WO2024223847A1 - Particule de vecteur rétroviral pseudotypé avec affichage anti-cd3 - Google Patents
Particule de vecteur rétroviral pseudotypé avec affichage anti-cd3 Download PDFInfo
- Publication number
- WO2024223847A1 WO2024223847A1 PCT/EP2024/061576 EP2024061576W WO2024223847A1 WO 2024223847 A1 WO2024223847 A1 WO 2024223847A1 EP 2024061576 W EP2024061576 W EP 2024061576W WO 2024223847 A1 WO2024223847 A1 WO 2024223847A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- antigen
- seq
- retroviral vector
- vector particle
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/005—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2809—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/60—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
- C07K2317/62—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
- C07K2317/622—Single chain antibody (scFv)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/03—Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/33—Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/13011—Gammaretrovirus, e.g. murine leukeamia virus
- C12N2740/13041—Use of virus, viral particle or viral elements as a vector
- C12N2740/13043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/13011—Gammaretrovirus, e.g. murine leukeamia virus
- C12N2740/13041—Use of virus, viral particle or viral elements as a vector
- C12N2740/13045—Special targeting system for viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/16011—Human Immunodeficiency Virus, HIV
- C12N2740/16041—Use of virus, viral particle or viral elements as a vector
- C12N2740/16043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/16011—Human Immunodeficiency Virus, HIV
- C12N2740/16041—Use of virus, viral particle or viral elements as a vector
- C12N2740/16045—Special targeting system for viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2760/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
- C12N2760/00011—Details
- C12N2760/18011—Paramyxoviridae
- C12N2760/18211—Henipavirus, e.g. hendra virus
- C12N2760/18222—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2760/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
- C12N2760/00011—Details
- C12N2760/18011—Paramyxoviridae
- C12N2760/18411—Morbillivirus, e.g. Measles virus, canine distemper
- C12N2760/18422—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2810/00—Vectors comprising a targeting moiety
- C12N2810/50—Vectors comprising as targeting moiety peptide derived from defined protein
- C12N2810/60—Vectors comprising as targeting moiety peptide derived from defined protein from viruses
- C12N2810/6072—Vectors comprising as targeting moiety peptide derived from defined protein from viruses negative strand RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2810/00—Vectors comprising a targeting moiety
- C12N2810/50—Vectors comprising as targeting moiety peptide derived from defined protein
- C12N2810/60—Vectors comprising as targeting moiety peptide derived from defined protein from viruses
- C12N2810/6072—Vectors comprising as targeting moiety peptide derived from defined protein from viruses negative strand RNA viruses
- C12N2810/6081—Vectors comprising as targeting moiety peptide derived from defined protein from viruses negative strand RNA viruses rhabdoviridae, e.g. VSV
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2810/00—Vectors comprising a targeting moiety
- C12N2810/50—Vectors comprising as targeting moiety peptide derived from defined protein
- C12N2810/80—Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates
- C12N2810/85—Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian
- C12N2810/859—Vectors comprising as targeting moiety peptide derived from defined protein from vertebrates mammalian from immunoglobulins
Definitions
- the present invention generally relates to the field of the generation of pseudotyped retroviral vector particles, in particular to the generation of pseudotyped retroviral vector particles with anti-CD3 display.
- CAR T cells play a key role in cell-mediated immunity and are therefore an interesting target for adoptive immunotherapy such as chimeric antigen receptor (CAR) T cell immunotherapy.
- CAR T manufacturing processes involve isolation of T cells from the human body, polyclonal activation, genetic modification with lentiviral vectors (LV) followed by an expansion phase. This complex process is time and cost intensive, but also results in a high degree of in vitro modification of the T cells. Decreasing the process complexity e.g. by omitting the activation step or by reducing the expansion phase therefore offers the potential to improve the cost efficiency of CAR T manufacture.
- CAR T cell function In addition, reduced ex vivo cultivation and production from a naive phenotype was shown to improve CAR T cell function (Ghassemi et al (2016); Arcangeli et al (2022)). In-situ generation of CAR T cells offers the potential to further reduce the complexity of manufacturing and to completely avoid in vitro manipulation of the T cells. To achieve this goal, efficient methods are required to transduce non-activated T cells and subsequently to induce sufficient proliferation in vivo.
- VSV-G LV Vesicular Stomatitis Virus glycoproteins
- MV-LV MV-LV
- NiV-LV Nipah virus glycoproteins
- T cells require simultaneous engagement of the T-cell receptor with the major histocompatibility complex peptide and co-stimulatory molecules on the antigen presenting cells for activation. In vitro this can be achieved by supplementing CD3 and CD28 agonists to the culture, e.g. anti-CD3 and anti-CD28 antibodies.
- T cells may be activated with anti-CD3 and anti-CD28 antibodies coupled to beads (Kalamasz et al (2004), Li et al (2010)).
- an in vitro method for polyclonal stimulation of T cells comprising contacting a population of T cells with a humanized anti- CD3 antibody fragment (a scFV of a mutated Oct3 clone) linked to solid particles.
- a humanized anti- CD3 antibody fragment a scFV of a mutated Oct3 clone
- Optimal stimulation of T cells needs additional presentation of an anti-CD28 antibody fragment, linked to particles.
- activation of T cells can be induced by retroviral vectors that present activating ligands within their envelope.
- Maurice et al (2002) presented a technology for displaying the anti-CD3 scFV (Okt3) on the envelope of VSV-G pseudotyped retroviral vectors. They demonstrated both activation of T cells and transduction of non-activated T cells, however proliferation was decreased compared to T cells activated with anti-CD3 an anti-CD28 antibodies.
- Derdak et al (2006) described the development of activating virus-like particles.
- a viral particle comprising a vector genome comprising a polynucleotide sequence encoding an anti-CD19 chimeric antigen receptor, wherein the viral particle transduces immune cells in vivo, and the particle may comprise a viral envelope that comprises an anti-CD3 single-chain variable fragment exposed on the surface and/or conjugated to the surface of the viral envelope.
- a multi ci str onic vector for surface-engineering retroviral particles comprising a polynucleotide operatively linked to a promoter, wherein the polynucleotide encodes a plurality of polypeptides joined by linkers comprising peptides capable of inducing ribosome skipping or self-cleavage, and wherein the plurality of polypeptides comprise a fusion glycoprotein or functional variant thereof and one or more non- viral proteins capable of viral surface display.
- Said non-viral protein may be specific for human CD3 (anti-CD3 scFV).
- a retroviral vector particle for activating and efficiently transducing T cells comprising a nucleic acid sequence encoding a small-molecule controllable T-cell/NK-cell activation receptor, the nucleic acid sequence operatively linked to a promoter, wherein the T-cell/NK-cell activation receptor is capable of being activated by a small molecule.
- Said retroviral vector particle may be a surface-engineered retroviral vector particle comprising a T-cell activation or co-stimulation molecule.
- Said T-cell activation or co-stimulation molecule may comprise one or more of an anti-CD3 antibody, CD28 ligand, and 41bb ligand.
- retroviral vector particles for activating and efficiently transducing T cells comprising a mitogenic T cell activating transmembrane protein which binds to mitogenic tetraspanins.
- Said retroviral vector may also comprise an additional transmembrane protein which binds CD3.
- NiV-LVs are retargeted to T cells by fusion of CD3-scFVs (clone TR66 and HuM291) to Nipah-G. They demonstrate successful transduction and activation of non-activated T cells. However, both, marker expression and cytokine secretion are reduced compared to state-of-the art T cell activation. Moreover, proliferation was only induced upon co-stimulation with CD28-specific antibodies.
- Huckaby et al (2019) described activation of T cells using Sindbis-pseudotyped LVs. Here, Sindbis-pseudotyped LV were retargeting a bispecific Fab molecule (a-CD3 and Sindbis E2). However, transduction efficiency on nonactivated T cells remained low and activation of T cells was not evaluated.
- a method of transducing immune cells in a subject in need thereof comprising: a) administering a multispecific antibody to render immune cells in the subject more transducible; and b) administering a vector, optionally a viral vector; wherein the method transduces the immune cells.
- Said multispecific antibody may comprise anti-CD3.
- retroviruses and also retroviral vectors incorporate host cell proteins or artificial proteins in their envelope.
- the process of protein incorporation into retroviral envelopes is not well understood, the required protein structure needs to be evaluated.
- Rasbach et al (2013) described the display of a third envelope protein in measles pseudotyped retroviral vector particles. They displayed an a-Her2 scFV on the viral particle by expressing it in the pDisplay vector system, however reproducibility with alternative scFVs was not shown.
- Taube et al (2008) describe the expression of antibodies in cells and on viral vectors.
- a scFV with FC region was designed in the context of a IgG Signal and gp41, CD8 or CD28 (short extracellular domain & transmembrane domain).
- gp41 incorporation signal was attached. The influence of the protein architecture on the cell surface expression and viral incorporation was described.
- the present invention makes use of the discovery that retroviral vector particles such as lentiviral vectors can display T cell activating ligands within their envelope.
- retroviral vector particles such as lentiviral vectors can display T cell activating ligands within their envelope.
- the inventors surprisingly identified a modulating protein, as disclosed herein, especially advantageous in the field of immunotherapy to combine activation and genetic modification of T cells in one step induced by the same viral particles.
- the modulating protein comprising SEQ ID NO: 1 (VH) and SEQ ID NO:2 (VL) has become manifest for example in SEQ ID NO:3 or SEQ ID NO:4 for the binding to CD3, in HEK293T cells was enhanced compared to the modulating protein, having SEQ ID NO:5 for the binding to CD3.
- the surface expression on HEK293T cells was influenced by the choice of signal peptide, cytoplasmic domain as well as transmembrane domain of the modulating protein. But of most importance is the selection of the transmembrane domain.
- the mechanisms of protein incorporation into LVs is poorly understood and the level of protein expression on a cell surface does not necessarily correlate with the virion-incorporated amount of the same protein. Hence, it was surprising to find improved incorporation of the modulating protein into the LV comprising SEQ ID NO: 1 and SEQ ID NO:2 for the binding to CD3.
- Activation of T cells is characterized by upregulation of surface markers like CD25 or CD69, as well as induction of proliferation.
- LV displaying an a-CD3 scFV comprising SEQ ID NO: 1 and SEQ ID NO:2 e g. SEQ ID NO:3 or SEQ ID NO:4
- activation markers were up-regulated and efficient proliferation was induced without the need of co-stimulatory signals.
- T cell targeted LVs were generated that comprise two targeting ligands, CD4/CD8 and CD3.
- Targeted LVs based on morbillivirus or henipavirus derived glycoproteins undergo pH- independent binding and fusion for transduction.
- the agonistic effect of anti-CD3 antibodies induce CD3 crosslinking and subsequent internalization, which was suspected to block efficient cell contact of CD3-targeted LVs.
- transduction efficiency with CD3-targeted LVs was low and could only be rescued with blocking agents, i.e. Dasatinib (A. Braun et at, 2022).
- CD3 -targeted morbillivirus pseudotyped retroviral vectors have not been described in the art.
- the pseudotyped retroviral vector particle as disclosed herein is especially well-suited for in vivo transduction of T cells in a subject but also may be used for in vitro transduction of T cells in a cell culture, not requiring additional reagents to activate as it combines activation and genetic modification in one entity.
- the retroviral vector particle for activating and transducing T cells comprises a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, comprises a nucleic acid encoding a CAR.
- the retroviral vector particle as disclosed herein may be formulated for administration to a subject by, for example, injection.
- the retroviral vector particle may be administered only once or multiple times.
- Such pharmaceutical compositions may be useful for transducing quiescent T cells, with the gene product of a desired protein such as a CAR that, if expressed in the T cell, leads to the prevention or the treatment of a particular medical condition.
- FIG 1 Schematic representation of the retroviral vector particles for activating and transducing T cells. To enable simultaneous activation and transduction of human T cells a modulating protein was designed that integrates into the retroviral envelope.
- retroviral vector particles of the disclosure which are pseudotyped with envelope proteins of Canine distemper virus (CDV), Measles virus (MV), Nipah virus (NiV) or Vesicular Stomatitis virus (VSV-G).
- CDV Canine distemper virus
- MV Measles virus
- NiV Nipah virus
- VSV-G Vesicular Stomatitis virus
- H or G the receptor binding protein
- H or G was mutated to ablate binding to the natural receptor and fused to an scFV specific for the antigen of choice.
- the modulating protein is incorporated in the viral envelope.
- TCR T cell receptor
- Treatment of T cells with these retroviral vectors results in increased proliferation, metabolism and activation marker expression.
- FIG 2 Schematic representation of the modulating protein and quantification of surface expression.
- the a-CD3 scFV is expressed under the control of a CMV promoter followed by a signal peptide (SP) to enable translocation to the cell membrane.
- SP signal peptide
- the variable domains of the scFV may be linked by a (G4S)s linker and the entire scFV may be flanked by two tags for detection. Different transmembrane and cytoplasmic domains were used to anchor the scFV in the LV envelope.
- B HEK293T cells were either left untreated (mock) or were transiently transfected with the plasmids encoding the modulating protein or GFP as positive control (GFP).
- Surface expression of the modulating protein comprising two different a-CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) and two different signal peptides (hCSF2R SP: SEQ ID NO 15; CD8 SP: SEQ ID NO 17) was determined by flow cytometry two days post transfection upon staining for a-CD3 scFV expression.
- FIG 3 Production of adapter-based retroviral vector particles and quantification of transfection efficiency.
- Second-generation LVs are generated via transient transfection of HEK293T using plasmids encoding the envelope proteins for attachment and fusion, the plasmid encoding the modulating protein (a-CD3 scFV), helper plasmids encoding gag, pol and rev and the plasmid encoding the transgene.
- the transfection efficiency can be determined by quantification of the transgene expression by flow cytometry.
- CDV H the viral glycoprotein
- a-CD3 scFV the a-CD3 scFV in presence (with a-CD3 display) or absence (without a-CD3 display) of the modulating protein compared to the negative control (untransfected).
- FIG 4 Functional titer of adapter-based retroviral vector particles.
- Retroviral vector particles were produced from transient transfected HEK293T cells. Functional titers were quantified by transducing biotinylated SupTl cells with serially diluted retroviral vector. Transduction efficiency was analyzed four days post transduction by quantification of marker positive cells. LV titers are represented as transducing units per volume (TU/mL), calculated by the ratio of transduced cells and applied LV volume.
- CDV-pseudotyped retroviral vector particles displaying the modulating protein comprising an a-CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) expressed with indicated signal peptides (hCSF2R SP : SEQ ID NO: 15; IgK SP : SEQ ID NO: 29; VSVG SP : SEQ ID NO: 30, LNGFR SP : SEQ ID NO: 31) and transmembrane domains (PDGFR TM : SEQ ID NO: 28; CD8 TM : SEQ ID NO: 7; CD28 TM : SEQ ID NO: 27; VSVG TM : SEQ ID NO: 33; LNGFR TM : SEQ ID NO: 32) is shown.
- CDV- pseudotyped retroviral particles (CDV-LV) without display of an a-CD3 scFV was used as positive control.
- CDV-pseudotyped retroviral vector particles displaying the modulating protein comprising an a-CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) expressed with indicated signal peptides (hCSF2R SP : SEQ ID NO: 15; CD8 SP : SEQ ID NO: 17) and transmembrane domains (CD8 TM : SEQ ID NO: 7; CD28 TM : SEQ ID NO: 27; VSVG TM : SEQ ID NO: 33) is shown.
- CDV-pseudotyped retroviral particles (CDV-LV) without display of an a-CD3 scFV was used as positive control.
- FIG 5 Incorporation of a-CD3 scFV within the envelope of adapter-based retroviral particles.
- GFP-encoding CDV- pseudotyped retroviral particles CDV-LV
- Unbound particles were removed by thorough washing with PBS.
- SupTl were added to the immobilized particles to allow transduction.
- the transduction efficiency was determined by quantification of GFP-expressing cells by flow cytometry seven days after transduction confirming presence of functional particles.
- CDV- pseudotyped LV without a-CD3 scFV display or wells without treatment of any LV (mock) were used as negative control.
- FIG 6 Analysis of activation marker expression, proliferation and transduction efficiency upon transduction with adapter-based retroviral particles.
- Non-activated cell-trace dye stained CD8+ T cells of two healthy donors were left untreated (mock) or were transduced with GFP-encoding CDV-pseudotyped retroviral vectors (CDV- LV) at a dose of 10 TU/cell in presence of CD8-specific adapter molecules.
- Retroviral vector particles displaying the modulating protein comprising an a-CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) expressed with different signal peptides (hCSF2R SP: SEQ ID NO: 15; IgK SP: SEQ ID NO: 29; VSVG SP: SEQ ID NO: 30; LNGFR SP: SEQ ID NO: 31) and transmembrane domains (PDGFR TM: SEQ ID NO: 28; CD8 TM: SEQ ID NO: 7; CD28 TM: SEQ ID NO: 27; VSVG TM: SEQ ID NO: 33; LNGFR TM: SEQ ID NO: 32).
- Transduction efficiency was analyzed by quantification of GFP-expressing cells using flow cytometry. Activation was quantified by staining for the activation marker CD25 and subsequent flow cytometry. T cells treated with TransactTM (mock TA) were used as positive control.
- CD25 expression of CD8+ T cells is shown five days after transduction with CDV-LV alone (CDV-LV), in presence of TransactTM (CDV-LV TA) or with CDV-LV containing a-CD3 scFV expressed with different signal peptides and transmembrane domains.
- FIG 7 Generation of CAR-T cells from non-activated PBMC with adapter-based retroviral particles.
- Non-activated PBMC of two healthy donors were left untreated (mock) or were transduced with CD20CAR encoding CDV-pseudotyped retroviral vectors (CDV-LV) at a dose of 2.5 TU/cell in presence or absence of CD8-specific adapter molecules.
- Retroviral vector particles displaying the modulating protein comprising an a-CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) expressed with different signal peptides (hCSF2R SP: SEQ ID NO: 15; IgK SP: SEQ ID NO: 29; VSVG SP: SEQ ID NO: 30) and transmembrane domains (PDGFR TM: SEQ ID NO: 28; CD8 TM: SEQ ID NO: 7; CD28 TM: SEQ ID NO: 27).
- the integrase inhibitor Raltegravir was used to confirm retroviral gene transfer. Transduction efficiency was analyzed by quantification of marker positive (LNGFR) cells using flow cytometry. In addition, the cell count of viable CD20-expressing target cells (B cells) was determined by flow cytometry.
- CD8+ T cells A Transduction efficiency of CD8+ T cells is shown seven days after transduction with CDV- LV alone (CDV-LV), in the presence of TransactTM (CDV-LV TA) or with CDV-LV containing a-CD3 scFV expressed with different signal peptides and transmembrane domains.
- PBMC activated with TransactTM (mock TA) were used as control.
- FIG 8 Surface expression of the modulating protein.
- HEK293T cells were either left untreated (mock) or were transiently transfected with the plasmids encoding the modulating protein.
- Surface expression of the modulating protein comprising an a-CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) expressed with different signal peptides (hCSF2R SP: SEQ ID NO: 15; IgK SP: SEQ ID NO: 29; VSVG SP: SEQ ID NO: 30; CD8 SP: SEQ ID NO: 17; LNGFR SP: SEQ ID NO: 31) and transmembrane domains (CD8 TM: SEQ ID NO: 7; LNGFR TM: SEQ ID NO: 32; VSVG TM: SEQ ID 33) was determined by flow cytometry two days post transfection upon staining for a- CD3 scFV expression.
- FIG 9 Functional titer of CD8-targeted retroviral vector particles.
- Retroviral vector particles were produced from transient transfected HEK293T cells. Functional titers were quantified by transducing SupTl cells with serially diluted retroviral vector. Transduction efficiency was analyzed four days post transduction by quantification of marker positive cells. LV titers are represented as transducing units per volume (TU/mL), calculated by the ratio of transduced cells and applied LV volume.
- CD8-targeted CDV-pseudotyped retroviral vector particles displaying a modulating protein comprising an a- CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) expressed with different signal peptides (hCSF2R SP: SEQ ID NO: 15; IgK SP: SEQ ID NO: 29; VSVG SP: SEQ ID NO: 30; CD8 SP: SEQ ID NO: 17; LNGFR SP: SEQ ID NO: 31) and transmembrane domains (CD8 TM: SEQ ID NO: 7; LNGFR TM: SEQ ID NO: 32; VSVG TM: SEQ ID 33) is shown.
- CD8-targeted CDV-pseudotyped retroviral particles aCD8-CDV-LV
- aCD8-CDV-LV without display of an a- CD3 scFV was used as positive control.
- FIG 10 Incorporation of a-CD3 scFV within the LV envelope of CD8-targeted retroviral particles.
- aCD8-CDV-LV CDV-pseudotyped retroviral particles
- CD8-targeted CDV-pseudotyped LV (aCD8-CDV-LV) without a-CD3 scFV display or wells without treatment of any LV (mock) were used as negative control.
- Incorporation of the modulating protein comprising an a-CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) expressed with different signal peptides (hCSF2R SP: SEQ ID NO: 15; IgK SP: SEQ ID NO: 29; VSVG SP: SEQ ID NO: 30; CD8 SP: SEQ ID NO: 17; LNGFR SP: SEQ ID NO: 31) and transmembrane domains (CD8 TM: SEQ ID NO: 7; LNGFR TM: SEQ ID NO: 32; VSVG TM: SEQ ID 33) in functional retroviral vector particles are shown.
- FIG 11 Analysis of activation marker expression, proliferation and transduction efficiency upon transduction with CD8-targeted retroviral particles.
- Non-activated cell-trace dye stained Pan T cells of three healthy donors were either left untreated (mock) or were transduced with GFP-encoding CD8-targeted CDV-pseudotyped retroviral vectors (aCD8-CDV-LV) with aCD8-CDV-LV alone (aCD8-CDV-LV), in presence of TransactTM (aCD8-CDV-LV TA) or with aCD8-CDV-LV containing a modulating protein comprising an a-CD3 scFV (CD3mut scFV: SEQ ID NO: 4; CD3 scFV: SEQ ID NO: 5) expressed with different signal peptides (hCSF2R SP: SEQ ID NO: 15; IgK SP: SEQ ID NO: 29; VSVG SP: SEQ ID NO: 30; CD8 SP: SEQ ID NO: 17; LNGFR SP: SEQ ID NO: 31) and transmembrane domains (CD8 TM:
- Transduction efficiency was analyzed by quantification of GFP-expressing cells using flow cytometry. Activation was quantified by staining for the activation marker CD25 and subsequent flow cytometry. T cells treated with TransactTM (mock TA) were used as positive control.
- CD25 expression of CD8+ and CD4+ T cells is shown two days after transduction.
- FIG 12 Incorporation of a-CD3 scFV within the envelope of measles virus, Nipah virus or VSV-G pseudotyped retroviral particles.
- a-CD3 scFV functional retroviral particles
- biotin-specific measles pseudotyped (MV-LV) or VSV-G pseudotyped (VSV-G-LV) retroviral vectors were immobilized via a N-terminal tag of the modulating protein in an ELISA plate. Unbound particles were removed by thorough washing with PBS. Subsequently SupTl (for VSV-G-LV and aCD8-NiV-LV) and biotinylated SupTl (for MV-LV) were added to the immobilized particles to allow transduction.
- the transduction efficiency was determined by quantification of GFP-expressing cells by flow cytometry six days after transduction confirming presence of functional particles.
- Retroviral vector particles without a-CD3 scFV display VSV-G-LV, aCD8-NiV-LV, MV-LV) or wells without treatment of any LV (mock) were used as negative control.
- a modulating protein comprising an a-CD3 scFV (SEQ ID NO: 4) expressed with a hCSF2R signal peptide (SP) (SEQ ID NO: 15) and CD8 transmembrane domains (TM) (SEQ ID NO: 7) with or without viral incorporation motif (gp41) (SEQ ID NO: 6) in functional retroviral vector particles are shown.
- SP hCSF2R signal peptide
- TM CD8 transmembrane domains
- gp41 viral incorporation motif
- FIG 13 Analysis of activation marker expression, proliferation and transduction efficiency upon transduction with retroviral particles of different pseudotypes.
- Non-activated cell-trace dye stained Pan T cells of three healthy donors were either left untreated (mock) or were transduced with GFP-encoding VSV-G-, biotin-specific CDV- or MV-pseudotyped retroviral vectors containing a modulating protein comprising an a-CD3- scFV (SEQ ID NO: 4) expressed with a hCSF2R signal peptide (SP) (SEQ ID NO: 15) and CD8 transmembrane domains (TM) (SEQ ID NO: 7) with or without viral incorporation motif (gp41) (SEQ ID NO: 6) at a dose of 5 TU/cell.
- a modulating protein comprising an a-CD3- scFV (SEQ ID NO: 4) expressed with a hCSF2R signal peptide (SP) (SEQ ID NO: 15) and CD8 transmembrane domains (TM) (SEQ ID NO: 7) with or without viral incorpor
- Retroviral vector particles without a-CD3 scFV display (VSV-G-LV, CDV-LV, MV-LV) were used as control. Transduction efficiency was analyzed by quantification of GFP-expressing cells using flow cytometry. Activation was quantified by staining for the activation marker CD25 and subsequent flow cytometry. T cells treated with TransactTM (mock TA) were used as positive control.
- CD25 expression of CD8+ and CD4+ T cells is shown five days after transduction.
- Retroviral vector particles without a-CD3 scFV display were used as control.
- FIG 14 Surface expression of the modulating protein with different scFV sequences.
- HEK293T cells were either left untreated (mock) or were transiently transfected with the plasmids encoding the modulating protein.
- Surface expression of the modulating protein comprising different a-CD3 scFV (SEQ ID NO: 4, SEQ ID NO: 34 and SEQ ID NO: 3), a hCSF2R2 signal peptide (SP) (SEQ ID NO: 15) and a CD8 transmembrane domain (TM) (SEQ ID NO: 7) with the viral incorporation motif (gp41) (SEQ ID NO: 6) was determined by flow cytometry two days post transfection upon staining for a-CD3 scFV expression.
- FIG 15 Functional titer of adapter-based and CD8-targeted retroviral vector particles displaying a modulation protein with varying scFV sequences.
- Retroviral vector particles displaying the modulating protein comprising different a-CD3 scFV (SEQ ID NO: 4, SEQ ID NO: 34 and SEQ ID NO: 3), a hCSF2R2 signal peptide (SP) (SEQ ID NO: 15) and a CD8 transmembrane domain (TM) (SEQ ID NO: 7) with the viral incorporation motif (gp41) (SEQ ID NO: 6) were produced from transient transfected HEK293T cells. Functional titers were quantified by transducing SupTl (CD 8 -targeted) and biotinylated SupTl (adapter-based) cells with serially diluted retroviral vector.
- a-CD3 scFV SEQ ID NO: 4, SEQ ID NO: 34 and SEQ ID NO: 3
- SP hCSF2R2 signal peptide
- TM CD8 transmembrane domain
- gp41 the viral incorporation motif
- LV titers are represented as transducing units per volume (TU/mL), calculated by the ratio of transduced cells and applied LV volume.
- Adapter-based (CDV-LV) and CD8-targeted CDV-pseudotyped retroviral particles (aCD8-CDV-LV) without display of an a-CD3 scFV were used as positive control.
- CDV- LV concentrated adapter-based CDV-pseudotyped retroviral vector particles
- FIG 16 Analysis of activation marker expression, proliferation and transduction efficiency upon transduction with retroviral particles displaying a modulation protein with varying scFV sequences.
- Non-activated cell-trace dye stained CD8+ T cells of four healthy donors were either left untreated (mock) or were transduced with GFP-encoding CD8-targeted CDV-pseudotyped retroviral vectors (aCD8-CDV-LV) or biotin-specific CDV-pseudotyped retroviral vector (CDV-LV) in presence (+) or absence (-) of CD8-sepcific adapter molecules at a dose of 5 TU/cell.
- Transduction with retroviral vector particles without modulating protein and with modulating protein comprising different a-CD3 scFV (SEQ ID NO: 4, SEQ ID NO: 34 and SEQ ID NO: 3), a hCSF2R2 signal peptide (SP) (SEQ ID NO: 15) and a CD8 transmembrane domain (TM) (SEQ ID NO: 7) with the viral incorporation motif (gp41) (SEQ ID NO: 6) was compared.
- Transduction efficiency was analyzed by quantification of GFP-expressing cells using flow cytometry. Activation was quantified by staining for the activation marker CD25 and subsequent flow cytometry.
- CD25 expression of CD8+ T cells is shown two days after transduction with aCD8-CDV- LV alone(aCD8-CDV-LV) or with aCD8-CDV-LV containing modulating proteins.
- CD25 expression of CD8+ T cells is shown two days after transduction with CDV-LV alone(CDV-LV) or with CDV-LV containing modulating proteins.
- FIG 17 In vivo generation of CD20 CAR-T cells with adapter-based retroviral vector particles. NSG mice were transplanted with human PBMC and left untreated or were i.p. injected with CD8-specific adapter molecules the next day.
- biotin-specific CDV-LVs encoding a CD20CAR with or without modulating protein (hCSF2R signal peptide (SP) SEQ ID NO: 15, a-CD3 scFV SEQ ID NO: 4; CD8 transmembrane domain (TM) SEQ ID NO: 7; with the viral incorporation motif (gp41) (SEQ ID NO: 6) were i.v. injected.
- Blood samples were collected 5 and 12 days post transduction. On day 15 post transduction, the mice were sacrificed and blood samples, bone marrow and spleen samples were analyzed for transduced T cells. Transduction efficiency was analyzed by quantification of marker positive (LNGFR) cells using flow cytometry.
- LNGFR marker positive
- CD4+ and CD8+ T cells isolated from blood, spleen and bone marrow on day 15 post LV injection.
- FIG 18 CD3 -targeted CDV-LVs.
- CDV pseudotyped retroviral vector particles targeted against CD3 were produced from transient transfected HEK293T cells. Functional titers were quantified by transducing Jurkat cells with serially diluted retroviral vector. CD4- targeted CDV-LVs (aCD4-CDV-LV) were used as positive control. Transduction efficiency was analyzed four days post transduction by quantification of marker positive cells. Exemplary dot plots of two retroviral vector particle dilutions are shown.
- the present invention provides a pseudotyped retroviral vector particle for activating and transducing T cells, wherein said retroviral vector particle comprises a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, wherein said retroviral vector particle comprises at least one nucleic acid sequence encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- Said transgene may be a CAR or a TCR.
- Said modulating protein may comprise a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 (VH) and SEQ ID NO:2 (VL), wherein the order of sequence is from N-terminus to C-terminus SEQ ID NO: 1 (VH) - SEQ ID NO:2 (VL).
- Said modulating protein may comprise a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain is a scFV specific for the antigen CD3 and comprises SEQ ID NO: 1 and SEQ ID NO:2.
- Said modulating protein may comprise a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain is a scFV specific for the antigen CD3 and comprises SEQ ID NO:3 or SEQ ID NO:4.
- the nucleic acid molecule encoding a transgene may be a nucleic acid molecule encoding a chimeric antigen receptor.
- a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, preferentially in the order of SEQ ID NO: 1 - SEQ ID NO:2,
- retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- Said pseudotyped retroviral vector particle as disclosed herein, wherein said cytoplasmic domain of said modulating protein comprises a viral incorporation motif.
- transmembrane domain of said modulating protein may be e.g. the transmembrane domain of CD8, CD28 or PDGFR.
- transmembrane domain of said modulating protein may be the transmembrane domain of CD8 comprising SEQ ID N0:7.
- transmembrane domain of said modulating protein may be the transmembrane domain of CD28 comprising SEQ ID NO:27.
- transmembrane domain of said modulating protein may be the transmembrane domain of PDGFR comprising SEQ ID NO:28.
- said signal peptide of said modulating protein may be a hCSF2R signal peptide, preferentially comprising SEQ ID NO:15, may be a CD8 signal peptide, preferentially comprising SEQ ID NO:17, may be a IgK signal peptide, preferentially comprising SEQ ID NO:29, or may be a VSV-G signal peptide , preferentially comprising SEQ ID NO:30, or may be a LNGFR signal peptide , preferentially comprising SEQ ID NO:31.
- envelope protein with antigen-binding activity wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a target antigen expressed on the surface of a T cell, and wherein said envelope protein is protein H of a virus of the Paramyxoviridae family, b) an envelope protein with fusion activity of a virus of the Paramyxoviridae family, c) said modulating protein wherein said target antigen expressed on the surface of said T cell is not CD3, and wherein said Paramyxoviridae virus is a virus of the morbillivirus genus, or
- Said polypeptide that specifically may bind a target antigen expressed on the surface of a T cell may be a polypeptide comprising an antigen binding domain specific for a target antigen expressed on the surface of a T cell.
- said retroviral vector particle of B) said protein H of a virus of the Paramyxoviridae family lacks at least one part of the cytoplasmic region of said protein H, and wherein said envelope protein with fusion activity lacks at least one part of the cytoplasmic region of said envelope protein; or wherein in said retroviral vector particle of C) said protein G of a virus of the Paramyxoviridae family lacks at least one part of the cytoplasmic region of said protein G, and wherein said envelope protein with fusion activity lacks at least one part of the cytoplasmic region of said envelope protein.
- Said pseudotyped retroviral vector particle as disclosed herein, wherein said polypeptide that specifically binds to said target antigen expressed on the surface of a T cell is specific for the antigen CD4, or wherein said polypeptide that specifically binds to said target antigen expressed on the surface of a T cell is specific for the antigen CD8.
- Said pseudotyped retroviral vector particle as disclosed herein, wherein said polypeptide comprising an antigen binding domain specific for said target antigen expressed on the surface of a T cell is specific for the antigen CD4 and comprises SEQ ID NO:8 (VL) and SEQ ID NO:9 (VH), preferentially in the order of sequence from N to C-terminus VL-VH, or wherein said polypeptide comprising an antigen binding domain specific for a target antigen expressed on the surface of a T cell is specific for the antigen CD8 and comprises SEQ ID NO: 10 (VL) and SEQ ID NO: 11 (VH), preferentially in the order of sequence from N to C-terminus VL-VH.
- Said pseudotyped retroviral vector particle as disclosed herein, wherein said polypeptide comprising an antigen binding domain specific for said target antigen expressed on the surface of a T cell is specific for the antigen CD4 and comprises SEQ ID NO:25 or wherein said polypeptide comprising an antigen binding domain specific for a target antigen expressed on the surface of a T cell is specific for the antigen CD8 and comprises SEQ ID NO:26.
- said virus of the morbillivirus genus is a measles virus or the Edmonston strain of measles virus, or is a canine distemper virus (CDV) or the5804P strain or the Madrid/16 strain of the CDV.
- CDV canine distemper virus
- Said pseudotyped retroviral vector particle as disclosed herein, wherein said at least one nucleic acid sequence encoding a transgene is a chimeric antigen receptor (CAR) specific for an antigen expressed on the surface of a second target cell such as a cancer cell, or wherein said at least one nucleic acid sequence encoding a transgene is a T cell receptor (TCR) with specificity for an antigen of a second target cell such as a cancer cell.
- CAR chimeric antigen receptor
- TCR T cell receptor
- pseudotyped retroviral vector particle as disclosed herein, wherein said pseudotyped retroviral vector particle comprises at least a second nucleic acid sequence encoding at least a second transgene such as a cytokine such as IL7, IL 15, or IL21 or a cytokine receptor, such as CD127.
- a second transgene such as a cytokine such as IL7, IL 15, or IL21
- a cytokine receptor such as CD127.
- the present invention provides a pseudotyped retroviral vector particle as disclosed herein for use in immunotherapy.
- the present invention provides a pseudotyped retroviral vector particle as disclosed herein for use in treatment of a disease in a subject in need thereof.
- the disease may be a cancer, an autoimmune disease or an infectious disease.
- the transgene may be a CAR specific for a tumor associated antigen (TAA) or a tumor specific antigen (TSA) expressed on the surface of a cancer cell.
- TAA tumor associated antigen
- TSA tumor specific antigen
- the present invention provides a composition
- a composition comprising i) a pseudotyped retroviral vector particle for activating and transducing T cells comprising a) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a tag of a tagged polypeptide, b) an envelope protein with fusion activity, c) a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, preferentially in the order from N-terminus to C-terminus of SEQ ID NO: 1 - SEQ ID NO:2, ii) said tagged polypeptide, wherein said tagged polypeptide binds specifically to a target antigen
- Said modulating protein may comprise a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain is a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, preferentially in the order from N-terminus to C-terminus of SEQ ID NO: 1 - SEQ ID NO:2.
- Said modulating protein may comprise a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain is a scFV specific for the antigen CD3 comprises SEQ ID NO:3 or SEQ ID NON.
- Said polypeptide that specifically binds to a tag of a tagged polypeptide may be a polypeptide comprising an antigen binding domain specific for a tag of a tagged polypeptide.
- composition as disclosed herein, wherein said modulating protein comprises (from N- terminus to C-terminus):
- composition as disclosed herein, wherein said modulating protein comprises (from N- terminus to C-terminus):
- a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, preferentially in the order of SEQ ID NO: 1 - SEQ ID NO:2,
- retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- composition as disclosed herein, wherein said viral incorporation motif is the incorporation motif of gp41 (ENV of HIV), preferentially comprising SEQ ID NO:6.
- transmembrane domain of said modulating protein may be e.g. the transmembrane domain of CD8, CD28 or PDGFR.
- said signal peptide of said modulating protein may be a hCSF2R signal peptide, preferentially comprising SEQ ID NO: 15, may be a CD8 signal peptide, preferentially comprising SEQ ID NO: 17, may be a IgK signal peptide, preferentially comprising SEQ ID NO:29, or may be a VSV-G signal peptide , preferentially comprising SEQ ID NO:30, or may be a LNGFR signal peptide , preferentially comprising SEQ ID N0:31.
- said pseudotyped retroviral vector particle comprises: a) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a tag of a tagged polypeptide, and wherein said envelope protein is protein H of a virus of the Paramyxoviridae family, b) an envelope protein with fusion activity of a virus of the Paramyxoviridae family, c) a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, ii) said tagged polypeptide, wherein said tagged polypeptide binds specifically to a target antigen expressed on the surface of a T cell, wherein said target antigen
- said pseudotyped retroviral vector particle comprises: a) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a tag of a tagged polypeptide, and wherein said envelope protein is protein G of a virus from the Paramyxoviridae family, b) an envelope protein with fusion activity of a virus of the Paramyxoviridae family, c) a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, ii) said tagged polypeptide, wherein said tagged polypeptide binds specifically to a target antigen expressed on the surface of a T cell, wherein said antigen is
- composition as disclosed herein, wherein in said retroviral vector particle of A) said protein H of a virus of the Paramyxoviridae family lacks at least one part of the cytoplasmic region of said protein H, and wherein said envelope protein with fusion activity lacks at least one part of the cytoplasmic region of said envelope protein; or wherein in said retroviral vector particle of B) said protein G of a virus of the Paramyxoviridae family lacks at least one part of the cytoplasmic region of said protein H, and wherein said envelope protein with fusion activity lacks at least one part of the cytoplasmic region of said envelope protein.
- Said virus of the morbillivirus genus may be a measles virus or the Edmonston strain of measles virus, or may be a canine distemper virus (CDV) or the 5804P strain or the Madrid/16 strain of the CDV.
- CDV canine distemper virus
- Said virus of the henpivirus genus may be a Nipah virus, or is a Hendravirus.
- Said virus of the morbillivirus genus may be a CDV, and wherein said protein H is aN-terminal truncated protein H, and wherein said truncated protein H is HcA21-A32, and wherein said truncated protein H comprises the amino acid substitutions of positions D526A, I527S, S528A, R529A, Y547A and T548A as compared to the unmodified protein H set forth in SEQ ID NO: 12, and wherein said protein with fusion activity (protein F) is a C-terminal truncated protein F (as compared to the unmodified protein F set forth in SEQ ID NO: 13), wherein said truncated protein is FcA26-FcA30, preferentially FcA30, and wherein the N-terminal signal peptide sequence comprising SEQ ID NO: 14 is present in said truncated protein F.
- protein F protein with fusion activity
- composition as disclosed herein, wherein said tagged polypeptide of A) or B) specific for said target antigen expressed on the surface of a T cell is specific for the antigen CD4 or for the antigen CD8.
- Said composition as disclosed herein, wherein said tagged polypeptide of A) and/or B) specific for said target antigen expressed on the surface of a T cell is specific for the antigen CD4 and comprises SEQ ID NO:8 (VL) and SEQ ID NO:9 VH), , or is specific for the antigen CD8 and comprises SEQ ID NO: 10 (VL) and SEQ ID NO: 11 (VH).
- composition wherein said at least one nucleic acid sequence encoding a transgene is a chimeric antigen receptor (CAR) specific for an antigen expressed on the surface of a second target cell such as a cancer cell, or wherein said at least one nucleic acid encoding a transgene is a T cell receptor (TCR) with specificity for an antigen of a second target cell such as a cancer cell.
- CAR chimeric antigen receptor
- TCR T cell receptor
- the present invention provides a composition as disclosed herein for use in immunotherapy.
- the present invention provides a composition as disclosed herein for use in treatment of a disease in a subject in need thereof.
- the disease may be a cancer, an autoimmune disease or an infectious disease.
- the transgene may be a CAR specific for a tumor associated antigen (TAA) or a tumor specific antigen (TSA) expressed on the surface of a cancer cell.
- TAA tumor associated antigen
- TSA tumor specific antigen
- the present invention provides a combination (or a combinations of compositions) comprising i) (a first composition comprising) a pseudotyped retroviral vector particle for activating and transducing T cells comprising a) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a tag of a tagged polypeptide, b) an envelope protein with fusion activity, c) a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, preferentially in the order from N-terminus to C-terminus of SEQ ID NO: 1 - SEQ ID NO:2, ii) (a second composition comprising) said tagged
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising a) a pseudotyped retroviral vector particle for activating and transducing T cells, wherein said retroviral vector particle comprises a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, wherein said retroviral vector particle comprises at least one nucleic acid sequence encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle; and optionally b) a pharmaceutically acceptable carrier.
- Pharmaceutically acceptable carriers, diluents or excipients may comprise buffers such as neutral buffered saline, phosphate buffered saline and the like; carbohydrates such as glucose, mannose, sucrose or dextrans, mannitol; proteins; polypeptides or amino acids such as glycine; antioxidants; chelating agents such as EDTA or glutathione; adjuvants (e.g., aluminum hydroxide); and preservatives.
- Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is well known in the art.
- Said pseudotyped retroviral vector particle may comprise a polypeptide comprising an antigen binding domain specific for a target antigen expressed on the surface of a T cell as disclosed herein, and wherein said polypeptide is specific for the antigen CD4 or for the antigen CD8.
- the present invention provides a pharmaceutical composition
- a pseudotyped retroviral vector particle for activating and transducing T cells comprising a) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a tag of a tagged polypeptide, b) an envelope protein with fusion activity, c) a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, ii) said tagged polypeptide, wherein said tagged polypeptide binds specifically to a target antigen expressed on the surface of a T cell, wherein said target antigen is not CD3, and wherein said retroviral vector particle comprises at least
- Said tagged polypeptide may comprise an antigen binding domain specific for a target antigen expressed on the surface of a T cell as disclosed herein, and wherein said polypeptide is specific for the antigen CD4 or for the antigen CD8.
- the present invention provides an in vitro method for the generation of a sample of genetically modified T cells comprising a) providing a sample comprising T cells b) genetic modification of the T cells by targeted transduction with a pseudotyped retroviral vector particle, wherein said retroviral vector particle comprises a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, wherein said retroviral vector particle comprises at least one nucleic acid sequence encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle; thereby generating a sample of genetically modified T cells.
- retroviral vector particle comprises
- envelope protein with antigen-binding activity wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a target antigen expressed on the surface of a T cell, and wherein said envelope protein is protein H of a virus of the Paramyxoviridae family, b) an envelope protein with fusion activity of a virus of the Paramyxoviridae family, c) said modulating protein, wherein said target antigen expressed on the surface of said T cell is not CD3, and wherein said Paramyxoviridae virus is a virus of the morbillivirus genus, or
- said retroviral vector particle comprises a) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a tag of a tagged polypeptide, b) an envelope protein with fusion activity, c) a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, and wherein said pseudotyped retroviral vector is in combination with said tagged polypeptide, wherein said tagged polypeptide binds specifically to a target antigen expressed on the surface of a T cell, wherein said target antigen is not CD3.
- said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused
- the present invention provides an in vivo method for treating a disease in a subject in need thereof comprising administering to said subject a pseudotyped retroviral vector particle, wherein said retroviral vector particle comprises a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, wherein said retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- Said pseudotyped retroviral vector particle may comprise a polypeptide that specifically binds to a target antigen expressed on the surface of a T cell as disclosed herein, and wherein said polypeptide is specific for the antigen CD4 or for the antigen CD8.
- the present invention provides an in vivo method for treating a disease in a subject in need thereof comprising i) administering to said subject a pseudotyped retroviral vector particle comprising a) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a tag of a tagged polypeptide, b) an envelope protein with fusion activity, c) a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2, and ii) administering to said subject said tagged polypeptide, wherein said tagged polypeptide binds specifically to a target antigen expressed on the surface of a T cell, wherein said target antigen is
- the administration of the pseudotyped retroviral vector particle to said subject may be performed before, simultaneously or after the administration of the tagged polypeptide.
- the present invention provides a plasmid vector system (a kit) for generation of a pseudotyped retroviral vector particle for activating and transducing T cells comprising a) a nucleic acid sequence encoding the envelope for a pseudotyped retroviral vector particle, b) a nucleic acid sequence encoding a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID N0:2, c) a nucleic acid sequence encoding gag/pol of HIV- 1, d) a nucleic acid sequence encoding a transgene, wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- the present invention provides a plasmid vector system (a kit) for generation of a pseudotyped retroviral vector particle for activating and transducing T cells comprising a) a nucleic acid sequence encoding the envelope for a pseudotyped retroviral vector particle, wherein said retroviral vector particle comprises i) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a tag of a tagged polypeptide, ii) an envelope protein with fusion activity, b) a nucleic acid sequence encoding a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID N0:2, c) a nucleic acid sequence
- the present invention provides a plasmid vector system (a kit) for generation of a pseudotyped retroviral vector particle for activating and transducing T cells comprising a) a nucleic acid sequence encoding the envelope for a pseudotyped retroviral vector particle, wherein said retroviral vector particle comprises i) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a target antigen expressed on the surface of a T cell, and wherein said envelope protein is protein H of a virus of the Paramyxoviridae family, ii) an envelope protein with fusion activity of a virus of the Paramyxoviridae family, b) a nucleic acid sequence encoding a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen
- the present invention provides a plasmid vector system (a kit) for generation of a pseudotyped retroviral vector particle for activating and transducing T cells comprising a) a nucleic acid sequence encoding the envelope for a pseudotyped retroviral vector particle, wherein said retroviral vector particle comprises i) an envelope protein with antigen-binding activity, wherein said envelope protein is a recombinant protein that does not interact with at least one of its native receptors and is fused at its ectodomain to a polypeptide that specifically binds to a target antigen expressed on the surface of a T cell, and wherein said envelope protein is protein G of a virus of the Paramyxoviridae family, ii) an envelope protein with fusion activity of a virus of the Paramyxoviridae family, b) a nucleic acid sequence encoding a modulating protein comprising a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen
- nucleic acid sequence encoding a pseudotyped retroviral vector particle wherein nucleic acid sequence encoding said modulating protein comprises:
- the pseudotyped retroviral vector particle for activating and transducing T cells comprises a nucleic acid sequence encoding in 5 'to 3 'order a modulating protein comprising
- a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2,
- the retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- the pseudotyped retroviral vector particle for activating and transducing T cells comprises a nucleic acid sequence encoding in 5 'to 3 'order a modulating protein comprising
- a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2,
- a cytoplasmic domain comprising an incorporation motif, preferentially the incorporation motif of gp41 of HIV-1, preferentially comprising SEQ ID NO:6 optionally a spacer, preferentially a CD8 spacer, between said functional ectodomain and said transmembrane domain, preferentially comprising SEQ ID NO: 16; wherein said retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- the pseudotyped retroviral vector particle for activating and transducing T cells comprises a nucleic acid sequence encoding in 5 'to 3 'order a modulating protein comprising
- a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2,
- a cytoplasmic domain comprising an incorporation motif, preferentially the incorporation motif of gp41 of HIV-1, preferentially comprising SEQ ID NO:6 optionally a spacer, preferentially a CD8 spacer, between said functional ectodomain and said transmembrane domain, preferentially comprising SEQ ID NO: 16; wherein said retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- the pseudotyped retroviral vector particle for activating and transducing T cells comprises a nucleic acid sequence encoding in 5 'to 3 'order a modulating protein comprising I) a signal peptide
- a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2,
- a cytoplasmic domain comprising a co-stimulatory domain, preferentially the costimulatory domain of 4-1BB, preferentially comprising the SEQ ID NO:23, and optionally a spacer between said functional ectodomain and said transmembrane domain; wherein said retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- the pseudotyped retroviral vector particle for activating and transducing T cells comprises a nucleic acid sequence encoding in 5 'to 3 'order a modulating protein comprising
- a functional ectodomain comprising an antigen binding domain specific for the antigen CD3, wherein said antigen binding domain such as a scFV specific for the antigen CD3 comprises SEQ ID NO: 1 and SEQ ID NO:2,
- a cytoplasmic domain comprising a co-stimulatory domain, preferentially the costimulatory domain of CD28, preferentially comprising the SEQ ID NO:24, and optionally a spacer between said functional ectodomain and said transmembrane domain; wherein said retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- a pseudotyped retroviral vector particle as disclosed herein may be used to transduce cells in vivo at an any effective dosage.
- the viral particle is administered to a subject in vivo by application to the tissue, the organ or to the blood circulation of a subject in need of therapy.
- the adapter (tagged polypeptide) may be administered to a subject in vivo - in addition to the viral particle - by application to the tissue, the organ or to the blood circulation of a subject in need of therapy, said administration of the said adapter may be before, simultaneous to or after the administration of said viral particle.
- the pseudotyped retroviral vector particle as disclosed herein may be administered via a route of parenteral, intravenous, intramuscular, subcutanous, intratumoral, intraperitoneal, or intralymphatic administration.
- the viral particle may be administered multiple times.
- the pseudotyped retroviral vector particle as disclosed herein may be administered intratumorally to a subject and thereby activates and transduces the T cell portion of the tumor-infiltrating lymphocytes at the tumor site.
- the pseudotyped retroviral vector particle as disclosed herein may be administered intravenously to a subject, and thereby activates and transduces the T cells in the circulatory blood system.
- the pseudotyped retroviral vector particle as disclosed herein may be administered by intra lymphnode injection to a subject, and thereby activates and transduces the T cells in the lymph node.
- the pseudotyped retroviral vector particle as disclosed herein may be administered by intra splenic injection to a subject, and thereby activates and transduces the T cells in the spleen.
- the pseudotyped retroviral vector particle as disclosed herein may also be delivered to a subject according to viral titer (TU/mL).
- the amount of the pseudotyped retroviral vector particle as disclosed herein directly injected may be determined by total TU and can vary based on both the volume that could be feasibly injected to the site and the type of tissue to be injected.
- the viral titer delivered is about 1 x 10 5 to 1 x 10 6 , about 1 x 10 5 to 1 x 10 7 , 1 x 10 5 to lx 10 7 , about 1 x 10 6 to 1 x 10 9 , about 1 x 10 7 to 1 x IO 10 , about 1 x 10 7 to 1 x 10 11 , or about 1 x 10 9 to 1 x 10 11 TU.
- the pseudotyped retroviral vector particle as disclosed herein may activate and transduce T cells that are provided in a cell culture, and thereby may activate and transduce the T cells of the cell culture.
- the transduced T cells may be expanded to a therapeutically effective amount.
- the expanded T cells may be subsequently administered to a subject in need thereof.
- the pseudotyped retroviral vector particle as disclosed herein may activate and transduce T cells that are provided in a closed system that may be an automated manufacturing system, and thereby may activate and transduce the T cells of the cell culture.
- the transduced T cells may be expanded to a therapeutically effective amount.
- the expanded T cells may be subsequently administered to a subject in need thereof.
- the pseudotyping glycoproteins of the pseudotyped retroviral vector particle as disclosed herein or functional fragments thereof may be a full-length polypeptide, a functional fragment, a homolog, or a functional variant of Human immunodeficiency virus (HIV) gpl60, Murine leukemia virus (MLV) gp70, Gibbon ape leukemia virus (GALV) gp70, Feline leukemia virus (RD114) gp70, Amphotropic retrovirus (Ampho) gp70, 10A1 MLV (10A1) gp70, Ecotropic retrovirus (Eco) gp70, Baboon ape leukemia virus (BaEV) gp70, Measles virus (MV) H and F, Nipah virus (NiV) H and F, Rabies virus (RabV) G, Mokola virus (MOKV) G, Ebola Zaire virus (EboZ) G, Lymphocytic choriomen
- HBV
- the viral envelope glycoprotein of the pseudotyped retroviral vector particle as disclosed herein may be a G protein from the Cocal vesiculovirus species or a functional fragment thereof.
- compositions of the present invention may comprise a combination of any number of the pseudotyped retroviral vector particle as disclosed herein, and optionally one or more additional pharmaceutical agents (polypeptides, polynucleotides, compounds etc.) formulated in pharmaceutically acceptable compositions for administration to a cell, tissue, organ, or a subject, either alone, or in combination with one or more other modalities of therapy.
- the one or more additional pharmaceutical agent further increases transduction efficiency of vectors.
- the pseudotyped retroviral vector particle for activating and transducing T cells comprises a second modulating protein comprising I) a signal peptide
- a functional ectodomain comprising an antigen binding domain specific for but not limited to CD28, CD137, 0X40 and ICOS
- retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- a functional ectodomain comprising a cytokine for example IL7 or IL21
- retroviral vector particle comprises at least one nucleic acid encoding a transgene, and wherein said retroviral vector particle is a lentiviral or gammaretroviral vector particle.
- compositions, methods, and respective component s) thereof that are essential to the method or composition, yet open to the inclusion of unspecified elements, whether essential or not.
- Rhabdoviridae is a family of negative-strand RNA viruses in the order Mononegavirales.
- the Rhabodivirdae family comprises to date 17 known taxonomic groups of which the Vesiculovirus is the best studies one.
- Indiana vesiculovirus (formerly Vesicular stomatitis Indiana virus (VSIV or VSV)) is the prototypic member of the genus Vesiculovirus. Its RNA genome encodes codes five major proteins: G protein (G), large protein (L), phosphoprotein (P), matrix protein (M) and nucleoprotein (N).
- the envelope glycoprotein (G) mediates viral attachment by binding to the LDL receptor (LDLR) and induces rapid endocytosis followed by fusion of the viral membrane with the endosomal membrane.
- LDL receptor LDL receptor
- protein G of the Vesicular stomatitis virus (VSV) of the Rhabdoviridae family is often used in retroviral vectors for pseudotyping.
- Cocal vesiculovirus that also belongs to the genus of vesiculoviruses in the family of rhabdoviridae, is the cause of vesicular stomatitis in mammals. It is most closely related to the Vesicular stomatitis Indiana virus, but serologically different. While they also provide a broad tropism, lentiviral vectors pseudotyped with Cocal glycoproteins were shown to be resistant against inactivation by human serum.
- Retroviridae is a virus family with a single-stranded, diploid, positive-sense RNA genome that is reverse-transcribed into a DNA intermediate that is then incorporated into the host cell genome.
- Aetrow'rzt/ae-derived viruses are enveloped particles with a diameter of 80-120 nm.
- (Retro- /lenti- /gammaretro-) viral vectors are replication-deficient viral particles that are derived from the corresponding virus family. They contain Gag and Pol proteins, a singlestranded RNA genome and are usually pseudotyped with heterologous envelope proteins derived from other viruses.
- the RNA genome of said viral vectors do not contain any viral gene to produce viral progeny, but psi elements and LTRs that are required for efficient packing and reverse transcription into DNA.
- the DNA intermediate may contain a gene of interest under the control of a suitable promoter, for example, the CMV promoter and the gene of interest is expressed upon integration of said DNA into the genome of the host cell.
- a suitable promoter for example, the CMV promoter
- the process of entering the host cell, delivering the RNA genome, integration and expression of the gene of interest is called transduction.
- transduction The minimal requirements of a gammaretrovirus or lentivirus based viral vector has been well-described in the art.
- ID-RVs integrase-deficient retroviral vectors
- ID-RVs are derived from conventional retroviral vectors but contain no or a mutated form of the retroviral integrase.
- ID- RVs are useful tools to express the gene of interest transiently.
- the definition of retroviral vectors and transduction also extents the integration-deficient retroviral vectors and its application.
- Lentivirus is a genus of Retroviridae that cause chronic and deadly diseases characterized by long incubation periods, in the human and other mammalian species.
- the best-known lentivirus is the Human Immunodeficiency Virus (HIV), which can efficiently infect nondividing cells, so lentiviral derived retroviral vectors are one of the most efficient methods of gene delivery.
- HIV Human Immunodeficiency Virus
- Gammaretroviridae is a genus of the Retroviridae family. Representative species are the murine leukemia virus (MLV) and the feline leukemia virus (FLV).
- MLV murine leukemia virus
- FLV feline leukemia virus
- Paramyxoviridae is a family of viruses in the order of Mononegavirales. There are currently 49 species in this family, divided among 7 genera. Diseases associated with this virus family include measles, mumps, and respiratory tract infections. Members of this virus family are enveloped viruses with a non-segmented, negative-strand RNA genome of about 16 kb. Two membrane proteins with two distinct functions appear as spikes on the virion surface. The H/HN/G proteins mediate binding to the receptor at the cell surface.
- virus envelope protein(s) that have antigen binding activity refers to protein(s) on the viral envelope that are responsible for binding to complementary receptors or antigens on the cell membrane of a target cell.
- virus envelope protein(s) that have antigen binding activity are virus envelope protein(s) that have antigen binding activity.
- H/HN/G proteins Upon binding the H/HN/G proteins change their conformation that induces a process called fusion helper function, leading to subsequent conformational changes within the F protein that is mediating the fusion of the viral and cellular membrane.
- the capsid and viral genome may now enter and infect or transduce the host cell.
- virus envelope proteins(s) that have fusion activity refers to protein(s) that initiate fusion of viral and cellular membrane.
- Paramyxoviridae F proteins refer to virus envelope protein(s) that have fusion activity.
- ectodomain“ or “extracellular part/domain” as used herein refers to a domain of a membrane protein that extends into the extracellular space (the space outside a cell or virion).
- activation refers to inducing physiological changes of a cell that increase target cell function, proliferation and/or differentiation.
- pseudotyping or “pseudotyped” as used herein refers to a viral vector particle bearing envelope glycoproteins derived from other viruses having envelopes.
- the host range of the lentiviral vectors or viral vector particles of the present invention can thus be expanded or altered depending on the type of cell surface receptor used by the glycoprotein.
- modulating protein of a retroviral vector particle refers to a protein that may modulate, e.g. activate a T cell. This modulation or specifically this activation may be due to the binding of an antigen binding domain of said modulating protein to stimulatory or co-stimulatory receptors of T cells, e.g. CD3 expressed on the surface of T cells.
- the gag, pol and env proteins needed to assemble the vector particle are provided in trans by means of a packaging cell line, for example, HEK293T. This is usually accomplished by transfection of the packaging cell line with one or more plasmids containing the gag, pol and env genes.
- the env gene originally derived from the same retrovirus as the gag and pol genes and as the RNA molecule or expression vector, is exchanged for the envelope protein(s) of a different enveloped virus.
- the F and H or HN or G protein of Paramyxoviridae is used.
- an exemplary pseudotyped vector particle based on the HIV-1 retrovirus comprises the (1) HIV-1 Gag and Pol proteins, (2) an RNA molecule derived from the HIV-1 genome that may be used to generate a retroviral vector particle based on the HIV-1 genome lacking the gag, env, pol, tat, vif, vpr, vpu and nef genes, but still comprising the LTRs, the psi element and a CMV promoter followed by the gene to be transduced, for example, a gene for the GFP protein, and (3) the F and H proteins of measles virus, for example, in a truncated form.
- native receptor or “originally receptor” as used herein may be used interchangeably and refer to the receptor or antigen expressed on the cell surface of a cell that is bound by the naturally occurring virus envelope protein with antigen (receptor) binding activity.
- the native measles virus receptors are SLAM, nectin-4 and CD46.
- the native canine distemper virus receptors are SLAM and nectin-4.
- Nipahvirus envelope proteins use ephrin-B2 and ephrin-B3 as receptors for entry.
- one envelope protein with antigen-binding activity that does not interact with at least one of its native receptor(s) means that said protein has reduced or ablated interaction with at least one receptor of a cell that is normally targeted by the virus having said protein as described elsewhere herein.
- Reduced interaction means that said truncated and/or mutated protein interacts with said at least one native receptor at least 50 % less efficient, at least 60 % less efficient, at least 70 % less efficient, at least 80 % less efficient, at least 90 % less efficient, at least 95 % less efficient, at least 99 % less efficient compared to the nonmutated protein.
- said protein does not interact anymore with said at least one of its native receptors.
- the interaction may be the binding of these two molecules to each other.
- the less efficient interaction may be a reduced affinity of said protein to its native receptor.
- Said envelope protein with antigen-binding activity may have more than one native receptors, then the reduction or ablation of interaction of one of these native receptors of said protein results in a reduced tropism of the vector particle.
- the more interactions of said protein with its native receptors are inhibited by mutation the more effective is the reduction of tropism of the vector particle.
- the retroviral vector particle as disclosed herein, e.g. when a native receptor is not expressed on any cell (target cells and non-target cells) in the environment of target cells that are intended to be transduced.
- an envelope protein with antigen-binding activity has more than 2 native receptors, e.g. 3 native receptors, then preferentially said protein does not interact with the majority of its native receptors, e.g. 2 from 3.
- the envelope protein with antigen-binding activity does not interact with all of its native receptors.
- tropism refers to the host range or specificity of a virus or retroviral vector.
- envelope protein with antigen-binding activity that is fused at its ectodomain to a polypeptide comprising an antigen binding domain defines the host range of the retroviral vector.
- the tagged polypeptide specific for antigen expressed on target cells defines the host range of the retroviral vector.
- target cell refers to a cell which expresses an antigen (a marker) on its cell surface that should be recognized (bound) by the pseudotyped retroviral vector particle as disclosed herein or the tagged polypeptide of the adaptable system as disclosed herein.
- the target cell may be T cell, a primary T cell or a cell line derived from a T cell.
- the target cell may be a mammalian cell such as a murine cell, preferentially the target cell is a human cell.
- the truncated protein H fused to the polypeptide comprising an antigen binding domain specific for an antigen expressed on a target cell or for a tag of a tagged polypeptide as disclosed herein must have mutations that generally reduce or ablate productive interactions with its native receptors. Such mutations are well-known in the art. A mutation that ablates interaction of measles H protein with CD46 is e.g.
- the following introduction of mutations ablates productive interaction of the measles H protein with CD46 and SLAM, respectively: Y481A R533A.
- the Hmut protein also includes the mutations S548L and F549S, which lead to a more complete ablation of residual infectivity via CD46.
- the mutation of the residues V451 and Y529 ablates productive interaction with CD46 and SLAM.
- a mutation that ablates interaction of canine distemper virus H protein with SLAM and Nectin 4 may be e.g. the point mutation at position D526, 1527, S528, R529; Y547 and T548 according to amino acid number of SEQ ID NO: 12, wherein amino these amino acids are replaced with another amino acid and this mutation prevents or assists in preventing interaction of the H protein with SLAM and Nectin-4 (Bah et al (2020); von Messing et al (2005)).
- E501, W504, Q530, E533 were either single mutated or in combination.
- the combined mutation of E501A, W504A, Q530A, E533A showed completely ablated receptor binding ability for both receptors ephrin- B2 and ephrin-B3.
- a pseudotyped retroviral vector particle "derived from”, for example, HIV-1, as used in the present invention, refers to a particle in which the genetic information for the RNA and/or the Gag and Pol proteins comprised by the vector particle originate from said retrovirus, in the above case, HIV-1.
- the original retroviral genome can comprise mutations, such as deletions, frame shift mutations and insertions.
- cytoplasmic domain refers to the portion of the respective protein that is adjacent to the transmembrane domain of the protein and, if the protein is inserted into the membrane under physiological conditions, extends into the cytoplasm or in case of viral particles reaching into the intravirion side.
- cytoplasmic domain refers to the portion of the respective protein that is adjacent to the transmembrane domain of the protein and, if the protein is inserted into the membrane under physiological conditions, extends into the cytoplasm or in case of viral particles reaching into the intravirion side.
- envelope proteins with antigen-binding function are characterized to date as type II membrane proteins, meaning that the cytoplasmic domain is located at the N-terminus of the envelope protein.
- the cytoplasmic domain refers to the portion of the protein that reaches into the intravirion side.
- the portion of the protein that reaches into the intravirion side may comprise at least 1, at least 2, at least 3 or at least 4 amino acids.
- the cytoplasmic domain of the modulating protein may comprise a viral incorporation motif. Said incorporation motif may be the incorporation motif of gp41.
- truncated refers to a deletion of amino acid residues of the designated protein. It is clear to the skilled person that a protein is encoded by a nucleic acid. Thus, “truncated” also refers to the corresponding coding nucleic acids in a nucleic acid molecule that codes for a given "truncated” protein.
- truncated H designates the Paramyxoviridae , preferably measles H protein, Nipah G protein and Nipah or measles F proteins, respectively, whose cytoplasmic portion has been partly or completely truncated, i.e. amino acid residues (or coding nucleic acids of the corresponding nucleic acid molecule encoding the protein) have been deleted.
- the cytoplasmic portion of the F protein (termed Fc) is located at the C-terminus of the protein.
- FcA30 For all envelope proteins with the cytoplasmic portion located at the C-terminus one begins counting from the C-terminal end of the protein when ascertaining the desired sequence.
- FcA30 would refer to an F protein having deleted the last 30 amino acids counting from the C-terminal end of the protein F set forth in SEQ ID NO: 18. Consequently, FcA30 would refer to an F protein having a cytoplasmic domain with the amino acid sequence SEQ ID NO: 19.
- FcA30 would refer to an F protein having deleted the last 30 amino acids counting from the C-terminal end of the protein F set forth in SEQ ID NO: 13. Consequently, for the F protein derived from Canine distemper virus strain 5840P FcA30 would refer to an F protein having a cytoplasmic domain comprising amino acid sequence of SEQ ID NO:20.
- the cytoplasmic portion of the H, HN or G protein is located at the N-terminus (termed He).
- truncated protein H is HcA21-A32” in the context of the protein H of a CDV as used herein refers to any truncated protein H of the CDV having deleted the first 2 Ito 32 amino acids counting from the N-terminal end of the protein H set forth in SEQ ID NO: 12 (the unmodified protein H): individually said truncated protein H may be:
- the cytoplasmic domain of HcA21 comprises the amino acids as in SEQ ID NO:21. Accordingly, the H protein derived from Canine Distemper virus strain 5840P the cytoplasmic domain of HcA30 comprises the amino acids as in SEQ ID NO:22.
- the cytoplasmic domain of the measles F protein can be truncated to comprise at least 1 positively charged amino acid residue and the cytoplasmic portion of the H protein can be truncated to comprise at least 9 consecutive amino acid residues of the C-terminal cytoplasmic portion of the H protein plus an additional methionine at the N- terminus.
- a further truncation of the cytoplasmic portion of the H protein is expected to be feasible, if the H protein is truncated to allow efficient pseudotyping and still has fusion support function.
- Modifications that allow truncation for efficient pseudotyping may be combined with modifications that ablate native receptor binding function.
- the proteins of the present invention further include functional homologs.
- a protein is considered a functional homolog of another protein for a particular function, if the homolog has a similar function as the original protein.
- the homolog can be, for example, a fragment of the protein, or a substitution, addition, or deletion mutant of the protein.
- Determining whether two amino acid sequences are substantially homologous is typically based on FASTA searches.
- the amino acid sequence of a first protein is considered to be homologous to that of a second protein if the amino acid sequence of the first protein shares at least about 70 % amino acid sequence identity, preferably at least about 80% identity, and more preferably at least about 85 %, 90 %, 95 % or 99 % identity, with the sequence of the second protein.
- Psi positive and psi negative refer to a nucleic acid molecule where the retroviral psi element is present and absent, respectively.
- the psi element is a cis-acting signal located near the 5’ end of the retroviral genome and designates a packaging signal, which is of importance during assembly of the viruses and leads to the incorporation of the viral RNA into the viral core.
- a psi negative RNA does not comprise the retroviral psi element and consequently will not be assembled into a vector particle of the present invention; in contrast, a psi positive RNA that does comprise said psi element will be effectively assembled into the vector particle.
- a (target) cell or "cell (surface) marker”, as used in the present invention, refers to a molecule present on the surface of a cell, preferentially on a target cell.
- molecules can be, inter alia, peptides or proteins that may comprise sugar chains or lipids, clusters of differentiation (CDs), antibodies or receptors. Since not all populations of cells express the same cell markers, a cell marker can thus be used to identify, select or isolate a given population of cells expressing a specific cell marker.
- CD4 is a cell marker expressed by T helper cells, regulatory T cells, and monocytes.
- T helper cells, regulatory T cells, and monocytes can be identified, selected or otherwise isolated, inter alia by a FACS cell sorter, by means of the CD4 cell marker.
- tagged polypeptide that is specific for binding to a target antigen expressed on the surface of a T cell as used herein refers to a polypeptide that has bound thereto directly or indirectly at least one additional component, i.e. the tag.
- the tagged polypeptide as used herein is able to bind a target antigen expressed on T cells, regularly CD4 or CD8.
- the polypeptide may be an antibody or antigen binding fragment thereof that binds to said antigen expressed on the surface of a target cell.
- the polypeptide of the tagged polypeptide alternatively may be a cytokine or a growth factor or another soluble polypeptide that is capable of binding to an antigen of a target cell.
- adapter or “adapter molecule” as used herein refers to a tagged polypeptide that can bind to a target antigen of a T cell, e.g. antibody or antigen binding fragment thereof such as a scFV, and has bound thereto directly or indirectly at least one additional component, i.e. the tag.
- the adapter or adapter molecule may by a tagged antibody or antigen binding fragment thereof, a cytokine or a growth factor or another soluble polypeptide that is capable of binding to a target antigen of a T cell.
- the retroviral vector particle specific for a tag as disclosed herein may bind to said adapter.
- Such an adaptable retroviral vector system that comprises a pseudotyped retroviral vector particle specific for a tag and said tagged polypeptide specific for an antigen expressed on the surface of a target cell is disclosed e.g. in WO2019086351A1.
- the tag of said tagged polypeptide may be e.g. a hapten or dextran and the hapten or dextran may be bound by the antigen binding domain of the polypeptide comprising an antigen binding domain specific for the tag.
- Said tag also may be a peptide such as a neopeptide.
- Haptens such as e.g. FITC, biotin, PE, streptavidin, thiamin or dextran are small molecules that elicit an immune response only when attached to a large carrier such as a protein; the carrier may be one that also does not elicit an immune response by itself.
- the small-molecule hapten may also be able to bind to the antibody, but it will usually not initiate an immune response; usually only the hapten-carrier adduct can do this.
- polypeptide comprising an antigen binding domain specific for a tag refers to a polypeptide that can bind a tag of a tagged polypeptide.
- the tagged polypeptide is different from the polypeptide that comprises the antigen binding domain specific for the tag.
- the polypeptide comprising the antigen binding domain specific for a tag may be an antibody or antigen binding fragment thereof that binds to said tag of the tagged polypeptide.
- polypeptide comprising an antigen binding domain specific for a tag refers to a polypeptide that can bind a tag of a tagged polypeptide.
- the polypeptide comprising the antigen binding domain specific for a tag may be an antibody or antigen binding fragment thereof such as a scFV or a nanobody that binds to said tag of the tagged polypeptide.
- antibody as used herein is used in the broadest sense to cover the various forms of antibody structures including but not being limited to monoclonal and polyclonal antibodies (including full length antibodies), multispecific antibodies (e.g. bispecific antibodies), antibody fragments, i.e. antigen binding fragments of an antibody, immunoadhesins and antibody - immunoadhesin chimeras, that specifically recognize (i.e. bind) an antigen.
- Antigen binding fragments comprise a portion of a full-length antibody, preferably the variable domain thereof, or at least the antigen binding site thereof (“an antigen binding fragment of an antibody”).
- antigen binding fragments include Fab (fragment antigen binding), scFV (single chain fragment variable), single domain antibodies, diabodies, dsFv, Fab’, single-chain antibody molecules, and multispecific antibodies formed from antibody fragments.
- the term “antigen” is intended to include substances that bind to or evoke the production of one or more antibodies and may comprise, but is not limited to, proteins, peptides, polypeptides, oligopeptides, lipids, carbohydrates such as dextran, and combinations thereof, for example a glycosylated protein or a glycolipid.
- antigen refers to a molecular entity that may be expressed on the surface of a target cell and that can be recognized by means of the adaptive immune system including but not restricted to antibodies or TCRs, or engineered molecules including but not restricted to endogenous or transgenic TCRs, CARs, scFVs or multimers thereof, Fab-fragments or multimers thereof, antibodies or multimers thereof, single chain antibodies or multimers thereof, or any other molecule that can execute binding to a structure with high affinity.
- expression is defined as the transcription and/or translation of a particular nucleotide sequence driven by its promoter in a cell.
- the term “subject” refers to an animal. Preferentially, the subject is a mammal such as mouse, rat, cow, pig, goat, chicken dog, monkey or human. More preferentially, the individual is a human.
- the subject may be a subject suffering from a disease such as cancer.
- the term “administering” refers to local and systemic administration, e.g., including enteral, parenteral, pulmonary, and topical/transdermal administration. The administration may be directly intratumoral.
- Routes of administration for pharmaceutical ingredients include, e.g., oral administration, nasal or inhalation administration, administration as a suppository, topical contact, transdermal delivery, intrathecal administration, intravenous administration, intraperitoneal administration, intramuscular administration, intralesional administration, or subcutaneous administration to a subject. Administration can be by any route including parenteral and transmucosal (e.g, oral, nasal, vaginal, rectal, or transdermal).
- Parenteral administration includes, e.g., intravenous, intramuscular, intraarterial, intrarenal, intraurethral, intracardiac, intracoronary, intramyocardial, intradermal, epidural, subcutaneous, intraperitoneal, intraventricular, ionophoretic and intracranial.
- a recombinant protein is a biotechnologically generated protein that does not occur naturally in a eukaryotic and/or prokaryotic cell. Often it is composed of different domains from different proteins, e.g. as used herein, a viral envelope protein is fused (at its ectodomain) to a polypeptide that comprises an antigen binding domain specific for an antigen or for a tag.
- the terms “having specificity for”, “specifically binds” or “specific for” with respect to an antigen-binding domain of an antibody or a fragment thereof refer to an antigen-binding domain which recognizes and binds to a specific antigen, but does not substantially recognize or bind other molecules in a sample.
- An antigen-binding domain that binds specifically to an antigen from one species may bind also to that antigen from another species. This cross-species reactivity is not contrary to the definition of that antigen-binding domain as specific.
- An antigen-binding domain that specifically binds to an antigen may bind also to different allelic forms of the antigen (allelic variants, splice variants, isoforms etc.). This cross reactivity is not contrary to the definition of that antigen-binding domain as specific.
- Immunotherapy is a medical term defined as the "treatment of disease by inducing, enhancing, or suppressing an immune response”. Immunotherapies designed to elicit or amplify an immune response are classified as activation immunotherapies, while immunotherapies that reduce or suppress are classified as suppression immunotherapies. Cancer immunotherapy as an activating immunotherapy attempts to stimulate the immune system to reject and destroy tumors. Adoptive cell transfer uses cell-based, preferentially T cell-based cytotoxic responses to attack cancer cells. T cells that have a natural or genetically engineered reactivity to a patient's cancer are generated in vitro and then transferred back into the cancer patient or are directly generated in vivo. Then the immunotherapy is referred to as “CAR T cell immunotherapy”.
- treatment means to reduce the frequency or severity of at least one sign or symptom of a disease.
- terapéuticaally effective amount or “therapeutically effective population” mean an amount of a cell population which provides a therapeutic benefit in a subject.
- engineered cell and “genetically modified cell” as used herein can be used interchangeably.
- the terms mean containing and/or expressing a foreign gene or nucleic acid sequence which in turn modifies the genotype or phenotype of the cell or its progeny.
- the terms refer to the fact that cells, preferentially T cells can be manipulated by recombinant methods well known in the art to express stably or transiently peptides or proteins which are not expressed in these cells in the natural state.
- T cells, preferentially human T cells are engineered to express an artificial construct such as a chimeric antigen receptor on their cell surface.
- automated method or “automated process” as used herein refer to any process being automated through the use of devices and/or computers and computer software. Methods (processes) that have been automated require less human intervention and less human time. In some instances the method of the present invention is automated if at least one step of the present method is performed without any human support or intervention. Preferentially the method of the present invention is automated if all steps of the method as disclosed herein are performed without human support or intervention other than connecting fresh reagents to the system. Preferentially the automated process is implemented on a closed system such as CliniMACS Prodigy® (Miltenyi Biotec).
- the closed system may comprise a) a sample processing unit comprising an input port and an output port coupled to a rotating container (or centrifugation chamber) having at least one sample chamber, wherein the sample processing unit is configured to provide a first processing step to a sample or to rotate the container so as to apply a centrifugal force to a sample deposited in the chamber and separate at least a first component and a second component of the deposited sample; and b) a sample separation unit coupled to the output port of the sample processing unit, the sample separation unit comprising a separation column holder, a pump, and a plurality of valves configured to at least partially control fluid flow through a fluid circuitry and a separation column positioned in the holder, wherein the separation column is configured to separate labeled and unlabeled components of sample flown through the column.
- This chamber may be flooded with defined gas mixes, provided by an attached gas mix unit (e.g. use of pressurized air/ N2 / CO2 or N2/CO2/O2).
- All agents may be connected to the closed system before process initiation. This comprises all buffers, solutions, cultivation media and supplements, MicroBeads, used for washing, transferring, suspending, cultivating, harvesting cells or immunomagnetic cell sorting within the closed system. Alternatively, such agents might by welded or connected by sterile means at any time during the process.
- the cell sample comprising T cells may be provided in transfer bags or other suited containers which can be connected to the closed system by sterile means.
- providing a (cell) sample comprising T cells means the provision of a cell sample, preferentially of a human cell sample of hematologic origin.
- the cell sample may be composed of hematologic cells from a donor or a patient.
- Such blood product can be in the form of whole blood, buffy coat, leukapheresis, PBMCs or any clinical sampling of blood product. It may be from fresh or frozen origin.
- cancer is known medically as a malignant neoplasm. Cancer is a broad group of diseases involving unregulated cell growth and includes all kinds of leukemia. In cancer, cells (cancerous cells) divide and grow uncontrollably, forming malignant tumors, and invading nearby parts of the body. The cancer may also spread to more distant parts of the body through the lymphatic system or bloodstream. There are over 200 different known cancers that affect humans.
- T cells may be characterized based on their function and marker expression.
- Two main subgroups have been defined: CD4 expressing T cells (i.e. T helper cells) and CD8 expressing T cells (i.e. cytotoxic T cells).
- CD8 positive specifically lyse e.g. virus infected or tumor cells by releasing perforin, granzyme and FasL upon specific binding to the respective peptide presented on the MHC I to the TCR.
- CD4+ T cells peptides presented on MHC II are bound specifically by the respective TCR inducing a signaling cascade triggering the release of several cytokines such as interferons and interleukins. Such cytokines may recruit other immune cells and may activate CD8+ T cells for a boosted and sustained cytolytic activity.
- T cells differentiate into different phenotypes showing a specific memory or effector function profile.
- Naive T cells have recently undergone positive and negative selection in the thymus and are considered to be early differentiated with high memory function but a low effector function. They can be identified by flow cytometry expressing CD45RA, CCR7 and CD62L and being negative for CD45RO, CD95 and IL-2Rbeta. Naive T cells in the blood are normally found in a quiescent state, which is characterized by small cell size, low proliferative capacity, low basal metabolic programs and low responsiveness to key cytokines, e.g. IL-2.
- the terms resting T cells”, “quiescent T cells”, “unstimulated T cells “and “non-activated T cells” may be used interchangeably.
- TSCM Stem cell memory T cells
- Central memory T cells are characterized by a low effector function profile and a long persistence. Upon antigen encounter, this T cell subset expands rapidly and differentiate into T cells with effector function. They can be identified by flow cytometry expressing CD45RO, CCR7, CD62L, CD95 and IL-2Rbeta.
- Effector memory T cells migrate to inflamed tissues and have an intermediate level of effector function. They can be identified by flow cytometry expressing CD45RO, CD95, IL- 2Rbeta and being negative for CCR7 and CD62L.
- TEFF Effector T cells
- a chimeric antigen receptor may comprise an extracellular domain (extracellular part) comprising the antigen binding domain, a transmembrane domain and a cytoplasmic signaling domain (intracellular signaling domain).
- the extracellular domain may be linked to the transmembrane domain by a linker or spacer.
- the extracellular domain may also comprise a signal peptide.
- the CAR may be an adaptable CAR system (similar to the adaptable retroviral vector system) and may be then referred to as “antitag” CAR or “adapterCAR” or “universal CAR” as disclosed e.g. in US9233125B2.
- a “signal peptide” refers to a peptide sequence that directs the transport and localization of the protein within a cell during or post translation, e.g. to a certain cell organelle (such as the endoplasmic reticulum) and/or the cell surface.
- an “antigen binding domain” of a CAR refers to the region of the CAR that specifically binds to an antigen, e.g. to a tumor associated antigen (TAA) or tumor specific antigen (TSA).
- TAA tumor associated antigen
- TSA tumor specific antigen
- the CARs of the invention may comprise one or more antigen binding domains (e.g. a tandem CAR).
- the targeting regions on the CAR are extracellular.
- the antigen binding domain of the CAR may comprise an antibody or an antigen binding fragment thereof.
- the antigen binding domain of the CAR may comprise, for example, full length heavy chain, Fab fragments, single chain Fv (scFV) fragments, nanobodies, divalent single chain antibodies or diabodies.
- any molecule that binds specifically to a given antigen such as affibodies or ligand binding domains from naturally occurring receptors may be used as an antigen binding domain.
- the antigen binding domain of a CAR is a scFV.
- a scFV Normally, in a scFV the variable regions of an immunoglobulin heavy chain and light chain are fused by a flexible linker to form a scFV.
- a linker may be for example the “(G4S)3-linker”.
- the antigen binding domain of the CAR it is beneficial for the antigen binding domain of the CAR to be derived from the same species in which the CAR will be used in.
- the antigen binding domain of the CAR when it is planned to use it therapeutically in humans, it may be beneficial for the antigen binding domain of the CAR to comprise a human or humanized antibody or antigen binding fragment thereof.
- Human or humanized antibodies or antigen binding fragments thereof can be made by a variety of methods well known in the art.
- Spacer refers to the hydrophilic region which is between the antigen binding domain of the CAR and the transmembrane domain.
- the CARs of the invention may comprise an extracellular spacer domain but is it also possible to leave out such a spacer.
- the spacer may include e.g. Fc fragments of antibodies or fragments thereof, hinge regions of antibodies or fragments thereof, CH2 or CH3 regions of antibodies, accessory proteins, artificial spacer sequences or combinations thereof.
- a prominent example of a spacer is the CD8alpha hinge.
- the transmembrane domain of the CAR may be derived from any desired natural or synthetic source for such domain.
- the domain When the source is natural the domain may be derived from any membrane-bound or transmembrane protein.
- the transmembrane domain may be derived for example from CD8alpha or CD28.
- the key signaling and antigen recognition modules domains
- the CAR may have two (or more) transmembrane domains. Splitting key signaling and antigen recognition modules enable for a small molecule-dependent, titratable and reversible control over CAR cell expression (e.g. WO2014127261A1) due to small molecule-dependent heterodimerizing domains in each polypeptide of the CAR.
- the cytoplasmic signaling domain (the intracellular signaling domain or the activating endodomain) of the CAR is responsible for activation of at least one of the normal effector functions of the immune cell in which the CAR is expressed, if the respective CAR is an activating CAR (normally, a CAR as described herein refers to an activating CAR).
- "Effector function" means a specialized function of a cell, e.g. in a T cell an effector function may be cytolytic activity or helper activity including the secretion of cytokines.
- the intracellular signaling domain refers to the part of a protein which transduces the effector function signal and directs the cell expressing the CAR to perform a specialized function.
- the intracellular signaling domain may include any complete, mutated or truncated part of the intracellular signaling domain of a given protein sufficient to transduce a signal which initiates or blocks immune cell effector functions.
- Prominent examples of intracellular signaling domains for use in the CARs include the cytoplasmic signaling sequences of the T cell receptor (TCR) and co-receptors that initiate signal transduction following antigen receptor engagement.
- TCR T cell receptor
- T cell activation can be mediated by two distinct classes of cytoplasmic signaling sequences, firstly those that initiate antigen-dependent primary activation through the TCR (primary cytoplasmic signaling sequences, primary cytoplasmic signaling domain) and secondly those that act in an antigen-independent manner to provide a secondary or costimulatory signal (secondary cytoplasmic signaling sequences, co-stimulatory signaling domain).
- primary cytoplasmic signaling sequences primary cytoplasmic signaling domain
- secondly those that act in an antigen-independent manner to provide a secondary or costimulatory signal secondary cytoplasmic signaling sequences, co-stimulatory signaling domain.
- an intracellular signaling domain of a CAR may comprise one or more primary cytoplasmic signaling domains and/or one or more secondary cytoplasmic signaling domains.
- Primary cytoplasmic signaling domains that act in a stimulatory manner may contain ITAMs (immunoreceptor tyrosine-based activation motifs).
- IT AM containing primary cytoplasmic signaling domains often used in CARs are that those derived from TCR ⁇ (CD3Q, FcRgamma, FcRbeta, CD3 gamma, CD3 delta, CD3epsilon, CD5, CD22, CD79a, CD79b, and CD66d. Most prominent is sequence derived from CD3 ⁇ .
- the cytoplasmic domain of the CAR may be designed to comprise the CD3 ⁇ signaling domain by itself or combined with any other desired cytoplasmic domain(s).
- the cytoplasmic domain of the CAR can comprise a CD3 ⁇ chain portion and a co-stimulatory signaling region (domain).
- the co-stimulatory signaling region refers to a part of the CAR comprising the intracellular domain of a co-stimulatory molecule.
- a co-stimulatory molecule is a cell surface molecule other than an antigen receptor or their ligands that is required for an efficient response of lymphocytes to an antigen.
- Examples for a co-stimulatory molecule are CD27, CD28, 4-1BB (CD137), 0X40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen- 1 (LFA- 1), CD2, CD7, LIGHT, NKG2C, B7-H3.
- LFA- 1 lymphocyte function-associated antigen- 1
- the cytoplasmic signaling sequences within the cytoplasmic signaling part of the CAR may be linked to each other with or without a linker in a random or specified order.
- a short oligo- or polypeptide linker which is preferably between 2 and 10 amino acids in length, may form the linkage.
- a prominent linker is the glycine-serine doublet.
- the cytoplasmic domain may comprise the signaling domain of CD3 ⁇ and the signaling domain of CD28.
- the cytoplasmic domain may comprise the signaling domain of CD3 ⁇ and the signaling domain of CD137.
- the cytoplasmic domain may comprise the signaling domain of CD3 ⁇ , the signaling domain of CD28, and the signaling domain of CD137.
- CAR is an inhibitory CAR (referred to normally as “iCAR”)
- said CAR may have the same extracellular and/or transmembrane domains as the activating CAR but differs from the activating CAR with regard to the endodmain.
- the at least one endodomain of the inhibitory CAR may be a cytoplasmic signaling domain comprising at least one signal transduction element that inhibits an immune cell or comprising at least one element that induces apoptosis.
- the CARs that may be transduced by the pseudotyped retroviral vector particle as disclosed herein present may be designed to comprise any portion or part of the above-mentioned domains as described herein in any order and/or combination resulting in a functional CAR.
- display refers to a protein or peptide that is incorporated into the viral envelope, thereby presenting the extracellular domain outside the viral particle.
- Example 1 Principle of the anti-CD3-displaying retroviral vector system
- an envelope protein was designed that confers T cell modulating capacity.
- This modulating protein can be incorporated in the envelope of pseudotyped retroviral particles.
- Suitable pseudotypes may be Canine distemper virus (CDV), Measles virus (MV), Nipah virus (NiV) and Vesicular stomatitis virus (FIG 1 A).
- CDV Canine distemper virus
- MV Measles virus
- NiV Nipah virus
- VFG 1 A Vesicular stomatitis virus
- the envelope glycoproteins of CDV, MV and NiV may be modified to ablate binding to their native receptors but bind to a desired antigen, e.g., CD4, CD8, or a tag, by fusion with an scFVs.
- the retroviral particles bind via the modulating protein, that is comprising an a-CD3 scFV, to the TCR. This results in activation of the T cells, which can be observed in activation marker upregulation, enhanced metabolism and proliferation of the T cells.
- binding of the envelope glycoproteins to their designated receptors will result in transduction of the T cells, characterized by integration and expression of the transgene, e.g. GFP or a CAR (FIG IB).
- HEK293T cells were seeded in 6 wells with a density of 8xl0 5 cells/well one day before transfection. The HEK293T cells were transfected with the plasmids encoding the modulating protein or GFP as a positive control.
- Pseudotyped retroviral vector particles specific for a tag of tagged polypeptide or specific for a target antigen expressed on a cell were generated by transient transfection of HEK293T cells.
- the supernatant was collected, centrifuged for 10 min at 1000 rpm, followed by filtration through a 0.45 pm filter. To concentrate, the filtered supernatant was centrifuged through a 20 % sucrose (Sigma Aldrich, Cat.No. 84097-250 g, 20 % w/v in PBS) cushion for 24 h at 4 °C with 5350xg. The pelleted retroviral vectors were resuspended in 250 pl precooled PBS, aliquoted and stored at -80 °C for later use. To confirm sufficient transfection for production, the producer cells were stained after vector harvest for expression of the a-CD3 scFV and the CDV H protein. Subsequently, transfection efficiency was determined by quantification of the a-CD3 scFV, H and GFP expressing cells using flow cytometry (FIG 3B, C).
- Pseudotyped retroviral vector particles were titrated on SupTl cells.
- Tag-specific LVs were titrated in the absence of the tagged polypeptide. Therefore, cell surface proteins were randomly labeled with LC-LC- biotin using by Biotin-LC-LC-NHS.
- SupTl resuspended in 1 ml PBS were supplemented with Biotin-LC-LC-NHS followed by an incubation at 4 °C at constant mixing. After removing cell-free supernatant, cells were washed and seeded.
- SupTl or biotinylated SupTl were seeded with 2xl0 5 cells/well in 96-well in media (RPMI) without FCS.
- the LV particles were serially diluted in RPMI and added to the SupTl cells.
- 96 h post transduction the transduction efficiency was determined by flow cytometry quantifying the ratio of GFP positive.
- the ratio of GFP positive cells, the dilution factor and the volume of retroviral applied is used to calculate the retroviral vector titer (i.e. transducing units per volume (TU/ml) (FIG 4A-B, FIG 9, FIG 15 A-B).
- CDV-pseudotyped retroviral vectors could be produced by HEK293T cells co-expressing the modulating proteins. Only, in presence of modulating proteins comprising a LNGFR transmembrane domain (SEQ ID: 32) resulted in a significant decrease in vector productivity. Importantly, the a-CD3 scFV did not influence the productivity (FIG 15 A-B).
- Pseudotyped retroviral particles were immobilized via a N-terminal tag comprised in the modulating protein to confirm the incorporation of the a-CD3 scFV in functional retroviral particles. Therefore, a 96-well ELISA plate was coated with anti-tag antibodies and washed with PBS containing 0.05 % Tween three times. After blocking with PBS containing 0.05 % Tween with 2 % BSA for two hours, GFP encoding retroviral vector particles diluted in PBS containing 2 % BSA were added to the wells and incubated at room temperature for 1.5 hours. Unbound particles were removed by washing three times with PBS containing 2 % BSA.
- Biotinylated SupTl were prepared as described in Example 4 and added to the wells with a density of 2xl0 5 cells/well.
- SupTl were added with a density of 2xl0 5 cells/well.
- the transduction efficiency was determined by quantification of GFP-expressing cells using flow cytometry six to eight days after transduction (FIG 5, FIG 10, FIG 12 A-C). The data suggest, that the modulating protein was incorporated into functional retroviral vectors. The level of immobilization varied with the architecture of the modulating protein.
- PBMCs were isolated from buffy coat of healthy donors by density gradient centrifugation.
- Non-activated primary CD4+ and CD8+ T cells were isolated from the PBMC using the Pan T cell Isolation kit (Miltenyi Biotec, Cat.No. 130-096-535) and CD8+ T cells were isolated from the PBMC using the CD8 T cell Isolation kit (Miltenyi Biotec, Cat.No. 130-096-495) and stained with CellTraceTM Violet following the manufacturer’s instructions (ThermoFisher, Cat.No. C34571).
- Pan T cells or CD8+ T cells were seeded with a density of 2xl0 5 cells/ well in a 96-well plate and transduced with GFP encoding retroviral vector particles. The medium was replaced two days after transduction with fresh complete medium.
- T cells were stained two, five and/or eight days after transduction for expression of the activation marker CD25. Subsequently, T cell proliferation transduction efficiency and activation marker expression were determined by flow cytometry (FIG 6, FIG 11, FIG 13, FIG 16). Treatment of T cells with retroviral vectors displaying the modulating proteins resulted in upregulation of CD25, suggesting an induction of activation. The level of CD25 expression was dependent on the architecture of the modulating protein displayed by the respective retroviral vectors. Highest level of CD25 expression was achieved with retroviral vectors displaying modulating proteins comprising CD28, PDGFR and CD8 transmembrane domains (FIG 6A, FIG 11 A).
- T cell activation was also achieved with VSV-G or MV-pseudotyped retroviral vectors displaying the modulating protein (FIG 13A).
- Analysis of proliferation showed that induction of proliferation in absence of CD28 costimulation was achieved (FIG 6B, FIG 11D, FIG 13C, FIG 16E).
- the highest level of proliferation was achieved with retroviral vectors displaying modulating proteins comprising CD8 transmembrane domains and the a-CD3 scFV of SEQ ID 4 (FIG 6B, 1 ID).
- the highest level of proliferation was achieved with modulating proteins comprising SEQ ID 4 as compared to SEQ ID 3 and SEQ ID 34 (FIG 16E).
- Proliferation was also induced by VSV-G or MV pseudotyped retroviral vectors displaying the modulating protein (FIG 13C).
- Retroviral vectors displaying the modulating protein efficiently transduced the non-activated T cells (FIG 6C, FIG 11B-C, FIG 13B, FIG 16C-D). Transduction efficiency was increased in presence of the modulating protein for VSV-G pseudotyped LV and adapter-specific retroviral vectors, resulting also in an increased number of transduced cells (FIG 16C-D). Importantly, the transduction remained to be selective as no transduction of CD4+ T cells was observed upon transduction with CD8-sepcific retroviral vectors comprising the modulating protein (FIG 11B- C).
- PBMCs of healthy donors were isolated from buffy coat by density gradient centrifugation.
- the PBMC were seeded with 2.5xl0 5 cells/well in TexMACSTM medium supplemented with 12.5 ng/ml IL7 and 12.5 ng/ml IL15 in a 96-well plate.
- 1 pM of Raltegravir - an integrase inhibitor - was added one hour before transduction and maintained in the medium throughout the culture to exclude pseudo-transduction.
- the cells were incubated with CD8- specific fab adapter molecules for 30 min at 4 °C, followed by transduction with CD20CAR encoding retroviral vector particles at a dose of 2.5 TU/cell.
- TransActTM was added together with the LV particles.
- the medium was replaced with fresh complete medium two days after transduction.
- Seven days post transduction the transduction efficiency was analyzed by staining for the transduction marker LNGFR and subsequent flow cytometry analysis.
- viable B cells were quantified by flow cytometry after staining for CD 19 (FIG 7A-B). Efficient transduction only in presence of adapter and in absence of Raltegravir shows that a selective gene delivery with retroviral vectors was achieved. Depletion of B cells upon transduction suggests generation of functional CD20CAR T cells.
- Example 8 In vivo generation of CD20 CAR-T cells with adapter-based retroviral vector particles.
- mice were left untreated or were i.p. injected with CD8-specific adapter molecules. Two-to three hours later, biotin-specific CDV-LV encoding a CD20CAR and the transduction marker LNGFR with or without displaying the modulating protein was i.v. injected with 7.5xl0 7 TU/mouse. Blood samples were taken 5 and 12 days post transduction via the vena facialis.
- mice 15 days post injection of retroviral vector particles the mice were sacrificed and blood, bone marrow and spleen tissue was harvested.
- the amount of human T cells and the level of CAR positive T cells was analyzed upon staining for respective markers and subsequent flow cytometry (FIG 17 A-D).
- the amount of human T cells in the mouse blood increased over time with a stronger increase in mice treated with retroviral vectors comprising the modulating protein (FIG 17B).
- the endpoint analysis revealed a higher amount of CD8+ T cells upon transduction with retroviral vectors comprising the modulating protein compared to untreated mice or mice treated with the retroviral vector without display of the modulating protein (FIG 17C).
- the level of transduced T cells was increased with retroviral vectors displaying the modulating protein compared the control treated mice (FIG 17D).
- CDV-pseudotyped retroviral vector particles targeted against CD3 were produced as described in Example 3. Subsequently, Jurkat cells were seeded with a density of 3xl0 5 cells/well in 48-well plate. aCD3-targeted retroviral vector particles were serially diluted in RMPI without FCS and added to the cells. After 96 hours, transduction efficiency was analyzed by quantification of GFP positive cells. CD4-targeted CDV-LV (aCD4-CDV-LV) was used as positive control. Interestingly, GFP expressing cells were not detected upon transduction with aCD3-CDV-LV (FIG 18), suggesting that retargeting of CDV pseudotyped LV to CD3 results in generation of non-functional particles.
- SEQ ID NO: 6 (env incorporation signal (gp41)):
- SEQ ID NO: 8 (CD4 scFV VL):
- SEQ ID NO: 10 (CD8 scFV VL):
- SEQ ID NO: 14 N-terminal signal Sequence F protein
- SEQ ID NO: 19 (cytoplasmic domain FcD30 measles):
- SEQ ID NO:20 cytoplasmic domain FcD30 of Canine distemper virus strain 5840P: ALLFLIYCCKRR
- SEQ ID NO:21 cytoplasmic domain of HcD21 of Canine distemper virus strain 5840P: MLSLVTEEQGGRRPPY
- SEQ ID NO:22 cytoplasmic domain of HcD30 of Canine distemper virus strain 5840P: MGRRPPY
- SEQ ID NO:23 (cytoplasmic domain of 4-1BB):
- SEQ ID NO:24 (cytoplasmic domain of CD28):
- SEQ ID NO: 28 (PDGFR transmembrane domain)
- SEQ ID NO: 32 (LNGFR transmembrane domain)
- SEQ ID NO: 33 (VSVG transmembrane domain)
- VSV-G-LVs do not allow efficient gene transfer into unstimulated T cells, B cells, and HSCs because they lack the LDL receptor.
- Tahara M Takeda M, Shirogane Y, Hashiguchi T, Ohno S, Yanagi Y. Measles virus infects both polarized epithelial and immune cells by using distinctive receptor-binding sites on its hemagglutinin. J Virol. 2008 May;82(9):4630-7. doi: 10. 1128/JVI.02691-07. Epub 2008 Feb 20. PMID: 18287234; PMCID: PMC2293038. Taube R, Zhu Q, Xu C, Diaz-Griffero F, Sui J, Kamau E, Dwyer M, Aird D, Marasco WA. Lentivirus display: stable expression of human antibodies on the surface of human cells and virus particles.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Genetics & Genomics (AREA)
- Immunology (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- General Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Virology (AREA)
- General Engineering & Computer Science (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Medicinal Chemistry (AREA)
- Biomedical Technology (AREA)
- Biotechnology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Plant Pathology (AREA)
- Microbiology (AREA)
- Physics & Mathematics (AREA)
- Gastroenterology & Hepatology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Peptides Or Proteins (AREA)
Abstract
La présente invention concerne une particule de vecteur rétroviral pseudotypé pour activer et transduire des lymphocytes T, ladite particule de vecteur rétroviral comprenant une protéine de modulation comprenant un ectodomaine fonctionnel comprenant un domaine de liaison à l'antigène spécifique de l'antigène CD3, ledit domaine de liaison à l'antigène spécifique de l'antigène CD3 comprenant une séquence scFV humanisée et optimisée, ladite particule de vecteur rétroviral comprenant au moins une séquence d'acide nucléique codant pour un transgène, et ladite particule de vecteur rétroviral étant une particule de vecteur lentiviral ou gammarétroviral. L'invention concerne également une composition pharmaceutique de celle-ci.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP23170224.2 | 2023-04-27 | ||
| EP23170224 | 2023-04-27 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024223847A1 true WO2024223847A1 (fr) | 2024-10-31 |
Family
ID=86282411
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2024/061576 Pending WO2024223847A1 (fr) | 2023-04-27 | 2024-04-26 | Particule de vecteur rétroviral pseudotypé avec affichage anti-cd3 |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024223847A1 (fr) |
Citations (13)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2008037458A2 (fr) | 2006-09-27 | 2008-04-03 | Bundesrepublik Deutschland, Letztvertreten Durch Den Präsidenten Des Paul-Ehrlich-Instituts | Pseudotypage de vecteurs rétroviraux, procédés de fabrication et d'utilisation de ceux-ci pour transfert de gène ciblé et criblage haut débit |
| WO2013188693A1 (fr) * | 2012-06-15 | 2013-12-19 | Imaginab, Inc. | Constructions de liaison à l'antigène pour cd3 |
| WO2014127261A1 (fr) | 2013-02-15 | 2014-08-21 | The Regents Of The University Of California | Récepteur d'antigène chimère et procédés d'utilisation de celui-ci |
| US9233125B2 (en) | 2010-12-14 | 2016-01-12 | University Of Maryland, Baltimore | Universal anti-tag chimeric antigen receptor-expressing T cells and methods of treating cancer |
| WO2016139463A1 (fr) * | 2015-03-02 | 2016-09-09 | Ucl Business Plc | Vecteurs rétroviraux et lentiviraux |
| WO2016180721A1 (fr) | 2015-05-08 | 2016-11-17 | Miltenyi Biotec Gmbh | Anticorps humanisé ou fragment de celui-ci spécifique pour cd3 |
| WO2018033726A1 (fr) | 2016-08-17 | 2018-02-22 | Autolus Limited | Vecteurs rétroviraux et lentiviraux |
| WO2019086351A1 (fr) | 2017-10-30 | 2019-05-09 | Miltenyi Biotec Gmbh | Système de vecteurs rétroviraux basé sur un adaptateur pour la transduction sélective de cellules cibles |
| WO2019200056A2 (fr) | 2018-04-12 | 2019-10-17 | Vivo-Til Therapeutics Inc. | Vecteurs viraux et lignées d'encapsidation |
| WO2020106992A1 (fr) | 2018-11-21 | 2020-05-28 | Umoja Biopharma, Inc. | Vecteur multicistronique pour ingénierie de surface de particules lentivirales |
| WO2021154839A1 (fr) | 2020-01-30 | 2021-08-05 | Umoja Biopharma, Inc. | Activateur de transduction bispécifique |
| EP3964585A1 (fr) * | 2020-09-03 | 2022-03-09 | Miltenyi Biotec B.V. & Co. KG | Particule de vecteur lentiviral spécifique de cd62l pour la transduction ciblée de sous-ensembles de lymphocytes t |
| WO2022164935A1 (fr) | 2021-01-27 | 2022-08-04 | Umoja Biopharma, Inc. | Lentivirus permettant de générer des cellules exprimant un récepteur antigénique chimérique anti-cd19 |
-
2024
- 2024-04-26 WO PCT/EP2024/061576 patent/WO2024223847A1/fr active Pending
Patent Citations (13)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2008037458A2 (fr) | 2006-09-27 | 2008-04-03 | Bundesrepublik Deutschland, Letztvertreten Durch Den Präsidenten Des Paul-Ehrlich-Instituts | Pseudotypage de vecteurs rétroviraux, procédés de fabrication et d'utilisation de ceux-ci pour transfert de gène ciblé et criblage haut débit |
| US9233125B2 (en) | 2010-12-14 | 2016-01-12 | University Of Maryland, Baltimore | Universal anti-tag chimeric antigen receptor-expressing T cells and methods of treating cancer |
| WO2013188693A1 (fr) * | 2012-06-15 | 2013-12-19 | Imaginab, Inc. | Constructions de liaison à l'antigène pour cd3 |
| WO2014127261A1 (fr) | 2013-02-15 | 2014-08-21 | The Regents Of The University Of California | Récepteur d'antigène chimère et procédés d'utilisation de celui-ci |
| WO2016139463A1 (fr) * | 2015-03-02 | 2016-09-09 | Ucl Business Plc | Vecteurs rétroviraux et lentiviraux |
| WO2016180721A1 (fr) | 2015-05-08 | 2016-11-17 | Miltenyi Biotec Gmbh | Anticorps humanisé ou fragment de celui-ci spécifique pour cd3 |
| WO2018033726A1 (fr) | 2016-08-17 | 2018-02-22 | Autolus Limited | Vecteurs rétroviraux et lentiviraux |
| WO2019086351A1 (fr) | 2017-10-30 | 2019-05-09 | Miltenyi Biotec Gmbh | Système de vecteurs rétroviraux basé sur un adaptateur pour la transduction sélective de cellules cibles |
| WO2019200056A2 (fr) | 2018-04-12 | 2019-10-17 | Vivo-Til Therapeutics Inc. | Vecteurs viraux et lignées d'encapsidation |
| WO2020106992A1 (fr) | 2018-11-21 | 2020-05-28 | Umoja Biopharma, Inc. | Vecteur multicistronique pour ingénierie de surface de particules lentivirales |
| WO2021154839A1 (fr) | 2020-01-30 | 2021-08-05 | Umoja Biopharma, Inc. | Activateur de transduction bispécifique |
| EP3964585A1 (fr) * | 2020-09-03 | 2022-03-09 | Miltenyi Biotec B.V. & Co. KG | Particule de vecteur lentiviral spécifique de cd62l pour la transduction ciblée de sous-ensembles de lymphocytes t |
| WO2022164935A1 (fr) | 2021-01-27 | 2022-08-04 | Umoja Biopharma, Inc. | Lentivirus permettant de générer des cellules exprimant un récepteur antigénique chimérique anti-cd19 |
Non-Patent Citations (25)
| Title |
|---|
| AMIRACHE FLEVY CCOSTA CMANGEOT PETORBETT BEWANG CXNEGRE DCOSSET FLVERHOEYEN E.: "Mystery solved: VSV-G-LVs do not allow efficient gene transfer into unstimulated T cells, B cells, and HSCs because they lack the LDL receptor", BLOOD, vol. 123, no. 9, 27 February 2014 (2014-02-27), pages 1422 - 4, XP055933750 |
| ARCANGELI SBOVE CMEZZANOTTE CCAMISA BFALCONE LMANFREDI FBEZZECCHI EEL KHOURY RNORATA RSANVITO F: "CAR T cell manufacturing from naive/stem memory T lymphocytes enhances antitumor responses while curtailing cytokine release syndrome", J CLIN INVEST., vol. 132, no. 12, 15 June 2022 (2022-06-15), pages e150807, XP093066306, DOI: 10.1172/JCI150807 |
| BAH ESNACE RAPENG KWMUNOA-ALIA MARUSSELL SJ.: "Retargeted and Stealth-Modified Oncolytic Measles Viruses for Systemic Cancer Therapy in Measles Immune Patients", MOL CANCER THER., vol. 19, no. 10, 26 August 2020 (2020-08-26), pages 2057 - 2067 |
| BENDER RRMUTH ASCHNEIDER ICFRIEDEL THARTMANN JPLUCKTHUN AMAISNER ABUCHHOLZ CJ.: "Receptor-Targeted Nipah Virus Glycoproteins Improve Cell-Type Selective Gene Delivery and Reveal a Preference for Membrane-Proximal Cell Attachment", PLOS PATHOG., vol. 12, no. 6, 9 June 2016 (2016-06-09), pages e1005641, XP055309520, DOI: 10.1371/journal.ppat.1005641 |
| DAI JAGOSTO LMBAYTOP CYU JJPACE MJLISZEWSKI MKO'DOHERTY U.: "Human immunodeficiency virus integrates directly into naive resting CD4+ T cells but enters naive cells less efficiently than memory cells", J VIROL., vol. 83, no. 9, 11 February 2009 (2009-02-11), pages 4528 - 37, XP055943099, DOI: 10.1128/JVI.01910-08 |
| DERDAK SVKUENG HJLEB VMNEUNKIRCHNER ASCHMETTERER KGBIELEK EMAJDIC OKNAPP WSEED BPICKL WF.: "Direct stimulation of T lymphocytes by immunosomes: virus-like particles decorated with T cell receptor/CD3 ligands plus costimulatory molecules", PROC NATL ACAD SCI USA, vol. 103, no. 35, 21 August 2006 (2006-08-21), pages 13144 - 9, XP055893683, DOI: 10.1073/pnas.0602283103 |
| FRANK AMBRAUN AHSCHEIB LAGARWAL SSCHNEIDER ICFUSIL FPERIAN SSAHIN UTHALHEIMER FBVERHOEYEN E: "Combining T-cell-specific activation and in vivo gene delivery through CD3-targeted lentiviral vectors", BLOOD ADV., vol. 4, no. 22, 24 November 2020 (2020-11-24), pages 5702 - 5715, XP055810142, DOI: 10.1182/bloodadvances.2020002229 |
| FRANK ANNIKA M ET AL, vol. 4, no. 22, 24 November 2020 (2020-11-24), pages 5702 - 5715, XP055810142, ISSN: 2473-9529, Retrieved from the Internet <URL:https://ashpublications.org/bloodadvances/article-pdf/4/22/5702/1790494/advancesadv2020002229.pdf> DOI: 10.1182/bloodadvances.2020002229 * |
| GHASSEMI SNUNEZ-CRUZ SO'CONNOR RSFRAIETTA JAPATEL PRSCHOLLER JBARRETT DMLUNDH SMDAVIS MMBEDOYA F: "Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells", CANCER IMMUNOL RES., vol. 6, no. 9, 20 July 2018 (2018-07-20), pages 1100 - 1109, XP055681222, DOI: 10.1158/2326-6066.CIR-17-0405 |
| HUCKABY JTLANDONI EJACOBS TMSAVOLDO BDOTTI GLAI SK.: "Bispecific binder redirected lentiviral vector enables in vivo engineering of CAR-T cells", J IMMUNOTHER CANCER, vol. 9, no. 9, September 2021 (2021-09-01), pages e002737, XP093152324, DOI: 10.1136/jitc-2021-002737 |
| KALAMASZ DLONG SATANIGUCHI RBUCKNER JHBERENSON RJBONYHADI M.: "Optimization of human T-cell expansion ex vivo using magnetic beads conjugated with anti-CD3 and Anti-CD28 antibodies", J IMMUNOTHER., vol. 27, no. 5, September 2004 (2004-09-01), pages 405 - 18, XP009067003, DOI: 10.1097/00002371-200409000-00010 |
| LI YKURLANDER RJ.: "Comparison of anti-CD3 and anti-CD28-coated beads with soluble anti-CD3 for expanding human T cells: differing impact on CD8 T cell phenotype and responsiveness to restimulation", J TRANSL MED., vol. 8, 26 October 2010 (2010-10-26), pages 104, XP021078842, DOI: 10.1186/1479-5876-8-104 |
| LIU YPRUSSELL SPAYALA-BRETON CRUSSELL SJPENG KW.: "Ablation of nectin4 binding compromises CD46 usage by a hybrid vesicular stomatitis virus/measles virus", J VIROL., vol. 88, no. 4, 11 December 2013 (2013-12-11), pages 2195 - 204, XP055224540, DOI: 10.1128/JVI.02628-13 |
| MASSE NAINOUZE MNEEL BWILD TFBUCKLAND RLANGEDIJK JP: "Measles virus (MV) hemagglutinin: evidence that attachment sites for MV receptors SLAM and CD46 overlap on the globular head", J VIROL., vol. 78, no. 17, September 2004 (2004-09-01), pages 9051 - 63 |
| MASSE NBARRETT TMULLER CPWILD TFBUCKLAND R.: "Identification of a second major site for CD46 binding in the hemagglutinin protein from a laboratory strain of measles virus (MV): potential consequences for wild-type MV infection", J VIROL., vol. 76, no. 24, December 2002 (2002-12-01), pages 13034 - 8 |
| MAURICE MVERHOEYEN ESALMON PTRONO DRUSSELL SJCOSSET FL: "Efficient gene transfer into human primary blood lymphocytes by surface-engineered lentiviral vectors that display a T cell-activating polypeptide", BLOOD, vol. 99, no. 7, 1 April 2002 (2002-04-01), pages 2342 - 50, XP002346143, DOI: 10.1182/blood.V99.7.2342 |
| NAKAMURA TPENG KWHARVEY MGREINER SLORIMER IAJAMES CDRUSSELL SJ.: "Rescue and propagation of fully retargeted oncolytic measles viruses", NAT BIOTECHNOL., vol. 23, no. 2, 30 January 2005 (2005-01-30), pages 209 - 14, XP037923888, DOI: 10.1038/nbt1060 |
| NAKAMURA TPENG KWVONGPUNSAWAD SHARVEY MMIZUGUCHI HHAYAKAWA TCATTANEO RRUSSELL SJ.: "Antibody-targeted cell fusion", NAT BIOTECHNOL., vol. 22, no. 3, 15 February 2004 (2004-02-15), pages 331 - 6, XP055337950, DOI: 10.1038/nbt942 |
| PATTERSON JBSCHEIFLINGER FMANCHESTER MYILMA TOLDSTONE MB.: "Structural and functional studies of the measles virus hemagglutinin: identification of a novel site required for CD46 interaction", VIROLOGY, vol. 256, no. 1, 30 March 1999 (1999-03-30), pages 142 - 51, XP004440003, DOI: 10.1006/viro.1999.9644 |
| RAN TAUBE ET AL: "Lentivirus Display: Stable Expression of Human Antibodies on the Surface of Human Cells and Virus Particles", PLOS ONE, vol. 3, no. 9, 11 September 2008 (2008-09-11), pages e3181, XP055027886, DOI: 10.1371/journal.pone.0003181 * |
| RASBACH AABEL TMUNCH RCBOILER KSCHNEIDER-SCHAULIES JBUCHHOLZ CJ.: "The receptor attachment function of measles virus hemagglutinin can be replaced with an autonomous protein that binds Her2/neu while maintaining its fusion-helper function", J VIROL., vol. 87, no. 11, 27 March 2013 (2013-03-27), pages 6246 - 56 |
| TAHARA MTAKEDA MSHIROGANE YHASHIGUCHI TOHNO SYANAGI Y.: "Measles virus infects both polarized epithelial and immune cells by using distinctive receptor-binding sites on its hemagglutinin", J VIROL., vol. 82, no. 9, 20 February 2008 (2008-02-20), pages 4630 - 7 |
| TAUBE RZHU QXU CDIAZ-GRIFFERO FSUI JKAMAU EDWYER MAIRD DMARASCO WA.: "Lentivirus display: stable expression of human antibodies on the surface of human cells and virus particles", PLOS ONE, vol. 3, no. 9, 11 September 2008 (2008-09-11), pages e3181 |
| VON MESSLING VOEZGUEN NZHENG QVONGPUNSAWAD SBRAUN WCATTANEO R: "Nearby clusters of hemagglutinin residues sustain SLAM-dependent canine distemper virus entry in peripheral blood mononuclear cells", J VIROL., vol. 79, no. 9, May 2005 (2005-05-01), pages 5857 - 62 |
| VONGPUNSAWAD SOEZGUN NBRAUN WCATTANEO R.: "Selectively receptor-blind measles viruses: Identification of residues necessary for SLAM- or CD46-induced fusion and their localization on a new hemagglutinin structural model", J VIROL., vol. 78, no. 1, January 2004 (2004-01-01), pages 302 - 13, XP055189849, DOI: 10.1128/JVI.78.1.302-313.2004 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US12258574B2 (en) | Methods and compositions for transducing lymphocytes and regulating the activity thereof | |
| JP7474281B2 (ja) | リンパ球に形質導入を行うための方法及び組成物、並びにその制御された増加 | |
| US11814641B2 (en) | Retroviral and lentiviral vectors | |
| US20240093232A1 (en) | Retroviral And Lentiviral Vectors | |
| KR102758204B1 (ko) | 표적 세포의 선택적 형질도입을 위한 어댑터-기반 레트로바이러스 벡터 시스템 | |
| ES2520024T3 (es) | Pseudotipificación de vectores retrovíricos, métodos para su producción y su uso para la transferencia de genes dirigida y la selección de alta capacidad de procesamiento | |
| US20230407330A1 (en) | Vector system for delivery of multiple polynucleotides and uses thereof | |
| CA3030003A1 (fr) | Procedes et compositions pour la transduction de lymphocytes et la regulation de l'activite correspondante | |
| CA3054064A1 (fr) | Procedes et compositions pour la transduction et l'expansion de lymphocytes et la regulation de l'activite de ces derniers | |
| US12018273B2 (en) | CD62L specific lentiviral vector particle for targeted transduction of T cell subsets | |
| US20230392139A1 (en) | Methods and compositions for transducing and expanding lymphocytes and regulating the activity thereof | |
| WO2024223870A1 (fr) | Particule de vecteur rétroviral pseudotypée avec des protéines d'enveloppe du virus de la maladie de carré | |
| WO2024223847A1 (fr) | Particule de vecteur rétroviral pseudotypé avec affichage anti-cd3 | |
| WO2025078492A1 (fr) | Particules de vecteur lentiviral nipah-pseudotypé ciblant cd3 | |
| US20220315894A1 (en) | Method for Transduction of T Cells in the Presence of Malignant Cells |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24723049 Country of ref document: EP Kind code of ref document: A1 |