVLN



LAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCE



YADETATIVEFLNRWITFCQSIISTLT (





(SEQ ID NO: 7 
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKL


(also referred to as H9-K43N)
TRMLTFNFYMPKKATELKHLQCLEEELKPLEEVLN



LAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCE



YADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 8
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKL


(H9-F42A/Y45A; H9-FYAA)
TRMLTAKFAMPKKATELKHLQCLEEELKPLEEVLN



LAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCE



YADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 9)
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKL


(H9-F42A/E62A; H9-FEAA)
TRMLTAKFYMPKKATELKHLQCLEEALKPLEEVLN



LAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCE



YADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 10;
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKL


H9-F42A/Y45A/E62A; H9-FYEAAA).
TRMLTAKFAMPKKATELKHLQCLEEALKPLEEVLN



LAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCE



YADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 20
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLARSKNFHLRPRD



LISNINVIVLELKGSETTFMCEYADETATIVEFLNRWITFCQ



SIISTLT





SEQ ID NO: 21
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLARSKNFHLRPRD





V
ISNINVIVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 22
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLARSKNFHLIPRD





V
ISNINVIVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 23
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLAHSKNFHLTPRD





VV
SNINVFILELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 24
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRD





VV
SNVNVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 25
APTSSSTKKTQLQLEHLLLDLQMVLNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLASSKNFHFDPRD





VV
SNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 26
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKHLEEVLNLANSKNFHVTPRD





VV
SNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 27
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLAHSKNFHFDPRD





VV
SNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 28
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLASSKNFHFDPRD





VV
SNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 29
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRD





VV
SNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 30
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLASSKNFHLTPRD





V
ISNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 31
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTF



KFYMPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRD





VV
SNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQ




SIISTLT





SEQ ID NO: 32
H9D10


IL-2 agonist
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFK



FYMPKKATELKHLQCLEEELKPLEEVLNLAHSKNFHFDPRDVVSNI



NVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 33
H9E10


IL-2 agonist
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYM



PKKATELKHLQCLEEELKPLEEVLNLASSKNFHFDPRDVVSNINVF



VLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 34
H9G8


IL-2 agonist
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYM



PKKATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNINVF



VLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 35
H9B1


IL-2 agonist
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYM



PKKATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNVNVF



VLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 146
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYM


(F42A, E62A, L80F, R81D, L85V,
PKKATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVF


I86V, 192F, and C125S)
VLELKGSETTFMCEYADETATIVEFLNRWITFSQSIISTLT









B. IL-2 Mutein Fusion Proteins


The IL-2 muteins can be prepared as fusion or chimeric polypeptides that include a subject IL-2 mutein and a heterologous polypeptide (i.e., a polypeptide that is not IL-2 or a mutant thereof) (see, e.g., U.S. Pat. No. 6,451,308), including for example, bispecific IL-2 cytokine fusions. Exemplary heterologous polypeptides can increase the circulating half-life of the chimeric polypeptide in vivo, and may, therefore, further enhance the properties of the mutant IL-2 polypeptides. In various embodiments, the polypeptide that increases the circulating half-life may be a serum albumin, such as human serum albumin, PEG, PEG-derivatives, or the Fc region of the IgG subclass of antibodies that lacks the IgG heavy chain variable region. Exemplary Fc regions can include a mutation that inhibits complement fixation and Fc receptor binding, or it may be lytic, i.e., able to bind complement or to lyse cells via another mechanism, such as antibody-dependent complement lysis (ADCC; U.S. Ser. No. 08/355,502 filed Dec. 12, 1994).


The “Fc region” can be a naturally occurring or synthetic polypeptide that is homologous to the IgG C-terminal domain produced by digestion of IgG with papain. IgG Fc has a molecular weight of approximately 50 kDa. The mutant IL-2 polypeptides can include the entire Fc region, or a smaller portion that retains the ability to extend the circulating half-life of a chimeric polypeptide of which it is a part. In addition, full-length or fragmented Fc regions can be variants of the wild-type molecule. In some embodiments, the IL-2 mutein fusion protein (e.g., an IL-2 mutein as described herein) includes an IgG1, IgG2, IgG3, or IgG4 Fc region (see, for example, sequences in FIG. 2A-2B). In some embodiments, the Fc region comprises the substitution N297A.


In some embodiments, the IL-2 mutein is linked directly or indirectly to the heterologous fusion polypeptide.


In some embodiments, the IL-2 mutein is linked directly to the Fc region. In some embodiments, the IL-2 mutein is linked to the Fc region via a linker peptide, such as GGGGS. In some embodiments, the linker is (GGGGS)n, wherein n is an integer between 1 and 10. In some embodiments, the linker is GGGGS. In some embodiments, the linker is GGGGSGGGGS (SEQ ID NO: 16). In some embodiments, the linker is GGGGSGGGGSGGGGS (SEQ ID NO:17). In some embodiments, the linker is GGGGSGGGGSGGGGSGGGGS (SEQ ID NO:18). In some embodiments, the linker is GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 19). In some embodiments, the linker contains one or more protease cleavage sites (e.g., is a protease cleavable linker). Linkers additionally can contain one or more protease cleavage sites or be sensitive to cleavage via oxidation and/or reduction. Peptide linkers that are susceptible to cleavage by enzymes of the complement system, urokinase, tissue plasminogen activator, trypsin, plasmin, caspases, kallikreins, cathepsins, legumain, MMPs, thrombin, urokinase-type plasminogen activator (uPA), matriptase, or another enzyme having proteolytic activity may be used in one example. According to another example, a linker may comprise disulfide bonds (for example, the disulfide bonds on a cysteine molecule). According to another example, a linker may comprise a protease-cleavable Val-Cit (VC) linker, a Phe-Arg linker, a Val-Lys linker, a Val-Ala linker, a Val-Arg linker, a Val-Leu-Lys linker, a Gly-Phe-Leu-Gly linker, an Ala-Phe-Lys linker, a pol-L-lysine linker, a beta-Ala-Leu-Ala-Leu linker, an Arg-Arg-Ala-Leu-Ala-Leu linker, a peptidomimetic linker, a legumain-cleavable Ala-Ala-Asn tripeptide linker, a peptide linker that is cleaved by cathepsin B and other lysosomal proteases, such as Gly-Phe-Leu-Gly and Ala-Leu-Ala-Leu, a caspase 3 DEVD sequence, or a self-immolative linker. For example, linkers disclosed in Poreba, M, FEBS J. 287(10):1936-1969 (2020), incorporated by reference herein, are contemplated by the present disclosure. Since many tumors naturally release high levels of glutathione (a reducing agent) this can reduce the disulfide bonds with subsequent release of the cargo moiety at the site of delivery. In some embodiments, the linkers is a protease-cleavable linkers is a linker cleavable by a matrix metalloprotease (MMP). MMPs are overexpressed in situ at tumors, and linkers cleavable in such contexts are contemplated by the presentation disclosure. For example, linkers disclosed in Hsu, E. J., et al., Nat. Commun. 12(2768):1-13 (2021), incorporated by reference herein, are contemplated by the present disclosure. In some embodiments, the MMP linker sequence is selected from the group consisting of SGARYRWLTA (SEQ ID NO: 234), SGRSYAILTA (SEQ ID NO: 235), SRSGRSPAIFTATG (SEQ ID NO: 236), GSSGRSPAIFTAGS (SEQ ID NO: 237), and SGFIANPVTA (SEQ ID NO: 238). In some embodiments, the MMP linker sequence is SGARYRWLTA. In some embodiments, the MMP linker sequence is SGRSYAILTA. In some embodiments, the MMP linker sequence is SRSGRSPAIFTATG. In some embodiments, the MMP linker sequence is GSSGRSPAIFTAGS. In some embodiments, the MMP linker sequence is SGFIANPVTA.


The Fc region can be “lytic” or “non-lytic,” but is typically non-lytic. A non-lytic Fc region typically lacks a high affinity Fc receptor binding site and a C′1q binding site. The high affinity Fc receptor binding site of murine IgG Fc includes the Leu residue at position 235 of IgG Fc. Thus, the Fc receptor binding site can be destroyed by mutating or deleting Leu 235. For example, substitution of Glu for Leu 235 inhibits the ability of the Fc region to bind the high affinity Fc receptor. The murine C′1q binding site can be functionally destroyed by mutating or deleting the Glu 318, Lys 320, and Lys 322 residues of IgG. For example, substitution of Ala residues for Glu 318, Lys 320, and Lys 322 renders IgG1 Fc unable to direct antibody-dependent complement lysis. In contrast, a lytic IgG Fc region has a high affinity Fc receptor binding site and a C′1q binding site. The high affinity Fc receptor binding site includes the Leu residue at position 235 of IgG Fc, and the C′1q binding site includes the Glu 318, Lys 320, and Lys 322 residues of IgG1. Lytic IgG Fc has wild-type residues or conservative amino acid substitutions at these sites. Lytic IgG Fc can target cells for antibody dependent cellular cytotoxicity or complement directed cytolysis (CDC). Appropriate mutations for human IgG are also known (see, e.g., Morrison et al., The Immunologist 2:119-124, 1994; and Brekke et al., The Immunologist 2: 125, 1994).


In other embodiments, the chimeric polypeptide can include a subject IL-2 mutein and a polypeptide that functions as an antigenic tag, such as a FLAG sequence. FLAG sequences are recognized by biotinylated, highly specific, anti-FLAG antibodies, as described herein (see also Blanar et al., Science 256:1014, 1992; LeClair et al., Proc. Natl. Acad. Sci. USA 89:8145, 1992). In some embodiments, the chimeric polypeptide further comprises a C-terminal c-myc epitope tag.


In other embodiments, the chimeric polypeptide includes the mutant IL-2 polypeptide and a heterologous polypeptide that functions to enhance expression or direct cellular localization of the mutant IL-2 polypeptide, such as the Aga2p agglutinin subunit (see, e.g., Boder and Wittrup, Nature Biotechnol. 15:553-7, 1997).


In other embodiments, a chimeric polypeptide including a mutant IL-2 and an antibody or antigen-binding portion thereof can be generated. The antibody or antigen-binding component of the chimeric protein can serve as a targeting moiety. For example, it can be used to localize the chimeric protein to a particular subset of cells or target molecule. Methods of generating cytokine-antibody chimeric polypeptides are described, for example, in U.S. Pat. No. 6,617,135.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that disrupts the interaction between the PD-1 receptor and its ligand, PD-L1, and/or is an antibody to a component of the PD-1/PD-L1 signaling pathway. Antibodies known in the art which bind to PD-1 and disrupt the interaction between the PD-1 and its ligand, PD-L1, and stimulate an anti-tumor immune response, are suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the antibody or antigen-binding portion thereof binds specifically to PD-1. For example, antibodies that target PD-1 and which can find used in the present invention include, e.g., but are not limited to nivolumab (BMS-936558, Bristol-Myers Squibb), pembrolizumab (lambrolizumab, MK03475 or MK-3475, Merck), humanized anti-PD-1 antibody JS001 (ShangHai JunShi), monoclonal anti-PD-1 antibody TSR-042 (Tesaro, Inc.), Pidilizumab (anti-PD-1 mAb CT-011, Medivation), anti-PD-1 monoclonal Antibody BGB-A317 (BeiGene), and/or anti-PD-1 antibody SHR-1210 (ShangHai HengRui), human monoclonal antibody REGN2810 (cemiplimab, Regeneron), human monoclonal antibody MDX-1106 (Bristol-Myers Squibb), and/or humanized anti-PD-1 IgG4 antibody PDR001 (Novartis). In some embodiments, the PD-1 antibody is from clone: RMP1-14 (rat IgG)-BioXcell cat #BP0146. Other suitable antibodies include anti-PD-1 antibodies disclosed in U.S. Pat. No. 8,008,449, herein incorporated by reference. In some embodiments, the antibody or antigen-binding portion thereof binds specifically to PD-L1 and inhibits its interaction with PD-1, thereby increasing immune activity. Any antibodies known in the art which bind to PD-L1 and disrupt the interaction between the PD-1 and PD-L1, and stimulates an anti-tumor immune response, are suitable for use in the chimeric polypeptides disclosed herein. For example, antibodies that target PD-L1 and are in clinical trials, include BMS-936559 (Bristol-Myers Squibb) and MPDL3280A (Genetech). Other suitable antibodies that target PD-L1 are disclosed in U.S. Pat. No. 7,943,743, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to PD-1 or PD-L1, disrupts the PD-1/PD-L1 interaction, and stimulates an anti-tumor immune response, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-PD-1 antibody. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-PD-L1 antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets CTLA-4 and disrupts its interaction with CD80 and CD86. Exemplary antibodies that target CTLA-4 include ipilimumab (MDX-010, MDX-101, Bristol-Myers Squibb), which is FDA approved, and tremelimumab (ticilimumab, CP-675, 206, Pfizer), currently undergoing human trials. Other suitable antibodies that target CTLA-4 are disclosed in WO 2012/120125, U.S. Pat. Nos. 6,984,720, 6,682,7368, and U.S. Patent Applications 2002/0039581, 2002/0086014, and 2005/0201994, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to CTLA-4, disrupts its interaction with CD80 and CD86, and stimulates an anti-tumor immune response, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-CTLA-4 antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets LAG-3 and disrupts its interaction with MHC class II molecules. An exemplary antibody that targets LAG-3 is IMP321 (Immutep), currently undergoing human trials. Other suitable antibodies that target LAG-3 are disclosed in U.S. Patent Application 2011/0150892, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to LAG-3, disrupts its interaction with MHC class II molecules, and stimulates an anti-tumor immune response, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-LAG-3 antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets B7-H3 or B7-H4. The B7 family does not have any defined receptors but these ligands are upregulated on tumor cells or tumor-infiltrating cells. An exemplary antibody that targets B7-H3 is MGA271 (Macrogenics) is currently undergoing human trials. Other suitable antibodies that target B7 family members are disclosed in U.S. Patent Application 2013/0149236, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to B7-H3 or H4, and stimulates an anti-tumor immune response, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-B7-H3 or B7-H4 antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets TIM-3 and disrupts its interaction with galectin 9. Suitable antibodies that target TIM-3 are disclosed in U.S. Patent Application 2013/0022623, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to TIM-3, disrupts its interaction with galectin 9, and stimulates an anti-tumor immune response, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-TIM-3 antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets 4-1BB/CD137 and disrupts its interaction with CD137L. It will be understood by one of ordinary skill that any antibody which binds to 4-1BB/CD137, disrupts its interaction with CD137L or another ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-4-1BB/CD137 antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets GITR and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to GITR, disrupts its interaction with GITRL or another ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-GITR antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets OX40 and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to OX40, disrupts its interaction with OX40L or another ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-OX40 antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets CD40 and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to CD40, disrupts its interaction with its ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-CD40 antibody


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets ICOS and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to ICOS, disrupts its interaction with its ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-ICOS antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets CD28 and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to CD28, disrupts its interaction with its ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-CD28 antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to an antibody or an antigen-binding portion thereof that targets IFNα and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to IFNα, disrupts its interaction with its ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the chimeric polypeptides disclosed herein. In some embodiments, the chimeric polypeptide comprises a fusion to an anti-IFNα antibody.


In some embodiments, the chimeric polypeptide comprises a fusion to a tumor antigen or polypeptide targeting a tumor antigen. Generally, tumor antigens allow for distinguishing the tumor cells from their normal cellular counterparts and can include, for example, tumor-specific antigens (TSA) as well as tumor-associated antigens (TAA). In some embodiments, a tumor antigen is a protooncogene and/or a tumor suppressor, as well as overexpressed or aberrantly expressed cellular proteins, tumor antigens produced by oncogenic viruses, oncofetal antigens, altered cell surface glycolipids and glycoproteins, and/or cell type-specific differentiation antigens. Such tumor antigens can include melanoma antigens, cancer-testis antigens, epithelial tumor antigens, cell cycle regulatory proteins, prostate specific antigens (including prostate carcinoma antigens, such as for example those disclosed in U.S. Pat. No. 5,538,866) lymphoma (U.S. Pat. Nos. 4,816,249; 5,068,177; and 5,227,159). Tumor antigens can include for example, but are not limited to, HMW mucins bound by 2G3 and 369F10, c-erbB-2 related tumor antigen (an approximately 42 kD or 55 kD glycoprotein), the approximately 40, 60, 100 and 200 kD antigens bound by 113F1, 9-O-acetyl GD3, p97, alphafetoprotein (AFP) (for example, for germ cell tumors and/or hepatocellular carcinoma), carcinoembryonic antigen (CEA) (for example, for bowel cancers occasional lung or breast cancer), CA-125 (for example, for ovarian cancer), MUC-1 (for example, for breast cancer), epithelial tumor antigen (ETA) (for example, for breast cancer), tyrosinase (for example, for malignant melanoma), melanoma-associated antigen (MAGE) (for example, for malignant melanoma), cancer/testis antigen 1 (CTAG1B), melanoma-associated antigen 1 (MAGEA1), abnormal Ras products, abnormal p53 products, overexpression of cyclins (including, for example, cyclin B1), mutation in fibronectin, posttranslational alteration in the MUC1 glycoprotein, secreted tumor antigens (including, for example, gangliosides).


Other fusions can include fusions with pro-apoptotic payloads. Such exemplary sequences are provided in the table below. In some embodiments, and IL-2 mutein as described herein is fused to a pro-apoptotic payload, for example a BAD, BAX, BAR, BIK, and/or BIDsequence. In some embodiments, the pro-apoptotic payload is a Bcl-2 domain containing peptide and/or a susequence of a BAD, BAX, BAR, BIK, and/or BID sequence. Exemplary pro-apoptotic fusions are provided below, in Table 3.









TABLE 3







List of Selected Pro-Apoptotic Fusion Partners








SEQ ID NO:



(Information)
Amino acid sequence





SEQ ID NO: 38
MFQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQ


BAD amino acid sequence
APGLLWDASHQQEQPTSSSHHGGAGAVEIRSRHSAYPAGT



EDDEGMGEEPSPFRGRSRAAPPNLWAAQRYGRELRRMSDE



FVDSFKKGLPRPKSAGTATQMRQSSSWTRVFQSWWDRNLG



RGSSAPSQ





SEQ ID NO: 39
MFQIPEFEPSEQEDSSSAERGLGPSPAGDGPSGSGKHHRQ


>HsBAD_Q92934-1(UniProtKB)
APGLLWDASHQQEQPTSSSHHGGAGAVEIRSRHSSYPAGT



EDDEGMGEEPSPFRGRSRSAPPNLWAAQRYGRELRRMSDE



FVDSFKKGLPRPKSAGTATQMRQSSSWTRVFQSWWDRNLG



RGSSAPSQ





SEQ ID NO: 40
MDGSGEQPRGGGPTSSEQIMKTGALLLQGFIQDRAGRMGG


>HsBAX_Q07812-1(UniProtKB)
EAPELALDPVPQDASTKKLSECLKRIGDELDSNMELQRMI



AAVDTDSPREVFFRVAADMFSDGNFNWGRVVALFYFASKL



VLKALCTKVPELIRTIMGWTLDFLRERLLGWIQDQGGWDG



LLSYFGTPTWQTVTIFVAGVLTASLTIWKKMG





SEQ ID NO: 41
MASGQGPGPPRQECGEPALPSASEEQVAQDTEEVFRSYVF


>HsBAK1_Q16611-1(UniProtKB)
YRHQQEQEAEGVAAPADPEMVTLPLQPSSTMGQVGRQLAI



IGDDINRRYDSEFQTMLQHLQPTAENAYEYFTKIATSLFE



SGINWGRVVALLGFGYRLALHVYQHGLTGF



LGQVTRFVVDFMLHHCIARWIAQRGGWVAALNLGNGPILN



VLVVLGVVLL



GQFVVRRFFKS





SEQ ID NO: 42
MSEVRPLSRDILMETLLYEQLLEPPTMEVLGMTDSEEDLD


>HsBIK_Q13323-1(UniProtKB)
PMEDFDSLEC



MEGSDALALRLACIGDEMDVSLRAPRLAQLSEVAMHSLGL



AFTYDQTEDI



RDVLRSFMDGFTTLKENIMRFWRSPNPGSWVSCEQVLLAL



LLLLALLLPL



LSGGLHLLLK





SEQ ID NO: 43
MDCEVNNGSSLRDECITNLLVFGFLQSCSDNSFRRELDAL


>HsBID_P55957-1(UniProtKB)
GHELPVLAPQ



WEGYDELQTDGNRSSHSRLGRIEADSESQEDIIRNIARHL



AQVGDSMDRS



IPPGLVNGLALQLRNTSRSEEDRNRDLATALEQLLQAYPR



DMEKEKTMLV



LALLLAKKVASHTPSLLRDVFHTTVNFINQNLRTYVRSLA



RNGMD









In some particular embodiments, an IL-2 antagonist can be fused to a pro-apoptotic payload for the treatment of cancer. An “antagonist” is a compound that opposes the actions of an agonist, e.g. by preventing, reducing, inhibiting, or neutralizing the activity of an agonist. An “antagonist” can also prevent, inhibit, or reduce constitutive activity of a target, e.g., a target receptor, even where there is no identified agonist. While typically IL-2 muteins with agonist or superagonist activity as compared to wild-type IL-2 are employed with the cancer treatment methods of the present invention, IL-2 muteins with antagonistic properties can be employed when such antagonists are fused to a pro-apoptotitic payload. In some embodiments, the IL-2 antagonist comprises the following amino acid substitutions L18R, Q22E, Q126T, and S130R as compared to the wild-type IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 antagonist comprises the following amino acid substitutions L18R, Q22E, L80F, R81D, L85V, I86V, and Q126T as compared to the wild-type IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 antagonist comprises the following amino acid substitutions L18R, Q22E, L80F, R81D, L85V, I86V, Q126T, and S130R as compared to the wild-type IL-2 of SEQ ID NO:2. Exemplary antagonists that can be fuses with pro-apoptotic payloads, such as those provided above, are provided below in Table 4.









TABLE 4







IL-2 Antagonsits for Fusion with Pro-Apoptotic Payloads








SEQ ID NO:



(Information)
Amino acid sequence





SEQ ID NO: 36
APTSSSTKKTQLQLEHLRLDLEMILNGINNYKNPKLTRMLTFKFYM


IL-2 antagonist
PKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPR



DVVSNINVFV LELKGSETTF MCEYADETATIVEFLNRWIT



FCTSIISTLT





SEQ ID NO: 37
APTSSSTKKTQLQLEHLRLDLEMILNGINNYKNPKLTRMLTFKFYM


IL-2 VARIANT (antagonist)
PKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVF



VLELKGSETTFMCEYADETATIVEFLNRWITFCTSIIRTLT





SEQ ID NO: 56
H9RET-Fc


IL-2 extended half-life fusion
APTSSSTKKTQLQLEHLRLDLEMILNGINNYKNPKLTRMLTFKFYM


(GS linker can be
PKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVF




GGGGSGGGGSGGGGS
 as shown

VLELKGSETTFMCEYADETATIVEFLNRWITFCTSIISTLTGGGGS


or anything other GS containingin


GGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT



linker)
PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRV



VSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV



YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT



PPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKS



LSLSPGK*





SEQ ID NO: 57
IL-2 VARIANT-Fc


IL-2 extended half-life fusion
APTSSSTKKTQLQLEHLRLDLEMILNGINNYKNPKLTRMLTFKFYM


(GS linker can be
PKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVF




GGGGSGGGGSGGGGS
 as shown

VLELKGSETTFMCEYADETATIVEFLNRWITFCTSIIRTLTGGGGS


or anything other GS containingin


GGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT



linker)
PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRV



VSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV



YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT



PPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKS



LSLSPGK*





SEQ ID NO: 58
H9RETFYAA-Fc


IL-2 extended half-life fusion
APTSSSTKKTQLQLEHLRLDLEMILNGINNYKNPKLTRMLTAKFAM


(GS linker can be
PKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVF




GGGGSGGGGSGGGGS
 as shown

VLELKGSETTFMCEYADETATIVEFLNRWITFCTSIISTLTGGGGS


or anything other GS containingin


GGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT



linker)
PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRV



VSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV



YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT



PPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKS



LSLSPGK*





SEQ ID NO: 59
IL-2 VARIANTFYAA-Fc


IL-2 extended half-life fusion
APTSSSTKKTQLQLEHLRLDLEMILNGINNYKNPKLTRMLTAKFAM


(GS linker can be
PKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVF




GGGGSGGGGSGGGGS
 as shown

VLELKGSETTFMCEYADETATIVEFLNRWITFCTSIIRTLTGGGGS


or any other GS containing linker)


GGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT




PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRV



VSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV



YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT



PPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKS



LSLSPGK*





SEQ ID NO: 60
H9RETFEAA-Fc


IL-2 extended half-life fusion
PKKATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVF


(GS linker can be
VLELKGSETTFMCEYADETATIVEFLNRWITFCTSIISTLTGGGGS




GGGGSGGGGSGGGGS
 as shown



GGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT



or anyt other GS containing linker)
PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRV



APTSSSTKKTQLQLEHLRLDLEMILNGINNYKNPKLTRMLTAKFYM



VSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV



YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT



PPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKS



LSLSPGK*





SEQ ID NO: 61
IL-2 VARIANTFEAA-Fc


IL-2 extended half-life fusion
APTSSSTKKTQLQLEHLRLDLEMILNGINNYKNPKLTRMLTAKFYM


(GS linker can be
PKKATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVF




GGGGSGGGGSGGGGS
 as shown

VLELKGSETTFMCEYADETATIVEFLNRWITFCTSIIRTLTGGGGS


or any other GS containing linker)


GGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRT




PEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRV



VSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV



YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT



PPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKS



LSLSPGK*









Other fusions can include fusions with anti-apoptotic payloads for use in prolonging activation of CD8 cells, NK cells and anergic NK cells as well, and such exemplary sequences are provided in the table below. Such prolong activation of T-cells can prove beneficial in cancer therapy treatment methods.









TABLE 5







List of Exemplary IL-2 Anti-Apoptotic Fusion Amino Acid Sequences








SEQ ID NO: 



(Information)
Amino acid sequence





SEQ ID NO: 40
H9-BclxL



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY



MPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG



GGSMSQSNRELVVDFLSYKLSQKGYSWSQFSDVEENRTEAPEGTE



SEMETPSAINGNPSWHLADSPAVNGATGHSSSLDAREVIPMAAVK



QALREAGDEFELRYRRAFSDLTSQLHITPGTAYQSFEQVVNELFR



DGVNWGRIVAFFSFGGALCVESVDKEMQVLVSRIAAWMATYLNDH



LEPWIQENGGWDTFVELYGNNAAAESRKGQERFNRWFLTGMTVAG



VVLLGSLFSRK*





SEQ ID NO: 41
H9FYAA-BclxL



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFA



MPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG



GGSMSQSNRELVVDFLSYKLSQKGYSWSQFSDVEENRTEAPEGTE



SEMETPSAINGNPSWHLADSPAVNGATGHSSSLDAREVIPMAAVK



QALREAGDEFELRYRRAFSDLTSQLHITPGTAYQSFEQVVNELFR



DGVNWGRIVAFFSFGGALCVESVDKEMQVLVSRIAAWMATYLNDH



LEPWIQENGGWDTFVELYGNNAAAESRKGQERFNRWFLTGMTVAG



VVLLGSLFSRK*





SEQ ID NO: 42
H9FEAA-BclxL



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFY



MPKKATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG



GGSMSQSNRELVVDFLSYKLSQKGYSWSQFSDVEENRTEAPEGTE



SEMETPSAINGNPSWHLADSPAVNGATGHSSSLDAREVIPMAAVK



QALREAGDEFELRYRRAFSDLTSQLHITPGTAYQSFEQVVNELFR



DGVNWGRIVAFFSFGGALCVESVDKEMQVLVSRIAAWMATYLNDH



LEPWIQENGGWDTFVELYGNNAAAESRKGQERFNRWFLTGMTVAG



VVLLGSLFSRK*





SEQ ID NO: 43
H9D10-BclxL



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY



MPKKATELKHLQCLEEELKPLEEVLNLAHSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG



GGSMSQSNRELVVDFLSYKLSQKGYSWSQFSDVEENRTEAPEGTE



SEMETPSAINGNPSWHLADSPAVNGATGHSSSLDAREVIPMAAVK



QALREAGDEFELRYRRAFSDLTSQLHITPGTAYQSFEQVVNELFR



DGVNWGRIVAFFSFGGALCVESVDKEMQVLVSRIAAWMATYLNDH



LEPWIQENGGWDTFVELYGNNAAAESRKGQERFNRWFLTGMTVAG



VVLLGSLFSRK*





SEQ ID NO: 44
H9E10-BclxL



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY



MPKKATELKHLQCLEEELKPLEEVLNLASSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG



GGSMSQSNRELVVDFLSYKLSQKGYSWSQFSDVEENRTEAPEGTE



SEMETPSAINGNPSWHLADSPAVNGATGHSSSLDAREVIPMAAVK



QALREAGDEFELRYRRAFSDLTSQLHITPGTAYQSFEQVVNELFR



DGVNWGRIVAFFSFGGALCVESVDKEMQVLVSRIAAWMATYLNDH



LEPWIQENGGWDTFVELYGNNAAAESRKGQERFNRWFLTGMTVAG



VVLLGSLFSRK*





SEQ ID NO: 45
H9G8-Bclx1



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY



MPKKATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG



GGSMSQSNRELVVDFLSYKLSQKGYSWSQFSDVEENRTEAPEGTE



SEMETPSAINGNPSWHLADSPAVNGATGHSSSLDAREVIPMAAVK



QALREAGDEFELRYRRAFSDLTSQLHITPGTAYQSFEQVVNELFR



DGVNWGRIVAFFSFGGALCVESVDKEMQVLVSRIAAWMATYLNDH



LEPWIQENGGWDTFVELYGNNAAAESRKGQERFNRWFLTGMTVAG



VVLLGSLFSRK*





SEQ ID NO: 46
H9B1-BclxL



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY



MPKKATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNVN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG



GGSMSQSNRELVVDFLSYKLSQKGYSWSQFSDVEENRTEAPEGTE



SEMETPSAINGNPSWHLADSPAVNGATGHSSSLDAREVIPMAAVK



QALREAGDEFELRYRRAFSDLTSQLHITPGTAYQSFEQVVNELFR



DGVNWGRIVAFFSFGGALCVESVDKEMQVLVSRIAAWMATYLNDH



LEPWIQENGGWDTFVELYGNNAAAESRKGQERFNRWFLTGMTVAG



VVLLGSLFSRK*









Other exemplary IL-2 fusions include those listed in the table below:









TABLE 6







List of Exemplary IL-2 Extended Half-Life Fusion Amino Acid Sequences








SEQ ID NO: 



(Information)
Amino acid sequence





SEQ ID NO: 47
H9-Fc (H9 at N-terminal only shown)


IL-2 extended half-life
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY


fusion
MPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG





GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM




ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA



STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK*





SEQ ID NO: 48
H9-Fc (″Knob-in-hole″ with H9 at N-terminus)


IL-2 extended half-life
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY


fusion
MPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG





GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM




ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA



STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK-[FC sequence]





SEQ ID NO: 49
H9-Fc (″Knob-in-hole″ with H9 at C-terminus)


IL-2 extended half-life
[FC sequence]-


fusion
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY



MPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG





GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM




ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA



STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK*





SEQ ID NO: 50
H9FYAA-Fc


IL-2 extended half-life
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFA


fusion
MPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNIN



VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG





GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM




ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA



STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK*





SEQ ID NO: 51
H9FEAA-Fc


IL-2 extended half-life
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFY


fusion
MPKKATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNIN


(GS linker can be
VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG




GGGGSGGGGSGGGGS
 as shown



GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM



or any other GS
ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA


containing linker)
STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK*





SEQ ID NO: 52
H9D10-Fc


IL-2 extended half-life
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY


fusion
MPKKATELKHLQCLEEELKPLEEVLNLAHSKNFHFDPRDVVSNIN


(GS linker can be
VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG




GGGGSGGGGSGGGGS
 as shown



GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM



or any other GS
ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA


containing linker)
STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK*





SEQ ID NO: 53
H9E10-Fc


IL-2 extended half-life
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY


fusion
MPKKATELKHLQCLEEELKPLEEVLNLASSKNFHFDPRDVVSNIN


(GS linker can be
VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG




GGGGSGGGGSGGGGS
 as shown



GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM



or any other GS
ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA


containing linker)
STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK*





SEQ ID NO: 54
H9G8-Fc


IL-2 extended half-life
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY


fusion
MPKKATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNIN


(GS linker can be
VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG




GGGGSGGGGSGGGGS
 as shown



GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM



or any other GS
ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA


containing linker)
STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK*





SEQ ID NO: 55
H9B1- Fc


IL-2 extended half-life
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFY


fusion
MPKKATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNVN


(GS linker can be
VFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGG




GGGGSGGGGSGGGGS
 as shown



GGSGGGGSGGGGS
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM



or any other GS
ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA


containing linker)
STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP



ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGK*





SEQ ID NO: 62
H9-Albumin (H9 at C-terminal shown)


(GS linker can be
DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA




GGGGSGGGGSGGGGS
 as shown

KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE


or any other GS
CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY


containing linker)
APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC



ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL



LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA



DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA



KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE



YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE



DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK



EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD



FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLGGGGSGGGGSGGGG





S
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKF




YMPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNI



NVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT*





SEQ ID NO: 63
H9FYAA-Albumin (H9FYAA at C-terminal shown)


(GS linker can be
DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA




GGGGSGGGGSGGGGS
 as shown

KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE


or any other GS
CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY


containing linker)
APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC



ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL



LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA



DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA



KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE



YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE



DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK



EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD



FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLGGGGSGGGGSGGGG





SAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFAMPKK






ATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGS





ETTFMCEYADETATIVEFLNRWITFCQSIISTLT






SEQ ID NO: 64
H9FEAA-Albumin (H9FEAA at C-terminal shown)


(GS linker can be
DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA




GGGGSGGGGSGGGGS
 as shown

KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE


or any other GS
CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY


containing linker)
APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC



ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL



LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA



DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA



KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE



YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE



DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK



EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD



FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLGGGGSGGGGSGGGG





SAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKK






ATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGS





ETTFMCEYADETATIVEFLNRWITFCQSIISTLT






SEQ ID NO: 65
H9D10-Albumin (H9D10 shown at N-terminal)


(GS linker can be
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKA




GGGGSGGGGSGGGGS
 as shown

TELKHLQCLEEELKPLEEVLNLAHSKNFHFDPRDVVSNINVFVLELKGSE


or any other GS
TTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGGSD


containing linker)
AHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAK



TCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNEC



FLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFYA



PELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKCA



SLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDLL



ECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPAD



LPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAK



TYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEY



KFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAED



YLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPKE



FNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDDF



AAFVEKCCKADDKETCFAEEGKKLVAASQAALGL





SEQ ID NO: 66
H9D10FEAA-Albumin




GGGGSGGGGSGGGGS
 as shown

APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKA


or any other GS
TELKHLQCLEEALKPLEEVLNLAHSKNFHFDPRDVVSNINVFVLELKGSE


containing linker)
TTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGGSD


(GS linker can be
AHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAK



TCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNEC



FLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFYA



PELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKCA



SLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDLL



ECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPAD



LPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAK



TYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEY



KFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAED



YLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPKE



FNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDDF



AAFVEKCCKADDKETCFAEEGKKLVAASQAALGL





SEQ ID NO: 67
H9E10-Albumin (H9E10 shown at N-terminal)


(GS linker can be
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKA




GGGGSGGGGSGGGGS
 as shown

TELKHLQCLEEELKPLEEVLNLASSKNFHFDPRDVVSNINVFVLELKGSE


or anything other GS
TTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGGSD


containingin linker)
AHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAK



TCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNEC



FLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFYA



PELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKCA



SLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDLL



ECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPAD



LPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAK



TYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEY



KFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAED



YLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPKE



FNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDDF



AAFVEKCCKADDKETCFAEEGKKLVAASQAALGL





SEQ ID NO: 68
H9G8-Albumin (H9G8 shown at N-terminal)


(GS linker can be
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKA




GGGGSGGGGSGGGGS
 as shown

TELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNINVFVLELKGSE


or anything other GS
TTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGGSD


containingin linker)
AHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAK



TCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNEC



FLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFYA



PELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKCA



SLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDLL



ECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPAD



LPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAK



TYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEY



KFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAED



YLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPKE



FNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDDF



AAFVEKCCKADDKETCFAEEGKKLVAASQAALGL





SEQ ID NO: 69
H9B1-Albumin (H9B1 shown at N-terminal)


(GS linker can be
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKA




GGGGSGGGGSGGGGS
 as shown

TELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNVNVFVLELKGSE


or anything other GS
TTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGGSDA


containingin linker)
HKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAKT



CVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNECF



LQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFYAP



ELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKCAS



LQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDLLE



CADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPADL



PSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAKT



YETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEYK



FQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAEDY



LSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPKEF



NAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDDFA



AFVEKCCKADDKETCFAEEGKKLVAASQAALGL





SEQ ID NO: 70
H9FEAA-Albumin (H9FEAA at N-terminal shown)


(GS linker can be
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKA




GGGGSGGGGSGGGGS
 as shown

TELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSE


or any other GS
TTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGGSD


containing linker)
AHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAK



TCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNEC



FLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFYA



PELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKCA



SLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDLL



ECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPAD



LPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAK



TYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEY



KFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAED



YLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPKE



FNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDDF



AAFVEKCCKADDKETCFAEEGKKLVAASQAALGL





SEQ ID NO: 71
H9D10-Albumin (H9D10 shown at C-terminal)


(GS linker can be
DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA




GGGGSGGGGSGGGGS
 as shown

KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE


or any other GS
CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY


containing linker)
APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC



ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL



LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA



DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA



KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE



YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE



DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK



EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD



FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLGGGGSGGGGSGGGG





S
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKK




ATELKHLQCLEEELKPLEEVLNLAHSKNFHFDPRDVVSNINVFVLELKGS



ETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 72
H9D10FEAA-Albumin (H9FEAA shown at C-


(GS linker can be
terminal)




GGGGGGGGSGGGGS
 as shown

DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA


or any other GS
KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE


containing linker)
CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY



APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC



ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL



LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA



DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA



KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE



YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE



DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK



EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD



FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLGGGGSGGGGSGGGG





S
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKK




ATELKHLQCLEEALKPLEEVLNLAHSKNFHFDPRDVVSNINVFVLELKGS



ETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 73
H9E10-Albumin (H9E10 shown at C-terminal)


(GS linker can be
DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA




GGGGSGGGGSGGGGS
 as shown

KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE


or anything other GS
CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY


containingin linker)
APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC



ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL



LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA



DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA



KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE



YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE



DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK



EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD



FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLGGGGSGGGGSGGGG





S
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKK




ATELKHLQCLEEELKPLEEVLNLASSKNFHFDPRDVVSNINVFVLELKGS



ETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 74
H9G8-Albumin (H9G8 shown at C-terminal)


(GS linker can be
DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA




GGGGSGGGGSGGGGS
 as shown

KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE


or anything other GS
CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY


containingin linker)
APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC



ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL



LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA



DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA



KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE



YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE



DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK



EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD



FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLGGGGSGGGGSGGGG





S
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKK




ATELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNINVFVLELKGS



ETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 75
H9B1-Albumin (H9B1 shown at C-terminal)


(GS linker can be
DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA




GGGGSGGGGSGGGGS
 as shown

KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE


or anything other GS
CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY


containingin linker)
APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC



ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL



LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA



DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA



KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE



YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE



DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK



EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD



FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLGGGGSGGGGSGGGGS



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKA



TELKHLQCLEEELKPLEEVLNLANSKNFHFDPRDVVSNVNVFVLELKGSE



TTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 147
MDNA11 (MDNA109FEAA-Albumin)


(GS linker can be
MYRMQLLSCIALSLALVTNS_APTSSSTKKTQLQLEHLLLDLQMI




GGGGSGGGGSGGGGS
 as shown

LNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEEALKPLEEV


or anything other GS
LNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATI


containingin linker)
VEFLNRWITFSQSIISTLTGGGGSGGGGSGGGGSDAHKSEVAHRF


MDNA11 (MDNA109FEAA-
KDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAKTCVAD


Albumin)
ESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNEC



FLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRH



PYFYAPELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKA



SSAKQRLKCASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVT



DLTKVHTECCHGDLLECADDRADLAKYICENQDSISSKLKECCEK



PLLEKSHCIAEVENDEMPADLPSLAADFVESKDVCKNYAEAKDVF



LGMFLYEYARRHPDYSVVLLLRLAKTYETTLEKCCAAADPHECYA



KVFDEFKPLVEEPQNLIKQNCELFEQLGEYKFQNALLVRYTKKVP



QVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAEDYLSVVLNQLC



VLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPKEFNAET



FTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDDF



AAFVEKCCKADDKETCFAEEGKKLVAASQAALGL*



Cys125 changed to Serine (underlined and



bolded









In some embodiments, the IL-2 mutein-Fc fusion comprises one of the following sequences:









TABLE 7







List of Amino Acid Sequences








SEQ ID NO: 



(Information)
Amino acid sequence





SEQ ID NO: 76

MYRMQLLSCIALSLALVTNS
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPK



(also listed

LTRMLTFKFYMPKKATELKHLQCLEE



herein as SEQ

ELKPLEEVLNLAQSKNFHLRPRDLISNINVIVLELKGSETTFMCEYADETATIVE



ID NO: 11)

FLNRWITFCQSIISTLTGGGGSGGGG




SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



DPEVKFNWYVDGVEVHNAKTKPREEQ



YASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYT



LPPSREEMTKNQVSLTCLVKGFYPSD



IAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEA



LHNHYTQKSLSLSPGK*





SEQ ID NO: 77

MYRMQLLSCIALSLALVTNS
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPK



(also listed

LTRMLTFKFYMPKKATELKHLQCLEE



herein as SEQ

ELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVE



ID NO: 12)

FLNRWITFCQSIISTLTGGGGSGGGG




SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



DPEVKFNWYVDGVEVHNAKTKPREEQ



YASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYT



LPPSREEMTKNQVSLTCLVKGFYPSD



IAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEA



LHNHYTQKSLSLSPGK*





SEQ ID NO: 78

MYRMQLLSCIALSLALVTNS
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPK



(also listed

LTRMLTAKFAMPKKATELKHLQCLEE



herein as SEQ

ELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVE



ID NO: 13)

FLNRWITFCQSIISTLTGGGGSGGGG




SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



DPEVKFNWYVDGVEVHNAKTKPREEQ



YASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYT



LPPSREEMTKNQVSLTCLVKGFYPSD



IAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEA



LHNHYTQKSLSLSPGK*





SEQ ID NO: 79

MYRMQLLSCIALSLALVTNS
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPK



(also listed

LTRMLTAKFYMPKKATELKHLQCLEE



herein as SEQ


ALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVE




ID NO: 14)

FLNRWITFCQSIISTLTGGGGSGGGG




SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



DPEVKFNWYVDGVEVHNAKTKPREEQ



YASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYT



LPPSREEMTKNQVSLTCLVKGFYPSD



IAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEA



LHNHYTQKSLSLSPGK*





SEQ ID NO: 80

MYRMQLLSCIALSLALVTNS
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPK



(also listed

LTRMLTAKFAMPKKATELKHLQCLEE



herein as SEQ

ALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVE



ID NO: 15)

FLNRWITFCQSIISTLTGGGGSGGGG




SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



DPEVKFNWYVDGVEVHNAKTKPREEQ



YASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYT



LPPSREEMTKNQVSLTCLVKGFYPSD



IAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEA



LHNHYTQKSLSLSPGK*









In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO: 12 through SEQ ID NO:15 and/or SEQ ID NO:20 through SEQ ID NO:80 (for example, any of the IL-2 sequences provided herein). In some embodiments, the IL-2 mutein sequence is 95% identical to any one of SEQ ID NO:12 through SEQ ID NO:15 and/or SEQ ID NO:20 through SEQ ID NO:80 (for example, any of the IL-2 sequences provided herein). In some embodiments, the IL-2 mutein sequence is 98% identical to any one of SEQ ID NO:12 through SEQ ID NO:15 and/or SEQ ID NO:20 through SEQ ID NO:80 (for example, any of the IL-2 sequences provided herein). In some embodiments, the IL-2 mutein sequence is 99% identical to any one of SEQ ID NO: 12 through SEQ ID NO:15 and/or SEQ ID NO:20 through SEQ ID NO:80 (for example, any of the IL-2 sequences provided herein).


C. IL-4, IL-13, IL-10, IL-12, IL15, and IL-18 for Use in Bispecific IL-2 Cytokine Fusions


In some embodiments, an IL-2 mutein can be fused to an IL-4 mutein as described herein. In some embodiments, an IL-2 mutein can be fused to an IL-13 mutein as described herein. In some embodiments, an IL-2 mutein can be fused to an IL-10. In some embodiments, an IL-2 mutein can be fused to an IL-12. In some embodiments, an IL-2 mutein can be fused to an IL-15. In some embodiments, an IL-2 mutein can be fused to an IL-18. In some embodiments, such fusions function to specifically target cancer cells and/or cancer stem cells and reduce or inhibit cancer stem cell growth, as well as targeting the immunosuppressive cells in the tumor microenvironment (TME).


Any IL-13 sequence or variant thereof can be used in a fusion with an IL-2 mutein as described herein. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO:10; C5 SEQ ID NO:11; D10 SEQ ID NO:12; E10 SEQ ID NO:13; G8 SEQ ID NO:14; H4 SEQ ID NO:15; and H9 SEQ ID NO:16. Exemplary IL-13 polypeptide sequences are provided in SEQ ID NO:81-SEQ ID NO:128, as well as the table below. In some embodiments, the IL-13 polypeptide sequence is as provided in any one of SEQ ID NO:81-SEQ ID NO:128. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:81. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:82. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:83. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:84. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:85. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:86. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:87. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:88. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:89. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:90. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:91. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:92. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:93. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:94. In some embodiments, the polypeptide sequence is SEQ ID NO:95. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:96. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:97. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:98. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:99. In some embodiments, the polypeptide sequence is SEQ ID NO: 100. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:101. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:102. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:103. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:104. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:105. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:106. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:107. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:108. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:109. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:110. In some embodiments, the polypeptide sequence is SEQ ID NO:111. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:112. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:113. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:114. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:115. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:116. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:117. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:118. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:119. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:120. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:121. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:122. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:123. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:124. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:125. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:126. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:127. In some embodiments, the IL-13 polypeptide sequence is SEQ ID NO:128. IL-13 In some embodiments, the IL-13 polypeptide sequence is 90% identical to any one of SEQ ID NO:81 through SEQ ID NO:128. In some embodiments, the IL-13 polypeptide sequence is 95% identical to any one of SEQ ID NO:81 through SEQ ID NO:128. In some embodiments, the IL-13 polypeptide sequence is 98% identical to any one of SEQ ID NO:81 through SEQ ID NO:128. In some embodiments, the IL-13 polypeptide sequence is 99% identical to any one of SEQ ID NO:81 through SEQ ID NO: 128.


In some embodiments, any one of SEQ ID NO:81-SEQ ID NO:128 are linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO: 81 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:82 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:83 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:84 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:85 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:86 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:87 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:88 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:89 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:90 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:91 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:92 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:93 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:94 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:94 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:96 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:97 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:98 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:99 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:100 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:101 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:102 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:103 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:104 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:105 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:106 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:107 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:108 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:109 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:110 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:111 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:112 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:113 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:114 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:115 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:116 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:117 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:118 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:119 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:120 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:121 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:122 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:123 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:124 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:125 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:126 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:127 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:128 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO: 11; D10 SEQ ID NO: 12; E10 SEQ ID NO: 13; G8 SEQ ID NO: 14; H4 SEQ ID NO:15; and H9 SEQ ID NO:16.


In some embodiments an IL-13 peptide of the invention comprises one or more of the amino acids substitutions: (1) L10F, L10I, L10V, L10A, L10D, L10T, L10H; (2) R11S, R11N, R11H, R11L, R11I; (3) 114L, 114F, 114V, 114M; (4) V18L, V18F, V181; (5) E12A, (6) R65D, (7) R86K, R86T, R86M; (8) D87E, D87K, D87R, D87G, D87S; (9) T881, T88K, T88R; (10) K89R, K89T, K89M; (11) L101F, L101I, L101Y, L101H, L101N; (12) K104R, K104T, K104M; (13) K105T, K105A, K105R, K105E; (14) F107L, F1071, F107V, F107M; and (15) R108K, R108T, R108M, which substitutions cause an altered affinity for one or both of IL-13Rα1 and IL-13Rα2. In other embodiments, modified residues are at two or more, three or more, four or more, five or more, and not more than 14 amino acids within the combined set of contact residues defined above. As described in International Patent Publication WO 2013/112871, the disclosure of which is incorporated by reference herein in its entirety. In some embodiments, amino acid substitutions include without limitation those provided in FIG. 4.


Sets of modifications may include the following specific changes: (1) L10H; L10A; (2) R11L; (4) V181; (7) R86M; R86K; R86T; (8) D87K; D87G; (9) T88R, T88S; T88K; (10) K89R; (11) L101N; (12) K104R; (13) K105A; K105E; (14) R108K. In some embodiments, the modification includes any one of the recited specific changes. In some embodiments, the modification includes L10H. In some embodiments, the modification includes L10A. In some embodiments, the modification includes R11L. In some embodiments, the modification includes V181. In some embodiments, the modification includes R86M. In some embodiments, the modification includes R86K. In some embodiments, the modification includes R86T. In some embodiments, the modification includes D87K. In some embodiments, the modification includes D87G. In some embodiments, the modification includes T88R. In some embodiments, the modification includes T88S. In some embodiments, the modification includes T88K. In some embodiments, the modification includes K89R. In some embodiments, the modification includes L101N. In some embodiments, the modification includes K104R. In some embodiments, the modification includes K1T05A. In some embodiments, the modification includes K105E. In some embodiments, the modification includes R108K. In some embodiments, the polypeptide comprising the one or more modifications is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, amino acid substitutions include without limitation those provided in FIG. 4. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO: 8; B1 SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO: 11; D10 SEQ ID NO:12; E10 SEQ ID NO: 13; G8 SEQ ID NO: 14; H4 SEQ ID NO: 15; and H9 SEQ ID NO:16.


Specific sets of modifications that provide for greater selectivity in binding to IL-13Rα2 versus IL-13Rα1 relative to a native IL-13 sequence may include, without limitation:

    • [L10D, R11, V18I, R86K, D87K, k89R, R108K] (for example, C2, e.g. SEQ ID NO:31 or SEQ ID NO:49)
    • [L10A, R86T, D87G, T88K, K89R, L101N, K104R, K105A, R108K] (for example, C3, e.g. SEQ ID NO:32 or SEQ ID NO:50)
    • [L10V, K89R, L101N, K105E, R108T] (for example, C4, e.g. SEQ ID NO:33 or SEQ ID NO:31)
    • [R11S, I14M, T88S, L101N, K105A, R108K] (for example, C7, e.g. SEQ ID NO:34 or SEQ ID NO:52)
    • [L10H, R11L, V18I, R86K, D87E, K89R, L101N, K105T, R108K] (C9, e.g. SEQ ID NO:53)
    • [L10H, R86T, D87G, T88R, R108K] (C11e.g. SEQ ID NO:38 or SEQ ID NO:55)
    • [L10A, V18F, R86K, D87K, K89R, L101I, K104R, R108K] (D7, e.g. SEQ ID NO:40 or SEQ ID NO:57)
    • [L10T/D; R11I; V181; R86K; D87K/G; T88S; K89R; L101Y; K104R; K105T; R108K]
    • [L10A/V; R86T; D87G; T88K; K89R; L101N; K104R; K105A/E; R108K/T]


In some embodiments, the set of modifications comprises L10V, K89R, L101N, K105E, R108T. In some embodiments, the set of modifications comprises R11S, 114M, T88S, L101N, K105A, and R108K (C7, e.g. SEQ ID NO:35 or SEQ ID NO:52). In some embodiments, the set of modifications comprises L10H, R11L, V18I, R86K, D87E, K89R, L101N, K105T, and R108K (C9, e.g. SEQ ID NO:36 or SEQ ID NO:53). In some embodiments, the set of modifications comprises L10H, R86T, D87G, T88R, and R108K (C11 e.g. SEQ ID NO:38 or SEQ ID NO:55). In some embodiments, the set of modifications comprises L10A, V18F, R86K, D87K, K89R, L101I, K104R, and R108K (D7, e.g. SEQ ID NO:40 or SEQ ID NO:57). In some embodiments, the set of modifications comprises L10T/D, R11I, V181, R86K, D87K/G, T88S, K89R, L101Y, K104R, K105T, and R108K. In some embodiments, the set of modifications comprises L10T, R11I, V181, R86K, D87K, T88S, K89R, L101Y, K104R, K105T, and R108K. In some embodiments, the set of modifications comprises L10T, R11I, V181, R86K, D87G, T88S, K89R, L101Y, K104R, K105T, and R108K. In some embodiments, the set of modifications comprises L10D, R11I, V18I, R86K, D87K, T88S, K89R, L101Y, K104R, K105T, and R108K. In some embodiments, the set of modifications comprises L10D, R11I, V18I, R86K, D87G, T88S, K89R, L101Y, K104R, K105T, R108K. In some embodiments, the set of modifications comprises L10A/V, R86T, D87G, T88K, K89R, L101N, K104R, K105A/E, and R108K/T. In some embodiments, the set of modifications comprises L10A, R86T, D87G, T88K, K89R, L101N, K104R, K105A, and R108K. In some embodiments, the set of modifications comprises L10A, R86T, D87G, T88K, K89R, L101N, K104R, K105E, and R108K. In some embodiments, the set of modifications comprises L10A, R86T, D87G, T88K, K89R, L101N, K104R, K105A, and R108T. In some embodiments, the set of modifications comprises L10A, R86T, D87G, T88K, K89R, L101N, K104R, K105E, and R108T. In some embodiments, the set of modifications comprises L10V, R86T, D87G, T88K, K89R, L101N, K104R, K105A, and R108K. In some embodiments, the set of modifications comprises L10V, R86T, D87G, T88K, K89R, L101N, K104R, K105E, and R108K. In some embodiments, the set of modifications comprises L10V, R86T, D87G, T88K, K89R, L101N, K104R, K105A, an dR108T. In some embodiments, the set of modifications comprises L10V, R86T, D87G, T88K, K89R, L101N, K104R, K105E, and R108T. In some embodiments, the amino acid sequence is 90% identical. In some embodiments, the amino acid sequence is 95% identical. In some embodiments, the amino acid sequence is 98% identical. In some embodiments, the amino acid sequence is 99% identical. In some embodiments, the polypeptide comprising the one or more modifications is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, amino acid substitutions include without limitation those provided in FIG. 4. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO:11; D10 SEQ ID NO: 12; E10 SEQ ID NO: 13; G8 SEQ ID NO:14; H4 SEQ ID NO:15; and H9 SEQ ID NO:16.


Specific sets of modifications that provide for greater selectivity in binding to IL-13Rα1 v IL-13Rα2 relative to a native IL-13 sequence may include, without limitation:

    • [L10V, V18I, D87S, D88S, L101F, K104R, K105T]
    • [R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T]
    • [L10V, V18I, D87S, T88S, L101F, K104R, K105T]
    • [L10V/I; D87S; T88S; K89R; L101H/F; K104R; K105T]
    • [L10I; V18I; R86T; D87G; T88S; K89R; L101Y/H; K104R; K105A]
    • [L10V; V18I; D87S; T88S; L101F; K104R; K105T]
    • [V181, R86T, D87G, T88S, L101Y, K104R, K105A]
    • [R11I, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M]
    • which substitutions are optionally combined with the substitutions [E12A/G/S, R65D/E].


In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, and K105T. In some embodiments, the set of modifications comprises R11S, V181, R86K, D87G, T88S, K89M, L101Y, K104R, and K105T. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, and K105T. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, and K105T. In some embodiments, the set of modifications comprises L10I, V181, R86T, D87G, T88S, K89R, L101Y/H, K104R, and K105A. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, and K105T. In some embodiments, the set of modifications comprises V18I, R86T, D87G, T88S, L101Y, K104R, and K105A. In some embodiments, the set of modifications comprises R11, V18I, R86K, D87G, T88S, L101H, K104R, K105A, and F107M. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises L10V, V181, D87S, T88S, L101F, K104R, K105T, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises V181, R86T, D87G, T88S, L101Y, K104R, K105A, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises R11I, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12A, and R65D/E. In some embodiments, the set of modifications comprises R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12A, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A, and R65D/E. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12A, and R65D/E. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12A, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A, and R65D/E. In some embodiments, the set of modifications comprises V18I, R86T, D87G, T88S, L101Y, K104R, K105A, E12A, and R65D/E. In some embodiments, the set of modifications comprises R11, V181, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12A, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12G, and R65D/E. In some embodiments, the set of modifications comprises R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12G, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12G, and R65D/E. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12G, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12G, and R65D/E. In some embodiments, the set of modifications comprises V181, R86T, D87G, T88S, L101Y, K104R, K105A, E12G, and R65D/E. In some embodiments, the set of modifications comprises R11I, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12G, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12S, and R65D/E. In some embodiments, the set of modifications comprises R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12A/G/S, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12S, and R65D/E. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12S, and R65D/E. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12S, and R65D/E. In some embodiments, the set of modifications comprises L10V, V181, D87S, T88S, L101F, K104R, K105T, E12S, and R65D/E. In some embodiments, the set of modifications comprises V18I, R86T, D87G, T88S, L101Y, K104R, K105A, E12S, and R65D/E. In some embodiments, the set of modifications comprises R11, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12S, and R65D/E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12A, and R65D. In some embodiments, the set of modifications comprises R11S, V181, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12A, and R65E. In some embodiments, the set of modifications comprises L10V, V181, D87S, T88S, L101F, K104R, K105T, E12A, and R65D. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12A, and R65D. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12A, and R65D. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A, and R65D. In some embodiments, the set of modifications comprises V18I, R86T, D87G, T88S, L101Y, K104R, K105A, E12A, and R65D. In some embodiments, the set of modifications comprises R11, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12A, and R65D. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12G, and R65D. In some embodiments, the set of modifications comprises R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12G, and R65D. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A/G/S, and R65D. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12G, and R65D. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12G, and R65D. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12G, and R65D. In some embodiments, the set of modifications comprises V181, R86T, D87G, T88S, L101Y, K104R, K105A, E12G, and R65D. In some embodiments, the set of modifications comprises R11, V181, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12G, and R65D. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12S, and R65D. In some embodiments, the set of modifications comprises R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12S, and R65D. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12S, and R65D. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12S, and R65D. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12S, and R65D. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12S, and R65D. In some embodiments, the set of modifications comprises V18I, R86T, D87G, T88S, L101Y, K104R, K105A, E12S, and R65D. In some embodiments, the set of modifications comprises Ri 11, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12S, and R65D. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12A, and R65E. In some embodiments, the set of modifications comprises R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12A, and R65E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A, and R65E. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12A, and R65E. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12A, and R65E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A, and R65E. In some embodiments, the set of modifications comprises V181, R86T, D87G, T88S, L101Y, K104R, K105A, E12A, and R65E. In some embodiments, the set of modifications comprises R11I, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12A, and R65E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, D88S, L101F, K104R, K105T, E12G, and R65E. In some embodiments, the set of modifications comprises R11S, V18I, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12G, and R65E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12A/G/S, and R65E. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12G, and R65E. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12G, and R65E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12G, and R65E. In some embodiments, the set of modifications comprises V18I, R86T, D87G, T88S, L101Y, K104R, K105A, E12G, and R65E. In some embodiments, the set of modifications comprises R11I, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12G, and R65E. In some embodiments, the set of modifications comprises L10V, V181, D87S, D88S, L101F, K104R, K105T, E12S, and R65E. In some embodiments, the set of modifications comprises R11S, V181, R86K, D87G, T88S, K89M, L101Y, K104R, K105T, E12A/G/S, and R65E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12S, and R65E. In some embodiments, the set of modifications comprises L10V/I, D87S, T88S, K89R, L101H/F, K104R, K105T, E12S, and R65E. In some embodiments, the set of modifications comprises L10I, V18I, R86T, D87G, T88S, K89R, L101Y/H, K104R, K105A, E12S, and R65E. In some embodiments, the set of modifications comprises L10V, V18I, D87S, T88S, L101F, K104R, K105T, E12S, and R65E. In some embodiments, the set of modifications comprises V18I, R86T, D87G, T88S, L101Y, K104R, K105A, E12S, and R65E. In some embodiments, the set of modifications comprises R11, V18I, R86K, D87G, T88S, L101H, K104R, K105A, F107M, E12S, and R65E. In some embodiments, the set of modifications comprises L10V, E12A, V18I, R65D, D87S, T88S, L101F, K104R, and K105T (see, for example, IL-13dn; SEQ ID NO:38). In some embodiments, the amino acid sequence is 90% identical. In some embodiments, the amino acid sequence is 95% identical. In some embodiments, the amino acid sequence is 98% identical. In some embodiments, the amino acid sequence is 99% identical. In some embodiments, the polypeptide comprising the one or more modifications is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, amino acid substitutions include without limitation those provided in FIG. 4. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5, 5-2 SEQ ID NO:6, 6-6 SEQ ID NO:7, A2 SEQ ID NO:8, BT SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO: 11, D10 SEQ ID NO: 12, E10 SEQ ID NO: 13; G8 SEQ ID NO: 14; H4 SEQ ID NO: 15; and H9 SEQ ID NO: 16.


Table of IL-13 sequences is provided below.









TABLE 8







List of IL-13 Amino Acid Sequences








SEQ ID NO: 



(Information)
Amino acid sequence





SEQ ID NO: 81
PGPVPPSTALRELIEELVNITQNQKAPLCNGSMVW


(IL-13 wildtype)
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVRDTKIEVAQFVKDLLLHLKK



LFREGQFN





SEQ ID NO: 82
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVW



SINRTAGMYCAALESLINVSGCSAIEKTQDMLSGF



CPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRT



LFREGQFN





SEQ ID NO: 83
PGPVPPSTAIRELIEELINITQNQKAPLCNGSMVW



SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVRGSKIEVAQFVKDLLHHLRA



LFREGQFN





SEQ ID NO: 84
PGPVPPSTAVRELIEELINITQNQKAPLCNGSMVW



SINRTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRT



LFREGQFN





SEQ ID NO: 85
PGPVPPSTALIELIEELINITQNQKAPLCNGSMVW



SINLTAGIYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVKGSKIEVAQFVKDLLHHLRA



LMREGQFN


SEQ ID NO: 86
PGPVPPSTAIRELIEELLNITQNQKAPLCNGSMVW






SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVMKSKIEVAQFVKDLLHHLRA



LFREGQFN





SEQ ID NO: 87
PGPVPPSTAIRELIEELINITQNQKAPLCNGSMVW



SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVRSSRIEVAQFVKDLLHHLRT



LFREGQFN





SEQ ID NO: 88
PGPVPPSTALRELIEELINITQNEKAPLCNGSMVW



SINLTAGIYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVTGSKIEVAQFVKDLLYHLRA



LFREGQFN





SEQ ID NO: 89
PGPVPPSTALSELIEELINITQNQKAPLCNGSMVW



SINPTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVAAGQFSSLHDKGSMIEVAQFVKDLLYHLRT



LFREGQFN





SEQ ID NO: 90
PGPVPPSTATRELIEELINITQNQKAPLCNGSMVW



SINLTADMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSVGQFSSLHVRGSKIEVAQFVKDLLYHLRT



LFREGQFN





SEQ ID NO: 91
PGPVPPSTADIELIAELINITQNQKAPLCNGSMVW



SINLTADMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVKKTRIEVAQFVKDLLLHLKK



LFKEGQFN





SEQ ID NO: 92
PGPVPPSTAARELIEELVNITQNQKAPLCNGSMVW



SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQLSSLHVTGKRIEVAQFVKDLLNHLRA



LFKEGQFN





SEQ ID NO: 93
PGPVPPSTAVRELIEELVNITQNQKAPLCNGSMVW



SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVRDTRIEVAQFVKDLLNHLKE



LFTEGQFN





SEQ ID NO: 94
PGPVPPSTALSELMEELVNITQNQKAPLCNGSMVW



SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVRDSKIEVAQFVKDLLNHLKA



LFKEGQFN





SEQ ID NO: 95
GPVPPSTAFRELIEELVNITQNQKAPLCNGSMVWS



INLTAGMYCAALESLINVSGCSAIEKTQRMLSGFC



PHKVSPGQFSSLHVTNSRIEVAQFVKDLLNHLKAL



FKEGQYN





SEQ ID NO: 96
GPVPPSTAHLELIEELINITQNQKAPLCNGSMVWS



INLTAGMYCAALESLINVSGCSAIEKTQRMLSGFC



PHKVSAGQFSSLHVKETRIEVAQFVKDLLNHLKTL



FKEGQFN





SEQ ID NO: 97
PGPVPPSTAHLELIEELINITQNQKAPLCNGSMVW



SINPTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVMDTRIEVAQFVKDLLLHLKK



LFKEGQFN





SEQ ID NO: 98
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVW



SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLHLKK



LFKEGQFN





SEQ ID NO: 99
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVW



RINRTAGMYCAALESLINVSGCSAIEKTQRMLSGF



CPHKVSAGQFSSLHVMDSRIEVAQFVKDLLNHLRA



LFKEGQFN





SEQ ID NO: 100
PGPVPPSTAARELIEELFNITQNQKAPLCNGSMVW



SINLTAGMYCAALESLINVSGCSAIEKTKRMLSGF



CPHKVSAGQFPSLHVKKTRIEVAQFVKDLLIHLRK



LFKEGQFN





SEQ ID NO: 101
PGPVPPSTALIELIEELINITQNQKAPLCNGSMVWS


(Exemplary sequence
INLTAGMYCAALESLINVSGCSAIEKTQRMLSGFCP


comprising R11I, V18I, R86K,
HKVSAGQFSSLHVKGSKIEVAQFVKDLLHHLRALMR


D87G, T88S, L101H, K104R,
EGQFN


K105A, F107M, referred to



herein as A5)






SEQ ID NO: 102
PGPVPPSTAIRELIEELLNITQNQKAPLCNGSMVWS


(Exemplary sequence
INLTAGMYCAALESLINVSGCSAIEKTQRMLSGFCP


comprising L10I, V18L, R86M,
HKVSAGQFSSLHVMKSKIEVAQFVKDLLHHLRALFR


D87K, T88S, L101H, K104R,
EGQFN


K105A, referred to herein



as A6)






SEQ ID NO: 103
PGPVPPSTAIRELIEELINITQNQKAPLCNGSMVWS


(Exemplary sequence
INLTAGMYCAALESLINVSGCSAIEKTQRMLSGFCP


comprising L10I, V18I, D87G,
HKVSAGQFSSLHVRGSKIEVAQFVKDLLHHLRALFR


T88S, L101H, K104R, K105A,
EGQFN


referred to herein as A7)






SEQ ID NO: 104
PGPVPPSTAIRELIEELINITQNQKAPLCNGSMVWS


(Exemplary sequence
INLTAGMYCAALESLINVSGCSAIEKTQRMLSGFCP


comprising L10I, V18I, D87S,
HKVSAGQFSSLHVRSSRIEVAQFVKDLLHHLRTLFR


T88S, K89R, L101H, K104R,
EGQFN


K105T; referred to herein



as A8)






SEQ ID NO: 105
PGPVPPSTAVRELIEELINITQNQKAPLCNGSMVWS


(Exemplary sequence
INcustom-character TAGMYCAALESLINVSGCSAIEKTQRMLSGFCP


comprising L10V, V18I, D87S,
HKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRTLFR


T88S, L101F, K104R, K105T,
EGQFN


referred to herein as A11



variant 1)






SEQ ID NO: 105
PGPVPPSTAVRELIEELINITQNQKAPLCNGSMVWS


(Exemplary sequence
INcustom-character TAGMYCAALESLINVSGCSAIEKTQRMLSGFCP


comprising L10V, V18I, D87S,
HKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRTLFR


T88S, L101F, K104R, K105T,
EGQFN


referred to herein as A11



variant 2)






SEQ ID NO: 106
PGPVPPSTALRELIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising V18I, R86T, D87G,
CPHKVSAGQFSSLHVTGSKIEVAQFVKDLLYHLRA


T88S, L101Y, K104R, K105A,
LFREGQFN


referred to herein as B2)






SEQ ID NO: 107
PGPVPPSTALSELIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising R11S, V18I, R86K,
CPHKVSAGQFSSLHVKGSMIEVAQFVKDLLYHLRT


D87G, T88S, K89M, L101Y,
LFREGQFN


K104R, K105T, referred to



herein as B4)






SEQ ID NO: 108
PGPVPPSTATRELIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10T, V18I, D87G,
CPHKVSAGQFSSLHVRGSKIEVAQFVKDLLYHLRT


T88S, K89K, L10Y1, K104R,
LFREGQFN


K105T, referred to herein as B6)






SEQ ID NO: 109
PGPVPPSTADIELIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10D, R11I, V18I,
CPHKVSAGQFSSLHVKKTRIEVAQFVKDLLLHLKK


R86K, D87K, K89R, R108K,
LFKEGQFN


referred to herein as C2)






SEQ ID NO: 110
PGPVPPSTAARELIEELVNITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10A, R86T, D87G,
CPHKVSAGQFSSLHVTGKRIEVAQFVKDLLNHLRA


T88K, K89R, L101N, K104R,
LFKEGQFN


K105A, R108K, referred to



herein as C3)






SEQ ID NO: 111
PGPVPPSTAVRELIEELVNITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10V, K89R, L101N,
CPHKVSAGQFSSLHVRDTRIEVAQFVKDLLNHLKE


K105E, R108T, referred to
LFTEGQFN


herein as C4)






SEQ ID NO: 112
PGPVPPSTALSELMEELVNITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising R11S, I14M, T88S,
CPHKVSAGQFSSLHVRDSKIEVAQFVKDLLNHLKA


L101N, K105A, R108K, referred
LFKEGQFN


to herein as C7)






SEQ ID NO: 113
PGPVPPSTAHLELIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10H, R11L, V18I,
CPHKVSAGQFSSLHVKETRIEVAQFVKDLLNHLKT


R86K, D87E, K89R, L101N,
LFKEGQFN


K105T, R108K, refered to herein



as C9)






SEQ ID NO: 114
PGPVPPSTAHLELIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10H, R11L, V18I,
CPHKVSAGQFSSLHVMDTRIEVAQFVKDLLLHLKK


R86M, K89R, R108K, referred to
LFKEGQFN


herein as C10)






SEQ ID NO: 115
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10H, R86T, D87G,
CPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLHLKK


T88R, R108K, referred to herein
LFKEGQFN


as C11)






SEQ ID NO: 116
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10H, R86M, T88S,
CPHKVSAGQFSSLHVMDSRIEVAQFVKDLLNHLRA


K89R, L101N, K104R, K105A,
LFKEGQFN


R108K, referred to herein as



C12)






SEQ ID NO: 117
PGPVPPSTAARELIEELFNITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQRMLSGF


comprising L10A, V18F, R86F,
CPHKVSAGQFSSLHVKKTRIEVAQFVKDLLIHLRK


D87F, K89R, L101I, K104R,
LFKEGQFN


R108K, referred to herein as D7)






SEQ ID NO: 118
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINLTAGMYCAALESLINVSGCSAIEKTQDMLSGF


comprising L10V, E12A, V18I,
CPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRT


R65D, D87S, T88S, L101F,
LFREGQFN


K104R, K105T, referred to



herein as IL-13dn)






SEQ ID NO: 119


MHPLLNPLLLALGLMALLLTTVIALTCLGGFAS
PG



**signal peptide**
PVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSI



NLTAGMYCAALESLINVSGCSAIEKTQRMLSGFCP



HKVSAGQFSSLHVTGRKIEVAQFVKDLLLHLKKLF



KEGQFN





SEQ ID NO: 120
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINcustom-character TAGMYCAALESLINVSGCSAIEKTQDMLSGF


comprising L10V, E12A, V18I,
CPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRT


R65D, D87S, T88S, L101F,
LFREGQFN


K104R, K105T, referred to



herein as IL-13DN variant 1)






SEQ ID NO: 121
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVW


(Exemplary sequence
SINcustom-character TAGMYCAALESLINVSGCSAIEKTQDMLSGF


comprising L10V, E12A, V18I,
CPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRT


R65D, D87S, T88S, L101F,
LFREGQFN


K104R, K105T, referred to



herein as IL-13DN variant 2)






SEQ ID NO: 122
MPGPVPPSTALRELIEELVNITQNQKAPLCNGSMV


wild-type IL-13 including an
WSINLTAGMYCAALESLINVSGCSAIEKTQRMLSG


additional methionine at the N-
FCPHKVSAGQFSSLHVRDTKIEVAQFVKDLLLHLK


terminus
KLFREGQFN





SEQ ID NO: 123
MYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSA


circularly permuted IL-13
GQFSSLHVRDTKIEVAQFVKDLLLHLKKLFREGQF



NGGSGPGPVPPSTALRELIEELVNITQNQKAPLCN



GSMVWSINLTAG





SEQ ID NO: 124
MYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSA


Circularly permuted IL-13
GQFSSLHVRDTKIEVAQFVKDLLLHLKKLFREGQF



NGGSGMPGPVPPSTALRELIEELVNITQNQKAPLC



NGSMVWSINLTAG





SEQ ID NO: 125
MYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSA


circularly permuted IL-13 ″A11″
GQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQF


variant
NGGSGPGPVPPSTAVRELIEELINITQNQKAPLCN



GSMVWSINRTAG





SEQ ID NO: 126
MYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSA


circularly permuted IL-13
GQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQF



NGGSGMPGPVPPSTAVRELIEELINITQNQKAPLC



NGSMVWSINRTAG





SEQ ID NO: 127
MYCAALESLINVSGCSAIEKTQDMLSGFCPHKVSA


circularly permuted IL-13 ″DN″
GQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQF


variant
NGGSGPGPVPPSTAVRALIEELINITQNQKAPLCN



GSMVWSINLTAG





SEQ ID NO: 128
MYCAALESLINVSGCSAIEKTQDMLSGFCPHKVSA


circular permuted IL-13
GQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQF



NGGSGMPGPVPPSTAVRALIEELINITQNQKAPLC



NGSMVWSINLTAG









Any IL-4 sequence or variant thereof can be used in a fusion with an IL-2 mutein or variant, including those as described herein. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO:10; C5 SEQ ID NO: 11; D10 SEQ ID NO:12; E10 SEQ ID NO:13; G8 SEQ ID NO: 14; H4 SEQ ID NO:15; and H9 SEQ ID NO:16. Exemplary polypeptide sequences are provided in SEQ ID NO:130-SEQ ID NO:135, including any of those provided herein. In some embodiments, the IL-4 polypeptide sequence is as provided in any one of SEQ ID NO:130 through SEQ ID NO:135. In some embodiments, the IL-4 polypeptide sequence is SEQ ID NO:130. In some embodiments, the IL-4 polypeptide sequence is SEQ ID NO:131. In some embodiments, the IL-4 polypeptide sequence is SEQ ID NO:132. In some embodiments, the IL-4 polypeptide sequence is SEQ ID NO:133. In some embodiments, the IL-4 polypeptide sequence is SEQ ID NO:134. In some embodiments, the IL-4 polypeptide sequence is SEQ ID NO:135. In some embodiments, the IL-4 polypeptide sequence is 98% identical to any one of SEQ ID NO: 130 through SEQ ID NO:135. In some embodiments, the IL-4 polypeptide sequence is 99% identical to any one of SEQ ID NO:130 through SEQ ID NO: 135. In some embodiments, any one of SEQ ID NO:130-SEQ ID NO:135 are linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:130 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:131 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:132 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:133 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:134 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO:135 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO:11; D10 SEQ ID NO: 12; E10 SEQ ID NO:13; G8 SEQ ID NO:14; H4 SEQ ID NO: 15; and H9 SEQ ID NO:16.


Table of IL-4 sequences is provided below.









TABLE 9







List of IL-4 Amino Acid Sequences








SEQ ID NO: 



(Information)
Amino acid sequence





SEQ ID NO: 129


MGLTSQLLPPLFFLLACAGNFVHGH
KCDITLQEII



(IL-4 wildtype with signal
KTLNSLTEQKTLCTELTVTDIFAASKNTTEKETFC


peptide)
RAATVLRQFYSHHEKDTRCLGATAQQFHRHKQLIR



FLKRLDRNLWGLAGLNSCPVKEANQSTLENFLERL



KTIMREKYSKCSS





SEQ ID NO: 130
MHKCDITLQEIIKTLNSLTEQKTLCTELTVTDIFA


IL-4 including an additional
ASKDTTEKETFCRAATVLRQFYSHHEKDTRCLGAT


methionine at the N-terminus″
AQQFHRHKQLIRFLKRLDRNLWGLAGLNSCPVKEA


starting
NQSTLENFLERLKTIMREKYSKCSS





SEQ ID NO: 131
KCDITLQEIIKTLNSLTEQKTLCTELTVTDIFAAS


KFR
KNTTEKETFCRAATVLRQFYSHHEKDTRCLGATAQ



QFHRHKQLIRFLKRLDRNLWGLAGLNSCPVKEANQ



STLENFLERLKTIMKEKFRKCSS





SEQ ID NO: 132
MDTTEKETFCRAATVLRQFYSHHEKDTRCLGATAQ


RGA
QFHRHKQLIRFLKRLDRNLWGLAGLNSCPVKEANQ



STLENFLERLRVIMQSKWFKCGAGGNGGHKCDITL



QEIIKTLNSLTEQKTLCTELTVTDIFAAS





SEQ ID NO: 133
MDTTEKETFCRAATVLRQFYSHHEKDTRCLGATAQ


cirularly permuted wild-type IL-4
QFHRHKQLIRFLKRLDRNLWGLAGLNSCPVKEANQ



STLENFLERLKTIMREKYSKCSSGGNGGHKCDITL



QEIIKTLNSLTEQKTLCTELTVTDIFAAS





SEQ ID NO: 134
MDTTEKETFCRAATVLRQFYSHHEKDTRCLGATAQ


circularly permuted ″KFR″ IL-4
QFHRHKQLIRFLKRLDRNLWGLAGLNSCPVKEANQ


variant
STLENFLERLKTIMKEKFRKCSSGGNGGHKCDITL



QEIIKTLNSLTEQKTLCTELTVTDIFAASRQFYSH



HEKDTRCLGATAQQFHRHKQLIRFLKRLDRNLWGL



AGLNSCPVKEANQSTLENFLERLRVIMQSKWFKCG



AGGNGGHKCDITLQEIIKTLNSLTEQKTLCTELTV



TDIFAAS





SEQ ID NO: 135
MDTTEKETFCRAATVLRQFYSHHEKDTRCLGATAQ


circularly permuted ″KF″ IL-4
QFHRHKQLIRFLKRLDRNLWGLAGLNSCPVKEANQ


variant
STLENFLERLKTIMKEKFKCSSGGNGGHKCDITLQ



EIIKTLNSLTEQKTLCTELTVTDIFAAS









In some embodiments, an IL-2 mutein can be fused to an IL-10, IL-12, IL-15, and/or IL-18 sequence. In some embodiments, such fusions function to specifically target the fusion construct to NK cells and/or CD8+ cells. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8, BT SEQ ID NO:9; BT 1 SEQ ID NO:10; C5 SEQ ID NO:11; D10 SEQ ID NO:12; ET SEQ ID NO:13; G8 SEQ ID NO:14; H4 SEQ ID NO: 15 and H9 SEQ ID NO 16 In some embodiments, SEQ ID NO 136 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO: 137 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO: 138 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO: 139 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO: 140 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, SEQ ID NO: 141 is linked to an IL-2 or IL-2 mutein as described herein. In some embodiments, the IL-2 mutein can be fused to an IL-IL-10, IL-12, IL-15, and/or IL-18 sequence as provided in the table below, in SEQ ID NOs: 136-141.









TABLE 10







Exemplary IL-10, IL-12, IL-15, and/or IL-18 Sequences








SEQ ID NO: 
Amino Acid Sequence





SEQ ID NO: 136
MHSSALLCCLVLLTGVRASPGQGTQSENSCTHFPGNLPNMLRDLRDAFS


IL-10
RVKTFFQMKDQLDNLLLKESLLEDFKGYLGCQALSEMIQFYLEEVMPQA


(Uniprot sp|P22301|)
ENQDPDIKAHVNSLGENLKTLRLRLRRCHRFLPCENKSKAVEQVKNAFN



KLQEKGIYKAMSEFDIFINYIEAYMTMKIRN





SEQ ID NO: 137
MCPARSLLLVATLVLLDHLSLARNLPVATPDPGMFPCLHHSQNLLRAVS


IL-12A
NMLQKARQTLEFYPCTSEEIDHEDITKDKTSTVEACLPLELTKNESCLN


(Uniprot sp|P29459|)
SRETSFITNGSCLASRKTSFMMALCLSSIYEDLKMYQVEFKTMNAKLLM



DPKRQIFLDQNMLAVIDELMQALNFNSETVPQKSSLEEPDFYKTKIKLC



ILLHAFRIRAVTIDRVMSYLNAS





SEQ ID NO: 138
MCHQQLVISWFSLVFLASPLVAIWELKKDVYVVELDWYPDAPGEMVVLT


IL-12B
CDTPEEDGITWTLDQSSEVLGSGKTLTIQVKEFGDAGQYTCHKGGEVLS


(Uniprot sp|P29460)
HSLLLLHKKEDGIWSTDILKDQKEPKNKTFLRCEAKNYSGRFTCWWLTT



ISTDLTFSVKSSRGSSDPQGVTCGAATLSAERVRGDNKEYEYSVECQED



SACPAAEESLPIEVMVDAVHKLKYENYTSSFFIRDIIKPDPPKNLQLKP



LKNSRQVEVSWEYPDTWSTPHSYFSLTFCVQVQGKSKREKKDRVFTDKT



SATVICRKNASISVRAQDRYYSSSWSEWASVPCS





SEQ ID NO: 139
MRISKPHLRSISIQCYLCLLLNSHFLTEAGIHVFILGCFSAGLPKTEAN


IL-15
WVNVISDLKKIEDLIQSMHIDATLYTESDVHPSCKVTAMKCFLLELQVI


(Uniprot sp|P40933|)
SLESGDASIHDTVENLIILANNSLSSNGNVTESGCKECEELEEKNIKEF



LQSFVHIVQMFINTS





SEQ ID NO: 140


MAAEPVEDNCINFVAMKFIDNTLYFIAEDDENLESD
YFGKLESKLSVIR



IL-18
NLNDQVLFIDQGNRPLFEDMTDSDCRDNAPRTIFIISMYKDSQPRGMAV


(Uniprot
TISVKCEKISTLSCENKIISFKEMNPPDNIKDTKSDIIFFQRSVPGHDN


sp|Q14116|)
KMQFESSSYEGYFLACEKERDLFKLILKKEDELGDRSIMFTVQNED





SEQ ID NO: 141
YFGKLESKLSVIRNLNDQVLFIDQGNRPLFEDMTDSDCRDNAPRTIFII


IL-18
SMYKDSQPRGMAVTISVKCEKISTLSCENKIISFKEMNPPDNIKDTKSD


(mature)
IIFFQRSVPGHDNKMQFESSSYEGYFLACEKERDLFKLILKKEDELGDR



SIMFTVQNED









The sequences of exemplary IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7, A2 SEQ ID NO:8, BT SEQ ID NO:9; BT 1 SEQ ID NO: 10, C5 SEQ ID NO: 11, D10 SEQ ID NO: 12, E10 SEQ ID NO: 13, G8 SEQ ID NO: 14; H4 SEQ ID NO: 15; and H9 SEQ ID NO: 16.


In some embodiments, the cytokine-cytokine fusion is one of those included in the table below.









TABLE 11







List of Exemplary IL-2 Fusion Amino Acid Sequences








SEQ ID NO: 



(Information)
Amino acid sequence





SEQ ID NO: 142
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMY


IL-13 variant-H9 (linker in
CAALESLINVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSS


bold and underlined)
KIEVAQFVKDLLFHLRTLFREGQFNGGGGSGGGGSGGGGSAPTS



SSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPK



KATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVF



VLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 143
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMY


IL-13 variant-H9 (linker in
CAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGR


bold and underlined)
KIEVAQFVKDLLLHLKKLFKEGQFNGGGGSGGGGSGGGGSAPTS



SSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPK



KATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVF



VLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLT





SEQ ID NO: 144
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKF


H9-IL-12 (linker in bold and
YMPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSN


underlined)
INVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTL



TGGGGSGGGGSGGGGSRNLPVATPDPGMFPCLHHSQNLLRAVSN



MLQKARQTLEFYPCTSEEIDHEDITKDKTSTVEACLPLELTKNE



SCLNSRETSFITNGSCLASRKTSFMMALCLSSIYEDLKMYQVEF



KTMNAKLLMDPKRQIFLDQNMLAVIDELMQALNFNSETVPQKSS



LEEPDFYKTKIKLCILLHAFRIRAVTIDRVMSYLNASIWELKKD



VYVVELDWYPDAPGEMVVLTCDTPEEDGITWTLDQSSEVLGSGK



TLTIQVKEFGDAGQYTCHKGGEVLSHSLLLLHKKEDGIWSTDIL



KDQKEPKNKTFLRCEAKNYSGRFTCWWLTTISTDLTFSVKSSRG



SSDPQGVTCGAATLSAERVRGDNKEYEYSVECQEDSACPAAEES



LPIEVMVDAVHKLKYENYTSSFFIRDIIKPDPPKNLQLKPLKNS



RQVEVSWEYPDTWSTPHSYFSLTFCVQVQGKSKREKKDRVFTDK



TSATVICRKNASISVRAQDRYYSSSWSEWASVPCS





SEQ ID NO: 145
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKF


H9-IL-18 (linker in bold and
YMPKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSN


underlined)
INVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTL



TGGGGSGGGGSGGGGSYFGKLESKLSVIRNLNDQVLFIDQGNRP



LFEDMTDSDCRDNAPRTIFIISMYKDSQPRGMAVTISVKCEKIS



TLSCENKIISFKEMNPPDNIKDTKSDIIFFQRSVPGHDNKMQFE



SSSYEGYFLACEKERDLFKLILKKEDELGDRSIMFTVQNED





SEQ ID NOs: 148 and 213
Gene1: 


KIH MDNA132-Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTA


(1:1:1:)
GMYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFS


Fc heteroduplex with MW of
SLHVTGRKIEVAQFVKDLLLHLKKLFKEGQFNGGGGSGGG


80.8 KDa
GSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI



SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPR



EEQYASTYRWVSVLTVLHQDWLNGKEYKCKVSNKALPAPI



EKTISKAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKG



FYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT



VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGG



SGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDL



QMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEEA



LKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTF



MCEYADETATIVEFLNRWITFCQSIISTLT (SEQ ID NO: 



148)



Gene2: 



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTC



VVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTY



RVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK



GQPREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAV



EWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQ



QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 



213)





SEQ ID NOs: 149 and 213
Gene1: 


MDNA413-Fc-MDNA109 (KIH)
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTA


Fc heteroduplex with MW of
GMYCAALESLINVSGCSAIEKTQDMLSGFCPHKVSAGQFS


80.8 KDa
SLHVRSSKIEVAQFVKDLLFHLRTLFREGQFNGGGGSGG



GGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTL



MISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTK



PREEQYASTYRWVSVLTVLHQDWLNGKEYKCKVSNKALP



APIEKTISKAKGQPREPQVYTLPPCREEMTKNQVSLWCLV



KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSK



LTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGG



GGSGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLL



DLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLE



EELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSET



TFMCEYADETATIVEFLNRWITFCQSIISTLT (SEQ ID NO: 



149)



Gene2



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTC



VVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTY



RVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK



GQPREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAV



EWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQ



QGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 



213)





SEQ ID NO: 150
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLT


MDNA109FEAA-Fc-MDNA413
AKFYMPKKATELKHLQCLEEALKPLEEVLNLAQSKNFHFD


Fc homodimer with MW of
PRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWI


109.8 KDa
TFCQSIISTLTGGGGSGGGGSGGGGSDKTHTCPPCPAPE



LLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV



KFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLAPTSSS



TKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMP



KKATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDWVS



NINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSII



STLTGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYV



DGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE



YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMT



KNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD



SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ



KSLSLSPGGGGGSGGGGSGGGGSPGPVPPSTAVRALIEE



LINITQNQKAPLCNGSMVWSINRTAGMYCAALESLINVSGC



SAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVK



DLLFHLRTLFREGQFN









In some embodiments, the cytokine-cytokine fusion is one of those included in the table below (see Table 12, as well as FIG. 54).









TABLE 12







List of Bispecific Fusion Constructs








Specific



Construct
Gene sequence





SEQ ID NOS: 151
Gene 1: 


and 214
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY


Fc-MDNA132 (1:1
VDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI


KIH)
SKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKT



TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGG



GSGGGGSGGGGSPGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYC



AALESLINVSGCSAIEKTORMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLHLK



KLFKEGQFN (SEQ ID NO: 151)



Gene 2: 



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY



VDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKT



TPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID



NO: 214)





SEQ ID NO: 152
Gene 1 and Gene 2: 


Fc-A11 (1:2)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY


version 1
VDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKT



TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGG



GSGGGGSGGGGSPGPVPPSTAVRELIEELINITQNQKAPLCNGSMVWSINRTAGMYC



AALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLR



TLFREGQFN





SEQ ID NO: 153
Gene 1 and Gene 2: 


Fc-A11 (1:2)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY


version 2
VDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKT



TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGG



GSGGGGSGGGGSPGPVPPSTAVRELIEELINITQNQKAPLCNGSMVWSINLTAGMYC



AALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLR



TLFREGQFN





SEQ ID NO: 154
Gene 1 and Gene 2: 


Fc-MDNA413
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY


(1:2)
VDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI


version 1
SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKT



TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGG



GSGGGGSGGGGSPGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYC



AALESLINVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLR



TLFREGQFN





SEQ ID NO: 155
Gene 1 and Gene 2: 


Fc-MDNA413
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY


(1:2)
VDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI


version 2
SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKT



TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGG



GSGGGGSGGGGSPGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINLTAGMYC



AALESLINVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLR



TLFREGQFN





SEQ ID NO: 156
GENE 1 & 2: 


Fc4-MDNA413
APPCPSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDGVE


(1:2)
VHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPR



EPQVYTLPPSQEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFF



LYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSPGKGGGSGSPGPVPPSTAVRAL



IEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLINVSGCSAIEKTQDMLSGFCPHK



VSAGQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQFN





SEQ ID NOS: 157
Gene 1: 


and 213
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLINVSGCSAIE


MDNA413-Fc (1:1
KTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQFNGGGGSGGGG


KIH)
SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF



NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID



NO: 157)



Gene 2: 



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG



VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 



213)





SEQ ID NO: 158
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEE


MDNA109-Fc
ELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWI


(2:1)
TFCQSIISTLTGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISR



TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEYASTYRVVSVLTVLHQDWLNGK



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIA



VEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK



SLSLSPGK





SEQ ID NO: 159
Gene 1 and Gene 2: 


MDNA-109FEAA-Fc
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEE


(2:1)
ALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWI



TFCQSIISTLTGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISR



TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNG



KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDI



AVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ



KSLSLSPGK





SEQ ID NO: 160
Gene 1 and Gene 2: 


Fc-MDNA109
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG


(1:2)
VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSAPT



SSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELK



PLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFC



QSIISTLT





SEQ ID NOS: 161
Gene 1: 


and 215
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEE


MDNA109FEAA-Fc-
ALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWI


MDNA132 (2:1:1
TFSQSIISTLTGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISR


KIH)
TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNG



KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDI



AVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ



KSLSLSPGGGGGSGGGGSGGGGSPGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSIN



LTAGMYCAALESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDL



LLHLKKLFKEGQFN (SEQ ID NO: 161)



Gene 2: 



APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEE



ALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWI



TFSQSIISTLTGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISR



TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNG



KEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDI



AVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ



KSLSLSPG (SEQ ID NO: 215)



MDNA109FEAA sequence in green.



MDNA132 sequence in blue.



Fc Sequence in black; (G4S)3 linker underlined.





SEQ ID NOS: 162
Gene 1: 


MDNA413-FC-
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLINVSGCSAIE


MDNA132 (2:1:1
KTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQFNGGGGSGGGG


KIH)

SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF



and 216
NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGG




GGSPGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLINVSGCS




AIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLHLKKLFKEGQFN (SEQ



ID NO: 162)



Gene 2: 



PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLINVSGCSAIE



KTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQFNGGGGSGGGG




SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF




NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 



216)



MDNA413 sequence in red.



MDNA132 sequence in blue.



Fc Sequence in black; (G4S)3 linker underlined.





SEQ ID NO: 163
MDNA109FEAA-Fc-MDNA413 (2:1:2)- SEQUENCE FROM CRO


MDNA109FEAA-Fc-
PR00376


MDNA413 (2:1:2)-
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEE


version 1
ALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWI



TFCQSIISTLTGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISR



TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNG



KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDI



AVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ



KSLSLSPGGGGGSGGGGSGGGGSPGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSIN



RTAGMYCAALESLINVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDL



LFHLRTLFREGQFN





SEQ ID NO: 164
Gene 1 and Gene 2


MDNA109FEAA-FC-
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEE


MDNA413 (2:1:2)-
ALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWI


version 2
TFSQSIISTLTGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISR



TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNG



KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDI



AVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQ



KSLSLSPGGGGGSGGGGSGGGGSPGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSIN



RTAGMYCAALESLINVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDL



LFHLRTLFREGQFN



MDNA109FEAA sequence in green.



MDNA413 sequence in red.



Fc Sequence in black; (G4S)3 linker underlined.





SEQ ID NOS: 165
MDNA132-Fc-MDNA109 (1:1:1 KiH) SEQUENCE FROM CRO


and 213
PR00328


MDNA132-FC-
Gene 1: 


MDNA109 (1:1:1
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLINVSGCSAIE


KIH)
KTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLHLKKLFKEGQFNGGGGSGGGG



SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF



NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGSGGGGGSGG



GGSGGGGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATEL



KHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATI



VEFLNRWITFCQSIISTLT (SEQ ID NO: 165)



Gene 2: 



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG



VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 213)





SEQ ID NOS: 166
MDNA413-Fc-MDNA109 (1:1:1 KiH)-SEQUENCE FROM CRO


and 213
PR00331


MDNA413-Fc-
Gene1: 


MDNA109 (1:1:1
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLINVSGCSAIE


KIH)
KTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQFNGGGGSGGGG



SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF



NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGSGGGGGSGG



GGSGGGGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATEL



KHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATI



VEFLNRWITFCQSIISTLT (SEQ ID NO: 166)



Gene2: 



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG



VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 



213)





SEQ ID NOS: 167
Gene 1


and 217
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLINVSGCSAIE


MDNA132-Fc-
KTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLHLKKLFKEGQFNGGGGSGGGG


MDNA109FEAA

SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF



(1:1:1KIH)
NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGSGGGGGSGG




GGSGGGGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATEL




KHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATI



VEFLNRWITFCQSIISTLT (SEQ ID NO: 167)



Gene 2



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG



VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 



217)



MDNA132 sequence in blue



MDNA109FEAA sequence in green



Fc (KIH) sequence in black; (G4S) 3 linker underlined





SEQ ID NO: 168
Gene 1 and Gene 2


Fc-MDNA413 (1:2)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG


Version A
VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ



PREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGScustom-character




custom-character
custom-character
custom-character





custom-character
custom-character






SEQ ID NO: 169
Gene 1


Fc-MDNA132 (1:1
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG


KIH)
VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ


Version A
PREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGScustom-character




custom-character
custom-character
custom-character





custom-character
custom-character






SEQ ID NO: 170
Gene 1


Fc-MDNA132 (1:1
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG


KIH)
VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ


Version B
PREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGScustom-character




custom-character
custom-character
custom-character





custom-character
custom-character






SEQ ID NO: 171
Gene 2


FC-MDNA132 (1:1
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG


KIH)
VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ


Version A
PREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS



FFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG





SEQ ID NO: 172
Gene 1


MDNA109FEAA-FC-
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEEALKPLEE


MDNA413 (2:1:2)
VLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFSQSIISTLTGGG


Version A
GSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN



WYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPR



EPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV



DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGScustom-character




custom-character
custom-character
custom-character





custom-character






SEQ ID NO: 173
Gene 1


MDNA109FEAA-FC-
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLEEALKPLEE


MDNA413 (2:1:2)
VLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRWITFCQSIISTLTGGG


Version B
GSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN



WYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPR



EPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV



DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGScustom-character




custom-character
custom-character
custom-character





custom-character






SEQ ID NO: 174
Gene 1


MDNA132-Fc-

custom-character
custom-character
custom-character



MDNA109 (1:1:1

custom-character
custom-character
custom-character



KIH)
SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF


Version A
NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGG



GGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQC



LEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLN



RWITFSQSIISTLT





SEQ ID NO: 175
Gene 1


MDNA132-Fc-

custom-character
custom-character
custom-character



MDNA109 (1:1:1

custom-character
custom-character
custom-character



KIH)
SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF


Version B
NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGG



GGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQC



LEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLN



RWITFCQSIISTLT





SEQ ID NO: 176
Gene 1


MDNA132-Fc-

custom-character
custom-character
custom-character



MDNA109 (1:1:1

custom-character
custom-character
custom-character



KIH)
SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF


Version C
NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGG



GGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQC



LEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLN



RWITFSQSIISTLT





SEQ ID NO: 177
Gene 1


MDNA132-Fc-

custom-character
custom-character
custom-character



MDNA109 (1:1:1

custom-character
custom-character
custom-character



KIH)
SGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF


Version D
NWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI



SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPV



LDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGG



GGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQC



LEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLN



RWITFCQSIISTLT





SEQ ID NO: 178
Gene 2


MDNA132-Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG


MDNA109 (1: 1: 1
VEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ


KIH)
PREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS


Version A
FFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK
















TABLE 13







List of Bispecific Fusion Constructs with Mutations Specified









Name
Sequence
Mutations





Fc-MDNA413
Gene 1 and Gene 2 (Version A)
MDNA413


(1:2)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
mutations:



DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
L10V, E12A,



YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
V18I, L39R,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
R65D, D87S,



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV
T88S, L101F,



PPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLIN
K104R, K105T



VSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFH




LRTLFREGQFN (SEQ ID NO: 168)



Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
L10V, E12A,


MDNA413R39/Q111
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
V18I, L39R,


(1:2)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
R65D, D87S,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
T88S, L101F,



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV
K104R, K105T




PPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALESLIN






VSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

H






L

RT

LFREGQFN (SEQ ID NO: 218)







Fc-MDNA132
Gene 1 (Version A)
MDNA132


(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
mutations:



DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
L10H, R86T,



YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
D87G, T88R,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
R108K



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV




PPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN




VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLH




LKKLFKEGQFN (SEQ ID NO: 169)






Fc-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
D87G, T88R,


(1:1
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
R108K


KIH)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC




LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV





PPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN






VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKDLLLH






LKKLF

K

EGQFN (SEQ ID NO: 219)







Fc-MDNA132L39/R111
Gene 1:
L10H, R86T,


(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
D87G, T88R,



DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
R108K



YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWC




LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV





PPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSIN

L

TAGMYCAALESLIN






VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKDLLLH






LKKLF

K

EGRFN (SEQ ID NO: 220)







Fc-MDNA132
Gene 1 (Version B)
MDNA132


(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
mutations:



DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
L10H, L39R,



YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
R86T, D87G,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
T88R, R108K



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV




PPSTAHRELIEELVNITQNQKAPLCNGSMVWSINRTAGMYCAALESLIN




VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLH




LKKLFKEGQFN (SEQ ID NO: 170)






Fc-MDNA132
Gene 2 (Version A)



(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 171)






Fc-
Gene 2:



MDNA132L39/Q111
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



(1:1
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE



KIH)
YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 221)






Fc -MDNA132L39/R111
Gene 2:



(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 222)






MDNA109FEA
Gene 1 (Version A)
MDNA109


A-Fc-
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKK
mutations:


MDNA413
ATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG
F42A, E62A,


(2:1:2)
SETTFMCEYADETATIVEFLNRWITFSQSIISTLTGGGGSGGGGSGGGG
L80F, R81D,



SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
L85V, I86V,



EDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK
I92F, C125S



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT
MDNA413



CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
L10V, E12A,



RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGP
V18I, L39R,



VPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLI
R65D, D87S,



NVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLF
T88S, L101F,



HLRTLFREGQFN (SEQ ID NO: 172)
K104R, K105T


MDNA109FEA

APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLT

A

KFYMPKK

MDNA109FEAA


AC125-Fc-

ATELKHLQCLEE

A

LKPLEEVLNLAQSKNFH

FD

PRD

VV

SNINV

F

VLELKG

mutations:


MDNA413R39/Q111

SETTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGG

mutations:


(2:1:2)
SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
F42A, E62A,



EDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK
L80F, R81D,



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT
L85V, I86V,



CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
I92F



RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGP
MDNA413




VPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINRTAGMYCAALESLI

mutations:




NVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F


L10V, E12A,




HL

RT

LFREGQFN (SEQ ID NO: 223)

L39R, V18I,




R65D, D87S,




T88S, L101F,




K104R, K105T





MDNA109FEA
Gene 1 (Version B)
MDNA109


A-Fc-
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKK
mutations:


MDNA413
ATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG
F42A, E62A,


(2:1:2)
SETTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGG
L80F, R81D,



SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
L85V, 186V,



EDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK
I92F



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT
MDNA413



CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
mutations:



RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGP
L10V, E12A,



VPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLI
V18I, L39R,



NVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLF
R65D, D87S,



HLRTLFREGQFN (SEQ ID NO: 173)
T88S, L101F,




K104R, K105T





MDNA132-
Gene 1 (Version A)
MDNA132


Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALE
mutations:


(1:1:1
SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKD
L10H, R86T,


KIH)
LLLHLKKLFKEGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS
D87G, T88R,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
R108K



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
MDNA109



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
mutations:



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
L80F, R81D,



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L85V, I86V,



ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL
I92F, C125S



AQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRW




ITFSQSIISTLT (SEQ ID NO: 174)



MDNA132L39/Q111-

PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALE

MDNA132


Fc-

SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKD

mutations:


MDNA109

LLLHLKKLF

K

EGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS

L10H, R86T,


(1:1:1
VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
D87G, T88R,


KIH)
TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
R108K



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
MDNA109



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
mutations:



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L80F, R81D,




ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL

L85V, I86V,




AQSKNFH

FD

PRD

VV

SNINV

F

VLELKGSETTFMCEYADETATIVEFLNRW

I92F




ITFCQSIISTLT (SEQ ID NO: 224)







MDNA132-
Gene 1 (Version B)
MDNA132


Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALE
mutations:


(1:1:1
SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKD
L10H, R86T,


KIH)
LLLHLKKLFKEGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS
D87G, T88R,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
R108K



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
MDNA109



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
mutations:



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
L80F, R81D,



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L85V, I86V,



ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL
I92F



AQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRW




ITFCQSIISTLT (SEQ ID NO: 175)






MDNA132-
Gene 1 (Version C)
MDNA132


Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINRTAGMYCAALE
mutations:


(1:1:1
SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKD
L10H, L39R,


KIH)
LLLHLKKLFKEGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS
R86T, D87G,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
T88R, R108K



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
MDNA109



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
mutations:



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
L80F, R81D,



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L85V, I86V,



ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL
I92F, C125S



AQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRW




ITFSQSIISTLT (SEQ ID NO: 174)






MDNA132-
Gene 1 (Version D)
MDNA132


Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINRTAGMYCAALE
mutations:


(1:1:1
SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKD
L10H, L39R,


KIH)
LLLHLKKLFKEGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS
R86T, D87G,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
T88R, R108K



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
MDNA109



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
mutations:



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
L80F, R81D,



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L85V, I86V,



ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL
I92F



AQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRW




ITFCQSIISTLT (SEQ ID NO: 175)






MDNA132-
Gene 2 (Version A)
Fc portion


Fc-MDNA109
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



(1:1:1
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE



KIH)
YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSREEMTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 213)






MDNA132
Gene 2:




L39/Q111-Fc-

DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



MDNA109
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE



(1:1:1
YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSREEMTKNQVSLSC



KIH)
AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 225)






MDNA413R39/Q111-
Gene 1:
MDNA413


Fc-

PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALE

mutations:


MDNA132L39/Q111

SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD

L10V, E12A,


(2:1:1

LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS

L39R, V18I,


KIH)
VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
R65D, D87S,



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
T88S, L101F,



KAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQ
K104R, K105T



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
MDNA132



YTQKSLSLSPGcustom-characterPGPVPPSTAHRELIEELVNITQN
mutations:




QKAPLCNGSMVWSINLTAGMYCAALESLINVSGCSAIEKTQRMLSGFCP

L10H, R86T,




HKVSAGQFSSLHV

TGR

KIEVAQFVKDLLLHLKKLF

K

EGQFN (SEQ ID

D87G, T88R,



NO: 177)
R108K



Gene 2:





PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALE






SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD






LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS





VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK




TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS




KAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDIAVEWESNGQ




PENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNH




YTQKSLSLSPG (SEQ ID NO: 178)






Fc4-
APPCPSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDP
L10V, E12A,


MDNA413R39/Q111
EVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYK
L39R, V18I,


(1:2)
CKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLV
R65D, D87S,



KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQ
T88S, L101F,



EGNVFSCSVMHEALHNHYTQKSLSLSPGKGGGSGSPGPVPPSTAVRALI
K104R, K105T




EEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALESLINVSGCSAIEKT






Q

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

HL

RT

LFREGQF






N (SEQ ID NO: 179)







MDNA413R39/Q111-
Gene 1:
MDNA413


Fc-

PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALE

mutations:


MDNA109

SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD

L10V, E12A,


(1:1:1

LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS

L39R, V18I,


KIH)
VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
R65D, D87S,



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
T88S, L101F,



KAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDIAVEWESNGQ
K104R, K105T



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
MDNA109



YTQKSLSLSPGcustom-characterAPTSSSTKKTQLQLEHLL
mutations:




LDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLE

L80F, R81D,




EVLNLAQSKNFH

FD

PRD

VV

SNINV

F

VLELKGSETTFMCEYADETATIVE

L85V, I86V,




FLNRWITFCQSIISTLT (SEQ ID NO: 180)

I92F



Gene 2:




DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSREEMTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 181)






Fc -
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
L10V, E12A,


MDNA413L39/Q111
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
V18I, R65D,


(1:2)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
D87S, T88S



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
L101F,



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV
K104R, K105T




PPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN






VSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

H






L

RT

LFREGQFN (SEQ ID NO: 182)







MDNA132L39/R111-
Gene 1:
L10H, R86T,


Fc

PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALE

D87G, T88R,


(1:1 KIH)

SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKD

R108K




LLLHLKKLF

K

EGRFN
custom-character DKTHTCPPCPAPELLGGPS





VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK




TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS




KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ




PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH




YTQKSLSLSPG (SEQ ID NO: 183)




Gene 2:




DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC




LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 184)






Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
L10H, R86T,


MDNA132L39/R111
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
D87G, T88R,


(1:2)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
R108K



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV





PPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN






VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKDLLLH






LKKLF

K

EGRFN (SEQ ID NO: 185)







Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
L10V, E12A,


MDNA413L39/R111
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
V18I, R65D,


(1:2)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
D87S, T88S,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
L101F,



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV
K104R, K105T




PPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN






VSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

H






L

RT

LFREGRFN (SEQ ID NO: 186)







MDNA413L39/Q111-

PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINLTAGMYCAALE

L10V, E12A,


Fc-

SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD

V18I, R65D,


MDNA413L39/Q111

LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS

D87S, T88S,


(2:1:2)
VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
L101F,



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
K104R, K105T



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ




PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH




YTQKSLSLSPGcustom-characterPGPVPPSTAVRALIEELINITQN





QKAPLCNGSMVWSINLTAGMYCAALESLINVSGCSAIEKTQ

D

MLSGFCP






HKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

HL

RT

LFREGQFN (SEQ





ID NO: 187)






H4FEAA-Fc-

APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAK

F

YMPKK

H4FEAA


MDNA413L39/Q111

ATELKHLQCLEE

A

LKPLEEVLNLA

S

SKNFHF

T

PRD

V

VSN

I

NVFVLELKG

mutations:


(2:1:2)

SETTFMCEYADETATIVEFLNRWITF

S

QSIISTLTGGGGSGGGGSGGGG

F42A, E62A,



SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
Q74S, R81T,



EDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK
L85V, I92F,



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT
C125S



CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
MDNA413



RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGP
mutations:




VPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINLTAGMYCAALESLI

L10V, E12A,




NVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F


V18I, R65D,




HL

RT

LFREGQFN (SEQ ID NO: 188)

D87S, T88S,




L101F,




K104R, K105T





mCD3 IgG-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
D87G, T88R,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R108K


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV



sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL






ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVK






DLLLHLKKLF

K

EGQFN (SEQ ID NO: 189)





Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 190)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 191)






huCD3 IgG-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
D87G, T88R,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R108K


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV



sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPcustom-character





custom-character
PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL






ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVK






DLLLHLKKLF

K

EGQFN (SEQ ID NO: 192)





Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 193)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 194)






mPD1 IgG-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
D87G, T88R,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R108K


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV



sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL






ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVK






DLLLHLKKLF

K

EGQFN (SEQ ID NO: 195)





Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 196)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 197)






huPD1 IgG-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
D87G, T88R,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R108K


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV



sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL






ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVK






DLLLHLKKLF

K

EGQFN (SEQ ID NO: 198)





Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 199)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 200)






mPD1 IgG-
Gene 1:
F42A, E62A,


MDNA109FEAAS125
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
L80F, R81D,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
L85V, I86V,


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
I92F, C125S


sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLT

A

KFYMPK






KATELKHLQCLEE

A

LKPLEEVLNLAQSKNFH

FD

PRD

VV

SNINV

F

VLELK






GSETTFMCEYADETATIVEFLNRWITF

S

QSIISTLT (SEQ ID





NO: 201)




Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 202)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 203)






huPD1 IgG-
Gene 1:
F42A, E62A,


MDNA109FEAAS125
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
L80F, R81D,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
L85V, I86V,


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
I92F, C125S


sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLT

A

KFYMPK






KATELKHLQCLEE

A

LKPLEEVLNLAQSKNFH

FD

PRD

VV

SNINV

F

VLELK






GSETTFMCEYADETATIVEFLNRWITF

S

QSIISTLT (SEQ ID





NO: 204)




Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 205)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 206)






mPD1 IgG-
Gene 1:
L10V, E12A,


MDNA413R39/Q111*
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
L39R, V18I,


(*partial
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R65D, D87S,


sequence)
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
T88S, L101F



DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW
K104R, K105T



LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQ




VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKcustom-character





custom-character
PGPVPPSTA

V

R

A

LIEEL

I

NITONQKAPLCNGSMVWSIN

R

TAGMYCAA






LESLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFV






KDLL

F

HL

RT

LFREGQFN (SEQ ID NO: 207)





Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 208)






huPD1 IgG-
Gene 1:
L10V, E12A,


MDNA413R39/Q111*
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
L39R, V18I,


(*partial
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R65D, D87S,


sequence)
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
T88S, L101F,



DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW
K104R, K105T



LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQ




VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKcustom-character





custom-character
PGPVPPSTA

V

R

A

LIEEL

I

NITONQKAPLCNGSMVWSIN

R

TAGMYCAA






LESLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFV






KDLL

F

HL

RT

LFREGQFN (SEQ ID NO: 209)





Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 210)






MDNA413R39/Q111-
Gene 1:
L10V, E12A,


Fc

PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALE

V18I, L39R,


(1:1 KIH)

SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD

R65D, D87S,




LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS

T88S, L101F,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
K104R, K105T



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS




KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ




PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH




YTQKSLSLSPG (SEQ ID NO: 211)




Gene 2:




DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 212)









D. Recombinant Expression of IL-2 Muteins, Expression Vectors and Host Cells


In various embodiments, polypeptides used in the practice of the instant invention are synthetic or are produced by expression of a recombinant nucleic acid molecule. In the event the polypeptide is a chimera (e.g., a fusion protein containing at least a mutant IL-2 polypeptide and a heterologous polypeptide, including a bispecific IL-2 cytokine fusion), it can be encoded by a hybrid nucleic acid molecule containing one sequence that encodes all or part of the IL-2 mutein, and a second sequence that encodes all or part of the heterologous polypeptide. For example, subject IL-2 muteins described herein may be fused to a hexa-histidine tag to facilitate purification of bacterially expressed protein, or to a hemagglutinin tag to facilitate purification of protein expressed in eukaryotic cells.


Methods for constructing a DNA sequence encoding the IL-2 muteins and expressing those sequences in a suitably transformed host include, but are not limited to, using a PCR-assisted mutagenesis technique. Mutations that consist of deletions or additions of amino acid residues to an IL-2 polypeptide can also be made with standard recombinant techniques. In the event of a deletion or addition, the nucleic acid molecule encoding IL-2 is optionally digested with an appropriate restriction endonuclease. The resulting fragment can either be expressed directly or manipulated further by, for example, ligating it to a second fragment. The ligation may be facilitated if the two ends of the nucleic acid molecules contain complementary nucleotides that overlap one another, but blunt-ended fragments can also be ligated. PCR-generated nucleic acids can also be used to generate various mutant sequences.


The complete amino acid sequence can be used to construct a back-translated gene. A DNA oligomer containing a nucleotide sequence coding for IL-2 mutein can be synthesized. For example, several small oligonucleotides coding for portions of the desired polypeptide can be synthesized and then ligated. The individual oligonucleotides typically contain 5′ or 3′ overhangs for complementary assembly.


In addition to generating mutant polypeptides via expression of nucleic acid molecules that have been altered by recombinant molecular biological techniques, subject IL-2 muteins can be chemically synthesized. Chemically synthesized polypeptides are routinely generated by those of skill in the art.


Once assembled (by synthesis, site-directed mutagenesis or another method), the DNA sequences encoding an IL-2 mutein will be inserted into an expression vector and operatively linked to an expression control sequence appropriate for expression of the IL-2 mutein in the desired transformed host. Proper assembly can be confirmed by nucleotide sequencing, restriction mapping, and expression of a biologically active polypeptide in a suitable host. As is well known in the art, in order to obtain high expression levels of a transfected gene in a host, the gene must be operatively linked to transcriptional and translational expression control sequences that are functional in the chosen expression host.


The DNA sequence encoding the IL-2 mutein, whether prepared by site directed mutagenesis, chemical synthesis or other methods, can also include DNA sequences that encode a signal sequence. Such signal sequence, if present, should be one recognized by the cell chosen for expression of the IL-2 mutein. It can be prokaryotic, eukaryotic or a combination of the two. It can also be the signal sequence of native IL-2. The inclusion of a signal sequence depends on whether it is desired to secrete the IL-2 mutein from the recombinant cells in which it is made. If the chosen cells are prokaryotic, it generally is preferred that the DNA sequence not encode a signal sequence. If the chosen cells are eukaryotic, it generally is preferred that a signal sequence be encoded and most preferably that the wild-type IL-2 signal sequence be used.


E. Oncolytic Viruses Targeting Moieties


In some examples, the bispecific IL-2 cytokine fusion and/or IL-2 muteins described herein can be employed to target an oncolytic virus (e.g., see Allen et al., Mol. Ther. 16:1556-64, 2008). In some examples, oncolytic virus can be used to target an IL-2 mutein to a tumor or TME. Numerous viruses can be employed as the oncolytic virus, including adenoviruses as well as self-replicating alphavirus, as well as oncolyctic vaccinia viruses (see, for example WO2013038066, incorporated herein by reference in its entirety; in particular FIG. 17). Other oncolytic viruses can include Seneca Valley Virus, Newcastle disease Virus (also referred to as Newcastle virus), Maraba virus, vesicular stomatitis virus (VSV), Herpes virus (including HSV-1), Measles virus, poliovirus, reovirus, coxsackie virus, a lentivirus, a morbillivirus, an influenza virus, Sinbis virus, myxoma virus and/or retrovirus (see, for example, Twumasi-Boateng, et al., “Oncolytic viruses as engineering platforms for combination immunotherapy”, Nature Reviews Cancer, 2018), and Kaufman et al., Cancer Immunotherapy, 14:642-662 (2015), all of which are incorporated by reference herein their entireties). In some embodiments, the oncolytic virus includes but is not limited to an adenovirus, a self-replicating alphavirus, a vaccinia virus, a Seneca Valley Virus, a Newcastle disease Virus, a Maraba virus, vesicular stomatitis virus (VSV), a Herpes virus (including HSV-1 and HSV-2), a measles virus, a poliovirus, a reovirus, a coxsackie virus, a lentivirus, a morbillivirus, an influenza virus, Sinbis virus, myxoma virus and/or a retrovirus. The IL-2 superkines (H9 and IL-2 variants as described herein) also can be used to direct T cells/OVs to the TME. An IL-2 variant (such as H9) can boost effector T cells and NK cells while IL-2 variant can suppress T reg activity. Other oncolytic viruses include can include, for example, oncoVex/T-VEC, which involves the intratumoral injection of replication-conditional herpes simplex virus which preferentially infects cancer cells. The virus, which is also engineered to express GM-CSF, is able to replicate inside a cancer cell causing its lysis, releasing new viruses and an array of tumor antigens, and secreting GM-CSF in the process. Such oncolytic virus vaccines enhance DCs function in the tumor microenvironment to stimulate anti-tumor immune responses. These oncolytic viruses can be used to target or deliver the IL-2 muteins described herein to the tumor. In some embodiments, the IL-2 mutein is any IL-2 mutein or variant disclosed herein. In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO:10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO:10; C5 SEQ ID NO: 11; D10 SEQ ID NO:12; E10 SEQ ID NO:13; G8 SEQ ID NO:14; H4 SEQ ID NO:15; and H9 SEQ ID NO:16. In some embodiments, the substitutions in the IL-2 mutein comprise L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the oncolytic virus comprises a transgene capable of expressing an IL-2 mutein as described herein. In some embodiments, the oncolytic virus comprises a transgene capable of expressing an IL-2 mutein comprising the following amino acid substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the oncolytic virus comprises a nucleic acid encoding an IL-2 mutein comprising the following amino acid substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the oncolytic virus comprises a transgene that is expressed as a therapeutic payload. In some embodiments, the therapeutic payload is an I1-2 as described herein. In some embodiments, the therapeutic payload is IL-2 mutein comprising the following amino acid substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2.


In some embodiments, the oncolytic virus is an oncolytic vaccinia virus. In some embodiments, the oncolytic vaccinia virus vector is characterized in that the virus particle is of the type intracellular mature virus (IMV), intracellular enveloped virus (IEV), cell-associated enveloped virus (CEV), or extracellular enveloped virus (EEV). In some embodiments, the oncolytic vaccinia virus particle is of the type EEV or IMV. In some embodiments, the oncolytic vaccinia virus particle is of the type EEV.


Generally, construction of oncolytic vaccinia virus recombinants and cells and pharmaceutical compositions comprising the vectors which preferentially replicate in tumor cells and express at least one transgene (for example, and IL-2 mutein as described herein) to facilitate antitumor efficacy and apoptosis induction and to modulate host immune responses in a subject. According to the present invention, oncolytic adenoviruses and oncolytic vaccinia viruses can be combined with IL-2 expression or targeting moieties as described herein in order to target the oncolytic vaccinia virus or the oncolytic adenovirus and/or express the IL-2 mutein. Oncolysis releases tumor antigens and provides costimulatory danger signals. However, arming the virus can improve efficacy further. For example, CD40 ligand (CD40L, CD154) is known to induce apoptosis of tumor cells and it also triggers several immune mechanisms. One of these is a T-helper type 1 (Th1) response that leads to activation of cytotoxic T-cells and reduction of immune suppression. The present invention provides for oncolytic viruses that express the IL-2 muteins of the present invention. In some embodiments, the present invention provides for oncolytic viruses that are targeted (for example, “armed”) with the IL-2 targeting moieties of the present invention.


In some embodiments, the oncolytic virus is a modified vaccinia virus vector, a virus particle, a host cell, a pharmaceutical composition and a kit comprising vaccinia virus genome wherein the thymidine kinase gene is inactivated by either a substitution in the thymidine kinase (TK) gene and/or an open reading frame ablating deletion of at least one nucleotide providing a partially deleted thymidine kinase gene, the vaccinia growth factor gene is deleted, and the modified vaccinia virus vector comprises at least one nucleic acid sequence encoding a non-viral protein (e.g., an IL-2 mutein as described herein which is capable of being expressed). In another aspect is provided the modified vaccinia virus vector, the virus particle, the pharmaceutical composition or the kit can be used for cancer therapy, for eliciting immune response in a subject, for use in a method of inhibiting malignant cell proliferation in a mammal, for use in a therapy or prophylaxis of cancer, for detecting the presence of the modified vaccinia virus in a subject, and as an in situ cancer vaccine, optionally in combination with adenovirus. In some embodiments, the invention provides method of producing a modified vaccinia virus comprising vaccinia virus genome wherein the thymidine kinase gene is inactivated by a substitution in the thymidine kinase (TK) gene and/or an open reading frame ablating deletion of at least one nucleotide providing a partially deleted thymidine kinase gene, the vaccinia growth factor gene is deleted, and the modified vaccinia virus vector comprises at least one nucleic acid sequence encoding a non-viral protein (e.g., an IL-2 mutein as described herein), comprising the steps of providing producer cells capable of sustaining production of vaccinia virus particles and carrying the modified vaccinia vector; culturing the producer cells in conditions suitable for virus replication and production; and harvesting the virus particles.


In some embodiments, the present invention provides methods of administering an oncolytic virus “armed” with or including an nucleic acid encoding an IL-2 mutein as described herein, wherein the IL-2 mutein is expressed at the tumor location or is expressed systemically in the subject. In some embodiments, the present invention also provides methods of administering an oncolytic virus “armed” or targeted with an IL-2 mutein as described herein. The routes of administration vary, naturally, with the location and nature of the tumor, and include, e.g., intradermal, transdermal, parenteral, intravenous, intramuscular, intranasal, subcutaneous, regional (e.g., in the proximity of a tumor, particularly with the vasculature or adjacent vasculature of a tumor), percutaneous, intratracheal, intraperitoneal, intraarterial, intravesical, intratumoral, inhalation, perfusion, lavage, and oral administration. Compositions are formulated relative to the particular administration route.


1. Oncolytic Vaccinia Virus


Vaccinia virus is a member of the Orthopoxvirus genus of the Poxviridae. It has large double-stranded DNA genome (˜200 kb, ˜200 genes) and a complex morphogenic pathway produces distinct forms of infectious virions from each infected cell. Viral particles contain lipid membranes(s) around a core. Virus core contains viral structural proteins, tightly compacted viral DNA genome, and transcriptional enzymes. Dimensions of vaccinia virus are ˜360×270×250 nm, and weight of ˜5-10 fg. Genes are tightly packed with little non-coding DNA and open-reading frames (ORFs) lack introns. Three classes of genes (early, intermediate, late) exists. Early genes (˜100 genes; immediate and delayed) code for proteins mainly related to immune modulation and virus DNA replication. Intermediate genes code for regulatory proteins which are required for the expression of late genes (e.g. transcription factors) and late genes code for proteins required to make virus particles and enzymes that are packaged within new virions to initiate the next round of infection. Vaccinia virus replicates in the cell cytoplasm.


Different strains of vaccinia viruses have been identified (as an example: Copenhagen, modified virus Ankara (MVA), Lister, Tian Tan, Wyeth (=New York City Board of Health), Western Reserve (WR)). The genome of WR vaccinia has been sequenced (Accession number AY243312). In some embodiments, the oncolytic vaccinia virus is a Copenhagen, modified virus Ankara (MVA), Lister, Tian Tan, Wyeth, or Western Reserve (WR) vaccinia virus.


Different forms of viral particles have different roles in the virus life cycle Several forms of viral particles exist: intracellular mature virus (IMV), intracellular enveloped virus (IEV), cell-associated enveloped virus (CEV), extracellular enveloped virus (EEV). EEV particles have an extra membrane derived from the trans-Golgi network. This outer membrane has two important roles: a) it protects the internal IMV from immune aggression and, b) it mediates the binding of the virus onto the cell surface.


CEVs and EEVs help virus to evade host antibody and complement by being wrapped in a host-derived membrane. IMV and EEV particles have several differences in their biological properties and they play different roles in the virus life cycle. EEV and IMV bind to different (unknown) receptors (1) and they enter cells by different mechanisms. EEV particles enter the cell via endocytosis and the process is pH sensitive. After internalization, the outer membrane of EEV is ruptured within an acidified endosome and the exposed IMV is fused with the endosomal membrane and the virus core is released into the cytoplasm. IMV, on the other hand, enters the cell by fusion of cell membrane and virus membrane and this process is pH-independent. In addition to this, CEV induces the formation of actin tails from the cell surface that drive virions towards uninfected neighboring cells.


Furthermore, EEV is resistant to neutralization by antibodies (NAb) and complement toxicity, while IMV is not. Therefore, EEV mediates long range dissemination in vitro and in vivo. Comet-inhibition test has become one way of measuring EEV-specific antibodies since even if free EEV cannot be neutralized by EEV NAb, the release of EEV from infected cells is blocked by EEV NAb and comet shaped plaques cannot be seen. EEV has higher specific infectivity in comparison to IMV particles (lower particle/pfu ratio) which makes EEV an interesting candidate for therapeutic use. However, the outer membrane of EEV is an extremely fragile structure and EEV particles need to be handled with caution which makes it difficult to obtain EEV particles in quantities required for therapeutic applications. EEV outer membrane is ruptured in low pH (pH ˜6). Once EEV outer membrane is ruptured, the virus particles inside the envelope retain full infectivity as an IMV.


Some host-cell derived proteins co-localize with EEV preparations, but not with IMV, and the amount of cell-derived proteins is dependent on the host cell line and the virus strain. For instance, WR EEV contains more cell-derived proteins in comparison to VV IHD-J strain. Host cell derived proteins can modify biological effects of EEV particles. As an example, incorporation of the host membrane protein CD55 in the surface of EEV makes it resistance to complement toxicity. In the present invention it is shown that human A549 cell derived proteins in the surface of EEV particles may target virus towards human cancer cells. Similar phenomenon has been demonstrated in the study with human immunodeficiency virus type 1, where host-derived ICAM-1 glycoproteins increased viral infectivity. IEV membrane contains at least 9 proteins, two of those not existing in CEV/EEV. F12L and A36R proteins are involved in IEV transport to the cell surface where they are left behind and are not part of CEV/EEV (9, 11). 7 proteins are common in (IEV)/CEV/EEV: F13L, A33R, A34R, A56R, B5R, E2, (K2L). For Western Reserve strain of vaccinia virus, a maximum of 1% of virus particles are normally EEV and released into the culture supernatant before oncolysis of the producer cell. 50-fold more EEV particles are released from International Health Department (IHD)-J strain of vaccinia. IHD has not been studied for use in cancer therapy of humans however. The IHD-W phenotype was attributed largely to a point mutation within the A34R EEV lectin-like protein. Also, deletion of A34R increases the number of EEVs released. EEV particles can be first detected on cell surface 6 hours post-infection (as CEV) and 5 hours later in the supernatant (IHD-J strain). Infection with a low multiplicity of infection (MOI) results in higher rate of EEV in comparison to high viral dose. The balance between CEV and EEV is influenced by the host cell and strain of virus.


Vaccinia has been used for eradication of smallpox and later, as an expression vector for foreign genes and as a live recombinant vaccine for infectious diseases and cancer. Vaccinia virus is the most widely used pox virus in humans and therefore safety data for human use is extensive. During worldwide smallpox vaccination programs, hundreds of thousands humans have been vaccinated safety with modified vaccinia virus strains and only very rare severe adverse events have been reported. Those are generalized vaccinia (systemic spread of vaccinia in the body), erythema multiforme (toxic/allergic reaction), eczema vaccinatum (widespread infection of the skin), progressive vaccinia (tissue destruction), and postvaccinia! encephalitis.


All together 44 melanoma patients have been treated in early clinical trials with wild type vaccinia virus in 1960s-1990s and the overall objective response rate of injected tumors was 50%. Also some beneficial immunological responses were seen (36). Wild type vaccinia virus has been used also for treatment of bladder cancer, lung and kidney cancer, and myeloma and only mild adverse events were seen. JX-594, an oncolytic Wyeth strain vaccinia virus coding for GM-CSF, has been successfully evaluated in three phase I studies and preliminary results from randomized phase II trial has been presented in the scientific meeting.


Vaccinia virus is appealing for cancer gene therapy due to several characteristics. It has natural tropism towards cancer cells and the selectivity can be significantly enhanced by deleting some of the viral genes. The present invention relates to the use of double deleted vaccinia virus (vvdd) in which two viral genes, viral thymidine kinase (TK) and vaccinia growth factor (VGF), are at least partially deleted. TK and VGF genes are needed for virus to replicate in normal but not in cancer cells. The partial TK deletion may be engineered in the TK region conferring activity.


TK deleted vaccinia viruses are dependent on cellular nucleotide pool present in dividing cells for DNA synthesis and replication. IN some embodiments, the TK deletion limits virus replication significantly in resting cells allowing efficient virus replication to occur only in actively dividing cells (e.g., cancer cells). VGF is secreted from infected cells and has a paracrine priming effect on surrounding cells by acting as a mitogen. Replication of VGF deleted vaccinia viruses is highly attenuated in resting (non-cancer) cells. The effects of TK and VGF deletions have been shown to be synergistic.


2. Oncolytic Adenovirus


Generally, adenovirus is a 36 kb, linear, double-stranded DNA virus (Grunhaus and Horwitz, 1992). The term “adenovirus” or “AAV” includes AAV type 1 (AAV1), AAV type 2 (AAV2), AAV type 3 (AAV3), AAV type 4 (AAV4), AAV type 5 (AAV5), AAV type 6 (AAV6), AAV type 7 (AAV7), AAV type 8 (AAV8), AAV type 9 (AAV9), AAV 9_hu14, avian AAV, bovine AAV, canine AAV, equine AAV, primate AAV, non-primate AAV, and ovine AAV. “Primate AAV” refers to AAV capable of infecting primates, “non-primate AAV” refers to AAV capable of infecting non-primate mammals, “bovine AAV” refers to AAV capable of infecting bovine mammals, etc.


Adenoviral infection of host cells results in adenoviral DNA being maintained episomally, which reduces the potential genotoxicity associated with integrating vectors. Also, adenoviruses are structurally stable, and no genome rearrangement has been detected after extensive amplification. Adenovirus can infect virtually all epithelial cells regardless of their cell cycle stage, (See, for example, US20060147420, incorporated by reference herein in its entirety.) Moreover, the Ela and E4 regions of adenovirus are essential for an efficient and productive infection of human cells. The Ela gene is the first viral gene to be transcribed in a productive infection, and its transcription is not dependent on the action of any other viral gene products. However, the transcription of the remaining early viral genes requires Ela gene expression. The Ela promoter, in addition to regulating the expression of the Ela gene, also integrates signals for packaging of the viral genome as well as sites required for the initiation of viral DNA replication. See, Schmid, S. I., and Hearing, P. in Current Topics in Microbiology and Immunology, vol. 199: pages 67-80 (1995).


In some embodiments, the oncolytic virus is an oncolytic adenovirus. It has been established that naturally occurring viruses can be engineered to produce an oncolytic effect in tumor cells (Wildner, 2001; Jacotat, 1967; Kim, 2001; Geoerger et al., 2002; Yan et al., 2003; Vile et al., 2002, each of which is incorporated herein by reference). In the case of adenoviruses, specific deletions within their adenoviral genome can attenuate their ability to replicate within normal quiescent cells, while they retain the ability to replicate in tumor cells. One such conditionally replicating adenovirus, 024, has been described by Fueyo et al. (2000), see also U.S. Patent Application No. 20030138405, each of which are incorporated herein by reference. The A24 adenovirus is derived from adenovirus type 5 (Ad-5) and contains a 24-base-pair deletion within the CR2 portion of the E1A gene. See, for example WO2001036650A2 (incorporated by reference herein in it's entirety.


Oncolytic adenoviruses include conditionally replicating adenoviruses (CRADs) such as Delta 24, which have several properties that make them candidates for use as biotherapeutic agents. One such property is the ability to replicate in a permissive cell or tissue, which amplifies the original input dose of the oncolytic virus and helps the agent spread to adjacent tumor cells providing a direct antitumor effect.


In some embodiments, the oncolytic component of Delta 24 with a transgene expression approach to produce an armed Delta 24. Armed Delta 24 adenoviruses may be used for producing or enhancing bystander effects within a tumor and/or producing or enhancing detection/imaging of an oncolytic adenovirus in a patient, or tumor associated tissue and/or cell. In some embodiments the combination of oncolytic adenovirus with various transgene strategies (e.g., expression of and IL-2 mutein) w ill improve the therapeutic potential, including for example, potential against a variety of refractory tumors, as well as provide for improved imaging capabilities. In certain embodiments, an oncolytic adenovirus may be administered with a replication defective adenovirus, another oncolytic virus, a replication competent adenovirus, and/or a wildtype adenovirus. Each of which nay be adminstered concurrently, before or after the other adenoviruses.


In some embodiments, an Ela adenoviral vectors involves the replacement of the basic adenovirus Ela promoter, including the CAAT box, TATA box and start site for transcription initiation, with a basic promoter that exhibits tumor specificity, and preferably is E2F responsive, and more preferably is the human E2F-1 promoter. Thus, this virus will be repressed in cells that lack molecules, or such molecules are non functional, that activate transcription from the E2F responsive promoter. Normal non dividing, or quiescent cells, fall in this class, as the transcription factor, E2F, is bound to pRb, or retinoblastoma protein, thus making E2F unavailable to bind to and activate the E2F responsive promoter. In contrast, cells that contain free E2F should support E2F based transcription. An example of such cells are neoplastic cells that lack pRb function, allowing for a productive viral infection to occur. In some embodiments, an E1a adenoviral vector is targeted use an IL-2 moiety as described herein.


Retention of the enhancer sequences, packaging signals, and DNA replication start sites which lie in the Ela promoter will ensure that the adenovirus infection proceeds to wild type levels in the neoplastic cells that lack pRb function. In essence, the modified ET a promoter confers tumor specific transcriptional activation resulting in substantial tumor specific killing, yet provides for enhanced safety in normal cells.


In some embodiments, an Ela adenoviral vector is prepared by substituting the endogenous E1a promoter with the E2F responsive promoter, the elements upstream of nucleotide 375 in the adenoviral 5 genome are kept intact. The nucleotide numbering is as described by See, Schmid, S. I., and Hearing, P. Current Topics in Microbiology and Immunology, vol. 199: pages 67-80 (1995). This includes all of the seven A repeat motifs identified for packaging of the viral genome. Sequences from nucleotide 375 to nucleotide 536 are deleted by a BsaAI to BsrBI restriction start site, while still retaining 23 base pairs upstream of the translational initiation codon for the ETA protein. An E2F responsive promoter, preferably human E2F-1 is substituted for the deleted endogenous Ela promoter sequences using known materials and methods. The E2F-1 promoter may be isolated as described in Example 1.


The E4 region has been implicated in many of the events that occur late in adenoviral infection, and is required for efficient viral DNA replication, late mRNA accumulation and protein synthesis, splicing, and the shutoff of host cell protein synthesis. Adenoviruses that are deficient for most of the E4 transcription unit are severely replication defective and, in general, must be propagated in E4 complementing cell lines to achieve high titers. The E4 promoter is positioned near the right end of the viral genome and governs the transcription of multiple open reading frames (ORF). A number of regulatory elements have been characterized in this promoter that are critical for mediating maximal transcriptional activity. In addition to these sequences, the E4 promoter region contains regulatory sequences that are required for viral DNA replication. A depiction of the E4 promoter and the position of these regulatory sequences can be seen in FIGS. 2 and 3 of U.S. Pat. No. 7,001,596, incorporated by reference herein in its entirety.


In some embodiments, the adenoviral vector that has the E4 basic promoter substituted with one that has been demonstrated to show tumor specificity, preferably an E2F responsive promoter, and more preferably the human E2F-1 promoter. The reasons for preferring an E2F responsive promoter to drive E4 expression are the same as were discussed above in the context of an Ela adenoviral vector having the E1a promoter substituted with an E2F responsive promoter. The tumor suppressor function of pRb correlates with its ability to repress E2F-responsive promoters such as the E2F-1 promoter (Adams, P. D., and W. G. Kaelin, Jr. 1995, Cancer Biol. 6:99-108; Sellers, W. R., and W. G. Kaelin. 1996, published erratum appears in Biochim Biophys Acta 1996 Dec. 9; 1288(3):E-1, Biochim Biophys Acta. 1288:M1-5. Sellers, W. R., J. W. Rodgers, and W. G. Kaelin, Jr. 1995, Proc Natl Acad Sci USA. 92:11544-8.) The human E2F-1 promoter has been extensively characterized and shown to be responsive to the pRb signaling pathway, including pRb/p107, E2F-1/-2/-3, and G1 cyclin/cdk complexes, and ETA (Johnson, D. G., K. Ohtani, and J. R. Nevins. 1994, Genes Dev. 8:1514-25; Neuman, E., E. K. Flemington, W. R. Sellers, and W. G. Kaelin, Jr. 1995. Mol Cell Biol. 15:4660; Neuman, E., W. R. Sellers, J. A. McNeil, J. B. Lawrence, and W. G. Kaelin, Jr. 1996, Gene. 173:163-9.) Most, if not all, of this regulation has been attributed to the presence of multiple E2F sites present within the E2F-1 promoter. Hence, a virus carrying this (these) modification(s) would be expected to be attenuated in normal cells that contain an intact (wild type) pRb pathway, yet exhibit a normal infection/replication profile in cells that are deficient for pRb's repressive function. In order to maintain the normal infection/replication profile of this mutant virus we have retained the inverted terminal repeat (ITR) at the distal end of the E4 promoter as this contains all of the regulatory elements that are required for viral DNA replication (Hatfield, L. and P. Hearing. 1993, J. Virol. 67:3931-9; Rawlins, D. R., P. J. Rosenfeld, R. J. Wides, M. D. Challberg, and T. J. Kelly, Jr. 1984, Cell. 37:309-19; Rosenfeld, P. J., E. A. O'Neill, R. J. Wides, and T. J. Kelly. 1987, Mol Cell Biol. 7:875-86; Wides, R. J., M. D. Challberg, D. R. Rawlins, and T. J. Kelly. 1987, Mol Cell Biol. 7:864-74). This facilitates attaining wild type levels of virus in pRb pathway deficient tumor cells infected with this virus.


In some embodiments, the E4 promoter is positioned near the right end of the viral genome and it governs the transcription of multiple open reading frames (ORFs) (Freyer, G. A., Y. Katoh, and R. J. Roberts. 1984, Nucleic Acids Res. 12:3503-19; Tigges, M. A., and H. J. Raskas. 1984. Splice junctions in adenovirus 2 early region 4 mRNAs: multiple splice sites produce 18 to 24 RNAs. J. Virol. 50:106-17; Virtanen, A. P. Gilardi, A. Naslund, J. M. LeMoullec, U. Pettersson, and M. Perricaudet. 1984, J. Virol. 51:822-31.) A number of regulatory elements have been characterized in this promoter that mediate transcriptional activity (Berk, A. J. 1986, Annu Rev Genet. 20:45-79; Gilardi, P., and M. Perricaudet. 1986, Nucleic Acids Res. 14:9035-49; Gilardi, P., and M. Perricaudet. 1984, Nucleic Acids Res. 12:7877-88; Hanaka, S., T. Nishigaki, P. A. Sharp, and H. Handa. 1987, Mol Cell Biol. 7:2578-87; Jones, C., and K. A. Lee. 1991, Mol Cell Biol. 11:4297-305; Lee, K. A., and M. R. Green. 1987, Embo J. 6:1345-53.) In addition to these sequences, the E4 promoter region contains elements that are involved in viral DNA replication (Hatfield, L., and P. Hearing. 1993, J Virol. 67:3931-9; Rawlins, D. R., P. J. Rosenfeld, R. J. Wides, M. D. Challberg, and T. J. Kelly, Jr. 1984, Cell. 37:309-19; Rosenfeld, P. J., E. A. O'Neill, R. J. Wides, and T. J. Kelly. 1987, Mol Cell Biol. 7:875-86; Wides, R. J., M. D. Challberg, D. R. Rawlins, and T. J. Kelly. 1987, Mol Cell Biol. 7:864-74.) A depiction of the E4 promoter and the position of these regulatory sequences can be seen in FIGS. 1 and 2. See, also, Jones, C., and K. A. Lee. Mol Cell Biol. 11:4297-305 (1991). With these considerations in mind, an E4 promoter shuttle was designed by creating two novel restriction endonuclease sites: a XhoI site at nucleotide 35,576 and a SpeI site at nucleotide 35,815 (see FIG. 3). Digestion with both XhoI and SpeI removes nucleotides from 35,581 to 35,817. This effectively eliminates bases −208 to +29 relative to the E4 transcriptional start site, including all of the sequences that have been shown to have maximal influence on E4 transcription. In particular, this encompasses the two inverted repeats of E4F binding sites that have been demonstrated to have the most significant effect on promoter activation. However, all three Sp1 binding sites, two of the five ATF binding sites, and both of the NF1 and NFIII/Oct-1 binding sites that are critical for viral DNA replication are retained.


In some embodiments, the E2F responsive promoter is the human E2F-1 promoter. Key regulatory elements in the E2F-1 promoter that mediate the response to the pRb pathway have been mapped both in vitro and in vivo (Johnson, D. G., K. Ohtani, and J. R. Nevins. 1994, Genes Dev. 8:1514-25; Neuman, E., E. K. Flemington, W. R. Sellers, and W. G. Kaelin, Jr. 1995, Mol Cell Biol. 15:4660; Parr, M. J., Y. Manome, T. Tanaka, P. Wen, D. W. Kufe, W. G. Kaelin, Jr., and H. A. Fine. 1997, Nat Med. 3:1145-9.) Thus, we isolated the human E2F-1 promoter fragment from base pairs −218 to +51, relative to the transcriptional start site, by PCR with primers that incorporated a SpeI and XhoI site into them. This creates the same sites present within the E4 promoter shuttle and allows for direct substitution of the E4 promoter with the E2F-1 promoter.


F. Nucleic Acid Molecules Encoding Mutant IL-2


In some embodiments the subject IL-2 mutein, either alone or as a part of a chimeric polypeptide (including a bispecific IL-2 cytokine fusion), such as those described above, can be obtained by expression of a nucleic acid molecule. Just as IL-2 muteins can be described in terms of their identity with wild-type IL-2 polypeptides, the nucleic acid molecules encoding them will necessarily have a certain identity with those that encode wild-type IL-2. For example, the nucleic acid molecule encoding a subject IL-2 mutein can be at least 50%, at least 65%, preferably at least 75%, more preferably at least 85%, and most preferably at least 95% (e.g., 99%) identical to the nucleic acid encoding wild-type IL-2 (e.g., SEQ ID NO:2).


The nucleic acid molecules provided can contain naturally occurring sequences, or sequences that differ from those that occur naturally, but, due to the degeneracy of the genetic code, encode the same polypeptide. These nucleic acid molecules can consist of RNA or DNA (for example, genomic DNA, cDNA, or synthetic DNA, such as that produced by phosphoramidite-based synthesis), or combinations or modifications of the nucleotides within these types of nucleic acids. In addition, the nucleic acid molecules can be double-stranded or single-stranded (i.e., either a sense or an antisense strand).


The nucleic acid molecules are not limited to sequences that encode polypeptides; some or all of the non-coding sequences that lie upstream or downstream from a coding sequence (e.g., the coding sequence of IL-2) can also be included. Those of ordinary skill in the art of molecular biology are familiar with routine procedures for isolating nucleic acid molecules. They can, for example, be generated by treatment of genomic DNA with restriction endonucleases, or by performance of the polymerase chain reaction (PCR). In the event the nucleic acid molecule is a ribonucleic acid (RNA), molecules can be produced, for example, by in vitro transcription.


Exemplary isolated nucleic acid molecules of the present disclosure can include fragments not found as such in the natural state. Thus, this disclosure encompasses recombinant molecules, such as those in which a nucleic acid sequence (for example, a sequence encoding a mutant IL-2) is incorporated into a vector (e.g., a plasmid or viral vector) or into the genome of a heterologous cell (or the genome of a homologous cell, at a position other than the natural chromosomal location).


As described above, the subject IL-2 mutein may exist as a part of a chimeric polypeptide. In addition to, or in place of, the heterologous polypeptides described above, a subject nucleic acid molecule can contain sequences encoding a “marker” or “reporter.” Examples of marker or reporter genes include β-lactamase, chloramphenicol acetyltransferase (CAT), adenosine deaminase (ADA), aminoglycoside phosphotransferase (neor, G418r), dihydrofolate reductase (DHFR), hygromycin-B-hosphotransferase (HPH), thymidine kinase (TK), lacz (encoding β-galactosidase), and xanthine guanine phosphoribosyltransferase (XGPRT). One of skill in the art will be aware of additional useful reagents, for example, of additional sequences that can serve the function of a marker or reporter.


The subject nucleic acid molecules can be obtained by introducing a mutation into IL-2-encoding DNA obtained from any biological cell, such as the cell of a mammal. Thus, the subject nucleic acids (and the polypeptides they encode) can be those of a mouse, rat, guinea pig, cow, sheep, horse, pig, rabbit, monkey, baboon, dog, or cat. In one embodiment, the nucleic acid molecules will be those of a human.


G. Chimerica Antigen Receptors (Cars)


Targeted immunotherapy has emerged as promising field of research in the treatment of malignancies and has received a great deal of interest in recent years. Indeed, cures have been reported of lymphoma patients with engineered or genetically modified T cells targeting CD19 malignant cells. This has increased the focus towards antigens present on cancer cells as targets for gene- and immunotherapy. These CARS can be used to target or deliver the bispecific IL-2 cytokine fusion and/or IL-2 muteins described herein to the tumor, or even allow for systemic IL-2 mutein expression. In some embodiments, the IL-2 mutein is any IL-2 mutein or variant disclosed herein. In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO: 10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO: 11; D10 SEQ ID NO: 12; E10 SEQ ID NO: 13; G8 SEQ ID NO: 14; H4 SEQ ID NO:15; and H9 SEQ ID NO: 16. In some embodiments, the substitutions in the IL-2 mutein comprise L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2.


Genetic manipulation of autologous or allogeneic T cells or NK cells to specifically target a particular tumor antigen provides a strategy to bypass the failure of cytotoxic immune response induction by most tumor cells. In some embodiments, these genetically manipulated T-cells or NK cells can be used to target the IL-2 muteins described herein to the tumor, for example, so that the IL-2 mutein is expressed at the tumor location. These technologies are based on the genetic modification of human immune cells, where the cells may be extracted from a patient or donor by leukapheresis. Specific cells, usually T-cells, are purified and engineered to express a receptor targeting a cancer antigen of interest. Engineering may utilize transduction by retroviral, lentiviral, transposon, mRNA electroporation, and the like. The immune cells may be expanded to the desired dose, and introduced into a patient. The engineered cells can specifically kill cancer cells through cell-mediated toxicity (cytotoxic T-cells) and/or eliciting an immune response to the cancer cell by immune recognition of tumor, cytokine release and immune cell recruitment.


For example, the application of chimeric antigen receptors (CAR) for immunogene therapy of malignant tumors is a promising strategy in which an antibody or ligand binding domain is fused with the zeta signaling chain of the T cell receptor. The resulting CAR immune cells are redirected by the neospecificity to attack tumors expressing the surface antigen or receptors recognized by the gene-modified T cell receptors and provide cellular therapy that attacks the tumor through normal host immune response in a highly regulated fashion. These cells are free to circulate throughout the brain and systemic circulation, making the need for colocalization and bioavailability less of a problem.


A number of generations of CAR immune cells have been developed. CARs are created by the fusion of a tumour-specific scFv antibody or other extracellular ligand binding domain to either the TCR-associated CD3ζ signalling domain or another intracellular signalling domains from co-stimulatory protein receptors. This structure allows CARs to have the tumor specificity of the B cell antigen receptor, and to activate T cells through the T cell antigen receptor independently of MHC binding. The first-generation CAR contained one intracellular signalling domain, typically with the CD3ζ signalling domain to allow for TCR signalling. Second-generation CARs have two intracellular signalling domains: a co-stimulatory domain comprising either a CD28 or a 4-1BB signalling domain, coupled with a CD3ζ signalling domain. This arrangement enables T-cell activation and proliferation upon antigen recognition by the scFv region of the CAR. The third-generation CARs have two co-stimulatory domains and a CD3ζ signalling domain. The first co-stimulatory domain is either a CD28 or a 4-1BB domain, with the second co-stimulatory domain consisting of either a CD28, a 4-1BB or a OX40 domain. Fourth-generation “armoured CAR T cells” combine a second-generation CAR with the addition of various genes, including cytokine and co-stimulatory ligands, to enhance the tumoricidal effect of the CAR T cells. See, for example, Batlevi et al. (2016) Nature Reviews Clinical Oncology 13:25-40. See also, U.S. Pat. No. 7,741,465 and International Patent Publication No. WO2014127261; all of which are incorporated by reference herein in their entireties.


Alternative approaches to T cell targeting include T cell antigen couplers, as described in International application WO2015/117229, entitled “Trifunctional T cell antigen Coupler and Methods and Uses thereof”, herein specifically incorporated by reference. The T cell antigen coupler system comprises three linked domains: a target-specific polypeptide ligand; a ligand that binds a protein associated with the TCR complex, for example an scFv binding to CD3 (TCR, T-cell receptor) to stimulate T cell activation; and a T cell receptor signaling domain, for example a CD4 transmembrane and intracellular domain to amplify T cell activation. By stimulating T cell activation through the TCR, TACs were engineered to work with the T cell's essential molecular machinery.


Antibody coupled T cell receptors are another approach to T cell targeting. ACTRs are a hybrid approach to CARs and the established monoclonal antibody oncology therapeutics. ACTRs are composed of a typical CAR construct that can bind the heavy chain of an antibody through a high-affinity variant of the Fc receptor CD16. ACTR-T cells can target tumours by binding a ligand targeted to a specific cancer antigen. T cell activation is performed by the CAR module.


Bispecific T cell exchangers (BiTEs) are bispecific antibodies that can bind the TCR of T cells and target tumour cells through two modules: a cancer targeting ligand; and a CD3-binding scFv domain that bridges T cells to the tumor. In some embodiments, bi-specific fusion proteins comprising a superkine may also be referred to as Bi-functional SuperKines as ImmunoTherapies (BiSKITs). In some embodiments, novel interleukin super-agonists, partial agonists, and super-antagonists are designed using directed evolution. In some embodiments, rational approaches are used to further design long-acting IL-2, IL-4, and IL-13 Superkines without masking functional activity.


In some embodiments, a platform using directed evolution together with yeast display results in tunable superkines. In some embodiments, such platform generates an extensive library of IL-2, IL-4, and IL-13 superkines with unique properties (FIG. 55). In some embodiments, MDNA109 is an engineered version of human IL-2 showing enhanced agonist activity (FIG. 56). In some embodiments, MDNA109 family of ‘IL-2 Superkines’ have been engineered to improve PK characteristics and enhance selectivity to further improve therapeutic window (FIG. 57).


In some embodiments, MDNA11 is a ‘Beta-Only’ superkine with uniquely enhanced affinity for CD122 (FIG. 58). In some embodiments, MDNA11 preferentially stimulates immune effector cells (FIG. 59). In some embodiments, MDNA11 shows monotherapy anti-tumor efficacy and combination effect with anti-PD1 in MC38 tumor model (FIG. 60). In some embodiments, MDNA11 together with Anti-CTLA4 induces tumor clearance, protects against re-challenges, and promotes antigen-specific CD8 T-cells (FIG. 61). In some embodiments, MDNA11 induces durable and sustained proliferation and expansion of immune effector cells but not Tregs in NHP (FIG. 62).


In some embodiments, Superkine Targeted with Antibody (STAb) enhances accumulation in tumors (FIG. 63). In some embodiments, STAb overcomes checkpoint resistance and ‘cold’ tumors (FIG. 64).


In some embodiments, IL-4 and IL-13 receptors play a role in cancer (FIG. 65). In some embodiments, MDNA55 is an empowered Superkine with a potent payload targeting Type 2 IL-4R expressed on tumor cells and tumor microenvironment (MDSC and TAM). In some embodiments, MDNA413 is a super-antagonist blocking IL-4 and IL-13 signaling via type 2 IL-4R to suppress MDSC and TAM. In some embodiments, MDNA 132 is a superkine that selectively targets decoy IL-13Rα2 that is overexpressed on solid tumors. In some embodiments, MDNA132 is an engineered version of human IL-13 targeting tumor specific antigen (FIG. 66). In some embodiments, MDNA132 plays a role in localizing T-cell engager and checkpoint inhibitor to tumors (FIG. 67).


In some embodiments, MDNA413 is an engineered version of human IL-13 showing antagonist activity (FIG. 68). In some embodiments, Fc-MDNA413 inhibits IL-4 and IL-13 induced signaling and function (FIG. 69).


In some embodiments, a dual specific cytokine (DUCK Cancer) is MDNA109FEAA-Fc-MDNA413 and has a mechanism of action as shown in FIG. 70.


Targeted therapies have been developed against IL13Rα2, including bacterial toxins conjugated to IL13, nanoparticles, oncolytic virus, as well as immunotherapies using monoclonal antibodies, IL13Rα2-pulsed dendritic cells, and IL13Rα2-targeted chimeric antigen receptors (see Kahlon et al. (2004) Cancer Research. 64(24):9160-9166; Kong et al. (2012) Clinical Cancer Research. 18(21):5949-5960; Thaci et al. (2014) Neuro-Oncology; and clinical trials NCT02208362, NCT00730613 and NCT01082926). In some emnodiemtns, these targeted therapies can be used to deliver the IL-2 muteins to the tumor.


Biologicals that provide for selective alteration of IL-13 activity are of interest for a number of therapeutic purposes, including the treatment of certain cancers with by engineering of T cell specificities. The present invention addresses this issue.


Methods and compositions are provided for enhancing anti-tumor immune effector cells, e.g. T cells, NK cells, etc. with targeted compositions, including without limitation chimeric antigen receptors (CARs); T cell antigen couplers (TACs); antibody coupled T cell receptors (ACTRs); and bispecific T cell exchangers (BiTEs), where an IL-13 or IL-4 superkine provides the target-specific ligand. In further embodiments, the immune effector cell expresses an IL-2 mutein.


Immune cell targeting or expression constructs comprising IL-2 superkine sequences are provided and can include any IL-2 sequence as described herein. Superkines are useful for targeting immune cells to cells, e.g. tumor cells, expressing the at least one receptor. In some embodiments, the IL-2 mutein is any IL-2 mutein or variant disclosed herein. In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO:10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO:11; D10 SEQ ID NO: 12; E10 SEQ ID NO: 13; G8 SEQ ID NO:14; H4 SEQ ID NO: 15; and H9 SEQ ID NO: 16. In some embodiments, the substitutions in the IL-2 mutein comprise L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2.


The IL-2 superkine or mutein component of the construct may be at least about 50 amino acids in length, at least about 75, at least about 100, at least about 110, at least about 115 amino acids in length, up to the full-length of the wild-type protein at the transmembrane domain, i.e. about 116 amino acids in length. For example, the superkine or mutein may be fused to the hinge, transmembrane or signaling domains of a CAR. Exemplary polypeptide sequences are provided


Included as superkines or muteins are amino acid and nucleic acid coding sequences that are 90%, 95%, 98% or 99% identical to these sequences, longer sequences that comprise those sequences but also include additional nucleotides at the 3′ or 5′ end, for example any number of additional nucleotides or codons, such as 3, 6, 9, 12 or more nucleotides, or up to about 12, 20, 50 or 100 additional nucleotides, and any sequence that encodes the same amino acid sequence as these nucleic acids due to the degeneracy of the genetic code. In particular, sequences that are codon optimized (CO) for expression by the desired host are contemplated as part of the invention. In some embodiments, the amino acid sequence is 90% identical. In some embodiments, the amino acid sequence is 95% identical. In some embodiments, the amino acid sequence is 98% identical. In some embodiments, the amino acid sequence is 99% identical. In some embodiments, the polypeptide is linked to an IL-2 superkine immune cell targeting or expression construct. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises one or more signaling domains derived from CD3-ζ, CD28, DAP10, OX-40, ICOS and CD137. In some embodiments, an IL-2 superkine immune cell targeting or expression construct or expression comprises one or more signaling domains derived from CD3-ζ. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises one or more signaling domains derived from CD28. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises one or more signaling domains derived from DAP10. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises one or more signaling domains derived from OX-40. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises one or more signaling domains derived from CD137. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises an IL-2 variant/IL-2 superkine including those provided herein. In some embodiments, an IL-2superkine immune cell targeting or expression construct comprises an IL-2 variant/IL-2 superkine including those provided in SEQ ID NO:2 through SEQ ID NO:38.


1. NK Cells


In some embodiments the immune cells are natural killer (NK) cells. NK cells recognize infected or transformed cells through multiple cell surface receptors including NKG2D, CD16, and natural cytotoxicity receptors (NCRs) such as NKp44, NKp46, and NKp30. These receptors activate signaling adapter proteins such as DAP10, DAP12, and CD3ζ, which contain immuno-tyrosine activation motifs (ITAMs) that initiate the release of cytolytic granules containing perforin and granzymes, as well as mediate production and release of cytokines and chemokines such as IFN-γ and TNF-α. Importantly, NK cell-mediated cytotoxicity does not rely on the presentation of self HLA. Therefore, NK cells hold significant clinical interest as a cell-based therapy for cancer because of their ability to be used in an allogeneic setting and potentially provide an off-the-shelf cellular product.


Natural killer cells provide an alternative to the use of T cells for adoptive immunotherapy since they do not require HLA matching, so can be used as allogeneic effector cells. Clinical trials of adoptively transferred allogeneic NK cells demonstrate these cells can survive in patients for several weeks to months. Additionally, expression of CARs in NK cells allow these cells to more effectively kill solid tumors that are often resistant to NK cell-mediated activity compared to hematologic malignancies (especially acute myelogenous leukemia) that are typically more NK cell-sensitive. CARs useful in NK cell targeting include, for example, first generation CAR constructs that contain CD3ζ as the sole signaling domain. Second and third generation CARs are also useful in NK cells. In some embodiments the ectodomain of NKG2D, an NK cell activation receptor, is linked directly to CD3ζ.


NK cells for modification include cell lines, or peripheral blood NK cells, which can be isolated from donors through simple blood draws or by apheresis if larger numbers of cells are needed. Activated PB-NK cells express a wider range of activating receptors, such as CD16, NKp44, and NKp46 as well as KIRs, which play an important role in NK cell licensing. In addition, PB-NK cells can be given without irradiating the cells so have the ability to expand in vivo. Another source of NK cells suitable for CAR expression are NK cells derived from human pluripotent stem cells—both induced pluripotent stem cells (iPSCs) or human embryonic stem cells (hESCs). These NK cells display a similar phenotype to PB-NK cells, and hESC/iPSC-NK cells can be grown on a clinical scale.


2. Chimerica Antigen Receptors (CARs)


In addition to the superkine sequence, CARs contain the signaling domain for CD3ζ and the signaling domains of one or more costimulatory receptors that further promote the recycling, survival and/or expansion of immune cells expressing the CARs. The signaling domains of the costimulatory receptors are the intracellular portions of each receptor protein that generate the activating signal in the cell. Examples are amino acids 180-220 of the native CD28 molecule and amino acids 214-255 of the native 4-1BB molecule.


Examples of suitable hinge and transmembrane regions to link the superkine to the signaling region may include without limitation the constant (Fc) regions of immunoglobins, human CD8a, and artificial linkers that serve to move the targeting moiety away from the cell surface for improved access to and binding on target cells. Examples of suitable transmembrane domains include the transmembrane domains of the leukocyte CD markers, preferably that of CD4 or CD28. Examples of intracellular receptor signaling domains include the T cell antigen receptor complex, preferably the zeta chain of CD3, however any transmembrane region sufficient to anchor the CAR in the membrane can be used. Persons of skill are aware of numerous transmembrane regions and the structural elements (such as lipophilic amino acid regions) that produce transmembrane domains in numerous membrane proteins and therefore can substitute any convenient sequence. T cell costimulatory signaling receptors suitable for improving the function and activity of CAR-expressing cells include, but are not limited to, CD28, CD137, and OX-40.


Signaling via CD28 is required for IL2 production and proliferation, but does not play a primary role in sustaining T cell function and activity. CD137 (a tumor necrosis factor-receptor family member expressed following CD28 activation) and OX-40 are involved in driving long-term survival of T cells, and accumulation of T cells. The ligands for these receptors typically are expressed on professional antigen presenting cells such as dendritic cells and activated macrophages, but not on tumor cells. Expressing a CAR that incorporates CD28 and/or 4-1BB signaling domains in CD4+ T cells enhances the activity and anti-tumor potency of those cells compared to those expressing a CAR that contains only the CD3ζ signaling domain, which constructs may be referred to as second or third generation CARs.


Included as CAR constructs of interest are tandem CARs, e.g. see Hegde et al. (2016) J. Clin. Invest 126(8):3036-3052, herein specifically incorporated by reference. In such constructs a binding moiety for a tumor specific antigen is combined in tandem with an IL-13 superkine. The binding moiety may be, for example, an scFv specific for a tumor cell antigen, including without limitation HER-2, EGFR, CD20, etc. as known in the art.


In various embodiments, the antigen binding domain binds to an antigen on a target cell, e.g., a cancer cell. The antigen binding domain can bind an antigen, such as but not limited to a tumor target antigen. In some case, the antigen binding domain binds one or more antigens. Exemplary antigen binding domains can bind to an antigen including, but not limited to, D19; CD123; CD22; CD30; CD171; CS-1 (also referred to as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A24); C-type lectin-like molecule-1 (CLL-1 or CLECL1); CD33; epidermal growth factor receptor variant III (EGFRvIII); ganglioside G2 (GD2); ganglioside GD3; TNF receptor family member B cell maturation (BCMA); Tn antigen ((Tn Ag) or (GalNAcu Ser/Thr)); prostate-specific membrane antigen (PSMA); Receptor tyrosine kinase-like orphan receptor 1 (ROR1); Fms-Like Tyrosine Kinase 3 (FLT3); Tumor-associated glycoprotein 72 (TAG72); CD3ε; CD44v6; Carcinoembryonic antigen (CEA); Epithelial cell adhesion molecule (EPCAM); B7H3 (CD276); KIT (CD117); Interleukin-13 receptor subunit alpha-2 (IL-13Rα2 or CD213A2); Mesothelin; Interleukin 11 receptor alpha (IL-IIRa); prostate stem cell antigen (PSCA); Protease Serine 21 (Testisin or PRSS21); vascular endothelial growth factor receptor 2 (VEGFR2); Lewis(Y) antigen; CD24; Platelet-derived growth factor receptor beta (PDGFR-beta); Stage-specific embryonic antigen-4 (SSEA-4); CD20; Folate receptor alpha; Receptor tyrosine-protein kinase ERBB2 (Her2/neu); Mucin 1, cell surface associated (MUC1); epidermal growth factor receptor (EGFR); neural cell adhesion molecule (NCAM); Prostase; prostatic acid phosphatase (PAP); elongation factor 2 mutated (ELF2M); Ephrin B2; fibroblast activation protein alpha (FAP); insulin-like growth factor 1 receptor (IGF-I receptor), carbonic anhydrase IX (CAIX); Proteasome (Prosome, Macropain) Subunit, Beta Type, 9 (LMP2); glycoprotein 100 (gp 100); oncogene fusion protein consisting of breakpoint cluster region (BCR) and Abelson murine leukemia viral oncogene homolog 1 (Abl) (bcr-abl); tyrosinase; ephrin type-A receptor 2 (EphA2); Fucosyl GM1; sialyl Lewis adhesion molecule (sLe); ganglioside GM3 (aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer); transglutaminase 5 (TGS5); high molecular weight-melanoma-associated antigen (HMWMAA); o-acetyl-GD2 ganglioside (OAcGD2); Folate receptor beta; tumor endothelial marker 1 (TEM1/CD248); tumor endothelial marker 7-related (TEM7R); claudin 6 (CLDN6); thyroid stimulating hormone receptor (TSHR); G protein-coupled receptor class C group 5, member D (GPRC5D); chromosome X open reading frame 61 (CXORF61); CD97; CD179a; anaplastic lymphoma kinase (ALK); Polysialic acid; placenta-specific 1 (PLAC1); hexasaccharide portion of globoH glycoceramide (GloboH); mammary gland differentiation antigen (NY-BR-1); uroplakin 2 (UPK2); Hepatitis A virus cellular receptor 1 (HAVCR1); adrenoceptor beta 3 (ADRB3); pannexin 3 (PANX3); G protein-coupled receptor 20 (GPR20); lymphocyte antigen 6 complex, locus K 9 (LY6K); Olfactory receptor 51E2 (OR51E2); TCR Gamma Alternate Reading Frame Protein (TARP); Wilms tumor protein (WT1); Cancer/testis antigen 1 (NY-ESO-1); Cancer/testis antigen 2 (LAGE-1a); Melanoma-associated antigen 1 (MAGE-A1); ETS translocation-variant gene 6, located on chromosome 12p (ETV6-AML); sperm protein 17 (SPA17); X Antigen Family, Member 1A (XAGE1); angiopoietin-binding cell surface receptor 2 (Tie 2); melanoma cancer testis antigen-1 (MAD-CT-1); melanoma cancer testis antigen-2 (MAD-CT-2); Fos-related antigen 1; tumor protein p53 (p53); p53 mutant; prostein; surviving; telomerase; prostate carcinoma tumor antigen-1 (PCTA-1 or Galectin 8), melanoma antigen recognized by T cells 1 (MelanA or MART 1); Rat sarcoma (Ras) mutant; human telomerase reverse transcriptase (hTERT); sarcoma translocation breakpoints; melanoma inhibitor of apoptosis (ML-IAP); ERG (transmembrane protease, serine 2 (TMPRSS2) ETS fusion gene); N-Acetyl glucosaminyl-transferase V (NA17); paired box protein Pax-3 (PAX3); androgen receptor; Cyclin B1; v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN); Ras Homolog Family Member C (RhoC); Tyrosinase-related protein 2 (TRP-2); cytochrome P450 1B1 (CYP1B1); CCCTC-Binding Factor (Zinc Finger Protein)-Like (BORIS or Brother of the Regulator of Imprinted Sites), Squamous Cell Carcinoma Antigen Recognized By T Cells 3 (SART3); Paired box protein Pax-5 (PAX5); proacrosin binding protein sp32 (OY-TES1); lymphocyte-specific protein tyrosine kinase (LCK); A kinase anchor protein 4 (AKAP-4); synovial sarcoma, X breakpoint 2 (SSX2); Receptor for Advanced Glycation Endproducts (RAGE-1); renal ubiquitous 1 (RU1); renal ubiquitous 2 (RU2); legumain; human papilloma virus E6 (HPV E6); human papilloma virus E7 (HPV E7); intestinal carboxyl esterase; heat shock protein 70-2 mutated (mut hsp70-2); CD79a; CD79b; CD72; Leukocyte-associated immunoglobulin-like receptor 1 (LAIR1); Fc fragment of IgA receptor (FCAR or CD89); Leukocyte immunoglobulin-like receptor subfamily A member 2 (LILRA2); CD300 molecule-like family member f (CD300LF); C-type lectin domain family 12 member A (CLECi2A); bone marrow stromal cell antigen 2 (BST2); EGF-like module-containing mucin-like hormone receptor-like 2 (EMR2); lymphocyte antigen 75 (LY75); Glypican-3 (GPC3); Fc receptor-like 5 (FCRL5); and immunoglobulin lambda-like polypeptide 1 (IGLL1).


In some embodiments, the antigen binding domain comprises a monoclonal antibody, a polyclonal antibody, a synthetic antibody, a human antibody, a humanized antibody, a non-human antibody, a nanobody, a single-chain variable fragment (scFv), F(ab′)2, Fab′, Fab, Fv, and the like. The antigen binding domain can be linked to the transmembrane domain of the CAR. In some embodiments, a nucleic acid encoding the antigen binding domain is operably linked to a nucleic acid encoding a transmembrane domain of the CAR.


In some embodiments, the transmembrane domain can be derived from a membrane-bound or transmembrane protein. In certain embodiments, the transmembrane domain comprises one or more, e.g., 1, 2, 3, 4, 5, 6, 7, 8 or more amino acid modifications (e.g., substitutions, insertions, and deletions) compared to the wild-type amino acid sequence of the transmembrane domain of the membrane-bound or transmembrane protein. Non-limiting examples of a transmembrane domain of a CAR include at least the transmembrane region(s) of the alpha, beta or zeta chain of the T-cell receptor, CD28, CD3 epsilon (CD34), CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, or an erythropoietin receptor. In some embodiments, the transmembrane domain includes a human immunoglobulin (Ig) hinge region, e.g., an IgG4Fc hinge. In other embodiments, the transmembrane domain is a recombinant or synthetic domain comprising hydrophobic amino acid residues (e.g., leucine and valine). In some cases, the transmembrane domain includes a phenylalanine, tryptophan and valine at one or both ends of the domain.


The transmembrane domain links the antigen binding domain to the intracellular signaling domain of the CAR. In some embodiments, the nucleic acid encoding the antigen binding domain is operably linked to the nucleic acid encoding the transmembrane domain that is operably linked to the nucleic acid encoding the intracellular signaling domain.


In some embodiments, the intracellular signaling domain of a CAR comprises a signal activation or signal transduction domain. As such, an intracellular signaling domain includes any portion of an intracellular signaling domain of a protein sufficient to transduce or transmit a signal, e.g., an activation signal or to mediate a cellular response within a cell. Non-limiting examples include TCR, CD2, CD3 zeta, CD3 gamma, CD3 delta, CD3 epsilon, CD7, CD27, CD86, common FcR gamma, FcR beta, CD79a, CD79b, Fcgamma RIIa, DAP10, DAP12, T cell receptor (TCR), CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand that specifically binds with CD83, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD127, CD160, CD19, CD4, CD8alpha, CD8 beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAMI, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMFI, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, any derivative, variant, or fragment thereof. In certain embodiments, the intracellular signaling domain comprises an intracellular domain of a co-stimulatory molecule such as from CD3, CD27, CD28, CD127, ICOS, 4-1BB (CD137), PD-1, T cell receptor (TCR), any derivative thereof, or any variant thereof. In some embodiments, the intracellular signaling domain of the CAR is selected from the group consisting of a MHC class I molecule, a TNF receptor protein, an Immunoglobulin-like protein, a cytokine receptor, an integrin, a signaling lymphocytic activation molecule (SLAM protein), an activating NK cell receptor, BTLA, a Toll ligand receptor, OX40, CD2, CD7, CD27, CD28, CD30, CD40, CDS, ICAM-1, LFA-1 (CD11a/CD18), 4-1BB (CD137), B7-H3, CDS, ICAM-1, ICOS (CD278), GITR, BAFFR, LIGHT, HVEM (LIGHTR), KIRDS2, SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD19, CD4, CD8alpha, CD8beta, IL2R beta, IL2R gamma, IL7R alpha, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, NKG2D, NKG2C, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAMI, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), CD69, SLAMF6 (NTB-A, Ly108), SLAM (SLAMFI, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, CD19a, and a ligand that specifically binds with CD83.


3. Bites


Bi-specific T-cell engagers (BiTEs) are fusion proteins comprising an IL-13 superkine fused to an antibody variable region that specifically binds to CD3. In some embodiments the antibody variable region in a single-chain variable fragments (scFvs). The superkine may be fused to the variable region through a linker. An Fc region is optionally provided.


4. TACs


A TAC construct comprises an IL-2 superkine fused to a ligand that binds a protein associated with the TCR complex; fused to a T cell receptor signaling domain polypeptide. The domains may be separated by linkers. The protein associated with the TCR complex may be CD3. The ligand that binds a protein associated with the TCR complex may be a single chain antibody. The ligand that binds a protein associated with the TCR complex may be UCHT1, or a variant thereof. The T cell receptor signaling domain polypeptide may comprise a cytosolic domain and a transmembrane domain. The cytosolic domain may be a CD4 cytosolic domain and the transmembrane domain is a CD4 transmembrane domain.


5. ACTRs


ACTRs are a hybrid approach to CARs and the established monoclonal antibody oncology therapeutics. ACTRs are composed of a typical CAR construct that can bind the heavy chain of an antibody through a high-affinity variant of the Fc receptor CD16. A superkine is fused to a moiety recognized by the CAR, which may include, without limitation, an Fc region of an antibody with high affinity for CD16.


An immune cell targeting or expression construct coding sequence can be produced by any means known in the art, including recombinant DNA techniques. Nucleic acids encoding the several regions of the chimeric receptor can be prepared and assembled into a complete coding sequence by standard techniques of molecular cloning known in the art (genomic library screening, PCR, primer-assisted ligation, site-directed mutagenesis, etc.) as is convenient. The resulting coding region may be inserted into an expression vector and used to transform a suitable expression host cell line, e.g. a population of allogeneic or autologous T lymphocytes, allogeneic or autologous NK cells, including primary cultures, cell lines, iPSC derived cells, etc. The methods can be used on cells in vitro (e.g., in a cell-free system), in culture, e.g. in vitro or ex vivo. For example, IL-2 superkine CAR-expressing cells can be cultured and expanded in vitro in culture medium.


An non-IL-2 superkine immune cell targeting or expression construct can also be used specifically direct immune cells to target specific tumor cells. Anti-tumor effector cells, e.g. CD4+ or CD8+ effector T cells, are generated to be re-directed to recognize such tumor cells by introducing into the T cells an superkine immune cell targeting or expression construct comprising one or more signaling domains derived from CD3-ζ, CD28, DAP10, OX-40, ICOS and CD137. In some embodiments, the cells can further comprise a transgene capable of expressing an IL-2 mutein as described herein. An IL-2 superkine immune cell targeting or expression construct can specifically direct immune cells to target IL-2R expressing cell, including tumor cells. Anti-tumor effector cells, e.g. CD4+ or CD8+ effector T cells, are generated to be re-directed to recognize such tumor cells by introducing into the T cells an IL-2 superkine immune cell targeting or expression construct comprising one or more signaling domains derived from CD3-ζ, CD28, DAP10, OX-40, ICOS and CD137.


The IL-2 superkine immune cell targeting or expression construct is infected or transfected into human immune cells, e.g. using a non-viral plasmid vector and electroporation methods; a viral vector and infection methods, etc. as known in the art. A CAR comprising co-stimulatory signaling domains may enhance the duration and/or retention of anti-tumor activity in a manner that can significantly improve the clinical efficacy of adoptive therapy protocols. CD4+ and CD8+ T cell effector functions, and NK cell functions can be triggered via these receptors, therefore these cell types are contemplated for use with the invention. CD8+ T cells expressing the IL13 superkine CARs of this invention may be used to lyse target cells and to produce IL-2 in the presence of target cells, among the other functions of these cells. Expression of the appropriate costimulatory CAR in either or both CD4+ and CD8+ T cells is used to provide the most effective population of cells for adoptive immunotherapy, consisting therefore of either or both professional helper and killer T cells that exhibit enhanced and/or long term viability and anti-tumor activity. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises an IL-2 variant/IL-2 superkine including those provided in FIG. 2. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises an IL-2 variant/IL-2 superkine including any of those provided herein.


Polypeptides of the present invention can be further modified, e.g., joined to a wide variety of other oligopeptides or proteins for a variety of purposes. For example, post-translationally modified, for example by prenylation, acetylation, amidation, carboxylation, glycosylation, pegylation, etc. Such modifications can also include modifications of glycosylation, e.g. those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes which affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes.


Methods which are well known to those skilled in the art can be used to construct T cell targeting construct expression vectors containing coding sequences and appropriate transcriptional/translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. Alternatively, RNA capable of encoding the polypeptides of interest may be chemically synthesized. One of skill in the art can readily utilize well-known codon usage tables and synthetic methods to provide a suitable coding sequence for any of the polypeptides of the invention. The nucleic acids may be isolated and obtained in substantial purity. Usually, the nucleic acids, either as DNA or RNA, will be obtained substantially free of other naturally-occurring nucleic acid sequences, generally being at least about 50%, usually at least about 90% pure and are typically “recombinant,” e.g., flanked by one or more nucleotides with which it is not normally associated on a naturally occurring chromosome. The nucleic acids of the invention can be provided as a linear molecule or within a circular molecule, and can be provided within autonomously replicating molecules (vectors) or within molecules without replication sequences. Expression of the nucleic acids can be regulated by their own or by other regulatory sequences known in the art. The nucleic acids of the invention can be introduced into suitable host cells using a variety of techniques available in the art.


According to the present invention, immune cell targeting or expression construct vectors and immune cell targeting or expression construct modified cells can be provided in pharmaceutical compositions suitable for therapeutic use, e.g. for human treatment. In some embodiments, pharmaceutical compositions of the present invention include one or more therapeutic entities of the present invention or pharmaceutically acceptable salts, esters or solvates thereof. In some other embodiments, pharmaceutical compositions of the present invention include one or more therapeutic entities of the present invention in combination with another therapeutic agent, e.g., another anti-tumor agent.


Therapeutic entities of the present invention are often administered as pharmaceutical compositions comprising an active therapeutic agent and another pharmaceutically acceptable excipient. Such formulations can include one or more non-toxic pharmaceutically acceptable carriers, diluents, excipients and/or adjuvants. The preferred form depends on the intended mode of administration and therapeutic application. The compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration. The diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological phosphate-buffered saline, Ringer's solutions, dextrose solution, and Hank's solution. In addition, the pharmaceutical composition or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, nonimmunogenic stabilizers and the like.


In still some other embodiments, pharmaceutical compositions of the present invention can also include large, slowly metabolized macromolecules such as proteins, polysaccharides such as chitosan, polylactic acids, polyglycolic acids and copolymers (such as latex functionalized Sepharose™, agarose, cellulose, and the like), polymeric amino acids, amino acid copolymers, and lipid aggregates (such as oil droplets or liposomes).


The maximum tolerated dose (MTD) of CAR immune cells may be determined during clinical trial development, for example at up to about 104 T cells/kg of body weight, up to about 105 cells/kg of body weight, up to about 106 cells/kg of body weight, up to about 5×106 cells/kg of body weight, up to about 107 cells/kg of body weight, up to about 5×107 cells/kg of body weight, or more, as empirically determined. In some embodiments, the maximum tolerated dose (MTD) of CAR immune cells is up to about 104 T cells/kg of body weight. In some embodiments, the maximum tolerated dose (MTD) of CAR immune cells is up to about 105 T cells/kg of body weight. In some embodiments, the maximum tolerated dose (MTD) of CAR immune cells is up to about 106 T cells/kg of body weight. In some embodiments, the maximum tolerated dose (MTD) of CAR immune cells is up to about 107 T cells/kg of body weight. In some embodiments, the maximum tolerated dose (MTD) of CAR immune cells is up to about 5×106 T cells/kg of body weight. In some embodiments, the maximum tolerated dose (MTD) of CAR immune cells is up to about 5×107 T cells/kg of body weight.


Toxicity of the cells described herein can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., by determining the LD50 (the dose lethal to 50% of the population) or the LD100 (the dose lethal to 100% of the population). The dose ratio between toxic and therapeutic effect is the therapeutic index. The data obtained from these cell culture assays and animal studies can be used in formulating a dosage range that is not toxic for use in human. The dosage of the described herein lies preferably within a range of circulating concentrations that include the effective dose with little or no toxicity. The dosage can vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition.


After a dose escalation phase, patients in the expansion cohort are treated with immune cells at the MTD. An exemplary treatment regime entails administration once every two weeks or once a month or once every 3 to 6 months. Therapeutic entities of the present invention are usually administered on multiple occasions. Intervals between single dosages can be weekly, monthly or yearly. Intervals can also be irregular as indicated by measuring blood levels of the therapeutic entity in the patient.


In prophylactic applications, e.g. to maintain remission in a patient, a relatively low dosage may be administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In other therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patent can be administered a prophylactic regime.


Examples of additional therapeutic agents that can be coadministered and/or coformulated with an immune cell targeting or expression construct include: anti-proliferative, or cytoreductive therapy, which is used therapeutically to eliminate tumor cells and other undesirable cells in a host, and includes the use of therapies such as delivery of ionizing radiation, and administration of chemotherapeutic agents. Chemotherapeutic agents are well-known in the art and are used at conventional doses and regimens, or at reduced dosages or regimens, including for example, topoisomerase inhibitors such as anthracyclines, including the compounds daunorubicin, adriamycin (doxorubicin), epirubicin, idarubicin, anamycin, MEN 10755, and the like. Other topoisomerase inhibitors include the podophyllotoxin analogues etoposide and teniposide, and the anthracenediones, mitoxantrone and amsacrine. Other anti-proliferative agent interferes with microtubule assembly, e.g. the family of vinca alkaloids. Examples of vinca alkaloids include vinblastine, vincristine; vinorelbine (NAVELBINE); vindesine; vindoline; vincamine; etc. DNA-damaging agent include nucleotide analogs, alkylating agents, etc. Alkylating agents include nitrogen mustards, e.g. mechlorethamine, cyclophosphamide, melphalan (L-sarcolysin), etc.; and nitrosoureas, e.g. carmustine (BCNU), lomustine (CCNU), semustine (methyl-CCNU), streptozocin, chlorozotocin, etc. Nucleotide analogs include pyrimidines, e.g. cytarabine (CYTOSAR-U), cytosine arabinoside, fluorouracil (5-FU), floxuridine (FUdR), etc.; purines, e.g. thioguanine (6-thioguanine), mercaptopurine (6-MP), pentostatin, fluorouracil (5-FU) etc.; and folic acid analogs, e.g. methotrexate, 10-propargyl-5,8-dideazafolate (PDDF, CB3717), 5,8-dideazatetrahydrofolic acid (DDATHF), leucovorin, etc. Other chemotherapeutic agents of interest include metal complexes, e.g. cisplatin (cis-DDP), carboplatin, oxaliplatin, etc.; ureas, e.g. hydroxyurea; and hydrazines, e.g. N-methylhydrazine.


For example, ionizing radiation (IR) is used to treat about 60% of cancer patients, by depositing energy that injures or destroys cells in the area being treated, and for the purposes of the present invention may be delivered at conventional doses and regimens, or at reduced doses. Radiation injury to cells is nonspecific, with complex effects on DNA. The efficacy of therapy depends on cellular injury to cancer cells being greater than to normal cells. Radiotherapy may be used to treat every type of cancer. Some types of radiation therapy involve photons, such as X-rays or gamma rays. Another technique for delivering radiation to cancer cells is internal radiotherapy, which places radioactive implants directly in a tumor or body cavity so that the radiation dose is concentrated in a small area. A suitable dose of ionizing radiation may range from at least about 2 Gy to not more than about 10 Gy, usually about 5 Gy. A suitable dose of ultraviolet radiation may range from at least about 5 J/m2 to not more than about 50 J/m2, usually about 10 J/m2. The sample may be collected from at least about 4 and not more than about 72 hours following ultraviolet radiation, usually around about 4 hours.


Treatment may also be combined with immunoregulatory modulating agents, including an agent that agonizes an immune costimulatory molecule, e.g. CD40, OX40, etc.; and/or (iii) an agent that antagonizes an immune inhibitory molecule, e.g. CTLA-4, PD-1, PD-L1, etc. The active agents are administered within a period of time to produce an additive or synergistic effect on depletion of cancer cells in the host. Methods of administration include, without limitation, systemic administration, intra-tumoral administration, etc.


In some embodiments, an individual cancer is selected for treatment with a combination therapy because the cancer is a cancer type that is responsive to a checkpoint inhibitor, e.g. a PD-1 antagonist, a PD-L1 antagonist, a CTLA4 antagonist, a TIM-3 antagonist, a BTLA antagonist, a VISTA antagonist, a LAG3 antagonist; etc. In some embodiments, such an immunoregulatory agent is a CTLA-4, PD1 or PDL1 antagonist, e.g. avelumab, nivolumab, pembrolizumab, ipilimumab, and the like. In some such embodiments the cancer is, without limitation, melanoma or small cell lung cancer. In some such embodiments, the cancer is a type that has a high neoantigen, or mutagenesis, burden (see Vogelstein et al. (2013) Science 339(6127):1546-1558, herein specifically incorporated by reference).


In some embodiments, an individual cancer is selected for treatment with a combination therapy of the present invention because the cancer is a cancer type that is responsive to an immune response agonist, e.g. a CD28 agonist, an OX40 agonist; a GITR agonist, a CD137 agonist, a CD27 agonist, an HVEM agonist, etc. In some embodiments, such an immunoregulatory agent is an OX40, CD137, or GITR agonist e.g. tremelimumab, and the like. In some such embodiments the cancer is, without limitation, melanoma or small cell lung cancer. In some such embodiments, the cancer is a type that has a high neoantigen, or mutagenesis, burden.


In some embodiments, the combination therapy includes an antibody known in the art which binds to PD-1 and disrupt the interaction between the PD-1 and its ligand, PD-L1, and stimulate an anti-tumor immune response. In some embodiments, the antibody or antigen-binding portion thereof binds specifically to PD-1. For example, antibodies that target PD-1 and which can find used in the present invention include, e.g., but are not limited to nivolumab (BMS-936558, Bristol-Myers Squibb), pembrolizumab (lambrolizumab, MK03475 or MK-3475, Merck), humanized anti-PD-1 antibody JS001 (ShangHai JunShi), monoclonal anti-PD-1 antibody TSR-042 (Tesaro, Inc.), Pidilizumab (anti-PD-1 mAb CT-011, Medivation), anti-PD-1 monoclonal Antibody BGB-A317 (BeiGene), and/or anti-PD-1 antibody SHR-1210 (ShangHai HengRui), human monoclonal antibody REGN2810 (Regeneron), human monoclonal antibody MDX-1106 (Bristol-Myers Squibb), and/or humanized anti-PD-1 IgG4 antibody PDR001 (Novartis). In some embodiments, the PD-1 antibody is from clone: RMP1-14 (rat IgG)-BioXcell cat #BP0146. Other suitable antibodies include anti-PD-1 antibodies disclosed in U.S. Pat. No. 8,008,449, herein incorporated by reference. In some embodiments, the antibody or antigen-binding portion thereof binds specifically to PD-L1 and inhibits its interaction with PD-1, thereby increasing immune activity. Any antibodies known in the art which bind to PD-L1 and disrupt the interaction between the PD-1 and PD-L1, and stimulates an anti-tumor immune response, are suitable for use in the combination treatment methods disclosed herein. For example, antibodies that target PD-L1 and are in clinical trials, include BMS-936559 (Bristol-Myers Squibb) and MPDL3280A (Genetech). Other suitable antibodies that target PD-L1 are disclosed in U.S. Pat. No. 7,943,743, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to PD-1 or PD-L1, disrupts the PD-1/PD-L1 interaction, and stimulates an anti-tumor immune response, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds CTLA-4 and disrupts its interaction with CD80 and CD86. Exemplary antibodies that target CTLA-4 include ipilimumab (MDX-010, MDX-101, Bristol-Myers Squibb), which is FDA approved, and tremelimumab (ticilimumab, CP-675, 206, Pfizer), currently undergoing human trials. Other suitable antibodies that target CTLA-4 are disclosed in WO 2012/120125, U.S. Pat. Nos. 6,984,720, 6,682,7368, and U.S. Patent Applications 2002/0039581, 2002/0086014, and 2005/0201994, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to CTLA-4, disrupts its interaction with CD80 and CD86, and stimulates an anti-tumor immune response, is suitable for use in the combination treatment methods. In some embodiments, the combination therapy includes an antibody known in the art which binds LAG-3 and disrupts its interaction with MHC class II molecules. An exemplary antibody that targets LAG-3 is IMP321 (Immutep), currently undergoing human trials. Other suitable antibodies that target LAG-3 are disclosed in U.S. Patent Application 2011/0150892, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to LAG-3, disrupts its interaction with MHC class II molecules, and stimulates an anti-tumor immune response, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds TIM-3 and disrupts its interaction with galectin 9. Suitable antibodies that target TIM-3 are disclosed in U.S. Patent Application 2013/0022623, herein incorporated by reference. It will be understood by one of ordinary skill that any antibody which binds to TIM-3, disrupts its interaction with galectin 9, and stimulates an anti-tumor immune response, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds 4-1BB/CD137 and disrupts its interaction with CD137L. It will be understood by one of ordinary skill that any antibody which binds to 4-1BB/CD137, disrupts its interaction with CD137L or another ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds GITR and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to GITR, disrupts its interaction with GITRL or another ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds OX40 and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to OX40, disrupts its interaction with OX40L or another ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds CD40 and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to CD40, disrupts its interaction with its ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds ICOS and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to ICOS, disrupts its interaction with its ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds CD28 and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to CD28, disrupts its interaction with its ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the combination treatment methods.


In some embodiments, the combination therapy includes an antibody known in the art which binds IFNα and disrupts its interaction with its ligand. It will be understood by one of ordinary skill that any antibody which binds to IFNα, disrupts its interaction with its ligand, and stimulates an anti-tumor immune response or an immune stimulatory response that results in anti-tumor activity overall, is suitable for use in the combination treatment methods.


An “anti-cancer therapeutic” is a compound, composition, or treatment (e.g., surgery) that prevents or delays the growth and/or metastasis of cancer cells. Such anti-cancer therapeutics include, but are not limited to, surgery (e.g., removal of all or part of a tumor), chemotherapeutic drug treatment, radiation, gene therapy, hormonal manipulation, immunotherapy (e.g., therapeutic antibodies and cancer vaccines) and antisense or RNAi oligonucleotide therapy. Examples of useful chemotherapeutic drugs include, but are not limited to, hydroxyurea, busulphan, cisplatin, carboplatin, chlorambucil, melphalan, cyclophosphamide, Ifosphamide, danorubicin, doxorubicin, epirubicin, mitoxantrone, vincristine, vinblastine, Navelbine® (vinorelbine), etoposide, teniposide, paclitaxel, docetaxel, gemcitabine, cytosine, arabinoside, bleomycin, neocarcinostatin, suramin, taxol, mitomycin C, Avastin, Herceptin®, flurouracil, and temozolamide and the like.


The compounds are also suitable for use with standard combination therapies employing two or more chemotherapeutic agents. It is to be understood that anti-cancer therapeutics includes novel compounds or treatments developed in the future.


The pharmaceutical compositions and/or formulations described above include one or more therapeutic entities in an amount effective to achieve the intended purpose. Thus the term “therapeutically effective dose” refers to the amount of the therapeutic entities that ameliorates the symptoms of cancer. Determination of a therapeutically effective dose of a compound is well within the capability of those skilled in the art. For example, the therapeutically effective dose can be estimated initially either in cell culture assays, or in animal models, such as those described herein. Animal models can also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in other animals, including humans, using standard methods known in those of ordinary skill in the art.


Also within the scope of the invention are kits comprising the compositions of the invention and instructions for use. The kit may further contain a least one additional reagent, e.g. a chemotherapeutic drug, anti-tumor antibody, etc. Kits typically include a label indicating the intended use of the contents of the kit. The term label includes any writing, or recorded material supplied on or with the kit, or which otherwise accompanies the kit.


The invention now being fully described, it will be apparent to one of ordinary skill in the art that various changes and modifications can be made without departing from the spirit or scope of the invention. In some embodiments, the kit comprises an IL-2 superkine immune cell targeting or expression construct comprising an IL-2 variant/IL-2 superkine as described herein. In some embodiments, the kit comprises an IL-2 superkine immune cell targeting or expression construct comprising an IL-2 variant/IL-2 superkine including those provided herein. In some embodiments, an IL-2 superkine immune cell targeting or expression construct comprises an IL-2 variant/IL-2 superkine including those provided herein.


6. Exemplary Immune Cell Targeting or Expression Construct Embodiments


An immune cell targeting or expression construct comprising: an interleukin-2 receptor β (IL-2Rβ) binding protein, wherein the equilibrium dissociation constant for the IL-2Rβ of the binding protein is less than that of wild-type human IL-2 (hIL-2); linked to an immune cell targeting or expression construct. In some embodiments, a bispecific IL-2 cytokine fusion is also an interleukin-2 receptor β (IL-2Rβ) binding protein.


In some embodiments, the immune cell targeting or expression construct exhibits a cyotoxic effect on a T-cell, for example a CD8+ T-cell or a CD4+ T-cell.


In some embodiments, the construct is a chimeric antigen receptor (CAR) and wherein the IL-2 superkine is fused to a transmembrane domain; linked to an intracellular signaling region.


In some embodiments, the intracellular signaling region comprises a CD3 signaling domain.


In some embodiments, the intracellular signaling region comprises one or more of a CD28 signaling domain, a CD137 signaling domain, an OX-40 signaling domain, an ICOS signaling domain, a DAP10 signaling domain.


In some embodiments, the construct is a T cell antigen coupler (TAC), wherein the IL-2 superkine is fused to a ligand that binds a protein associated with the TCR complex; fused to a T cell receptor signaling domain polypeptide.


In some embodiments, the protein associated with the TCR complex is CD3.


In some embodiments, the T cell receptor signaling domain polypeptide comprises CD4 cytosolic domain and CD4 transmembrane domain.


In some embodiments, the construct is an antibody coupled T cell receptors (ACTR), comprising a chimeric antigen receptor component that binds to the IL-2 superkine at a high affinity.


In some embodiments, the CAR component comprises CD16, and the IL-2 superkine is fused to an Fc sequence.


In some embodiments, the construct is a bispecific T cell exchanger (BiTE) comprising an IL-2 superkine fused to a variable region of an antibody that binds to a component of a T cell receptor.


In some embodiments, the BiTE component of a T cell receptor is CD3.


In some embodiments, the IL-2Rβ binding protein comprises the following amino acid substitutions: L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type hIL-2.


In some embodiments, the nucleic acid encoding and IL-2 described herein is provided. In some embodiments, the vector comprising the nucleic acid is provided.


In some embodiments, a T cell comprising a construct according to any of the above is provided. In some embodiments, an NK cell comprising a construct according to any of the above is provided. In some embodiments, the T cell is a CD4+ T cell. In some embodiments, the T cell is a CD8+ T cell.


Also provide are an isolated population of immune cells described above. Also provided are pharmaceutical formulations comprising the immune cell population described above.


H. Expression of Mutant IL-2 Gene Products


The nucleic acid molecules described above can be contained within a vector that is capable of directing their expression in, for example, a cell that has been transduced with the vector. Accordingly, in addition to the subject IL-2 muteins and/or bispecific IL-2 cytokine fusions, expression vectors containing a nucleic acid molecule encoding a subject IL-2 mutein and cells transfected with these vectors are among the preferred embodiments.


It should of course be understood that not all vectors and expression control sequences will function equally well to express the DNA sequences described herein. Neither will all hosts function equally well with the same expression system. However, one of skill in the art may make a selection among these vectors, expression control sequences and hosts without undue experimentation. For example, in selecting a vector, the host must be considered because the vector must replicate in it. The vector's copy number, the ability to control that copy number, and the expression of any other proteins encoded by the vector, such as antibiotic markers, should also be considered. For example, vectors that can be used include those that allow the DNA encoding the IL-2 muteins to be amplified in copy number. Such amplifiable vectors are well known in the art. They include, for example, vectors able to be amplified by DHFR amplification (see, e.g., Kaufman, U.S. Pat. No. 4,470,461, Kaufman and Sharp, “Construction of a Modular Dihydrafolate Reductase cDNA Gene: Analysis of Signals Utilized for Efficient Expression”, Mol. Cell. Biol., 2, pp. 1304-19 (1982)) or glutamine synthetase (“GS”) amplification (see, e.g., U.S. Pat. No. 5,122,464 and European published application 338,841).


In some embodiments, the human IL-2 muteins of the present disclosure will be expressed from vectors, preferably expression vectors. The vectors are useful for autonomous replication in a host cell or may be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome (e.g., nonepisomal mammalian vectors). Expression vectors are capable of directing the expression of coding sequences to which they are operably linked. In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids (vectors). However, other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses, and adeno-associated viruses) are included also.


Exemplary recombinant expression vectors can include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, operably linked to the nucleic acid sequence to be expressed.


The expression constructs or vectors can be designed for expression of an IL-2 mutein or variant thereof in prokaryotic or eukaryotic host cells.


Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. Suitable methods for transforming or transfecting host cells can be found in Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y.) and other standard molecular biology laboratory manuals.


Expression of proteins in prokaryotes is most often carried out in Escherichia coli with vectors containing constitutive or inducible promoters. Strategies to maximize recombinant protein expression in E. coli can be found, for example, in Gottesman (1990) in Gene Expression Technology: Methods in Enzymology 185 (Academic Press, San Diego, Calif), pp. 119-128 and Wada et al. (1992) Nucleic Acids Res. 20:2111-2118. Processes for growing, harvesting, disrupting, or extracting the IL-2 mutein or variant thereof from cells are substantially described in, for example, U.S. Pat. Nos. 4,604,377; 4,738,927; 4,656,132; 4,569,790; 4,748,234; 4,530,787; 4,572,798; 4,748,234; and 4,931,543, herein incorporated by reference in their entireties.


In some embodiments the recombinant IL-2 muteins or biologically active variants thereof can also be made in eukaryotes, such as yeast or human cells. Suitable eukaryotic host cells include insect cells (examples of Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., Sf9 cells) include the pAc series (Smith et al. (1983) Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39)); yeast cells (examples of vectors for expression in yeast S. cerenvisiae include pYepSecl (Baldari et al. (1987) EMBO J. 6:229-234), pMFa (Kurjan and Herskowitz (1982) Cell 30:933-943), pJRY88 (Schultz et al. (1987) Gene 54:113-123), pYES2 (Invitrogen Corporation, San Diego, Calif), and pPicZ (Invitrogen Corporation, San Diego, Calif.)); or mammalian cells (mammalian expression vectors include pCDM8 (Seed (1987) Nature 329:840) and pMT2PC (Kaufman et al. (1987) EMBO J. 6:187:195)). Suitable mammalian cells include Chinese hamster ovary cells (CHO) or COS cells. In mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus, and Simian Virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells, see Chapters 16 and 17 of Sambrook et al. (1989) Molecular Cloning: A Laboratory Manual (2nd ed., Cold Spring Harbor Laboratory Press, Plainview, N.Y.). See, Goeddel (1990) in Gene Expression Technology: Methods in Enzymology 185 (Academic Press, San Diego, Calif).


The sequences encoding the human IL-2 muteins of the present disclosure can be optimized for expression in the host cell of interest. The G-C content of the sequence can be adjusted to levels average for a given cellular host, as calculated by reference to known genes expressed in the host cell. Methods for codon optimization are well known in the art. Codons within the IL-2 mutein coding sequence can be optimized to enhance expression in the host cell, such that about 1%, about 5%, about 10%, about 25%, about 50%, about 75%, or up to 100% of the codons within the coding sequence have been optimized for expression in a particular host cell.


Vectors suitable for use include T7-based vectors for use in bacteria (see, for example, Rosenberg et al., Gene 56:125, 1987), the pMSXND expression vector for use in mammalian cells (Lee and Nathans, J. Biol. Chem. 263:3521, 1988), and baculovirus-derived vectors (for example, the expression vector pBacPAK9 from Clontech, Palo Alto, Calif) for use in insect cells.


In some embodiments nucleic acid inserts, which encode the subject IL-2 muteins in such vectors, can be operably linked to a promoter, which is selected based on, for example, the cell type in which expression is sought.


In selecting an expression control sequence, a variety of factors should also be considered. These include, for example, the relative strength of the sequence, its controllability, and its compatibility with the actual DNA sequence encoding the subject IL-2 mutein, particularly as regards potential secondary structures. Hosts should be selected by consideration of their compatibility with the chosen vector, the toxicity of the product coded for by the DNA sequences of this invention, their secretion characteristics, their ability to fold the polypeptides correctly, their fermentation or culture requirements, and the ease of purification of the products coded for by the DNA sequences.


Within these parameters one of skill in the art may select various vector/expression control sequence/host combinations that will express the desired DNA sequences on fermentation or in large scale animal culture, for example, using CHO cells or COS 7 cells.


The choice of expression control sequence and expression vector, in some embodiments, will depend upon the choice of host. A wide variety of expression host/vector combinations can be employed. Useful expression vectors for eukaryotic hosts, include, for example, vectors with expression control sequences from SV40, bovine papilloma virus, adenovirus and cytomegalovirus. Useful expression vectors for bacterial hosts include known bacterial plasmids, such as plasmids from E. coli, including col E1, pCRI, pER32z, pMB9 and their derivatives, wider host range plasmids, such as RP4, phage DNAs, e.g., the numerous derivatives of phage lambda, e.g., NM989, and other DNA phages, such as M13 and filamentous single stranded DNA phages. Useful expression vectors for yeast cells include the 2μ plasmid and derivatives thereof. Useful vectors for insect cells include pVL 941 and pFastBac™ 1 (GibcoBRL, Gaithersburg, Md.). Cate et al., “Isolation Of The Bovine And Human Genes For Mullerian Inhibiting Substance And Expression Of The Human Gene In Animal Cells”, Cell, 45, pp. 685-98 (1986).


In addition, any of a wide variety of expression control sequences can be used in these vectors. Such useful expression control sequences include the expression control sequences associated with structural genes of the foregoing expression vectors. Examples of useful expression control sequences include, for example, the early and late promoters of SV40 or adenovirus, the lac system, the trp system, the TAC or TRC system, the major operator and promoter regions of phage lambda, for example PL, the control regions of fd coat protein, the promoter for 3-phosphoglycerate kinase or other glycolytic enzymes, the promoters of acid phosphatase, e.g., PhoA, the promoters of the yeast a-mating system, the polyhedron promoter of Baculovirus, and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells or their viruses, and various combinations thereof.


A T7 promoter can be used in bacteria, a polyhedrin promoter can be used in insect cells, and a cytomegalovirus or metallothionein promoter can be used in mammalian cells. Also, in the case of higher eukaryotes, tissue-specific and cell type-specific promoters are widely available. These promoters are so named for their ability to direct expression of a nucleic acid molecule in a given tissue or cell type within the body. Skilled artisans are well aware of numerous promoters and other regulatory elements which can be used to direct expression of nucleic acids.


In addition to sequences that facilitate transcription of the inserted nucleic acid molecule, vectors can contain origins of replication, and other genes that encode a selectable marker. For example, the neomycin-resistance (neor) gene imparts G418 resistance to cells in which it is expressed, and thus permits phenotypic selection of the transfected cells. Those of skill in the art can readily determine whether a given regulatory element or selectable marker is suitable for use in a particular experimental context.


Viral vectors that can be used in the invention include, for example, retroviral, adenoviral, and adeno-associated vectors, herpes virus, simian virus 40 (SV40), and bovine papilloma virus vectors (see, for example, Gluzman (Ed.), Eukaryotic Viral Vectors, CSH Laboratory Press, Cold Spring Harbor, N.Y.).


Prokaryotic or eukaryotic cells that contain and express a nucleic acid molecule that encodes a subject IL-2 mutein disclosed herein are also features of the invention. A cell of the invention is a transfected cell, i.e., a cell into which a nucleic acid molecule, for example a nucleic acid molecule encoding a mutant IL-2 polypeptide, has been introduced by means of recombinant DNA techniques. The progeny of such a cell are also considered within the scope of the invention.


The precise components of the expression system are not critical. For example, an IL-2 mutein can be produced in a prokaryotic host, such as the bacterium E. coli, or in a eukaryotic host, such as an insect cell (e.g., an Sf21 cell), or mammalian cells (e.g., CHO, HEK293, COS cells, NIH 3T3 cells, or HeLa cells). These cells are available from many sources, including the American Type Culture Collection (Manassas, Va.). In selecting an expression system, it matters only that the components are compatible with one another. Artisans or ordinary skill are able to make such a determination. Furthermore, if guidance is required in selecting an expression system, skilled artisans may consult Ausubel et al. (Current Protocols in Molecular Biology, John Wiley and Sons, New York, N.Y., 1993) and Pouwels et al. (Cloning Vectors: A Laboratory Manual, 1985 Suppl. 1987).


The expressed polypeptides can be purified from the expression system using routine biochemical procedures, and can be used, e.g., as therapeutic agents, as described herein.


In some embodiments, IL-2 muteins obtained will be glycosylated or unglycosylated depending on the host organism used to produce the mutein. If bacteria are chosen as the host then the IL-2 mutein produced will be unglycosylated. Eukaryotic cells, on the other hand, will glycosylate the IL-2 muteins, although perhaps not in the same way as native-IL-2 is glycosylated. The IL-2 mutein produced by the transformed host can be purified according to any suitable method. Various methods are known for purifying IL-2. See, e.g. Current Protocols in Protein Science, Vol 2. Eds: John E. Coligan, Ben M. Dunn, Hidde L. Ploehg, David W. Speicher, Paul T. Wingfield, Unit 6.5 (Copyright 1997, John Wiley and Sons, Inc. IL-2 muteins can be isolated from inclusion bodies generated in E. coli, or from conditioned medium from either mammalian or yeast cultures producing a given mutein using cation exchange, gel filtration, and/or reverse phase liquid chromatography.


Another exemplary method of constructing a DNA sequence encoding the IL-2 muteins is by chemical synthesis. This includes direct synthesis of a peptide by chemical means of the protein sequence encoding for an IL-2 mutein exhibiting the properties described. This method can incorporate both natural and unnatural amino acids at positions that affect the interactions of IL-2 with the IL-2Rα, the IL-2Rβ and/or the IL-2Rγ. Alternatively a gene which encodes the desired IL-2 mutein can be synthesized by chemical means using an oligonucleotide synthesizer. Such oligonucleotides are designed based on the amino acid sequence of the desired IL-2 mutein, and preferably selecting those codons that are favored in the host cell in which the recombinant mutein will be produced. In this regard, it is well recognized that the genetic code is degenerate—that an amino acid may be coded for by more than one codon. For example, Phe (F) is coded for by two codons, TIC or TTT, Tyr (Y) is coded for by TAC or TAT and his (H) is coded for by CAC or CAT. Trp (W) is coded for by a single codon, TGG. Accordingly, it will be appreciated that for a given DNA sequence encoding a particular IL-2 mutein, there will be many DNA degenerate sequences that will code for that IL-2 mutein. For example, it will be appreciated that in addition to the preferred DNA sequence for mutein H9, there will be many degenerate DNA sequences that code for the IL-2 mutein shown. These degenerate DNA sequences are considered within the scope of this disclosure. Therefore, “degenerate variants thereof” in the context of this invention means all DNA sequences that code for and thereby enable expression of a particular mutein.


The biological activity of the IL-2 muteins can be assayed by any suitable method known in the art. Such assays include PHA-blast proliferation and NK cell proliferation.


I. Anti-Pd-1 Antibodies and Combinations


Anti-PD-1 antibodies for use according to the invention and methods described herein include but are not limited to nivolumab, BMS-936558, MDX-1106, ONO-4538, AMP224, CT-011, and MK-3475 (pembrolizumab), cemiplimab (REGN2810), SHR-1210 (CTR20160175 and CTR20170090), SHR-1210 (CTR20170299 and CTR20170322), JS-001 (CTR20160274), IBI308 (CTR20160735), BGB-A317 (CTR20160872) and/or a PD-1 antibody as recited in U.S. Patent Publication No. 2017/0081409 (see, e.g., Table 38). There are two approved anti-PD-1 antibodies, pembrolizumab (Keytruda®; MK-3475-033) and nivolumab (Opdivo®; CheckMate078) and many more in development which can be used in combination described herein. Exemplary anti-PD-1 antibody sequences are shown in FIG. 10 and any of these can be used with the combination methods with the IL-2 muteins as described herein.









TABLE 38







PD-1 antibody sequences from U.S. Patent Publication No. 2017/0081409









SEQ ID




NO:

Sequence





286
>US20170081409A1_
SYGMH



1






287
>US20170081409A1_
VIWYDGSNKYYADSVKG



2






288
>US20170081409A1_
NVDY



3






289
>US20170081409A1_
RASQSVSSNLA



4






290
>US20170081409A1_
GASTRAT



5






291
>US20170081409A1_
QQYNNWPRT



6






292
>US20170081409A1_
NGDH



7






293
>US20170081409A1_
GFTFSSYG



8






294
>US20170081409A1_
IWYDGSNK



9






295
>US20170081409A1_
ASNVDY



10






296
>US20170081409A1_
QSVSSN



11






297
>US20170081409A1_
GAS



12






298
>US20170081409A1_
QQYNNWPRT



13






299
>US20170081409A1_
ASNGDH



14






300
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



15
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNVDYWGQGTLVTVSS





301
>US20170081409A1_
EIVMTQSPATLSVSPGERATLSCRASQSVSSNLAWYQQKPGQAPRLLIYG



16
ASTRATGIPARFSGSGSGTEFTLTISSLQSEDFAVYYCQQYNNWPRTFGQ




GTKVEIK





302
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



17
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSS





303
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



18
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNVDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYF




PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCN




VDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISR




TPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVV




SVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPS




QEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS




FFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK





304
>US20170081409A1_
EIVMTQSPATLSVSPGERATLSCRASQSVSSNLAWYQQKPGQAPRLLIYG



19
ASTRATGIPARFSGSGSGTEFTLTISSLQSEDFAVYYCQQYNNWPRTFGQ




GTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV




DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG




LSSPVTKSFNRGEC





305
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



20
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYF




PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCN




VDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISR




TPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVV




SVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPS




QEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS




FFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLGK





306
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



21
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNVDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF




PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICN




VNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTL




MISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNST




YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVY




TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD




SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK





307
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



22
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY




FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC




NVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDT




LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNST




YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVY




TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD




SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK





308
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



23
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY




FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC




NVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDT




LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYAST




YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVY




TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD




SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK





309
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



24
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY




FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC




NVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDT




LMISRTPEVTCVVVDVEHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS




TYRVVSVLTVLHQDWLNGKEYKCKVSNKAFPAPIEKTISKAKGQPREPQV




YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL




DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK





310
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



25
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY




FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC




NVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPDVFLFPPKPKDT




LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNST




YRVVSVLTVLHQDWLNGKEYKCKVSNKALPLPEEKTISKAKGQPREPQVY




TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD




SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK





311
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



26
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ATNGDYWGQGTLVTVSS





312
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



27
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDYWGQGTLVTVSS





313
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



28
VAVIWYDGSNEYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSS





314
>US20170081409A1_
QVQLVESGGGMVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLE



29
WVAVIWFDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYY




CASNGDHWGQGTLVTVSS





315
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



30
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGHGTLVTVSS





316
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



31
VAVIWYDGSNKYYADSVMGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSS





317
>US20170081409A1_
VIWXDGSNXYYADSVXG



32






318
>US20170081409A1_
NXDX



33






319
>US20170081409A1_
VIWYDGSNEYYADSVKG



34






320
>US20170081409A1_
VIWFDGSNKYYADSVKG



35






321
>US20170081409A1_
VIWYDGSNKYYADSVMG



36






322
>US20170081409A1_
NGDY



37






323
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFS



38






324
>US20170081409A1_
QVQLVESGGGMVQPGRSLRLSCAASGFTFS



39






325
>US20170081409A1_
WVRQAPGKGLEWVA



40






326
>US20170081409A1_
RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAS



41






327
>US20170081409A1_
RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAT



42






328
>US20170081409A1_
WGQGTLVTVSS



43






329
>US20170081409A1_
WGHGTLVTVSS



44






330
>US20170081409A1_
EIVMTQSPATLSVSPGERATLSC



45






331
>US20170081409A1_
WYQQKPGQAPRLLIY


T
46






332
>US20170081409A1_
GIPARFSGSGSGTEFTLTISSLQSEDFAVYYC



47






333
>US20170081409A1_
FGQGTKVEIK



48






334
>US20170081409A1_
QVQLVESGGGXVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



49
VAVIWXDGSNXYYADSVXGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




AXNXDXWGXGTLVTVSS





335
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



50
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




AR





336
>US20170081409A1_
EIVMTQSPATLSVSPGERATLSCRASQSVSSNLAWYQQKPGQAPRLLIYG



51
ASTRATGIPARFSGSGSGTEFTLTISSLQSEDFAVYYCQQYNNWP





337
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



52
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNVDYWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYF




PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCN




VDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISR




TPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVV




SVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPS




QEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS




FFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLG





338
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



53
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVKDYF




PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCN




VDHKPSNTKVDKRVESKYGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISR




TPEVTCVVVDVSQEDPEVQFNWYVDGVEVHNAKTKPREEQFNSTYRVV




SVLTVLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPS




QEEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS




FFLYSRLTVDKSRWQEGNVFSCSVMHEALHNHYTQKSLSLSLG





339
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



54
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNVDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF




PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICN




VNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTL




MISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNST




YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVY




TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD




SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG





340
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



55
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY




FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC




NVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDT




LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNST




YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVY




TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD




SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG





341
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



56
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY




FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC




NVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDT




LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYAST




YRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVY




TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD




SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG





342
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



57
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY




FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC




NVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDT




LMISRTPEVTCVVVDVEHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS




TYRVVSVLTVLHQDWLNGKEYKCKVSNKAFPAPIEKTISKAKGQPREPQV




YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL




DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG





343
>US20170081409A1_
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEW



58
VAVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYC




ASNGDHWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDY




FPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC




NVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPDVFLFPPKPKDT




LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNST




YRVVSVLTVLHQDWLNGKEYKCKVSNKALPLPEEKTISKAKGQPREPQVY




TLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLD




SDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG





344
>US20170081409A1_
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGV



59
HTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPK




SCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG




KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTC




LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW




QQGNVFSCSVMHEALHNHYTQKSLSLSPG





345
>US20170081409A1_
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGV



60
HTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPK




SCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG




KEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTC




LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW




QQGNVFSCSVMHEALHNHYTQKSLSLSPGK





346
>US20170081409A1_
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGV



61
HTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPK




SCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK




EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCL




VKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW




QQGNVFSCSVMHEALHNHYTQKSLSLSPG





347
>US20170081409A1_
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGV



62
HTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKRVEPK




SCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK




EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCL




VKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW




QQGNVFSCSVMHEALHNHYTQKSLSLSPGK





348
>US20170081409A1_
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGV



63
HTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESK




YGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPE




VQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEY




KCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVK




GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQE




GNVFSCSVMHEALHNHYTQKSLSLSLG





349
>US20170081409A1_
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGV



64
HTFPAVLQSSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESK




YGPPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPE




VQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEY




KCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLVK




GFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQE




GNVFSCSVMHEALHNHYTQKSLSLSLGK





350
>US20170081409A1_
RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS



65
GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTK




SFNRGEC





351
>US20170081409A1_
EVQLVESGGGLTQPGKSLKLSCEASGFTFSSFTMHWVRQSPGKGLEWV



66
AFIRSGSGIVFYADAVRGRFTISRDNAKNLLFLQMNDLKSEDTAMYYCAR




RPLGHNTFDSWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLV




KDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQT




YICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPK




DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY




NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP




QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP




PVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS




PG





352
>US20170081409A1_
DIVMTQSPSSLAVSPGEKVTMTCKSSQSLYYSGVKENLLAWYQQKPGQS



67
PKLLIYYASIRFTGVPDRFTGSGSGTDYTLTITSVQAEDMGQYFCQQGINN




PLTFGDGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKV




QWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACE




VTHQGLSSPVTKSFNRGEC





353
>US20170081409A1_
EVQLQQSGPELVKPGTSMKISCKASGYSFTGHLMNWVKQSHGKNLEWI



68
GLIIPYNGGTSYNQKFKGKATLTVDKSSSTAYMELLSLTSDDSAVYFCSRGL




RGFYAMDYWGQGTSVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVK




DYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTY




ICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPK




DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQY




NSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP




QVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP




PVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLS




PG





354
>US20170081409A1_
DVVLTQTPLSLSVSFGDQVSISCRSSQSLVNSYGNTFLSWYLHKPGQSPQ



69
LLIFGISNRFSGVPDRFSGSGSGTDFTLKISTIKPEDLGMYYCLQGTHQPPT




FGPGTKLEVKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQ




WKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVT




HQGLSSPVTKSFNRGEC





355
>US20170081409A1_
QVQLQQWGAGLLKPSETLSLTCAVYGGSFSGYYWSWIRQSPEKGLEWI



70
GEINHGGYVTYNPSLESRVTISVDTSKNQFSLKLSSVTAADTAVYYCARDY




GPGNYDWYFDLWGRGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGC




LVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGT




QTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPK




PKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREE




QYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPR




EPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKT




TPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLS




LSPG





356
>US20170081409A1_
EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLIYDA



71
SNRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSNWPPALTFGG




GTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV




DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG




LSSPVTKSFNRGEC





357
>US20170081409A1_
QVQLVQSGSELKKPGASVKVSCKASGYTFTDYSMHWVRQAPGQGLKW



72
MGWINTETGEPTYADDFKGRFVFSLDTSVSTAYLQISSLKAEDTAVYYCA




NPYYDYVSYYAMDYWGQGTTVTVSSASTKGPSVFPLAPSSKSTSGGTAA




LGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSS




LGTQTYICNVNHKPSNTKVDKRVEPKSCDKTHTCPPCPAPELLGGPSVFLF




PPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPR




EEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ




PREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNY




KTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKS




LSLSPG





358
>US20170081409A1_
DIQMTQSPSSLSASVGDRVTITCKASQDVSTAVAWYQQKPGKAPKLLIYS



73
ASYLYTGVPSRFSGSGSGTDFTFTISSLQPEDIATYYCQQHYSTPRTFGQG




TKLEIKRSVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDN




ALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSS




PVTKSFNRGEC





359
>US20170081409A1_
PGWFLDSPDRPWNPPTFSPALLVVTEGDNATFTCSFSNTSESFVLNWYR



74
MSPSNQTDKLAAFPEDRSQPGQDCRFRVTQLPNGRDFHMSVVRARRN




DSGTYLCGAISLAPKAQIKESLRAELRVTERRAEVPTAHPSPSPRPAGQFQ




TLVVGVVGGLLGSLVLLVWVLAVICSRAARGTIGARRTGQPLKEDPSAVP




VFSVDYGELDFQWREKTPEPPVPCVPEQTEYATIVFPSGMGTSSPARRGS




ADGPRSAQPLRPEDGHCSWPL





360
>US20170081409A1_
SLAPKAQIKESLRAEL



75






361
>US20170081409A1_
LDSPDRPWNPPTFSPALL



76






362
>US20170081409A1_
DSPDRPWNPP



77






363
>US20170081409A1_
EVPTAHPSP



78






364
>US20170081409A1_
ISLAPKAQ



79









In some embodiments, the IL-2 mutein portion of the bispecific IL-2 cytokine fusion comprises substitutions L801F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with an anti-PD-1 antibody or inhibitor. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, 186V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with nivolumab. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with pembrolizumab. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with cemiplimab. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination BMS-936558. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination MDX-1106. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination ONO-4538. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination AMP224. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, 186V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination CT-011. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination MK-3475. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


In some embodiments, the IL-2 mutein of the bispecific IL-2 cytokine fusion comprises substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with any of the referenced antibodies. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein is any IL-2 mutein or variant disclosed herein. In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO:10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO:10; C5 SEQ ID NO:11; D10 SEQ ID NO: 12; E10 SEQ ID NO: 13; G8 SEQ ID NO:14; H4 SEQ ID NO:15; and H9 SEQ ID NO: 16. In some embodiments, the IL-2 mutein used in combination with an anti-PD-1 antibody is a fusion mutein as described herein. In some embodiments, the IL-2 mutein used in combination with an anti-PD-1 antibody is a fusion mutein as described herein.


J. Anti-Pd-L1 Antibodies and Combinations


In some embodiments, any of the IL-2 muteins or the bispecific IL-2 cytokine fusions described herein can be used in combination with an anti-PD-1 antibody. There are three approved anti-PD-L1 antibodies, atezolizumab (TECENTRIQ®; MPDL3280A), avelumab (BAVENCIO®; MSB001071 8C), and Durvalumab (MED14736), as well as other anti-PD-L1 antibodies in development. Numerous anti-PD-L1 antibodies are available and many more in development which can be used in combination with the IL-2 muteins as described herein. In some embodiments, the PD-L1 antibody is one described in U.S. Patent Publication No. 2017/0281764 as well as International Patent Publication No. WO 2013/079174 (avelumab) and WO 2010/077634 (or U.S. Patent Application No. 20160222117 or U.S. Pat. No. 8,217,149; atezolizumab). In some embodiments, the PD-L1 antibody comprises a heavy chain sequence of SEQ ID NO:34 and a light chain sequence of SEQ ID NO:36 (from US 2017/281764). In some embodiments, the PD-L1 antibody is atezolizumab (TECENTRIQ®; MPDL3280A; IMpower110). In some embodiments, the PD-L1 antibody is avelumab (BAVENCIO®; MSB001071 8C). In some embodiments, the PD-L1 antibody is durvalumab (MEDI4736). In some embodiments, the PD-L1 antibody includes, for example, Atezolizumab (IMpower133), BMS-936559/MDX-1105, and/or RG-7446/MPDL3280A, and/or YW243.55.S70, as well as any of the exemplary anti-PD-L1 antibodies provided herein in FIG. 17. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, 186V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with any of the referenced antibodies. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein is any IL-2 mutein or variant disclosed herein. In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO:10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO:11; D10 SEQ ID NO: 12; E10 SEQ ID NO:13; G8 SEQ ID NO:14; H4 SEQ ID NO: 15; and H9 SEQ ID NO:16. In some embodiments, the IL-2 mutein used in combination with an anti-PD-L1 antibody is a fusion mutein as described herein. In some embodiments, the IL-2 mutein used in combination with an anti-PD-L1 antibody is a fusion mutein as described herein.


K. Other Immunotherapy Combinations


Other antibodies and/or immunotherapies for use according to the methods of the present invention include but are not limited to, anti-CTLA4 mAbs, such as ipilimumab, tremelimumab; anti-PD-L1 antagonistic antibodies such as BMS-936559/MDX-1105, MEDI4736, RG-7446/MPDL3280A; anti-LAG-3 such as IMP-321; agonistic antibodies targeting immunostimulatory proteins, including anti-CD40 mAbs such as CP-870,893, lucatumumab, dacetuzumab; anti-CD137 mAbs (anti-4-1-BB antibodies) such as BMS-663513 urelumab (anti-4-1BB antibody; see, for example, U.S. Pat. Nos. 7,288,638 and 8,962,804, incorporated by reference herein in their entireties); lirilumab (anti-KIR mAB; IPH2102/BMS-986015; blocks NK cell inhibitory receptors) and PF-05082566 (utomilumab; see, for example, U.S. Pat. Nos. 8,821,867; 8,337,850; and 9,468,678, as well as International Patent Application Publication No. WO 2012/032433, incorporated by reference herein in their entireties); anti-OX40 mAbs (see, for example, WO 2006/029879 or WO 2010/096418, incorporated by reference herein in their entireties); anti-GITR mAbs such as TRX518 (see, for example, U.S. Pat. No. 7,812,135, incorporated by reference herein in its entirety); anti-CD27 mAbs, such as varlilumab CDX-1127 (see, for example, WO 2016/145085 and U.S. Patent Publication Nos. US 2011/0274685 and US 2012/0213771, incorporated by reference herein in their entireties) anti-ICOS mAbs (for example, MEDI-570, JTX-2011, and anti-TIM-3 antibodies (see, for example, WO 2013/006490 or U.S. Patent Publication No US 2016/0257758, incorporated by reference herein in their entireties). In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, 186V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with any of the referenced antibodies. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein is any IL-2 mutein or variant disclosed herein. In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO: 10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO: 5; 5-2 SEQ ID NO: 6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO: 10; C5 SEQ ID NO: 11; D10 SEQ ID NO: 12; E10 SEQ ID NO: 13; G8 SEQ ID NO: 14; H4 SEQ ID NO: 15; and H9 SEQ ID NO: 16.


Other antibodies can also include monoclonal antibodies to prostate cancer, ovarian cancer, breast cancer, endometrial cancer, multiple myeloma, melanoma, lymphomas, lung cancers including small cell lung cancer, kidney cancer, colorectal cancer, pancreatic cancer, gastric cancer, brain cancer (see, generally www.clinicaltrials.gov). In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, 186V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with any of the referenced antibodies. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein is any IL-2 mutein or variant disclosed herein. In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO:10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO: 6; 6-6 SEQ ID NO: 7; A2 SEQ ID NO: 8; B1 SEQ ID NO: 9; B11 SEQ ID NO: 10; C5 SEQ ID NO: 11; D10 SEQ ID NO: 12; E10 SEQ ID NO: 13; G8 SEQ ID NO: 14; H4 SEQ ID NO: 15; and H9 SEQ ID NO: 16.


Antibodies can also include antibodies for antibody-dependent cell-mediated cytotoxicity (ADCC). In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, 186V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with an antibody for antibody-dependent cell-mediated cytotoxicity (ADCC). In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


L. Methods of Treatment


In some embodiments, subject IL-2 muteins and/or bispecific IL-2 cytokine fusions, and/or nucleic acids expressing them, can be administered to a subject to treat a disorder associated with abnormal apoptosis or a differentiative process (e.g., cellular proliferative disorders or cellular differentiative disorders, such as cancer, by, for example, producing an active or passive immunity). In the treatment of such diseases, the disclosed IL-2 muteins may possess advantageous properties, such as reduced vascular leak syndrome. In some embodiments, the IL-2 mutein is any IL-2 mutein or variant disclosed herein. In some embodiments, the IL-2 mutein sequence is 90% identical to any one of SEQ ID NO:2 or SEQ ID NO:6 through SEQ ID NO: 10 or SEQ ID NO:16. In some embodiments, the IL-2 mutein incudes any one of 5-1 SEQ ID NO:5; 5-2 SEQ ID NO:6; 6-6 SEQ ID NO:7; A2 SEQ ID NO:8; B1 SEQ ID NO:9; B11 SEQ ID NO:10; C5 SEQ ID NO: 11; D10 SEQ ID NO: 12; E10 SEQ ID NO: 13; G8 SEQ ID NO: 14; H4 SEQ ID NO:15; and H9 SEQ ID NO:16. In some embodiments, the substitutions in the IL-2 mutein comprise L80F, R81D, L85V, I86V, and I92F, numbered in accordance with wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein is a fusion protein. In some embodiments, the IL-2 mutein is associated with and/or expressed by a CAR-T construct. In some embodiments, the IL-2 mutein is expressed by and/or associated with an oncolytic virus.


Examples of cellular proliferative and/or differentiative disorders include cancer (e.g., carcinoma, sarcoma, metastatic disorders or hematopoietic neoplastic disorders, e.g., leukemias). A metastatic tumor can arise from a multitude of primary tumor types, including but not limited to those of prostate cancer, ovarian cancer, breast cancer, endometrial cancer, multiple myeloma, melanoma, lymphomas, lung cancers including small cell lung cancer, kidney cancer, liver cancer, colon cancer, colorectal cancer, pancreatic cancer, gastric cancer, and brain cancer.


The mutant IL-2 polypeptides can be used to treat patients who have, who are suspected of having, or who may be at high risk for developing any type of cancer, including renal carcinoma or melanoma, or any viral disease. Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary. The term also includes carcinosarcomas, which include malignant tumors composed of carcinomatous and sarcomatous tissues.


Additional examples of proliferative disorders include hematopoietic neoplastic disorders.


Alternatively, or in addition to methods of direct administration to patients, in some embodiments, mutant IL-2 polypeptides can be used in ex vivo methods. For example, cells (e.g., peripheral blood lymphocytes or purified populations of lymphocytes isolated from a patient and placed or maintained in culture) can be cultured in vitro in culture medium and the contacting step can be affected by adding the IL-2 mutant to the culture medium. The culture step can include further steps in which the cells are stimulated or treated with other agents, e.g., to stimulate proliferation, or to expand a population of cells that is reactive to an antigen of interest (e.g., a cancer antigen or a viral antigen). The cells are then administered to the patient after they have been treated.


Anti-PD-1 antibodies for use in combination with the IL-2 muteins disclosed herein for the treatment methods include but are not limited to nivolumab, BMS-936558, MDX-1106, ONO-4538, AMP224, CT-011, and MK-3475.


In some embodiments, the IL-2 mutein of the bispecific IL-2 cytokine fusion comprises substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with an anti-PD-1 antibody or inhibitor for the treatment of cancer. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with nivolumab for the treatment of cancer. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination BMS-936558 for the treatment of cancer. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, 186V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination MDX-1106 for the treatment of cancer. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination ONO-4538 for the treatment of cancer. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination AMP224 for the treatment of cancer. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination CT-011 for the treatment of cancer. In some embodiments, the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination MK-3475 for the treatment of cancer. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises K43N substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


In some embodiments, the IL-2 mutein of the bispecific IL-2 cytokine fusion comprises substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with an antibody and/or immunotherapy including but not limited to, anti-CTLA4 mAbs, such as ipilimumab, tremelimumab; anti-PD-L1 antagonistic antibodies such as BMS-936559/MDX-1105, MEDI4736, RG-7446/MPDL3280A; anti-LAG-3 such as IMP-321; agonistic antibodies targeting immunostimulatory proteins, including anti-CD40 mAbs such as CP-870,893, lucatumumab, dacetuzumab; anti-CD137 mAbs (anti-4-1-BB antibodies) such as BMS-663513 urelumab (anti-4-1BB antibody; see, for example, U.S. Pat. Nos. 7,288,638 and 8,962,804, incorporated by reference herein in their entireties); lirilumab (anti-KIR mAB; IPH2102/BMS-986015; blocks NK cell inhibitory receptors) and PF-05082566 (utomilumab; see, for example, U.S. Pat. Nos. 8,821,867; 8,337,850; and 9,468,678, as well as International Patent Application Publication No. WO 2012/032433, incorporated by reference herein in their entireties); anti-OX40 mAbs (see, for example, WO 2006/029879 or WO 2010/096418, incorporated by reference herein in their entireties); anti-GITR mAbs such as TRX518 (see, for example, U.S. Pat. No. 7,812,135, incorporated by reference herein in its entirety); anti-CD27 mAbs, such as varlilumab CDX-1127 (see, for example, WO 2016/145085 and U.S. Patent Publication Nos. US 2011/0274685 and US 2012/0213771, incorporated by reference herein in their entireties) anti-ICOS mAbs (for example, MEDI-570, JTX-2011, and anti-TIM-3 antibodies (see, for example, WO 2013/006490 or U.S. Patent Publication No US 2016/0257758, incorporated by reference herein in their entireties) for the treatment of cancer.


In some embodiments, the IL-2 mutein of the bispecific IL-2 cytokine fusion comprises substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with another antibody which can include monoclonal antibodies to prostate cancer, ovarian cancer, breast cancer, endometrial cancer, multiple myeloma, melanoma, lymphomas, lung cancers including small cell lung cancer, kidney cancer, colorectal cancer, pancreatic cancer, gastric cancer, brain cancer (see, generally www.clinicaltrials.gov), for the treatment of cancer.


In some embodiments, the IL-2 mutein of the bispecific IL-2 cytokine fusion comprises substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2) is used in combination with antibodies for antibody-dependent cell-mediated cytotoxicity (ADCC) for the treatment of cancer.


M. Pharmaceutical Compositions and Methods of Administration


In some embodiments, subject IL-2 muteins and/or the bispecific IL-2 cytokine fusions and nucleic acids can be incorporated into compositions, including pharmaceutical compositions. Such compositions typically include the polypeptide or nucleic acid molecule and a pharmaceutically acceptable carrier. Such compositions can also comprise anti-PD-1 antibodies. In some embodiments, the composition comprises an IL-2 mutein that is a fusion protein and/or is associated with a CAR-T construct and/or expressed by or associated with an oncolytic virus.


The anti-PD-1 antibodies and IL-2 muteins and/or the bispecific IL-2 cytokine fusions can be administered as a co-composition, simultaneously as two separate compositions, and/or sequentially as two separate compositions. In some embodiments, the anti-PD-1 antibody or inhibitor and IL-2 mutein are administered together as a single co-composition (i.e., co-formulated). In some embodiments, the anti-PD-1 antibody or inhibitor and IL-2 mutein are administered simultaneously as two separate compositions (i.e., separate formulations). In some embodiments, the anti-PD-1 antibody or inhibitor and IL-2 mutein are administered sequentially as separate compositions (i.e., separate formulations). In some embodiments, when the anti-PD-1 antibody or inhibitor and IL-2 mutein are administered sequentially as separate compositions, the anti-PD-1 antibody or inhibitor is administered before the IL-2 mutein. In some embodiments, when the anti-PD-1 antibody or inhibitor and IL-2 mutein are administered sequentially as separate compositions, the IL-2 mutein is administered before the anti-PD-1 antibody or inhibitor. In some embodiments, the anti-PD-1 antibodies include but are not limited to nivolumab, BMS-936558, MDX-1106, ONO-4538, AMP224, CT-011, and MK-3475. In some embodiments, the IL-2 mutein is the IL-2 mutein comprising substitutions L80F, R81D, L85V, I86V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2). In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises K43N substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


The other immunotherapy agents as described and IL-2 muteins and/or the bispecific IL-2 cytokine fusions can be administered as a co-composition, simultaneously as two separate compositions, and/or sequentially as two separate compositions. In some embodiments, the other immunotherapy agents and IL-2 mutein are administered together as a single co-composition (i.e., co-formulated). In some embodiments, the other immunotherapy agents and IL-2 mutein are administered simultaneously as two separate compositions (i.e., separate formulations). In some embodiments, the other immunotherapy agents and IL-2 mutein are administered sequentially as separate compositions (i.e., separate formulations). In some embodiments, when the other immunotherapy agents and IL-2 mutein are administered sequentially as separate compositions, the anti-PD-1 antibody or inhibitor is administered before the IL-2 mutein. In some embodiments, when other immunotherapy agents and IL-2 mutein are administered sequentially as separate compositions, the IL-2 mutein is administered before other immunotherapy agents. In some embodiments, the IL-2 mutein is the IL-2 mutein comprising substitutions L80F, R81D, L85V, 186V, and I92F, numbered in accordance with human wild-type IL-2 (SEQ ID NO:2). In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises K43N substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2. In some embodiments, the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.


A pharmaceutical composition is formulated to be compatible with its intended route of administration. The anti-PD-1 antibodies and/or mutant IL-2 polypeptides (including bispecific IL-2 cytokine fusions) of the invention may be given orally, but it is more likely that they will be administered through a parenteral route, including for example intravenous administration. Examples of parenteral routes of administration include, for example, intravenous, intradermal, subcutaneous, transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as mono- and/or di-basic sodium phosphate, hydrochloric acid or sodium hydroxide (e.g., to a pH of about 7.2-7.8, e.g., 7.5). The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.


Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™. (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all cases, the composition should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants, e.g., sodium dodecyl sulfate. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.


Oral compositions, if used, generally include an inert diluent or an edible carrier. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules, e.g., gelatin capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel™, or corn starch; a lubricant such as magnesium stearate or Sterotes™; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.


In the event of administration by inhalation, anti-PD-1 antibodies and/or IL-2 muteins, or the nucleic acids encoding them, are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer. Such methods include those described in U.S. Pat. No. 6,468,798.


Systemic administration of the anti-PD-1 antibodies and/or IL-2 muteins or nucleic acids can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.


In some embodiments, compounds (anti-PD-1 antibodies and/or mutant IL-2 polypeptides or nucleic acids) can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.


In some embodiments, compounds (subject IL-2 muteins or nucleic acids) can also be administered by transfection or infection using methods known in the art, including but not limited to the methods described in McCaffrey et al. (Nature 418:6893, 2002), Xia et al. (Nature Biotechnol. 20: 1006-1010, 2002), or Putnam (Am. J. Health Syst. Pharm. 53: 151-160, 1996, erratum at Am. J. Health Syst. Pharm. 53:325, 1996).


In one embodiment, the anti-PD-1 antibodies and/or IL-2 muteins and/or bispecific IL-2 cytokine fusions or nucleic acids are prepared with carriers that will protect the anti-PD-1 antibodies and/or mutant IL-2 polypeptides against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Such formulations can be prepared using standard techniques. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.


Dosage, toxicity and therapeutic efficacy of such anti-PD-1 antibodies, IL-2 muteins, or nucleic acids compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit high therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.


The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.


As defined herein, a therapeutically effective amount of a subject IL-2 mutein and/or of the bispecific IL-2 cytokine fusions (i.e., an effective dosage) and/or the anti-PD-1 antibody or inhibitor depends on the polypeptide or antibody selected. In some embodiments, single dose amounts of the IL-2 mutein can be in the range of approximately 0.001 mg/kg to 0.1 mg/kg of patient body weight can be administered. In some embodiments, single dose amounts of the anti-PD-1 antibody or inhibitor can be in the range of approximately 1 mg/kg to 20 mg/kg, or about 5 mg/kg to about 15 mg/kg, or about 10 mg/kg of patient body weight can be administered. In some embodiments, doses of the anti-PD-1 antibody or inhibitor and/or the IL-2 mutein of about 0.005 mg/kg, 0.01 mg/kg, 0.025 mg/kg, 0.05 mg/kg, 0.1 mg/kg, 0.25 mg/kg, 0.5 mg/kg, 1.0 mg/kg, 5.0 mg/kg, 10.0 mg/kg may be administered. In some embodiments, 600,000 IU/kg is administered (IU can be determined by a lymphocyte proliferation bioassay and is expressed in International Units (IU) as established by the World Health Organization 1St International Standard for Interleukin-2 (human)). The dosage may be similar to, but is expected to be less than, that prescribed for PROLEUKIN®. The compositions can be administered one from one or more times per day to one or more times per week; including once every other day. The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of the subject IL-2 muteins can include a single treatment or, can include a series of treatments. In one embodiment, the compositions are administered every 8 hours for five days, followed by a rest period of 2 to 14 days, e.g., 9 days, followed by an additional five days of administration every 8 hours. In some embodiments, administration is 3 doses administered every 4 days.


The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.


The following examples are provided to describe certain embodiments of the invention provided herein and are not to be construed to as limiting.


EXAMPLES
Example 1: Emergence of Novel Long-Acting Mono- and Bi-Specific IL-2/IL-13 Superkines as Potent Immune Modulators

This example provides data showing that combination therapy produces robust responses in a dose-dependent fashion.


BACKGROUND

Use of IL-2 (Proleukin®) remains limited due to a short half-life, toxicity, and preferential activation of Tregs driven by a high affinity for IL2Ra over IL2RP. To bypass these limitations, we further engineered MDNA109, an IL-2 superkine, to generate MDNA11 by (1) addition of mutations to abrogate IL2Ra binding and (2) fusion with albumin to extend half-life. We also leveraged the versatility of our superkine platforms to engineer long-acting bispecific constructs to simultaneously activate IL-2 signaling (i.e. pro-inflammatory) and suppress IL-4/IL-13 function (i.e., anti-inflammatory) and/or enable accumulation in tumors.


Materials and Methods: In vitro and in vivo studies included IL-2 signaling in human PBMCs, BLI/Octet binding analyses, PK studies in mice, and efficacy studies in syngeneic tumor models with or without immune checkpoint inhibitors (ICIs). A dose ranging finding study in cynomolgus monkeys (NHP) was performed to characterize the safety and PK/PD profiles of MDNA11.


Results

MDNA11 and bispecific constructs displayed enhanced STAT5 signaling in human NK and resting CD8 T cells with diminished Treg activity compared to IL-2. In spite of qwk dosing for two weeks, MDNA11 monotherapy or in combination with ICI demonstrated potent efficacy in a CT26 tumor model, resulting in long-term tumor regression and strong memory response. MDNA11 also inhibited growth of B16F10 melanomas with durable increase in tumor infiltrating CD8 T and NK cells. Preliminary studies with bispecific constructs demonstrated therapeutic responses in both CT26 and B16F10 tumor models. In NHP, MDNA11 induced durable proliferation and expansion of NK and CD8 T cells with limited effects on Tregs. Repeat dose of MDNA11 did not trigger cytokine storm, anti-drug antibody response nor eosinophilia (associated with vascular leak syndrome; VLS). Histopathological evaluation confirmed an absence of pulmonary edema, a major IL-2 induced toxicity.


Conclusions

MDNA11 is a long-acting IL-2 superkine with superior potency at activation of naïve CD8 T cells and NK cells, and diminished activity on Tregs. MDNA11 potently inhibited growth of tumors and induced durable regression and strong memory response. Novel bispecific constructs also demonstrated efficacy in these in vivo tumor models. In NHP, MDNA11 induced durable proliferation and expansion of immune effector cells without adverse side effects. These data demonstrate the potency of MDNA11 and underscore the versatility of our superkine platforms to the design of multi-functional therapeutics for immuno-oncological indications.


Example 2: MDNA132-Fc-MDNA109 (KIH) Bi-Specific Superkine
Introduction

MDNA109 is an IL-2 superkine with high selectivity towards the intermediate affinity IL2 receptor (IL2R) consisting of IL-2Rβ(CD122) and IL-2Rγc (CD132) that are expressed on naïve CD8 T-cells and NK cells. MDNA109 is therefore a potent activator of NK and CD8 T cells and can induce a superior anti-cancer response compared to IL-2. However, MDNA109 can still bind to IL-2Rα(CD25) resulting in strong activation of the high affinity IL-2R (IL-2Rαβγc) expressed on immune-suppressive Tregs, therefore impairing the anti-cancer effects of effector T cells. Addition of the FEAA mutations to MDNA109 abolishes binding to CD25 without affecting the binding capacity of MDNA109 to CD122, thereby shifting the balance towards effector immune cells to control or eradicate cancer cells.


MDNA109 and MDNA109FEAA are versatile molecules that can be coupled to protein scaffolds, such as hIgG1 Fc (with a N297A mutation to suppress effector function), to extend in vivo half-life. These Fc fusion constructs can be further fused with additional cytokines or superkines to generate bi-specific molecules with unique properties and potentially novel functions. Medicenna has to date engineered and manufactured 3 unique bi-specific superkines (with 6 more in development) and will evaluate their therapeutic potential in immuno-oncology. Description and rationale for the development of these bi-specifics are provided in Table 14, and in following sections. The sequences of these bi-specific superkines are listed in Table 15, as well as FIG. 54.


MDNA132-Fc-MDNA109 (KIH) Bi-Specific Superkine:

IL13Rα2 is a decoy receptor for the IL-13 cytokine, and while not being bound by theory, is believed to act by dampening IL13-mediated responses in inflamed tissues. It is minimally expressed in normal tissues, but is overexpressed in certain tumors, including glioblastoma, colorectal, pancreatic and basal-like breast tumors. Therefore, IL13Rα2 can be considered as a potential immunotherapeutic target for these cancer types and can be exploited as a means to increase localization of cytokines, such as MDNA109, to tumors sites in order to enhance their therapeutic activities.


MDNA132 is an IL-13 superkine engineered to selectively bind IL13Rα2 with a 10-fold increase in affinity compared to IL-13. The bi-specific superkine MDNA132-Fc-MDNA109 (see Table 12) was designed with the objective of exploiting MDNA132 for localizing to tumors overexpressing IL13Rα2 in order to enhance the therapeutic effect of MDNA109 by increasing bioavailability at the target sites. Variant constructs containing MDNA109 engineered with the FEAA mutations (e.g., MDNA132-Fc-MDNA109FEAA) were manufactured for testing.


MDNA413-Fc-MDNA109 (KIH) and MDNA109FEAA-Fc-MDNA413 (2:1:2) Bi-Specific Superkines: Interleukin 4 receptor (IL4R) is an important component of the immune system. It is heavily involved in T-cell differentiation, antibody isotype switching and other immune reactions.


Interestingly, the IL4R can be either classified as Type I or Type II receptor. In the context of type I receptor, the IL4Rα-chain binds to the γc chain (normally expressed on immune cells). In contrast, the type II receptor consists of the IL4Ra chain binding to IL13Rα1. Type II IL4R is particularly relevant to cancer immunotherapy as it is highly prevalent on various tumor types. For instance, it is overexpressed in more than 50% of pancreatic and ovarian tumors, more than 70% of colorectal, breast, head and neck, and lung cancer (i.e. NSCLC). In addition to solid tumors, myeloid derived suppressor cells (MDSCs) and tumor associated macrophage (TAMs), which constitute important components of the tumor microenvironment (TME), are positive for the type II IL4R. In this context, the IL4/IL4R axis induces cancer-promoting phenotypes in TAMs and boost MDSCs among other effects including promotion of proliferation, inhibition of apoptosis, and enhances tumor metabolism. Therefore, inhibition of this pathway could dramatically alter the TME and limit tumor growth.


MDNA413 is a dual IL4/IL-13 super-antagonist designed to efficiently inhibit IL-4 and IL-13 induced activities by selectively blocking their binding to IL4R Type-II. MDNA413 could therefore inhibit MDSC function and minimize the skewing of TAM toward the immune-suppressive M2 phenotype, thereby shifting the balance in favor of anti-cancer responses. The fusion of MDNA413 to MDNA109-Fc or MDNA109FEAA-Fc (Table 15 as well as FIG. 54) have the combined benefits of diminishing the tumor-promoting functions of MDSC and M2/TAMs while stimulating the anti-cancer activity of effector immune cells (i.e. CD8 T- and NK cells).









TABLE 14







List of bi-specific constructs manufactured and rationale for their production









Bi-specific




Construct
Description
Activities





KIH MDNA132-Fc-
Fc heterodimer in knob-
MDNA109 activates immune effector cells


MDNA109
in-hole (KIH) format
and MDNA132 targets the IL13a2 receptor


(1:1:1:)
containing 1
expressed on solid tumors; MDNA109



MDNA132 at N-
fusion to MDNA132 will help localizing



terminus, 1 Fc and 1
MDNA109 to tumor cells; fusion to hIgG1



MDNA109. Fusion to
Fc* would extend the in vivo half-life of the



Fc via a (G4S)3 linker.
construct.


KIH MDNA413-Fc-
Fc heterodimer in knob-
MDNA109 activates immune effector cells


MDNA109
in-hole (KIH) format
while MDNA413 targets the IL-4 type II


(1:1:1)
containing 1
receptor; fusion to MDNA413 may allow



MDNA413 at 7
MDNA109 localization to the tumor



terminus, 1 Fc and 1
microenvironment (TME) and inhibition of



MDNA109. Fusion to
the immunosuppressive and tumor-



Fc via a (G4S)3 linker.
promoting cells (M2 polarized tumor




associated macrophages, MDSCs and Treg




cells); fusion with hIgG1 Fc* extends the in




vivo half-life of the compound (Fc


MDNA109FEAA-
Fc homodimer
F42A and E62A mutations in


Fc-MDNA413 (2:1:2)
containing 2
MDNA109FEAA decrease affinity for



MDNAFEAA at N-
CD25, therefore limiting stimulation of



terminus, 1 Fc and 2
Tregs, while retain binding to CD122 for



MDNA413. Fusion to
potent activation of effector immune cells;



Fc via a (G4S)3
MDNA413 allows for localization to TME




and inhibition of tumor-promoting cells;




fusion with hIgG1 Fc* to extend in vivo half-




life of construct; a 2:1:2 stoichiometry may




lead to enhanced pharmacology.





*Fc contains N297A mutation to suppress effector function













TABLE 15







Sequence of bi-specific constructs








Bi-Specific



Construct
Sequence





KIH MDNA132-Fc-
MDNA132-FC-MDNA109 (1:1:1 KIH)


MDNA109
Gene 1:


(1:1:1:)
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLINVSGCSAI


Fc heteroduplex
EKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLHLKKLFKEGQFNGGGGSGG


with MW of 80.8
GGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPE


KDa
VKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPI



EKTISKAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYK



TTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGSGG



GGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYM



PKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCE



YADETATIVEFLNRWITFCQSIISTLT (SEQ ID NO: 226)



Gene 2:



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVD



GVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK



GQPREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS



DGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:



227)





MDNA413-Fc-
MDNA413-Fc-MDNA109 (1:1:1 KIH)


MDNA109 (KIH)
Gene1:


Fc heteroduplex
PGPVPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLINVSGCSAI


with MW of 80.8
EKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFHLRTLFREGQFNGGGGSGG


KDa
GGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPE



VKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPI



EKTISKAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDIAVEWESNGQPENNYK



TTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGSGG



GGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTFKFYM



PKKATELKHLQCLEEELKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCE



YADETATIVEFLNRWITFCQSIISTLT (SEQ ID NO: 228)



Gene2:



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVD



GVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK



GQPREPQVCTLPPSREEMTKNQVSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS



DGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO:



229)





MDNA109FEAA-
MDNA109FEAA-Fc-MDNA413 (2:1:2)


Fc-MDNA413
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKKATELKHLQCLE


Fc homodimer with
EALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNR


MW of 109.8 KDa
WITFCQSIISTLTGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM



ISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQD



WLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGF



YPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL



HNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPVPPSTAVRALIEELINITQNQKAPLCNG



SMVWSINRTAGMYCAALESLINVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIE



VAQFVKDLLFHLRTLFREGQFN (SEQ ID NO: 230)









Manufacturing and Purification of Bi-Specific Superkines

Bi-specific superkines were manufactured in HEK293 cells. MDNA132-Fc-MDNA109 (KIH) was purified by affinity chromatography (Protein A followed by cation exchange), and was analyzed by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis (PAGE) and high-performance liquid chromatography HPLC (FIG. 4). MDNA413-Fc-MDNA109 (KIH) and MDNA413FEAA-Fc-MDNA109 (2:1:2) were purified by affinity chromatography and size exclusion chromatography (SEC), and were analyzed by SDS-PAGE and HPLC (FIGS. 5 and 6 respectively). Endotoxin levels were measured by the LAL chromogenic endotoxin assay, and are reported in Table 16 with information on purity and buffer.









TABLE 16







Information on available bi-specific superkines.










Bi-specific

Purity



Construct
Buffer
(% monomer)
Endotoxin





MDNA132-Fc-
20 mM Histidine,
 >95%
 0.42 EU/mg


MDNA109 (KIH)
5% sorbitol, ph 6




MDNA413-Fc-
20 mM Histidine,
  100%
 0.07 EU/mg


MDNA109 (KIH)
5% sorbitol, ph 6




MDNA109FEAA-
20 mM Histidine,
 >89%
0.134 EU/mg


Fc-MDNA413
5% sorbitol, ph 6




(2:1:2)









Signaling in Human PBMC

In vitro signaling assays based on STAT5 phosphorylation (pSTAT5) provide important functional readouts and were used to evaluate the potency of the MDNA109 superkine at activating different subsets of immune cells expressing different forms of the IL-2 receptor (IL-2R) (Levin et al., 2012). Dose-response experiments were performed in PBMCs derived from healthy human donors to measure the potencies of bi-specific superkines in different subsets of immune cells within the same experimental condition. These studies were used to provide insights into the pharmacological profiles of bi-specific superkines and to determine whether fusions with MDNA413 and MDNA132 affect the activity of MDNA109 and MDNA109FEAA. A major strength with the use of human primary PBMCs is the ability to perform tests on multiple donors to understand the potential effect of genetic heterogeneity on the immune responses triggered by test constructs.


PBMCs were isolated over a density gradient, allowed to rest in complete media, and then stimulated for 15 minutes with 10-point 5-fold dilutions of IL-2, different MDNA109 variants, or bi-specific superkines. Controls included non-stimulated PBMC cells (i.e. no addition of MDNA109 variants or IL-2). Cells were fixed immediately following stimulation, and samples were analyzed by flow cytometry after intracellular staining for phosphorylated STAT5 (pSTAT5) in the immune subsets as listed in Table 17. The gating strategy employed to enable pSTAT5 analysis in different immune subsets is depicted in FIG. 7 fluorescence minus one (FMO) controls are shown in FIG. 8.









TABLE 17







Flow cytometry markers for different immune subsets










Cell type
Markers







Treg cells
CD3+CD4+CD25+FOXP3+



CD8 T cells (naive)
CD3+CD8+ CD25



CD8 T cells (activated)
CD3+CD8+ CD25+



NK cells
CD3CD56+










Representative dose-response curves for each immune subset following stimulation with the indicated test construct are shown in FIG. 9, and a summary of EC50 values from analyses of different PBMC donors are indicated in the chart under the plots. In comparison to IL-2 or IL2-Fc, MDNA109-Fc and MDNA109FEAA-Fc were clearly more potent at activation of naïve (CD25−) CD8 T-cells, as indicated by lower EC50 values. EC50 of MDNA109-Fc (152.2±35.8 pM) and MDNA109FEAA-Fc (370.6±131.1 pM) are 14- and 6-folds lower than IL2-Fc (2189.2±435.3 pM) respectively. With respect to NK cells, MDNA109-Fc and MDNA109FEAA-Fc are both more potent than IL2, but the comparison to IL2-Fc is less clear with MDNA109-Fc and MDNA109FEAA-Fc showing superior or similar potencies respectively.


Both IL2-Fc and MDNA109-Fc showed similar potency toward the Treg population with EC50 of 0.85±0.4 and 0.9±0.42 μM respectively. In contrast, MDNA109FEAA-Fc demonstrated a dramatic decrease in Treg activation, with an EC50 of 135.5±53.9 pM or an ˜150-fold reduction in comparison to IL2-Fc and MDNA109-Fc.


The data clearly showed that the bi-specific superkines retain their capacity to effectively stimulate naïve CD8 T-cells compared to IL-2 or IL2-Fc. In comparison to IL2-Fc, MDNA132-Fc-MDNA109 (EC50=258±58.8 pM) is ˜8-fold more potent, MDNA413-Fc-MDNA109 (EC50=378.4±123.1 pM) is ˜6-fold more potent, and MDNA109FEAA-Fc-MDNA413 (EC50=575.8 119 pM) is ˜4-fold more potent. In addition, all three bi-specific superkines are more potent than IL-2 at stimulating NK cells.


While MDNA109FEAA-Fc-MDNA413 is less potent than the other bi-specific superkines at activation of CD8 T-cells, it remains more effective than IL2-Fc and has the unique property of exhibiting a reduced capacity to stimulate Tregs compared to all other variants. In comparison to IL2-Fc (EC50=0.85±0.4 pM), MDNA109FEAA-Fc-MDNA413 (EC50=210.3±58), which is almost 250-fold weaker at stimulating Tregs.


A useful metric to predict the potential therapeutic activity of these constructs is the ratio of pSTAT5 EC50 of naïve CD8 T-cells and NK cells to Tregs. As shown in FIG. 10, the CD8 T-cells/Tregs EC50 ratio for IL-2 and IL-2-Fc are higher (694 and 3008 respectively), consistent with the fact that these constructs are better activators of Tregs than they are of naïve CD8 T-cells. This ratio is significantly reduced for MDNA109-Fc as well as MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109, consistent with their increased potencies towards naïve CD8 T-cells. The most significant changes are obtained with MDNA109FEAA-Fc and MDNA109FEAA-Fc-MDNA413, for which the CD8 T-cells/Tregs EC50 ratio dropped dramatically to low single digits. Ratios of EC50 of NK cells to Tregs show a similar trend, with MDNA109-Fc, MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 displaying lower ratios than IL-2 and IL2-Fc. MDNA109FEAA-Fc and MDNA109FEAA-Fc-MDNA413 again have the lowest NK cells/Treg EC50 ratios. These results reflect the fact that these constructs not only have increased potencies towards NK and naïve CD8 T-cells, but also have diminished potencies towards Tregs. A combination of increased pro-immune CD8 T-cells and NK cells and reduced immune-suppressive Tregs is anticipated to drive potent anti-cancer immune responses.


Conclusions:

Bi-specific superkines harboring an MDNA109 or MDNA109FEAA core component maintain enhanced potency over IL2-Fc at activation of CD8 T-cells. While MDNA132-Fc-MDNA109 (KIH) and MDNA413-Fc-MDNA109 (KIH) have similar potency to IL2-Fc towards Tregs activation, MDNA109FEAA-FC-MDNA413 (2:1:2) display a reduction of almost 250-fold at stimulating the Treg population.


MTD (Maximum Tolerated Dose) and Dosing Schedule Studies

MTD is defined as the highest dose of a drug that does not cause significant side effects or overt toxicity. MTD studies were performed in two mouse strains, BALB/c and C57BL/6, which were subsequently used to evaluate anti-cancer efficacy of bi-specific superkines. The use of these two strains enabled efficacy studies using various syngeneic cancer models that are either compatible with BALB/c or C57BL/6 mice. These data were also important for the design of combination studies where the administration of bi-specific superkines were aligned with that of the companion drug for optimal efficacy as well as practicality. The dosing schedule for immune checkpoint inhibitors (ICIs), as an example, in mice and in the clinic is well documented.


Bi-specific superkines were administered by intraperitoneal (IP) injection, which is also a common route for delivery of ICIs in mice. There were 5 mice in each treatment group depending on the study. In these studies, MTD was strictly defined as any incidence of spontaneous mortality or mandatory termination due to weight loss or moribund during the study period. To date, MTD studies have been conducted for MDNA132-Fc-MDNA109 (KIH) and MDNA413-Fc-MDNA109 (KIH).


MTD Study in BALB/c Mice:

A description of the design of the MTD study conducted in BALB/c mice is shown in Table 18. Studies were performed with MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109; MDNA109FEAA-Fc-MDNA413 was not available when these experiments were conducted.









TABLE 18







MTD study in BALB/c strain of mice










Bi-specific Superkine
# of Mice
Dose
Regimen





MDNA132-Fc-
5
1.0 mg/kg
2x/week for 2 weeks


MDNA109
5
2.5 mg/kg
2x/week for 2 weeks


MDNA413-Fc-
5
1.0 mg/kg
2x/week for 2 weeks


MDNA109
5
2.5 mg/kg
2x/week for 2 weeks









As shown in FIG. 11, BALB/c mice treated with either 1 mg/kg or 2.5 mg/kg of MDNA132-Fc-MDNA109 or MDNA413-Fc-MDNA109 twice weekly for two weeks were all viable at the end of the study. Mice in both groups appeared clinically normal and maintained their weights during the study period. In summary, MTD was not reached at the highest dose tested (2.5 mg/kg) on a twice weekly dosing schedule for two weeks for both MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 in BALB/c mice.


MTD Study in C57Bl/6 Mice:

Table 19 provides an outline of the design of MTD study performed in C57Bl/6 mice. A lower range of dose was used because C57Bl/6 has been found to be more sensitive to IL-2 than BALB/c mice (Chen et al., 2005).









TABLE 19







MTD Study in C57Bl/6 strain of mice










Bi-specific Superkine
# of Mice
Dose
Regimen





MDNA132-Fc-
5
0.2 mg/kg
2x/week for 2 weeks


MDNA109
5
0.5 mg/kg
2x/week for 2 weeks



5
1.0 mg/kg
2x/week for 2 weeks


MDNA413-Fc-
5
0.2 mg/kg
2x/week for 2 weeks


MDNA109
5
0.5 mg/kg
2x/week for 2 weeks



5
1.0 mg/kg
2x/week for 2 weeks









As shown in FIG. 12, C57Bl/6 mice treated with MDNA132-Fc-MDNA109 or MDNA413-Fc-MDNA109 twice weekly for two weeks at 0.2 mg/kg, 0.5 mg/kg and 1.0 mg/kg were all viable at the end of the study. Mice in both groups appeared clinically normal and maintained their weights during the study period. In summary, MTD was not reached at the highest dose tested (1.0 mg/kg) on a twice weekly dosing schedule for two weeks for both MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 in C57Bl/6 mice.



FIG. 12: Survival C57Bl/6 mice receiving different doses of MDNA132-Fc-MDNA109 (left) and MDNA413-Fc-MDNA109 (right).


Conclusions:

MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 were well tolerated by BALB/c mice when administered twice weekly for two weeks at 1.0 mg/kg and 2.5 mg/kg, indicating that MTD was not reached at the highest dose (2.5 mg/kg) tested.


MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 were well tolerated by C57B/6 mice when administered twice weekly for two weeks at 0.2 mg/kg, 0.5 mg/kg and 1.0 mg/kg, indicating that MTD was not reached at the highest dose (1.0 mg/kg) tested.


In Vivo Efficacy Studies

Translatability of strong in vitro functional data to clinically relevant anti-cancer phenotypes was tested using in vivo tumor models. These in vivo efficacy studies were performed using bi-specific superkines as single agents or in combination with ICI. Since MTD was not reached, higher doses than those tested were used, albeit at a less frequent dosing schedule. In addition, preliminary experiments were also performed in which bi-specific superkines were delivered directly into the tumors to assess the effects of localized administration on efficacy.


Efficacy Studies in CT26 Colon Cancer Model:

The CT26 syngeneic colon cancer model was selected to evaluate the therapeutic potential of bi-specific superkines for a number of reasons:

    • The CT26 cancer model is less aggressive compared to many other cancer models (e.g. B16F10 melanoma model, 4T1 metastatic breast cancer model or the Panc2 pancreatic cancer model), therefore providing a larger window to follow the therapeutic effects of MDNA109 variants.
    • CT26 cancer model is responsive to ICIs in contrast to several other models, therefore providing an opportunity to evaluate combinatory treatments to determine potential synergy.
    • The relatively slow growth characteristic of CT26 tumors provides an opportunity to evaluate the anti-tumor activity of MDNA109 variants in the context of early stage (i.e. small) versus late stage (i.e. large) tumors.


The efficacy study that has been conducted in the CT26 model with MDNA132-Fc-MDNA109 (KIH) has the following study design:

    • Female (8-10 weeks old) BALB/c mice were implanted with 2×106 CT26 cells subcutaneously in the right flank, and tumors were allowed to grow until average tumor size reached 60 mm3.
    • Mice were randomized into groups of either 6 (vehicle control) or 8 (MDNA132-Fc-MDNA109) mice each based on tumor size.
    • Mice were dosed by IP with either vehicle (PBS) or MDNA132-Fc-MDNA109 at 5 mg/kg once weekly for 2 weeks
    • Study measurements to include:
      • Daily clinical cage-side observations.
      • Twice weekly body weights and tumor measurements.
      • Monitor food and water consumption.
    • Animals to be prematurely terminated if any of the following criteria are met:
      • Weight loss exceeding 20% of the maximum weight for that animal.
      • Tumor volume exceeding 2000 mm3.
      • Animal appearing moribund.


As shown in FIG. 13 (a), treatment with MDNA132-Fc-MDNA109 (KIH) resulted in potent inhibition of CT26 tumors growth. In fact, 3 of 8 mice treated with MDNA132-Fc-MDNA109 (KIH) resulted in complete tumor regression whereas this was not observed with control mice (FIG. 13 (b)). These three mice have not shown any sign of tumor relapse for more than 120 days post-implant or more than 3 months since treatment was stopped, indicating that they have been cured of cancer.


To validate whether mice that were cured of their primary CT26 tumors following 2 doses of MDNA132-Fc-MDNA109 are protected against subsequent re-challenges, the three mice were implanted with CT26 on their opposite flank on Day 49 of the study and were not given any further treatment. As controls, naïve untreated mice were also implanted with CT26 tumor cells. As shown in FIG. 14(a), naïve BALB/c mice showed robust CT26 tumor growth. In contrast, the mice treated with MDNA132-Fc-MDNA109 and cured of their primary tumors did not show any sign of tumor growth at the re-challenge site, suggesting that they have developed a strong memory response against CT26 tumor cells. These mice have undergone a second re-challenge and are continued to be monitored. MDNA132-Fc-MDNA109 therefore provides these mice with overall survival benefits in spite of multiple re-challenges (FIG. 14(b)).


Efficacy Studies in B16F10 Melanoma Model:

The B16F10 syngeneic melanoma model in C57Bl/6 mice is an aggressive in vivo tumor model that has withstood many therapeutic efforts due in part to the speed at which these tumors grow and metastasize in mice. In comparison to the CT26 tumor model, this is a significantly more difficult model to treat and therefore provides an important challenge to evaluate the potential therapeutic activity of bi-specific superkines. Two efficacy studies were separately conducted to evaluate the efficacy of MDNA413-Fc-MDNA109 (KIH) and MDNA109FEAA-Fc-MDNA109. Both studies followed a similar protocol as outlined below:

    • Female (8-10 weeks old) C57Bl/6 mice were implanted with 3×105 C57Bl/6 cells subcutaneously in the right flank, and tumors were allowed until tumor growth has been established before initiation of dosing.
    • Mice were randomized into treatment groups based on tumor size such that average tumor size of each group are similar.
    • Mice were dosed by IP at dose and schedule specific to each study (see below)
    • Study measurements to include:
      • Daily clinical cage-side observations
      • Twice weekly body weights and tumor measurements
      • Monitor food and water consumption.
    • Animals to be prematurely terminated if any of the following criteria are met:
      • Weight loss exceeding 20% of the maximum weight for that animal
      • Tumor volume exceeding 2000 mm3
      • Animal appearing moribund


        B16F10 Efficacy Study with MDNA413-Fc-MDNA109 (KIH):


Average tumor size was ˜20 mm3 at the initiation of dosing. Mice were IP injected with PBS (n=9) or MDNA413-Fc-MDNA109 at 1 mg/kg (n=7) or 2.5 mg/kg (n=9) twice weekly for 2 weeks. As shown in FIG. 15, treatment with MDNA413-Fc-MDNA109 resulted in inhibition of B16F10 tumor growth in a dose-dependent manner. MDNA413-Fc-MDNA109 given at 2.5 mg/kg was more potent at growth inhibition than the 1 mg/kg dose.


B16F10 Efficacy with MDNA109FEAA-Fc-MDNA413.


In this study, tumors were grown to an average size of ˜15 mm3 at the start of dosing. Mice were dose with PBS (n=8) or MDNA109FEAA-Fc-MDNA413 (n=8) once weekly for 3 weeks at 5 mg/kg. As shown in FIG. 16A-16B, MDNA109FEAA-Fc-MDNA413 had no effect on the growth of B16F10 melanomas with tumors growing similarly as with the control mice. These data suggest that a more frequent dosing schedule (twice weekly) may be necessary to achieve a therapeutic response with MDNA109FEAA-Fc-MDNA413.


Efficacy Studies in the 4T1 Breast Cancer Model:

The 4T1 breast cancer model is also an aggressive model with a high incidence of tumor metastasis. This model has been used to test the therapeutic activity of drugs aim at modulating myeloid-derived suppressor cells (MDSC) and M1 to M2 skewing of tumor associated fibroblasts, both of which work to support tumor growth.


To date, one 4T1 efficacy study has been conducted to evaluate the therapeutic potential of MDNA132-Fc-MDNA109 (KIH) and MDNA413-Fc-MDNA109 (KIH) as monotherapies (i.e. with isotype control; BioXcel BE0089) or in combination with anti-PD1 (Clone RMP1-4; BioXcell BE0146). The study design was as follow:

    • 8-weeks old female BALB/c mice were implanted with 2×106 4T1 cells subcutaneously, and tumors were allowed to grow for 14 days prior to initiation of dosing.
    • Animals were randomized into groups of 8 mice each based on tumor size.
    • Treatment were as outlined in Table 20.
    • Study measurements to include:
      • Daily clinical cage-side observations
      • Twice weekly body weights and tumor measurements
      • Monitor food and water consumption.
    • Animals to be prematurely terminated if any of the following criteria are met:
      • Weight loss exceeding 20% of the maximum weight for that animal
      • Tumor volume exceeding 2000 mm3
      • Animal appearing moribund









TABLE 20







Design of 4T1 efficacy study.














# of




Dosing


Group
mice
Compound
Dose
Route
Frequency
Days





1
8
Vehicle
N/A
IP
Twice
4







weekly for








2 weeks

















Isotype
200
μg
IP
Twice
4








weekly for









2 weeks



2
8
PD1 Mab
200
μg
IP
Twice
4








weekly for









2 weeks



3
8
MDNA132-Fc-
5
mg/kg
IP
Once
2




MDNA109



weekly for





(KIH) C



2 weeks





Isotype
200
μg
IP
Twice
4








weekly for









2 weeks



4
8
MDNA132-Fc-
5
mg/kg
IP
Once
2




MDNA109



weekly for





(KIH) C



2 weeks





PD1 Mab
200
μg
IP
Twice
4








weekly for









2 weeks



5
8
MDNA413-Fc-
5
mg/kg
IP
Once
2




MDNA109



weekly for





(KIH)



2 weeks





Isotype
200
μg
IP
Twice
4








weekly for









2 weeks



6
8
MDNA413-Fc-
5
mg/kg
IP
Once
2




MDNA109



weekly for





(KIH)



2 weeks





PD1 Mab
200
μg
IP
Twice
4


















weekly for








2 weeks









As shown in FIG. 17, mice in the MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 monotherapy and combination groups were found dead following the second dose, indicating adverse toxicity at the 5 mg/kg once weekly doing regimen in this model. In a previous study in the CT26 model (see above), also in BALB/c mice, treatment with MDNA132-Fc-MDNA109 on the same regimen did not result in toxicity, suggesting that this may be related to the 4T1 tumor model itself.


Preliminary Efficacy Studies with Intra-Tumoral Delivery of Bi-specific Superkines:


Preliminary studies were conducted in a limited number of mice with established CT26 or 1B6F10 tumors to determine whether intratumoral delivery of bi-specific superkies into the tumors could have a strong therapeutic effect.


B16F10 Intra-Tumoral Treatment with MDNA132-Fc-MDNA109:


Established B16F10 tumors were intratumorally injected with PBS (n=2) or with 10 μg of MDNA132-Fc-MDNA109 (KIH) for 3 doses 3 days apart (n=2). While sample size was very small, the data suggested that intra-tumoral injection with MDNA132-Fc-MDNA109 delayed the growth of B16F10 melanomas (FIG. 18).


CT26 Intra-Tumoral Treatment with MDNA109FEAA-Fc-MDNA413:


Mice with established CT26 tumors (n=5) were injected intra-tumorally with MDNA109FEAA-Fc-MDNA413 at 10 μg per dose for 3 days apart for a total of 3 doses. As shown in FIG. 19, tumors injected with MDNA109FEAA-Fc-MDNA413 continued to grow steadily during the study. However, it is not clear whether treatment did have an effect on the rate of growth because PBS controls were not available.


SUMMARY

This report provides a summary of information available to date on bi-specific superkines (i.e. MDNA132-Fc-MDNA109 (KIH), MDNA413-Fc-MDNA109 (KIH), and MDNA109FEAA-Fc-MDNA109 (2:1:2)), including their manufacturing and in vitro and in vivo studies that have been performed to evaluate their biological activities.


Studies in human PBMC showed that MDNA132-Fc-MDNA109, MDNA413-Fc-MDNA109 and MDNA109FEAA-Fc-MDNA413 are more potent than IL-2 and IL2-Fc at activation of naïve CD8 T-cells, indicating that the activity of the core MDNA109 or MDNA109FEAA component of these bi-specific superkines are preserved. Whereas MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 displayed similar activity on the Treg population as IL-2 and IL2-Fc, MDNA109FEAA-Fc-MDNA413 showed a dramatic reduction in ability to stimulate these immune-suppressive cells, as anticipated due to the FEAA mutations. These data indicate that these bi-specific superkines, and particularly MDNA109FEAA-Fc-MDNA143, have a more favorable anti-cancer immunological profile than IL-2 and IL2-Fc.


MTD studies in BALB/c and C57Bl/6 mice showed that MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 were well tolerated at the highest dose tested (2.5 mg/kg in BALB/c and 1.0 mg/kg in C57Bl/6) when administered on a twice weekly schedule for 2 weeks. In both cases, MTD were not reached indicating that these bi-specific superkines can be safely administered at even higher doses.


MDNA132-Fc-MDNA109 (KIH) was tested in a CT26 colon tumor model, in which the treatment schedule was reduced to once weekly for 2 weeks, while the dose was increased to 5 mg/kg. This doing regimen was well tolerated. Data showed that MDNA132-Fc-MDNA109 (KIH) monotherapy potently inhibited the growth and CT26 tumors, and in fact induced complete and durable tumor regression in 3 of 8 mice. These mice are still viable and remaining tumor free for more than 3 months since treatment was stopped. Importantly, these mice have undergone a re-challenge with CT26 tumor cells and were found to be resistance against tumor growth, suggesting that they have developed a strong memory response against CT26. An additional re-challenge has been initiated with these mice.


MDNA413-Fc-MDNA109 (KIH) inhibited the growth the B16F10 melanomas when administered twice weekly for two weeks. The inhibitory effect was dose-dependent with mice receiving a dose of 2.5 mg/kg displaying slower tumor growth than mice receiving a dose of 1 mg/kg.


MDNA109FEAA-Fc-MDNA109 had little to no effect on the growth of B16F10 melanomas when administered at 5 mg/kg once weekly for 3 weeks. It is possible that a higher dose or more frequent dosing schedule is needed to achieve a therapeutic effect.


While MDNA132-Fc-MDNA109 was well tolerated in the CT26 tumor model in BALB/c mice (see above), it was not well tolerated well in the 4T1 model, also in BALB/c mice, under the exact dosing regimen of 5 mg/kg once weekly for two weeks. Mice died soon after receiving the second dose of MDNA132-Fc-MDNA109 irrespective of whether treatment was monotherapy or in combination with anti-PD1. Similarly, 4T1 tumor bearing mice treated once weekly with MDNA413-Fc-MDNA109 at 5 mg/kg also died soon after receiving the second dose. These data suggest the lethality associated with these bi-specific superkines may be related to the 4T1 model itself rather than the strain of mice used in the experiment.


Preliminary studies with intra-tumoral administration of bi-specific superkines were not conclusive due to a lack of controls and/or small sample size. CT26 tumors injected with MDNA109FEAA-Fc-MDNA413 continued to grow steadily during the study, however it was not clear whether there was an effect on growth rate since control tumors injected with vehicle were not available for comparison. MDNA132-Fc-MDNA109 did appear to delay the growth of B16F10 tumors when injected intra-tumorally compared to PBS injected control tumors, but the sample size was too small (n=2 per group) for meaningful comparison.


Antagonistic Effect of MDNA413-Fc-MDNA109 (KIH) and MDNA109-Fc-MDNA413

The capacity of MDNA413-Fc-MDNA109 (KIH) and MDNA109-Fc-MDNA413 to inhibit IL-4 and IL-13 induced STAT6 activation was assayed. by using HEK-Blue™ IL-4/IL-13 cells. These cells were generated by stably introducing the human STAT6 gene into HEK293 cells to obtain a fully active STAT6 signaling pathway. The other genes of the pathway are naturally expressed in sufficient amounts. Furthermore, HEK-Blue™ IL-4/IL-13 Cells stably express the reporter gene secreted embryonic alkaline phosphatase (SEAP) under the control of the IFNb minimal promoter fused to four STAT6 binding sites. Activation of the STAT6 pathway in HEK-Blue™ IL-4/IL-13 Cells by IL-4 or IL-13 induced the expression of the reporter gene. SEAP which was secreted in the supernatant was detected when using QUANTI-Blue™ a medium that turns purple/blue in the presence of SEAP.


The HEK Blue™ IL-4/IL-13 cells were treated with human IL-4 (hIL4), human IL-13 (hIL13) or mouse IL-13 (mIL13) with or without an indicated fusion protein for 24 hours. 20 μl of the cell culture supernatant was then taken and mixed with 180 μl of QUANTI-Blue™ solution and incubated for 2 hours at 37° C. Absorbance was measured at 650 nm.


As shown in FIG. 20, human IL-4 (hIL4), human IL-13 (hIL13) and mouse IL-13 (mIL13) activated STAT6 signaling pathway in a dose dependent manner. The EC50 was shown in the figure for each interleukin protein.


After HEK Blue™ IL-4/IL-13 cells were treated with human IL-13 (hIL13) together with MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, Fc-MDNA413, or Fc-MDNA109, the activation of STAT signaling pathway by hIL13 was antagonized by MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, and Fc-MDNA413 (FIG. 21). Among the above three fusion proteins, MDNA109-Fc-MDNA413 more effectively inhibits hIL-13 induced STAT6 activation.


After HEK Blue™ IL-4/IL-13 cells were treated with human IL-13 (hIL13) together with MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, Fc-MDNA413, or Fc-MDNA109, the activation of STAT signaling pathway by hIL13 was antagonized by MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, and Fc-MDNA413 (FIG. 21). Among the above three fusion proteins, MDNA109-Fc-MDNA413 and Fc-MDNA413 mostly effectively inhibit hIL13 induced STAT6 activation. MDNA413-Fc-MDNA109 (KIH) inhibits hIL13 induced STAT6 activation, but to a lesser degree than MDNA109-Fc-MDNA413 and Fc-MDNA413.


After HEK Blue™ mIL-4/IL-13 cells were treated with mouse IL-13 (mIL13) together with MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, Fc-MDNA413, or Fc-MDNA109, the activation of STAT signaling pathway by mIL13 was antagonized by MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, and Fc-MDNA413 (FIG. 22). Among the above three fusion proteins, MDNA109-Fc-MDNA413 and Fc-MDNA413 mostly effectively inhibit mIL13 induced STAT6 activation. MDNA413-Fc-MDNA109 (KIH) inhibits mIL13 induced STAT6 activation, but to a lesser degree than MDNA109-Fc-MDNA413 and Fc-MDNA413.


After HEK Blue™ mIL-4/IL-13 cells were treated with human IL-4 (hIL4) together with MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, Fc-MDNA413, or Fc-MDNA109, the activation of STAT signaling pathway by hIL4 was antagonized by MDNA413-Fc-MDNA109 (KIH), MDNA109-Fc-MDNA413, and Fc-MDNA413 (FIG. 23). Among the above three fusion proteins, MDNA109-Fc-MDNA413 and Fc-MDNA413 mostly effectively inhibit hIL4 induced STAT6 activation. MDNA413-Fc-MDNA109 (KIH) inhibits hIL4 induced STAT6 activation, but to a lesser degree than MDNA109-Fc-MDNA413 and Fc-MDNA413.


MDNA413-Fc-MDNA109 (KIH), MDNA109FEAA-Fc-MDNA413 and MDNA132-Fc-MDNA109 (KIH) Maintain IL-2 Mutein Activity

The IL-2 activity was assessed in the bispecific fusion proteins comprising an IL-2 mutein. The murine T cell line CTLL2 was used. This line is IL-2 dependent and constitutively expresses the αβγ form of IL-2R. The cells were cultured in RPMI 1640 supplemented with 10% heat-inactivated FBS, 2 mM L-glutamine, 50 U/ml penicillin, and 50 mg/ml streptomycin. Recombinant human IL-2 (rhIl2) or various fusion proteins were supplemented to the cell culture and the proliferation of the cells were assayed.


As shown in FIG. 24A, cell proliferation was dependent on IL-2. Cell proliferation was nonexistent when the cell culture was supplemented with Fc-MDNA413 instead of IL-2 (FIG. 24B). Cell proliferation was maintained or increased when the cell culture was supplemented with a bispecific fusion protein comprising an IL-2 mutein (MDNA413-Fc-MDNA109 (KIH), MDNA109FEAA-Fc-MDNA413 or MDNA132-Fc-MDNA109 (KIH), FIG. 24C). Therefore, MDNA413-Fc-MDNA109 (KIH), MDNA109FEAA-Fc-MDNA413 and MDNA132-Fc-MDNA109 (KIH) maintained IL-2 mutein activity.


P-STAT5 Assay in Human PBMC:

Briefly, the experimental design (carried out at CRL, Portishead, UK) was as follows: PBMCs were isolated over a density gradient, allowed to rest in complete media, and then stimulated for 15 minutes with 10-point 5-fold dilutions (from 25,000 pM to 0.013 pM) of each test construct to generate dose-response curves in order to determine EC50. Cells were fixed immediately following stimulation, and samples analyzed by flow cytometry after intracellular staining for phosphorylated STAT5 (pSTAT5) in the immune cell subsets listed in Table 21. The gating strategy employed to enable pSTAT5 analysis in these subsets is depicted in FIG. 7 and representative fluorescence minus one (FMO) controls are shown in FIG. 8.









TABLE 21







Flow cytometry markers for different immune cell subsets










Cell type
Markers







Treg cells
CD3+CD4+CD25+FOXP3+



CD8 T-cells (naive)
CD3+CD8+ CD25



CD8 T-cells (activated)
CD3+CD8+ CD25+



NK cells
CD3CD56+










As provided in FIG. 7, gating strategy. PBMCs were stimulated with IL-2 (25 nM) for 15 minutes prior to staining and analysis by flow-cytometry. Single lymphocytes were identified from which NK cells (CD3−CD56+), CD8+ T-cells (CD3+CD8+), Tregs (CD3+CD4+CD25+FOXP3+) and non-Treg CD4+ T-cells (CD3+CD25−CD4+FOXP3−) were gated. Frequency of pSTAT5 positive cells was determined for each immune cell subset.


As provided in FIG. 8, fluorescence minus one (FMO) controls. A) Following stimulation and gating strategy described in FIG. 4, all antibodies used to identify the different immune subsets were included except for pSTAT5 to provide an FMO control that could be used to create a pSTAT5+ gate for each population. B) FMOs for FOXP3 and CD25 were used to determine Treg population.


MTD (Maximum Tolerated Dose):

As provided in FIGS. 11 and 12, female BALB/c (Table 22) or C57BL/6 (Table 23) mice of 6-8 weeks of age were acclimatized for at least 7 days prior to start of experiment. Mice were administered with constructs by intraperitoneal (IP) inject following the dose and schedule indicated below. During the 2-week study, mice were monitored daily by cage-side observation and body weight measurements twice weekly. In these studies, MTD was strictly defined as any incidence of spontaneous mortality or mandatory termination due to weight loss (>20% of the pre-dose weight) or moribund during the study period. At end of the study, mice were euthanized and necropsy was performed.


Studies in BALB/c Mice:

BALB/c mice treated with either 1 mg/kg or 2.5 mg/kg of MDNA132-Fc-MDNA109 or MDNA413-Fc-MDNA109 twice weekly for two weeks were all viable at the end of the study. Mice in both groups appeared clinically normal and maintained their weights during the study period. In summary, MTD was not reached at the highest dose tested (2.5 mg/kg) on a twice weekly dosing schedule for two weeks for both MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 in BALB/c mice.









TABLE 22







MTD study in BALB/c strain of mice.










Bi-specific Superkine
# of Mice
Dose
Regimen





MDNA132-Fc-
5
1.0 mg/kg
2×/week for 2 weeks


MDNA109
5
2.5 mg/kg
2×/week for 2 weeks


MDNA413-Fc-
5
1.0 mg/kg
2×/week for 2 weeks


MDNA109









Studies in C57BL/6 Mice:

A lower range of dose was used because C57Bl/6 has been found to be more sensitive to IL-2 than BALB/c mice (Chen et al., 2005). C57Bl/6 mice treated with MDNA132-Fc-MDNA109 or MDNA413-Fc-MDNA109 twice weekly for two weeks at 0.2 mg/kg, 0.5 mg/kg and 1.0 mg/kg were all viable at the end of the study. Mice in both groups appeared clinically normal and maintained their weights during the study period. In summary, MTD was not reached at the highest dose tested (1.0 mg/kg) on a twice weekly dosing schedule for two weeks for both MDNA132-Fc-MDNA109 and MDNA413-Fc-MDNA109 in C57Bl/6 mice.









TABLE 23







MTD Study in C57Bl/6 strain of mice.










Bi-specific Superkine
# of Mice
Dose
Regimen





MDNA132-Fc-
5
0.2 mg/kg
2×/week for 2 weeks


MDNA109
5
0.5 mg/kg
2×/week for 2 weeks



5
1.0 mg/kg
2×/week for 2 weeks


MDNA413-Fc-
5
0.2 mg/kg
2×/week for 2 weeks


MDNA109
5
0.5 mg/kg
2×/week for 2 weeks



5
1.0 mg/kg
2×/week for 2 weeks









Note: Chen X, Oppenheim J J, Howard O M. BALB/c mice have more CD4+CD25+ T regulatory cells and show greater susceptibility to suppression of their CD4+CD25− responder T cells than C57BL/6 mice. J Leukoc Biol. (2005) 78(1):114-21.


CT26 Efficacy Study:

The CT26 syngeneic colon cancer model was selected to evaluate the therapeutic potential of bi-specific superkines for a number of reasons:


The CT26 cancer model is less aggressive compared to many other cancer models (e.g. B16F10 melanoma model, 4T1 metastatic breast cancer model or the Panc2 pancreatic cancer model), therefore providing a larger window to follow the therapeutic effects of MDNA109 variants.


CT26 cancer model is responsive to ICIs in contrast to several other models, therefore providing an opportunity to evaluate combinatory treatments to determine potential synergy.


The relatively slow growth characteristic of CT26 tumors provides an opportunity to evaluate the anti-tumor activity of MDNA109 variants in the context of early stage (i.e. small) versus late stage (i.e. large) tumors.


To date, one efficacy study has been conducted in the CT26 model with MDNA132-Fc-MDNA109 (KIH). Study design is as follow:


Female (8-10 weeks old) BALB/c mice were implanted with 2×106 CT26 cells subcutaneously in the right flank, and tumors were allowed to grow until average tumor size reached 60 mm3.


Mice were randomized into groups of either 6 (vehicle control) or 8 (MDNA132-Fc-MDNA109) mice each based on tumor size.


Mice were dosed by IP with either vehicle (PBS) or MDNA132-Fc-MDNA109 at 5 mg/kg once weekly for 2 weeks. This was based on dose and schedule of other MDNA109 constructs under investigation at that time.


Study measurements to include:

    • Daily clinical cage-side observations
    • Twice weekly body weights and tumor measurements
    • Monitor food and water consumption.


Animals to be prematurely terminated if any of the following criteria are met:

    • Weight loss exceeding 20% of the maximum weight for that animal
    • Tumor volume exceeding 2000 mm3
    • Animal appearing moribund


As provided in FIG. 13, treatment with MDNA132-Fc-MDNA109 (KIH) resulted in potent inhibition of CT26 tumors growth. In fact, 3 of 8 mice treated with MDNA132-Fc-MDNA109 (KIH) resulted in complete tumor regression whereas this was not observed with control mice. These three mice have not shown any sign of tumor relapse for more than 120 days post-implant or more than 3 months since treatment was stopped, indicating that they have been cured of cancer.


As provided in FIG. 14, to validate whether mice that were cured of their primary CT26 tumors following 2 doses of MDNA132-Fc-MDNA109 are protected against subsequent re-challenges, the three mice were implanted with CT26 on their opposite flank on Day 49 of the study and were not given any further treatment. As controls, naïve untreated mice were also implanted with CT26 tumor cells. Naïve BALB/c mice showed robust CT26 tumor growth. In contrast, the mice treated with MDNA132-Fc-MDNA109 and cured of their primary tumors did not show any sign of tumor growth at the re-challenge site, suggesting that they have developed a strong memory response against CT26 tumor cells. These mice have undergone a second re-challenge and are continued to be monitored. MDNA132-Fc-MDNA109 therefore provides these mice with overall survival benefits in spite of multiple re-challenges.


B16F10 Efficacy Study

The B16F10 syngeneic melanoma model in C57Bl/6 mice is an aggressive in vivo tumor model that has withstood many therapeutic efforts due in part to the speed at which these tumors grow and metastasize in mice. In comparison to the CT26 tumor model, this is a significantly more difficult model to treat and therefore provides an important challenge to evaluate the potential therapeutic activity of bi-specific superkines. To date, two efficacy studies have been separately conducted to evaluate the efficacy of MDNA413-Fc-MDNA109 (KIH) and MDNA109FEAA-Fc-MDNA109. Both studies followed a similar protocol as outlined below:


Female (8-10 weeks old) C57Bl/6 mice were implanted with 3×105 C57Bl/6 cells subcutaneously in the right flank, and tumors were allowed until tumor growth has been established before initiation of dosing.


Mice were randomized into treatment groups based on tumor size such that average tumor size of each group are similar.


Mice were dosed by IP at dose and schedule specific to each study (see below)


Study measurements to include:

    • Daily clinical cage-side observations
    • Twice weekly body weights and tumor measurements
    • Monitor food and water consumption.


Animals to be prematurely terminated if any of the following criteria are met:

    • Weight loss exceeding 20% of the maximum weight for that animal
    • Tumor volume exceeding 2000 mm3
    • Animal appearing moribund


      B16F10 Efficacy Study with MDNA413-Fc-MDNA109 (KIH):


As provided in FIG. 15, average tumor size was ˜20 mm3 at the initiation of dosing. Mice were IP injected with PBS (n=9) or MDNA413-Fc-MDNA109 at 1 mg/kg (n=7) or 2.5 mg/kg (n=9) twice weekly for 2 weeks. Treatment with MDNA413-Fc-MDNA109 resulted in inhibition of B16F10 tumor growth in a dose-dependent manner. MDNA413-Fc-MDNA109 given at 2.5 mg/kg was more potent at growth inhibition than the 1 mg/kg dose.


B16F10 Efficacy with MDNA109FEAA-Fc-MDNA413.


As provided in FIG. 16, tumors were grown to an average size of ˜15 mm3 at the start of dosing. Mice were dose with PBS (n=8) or MDNA109FEAA-Fc-MDNA413 (n=8) once weekly for 3 weeks at 5 mg/kg. As shown in FIG. 13, MDNA109FEAA-Fc-MDNA413 had no effect on the growth of B16F10 melanomas with tumors growing similarly as with the control mice. These data suggest that a more frequent dosing schedule (twice weekly) may be necessary to achieve a therapeutic response with MDNA109FEAA-Fc-MDNA413.


HEK Blue IL-4/IL-13 Assay:

HEK-Blue IL-4/IL-13 reporter cells were purchased from InvivoGen (Catalog bkb0il413). The kit contained all necessary contents for execution. Detailed description of methodology is provided by the InvivoGen. This cell line provides readout of activation of IL-4/IL-13 signaling in response to these cytokines.


Dose-Response to Human (h) IL13, Mouse (m) IL13 and Human (h) IL4

As provided in FIG. 20, cells were plated at 50,000 cells per well in the test medium per the manufacturer's instructions and treated with serially diluted hIL13, mIL13, and hIL4 for 24 hours. After incubation, the cell supernatant (20 μL) was removed to a new plate and then 180 of QUANTI-Blue solution was added and incubated for 2 hours at 37 C. Plates were scanned on a conventional plate reader for absorbance at 650 nm.


Blocking Assay with Bi-Specific Constructs


Variants tested are presented in Table 24. Cells were plated at 50,000 cells per well in the test medium per the manufacturer's instructions and treated with increasing concentrations of hIL13, mIL13 and hIL4 for 24 hours. In addition, wells containing hIL13 (at 10 and 30 ng/mL), mIL13 (at 10 ng/mL) and hIL4 (at 0.1 ng/mL) were treated with increasing concentrations of the test constructs listed in Table 25. Two concentrations were selected for hIL13 to examine the difference between running the assay at saturating or just sub-saturating levels of hIL13. After incubation, the cell supernatant (20 μL) was removed to a new plate and then 180 of QUANTI-Blue solution was added and incubated for 2 hours at 37 C. Plates were scanned on a conventional plate reader for absorbance at 650 nm.









TABLE 24







Test Constructs











Construct
Source
Note





1
MDNA413-Fc-MDNA109
At MDS
MDNA413 = IL4/IL13 antagonist



(KIH) (1:1:1) T1912B03

MDNA109 = IL2





superkine/agonist


2
MDNA109FEAA-Fc-
At MDS
MDNA413 = IL4/IL13 antagonist



MDNA413

MDNA109FEAA = IL2



(2:1:2) T1924B08

superkine/agonist


3
Fc4-413 T1918B04
At MDS
MDNA413 = IL4/IL13 antagonist


4
Fc-MDNA109 T1912B04
At MDS
MDNA109 = IL2





superkine/agonist









As provided in FIG. 21, For hIL13 the addition of up to 10,000 ng/mL of the various test proteins had a clear inhibitory effect on the hIL13 response with Fc4-MDNA413 and MDNA109FEAA-Fc-413 when hIL13 was tested at 10 ng/mL (˜EC80). MDNA413-Fc-MDNA109 KIH had a weak effect, and no obvious effect was observed with Fc-MDNA109. When hIL13 was included at 30 ng/mL (EC100), the responses were similar although the overall level of inhibition observed was much lower due to the higher level hIL13 included in the reactions.


As described in FIGS. 22 (mIL13) and 23 (hIL4): for mIL13 (tested at 10 ng/mL or EC60 for the standard curve in this experiment) and hIL4, (tested at 0.1 ng/mL or EC55 for this standard curve) the trends were also similar. Clear inhibition of the mIL13 activation response (see, FIGS. 22 and 23) and the hIL4 activation was observed with Fc4-MDNA413 and MDNA109FEAA-Fc-413 while MDNA413-Fc-MDNA109 KIH had a weaker overall effect. Again, little inhibition was observed with Fc-MDNA109 alone.


CTLL-2 Proliferation Assay

As described in FIG. 24, CTLL-2 is a mouse cytotoxic T lymphocyte cell line that is dependent on IL-2 for growth, and used as an in vitro functional model to measure potency of IL-2 as they express high affinity IL-2 receptor (IL-2R) consisting of CD122, CD132 and CD25, shown to be highly sensitive to IL-2 treatment (Carmenate et al., 2013).


Carmenate T, Pacios A, Enamorado M, Moreno E, Garcia-Martinez K, Fuente D, Leon K. Human IL-2 mutein with higher antitumor efficacy than wild-type IL-2. The Journal of Immunology (2013) 190:6230-6238.


CTTL2 cells were plated into 96 well plates at 30,000 cells per well in media lacking the TSTIM proliferation supplement. Following plating, cells were treated increasing concentrations of the various samples listed in Table 25 for 48 hours. After treatment, Cell Titer Blue viability reagent (Promega G8080) was added to each well and the plates were scanned after 3 hours at 560Ex/590Em for development of the fluorescent viability signal. Triplicate wells were tested at each concentration of the test samples.









TABLE 25







Test constructs in CTLL-2 assay










Construct
Concentrations tested





1
IL2 TP20284F
0.3 pM to 0.33 nM by ~½ log


2
MDNA413-Fc-MDNA109 (KIH)
10 pM to 10 nM by ~½ log



(1:1:1) T1912B03



3
MDNA109FEAA-Fc-MDNA413
10 pM to 10 nM by ~½ log



(2:1:2) T1924B08



4
Fc4-413 T1918B04
10 pM to 10 nM by ~½ log


5
MDNA132-Fc-MDNA109 (KIH)
10 pM to 10 nM by ~½ log



(1:1:1) T192B02









Results:

rhIL-2: shows dose-response of CTLL-2 cell proliferation to increasing concentration of rhIL-2 (as expected). rhIL-2 was used as a control.


MDNA109FEAA-Fc-MDNA413:

Shows dose-response of CTLL-2 cell proliferation to increasing concentration of this construct. There is reduced potency with this construct (Ec50=53 pM) compared to rhIL2 (EC50=6.4 pM). This is expected since FEAA mutations abrogate binding to CD25, which is expressed by CTLL2.


MDNA132-Fc-MDNA109 (KIH) and MDNA413-Fc-MDNA109 (KIH):

Very strong effect for both constructs as there was similar signals across the entire range of concentrations tested. This is expected since both constructs do not contain FEAA mutations and therefore can engage CD25. Additional, the MDNA109 components have increased binding CD122 and therefore much more potent than rhIL2 (which also binds CD25). To get dose-response curves, we will need to further reduce the concentrations of these 2 constructs in this assay.


Fc4-MDNA413:

Does not induce CTLL2 proliferation as expected since there is no IL-2/MDNA109 moiety.


rhIL2 & MDNA19:


Historical data included here for comparison.


REFERENCES



  • Chen X, Oppenheim J J, Howard O M. BALB/c mice have more CD4+CD25+ T regulatory cells and show greater susceptibility to suppression of their CD4+CD25− responder T cells than C57BL/6 mice. J Leukoc Biol. (2005) 78(1):114-21.

  • Levin A M, Bates D L, Ring A M, Krieg C, Lin J T, Su L, Moraga I, Raeber M E, Bowman G R, Novick P, Pande V S, Fathman C G, Boyman O, Garcia K C. Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine’. Nature (2012) 484(7395):529-33.



Example 3: Emergence of Novel Long-Acting Mono- and Bi-Specific IL-2/IL-13 Superkines as Potent Immune Modulators
Background:

Use of IL-2 (Proleukin) to treat renal carcinoma and metastatic melanoma achieved durable response in ˜15% of cases, but its application is limited due to its short half-life, toxicity and undesirable activation of Tregs, the latter due to binding to IL-2Rαβγ.


MDNA11 is a long-acting IL-2 Superkine with enhanced affinity to IL-2Rβγ (expressed by CD8+ T & NK cells) and diminished binding to IL-2Rαβγ (expressed by Tregs).


Conclusions

MDNA11 demonstrated potent therapeutic efficacy in mouse tumor models as monotherapy and in combination with immune checkpoint inhibitor.


In NHP, MDNA11 exhibited prolonged half-life and induced durable proliferation and expansion of CD8 T cells, CD4 T helper cells, NK cells but not Tregs.


MDNA11 did not cause vascular leak syndrome (i.e pulmonary edema), hypertension, cytokine storm and anti-drug antibody response.


The versatility of IL-2 and IL-13 superkine platforms enables engineering of long-acting bi-specific constructs to simultaneously activate IL-2 signaling (i.e. pro-inflammatory) and suppress IL-4/IL-13 function (i.e. anti-inflammatory) to target immunologically ‘cold’ tumors.


Targeting immunologic “cold tumors” by modulation of TME (tumor microenvironment) with IL-2/IL-13 Bi-specific Superkines.


“Cold” tumors are not responsive to check-point inhibitors because of a pro-tumoral TME, including low CD8+& NK cell counts; high Treg counts and high number of immune-suppressive myeloid cells (i.e. TAM & MDSC).


Example 4: MDNA132/MDNA413 Studies

The binding of MDNA132 and MDNA413 to IL-13Rα2 and IL-13Rα1 was assessed. Briefly, Receptors listed in Table 26 below were adsorbed to 96 well plates (at 1 μg per well) for 1 hour at room temperature in coating buffer (Carbonate pH 9.6). After coating, wells were washed 5 times with 250 μL of Tris-buffered Saline solution with Tween (TBST: 10 mM Tris HCl, 0.15 M NaCl, 0.1% Tween 20, pH 8.0) and then blocked with 250 μL of blocking buffer (1% BSA in TBS-T) for one hour at room temperature. Wells receiving no receptor were simply blocked with blocking buffer for one hour at room temperature. Wells were washed 5 times with TBST prior to sample addition. Samples were diluted as described and then 100 μL of each dilution was added to either receptor coated or albumin blocked wells for 1 hour at room temperature. Wells were then washed 5 times with TBST and probed with 100 μL of the detector antibody (1:5,000 dilution of anti-human IgG Fc Sigma SAB3701284) in blocking buffer (100 μL per well) at room temperature for 45 minutes. Wells were washed again and then incubated with an HRP-conjugated anti-goat IgG (Millipore 401515) for 45 minutes at room temperature. After a final series of washes, 100 μL of developer (0.1 mg/mL TMB, 0.006% hydrogen peroxide in TMB substrate buffer) was added. Plates were quenched after 10 minutes with 100 μL 1M HCl, and plates were scanned for absorbance at 450 nm to obtain optical density units (OD) for each well.









TABLE 26







Reagents









Reagent
Source
Catalog





IL13Rα2 Protein, Human, Recombinant, His
Sino
10350-H08H


Tag Source: HEK293
Biological
100 ug


IL13Rα2 Protein, Mouse, Recombinant, His
Sino
50061-M08H


Tag Source: HEK293
Biological
100 ug


IL13Rα Protein, Human, Recombinant, His
Sino
10943-H08H


Tag Source: HEK293
Biological
100 ug


IL13Rα1 Protein, Mouse, Recombinant, His
Sino
50088-M08H


Tag Source: HEK293
Biological
100 ug


IL4Rαa Protein, Human, Recombinant (His
Sino
10402-H08H


Tag) Source: HEK293 (aa 1-232, ECD)
Biological
10 ug


IL4Rα Protein, Mouse, Recombinant (ECD,
Sino
51180-M08H


His Tag)
Biological
100 ug


Source: HEK293 (aa 1-233, ECD)




Human recombinant Fc-IL13 *Control)
Sino
10369-H01H-



Biological
20 ug









As shown in FIG. 35, good binding of Fc-hIL13 was observed on human and mouse IL13Rα2 receptor coated wells, although the mIL13Rα2 coated wells had overall higher background, possibly due to non-specific interactions either with the detector or the HRP-conjugated secondary antibodies. Low binding was observed on the mIL13Rα1 and bindings similar to uncoated wells were observed with hIL13Rα1, and the human and mouse IL4R receptors.


As shown in FIGS. 36A and 36B, MDNA132-Fc-MDNA109 (1:1:1) molecule does in fact bind to the human and mouse IL13Rα2 receptors with similar affinity as Fc-IL13.


As shown in FIGS. 37A and 37B, all MDNA413 variants showed binding on the human and mouse IL13Rα1 receptor. Binding was strongest and similar for the MDNA109FEAA-Fc-MDNA413 (2:1:2) and Fc4-MDNA413 molecules, while binding of MDNA109-Fc-MDNA413 (1:1:1) was similar to Fc-IL13.


Example 5: MDNA132/MDNA413 Studies

Receptor binding affinity of MDNA132 and MDNA413 was assessed by surface plasmon resonance using Biacore T200 instrument. Briefly, Protein A (or anti-human Fc) was pre-immobilised on the sensor chip and captured each construct via its Fc portion. A receptor was flown over the surface to measure association, followed by a dissociation. Multiple concentrations of the receptors were tested in separate cycle, multi cycle kinetics (MCK).


As shown in FIG. 38, Fc-MDNA132 (1:1 KIH) bound to IL13Rα2 and not IL13Rα1. The KD Fc-MDNA132 for IL13Rα2 is estimated to be 1.30E−09 M.


As shown in FIG. 39, Fc-MDNA413 (1:2) bound to IL13Rα1 with higher affinity when compared to wild type IL13. As shown in FIG. 40, Fc-MDNA413 (1:2) bound with lower affinity to IL13Rα2 in comparison to wild type IL13. The estimated KD values from the assay are depicted in Table 27 below.









TABLE 27







Affinities









Antigen
Ligand
KD (M)





IL13Ra1
Fc-WT IL13
6.80E−08



Fc-MDNA413(1:2)
1.94E−08


IL 13Ra2
Fc-WT IL13
1.44E−10



Fc-MDNA413(1:2)
9.73E−09









Example 6: Modulation of Immune Responses to Cancer by Bi-Specific IL-2/IL-13 Superkines Background

The IL-2 pathway plays a vital role in stimulating a pro-inflammatory (Th1) response against cancer through expansion and activation of effector CD8 T and NK cells. In contrast, the IL-4/IL-13 pathway stimulates myeloid derived suppressor cells (MDSCs) and M2 skewing of tumor associated macrophage (TAM) to foster an anti-inflammatory (Th2) response that is often exploited by cancers as a means to dampen the effects of the Th1 pathway. Therefore, suppression of MDSC and M2 TAM through inhibition of the IL-4/IL-13 pathway together with stimulation of effector immune cells through activation of the IL-2 pathway have the potential to invigorate a pro-inflammatory response in an otherwise immune suppressive tumor microenvironment (TME). To achieve this goal, the versatility of IL-2 and IL-13 superkine platforms was leveraged to engineer long-acting bi-specific constructs to co-target surface receptors of the respective pathways.


Materials and Methods

Studies included binding analyses by BLI/Octet and Biacore/SPR, signaling analyses using IL-2 and IL-4/IL-13 reporter assays, signaling analysis in human PBMC, and in vitro M1/M2 macrophage polarization assay.


The binding affinity to IL13Rα1 and IL13Rα2 by SPR (MCK, multi-cycle kinetics) was measured using the human IgG (Fc) Capture Chip principle, which utilizes the following steps: (1) Immobilization: Immobilized Anti-Human IgG (Fc) antibody on CM5 chip; (2) Capture: capture the IL-13 Protein, Fc Tag or sample; (3) Analyte: the analyte is Human IL-13 R alpha 1 Protein or Human IL-13 R alpha 2 Protein; and (4) Regeneration: 3 M magnesium chloride.


A pSTAT5 signaling assay in human PBMC was implemented as follows. PBMCs were isolated over a density gradient, allowed to rest in complete media, and then stimulated for 15 minutes with IL2, MDNA109FEAA-Fc or MDNA109FEAA-Fc-MDNA413. Controls included non-stimulated PBMC cells. Cells were fixed immediately following stimulation, and samples analyzed by flow cytometry after intracellular staining for phosphorylated STAT5 (P-STAT5) in the immune subsets, Naïve CD8 T cells (CD8+CD25−), NK cells and Tregs.


An assay was performed in HEK Blue IL-4/IL-13 reporter cells (from InvivoGen) to measure pSTAT6 signaling as follows. HEK-Blue IL-4/IL-13 reporter cells (Invivogen) were plated at 50,000 cells per well in the test medium per the manufacturer's instructions and treated with increasing concentrations of either hIL4 or hIL13 for 24 hours (standard curve. In addition, wells containing hIL13 or hIL4 were treated with increasing concentrations of the test samples. Each standard or test sample serial dilution was assayed in duplicate wells. After incubation, the cell supernatants (20 μL) were removed to a new plate and then 180 of QUANTI-Blue solution was added and incubated for 2 hours at 37° C. Plates were scanned on a conventional plate reader for absorbance at 650 nm.


A macrophage polarization assay was performed as follows. Monocytes were first isolated from fresh or thawed frozen vial of human peripheral blood mononuclear cells (PBMCs) from 2 donors using EasySep™ Human Monocyte Enrichment Kit without CD16 Depletion on RoboSep. Cells were then seeded onto low-bind 6-well plates at 2×106 cells/well. Maturation of monocytes were then induced for 72 hrs using 50 ng/mL M-CSF exposure in media containing RPMI with L-glutamine (Gibco 21875-034), 10% FBS, 2% Pen-Strep, and incubated at 37° C. and 5% CO2. Polarization was subsequently done using the IL-13 agonist or control for another 72 hrs, following which they were dissociated, fixed using BD CellFIX, stained and examined using flow cytometry.


A CT26 colon cancer syngeneic mouse model was developed and used as follows. Female BALB/c mice were implanted with 2×106 CT26 cells subcutaneously in the right flank (study day 0) and allowed to grow for 11 days prior to the initiation of dosing. The average tumor size in the treatment group at time of dosing was about 60 mm3. MDNA132-Fc-MDNA109 was used at 5 mg/kg and the IP was QWx2.


Results


FIG. 41A-41C, along with the KD values in FIG. 41D, shows that Fc-MDNA413 binds to the functional receptor IL13Rα1 with a higher affinity than Fc-IL13. Also, the affinity to the decoy receptor, IL13Rα2 is reduced for Fc-MDNA413 in comparison to Fc-IL13. The higher binding affinity of Fc-MDNA413 for IL13Rα1 is also observed in the bispecific construct MDNA109FEAA-Fc-MDNA413 indicating that it can bind more strongly to IL13Rα1 receptor than Fc-IL-13.



FIG. 42A-42B shows that MDNA109FEAA-Fc-MDNA413 is able to activate signaling in naïve CD8 T cells and NK cells with a higher potency than rhIL-2. However, the potency is greatly reduced for pSTAT5 signaling in Tregs with MDNA109FEAA-Fc-MDNA413 in comparison to rhIL-2. These data show that MDNA109FEAA-Fc-MDNA413 is superior to rhIL-2 at activating immune effectors cell response required for achieve anti-cancer efficacy.



FIG. 43A-43C shows that both Fc-MDNA413 (monospecific) and MDNA109FEAA-Fc-MDNA413 (bispecific) are able to inhibit the IL4 (top panel) and IL13 (bottom panel) induced pSTAT6 signaling in the HEK Blue reporter cells with EC50 as shown in the table above. These data showed that Fc-MDNA413 and MDNA109FFEAA-Fc-MDNA413 can suppress IL-4 and IL-13 induced signaling.



FIG. 44A-44B shows that CD209 is a marker for M2 macrophages and is induced upon IL13 stimulation. As seen from the figure above, both Fc-MDNA413 (monospecific) and MDNA109FEAA-Fc-MDNA413 (bispecific) are able to inhibit the IL13 induced M2 polarization of macrophages in a dose dependent manner.



FIG. 45A-45D, along with the KD values in FIG. 45E, shows that Fc-MDNA132 has preferential binding affinity for the decoy receptor, IL13Rα2 wherein no binding is observed to the functional receptor, IL13Rα1. In contrast, Fc-IL13 binds to both the functional receptor, IL13Rα1 and the decoy receptor, IL13Rα2. These data emphasize the strong receptor selectivity profile of Fc-MDNA132.



FIG. 46A-46C shows that the bispecific construct MDNA132-Fc-MDNA109 is able to inhibit the tumor growth in the CT26 colon cancer model and also, extend overall survival in comparison to the vehicle control.


sIL2M-Fc-sIL13M is a bi-specific superkine composed of an IL-2 super-agonist (sIL2M) and IL-13 super-antagonist (sIL13M) linked together by human IgG1 Fc. sIL2M binds CD122 with superior affinity over IL-2 but does not engage CD25, which is expressed on immune-suppressive Tregs. sIL13M has higher affinity than IL-13 for IL13Rα1, which together with IL-4Rα forms a functional receptor complex. In pSTAT5 assay using primary human PBMCs, sIL2M-Fc-sIL13M showed enhanced potency over IL-2 in the activation of CD8 T-cells and NK cells while exhibiting limited activity on Treg. In an IL-4/IL-13 dependent pSTAT6 reporter assay, sIL2M-Fc-sIL13M demonstrated dose-dependent antagonism against stimulatory activity of both IL-4 and IL-13. This antagonistic effect was further validated with inhibition of IL-13 induced M2 polarization of macrophages in vitro.


Conclusions

sIL2M-Fc-sIL13M is a bi-specific superkine capable of concomitantly stimulating a Th1 response through activation of the IL-2 pathway and suppressing a Th2 response through inhibition of the IL-4/IL-13 pathway. Additional bi-specific superkine constructs will be tested, including those designed to enable accumulation in TME by engaging the decoy IL-13Rα2 that is overexpressed on a number of different tumor types.









TABLE 28







List of Bispecific Fusion Constructs with Mutations Specified









Name
Sequence
Mutations





Fc-MDNA413
Gene 1 and Gene 2 (Version A)
MDNA413


(1:2)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
mutations:



DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
L10V, E12A,



YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
V18I, L39R,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
R65D, D87S,



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV
T88S, L101F,



PPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLIN
K104R, K105T



VSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLFH




LRTLFREGQFN (SEQ ID NO: 168)



Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
L10V, E12A,


MDNA413R39/Q111
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
V18I, L39R,


(1:2)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
R65D, D87S,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
T88S, L101F,



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV
K104R, K105T




PPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALESLIN






VSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

H






L

RT

LFREGQFN (SEQ ID NO: 231)







Fc-MDNA132
Gene 1 (Version A)
MDNA132


(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
mutations:



DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
L10H, R86T,



YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
D87G, T88R,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
R108K



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV




PPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN




VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLH




LKKLFKEGQFN (SEQ ID NO: 169)






Fc-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
D87G, T88R,


(1:1
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
R108K


KIH)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQVSLWC




LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV





PPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN






VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKDLLLH






LKKLF

K

EGQFN (SEQ ID NO: 232)







Fc-MDNA132L39/R111
Gene 1:
L10H, R86T,


(1:1 KIH)

DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE

D87G, T88R,




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE

R108K




YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC






LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR






WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV







PPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN








VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLH








LKKLFKEGRFN
 (SEQ ID NO: 233)







Fc-MDNA132
Gene 1 (Version B)
MDNA132


(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
mutations:



DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
L10H, L39R,



YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
R86T, D87G,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
T88R, R108K



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV




PPSTAHRELIEELVNITQNQKAPLCNGSMVWSINRTAGMYCAALESLIN




VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKDLLLH




LKKLFKEGQFN (SEQ ID NO: 170)






Fc-MDNA132
Gene 2 (Version A)



(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 171)






Fc -
Gene 2:



MDNA132L39/Q111
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



(1:1
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE



KIH)
YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 234)






Fc-MDNA132L39/R111
Gene 2:



(1:1 KIH)
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 235)






MDNA109FEAA-
Gene 1 (Version A)
MDNA109


Fc-
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKK
mutations:


MDNA413
ATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG
F42A, E62A,


(2:1:2)
SETTFMCEYADETATIVEFLNRWITFSQSIISTLTGGGGSGGGGSGGGG
L80F, R81D,



SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
L85V, I86V,



EDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK
I92F, C125S



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT
MDNA413



CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
mutations:



RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGP
L10V, E12A,



VPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLI
V18I, L39R,



NVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLF
R65D, D87S,



HLRTLFREGQFN (SEQ ID NO: 172)
T88S, L101F,




K104R, K105T





MDNA109FEAAC125-

APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLT

A

KFYMPKK

MDNA109FEAA


Fc-

ATELKHLQCLEE

A

LKPLEEVLNLAQSKNFH

FD

PRD

VV

SNINV

F

VLELKG

mutations:


MDNA413R39/Q111

SETTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGG

F42A, E62A,


(2:1:2)
SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
L80F, R81D,



EDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK
L85V, 186V



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT
I92F



CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
MDNA413



RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGP
mutations:




VPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALESLI

L10V, E12A,




NVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F


L39R, V18I,




HL

RT

LFREGQFN (SEQ ID NO: 236)

R65D, D87S,




T88S, L101F,




K104R, K105T





MDNA109FEAA-
Gene 1 (Version B)
MDNA109


Fc-
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAKFYMPKK
mutations:


MDNA413
ATELKHLQCLEEALKPLEEVLNLAQSKNFHFDPRDVVSNINVFVLELKG
F42A, E62A,


(2:1:2)
SETTFMCEYADETATIVEFLNRWITFCQSIISTLTGGGGSGGGGSGGGG
L80F, R81D,



SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
L85V, I86V,



EDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK
I92F



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT
MDNA413



CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
mutations:



RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGP
L10V, E12A,



VPPSTAVRALIEELINITQNQKAPLCNGSMVWSINRTAGMYCAALESLI
V18I, L39R,



NVSGCSAIEKTQDMLSGFCPHKVSAGQFSSLHVRSSKIEVAQFVKDLLF
R65D, D87S,



HLRTLFREGQFN (SEQ ID NO: 173)
T88S, L101F,




K104R, K105T





MDNA132-
Gene 1 (Version A)
MDNA132


Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALE
mutations:


(1:1:1
SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKD
L10H, R86T,


KIH)
LLLHLKKLFKEGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS
D87G, T88R,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
R108K



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
MDNA109



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
mutations:



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
L80F, R81D,



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L85V, I86V,



ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL
I92F C125S



AQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRW




ITFSQSIISTLT (SEQ ID NO: 174)






MDNA132L39/Q111-

PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALE

MDNA132


Fc-

SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKD

mutations:


MDNA109

LLLHLKKLF

K

EGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS

L10H, R86T,


(1:1:1
VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
D87G, T88R,


KIH)
TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
R108K



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
MDNA109



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
mutations:



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L80F, R81D,




ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL

L85V, I86V,




AQSKNFH

FD

PRD

VV

SNINVFVLELKGSETTFMCEYADETATIVEFLNRW

I92F




ITFCQSIISTLT (SEQ ID NO: 237)







MDNA132-
Gene 1 (Version B)
MDNA132


Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALE
mutations:


(1:1:1
SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKD
L10H, R86T,


KIH)
LLLHLKKLFKEGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS
D87G, T88R,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
R108K



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
MDNA109



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
mutations:



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
L80F, R81D,



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L85V, I86V,



ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL
I92F



AQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRW




ITFCQSIISTLT (SEQ ID NO: 175)






MDNA132-
Gene 1 (Version C)
MDNA132


Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINRTAGMYCAALE
mutations:


(1:1:1
SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKD
L10H, L39R,


KIH)
LLLHLKKLFKEGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS
R86T, D87G,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
T88R, R108K



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
MDNA109



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
mutations:



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
L80F, R81D,



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L85V, I86V,



ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL
I92F, C125S



AQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRW




ITFSQSIISTLT (SEQ ID NO: 174)






MDNA132-
Gene 1 (Version D)
MDNA132


Fc-MDNA109
PGPVPPSTAHRELIEELVNITQNQKAPLCNGSMVWSINRTAGMYCAALE
mutations:


(1:1:1
SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHVTGRKIEVAQFVKD
L10H, L39R,


KIH)
LLLHLKKLFKEGQFNGGGGSGGGGSGGGGSDKTHTCPPCPAPELLGGPS
R86T, D87G,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
T88R, R108K



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
MDNA109



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ
mutations:



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
L80F, R81D,



YTQKSLSLSPGGGGGSGGGGSGGGGSAPTSSSTKKTQLQLEHLLLDLQM
L85V, I86V,



ILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLEEVLNL
I92F



AQSKNFHFDPRDVVSNINVFVLELKGSETTFMCEYADETATIVEFLNRW




ITFCQSIISTLT (SEQ ID NO: 175)






MDNA132-
Gene 2 (Version A)
Fc portion


Fc-MDNA109
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



(1:1:1
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE



KIH)
YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSREEMTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 213)






MDNA132L39/Q111-
Gene 2:



Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE



MDNA109
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE



(1:1:1
YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSREEMTKNQVSLSC



KIH)
AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 238)






MDNA413R39/Q111-
Gene 1:
MDNA413


Fc-

PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALE

mutations:


MDNA132L39/Q111

SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD

L10V, E12A,


(2:1:1

LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS

L39R, V18I,


KIH)
VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
R65D, D87S,



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
T88S, L101F



KAKGQPREPQVYTLPPCRDELTKNQVSLWCLVKGFYPSDIAVEWESNGQ
K104R, K105T



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
MDNA132



YTQKSLSLSPGcustom-characterPGPVPPSTAHRELIEELVNITQN
mutations:




QKAPLCNGSMVWSINLTAGMYCAALESLINVSGCSAIEKTQRMLSGFCP

L10H, R86T,




HKVSAGQFSSLHV

TGR

KIEVAQFVKDLLLHLKKLF

K

EGQFN (SEQ ID

D87G, T88R,



NO: 177)
R108K



Gene 2:





PGPVPPSTA

V

R

A

LIEEL

I

NITONQKAPLCNGSMVWSIN

R

TAGMYCAALE






SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD






LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS





VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK




TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS




KAKGQPREPQVCTLPPSRDELTKNQVSLSCAVKGFYPSDIAVEWESNGQ




PENNYKTTPPVLDSDGSFFLVSKLTVDKSRWQQGNVFSCSVMHEALHNH




YTQKSLSLSPG (SEQ ID NO: 178)






Fc4-
APPCPSCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDP
L10V, E12A,


MDNA413R39/Q111
EVQFNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYK
L39R, V18I,


(1:2)
CKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLV
R65D, D87S,



KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQ
T88S, L101F,



EGNVFSCSVMHEALHNHYTQKSLSLSPGKGGGSGSPGPVPPSTAVRALI
K104R, K105T




EEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALESLINVSGCSAIEKT






Q

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

HL

RT

LFREGQF






N (SEQ ID NO: 179)







MDNA413R39/Q111-
Gene 1:
MDNA413


Fc-

PGPVPPSTA

V

R

A

LIEEL

I

NITONQKAPLCNGSMVWSIN

R

TAGMYCAALE

mutations:


MDNA109

SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD

L10V, E12A,


(1:1:1

LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS

L39R, V18I,


KIH)
VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
R65D, D87S,



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
T88S, L101F



KAKGQPREPQVYTLPPCREEMTKNQVSLWCLVKGFYPSDIAVEWESNGQ
K104R, K105T



PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH
MDNA109



YTQKSLSLSPGcustom-characterAPTSSSTKKTQLQLEHLL
mutations:




LDLQMILNGINNYKNPKLTRMLTFKFYMPKKATELKHLQCLEEELKPLE

L80F, R81D,




EVLNLAQSKNFH

FD

PRD

VV

SNINVFVLELKGSETTFMCEYADETATIVE

L85V, I86V,




FLNRWITFCQSIISTLT (SEQ ID NO: 180)

I92F



Gene 2:




DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSREEMTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 181)






Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
L10V, E12A,


MDNA413L39/Q111
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
V18I, R65D,


(1:2)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
D87S, T88S,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
L101F,



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV
K104R, K105T




PPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN






VSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

H






L

RT

LFREGQFN (SEQ ID NO: 182)







MDNA132L39/R111-
Gene 1:
L10H, R86T,


Fc

PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALE

D87G, T88R,


(1:1 KIH)

SLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKD

R108K




LLLHLKKLF

K

EGRFN
custom-character DKTHTCPPCPAPELLGGPS





VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK




TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS




KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ




PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH




YTQKSLSLSPG (SEQ ID NO: 183)




Gene 2:




DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC




LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 184)






Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
L10H, R86T,


MDNA132L39/R111
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
D87G, T88R,


(1:2)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
R108K



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV





PPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN






VSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVKDLLLH






LKKLF

K

EGRFN (SEQ ID NO: 185)







Fc-
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE
L10V, E12A,


MDNA413L39/R111
DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE
V18I, R65D,


(1:2)
YKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTC
D87S, T88S,



LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
L101F,



WQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGPV
K104R, K105T




PPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINLTAGMYCAALESLIN






VSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

H






L

RT

LFREGRFN (SEQ ID NO: 186)







MDNA413L39/Q111-

PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINLTAGMYCAALE

L10V, E12A,


Fc-

SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD

V18I, R65D,


MDNA413L39/Q111

LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS

D87S, T88S,


(2:1:2)
VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
L101F,



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS
K104R, K105T



KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ




PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH




YTQKSLSLSPGcustom-characterPGPVPPSTAVRALIEELINITQN





QKAPLCNGSMVWSINLTAGMYCAALESLINVSGCSAIEKTQ

D

MLSGFCP






HKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F

HL

RT

LFREGQFN (SEQ





ID NO: 187)






H4FEAA-Fc-

APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLTAK

F

YMPKK

H4FEAA


MDNA413L39/Q111

ATELKHLQCLEE

A

LKPLEEVLNLA

S

SKNFHFTPRD

V

VSN

I

NVFVLELKG

mutations:


(2:1:2)

SETTFMCEYADETATIVEFLNRWITF

S

QSIISTLTGGGGSGGGGSGGGG

F42A, E62A



SDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH
Q74S, R81T,



EDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGK
L85V, I92F,



EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLT
C125S



CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS
MDNA413



RWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSGGGGSGGGGSPGP
mutations:




VPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSINLTAGMYCAALESLI

L10V, E12A,




NVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKDLL

F


V18I, R65D,




HL

RT

LFREGQFN (SEQ ID NO: 188)

D87S, T88S,




L101F,




K104R, K105T





mCD3 IgG-
Gene 1:
L10H, R86T


MDNA132L39/Q111
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
D87G, T88R,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R108K


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV



sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL






ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVK






DLLLHLKKLF

K

EGQFN (SEQ ID NO: 189)





Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 190)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 191)






huCD3 IgG-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
D87G, T88R,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R108K


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV



sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPcustom-character





custom-character
PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL






ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVK






DLLLHLKKLF

K

EGQFN (SEQ ID NO: 192)





Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 193)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 194)






mPD1 IgG-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
D87G, T88R,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R108K


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV



sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL






ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVK






DLLLHLKKLF

K

EGQFN (SEQ ID NO: 195)





Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 196)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC SEQ ID NO: 197)






huPD1 IgG-
Gene 1:
L10H, R86T,


MDNA132L39/Q111
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
D87G, T88R,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R108K


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV



sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
PGPVPPSTA

H

RELIEELVNITQNQKAPLCNGSMVWSINLTAGMYCAAL






ESLINVSGCSAIEKTQRMLSGFCPHKVSAGQFSSLHV

TGR

KIEVAQFVK






DLLLHLKKLF

K

EGQFN (SEQ ID NO: 198)





Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 199)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 200)






mPD1 IgG-
Gene 1:
F42A, E62A,


MDNA109FEAAS125
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
L80F, R81D,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
L85V, I86V,


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
I92F, C125S


sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLT

A

KFYMPK






KATELKHLQCLEE

A

LKPLEEVLNLAQSKNFH

FD

PRD

VV

SNINV

F

VLELK






GSETTFMCEYADETATIVEFLNRWITF

S

QSIISTLT (SEQ ID





NO: 201)




Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 202)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 203)






huPD1 IgG-
Gene 1:
F42A, E62A,


MDNA109FEAAS125
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
L80F, R81D,


(KiH)*
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
L85V, I86V,


(*partial
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
I92F, C125S


sequence)
DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPCRDELTKNQ




VSLWCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGcustom-character





custom-character
APTSSSTKKTQLQLEHLLLDLQMILNGINNYKNPKLTRMLT

A

KFYMPK






KATELKHLQCLEE

A

LKPLEEVLNLAQSKNFH

FD

PRD

VV

SNINV

F

VLELK






GSETTFMCEYADETATIVEFLNRWITF

S

QSIISTLT (SEQ ID





NO: 204)




Gene 2:




ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG




VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV




EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV




DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW




LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQ




VSLSCAVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK (SEQ ID




NO: 205)




Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 206)






mPD1 IgG-
Gene 1:
L10V, E12A,


MDNA413R39/Q111*
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
L39R, V18I,


(*partial
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R65D, D87S,


sequence)
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
T88S, L101F,



DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW
K104R, K105T



LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQ




VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKcustom-character





custom-character
PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAA






LESLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFV






KDLL

F

HL

RT

LFREGQFN (SEQ ID NO: 207)





Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 208)






huPD1 IgG-
Gene 1:
L10V, E12A,


MDNA413R39/Q111*
ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSG
L39R, V18I,


(*partial
VHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKV
R65D, D87S,


sequence)
EPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVV
T88S, L101F,



DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDW
K104R, K105T



LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQ




VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT




VDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKcustom-character





custom-character
PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAA






LESLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFV






KDLL

F

HL

RT

LFREGQFN (SEQ ID NO: 209)





Gene 3:




RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQS




GNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPV




TKSFNRGEC (SEQ ID NO: 210)






MDNA413R39/Q111-
Gene 1:
L10V, E12A,


Fc

PGPVPPSTA

V

R

A

LIEEL

I

NITQNQKAPLCNGSMVWSIN

R

TAGMYCAALE

V18I, L39R,


(1:1 KIH)

SLINVSGCSAIEKTQ

D

MLSGFCPHKVSAGQFSSLHVR

SS

KIEVAQFVKD

R65D, D87S,




LL

F

HL

RT

LFREGQFN
custom-character DKTHTCPPCPAPELLGGPS

T88S, L101F,



VFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK
K104R, K105T



TKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTIS




KAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQ




PENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH




YTQKSLSLSPG (SEQ ID NO: 211)




Gene 2:




DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE




DPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKE




YKCKVSNKALPAPIEKTISKAKGQPREPQVCTLPPSRDELTKNQVSLSC




AVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSKLTVDKSR




WQQGNVFSCSVMHEALHNHYTQKSLSLSPG (SEQ ID NO: 212)









Example 7: Assays Using Bispecific Fusion Constructs
HEK Blue IL2 Assay

HEK-Blue IL2 reporter cells were purchased from InvivoGen and the studies were done at MDS, San Diego, USA. The kit contained all necessary contents for execution. Cells were expanded as per the manufacturer's instructions. An assay was performed with control compounds to show proof of principle and establish dose response ranges previously. Subsequently, assays were run to test the potency of the Bispecific constructs harboring IL-2 agonists in the HEK Blue IL-2 pSTAT5 reporter system.


HEK-Blue IL2 reporter cells were plated at 50,000 cells per well in the test medium per the manufacturer's instructions and treated with the constructs noted in Table 29 and at the concentrations noted for 24 hours. After incubation, the cell supernatant (20 μL) was removed to a new plate and then 180 μL of QUANTI-Blue solution was added and incubated for 2 hours at 37° C. Plates were scanned on a conventional plate reader for absorbance at 650 nm.


MDNA109FEAAC125-Fc-MDNA413R39/Q111(2:1:2), MDNA132L39/Q111-Fc-MDNA109C125 (1:1 KIH), MDNA413R39/Q111-Fc-MDNA109C125 (1:1:1 KIH) and H4FEAA-Fc-MDNA413L39/Q111 (2:1:2) with rhIL2 (11979-828879, LP) and MDNA19 (MDNA019FEAA-Fc) were used as experimental control. All the molecules except H4FEAA-Fc-MDNA413L39/Q111 (2:1:2) were observed to have EC50s that were 1.4-1.8 fold lower than rhIL2 (11979-828879, LP) indicating that they are more potent than rhIL2 (11979-828879, LP) in inducing pSTAT5 in the HEK Blue IL2 reporter system (FIG. 47).









TABLE 29







The compiled EC50s of the constructs are listed below:










Construct
EC50 (pM)














MDNA132L39/Q111-Fc-MDNA109C125(1:1:1 KIH)
2.3



MDNA109FEAAC125-Fc-MDNA413R39/Q111
2.3



MDNA413R39/Q111-Fc-MDNA109C125 (1:1:1 KIH)
2.8



H4FEAA-Fc-MDNA413L39/Q111 (2:1:2)
179.4



Human anti-PD1-IgG-MDNA109FEAAS125
3.8



Mouse anti-PD1-IgG-MDNA109FEAAS125
2.3










CTTL-2 Assay

CTLL2 cells were plated into 96 well plates at 30,000 cells per well in media lacking the TSTIM proliferation supplement. Following plating, cells were treated increasing concentrations of the constructs listed in Table 30 below for 48 hours. As a comparator of activity, each plate also contained MDNA11 (TP28767F). After treatment, Cell Titer Blue viability reagent (Promega G8080) was added to each well and the plates were scanned after 3-6 hours at 560Ex/590Em for development of the fluorescent viability signal. Six plates were run for the assay, which each plate containing serial dilutions of the MDNA11 comparator and 2 test constructs run in duplicate wells.


Of the constructs tested for their ability to induce the proliferation of CTTL-2 assay, MDNA109FEAAC125-Fc-MDNA413R39/Q111 (2:1:2), MDNA132L39/Q111-Fc-MDNA109C125(1:1:1 KIH), MDNA413R39/Q111-Fc-MDNA109C125 (1:1:1 KIH) and H4FEAA-Fc-MDNA413L39/Q111 (2:1:2) with rhIL2 (11979-828879, LP) and MDNA11 as experimental control, MDNA413R39/Q111-Fc-MDNA109C125 (1:1:1 KIH) and MDNA132L39/Q111-Fc-MDNA109C125 (1:1:1 KIH) had similar relative potencies. (47 times and 38 times MDNA11 respectively); MDNA19 (MDNA019FEAA-Fc) and MDNA109FEAAC125-Fc-MDNA413R39/Q111 (2:1:2) had similar relative potencies to each other (2.89 and 2.63 times MNDA11 respectively) and were ˜ 1.5 log less active than rhIL2. The lowest potency observed was with H4FEAA-Fc-MDNA413L39/Q111 (2:1:2) which was ˜2% of the MDNA11 potency and ˜1600 times less active than rhIL2 (FIG. 48).









TABLE 30







The compiled list of EC50s of the constructs is listed below:










Construct
EC50 (pM)














MDNA132L39/Q111-Fc-MDNA109C125(1:1:1 KIH)
9.5



MDNA109FEAAC125-Fc-MDNA413R39/Q111
154



MDNA413R39/Q111-Fc-MDNA109C125 (1:1:1 KIH)
8.5



H4FEAA-Fc-MDNA413L39/Q111 (2:1:2)
20900










HEK Blue IL4/IL3

HEK-Blue IL4/IL13 reporter cells were purchased from InvivoGen and the studies were done at MDS, San Diego, USA. The kit contained all necessary contents for execution. Cells were expanded as per the manufacturer's instructions. An assay was performed with control compounds to show proof of principle and establish dose response ranges previously. Subsequently, assays were run to test the potency of the MDNA413 variants in the HEK Blue IL-4/IL-13 pSTAT6 reporter system.


HEK Blue IL4

For IL-4 competitive assay, HEK-Blue IL-4/IL-13 reporter cells (Invivogen) were plated at 50,000 cells per well in the test medium per the manufacturer's instructions and treated with increasing concentrations of hIL4 (R&D Systems 204-IL-010/CF) for 24 hours (standard curve from 0.2 nM down to 0.0002 nM hIL4 serially diluted ˜0.5 log intervals). In addition, wells containing hIL4 (at 0.1 nM), were treated with increasing concentrations of the test samples listed in Table 30 (150 nM down to 0.15 nM by 0.5 log intervals). Each standard or test sample serial dilution was assayed in duplicate wells. A total of 4 plates were run in the assay. After incubation, the cell supernatants (20 μL) were removed to a new plate and then 180 of QUANTI-Blue solution was added and incubated for 2 hours at 37 C. Plates were scanned on a conventional plate reader for absorbance at 650 nm (FIG. 49).


HEK Blue IL13

For the IL-13 competitive assay, HEK-Blue IL-4/IL-13 reporter cells (Invivogen) were plated at 50,000 cells per well in the test medium per the manufacturer's instructions and treated with increasing concentrations of hIL13 (R&D Systems 213-ILB-005/CF) for 24 hours (standard curve from 8 nM down to 0.033 nM hIL13 serially diluted by a factor of 2.5×). In addition, wells containing hIL13 (at 0.8 nM, 10 ng/mL), were treated with increasing concentrations of the test samples listed in Table 30 (150 nM down to 0.11 nM by a factor of 3.33×). Each standard or test sample serial dilution was assayed in duplicate wells. A total of 4 plates were run in the assay. After incubation, the cell supernatants (20 μL) were removed to a new plate and then 180 of QUANTI-Blue solution was added and incubated for 24 hours at 37° C. Plates were scanned on a conventional plate reader for absorbance at 650 nm (FIG. 50).


The data obtained from screening of constructs for their ability to inhibit the IL13 induced pSTAT6 activation in the HEK Blue IL13 reporter system establishes: (1) the leucine, L at 39 position seem to make the molecule less potent than arginine, R at the same position; and (2) the MDNA413 at the C-terminus of Fc seems to be more potent than MDNA413 at the N-terminus of Fc. Also, the EC50s obtained from the assay could enable the rank ordering of molecules with respect to functional in vitro potency as below:









TABLE 31







EC50 rank ordering










IL 13
IL4


Construct
EC50 (nM)
EC50 (nM)












MDNA109FEAAC125-Fc-MDNA413R39/Q111
12.3
8.05


(2:1:2)




Fc4-MDNA413R39/Q111 (1:2)
37.8
7.5


Fc-MDNA413R39/Q111 (1:2)
39.0
26.48


huPD1 IgG-MDNA413R39/Q111 (1:2)
57.9
34.18


MDNA413R39/Q111-Fc (1:1 KIH)
138.7
96.75


mPD1 IgG-MDNA413R39/Q111 (1:2)
148.5
86.4


MDNA413R39/Q111-Fc-MDNA109 (1:1:1
203.0
92.17


KIH)




Fc-MDNA413L39/Q111(1:2)
268.0
211.6


Fc-MDNA413L39/R111 (1:2)
270.0
150.5


MDNA413L39/Q111-Fc-MDNA413L39/Q111
600.8
326.2


(2:1:2)




MDNA413R39/Q111-Fc-MDNA132L39/Q111
890.8
852.3


(2:1:1)




H4FEAA-Fc-MDNA413L39/Q111 (2:1:2)
1119.0
No Inhibition









TF-1 Assay

TF-1 cells were cultured in RPMI media (ATCC) supplemented with 10% FBS (Gibco) and 2 ng/mL GM-CSF (ThermoFisher). TF-1 cells were harvest and washed twice with PBS before being placed into media lacking GM-CSF. Cells were added at 30,000 cells per well on the day of the experiment in a volume of 100 μL. To perform the antagonist assay, the TF-1 proliferation assay, as outlined above, was setup using rhIL13 EC50 or EC80 in the proliferation assay. Reactions were also supplemented with increasing concentrations of the anti-IL13Rα1 antibody or constructs. Controls included in the plate consisted of 1) cells grown in complete media, 2) cells grown in un-supplemented media (no IL13 or GM-CSF), 3) cells supplemented with EC50 or EC80 rhIL13, and cells treated with an IL13 standard curve. Triplicate wells were treated under each condition. Cells were incubated for 96 hours and developed with CyQUANT. For development a 3× solution of the CyQUANT assay reagent was prepared according the manufacturer's instructions and 100 μL of this solution was added to each well. Plates were incubated for 1 hour at 37° C., mixed and then centrifuged to settle the cells. Plates were read at 485Ex/535Em.


All constructs showed dose dependent inhibition of TF-1 proliferation with MDNA019FEAAC125-Fc-MDNA413R39/Q111 (2:1:2) showing maximum inhibition while Fc-MDNA413L39/Q111 showing the least. MDNA019FEAAC125-Fc-MDNA413R39/Q111 (2:1:2) showed inhibition of TF-1 proliferation at both EC50 and EC80 of rhIL13 whereas, Fc-MDNA413L39/Q111 (1:2) and Fc-MDNA413L39/R111 (1:2) showed significant inhibition at only EC50 (FIG. 51).


TF-1 cells were harvest and washed twice with PBS before being placed into media lacking GM-CSF. Cells were added at 30,000 cells per well on the day of the experiment in a volume of 100 μL. To perform the antagonist assay, the TF-1 proliferation assay was setup using rhIL13 EC80 in the proliferation assay.


MDNA109FEAAC125-Fc-MDNA413R39/Q111 showed dose dependent inhibition of TF-1 proliferation with an IC50 of 37 nM (FIG. 71).


pSTAT5 Assay in Human PBMCs


PBMCs were isolated over a density gradient, allowed to rest in complete media, and then stimulated for 15 minutes with IL2, MDNA109FEAA-Fc or MDNA109FEAAC125-Fc-MDNA413R39/Q111. Controls included non-stimulated PBMC cells. Cells were fixed immediately following stimulation, and samples analyzed by flow cytometry after intracellular staining for phosphorylated STAT5 (P-STAT5) in the immune subsets, Naïve CD8 T cells (CD8+CD25−), NK cells and Tregs (FIG. 52).


MDNA109FEAAC125-Fc-MDNA413R39/Q111 human anti-PD1-MDNA109FEAAS125 mouse anti-PD1-MDNA109FEAAS125 are able to activate signaling in naïve CD8 T cells and NK cells with a higher potency than rhIL-2. However, the potency is greatly reduced for pSTAT5 signaling in Tregs in comparison to rhIL-2. These data show that MDNA109FEAAC125-Fc-MDNA413R39/Q111 human anti-PD1-MDNA109FEAAS125, mouse anti-PD1-MDNA109FEAAS125 are superior to rhIL-2 at activating immune effectors cell response required for achieve anti-cancer efficacy. The construct H4FEAA-Fc-MDNA413L39/Q111 (2:1:2) is not potent though in inducing pSTAT5 induction in human PBMCs.


SPR Study

Human IgG (Fc) Capture Chip principle: (1) Immobilization: Immobilized Anti-Human IgG (Fc) antibody on CM5 chip; (2) Capture: capture the IL-13 Protein, Fc Tag or sample; (3) Analyte: the analyte is Human IL-13 R alpha 1 Protein or Human IL-13 R alpha 2 Protein; and (4) Regeneration: 3 M magnesium chloride (FIG. 53).


The binding to IL13Rα1 and IL13Rα2 is shown in FIG. 72 and Table 32.









TABLE 32







Binding affinity KD values of the molecules for IL13Ra1 and IL13Ra2










IL13Ra1, KD (nM)
IL13Ra2, KD (nM)





Human anti-PD1-
No binding
1.4 


MDNA132L39/Q111 (1:1 KIH)




Mouse anti-PD1-
No binding
1.56


MDNA132L39/Q111(1:1 KIH)









The binding to CD3 epitopes, CD3epsilon/delta is shown in FIG. 73 and Table 33. The results showed that human and mouse anti-CD3-MDNA132 bind to human and mouse CD3 epitopes respectively with no cross reactivity.









TABLE 33







Binding affinity KD values of the molecules for CD3 epitopes










Human CD3 epsilon delta
Mouse CD3 epsilon delta



heterodimer, KD (nM)
heterodimer, KD (nM)





Human anti-CD3-
1530
No binding


MDNA132L39/Q111




(1:1 KIH)




Mouse anti-CD3-
No binding
50.9


MDNA132L39/Q111




(1:1 KIH)









The binding to IL2R alpha (CD25) and beta (CD122) is shown in FIG. 74 and Table 34.









TABLE 34







Binding affinity KD values of the molecules for IL2Rα and IL2Rβ










IL2Rα, KD (nM)
IL2Rβ, KD (nM)





MDNA109FEAAC125-Fc-
No binding
 9.3


MDNA413R39/Q111 (2:1:2)




Mouse anti-PD1-MDNA109FEAAS125
No binding
11.3


(1:1 KIH)









The binding to PD1 is shown in FIG. 75 and Table 35.









TABLE 35







Binding affinity KD values of the molecules for PD1










Human PD1
Mouse PD1



(nM)
(nM)





Human anti-PD1-MDNA132L39/Q111 (1:1
4.36
No binding


KIH)




Mouse anti-PD1-MDNA132L39/Q111 (1:1
No binding
41.4


KIH)




Human anti-PD1-MDNA413R39/Q111 (1:2)
4.89
No binding


Mouse anti-PD1-MDNA109FEAAS125
No binding
102


(1:1 KIH)











Example 8: Inhibition of IL-4 and IL-13 Induced M2 Polarization

Experimental Protocol:

    • PBMCs were isolated from whole blood.
    • CD14+ monocytes were separated using magnetic selection and differentiated into M0 macrophage in the presence of AB serum over 6 days using M-CSF.
    • On day 6, macrophages were polarised towards M2 using the EC80 value of either IL-13 or IL-4 in the presence of a reference or test antagonist (10 point 3-fold dilution).
    • Following treatment, macrophages were collected for phenotyping by flow cytometry, with Fc receptors blocked prior to antibody staining.


The inhibition of IL-4 induced M2 polarization is shown in FIG. 76 and Table 36.



FIG. 76A shows phenotyping of IL-4 treated macrophages in the presence of Fc-MDNA413R39/Q111(1:2); FIG. 76B shows phenotyping of IL-4 treated macrophages in the presence of Fc4-MDNA413R39/Q111(1:2); FIG. 76C shows phenotyping of IL-4 treated macrophages in the presence of MDNA109FEAAC125-Fc-MDNA413R39/Q111(2:1:2); FIG. 76D shows phenotyping of IL-4 treated macrophages in the presence of hu anti-PD1-MDNA413R39/Q111(1:2); and FIG. 76E shows phenotyping of IL-4 treated macrophages in the presence of mouse anti-PD1-MDNA413R39/Q111(1:2). In each case, dotted lines represent: IL-4 only control, dashed lines represent: M0 control.









TABLE 36







Inhibition of IL-4 Mediated M2 Polarization (IC50 values from 2 donors


separated by comma)










IL-4 Mediated M2
IL-4 Mediated M2



polarization
polarization


Construct
CD206 (IC50 nM)
CD209 (IC50 nM)





Fc-MDNA413R39/Q111 (1:2)
ND, 1.7 
<0.01, 3.28 


Fc4-MDNA413R39/Q111(1:2)
ND, 2.01
0.59, 2.3 


MDNA109FEAAC125-Fc-
3.8, 3.1
6.3, 4.2


MDNA413R39/Q111(2:1:2)




Human anti-PD1-
2.0, 6.2
7.6, 5.2


MDNA413R39/Q111(1:2)




Mouse anti-PD1-
 4.5, 5.97
7.29, 5.37


MDNA413R39/Q111(1:2)









The inhibition of IL-13 induced M2 polarization is shown in FIG. 77 and Table 37.



FIG. 77A shows phenotyping of IL-13 treated macrophages in the presence of Fc-MDNA413R39/Q111 (1:2); FIG. 77B shows phenotyping of IL-13 treated macrophages in the presence of Fc4-MDNA413R39/Q111(1:2); FIG. 77C shows phenotyping of IL-13 treated macrophages in the presence of MDNA109FEAAC125-FcMDNA413R39/Q111(2:1:2); FIG. 77D shows phenotyping of IL-13 treated macrophages in the presence of hu anti-PD1-MDNA413R39/Q111(1:2); and FIG. 77E shows phenotyping of IL-13 treated macrophages in the presence of mouse anti-PD1-MDNA413R39/Q111(1:2). In each case, dotted lines represent: IL-13 only control, dashed lines represent: M0 control.









TABLE 37







Inhibition of IL-13 Mediated M2 Polarization (IC50 values from 2 donors


separated by comma)










IL-13 Mediated M2
IL-13 Mediated M2



polarization
polarization


Construct
CD206 (IC50 nM)
CD209 (IC50 nM)





Fc-MDNA413R39/Q111(1:2)
 ND, 12.78
0.036, 11.97


Fc4-MDNA413R39/Q111(1:2)
0.48, ND
 1.60, 0.492


MDNA109FEAAC125-Fc-
1.45, 2.48
0.49, 2.4 


MDNA413R39/Q111(2:1:2)




Human anti-PD1-
ND, 13.2
 6.7, 12.1


MDNA413R39/Q111(1:2)




Mouse anti-PD1-
ND, 10.8
 6.2, 7.05


MDNA413R39/Q111(1:2)









The examples set forth above are provided to give those of ordinary skill in the art a complete disclosure and description of how to make and use the embodiments of the compositions, systems and methods of the invention, and are not intended to limit the scope of what the inventors regard as their invention. Modifications of the above-described modes for carrying out the invention that are obvious to persons of skill in the art are intended to be within the scope of the following claims. All patents and publications mentioned in the specification are indicative of the levels of skill of those skilled in the art to which the invention pertains. All references cited in this disclosure are incorporated by reference to the same extent as if each reference had been incorporated by reference in its entirety individually.


All headings and section designations are used for clarity and reference purposes only and are not to be considered limiting in any way. For example, those of skill in the art will appreciate the usefulness of combining various aspects from different headings and sections as appropriate according to the spirit and scope of the invention described herein.


All references cited herein are hereby incorporated by reference herein in their entireties and for all purposes to the same extent as if each individual publication or patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety for all purposes.


Many modifications and variations of this application can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. The specific embodiments and examples described herein are offered by way of example only, and the application is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which the claims are entitled.

Claims
  • 1. A bispecific IL-2 cytokine fusion, comprising an IL-2 mutein fused to a second cytokine, wherein the IL-2 mutein comprises the following amino acid substitutions: L80F, R81D, L85V, I86V, and I92F, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.
  • 2. (canceled)
  • 3. The bispecific IL-2 cytokine fusion of claim 1, wherein the IL-2 mutein further comprises F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.
  • 4. The bispecific IL-2 cytokine fusion of claim 1, wherein the IL-2 mutein further comprises K43N substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.
  • 5. The bispecific IL-2 cytokine fusion of claim 1, wherein the IL-2 mutein further comprises Y45A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.
  • 6. The bispecific IL-2 cytokine fusion of claim 1, wherein the IL-2 mutein further comprises E62A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.
  • 7. The bispecific IL-2 cytokine fusion of claim 1, wherein the IL-2 mutein further comprises an E62A substitution and an F42A substitution, wherein numbering is in accordance with the wild-type human IL-2 of SEQ ID NO:2.
  • 8. The bispecific IL-2 cytokine fusion of claim 1, wherein the IL-2 mutein is an IL-2 mutein sequence as described in Tables 2, 4, 5, and/or 6.
  • 9. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion comprises a sequence as described in Tables 2, 4, 5, 6, 10, and/or 12, and/or FIG. 54.
  • 10. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion further comprises an Fc antibody fragment.
  • 11. The bispecific IL-2 cytokine fusion of claim 10, wherein the Fc antibody fragment is a human Fc antibody fragment.
  • 12. The bispecific IL-2 cytokine fusion of claim 10, wherein the Fc antibody fragment comprises a N297A substitution.
  • 13. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion further comprises albumin.
  • 14. The bispecific IL-2 cytokine fusion of claim 1, wherein the second cytokine is selected from the group consisting of IL-4, IL-13, IL-10, IL-12, IL15, and IL-18.
  • 15. The bispecific IL-2 cytokine fusion of claim 1, wherein the second cytokine is IL-4 or IL-13.
  • 16. The bispecific IL-2 cytokine fusion of claim 1, where the second cytokine is as described in Table 7 and/or Table 8 and/or Table 12 and/or Table 28 and/or FIG. 54.
  • 17. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion exhibits increased binding capacity for IL-2Rβ as compared to wild-type human IL-2.
  • 18. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion exhibits a greater binding affinity for IL-2Rβ as compared to wild-type human IL-2.
  • 19. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion exhibits abrogated IL2Ra binding (i.e., does not significantly bind to IL2Rα).
  • 20. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion exhibits decreased binding affinity for CD25 as compared to wild-type human IL-2.
  • 21. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion comprises SEQ ID NO:146, SEQ ID NO:147, SEQ ID NOs:148 and 213, SEQ ID NOs:149 and 213, SEQ ID NO:150, SEQ ID NOs:151 and 214, SEQ ID NO:152, SEQ ID NO:153, SEQ ID NO:154, SEQ ID NO:155, SEQ ID NO:156, SEQ ID NOs:157 and 213, SEQ ID NO:158, SEQ ID NO:159, SEQ ID NO:160, SEQ ID NOs:161 and 215, SEQ ID NOs:162 and 216, SEQ ID NO:163, SEQ ID NO:164, SEQ ID NOs:165 and 213, SEQ ID NOs:166 and 213, or SEQ ID NOs:167 and 217, or a sequence from Table 12 or Table 28 or FIG. 54.
  • 22. The bispecific IL-2 cytokine fusion of claim 1, wherein the bispecific IL-2 cytokine fusion comprises MDNA413-Fc-MDNA109, MDNA109FEAA-Fc-MDNA413, Fc-MDNA132 (1:1 KiH), Fc-A11 (1:2), Fc-A11 (1:2), Fc-MDNA413 (1:2), Fc-MDNA413 (1:2), Fc4-MDNA413 (1:2), MDNA413-Fc (1:1 KIH), MDNA109-Fc (2:1), MDNA-109FEAA-Fc (2:1), Fc-MDNA109 (1:2), MDNA109FEAA-Fc-MDNA132 (2:1:1 KiH), MDNA413-Fc-MDNA132 (2:1:1 KiH), MDNA109FEAA-Fc-MDNA413 (2:1:2)—version 1, MDNA109FEAA-Fc-MDNA413 (2:1:2)—version 2, MDNA132-Fc-MDNA109 (1:1:1 KIH), MDNA413-Fc-MDNA109 (1:1:1 KIH), MDNA132-Fc-MDNA109FEAA (1:1:1 KIH), or MDNA109-Fc-MDNA413.
  • 23. A method of treating cancer comprising administering a bispecific IL-2 cytokine fusion of claim 1.
  • 24. The method of claim 23, wherein the cancer is selected from the group consisting of prostate cancer, ovarian cancer, breast cancer, endometrial cancer, multiple myeloma, melanoma, lymphomas, lung cancers including small cell lung cancer, kidney cancer, liver cancer, colon cancer, colorectal cancer, pancreatic cancer, gastric cancer, and brain cancer.
  • 25. The method of claim 24, wherein the cancer is colon cancer.
  • 26. A method of treating cancer comprising administering a combination treatment comprising: (i) an anti-PD-1 antibody or inhibitor or an anti-PD-L1 antibody or inhibitor, and(ii) a bispecific IL-2 cytokine fusion of claim 1.
  • 27. The method of claim 26, wherein the anti-PD-1 antibody or inhibitor is selected from the group consisting of nivolumab, BMS-936558, MDX-1106, ONO-4538, AMP224, CT-011, and MK-3475(pembrolizumab), cemiplimab (REGN2810), SHR-1210 (CTR20160175 and CTR20170090), SHR-1210 (CTR20170299 and CTR20170322), JS-001 (CTR20160274), IBI308 (CTR20160735), BGB-A317 (CTR20160872) and a PD-1 antibody as recited in Table 38.
  • 28. The method of claim 27, wherein the anti-PD-L1 antibody or inhibitor is selected from the group consisting of atezolizumab, avelumab, and Durvalumab.
  • 29. The method of claim 26, wherein the cancer is selected from the group consisting of prostate cancer, ovarian cancer, breast cancer, endometrial cancer, multiple myeloma, melanoma, lymphomas, lung cancers including small cell lung cancer, kidney cancer, liver cancer, colon cancer, colorectal cancer, pancreatic cancer, gastric cancer, and brain cancer.
  • 30. The method of claim 27, wherein the cancer is colon cancer.
  • 31. A pharmaceutical composition comprising a bispecific IL-2 cytokine fusion of claim 1, and a pharmaceutically acceptable carrier.
  • 32. A pharmaceutical composition comprising an anti-PD-1 antibody or inhibitor, a bispecific TL-2 cytokine fusion of claim 1, and a pharmaceutically acceptable carrier.
Parent Case Info

This application is a 371 National Phase of PCT/CA21/50872 filed Jun. 24, 2021, which claims priority under 35 U.S.C. § 119 to U.S. Patent Application Nos. 63/043,227, filed on Jun. 24, 2020, 63/093,724, filed on Oct. 19, 2020, 63/094,322, filed on Oct. 20, 2020, 63/165,600, filed on Mar. 24, 2021, and 63/165,738, filed on Mar. 24, 2021, all of which are expressly incorporated herein by reference in their entireties.

PCT Information
Filing Document Filing Date Country Kind
PCT/CA21/50872 6/24/2021 WO
Provisional Applications (5)
Number Date Country
63165738 Mar 2021 US
63165600 Mar 2021 US
63094322 Oct 2020 US
63093724 Oct 2020 US
63043227 Jun 2020 US