VSV CHIMERIC VECTORS

Abstract
The present invention relates to VSV chimeric vectors, characterized in that the vectors comprise a gene coding for a glycoprotein GP of the Dandenong virus (DANDV) or Mopeia vims (MOPV) and lack a functional gene coding for envelope protein G of the VSV. The invention also provides VSV chimeric vector systems. In addition, the invention relates to uses of the VSV chimeric vectors and systems of the invention, including the use in medicine such as in the treatment of solid tumors.
Description

The present invention relates to VSV chimeric vectors, characterized in that the vectors comprise a gene coding for a glycoprotein GP of the Dandenong virus (DANDV) or Mopeia virus (MOPV) and lack a functional gene coding for envelope protein G of the VSV. The invention also provides VSV chimeric vector systems. In addition, the invention relates to uses of the VSV chimeric vectors and systems of the invention, including the use in medicine such as in the treatment of solid tumors.


DESCRIPTION OF THE PRIOR ART

The use of viruses in cancer therapy has been investigated intensively over the last decades. Oncolytic viruses (OVs) are considered as important agents in cancer treatment. The oncolytic viruses offer the attractive therapeutic combination of tumor-specific cell lysis together with immune stimulation. Moreover, OVs can be genetically modified for optimization of tumor selectivity and enhanced immune stimulation and can be readily combined with other agents such as checkpoint inhibitory antibody molecules and other immune therapeutics. The effectiveness of OVs has been demonstrated in many preclinical studies and recently in humans, with US Food and Drug Administration approval of the oncolytic herpesvirus talimogene laherparepvec [1, 2].


To date, a wide range of viruses with diverse properties are under preclinical and clinical investigation. Oncolytic viruses range in size and complexity from large, double-stranded DNA viruses such as vaccinia virus (190 kb) [3] and herpes simplex virus type 1 (152 kb) [4] to small single stranded RNA viruses such as vesicular stomatitis virus (VSV) [5,6], measles virus (MV) [7] or Newcastle disease virus (NDV) [8] with a genome size between 11 and 16 kb. However, every oncolytic virus platform has advantages and disadvantages. Clearly, a promising approach to oncolysis is to match different viruses with tumor types naturally permissive for their replication. On the other hand, the broad natural receptor tropism of some platforms, for example, vaccinia- and vesicular stomatitis virus-based vectors, allows application for many different types of cancer.


VSV-based vector platforms are considered as very promising virotherapy not only due to its broad receptor tropism but also because of its enormous capacity to replicate in permissive cells and its ability to induce a strong CPE causing local inflammation mounting an immune response within the infected tumor tissue. However, there are several drawbacks using wild type VSV in oncolytic virotherapy. First, wildtype VSV stains are considered to be neurotoxic. Wild type VSV can cause severe encephalitis leading to death if the virus accidentally crosses the blood-brain-barrier and spreads within the brain of the treated individual. Second, VSV infected individuals are able to rapidly mount a strong humoral response with high antibody titers directed mainly against the nucleo- and the glycoprotein. In addition to a strong CTL response with CD8+ cells specific to an epitope located within the nucleoprotein, neutralizing antibodies binding to the envelope glycoprotein G of VSV are considered to be important for the control of VSV infection. Neutralizing antibodies targeting the glycoprotein G of VSV are able to limit virus spread and they are able to mediate protection of individuals from VSV re-infection. Vector neutralization, however, limits repeated application of the oncolytic agent to the cancer patient.


To eliminate these drawbacks of VSV wildtype, a recombinant VSV vector named VSV-GP has been described in WO 2010/040526. In VSV-GP the coding region of the endogenous glycoprotein VSV-G, which is considered to be the main determinant for neurotoxicity and vector neutralization, was replaced by the envelope glycoprotein GP of lymphocytic choriomeningitis virus (LCMV), strain WE-HPI. Advantages offered by the replacement of VSV viral envelope protein G with LCMV-GP (WE HPI) are (i) the loss of VSV-G mediated neurotoxicity [9] and (ii) a lack of vector neutralization by antibodies in the mouse model only [10,11]. However, there is still a need for vectors with reduced induction of neutralizing antibodies and thus an improved suitability for use in therapy.


The technical problem to be solved by the present invention is thus the provision of improved VSV chimeric vectors and corresponding uses and methods. The technical problem is solved by the embodiments provided herein and as claimed.


SUMMARY OF THE INVENTION

The present invention provides VSV chimeric vectors, characterized in that the vectors comprise a gene coding for a glycoprotein GP of the Dandenong virus (DANDV) or Mopeia (MOPV) virus or a functional fragment or variant thereof and lack a functional gene coding for envelope protein G of the VSV. It is preferred within the present invention that the envelope protein G of VSV is replaced by GP of the DANDV or MOPV or a functional fragment or variant thereof. In an alternative embodiment of the invention, VSV chimeric vectors are provided, characterized in that the vectors comprise a gene coding for a glycoprotein GP of the Ippy virus (IPPYV), Latino virus (LATV) or Olivero virus (OLIVV) or a functional fragment or variant thereof and lack a functional gene coding for envelope protein G of the VSV.


In an attempt to improve the VSV-based OVs in the art, the inventors surprisingly found that a surface protein GP of DANDV or MOPV in a VSV vector leads to a chimeric vector which shows no or only low titers of neutralizing antibodies and maintains replicative capacity of the underlying VSV vector in tumor cells. In addition, both the tropism and the lack of neurotoxicity of the VSV-GP vector are preserved.


The inventors have thus surprisingly found that envelope protein GP of arenaviruses, in particular the DANDV and the MOPV, when used in the framework of VSV leads to recombinant vectors that do not show neurototoxicity after intra-cranial application in mice. At the same time, the chimeric VSV vectors surprisingly maintain cellular tropism of the VSV vector of the prior art. Much to the surprise of the inventors, improved killing of selected human tumor cell lines due to the virus induced cytopathic effect (CPE) could be shown when compared to VSV with GP of LCMV. This effect was confirmed in vivo, in particular the efficacy of VSV-G(x)-DANDV und VSV-G(x)-MOPV in a human lung cancer xenograft model was unexpectedly improved in comparison to VSV-LCMV-GP. Additionally, VSV-G(x)-DANDV und VSV-G(x)-MOPV showed a significantly delayed induction of vector-neutralizing antibodies in the rabbit model after intra-venous immunization.


Thus, in one embodiment, the invention relates to VSV chimeric vectors, characterized in that the vectors comprise a gene coding for a glycoprotein GP of the Dandenong virus (DANDV) or Mopeia virus (MOPV) and lack a functional gene coding for envelope protein G of the VSV, wherein the vectors show reduced induction of neutralizing antibodies as compared to a VSV vector pseudotyped with GP of LCMV under identical conditions. In a further embodiment of the invention, the vector comprises a gene coding for a glycoprotein GP of the Ippy, Olivero or Latino virus. A “neutralizing antibody” is an antibody that binds to an antigen and thereby prevents biological effects imparted by the antigen. Many antigens that are foreign to a host induce a reaction of the hosts' immune system including the production of neutralizing antibodies. The VSV chimeric vectors of the invention, however, are improved over known chimeric VSV vectors in that they only lead to a reduced induction of neutralizing antibodies, and may thus show improved biological effects on the targeted cells of the host. The skilled person is well aware of means and methods how to determine whether neutralizing antibodies are induced and to what extent. Thus, the skilled person can readily determine whether the VSV chimeric vectors of the invention induce neutralizing antibodies and whether the induction is reduced in comparison to a VSV chimeric vector pseudotyped with GP of LCMV. Such assays may provide a quantitative or qualitative result. For example, assays may provide an absolute measure of the neutralizing antibody induction ability which may then be compared to a pre-determined number for VSV chimeric vector pseudotyped with GP of LCMV. Additionally or alternatively, an assay may quantitatively provide the relative ability to induce neutralizing antibodies. An exemplary assay which may be employed within the present invention is provided in Example 5. As shown in FIG. 9, the results surprisingly and unexpectedly show that the vectors of the invention show significantly reduced induction of neutralizing antibodies. As such, an exemplary assay may comprise use of p-nitro-phenylphosphate (pNPP) in the determination of the presence of neutralizing antibodies that is added to serum samples from different time points post immunization. In the absence of nAbs, a GP-pseutotyped virus (LCMV-GP, DANDV-GP or MOPV-GP, respectively), that harbors the gene of the secreted embryonic alkaline phosphatase (SEAP) instead of the surface protein, is able to infected BHK21CI.13 cells resulting in the expression of SEAP. In contrast, if the virus is neutralized by antibodies in the sera, the virus was not able to infect the cells and hence SEAP is not expressed. Thus, nAb activity can be determined as a function of SEAP activity. By non-linear curve fitting EC50 values can be calculated.


In one embodiment of the invention, the improved biological effect may be an increased killing of tumor cells as compared to a VSV vector pseudotyped with GP of LCMV under identical conditions. Thus, the invention, in one embodiment relates to VSV chimeric vectors, characterized in that the vectors comprise a gene coding for a glycoprotein GP of the Dandenong (DANDV) virus or Mopeia (MOPV) virus and lack a functional gene coding for envelope protein G of the VSV, wherein the vectors show increased killing of tumor cells as compared to a VSV vector pseudotyped with GP of LCMV under identical conditions. The skilled person is well aware of means and methods how to determine whether tumor cells are killed and to what extent. Thus, the skilled person can readily determine whether the VSV chimeric vectors of the invention kill tumor cells and whether the killing is increased in comparison to a VSV chimeric vector that expresses GP of LCMV. Such assays may provide a quantitative or qualitative result. For example, assays may provide an absolute measure of the tumor cell killing ability which may then be compared to a pre-determined number for VSV chimeric vector that expresses GP of LCMV. Additionally or alternatively, an assay may quantitatively provide the relative ability to kill tumor cells. An exemplary assay is provided in Example 4. Such an assay may comprise the pre-incubation of cells to be infected with IFN and the subsequent infection with the VSV chimeric vectors of the invention and/or VSV-GP of LCMV. After incubation, e.g. for three days, cells may be analyzed for viability using an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide)-based in vitro cytotoxicity assay, according to the manufacturer's recommendations. Samples can then be measured in a conventional microplate reader at 550 nm. Values can be normalized to mock-infected cells that were not pre-treated with interferon (IFN), and represented as a percentage of viable cells.


The Dandenong virus (DANDV) is an old world arenavirus [12]. To date, there is only a single strain known to the person skilled in the art, which comprise a glycoprotein GP and which may be employed within the present invention as donor of the GP comprised in the VSV chimeric vectors of the invention. The DANDV GP comprised in the VSV chimeric vectors of the invention has more than 6 glycosylation sites, in particular 7 glycosylation sites. An exemplary preferred GP is that as comprised in DANDV as accessible under Genbank number EU136038. Thus, in one embodiment, the gene coding for GP of the DANDV has a nucleic acid sequence as shown in SEQ ID NO:1 or a sequence having at least 80, 85, 90, 95, 96, 97, 98, 99 or 100% sequence identity to the nucleic acid sequence of SEQ ID NO:1 while the functional properties of the chimeric VSV vector comprising a GP encoded by a nucleic acid sequence as shown in SEQ ID NO:1 are maintained.


The Mopeia virus (MOPV) is an old world arenavirus [13]. There are several strains known to the person skilled in the art, which comprise a glycoprotein GP and which may be employed within the present invention as donor of the GP comprised in the VSV chimeric vectors of the invention. The MOPV GP comprised in the VSV chimeric vectors of the invention has more than 6 glycosylation sites, in particular 7 glycosylation sites. An exemplary preferred GP is that as comprised in Mopeia virus as accessible under Genbank number AY772170. Thus, in one embodiment, the gene coding for GP of the MOPV has a nucleic acid sequence as shown in SEQ ID NO:3 or a sequence having at least 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% sequence identity to the nucleic acid sequence of SEQ ID NO:3 while the functional properties of the chimeric VSV vector comprising a GP encoded by a nucleic acid sequence as shown in SEQ ID NO:3 are maintained.


The Ippy virus (IPPYV) is an old world arenavirus that was first isolated from a wild caught rodent, Arvicanthis sp. [14]. There are several strains known to the person skilled in the art, which comprise a glycoprotein GP and which may be employed within the present invention as donor of the GP comprised in the VSV chimeric vectors of the invention. The IPPYV GP comprised in the VSV chimeric vectors of the invention has more than 6 glycosylation sites, preferably more than 8 glycosylation sites, in particular 10 glycosylation sites. An exemplary preferred GP is that as comprised in IPPYV as accessible under Genbank number DQ328877. Thus, in one embodiment, the gene coding for GP of the Ippy virus has a nucleic acid sequence as shown in SEQ ID NO:5 or a sequence having at least 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% sequence identity to the nucleic acid sequence of SEQ ID NO:5 while the functional properties of the chimeric VSV vector comprising a GP encoded by a nucleic acid sequence as shown in SEQ ID NO:5 are maintained.


The Latino virus (LATV) is a new world arenavirus that was first isolated from an adult pregnant female Calomys callosus [15]. There are several strains known to the person skilled in the art, which comprise a glycoprotein GP and which may be employed within the present invention as donor of the GP comprised in the VSV chimeric vectors of the invention. The LATV GP comprised in the VSV chimeric vectors of the invention has more than 6 glycosylation sites, preferably more than 8 glycosylation sites, in particular 10 glycosylation sites. An exemplary preferred GP is that as comprised in LATV as accessible under Genbank number AF485259. Thus, in one embodiment, the gene coding for GP of the LATV has a nucleic acid sequence as shown in SEQ ID NO:7 or a sequence having at least 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% sequence identity to the nucleic acid sequence of SEQ ID NO:7 while the functional properties of the chimeric VSV vector comprising a GP encoded by a nucleic acid sequence as shown in SEQ ID NO:7 are maintained.


The Olivero virus (OLIVV) is a new world arenavirus that was first isolated from the rodent Bolomys obscures [16]. There are several strains known to the person skilled in the art, which comprise a glycoprotein GP and which may be employed within the present invention as donor of the GP comprised in the VSV chimeric vectors of the invention. The OLIW GP comprised in the VSV chimeric vectors of the invention has more than 6 glycosylation sites, preferably more than 8 glycosylation sites, in particular 9 glycosylation sites. An exemplary preferred GP is that as comprised in OLIW as accessible under Genbank number U34248. Thus, in one embodiment, the gene coding for GP of the OLIW has a nucleic acid sequence as shown in SEQ ID NO:9 or a sequence having at least 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% sequence identity to the nucleic acid sequence of SEQ ID NO:9 while the functional properties of the chimeric VSV vector comprising a GP encoded by a nucleic acid sequence as shown in SEQ ID NO:9 are maintained.


In view of the surprising and unexpected technical advantages of the VSV chimeric vectors of the invention described above and as shown in the appended examples, it is within the scope of the present invention to provide the vectors of the invention for medical uses and, thus, vectors specifically designed to be suitable for such uses. Thus, in one embodiment of the invention, the vectors of the invention are provided comprising at least one transgene. A “transgene” is a segment of DNA comprising a gene sequence that has been isolated from one organism and is introduced into a different organism. Within the present invention, the transgene or the transgenes comprised in the vectors of the invention may be of any origin and may have any biological effect on the targeted cells.


In a further embodiment, the invention relates to a VSV chimeric vector system, characterized in that the system comprises at least two complementary replicating VSV vectors, wherein the system comprises genes n, l, p and m coding for proteins N, L, P and M of the VSV, a gene gp coding for Dandenong-GP or Mopeia-GP and lacks a functional gene coding for G protein of the VSV, wherein each vector of the system lacks one of the genes n, l, p, m and gp, and wherein the lacking gene is present on any other vector of the system. The respective GP gene comprised in the VSV chimeric vector system of the invention is one of the genes encoding a GP protein as comprised in the VSV chimeric vector of the invention described above.


The invention, in a further embodiment, relates to chimeric VSV virions, characterized in that the virion comprises a GP protein of the Dandenong virus or Mopeia virus as envelope protein. In a further embodiment, the virion may comprise the GP of the Ippy virus, Latino virus, or Olivero virus as envelope protein. Thus, the invention also provides the virions, viral particles, comprising any of the products of the GP genes described above as envelope protein. In this respect, the skilled person is aware that the envelope protein of a virion serves to identify and bind to receptor sites on the host's cell membrane. Thus, the virions of the invention comprise a chimeric GP protein on their surface as envelope protein, i.e. a GP protein different from the GP protein of VSV. However, the composition of the virions nucleic acid sequence content is not particularly limited. That is, for example, the chimeric VSV virons may comprise a non-functional gene coding for the GP of VSV. The chimeric VSV virons may also lack a gene coding for the GP of VSV. The GP expressed on the surface of the chimeric VSV virion of the invention is a gene product of the gene coding for a GP protein of the Dandenong virus or Mopeia virus as described further above. In a further embodiment, the GP expressed on the surface of the chimeric VSV virion of the invention may be a gene product of the gene coding for a GP of the Ippy virus, Latino virus, or Olivero virus as described further above. Thus, the GP protein may comprise an amino acid sequence as shown in any one of SEQ ID NOs: 2, or 4. Alternatively, the GP protein may comprise an amino acid sequence as shown in any one of SEQ ID NOs: 6, 8 or 10. The chimeric VSV virions of the invention may also comprise as envelope protein a protein comprising an amino acid sequence having 60, 65, 70, 75, 80, 85, 90 or 95% sequence identity to SEQ ID NO:2, or 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% sequence identity to SEQ ID NO:4, or 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% sequence identity to SEQ ID NO:6, or 92, 93, 94, 95, 96, 97, 98 or 99% sequence identity to SEQ ID NO:8 or 60, 65, 70, 75, 80, 85, 90 or 95% sequence identity to SEQ ID NO:10, wherein the envelope protein maintains the cellular tropism and functionality of a chimeric VSV virion comprising as envelope protein a GP comprising an amino acid sequence as shown in any one of SEQ ID NOs:2, 4, 6, 8 or 10.


The invention also provides a virus producing cell, characterized in that the cell produces a chimeric VSV virion of the invention. The cell may be of any origin and may be present as isolated cell or as a cell comprised in a cell population. It is preferred that the cell producing a pseudotyped VSV virion of the invention is a mammalian cell. In a more preferred embodiment, the virus producing cell of the invention is characterized in that the mammalian cell is a multipotent adult progenitor cell (MAPC), a neural stem cell (NSC), a mesenchymal stem cell (MSC), a HeLa cell, any HEK293 cell, a Vero cell or a bone marrow derived tumor infiltrating cell (BM-TIC). Alternatively, the virus producing cell may be a human cell, monkey cell, mouse cell or hamster cell. The skilled person is aware of methods suitable for use in testing whether a given cell produces a virus and, thus, whether a particular cell falls within the scope of this invention. In this respect, the amount of virus produced by the cell of the invention is not particularly limited. Preferred viral titers are ≥1×107 TCID50/ml or ≥1×108 genome copies/ml in the crude supernatants of the given cell culture after infection without further downstream processing.


In a particular embodiment, the virus producing cell of the invention is characterized in that the cell comprises one or more expression cassettes for the expression of at least one of the genes selected from the group consisting of genes n, l, p and m coding for proteins N, L, P and M of the VSV and a gene gp coding for Dandenong-GP or Mopeia-GP glycoprotein.


The virus producing cells of the invention may be used, for example, in gene therapy. For such purposes, a virus producing cell is provided, which is characterized in that the cell comprises a gene transfer vector for the packaging into a VSV virion pseudotyped with GP of Dandenong or Mopeia virus, wherein the gene transfer vector comprises a transgene. In this respect, the transgene may be any gene addressing a particular need of the host.


Transgenes within the meaning of the present invention may be transferred into cells using the means provided herein. Thus, in one embodiment, the invention relates to a method for transfer of a transgene into a cell in vitro or in vivo, characterized in that the cell is transduced with a chimeric virion of the invention, wherein the virion comprises a transgene. Transgenes may also be transferred using the virus producing cells of the invention. Thus, in a further embodiment, a method for transfer of a transgene into a cell in vitro, characterized in that the cell is contacted with a virus producing cell of the invention is provided. It is preferred that the cell into which the transgene is transferred is a tumor cell.


In a further embodiment, the invention relates to a pharmaceutical composition comprising any of the means provided herein. The pharmaceutical composition may thus be characterized in that the composition comprises the VSV chimeric vector of the invention, the VSV chimeric vector system of the invention, the chimeric VSV virion of the invention, or the virus producing cell of the invention.


In a further embodiment, the means provided herein, in particular the VSV chimeric vector of the invention, the VSV chimeric vector system of the invention, the chimeric VSV virion of the invention, the virus producing cell of the invention, or the pharmaceutical composition of the invention, are provided for use as medicament.


The invention further relates to the VSV chimeric vector of the invention, the VSV chimeric vector system of the invention, the chimeric VSV virion of the invention, the virus producing cell of the invention, or the pharmaceutical composition of the invention for use in treating cancer. In a preferred embodiment, the cancer is a solid cancer. In a more preferred embodiment, the solid cancer may be brain cancer, colorectal cancer, oropharyngeal squamous cell carcinoma, gastric cancer, gastroesophageal junction adenocarcinoma, esophageal carcinoma, hepatocellular carcinoma, pancreatic adenocarcinoma, cholangiocarcinoma, bladder urothelial carcinoma, metastatic melanoma, prostate carcinoma, breast carcinoma, glioblastoma, non-small cell lung cancer, brain tumor or small cell lung cancer.


In a further embodiment of the invention, the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition of the invention is provided, wherein the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition is combined with a PD-1 or PD-L1 antagonist. Such combination may occur prior to administration, i.e. the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition of the invention may be combined with the PD-1 or PD-L1 antagonist prior to administration to a patient in need thereof to form a combination formulation. Alternatively, the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition of the invention may be administered separately to form a combination treatment. Thus, the invention also relates to a pharmaceutical composition comprising the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition of the invention and a PD-1 or PD-L1 antagonist for uses provided herein. Moreover, the invention relates to a treatment method comprising the administration of the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition of the invention and administration of a PD-1 or PD-L1 antagonist.


Within the present invention, the skilled person, in particular a physician, may chose the appropriate route of administration for the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition of the invention. In a particular embodiment, the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition of the invention are provided, wherein the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition is to be administered intratumorally or intravenously. In a another embodiment, the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition for use according to the invention are provided, wherein the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition is to be administered intratumorally and subsequently intravenously. That is, the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition of the invention may be suitably formulated for intravenous or intratumoral administration.


The invention also provides methods for treating a subject, in particular a human subject, wherein the subject has cancer, in particular a solid tumor, the method comprising administering the VSV chimeric vector of the invention, the VSV chimeric vector system of the invention, the chimeric VSV virion of the invention, the virus producing cell of the invention, or the pharmaceutical composition of the invention in a therapeutically effective amount. In a particular embodiment, the solid cancer is brain cancer, colorectal cancer, oropharyngeal squamous cell carcinoma, gastric cancer, gastroesophageal junction adenocarcinoma, esophageal carcinoma, hepatocellular carcinoma, pancreatic adenocarcinoma, cholangiocarcinoma, bladder urothelial carcinoma, metastatic melanoma, prostate carcinoma, breast carcinoma, glioblastoma, non-small cell lung cancer, brain tumor or small cell lung cancer.


The invention furthermore relates to kits comprising the means provided herein.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 is a table (Table 1): Comparison of Old World Arenavirus (A) and New World Arenavirus clade C (B) GPC sequences.


Sequences were obtained from NCBI. The numbers of N-linked glycosylation signals N×T or N×S were inferred from the translated nucleotide sequence and ranges from 6 (LCMV) to 10 (IPPV, OLIW) signals. Given amino acid positions correspond to the sequence position of the individual sequences starting at the methionine of the translation start. GPC sequences were compared to LCMV-GP WE HPI by pairwise MUSCLE alignments [17]. Relative Sequence Identity (% Seq Id.), absolute identity (n) and sequence similarity is shown. aa: amino acid, nt: nucleotide.



FIG. 2: Multiple sequence alignment of arenavirus glycoproteins.


Amino acid sequences of LCMV-GP WE HPI, DANVD GP, IPPYV GP, MOPV GP, OLIW GP and LATV GP were aligned using MUSCLE [17]. Black arrows indicate the cleavage sites of the signal peptidase and the cellular subtilisin kexin isozyme 1 (SKI-1)/site 1 protease (S1P), that cleave the precursor glycoprotein GPC into SSP, GP1 and GP2. Predicted N-linked glycosylation signal sequences N×T or N×S within the GP1 peptides of the arenavirus GP were boxed by dashed lines.



FIG. 3 is a table (Table 2): Nucleotide and amino acid sequence identity in the GPC region among the selected Old World and New World clade C arenaviruses.



FIG. 4: GFP expression in BHK21CI.13 cells infected with trans-complemented VSV*MQΔG virus.


Non-cytopathic VSV*MQΔG virus, carrying the mutations M33A, M51R, V221F and S226R within the VSV matrix protein and with replacement of the entire coding sequence for the G protein by the eGFP sequence [18], was trans-complemented in BHK21CI.13 cells. 24 h before infection with VSV*MQΔG at a multiplicity of infection (MOI) of 3, BHK2101.13 were transiently transfected with expression plasmids pCAG-DANDV-GP, pCAG-IPPYV-GP, pCAG-LATV-GP, pCAG-MOPV-GP or pCAG-OLIW-GP to express the respective arenavirus GPs in the cells. For control, cells were mock transfected. Supernatants containing trans-complemented viruses were harvested at 24 h post infection and passaged undiluted (neat) or in serial ten-fold dilutions in a range of 1:10 to 1:1000 either on BHK21CI.13 cells (A) or on BHK-556 cells (B). In BHK-566 cells which stably expresses the LCMV-GP, VSV □G GFP viruses trans-complemented with DANDV-GP, LATV-GP, MOPV-GP or OLIW-GP were able to spread within the cell culture even at high dilutions, leading to a ubiquitous GFP expression after 48 h post infection. IPPY-GP did not trans-complement VSV*MQΔG virus, hence BHK-566 cells did not express GFP at levels higher than the mock control. GFP fluorescence was imaged at 48 h post infection with identical exposure times on a Leica DM2500 fluorescent microscope.



FIG. 5 is a table (Table 3): Cytopathic effects of VSV*McpΔG trans-complemented virus.


VSV*McpΔG virus [18] was trans-complemented in BHK21CI.13 cells. 24 h before infection with VSV*McpΔG at a M01=3, BHK21CI.13 were transiently transfected with expression plasmids pCAG-DANDV-GP, pCAG-IPPYV-GP, pCAG-LATV-GP, pCAG-MOPV-GP or pCAG-OLIW-GP. For control, cells were mock transfected. Supernatants containing trans-complemented viruses were harvested after 24 hpi and passaged undiluted (neat) or in serial ten-fold dilutions in a range of 1:10 to 1:1000 either on BHK21CI.13 cells (A) or on BHK-556 cells (B). In BHK-566 cells which stably expresses the LCMV-GP, VSV*McpΔG viruses trans-complemented with DANDV-GP, LATV-GP, MOPV-GP or OLIVV-GP were able to spread within the cell culture even at high dilutions. Cytopathic effect (CPE) was monitored at 48 h post infection under bright-field microscope. Classification of CPE ranges from strong CPE (++++) to no CPE visible (−).



FIG. 6: VSV-G(x) DANDV and MOPV replication kinetics.


Vero cells were seeded for infection in T75 cell culture flasks at a density of 4×104 cells/cm2, and infected 24 h later with the indicated MOI (Multiplicity of Infection, 0.05 or 0.0005) of VSV-GP as control and the two variants VSV-G(x) DANDV and VSV-G(x) MOPV. Samples were taken after 24, 30 and 42 h and the infectious titer was determined by TCID50 (A) and the genomic titer (B) was determined by qPCR.



FIG. 7: Type I interferon (IFN) limited VSV-G(x) replication in human tumor cell lines.


Human Calu6 (A) and 22Rv1 (B) cells were pre-incubated with indicated amounts of IFN for 16 hours. Subsequently, cells were infected in quadruplicates with VSV-GP, VSV-G(x) DANDV, VSV-G(x) MOPV or VSV-G(x) OLIVV at an MOI of 0.1, 1, or 10, or left uninfected as a negative control. Three days after infection, cells were analyzed for viability using an MTT assay. The graph shows mean±SEM, the viability of non-infected cells that were not pre-treated with IFN was normalized to 100%.



FIG. 8: Type I interferon (IFN) limited VSV-G(x) replication in murine tumor cell lines.


Murine SCCVII (A), Ct26CI.25 (B) or LLC1 (C) cells were pre-incubated with indicated amounts of IFN for 16 hours. Subsequently, cells were infected in quadruplicates with VSV-GP, VSV-G(x) DANDV, VSV-G(x) MOPV or VSV-G(x)


OLIVV at an MOI of 0.1, 1, or 10, or left uninfected as a negative control. Three days after infection, cells were analyzed for viability using an MTT assay. The graph shows mean±SEM, the viability of non-infected cells that were not pre-treated with IFN was normalized to 100%.



FIG. 9: Schedule of rabbit treatments and monitoring.


The schedule shows the design of one treatment out of three. All cycles of treatment were identical and started at D0, D14 and D28 with the i.v. injection of VSV-GP or VSV-G(x) viruses. Time points at which rabbits were handled are indicated by arrows. nAb, neutralizing antibody; BW, body weight; BT, body temperature; WBC, whole blood count.



FIG. 10: Neutralizing antibody induction in rabbits immunized with LCMV-GP, DANDV-GP and MOPV-GP recombinant VSV vectors.


Sera collected at day −4, at day 10 after each virus treatment (prime, 1st boost, 2nd boost) and at the end of the experiment were analysed for neutralizing antibodies against the autologous arenavirus glycoprotein. Sera were tested for their neutralizing capacities using VSV ΔG SEAP pseudo-typed with either LCMV-GP WE HPI, DANDV-GP or MOPV-GP described in detail in the method section. (A) The infection rate [in %], normalized to the no-serum control, was plotted versus the serum dilutions ranging from 1:10 to 1:31.250. After the prime immunization (open squares), none of the rabbits show nAbs directed against the arenavirus GPs of LCMV, DANDV or MOPV. After the first boost sera of rabbits immunized with VSV-G(x) DANDV and MOPV show only partial neutralization of the autologous VSV ΔG SEAP virus when compared to sera of VSV-GP immunized rabbits, hence calculation of the corresponding EC50 values was not applicable (n.a.). EC50 values were calculated after non-linear curve fit (B). Neutralization of VSV-ΔG SEAP GP by the LCMV-GP neutralizing antibody KL25 was used as inter assay control (C).



FIG. 11: Efficacy of intratumoral treatment with different arenavirus glycoprotein GP recombinant VSV vectors in a lung cancer xenograft mouse model.


5×106 Calu6 lung cancer cells were injected subcutaneously into the right flank of 8 week old NMRI nude mice. Treatment started, when the mean size of tumors reached a volume between 0.05 and 0.07 cm3. Mice were treated three times 4 days apart either with PBS (n=8) mice intratumorally (A), or with 1×107 TCID50 VSV-GP (B), VSV-G(x) DANDV or VSV-G(x) MOPV (D) Animals were monitored for tumor growth every 2-3 days after start of treatment, and sacrificed when tumor volume reached 0.8 cm3 or tumors ulcerated. Kaplan-Meier survival curve (E). Dotted lines indicate time points of virus injection.



FIG. 12 is a table (Table 4): Scores for the evaluation of virus induced neurotoxicity.


Neurotoxicity scoring of infected mice is based on different categories such as general appearance, clinical observations, body condition provoked behaviour motility and respiration. The scores in the different categories range from 0-3. The cumulative tox score shown in FIG. 12 is calculated by addition of the scores of each category.



FIG. 13: Neurotoxicity in Swiss CD1 mice.


Swiss CD-1 mice received a single intracranial injection of 3 μl containing 1×106 TCID50 via stereotactic injection into the right striatum. PBS was administered i.c. in the control group. Animals were monitored daily for signs of neurotoxicity and general well-being according to FIG. 12 for 42 days. (A) Mouse survival of PBS (diamonds), VSV-G DsRed (squares), VSV-GP (dots), VSV-GP(x) Dandenong (triangles) and VSV-GP(x) Mopeia (reversed triangles) experimental groups were plotted as Kaplan-Meier curves. Kaplan-Meier analysis indicates that none of the tested virus variants show neurotoxicity in mice. (B) Cumulative tox score, calculated according to FIG. 12, indicates that there is no neurotoxicity observed within in the groups. Only the VSV-G DsRed control group, which contains the wildtype VSV glycoprotein at the virion surface developed neurological signs leading to euthanasia within the first week after i.c. infection.





DETAILED DESCRIPTION

As described above, the invention generally relates to the provision of chimeric VSV vectors, wherein the VSV vectors lack a functional gene coding for the envelope protein GP of VSV and, instead, comprise a gene coding for a glycoprotein GP of an arenavirus, in particular the Dandenong virus or Mopeia (MOPV) virus. Alternatively, the vectors may comprise the GP of the Ippy virus, Latino virus or Olivero virus. As the skilled person is aware, the glycoprotein GP is an envelope protein present on the surface of virions, which is responsible for binding between the virion and a cell of a host organism. Thus, the envelope protein determines the tropism of the virion. By altering the tropism of VSV, chimeric VSV vectors may be prepared that are suitable for use in medicine. Such chimeric vectors/virions have been provided in the prior art, e.g. in WO 2010/040526. However, there is a need for further improved chimeric vectors that provide even more efficient treatment options and an even better suitability for medical use. This need is fulfilled by the present invention.


Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs and by reference to published texts, which provide one skilled in the art with a general guide to many of the terms used in the present application. The following definitions are provided for clarity only and are not intended to limit the claimed invention.


The terms “a” or “an” refers to one or more, for example, “a gene” is understood to represent one or more such genes. As such, the terms “a” (or “an”), “one or more,” and “at least one” are used interchangeably herein. As used herein, the term “about” means a variability of 10% from the reference given, unless otherwise specified.


With regard to the following description, it is intended that each of the compositions herein described, is useful, in another embodiment, in the methods of the invention. In addition, it is also intended that each of the compositions herein described as useful in the methods, is, in another embodiment, itself an embodiment of the invention. While various embodiments in the specification are presented using “comprising” language, under other circumstances, a related, embodiment is also intended to be interpreted and described using “consisting of or “consisting essentially of” language.


In this respect, the vectors of the invention are based on the virus of the vesicular stomatitis and have at least the below general advantages over retroviral vectors:

    • (i) VSV vectors are oncolytic and have a particularly high oncolytic activity compared to other oncolytic viral vectors.
    • (ii) VSV vectors replicate preferentially in tumor cells and have a particularly high replication capability compared to other oncolytic viral vectors.
    • (iii) VSV vectors infect actively dividing cells as well as resting cells.
    • (iv) VSV vectors induce a strong innate humoral and cellular immune response.
    • (v) VSV vectors replicate purely cytoplasmatically, i.e., as RNA viruses they cannot integrate into the host cell genome or recombine into replication-competent viruses.
    • (vi) VSV vectors are easy to package.
    • (vii) The VSV glycoprotein is interchangeable with a foreign envelope protein. Examples for glycoproteins which were previously incorporated in the VSV envelope are: HIVgp160 [19], HCVE1/E2 [20], SARS S [21], Lassa GP [22] or the GP of LCMV.


However, the vectors of the invention have additional advantages over the prior art and, in particular, VSV pseudotyped with GP of LCMV.


Specifically, the vectors of the invention surprisingly show a reduced induction of neutralizing antibodies as compared to a VSV vector pseudotyped with GP of LCMV under identical conditions. The skilled person is aware of assays that may be employed in order to determine induction of neutralizing antibodies.


By using the assay as described in Example 5, the inventors surprisingly found that induction of neutralizing antibodies is reduced. Such an effect could not have been reasonably expected by the skilled person. Moreover, the significance of this effect proves that the vectors and further means provided herein provide clear advantages. In this respect, it is preferred that induction of neutralizing antibodies by the vectors/virions provided herein is reduced as compared to a VSV vector pseudotyped with GP of LCMV.


The vectors/virions of the invention have not only proven to reduce neutralizing antibodies, but additionally show increased killing of tumor cells as compared to a VSV vector pseudotyped with GP of LCMV under identical conditions. The skilled person is aware of assays that may be employed to determine/quantify killing of tumor cells. It is preferred that killing of tumor cells by the vectors/virions of the invention is increased as compared to a VSV vector pseudotyped with GP of LCMV.


In order to achieve the above-described surprising effects, the GP of VSV is non-functional while GP of Dandenong virus or Mopeia (MOPV) virus is incorporated or the GP of VSV is replaced by the GP of Dandenong virus or Mopeia (MOPV) virus. Alternatively, the GP of VSV is non-functional while the GP of Ippy virus, Latino virus or Olivero virus is incorporated or the GP of VSV is replaced by the GP of Ippy virus, Latino virus or Olivero virus. There are various possibilities how a VSV vector can be pseudotyped with any of the above GPs. Thus, the precise nucleic acid sequence of the vectors of the invention may vary as long as the GP of VSV is essentially absent from the surface of virions while the GP of any of the viruses above is expressed on the surface of the virions. Within the present invention, exemplary preferred vectors comprise a nucleic acid sequence comprising SEQ ID NOs: 1 or 3, 5, 7 or 9 or a nucleic acid sequence comprising a nucleic acid sequence at least 70, 75, 80, 85, 90, 95, 96, 97, 98, 99 or 100% identical to any one of SEQ ID NOs: 1, 3, 5, 7 or 9. In this respect, SEQ ID NO:1 corresponds to a preferred nucleic acid sequence coding for the GP of the Dandenong virus, SEQ ID NO:3 corresponds to a preferred nucleic acid sequence coding for the GP of the Mopeia virus, SEQ ID NO:5 corresponds to a preferred nucleic acid sequence coding for the GP of the Ippy virus, SEQ ID NO:7 corresponds to a preferred nucleic acid sequence coding for the GP of the Latino virus and SEQ ID NO:9 corresponds to a preferred nucleic acid sequence coding for the GP of the Olivero virus.


“Percent (%) nucleic acid sequence identity” with respect to a reference nucleic acid sequence is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in the reference sequence. Methods to determine the percentage of identical nucleotides are known to the person skilled in the art. Within the present invention, it is preferred to use computer software such as BLAST. Parameters can readily be determined by the skilled person.


While the nucleic acid sequences coding for any of the GP genes used for pseudotyping VSV may vary, it is important that the functionality conferred by the GP encoded by the preferred gene sequence provided above is maintained. The functionality parameters that are maintained are, preferably, the induction of neutralizing antibodies, the killing of tumor cells and/or the tropism. In this respect, the above provided assays can be employed. Moreover, the skilled person is aware of assays that may be employed in order to determine tropism of a viral particle/virion.


As the skilled person will appreciate, a nucleic acid sequence may be varied with or without changing the primary sequence of the encoded polypeptide. It is preferred within the present invention that the polypeptide sequences encoded by the genes used for pseudotyping remain unchanged with respect to the GP proteins encoded by SEQ ID NOs: 1, 3, 5, 7 or 9, respectively. That is, it is preferred that the GP protein comprised in the virions of the invention corresponds to the GP protein of the Dandenong or Mopeia virus. More particularly, it is preferred that the virions of the invention comprise GP proteins comprising any one of the amino acid sequences shown by SEQ ID NOs: 2, 4, 6, 8 or 10. However, the skilled person is aware that the amino acid sequence of a polypeptide may be altered without affecting its functionality. Thus, polypeptides comprising alternative sequences are also encompassed by the present invention as long as the functionality of any one of the above amino acid sequences is essentially maintained.


Thus, in certain embodiments, virions comprising amino acid sequence variants of the GP proteins provided herein are contemplated. Amino acid sequence variants may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the respective GP, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the GP. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., induction of neutralizing antibodies, killing of tumor cells and/or tropism.


In certain embodiments, variants having one or more amino acid substitutions are provided. Conservative substitutions are shown in Table 5 under the heading of “preferred substitutions.” More substantial changes are provided in Table 5 under the heading of “exemplary substitutions,” and as further described below in reference to amino acid side chain classes. Amino acid substitutions may be introduced into a GP of interest and the products/virions screened for a desired activity.


Amino acids may be grouped according to common side-chain properties:

  • (1) hydrophobic: Norleucine, Met, Ala, Val, Leu, Ile;
  • (2) neutral hydrophilic: Cys, Ser, Thr, Asn, Gln;
  • (3) acidic: Asp, Glu;
  • (4) basic: His, Lys, Arg;
  • (5) residues that influence chain orientation: Gly, Pro;
  • (6) aromatic: Trp, Tyr, Phe.









TABLE 5







Amino acid substitutions









Original




Residue
Exemplary Substitutions
Preferred Substitutions





Ala (A)
Val; Leu; Ile
Val


Arg (R)
Lys; Gln; Asn
Lys


Asn (N)
Gln; His; Asp, Lys; Arg
Gln


Asp (D)
Glu; Asn
Glu


Cys (C)
Ser; Ala
Ser


Gin (Q)
Asn; Glu
Asn


Glu (E)
Asp; Gln
Asp


Gly (G)
Ala
Ala


His (H)
Asn; Gln; Lys; Arg
Arg


Ile (I)
Leu; Val; Met; Ala; Phe; Norleucine
Leu


Leu (L)
Norleucine; Ile; Val; Met; Ala; Phe
Ile


Lys (K)
Arg; Gln; Asn
Arg


Met (M)
Leu; Phe; Ile
Leu


Phe (F)
Trp; Leu; Val; Ile; Ala; Tyr
Tyr


Pro (P)
Ala
Ala


Ser (S)
Thr
Thr


Thr (T)
Val; Ser
Ser


Trp (W)
Tyr; Phe
Tyr


Tyr (Y)
Trp; Phe; Thr; Ser
Phe


Val (V)
Ile; Leu; Met; Phe; Ala; Norleucine
Leu









Non-conservative substitutions will entail exchanging a member of one of these classes for another class. Within the scope of the present invention, the variant GP proteins comprised in the virions of the invention maintain the functionality conferred on the virions by the non-altered GP proteins of Dandenong virus, Mopeia virus, Ippy virus, Latino virus or Olivero virus, respectively. Such variant GP proteins may have 60, 65, 70, 75, 80, 85, 90 or 95% sequence identity to SEQ ID NO:2, or 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% sequence identity to SEQ ID NO:4, or 45, 50, 55, 60, 65, 70, 75, 80, 85, 90 or 95% sequence identity to SEQ ID NO:6, or 92, 93, 94, 95, 96, 97, 98 or 99% sequence identity to SEQ ID NO:8 or 60, 65, 70, 75, 80, 85, 90 or 95% sequence identity to SEQ ID NO:10, wherein the envelope protein maintains the cellular tropism and functionality of a chimeric VSV virion comprising as envelope protein a GP comprising an amino acid sequence as shown in any one of SEQ ID NOs:2, 4, 6, 8 or 10.


“Percent (%) amino acid acid sequence identity” with respect to a reference amino acid sequence is defined as the percentage of amino acids in a candidate sequence that are identical with the amino acids in the reference polypeptide sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.


In a further embodiment, the invention relates to variant GP sequences having the functionality of any one of the Dandenong or Mopeia GP proteins, or the Ippy, Latino or Olivero GP, wherein the variant GP protein comprises 20, 15, 10, 5 or preferably 4, 3, 2 or 1 alteration(s).


In addition to their inherent oncolytic properties, the VSV chimeric vectors or vector systems based thereon and as provided herein can further be improved by introducing at least one transgene. The skilled person is aware of techniques that can be employed to introduce such transgenes into the vectors of the invention as well as where to introduce them. One exemplary approach comprises the steps of restriction and ligation or PCR and Gibson assembly. Transgenes that may be employed within the present invention are not particularly limited as long as a particular need is addressed by delivery of the transgene. Non-limiting, yet preferred, examples include transgenes coding for a suicide protein, a cytokine/chemokine, an antibody or antibody fragment binding to immune related receptors, a protein that serves for the purpose of vaccination, a viral fusion protein, a marker protein or fusions thereof.


To increase the safety during the use of replicable viruses in therapeutic uses, a vector system is provided which ensures that replication, oncolysis and the production of VSV viruses takes place only in cells which are infected by at least two replication-deficient mutually complementing vectors.


The invention thus, in one embodiment, relates to a VSV chimeric vector system, characterized in that the system comprises at least two complementary replicating VSV vectors, wherein the system comprises genes n, l, p and m coding for proteins N, L, P and M of the VSV, a gene gp coding for Dandenong-GP or Mopeia-GP, or alternatively Ippy-GP, Latino-GP or Olivero-GP, and lacks a functional gene coding for G protein of the VSV, wherein each vector of the system lacks one of the genes n, l, p, m and gp, and wherein the lacking gene is present on any other vector of the system. Such complementary replicating (cr) VSV vectors can spread to a limited extent within the targeted cell, for example a tumor cell, which increases the efficiency of the gene transfer and the oncolysis. Thus, the vector system according to the invention allows for the preparation of oncolytic VSV chimeric vectors with limited reproduction capability for the gene transfer in targeted cells, in particular tumor cells. The gene gp coding for LCMV-GP as well as possible additional genes such as therapy genes and/or marker genes can be present on any vector of the system.


Different variants of the vector system according to the invention are possible. For example, the vector system can consist of two vectors or more than two vectors. When the vector system consist of two vectors, a first vector may comprise GP of the Dandenong virus or Mopeia virus, or alternatively GP of the Ippy virus, Latino virus or Olivero virus, instead of GP of the VSV and a deletion of the gene p coding for the P protein. Also a second vector may comprise no VSV-G but express the P protein of the VSV. Each vector may express nucleoprotein (N) and polymerase (L) of the VSV as well as a less cytopathogenic variant of the M protein (Mncp). The first vector may carry in addition the marker gene rfp, whereas the second vector may carry a transgene.


The invention also provides a vector system comprising two vectors, wherein one vector comprises the GP of the Dandenong virus or Mopeia virus, or alternatively the GP of Ippy virus, Latino virus or Olivero virus, as defined herein and a second vector comprises the GP of LCMV. The second vector comprising the GP of LCMV may be a vector as described in WO 2010/040526.


The invention furthermore relates to cells producing the chimeric virions of the invention. Virus producing cells in the meaning of the invention include classical packaging cells for the production of virions from non-replicable vectors as well as producer cells for the production of virions from vectors capable of reproduction. Packaging cells usually comprise one or more plasmids for the expression of essential genes which lack in the respective vector to be packaged and/or are necessary for the production of virions. Such cells may be mammalian cells, in particular human cells, monkey cells, mouse cells or hamster cells, more particular HEK293 cells, HeLa cells, or Vero cells. Such cells are known to the skilled person who can select appropriate cell lines suitable for the desired purpose.


In previous studies, packaging cells were used for transferring viral vectors; however, this involved mainly fibroblasts which do not migrate within the tumor (Short et al., 1990, Culver et al., 1992). In contrast, adult stem cells, in particular neuronal (NSC), multipotent adult progenitor cells (MAPC) and mesenchymal stem cells (MSC) have a high migratory potential. They remain confined to the tumor tissue, whereby a very efficient but also specific gene transfer into the tumor tissue is achieved. However, these stem cells have limited passage capacity in vitro.


A subpopulation of adult mesenchymal stem cells, so-called BM-TIC (bone marrow derived tumor infiltrating cells) infiltrate, after injection into experimentally induced gliomas, the entire tumor and, in addition, track individual tumor cells remote from the tumor mass [23]. BM-TIC are isolated from adult bone marrow, have a high expansion potential and can be used as migrating producer cells for MLV [24] and VSV vectors.


The subject matter of the invention is thus virus producing cells which produce VSV chimeric vectors of the invention. In particular, these are tumor-infiltrating producer cells which release the said vectors during their migration within the tumor. Preferred cells are adult stem cells, in particular neuronal (NSC) and mesenchymal stem cells (MSC). Particularly preferred cells are BM-TIC cells derived from MSC.


The virus producing cells of the invention and hence also the VSV chimeric vectors produced by said cells may comprise a gene coding for a mutated M protein. This vector variant is selectively oncolytic for tumor cells, whereas it is not toxic for healthy cells. M variants with amino acid exchange in the 37PSAP40 region of the M protein or with single (M51R) or multiple (V221F and S226R; M33A and M51A) mutations outside of the PSAP region of the M protein are preferred. An M protein with mutations M33A, M51R, V22F and S226R is particularly preferred. In order to ensure an efficient virus production in packaging cells, the M variant can be stably transfected with a viral interferon antagonist.


In one embodiment, the virus producing is characterized in that the cell comprises one or more expression cassettes for the expression of at least one of the genes selected from the group consisting of genes n, l, p and m coding for proteins N, L, P and M of the VSV and a gene gp coding for Dandenong-GP or Mopeia-GP glycoprotein, or alternatively Ippy-GP, Latino-GP or Olivero-GP. The cell may furthermore comprise a gene transfer vector for the packaging into a VSV virion pseudotyped with GP of Dandenong or Mopeia virus, or alternatively Ippy, Latino or Olivero virus, wherein the gene transfer vector comprises a transgene.


In addition, subject matter of the invention is an in vitro method for gene transfer, wherein a VSV chimeric vector according to the invention or a VSV chimeric vector system according to the invention comprising a transgene is introduced into a cell either directly or by means of virus producing cells (packaging cells) according to the invention. If a cr vector system with at least two vectors is used, at least two packaging cells are used, wherein each of the cells produces one of the (replication-incompetent) cr vectors. The production of VSV viruses takes place only in cells which are infected with all vectors of the cr vector system and hence comprise all essential viral genes.


In addition, the invention relates to the use of vectors and virus producing cells according to the invention as drugs in therapeutic methods. In particular, the vectors and virus producing cells according to the invention are used for the therapy of solid cancers. The therapeutic effect is caused by the oncolytic properties of the recombinant vectors and viruses as well as by the use of therapeutic genes, without being bound by theory.


Solid cancer can be brain cancer, colorectal cancer, oropharyngeal squamous cell carcinoma, gastric cancer, gastroesophageal junction adenocarcinoma, esophageal carcinoma, hepatocellular carcinoma, pancreatic adenocarcinoma, cholangiocarcinoma, bladder urothelial carcinoma, metastatic melanoma, prostate carcinoma, breast carcinoma, glioblastoma, non-small cell lung cancer, brain tumor or small cell lung cancer.


The subject matter of the invention is further a pharmaceutical composition which comprises the vector, the virion, or the virus producing cell of the invention and optionally additives such as a pharmaceutically acceptable carrier and auxiliary substances.


In order to increase the viral oncolysis and the transfer efficiency of the therapeutic genes, tumor-infiltrating virus producing cells which continuously release vectors may be formulated for direct implantation into the tumors. The means provided herein may thus be formulated for intratumoral administration or intravenous administration.


The term “pharmaceutical formulation” or “pharmaceutical composition” refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.


A “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.


As used herein, “treatment” (and grammatical variations thereof such as “treat” or “treating”) refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.


Pharmaceutical formulations of the vectors, virions or cells as described herein are prepared by mixing such vectors, virions or cells having the desired degree of purity with one or more optional pharmaceutically acceptable carriers [25], in the form of lyophilized formulations or aqueous solutions. Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Exemplary pharmaceutically acceptable carriers herein further include interstitial drug dispersion agents such as soluble neutral-active hyaluronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaluronidase glycoproteins, such as rHuPH20 (HYLENEX®, Baxter International, Inc.). Certain exemplary sHASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968. In one aspect, a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.


The formulation herein may also contain more than one active ingredient as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.


Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in [25]. Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody or immunoconjugate, which matrices are in the form of shaped articles, e.g. films, or microcapsules. The formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.


In one aspect, the vector, virion, cell or pharmaceutical composition according to the invention for use as a medicament is provided. In further aspects, the vector, virion, cell or pharmaceutical composition according to the invention for use in a method of treatment are provided. In certain embodiments, the vector, virion, cell or pharmaceutical composition according to the invention for use in treating cancer are provided. In certain embodiments, the invention provides the vector, virion, cell or pharmaceutical composition of the invention for use in a method of treating an individual having cancer, the method comprising administering to the individual an effective amount of the vector, virion, cell or pharmaceutical composition. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.


In a further aspect, the invention provides for the use of a vector, virion, cell or pharmaceutical composition of the invention in the manufacture or preparation of a medicament. In one embodiment, the medicament is for treatment of cancer, in particular a solid tumor. In a further embodiment, the medicament is for use in a method of treating cancer, in particular a solid cancer, the method comprising administering to an individual having cancer, in particular a solid cancer, an effective amount of the medicament. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g. a therapeutic agent that is an antagonist of PD-1 or PD-L1.


In a further aspect, the invention provides a method for treating cancer, in particular a solid cancer. In one embodiment, the method comprises administering to an individual having cancer an effective amount of the vector, virion, cell or pharmaceutical composition of the invention. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, as described below.


An “individual” according to any of the above embodiments may be a human. However, included is any mammal in need of these methods of treatment or prophylaxis, including particularly humans. Other mammals in need of such treatment or prophylaxis include dogs, cats, or other domesticated animals, horses, livestock, laboratory animals, including non-human primates, etc. The subject may be male or female. In one embodiment, the subject has, or is at risk of developing, cancer and more particularly, a solid tumor.


As described above, the invention provides pharmaceutical compositions comprising any of the vector, virion or cell provided herein, e.g., for use in any of the above therapeutic methods. In one embodiment, a pharmaceutical formulation comprises any of the vector, virion, or cell provided herein and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical formulation comprises any of the vector, virion, or cell provided herein and at least one additional therapeutic agent, e.g., as described below.


The vector, virion, cell or pharmaceutical composition of the invention can be used either alone or in combination with other agents in a therapy. For instance, the vector, virion, cell or pharmaceutical composition of the invention may be co-administered with at least one additional therapeutic agent.


“Therapeutic agents” within the meaning of the invention are molecules including, without limitation, polypeptides, peptides, glycoproteins, nucleic acids, synthetic and natural drugs, peptides, polyenes, macrocyles, glycosides, terpenes, terpenoids, aliphatic and aromatic compounds, and their derivatives. In a preferred embodiment, the therapeutic agent is an antagonist of PD-1 or PD-L1. In another preferred embodiment, the therapeutic agent is a checkpoint inhibitory antibody or any other immune therapeutic that stimulates the immune response of the individual. In yet another embodiment, the therapeutic agent is a chemotherapeutic agent. The chemotherapeutic agent may be substantially any agent which exhibits an oncolytic effect against tumor cells in an individual and which does not inhibit or diminish the oncolytic effect of the oncolytic virus of the invention. The agent may be any known or subsequently discovered chemotherapeutic agent. By way of example, known types chemotherapeutic agents include, for example, anthracyclines, alkylating agents, alkyl sulfonates, aziridines, ethylenimines, methylmelamines, nitrogen mustards, nitrosoureas, antibiotics, antimetabolites, folic acid analogs, purine analogs, pyrimidine analogs, enzymes, podophyllotoxins, platinum-containing agents, interferons, and interleukins.


Suitable therapeutic agents include, without limitation, those presented in Goodman and Oilman's The Pharmacological Basis of Therapeutics (e.g., 9th Ed.) or The Merck Index (e.g., 12th Ed.). Genera of therapeutic agents include, without limitation, drugs that influence inflammatory responses, drugs that affect the composition of body fluids, drugs affecting electrolyte metabolism, chemotherapeutic agents (e.g., for hyperproliferative diseases, particularly cancer, for parasitic infections, and for microbial diseases), antineoplastic agents, drugs affecting the blood and blood-forming organs, hormones and hormone antagonists, vitamins and nutrients, vaccines, oligonucleotides and gene therapies. It will be understood that compositions comprising combinations, e.g. mixtures or blends of two or more active agents, such as two drugs, are also encompassed by the invention.


Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the vector, virion, cell or pharmaceutical composition of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant. The vector, virion, cell or pharmaceutical composition of the invention can also be used in combination with radiation therapy.


The vector, virion, cell or pharmaceutical composition of the invention (and any additional therapeutic agent) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional, intrauterine or intravesical administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Also envisaged is direct intratumoral administration. Preferred is intravenous or intratumoral administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time-points, bolus administration, and pulse infusion are contemplated herein.


Intratumoral injection, or injection directly into the tumor vasculature is specifically contemplated for discrete, solid, accessible tumors. Local, regional or systemic administration also may be appropriate. For example, for tumors of >4 cm, the volume to be administered may be about 4-10 mL (suitably 10 mL), while for tumors of <4 cm, a volume of about 1-3 mL may be used (suitably 3 ml). In some embodiments, the volume of agent administered can be up to 25% or up to 33% of the tumor volume. Multiple injections delivered as single dose comprise about 0.1 to about 0.5 mL volumes. The viral particles may advantageously be contacted by administering multiple injections to the tumor, spaced at approximately 1 cm intervals. In the case of surgical intervention, the present compositions may be used preoperatively, to render an inoperable tumor subject to resection. Continuous administration also may be applied where appropriate, for example, by implanting a catheter into a tumor or into tumor vasculature. Such continuous perfusion may take place for a period from about 1-2 hours, to about 2-6 hours, to about 6-12 hours, to about 12-24 hours, to about 1-2 days, to about 1-2 wk or longer following the initiation of treatment. Generally, the dose of the therapeutic composition via continuous perfusion will be equivalent to that given by a single or multiple injections, adjusted over a period of time during which the perfusion occurs. It is further contemplated that limb perfusion may be used to administer therapeutic compositions, particularly in the treatment of melanomas and sarcomas.


The vector, virion, cell or pharmaceutical composition of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The vector, virion, cell or pharmaceutical composition need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of the vector, virion, cell or pharmaceutical composition present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.


For the prevention or treatment of disease, the appropriate dosage of the vector, virion, cell or pharmaceutical composition of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the type of the vector, virion, cell or pharmaceutical composition, the severity and course of the disease, whether the vector, virion, cell or pharmaceutical composition is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the vector, virion, cell or pharmaceutical composition, and the discretion of the attending physician. The vector, virion, cell or pharmaceutical composition is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. 0.1 mg/kg-10 mg/kg) of the vector, virion, cell or pharmaceutical composition can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the vector, virion, cell or pharmaceutical composition would be in the range from about 0.05 mg/kg to about 10 mg/kg. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the vector, virion, cell or pharmaceutical composition). An initial higher loading dose, followed by one or more lower doses may be administered. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.


Alternatively, the vector, virion, cell or pharmaceutical composition of the invention may be delivered in a volume of from about 50 μL to about 10 mL including all numbers within the range, depending on the size of the area to be treated, the viral titer used, the route of administration, and the desired effect of the method. In one embodiment, the volume is about 50 μL. In another embodiment, the volume is about 70 μL. In another embodiment, the volume is about 100 μl another embodiment, the volume is about 125 μL. In another embodiment, the volume is about 150 μL. In another embodiment, the volume is about 175 μL. In yet another embodiment, the volume is about 200 μL. In another embodiment, the volume is about 250 μl. In another embodiment, the volume is about 300 μL. In another embodiment, the volume is about 450 μL. In another embodiment, the volume is about 500 μL. In another embodiment, the volume is about 600 μL. In another embodiment, the volume is about 750 μL. In another embodiment, the volume is about 850 μL. In another embodiment, the volume is about 1000 μL. In another embodiment, the volume is about 2000 μL. In another embodiment, the volume is about 3000 μL. In another embodiment, the volume is about 4000 μL. In another embodiment, the volume is about 5000 μL. In another embodiment, the volume is about 6000 μL. In another embodiment, the volume is about 7000 μL. In another embodiment, the volume is about 8000 μL. In another embodiment, the volume is about 9000 μL. In another embodiment, the volume is about 10000 μL. An effective concentration of a virion carrying a nucleic acid sequence encoding the desired transgene under the control of the cell-specific promoter sequence desirably ranges between about 108 and 1013 vector genomes per milliliter (vg/mL). The infectious units may be measured as described in S. K. McLaughlin et al [26]. Preferably, the concentration is from about 1.5×109 vg/mL to about 1.5×1012 vg/mL, and more preferably from about 1.5×109 vg/mL to about 1.5×1011 vg/mL. In one embodiment, the effective concentration is about 1.5×1010 vg/mL. In another embodiment, the effective concentration is about 1.5×1011 vg/mL. In another embodiment, the effective concentration is about 2.8×1011 vg/mL. In yet another embodiment, the effective concentration is about 1.5×1012 vg/mL. In another embodiment, the effective concentration is about 1.5×1013 vg/mL. It is desirable that the lowest effective concentration be utilized in order to reduce the risk of undesirable effects. Still other dosages in these ranges may be selected by the attending physician, taking into account the physical state of the subject, preferably human, being treated, the age of the subject, the particular type of cancer and the degree to which the cancer, if progressive, has developed.


It is understood that any of the above formulations or therapeutic methods may be carried out using any one of the vector, virion, cell or pharmaceutical composition of the invention.


In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the disorder and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is the vector, virion, cell or pharmaceutical composition of the invention. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises the vector, virion, cell or pharmaceutical composition of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition, in particular cancer. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically-acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution or dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.


EXAMPLES
Example 1: Arenavirus GP Sequence Alignments

Arena virus RNA sequences of the S segment were retrieved from the NCBI nucleotide library in gene bank flat file format as follows: LCMV GP WE HPI (accession: AJ297484), MOPV GP (accession: JN561684), DANDV GP (accession: EU136038), IPPYV GP (accession: DQ328877), OLIVV GP (accession: U34248) and LATV (accession: AF485259). GPC glycoprotein sequences were obtained by translation of the GPC open reading frame coded by the S segment. Old World and New World clade C arenavirus GPC sequences were compared on nucleotide and protein level using the Geneious software package version 11.0.5 (Biomatters Ltd.).


N-linked glycosylation signals in the GP1 peptide of the glycoprotein GP were identified by the glycosylation signal sequence N×S or N×T. Numbering of the glycosylation signal sites corresponds to the parental sequence starting at the ATG methionine translation initiation codon (FIG. 1).


Absolute and relative sequence identities (% Seq Id) were calculated after multiple alignment of the nucleotide sequences or the corresponding GPC amino acid sequences (FIGS. 2 and 3). Sequences were aligned using MUSCLE [17] with a maximum number of eight iterations using a distance matrix by k-mer and pctid clustering. Subsequently, sequences were clustered using the UPMGA method. Sequence similarities of every arenavirus GPC protein compared to LCMV WE HPI were calculated from pairwise alignments using the blosum62 matrix with a threshold≥1.


Example 2: Arenavirus GPs can Complement for VSV-G

The GP glycoproteins of DANDV, MOPV as well as LATV and OLIVV were able to trans-complement VSV*MQΔG virus. The resulting VSV pseudotypes were able to infect BHK21CI.13 cells in a subsequent passage and showed increased GFP signals at different dilutions when compared to the mock control (FIG. 4A). The GFP signal that could be observed in BHK21CI.13 cells infected with supernatants of IPPYV GP-complemented VSV*MQΔG virus was comparable to the mock control and results from the input VSV*MQΔG virus. To confirm these results, BHK-566 cells which stably expresses the LCMV-GP were infected with the pseudotyped VSV*MQΔG viruses. Viruses trans-complemented with DANDV-GP, LATV-GP, MOPV-GP or OLIVV-GP were able to spread within the cell culture even at high dilutions, leading to a ubiquitous GFP expression after 48 h post infection (FIG. 4B). Cythopathic effects (CPE) of VSV*McpΔG viruses trans-complemented with the GPs of DANDV, MOPW, IPPYV OLIVV and LATV confirmed the results obtained with the corresponding VSV*MQΔG pseudotypes and are summarized in FIG. 5.


For trans-complementation, 0.8-1×106 BHK21CI.13 cells were seeded in six-well plates one day before transfection. On the next day, BHK21CI.13 were transfected with 2.5 μg pCAG-DANDV-GP, pCAG-IPPYV-GP, pCAG-LATV-GP, pCAG-MOPV-GP or pCAG-OLIW-GP using TransIT-LT1 transfection reagent (Mirus Bio LCC, Madison, Wis. USA) according to the manufacturer's recommendations. 24 h post transfection, transiently transfected BHK21CI.13 cells were infected with VSV*McpΔG or VSV*MQΔG [9], carrying the mutations M33A, M51R, V221F and S226R within the VSV matrix protein at an MOI of 3. One hour post infection, cells were washed twice with cGMEM (containing 10% FCS, 2% Glutamine and 1% Tryptose Phosphate Broth) and incubated for another 24 hours. Supernatants were harvested and cell debris was removed by centrifugation at 8000 rpm for 5 min in a bench-top centrifuge. Supernatants were then transferred in serial ten-fold dilutions to fresh BHK21CI.13 or BHK-566 cells, which stably expresses LCMV-GP WE HPI after lentiviral transduction, in 24-well plates. GFP expression from the vector or cytopathic effect was monitored after 24 h and 48 h on a Leica DM2500 fluorescent microscope. Pictures were taken using identical exposure times.


Example 3—Replication Kinetics

VSV-chimeric vectors that comprises the VSV vector backbone as described in WO 2010/040526 and the GPs of DANDV, MOPV, OLIVV and LATV were cloned. The GPs in the resulting chimeric VSV-G(x)-DANDV, -MOPV, -OLIVV and LATV vectors replace the GP of LCMV without any changes in the remaining VSV vector backbone. Although there were slightly differences in the replication kinetics of VSV-G(x)-DANDV and -MOPV (VSV-G(x)-LATV and -OLIVV were not tested), both viruses replicate to titers higher than 1×107 TCID50/ml (FIG. 6A) or 1×109 genome copies/ml (FIG. 6B) after 30 hpi. With regard to their replication potential, VSV-G(x)-DANDV and -MOPV might be particularly suitable for OV cancer therapy.


For comparison of replication kinetics, Vero cell monolayers in a T75 cell culture flasks were infected with VSV-GP, VSV-G(x) DANDV or VSV-G(x) MOPV at an MOI of 0.05 or 0.0005. 500 μl supernatant of infected cells was collected at the indicated time points. Supernatant was centrifuged at approx. 2000 rpm for 5 min in a bench-top centrifuge to remove cell debris and stored at −80° C. until further processing. Analysis of viral titers by TCID50 assay [27] and of viral genomes by qPCR [28] was performed as described elsewhere.


Example 4—Tumor Cell Killing of VSV-G(x) Variants (FIG. 7)

Cell lines. Human Calu6 lung carcinoma cells were obtained from Dr. Edith Lorenz, OncoTyrol (Department of Internal Medicine, Hematology and Oncology, ΔG Zwierzina, Innsbruck). Cells were grown in DMEM medium with 10% fetal bovine serum, 2 mM L-Glutamine and 1% Penicillin/Streptomycin (Pen/Strep). The cells were sub-cultured every 2-3 days when they reach 80% confluency using EDTA-Trypsin 0.05%. 22Rv1 cells derived from a human prostate cancer were kindly provided by Prof. Z. Culig (Department of Urology, Medical University of Innsbruck). Cells were sub-cultured twice a week in RPMI1640 containing 10% FCS, 2 mM Glutamine, 10 mM HEPES, 1 mM sodium-pyruvate and 1% Pen/Strep. Adherent murine squamous cell carcinoma cells (SCCVII) were obtained from Dr. Lukas Mach (Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna) Cells were grown in DMEM containing 10% FCS, 2 mM Glutamine, 0.1 mM non-essential amino-acids (NEAA), 1 mM sodium-pyruvate and 1% Pen/Strep. SCCVII cells were sub-cultured at a ratio of 1:10 three times a week using EDTA-Trypsin 0.05%. CT26CI.25 murine colon carcinoma cells derived from Balb/c that were stable transduced with a LacZ cassette disrupting the IFN-I antiviral response were obtained from ATCC (#CRL-2639). Cells were sub-cultured twice a week at a ratio of 1:10 in RPMI1640 containing 10% FCS, 2% Glutamine, 10 mM HEPES, 0.1 mM NEAA, 1 mM sodium-pyruvate 1% P/S and 400 μg/ml G418. LLC1 cells were established from a C57BL/6 mouse bearing a lung tumor after implantation of a primary Lewis Lung Carcinoma. The cells were obtained from ATCC (#CRL-1642) and cultured in DMEM containing 10% FCS, 4 mM Glutamine and 1% P/S. Confluent cell cultures were sub-cultured 1:6 to 1:10 every 3-4 days by resuspension of loosely attached cells.


VSV killing assay. Cells were seeded in 96-well plates and pre-incubated overnight with 10, 100, 500 and 1000 units of universal type-1 IFN (PBL, Piscataway, N.J., USA) in a volume of 100 μl/well. The following morning, cells were infected with VSV-GP, VSV-G(x) DANDV, VSV-G(x) MOPV or VSV-G(x) OLIW at an MOI of 0.1, 1, or 10 in a final volume of 120 μl/well. For each condition, quadruplicate samples were performed. As a positive, killing control cells were incubated with a final concentration of 6.67 mM of H2O2. Three days after infection, cells were analyzed for viability using an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide)-based in vitro cytotoxicity assay (Sigma-Aldrich, Saint Louis, Mich., USA), according to the manufacturer's recommendations. Plates were measured in a conventional microplate reader at 550 nm, and blank values of wells without cells were subtracted. Values were normalized to mock-infected cells that were not pre-treated with interferon (IFN), and represented as a percentage of viable cells.


Example 5: Neutralizing Antibodies after i.v. Treatment of NZW Rabbits

The study attempted to evaluate whether the VSV G(x) variants induce no neutralizing antibodies (nAb) or lower levels of nAbs compared to VSV-GP following 3 intravenous (i.v.) administrations in healthy tumor-free New Zealand White (NZW) rabbits. NZW rabbits were treated i.v. three times 14 days apart with 1×109 TCID50 of VSV-GP or different VSV-G(x) candidates. nAb induction as well as body weight and temperature, viremia, whole blood count and blood chemistry were monitored at regular intervals following treatment as depicted in FIG. 9. The primary objective of this study was to monitor nAb induction by measuring nAb levels 10 days after each administration. The nAb assay is based on previously published work by Kaku et al. [29] J. Virol Methods (2012) 179(1):226-32. The VSV ΔG SEAP virus used in the nAb assays expresses the secreted embryonic alkaline phosphatase (SEAP) instead of the envelope glycoprotein G. This virus was complemented by LCMV GP or the respective other arenavirus glycoproteins ectopically expressed on the cell surface of the producer cell. These trans-complemented viruses were than tested for neutralization by the collected rabbit (FIG. 10).


Safety parameters such as the health status of the animals, including body weight (BW) and body temperature (BT), white blood cell (WBC) counts, blood chemistry (optional) and viremia (TCID50 and N-specific qPCR in plasma/blood) were secondary objectives.


Rabbit handling and housing: Crl:KBL(NZW) rabbits were purchased from Charles River Laboratories, (France). At arrival in the animal facility, rabbits were approx. 7 weeks old. The animals were housed in groups of two individuals in rabbit cages (R-Suite Enriched Rabbit Housing, Techniplast). Individuals were identified by ear tattoos. Rabbit housing was controlled daily and the overall health status was monitored. Food supply ad libitum and daily change of drinking water was guaranteed for the time period of the experiment. The rabbits were acclimated a minimum of 4 days before the rabbits were adapted to handling. Virus treatment started after the rabbits were acclimated and handled for at least one week.


Viruses and treatment of NZW rabbits: Virus dilutions were done with PBS. The final concentration of VSV-GP or VSV-G(x) solutions was 1×109 TCID50 per ml. Virus dilutions were kept on ice until shortly before injection. Rabbits were fixed by wrapping the animal in a cotton towel. The injection site of the tattooed left ear was shaved properly and then cleaned using a sterile alcohol swab. 1×109TCID50 virus in 1 ml PBS were injected slowly into the left ear vein by a 29 gauge needle attached to a 1 ml syringe.


Rabbit monitoring: Body weight and body temperature were measured regularly according to the Schedule presented in FIG. 9. Temperature was measured rectally with a digital thermometer. Body weight was recorded using an Ultra MBSC balance.


Blood sampling and processing: Blood was collected from the ear artery using a 23G micro needle (Sarstedt, Germany). Before puncture of the artery, the ear was shaved properly and disinfected using an alcohol pad. A minimum of 2 ml blood was collected in a 15 ml tube containing 50 μl heparin (5000 IE/ml). 100 μl heparin blood was stored at −80° C. for analysis of viral titers by qPCR, the remaining blood was processed to heparin plasma (200 μl) for TCID50 determination or clinical chemistry (300 μl). At day −4 before the first treatment and at day 10 after each treatment cycle additional 2-3 ml blood was collected in a 15 ml Falcon for serum preparation. In addition, approx. 500 μl blood was collected in two pre-coated EDTA microvettes (Microvette® CB 300 K2E, Sarstedt, Germany). EDTA blood was used for whole blood count and a minimum of 100 μl was archived and used for qPCR. EDTA plasma was prepared from the second microvette and stored at −80° C. until use.


Viremia by TCID50 and VSV-N specific qPCR: Viremia was measured by TCID50 assay in blood plasma after 8 h and consecutive three days after virus application of each treatment cycle (as described by Urbiola et al. [27]). For VSV-N specific qPCR, 100 μl of EDTA and Heparin blood frozen and stored at −80° C. for qPCR was used. RNA was isolated using a commercial available RNA isolation kit for blood samples. Determination of viral titers by qPCR was performed as described e.g. by Jenks et al. [28].


White Blood Count: White blood counts using EDTA blood were measured using the ScilVet ABC blood counter. Approximately 10-20 μl of fresh EDTA-blood was needed for the assay. Measurements were performed according to the manufacturer.


Sacrifice: Rabbits were anesthetized by Ketamine/Xylazine according to FELASA and GV SOLAS recommendations. After anesthesia the ear right ear vein was catheterized and rabbits were terminal bleed by heart puncture. Blood without anticoagulants and heparin blood was collected. Rabbits were released from anesthesia with a lethal dose of pentobarbital through the vein catheter according to FELASA and GV SOLAS recommendations. After the rabbits had been sacrificed, spleens were removed and splenocytes were isolated by standard procedures for optional downstream analysis (e.g. VSV-N specific T cell responses). Blood was processed to serum or plasma and aliquots were stored at −80° C. until use.


Neutralizing antibody (nAb) assay: Pre-immune sera collected before the first treatment at D-4 and serum samples taken 10d after each treatment cycle were analyzed for the presence of nAb directed against VSV-ΔG SEAP GP virus or pseudo-typed virus with a different glycoprotein variant. SEAP activity in the supernatant of infected BHK21CI.13 cells was measured by the enzymatic conversion of p-nitro-phenylphosphate (pNPP) to p-nitrophenyl which has a yellow color in solution by the virus coded SEAP. The color reaction was read in a conventional micro plate reader at 405 nm and served as indirect measurement of the infection rate by the SEAP expressing virus. Comparing serum and no serum-treated control samples the nAb assay indicated whether a serum sample contained neutralizing antibodies or not. In the absence of nAbs, the virus was not neutralized and able to infected BHK21CI.13 cells resulting in the expression of SEAP. In contrast, if the virus was neutralized by antibodies in the sera, the virus was not able to infect the cells and hence SEAP was not expressed.


One day before virus neutralization, BHK21CI.13 cells were seeded in a 96-well cell culture plate at a density of 1×104 cells in 100 μl cGMEM per well using a multi-channel dispenser pipette. To guarantee low inter assay variation cells were counted using the Luna cell counter (Logos Biosystems). Any other cell counting device that is based on the photographic analysis of cell numbers (e.g. Tecan reader) could be used. Counting cells with a CASY counter (OLS OMNI Life Science) or Kovar Glasstic slides is not recommended. Cells were incubate over night at 37° C., 6% CO2, 95% humidity. Cell culture plates were kept in a moisture chamber to prevent evaporation of the wells at the plate borders.


The next day, serial 1:5 serum dilutions starting at a lowest dilution of 1:5 were prepared first. Briefly, 200 μl cGMEM per well were added to a 96-well plate using a multi-channel dispenser pipette. 50 μl KL25 monoclonal ΔLCMV GP antibody at a concentration of 100 μg/ml (inter assay control) or heat inactivated (1 h, 56° C.) serum sample were added to 200 μl GMEM into the wells of row C of the 96-well plate and mixed by pipetting up and down several times. Five-fold serial dilutions starting at row C down to row H of the 96 well plate were done using a multichannel pipette: 50 μl from the wells in row C were transferred to the wells of the next row and mixed by pipetting several times up and down. 50 μl from the wells of row D were then added to the wells of the next row etc. until finished at row H. 175 μl of the serum dilutions of each well were transferred into a clean 96-well plate using a multichannel pipette. Serum dilutions were kept on ice. Row A and B of the 96-well plate served as no-infection control (row A) or no-serum control (row B).


Dilutions of pseudo-typed VSV ΔG SEAP virus were prepared in cGMEM at a MOI of 0.1 or 1, corresponding to 1×103 or 1×104 infectious particles per well, respectively, when 1×104 BHK21CI.13 cells were seeded one day before. The amount of virus needed for each triplicate in the nAb assay was calculated according to the following formulas:







Volume





of





virus





stock





needed






in




[

in





µl

]


=



3.5
*

10
4






infectious





particles






(

MOI
=
1

)




titer





of





virus






stock




[

TCID






50
/
ml


]


)


*
1000

µl
*
number





of





wells






and






Volume





of






cGMEM




[

in





µl

]


=


(

175

µl
×
number





of





wells

)

-

Volume





of





virus





stock





needed






in




[

in





µl

]







175 μl virus solution were then added to each well of row B-H containing the 175 μl serial serum dilutions to make 350 μl serum/virus sample. Virus/serum samples were mix by pipetting up and down several times using a multichannel pipette. 175 μl cGMEM were added to the wells of row A (non-infection control). Serum/virus mixtures were incubated for 1 h on ice.


After incubation, 100 μl of the serum/virus sample was added in triplicate to BHK21CI.13 cells seeded in 96 well plates one day before. Cells should be at 80-90% confluency. BHK21CI.13 were incubated for additional 24 h at 37° C., 6% CO2 and 95% humidity.


On the next day, the nAb activity was measured as a function of SEAP activity. Briefly, the p-nitro-phenylphosphate (pNPP) substrate solution (SIGMA-FAST, Sigma Aldrich) was prepared according to the manufactures recommendations For each 96-well cell culture pNPP substrate solution was prepared by dissolving one buffer tablet and one pNPP substrate tablet in 21 ml sterile H2O. 40 μl cell culture supernatant from the cells infected one day before were transferred into a new 96 well plate using a multichannel pipette. Then 200 μl of the pNNP substrate solution was added to the cell culture supernatant using a multichannel dispenser pipette. Plates were incubated at least 1 h at room temperature in the dark. The OD of the wells was read in a conventional micro plate reader at 405 nm. If the OD405 of the no-serum control was lower than 1.0, incubation time was prolonged for an additional hour or up to 4 h, before OD measurements were repeated. Measured OD values were plotted versus serum dilutions. By non-linear curve fitting EC50 values were calculated using the GraphPad Prism 5 Software version 5.03 (GraphPad, San Diego, Calif. USA).


Example 6: Treatment of Calu6 Lung Tumor Xenograft Model (FIG. 11)

Recipient mice: Eight-week old female NMRI-Nude mice were purchased from Janvier (France). Animals were housed in individually ventilated cages in groups of 8 animals. For identification purpose, the mice were ear clipped. Before tumor engraftment the mice were acclimated for at least one week.


Tumor cells: Calu6 lung carcinoma cell line were obtained from Dr. Edith Lorenz, OncoTyrol (Department of Internal Medicine, Hematology and Oncology, ΔG Zwierzina, Innsbruck). Cells were grown in DMEM medium with 10% fetal bovine serum, 2 mM L-Glutamine and 1% Penicillin/Streptomycin. The cells were sub-cultured every 2-3 days when they reach 80% confluency using EDTA-Trypsin 0.05%. Cells were seeded at low cell numbers. 24 hrs before grafting into NRMI nude mice, cells were harvested, pooled and then passaged at a ratio 1:2 in T175 flasks.


On the day of engraftment, cells were detached using EDTA-Trypsin 0.05%, complete medium was added and cells were counted in a KOVA Glasstic slide (FisherScientific). To determine cell numbers, 10 μl cell suspension were mixed with 90 μl trypan blue solution by repeated pipetting in a 96-well plate. 20 μl of the counting mix was transferred to a KOVA Glasstic slide and cells in 3 squares (a, b, c) including the top and right gridline were counted omitting the blue labeled dead cells. The cell number per ml was determined by the following equation: ((count (a)+count (b)+count (c)))/3)×104×10=cells per ml. 5×106 cells per animal were transferred in a 50 ml Falcon tube and washed with PBS. The cells were resuspended in PBS to obtain a final concentration of 1×108 cells per ml. Subsequently, the cell suspension was divided in 1 ml aliquots and stored on ice for maximum 30 minutes before injection.


Engraftment of Calu6 cells into NMRI-Nude recipient mice: Calu6 cells were prepared as described above and resuspended by gently flicking the tube. Mice were anesthetized by isoflurane inhalation, the injection site was cleaned using a sterile alcohol swab. Using a 0.5 ml syringe with a 27G needle, 5×106 cells in 50 μl were administered subcutaneously (s.c.) in the right flank. Following injection of tumor cells, animals were checked bi-weekly for palpable lesions. After detecting a lesion, the animal was weighed and tumor size was measured. Tumors were measured using a caliper, values for length (cm) and width (cm) of the tumor were used to calculate the approximate tumor volume (cm3) using the following formula: Volume=Length×(Width)2×0.4. Length was defined regardless of orientation to the animal axes as being the longer dimension.


Treatment by recombinant VSV vectors: When the mean size of tumors reached a volume between 0.05 and 0.07 cm3, tumors were treated. 1 cage of 8 mice was allocated per group. Allocation of the cages was done to obtain similar mean and distribution of tumor size between the different groups. For the negative control group, 30 μl of PBS was injected i.t. For the other three treatment groups the final concentration of VSV-GP, VSV-GP(x) DANDV and VSV-GP(x) MOPV was 3.3×108 TCID50 per ml. Dilutions of virus were done with PBS. Before injection, mice were anesthetized by isoflurane inhalation and the injection site was cleaned using a sterile alcohol swab. With a 0.1 ml syringe with a 29G needle, 1×107TCID50 VSV-GP, VSV-GP(x) DANDV or VSV-GP(x) MOPV in 30 μl was injected i.t. per tumor and animal. Treatments were repeated accordingly two times four days apart.


Monitoring Tumor growth: After treatment, the animal were examined and tumor size was measured at least twice a week with a maximal interval of 4 days. Tumors were measured as described above. If tumor size of one animal was measured in between for ethical reasons (e.g. just before sacrifice), at least all animal of this group were measured. Body weight of mice was determined once a week. The following endpoint criteria require euthanasia: (i) tumor volume exceeds 0.8 cm3 (ii) mice show weight drop >20% or (iii) tumor ulcerates. Dates of sacrifice were used to calculate Kaplan-Mayer survival curves.


Example 7: Neurotoxicity of VSV-G(x)-DANDV and -MOPV

Wild type VSV infections can cause neurological symptoms when the virus gets access to the brain. These neurological complications include a severe encephalitis that can lead to death of the infected subject. The advantage of using a chimeric VSV-GP is that neuronal infection has been shown to be nearly completely absent rendering the VSV-backbone a safe oncolytic agent. The reason for the attenuated phenotype is thought to be due to an altered virus tropism facilitated via the viral envelope glycoprotein. Alterations or modifications of the GP glycoprotein might affect the tropism profile of VSV-GP therefore a neurotoxicity assessment of VSV-G(x) DANV and of VSV-G(x) MOPV was done. Both VSV-G(x) DANDV and VSV-G(x)MOPV did not show any signs of neurotoxicity after direct intracranial injection of Swiss CD1 mice (FIG. 13) as shown for VSV-LCMV GP. All control mice that received VSV-G DsRed died within the first week post infection.


Mice. 8 weeks old female Swiss CD-1 mice were purchased from Janvier (France). The animals were housed in individually ventilated cages in groups of 5 animals. Mice were ear clipped for identification purpose. The animals were acclimated for at least one week before start of the experiment.


Viruses. For each experimental group the designated virus stock was thawed on ice. The viruses were diluted to 1×106TCID50/3 μl in PBS. The overall volume was 100 μl. 80 μl of the virus suspension was used for stereotactic injection of one group of mice (n=5). The remaining 20 μl were used for titration according to standard procedure. Serial ½ log dilutions of the sample covered the range of 1×105-1×1010 TCID50/ml. Virus titration was done within one hour after preparation of the virus dilution for stereotactic injection.


Stereotactic Injection. Mice were weighted and an appropriate dose of Ketamine and Xylazine (100 μl per 10 g body weight) was injected i.p. After anesthesia had started, the skull between eye line and occiput was shaved properly and the eyes were protected with ointment (bepanthen crème). The mouse was placed on the stereotactic frame by fixing the ear bars into the external auditory canal bilaterally and enclosing the upper jaw with the jaw bar. The skin was antiseptically cleaned with betaiodine in ethanol before a linear skin incision over the midline along the antero-posterior axis was done. The galea-periosteum was scraped with centrifugal motion away from the midline and the bone was dried with a cotton swab with the same motion. After identification of the Bregma, the needle tip of the Hamilton syringe fixed in the stereotactic frame was adjusted directly over the Bregma. Then the needle was moved to the target location 0.4 mm rostral and 2 mm right lateral of the Bregma and the position was marked with a scalpel tip and the needle was removed again. Using an electric drill a burr hole was made at the target location and bone chips and dust was removed from the lesion with a PBS moistened cotton swab.


The syringe was filled up with prepared virus solution using the fast reverse mode of the automated injector. After fast forwarding the syringe plunger until the fluid forms a little droplet at the tip of the needle, it was cleaned with a moistened cotton swab. After the needle tip was positioned to the burr hole 0.4 mm rostral and 2 mm right lateral of the Bregma, the needle was lowered down to the desired coordinate at 3 mm deep. Then automated injection procedure was started. After the injection was finished, the needle was left in place for 1 min to prevent back-flow through the needle trajectory. Then the needle was slowly withdrawed from brain and scull. Before the wound was closed using Vetbond 3M, the lesion was cleaned with PBS. Then the wound was wiped with betaiodine to prevent infection and the animal was transferred to recovery cage equipped with a heating pad to prevent hypothermia.


Post-operative analgesia was provided in accordance with FELASA requirements for postoperative analgesia in rodents. Briefly, 30 mg/kg/day of Ibuprofen was supplied per os in the drinking water for 72 hours. The animals were monitored from the completion of surgery until recumbence and full mobility was achieved. In the first day animals were monitor several times and then daily for opening, discharge, redness or other possible signs of infection at the wound site.


Evaluation of neurotoxicity. After treatment, the animals were examined twice a day for one week or until all VSV-G animals had been sacrificed. After that, animals were monitored daily until day 40 post treatment. Scoring parameters include body weight, motility, appearance and body condition as well as clinical and provoked behavioral signs of neurotoxicity (FIG. 12). A cumulative toxicity score was calculated by addition of the particular scores. Euthanasia was applied according to defined endpoint criteria.










SEQUENCES



SEQ ID NO: 1










   1
atggggcagc tcataacaat gtttgaggct ttgccccaca tcatcgatga ggttatcaac






  61
atagttataa ttgtgcttgt gataataaca agcataaagg ctgtgtacaa ctttgctacc





 121
tgtggcatta ttgcactgat cagcttttgc ctcctagctg gcagatcttg tggtttatat





 181
ggtgtcaccg gtcctgacat ttacaaagga ctctaccaat tcaagtccgt ggagttcaac





 241
atgtcacagc tgaatttgac gatgcccaat gcatgttcag ccaacaactc ccaccattac





 301
atcagtatgg ggaagtctgg cttggagctg acttttacaa atgactccat catcagtcac





 361
aacttttgca acctgactga tgggttcaag aaaaaaacct ttgaccacac gctcatgagt





 421
atagtggcaa gcctacacct tagcatcaga ggaaacacca actataaggc tgtttcctgt





 481
gattttaaca atggaatcac catccaatac aacttgtctt tctctgatgc acaaagtgcc





 541
atcaatcaat gcagaacttt tagaggtaga gttctagaca tgttcaggac agctttcgga





 601
gggaagtaca tgaggtccgg ctatggttgg aagggctctg atgggaaaac tacttggtgc





 661
agtcagacca gttatcaata cctaatcata cagaacagaa catgggagaa tcactgtgag





 721
tacgccggtc cttttggcct ctcaagggtt ctctttgctc aggaaaaaac aaaatttctc





 781
actaggagat tggcagggac ttttacttgg acactgtcgg attcctcagg aactgagaac





 841
ccaggtgggt actgtctgac aaagtggatg cttatagctg ctgaactcaa gtgttttgga





 901
aacactgctg ttgctaaatg caatatcaat catgatgagg aattctgtga catgttgagg





 961
ctaattgatt acaacaaggc tgctctgaag aaattcaaag aagacgtaga gtctgccctt





1021
catctgttca agacaactgt gaattctcta atatctgacc agctgttaat gagaaatcat





1081
ttgagggact taatgggtgt gccctattgc aactactcga aattctggta cttggagcat





1141
gtgaaaacag gtgatacaag tgttccaaaa tgctggttgg tttctaacgg ctcatactta





1201
aatgaaacac atttcagtga ccagatagag caggaagcag acaacatgat cactgagatg





1261
ctcaggaaag attacatcaa gaggcaggga agcactcctt tggcactgat ggacctatta





1321
atgttctcta catcggcata tttaatcagt gttttccttc atctgatgaa aatcccaact





1381
catagacaca ttaaaggtgg cacatgccct aagccacaca ggttaactag taaaggcatc





1441
tgtagctgtg gtgcgttcaa agtgccagga gtgaaaacag tctggaagag acgctga











>Danedong NC_010248.1



SEQ ID NO: 2



MGQLITMFEALPHIIDEVINIVIIVLVIITSIKAVYNFATCGIIALISFCLLAGRSCGLYGVTGPDIYKGLYQFKSVEFNMSQLNLTMPNACSA






NNSHHYISMGKSGLELTFTNDSIISHNFCNLTDGFKKKTFDHTLMSIVASLHLSIRGNTNYKAVSCDFNNGITIQYNLSFSDAQSAINQCRTFR





GRVLDMFRTAFGGKYMRSGYGWKGSDGKTTWCSQTSYQYLIIQNRTWENHCEYAGPFGLSRVLFAQEKTKFLTRRLAGTFTWTLSDSSGTENPG





GYCLTKWMLIAAELKCFGNTAVAKCNINHDEEFCDMLRLIDYNKAALKKFKEDVESALHLFKTTVNSLISDQLLMRNHLRDLMGVPYCNYSKFW





YLEHVKTGDTSVPKCWLVSNGSYLNETHFSDQIEQEADNMITEMLRKDYIKRQGSTPLALMDLLMFSTSAYLISVFLHLMKIPTHRHIKGGTCP





KPHRLTSKGICSCGAFKVPGVKTVWKRR





SEQ ID NO: 3










   1
    atgggg cagatagtca ccttctttca agaggtgcca cacatccttg aagaagtgat






  61
gaacattgtg ctgatgaccc tctcaatctt ggccatccta aagggcatct acaatgtgat





 121
gacctgtgga atcatcggtt tgataacatt tttgttcttg tgtgggagat catgctcaag





 181
catctataag gacaactatg agttcttctc tctcgacctc gacatgtctt cactgaatgc





 241
aacgatgcct ctctcctgct caaagaacaa ctcccatcac tacatccaag ttgggaatga





 301
gacaggccta gagctgacat tgacaaacac tagcataatt gatcataagt tttgcaacct





 361
gtctgatgct cacaggagga atctttatga caaagcactt atgtcaatct tgacaacatt





 421
ccacctaagc attccagatt ttaaccaata tgaggcaatg tcatgtgatt ttaacggagg





 481
gaagatctca attcaataca acctgtccca ctcaaattat gtggatgccg gcaaccactg





 541
tggcactatt gcaaatggca ttatggatgt ttttagaaga atgtattgga gcacctccct





 601
ttcggttgcc tctgacataa gtgggactca atgcatacag accgattata agtatttgat





 661
tattcagaac acatcatggg aggatcattg catgttctca agaccctcac ccatgggatt





 721
tttaagcctt ctgtcacaga gaaccaggaa tttctacatc tcaagaagac ttttgggtct





 781
ttttacatgg actttgagtg actcggaagg gaacgacatg ccaggtggtt actgtctcac





 841
aaggtctatg ctaatagggc ttgatctgaa atgctttggg aacactgcca ttgcgaagtg





 901
taatcaggca catgatgaag aattctgtga catgctccgc ctttttgatt ttaataaaca





 961
agcaattagc aaactgaggt ctgaagttca gcagagcata aatttgataa ataaagctgt





1021
caacgccctc atcaatgatc aactggtcat gaggaaccat ttgagagatc taatgggtat





1081
tccttattgc aactactcta agttttggta cttgaatgat actaggacag ggagaacatc





1141
cctcccaaag tgttggcttg tgaccaatgg gtcataccta aatgaaaccc agttctcaac





1201
agaaattgag caagaagcca ataacatgtt cactgacatg ttgaggaagg agtatgagaa





1261
aaggcagagc acaacacctc tggggttagt agatcttttt gttttctcca ctagctttta





1321
tttgatctcc gtgttcctcc acctaatcaa aattccaaca catagacaca taaaaggcaa





1381
gccttgcccg aaaccacaca gactcaatca catggcaatc tgttcctgtg gcttctataa





1441
gcaaccaggt ctccccacac aatggaaaag gtga











>Mopeia JN_561684.1



SEQ ID NO: 4



MGQIVTFFQEVPHILEEVMNIVLMTLSILAILKGIYNVMTCGIIGLITFLFLCGRSCSSIYKDNYEFFSLDLDMSSLNATMPLSCSKNNSHHYI






QVGNETGLELTLTNTSIIDHKFCNLSDAHRRNLYDKALMSILTTFHLSIPDFNQYEAMSCDFNGGKISIQYNLSHSNYVDAGNHCGTIANGIMD





VFRRMYWSTSLSVASDISGTQCIQTDYKYLIIQNTSWEDHCMFSRPSPMGFLSLLSQRTRNFYISRRLLGLFTWTLSDSEGNDMPGGYCLTRSM





LIGLDLKCFGNTAIAKCNQAHDEEFCDMLRLFDFNKQAISKLRSEVQQSINLINKAVNALINDQLVMRNHLRDLMGIPYCNYSKFWYLNDTRTG





RTSLPKCWLVTNGSYLNETQFSTEIEQEANNMFTDMLRKEYEKRQSTTPLGLVDLFVFSTSFYLISVFLHLIKIPTHRHIKGKPCPKPHRLNHM





AICSCGFYKQPGLPTQWKR





SEQ ID NO: 5










   1
                                                     a tgggacagat






  61
aataaccttc tttcaggaag tgcctcacat tatagaggaa gtcatgaaca ttgttctgat





 121
caccctatcc ctcttggcta tcttgaaggg tgtgtacaat gtcatgactt gtggtctgat





 181
tggattgatt tcctttctct tactgtgtgg gaagtcctgc tctctaatct acaaggacac





 241
atataacttc tcatccattg aacttgacct ctcacacctc aacatgacat tgcccatgtc





 301
ctgcagccga aacaactcac accactatgt cttcttcaac ggatctggct tagagatgac





 361
attcactaac gattcacttc ttaaccacaa attctgcaat ctttcagacg cccataaaaa





 421
gaatctttac gatcatgcgc taatgggcat tgtcacaact ttccacttgt caatcccgaa





 481
cttcaatcaa tacgaagcaa tggcctgtga cttcaatgga gggaatatta gcattcaata





 541
caacttgagc cacaatgatc gaacagatgc aatgaaccac tgtggcactg tagctaatgg





 601
tgtactggat gctttttata ggttccattg gggcagaaat ataacctaca tcgctcagtt





 661
gccaaatggc gacggcactg gcagatggac attctgttat gccacaagtt ataagtacct





 721
ggttatccaa aacattagct gggctgatca ttgtcaaatg tcaagaccaa ctcccattgg





 781
ctttgctagc attctgagcc aacggattag gagcatttac atttcaagac gactgatgag





 841
cacattcact tggtctctat ccgacagttc aggtactgag aatccaggtg gctattgtct





 901
caccagatgg atgctttttg cggctgactt gaaatgcttt ggaaacacag ccattgccaa





 961
atgtaacctg aatcatgatg aagagttctg cgacatgttg aggcttattg acttcaacaa





1021
gcaagccttg aaaacattca aatcggaggt taaccatggt ctacaactaa ttacaaaggc





1081
gattaatgcc ttgatcaatg accaactcat tatgaagaat cacttaaggg acctaatggg





1141
tatcccttat tgcaattatt caaaattctg gtatctaaat gacacaagaa caggtagagt





1201
ctctctgcct aagtgctgga tgatcagcaa cgggacctat ctaaatgaga ctcatttttc





1261
tgatgagatt gaacaggaag cagacaatat gatcacagaa atgttgagaa aagagtatca





1321
agaaagacaa gggaagaccc cattgggtct agttgatctt tttattttta gcacaagctt





1381
ttattcaatc acagtgttcc tccatctcat aaagattcca acgcacagac acatcgtggg





1441
acaagggtgt cccaagccac acagactaaa cagtagagca atctgctcct gtggtgccta





1501
taaacagccg ggactaccca ccaagtggaa gcgctga











>Ippy NC_007905.1



SEQ ID NO: 6



MGQIITFFQEVPHIIEEVMNIVLITLSLLAILKGVYNVMTCGLIGLISFLLLCGKSCSLIYKDTYNFSSIELDLSHLNMTLPMSCSRNNSHHYV






FFNGSGLEMTFTNDSLLNHKFCNLSDAHKKNLYDHALMGIVTTFHLSIPNFNQYEAMACDFNGGNISIQYNLSHNDRTDAMNHCGTVANGVLDA





FYRFHWGRNITYIAQLPNGDGTGRWTFCYATSYKYLVIQNISWADHCQMSRPTPIGFASILSQRIRSIYISRRLMSTFTWSLSDSSGTENPGGY





CLTRWMLFAADLKCFGNTAIAKCNLNHDEEFCDMLRLIDFNKQALKTFKSEVNHGLQLITKAINALINDQLIMKNHLRDLMGIPYCNYSKFWYL





NDTRTGRVSLPKCWMISNGTYLNETHFSDEIEQEADNMITEMLRKEYQERQGKTPLGLVDLFIFSTSFYSITVFLHLIKIPTHRHIVGQGCPKP





HRLNSRAICSCGAYKQPGLPTKWKR





SEQ ID NO: 7










  61
                               a tgggtcaagt gattggattc ttccaatcat






 121
tgcccgagat tattaatgaa gcgttgaaca ttgctctgat ctgtgtggct ctgcttgcca





 181
caatcaaggg aatggtcaac atctggaaat ctggtttgat acagctcctc ttcttcctca





 241
cattggcagg aagaagttgt tctcattctt ttacaattgg aagatttcat gaatttcaga





 301
gtgtgacagt taactttaca cagttcatgt cctatgctcc aagctcttgc tcagtaaaca





 361
acacccacca ttatttcaaa ggtccccaaa acaccacctg gggtcttgag ttgactttga





 421
ctaatgaatc aatgataaac atcaccaatt ccatgagggt cttcactaac attcatcaca





 481
atgtaaccaa ttgcgtgcag aacatttccg aacatgaggg tgtgctcaaa tggctgcttg





 541
agacaatgca cctaagcatc agcaagccag gaaaacacat cgcacctgtc atgtgtgaga





 601
gacaaaaggg gttgctcatc gagtacaatc tcactatgac caaagaccac cacccaaatt





 661
attggaatca agtcttgtat ggactggcta aattattggg ctcaagtaag cgtttgtggt





 721
ttggcgcatg taacaaggct gactgtcaga tgcaatcaga ccaccagcac ataaaatgca





 781
attactcgaa ttgcaaaggt tacaccagct ttaaatactt aattattcaa aataccactt





 841
gggaaaacca ttgtgagtat aaccatctga acaccatcca ccttctcatg agctctattg





 901
gccaatcctt tatcacaaga aggcttcaag cctttttaac ctggacactt tctgatgcac





 961
tagggaatga ccttcctggg ggatactgtc ttgaacagtg ggctgtggtg tggtttggca





1021
taaagtgttt tgataacaca gcaatggcca agtgcaatca gaatcatgat tcagagtttt





1081
gtgatatgct aaggcttttt gattacaata gaaatgctat tcagagctta aatgatcaat





1141
ctcaagccag attaaacctt ttaactaaca ccattaattc acttgtctct gacaatttac





1201
taatgaaaaa taagctaaga gaactaatga acgtgccata ctgtaattac actagattct





1261
ggtttattaa tgatacaaag aatggtagac acactcttcc acaatgttgg ttggtatcag





1321
atggttcgta cttgaatgaa accagattca gaactcaatg gctaagtgaa agtaactctt





1381
tatacacaga aatgctgact gaagaatatg aaaaaagaca gggtagaaca cctctttctt





1441
tagtggacct atgcttttgg tcaactctct tttacatatc aacattattt gcgcatttag





1501
tgggtttccc aactcacaga caccttatag gtgagggctg cccaaaacca cacagattaa





1561
cagggtcagg gatctgctca tgtggacatt atggtattcc aggtaaacca gtcagatgga





1621
caaagatgtc aaggtga











>Latino NC_010758.1



SEQ ID NO: 8



MGQVIGFFQSLPEIINEALNIALICVALLATIKGMVNIWKSGLIQLLFFLTLAGRSCSHSFTIGRFHEFQSVTVNFTQFMSYAPSSCSVNNTHH






YFKGPQNTTWGLELTLTNESMINITNSMRVFTNIHHNVTNCVQNISEHEGVLKWLLETMHLSISKPGKHIAPVMCERQKGLLIEYNLTMTKDHH





PNYWNQVLYGLAKLLGSSKRLWFGACNKADCQMQSDHQHIKCNYSNCKGYTSFKYLIIQNTTWENHCEYNHLNTIHLLMSSIGQSFITRRLQAF





LTWTLSDALGNDLPGGYCLEQWAVVWFGIKCFDNTAMAKCNQNHDSEFCDMLRLFDYNRNAIQSLNDQSQARLNLLTNTINSLVSDNLLMKNKL





RELMNVPYCNYTRFWFINDTKNGRHTLPQCWLVSDGSYLNETRFRTQWLSESNSLYTEMLTEEYEKRQGRTPLSLVDLCFWSTLFYISTLFAHL





VGFPTHRHLIGEGCPKPHRLTGSGICSCGHYGIPGKPVRWTKMSR





SEQ ID NO: 9










  61
                                                             atgg






 121
gtcaagtcat tgggttcttc caatccttac ccaacatcat taatgaggca ctaaacattg





 181
ccctcatctg tgtggcatta attgccatat tgaaaggtat tgttaacata tggaaatcag





 241
gtctgattca gctgttcata ttcctcatcc tagcggggag gagctgttct cacacctttc





 301
agattggccg aaaccatgag ttccaaagta tcactctaaa cttcacacag tttttgggct





 361
acgcacccag ctcctgttct gttaacaaca ctcatcacta ctttagaggt cctggcaatg





 421
tctcttgggg catcgagctc acattgacca acaactccgt gatcaatgca agcaactcac





 481
tgaaggtgtt caccaacatt caccacaaca tcaccaactg cgttcagaat atcgatgagc





 541
aagaccacct aatgaaatgg ctcatagaaa caatgcatct acagatcatg aaaccaggaa





 601
agagactacc accaatcctg tgtgagaaag acaaagggtt actaattgaa tacaacctaa





 661
ccaacattgc atctcgcgaa gaaaagcact ctgagtactg gagccaactt ctctatggtc





 721
tatcaaaact attaggctcc agtaagtcat tgtggtttga ttactgtcag agagcagatt





 781
gcatgatgca agagcacagc tcacacttga agtgcaacta ctcagagtgt tctggccata





 841
ccaccttcaa atacctcatt ctacaaaaca ccacttggga gaaccattgt gagttcaacc





 901
atctaaacac aattcacctg ttgatgagct ctactggtca gtctttcata actagaaggt





 961
tgcaggcttt tctaacttgg actttgtcag atgctacagg caatgacctc cctgggggtt





1021
actgcctaga gcagtgggcc atagtttggg caggcatcaa gtgctttggg aacacagcag





1081
ttgcaaagtg taatcaaaat catgactctg aattctgtga catgttgagg ctctttgatt





1141
acaataggaa tgctattaag agtctgaatg atcagtctca aagtcgtcta aatcttctaa





1201
caaatacaat aaattcactg atttcggata atctgttgat gaaaaacaag ttggctgaga





1261
taatgaacat tccgtattgc aattacacaa aattctggta catcaacgac actaggactg





1321
gaaggcacac ccttccccaa tgctggttaa tctcaaatgg gtcttacttg aatgagacta





1381
aattcagaac tcagtggctg tcagagagca atgcactcta tacagaaatg ctaacagaag





1441
attatgacaa aaggcagggt agcacaccac tctccttggt tgatctatgt ttttggtcaa





1501
ccttattcta tgtgaccaca ttgtttgctc atcttgttgg tttcccaaca cacagacaca





1561
tcttagatgg tccttgccct aaaccccaca gattaacaaa gaaaggaatt tgttcctgtg





1621
gacattttgg aataccgggc aaacctgttc gatgggtgaa acgatcacgg tga











>Oliveros NC_010248.1



SEQ ID NO: 10



MGQVIGFFQSLPNIINEALNIALICVALIAILKGIVNIWKSGLIQLFIFLILAGRSCSHTFQIGRNHEFQSITLNFTQFLGYAPSSCSVNNTHH






YFRGPGNVSWGIELTLTNNSVINASNSLKVFTNIHHNITNCVQNIDEQDHLMKWLIETMHLQIMKPGKRLPPILCEKDKGLLIEYNLTNIASRE





EKHSEYWSQLLYGLSKLLGSSKSLWFDYCQRADCMMQEHSSHLKCNYSECSGHTTFKYLILQNTTWENHCEFNHLNTIHLLMSSTGQSFITRRL





QAFLTWTLSDATGNDLPGGYCLEQWAIVWAGIKCFGNTAVAKCNQNHDSEFCDMLRLFDYNRNAIKSLNDQSQSRLNLLTNTINSLISDNLLMK





NKLAEIMNIPYCNYTKFWYINDTRTGRHTLPQCWLISNGSYLNETKFRTQWLSESNALYTEMLTEDYDKRQGSTPLSLVDLCFWSTLFYVTTLF





AHLVGFPTHRHILDGPCPKPHRLTKKGICSCGHFGIPGKPVRWVKRSR






BIBLIOGRAPHY



  • [1] Sean E. Lawler, Maria-Carmela Speranza, Choi-Fong Cho et al. Oncolytic Viruses in Cancer Treatment JAMA Oncol. 2017; 3(6):841-849.

  • [2] Liu B L, Robinson M, Han Z Q, et al. ICP34.5 deleted herpes simplex virus with enhanced oncolytic, immune stimulating, and anti-tumour properties. Gene Ther. 2003; 10(4):292-303.

  • [3] Guse K, Cerullo V, Hemminki A. Oncolytic vaccinia virus for the treatment of cancer. Expert Opin Biol Ther. 2011; 11(5):595-608.

  • [4] Weller S K, Coen D M. Herpes simplex viruses: mechanisms of DNA replication. Cold Spring Harb Perspect Biol. 2012; 4(9):a013011.

  • [5] Lichty B D, Power A T, Stojdl D F, Bell J C. Vesicular stomatitis virus: re-inventing the bullet. Trends Mol Med. 2004; 10(5):210-216.

  • [6] Hastie E, Grdzelishvili V Z. Vesicular stomatitis virus as a flexible platform for oncolytic virotherapy against cancer. J Gen Virol. 2012; 93(pt 12):2529-2545.

  • [7] Russell S J, Peng K W. Measles virus for cancer therapy. Curr Top Microbiol Immunol. 2009; 330:213-241.

  • [8] Zamarin D, Palese P. Oncolytic Newcastle disease virus for cancer therapy: old challenges and new directions. Future Microbiol. 2012; 7(3):347-367.

  • [9] Alexander Muik, Inna Kneiske, Marina Werbizki, et al. Pseudotyping Vesicular Stomatitis Virus with Lymphocytic Choriomeningitis Virus Glycoproteins Enhances Infectivity for Glioma Cells and Minimizes Neurotropism. J Virol. 2011 June; 85(11): 5679-5684.

  • [10] Reinhard Tober, Zoltan Banki, Lisa Egerer, et al. VSV-GP: a Potent Viral Vaccine Vector That Boosts the Immune Response upon Repeated Applications J Virol. 2014 May; 88(9): 4897-4907.

  • [11] R Tober, Z Banki, A Ejaz, et al. The viral vector vaccine VSV-GP boosts immune response upon repeated applications. Retrovirology. 2012; 9 (Suppl 2): P301. Published online 2012 Sep. 13.

  • [12] Palacios G, Druce J, Du L, et al. A New Arenavirus in a cluster of fatal transplant-associated diseases. N Engl J Med. 2008; 358:991-998

  • [13] Georges A J, Gonzalez J P, Abdul-Wahid S, Saluzzo J F, Meunier D M, McCormick J B. Antibodies to Lassa and Lassa-like viruses in man and mammals in the Central African Republic. Trans R Soc Trop Med Hyg. 1985; 79(1):78-9.

  • [14] Digoutte, Rapport de l'Institut Pasteur de Bangui (1970), p. 59.

  • [15] Murphy F A, Webb P A, Johnson K M, Whitfield S G, Chappell W A. Murphy et al. Arenoviruses in Vero cells: ultrastructural studies. J. Virol 6 (1970), pp. 507-518.

  • [16] Bowen M D, Peters C J, Mills J N, Nichol S T. Oliveros virus: a novel arenavirus from Argentina. Virology. 1996 Mar. 1; 217(1):362-6.

  • [17] Edgar R C. MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res. 2004 Mar. 19; 32(5):1792-7.

  • [18] Beyer W R, Miletic H, Ostertag W, von Laer D. Recombinant expression of lymphocytic choriomeningitis virus strain W E glycoproteins: a single amino acid makes the difference. J Virol. 2001 January; 75(2):1061-4.

  • [19] Owens R J, Rose J K. Cytoplasmic domain requirement for incorporation of a foreign envelope protein into vesicular stomatitis virus. J Virol. 1993 January; 67(1):360-5.

  • [20] Tani H, Komoda Y, Matsuo E, Suzuki K, Hamamoto I, Yamashita T, Moriishi K, Fujiyama K, Kanto T, Hayashi N, Owsianka A, Patel A H, Whitt M A, Matsuura Y. Replication-competent recombinant vesicular stomatitis virus encoding hepatitis C virus envelope proteins. J Virol. 2007 August; 81(16):8601-12. Epub 2007 Jun. 6.

  • [21] Ge J, Wen Z, Wang X, Hu S, Liu Y, Kong X, Chen H, Bu Z. Generating vesicular stomatitis virus pseudotype bearing the severe acute respiratory syndrome coronavirus spike envelope glycoprotein for rapid and safe neutralization test or cell-entry assay. Ann N Y Acad Sci. 2006 October; 1081:246-8.

  • [22] Garbutt M, Liebscher R, Wahl-Jensen V, Jones S, Möller P, Wagner R, Volchkov V, Klenk H D, Feldmann H, Ströher U. Garbutt et al. Properties of replication-competent vesicular stomatitis virus vectors expressing glycoproteins of filoviruses and arenaviruses. J Virol. 2004 May; 78(10):5458-65.

  • [23] Miletic H, Fischer Y, Litwak S, Giroglou T, Waerzeggers Y, Winkeler A, Li H, Himmelreich U, Lange C, Stenzel W, Deckert M, Neumann H, Jacobs A H, von Laer D. Bystander killing of malignant glioma by bone marrow-derived tumor-infiltrating progenitor cells expressing a suicide gene. Mol Ther. 2007 July; 15(7):1373-81.

  • [24] Fischer Y H, Miletic H, Giroglou T, Litwak S, Stenzel W, Neumann H, von Laer D. A retroviral packaging cell line for pseudotype vectors based on glioma-infiltrating progenitor cells. J Gene Med. 2007 May; 9(5):335-44.

  • [25] Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)

  • [26] McLaughlin S K1, Collis P, Hermonat P L, Muzyczka N. Adeno-associated virus general transduction vectors: analysis of proviral structures. J Virol. 1988 June; 62(6):1963-73.

  • [27] Urbiola C, Santer F R, Petersson M, van der Pluijm G, Horninger W, Erlmann P, Wollmann G, Kimpel J, Culig Z, von Laer D. Oncolytic activity of the rhabdovirus VSV-GP against prostate cancer. Int J Cancer. 2018 Apr. 26.

  • [28] Jenks N, Myers R, Greiner S M, Thompson J, Mader E K, Greenslade A, Griesmann G E, Federspiel M J, Rakela J, Borad M J, Vile R G, Barber G N, Meier T R, Blanco M C, Carlson S K, Russell S J, Peng K W. Safety studies on intrahepatic or intratumoral injection of oncolytic vesicular stomatitis virus expressing interferon-beta in rodents and nonhuman primates. Hum Gene Ther. 2010 April; 21(4):451-62.

  • [29] Kaku Y, Noguchi A, Marsh G A, Barr J A, Okutani A, Hotta K, Bazartseren B, Fukushi S, Broder C C, Yamada A, Inoue S, Wang L F Second generation of pseudotype-based serum neutralization assay for Nipah virus antibodies: sensitive and high-throughput analysis utilizing secreted alkaline phosphatase. J Virol Methods. 2012 January; 179(1):226-32.


Claims
  • 1. VSV chimeric vector, characterized in that the vector comprises a gene coding for a glycoprotein GP of the Dandenong virus or Mopeia virus and lacks a functional gene coding for envelope protein G of the VSV.
  • 2. The vector of claim 1, wherein the vector shows increased killing of tumor cells and reduced induction of neutralizing antibodies as compared to a VSV vector pseudotyped with GP of LCMV under identical conditions.
  • 3. The vector of claim 1 or 2, characterized in that the envelope protein G of VSV is replaced by GP of the Dandenong virus or Mopeia virus.
  • 4. The vector of any one of claims 1 to 3, characterized in that the vector further comprises at least one transgene.
  • 5. VSV chimeric vector system, characterized in that the system comprises at least two complementary replicating VSV vectors, wherein the system comprises genes n, l, p and m coding for proteins N, L, P and M of the VSV, a gene gp coding for Dandenong-GP or Mopeia-GP and lacks a functional gene coding for G protein of the VSV, wherein each vector of the system lacks one of the genes n, l, p, m and gp, and wherein the lacking gene is present on any other vector of the system.
  • 6. Chimeric VSV virion, characterized in that the virion comprises a GP protein of the Dandenong or Mopeia virus as envelope protein.
  • 7. Virus producing cell, characterized in that the cell produces a chimeric VSV virion of claim 6.
  • 8. The virus producing cell of claim 7, characterized in that the cell is a mammalian cell.
  • 9. The virus producing cell of claim 8, characterized in that the mammalian cell is a multipotent adult progenitor cell (MAPC), a neural stem cell (NSC), a mesenchymal stem cell (MSC), a HeLa cell, any HEK293 cell, a Vero cell or a bone marrow derived tumor infiltrating cell (BM-TIC).
  • 10. The virus producing cell of any one of claims 7 to 9, characterized in that the cell comprises one or more expression cassettes for the expression of at least one of the genes selected from the group consisting of genes n, l, p and m coding for proteins N, L, P and M of the VSV and a gene gp coding for Dandenong or Mopeia GP glycoprotein.
  • 11. The virus producing cell of any one of claims 7 to 10, characterized in that the cell comprises a gene transfer vector for the packaging into a VSV virion pseudotyped with GP of Dandenong or Mopeia virus, wherein the gene transfer vector comprises a transgene.
  • 12. Method for transfer of a transgene into a cell in vitro, characterized in that the cell is transduced with a chimeric virion of claim 6, wherein the virion comprises a transgene.
  • 13. Method for transfer of a transgene into a cell in vitro, characterized in that the cell is contacted with a virus producing cell of any one of claims 7 to 11.
  • 14. The method of claim 12 or 13, characterized in that the cell is a tumor cell.
  • 15. Pharmaceutical composition, characterized in that the composition comprises the VSV chimeric vector of any one of claims 1 to 4, the VSV chimeric vector system of claim 5, the chimeric VSV virion of claim 6, or the virus producing cell of any one of claims 7 to 11.
  • 16. A VSV chimeric vector of any one of claims 1 to 4, a VSV chimeric vector system of claim 5, a chimeric VSV virion of claim 6, a virus producing cell of any one of claims 7 to 11, or a pharmaceutical composition of claim 15 for use as medicament.
  • 17. A VSV chimeric vector of any one of claims 1 to 4, a VSV chimeric vector system of claim 5, a pseudotyped VSV virion of claim 6, a virus producing cell of any one of claims 7 to 11, or a pharmaceutical composition of claim 15 for use in treating cancer.
  • 18. The VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition for use according to claim 17, wherein the cancer is a solid cancer.
  • 19. The VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition for use according to claim 18, characterized in that the solid cancer is brain cancer, colorectal cancer, oropharyngeal squamous cell carcinoma, gastric cancer, gastroesophageal junction adenocarcinoma, esophageal carcinoma, hepatocellular carcinoma, pancreatic adenocarcinoma, cholangiocarcinoma, bladder urothelial carcinoma, metastatic melanoma, prostate carcinoma, breast carcinoma, glioblastoma, non-small cell lung cancer, brain tumor or small cell lung cancer.
  • 20. The VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition for use according to any one of claims 16 to 19, wherein the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition is combined with a PD-1 or PD-L1 antagonist.
  • 21. The VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition for use according to any one of claims 16 to 20, wherein the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition is to be administered intratumorally or intravenously.
  • 22. The VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition for use according to claim 21, wherein the VSV chimeric vector, the VSV chimeric vector system, the chimeric VSV virion, the virus producing cell or the pharmaceutical composition is to be administered intratumorally and subsequently intravenously.
Priority Claims (1)
Number Date Country Kind
18208074.7 Nov 2018 EP regional
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2019/082328 11/22/2019 WO 00