RNA VIRUSES EXPRESSING IL-12 FOR IMMUNOVIROTHERAPY

The present invention relates to a recombinant virus of the family Paramyxoviridae, comprising at least one expressible polynucleotide encoding an IL-12 polypeptide, wherein said IL-12 polypeptide is an IL-12 fusion polypeptide comprising a p35 subunit of an IL-12 and a p40 subunit of an IL-12; to a polynucleotide encoding the same, and to a kit comprising the same. Moreover, the present invention relates to a method for treating cancer in a subject afflicted with cancer, comprising contacting said subject with a recombinant virus of the family Paramyxoviridae of the invention, and thereby, treating cancer in a subject afflicted with cancer.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATION

This application is a Continuation of U.S. application Ser. No. 15/441,545, filed Feb. 24, 2017; which claims the benefit of U.S. Provisional Application No. 62/299,788, filed Feb. 25, 2016, which application is incorporated herein by reference.

FIELD OF THE INVENTION

This invention relates to biotechnology, including modified viruses, especially for the treatment or prevention of a human disease.

BACKGROUND

Interleukin 12 (IL-12) is a heterodimeric polypeptide interleukin consisting of two subunits, p35 and p40, encoded by two separate genes, IL-12A and IL-12B, respectively. IL-12 is produced in response to immune stimuli by dendritic cells, macrophages, neutrophils, and by human B-lymphoblastoid cells, and has been known as an important stimulator of immune cell activity, in particular of T cells and natural killer cells, which are, among other effects, stimulated to secrete IFN-γ by IL-12. IFN-γ, in turn, is known to stimulate expression of immune checkpoint blockade proteins on non-immune cells, e.g. of PD-L1, a mechanism used by cancer cells to evade the immune system (Abiko et al. (2015), British journal of cancer 112(9): 1501; Quetglas et al. (2015) Cancer Research 75(15 Supplement): 281). Due to the known immunostimulatory effects of IL-12, it was attempted to use a recombinant measles virus expressing both subunits of IL-12 as a vaccine to improve immune response against measles virus. However, it was found that the transgene had a detrimental effect on the neutralizing antibody response and that lymphoproliferative responses were not improved (Hoffman et al. (2003), J Infect Dis 188:1553).

Oncolytic viruses (OV) which replicate selectively in tumor cells are an emerging modality of cancer treatment. Aside from direct cytopathic effects and lysis of tumor cells, interactions of OV with the immune system can trigger systemic anti-tumor immunity. OV have been modified to express immunomodulatory transgenes to further enhance these effects (Melcher et al., Mol Ther. 2011, 19: 1008-1016). The vaccinia virus JX-594 and herpesvirus talimogene laherpavec (TVEC), both harboring GM-CSF, have shown promising results in clinical phase II and III trials (Heo et al., Nat Med. 2013,19: 329-336 and Andtbacka et al. J Clin Oncol. 2013, 31, suppl; abstr LBA9008).

RNA viruses, in particular members of the family Paramyxoviridae like, e.g. measles virus (MV), have also shown potential use in oncolysis. Viruses of the family Paramyxoviridae are negative-sense single-stranded RNA viruses and include human pathogens like, e.g. human parainfluenza viruses, mumps virus, human respiratory syncytial virus, and measles virus. From wild type measles virus, several non-pathogenic strains, including a vaccine strain, have been derived, which have been shown to remain oncolytic. The measles virus vaccine strain has been developed as a vector platform to target multiple tumor entities and several clinical trials are ongoing (Russell et al., Nat Biotechnol. 2012, 30: 658-670). Recently, the capacity of oncolytic MV encoding GM-CSF to support the induction of a specific anti-tumor immune response in terms of a tumor vaccination effect was demonstrated (Grossardt et al. Hum Gene Ther. 2013, 24: 644-654.).

There is, however, still a need in the art for improved cancer therapies, in particular for improved oncolytic virus therapies. It is therefore an objective of the present invention to provide an improved oncolytic virus, which fully or partially avoids the short-comings of known oncolytic viruses.

SUMMARY OF THE INVENTION

The present invention relates to a recombinant virus of the family Paramyxoviridae, comprising at least one expressible polynucleotide encoding an IL-12 polypeptide, wherein said IL-12 polypeptide is an IL-12 fusion polypeptide comprising a p35 subunit of an IL-12 and a p40 subunit of an IL-12; to a polynucleotide encoding the same, and to a kit comprising the same. Moreover, the present invention relates to a method for treating cancer in a subject afflicted with cancer, comprising contacting said subject with a recombinant virus of the family Paramyxoviridae of the invention, and, thereby, treating cancer in a subject afflicted with cancer.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1: One step growth curves in Vero-αHis (A) and MC38cea (B) cells: Cells were transduced with MeVac encoding the respective transgenes at MOI=3. Cell suspensions were collected by scraping in the culture medium and titre determined at the depicted time points.

FIG. 2: Cytotoxic effect in the target MC8cea cells: Cells were transduced with MeVac encoding the respective transgenes at MOI=5 and cell viability was determined by XTT assay at the depicted time points. Mean results of triplicate infections per time point with standard errors of the mean (not visible for some data points) are shown.

FIG. 3: Expression of MeVac encoded immunomodulators in MC38cea cells. MC38cea cells were transduced with MeVac encoding the respective immunomodulators and eGFP or IgG1-Fc as control vectors at MOI=3. Supernatant samples were collected at the depicted time points and transgene expression detected by ELISA. Unspecific binding was controlled by IgG1-Fc (upper panels) or eGFP (lower panels) supernatants and subtracted from the specific measurements. In case of mIP-10 an increase of the signal was observed in the eGFP controls which was not subtracted from the specific measurements and is depicted accordingly.

FIG. 4: MeVac encoded anti-PD-L1 binding to MC38cea cells. MC38cea cells were incubated with supernatant from Vero-αHis infected with MeVac encoding anti-PD-L1 or IgG1-Fc. For detection of bound anti-PD-L1, cells were stained with primary Ab specific for HA tag and secondary Ab coupled to PE. DAPI staining was used to exclude dead cells and samples were analyzed by flow cytometry. (A) Overlay histogram for PE of DAPI- MC38cea populations from one of three independent experiments is shown on panel (B). (B)Average median fluorescence intensity (MFI) of PE for DAPI-populations with standard error of the means from the three independent experiments is shown on the left panel.

FIG. 5: Functionality of MeVac encoded immunomodulators. (A) MC38cea cells were treated with supernatants from Vero-αHis cells infected with MeVac encoding the respective immunomodulators and cocultured in ratio 2:1 with murine splenocytes in the presence of PMA and ionomycin in 96-well plate. After 24 h the supernatants were collected and IFN-γ concentration measured by ELISA. Relative activation corresponds to ratio of the optical density (absorbance at 450 nm minus 570 nm) of the respective samples to activated splenocytes. Data for one of three independent experiments are shown; (B) splenocytes were stimulated with recombinant murine IL-2 and cultivated in the presence of medium from Vero-αHis infected with MeVac encoding FmIL-12 or eGFP. After 48 h the supernatants were collected and IFN-γ concentration measured by ELISA. Mean results with standard error of the mean of triplicate splenocyte cultures per FmIL-12 concentration are shown. IFN-γ concentration in the eGFP controls was close to background.

FIG. 6: Therapeutic efficacy of immunomodulatory MeVac in vivo: Breaking immunosuppression: MC38cea cells were implanted subcutaneously (s.c.) into the right flank of C57BL/6J mice (6-9 animals per group). When tumors reached an average volume of 50 mm3 mice received intratumoral injections with 1×106 cell infectious units (ciu) with the respective viruses on four consecutive days in 100 Tumor volume was determined every third day and mice were sacrificed when tumor volumes exceeded 1500mm3 or when ulceration occurred.

FIG. 7: Therapeutic efficacy of immunomodulatory MeVac in vivo: Activating DCs and effector cells: Therapeutic. MC38cea cells were implanted subcutaneously (s.c.) into the right flank of C57BL/6J mice (6-9 animals per group). When tumors reached an average volume of 50 mm3 mice received intratumoral injections with 5×105 ciu with the respective viruses on five consecutive days in 100 μl. Tumor volume was determined every third day and mice were sacrificed when tumor volumes exceeded 1500 mm3 or when ulceration occurred.

FIG. 8: Rechallenge of long term survivors with MC38cea. Mice experiencing complete tumor remission in the experiments identifying the most effective MeVac vectors and were rechallenged with MC38cea cells 3 to 6 months after the initinal tumor cell implantation. Eight I mice served as a control group. 1×105 MC38cea cells were implanted subcutaneously (s.c.) in the left flank of the mice. Tumor engraftment rates were monitored.

FIG. 9: Comparison of therapeutic efficacy of MeVac encoding FmIL-12 and anti-PD-L1. MC38cea cells were implanted subcutaneously (s.c.) into the right flank of C57BL/6J mice (10 animals per group). When tumors reached an average volume of 50 mm3 mice received intratumoral injections with 1×106 ciu with the respective viruses on four consecutive days in 100 μl. Tumor volume was determined every third day and mice were sacrificed when tumor volumes exceeded 1500 mm3 or when ulceration occurred.

FIG. 10: Rechallenge of long term survivors from the experiment comparing efficacy of FmIL-12 and anti-PD-L1 encoding vectors with MC38cea. Mice were rechallenged with MC38cea cells ca. 6 months after the initinal tumor cell implantation. Ten I mice served as a control group. 1×105 MC38cea cells were implanted subcutaneously (s.c.) in the left flank of the mice. Tumor engraftment rates were monitored.

FIG. 11: IFN-γ memory recall in murine splenocytes from mice experiencing complete tumor remissions in MeVac FmIL-12 versus MeVac anti-PD-L1 efficacy experiment. Anti-tumor: Freshly isolated splenocytes from mice treated with MeVac encoding the respective immunomodulators or naïve mice were stimulated with recombinant murine IL-2 and cocultivated with MC38cea (A) or MC38 (B) and B16 (B) tumor cells or irrelevant human cell lysate (DLD-1). After 48 h of cultivation cell culture medium was collected and IFN-γ concentration was measured by ELISA. IFN-γ concentrations in the individual cocultures with median in the group (A) or average concentration from two replicate measurements with standard error of the mean (SEM) are shown (B).

FIG. 12: IFN-γ memory recall in murine splenocytes from mice experiencing complete tumor remissions in MeVac FmIL-12 versus MeVac anti-PD-L1 efficacy experiment: Anti-MeVac: Freshly isolated splenocytes from mice treated with MeVac encoding the respective immunomodulators or naïve mice were stimulated with recombinant murine IL-2 and cocultivated with MeVac (A) or as controls with an irrelevant human cell lysate (DLD-1) (B) or as a negative control splenocytes were cultivated alone. After 48 h of cultivation cell culture medium was collected and IFN-γ concentration was measured by ELISA. IFN-γ concentrations in the individual cocultures with median in the group (A) or average concentration from two replicate measurements with standard error of the mean (SEM) are shown (B).

FIG. 13: Comparison of therapeutic efficacy of combination of MeVac encoding Fm-IL-12 and anti-PD-L1 with either vector in combination with a control vector encoding IgG1-Fc. MC38cea cells were implanted subcutaneously (s.c.) into the right flank of C57BL/6J mice (8-10 animals per group). When tumors reached an average volume of 50 mm3 mice received intratumoral injections with 1×106 ciu with the respective viruses on four consecutive days in 100 Tumor volume was determined every third day and mice were sacrificed when tumor volumes exceeded 1500 mm3 or when ulceration occurred.

FIG. 14: Rechallenge of long term survivors from the experiment comparing efficacy of combination of MeVac encoding Fm-IL-12 and anti-PD-L1 with either vector in combination with a control vector encoding IgG1-Fc. Mice were rechallenged with MC38cea cells ca. 5 months after the initinal tumor cell implantation. Ten I mice served as a control group. 1×105 MC38cea cells were implanted subcutaneously (s.c.) in the left flank of the mice. Tumor engraftment rates were monitored.

FIG. 15: IFN-γ memory recall in murine splenocytes from mice experiencing complete tumor remissions in MeVac FmIL-12 and MeVac anti-PD-L1 combination experiment: Anti-tumor: Freshly isolated splenocytes from mice treated with MeVac encoding the respective immunomodulators or naïve mice were stimulated with recombinant murine IL-2 and cocultivated with MC38cea (A), MC38 (B) or B16 (C) tumor cells. After 48 h of cultivation cell culture medium was collected and IFN-γ concentration was measured by ELISA. IFN-γ concentrations in the individual cocultures with median in the group (A) or average concentration from two replicate measurements with standard error of the mean (SEM) are shown (B).

FIG. 16: IFN-γ memory recall in murine splenocytes from mice experiencing complete tumor remissions in MeVac FmIL-12 and MeVac anti-PD-L1 combination experiment: Anti-MeVac: Freshly isolated splenocytes from mice treated with MeVac encoding the respective immunomodulators or naïve mice were stimulated with recombinant murine IL-2 and cocultivated with MeVac (A) or Vero-αHis lysate (B). After 48 h of cultivation cell culture medium was collected and IFN-γ concentration was measured by ELISA. IFN-γ concentrations in the individual cocultures with median in the group (A) or average concentration from two replicate measurements with standard error of the mean (SEM) are shown (B).

FIG. 17: Comparison of therapeutic efficacy of combination of MeVac encoding Fm-IL-12 and anti-PD-L1 with either vector in combination with a control vector encoding IgG1-Fc. B16-CD20 cells were implanted subcutaneously (s.c.) into the right flank of C57BL/6J mice (8-10 animals per group). When tumors reached an average volume of 50 mm3 mice received intratumoral injections with 1×106 ciu with the respective viruses on four consecutive days in 100 μl. Tumor volume was determined every third day and mice were sacrificed when tumor volumes exceeded 1500 mm3 or when ulceration occurred.

FIG. 18: Schemes of the constructed recombinant MeVac genomes. Transgenes encoding different immunomodulators as well as eGFP and IgG1-Fc as controls were inserted in different positions of MeVac genome. Murine IL-12 was inserted as a fusion protein consisting of p40 and p35 protein subunits linked by a (Gly4Ser)3 linker (FmIL-12). Murine CD80 was inserted as a soluble form of the protein consisting of the extracellular part of the protein fused to a human IgG1-Fc (CD80-Fc). The MeVac H gene in the novel constructs was fully retargeted to human CEA (hCEA) antigen by ablating attachment to the natural receptors, fusing the H protein to a single chain antibody (7cab) against the hCEA and including a six-histidine tag at the C terminus to allow specific transduction of murine MC38cea cells via human CEA antigen and Vero-αHis cells via anti-His 7cab.

FIG. 19: Scheme of MeVac genomes encoding FmIL-12 or anti-PD-L1 or IgG1-Fc retargeted to human CD20. MeVac H gene was fully retargeted to human CD20 antigen by ablating attachment to the natural receptors, fusing the H protein to a single chain antibody (7cab) against the CD20 and including a six-histidine tag at the C terminus to allow specific transduction of murine melanoma B16-CD20 cells via human CD20 antigen and Vero-αHis cells via anti-His 7cab.

DETAILED DESCRIPTION OF THE INVENTION

Accordingly, the present invention relates to a recombinant virus of the family Paramyxoviridae, comprising an expressible polynucleotide encoding an IL-12 polypeptide, wherein said IL-12 polypeptide is an IL-12 fusion polypeptide comprising a p35 subunit of an IL-12 and a p40 subunit of an IL-12.

As used in the following, the terms “have”, “comprise” or “include” or any arbitrary grammatical variations thereof are used in a non-exclusive way. Thus, these terms may both refer to a situation in which, besides the feature introduced by these terms, no further features are present in the entity described in this context and to a situation in which one or more further features are present. As an example, the expressions “A has B”, “A comprises B” and “A includes B” may both refer to a situation in which, besides B, no other element is present in A (i.e. a situation in which a solely and exclusively consists of B) and to a situation in which, besides B, one or more further elements are present in entity A, such as element C, elements C and D or even further elements.

Further, as used in the following, the terms “preferably”, “more preferably”, “most preferably”, “particularly”, “more particularly”, “specifically”, “more specifically” or similar terms are used in conjunction with optional features, without restricting alternative possibilities. Thus, features introduced by these terms are optional features and are not intended to restrict the scope of the claims in any way. The invention may, as the skilled person will recognize, be performed by using alternative features. Similarly, features introduced by “in an embodiment of the invention” or similar expressions are intended to be optional features, without any restriction regarding alternative embodiments of the invention, without any restrictions regarding the scope of the invention and without any restriction regarding the possibility of combining the features introduced in such way with other optional or non-optional features of the invention. Moreover, if not otherwise indicated, the term “about” relates to the indicated value with the commonly accepted technical precision in the relevant field, preferably relates to the indicated value ±20%.

The terms “virus” and “virus of the family Paramyxoviridae” are known to the skilled person. Preferably, the virus of the family Paramyxoviridae is a member of the genus Morbillivirus. More preferably, the virus of the family Paramyxoviridae is a measles virus (MV), still more preferably an MV of strain Edmonston A or B, preferably B. Most preferably, the virus of the family Paramyxoviridae is an MV of vaccine strain Schwarz/Moraten.

The term “recombinant virus”, as used herein, relates to a virus comprising a genome modified by biotechnological means as compared to known, naturally occurring, virus genomes. Preferably, the recombinant virus is a virus comprising a genome modified as compared to naturally occurring virus genomes. Preferred biotechnological means for modifying a viral genome are known to the skilled person and include any of the methods of molecular cloning, in particular recombinant DNA techniques including, without limitation, cleavage of DNA by restriction enzymes, ligation of DNA, polymerase chain reaction (PCR), cloning of viral genomes, and the like. It is understood by the skilled person that viruses of the family Paramyxoviridae have a single-stranded (−)-RNA as a genome. Accordingly, the genome of the recombinant virus of the present invention, preferably, is obtained by cloning an expression vector as described herein below comprising an expressible nucleotide sequence encoding said recombinant virus genome, followed by expressing said expressible nucleotide sequence encoding said recombinant virus in a permissive host cell. Alternatively, the recombinant virus genome may also be expressed in non-permissive host cells, e.g., preferably, from rodents or other higher eukaryotes. Preferably, the recombinant virus of the present invention is a recombinant virus of the family Paramyxoviridae, more preferably a recombinant Morbillivirus, most preferably, a recombinant measles virus (MV). As will be understood by the skilled person, the recombinant virus of he present invention may comprises further modifications as compared to a naturally occurring virus. Preferably, the recombinant virus comprises a polypeptide mediating a modified tropism and/or a polynucleotide encoding the same. More preferably, said polypeptide mediating a modified tropism is a fusion polypeptide of a viral membrane integral polypeptide or of a viral membrane associated polypeptide with a polypeptide mediating binding to a target, e.g. a cell, preferably a specific kind of cell, more preferably a cancer cell. Preferably, said fusion polypeptide comprises a viral hemagglutinin or a fragment thereof, preferably a membrane integral fragment thereof. Preferably, said fusion polypeptide comprises a single-chain antibody specifically binding to a target molecule, e.g. to Carcinoembryonic antigen (CEA) or CD20. Most preferably, said fusion polypeptide is a fusion polypeptide of a truncated viral hemagglutinin with an anti-CD20 single-chain antibody or with an anti-CEA single-chain antibody. Preferably, the recombinant virus comprises a polynucleotide comprising the nucleic acid sequence of any one of SEQ ID Nos: 4 to 7, 14, and 15. SEQ ID NO: 4 is an artificial MV genome encoding an IL-12 fusion polypeptide comprising the mouse p40 subunit of IL-12 and the mouse p35 subunit of IL-12 as specified elsewhere herein. SEQ ID NO: 5 is an artificial MV genome encoding an IL-12 fusion polypeptide comprising the human p40 subunit of IL-12 and the human p35 subunit of IL-12 as specified elsewhere herein. SEQ ID NO: 6 is an artificial MV genome encoding an IL-12 fusion polypeptide comprising the mouse p40 subunit of IL-12 and the mouse p35 subunit of IL-12 as specified elsewhere herein, and a fusion polypeptide comprising a viral hemagglutinin and an anti-human-CEA single-chain antibody. SEQ ID NO: 7 is an artificial MV genome encoding an IL-12 fusion polypeptide comprising the mouse p40 subunit of IL-12 and the mouse p35 subunit of IL-12 as specified elsewhere herein, and a fusion polypeptide comprising a viral hemagglutinin and an anti-human-CD20 single-chain antibody. SEQ ID NO: 14 is an artificial MV genome derived from strain Edmonston B encoding an IL-12 fusion polypeptide comprising the mouse p40 subunit of IL-12 and the mouse p35 subunit of IL-12 as specified elsewhere herein. SEQ ID NO: 15 is an artificial MV genome encoding an IL-12 fusion polypeptide comprising the human p40 subunit of IL-12 and the human p35 subunit of IL-12 as specified elsewhere herein.

As used herein, the term “IL-12” relates to an interleukin 12 which is, in principle, known to the skilled person. Preferably, IL-12 is the heterodimeric IL-12 having the activity of stimulating the immune response of a subject. Preferably, the IL-12 is an IL-12 of a vertebrate species, more preferably of a mammal, even more preferably of a rat, a mouse, or a human, most preferably of a human. Preferably, the IL-12 has the subunits of rat IL-12, i.e. p35 comprising the amino acid sequence of Genbank Acc. No: NP_445842.1 GI:16758120, and p40 comprising the amino acid sequence of Genbank Acc. No: NP_072133.1 GI:12018288. Preferably, the subunits of the rat IL-12 are encoded by a polynucleotide comprising the nucleic acid sequence of Genbank Acc. No: NM_053390.1 GI:16758119 (rat mRNA expressed from the rat IL-12A gene) and/or of Genbank Acc. No: NM_022611.1 GI:12018287 (rat mRNA expressed from the rat IL-12B gene). More preferably, the IL-12 has the subunits of mouse IL-12, i.e. p35 comprising the amino acid sequence of Genbank Acc. No: NP_001152896.1 GI:226874945, and p40 comprising the amino acid sequence of Genbank Acc. No: NP_001290173.1 GI:735997434. Preferably, the subunits of the mouse IL-12 are encoded by a polynucleotide comprising the nucleic acid sequence of Genbank Acc. No: NM_001159424.2 GI:746816821 (mouse mRNA expressed from the mouse IL-12A gene) and/or of Genbank Acc. No: NM_001303244.1 GI:735997433 (mouse mRNA expressed from the mouse IL-12B gene). Most preferably, the IL-12 has the subunits of human IL-12, i.e. p35 comprising the amino acid sequence of Genbank Acc. No: NP_000873.2 GI:24430219, and p40 comprising the amino acid sequence of Genbank Acc. No: NP_002178.2 GI:24497438. Preferably, the subunits of the human IL-12 are encoded by a polynucleotide comprising the nucleic acid sequence of Genbank Acc. No: NM_000882.3 GI:325974478 (human mRNA expressed from the human IL-12A gene) and/or of Genbank Acc. No: NM_002187.2 GI:24497437 (human mRNA expressed from the human IL-12B gene). In its natural form, IL-12 is a secreted interleukin, i.e. it is processed and transported from the interior of the producing cell to the exterior of the producing cell by said producing cell. Accordingly, IL-12 preferably is a secreted IL-12.

More preferably, the IL-12 according to the present invention is an IL-12 fusion polypeptide comprising a p40 subunit of an IL-12 and a p35 subunit of an IL-12, preferably comprising subunits as specified herein above. More preferably, the p40 subunit and the p35 subunit of said IL-12 fusion polypeptide are from the same species; i.e. preferably, the p40 subunit and the p35 subunit of said IL-12 fusion polypeptide are a rat p40 subunit and a rat p35 subunit, more preferably are a mouse p40 subunit and a mouse p35 subunit, most preferably are a human p40 subunit and a human p35 subunit. Preferably, said p40 subunit and said p35 subunit are comprised in the order N-terminus- p40 subunit- p35 subunit—C-terminus in said fusion polypeptide. Preferably, said p40 subunit and said p35 subunit are separated by a linker, i.e., the fusion polypeptide comprises the structure p40-linker-35.

The term “linker” is known to the skilled person and, preferably, relates to a short sequence of amino acids separating two domains of a polypeptide or two components of a fusion polypeptide. The skilled person knows how to select appropriate linker sequences in order to construct functional fusion polypeptides, e.g. from Xue et al. (2004), NAR 32 (Web server issue):W562. Preferably, said linker comprises of from 1 to 50, more preferably of from 2 to 25, most preferably of from 10 to 20 amino acids. Preferably, the amino acids of the linker are small amino acids and/or amino acids promoting turns in protein structure; accordingly, the amino acids comprised in the linker, preferably, are glycine, alanine, serine, and/or proline. Preferably, the linker has a repetitive structure; thus, preferably, the linker comprises of from 1 to 10, more preferably of from 2 to 5, most preferably 3 repetitions of an amino acid sequence comprising 3 or 4, preferably 5 amino acids. Preferably, the repetitive sequence of the linker comprises the sequence (glycinex-serine), with x=3 to 6, preferably 4 to 6, more preferably 4 or 6. More preferably, the repetitive sequence of the linker comprises the sequence (glycine4-serine), i.e. gly-gly-gly-gly-ser (SEQ ID NO:1), preferably repeated as specified above. Thus, preferably the linker comprises or consists of the amino acid sequence -(glycine4-serine)n-, with n=1 to 10, preferably n=2 to 5, more preferably n=3. Most preferably, the linker has the amino acid sequence of SEQ ID NO:8. Also more preferably, the repetitive sequence of the linker comprises the sequence (glycine6-serine), i.e. gly-gly-gly-gly-gly-gly-ser (SEQ ID NO 9).

As used herein, the term “fusion polypeptide” relates to a polypeptide wherein all components, e.g. p35 subunit, linker, and p40 subunit, are covalently linked and, preferably, are produced as a contiguous polypeptide chain. Thus, preferably, the fusion polypeptide of the present invention, preferably, is expressed from a single gene. Thus, the IL-12 of the present invention preferably is fused mouse IL-12 (FmIL-12), preferably comprising the amino acid sequence of SEQ ID NO:10, preferably encoded by a polynucleotide comprising the nucleic acid sequence of SEQ ID NO:11. More preferably, the IL-12 of the present invention preferably is fused human IL-12 (FhIL-12), preferably comprising the amino acid sequence of SEQ ID NO:12, preferably encoded by a polynucleotide comprising the nucleic acid sequence of SEQ ID NO:13.

The terms “polypeptide” and “fusion polypeptide”, as used herein, preferably encompass variants of said polypeptides and fusion polypeptides as specified elsewhere herein.

Preferably, the recombinant virus of the family Paramyxoviridae of the present invention further comprises at least one expressible polynucleotide encoding a further activator of the immune response, preferably an immunoglobulin or part thereof, preferably a secreted immunoglobulin.

As used herein, the term “further activator of the immune response” relates to a compound which, when contacted with a mixture of immune cells and immune-response inducing cells, e.g. cancer cells, causes at least one type of immune cell to be more active as compared to an immune cell of the same type comprised in the same mixture but lacking said compound. As used herein, the term IL-12 as specified elsewhere herein relates to an activator of the immune response, but not to a further activator of the immune response. Preferably, the immune cell activated is a cell mediating a response increasing a subject's resistance to an antigen, i.e. preferably, said immune cell is not a tolerance-mediating immune cell. Preferably, the immune cell activated by the further activator of the immune response is a T-cell, more preferably a helper T-cell ora cytotoxic T-cell. Most preferably, the immune cell activated by the further activator of the immune response is a cytotoxic T-cell expressing PD-1. Measures of immune cell activity are known to the skilled person and include, preferably, expression of activation markers, production of antibodies, excretion of cytokines, and release of cytotoxins, e.g. perforin, granzymes, and/or granolysin.

Preferably, the further activator of the immune response is an antagonist of a signaling pathway causing at least one type of immune cell to become inhibited. Accordingly, preferably, the further activator of the immune response is a ligand for an immune checkpoint blockade protein. More preferably, the further activator of the immune response is a ligand for an immune checkpoint blockade protein. Still more preferably, the activator of the immune response is an inhibitor of PD-1 receptor signaling. It is understood by the skilled person that signaling through a receptor signaling pathway can be inhibited by either preventing the receptor from being activated, or by preventing the signal generated by the activated receptor from being further transmitted. Accordingly, preferably, the further activator of the immune response is a PD-L1 antagonist, the term “antagonist” relating to a compound binding to the molecule the effect of which is antagonized and through said binding preventing said molecule from interacting with its native binding partner in a productive, i.e. signaling-inducing, way. Preferred assays for said activity are described e.g. in WO 2015/128313 A1.

Preferably, the further activator of the immune response is an antagonist as described above selected from the list of molecule types consisting of a peptide aptamer, an anticalin, a Designed Ankyrin Repeat Protein (DARPin), an inhibitory peptide, and, preferably, an immunoglobulin, more preferably, an antibody.

In the context of this invention, a “peptide aptamer” is a peptide specifically binding its interaction partner and having the activity of activating the immune response as specified herein above, preferably, the activity of being an antagonist of PD-L1 as specified herein above. Peptide aptamers, preferably, are peptides comprising 8-80 amino acids, more preferably 10-50 amino acids, and most preferably 15-30 amino acids. They can e.g. be isolated from randomized peptide expression libraries in a suitable host system like baker's yeast (see, for example, Klevenz et al., Cell Mol Life Sci. 2002, 59: 1993-1998). A peptide aptamer, preferably, is a free peptide; it is, however, also contemplated by the present invention that a peptide aptamer is fused to a polypeptide serving as “scaffold”, meaning that the covalent linking to said polypeptide serves to fix the three-dimensional structure of said peptide aptamer to one specific conformation. More preferably, the peptide aptamer is fused to a transport signal, in particular a peptide export signal.

As used herein, the term “anticalin” relates to an artificial polypeptide derived from a lipocalin specifically binding its interaction partner. Similarly, a “Designed Ankyrin Repeat Protein” or “DARPin”, as used herein, is an artificial polypeptide comprising several 14cab14i14 repeat motifs and specifically binding its interaction partner. The anticalins and the DARPins of the present invention have the activity of activating the immune response as specified herein above, preferably, the activity of being an antagonist of PD-L1 as specified herein above.

As used herein, the term “inhibitory peptide” relates to any chemical molecule comprising at least one peptide having the activity of activating the immune response as specified herein above, preferably, the activity of being an antagonist PD-L1 as specified herein above. Preferably, the inhibitory peptide comprises a peptide having an amino acid sequence corresponding to an amino acid sequence of at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, at least 13, at least 14, or at least 15 consecutive amino acids comprised in a PD-L1 polypeptide. Preferably, the inhibitory peptide comprises a peptide having an amino acid sequence corresponding to an amino acid sequence of 5 to 200, more preferably 6 to 100, even more preferably 7 to 50, or, most preferably, 8 to 30 consecutive amino acids comprised in a PD-L1 polypeptide. Moreover, also encompassed are variants of the aforementioned inhibitory peptides. Such variants have at least the same essential biological activity as the specific inhibitory peptides.

As used herein, the term “immunoglobulin” relates to a polypeptide being a soluble immunoglobulin, preferably an antibody from any of the classes IgA, IgD, IgE, IgG, or IgM, preferably having the activity of binding, more preferably specifically binding, a molecule of interest. Immunoglobulins against antigens of interest can be prepared by well known methods using, e.g., a purified molecule of interest or a suitable fragment derived therefrom as an antigen. A fragment which is suitable as an antigen may be identified by antigenicity determining algorithms well known in the art. Such fragments may be obtained either from one of the molecules of interest by proteolytic digestion, may be a synthetic peptide, or may be obtained by recombinant expression. Preferably, a peptide of a molecule of interest used as an antigen is located at the exterior of a cell expressing the molecule of interest; i.e. preferably, the epitope the binding domain interacts with, preferably, is an extracellular domain. Preferably, the immunoglobulin of the present invention is a monoclonal antibody, a human or humanized antibody or primatized, chimerized antibody or a fragment thereof, so long as they exhibit the desired binding activity as specified elsewhere herein. Also comprised as antibodies of the present invention are a bispecific antibody, a synthetic antibody, or a chemically modified derivative of any of these. Preferably, the antibody of the present invention shall specifically bind (i.e. does only to a negligible extent or, preferably, not cross react with other polypeptides or peptides) to a molecule of interest as specified above. Specific binding can be tested by various well known techniques. Antibodies or fragments thereof can be obtained by using methods which are described, e.g., in Harlow and Lane “Antibodies, A Laboratory Manual”, CSH Press, Cold Spring Harbor, 1988. Monoclonal antibodies can be prepared by the techniques originally described in Köhler and Milstein, Nature. 1975. 256: 495; and Galfré, Meth. Enzymol. 1981, 73: 3, which comprise the fusion of mouse myeloma cells to spleen cells derived from immunized mammals. As will be understood by the skilled person, a molecule of interest, bound by an immunoglobulin of the present invention, may also be an Fc receptor or a complement protein binding an Fc part of an antibody; accordingly, the immunoglobulin preferably is an Fc domain of an antibody, more preferably a soluble Fc domain of an antibody, most preferably a secreted soluble Fc domain of an antibody. Preferably, said antibody the Fc domain is derived from is an IgG, more preferably an IgG1, most preferably a human IgG1. Preferably, the secreted soluble Fc domain comprises the amino acid sequence of SEQ ID NO: 16 or a variant thereof, preferably encoded by the nucleic acid sequence of SEQ ID NO: 17. More preferably, the immunoglobulin is an antagonistic anti-PD-L1 antibody, still more preferably comprising the amino acid sequence of SEQ ID NO:2 or a variant thereof, preferably encoded by a polynucleotide comprising the nucleic acid sequence of SEQ ID NO:3 or a variant thereof. More preferably, the immunoglobulin is an antagonistic anti-PD-L1 antibody, more preferably comprising the amino acid sequence of SEQ ID NO:2, preferably encoded by a polynucleotide comprising the nucleic acid sequence of SEQ ID NO:3.

“Immunoglobulin fragments” comprise a portion of an intact immunoglobulin, preferably of an antibody, in an embodiment, comprise the antigen-binding region thereof. Examples of antibody fragments and fusion proteins of variable regions include Fab, Fab′, F(ab′)2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; single-domain-antibodies (VHH), also known as nanobodies, and multispecific antibodies formed from antibody fragments. Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual “Fc” fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen-combining sites and is still capable of cross-linking antigen. “Fv” is the minimum antibody fragment which contains a complete antigen-binding site. Preferably, a two-chain Fv species consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. In a single-chain Fv (scFv) species, one heavy- and one light-chain variable domain can be covalently linked by a flexible peptide linker such that the light and heavy chains can associate in a “dimeric” structure analogous to that in a two-chain Fv species. It is in this configuration that the three hypervariable regions (HVRs, also referred to as complementarity determining regions (CDRs)) of each variable domain interact to define an antigen-binding site. Collectively, the six HVRs of one scFv confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three HVRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site. The term “diabodies” refers to antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) in the same polypeptide chain (VH-VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies may be bivalent or bispecific. Diabodies are described more fully in, for example, EP 0 404 097; WO 1993/01161; Hudson et al., Nat. Med. 9 (2003) 129-134; and Hollinger et al., PNAS USA 90 (1993) 6444-6448. Triabodies and tetrabodies are also described in Hudson et al., Nat. Med. 9 (2003) 129-134.

The term “secreted”, as used herein, relates to a compound being transferred from the interior of a host cell to the exterior of said host cell by a mechanism intrinsic to said host cell. Preferably, secretion of a polypeptide or fusion polypeptide is mediated by a, preferably eukaryotic, signal peptide mediating import of said peptide or polypeptide into the lumen of the endoplasmic reticulum and, more preferably, by the absence of retention signals. Signal peptides causing secretion of peptides or polypeptides are known in the art. Preferably, the signal peptide is an IL-12 signal peptide. Also preferably, the signal peptide is or comprises an Ig leader sequence. More preferably, the signal peptide is or comprises a human Ig leader sequence. Still more preferably, the signal peptide is or comprises a matching leader sequence, i.e. a leader sequence selected from the same Ig kappa subgroup as the variable light chain of the antibody, preferably, of the single-chain antibody.

As used herein, the terms “polypeptide variant” relates to any chemical molecule comprising at least one polypeptide or fusion polypeptide as specified elsewhere herein, having the indicated activity, but differing in primary structure from said polypeptide or fusion polypeptide indicated above. Thus, the polypeptide variant, preferably, is a mutein having the indicated activity. Preferably, the polypeptide variant comprises a peptide having an amino acid sequence corresponding to an amino acid sequence of 5 to 200, more preferably 6 to 100, even more preferably 7 to 50, or, most preferably, 8 to 30 consecutive amino acids comprised in a polypeptide as specified above. Moreover, also encompassed are further polypeptide variants of the aforementioned polypeptides. Such polypeptide variants have at least essentially the same biological activity as the specific polypeptides. Moreover, it is to be understood that a polypeptide variant as referred to in accordance with the present invention shall have an amino acid sequence which differs due to at least one amino acid substitution, deletion and/or addition, wherein the amino acid sequence of the variant is still, preferably, at least 50%, 60%, 70%, 80%, 85%, 90%, 92%, 95%, 97%, 98%, or 99% identical with the amino acid sequence of the specific polypeptide. The degree of identity between two amino acid sequences can be determined by algorithms well known in the art. Preferably, the degree of identity is to be determined by comparing two optimally aligned sequences over a comparison window, where the fragment of amino acid sequence in the comparison window may comprise additions or deletions (e.g., gaps or overhangs) as compared to the sequence it is compared to for optimal alignment. The percentage is calculated by determining, preferably over the whole length of the polypeptide, the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity. Optimal alignment of sequences for comparison may be conducted by the local homology algorithm of Smith and Waterman (1981), by the homology alignment algorithm of Needleman and Wunsch (1970), by the search for similarity method of Pearson and Lipman (1988), by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, PASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by visual inspection. Given that two sequences have been identified for comparison, GAP and BESTFIT are preferably employed to determine their optimal alignment and, thus, the degree of identity. Preferably, the default values of 5.00 for gap weight and 0.30 for gap weight length are used. Polypeptide variants referred to herein may be allelic variants or any other species specific homologs, paralogs, or orthologs. Moreover, the polypeptide variants referred to herein include fragments of the specific polypeptides or the aforementioned types of polypeptide variants as long as these fragments and/or variants have the biological activity as referred to above. Such fragments may be or be derived from, e.g., degradation products or splice variants of the polypeptides. Further included are variants which differ due to posttranslational modifications such as phosphorylation, glycosylation, ubiquitinylation, sumoylation, or myristylation, by including non-natural amino acids, and/or by being peptidomimetics.

The term “expressible polynucleotide”, as used herein, relates to a polynucleotide operatively linked to at least one expression control sequence causing transcription of the nucleic acid sequence comprised in said polynucleotide to occur, preferably in eukaryotic cells or isolated fractions thereof, preferably into a translatable mRNA or into a viral genome. Regulatory elements ensuring expression in eukaryotic cells, preferably mammalian cells, are well known in the art. They, preferably, comprise regulatory sequences ensuring initiation of transcription and, optionally, poly-A signals ensuring termination of transcription and stabilization of the transcript. Additional regulatory elements may include transcriptional as well as translational enhancers. Preferably, the aforesaid at least one expression control sequence is an expression control sequence of a (−)strand RNA virus, more preferably of a Paramyxovirus as described herein above, most preferably of an MV. Thus, preferably, at least one expression control sequence comprises a (−)strand RNA viral regulatory sequence ensuring initiation of transcription (consensus “gene start signal”, preferably consensus MV “gene start signal”) and termination signals (consensus “gene stop signal”, preferably, consensus MV “gene stop signal”) ensuring termination of transcription and stabilization of the transcript. It is known in the art that production of viral particles in permissive host cells can be initiated by transfecting into said permissive host cells one or more expressible DNA constructs encoding (i) a recombinant viral anti-genome, (ii) the viral L gene, (iii) the viral P gene, and (iv) the viral N gene. It is also understood by the skilled person that, once a viral genome and the aforesaid viral genes were expressed in said host cell, replication and assembly of viral particles occurs in the cytoplasm of the host cell and is, therefore, solely dependent on viral regulatory signals. The term polynucleotide, as used herein, preferably encompasses polynucleotide variants as specified elsewhere herein. Preferably, the expressible polynucleotide encoding an IL-12 is comprised in the genome of the recombinant virus of the family Paramyxoviridae in a region corresponding to the region intervening the P and the M gene of measles virus.

The term “polynucleotide encoding a recombinant virus”, as used herein, relates to a polynucleotide comprising a nucleic acid sequence or nucleic acid sequences required for generating a virus particle or a virus-like particle in a host cell. It is understood by the skilled person that a virus is constituted by a polynucleotide genome and at least one kind of capsid polypeptide. Accordingly, the polynucleotide encoding a recombinant virus of the present invention, preferably, comprises a recombinant virus genome. As will be understood by the skilled person, in case the polynucleotide encoding a recombinant virus is comprised in a virus according to the present invention, i.e. a virus of the family Paramyxoviridae, the polynucleotide is (−)strand RNA. It is also understood by the skilled person that in case the polynucleotide is DNA comprised in a host cell, at least an RNA-dependent RNA polymerase activity will additionally be required to produce viral particles from said DNA polynucleotide. Preferably, the polynucleotide encoding a recombinant virus comprises or consists of the nucleic acid sequence as specified elsewhere herein. As annotated herein, the sequence of the DNA copy of negative-strand (−)RNA viruses is annotated in the usual 5′→3′-orientation; this corresponds to the viral sequence in antigenomic (+)RNA orientation with respect to the natural 3′→5′-orientation of negative-strand (−)RNA viruses.

The term “polynucleotide variant”, as used herein, relates to a variant of a polynucleotide related to herein comprising a nucleic acid sequence characterized in that the sequence can be derived from the aforementioned specific nucleic acid sequence by at least one nucleotide substitution, addition and/or deletion, wherein the polynucleotide variant shall have the activity as specified for the specific polynucleotide. Preferably, said polynucleotide variant is an ortholog, a paralog or another homolog of the specific polynucleotide. Also preferably, said polynucleotide variant is a naturally occurring allele of the specific polynucleotide. Polynucleotide variants also encompass polynucleotides comprising a nucleic acid sequence which is capable of hybridizing to the aforementioned specific polynucleotides, preferably, under stringent hybridization conditions. These stringent conditions are known to the skilled worker and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. A preferred example for stringent hybridization conditions are hybridization conditions in 6× sodium chloride/sodium citrate (=SSC) at approximately 45° C., followed by one or more wash steps in 0.2× SSC, 0.1% SDS at 50 to 65° C. The skilled worker knows that these hybridization conditions differ depending on the type of nucleic acid and, for example when organic solvents are present, with regard to the temperature and concentration of the buffer. For example, under “standard hybridization conditions” the temperature differs depending on the type of nucleic acid between 42° C. and 58° C. in aqueous buffer with a concentration of 0.1× to 5× SSC (pH 7.2). If organic solvent is present in the abovementioned buffer, for example 50% formamide, the temperature under standard conditions is approximately 42° C. The hybridization conditions for DNA:DNA hybrids are preferably for example 0.1x SSC and 20° C. to 45° C., preferably between 30° C. and 45° C. The hybridization conditions for DNA:RNA hybrids are preferably, for example, 0.1× SSC and 30° C. to 55° C., preferably between 45° C. and 55° C. The abovementioned hybridization temperatures are determined for example for a nucleic acid with approximately 100 bp (=base pairs) in length and a G+C content of 50% in the absence of formamide. The skilled worker knows how to determine the hybridization conditions required by referring to textbooks such as the textbook mentioned above, or the following textbooks: Sambrook et al., “Molecular Cloning”, Cold Spring Harbor Laboratory, 1989; Hames and Higgins (Ed.) 1985, “Nucleic Acids Hybridization: A Practical Approach”, IRL Press at Oxford University Press, Oxford; Brown (Ed.) 1991, “Essential Molecular Biology: A Practical Approach”, IRL Press at Oxford University Press, Oxford. Alternatively, polynucleotide variants are obtainable by PCR-based techniques such as mixed oligonucleotide primer-based amplification of DNA, i.e. using degenerated primers against conserved domains of a polypeptide of the present invention. Conserved domains of a polypeptide may be identified by a sequence comparison of the nucleic acid sequence of the polynucleotide or the amino acid sequence of the polypeptide of the present invention with sequences of other organisms. As a template, DNA or cDNA from bacteria, fungi, or plants preferably, from animals may be used. Further, variants include polynucleotides comprising nucleic acid sequences which are at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or at least 99% identical to the specifically indicated nucleic acid sequences. Moreover, also encompassed are polynucleotides which comprise nucleic acid sequences encoding amino acid sequences which are at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98% or at least 99% identical to the amino acid sequences specifically indicated. The percent identity values are, preferably, calculated over the entire amino acid or nucleic acid sequence region. A series of programs based on a variety of algorithms is available to the skilled worker for comparing different sequences. In this context, the algorithms of Needleman and Wunsch or Smith and Waterman give particularly reliable results. To carry out the sequence alignments, the program PileUp (J. Mol. Evolution., 25, 351-360, 1987, Higgins et al., CABIOS, 5 1989: 151-153) or the programs Gap and BestFit (Needleman and Wunsch (J. Mol. Biol. 48; 443-453 (1970)) and Smith and Waterman (Adv. Appl. Math. 2; 482-489 (1981))), which are part of the GCG software packet (Genetics Computer Group, 575 Science Drive, Madison, Wis., USA 53711 (1991)), are to be used. The sequence identity values recited above in percent (%) are to be determined, preferably, using the program GAP over the entire sequence region with the following settings: Gap Weight: 50, Length Weight: 3, Average Match: 10.000 and Average Mismatch: 0.000, which, unless otherwise specified, shall always be used as standard settings for sequence alignments.

A polynucleotide comprising a fragment of any of the specifically indicated nucleic acid sequences is also encompassed as a variant polynucleotide of the present invention. The fragment shall still encode a polypeptide or fusion polypeptide which still has the activity as specified. Accordingly, the polypeptide encoded may comprise or consist of the domains of the polypeptide of the present invention conferring the said biological activity. A fragment as meant herein, preferably, comprises at least 50, at least 100, at least 250 or at least 500 consecutive nucleotides of any one of the specific nucleic acid sequences or encodes an amino acid sequence comprising at least 20, at least 30, at least 50, at least 80, at least 100 or at least 150 consecutive amino acids of any one of the specific amino acid sequences.

The polynucleotides of the present invention either consist of, essentially consist of, or comprise the aforementioned nucleic acid sequences. Thus, they may contain further nucleic acid sequences as well. Specifically, the polynucleotides of the present invention may encode fusion proteins wherein one partner of the fusion protein is a polypeptide being encoded by a nucleic acid sequence recited above. Such fusion proteins may comprise as additional part polypeptides for monitoring expression (e.g., green, yellow, blue or red fluorescent proteins, alkaline phosphatase and the like) or so called “tags” which may serve as a detectable marker or as an auxiliary measure for purification purposes. Tags for the different purposes are well known in the art and are described elsewhere herein.

The polynucleotide of the present invention shall be provided, preferably, either as an isolated polynucleotide (i.e. isolated from its natural context) or in genetically modified form. The polynucleotide, preferably, is DNA, including cDNA, or RNA. The term encompasses single as well as double stranded polynucleotides. Moreover, preferably, comprised are also chemically modified polynucleotides including naturally occurring modified polynucleotides such as glycosylated or methylated polynucleotides or artificial modified one such as biotinylated polynucleotides.

As used herein, the term “host cell” relates to a vertebrate cell. Preferably, the cell is a mammalian cell, more preferably, a mouse, rat, cat, dog, hamster, guinea pig, sheep, goat, pig, cattle, or horse cell. Still more preferably, the host cell is a primate cell. Most preferably, the host cell is a human cell. Preferably, the host cell is a tumor cell, more preferably a cancer cell.

Advantageously, it was found in the work underlying the present invention that oncolytic measles virus can be engineered to express IL-12, in particular an IL-12 fusion polypeptide, while infecting cancer cells and that IL-12 expression strongly enhances the immune response induced by the measles virus against said cancer cells. Moreover, it was found that by further expressing immunoglobulins, in particular an anti-PD-L1 antibody, measles virus can further augment the immunological response to cancer cells, thus further contributing to their elimination.

The definitions made above apply mutatis mutandis to the following. Additional definitions and explanations made further below also apply for all embodiments described in this specification mutatis mutandis.

The present invention further relates to a polynucleotide encoding the recombinant virus of the family Paramyxoviridae according to the present invention.

The present invention further relates to a host cell comprising the recombinant virus of the family Paramyxoviridae of the present invention and/or the polynucleotide encoding the recombinant virus of the family Paramyxoviridae of the present invention.

As used herein, the term “host cell” relates to a host cell as specified herein above. Moreover, the host cell comprising the polynucleotide encoding the recombinant virus of the family Paramyxoviridae of the present invention may also be a bacterial, yeast, or insect cell, preferably a bacterial cell of the genus Escherichia, more preferably an Escherichia coli cell.

The present invention also relates to a medicament comprising (a) (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist; (ii) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 fusion polypeptide of the present invention; (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii), (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae and/or the polynucleotide encoding the recombinant virus of the family Paramyxoviridae according to; or (v) any combination of (i) to (iv); and (b) at least one pharmacologically acceptable excipient.

The terms “medicament” and “pharmaceutical composition”, as used herein, relate to the compounds of the present invention and optionally one or more pharmaceutically acceptable carrier, i.e. excipient. The compounds of the present invention can be formulated as pharmaceutically acceptable salts. Acceptable salts comprise acetate, methyl ester, HCI, sulfate, chloride and the like. The pharmaceutical compositions are, preferably, administered locally, topically or systemically. Suitable routes of administration conventionally used for drug administration are oral, intravenous, or parenteral administration as well as inhalation. A preferred route of administration is intra-tumoral administration. However, depending on the nature and mode of action of a compound, the pharmaceutical compositions may be administered by other routes as well. For example, polynucleotide compounds may be administered in a gene therapy approach by using viral vectors or viruses or liposomes.

Moreover, the compounds can be administered in combination with other drugs either in a common pharmaceutical composition or as separated pharmaceutical compositions wherein said separated pharmaceutical compositions may be provided in form of a kit of parts. The compounds are, preferably, administered in conventional dosage forms prepared by combining the drugs with standard pharmaceutical carriers according to conventional procedures. These procedures may involve mixing, granulating and compressing or dissolving the ingredients as appropriate to the desired preparation. It will be appreciated that the form and character of the pharmaceutically acceptable carrier or diluent is dictated by the amount of active ingredient with which it is to be combined, the route of administration and other well-known variables.

The excipient(s) must be acceptable in the sense of being compatible with the other ingredients of the formulation and being not deleterious to the recipient thereof. The excipient employed may be, for example, a solid, a gel or a liquid carrier. Exemplary of solid carriers are lactose, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, stearic acid and the like. Exemplary of liquid carriers are phosphate buffered saline solution, syrup, oil such as peanut oil and olive oil, water, emulsions, various types of wetting agents, sterile solutions and the like. Similarly, the carrier or diluent may include time delay material well known to the art, such as glyceryl mono-stearate or glyceryl distearate alone or with a wax. Said suitable carriers comprise those mentioned above and others well known in the art, see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. The diluent(s) is/are selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological saline, Ringer's solutions, dextrose solution, and Hank's solution. In addition, the pharmaceutical composition or formulation may also include other carriers, adjuvants, or nontoxic, nontherapeutic, non-immunogenic stabilizers and the like.

A therapeutically effective dose refers to an amount of the compounds to be used in a pharmaceutical composition of the present invention which prevents, ameliorates or treats the symptoms accompanying a disease or condition referred to in this specification. Therapeutic efficacy and toxicity of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., ED50 (the dose therapeutically effective in 50% of the population) and LD50 (the dose lethal to 50% of the population). The dose ratio between therapeutic and toxic effects is the therapeutic index, and it can be expressed as the ratio, LD50/ED50.

The dosage regimen will be determined by the attending physician and other clinical factors; preferably in accordance with any one of the above described methods. As is well known in the medical arts, dosages for any one patient depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, time and route of administration, general health, and other drugs being administered concurrently. Progress can be monitored by periodic assessment. A typical dose can be, for example, in the range of 1 to 1000 μg for a polypeptide or polynucleotide, or 104-108 viral particles for a virus or a virus-like particle; however, doses below or above this exemplary range are envisioned, especially considering the aforementioned factors. Progress can be monitored by periodic assessment. The pharmaceutical compositions and formulations referred to herein are administered at least once in order to treat or ameliorate or prevent a disease or condition recited in this specification. However, the said pharmaceutical compositions may be administered more than one time, for example from one to four times daily up to a non-limited number of days. Specific pharmaceutical compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one active compound referred to herein above in admixture or otherwise associated with a pharmaceutically acceptable carrier or diluent. For making those specific pharmaceutical compositions, the active compound(s) will usually be mixed with a carrier or the diluent, or enclosed or encapsulated in a capsule, sachet, cachet, paper or other suitable containers or vehicles. The resulting formulations are to be adapted to the mode of administration, i.e. in the forms of tablets, capsules, suppositories, solutions, suspensions or the like. Dosage recommendations shall be indicated in the prescribers or users instructions in order to anticipate dose adjustments depending on the considered recipient.

Accordingly, the present invention also relates to a method for treating cancer in a subject afflicted with cancer, comprising

  • a) contacting said subject with
    • (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
    • (ii) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 fusion polypeptide of the present invention;
    • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
    • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae of (i) and/or (ii) and/or the polynucleotide encoding the recombinant virus of the family Paramyxoviridae according to (iii); or
    • (v) any combination of (i) to (iv), and thereby,
  • b) treating cancer in a subject afflicted with cancer.

The methods of treatment of the present invention, preferably, may comprise steps in addition to those explicitly mentioned above. For example, further steps may relate, e.g., to localizing a tumor and/or diagnosing cancer for step a), or administration of additional medication for step b). Moreover, one or more of said steps may be performed by automated equipment. The method of the present invention, preferably, is an in vivo method of treatment.

The term “treatment” refers to an amelioration of the diseases or disorders referred to herein or the symptoms accompanied therewith to a significant extent. Said treating as used herein also includes an entire restoration of the health with respect to the diseases or disorders referred to herein. It is to be understood that treating as used in accordance with the present invention may not be effective in all subjects to be treated. However, the term shall require that, preferably, a statistically significant portion of subjects suffering from a disease or disorder referred to herein can be successfully treated. Whether a portion is statistically significant can be determined without further ado by the person skilled in the art using various well known statistic evaluation tools, e.g., determination of confidence intervals, p-value determination, Student's t-test, Mann-Whitney test etc. Preferred confidence intervals are at least 90%, at least 95%, at least 97%, at least 98% or at least 99%. The p-values are, preferably, 0.1, 0.05, 0.01, 0.005, or 0.0001. Preferably, the treatment shall be effective for at least 10%, at least 20% at least 50%at least 60%, at least 70%, at least 80%, or at least 90% of the subjects of a given cohort or population. Preferably, treating cancer is reducing tumor burden in a subject. As will be understood by the skilled person, effectiveness of treatment of e.g. cancer is dependent on a variety of factors including, e.g. cancer stage and cancer type.

As used herein, the term “subject” relates to a vertebrate. Preferably, the subject is a mammal, more preferably, a mouse, rat, cat, dog, hamster, guinea pig, sheep, goat, pig, cattle, or horse. Still more preferably, the subject is a primate. Most preferably, the subject is a human. Preferably, the subject is afflicted with a disease caused or aggravated by an insufficient response of the immune response of said subject, more preferably, the subject is afflicted with cancer.

The term “cancer”, as used herein, relates to a disease of an animal, including man, characterized by uncontrolled growth by a group of body cells (“cancer cells”). This uncontrolled growth may be accompanied by intrusion into and destruction of surrounding tissue and possibly spread of cancer cells to other locations in the body. Preferably, also included by the term cancer is a relapse. Thus, preferably, the cancer is a solid cancer, a metastasis, or a relapse thereof.

Preferably, the cancer is selected from the list consisting of acute lymphoblastic leukemia, acute myeloid leukemia, adrenocortical carcinoma, aids-related lymphoma, anal cancer, appendix cancer, astrocytoma, atypical teratoid, basal cell carcinoma, bile duct cancer, bladder cancer, brain stem glioma, breast cancer, burkitt lymphoma, carcinoid tumor, cerebellar astrocytoma, cervical cancer, chordoma, chronic lymphocytic leukemia, chronic myelogenous leukemia, colon cancer, colorectal cancer, craniopharyngioma, endometrial cancer, ependymoblastoma, ependymoma, esophageal cancer, extracranial germ cell tumor, extragonadal germ cell tumor, extrahepatic bile duct cancer, fibrosarcoma, gallbladder cancer, gastric cancer, gastrointestinal stromal tumor, gestational trophoblastic tumor, hairy cell leukemia, head and neck cancer, hepatocellular cancer, 27cab27i27 lymphoma, hypopharyngeal cancer, hypothalamic and visual pathway glioma, intraocular melanoma, 27cab27i sarcoma, laryngeal cancer, medulloblastoma, medulloepithelioma, melanoma, merkel cell carcinoma, mesothelioma, mouth cancer, multiple endocrine neoplasia syndrome, multiple myeloma, mycosis fungoides, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma, non-hodgkin lymphoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, osteosarcoma, ovarian cancer, ovarian epithelial cancer, ovarian germ cell tumor, ovarian low malignant potential tumor, pancreatic cancer, papillomatosis, paranasal sinus and nasal cavity cancer, parathyroid cancer, penile cancer, pharyngeal cancer, pheochromocytoma, pituitary tumor, pleuropulmonary blastoma, primary central nervous system lymphoma, prostate cancer, rectal cancer, renal cell cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, sézary syndrome, small cell lung cancer, small intestine cancer, soft tissue sarcoma, squamous cell carcinoma, squamous neck cancer, testicular cancer, throat cancer, thymic carcinoma, thymoma, thyroid cancer, urethral cancer, uterine sarcoma, vaginal cancer, vulvar cancer, waldenström macroglobulinemia, and wilms tumor. More preferably, the cancer is a solid cancer, a metastasis, or a relapse thereof. Most preferably, the cancer is a tumor derived from malignant melanoma, head and neck cancer, hepatocellular carcinoma, pancreatic carcinoma, prostate cancer, renal cell carcinoma, gastric carcinoma, colorectal carcinoma, lymphomas or leukemias.

Preferably, the method of treatment of the present invention comprises contacting a subject with a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist. Thus, preferably, the method comprises contacting a subject with a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12, wherein said recombinant virus of the family Paramyxoviridae is not a virus disclosed in WO 2015/128313 A1. Preferably, said recombinant virus of the family Paramyxoviridae of (i) is a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 polypeptide and not comprising an expressible polynucleotide encoding a ligand for an immune checkpoint blockade protein. More preferably, the recombinant virus of the family Paramyxoviridae of (i) is a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 polypeptide as the only expressible polynucleotide encoding an activator of the immune response, i. e. preferably, the recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 does not comprise an expressible polynucleotide encoding a further activator of the immune response.

The present invention further relates to an in vitro method for activating immune cells with antitumor activity in a sample comprising cancer cells and immune cells, comprising

  • a) contacting said sample comprising cancer cells and immune cells with
    • (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
    • (ii) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 fusion polypeptide of the present invention;
    • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
    • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae of (i) and/or (ii) and/or the polynucleotide encoding the recombinant virus of the family Paramyxoviridae according to (iii); or
    • (v) any combination of (i) to (iv), and thereby,
  • b) activating immune cells with antitumor activity comprised in said sample.

The method for activating immune cells with antitumor activity may comprise steps in addition to those explicitly mentioned above. For example, further steps may relate, e.g., to providing the recombinant virus of the family Paramyxoviridae for step a), administering further activating compounds, e.g. cytokines, to the immune cells in step b), or separating immune cells from cancer cells after step b). Moreover, one or more of said steps may be performed by automated equipment.

Moreover, the present invention relates to a recombinant virus of the family Paramyxoviridae of the present invention for use in treatment of inappropriate cell proliferation.

The term “inappropriate cell proliferation” relates to any proliferation of cells of a subject which is not appropriate to the physiological state of said subject and/or to the tissue context of said cells. Preferably, inappropriate cell proliferation is caused or aggravated by an inhibition or insufficient activation of the immune system, more preferably inhibition or insufficient activation of T cells. Also preferably, inappropriate cell proliferation is cancer.

The present invention further relates to a kit comprising at least

    • (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
    • (ii) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 fusion polypeptide of the present invention;
    • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
    • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae of (i) and/or (ii) and/or the polynucleotide encoding the recombinant virus of the family Paramyxoviridae according to (iii); or
    • (v) any combination of (i) to (iv),
    • housed in a container.

The term “kit”, as used herein, refers to a collection of the aforementioned components. Preferably, said components are combined with additional components, preferably within an outer container. The outer container, also preferably, comprises instructions for carrying out a method of the present invention. Examples for such the components of the kit as well as methods for their use have been given in this specification. The kit, preferably, contains the aforementioned components in a ready-to-use formulation. Preferably, the kit may additionally comprise instructions, e.g., a user's manual for applying the recombinant virus of the family Paramyxoviridae with respect to the applications provided by the methods of the present invention. Details are to be found elsewhere in this specification. Additionally, such user's manual may provide instructions about correctly using the components of the kit. A user's manual may be provided in paper or electronic form, e.g., stored on CD or CD ROM. The present invention also relates to the use of said kit in any of the methods according to the present invention.

Moreover, the present invention relates to a use of

    • (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
    • (ii) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 fusion polypeptide of the present invention;
    • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
    • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae of (i) and/or (ii) and/or the polynucleotide encoding the recombinant virus of the family Paramyxoviridae according to (iii); or
    • (v) any combination of (i) to (iv),
    • for the manufacture of a medicament for treating inappropriate cell proliferation, preferably for treating cancer.

Summarizing the findings of the present invention, the following embodiments are preferred:

1. A recombinant virus of the family Paramyxoviridae, comprising an expressible polynucleotide encoding an IL-12 polypeptide, wherein said IL-12 polypeptide is an IL-12 fusion polypeptide comprising a p40 subunit of an IL-12 and a p35 subunit of an IL-12.

2. The recombinant virus of the family Paramyxoviridae of embodiment 1, wherein said p40 subunit and said p35 subunit of said IL-12 fusion polypeptide are from the same species.

3. The recombinant virus of the family Paramyxoviridae of embodiment 1 or 2, wherein said p40 subunit and said p35 subunit of said IL-12 fusion polypeptide are a mouse p40 subunit and a mouse p35 subunit or a variant thereof, preferably are a human p40 subunit and a human p35 subunit or a variant thereof.

4. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 3, wherein said p40 subunit and said p35 subunit of said IL-12 fusion polypeptide are a Claim subunit and a mouse p35 subunit, preferably are a human p40 subunit and a human p35 subunit.

5. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 4, wherein said IL-12 fusion polypeptide comprises the structure p40-linker-p35.

6. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 5, wherein said linker is -(glycine4-serine)n-, with n=1 to 10, preferably n=2 to 5, more preferably n=3.

7. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 5, wherein said linker is -(glycine6-serine)-.

8. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 7, wherein said expressible polynucleotide encoding an IL-12 is comprised in the genome of the recombinant virus of the family Paramyxoviridae in a region corresponding to the region intervening the P and the M gene of measles virus.

9. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 8, further comprising at least one expressible polynucleotide encoding a further activator of the immune response.

10. The recombinant virus of the family Paramyxoviridae of embodiment 9, wherein said further activator of the immune response is an immunoglobulin or fragment thereof.

11. The recombinant virus of the family Paramyxoviridae of embodiments 9 or 10, wherein said further activator of the immune response is a secreted immunoglobulin.

12. The recombinant virus of the family Paramyxoviridae of any one of embodiments 9 to 11, wherein said further activator of the immune response is an Fc domain of an antibody.

13. The recombinant virus of the family Paramyxoviridae of any one of embodiments 9 to 12, wherein said further activator of the immune response is a secreted soluble Fc domain of a human IgG1 antibody.

14. The recombinant virus of the family Paramyxoviridae of any one of embodiments 9 to 13, wherein said further activator of the immune response is a secreted soluble activator of the immune response.

15. The recombinant virus of the family Paramyxoviridae of any one of embodiments 9 to 14, wherein said further activator of the immune response is a single-chain antibody or a nanobody.

16. The recombinant virus of the family Paramyxoviridae of any one of embodiments 9 to 15, wherein said further activator of the immune response is a secreted soluble anti-PD-L1 antibody.

17. The recombinant virus of the family Paramyxoviridae of embodiment 16, wherein said secreted soluble anti-PD-L1 antibody comprises an amino acid sequence according to SEQ ID NO: 2.

18. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 17, wherein said recombinant virus is a recombinant Morbillivirus, preferably, a recombinant measles virus (MV).

19. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 18, wherein said recombinant MV is derived from MV strain Edmonston A or B, preferably B, more preferably from MV vaccine strain Schwarz/Moraten.

20. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 19, wherein the at least one expressible polynucleotide encoding an IL-12 polypeptide is comprised in a polynucleotide encoding the recombinant virus of the family Paramyxoviridae.

21. The recombinant virus of the family Paramyxoviridae of any one of embodiments 1 to 20, wherein said polynucleotide encoding the recombinant virus of the family Paramyxoviridae comprises the nucleic acid sequence of any one of SEQ ID Nos: 4 to 7, 14, and 15.

22. A polynucleotide encoding the recombinant virus of the family Paramyxoviridae according to any one of embodiments 1 to 21.

23. The polynucleotide according to embodiment 22, wherein said polynucleotide comprises the nucleic acid sequence any one of SEQ ID Nos: 4 to 7, 14, and 15.

24. A host cell comprising the recombinant virus of the family Paramyxoviridae according to any one of embodiments 1 to 21 and/or the polynucleotide encoding the recombinant virus of the family Paramyxoviridae according to embodiment 21 or 22.

25. A medicament comprising

    • (a) (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
      • (ii) a recombinant virus of the family Paramyxoviridae according of any one of embodiments 1 to 21,
      • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
      • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae according to (i) or (ii) and/or a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (iii); or
      • (v) any combination of (i) to (iv); and
    • (b) at least one pharmacologically acceptable excipient.

26. A method for treating cancer in a subject afflicted with cancer, comprising

    • a) contacting said subject with
      • (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
      • (ii) a recombinant virus of the family Paramyxoviridae according of any one of embodiments 1 to 21,
      • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
      • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae according to (i) or (ii) and/or a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (iii); or
      • (v) any combination of (i) to (iv); and, thereby,
    • b) treating cancer in a subject afflicted with cancer.

27. The method of embodiment 26, wherein said recombinant virus of the family Paramyxoviridae of (i) is a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 polypeptide and not comprising an expressible polynucleotide encoding a ligand for an immune checkpoint blockade protein.

28. The method of embodiment 26 or 27, wherein said recombinant virus of the family Paramyxoviridae of (i) is a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 polypeptide as the only expressible polynucleotide encoding an activator of the immune response.

29. The method of any one of embodiments 26 to 28, wherein said cancer is a solid cancer, a metastasis, or a relapse thereof.

30. The method of any one of embodiments 26 to 29, wherein treating cancer is reducing tumor burden.

31. The method of any one of embodiments 26 to 30, wherein said cancer is malignant melanoma, head and neck cancer, hepatocellular carcinoma, pancreatic carcinoma, prostate cancer, renal cell carcinoma, gastric carcinoma, colorectal carcinoma, lymphomas or leukemias.

32. An in vitro method for activating immune cells with antitumor activity in a sample comprising cancer cells and immune cells, comprising

a) contacting said sample comprising cancer cells and immune cells with

    • (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
    • (ii) a recombinant virus of the family Paramyxoviridae according of any one of embodiments 1 to 21,
    • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
    • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae according to (i) or (ii) and/or a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (iii); or
    • (v) any combination of (i) to (iv); and thereby,

b) activating immune cells with antitumor activity comprised in said sample.

33. Use of

    • (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
    • (ii) a recombinant virus of the family Paramyxoviridae according of any one of embodiments 1 to 21,
    • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
    • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae according to (i) or (ii) and/or a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (iii); or
    • (v) any combination of (i) to (iv);

for the manufacture of a medicament for treating cancer.

34. A recombinant virus of the family Paramyxoviridae according to any one of embodiments 1 to 21 and/or a polynucleotide according to embodiment 22 or 23 for use in medical treatment.

35. A recombinant virus of the family Paramyxoviridae

(i) comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist; and/or

(ii) according of any one of embodiments 1 to 21,

for use in treatment of inappropriate cell proliferation.

36. The recombinant virus of the family Paramyxoviridae for use of embodiment 35, wherein treatment of inappropriate cell proliferation is cancer treatment.

37. A kit comprising

    • (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist;
    • (ii) a recombinant virus of the family Paramyxoviridae according of any one of embodiments 1 to 21,
    • (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii),
    • (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae according to (i) or (ii) and/or a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (iii); or
    • (v) any combination of (i) to (iv);

housed in a container.

All references cited in this specification are herewith incorporated by reference with respect to their entire disclosure content and the disclosure content specifically mentioned in this specification.

The following Examples shall merely illustrate the invention. They shall not be construed, whatsoever, to limit the scope of the invention.

EXAMPLES Example 1 Cell Culture

Vero African green monkey kidney cells were obtained from the American Type Culture Collection (Manassas, VA). Vero-αHis cell line stably expressing a single chain antibody (38cab) against His6tag (Nakamura et al. 2005) was a kind gift of S. J. Russel (Mayo Clinic, Rochester, Minn.). Murine colon adenocarcinoma cells MC38cea (transduced for stable expression of human CEA antigen) and the parental MC38 cell line were a gift of R. Cattaneo (Mayo Clinic, Rochester, Minn.). B16-CD20 have previously been generated by transducing the parental cell line with a lentiviral vector encoding human CD20 (Engeland et al. 2014). All cell lines were cultivated in either Dulbecco's modified Eagle's medium (DMEM; Life Technologies, Darmstadt, Germany) or Roswell Park Memorial Institute 1640 medium (RPMI 1640; Life Technologies) supplemented with 10% Fetal Calf Serum (FCS) at 37° C. in a humidified atmosphere with 5% CO2 and routinely tested for mycoplasma contamination.

Example 2 Cloning of Recombinant MeVac Genomes

The cDNA plasmids encoding recombinant MeV genomes were constructed on the basis of the commercially used Schwarz/Moraten vaccine strain (MeVac) (Combredet et al. 2003). Transgenes were inserted in additional transcription units (ATUs) containing additional gene-end gene-start signals and a unique cloning site. Transgenes smaller than 1 kbp including murine GM-CSF (426 bp), murine IP-10 (312 bp) and eGFP (720 bp) were inserted into the leader position of pcMeVac as Mlul-Ascl fragments via the unique Ascl restriction site. The mGM-CSF and eGFP fragments were amplified from the MeV Edmonston B (Nse) vaccine strain genomes encoding the respective transgenes (Grossardt et al. 2013). The mIP-10 (mCxcl10) gene was amplified with primers flanking the novel construct and adding a Mlul site and a Kozak sequence (GCCACC) in the 5′-end and a two nucleotide TA spacer and Ascl site in the 3′-end using cDNA obtained from murine splenocytes.

Cassette encoding a murine IL-12 fusion protein (FmIL-12) consisting of murine IL-12 p40 and p35 subunits linked by a (Gly4Ser)3 for insertion into MeV genome had previously been constructed by C. Grossardt (Grossardt 2013) based on results of Lieschke and colleagues (Lieschke et al. 1997). FmIL-12 construct (1650 bp) was excised from pCG expression vector (constructed by C. Grossardt) as Paul-Mlul fragment and inserted into the MeVac genome downstream the P ORF via the unique MauBl cloning site.

Antibodies against negative murine T cell regulators CTLA-4 and PD-L1 as well as soluble form of murine CD80 T cell costimulatory molecule and human IgG1-Fc fragment for use as a control were inserted into the ATU downstream the H gene. Cassettes encoding antibodies against murine CTLA-4 and PD-L1 and human IgG1-Fc fragment previously designed by C. E. Engeland (Engeland et al. 2014) were used as templates. The respective constructs were excised from pCG expression vectors as Miul-Paul fragments and inserted into the pcMeVac H-ATU via the unique MauBl cloning site.

Murine CD80 molecule was inserted for expression from MeVac in a soluble form. CD80-Fc was constructed by fusing the extracellular part of the murine CD80 with the same human IgG1-Fc region as used in both anti-CTLA-4 and anti-PD-L1 constructs via fusion PCR. The first PCR fragment consisting of the Mlul restriction site followed by Kozak sequence (GCCACC), murine CD80 signal peptide, extracellular part (up to the asparagine in position 246) of murine CD80 and first 26 nucleotides of the hinge of IgG1-Fc was synthesized using pCG vector encoding murine CD80 as a template. The second PCR fragment consisting of the human IgG1-Fc region followed by myc tag, stop codon and Ascl restriction site was synthesized using pCG vector encoding human IgG1-Fc as a template. The obtained PCR products were fused with flanking primers in an overlap PCR obtaining the mCD80-Fc construct of 1614 bp. The mCD80-Fc was inserted into the pcMeVac H-ATU as a Mlul-Ascl fragment via the unique MauBl cloning site.

MeVac genomes encoding the previously described transgenes with a fully retargeted MeV H attachment gene were constructed to allow targeted transduction of murine MC38cea and B16-CD20 cells. MeVac H gene was exchanged for H gene with mutated attachment sites to the natural MeV receptors CD46 and CD150, fused to a single chain antibody (39cab) against human CEA or CD20 and containing a C-terminal His6tag. The retargeting system allows a flexible change of the targeted antigen by exchanging the specific 39cab as a Sfil-Notl fragment.

Example 3 Virus Propagation and Titration

Recombinant MeVac particles were obtained from cDNA constructs according to Radecke et al. (Radecke et al. 1995) and propagated on Vero-αHis cells according to Nakamura et al. (Nakamura et al. 2005). For propagation Vero-αHis cells were infected at a multiplicity of infection (MOI) of 0.03 and cultivated at 37° C. 5% CO2 until syncytia had spread across the whole cell layer (36-48 h post infection). Subsequently culture medium was completely removed, cells were scraped and collected and viral particles released by one freeze-thaw cycle. Cellular debris was removed by centrifugation at 6000×g for 5 min. The amount of viral particles was determined by 1:10 serial dilution titrations in octuplicates on 1.5×104Vero-αHis cells per well in 96-well cell culture plates. Individual syncytia were counted 72 h post infection and titers calculated as cell infectious units per ml (ciu/ml).

Example 4 Statistical Analyses

Statistical analyses were performed using GraphPad Prism software (version 5.04; GraphPad Software, La Jolla, Calif.). Tumor volumes and ELISA results in restimulation experiments were analysed by one-way ANOVA with Tukey's multiple comparison test. Survival curves were analyzed by log-rank (Mantel-Cox) test with Bonferroni-Holm correction for multiple comparisons. Result was considered statistically significant if p value was lower than 0.05 after correcting for multiple comparisons.

Example 5 Characterization of Virus Replication

Vero-αHis and MC38cea cells were seeded in 12-well plates (1×105 cells per well). After 12 h the cell culture medium was removed and cells were infected with the respective viruses at MOI=3 in 300 μl OptiMEM in triplicates for each time point and cultivated at 37° C. 5% CO2. After adsorption for ca. 2 h the inoculum was removed and substituted with 1 ml DMEM+10% FCS per well. Cells were scraped in the culture medium at the designated time points, collected and snap frozen in liquid nitrogen. The amount of viral particles was determined by 1:10 serial dilution titrations in quadruplicates on 1.5×104 Vero-αHis cells per well in 96-well cell culture plates. Individual syncytia were counted 72 h post infection and titers calculated as ciu/ml.

Example 6 Assessment of Virus Cytotoxic Potential In Vitro

MC38cea cells were seeded in 6-well plates (2×105 cells per well). After 12 h the cell culture medium was removed and cells were infected with the respective viruses at MOI=5 in 800 μl OptiMEM in triplicates for each time point and cultivated at 37° C. 5% CO2. After adsorption for ca. 2 h the inoculum was removed and substituted with 2 ml DMEM+10% FCS per well. At the designated time points cell viability was determined using Colorimetric Cell Viability Kit III (XTT) (PromoKine, Heidelberg, Germany) according to instructions of the manufacturer.

Example 7 Characterization of Transgene Expression

MC38cea cells were seeded in 12-well plates (1×105 cells per well). After 12 h the cell culture medium was removed and cells were infected with the respective viruses at MOI=3 in 300 μl OptiMEM in triplicates for each time point and cultivated at 37° C. 5% CO2. After adsorption for ca. 2 h 700 μl DMEM+10% FCS per well was added. Supernatants were collected at the designated time points. Time point Oh was represented by inoculum in OptiMEM used for infection. Expression of the respective immunomodulators was detected by ELISA. Commercially available ELISA kits were used for detection of mGM-CSF, FmIL-12, mIP-10 (R&D Systems, Wiesbaden, Germany) and CD80-Fc (Boster Biological Technology, Offenbach, Germany) according to instructions of the manufacturer. Anti-CTLA-4 and anti-PD-L1 were detected by binding to their respective murine proteins. Ninety-six well plates (Nunc Maxisorp, Thermo Fisher Scientific, Schwerte, Germany) were coated with 100 ng recombinant His-tagged murine CTLA-4 or PD-L1 (Life Technologies). Wells were blocked and 100 μl of the respective samples were added and incubated for 2 h. After washing the antibodies were detected with anti-human IgG-Fc Biotin (clone HP-6071; Sigma-Aldrich, Taufkirchen, Germany), Peroxidase conjugated Streptavidin (Dianova, Hamburg, Germany) and 1-Step Ultra-TMB ELISA Substrate Solution (Thermo Scientific, Karlsruhe, Germany). Absorbance was measured using Infinite M200 Pro microplate reader and i-control software (Tecan, Mannedorf, Switzerland).

Example 8 Flow Cytometry for Detection of anti-PD-L1 Binding to MC38cea Cells

Vero-αHis cells were seeded in 15 cm cell culture dishes and infected with MeVac encoding anti-PD-L1 or IgG1-Fc with MOI=0.03. Supernatants were collected (15m1 per plate) when syncytia had spread over the whole cell layer (ca. 36 h post infection). 1×106 MC38cea cells were incubated with anti-PD-L1 or IgG1-Fc containing supernatant previously collected from one fully infected 15 cm dish for 1 h with rotation at room to. After washing the bound anti-PD-L1 was detected by staining with anti-HA (clone HA-7; Sigma-Aldrich) and goat anti-mouse IgG PE (polyclonal; BD Biosciences, Heidelberg, Germany). The stained cells were resuspended in DPBS with 0.2 μg/ml DAPI (Sigma-Aldrich) and directly acquired on LSRII flow cytometer (BD Biosciences) collecting at least 10000 events per sample.

Example 9 Isolation of Murine Splenocytes

Spleens were aseptically isolated and maintained in RPMI 1640 (Life Technologies, Darmstadt, Germany) at 4° C. until further processing. Spleen was passed through a 100 μm nylon cell strainer (BD Biosciences, Heidelberg, Germany) into 10 ml RPMI 1640 and cells were pelleted at 300×g for 5 min. For red blood cell lysis pellet was resuspended in 1 ml ACK Lysing solution (Life Technologies), incubated 10 min at room to and centrifuged at 300×g for 5 min. Cells were resuspended in DPBS (Life Technologies) and cell concentration determined using Neubauer hemocytometer and Trypan blue (Sigma-Aldrich) staining for dead cell exclusion.

Example 10 Functional Assay for MeVac Encoded Anti-PD-L1, CD80-Fc and Anti-CTLA-4

Vero-αHis cells were seeded in 15 cm cell culture dishes and infected with MeVac encoding anti-PD-L1, anti-CTLA-4, CD80-Fc or IgG1-Fc with MOI=0.03. Supernatants were collected (15 ml per plate) when syncytia had spread over the whole cell layer (ca. 36 h post infection). 2×105 MC38cea cells were incubated with 2 ml medium collected from the Vero-αHis infected with the respective viruses for 5 min with rotation at room to and pelleted by centrifugation 5 min at 300×g. The procedure was repeated six times. The treated cells were resuspended in 100 μl activation medium—RPMI 1640 supplemented with 5% FCS, 1% Penicillin-Streptomycin (Life Technologies), 500 μM ionomycin (Cayman Chemical Company, Hamburg, Germany) and 5 μM PMA (Cayman Chemical Company) and seeded in 96-well plate. 2×105 freshly isolated splenocytes from C57BL/6J mouse in 100 μl activation medium were added per each well with the treated MC38cea cells. Cells were cocultivated 24 h at 37° C. 5% CO2 and supernatants collected subsequently. IFN-γ concentration was determined using mouse IFN gamma ELISA Ready-SET-Go!® (eBioscience, Frankfurt am Main, Germany) according to the instructions of the manufacturer.

Example 11 Functional Assay for MeVac Encoded FmIL-12

Vero-αHis cells were seeded in 15 cm cell culture dishes and infected with MeVac encoding FmIL-12 or eGFP. Supernatants were collected (15 ml per plate) when syncytia had spread over the whole cell layer (ca. 36 h post infection). FmIL-12 concentration was assessed using Mouse IL-12 p70 Quantikine ELISA Kit (R&D Systems). 2×106 freshly isolated splenocytes from a C57BL/6J mouse were resuspended in RPMI 1640 supplemented with 10% FCS, 1% Penicillin-Streptomycin solution and 50 U/ml recombinant murine IL-2 (Miltenyi, Bergisch Gladbach, Germany) with varying concentrations of MeVac encoded FmIL-12 or respective parts of supernatant from cells infected with eGFP encoding MeVac. Splenocytes were seeded in 12-well plates and incubated 48 h at 37° C. 5% CO2. Supernatants were collected and IFN-γ concentration assessed using mouse IFN gamma ELISA Ready-SET-Go!® (eBioscience) according to the instructions of the manufacturer.

Example 12 Assessment of Therapeutic Efficacy In Vivo

MC38cea cells were subcutaneously (s.c.) implanted into six to eight weeks old C57BI/6J mice (Harlan Laboratories, Rossdorf Germany or DKFZ, Heidelberg, Germany). When average tumor volume reached 50-100 mm3 (depending on experiment) treatment was initiated. Mice received intratumoral (i.t.) injections with the respective viruses on four or five consecutive days with 5×105 or 1×106 ciu in 100 μl. Mice in the mock group received treatment with 100 μl OptiMEM. Tumor volume was determined every third day measuring largest and smallest diameter with a caliper and calculating the volume using a formula: largest diameterx(smallest diameter)2×0.5. Mice were sacrificed when tumor volume exceeded 1500 mm3, ulceration occurred or signs of severe illness were observed.

Example 13 Antigen Specific IFN-γ Memory Recall with Murine Splenocytes

MC38cea, MC38 and B16 cells were treated with 20 μg/ml mitomycin-C (Sigma-Aldrich) for 2 h with shaking at 37° C. After subsequent washing three times with DPBS cells were resuspended in activation medium containing RPMI 1640 supplemented with 10% FCS, 1% Penicillin-Streptomycin and 50 U/ml recombinant murine IL-2. Freshly isolated murine splenocytes were resuspended in the same activation medium. Cocultures were prepared in 24-well plates seeding 1×105 mitomycin-c treated tumor cells or 1×106 ciu MeVac with 1×106 splenocytes per well in 0.5 ml total volume of activation medium. As controls 1×106 splenocytes were cocultivated also with Vero-αHis or DLD-1 cell lysates prepared by lysis of 1×106cells per ml with one freeze-thaw cycle. Cells were cocultivated for 48 h, supernatants collected and IFN-y concentration assessed using mouse IFN gamma ELISA Ready-SET-Go!® (eBioscience) according to the instructions of the manufacturer.

REFERENCES

  • Combredet, C. et al., 2003. A Molecularly Cloned Schwarz Strain of Measles Virus Vaccine Induces Strong Immune Responses in Macaques and Transgenic Mice A Molecularly Cloned Schwarz Strain of Measles Virus Vaccine Induces Strong Immune Responses in Macaques and Transgenic Mice.
  • Engeland, C. E. et al., 2014. CTLA-4 and PD-L1 Checkpoint Blockade Enhances Oncolytic Measles Virus Therapy. Molecular Therapy, 22(11), pp. 1949-1959.
  • Grossardt, C., 2013. Engineering Targeted and Cytokine-armed Oncolytic Measles Viruses. Ruperto-Carola University of Heidelberg.
  • Grossardt, C. et al., 2013. Granulocyte-Macrophage Colony-Stimulating Factor-Armed Oncolytic Measles Virus Is an Effective Therapeutic Cancer Vaccine. Human Gene Therapy, 24(7), pp. 644-654.
  • Lieschke, G. J . et al., 1997. Bioactive murine and human interleukin-12 fusion proteins which retain antitumor activity in vivo. Nature biotechnology, 15(1), pp. 35-40.
  • Nakamura, T. et al., 2005. Rescue and propagation of fully retargeted oncolytic measles viruses. Nature biotechnology, 23(2), pp. 209-14.
  • Radecke, F. et al., 1995. Rescue of measles viruses from cloned DNA. The EMBO journal, 14(23), pp. 5773-84. Available at:

Claims

1. A recombinant virus of the family Paramyxoviridae, comprising an expressible polynucleotide encoding an IL-12 polypeptide, wherein said IL-12 polypeptide is an IL-12 fusion polypeptide comprising a p35 subunit of an IL-12 and a p40 subunit of an IL-12.

2. The recombinant virus of the family Paramyxoviridae of claim 1, wherein said p35 subunit and said p40 subunit of said IL-12 fusion polypeptide are from the same species.

3. The recombinant virus of the family Paramyxoviridae of claim 1, wherein said IL-12 fusion polypeptide comprises the structure p35-linker-p40.

4. The recombinant virus of the family Paramyxoviridae of claim 1, wherein said linker is -(glycine4-serine)n-, with n=1 to 10.

5. The recombinant virus of the family Paramyxoviridae of claim 1, further comprising at least one expressible polynucleotide encoding a further activator of the immune response.

6. The recombinant virus of the family Paramyxoviridae of claim 5, wherein said further activator of the immune response is a secreted immunoglobulin.

7. The recombinant virus of the family Paramyxoviridae of claim 5, wherein said further activator of the immune response is an Fc domain of an antibody.

8. The recombinant virus of the family Paramyxoviridae of claim 5, wherein said further activator of the immune response is a secreted soluble activator of the immune response.

9. The recombinant virus of the family Paramyxoviridae of claim 5, wherein said further activator of the immune response is a secreted soluble anti-PD-L1 antibody.

10. The recombinant virus of the family Paramyxoviridae of claim 1, wherein said recombinant virus is a recombinant Morbillivirus.

11. The recombinant virus of the family Paramyxoviridae of claim 1, wherein said recombinant virus is derived from a measles virus strain Edmonston A or B.

12. The recombinant virus of the family Paramyxoviridae of claim 1, wherein the at least one expressible polynucleotide encoding an IL-12 polypeptide is comprised in a polynucleotide encoding the recombinant virus of the family Paramyxoviridae.

13. The recombinant virus of the family Paramyxoviridae of claim 1, wherein said polynucleotide encoding the recombinant virus of the family Paramyxoviridae comprises the nucleic acid sequence of any one of SEQ ID NOs:4 to 7, 14, and 15.

14. A medicament comprising

(a) (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist; (ii) a recombinant virus of the family Paramyxoviridae according to claim 1, (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii), (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae according to (i) or (ii) and/or a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (iii); or (v) any combination of (i) to (iv); and
(b) at least one pharmacologically acceptable excipient.

15. A method for treating cancer in a subject afflicted with cancer, comprising

a) contacting said subject with (i) a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 and not comprising an expressible polynucleotide encoding a CTLA-4 antagonist, a PD-1 antagonist, a CD80 antagonist, a CD86 antagonist, or a PD-L1 antagonist; (ii) a recombinant virus of the family Paramyxoviridae according of claim 1, (iii) a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (i) and/or (ii), (iv) a host cell comprising the recombinant virus of the family Paramyxoviridae according to (i) or (ii) and/or a polynucleotide encoding the recombinant virus of the family Paramyxoviridae of (iii); or (v) any combination of (i) to (iv); and, thereby,
b) treating cancer in a subject afflicted with cancer.

16. The method of claim 15, wherein said recombinant virus of the family Paramyxoviridae of (i) is a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 polypeptide and not comprising an expressible polynucleotide encoding a ligand for an immune checkpoint blockade protein.

17. The method of claim 15, wherein said recombinant virus of the family Paramyxoviridae of (i) is a recombinant virus of the family Paramyxoviridae comprising an expressible polynucleotide encoding an IL-12 polypeptide as the only expressible polynucleotide encoding an activator of the immune response.

18. The method of claim 15, wherein said cancer is a solid cancer, a metastasis, or a relapse thereof.

19. The method of claim 15, wherein treating cancer is reducing tumor burden.

20. The method of claim 15, wherein said cancer is malignant melanoma, head and neck cancer, hepatocellular carcinoma, pancreatic carcinoma, prostate cancer, renal cell carcinoma, gastric carcinoma, colorectal carcinoma, lymphomas or leukemias.

21. The recombinant virus of the family Paramyxoviridae of claim 1, wherein said p35 subunit and said p40 subunit of said IL-12 fusion polypeptide are a mouse p35 subunit and a mouse p40 subunit or a variant thereof, or a human p35 subunit and a human p40 subunit or a variant thereof.

Patent History
Publication number: 20190315825
Type: Application
Filed: Jun 28, 2019
Publication Date: Oct 17, 2019
Applicants: DEUTSCHES KREBSFORSCHUNGSZENTRUM (Heidelberg), RUPRECHT-KARLS UNIVERSITAET (Heidelberg)
Inventors: Guy UNGERECHTS (Heidelberg), Christine ENGELAND (Heidelberg), Ruta VEINALDE (Heidelberg)
Application Number: 16/456,773
Classifications
International Classification: C07K 14/54 (20060101); C07K 16/30 (20060101); C07K 16/28 (20060101);