HEAT-INACTIVATED VACCINIA VIRUS AS A VACCINE IMMUNE ADJUVANT

Abstract
The technology of the present disclosure relates to the use of Heat-inactivated modified vaccinia Ankara (MVA) vims (Heat-iMVA) or Heat-inactivated vaccinia vims as a vaccine immune adjuvant. In particular, the present technology relates to the use of Heat-iMVA as a vaccine adjuvant for tumor antigens in cancer vaccines alone or in combination with immune checkpoint blockade (ICB) antibodies for use as a cancer immunotherapeutic.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Dec. 7, 2018, is named 115872-0795 SL.txt and is 233,215 bytes in size.


TECHNICAL FIELD

The technology of the present disclosure relates generally to the fields of oncology, virology, and immunotherapy. The present technology relates to the use of Heat-inactivated vaccinia virus as a vaccine immune adjuvant. In particular, the present technology relates to the use of Heat-inactivated modified vaccinia Ankara (MVA) virus or “Heat-iMVA” as a vaccine adjuvant for tumor antigens in cancer vaccines alone or in combination with immune checkpoint blockade (ICB) antibodies for use as a cancer immunotherapeutic.


BACKGROUND

The following description is provided to assist the understanding of the reader. None of the information provided or references cited is admitted to be prior art to the present technology.


Malignant tumors are inherently resistant to conventional therapies and present significant therapeutic challenges. Immunotherapy is an evolving area of research and an additional option for the treatment of certain types of cancers. The immunotherapy approach rests on the rationale that the immune system may be stimulated to identify tumor cells, and target them for destruction. Despite presentation of antigens by cancer cells and the presence of immune cells that could potentially react against tumor cells, in many cases, the immune system is not activated or is affirmatively suppressed. Key to this phenomenon is the ability of tumors to protect themselves from immune response by coercing cells of the immune system to inhibit other cells of the immune system. Tumors develop a number of immunomodulatory mechanisms to evade antitumor immune responses. Thus, improved immunotherapeutic approaches are needed to enhance host antitumor immunity and target tumor cells for destruction.


SUMMARY

In one aspect, the present disclosure provides a method for treating a solid tumor in a subject in need thereof, the method comprising administering to the subject an immunogenic composition comprising an antigen and a therapeutically effective amount of an adjuvant comprising an inactivated modified vaccinia Ankara virus and/or an inactivated vaccinia virus. In some embodiments, the inactivated modified vaccinia Ankara virus is either a Heat-inactivated modified vaccinia Ankara virus (Heat-iMVA) or a UV-inactivated MVA, and the inactivated vaccinia virus is either a Heat-inactivated vaccinia virus or a UV-inactivated vaccinia virus. In some embodiments, the inactivated modified vaccinia virus is Heat-iMVA.


In some embodiments of the methods disclosed herein, the antigen is selected from the group consisting of tumor differentiation antigens, cancer testis antigens, neoantigens, viral antigens in the case of tumors associated with oncogenic virus infection, GPA33, HER2/neu, GD2, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, MUM-1, CDK4, N-acetylglucosaminyltransferase, p15, gp75, beta-catenin, ErbB2, cancer antigen 125 (CA-125), carcinoembryonic antigen (CEA), RAGE, MART (melanoma antigen), MUC-1, MUC-2, MUC-3, MUC-4, MUC-5ac, MUC-16, MUC-17, tyrosinase, tyrosinase-related proteins 1 and 2, Pmel 17 (gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate cancer psm, PRAME (melanoma antigen), β-catenin, EBNA (Epstein-Barr Virus nuclear antigen) 1-6, p53, kras, lung resistance protein (LRP) Bc1-2, prostate specific antigen (PSA), Ki-67, CEACAM6, colon-specific antigen-p (CSAp), NY-ESO-1, human papilloma virus E6 and E7, and combinations thereof. In some embodiments, the antigen comprises a neoantigen selected from the group consisting of M27 (REGVELCPGNKYEMRRHGTTHSLVIHD) (SEQ ID NO: 4), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL) (SEQ ID NO: 5), M48 (SHCHWNDLAVIPAGVVHNWDFEPRKVS) (SEQ ID NO: 6), and combinations thereof.


In some embodiments of the methods disclosed herein, the administration step comprises administering the immunogenic composition in one or more doses.


In some embodiments, the methods disclosed herein further comprise administering to the subject an immune checkpoint blockade agent selected from the group consisting of cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors, programmed death 1 (PD-1) inhibitors, PD-L1 inhibitors, and PD-L2 inhibitors.


In some embodiments, the immunogenic composition is delivered to the subject separately, sequentially, or simultaneously with the administration of the immune checkpoint blockade agent.


In some embodiments, the PD-L1 inhibitor is an anti-PD-L1 antibody.


In some embodiments, treatment comprises one or more of the following: inducing an immune response in the subject against the tumor or enhancing or promoting an ongoing immune response against the tumor in the subject, reducing the size of the tumor, eradicating the tumor, inhibiting growth of the tumor, inhibiting metastatic growth of the tumor, inducing apoptosis of the tumor cells, or prolonging survival of the subject.


In some embodiments, the induction, enhancement, or promotion of the immune response comprises one or more of the following: increased levels of interferon gamma (IFN-γ) expression in T-cells in the spleen, draining lymph nodes, and/or serum as compared to an untreated control sample; increased levels of antigen-specific T-cells in the spleen, draining lymph nodes, and/or serum as compared to an untreated control sample; and increased levels of antigen-specific immunoglobulin in serum as compared to an untreated control sample. In some embodiments, the antigen-specific immunoglobulin is IgG1 or IgG2.


In some embodiments, the immunogenic composition is formulated to be administered intratumorally, intramuscularly, intradermally, or subcutaneously.


In some embodiments, the tumor is selected from the group consisting of melanoma, colorectal cancer, breast cancer, prostate cancer, lung cancer, pancreatic cancer, ovarian cancer, squamous cell carcinoma of the skin, Merkel cell carcinoma, gastric cancer, liver cancer, and sarcoma.


In some embodiments, the inactivated modified vaccinia Ankara virus or inactivated vaccinia virus is administered at a dosage per administration of about 105 to about 1010 plaque-forming units (pfu).


In some embodiments of the methods disclosed herein, the subject is human.


In one aspect, the present disclosure provides an immunogenic composition comprising an antigen and an adjuvant comprising an inactivated modified vaccinia Ankara virus and/or an inactivated vaccinia virus. In some embodiments, the inactivated modified vaccinia Ankara virus is either a Heat-inactivated modified vaccinia Ankara virus (Heat-iMVA) or a UV-inactivated MVA, and the inactivated vaccinia virus is either a Heat-inactivated vaccinia virus or a UV-inactivated vaccinia virus. In some embodiments, the inactivated modified vaccinia virus is Heat-iMVA.


In some embodiments, the immunogenic compositions of the present technology further comprise a pharmaceutically acceptable carrier.


In some embodiments, the antigen of the immunogenic compositions of the present technology is selected from the group consisting of tumor differentiation antigens, cancer testis antigens, neoantigens, viral antigens in the case of tumors associated with oncogenic virus infection, GPA33, HER2/neu, GD2, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, MUM-1, CDK4, N-acetylglucosaminyltransferase, p15, gp75, beta-catenin, ErbB2, cancer antigen 125 (CA-125), carcinoembryonic antigen (CEA), RAGE, MART (melanoma antigen), MUC-1, MUC-2, MUC-3, MUC-4, MUC-5ac, MUC-16, MUC-17, tyrosinase, tyrosinase-related proteins 1 and 2, Pmel 17 (gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate cancer psm, PRAME (melanoma antigen), β-catenin, EBNA (Epstein-Barr Virus nuclear antigen) 1-6, p53, kras, lung resistance protein (LRP) Bc1-2, prostate specific antigen (PSA), Ki-67, CEACAM6, colon-specific antigen-p (CSAp), NY-ESO-1, human papilloma virus E6 and E7, and combinations thereof. In some embodiments, the antigen comprises a neoantigen selected from the group consisting of M27 (REGVELCPGNKYEMRRHGTTHSLVIHD) (SEQ ID NO: 4), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL) (SEQ ID NO: 5), M48 (SHCHWNDLAVIPAGVVHNWDFEPRKVS) (SEQ ID NO: 6), and combinations thereof.


In some embodiments, the immunogenic compositions of the present technology further comprise an immune checkpoint blockade agent selected from the group consisting of cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors, programmed death 1 (PD-1) inhibitors, PD-L1 inhibitors, and PD-L2 inhibitors. In some embodiments, the PD-L1 inhibitor is an anti-PD-L1 antibody.


In some embodiments, of the immunogenic compositions of the present technology, the inactivated modified vaccinia Ankara virus or inactivated vaccinia virus is administered at a dosage per administration of about 105 to about 1010 plaque-forming units (pfu).


In one aspect, the present disclosure provides a kit comprising instructions for use, a container means, and a separate portion of each of: (a) an antigen; and (b) an adjuvant comprising inactivated modified vaccinia Ankara virus and/or inactivated vaccinia virus. In some embodiments, the inactivated modified vaccinia Ankara virus is either a Heat-inactivated modified vaccinia Ankara virus (Heat-iMVA) or a UV-inactivated MVA, and the inactivated vaccinia virus is either a Heat-inactivated vaccinia virus or a UV-inactivated vaccinia virus. In some embodiments, the inactivated modified vaccinia virus is Heat-iMVA. In some embodiments, the antigen is selected from the group consisting of tumor differentiation antigens, cancer testis antigens, neoantigens, viral antigens in the case of tumors associated with oncogenic virus infection, GPA33, HER2/neu, GD2, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, MUM-1, CDK4, N-acetylglucosaminyltransferase, p15, gp75, beta-catenin, ErbB2, cancer antigen 125 (CA-125), carcinoembryonic antigen (CEA), RAGE, MART (melanoma antigen), MUC-1, MUC-2, MUC-3, MUC-4, MUC-5ac, MUC-16, MUC-17, tyrosinase, tyrosinase-related proteins 1 and 2, Pmel 17 (gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate cancer psm, PRAME (melanoma antigen), β-catenin, EBNA (Epstein-Barr Virus nuclear antigen) 1-6, p53, lung resistance protein (LRP) Bcl-2, prostate specific antigen (PSA), Ki-67, CEACAM6, colon-specific antigen-p (CSAp), NY-ESO-1, and combinations thereof. In some embodiments, the antigen comprises a neoantigen selected from the group consisting of M27 (REGVELCPGNKYEMRRHGTTHSLVIHD) (SEQ ID NO: 4), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL) (SEQ ID NO: 5), M48 (SHCHWNDLAVIPAGVVHNWDFEPRKVS) (SEQ ID NO: 6), and combinations thereof. In some embodiments, the kit further comprises an immune checkpoint blockade agent selected from the group consisting of cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors, programmed death 1 (PD-1) inhibitors, PD-L1 inhibitors, and PD-L2 inhibitors. In some embodiments, the immune checkpoint blockade agent comprises a PD-L1 inhibitor, which is an anti-PD-L1 antibody.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-1K are a series of graphs showing antigen-specific T-cell and antibody responses after intramuscular (IM) vaccination of C57BL/6J mice with chicken ovalbumin (OVA) in the presence or absence of the immune adjuvant heat-inactivated MVA (Heat-iMVA). FIG. 1A: OVA intramuscular vaccination strategy. On day 0 and day 14, mice were intramuscularly injected with OVA (10 μg/mouse)+/−Heat-iMVA (an equivalent amount of 107 pfu/mouse). Spleens, lymph nodes and serum were collected on day 21. FIG. 1B and FIG. 1D: Splenocytes were stimulated with OVA257-264(SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml) for 12 h. The expression of IFN-γ by CD8+ T-cells was measured by flow cytometry. FIG. 1D: Dot plots of IFN-γ+CD8+ T-cells in the spleens of PBS, OVA, or OVA+Heat-iMVA-vaccinated mice. FIG. 1C and FIG. 1E: Splenocytes were stimulated with OVA323-339 (ISQAVHAAHAEINEAGR) peptide (SEQ ID NO: 2) (10 μg/ml) for 12 h. The expression of IFN-γ by CD4+ T-cells was measured by flow cytometry. FIG. 1E: Dot plots of IFN-γ+CD4+ T-cells in the spleens of PBS, OVA, or OVA+Heat-iMVA-vaccinated mice. FIG. 1F and FIG. 1H: Cells from the draining lymph nodes (dLNs) were stimulated with OVA257-264 (SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml) for 12 h. The expression of IFN-γ by CD8+ T-cells was measured by flow cytometry. FIG. 1G and FIG. 1I: Cells from the dLNs were stimulated with OVA323-339 (ISQAVHAAHAEINEAGR) peptide (SEQ ID NO: 2) (10 μg/ml) for 12 h. The expression of IFN-γ by CD4+ T-cells was measured by flow cytometry. FIG. 1J and FIG. 1K: OVA-specific immunoglobulin G1 (IgG1) or OVA-specific immunoglobulin G2c (IgG2c) titers in the serum from PBS, OVA, or OVA+Heat-iMVA-vaccinated mice were determined by ELISA (*p<0.05; **p<0.01; ***p<0.001; n=5).



FIGS. 2A-2G are a series of graphs showing antigen-specific T-cell and antibody responses after intramuscular (IM) or subcutaneous (SC) vaccination with OVA+/−Heat-iMVA or complete Freund adjuvant (CFA) in C57BL/6J mice. FIG. 2A: OVA vaccination strategy. On day 0 and day 14, mice were intramuscularly or subcutaneously injected with OVA (10 μg/mouse)+/−Heat-iMVA (an equivalent amount of 107 pfu/mouse). Spleens, lymph nodes and serum were collected on day 21. FIG. 2B: Splenocytes were stimulated with OVA257-264 (SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml) for 12 h. The expression of IFN-γ by CD8+ T-cells was measured by flow cytometry. FIG. 2C: Splenocytes were stimulated with OVA323-339 (ISQAVHAAHAEINEAGR) peptide (SEQ ID NO: 2) (10 μg/ml) for 12 h. The expression of IFN-γ by CD4+ T-cells was measured by flow cytometry. FIG. 2D: Cells from the dLNs were stimulated with OVA257-264(SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml) for 12 h. The expression of IFN-γ by CD8+ T-cells was measured by flow cytometry. FIG. 2E: dLNs were stimulated with OVA323-339 (ISQAVHAAHAEINEAGR) peptide (SEQ ID NO: 2) (10 μg/ml) for 12 h. The expression of IFN-γ by CD4+ T-cells was measured by flow cytometry. FIG. 2F and FIG. 2G: OVA-specific immunoglobulin G1 (IgG1) or OVA-specific immunoglobulin G2c (IgG2c) titers in the serum from PBS, OVA, or OVA+Heat-iMVA, or OVA+CFA-vaccinated mice were determined by ELISA (*p<0.05; **p<0.01; ***p<0.001; ns: not significant; n=5 except for the PBS group).



FIGS. 3A-3G are a series of graphs showing T-cell and antibody responses after subcutaneous (SC) vaccination with OVA+/−Heat-iMVA in C57BL/6J, STINGGt/Gt and Batf3−/− mice. FIG. 3A: OVA vaccination strategy. On day 0 and day 14, mice were subcutaneously injected with OVA (10 μg/mouse) plus Heat-iMVA (an equivalent amount of 107 pfu/mouse). Spleens, lymph nodes, and serum were collected on day 21. FIG. 3B: Splenocytes were stimulated with OVA257-264(SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml). The expression of IFN-γ by CD8+ T-cells was measured by flow cytometry. FIG. 3C: Splenocytes were stimulated with OVA323-339 (ISQAVHAAHAEINEAGR) peptide (SEQ ID NO: 2) (10 μg/ml). The expression of IFN-γ by CD4+ T-cells was measured by flow cytometry. FIG. 3D: Cells from the dLNs were stimulated with OVA257-264 (SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml). The expression of IFN-γ by CD8+ T-cells was measured by flow cytometry. FIG. 3E: Cells from the dLNs were stimulated with OVA323-339 (ISQAVHAAHAEINEAGR) peptide (SEQ ID NO: 2) (10 μg/ml). The expression of IFN-γ by CD4+ T-cells was measured by flow cytometry. FIG. 3F and FIG. 3G: OVA-specific immunoglobulin G1 (IgG1) or OVA-specific immunoglobulin G2c (IgG2c) titers in the serum from OVA+Heat-iMVA-vaccinated WT, STINGGt/Gt and Batf3−/− mice were determined by ELISA (*p<0.05; **p<0.01; ***p<0.001; ns: not significant; n=5 except for the PBS group).



FIGS. 4A-4C are a series of graphs showing T-cell and antibody responses after skin scarification vaccination with MVA-OVA+/−Heat-iMVA in C57BL/6J, STINGGt/Gt and Batf3−/− mice. FIG. 4A: MVA-OVA vaccination strategy. On day 0, C57BL/6J mice were vaccinated with different doses of MVA-OVA (105, 106, 107 pfu/mouse) in the presence or absence of Heat-iMVA (an equivalent of 105 pfu/mouse). Spleens, lymph nodes, and serum were harvested from euthanized mice one week later. With respect to STINGGt/Gt and Batf3−/− mice, they were vaccinated with MVA-OVA (106 pfu/mouse). FIG. 4B: Splenocytes were co-cultured with MVA-OVA infected BMDCs. Expression of IFN-γ by CD8+ T-cells is measured by flow cytometry. FIG. 4C: Splenocytes were co-cultured with OVA257-264(SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml) pulsed BMDCs for 12 h. The expression of IFN-γ by CD8+ T-cells is measured by flow cytometry. (*p<0.05; **p<0.01; ns: not significant; n=5 except for the PBS group).



FIGS. 5A-5D are a series of graphs showing cell surface MHC-I (H-2Kb) expression of GM-CSF-cultured bone marrow-derived dendritic cells (BMDCs) and their capacity for uptake of fluorescent-labeled model antigen OVA (OVA-647) after Heat-iMVA treatment. FIG. 5A: BMDCs were incubated with OVA (1 mg/ml)+/−Heat-iMVA (MOI of 1) or poly IC (5 μg/ml) for 16 h. Then, the cell surface H-2Kb expression was determined by FACS using anti-H-2Kb antibody. FIG. 5B: The mean fluorescence intensities of H2-Kb of BMDCs are shown. FIG. 5C: BMDC were infected with Heat-iMVA (MOI of 1) for 1 h and then incubated with OVA-647 (0.5 mg/ml) for 1 h. The fluorescence intensities of phagocytosed OVA-647 in BMDC were measured by flow cytometry. FIG. 5D: BMDC were infected with Heat-iMVA (MOI of 1) for 16 h and then incubated with OVA-647 (0.5 mg/ml) for 1 h. The fluorescence intensities of phagocytosed OVA-647 in BMDCs were determined by flow cytometry.



FIGS. 6A-6B are a series of graphs showing the proliferation of Carboxyfluorescein Diacetate Succinimidyl Ester (CFSE)-labeled OT-I T-cells after incubation with GM-CSF-cultured BMDCs pulsed with OVA+/−Heat-iMVA. BMDCs were incubated with OVA (0.1, 0.2, 0.5 mg/ml)+/−Heat-iMVA (MOI of 1) for 3 h and then washed and co-cultured with CFSE-labeled OT-I cells for 3 days (BMDC:OT-I T-cells=1:5). Flow cytometry was applied to measure CFSE intensities of OT-I cells. FIG. 6A: CFSE of OT-I T-cells incubated with BMDCs pulsed with OVA alone. FIG. 6B CFSE of OT-1 cells incubated with BMDCs pulsed with OVA and Heat-iMVA.



FIGS. 7A-7B are a series of graphs showing the proliferation of CFSE labeled OT-II T-cells after incubation with GM-CSF-cultured BMDCs pulsed with OVA in the presence or absence of Heat-iMVA. BMDCs were incubated with OVA (0, 0.1, 0.2, 0.5 mg/ml)+/−Heat-iMVA (MOI of 1) or poly IC (5 μg/ml) for 3 h, then washed and co-cultured with CFSE-labeled OT-II cells for 3 days (BMDC:OT-II T-cells=1:5). Flow cytometry was applied to measure CFSE intensities of OT-II cells. FIG. 7A: CFSE of OT-II T-cells incubated with BMDCs pulsed with OVA alone. FIG. 7B: CFSE of OT-I cells incubated with BMDCs pulsed with OVA plus Heat-iMVA or poly IC.



FIG. 8 is a series of graphs showing the proliferation of CFSE-labeled OT-I cells after incubation with FMS-like tyrosine kinase 3 ligand (Flt3L)-cultured BMDCs from C57B/6J pulsed with OVA in the presence or absence of Heat-iMVA. Bone marrow cells were differentiated in cell culture medium in the presence of Flt3L (100 ng/ml) for 9 days. Flt3L-cultured BMDCs were incubated with OVA (0.01, 0.03 mg/ml)+/−Heat-iMVA (MOI of 1) for 3 h, then co-cultured with CFSE-labeled OT-I cells for 3 days (BMDC:OT-I=1:5). Flow cytometry was applied to measure CFSE intensities of OT-I cells.



FIGS. 9A-9C are a series of graphs showing murine pDCs are important for Heat-iMVA-elicited vaccine adjuvant effects. FIG. 9A: OVA vaccination strategy with or without Heat-iMVA in the presence or absence of pDC-depleting antibody anti-PDCA-1. On day 0 and day 14, C57BL/6J mice were intradermally immunized with OVA (10 μg/mouse)+/−Heat-iMVA (an equivalent amount of 107 pfu/mouse). Anti-PDCA1 antibody (500 μg/mouse) which selectively depletes pDCs or control IgG (500 μg/mouse) were administered intraperitoneally on Day −1, Day 1, Day 13, and Day 15. Spleens and lymph nodes were collected on day 21 for antigen-specific CD8+ T-cell analyses. FIG. 9B: Splenocytes were stimulated with OVA257-264 (SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml) for 12 h. The expression of IFN-γ by CD8+ T-cells was measured by flow cytometry. FIG. 9C: Cells from the draining lymph nodes (dLNs) were stimulated with OVA257-264(SIINFEKL) peptide (SEQ ID NO: 1) (10 μg/ml) for 12 h. The expression of IFN-γ by CD8+ T-cells was measured by flow cytometry (*p<0.05; ***p<0.001; n=5 except for the PBS group).



FIGS. 10A-10D are a series of graphs showing OVA-647 uptake in different dendritic cell populations in the draining lymph nodes. FIG. 10A: Murine inguinal lymph nodes were digested and single cell suspensions were obtained and labeled with cell surface markers. Migratory dendritic cell populations were marked as MHC-II+CD11c+. Resident dendritic cell populations were marked as MHC-IIIntCD11c+. The migratory dendritic cells were separated into CD11b+ DC, Langerin CD11b DC, and Langerin+ DC. Langerin+ DCs were divided into two populations: CD103+ DC and Langerhans cells. The resident dendritic cells were composed of two populations: CD8α+ resident DC and CD8α resident DC. FIG. 10B: C57/B6J mice were vaccinated with OVA-647 (10 μg/mice) by intradermal injection. After 24 h, dLNs were harvested and OVA-647 intensities in different dendritic cells populations from dLNs were measured by flow cytometry. FIG. 10C: C57/B6 mice were vaccinated with OVA-647 (10 μg/mice)+Heat-iMVA (an equivalent amount of 107 pfu) or Addavax (25 μl/mice) by intradermal injection. After 24 h, dLNs were harvested and the percentages of OVA-647+ cells among CD103CD11b DCs population from dLNs were measured by flow cytometry (**p<0.01; n=3). FIG. 10D: the percentages of OVA-647+ cells among CD11b+ DCs from dLNs were measured by flow cytometry (*p<0.05; n=3).



FIGS. 11A-11D are a series of graphs showing the efficacy of irradiated whole cell vaccination in the presence or absence of adjuvant and immune checkpoint blockade antibody anti-PD-L1 in a therapeutic murine B16-OVA tumor model. FIG. 11A: Irradiated B16-OVA vaccination strategy with or without Heat-iMVA or poly IC in the presence or absence of anti-PD-L1. C57BL/6J mice were implanted intradermally with B16-OVA cells (5×104) on the right flank. On day 3, 6, and 9, mice were immunized intradermally with irradiated B16-OVA cells (1×106) with or without immune adjuvant Heat-iMVA (an equivalent amount of 107 pfu per mouse) or with TLR3 agonist poly IC (50 μg per mouse) for a total of three times on the left flank. Anti-PD-L1 antibody (200 μg per mouse) was administered intraperitoneally to the indicated groups on day 3, 6 and 9. Mice were monitored for tumor sizes and survival. FIG. 11B and FIG. 11C: Kaplan-Meier survival curve of tumor-bearing mice vaccinated with PBS (n=5), Irradiated B16-OVA (n=5), Irradiated B16-OVA+Heat-iMVA (n=5), Irradiated B16-OVA+poly IC (n=5), PBS+anti-PD-L1 (n=10), Irradiated B16-OVA+anti-PD-L1 (n=10), Irradiated B16-OVA+Heat-iMVA+anti-PD-L1 (n=10), Irradiated B16-OVA+poly IC+anti-PD-L1 (n=10) **, p<0.01. FIG. 11D: Individual tumor volumes at different treatment groups over days post tumor implantation.



FIGS. 12A-12C show co-administration of melanoma neoantigen peptides with Heat-iMVA elicits antitumor effects in a therapeutic vaccination tumor model. FIG. 12A: vaccination model. FIG. 12B and FIG. 12C: subcutaneous (SC) vaccination with melanoma neoantigen peptide mix (M27/M30/M48) delayed B16-F10 tumor growth and prolonged survival of the mice. The antitumor effect is enhanced when neoantigen peptide mix were co-administered with Heat-iMVA.



FIG. 13 shows the complete genome sequence of vaccinia virus strain Ankara (GenBank Accession No.: U94848.1; SEQ ID NO: 3).





DETAILED DESCRIPTION

It is to be appreciated that certain aspects, modes, embodiments, variations, and features of the present technology are described below in various levels of detail in order to provide a substantial understanding of the present technology.


I. Definitions

The definitions of certain terms as used in this specification are provided below. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this present technology belongs.


As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the content clearly dictates otherwise. For example, reference to “a cell” includes a combination of two or more cells, and the like.


As used herein, the term “about” encompasses the range of experimental error that may occur in a measurement and will be clear to the skilled artisan.


As used herein, the term “adjuvant” refers to a substance that enhances, augments, or potentiates the host's immune response to antigens, including tumor antigens.


As used herein, the “administration” of an agent or drug to a subject includes any route of introducing or delivering to a subject a compound to perform its intended function. Administration can be carried out by any suitable route, including but not limited to, orally, intranasally, parenterally (intravenously, intramuscularly, intradermally, intraperitoneally, or subcutaneously), rectally, intrathecally, intratumorally, or topically. Administration includes self-administration and the administration by another.


As used herein, the term “antigen” refers to a molecule to which an antibody (or antigen binding fragment thereof) can selectively bind. The target antigen may be a protein, carbohydrate, nucleic acid, lipid, hapten, or other naturally occurring or synthetic compound. In some embodiments, the antigen is contained within a whole cell, such as in a tumor antigen-containing whole cell vaccine. In some embodiments, the target antigen encompasses cancer-related antigens or neoantigens and includes proteins or other molecules expressed by tumor or non-tumor cancers, such as molecules that are present in cancer cells but absent in non-cancer cells, and molecules that are up-regulated in cancer cells as compared to non-cancer cells.


As used herein, the term “effective amount” refers to a quantity of an agent, which when administered at one or more dosages and for a period of time, is sufficient to achieve a desired therapeutic and/or prophylactic effect, e.g., an amount which results in the prevention of, or a decrease in a disease or condition described herein or one or more signs or symptoms associated with a disease or condition described herein. In the context of therapeutic or prophylactic applications, the amount of a composition administered to the subject will vary depending on the composition, the degree, type, and severity of the disease and on the characteristics of the individual, such as general health, age, sex, body weight and tolerance to drugs. The skilled artisan will be able to determine appropriate dosages depending on these and other factors. The compositions can also be administered in combination with one or more additional therapeutic compounds. In the methods described herein, the therapeutic compositions may be administered to a subject having one or more signs or symptoms of a disease or condition described herein. As used herein, a “therapeutically effective amount” of a composition refers to composition levels in which the physiological effects of a disease or condition are ameliorated or eliminated. A therapeutically effective amount can be given in one or more administrations.


As used herein, “immune response” refers to the action of one or more of lymphocytes, antigen presenting cells, phagocytic cells, granulocytes, and soluble macromolecules produced by the above cells or the liver (including antibodies, cytokines, and complement) that results in selective damage to, destruction of, or elimination from the human body of cancerous cells, metastatic tumor cells, etc. An immune response may include a cellular response, such as a T-cell response that is an alteration (modulation, e.g., significant enhancement, stimulation, activation, impairment, or inhibition) of cellular, i.e., T-cell function. A T-cell response may include generation, proliferation or expansion, or stimulation of a particular type of T-cell, or subset of T-cells, for example, effector CD4+, CD4+ helper, effector CD8+, CD8+ cytotoxic, or natural killer (NK) cells. Such T-cell subsets may be identified by detecting one or more cell receptors or cell surface molecules (e.g., CD or cluster of differentiation molecules). A T-cell response may also include altered expression (statistically significant increase or decrease) of a cellular factor, such as a soluble mediator (e.g., a cytokine, lymphokine, cytokine binding protein, or interleukin) that influences the differentiation or proliferation of other cells. For example, interferon- (IFN-) γ is an essential cytokine for immunity against intracellular pathogens and cancer. IFN-γ is produced by cells that mediate both innate and adaptive immune responses. Natural killer (NK) and natural killer T (NKT) cells are the innate cell sources of this cytokine and rapidly produce IFN-γ upon activation. An immune response may also include humoral (antibody) response.


The term “immunogenic composition” is used herein to refer to a composition that will elicit an immune response in a mammal that has been exposed to the composition. In some embodiments, an immunogenic composition comprises an antigen and an adjuvant comprising Heat-iMVA, alone or in combination with immune checkpoint blockade inhibitors. As used herein, an immunogenic composition encompasses vaccines. In some embodiments, the immunogenic composition comprises a tumor antigen-containing whole cell vaccine (e.g., an irradiated whole cell vaccine).


As used herein, the term “inactivated MVA” refers to heat-inactivated MVA (Heat-iMVA) and/or UV-inactivated MVA which are infective, nonreplicative, and do not suppress IFN Type I production in infected DC cells. As used herein, the term “inactivated vaccinia virus” includes heat-inactivated vaccinia virus and/or UV-inactivated vaccinia virus. MVA or vaccinia virus inactivated by a combination of heat and UV radiation is also within the scope of the present disclosure.


As used herein, “Heat-inactivated MVA” (Heat-iMVA) and “inactivated vaccinia virus” refer to MVA and vaccinia virus, respectively, which have been exposed to heat treatment under conditions that do not destroy its immunogenicity or its ability to enter target cells (tumor cells) but remove residual replication ability of the virus as well as factors that inhibit the host's immune response. An example of such conditions is exposure to a temperature within the range of about 50 to about 60° C. for a period of time of about an hour. Other times and temperatures can be determined by one of skill in the art.


As used herein, “UV-inactivated MVA” and “UV-inactivated vaccinia virus” refer to MVA and vaccinia virus, respectively, that have been inactivated by exposure to UV under conditions that do not destroy its immunogenicity or its ability to enter target cells (tumor cells) but remove residual replication ability of the virus. An example of such conditions, which can be useful in the present methods, is exposure to UV using, for example, a 365 nm UV bulb for a period of about 30 min to about 1 hour. Other limits of these conditions of UV wavelength and exposure can be determined by one of skill in the art.


As used herein, the terms “individual”, “patient”, or “subject” can be an individual organism, a vertebrate, a mammal, or a human. In some embodiments, “subject” means any animal (mammalian, human, or other) patient that can be afflicted with cancer and when thus afflicted is in need of treatment. In some embodiments, the individual, patient or subject is a human.


As used herein, “metastasis” refers to the spread of cancer from its primary site to neighboring tissues or distal locations in the body. Cancer cells (including cancer stem cells) can break away from a primary tumor, penetrate lymphatic and blood vessels, circulate through the bloodstream, and grow in normal tissues elsewhere in the body. Metastasis is a sequential process, contingent on tumor cells (or cancer stem cells) breaking off from the primary tumor, traveling through the bloodstream or lymphatics, and stopping at a distant site. Once at another site, cancer cells re-penetrate through the blood vessels or lymphatic walls, continue to multiply, and eventually form a new tumor (metastatic tumor). In some embodiments, this new tumor is referred to as a metastatic (or secondary) tumor.


As used herein, “MVA” means “modified vaccinia Ankara” and refers to a highly attenuated strain of vaccinia derived from the Ankara strain and developed for use as a vaccine and vaccine adjuvant. The original MVA was isolated from the wild-type Ankara strain by successive passage through chicken embryonic cells. Treated thus, it lost about 15% of the genome of wild-type vaccinia including its ability to replicate efficiently in primate (including human) cells. MVA sequences are disclosed in Genbank U94848.1 (FIG. 13; SEQ ID NO: 3). Clinical grade MVA is commercially and publicly available from Bavarian Nordic A/S Kvistgaard, Denmark. Additionally, MVA is available from ATCC, Rockville, Md. and from CMCN (Institut Pasteur Collection Nationale des Microorganismes) Paris, France.


As used herein, the term “pharmaceutically-acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal compounds, isotonic and absorption delaying compounds, and the like, compatible with pharmaceutical administration. Pharmaceutically-acceptable carriers and their formulations are known to one skilled in the art and are described, for example, in Remington's Pharmaceutical Sciences (20th edition, ed. A. Gennaro, 2000, Lippincott, Williams & Wilkins, Philadelphia, Pa.).


As used herein, “pharmaceutically acceptable excipient” refers to substances and compositions that do not produce an adverse, allergic, or other untoward reaction when administered to an animal or a human. As used herein, the term includes all inert, non-toxic, liquid or solid fillers or diluents, as long as they do not react with the therapeutic substance of the invention in an inappropriate negative manner, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, preservatives and the like, for example liquid pharmaceutical carriers e.g., sterile water, saline, sugar solutions, Tris buffer, ethanol and/or certain oils.


As used herein, “prevention,” “prevent,” or “preventing” of a disorder or condition refers to one or more compounds that, in a statistical sample, reduces the occurrence of the disorder or condition in the treated sample relative to an untreated control sample, or delays the onset of one or more symptoms of the disorder or condition relative to the untreated control sample.


As used herein, the term “separate” therapeutic use refers to an administration of at least two active ingredients at the same time or at substantially the same time by different routes.


As used herein, the term “sequential” therapeutic use refers to administration of at least two active ingredients at different times, the administration route being identical or different. More particularly, sequential use refers to the whole administration of one of the active ingredients before administration of the other or others commences. It is thus possible to administer one of the active ingredients over several minutes, hours, or days before administering the other active ingredient or ingredients. There is no simultaneous treatment in this case.


As used herein, the term “simultaneous” therapeutic use refers to the administration of at least two active ingredients by the same route and at the same time or at substantially the same time.


As used herein, “solid tumor” refers to all neoplastic cell growth and proliferation, and all pre-cancerous and cancerous cells and tissues, except for hematologic cancers such as lymphomas, leukemias, and multiple myeloma. Examples of solid tumors include, but are not limited to: soft tissue sarcoma, such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor and other bone tumors (e.g., osteosarcoma, malignant fibrous histiocytoma), leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, brain/CNS tumors (e.g., astrocytoma, glioma, glioblastoma, childhood tumors, such as atypical teratoid/rhabdoid tumor, germ cell tumor, embryonal tumor, ependymoma) medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningioma, melanoma, neuroblastoma, and retinoblastoma. Some of the most common solid tumors for which the compositions and methods of the present disclosure would be useful include: head-and-neck cancer, rectal adenocarcinoma, glioma, medulloblastoma, urothelial carcinoma, pancreatic adenocarcinoma, uterine (e.g., endometrial cancer, fallopian tube cancer) ovarian cancer, cervical cancer prostate adenocarcinoma, non-small cell lung cancer (squamous and adenocarcinoma), small cell lung cancer, melanoma, breast carcinoma, ductal carcinoma in situ, renal cell carcinoma, and hepatocellular carcinoma. adrenal tumors (e.g., adrenocortical carcinoma), esophageal, eye (e.g., melanoma, retinoblastoma), gallbladder, gastrointestinal, Wilms' tumor, heart, head and neck, laryngeal and hypopharyngeal, oral (e.g., lip, mouth, salivary gland), nasopharyngeal, neuroblastoma, peritoneal, pituitary, Kaposi's sarcoma, small intestine, stomach, testicular, thymus, thyroid, parathyroid, vaginal tumor, and the metastases of any of the foregoing.


“Treating” or “treatment” as used herein covers the treatment of a disease or disorder described herein, in a subject, such as a human, and includes: (i) inhibiting a disease or disorder, i.e., arresting its development; (ii) relieving a disease or disorder, i.e., causing regression of the disorder; (iii) slowing progression of the disorder; and/or (iv) inhibiting, relieving, or slowing progression of one or more symptoms of the disease or disorder. In some embodiments, treatment means that the symptoms associated with the disease are, e.g., alleviated, reduced, cured, or placed in a state of remission.


It is also to be appreciated that the various modes of treatment of tumors as described herein are intended to mean “substantial,” which includes total but also less than total treatment, and wherein some biologically or medically relevant result is achieved. The treatment may be a continuous prolonged treatment for a chronic disease or a single, or few time administrations for the treatment of an acute condition.


As used herein, “T-cell” refers to a thymus derived lymphocyte that participates in a variety of cell-mediated adaptive immune reactions.


As used herein, “helper T-cell” refers to a CD4+ T-cell; helper T-cells recognize antigen bound to WIC Class II molecules. There are at least two types of helper T-cells, Th1 and Th2, which produce different cytokines.


As used herein, “cytotoxic T-cell” refers to a T-cell that usually bears CD8 molecular markers on its surface (CD8+) and that functions in cell-mediated immunity by destroying a target T-cell having a specific antigenic molecule on its surface. Cytotoxic T-cells also release Granzyme, a serine protease that can enter target T-cells via the perforin-formed pore and induce apoptosis (cell death). Granzyme serves as a marker of cytotoxic phenotype. Other names for cytotoxic T-cell include CTL, cytolytic T-cell, cytolytic T lymphocyte, killer T-cell, or killer T lymphocyte. Targets of cytotoxic T-cells may include virus-infected cells, cells infected with bacterial or protozoal parasites, or cancer cells. Most cytotoxic T-cells have the protein CD8 present on their cell surfaces. CD8 is attracted to portions of the Class I MEW molecule. Typically, a cytotoxic T-cell is a CD8+ cell.


II. Immune System and Cancer

Malignant tumors are inherently resistant to conventional therapies and present significant therapeutic challenges. Immunotherapy has become an evolving area of research and an additional option for the treatment of certain types of cancers. The immunotherapy approach rests on the rationale that the immune system may be stimulated to identify tumor cells, and target them for destruction.


Numerous studies support the importance of the differential presence of immune system components in cancer progression (Jochems et al., Exp Biol Med, 236(5): 567-579 (2011)). Clinical data suggest that high densities of tumor-infiltrating lymphocytes are linked to improved clinical outcome (Mlecnik et al., Cancer Metastasis Rev.; 30: 5-12, (2011)). The correlation between a robust lymphocyte infiltration and patient survival has been reported in various types of cancer, including melanoma, ovarian, head and neck, breast, urothelial, colorectal, lung, hepatocellular, gallbladder, and esophageal cancer (Angell et al., Current Opinion in Immunology, 25:1-7, (2013)). Tumor immune infiltrates include macrophages, dendritic cells (DC), monocytes, neutrophils, natural killer (NK) cells, naïve and memory lymphocytes, B cells and effector T-cells (T lymphocytes), primarily responsible for the recognition of antigens expressed by tumor cells and subsequent destruction of the tumor cells by cytotoxic T-cells.


Despite presentation of antigens by cancer cells and the presence of immune cells that could potentially react against tumor cells, in many cases the immune system does not get activated or is affirmatively suppressed. Key to this phenomenon is the ability of tumors to protect themselves from immune response by coercing cells of the immune system to inhibit other cells of the immune system. Tumors develop a number of immunomodulatory mechanisms to evade antitumor immune responses. For example, tumor cells secrete immune inhibitory cytokines (such as TGF-β) or induce immune cells, such as CD4+ T regulatory cells and macrophages, in tumor lesions to secrete these cytokines. Tumors also have the ability to bias CD4+ T-cells to express the regulatory phenotype. The overall result is impaired T-cell responses and impaired induction of apoptosis or reduced anti-tumor immune capacity of CD8+ cytotoxic T-cells. Additionally, tumor-associated altered expression of MEW class I on the surface of tumor cells makes them “invisible” to the immune response (Garrido et al. Cancer Immunol. Immunother. 59(10), 1601-1606 (2010)). Inhibition of antigen-presenting functions and dendritic cell (DC) additionally contributes to the evasion of anti-tumor immunity (Gerlini et al. Am. J. Pathol. 165(6), 1853-1863 (2004)).


Moreover, the local immunosuppressive nature of the tumor microenvironment, along with immune editing, can lead to the escape of cancer cell subpopulations that do not express the target antigens. Thus, finding an approach that would promote the preservation and/or restoration of anti-tumor activities of the immune system would be of considerable therapeutic benefit.


III. Modified Vaccinia Ankara (MVA)

Modified Vaccinia Ankara (MVA) virus is a member of the genera Orthopoxvirus in the family of Poxviridae. MVA was generated by approximately 570 serial passages on chicken embryo fibroblasts (CEF) of the Ankara strain of vaccinia virus (CVA) (Mayr et al., Infection 3, 6-14 (1975)). As a consequence of these long-term passages, the resulting MVA virus contains extensive genome deletions and is highly host cell restricted to avian cells (Meyer et al., J. Gen. Virol. 72, 1031-1038 (1991)). It was shown in a variety of animal models that the resulting MVA is significantly avirulent (Mayr et al., Dev. Biol. Stand. 41, 225-34 (1978)).


The safety and immunogenicity of MVA has been extensively tested and documented in clinical trials, particularly against the human smallpox disease. These studies included over 120,000 individuals and have demonstrated excellent efficacy and safety in humans. Moreover, compared to other vaccinia based vaccines, MVA has weakened virulence (infectiousness) while it triggers a good specific immune response. Thus, MVA has been established as a safe vaccine vector, with the ability to induce a specific immune response.


Due to the above mentioned characteristics, MVA became an attractive candidate for the development of engineered MVA vectors, used for recombinant gene expression and vaccines. As a vaccine vector, MVA has been investigated against numerous pathological conditions, including HIV, tuberculosis and malaria, as well as cancer (Sutter et al., Curr Drug Targets Infect Disord 3: 263-271(2003); Gomez et al., Curr Gene Ther 8: 97-120 (2008)).


It has been demonstrated that MVA infection of human monocyte-derived dendritic cells (DC) causes DC activation, characterized by the upregulation of co-stimulatory molecules and secretion of proinflammatory cytokines (Drillien et al., J Gen Virol 85: 2167-2175 (2004)). In this respect, MVA differs from standard wild type Vaccinia virus (WT-VAC), which fails to activate DCs. Dendritic cells can be classified into two main subtypes: conventional dendritic cells (cDCs) and plasmacytoid dendritic cells (pDCs). The former, especially the CD103+/CD8α+ subtype, are particularly adapted to cross-presenting antigens to T-cells; the latter are strong producers of Type I IFN.


Viral infection of human cells results in activation of an innate immune response (the first line of defense) mediated by type I interferons, notably interferon-alpha (a). This normally leads to activation of an immunological “cascade,” with recruitment and proliferation of activated T-cells (both CTL and helper) and eventually with antibody production. However, viruses express factors that dampen immune responses of the host. MVA is a better immunogen than WT-VAC and replicates poorly in mammalian cells. (See, e.g., Brandler et al., J Virol. 84, 5314-5328 (2010)).


The MVA genome sequence (SEQ ID NO: 3) given by GenBank Accession No. U94848.1 is provided in FIG. 13.


IV. Delivery of Heat-Inactivated MVA (Heat-iMVA) as an Adjuvant to a Subject to Treat Cancer

A. Compositions


Immune-Activating Cancer Vaccine Adjuvants


Recent discoveries of cancer neoantigens have generated a renewed interest in cancer vaccination and the combination of cancer vaccination with immune checkpoint blockade to enhance vaccination effects. Developing effective vaccine adjuvants that can maximize antitumor immune responses is critical for the success of cancer vaccines.


Cancer vaccines comprise cancer antigens and immune adjuvants. Cancer antigens generally include tumor differentiation antigens, cancer testis antigens, neoantigens, and viral antigens in the case of tumors associated with oncogenic virus infection. Cancer antigens can be provided in the form of irradiated tumor cells, dendritic cells (DCs) loaded with tumor cell lysates or peptides, DNA or RNA encoding antigen, as well as oncolytic virus with transgene(s) encoding cancer antigen(s). Dendritic cells (DCs) are professional antigen-presenting cells that are important for priming naive T-cells to generate antigen-specific T-cell responses. Immune adjuvants are agents that promote antigen uptake by DCs and/or DC maturation and activation. Several immune adjuvants, including toll-like receptor (TLR) agonists, poly (I:C) (TLR3 agonist), CpG (TLR9 agonist), Imiquimod (TLR7 agonist), as well as STING agonists, have been shown to improve vaccine efficacy in preclinical models and clinical settings.


Heat-iMVA as Adjuvant Therapy


The disclosure of the present technology relates to the use of Heat-inactivated MVA (Heat-iMVA) as a vaccine adjuvant. Heat-iMVA or Heat-inactivated vaccinia has been shown to induce type I IFN in conventional DCs (cDCs) via the cGAS/STING-dependent pathway and also induces type I IFN in plasmacytoid DCs (pDCs) via the TLR7/MyD88-dependent mechanism. Moreover, intratumoral injection of Heat-iMVA eradicates injected tumors and leads to the generation of systemic antitumor immunity either as monotherapy or in combination with immune checkpoint blockade (ICB). However, the use of Heat-iMVA as a vaccine adjuvant for peripheral vaccination outside the tumor beds has yet to be described.


Target Antigens


The compositions and methods disclosed herein are not intended to be limited by the choice of antigen or neoantigen. While numerous examples of antigens and neoantigens are provided, the skilled artisan can easily utilize the adjuvant disclosed herein with an antigen or neoantigen of choice. Exemplary, non-limiting target antigens that may be used in therapeutic regimens of the present technology include tumor differentiation antigens, cancer testis antigens, neoantigens, viral antigens in the case of tumors associated with oncogenic virus infection, GPA33, HER2/neu, GD2, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, MUM-1, CDK4, N-acetylglucosaminyltransferase, p15, gp75, beta-catenin, ErbB2, cancer antigen 125 (CA-125), carcinoembryonic antigen (CEA), RAGE, MART (melanoma antigen), MUC-1, MUC-2, MUC-3, MUC-4, MUC-5ac, MUC-16, MUC-17, tyrosinase, tyrosinase-related proteins 1 and 2, Pmel 17 (gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate cancer psm, PRAME (melanoma antigen), β-catenin, EBNA (Epstein-Barr Virus nuclear antigen) 1-6, p53, lung resistance protein (LRP) Bc1-2, prostate specific antigen (PSA), Ki-67, CEACAM6, colon-specific antigen-p (CSAp), and NY-ESO-1. In some embodiments, the antigen is a neoantigen selected from the group consisting of M27 (REGVELCPGNKYEMRRHGTTHSLVIHD) (SEQ ID NO: 4), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL) (SEQ ID NO: 5), M48 (SHCHWNDLAVIPAGVVHNWDFEPRKVS) (SEQ ID NO: 6), and combinations thereof. The target antigen may also be a fragment or fusion polypeptide comprising an immunologically active portion of the antigens listed above.


Immune Checkpoint Blockade (ICB)


In some embodiments, the immunogenic compositions of the present technology further comprise one or more immune checkpoint blockade agents. Immune checkpoint blockade (ICB) antibodies have been at the forefront of immunotherapy and have been accepted as one of the pillars of cancer management options, including surgery, radiation, and chemotherapy. Because immune checkpoints have been implicated in the downregulation of antitumor immunity, agents and antibodies targeting immune checkpoint proteins or their ligands (CTLA-4, PD-1, or PD-L1) have been successful in disinhibiting antitumor T-cells, thereby leading to proliferation and survival of activated T-cells. This has led to the FDA approval of multiple immune checkpoint blockade (ICB) agents for patients with advanced cancers of various histological types, including melanoma, non-small cell lung cancer, renal cell carcinoma, Hodgkin lymphoma, head-and-neck cancer, urothelial carcinoma, Merkel cell carcinoma, PD-L1+ gastric adenocarcinoma, as well as mismatch repair deficient and microsatellite instability (MSI) high metastatic solid tumors.


Non-limiting examples of immune checkpoint blocking agents include agents or antibodies that modulate the activity of one or more checkpoint proteins including cytotoxic T-lymphocyte antigen-4 (CTLA-4) or its ligands, and programmed death 1 (PD-1) or its ligands, PD-L1, and PD-L2.


Pharmaceutical Compositions and Preparations of the Present Technology

Disclosed herein are pharmaceutical compositions comprising an antigen and Heat-iMVA as an adjuvant that may contain a carrier or diluent, which can be a solvent or dispersion medium containing, for example, water, saline, Tris buffer, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. 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. Prevention of the action of microorganisms can be effected by various antibacterial and antifungal agents and preservatives, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In some embodiments, isotonic agents, for example, sugars or sodium chloride, and buffering agents are included. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin or carrier molecules. Other excipients may include wetting or emulsifying agents. In general, excipients suitable for injectable preparations can be included as apparent to those skilled in the art.


Pharmaceutical compositions and preparations comprising an antigen and Heat-iMVA as an adjuvant may be manufactured by means of conventional mixing, dissolving, granulating, emulsifying, encapsulating, entrapping or lyophilizing processes. Pharmaceutical compositions may be formulated in conventional manner using one or more physiologically acceptable carriers, diluents, excipients or auxiliaries that facilitate formulating preparations suitable for in vitro, in vivo, or ex vivo use. The compositions can be combined with one or more additional biologically active agents (for example parallel administration of GM-CSF) and may be formulated with a pharmaceutically acceptable carrier, diluent or excipient to generate pharmaceutical (including biologic) or veterinary compositions of the instant disclosure suitable for parenteral or intra-tumoral administration.


Many types of formulation are possible as is appreciated by those skilled in the art. The particular type chosen is dependent upon the route of administration chosen, as is well-recognized in the art. For example, systemic formulations will generally be designed for administration by injection, e.g., intravenous, as well as those designed for intratumoral delivery. In some embodiments, the systemic or intratumoral formulation is sterile.


Sterile injectable solutions are prepared by incorporating an antigen and Heat-iMVA as an adjuvant in the required amount of the appropriate solvent with various other ingredients enumerated herein, as required, followed by suitable sterilization means. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle that contains the 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 techniques, which yield a powder of the virus plus any additional desired ingredient from a previously sterile-filtered solution thereof.


In some embodiments, an antigen and Heat-iMVA compositions of the present disclosure may be formulated in aqueous solutions, or in physiologically compatible solutions or buffers such as Hanks's solution, Ringer's solution, mannitol solutions or physiological saline buffer. In certain embodiments, any of the antigen and Heat-iMVA compositions may contain formulator agents, such as suspending, stabilizing, penetrating or dispersing agents, buffers, lyoprotectants or preservatives such as polyethylene glycol, polysorbate 80, 1-dodecylhexahydro-2H-azepin-2-one (laurocapran), oleic acid, sodium citrate, Tris HCl, dextrose, propylene glycol, mannitol, polysorbate polyethylenesorbitan monolaurate (Tween®-20), isopropyl myristate, benzyl alcohol, isopropyl alcohol, ethanol sucrose, trehalose and other such generally known in the art may be used in any of the compositions of the instant disclosure.


In some embodiments, the compositions of the present technology can be stored at −80° C. For the preparation of vaccine shots, e.g., 102-108 or 102-109 viral particles can be lyophilized, for example, in 100 ml of phosphate-buffered saline (PBS) in the presence of 2% peptone and 1% human albumin in an ampoule, preferably a glass ampoule. Alternatively, the injectable preparations can be produced by stepwise freeze-drying of the recombinant virus in a formulation. This formulation can contain additional additives such as mannitol, dextran, sugar, glycine, lactose or polyvinylpyrrolidone or other additives such as antioxidants or inert gas, stabilizers or recombinant proteins (e.g., human serum albumin) suitable for in vivo administration. The glass ampoule is then sealed and can be stored between 4° C. and room temperature for several months. In some embodiments, the ampoule is stored at temperatures below −20° C.


For therapy, the lyophilisate can be dissolved in an aqueous solution, such as physiological saline or Tris buffer, and administered either systemically or intratumorally. The mode of administration, the dose, and the number of administrations can be optimized by those skilled in the art.


The pharmaceutical compositions comprising an antigen and Heat-iMVA as an adjuvant according to the present disclosure may comprise an additional adjuvant including aluminum salts, such as aluminum hydroxide or aluminum phosphate, Quil A, bacterial cell wall peptidoglycans, virus-like particles, polysaccharides, toll-like receptors, nano-beads, etc.


Vaccines


In some embodiments, compositions comprising a Heat-iMVA adjuvant and one or more antigens are formulated into vaccines. In some embodiments, the vaccines are tumor antigen-containing whole cell vaccines (e.g., an irradiated whole cell vaccine). In some embodiments, the vaccines are administered to a subject to elicit an immune response against the antigens formulated therewith.


Effective Amount and Dosage of Heat-iMVA as a Cancer Vaccine Immune Adjuvant


In general, the subject is administered a dosage Heat-iMVA in the range of about 106 to about 1010 plaque forming units (pfu), although a lower or higher dose may be administered. In some embodiments, the dosage ranges from about 102 to about 1010 pfu. In some embodiments, the dosage ranges from about 103 to about 1010 pfu. In some embodiments, the dosage ranges from about 104 to about 1010 pfu. In some embodiments, the dosage ranges from about 105 to about 1010 pfu. In some embodiments, the dosage ranges from about 106 to about 1010 pfu. In some embodiments, the dosage ranges from about 103 to about 1010 pfu. In some embodiments, the dosage ranges from about 108 to about 1010 pfu. In some embodiments, the dosage ranges from about 109 to about 1010 pfu. In some embodiments, dosage is about 107 to about 109 pfu. The equivalence of pfu to virus particles can differ according to the specific pfu titration method used. Generally, a pfu is equal to about 5 to 100 virus particles and 0.69 PFU is about 1 TCID50. A therapeutically effective amount of Heat-iMVA can be administered in one or more divided doses for a prescribed period of time and at a prescribed frequency of administration.


For example, as is apparent to those skilled in the art, a therapeutically effective amount of Heat-iMVA as an adjuvant in accordance with the present disclosure may vary according to factors such as the disease state, age, sex, weight, and general condition of the subject, and the ability of Heat-iMVA to elicit a desired immunological response in the particular subject (the subject's response to therapy). In delivering Heat-iMVA to a subject, the dosage will also vary depending upon such factors as the general medical condition, previous medical history, disease type and progression, tumor burden, the presence or absence of tumor infiltrating immune cells in the tumor, and the like.


In some embodiments, it may be advantageous to formulate compositions of the present disclosure in dosage unit form for ease of administration and uniformity of dosage. “Dosage unit form as used herein” refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required pharmaceutically or veterinary acceptable carrier.


Administration and Therapeutic Regimen of Heat-iMVA as a Cancer Vaccine Immune Adjuvant


A pharmaceutical composition is typically formulated to be compatible with its intended route of administration. Administration of Heat-iMVA as an adjuvant in an immunogenic composition (e.g., vaccine) can be achieved using more than one route. Examples of routes of administration include, but are not limited to parenteral (e.g., intravenous, intramuscular, intraperitoneal, intradermal, subcutaneous), intratumoral, intrathecal, intranasal, systemic, transdermal, iontophoretic, intradermal, intraocular, or topical administration. In one embodiment, the pharmaceutical composition of the present technology comprising an antigen and Heat-iMVA as an adjuvant is administered directly into the tumor, e.g. by intratumoral injection, where a direct local reaction is desired. In some embodiments, the pharmaceutical composition of the present technology comprising an antigen and Heat-iMVA as an adjuvant is administered peripherally relative to tumor beds. Additionally, the administration routes can vary, e.g., first administration using an intratumoral injection, and subsequent administration via an intravenous injection, or any combination thereof. A therapeutically effective amount of Heat-iMVA as an adjuvant in a cancer vaccine injection can be administered for a prescribed period of time and at a prescribed frequency of administration. In certain embodiments, the pharmaceutical compositions of the present technology can be used in conjunction with other therapeutic treatments such as chemotherapy or radiation. In some embodiments, the pharmaceutical compositions of the present technology comprising a therapeutically effective amount of Heat-iMVA as an adjuvant can be used in conjunction with immune checkpoint blockade therapy, such as antibodies targeting immune checkpoint proteins CTLA-4, PD-1, PD-L1, and/or PD-L2.


In certain embodiments, the pharmaceutical composition comprising an antigen and Heat-iMVA as an adjuvant is administered at least once weekly or monthly but can be administered more often if needed, such as two times weekly for several weeks, months, years or even indefinitely as long as benefits persist. More frequent administrations are contemplated if tolerated and if they result in sustained or increased benefits. Benefits of the present methods include but are not limited to the following: reduction of the number of cancer cells, reduction of the tumor size (e.g., tumor volume), eradication of tumor, inhibition of cancer cell infiltration into peripheral organs, inhibition or stabilization or eradication of metastatic growth, inhibition or stabilization of tumor growth, and stabilization or improvement of quality of life. Furthermore, the benefits may include induction of an immune response against the tumor, increased IFN-γ+CD8+ T-cells, increased IFN-γ+CD4+ T-cells, activation of effector CD4 T-cells, an increase of effector CD8+ T-cells, or reduction of regulatory CD4+ cells. For example, in the context of melanoma, a benefit may be a lack of recurrences or metastasis within one, two, three, four, five or more years of the initial diagnosis of melanoma. Similar assessments can be made for colon cancer and other solid tumors.


B. Methods


In one aspect, the present disclosure provides for a method for treating solid tumor by enhancing an immune response in a subject in need thereof, the method comprising administering to the subject an immunogenic composition comprising one or more antigens and an adjuvant comprising a Heat-inactivated modified vaccinia Ankara virus (Heat-iMVA), thereby treating the tumor by enhancing immune response.


In some embodiments, the disclosure provides methods comprising administering the immunogenic composition comprising one or more antigens and Heat-iMVA as an adjuvant to a subject in order to elicit an immune response against the antigens.


In some embodiments of the methods disclosed herein, the administration step comprises administering the immunogenic composition in multiple doses.


In some embodiments, the methods described herein further comprise administering to the subject an immune checkpoint blockade agent selected from the group consisting of cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors, programmed death 1 (PD-1) inhibitors, PD-L1 inhibitors, and PD-L2 inhibitors. In some embodiments, the immunogenic composition is delivered to the subject separately, sequentially, or simultaneously with the administration of the immune checkpoint blockade agent. In some embodiments, the PD-L1 inhibitor is an anti-PD-L1 antibody.


C. Kits


In some embodiments, kits are provided. In some embodiments, the kit includes a container means and a separate portion of each of: (a) an antigen and (b) an adjuvant comprising Heat-iMVA.


V. Type I IFN and the Cytosolic DNA-Sensing Pathway in Tumor Immunity

Type I IFN plays important roles in host antitumor immunity (Fuertes et al., Trends Immunol 34, 67-73 (2013)). IFNAR1-deficent mice are more susceptible to developing tumors after implantation of tumor cells; spontaneous tumor-specific T-cell priming is also defective in IFNAR1-deficient mice (Diamond et al., J Exp Med 208, 1989-2003 (2011); Fuertes et al., J Exp Med 208, 2005-2016 (2011)). More recent studies have shown that the cytosolic DNA-sensing pathway is important in the innate immune sensing of tumor-derived DNA, which leads to the development of antitumor CD8+ T-cell immunity (Woo et al., Immunity 41, 830-842 (2014)). This pathway also plays a role in radiation-induced antitumor immunity (Deng et al., Immunity 41, 843-852 (2014)). Although spontaneous anti-tumor T-cell responses can be detected in patients with cancers, cancers eventually overcome host antitumor immunity in most patients. Novel strategies to alter the tumor immune suppressive microenvironment would be beneficial for cancer therapy.


VI. Immune Response

In addition to induction of the immune response by up-regulation of particular immune system activities (such as antibody and/or cytokine production, or activation of cell mediated immunity), immune responses may also include suppression, attenuation, or any other downregulation of detectable immunity, so as to reestablish homeostasis and prevent excessive damage to the host's own organs and tissues. In some embodiments, an immune response that is induced according to the methods of the present disclosure generates effector CD8+ (antitumor cytotoxic CD8+) T-cells or activated T helper cells or both that can bring about directly or indirectly the death, or loss of the ability to propagate, of a tumor cell.


Induction of an immune response by the compositions and methods of the present disclosure may be determined by detecting any of a variety of well-known immunological parameters (Takaoka et al., Cancer Sci. 94:405-11 (2003); Nagorsen et al., Crit. Rev. Immunol. 22:449-62 (2002)). Induction of an immune response may therefore be established by any of a number of well-known assays, including immunological assays. Such assays include, but need not be limited to, in vivo, ex vivo, or in vitro determination of soluble immunoglobulins or antibodies; soluble mediators such as cytokines, chemokines, hormones, growth factors and the like as well as other soluble small peptide, carbohydrate, nucleotide and/or lipid mediators; cellular activation state changes as determined by altered functional or structural properties of cells of the immune system, for example cell proliferation, altered motility, altered intracellular cation gradient or concentration (such as calcium); phosphorylation or dephosphorylation of cellular polypeptides; induction of specialized activities such as specific gene expression or cytolytic behavior; cellular differentiation by cells of the immune system, including altered surface antigen expression profiles, or the onset of apoptosis (programmed cell death); or any other criterion by which the presence of an immune response may be detected. For example, cell surface markers that distinguish immune cell types may be detected by specific antibodies that bind to CD4+, CD8+, or NK cells. Other markers and cellular components that can be detected include but are not limited to interferon γ (IFN-γ), tumor necrosis factor (TNF), IFN-α, IFN-β (IFNB), IL-6, and CCLS. Common methods for detecting the immune response include, but are not limited to flow cytometry, ELISA, immunohistochemistry. Procedures for performing these and similar assays are widely known and may be found, for example in Letkovits (Immunology Methods Manual: The Comprehensive Sourcebook of Techniques, Current Protocols in Immunology, 1998).


VII. The Role of Dendritic Cells in Vaccine Efficacy

Dendritic cells (DCs) are professional antigen-presenting cells that play important roles in linking innate immunity with adaptive immunity. DCs can efficiently capture antigens, undergo maturation, and migrate to lymphoid organs to prime naïve T-cells to generate antigen-specific T-cell immune responses. DCs comprise several heterogeneous populations, each of which plays a distinct role in antigen presentation. For example, Batf3-dependent CD103+/CD8 α DCs are most efficient in cross-presenting antigens that expand and activate CD8+ T-cells. CD11b+ DCs, which are important in generating Th2. By contrast, pDCs are potent type I IFN producing cells and can cooperate with CD103+/CD8α DCs for antigen cross-presentation, possibly through the production of type I IFN. Type I IFN signaling is important for the function of CD103+/CD8 α DCs.


VIII. The Role of the STING Pathway in Vaccine Efficacy

STING (stimulator of IFN genes), also known as transmembrane protein 173 (TMEM 173), is an endoplasmic reticulum-localized critical adaptor for innate immunity. The STING pathway is activated by interacting with cyclic dinucleotides, which include cyclic GMP-AMP (cGAMP), which is produced by the mammalian cytosolic DNA sensor cGAS as well as cyclic dinucleotides (CDN) produced by bacteria. Recent reports indicate that tumor DNA could be detected by the cytosolic DNA-sensing pathway mediated by STING/IRF3, which leads to spontaneous CD8+ T-cell priming. Mice deficient in STING or IRF3 were incapable of rejecting immunogenic tumors. Furthermore, STING-deficient mice were resistant to the combination immunotherapy with anti-CTLA-4 and anti-PD-L1, partly because tumor-specific T-cells failed to expand in the STING-deficient host. Moreover, intratumoral delivery of murine STING agonist, DMXAA, also showed efficacy in tumor eradication in a B16.SIY model, which is dependent on STING. However, human STING is insensitive to DMAXX stimulation, which explains the failure of DMXAA in clinical trials. By contrast, synthetic cyclic dinucleotides (CDN) can act as human STING agonists, and preclinical studies showed that intratumoral delivery of CDN elicits antitumor effects in a B16 melanoma model in a STING-dependent manner. STING is also important for iMVA-induced anti-tumor therapeutic effects. STING-deficient mice are much less adept than WT mice at eradicating tumors in response to Heat-iMVA. Immune profiling of injected and non-injected tumors revealed that Heat-iMVA-treatment leads to the increase of CD8+ T-cells, which is reduced in STING-deficient mice.


EXPERIMENTAL EXAMPLES

The present technology is further illustrated by the following examples, which should not be construed as limiting in any way.


In summary, the examples described herein show that co-administration of Heat-iMVA with an antigen (e.g., chicken ovalbumin (OVA)) increased the percentage of antigen-specific (e.g., OVA-specific) CD8+ T-cells and CD4+ T-cells in spleens and draining lymph nodes and boosted serum levels of antigen-specific (e.g., OVA-specific) immunoglobulins (e.g., IgG2c and IgG1. These results also show that the induction of OVA-specific CD8+ T-cells was significantly reduced in Batf3−/− mice as well as in mice with deletion of plasmacytoid dendritic cells (pDCs). In addition, these results also show that in a murine therapeutic vaccination model, vaccination with irradiated B16-OVA cells with Heat-iMVA extended the median survival, which was further extended in the presence of immune checkpoint blockade antibodies. Moreover, the results presented herein demonstrate that Heat-iMVA enhances antigen presentation by DCs, and demonstrate that Heat-iMVA is useful as a vaccine adjuvant for peptide, viral-based vaccine vector, and irradiated whole cell vaccinations.


General Materials and Methods

Viruses and Cell lines. MVA and MVA-OVA viruses were kindly provided by Gerd Sutter (University of Munich), and propagated in BHK-21 (baby hamster kidney cell, ATCC CCL-10) cells. MVA is commercially and/or publicly available. The viruses were purified through a 36% sucrose cushion. Heat-iMVA was generated by incubating purified MVA virus at 55° C. for 1 hour. BHK-21 cells were cultured in Eagle's Minimal Essential Medium (Eagle's MEM, can be purchased from Life Technologies, Cat #11095-080) containing 10% FBS, 0.1 mM nonessential amino acids (NEAA), and 50 mg/ml gentamycin. The murine melanoma cell line B16-F10 was originally obtained from I. Fidler (MD Anderson Cancer Center). B16-OVA cells were kindly provided by G. Dranoff (Dana Farber Cancer Center). B16-F10 cells and B16-OVA were maintained in RPMI 1640 medium supplemented with 10% FBS, 100 Units/ml penicillin, 100 μg/ml streptomycin, 0.1 mM NEAA, 2 mM L-glutamine, 1 mM sodium pyruvate, and 10 mM HEPES buffer. All cells were grown at 37° C. in a 5% CO2 incubator.


Cells and cell lines used herein are commercially or publicly available unless otherwise indicated.


Mice. Female C57BL/6J mice between 6 and 10 weeks of age were purchased from the Jackson Laboratory and were used for the preparation of bone marrow-derived dendritic cells and for in vivo experiments. These mice were maintained in the animal facility at the Sloan Kettering Institute. All procedures were performed in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institute of Health. The protocol was approved by the Committee on the Ethics of Animal Experiments of Sloan-Kettering Cancer Institute. Batf3−/−, and STINGGt/Gt mice were generated in the laboratories of K. Murphy (Washington University; Batf3−/−), and R. Vance (University of California, Berkeley; STINGGt/Gt). These mice were bred and maintained in the animal facility at the Sloan Kettering Institute.


For chicken ovalbumin (OVA) immunization experiments, mice were injected with OVA (10 μg dissolved in 50 μl of PBS per mouse) with or without heat-inactivated MVA (Heat-iMVA; an equivalent amount of 107 pfu per mouse), or with Complete Freund Adjuvant (CFA) on Day 0 and Day 14. For vaccination with OVA alone or OVA+Heat-MVA, intramuscular (IM), subcutaneous (SC), or intradermal (ID) delivery methods were tested as detailed in vaccination strategies. For vaccination with OVA+CFA, only subcutaneous administration was used due to painful reactions and risks of tissue damage at the site of injection. On day 21, spleens, draining lymph nodes (dLNs), and blood were collected for evaluation of antigen-specific T cell and antibody responses.


In some experiments, STINGGt/Gt, Batf3−/− mice and WT age-matched controls were used for SC vaccination with OVA+Heat-iMVA.


For the depletion of plasmacytoid dendritic cells (pDCs), mice were injected intraperitoneally (i.p.) four times on Day −1, Day 1, Day 13, and Day 14, with 500 μg of BX444 anti-pDC antibody targeting CD317 (BioXCell) in 500 μl of PBS. CD317 is also known as BST2 and PDCA-1, which is exclusively expressed on pDCs from naïve mice. Control mice were injected i.p. with 500 μg of isotype control rat IgG1 anti-horseradish peroxidase (HRPN) antibody (BioXCell) in 500 μl of PBS following the same schedule. Immunizations with OVA+Heat-iMVA were administered on Day 0 and Day 14 as described before. On Day 21, spleens, dLNs, and blood were collected for evaluation of antigen-specific T cell and antibody responses.


For skin scarification (SS) with MVA-OVA with or without Heat-iMVA, 6-8 week old female C57BL/6J mice were anesthetized and 5 μl of viruses were applied to the tail skin 1 cm from the base of the tails. SS was accomplished by gently scratching the skin with a 28 1/2G needle 25 times (Liu et al. Immunity 2006). Increasing doses of MVA-OVA (105, 106, 107 pfu) were applied. For the combination, MVA-OVA at 107 pfu and Heat-iMVA at an equivalent amount of 105 pfu were mixed prior to SS. Spleens, dLNs, and blood were harvested from euthanized mice one week later.


In some experiments, STINGGt/Gt, Batf3−/− mice and WT age-matched controls were used for SS vaccination with MVA-OVA at 106 pfu.


Flow cytometry analysis of IFN-γ secreting antigen-specific T cells. Spleen or dLNs were minced into single cell suspensions and subjected into analysis. For OVA257-264 (SIINFEKL) (SEQ ID NO: 1) or OVA323-339 (ISQAVHAAHAEINEAGR) (SEQ ID NO: 2)-specific T cells analysis, splenocyetes or dLN cells were incubated with 10 μg/ml of respective peptide (Invivogen) overnight, then were stained with following antibodies: PE-Cy7 anti-CD3, APC-Cy7 anti-CD4, PE-Cy5.5 anti-CD8, and APC anti-IFN-γ antibodies. All antibodies are purchased from eBioscience. Live cells are distinguished from dead cells by using the fixable dye eFluor506 (eBioscience). Data were acquired using the LSR II flow cytometer (BD Biosciences). Data were analyzed with FlowJo software (Tree Star). For MVA-OVA-specific T cell analysis, BMDC were firstly infected with MVA-OVA (MOI=5) for 6 h, and they were then co-incubated with splenocytes or lymph node cells overnight prior to intracellular cytokine staining for IFN-γ production.


ELISA analysis of OVA-specific antibodies. ELISA plates were coated with 10 μg/ml of OVA protein overnight. Then plates were blocked with 1% BSA in PBST at 37° C. for 1 h. Serum was two-fold serially diluted and were added to each well and incubated at 37° C. for 1 h. After three washes with PBST, plates were incubated with HRP-labeled secondary antibodies (HRP-anti-mouse IgG1 or HRP-anti-mouse IgG2c) at 37° C. for 1 h. Then TMB substrates (Sigma) were added and A450 absorbance was measured.


Generation of bone marrow-derived dendritic cells (BMDCs). The bone marrow cells from the tibia and femur of mice were collected by first removing muscles from the bones, and then flushing the cells out using 0.5 cc U-100 insulin syringes (Becton Dickinson) with RPMI with 10% FCS. After centrifugation, cells were re-suspended in ACK Lysing Buffer (Lonza) for red blood cells lysis by incubating the cells on ice for 1-3 min. Cells were then collected, re-suspended in fresh medium, and filtered through a 40-μm cell strainer (BD Biosciences). The number of cells was counted. For the generation of GM-CSF-BMDCs, the bone marrow cells (5 million cells in each 15 cm cell culture dish) were cultured in CM in the presence of GM-CSF (30 ng/ml, produced by the Monoclonal Antibody Core facility at the Sloan Kettering Institute) for 10-12 days. CM is RPMI 1640 medium supplemented with 10% fetal bovine serum (FBS), 100 Units/ml penicillin, 100 μg/ml streptomycin, 0.1 mM essential and nonessential amino acids, 2 mM L-glutamine, 1 mM sodium pyruvate, and 10 mM HEPES buffer. Cells were fed every 2 days by replacing 50% of the old medium with fresh medium and re-plated every 3-4 days to remove adherent cells. Only non-adherent cells were used for experiments. For the generation of fms-like tyrosine kinase-3 ligand-cultured murine bone marrow-derived dendritic cells (Flt3L-BMDCs), the bone marrow cells (5×106 cells in each well of 6-well plates) were cultured in the presence of Flt3L (100 ng/ml; R&D Systems) for 7 to 9 days. Cells were fed every 2 to 3 days by replacing 50% of the old medium with fresh medium.


OT-I or OT-II proliferation assay. Spleens were harvested from OT-I or OT-II mice and T cells were purified with mouse CD8+ or CD4+ isolation kit (Miltenyi). OT-1 or OT-II cells were labeled with 10 μM of Carboxyfluorescein Diacetate Succinimidyl Ester (CF SE) (ThermoFisher) for 15 min at RT. GM-CSF-cultured or Flt3L-cultured BMDC were incubated with OVA (0.1, 0.2, 0.5 mg/ml)+/−Heat-iMVA (MOI of 1) for 3 h, then washed and co-cultured with CFSE-labeled OT-I or OT-II for 3 days (BMDC:OT-I or OT-II T cells=1:5). After staining with anti-CD3, anti-CD4, and anti-CD8 antibodies, CFSE intensities were determined using the LSR II flow cytometer (BD Biosciences). In some experiments, toll-like receptor 3 (TLR3) agonist poly IC (5 μg/ml) was used in lieu of Heat-iMVA.


MHC-I (H-2Kb) expression on BMDCs and OVA uptake by BMDCs. GM-CSF cultured BMDC are incubated with OVA (1 mg/ml)+/−Heat-iMVA (MOI of 1), or poly IC (5 μg/ml) for 16 h, then the cell surface expression of H2-Kb at cellular surface was measured by FACS using anti-H2-Kb antibody (eBioscience). For OVA-647 uptake, GM-CSF-cultured BMDCs were infected or mock infected with Heat-iMVA (MOI of 1) for 1 h or 16 h, and then incubated with Alexa Fluor™ 647 Conjugated Ovalbumin (OVA-647; ThemoFisher) for 1 h. The fluorescence intensities of phagocytosed OVA-647 in BMDCs were determined by flow cytometry. Data were analyzed with Flowjo software (Treestar).


OVA-647 uptake and transport by migratory dendritic cells. 10 μg of OVA-647 with or without Heat-iMVA (an equivalent of 107 pfu) was intradermally injected into right flank of C57Bl/6J mice. After 24 h, the draining inguinal lymph nodes were collected. Single cell suspensions were generated and stained with the following antibodies: efluor450-CD19 (1D3), TER119 (TER119), CD49b (DX5), PE-Cy7-CD3e (145-2C11), Alexa Fluor-700-CD11c (N418), PE-Texa Red-MI-IC-II (M5/114.15.2), APC-Cy7-CD11b (M1/70), FITC-CD103 (2E7), PE-Cy5.5-CD8α (53-6.7), and PE-Langerin (eBioL31). All antibodies are from eBioscience. The fluorescence intensities of phagocytosed OVA-647 in DC subsets were determined by flow cytometry. Data were analyzed with FlowJo software (Treestar).


In some experiments, vaccine adjuvant Addavax (25 μl per mouse) was used in lieu of Heat-iMVA.


Tumor Implantation Model and Intradermal Vaccination with Irradiated B16-OVA Cells. Briefly, 5×104 B16-OVA melanoma cells were implanted intradermally (i.d.) to the right flanks of C57BL/6J mice on day 0. 3, 6, and 9 days after tumor implantation, the mice were intradermally vaccinated on the left flanks with γ-irradiated (150 Gy) B16-OVA (1×106 cells per mouse)+/−Heat-iMVA (107 pfu per mouse). The tumor sizes were measured and the survival of mice was monitored.


In some experiments, poly IC (50 μg per mouse) was used in lieu of Heat-iMVA.


In some experiments, anti-PD-L1 antibody (200 μg per mouse; clone 10F.9G2 from BioXcell) was administered intraperitoneally during immunization on Day 3, 6, and 9.


Statistics. Two-tailed unpaired Student's t test was used for comparisons of two groups in the studies. Survival data were analyzed by log-rank (Mantel-Cox) test. The p values deemed significant are indicated in the figures as follows: *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001. The numbers of animals included in the study are discussed in each figure legend.


Example 1: Co-Administration of Heat-iMVA with Model Antigen, Chicken Ovalbumin (OVA), Enhances the Generation of OVA-Specific CD8+ and CD4+ T-Cells in the Spleen and Draining Lymph Nodes (dLNs), and Serum Anti-OVA IgG Antibodies in Immunized Mice

This example demonstrates that Heat-iMVA can act as a vaccine adjuvant to enhance antigen presentation by dendritic cells (DCs). Mice were immunized intramuscularly (IM) with OVA (10 μg) with or without Heat-iMVA (1×107 pfu) twice, 2 weeks apart. Mice were euthanized 1 week after the second vaccination, with spleens, draining lymph nodes (dLNs), and blood subsequently collected for OVA-specific T-cell and antibody assessment (FIG. 1A). To determine anti-OVA CD8+ T-cell responses, splenocytes (500,000 cells) were incubated with OVA 257-264 (SIINFEKL) peptide (SEQ ID NO: 1), which is a MHC class I (Kb)-restricted peptide epitope of OVA, for 12 h before they were stained for anti-CD8 and anti-IFN-γ antibodies. To test anti-OVA CD4+ T-cell responses, splenocytes (500,000 cells) were incubated with OVA 323-339 (ISQAVHAAHAEINEAGR) peptide (SEQ ID NO: 2), which is a MHC class II I-Ad-restricted peptide epitope of OVA, for 12 h before they were stained for anti-CD4 and anti-IFN-γ antibodies. Co-administration of Heat-iMVA with OVA intramuscularly resulted in the increase of anti-OVA IFN-γ+CD8+ T-cells and anti-OVA IFN-γ+CD4+ T-cells in the spleens compared with OVA alone. The percentages of IFN-γ+ T-cells among CD8+ T-cells in spleens increased from 1% in the OVA-treated mice to 1.8% in OVA+Heat-iMVA-treated ones (P<0.01; n=5; FIGS. 1B, D). The percentages of IFN-γ+ T-cells among CD4+ T-cells in spleens increased from 0.5% in the OVA-treated mice to 1.5% in OVA+Heat-iMVA-treated mice (P<0.01; n=5; FIG. 1C, E).


Similar induction of anti-OVA IFN-γ+CD8+ T-cells and anti-OVA IFN-γ+CD8+ T-cells after IM OVA plus Heat-iMVA was observed in the dLNs. Briefly, single cell suspensions were generated from dLNs, and 500,000 cells were incubated with either OVA 257-264 or OVA 323-339 peptides. The percentages of IFN-γ+ T-cells among CD8+ T-cells in dLNs increased from 0.5% in the OVA-treated mice to 2.5% in OVA+Heat-iMVA-treated ones (P<0.01; n=5; FIG. 1E, H). The percentages of IFN-γ+ T-cells among CD4+ T-cells in dLNs increased from 0.25% in the OVA-treated mice to 0.65% in OVA+Heat-iMVA-treated mice (P<0.05; n=5; FIG. 1G, I). In addition, intramuscular co-administration of OVA and Heat-iMVA induced higher titers of anti-OVA IgG1 and IgG2c compared with OVA alone (P<0.01; n=5; FIG. 1.1 for IgG1; P<0.001; n=5; FIG. 1K for IgG2).


Example 2: Heat-iMVA is Superior to Complete Freund Adjuvant (CFA) in Generating Antigen-Specific CD8+ and CD4+ T-cell Responses

Complete Freund adjuvant (CFA) comprises heat-killed Mycobacterium tuberculosis in non-metabolizable oils (paraffin oil and mannide monooleate). It also contains ligands for TLR2, TLR4, and TLR9. Injection of antigen with CFA induces a Th1-dominant immune response. CFA's use in humans is currently impermissible due to its toxicity profile, and its use in animals is limited to subcutaneous or intraperitoneal routes due to painful reactions and risks of tissue damage at the site of injection. To test whether Heat-iMVA is superior to CFA, mice were vaccinated subcutaneously with OVA antigen plus Heat-iMVA or OVA plus CFA twice, 2 weeks apart, and subsequently harvested spleens, dLNs, and blood were harvested for anti-OVA CD8+ and CD4+ T-cell and antibody responses as described in Example 1. Subcutaneous co-administration of OVA with Heat-iMVA induced higher levels of antigen-specific CD8+ and CD4+ T-cells compared with immunization with OVA plus CFA in the spleens of vaccinated mice. The percentages of IFN-γ+ T-cells among CD8+ T-cells in the spleens increased from 0.8% in the OVA-treated mice to 1.6% in OVA+Heat-iMVA-treated mice as opposed to 1.0% in OVA+CFA-treated mice (P<0.01; n=5; OVA+Heat-iMVA vs. OVA+CFA; FIG. 2B). The percentages of IFN-γ+ T-cells among CD4+ T-cells in the spleens increased from 0.75% in the OVA-treated mice to 1.6% in OVA+Heat-iMVA-treated mice as opposed to 0.75% in OVA+CFA group (P<0.001; n=5; OVA+Heat-iMVA vs. OVA+CFA; FIG. 2C). Similar differences were observed in dLNs revealing that the subcutaneous co-administration of OVA with Heat-iMVA induced higher levels of antigen-specific CD8+ and CD4+ T-cells compared with immunization with OVA plus CFA in the dLNs of vaccinated mice (P<0.05; n=5; OVA+Heat-iMVA vs. OVA+CFA for IFN-γ CD8+ T-cells; FIG. 2D; P<0.001; n=5; OVA+Heat-iMVA vs. OVA+CFA for IFN-γ CD4+ T-cells; FIG. 2E). IgG1 titers in the serum of OVA+CFA-immunized mice were higher than those in the serum of OVA+Heat-iMVA-immunized mice (P<0.01; n=5; OVA+Heat-iMVA vs. OVA+CFA; FIG. 2F), whereas IgG2c titers in the serum of OVA+CFA-immunized mice were lower than those in the serum of OVA+Heat-iMVA-immunized mice (P<0.01; n=5; OVA+Heat-iMVA vs. OVA+CFA; FIG. 2G). IgG1 is considered as a “Th2-like” isotype, whereas IgG2c is considered as a “Th1-like” isotype. These results indicate that co-administration of OVA with Heat-iMVA promotes the production of the IgG2c isotype. Subcutaneous co-administration of OVA+Heat-iMVA induced higher levels of anti-OVA CD8+ T-cells in the spleens compared with intramuscular co-administration of OVA+Heat-iMVA (P<0.05; n=5; SC OVA+Heat-iMVA vs. IM OVA+Heat-iMVA;



FIG. 2B).


Example 3: The Effect of Heat-iMVA-Mediated Vaccine Adjuvant on Antigen-Specific T-Cell Responses Require Batf3-Dependent DCs

Batf3 is a transcription factor that is critical for the development of CD103+/CD8α+ lineage DCs, which play an important role in cross-presentation of viral and tumor antigens. Batf3-deficient mice are unable to reject highly immunogenic tumors. To test whether STING or Batf3 plays a role in Heat-iMVA-mediated vaccine adjuvant effects, WT C57B/6, STINGGt/Gt, or Batf3−/− mice were subcutaneously vaccinated with OVA+Heat-iMVA twice, two weeks apart. The spleens, dLNs, and blood were then harvested one-week post last vaccination for analyses of cellular and humoral immune responses (FIG. 3A). It was found that the percentages of anti-OVA IFN-γ+ T-cells among CD8+ T-cells induced by Heat-iMVA in the spleens were reduced from 1.2% in immunized WT mice to 0.38% in immunized Batf3−/− mice (P<0.01; n=5; WT vs. Batf3−/−; FIG. 3B). In addition, the percentages of anti-OVA IFN-γ+ T-cells among CD8+ T-cells induced by Heat-iMVA in the dLNs were reduced from 1.3% in immunized WT mice to 0.4% in immunized Batf3−/− mice (P<0.0001; n=5; WT vs. Batf3−/−; FIG. 3D). Batf3-deficiency does not seem to affect the generation of anti-OVA IFN-γ+CD4+ T-cells in the spleens or dLNs (FIGS. 3C and E). These results support the role of Batf3-dependent DCs in cross-presenting OVA antigen to generate OVA-specific CD8+ T-cells in the spleens and dLNs in our vaccination model.


It was also observed that the percentage of anti-OVA IFN-γ+ T-cells among CD8+ T-cells induced by Heat-iMVA in the spleens was reduced from 1.2% in immunized WT mice to 0.97% in immunized STINGGt/Gt mice (P=0.31; n=5; WT vs. STINGGt/Gt; FIG. 3B). The percentage of anti-OVA IFN-γ+ T-cells among CD8+ T-cells induced by Heat-iMVA in the dLNs was reduced from 1.3% in immunized WT mice to 0.98% in immunized STINGGt/Gt mice (P=0.0564; n=5; WT vs. STINGGt/Gt; FIG. 3D). STING-deficiency does not seem to affect the generation of anti-OVA IFN-γ+CD4+ T-cells in the spleens or dLNs (FIGS. 3C and E). The serum IgG2c titers were reduced in the immunized STINGGt/Gt mice compared with WT mice, whereas the serum IgG1 titers were not significantly different between the two groups (FIGS. 3F and G).


Example 4: Co-Administration of MVA-OVA with Heat-iMVA During Scarification Enhances the Generation of OVA-Specific CD8+ T-Cells

MVA is a highly attenuated, non-replicative, safe, and efficacious vaccine vector for various infectious agents and cancers. The optimal dosage for MVA vaccination was tested via skin scarification. MVA-OVA (which encodes full-length of OVA under the control of P7.5 promoter) at doses of 105, 106, and 107 pfu were administered to the tails of 6-8 week old female C57BL/6J mice after skin scarification. One week after vaccination, mice were euthanized and the spleens were isolated for testing antigen-specific CD8+ T-cell responses. Bone marrow-derived DCs (BMDCs) were infected with MVA-OVA at MOI of 5 for 1 h and then incubated for 5 h before the BMDCs were incubated with splenocytes for 12 h. Cells were processed for intracellular cytokine staining (ICS) for IFN-γ+CD8+ T-cells. Alternatively, BMDCs were incubated the SIINFEKL peptide (SEQ ID NO: 1) for 1 h and then incubated with splenocytes for 12 h. ICS was performed for IFN-γ+CD8+ T-cells reactive to SIINFEKL peptide (SEQ ID NO: 1). It was found that with either assay, skin scarification with MVA-OVA at 107 pfu elicited the highest percentages of IFN-γ+CD8+ T-cells among the three doses (P<0.01; n=5; 107 vs. 105 pfu; FIG. 4B, C).


To test whether STING or Batf3-dependent DCs play a role in MVA-induced vaccination effects, MVA at a dose of 106 pfu was also administered to the tails of STINGGt/Gt or Batf3−/− mice after skin scarification. It was found that MVA-OVA induced anti-viral and anti-OVA IFN-γ+CD8+ T-cells were reduced compared with those in immunized WT mice (P<0.05; n=5; WT vs. Batf3−/−; FIG. 4B). A significant defect of generating antigen-specific IFN-γ+CD8+ T-cells in the spleen of STING-deficient mice after MVA-OVA vaccination was not observed (FIGS. 4B and C). To test whether Heat-iMVA provides an adjuvant effect on recombinant MVA-mediated vaccination, Heat-iMVA (with an equivalent amount of 105 pfu) was co-administered with MVA-OVA (107 pfu) to the tails of WT mice after skin scarification. It was found that co-administration of Heat-iMVA (105 pfu) and MVA-OVA (107 pfu) increased anti-viral and anti-OVA IFN-γ+CD8+ T-cells from 0.74% to 0.90% (P=0.461; n=5; MVA-OVA 107 vs. MVA-OVA 107+Heat-iMVA 105; FIG. 4B). In addition, co-administration of Heat-iMVA (105 pfu) and MVA-OVA (107 pfu) increased anti-SIINFEKL (SEQ ID NO: 1) IFN-γ+CD8+ T-cells from 0.87% to 1.57% (P=0.094; n=5; MVA-OVA 107 vs. MVA-OVA 107+Heat-iMVA 105; FIG. 4C). These results indicate that Batf3-dependent DCs are also important for recombinant MVA-induced antigen-specific CD8+ T-cell responses. Accordingly, Heat-iMVA can function as an adjuvant for recombinant MVA-mediated vaccine effects to enhance antigen-specific CD8+ T-cells.


Example 5: Heat-iMVA Induces MHC-I Expression of GM-CSF-Cultured Bone Marrow-Derived Dendritic Cells (BMDCs), but it does not Increase Phagocytosis of Antigen

Infection of BMDCs with Heat-iMVA induces DC maturation that is dependent on the STING-mediated cytosolic DNA-sensing pathway (Dai et al., Science Immunology 2017). In this example, the induction of MHC-I expression on the cell surface of BMDCs by Heat-iMVA was compared with poly I:C. BMDCs were incubated with OVA in the presence or absence of Heat-iMVA for 3 or 16 h, or with poly IC for 16 h. The cell surface MHC-I (H-2Kb) expression was determined by FACS using anti-H-2Kb antibody. It was found that while co-incubation with Heat-iMVA for 3 h did not increase H-2Kb expression, co-incubation with Heat-iMVA for 16 h dramatically increased the cell surface expression of H-2Kb (P<0.001; n=3; OVA+Heat-iMVA 3 h vs. OVA+Heat-iMVA 16 h; FIG. 5A, B). The mean fluorescence intensities of H-2Kb was increased from 1778 on BMDCs co-incubated with OVA alone to 5900 on BMDCs co-incubated with OVA+Heat-iMVA for 16 h vs. 3900 on BMDCs co-incubated with OVA+poly IC (P<0.05; n=3; OVA+Heat-iMVA 16 h vs. OVA+poly IC 16 h; FIG. 5A, B). This result suggests that Heat-iMVA is a stronger inducer of MHC-I expression on BMDCs compared with poly IC.


To assess whether BMDCs' capacity for uptake of fluorescent-labeled model antigen OVA (OVA-647) is affected by Heat-iMVA treatment, BMDC were infected with Heat-iMVA (MOI of 1) for 1 h and then incubated with OVA-647 for 1 h. The fluorescence intensities of phagocytosed OVA-647 in BMDC were measured by flow cytometry. It was found that pre-incubation with Heat-iMVA for 1 h did not affect their capacity to phagocytose OVA-647 (FIG. 5C). By contrast, when BMDC were infected or mock-infected with Heat-iMVA (MOI of 1) for 16 h and then incubated with OVA-647 for 1 h, the fluorescence intensity of phagocytosed OVA-647 in Heat-iMVA-treated BMDCs was reduced compared with that in mock-treated BMDCs (FIG. 5D). These results indicate that although Heat-iMVA-treated BMDCs undergo maturation, their capacity to phagocytose antigen is reduced as a consequence of maturation.


Example 6: Co-Incubation of GM-CSF-Cultured BMDCs with Heat-iMVA and OVA Enhances the Proliferation of OT-I and OT-II T-Cells In Vitro

Infection of epidermal dendritic cells with live WT vaccinia inhibits DCs' capacity to activate antigen-specific T-cells (Deng et al., JVI, 2006). To test whether Heat-iMVA infection of BMDCs enhances the proliferation of antigen-specific OT-I and OT-II T-cells, BMDCs were incubated with OVA at various concentrations in the presence or absence of Heat-iMVA for 3 h. Cells were washed to remove unabsorbed OVA or virus, and then co-cultured with Carboxyfluorescein Diacetate Succinimidyl Ester (CFSE)-labeled OT-I T-cells for 3 days (BMDC:OT-I T-cells=1:5). Flow cytometry was applied to measure CFSE intensities of OT-I cells. It was found that pre-incubation with Heat-iMVA enhanced the capacity of DCs to stimulate the proliferation of OT-I T-cells, as indicated by CSFE dilution in dividing cells (FIGS. 6A and B). It was also found that pre-treatment with heat-iMVA or poly IC modestly enhances DCs' capacity to stimulate the proliferation of OT-II T-cells that recognize OVA-antigen presented by MHC-II on DCs (FIGS. 7A and B).


Example 7: Co-Incubation of Flt3L-Cultured BMDCs with Heat-iMVA and OVA Dramatically Enhances the Proliferation of OT-I T-Cells In Vitro

FMS-like tyrosine kinase 3 ligand (Flt3L) is a critical growth factor for the differentiation of Batf3-dependent CD103+/CD8α+ DCs and plasmacytoid DCs (pDCs). Flt3L-cultured BMDCs were pulsed with OVA in the presence or absence of Heat-iMVA, and then co-cultured with CFSE-labeled OT-I cells for 3 days (BMDC:OT-I=1:5). Flow cytometry was applied to measure CFSE intensities of OT-I cells. It was found that Heat-iMVA potently stimulated the proliferation of OT-I cells, which recognizes OVA257-264(SIINFEKL) peptide (SEQ ID NO: 1) presented on MHC-I, even at very low concentrations of OVA (FIG. 8).


Example 8: Plasmacytoid Dendritic Cells (pDCs) Play Important Role in Heat-iMVA-Mediated Vaccine Adjuvant Effects

The results described herein indicate that Flt3L-cultured DCs are more efficient in cross-present OVA antigen to stimulate OT-I T-cell proliferation than GM-CSF-cultured DCs. Flt3L-cultured DCs generate plasmacytoid DCs (pDCs), which are potent type I IFN producing cells that can be activated by Heat-inactivated vaccinia via the MyD88-dependent endosomal toll-like receptor 7 and 9 (Cao et al., 2012, PLoS One). pDCs can also cross-present antigen to stimulate CD8+ T-cell responses. To test whether pDCs play a role in Heat-iMVA-mediated adjuvant effect in vivo, anti-PDCA-1 antibody was used one day prior and one day post intradermal immunization with OVA+Heat-iMVA, which were performed on Day 0 and Day 14. Spleens and dLNs were isolated on day 21 for antigen-specific CD8+ T-cell analyses (FIG. 9A). It was found that intradermal co-administration of OVA+Heat-iMVA increased the percentage of IFN-γ+ T-cells among CD8+ T-cells in the spleens from 0.097% in the OVA-treated mice to 0.16% in OVA+Heat-iMVA-treated mice (P<0.001; n=5; OVA+Heat-iMVA; FIG. 9B). Depletion of pDCs resulted in dramatic decrease in the percentage of IFN-γ+ T-cells among CD8+ T-cells in the spleens (P<0.001; n=5; OVA+Heat-iMVA+control IgG vs. OVA+Heat-iMVA+anti-PDCA-1; FIG. 9B). Similar results were obtained in dLNs (FIG. 9C). These results support a critical role of pDCs in Heat-iMVA-elicited vaccine adjuvant effects in a peptide vaccination model in vivo.


Example 9: Migratory Dendritic Cell Subsets LangerinCD11b and CD11b+ DCs are Efficient in OVA Antigen Uptake

Many DC subsets are present in the lymph nodes, which include migratory DCs and resident DCs. Migratory DCs are MHC-II+CD11c+. Resident dendritic cell populations are MHC-IIIntCD11c+. Migratory DCs can be further separated into CD11b+ DC, Langerin CD11b DC, and Langerin+ DC. Langerin+ DCs comprise of CD103+ DC and Langerhans cells, whereas resident DCs are composed of CD8α+ resident DC and CD8α″ resident DC (FIG. 10A). To test which DCs subsets are efficient in phagocytosing OVA antigen labeled with fluorescent dye (OVA-647) and have the capacity to migrate to the dLNs, OVA-647 were injected intradermally (ID) to the right flank and harvested the dLNs at 24 h post injection. It was found that Langerin CD11b and CD11b+ DCs were two prominent migratory DC subsets that carry OVA-647 to the dLNs (FIG. 10B). To compare whether co-administration of OVA-647 with or without vaccine adjuvants Addavax or Heat-iMVA affected the percentages of OVA-647+ cells among LangerinCD11b and CD11b+ DCs, OVA-647 was intradermally (ID) injected with or without Addavax or Heat-iMVA, and analyzed OVA-647+ DCs among LangerinCD11b and CD11b+ DCs. It was found that co-administration of OVA with Heat-iMVA increased the percentages of OVA-647+ cells among LangerinCD11b and CD11b+ DCs, whereas co-administration of OVA with Addavax failed to do the same (**P<0.01; n=3; OVA+Heat-iMVA vs. OVA+Addavax; FIG. 10C, *P<0.05; n=3; OVA+Heat-iMVA vs. OVA+Addavax; FIG. 10D). Addavax is a well-accepted squalene-based oil-in-water nano-emulsion with a formulation similar to MF59 that has been licensed in Europe for adjuvanted flu vaccines. These results suggest that co-administration of OVA-647 with Heat-iMVA enhances migratory DCs' capacity to transport phagocytosed antigen to the dLNs.


Example 10: Heat-iMVA is a Potent Immune Adjuvant for Irradiated Whole Cell Vaccine

The advantage of using irradiated whole cell vaccines rather than peptide tumor antigen or neoantigen include: (i) tumor cells provide multiple tumor antigens that can be recognized by the host immune system; and (ii) can bypass the need or time to identify tumor antigens or neoantigens. Whether the addition of Heat-iMVA with irradiated B16-OVA improves vaccination efficacy, and whether systemic delivery of anti-PD-L1 would further improve vaccination efficacy was analyzed. Mice were intradermally implanted with B16-OVA, they were vaccinated intradermally with irradiated B16-OVA, B16-OVA+Heat-iMVA, or B16-OVA+poly IC three times at day 3, 6, and 9 on the contralateral flank. It was found that vaccination with irradiated B16-OVA+Heat-iMVA extended the median survival from 16 days (with irradiated B16-OVA vaccination) to 23 days (**, P<0.01; N=5; Irradiated B16-OVA+Heat-iMVA vs. Irradiated B16-OVA alone; FIG. 11B). In the presence of anti-PD-L1 antibody, vaccination with irradiated B16-OVA+Heat-iMVA extended the median survival from 20 days to 27 days (**, P<0.01; N=10; Irradiated B16-OVA+Heat-iMVA+anti-PD-L1 vs. Irradiated B16-OVA+anti-PD-L1; FIG. 11C). No statistic difference in median survival was observed between Irradiated B16-OVA+Heat-iMVA and Irradiated B16-OVA+poly IC groups. Mice received poly IC lost more weight than Heat-iMVA-treated mice, which is suggestive of systemic inflammation and toxicity (data not shown). These results indicate that Heat-iMVA is a potent and safe vaccine adjuvant for irradiated whole cell vaccination.


Example 11: Heat-iMVA is an Immune Adjuvant for Neoantigen Peptide Vaccination

To test whether Heat-iMVA can act as a vaccine adjuvant for neoantigen peptide vaccination, a subcutaneous vaccination model was used in which mice were first implanted with B16-F10 cells (7.5×104 cells per mouse) intradermally. At day 3 and 7 post implantation, mice were vaccinated at the contralateral flank subcutaneously (SC) with a mixture of neoantigen peptides (M27 (REGVELCPGNKYEMRRHGTTHSLVIHD) (SEQ ID NO: 4), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL) (SEQ ID NO: 5), and M48 (SHCHWNDLAVIPAGVVHNWDFEPRKVS) (SEQ ID NO: 6)) with or without either Heat-iMVA or poly I:C. Tumor growth and mice survival were monitored. It was found that SC vaccination with neoantigen peptides alone generates systemic antitumor immunity (FIGS. 12A-12C). The antitumor effect is enhanced when neoantigen peptide mix were co-administered with Heat-iMVA (FIGS. 12A-12C).


Example 12: Heat-iMVA is an Immune Adjuvant for Viral Antigen Peptide Vaccination

Viral antigens are potent immunogens that can be recognized by the host immune system. To test whether the combination of Heat-iMVA or Heat-inactivated vaccinia and viral antigen (such as synthetic long peptide (SLP) of human papilloma virus E7) elicits antiviral T cells, mice are subcutaneously vaccinated with E7 SLP alone, or E7 SLP plus Heat-iMVA, or E7 plus poly I:C twice, 2 weeks apart, and subsequently harvested spleens, dLNs, and blood are harvested for anti-CD8+ and CD4+ T-cell and antibody responses. To test the role of Heat-iMVA in the therapeutic vaccination model, E7-expressing cancer cells are implanted intradermally, and then the vaccination is performed with or without adjuvant two weeks apart, and tumor volumes are analyzed in mice.


Example 13: Determining Whether Intratumoral (IT) Vaccination is Superior to Subcutaneous (SC) Vaccination in Generating Antigen-Specific Immune Responses

It has been shown that intratumoral (IT) injection of Heat-iMVA eradicates injected tumors and induces systemic antitumor immunity, which requires Batf3-dependent CD103+/CD8 □+ DCs and STING-mediated cytosolic DNA-sensing pathway. IT delivery of Heat-iMVA alters the tumor immunosuppressive microenvironment partially through activating cGAS/STING pathway and promotes tumor antigen presentation by the CD103+ DCs. It is anticipated that IT delivery of Heat-iMVA plus model antigen or neoantigen will enhance antigen presentation by tumor-infiltrating DCs and generate superior adaptive immunity compared with SC delivery of Heat-iMVA plus antigen.


To test whether IT vaccination is superior to SC vaccination in generating antigen-specific immune responses, B16-F10 melanoma cells (5×105 cells) are implanted intradermally at the right flank. At day 7 post implantation, when the tumors are 2-3 mm in diameter, Heat-iMVA and OVA protein will either be directly injected into the tumors or injected SC 1 cm away from the tumors on the right flank. At one-week post injection, TDLNs and spleens will be collected and anti-OVA CD4 and CD8 T cells will be analyzed by FACS.


Alternatively, B16-F10 neoantigen peptide mix (M27/M30/M48) will be co-injected with Heat-iMVA either directly into the tumors on the right flank, or injected SC 1 cm away from the tumors on the right flank. At one-week post injection, TDLNs and spleens will be collected and co-cultured with either M27, M30, or M48 peptide for 16 h for ELISPOT analysis.


EQUIVALENTS

The present technology is not to be limited in terms of the particular embodiments described in this application, which are intended as single illustrations of individual aspects of the present technology. Many modifications and variations of this present technology can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods and apparatuses within the scope of the present technology, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. The present technology is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. It is to be understood that this present technology is not limited to particular methods, reagents, compounds compositions or biological systems, which can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting.


In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.


As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like, include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 cells refers to groups having 1, 2, or 3 cells. Similarly, a group having 1-5 cells refers to groups having 1, 2, 3, 4, or 5 cells, and so forth.


All patents, patent applications, provisional applications, and publications referred to or cited herein are incorporated by reference in their entirety, including all figures and tables, to the extent they are not inconsistent with the explicit teachings of this specification.


Other embodiments are set forth within the following claims.

Claims
  • 1. A method for treating a solid tumor in a subject in need thereof, the method comprising administering to the subject an immunogenic composition comprising an antigen and a therapeutically effective amount of an adjuvant comprising an inactivated modified vaccinia Ankara virus and/or an inactivated vaccinia virus.
  • 2. The method of claim 1, wherein the inactivated modified vaccinia Ankara virus is either a Heat-inactivated modified vaccinia Ankara virus (Heat-iMVA) or a UV-inactivated MVA, and the inactivated vaccinia virus is either a Heat-inactivated vaccinia virus or a UV-inactivated vaccinia virus.
  • 3. The method of claim 2, wherein the inactivated modified vaccinia virus is Heat-iMVA.
  • 4. The method of claim 1, 2, or 3, wherein the antigen is selected from the group consisting of tumor differentiation antigens, cancer testis antigens, neoantigens, viral antigens in the case of tumors associated with oncogenic virus infection, GPA33, HER2/neu, GD2, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, MUM-1, CDK4, N-acetylglucosaminyltransferase, p15, gp75, beta-catenin, ErbB2, cancer antigen 125 (CA-125), carcinoembryonic antigen (CEA), RAGE, MART (melanoma antigen), MUC-1, MUC-2, MUC-3, MUC-4, MUC-5ac, MUC-16, MUC-17, tyrosinase, tyrosinase-related proteins 1 and 2, Pmel 17 (gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate cancer psm, PRAME (melanoma antigen), β-catenin, EBNA (Epstein-Barr Virus nuclear antigen) 1-6, p53, kras, lung resistance protein (LRP) Bcl-2, prostate specific antigen (PSA), Ki-67, CEACAM6, colon-specific antigen-p (CSAp), NY-ESO-1, human papilloma virus E6 and E7, and combinations thereof.
  • 5. The method of any one of claims 1-4, wherein the administration step comprises administering the immunogenic composition in one or more doses, wherein the administration step comprises administering the immunogenic composition in one or more doses, and/or wherein the antigen and the adjuvant are administered separately, sequentially, or simultaneously.
  • 6. The method of any one of claims 1-5, further comprising administering to the subject an immune checkpoint blockade agent selected from the group consisting of cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors, programmed death 1 (PD-1) inhibitors, PD-L1 inhibitors, and PD-L2 inhibitors.
  • 7. The method of claim 6, wherein the immunogenic composition is delivered to the subject separately, sequentially, or simultaneously with the administration of the immune checkpoint blockade agent.
  • 8. The method of claim 6 or 7, wherein the PD-L1 inhibitor is an anti-PD-L1 antibody.
  • 9. The method of any one of claims 1-8, wherein treatment comprises one or more of the following: inducing an immune response in the subject against the tumor or enhancing or promoting an ongoing immune response against the tumor in the subject, reducing the size of the tumor, eradicating the tumor, inhibiting growth of the tumor, inhibiting metastatic growth of the tumor, inducing apoptosis of the tumor cells, or prolonging survival of the subject.
  • 10. The method of claim 9, wherein the induction, enhancement, or promotion of the immune response comprises one or more of the following: increased levels of interferon gamma (IFN-γ) expression in T-cells in the spleen, draining lymph nodes, and/or serum as compared to an untreated control sample;increased levels of antigen-specific T-cells in the spleen, draining lymph nodes, and/or serum as compared to an untreated control sample; andincreased levels of antigen-specific immunoglobulin in serum as compared to an untreated control sample.
  • 11. The method of claim 10, wherein the antigen-specific immunoglobulin is IgG1 or IgG2.
  • 12. The method of any one of claims 1-11, wherein the immunogenic composition is formulated to be administered intratumorally, intramuscularly, intradermally, or subcutaneously.
  • 13. The method of any one of claims 1-12, wherein the tumor is selected from the group consisting of melanoma, colorectal cancer, breast cancer, prostate cancer, lung cancer, pancreatic cancer, ovarian cancer, squamous cell carcinoma of the skin, Merkel cell carcinoma, gastric cancer, liver cancer, and sarcoma.
  • 14. The method of any one of claims 1-13, wherein the inactivated modified vaccinia Ankara virus or inactivated vaccinia virus is administered at a dosage per administration of about 105 to about 1010 plaque-forming units (pfu).
  • 15. The method of any one of claims 1-14, wherein the subject is human.
  • 16. An immunogenic composition comprising an antigen and an adjuvant comprising an inactivated modified vaccinia Ankara virus and/or an inactivated vaccinia virus.
  • 17. The immunogenic composition of claim 16, wherein the inactivated modified vaccinia Ankara virus is either a Heat-inactivated modified vaccinia Ankara virus (Heat-iMVA) or a UV-inactivated MVA, and the inactivated vaccinia virus is either a Heat-inactivated vaccinia virus or a UV-inactivated vaccinia virus.
  • 18. The immunogenic composition of claim 17, wherein the inactivated modified vaccinia virus is Heat-iMVA.
  • 19. The immunogenic composition of claim 16, 17, or 18, further comprising a pharmaceutically acceptable carrier.
  • 20. The immunogenic composition of any one of claims 16-19, wherein the antigen is selected from the group consisting of tumor differentiation antigens, cancer testis antigens, neoantigens, viral antigens in the case of tumors associated with oncogenic virus infection, GPA33, HER2/neu, GD2, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, MUM-1, CDK4, N-acetylglucosaminyltransferase, p15, gp75, beta-catenin, ErbB2, cancer antigen 125 (CA-125), carcinoembryonic antigen (CEA), RAGE, MART (melanoma antigen), MUC-1, MUC-2, MUC-3, MUC-4, MUC-5ac, MUC-16, MUC-17, tyrosinase, tyrosinase-related proteins 1 and 2, Pmel 17 (gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate cancer psm, PRAME (melanoma antigen), β-catenin, EBNA (Epstein-Barr Virus nuclear antigen) 1-6, p53, kras, lung resistance protein (LRP) Bcl-2, prostate specific antigen (PSA), Ki-67, CEACAM6, colon-specific antigen-p (CSAp), NY-ESO-1, human papilloma virus E6 and E7, and combinations thereof.
  • 21. The immunogenic composition of any one of claims 16-20, further comprising an immune checkpoint blockade agent selected from the group consisting of cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors, programmed death 1 (PD-1) inhibitors, PD-L1 inhibitors, and PD-L2 inhibitors.
  • 22. The immunogenic composition of claim 21, wherein the PD-L1 inhibitor is an anti-PD-L1 antibody.
  • 23. The immunogenic composition of any one of claims 16-22, wherein the inactivated modified vaccinia Ankara virus or inactivated vaccinia virus is administered at a dosage per administration of about 105 to about 1010 plaque-forming units (pfu).
  • 24. A kit comprising instructions for use, a container means, and a separate portion of each of: (a) an antigen; and (b) an adjuvant comprising inactivated modified vaccinia Ankara virus and/or inactivated vaccinia virus.
  • 25. The kit of claim 24, wherein the inactivated modified vaccinia Ankara virus is either a Heat-inactivated modified vaccinia Ankara virus (Heat-iMVA) or a UV-inactivated MVA, and the inactivated vaccinia virus is either a Heat-inactivated vaccinia virus or a UV-inactivated vaccinia virus.
  • 26. The kit of claim 25, wherein the inactivated modified vaccinia virus is Heat-iMVA.
  • 27. The kit of any one of claims 24-26, wherein the antigen is selected from the group consisting of tumor differentiation antigens, cancer testis antigens, neoantigens, viral antigens in the case of tumors associated with oncogenic virus infection, GPA33, HER2/neu, GD2, MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, MUM-1, CDK4, N-acetylglucosaminyltransferase, p15, gp75, beta-catenin, ErbB2, cancer antigen 125 (CA-125), carcinoembryonic antigen (CEA), RAGE, MART (melanoma antigen), MUC-1, MUC-2, MUC-3, MUC-4, MUC-5ac, MUC-16, MUC-17, tyrosinase, tyrosinase-related proteins 1 and 2, Pmel 17 (gp100), GnT-V intron V sequence (N-acetylglucoaminyltransferase V intron V sequence), Prostate cancer psm, PRAME (melanoma antigen), β-catenin, EBNA (Epstein-Barr Virus nuclear antigen) 1-6, p53, kras, lung resistance protein (LRP) Bcl-2, prostate specific antigen (PSA), Ki-67, CEACAM6, colon-specific antigen-p (CSAp), NY-ESO-1, human papilloma virus E6 and E7, and combinations thereof.
  • 28. The kit of any one of claims 24-27, wherein the kit further comprises an immune checkpoint blockade agent selected from the group consisting of cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors, programmed death 1 (PD-1) inhibitors, PD-L1 inhibitors, and PD-L2 inhibitors.
  • 29. The kit of claim 28, wherein the immune checkpoint blockade agent comprises is a PD-L1 inhibitor, which is an anti-PD-L1 antibody.
  • 30. The method of any one of claims 4-14, wherein the antigen comprises a neoantigen selected from the group consisting of M27 (REGVELCPGNKYEMRRHGTTHSLVIHD) (SEQ ID NO: 4), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL) (SEQ ID NO: 5), M48 (SHCHWNDLAVIPAGVVHNWDFEPRKVS) (SEQ ID NO: 6), and combinations thereof.
  • 31. The immunogenic composition of any one of claims 20-23, wherein the antigen comprises a neoantigen selected from the group consisting of M27 (REGVELCPGNKYEMRRHGTTHSLVIHD) (SEQ ID NO: 4), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL) (SEQ ID NO: 5), M48 (SHCHWNDLAVIPAGVVHNWDFEPRKVS) (SEQ ID NO: 6), and combinations thereof.
  • 32. The kit of any one of claims 27-29, wherein the antigen comprises a neoantigen selected from the group consisting of M27 (REGVELCPGNKYEMRRHGTTHSLVIHD) (SEQ ID NO: 4), M30 (PSKPSFQEFVDWENVSPELNSTDQPFL) (SEQ ID NO: 5), M48 (SHCHWNDLAVIPAGVVHNWDFEPRKVS) (SEQ ID NO: 6), and combinations thereof.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a National Stage Application of PCT/US2018/059476, filed Nov. 6, 2018, which claims the benefit of and priority to U.S. Provisional Patent Application No. 62/582,263, filed Nov. 6, 2017, the entire contents of which are incorporated herein by reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/US18/59476 11/6/2018 WO 00
Provisional Applications (1)
Number Date Country
62582263 Nov 2017 US