WO2014071963A1 - Procédé pour l'expression d'arn cellulaire - Google Patents

Procédé pour l'expression d'arn cellulaire Download PDF

Info

Publication number
WO2014071963A1
WO2014071963A1 PCT/EP2012/004673 EP2012004673W WO2014071963A1 WO 2014071963 A1 WO2014071963 A1 WO 2014071963A1 EP 2012004673 W EP2012004673 W EP 2012004673W WO 2014071963 A1 WO2014071963 A1 WO 2014071963A1
Authority
WO
WIPO (PCT)
Prior art keywords
rna
cells
cell
ifn
pkr
Prior art date
Application number
PCT/EP2012/004673
Other languages
English (en)
Inventor
Ugur Sahin
Tim Beissert
Marco Poleganov
Stephanie Herz
Original Assignee
Biontech Ag
Tron - Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Gemeinnützige Gmbh
Universitätsmedizin Der Johannes Gutenberg-Universität Mainz
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by Biontech Ag, Tron - Translationale Onkologie An Der Universitätsmedizin Der Johannes Gutenberg-Universität Mainz Gemeinnützige Gmbh, Universitätsmedizin Der Johannes Gutenberg-Universität Mainz filed Critical Biontech Ag
Priority to PCT/EP2012/004673 priority Critical patent/WO2014071963A1/fr
Priority to AU2013343864A priority patent/AU2013343864B2/en
Priority to ES13792854.5T priority patent/ES2676470T3/es
Priority to TR2018/09547T priority patent/TR201809547T4/tr
Priority to EP13792854.5A priority patent/EP2917350B1/fr
Priority to PCT/EP2013/003362 priority patent/WO2014072061A1/fr
Priority to CA2890529A priority patent/CA2890529C/fr
Priority to JP2015541037A priority patent/JP6353846B2/ja
Publication of WO2014071963A1 publication Critical patent/WO2014071963A1/fr
Priority to US14/706,228 priority patent/US10207009B2/en
Priority to US16/245,353 priority patent/US10729784B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/24Interferons [IFN]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/602Sox-2
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/603Oct-3/4
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/604Klf-4
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/605Nanog
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/606Transcription factors c-Myc
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/608Lin28
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/70Enzymes
    • C12N2501/72Transferases (EC 2.)
    • C12N2501/727Kinases (EC 2.7.)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates to expressing RNA in cells and, in particular, enhancing viability of cells in which RNA is to be expressed.
  • the cells are preferably transfected with the RNA such as by repetitive transfection.
  • the present invention provides methods for expressing RNA in cells comprising the steps of preventing engagement of IFN receptor by extracellular IFN and inhibiting intracellular IFN signalling in the cells. Preventing engagement of IFN receptor by extracellular IFN and inhibiting intracellular IFN signalling in the cells allows stable expression of RNA in the cells, in particular, if cells are transfected repetitively with RNA.
  • preventing engagement of IFN receptor by extracellular IFN and inhibiting intracellular IFN signalling enhances survival of the cells, in particular, if cells are transfected repetitively with RNA.
  • preventing engagement of IFN receptor by extracellular IFN and inhibiting intracellular IFN signalling in the cells allows repetitive transfer of RNA into the cells.
  • RNA does not need to enter the nucleus in order to be expressed and moreover cannot integrate into the host genome, thereby eliminating the risk of oncogenesis.
  • Transfection rates attainable with RNA are relatively high, for many cell types even >90%, and therefore, there is no need for selection of transfected cells.
  • the amounts of protein achieved correspond to those in physiological expression.
  • RNA has been described for as being useful in de-differentiating somatic cells into stem-like cells without generating embryos or fetuses.
  • De-differentiation of somatic cells to cells having stem cell characteristics, in particular pluripotency can be effected by introducing RNA encoding factors inducing the de-differentiation of somatic cells into the somatic cells (also termed reprogramming transcription factors (rTF)) and culturing the somatic cells allowing the cells to de-differentiate.
  • rTF reprogramming transcription factors
  • the cells could be induced to re- differentiate into the same or a different somatic cell type such as neuronal, hematopoietic, muscle, epithelial, and other cell types.
  • stem-like cells have medical applications for treatment of degenerative diseases by "cell therapy” and may be utilized in novel therapeutic strategies in the treatment of cardiac, neurological, endocrinological, vascular, retinal, dermatological, muscular-skeletal disorders, and other diseases.
  • RNA provides an attractive alternative to circumvent the potential risks of DNA based vaccines.
  • transfer of RNA into cells can also induce both the cellular and humoral immune responses in vivo.
  • two different strategies have been pursued for immunotherapy with in vitro transcribed RNA (IVT-RNA), which have both been successfully tested in various animal models.
  • RNA is directly injected into the patient by different immunization routes or cells are transfected with IVT-RNA using conventional transfection methods in vitro and then the transfected cells are administered to the patient.
  • RNA may, for example, be translated and the expressed protein presented on the MHC molecules on the surface of the cells to elicit an immune response. It has been attempted to stabilize IVT-RNA by various modifications in order to achieve higher and prolonged expression of transferred IVT-RNA.
  • RNA transfection-based strategies to express peptides and proteins in cells there remain issues related to RNA stability, sustained expression of the encoded peptide or protein and cytotoxicity of the RNA.
  • RNA-based gene transfer is accompanied with an induction of the IFN-response which hinders the continuous expression of rTF when delivered as mRNA and therefore successful RNA-based reprogramming.
  • the invention relates to a method for expressing RNA in a cell comprising the steps of (i) preventing engagement of IFN receptor by extracellular IFN and (ii) inhibiting intracellular IFN signalling.
  • the RNA is or has been introduced into the cell such as by electroporation or lipofection.
  • the RNA is or has been introduced into the cell repetitively.
  • the RNA is in vitro transcribed RNA.
  • preventing engagement of IFN receptor by extracellular IFN inhibits autocrine and/or paracrine IFN functions. In one embodiment, preventing engagement of IFN receptor by extracellular IFN comprises providing a binding agent for extracellular IFN such as a viral binding agent for extracellular IFN. In one embodiment, the viral binding agent for extracellular IFN is a viral interferon receptor. In one embodiment, the viral binding agent for extracellular IFN is vaccinia virus B18R. In one embodiment, the viral binding agent for extracellular IFN is provided to the cell in the form of a nucleic acid encoding the binding agent, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the cell together with the RNA which is to be expressed in the cell.
  • the intracellular IFN signalling if not inhibited according to the invention results in inhibition of translation and/or RNA degradation.
  • inhibiting intracellular IFN signalling comprises inhibiting one or more IFN-inducible antivirally active effector proteins.
  • the IFN-inducible antivirally active effector protein is selected from the group consisting of RNA-dependent protein kinase (P R), 2',5'- oligoadenylate synthetase (OAS) and RNaseL.
  • inhibiting intracellular IFN signalling comprises inhibiting the PKR- dependent pathway and/or the OAS-dependent pathway.
  • inhibiting the P R-dependent pathway comprises inhibiting eIF2-alpha phosphorylation. In one embodiment, inhibiting eIF2-alpha phosphorylation comprises inhibiting PKR and/or providing a pseudosubstrate mimicking eIF2-alpha. In one embodiment, the pseudosubstrate mimicking eIF2-alpha is a viral pseudosubstrate mimicking eIF2-alpha. In one embodiment, the viral pseudosubstrate mimicking eIF2-alpha is vaccinia virus K3.
  • the viral pseudosubstrate mimicking eIF2-alpha is provided to the cell in the form of a nucleic acid encoding the viral pseudosubstrate, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the cell together with the RNA which is to be expressed in the cell.
  • inhibiting PKR comprises treating the cell with at least one PKR inhibitor.
  • the PKR inhibitor inhibits RNA-induced PKR autophosphorylation.
  • the PKR inhibitor is an ATP-binding site directed inhibitor of PKR.
  • the PKR inhibitor is an imidazolo-oxindole compound.
  • the PKR inhibitor is 6,8-dihydro-8-(lH-imidazol-5-ylmethylene)-7H- pyrrolo[2,3-g]benzothiazol-7-one and/or 2-aminopurine.
  • the PKR inhibitor is a viral inhibitor of PKR.
  • the viral inhibitor of PKR is vaccinia virus E3.
  • the viral inhibitor of PKR is provided to the cell in the form of a nucleic acid encoding the inhibitor, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the cell together with the RNA which is to be expressed in the cell.
  • inhibiting PKR comprises silencing expression of the PKR gene.
  • inhibiting the OAS-dependent pathway comprises inhibiting activation of RNaseL. In one embodiment, inhibiting the OAS-dependent pathway comprises inhibiting OAS.
  • inhibiting OAS comprises treating the cell with at least one OAS inhibitor.
  • the OAS inhibitor is a viral inhibitor of OAS.
  • the viral inhibitor of OAS is vaccinia virus E3.
  • the viral inhibitor of OAS is provided to the cell in the form of a nucleic acid encoding the inhibitor, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the cell together with the RNA which is to be expressed in the cell.
  • the RNA expressed in the cell is not modified by pseudouridine and/or 5- methylcytidine.
  • the steps of (i) preventing engagement of IFN receptor by extracellular IFN and (ii) inhibiting intracellular IFN signalling result in an enhancement of stability and/or an enhancement of expression of the RNA in the cell compared to the situation where engagement of IFN receptor by extracellular IFN is not prevented and/or intracellular IFN signalling is not inhibited.
  • the enhancement of expression of the RNA in the cell preferably comprises an increase in the level of expression and/or an increase in the duration of expression of the RNA in the cell.
  • the steps of (i) preventing engagement of IFN receptor by extracellular IFN and (ii) inhibiting intracellular IFN signalling result in an enhancement of cell viability compared to the situation where engagement of IFN receptor by extracellular IFN is not prevented and/or intracellular IFN signalling is not inhibited.
  • the steps of (i) preventing engagement of IFN receptor by extracellular IFN and (ii) inhibiting intracellular IFN signalling comprise treating the cell with (i) vaccinia virus B18R and (ii) vaccinia virus E3 or vaccinia virus K3, or both.
  • the vaccinia virus B18R, vaccinia virus E3 and/or vaccinia virus 3 are provided to the cell in the form of nucleic acid encoding the vaccinia virus B18R, vaccinia virus E3 and/or vaccinia virus K.3, either on the same or on two or more different nucleic acid molecules, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the cell together with the RNA which is to be expressed in the cell.
  • the cell is a cell having a barrier function.
  • the cell is a fibroblast, a keratinocyte, an epithelial cell, or an endothelial cell, wherein the endothelial cell preferably is an endothelial cell of the heart, an endothelial cell of the lung, or an umbilical vein endothelial cell.
  • the cell is a human cell.
  • the invention also relates to the use of (i) means which are suitable for preventing engagement of IFN receptor by extracellular IFN and (ii) means which are suitable for inhibiting intracellular IFN signalling, such as the means described herein, for treating a cell in which RNA is to be expressed.
  • means which are suitable for preventing engagement of IFN receptor by extracellular IFN and (ii) means which are suitable for inhibiting intracellular IFN signalling, such as the means described herein, for treating a cell in which RNA is to be expressed.
  • the invention also relates to a method for providing cells having stem cell characteristics comprising the steps of (i) providing a cell population comprising somatic cells, (ii) preventing engagement of IFN receptor of the somatic cells by extracellular IFN, (iii) inhibiting intracellular IFN signalling in the somatic cells, (iv) introducing RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics into the somatic cells, and (v) allowing the development of cells having stem cell characteristics.
  • the method further comprises introducing into the somatic cells miRNA enhancing reprogramming of the somatic cells to cells having stem cell characteristics.
  • the one or more factors comprise OCT4 and SOX2.
  • the one or more factors may further comprise LF4 and/or c-MYC and/or NANOG and/or LIN28.
  • the one or more factors comprise OCT4, SOX2, .LF4 and c-MYC and may further comprise LIN28 and optionally NANOG.
  • the one or more factors comprise OCT4, SOX2, NANOG and LIN28.
  • the method further comprises the step of culturing the somatic cells in the presence of at least one histone deacetylase inhibitor, wherein the at least one histone deacetylase inhibitor preferably comprises valproic acid, sodium butyrate, trichostatin A and/or scriptaid.
  • step (v) comprises culturing the somatic cells under embryonic stem cell culture conditions.
  • the stem cell characteristics comprise an embryonic stem cell morphology.
  • the cells having stem cell characteristics have normal karyotypes, express telomerase activity, express cell surface markers that are characteristic for embryonic stem cells and/or express genes that are characteristic for embryonic stem cells.
  • the cells having stem cell characteristics exhibit a pluripotent state. In one embodiment, the cells having stem cell characteristics have the developmental potential to differentiate into advanced derivatives of all three primary germ layers.
  • the somatic cells are fibroblasts such as lung fibroblasts, foreskin fibroblasts or dermal fibroblasts.
  • the somatic cells are human cells.
  • the RNA is introduced into the somatic cells by electroporation or lipofection.
  • the RNA is introduced into the somatic cells repetitively.
  • the RNA is in vitro transcribed RNA.
  • preventing engagement of IFN receptor by extracellular IFN inhibits autocrine and/or paracrine IFN functions. In one embodiment, preventing engagement of IFN receptor by extracellular IFN comprises providing a binding agent for extracellular IFN such as a viral binding agent for extracellular IFN. In one embodiment, the viral binding agent for extracellular IFN is a viral interferon receptor. In one embodiment, the viral binding agent for extracellular IFN is vaccinia virus B18R.
  • the viral binding agent for extracellular IFN is provided to the somatic cells in the form of a nucleic acid encoding the binding agent, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the somatic cells together with the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics.
  • the nucleic acid is preferably RNA which preferably is or has been introduced into the somatic cells together with the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics.
  • the intracellular IFN signalling if not inhibited according to the invention results in inhibition of translation and/or RNA degradation.
  • inhibiting intracellular IFN signalling comprises inhibiting one or more IFN-inducible antivirally active effector proteins.
  • the IFN-inducible antivirally active effector protein is selected from the group consisting of RNA-dependent protein kinase (P R), 2',5'- oligoadenylate synthetase (OAS) and RNaseL.
  • inhibiting intracellular IFN signalling comprises inhibiting the PKR- dependent pathway and/or the OAS-dependent pathway.
  • inhibiting the P R-dependent pathway comprises inhibiting eIF2-alpha phosphorylation. In one embodiment, inhibiting eIF2-alpha phosphorylation comprises inhibiting PKR and/or providing a pseudosubstrate mimicking eIF2-alpha. In one embodiment, the pseudosubstrate mimicking eIF2-alpha is a viral pseudosubstrate mimicking eIF2-alpha. In one embodiment, the viral pseudosubstrate mimicking eIF2-alpha is vaccinia virus K3.
  • the viral pseudosubstrate mimicking eIF2-alpha is provided to the somatic cells in the form of a nucleic acid encoding the viral pseudosubstrate, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the somatic cells together with the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics.
  • the nucleic acid is preferably RNA which preferably is or has been introduced into the somatic cells together with the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics.
  • inhibiting P R comprises treating the cell with at least one P R inhibitor.
  • the PKR inhibitor inhibits RNA-induced PKR autophosphorylation.
  • the PKR inhibitor is an ATP -binding site directed inhibitor of PKR.
  • the PKR inhibitor is an imidazolo-oxindole compound.
  • the PKR inhibitor is 6,8-dihydro-8-(lH-imidazol-5-ylmethylene)-7H- pyrrolo[2,3-g]benzothiazol-7-one and/or 2-aminopurine.
  • the PKR inhibitor is a viral inhibitor of PKR.
  • the viral inhibitor of PKR is vaccinia virus E3.
  • the viral inhibitor of PKR is provided to the somatic cells in the form of a nucleic acid encoding the inhibitor, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the somatic cells together with the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics.
  • inhibiting PKR comprises silencing expression of the PKR gene.
  • inhibiting the OAS-dependent pathway comprises inhibiting activation of RNaseL.
  • inhibiting the OAS-dependent pathway comprises inhibiting OAS.
  • inhibiting OAS comprises treating the somatic cells with at least one OAS inhibitor.
  • the OAS inhibitor is a viral inhibitor of OAS.
  • the viral inhibitor of OAS is vaccinia virus E3.
  • the viral inhibitor of OAS is provided to the somatic cells in the form of a nucleic acid encoding the inhibitor, wherein the nucleic acid is preferably RNA which preferably is or has been introduced into the somatic cells together with the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics.
  • the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics is not modified by pseudouridine and/or 5-methylcytidine.
  • the steps of (i) preventing engagement of IFN receptor by extracellular IFN and (ii) inhibiting intracellular IFN signalling result in an enhancement of stability and/or an enhancement of expression of the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics in the somatic cells compared to the situation where engagement of IFN receptor by extracellular IFN is not prevented and/or intracellular IFN signalling is not inhibited.
  • the enhancement of expression of the RNA in the somatic cells preferably comprises an increase in the level of expression and/or an increase in the duration of expression of the RNA in the somatic cells.
  • the steps of (i) preventing engagement of IFN receptor by extracellular IFN and (ii) inhibiting intracellular IFN signalling result in an enhancement of cell viability compared to the situation where engagement of IFN receptor by extracellular IFN is not prevented and/or intracellular IFN signalling is not inhibited.
  • the steps of (i) preventing engagement of IFN receptor by extracellular IFN and (ii) inhibiting intracellular IFN signalling comprise treating the somatic cells with (i) vaccinia virus B18R and (ii) vaccinia virus E3 or vaccinia virus 3, or both.
  • the vaccinia virus B18R, vaccinia virus E3 and/or vaccinia virus K.3 are provided to the somatic cells in the form of nucleic acid encoding the vaccinia virus B18R, vaccinia virus E3 and/or vaccinia virus K.3, either on the same or on two or more different nucleic acid molecules, wherein the nucleic acid is preferably RNA which preferably is introduced or has been introduced into the somatic cells together with the RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics.
  • the invention also relates to a method for providing differentiated cell types comprising the steps of (i) providing cells having stem cell characteristics using the method for providing cells having stem cell characteristics according to the invention, and (ii) culturing the cells having stem cell characteristics under conditions that induce or direct partial or complete differentiation to a differentiated cell type.
  • the invention also relates to a composition comprising (i) an agent useful for preventing engagement of IFN receptor by extracellular IFN and (ii) an agent useful for inhibiting intracellular IFN signalling.
  • a kit comprising the composition of the invention.
  • Various embodiments of the composition or kit of the invention are described above for the methods of the invention. In one embodiment, the composition or kit of the invention is useful in the methods of the invention.
  • the composition or kit of the invention comprises RNA to be introduced into a cell for expression, e.g. RNA capable of expressing one or more factors allowing the reprogramming of somatic cells to cells having stem cell characteristics as described above.
  • an agent useful for preventing engagement of IFN receptor by extracellular IFN inhibits autocrine and/or paracrine IFN functions.
  • an agent useful for preventing engagement of IFN receptor by extracellular IFN comprises a binding agent for extracellular IFN such as a viral binding agent for extracellular IFN.
  • the viral binding agent for extracellular IFN is a viral interferon receptor.
  • the viral binding agent for extracellular IFN is vaccinia virus B18R.
  • the viral binding agent for extracellular IFN is present in the form of a nucleic acid encoding the binding agent, wherein the nucleic acid is preferably RNA.
  • an agent useful for inhibiting intracellular IFN signalling comprises an agent inhibiting one or more IFN-inducible antivirally active effector proteins.
  • the IFN-inducible antivirally active effector protein is selected from the group consisting of RNA-dependent protein kinase (P R), 2',5'-oligoadenylate synthetase (OAS) and RNaseL.
  • an agent useful for inhibiting intracellular IFN signalling comprises an agent useful for inhibiting the PKR-dependent pathway and/or the OAS-dependent pathway.
  • an agent useful for inhibiting the PKR-dependent pathway comprises an agent useful for inhibiting eIF2-alpha phosphorylation.
  • an agent useful for inhibiting eIF2-alpha phosphorylation comprises an agent useful for inhibiting PKR and/or a pseudosubstrate mimicking eIF2-alpha.
  • the pseudosubstrate mimicking eIF2-alpha is a viral pseudosubstrate mimicking eIF2-alpha.
  • the viral pseudosubstrate mimicking eIF2-alpha is vaccinia virus K3.
  • the viral pseudosubstrate mimicking eIF2-alpha is present in the form of a nucleic acid encoding the viral pseudosubstrate, wherein the nucleic acid is preferably RNA.
  • an agent useful for inhibiting PKR comprises at least one PKR inhibitor.
  • the PKR inhibitor inhibits RNA-induced PKR autophosphorylation.
  • the PKR inhibitor is an ATP-binding site directed inhibitor of PKR.
  • the PKR inhibitor is an imidazolo-oxindole compound.
  • the PKR inhibitor is 6,8-dihydro-8-(lH-imidazol-5-ylmethylene)-7H-pyrrolo[2,3-g]benzothiazol- 7-one and/or 2-aminopurine.
  • the PKR inhibitor is a viral inhibitor of PKR.
  • the viral inhibitor of PKR is vaccinia virus E3. In one embodiment, the viral inhibitor of PKR is present in the form of a nucleic acid encoding the inhibitor, wherein the nucleic acid is preferably RNA. In one embodiment, an agent useful for inhibiting PKR comprises an agent useful for silencing expression of the PKR gene.
  • an agent useful for inhibiting the OAS-dependent pathway comprises an agent useful for inhibiting activation of RNaseL. In one embodiment, an agent useful for inhibiting the OAS-dependent pathway comprises an agent useful for inhibiting OAS.
  • an agent useful for inhibiting OAS comprises at least one OAS inhibitor.
  • the OAS inhibitor is a viral inhibitor of OAS.
  • the viral inhibitor of OAS is vaccinia virus E3.
  • the viral inhibitor of OAS is present in the form of a nucleic acid encoding the inhibitor, wherein the nucleic acid is preferably RNA.
  • an agent useful for preventing engagement of IFN receptor by extracellular IFN and an agent useful for inhibiting intracellular IFN signalling comprise (i) vaccinia virus B18R and (ii) vaccinia virus E3 or vaccinia virus K3, or both.
  • the vaccinia virus B18R, vaccinia virus E3 and/or vaccinia virus K3 are present in the form of nucleic acid encoding the vaccinia virus B 18R, vaccinia virus E3 and/or vaccinia virus 3, either on the same or on two or more different nucleic acid molecules, wherein the nucleic acid is preferably RNA.
  • Fig. 1 Viral escape mechanism
  • Viruses have evolved many escape mechanism that are mediated by viral proteins or viral nucleic acids. RNA that codes for these viral escape proteins can easily be co-transfered with RNA coding for rTF.
  • Antagonistic protein E3 Vaccinia virus
  • P R & OAS P R & OAS
  • 3 Vaccinia virus
  • B18R Vaccinia virus
  • IFN IFN
  • E3 and K3 are acting intracellular
  • B18R protein coded by IVT-RNA is secreted from the cell where it binds extracellular type I IFNs and prevents engagement of IFN receptors.
  • Fig. 2 Repetitive transfer of IVT-RNA (Reprogramming-TF)
  • CCD1079Sk fibroblasts were electroporated as indicated in the side panel either with 15 ⁇ g or 5 ⁇ g of each in vitro transcribed (IVT)-RNA encoding the transcription factors OCT4 (O), SOX2 (S), LF4 ( ) and cMYC (M) and cultivated in human embryonic stem (ES) cell medium. Electroporations were performed in 4 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts.
  • CCD1079Sk fibroblasts were electroporated as indicated in the side panel with 15 g of each IVT-RNA encoding the transcription factors OSKM and cultivated in human ES cell medium. Electroporations were performed in 4 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts. At the indicated time points remaining cells were counted and survival rate in relation to the starting cells was calculated.
  • CCD1079Sk fibroblasts were electroporated with 1 ⁇ g IVT RNA encoding for firefly luciferase (Luc) and 5 ⁇ g IVT RNA encoding for green fluorescent protein (GFP). Electroporations were performed in 2 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts. 24h post electroporation, cells were pelleted, total RNA was isolated and mRNA-expression of Interferon (IFN)-a and -b was quantified by qRT-PCR.
  • IFN Interferon
  • CCD1079Sk fibroblasts were electroporated with 33,4 ⁇ g IVT RNA encoding reprogramming mixture (29,5 ⁇ & rTF (OSKM NANOG (N) LIN28 (L) ( 1 : 1 : 1 : 1 : 1 : 1 )), l ⁇ g SV40 largeT antigen (lgT), l ⁇ g HTLV E6 and l ,25 g GFP). Electroporations were performed in 4 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts.
  • CCD 1079Sk fibroblasts were electroporated once with the indicated amounts of IVT-RNA encoding the reporter genes Luc, GFP or the Protein Kinase R (PKR) wild type. Electroporations were performed in 4 mm gap cuvettes using optimized parameters for CCD 1079Sk fibroblasts. Cells were lysed 24h post electroporation and expression and phosphorylation status of the PKR target eukaryotic initiation factor 2a (eIF2a) was monitored by Western Blotting using specific antibodies.
  • eIF2a eukaryotic initiation factor 2a
  • Fig. 3 Use of E3, K3 and B18R in RNA-based gene transfer
  • AB CCD1079Sk fibroblasts were plated into 6 wells (100,000 cells/well) and lipofected the next day using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • IVT- RNA encoding for Luc was used to sum up the mixtures to l ⁇ g. Lipofections were performed according to the manufacturers instructions and cells were harvested 48h post transfection.
  • C CCD1079Sk fibroblasts were plated into 6 wells (100.000 cells/well) and lipofected the next four consecutive days using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • the IVT-RNA mixture was thereby composed of 0 ⁇ g GFP with 0 ⁇ g of each B18R, E3 or K3 (as indicated).
  • IVT-RNA encoding for Luc was used to sum up the mixture to 1 ⁇ g total IVT-RNA.
  • RNAs were composed of 100% pseudouridine (psi) and 100% 5-methylcytidine (5mC) instead of uridine and cytidine which display less immunstimulatory characteristics. Lipofections were performed according to the manufacturers instructions. 24h after the last lipofection, cell viability was assayed using the Cell Proliferation Kit II (Roche) and normalized to the mock transfected cells.
  • Fig. 4 Use of E3, K3 and B18R in RNA-based gene transfer for reprogramming
  • CCD1079Sk fibroblasts were plated into 6 wells (80,000 cells/well) and lipofected the next four consecutive days using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • the IVT-RNA mixtures were thereby composed of 0 ⁇ g unmodified GFP or 0 ⁇ g OSKMNL (1 : 1 : 1 : 1 :1 : 1) either unmodified or modified and either with O ⁇ g of each B18R, E3 and K3 unmodified or modified. If necessary IVT-RNA encoding for Luc was used to sum up the mixture to 1 ⁇ g total IVT-RNA.
  • RNAs were composed of 100% psi and 100% 5mC instead of uridine and cytidine which display less immunstimulatory characteristics. Lipofections were performed according to the manufacturers instructions. 24h after the last lipofection, cell viability was assayed using the Cell Proliferation Kit II (Roche) with normalization to mock transfected cells (A) and by microscopy (B). After that, cells were pelleted, total RNA was isolated and mRNA-expression of IFNb and OAS1 was quantified by qRT-PCR (C).
  • Fig. 5 Translation of rTF after repetitive lipofection in the presence of E3, K3 and B18R
  • CCD1079Sk fibroblasts were plated into 6 wells (100,000 cells/well) and lipofected the next three consecutive days using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • the IVT-RNA mixtures was thereby composed of 0 ⁇ g GFP with 0 ⁇ g OCT4 or SOX2 or NANOG either unmodified or modified and 0 ⁇ g of each B18R, E3 and K3 (EKB) either unmodified or modified.
  • Modified RNAs were composed of 100% psi and 100% 5mC instead of uridine and cytidine which display less immunstimulatory characteristics. Lipofections were performed according to the manufacturers instructions. 24h after the last lipofection, intracellular expression of OSN was monitored by FACS analysis using the human pluripotent stem cell transcription factor analysis kit (BD 560589).
  • Fig. 6 Reprogramming of HFF using rTF and microRNA in the presence of EKB
  • HFF fibroblasts (System Bioscience) were plated into 6 wells (100,000 cells/well) and lipofected 5 times a week (Monday to Friday) for two weeks using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT-RNA (A).
  • the IVT-RNA mixtures were thereby composed of 0 ⁇ g unmodified GFP or 0 ⁇ g OSKMNL (1 : 1 : 1 : 1 : 1 : 1) either unmodified or modified with either 0 ⁇ g of each B 18R, E3 and K3 (EKB) either unmodified or modified and 0 ⁇ g of a miRNA mixture composed of miRNAs 302a-d and 367 [0.4 ⁇ each].
  • RNAs were composed of 100% psi and 100% 5mC instead of uridine and cytidine which display less immunstimulatory characteristics.
  • Lipofections in stem cell media were performed according to the manufacturers instructions.
  • stem cell media Nutristem media, Stemgent
  • Fig. 7 Reprogramming of HFF using rTF and micro R A in the presence of EKB (splitting 1:8)
  • HFF fibroblasts (System Bioscience) were plated into 6 wells ( 100,000 cells/well) and lipofected 5 times a week (Monday to Friday) for two weeks using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT-RNA (A).
  • the IVT-RNA mixtures were thereby composed of 0 ⁇ g OSKMNL (1 : 1 : 1 : 1 : 1 : 1) either unmodified or modified with either 0 ⁇ g of each B18R, E3 and K3 (EKB) unmodified or modified and 0 ⁇ g of a miRNA mixture composed of miRNAs 302a-d and 367 [0.4 ⁇ each].
  • RNAs were composed of 100% psi and 100% 5mC instead of uridine and cytidine which display less immunstimulatory characteristics.
  • Lipofections in stem cell media were performed according to the manufacturers instructions. On day 5 and day 12, cells were pelleted, total RNA was isolated and mRNA-expression of the human ES-marker TERT, DPPA4, GDF3, LIN28 (endogenous) and REX1 was quantified by qRT-PCR (B).
  • Colony growth was observed by microscopy and for further analysis, colonies were stained for the ES surface marker TRA-1-60 using the StainAlive TRA-1-60 antibody (Stemgent) (C) or for the activity of alkaline phosphatase (Vector Red staining kit) following the manufacturers instructions (D).
  • C StainAlive TRA-1-60 antibody
  • D Vector Red staining kit
  • HFF fibroblasts (System Bioscience) were plated into 6 wells (100,000 cells/well) and lipofected the next four consecutive days using 6 ⁇ 1 RNAiMAX (Invitrogen) and 1 ⁇ g IVT.
  • the IVT-RNA mixtures were thereby composed of 0 ⁇ g unmodified OSKMNL (1 : 1 : 1 : 1 : 1 : 1 ) with variable amounts of unmodfied B18R, E3 and K3 as indicated.
  • IVT-RNA encoding for Luc was used to sum up the mixture to 1.4 ⁇ total IVT-RNA. According to the reprogramming experiments 0 ⁇ g of a miRNA mixture composed of miRNAs 302a-d and 367 [0.4 ⁇ each] was added to the samples.
  • 1.4pg modified (mod.) IVT-RNA encoding for Luc (0 ⁇ g) and OSKMNL (0 ⁇ g; 1 : 1 : 1 : 1 : 1 : 1 ) was used.
  • These RNAs were composed of 100% psi and 5mC instead of uridine and cytidine which display less immunstimulatory characteristics.
  • Lipofections were performed according to the manufacturers instructions. 24h after the last lipofection, cell viability was assayed using the Cell Proliferation Kit II (Roche) (A). After that, cells were pelleted, total RNA was isolated and mRNA-expression of IFNb and OAS1 was quantified by qRT-PCR (B).
  • CCD1079SK fibroblasts were electroporated with IVT RNA encoding Luc (1 ⁇ g), GFP (5 ⁇ ) and 3 ⁇ g of E3 or K3 or both as indicated. Electroporations were performed in 2 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts.
  • RNA comprises a poly(A)-tail consisting of 120 nucleotides and in another preferred embodiment the RNA molecule comprises a 5 '-cap analog, then in a preferred embodiment, the RNA comprises the poly(A)- tail consisting of 120 nucleotides and the 5'-cap analog.
  • the terms used herein are defined as described in "A multilingual glossary of biotechnological terms: (IUPAC Recommendations)", H.G.W. Leuenberger, B. Nagel, and H. olbl, Eds., Helvetica Chimica Acta, CH-4010 Basel, Switzerland, (1995).
  • Terms such as “increasing”, “enhancing”, or “prolonging” preferably relate to an increase, enhancement, or prolongation by about at least 10%, preferably at least 20%, preferably at least 30%, preferably at least 40%, preferably at least 50%, preferably at least 80%, preferably at least 100%, preferably at least 200% and in particular at least 300%. These terms may also relate to an increase, enhancement, or prolongation from zero or a non-measurable or non- detectable level to a level of more than zero or a level which is measurable or detectable.
  • recombinant in the context of the present invention means "made through genetic engineering".
  • a "recombinant entity” such as a recombinant protein in the context of the present invention is not occurring naturally, and preferably is a result of a combination of entities such as amino acid or nucleic acid sequences which are not combined in nature.
  • a recombinant protein in the context of the present invention may contain several amino acid sequences derived from different proteins fused together, e.g., by peptide bonds.
  • naturally occurring refers to the fact that an object can be found in nature.
  • a protein or nucleic acid that is present in an organism (including viruses) and can be isolated from a source in nature and which has not been intentionally modified by man in the laboratory is naturally occurring.
  • a nucleic acid is according to the invention preferably deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), more preferably RNA, most preferably in vitro transcribed RNA (IVT RNA).
  • Nucleic acids include according to the invention genomic DNA, cDNA, mRNA, recombinantly produced and chemically synthesized molecules.
  • a nucleic acid may be present as a single-stranded or double-stranded and linear or covalently circularly closed molecule.
  • a nucleic acid can, according to the invention, be isolated.
  • isolated nucleic acid means, according to the invention, that the nucleic acid (i) was amplified in vitro, for example via polymerase chain reaction (PCR), (ii) was produced recombinantly by cloning, (iii) was purified, for example, by cleavage and separation by gel electrophoresis, or (iv) was synthesized, for example, by chemical synthesis.
  • a nucleic can be employed for introduction into, i.e. transfection of, cells, in particular, in the form of RNA which can be prepared by in vitro transcription from a DNA template.
  • the RNA can moreover be modified before application by stabilizing sequences, capping, and polyadenylation.
  • nucleic acid for expression of more than one peptide or protein
  • RNA for expression of more than one peptide or protein
  • either of a nucleic acid type in which the different peptides or proteins are present in different nucleic acid molecules or a nucleic acid type in which the peptides or proteins are present in the same nucleic acid molecule can be used.
  • RNA relates to a molecule which comprises ribonucleotide residues and preferably being entirely or substantially composed of ribonucleotide residues.
  • “Ribonucleotide” relates to a nucleotide with a hydroxyl group at the 2'-position of a ⁇ -D-ribofuranosyl group.
  • the term “RNA” comprises double-stranded RNA, single-stranded RNA, isolated RNA such as partially or completely purified RNA, essentially pure RNA, synthetic RNA, and recombinantly generated RNA such as modified RNA which differs from naturally occurring RNA by addition, deletion, substitution and/or alteration of one or more nucleotides.
  • Such alterations can include addition of non-nucleotide material, such as to the end(s) of a RNA or internally, for example at one or more nucleotides of the RNA.
  • Nucleotides in RNA molecules can also comprise non-standard nucleotides, such as non-naturally occurring nucleotides or chemically synthesized nucleotides or deoxynucleotides. These altered RNAs can be referred to as analogs or analogs of naturally- occurring RNA.
  • RNA includes and preferably relates to "mRNA".
  • mRNA means "messenger-RNA” and relates to a "transcript” which is generated by using a DNA template and encodes a peptide or protein.
  • an mRNA comprises a 5'-UTR, a protein coding region, and a 3'-UTR.
  • mRNA only possesses limited half-life in cells and in vitro.
  • mRNA may be generated by in vitro transcription from a DNA template.
  • the in vitro transcription methodology is known to the skilled person. For example, there is a variety of in vitro transcription kits commercially available.
  • RNA may be stabilized and its translation increased by one or more modifications having a stabilizing effects and/or increasing translation efficiency of RNA.
  • modifications are described, for example, in PCT/EP2006/009448 incorporated herein by reference.
  • the RNA used according to the present invention it may be modified within the coding region, i.e. the sequence encoding the expressed peptide or protein, preferably without altering the sequence of the expressed peptide or protein, so as to increase the GC-content to increase mRNA stability and to perform a codon optimization and, thus, enhance translation in cells.
  • modification in the context of the RNA used in the present invention includes any modification of an RNA which is not naturally present in said RNA.
  • the RNA used according to the invention does not have uncapped 5'-triphosphates. Removal of such uncapped 5'-triphosphates can be achieved by treating RNA with a phosphatase.
  • RNA according to the invention may have modified ribonucleotides in order to increase its stability and/or decrease cytotoxicity.
  • 5-methylcytidine is substituted partially or completely, preferably completely, for cytidine.
  • pseudouridine is substituted partially or completely, preferably completely, for uridine.
  • the term "modification” relates to providing an RNA with a 5'-cap or 5'- cap analog.
  • the term “5'-cap” refers to a cap structure found on the 5'-end of an mRNA molecule and generally consists of a guanosine nucleotide connected to the mRNA via an unusual 5' to 5' triphosphate linkage. In one embodiment, this guanosine is methylated at the 7-position.
  • the term “conventional 5'-cap” refers to a naturally occurring RNA 5'-cap, preferably to the 7-methylguanosine cap (m 7 G).
  • 5'-cap includes a 5'-cap analog that resembles the RNA cap structure and is modified to possess the ability to stabilize RNA and/or enhance translation of RNA if attached thereto, preferably in vivo and/or in a cell.
  • the 5' end of the RNA includes a Cap structure having the following general formula:
  • Ri and R 2 are independently hydroxy or methoxy and W “ , X “ and Y “ are independently oxygen, sulfur, selenium, or BH 3 .
  • Ri and R 2 are hydroxy and W “ , X “ and Y “ are oxygen.
  • one of Ri and R 2 preferably R ⁇ is hydroxy and the other is methoxy and W “ , X “ and Y “ are oxygen.
  • and R 2 are hydroxy and one of W ⁇ X " and Y " , preferably X " is sulfur, selenium, or BH 3 , preferably sulfur, while the other are oxygen.
  • one of Ri and R 2 preferably R 2 is hydroxy and the other is methoxy and one of W “ , X “ and Y “ , preferably X " is sulfur, selenium, or BH 3 , preferably sulfur while the other are oxygen.
  • the nucleotide on the right hand side is connected to the RNA chain through its 3' group.
  • the Cap structure having the above structure wherein Ri is methoxy, R 2 is hydroxy, X " is sulfur and W " and Y " are oxygen exists in two diastereoisomeric forms (Rp and Sp).
  • Rp and Sp diastereoisomeric forms
  • Dl and D2 diastereoisomeric forms
  • the Dl isomer of m 2 7 ' 2 " °GppspG is particularly preferred.
  • RNA with a 5'-cap or 5'-cap analog may be achieved by in vitro transcription of a DNA template in presence of said 5'-cap or 5'-cap analog, wherein said 5'-cap is co- transcriptionally incorporated into the generated RNA strand, or the RNA may be generated, for example, by in vitro transcription, and the 5'-cap may be attached to the RNA post- transcriptionally using capping enzymes, for example, capping enzymes of vaccinia virus.
  • RNA may comprise further modifications.
  • a further modification of the RNA used in the present invention may be an extension or truncation of the naturally occurring poly(A) tail or an alteration of the 5'- or 3 '-untranslated regions (UTR) such as introduction of a UTR which is not related to the coding region of said RNA, for example, the exchange of the existing 3 -UTR with or the insertion of one or more, preferably two copies of a 3'-UTR derived from a globin gene, such as alpha2-globin, alpha 1 -globin, beta-globin, preferably beta-globin, more preferably human beta-globin.
  • UTR 5'- or 3 '-untranslated regions
  • RNA having an unmasked poly-A sequence is translated more efficiently than RNA having a masked poly-A sequence.
  • poly(A) tail or "poly-A sequence” relates to a sequence of adenyl (A) residues which typically is located on the 3 '-end of a RNA molecule and "unmasked poly-A sequence” means that the poly-A sequence at the 3' end of an RNA molecule ends with an A of the poly-A sequence and is not followed by nucleotides other than A located at the 3' end, i.e. downstream, of the poly-A sequence.
  • a long poly-A sequence of about 120 base pairs results in an optimal transcript stability and translation efficiency of RNA.
  • the RNA used according to the present invention may be modified so as to be present in conjunction with a poly-A sequence, preferably having a length of 10 to 500, more preferably 30 to 300, even more preferably 65 to 200 and especially 100 to 150 adenosine residues.
  • the poly-A sequence has a length of approximately 120 adenosine residues.
  • the poly-A sequence can be unmasked.
  • incorporation of a 3 '-non translated region (UTR) into the 3 '-non translated region of an RNA molecule can result in an enhancement in translation efficiency.
  • a synergistic effect may be achieved by incorporating two or more of such 3 '-non translated regions.
  • the 3 '-non translated regions may be autologous or heterologous to the RNA into which they are introduced.
  • the 3 '-non translated region is derived from the human ⁇ -globin gene.
  • RNA relates to the "half-life" of RNA.
  • "Half-life” relates to the period of time which is needed to eliminate half of the activity, amount, or number of molecules.
  • the half-life of an RNA is indicative for the stability of said RNA.
  • the half-life of RNA may influence the "duration of expression" of the RNA. It can be expected that RNA having a long half-life will be expressed for an extended time period.
  • RNA if according to the present invention it is desired to decrease stability and/or translation efficiency of RNA, it is possible to modify RNA so as to interfere with the function of elements as described above increasing the stability and/or translation efficiency of RNA.
  • expression is used according to the invention in its most general meaning and comprises the production of RNA and/or peptides or proteins, e.g. by transcription and/or translation.
  • expression or “translation” relates in particular to the production of peptides or proteins. It also comprises partial expression of nucleic acids. Moreover, expression can be transient or stable.
  • RNA expression refers to the production of peptide or protein encoded by the RNA.
  • RNA expression refers to the translation of RNA so as to express, i.e. produce peptide or protein encoded by the RNA.
  • the term “transcription” relates to a process, wherein the genetic code in a DNA sequence is transcribed into RNA. Subsequently, the RNA may be translated into protein.
  • the term “transcription” comprises "in vitro transcription", wherein the term “in vitro transcription” relates to a process wherein RNA, in particular mRNA, is in vitro synthesized in a cell-free system, preferably using appropriate cell extracts.
  • cloning vectors are applied for the generation of transcripts. These cloning vectors are generally designated as transcription vectors and are according to the present invention encompassed by the term "vector".
  • the RNA used in the present invention preferably is in vitro transcribed RNA (IVT-RNA) and may be obtained by in vitro transcription of an appropriate DNA template.
  • the promoter for controlling transcription can be any promoter for any RNA polymerase.
  • RNA polymerases are the T7, T3, and SP6 RNA polymerases.
  • the in vitro transcription according to the invention is controlled by a T7 or SP6 promoter.
  • a DNA template for in vitro transcription may be obtained by cloning of a nucleic acid, in particular cDNA, and introducing it into an appropriate vector for in vitro transcription.
  • the cDNA may be obtained by reverse transcription of RNA.
  • the cDNA containing vector template may comprise vectors carrying different cDNA inserts which following transcription results in a population of different RNA molecules optionally capable of expressing different peptides or proteins or may comprise vectors carrying only one species of cDNA insert which following transcription only results in a population of one RNA species capable of expressing only one peptide or protein.
  • RNA capable of expressing a single peptide or protein only or to produce compositions of different RNAs such as RNA libraries and whole-cell RNA capable of expressing more than one peptide or protein, e.g. a composition of peptides or proteins.
  • the present invention envisions the introduction of all such RNA into cells.
  • translation relates to the process in the ribosomes of a cell by which a strand of messenger RNA directs the assembly of a sequence of amino acids to make a peptide or protein.
  • Expression control sequences or regulatory sequences which according to the invention may be linked functionally with a nucleic acid, can be homologous or heterologous with respect to the nucleic acid.
  • a coding sequence and a regulatory sequence are linked together "functionally” if they are bound together covalently, so that the transcription or translation of the coding sequence is under the control or under the influence of the regulatory sequence. If the coding sequence is to be translated into a functional protein, with functional linkage of a regulatory sequence with the coding sequence, induction of the regulatory sequence leads to a transcription of the coding sequence, without causing a reading frame shift in the coding sequence or inability of the coding sequence to be translated into the desired protein or peptide.
  • control sequence comprises, according to the invention, promoters, ribosome-binding sequences and other control elements, which control the transcription of a nucleic acid or the translation of the derived RNA.
  • the regulatory sequences can be controlled.
  • the precise structure of regulatory sequences can vary depending on the species or depending on the cell type, but generally comprises 5'-untranscribed and 5'- and 3 '-untranslated sequences, which are involved in the initiation of transcription or translation, such as TATA-box, capping- sequence, CAAT-sequence and the like.
  • 5'-untranscribed regulatory sequences comprise a promoter region that includes a promoter sequence for transcriptional control of the functionally bound gene.
  • Regulatory sequences can also comprise enhancer sequences or upstream activator sequences.
  • Terms such as “enhancement of expression”, “enhanced expression” or “increased expression” mean in the context of the present invention that the amount of peptide or protein expressed by a given number of RNA molecules is higher than the amount of peptide or protein expressed by the same number of RNA molecules, wherein expression of the RNA molecules is performed under the same conditions except the condition which results in the enhanced or increased expression of the RNA, such as preventing engagement of IFN receptor by extracellular IFN and inhibiting intracellular IFN signalling in a cell versus not preventing engagement of IFN receptor by extracellular IFN and not inhibiting intracellular IFN signalling in a cell.
  • “same conditions” refer to a situation wherein the same RNA sequences encoding the same peptide or protein are introduced by the same means into the same cells, the cells are cultured under the same conditions (except the condition which results in the enhanced or increased expression) and the amount of peptide or protein is measured by the same means.
  • the amount of peptide or protein may be given in moles, or by weight, e.g. in grams, or by mass or by polypeptide activity, e.g. if the peptide or protein is an enzyme it may be given as catalytic activity or if the peptide or protein is an antibody or antigen or a receptor it may be given as binding affinity.
  • terms such as “enhancement of expression”, “enhanced expression” or “increased expression” mean in the context of the present invention that the amount of peptide or protein expressed by a given number of RNA molecules and within a given period of time is higher than the amount of peptide or protein expressed by the same number of RNA molecules and within the same period of time.
  • the maximum value of peptide or protein expressed by a given number of RNA molecules at a particular time point may be higher than the maximum value of peptide or protein expressed by the same number of RNA molecules.
  • the maximum value of peptide or protein expressed by a given number of RNA molecules does not need to be higher than the maximum value of peptide or protein expressed by the same number of RNA molecules, however, the average amount of peptide or protein expressed by the given number of RNA molecules within a given period of time may be higher than the average amount of peptide or protein expressed by the same number of RNA molecules.
  • the latter cases are referred to herein as "higher level of expression” or “increased level of expression” and relate to higher maximum values of expression and/or higher average values of expression.
  • RNA molecules may be longer than the time in which the peptide or protein is expressed by the same RNA molecules.
  • terms such as “enhancement of expression”, “enhanced expression” or “increased expression” mean in the context of the present invention also that the amount of peptide or protein expressed by a given number of RNA molecules is higher than the amount of peptide or protein expressed by the same number of RNA molecules since the period of time in which the RNA is stably present and expressed is longer than the period of time in which the same number of RNA molecules is stably present and expressed. These cases are referred to herein also as “increased duration of expression”.
  • such longer time periods refer to expression for at least 48 h, preferably for at least 72 h, more preferably for at least 96 h, in particular for at least 120 h or even longer following introduction of RNA or following the first introduction (e.g. in case of repeated transfections) of RNA into a cell.
  • the level of expression and/or duration of expression of RNA may be determined by measuring the amount, such as the total amount expressed and/or the amount expressed in a given time period, and/or the time of expression of the peptide or protein encoded by the RNA, for example, by using an ELISA procedure, an immunohistochemistry procedure, a quantitative image analysis procedure, a Western Blot, mass spectrometry, a quantitative immunohistochemistry procedure, or an enzymatic assay.
  • the RNA according to the invention comprises a population of different RNA molecules, e.g. a mixture of different RNA molecules optionally encoding different peptides and/or protein, whole-cell RNA, an RNA library, or a portion of thereof, e.g. a library of RNA molecules expressed in a particular cell type, such as undifferentiated cells, in particular stem cells such as embryonic stem cells, or a fraction of the library of RNA molecules such as RNA with enriched expression in undifferentiated cells, in particular stem cells such as embryonic stem cells relative to differentiated cells.
  • a population of different RNA molecules e.g. a mixture of different RNA molecules optionally encoding different peptides and/or protein, whole-cell RNA, an RNA library, or a portion of thereof, e.g. a library of RNA molecules expressed in a particular cell type, such as undifferentiated cells, in particular stem cells such as embryonic stem cells, or a fraction of the library of RNA molecules such as RNA
  • RNA may include a mixture of RNA molecules, whole-cell RNA or a fraction thereof, which may be obtained by a process comprising the isolation of RNA from cells and/or by recombinant means, in particular by in vitro transcription.
  • the RNA to be expressed in a cell is introduced into said cell, either in vitro or in vivo, preferably in vitro.
  • RNA may be introduced into a cell either prior to, after and/or simultaneously with preventing engagement of IFN receptor by extracellular IFN and/or inhibiting intracellular IFN signalling in the cell.
  • engagement of IFN receptor by extracellular IFN is prevented and intracellular IFN signalling is inhibited as long as expression of the RNA in the cell is desired.
  • the RNA that is to be introduced into a cell is obtained by in vitro transcription of an appropriate DNA template.
  • the RNA used according to the present invention may have a known composition (in this embodiment it is preferably known which peptides or proteins are being expressed by the RNA) or the composition of the RNA may be partially or entirely unknown.
  • the RNA used according to the present invention may have a known function or the function of the RNA may be partially or entirely unknown.
  • RNA capable of expressing and "RNA encoding” are used interchangeably herein and with respect to a particular peptide or protein mean that the RNA, if present in the appropriate environment, preferably within a cell, can be expressed to produce said peptide or protein.
  • RNA according to the invention is able to interact with the cellular translation machinery to provide the peptide or protein it is capable of expressing.
  • RNA may be introduced into cells either in vitro or in vivo, preferably in vitro.
  • the cells into which the RNA has been introduced in vitro may, preferably following expression of the RNA in vitro by the methods of the invention, be administered to a patient.
  • transferring refers to the introduction of nucleic acids, in particular exogenous or heterologous nucleic acids, in particular RNA, into a cell.
  • Said terms also include the repetitive introduction of nucleic acids, in particular RNA, into a cell, wherein repetitive mean more than once, e.g. two times or more, three times or more, four times or more, five times or more, six times or more, seven times or more, eight times or more.
  • the time interval between said repetitive introductions of nucleic acids may be 3 days or less, 2 days or less, 24 hours or less or even lower.
  • a cell can be an isolated cell or it can form part of an organ, a tissue and/or an organism.
  • any technique which is suitable to introduce RNA into cells may be used.
  • the RNA is introduced into cells by standard techniques. Such techniques comprise transfection of nucleic acid calcium phosphate precipitates, transfection of nucleic acids which are associated with DEAE, the transfection or infection with viruses which carry the nucleic acids of interest, electroporation, lipofection, and microinjection.
  • the administration of a nucleic acid is either achieved as naked nucleic acid or in combination with an administration reagent.
  • administration of nucleic acids is in the form of naked nucleic acids.
  • the RNA is administered in combination with stabilizing substances such as RNase inhibitors.
  • the present invention also envisions the repeated introduction of nucleic acids into cells to allow sustained expression for extended time periods.
  • Cells can be transfected, for example, using commercially available liposome-based transfection kits such as LipofectamineTM (Invitrogen) and can be transfected with any carriers with which RNA can be associated, e.g. by forming complexes with the RNA or forming vesicles in which the RNA is enclosed or encapsulated, resulting in increased stability of the RNA compared to naked RNA.
  • Carriers useful according to the invention include, for example, lipid-containing carriers such as cationic lipids, liposomes, in particular cationic liposomes, and micelles. Cationic lipids may form complexes with negatively charged nucleic acids. Any cationic lipid may be used according to the invention.
  • RNA which encodes a peptide or protein results in expression of said peptide or protein in the cell.
  • the targeting of the nucleic acids to particular cells is preferred.
  • a carrier which is applied for the administration of the nucleic acid to a cell exhibits a targeting molecule.
  • a molecule such as an antibody which is specific for a surface membrane protein on the target cell or a ligand for a receptor on the target cell may be incorporated into the nucleic acid carrier or may be bound thereto.
  • proteins which bind to a surface membrane protein which is associated with endocytosis may be incorporated into the liposome formulation in order to enable targeting and/or uptake.
  • proteins encompass capsid proteins of fragments thereof which are specific for a particular cell type, antibodies against proteins which are internalized, proteins which target an intracellular location etc.
  • Electroporation or electropermeabilization relates to a significant increase in the electrical conductivity and permeability of the cell plasma membrane caused by an externally applied electrical field. It is usually used in molecular biology as a way of introducing some substance into a cell. Electroporation is usually done with electroporators, appliances which create an electro-magnetic field in the cell solution.
  • the cell suspension is pipetted into a glass or plastic cuvette which has two aluminum electrodes on its sides.
  • electroporation tyically a cell suspension of around 50 microliters is used. Prior to electroporation it is mixed with the nucleic acid to be transfected. The mixture is pipetted into the cuvette, the voltage and capacitance is set and the cuvette inserted into the electroporator.
  • liquid medium is added immediately after electroporation (in the cuvette or in an eppendorf tube), and the tube is incubated at the cells' optimal temperature for an hour or more to allow recovery of the cells and optionally expression of antibiotic resistance.
  • a nucleic acid such as RNA encoding a peptide or protein once taken up by or introduced into a cell which cell may be present in vitro or in a subject results in expression of said peptide or protein.
  • the cell may express the encoded peptide or protein intracellularly (e.g. in the cytoplasm and/or in the nucleus), may secrete the encoded peptide or protein, or may express it on the surface. If a peptide or protein (e.g. B18R) is to prevent engagement of IFN receptor by extracellular IFN, secretion of the peptide or protein is preferred. If a peptide or protein (e.g.
  • RNA capable of expressing certain factors for reprogramming of somatic cells is introduced into somatic cells, it is preferred that this introduction of RNA results in expression of said factors for a time period to complete the reprogramming process and in the development of cells having stem cell characteristics.
  • introduction of RNA capable of expression certain factors as disclosed herein into somatic cells results in expression of said factors for an extended period of time, preferably for at least 10 days, preferably for at least 11 days and more preferably for at least 12 days.
  • RNA is preferably periodically (i.e. repetitively) introduced into the cells more than one time, preferably using electroporation.
  • RNA is introduced into the cells at least twice, more preferably at least 3 times, more preferably at least 4 times, even more preferably at least 5 times up to preferably 6 times, more preferably up to 7 times or even up to 8, 9 or 10 times, preferably over a time period of at least 10 days, preferably for at least 1 1 days and more preferably for at least 12 days to ensure expression of one or more factors for an extended period of time.
  • the time periods elapsing between the repeated introductions of the RNA are from 24 hours to 120 hours, preferably 48 hours to 96 hours. In one embodiment, time periods elapsing between the repeated introductions of the RNA are not longer than 72 hours, preferably not longer than 48 hours or 36 hours.
  • the time periods elapsing between the repeated introductions of the RNA are at least 72 hours, preferably at least 96 hours, more preferably at least 120 hours.
  • the conditions should be selected so that the factors are expressed in the cells in amounts and for periods of time which support the reprogramming process.
  • previously not electroporated cells are added as carrier cells.
  • previously not electroporated cells are added prior to, during or after one or more of the 4 th and subsequent, preferably, the 5 th
  • previously not electroporated cells are added prior to, during or after the 4 th or 5 th and each subsequent electroporation.
  • the previously not electroporated cells are the same cells as those into which RNA is introduced.
  • “same conditions” refer to a situation wherein the same cells are used, the cells are cultured under the same conditions (except the condition which results in the enhanced or increased cell viability) and the cell viability is measured by the same means. “Same conditions” also encompasses the introduction or repetitive introduction of RNA into cells.
  • Double-stranded RNA (dsRNA) not only constitutes the genetic material of dsRNA viruses but is also produced in infected cells by positive-strand RNA viruses and some DNA viruses.
  • PTR RNA-dependent protein kinase
  • OFAS oligoadenylate synthetase
  • TLR3 Toll-like receptor 3
  • RIG-I and MDA5 serve as sensors for dsRNA; cf. Fig. 1.
  • IFNs type I interferons
  • IRFs interferon regulatory factors
  • Interferons are important cytokines characterized by antiviral, antiproliferative and immunomodulatory activities. Interferons are proteins that alter and regulate the transcription of genes within a cell by binding to interferon receptors on the regulated cell's surface, thereby preventing viral replication within the cells. According to the invention, the phrase "engagement of IFN receptor by extracellular IFN" relates to the binding of IFNs, in particular type I IFNs, to interferon receptors on the cell surface.
  • the interferons can be grouped into two types. IFN-gamma is the sole type II interferon; all others are type I interferons.
  • Type I and type II interferons differ in gene structure (type II interferon genes have three exons; type I, one), chromosome location (in humans, type II is located on chromosome- 12; the type I interferon genes are linked and on chromosome-9), and the types of tissues where they are produced (type I interferons are synthesized ubiquitously, type II by lymphocytes). Type I interferons competitively inhibit each others binding to cellular receptors, while type II interferon has a distinct receptor.
  • the term "interferon” or "IFN” preferably relates to type I interferons, in particular IFN-alpha and IFN-beta.
  • Human IFN-alpha's are encoded by a multigene family consisting of about 20 genes; each gene encodes a single subtype of the human IFN-alpha.
  • Human IFN-alpha polypeptides are produced by a number of human cell lines and human leukocyte cells after exposure to viruses or double-stranded RNA, or in transformed leukocyte cell lines (e.g., lymphoblastoid lines). IFN-alpha's interact with cell-surface receptors and induce the expression, primarily at the transcriptional level, of a broad but specific set of cellular genes.
  • Human IFN-beta is a regulatory polypeptide with a molecular weight of 22 kDa consisting of 166 amino acid residues. It can be produced by most cells in the body, in particular fibroblasts, in response to viral infection or exposure to other biologies. It binds to a multimeric cell surface receptor, and productive receptor binding results in a cascade of intracellular events leading to the expression of IFN-beta inducible genes which, in turn, produces effects which can be classified as antiviral, antiproliferative, or immunomodulatory. IFNs induce the expression of a plethora of antiviral genes, which can interfere with the viral replication cycle.
  • antivirally active effector protein relates to a group of proteins encoded by IFN-stimulated genes (ISGs) the transcription of which is signaled by type I IFNs. These proteins target distinct viral components and distinct stages of the viral life cycle, aiming to eliminate invading viruses. "Antivirally active effector proteins” are involved in different effector pathways individually blocking viral transcription, degrading viral RNA, inhibiting translation, and modifying protein function to control all steps of viral replication.
  • ISGs IFN-stimulated genes
  • Such proteins include 2',5'-oligoadenylate synthetase (OAS), in particular 2',5'-oligoadenylate synthetase 1 (OAS 1 ), RNA-dependent protein kinase R (P R), and RNaseL. Both PKR and OAS are directly activated by dsRNA. Hence, dsRNA induces the expression of these antivirally active effector proteins and is also necessary for their activation.
  • OFAS 2',5'-oligoadenylate synthetase
  • OAS 1 2',5'-oligoadenylate synthetase 1
  • P R RNA-dependent protein kinase R
  • RNaseL RNA-dependent protein kinase R
  • PKR is constitutively expressed, and induced by type I IFNs. Upon binding to dsRNA, PKR dimerizes and undergoes autophosphorylation to gain full catalytic activity. Once activated, PKR phosphorylates the eukaryotic translation initiation factor eIF2-alpha. In its phosphorylated state, eIF2-alpha forms a stable complex with the nucleotide exchange factor eIF2-beta, which is then no longer recycled for initiation of protein translation by GDP/GTP exchange. Consequently, PKR activation leads to a global block to protein synthesis in the infected cell, which can hamper the production of virus progeny. In this way, PKR in combination with eIF2-alpha constitutes an antiviral pathway (PKR-dependent pathway).
  • RNA-dependent protein kinase protein kinase RNA-activated; PKR
  • PKR protein kinase RNA-activated
  • Human PKR is 68 kDa with an about 20 kDa N-terminal dsRNA-binding domain and a C-terminal protein kinase domain. In vitro PKR is activated by binding to RNA molecules with extensive duplex secondary structure.
  • RNA-stimulated autophosphorylation increases cellular sensitivity to apoptotic and pro-inflammatory stimuli through a number of putative pathways, including phosphorylation of its known substrate eukaryotic initiation factor 2 (eIF2-alpha).
  • eIF2-alpha eukaryotic initiation factor 2
  • PPR preferably relates to human PKR, and, in particular, to a protein comprising the amino acid sequence according to SEQ ID NO: 14 of the sequence listing or a variant of said amino acid sequence.
  • the term “PKR” relates to a protein comprising an amino acid sequence encoded by the nucleic acid sequence according to SEQ ID NO: 13.
  • the term “PKR” includes any variants, in particular mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present.
  • An allelic variant relates to an alteration in the normal sequence of a gene, the significance of which is often unclear. Complete gene sequencing often identifies numerous allelic variants for a given gene.
  • a species homolog is a nucleic acid or amino acid sequence with a different species of origin from that of a given nucleic acid or amino acid sequence.
  • PKR cDNA sequence of PKR as described above would be equivalent to PKR mRNA, and can be used for the generation of inhibitory nucleic acids against PKR.
  • Protein kinase activity including protein kinase autophosphorylation can be measured by a variety of techniques known to the skilled person.
  • One method involves separation of unreacted ATP from the phosphorylated kinase substrate by e.g. precipitating phosphoprotein onto cellulose strips by trichloroacetic acid followed by washing, or adsorption of phosphoprotein onto phosphocellulose strips.
  • dephosphoPKR can be activated by incubation with poly[I:C] and autophosphorylation can be allowed to proceed in the presence of [ ⁇ -32 ⁇ ] ⁇ . The ability of compounds to block this RNA-induced PKR autophosphorylation can be tested.
  • Another method involves detection and quantification of phospho-PKR in relation to the total amount of PKR in the same lysate of cells by Western blotting with antibodies specific for phospho-PKR or full length PKR.
  • Another method involves detection and quantification of the phosphorylated substrate of PKR, e.g. phospho- eIF2-alpha in relation to the total amount of eIF2-alpha in the same lysate of cells by Western blotting with antibodies specific for phospho-eIF2-alpha or full length eIF2-alpha.
  • eIF2-alpha preferably relates to human eIF2-alpha, and, in particular, to a protein comprising the amino acid sequence according to SEQ ID NO: 16 of the sequence listing or a variant of said amino acid sequence.
  • the term “eIF2-alpha” relates to a protein comprising an amino acid sequence encoded by the nucleic acid sequence according to SEQ ID NO: 15.
  • the term “eIF2-alpha” includes any variants, in particular mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present.
  • OAS is expressed at low constitutive levels and is induced by type I IFNs.
  • the protein accumulates in the cell cytoplasm as inactive monomers.
  • the enzyme oligomerizes to (in the instance of OAS 1) form a tetramer that is able to condense ATP molecules via unusual 2',5'-phosphodiester linkages and synthesizes 2 ',5'- oligoadenlylates that, in turn, activate the constitutively expressed inactive RNaseL.
  • Binding of 2',5'-oligoadenlylates to RNaseL triggers dimerization of enzyme monomers, via their kinase-like domains, which then cleaves cellular (and viral) RNAs.
  • OAS in combination with RNaseL constitutes an antiviral RNA decay pathway (OAS-RNaseL antiviral pathway or OAS-dependent pathway).
  • OASl The four OAS genes identified in humans, termed OASl, OAS2, OAS3 and OASL (OAS- like), have been mapped to chromosome 12 (chromosome 5 in mice).
  • OASl has two spliced forms in humans (eight in mice) that produce two, 40 and 46 kDa, proteins that differ at their C-termini by 18 and 54 amino acids, respectively.
  • OAS2 produces four alternatively spliced transcripts that encode two proteins of 69 and 71 kDa.
  • OAS3 encodes a single transcript that produces a 100 kDa protein. These proteins have considerable homology to each other, with OASl , OAS2 and OAS3 encoding one, two and three, respectively, "OAS" domains.
  • OAS proteins The most distinctive of the OAS proteins is OASL.
  • Two OASL transcripts are expressed producing two proteins of 30 and 59 kDa.
  • the higher molecular weight OASL contains a putative nucleolar localization signal at its C-terminus that, probably, accounts for its unique (from the other OAS isoforms) distribution in the cell.
  • the OASL protein also has an OAS domain, however, mutations at key residues disable the catalytic function of this human protein.
  • one of the two mouse homologues retains its 2',5 '-polymerase activity.
  • OASL has a unique 160 amino acid C-terminus that encodes a ubiquitin-like domain that is homologous to ISG15.
  • OASL becomes conjugated (ISGylation) to cellular proteins following the treatment of cells with type I IFNs.
  • ISGylation conjugated
  • cellular proteins There appears to be differential expression and induction of each form of the human OAS proteins.
  • each of the three functional OAS proteins has unique biological functions.
  • a tripeptide motif (CFK) within the OAS domains of OASl and OAS2 mediate oligomerization, so the catalytically active form of these enzymes is a tetramer and dimer, for OASl and OAS2, respectively.
  • This tripeptide motif is not conserved in the OAS domains of OAS3 and OASL and therefore these proteins function as monomers.
  • OAS monomers influences their processivity, with OAS3 synthesizing dimeric molecules of 2',5'-linked oligomers, whereas OASl and OAS2 are capable of synthesizing trimeric and tetrameric oligomers.
  • the dimeric 2',5'-linked oligomers are not efficient activators of RNaseL and, consequently, are thought to regulate alternative processes, with one report suggesting a role in gene expression by regulating DNA topoisomerase I.
  • the term "2',5'-oligoadenylate synthetase" or "OAS" preferably relates to molecules which are activators of RNaseL and preferably to OASl and OAS2.
  • OAS preferably relates to human OAS, and, in particular, to a protein comprising the amino acid sequence according to SEQ ID NO: 18 or 20 of the sequence listing or a variant of said amino acid sequence.
  • OAS relates to a protein comprising an amino acid sequence encoded by the nucleic acid sequence according to SEQ ID NO: 17 or 19.
  • OAS includes any variants, in particular mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present.
  • the 2 ',5 '-dependent RNaseL is expressed as an 80 kDa protein with two kinase-like domains (PUG and STYKc) and eight ankyrin repeats.
  • the enzyme is constitutively expressed as an inactive monomer. Autoinhibition of the enzyme is relieved upon binding of 2 ',5 '-oligomers (generated by OAS proteins) to the ankyrin repeats, and subsequent homodimerization. The active dimeric enzyme then degrades ssRNA.
  • RNaseL preferably relates to human RNaseL, and, in particular, to a protein comprising the amino acid sequence according to SEQ ID NO: 22 of the sequence listing or a variant of said amino acid sequence.
  • RNaseL relates to a protein comprising an amino acid sequence encoded by the nucleic acid sequence according to SEQ ID NO: 21.
  • RNaseL includes any variants, in particular mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present.
  • the term "preventing engagement of IFN receptor by extracellular IFN” relates to an inhibition, i.e. blocking, or reduction, of the interaction of IFNs, in particular type I IFNs, with their specific receptors thus, inhibiting or reducing IFN function.
  • Engagement of IFN receptor by extracellular IFN may be prevented, for example, by the provision of a binding agent for extracellular IFN.
  • the B18R protein is a vaccinia virus-encoded type I interferon receptor with specificity for mouse, human, rabbit, pig, rat, and cow type I interferons which has potent neutralizing activity.
  • the B18R protein encoded by the B18R gene of the Western Reserve vaccinia virus strain is a vaccinia virus-encoded type I interferon receptor with specificity for mouse, human, rabbit, pig, rat, and cow type I interferons which has potent neutralizing activity.
  • the 60-65 kD glycoprotein is related to the interleukin- 1 receptors and is a member of the immunoglobulin superfamily, unlike other type I IFN-receptors, which belong to the class II cytokine receptor family.
  • the B18R protein has a high affinity (KD, 174 pM) for human IFN alpha.
  • KD 174 pM
  • the B18R protein is unique in that it exists as a soluble extracellular, as well as a cell surface protein, enabling blockage of both autocrine and paracrine IFN functions.
  • the B 18R protein has been shown to inhibit the antiviral potency of IFN-alphal, IFN-alpha2, IFN-alpha-8/1/8, and IFN-omega on human cells.
  • the soluble B18R protein is highly potent for neutralizing type I interferons, which include IFN-alpha, beta, delta, kappa.
  • B18R preferably relates to a protein comprising the amino acid sequence according to SEQ ID NO: 24 of the sequence listing or a variant of said amino acid sequence.
  • the term “B18R” relates to a protein comprising an amino acid sequence encoded by the nucleic acid sequence according to SEQ ID NO: 23.
  • the term “B18R” includes any variants, in particular mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present.
  • intracellular IFN signalling relates to the intracellular signaling events and effector functions, in particular antiviral functions, activated by IFNs interacting with their specific receptors and includes the functions of proteins that are induced by IFN, in particular antivirally active effector proteins.
  • intracellular IFN signalling includes the signal propagation through and effector functions, in particular antiviral functions, exerted by proteins which are part of the P R-dependent pathway, in particular P R and eIF2-alpha, and/or the OAS-dependent pathway, in particular OAS and RNaseL.
  • the term "inhibiting intracellular IFN signalling” relates to an inhibition or reduction of intracellular IFN signalling and may be achieved by inhibiting expression, activity or activation of proteins which are involved in intracellular IFN signaling, in particular proteins which are part of the PKR-dependent pathway and/or the OAS-dependent pathway.
  • proteins which are involved in intracellular IFN signaling in particular proteins which are part of the PKR-dependent pathway and/or the OAS-dependent pathway.
  • many viruses have evolved mechanisms for counteracting the PKR and OAS/RNase L pathways. These mechanisms may be used according to the invention for inhibiting intracellular IFN signaling.
  • the P R-dependent pathway may be inhibited by an agent inhibiting or reducing the activity or activation of PKR or by an agent dephosphorylating eIF2-alpha or preventing its phosphorylation, thereby terminating the PKR-induced signal.
  • intracellular IFN signalling may be inhibited according to the invention by utilizing any of the viral defense mechanisms against the PKR signaling cascade.
  • the invention may involve the use of decoy dsRNA (e.g. adenovirus VAI RNA; Epstein-Barr virus EBER; HIV TAR), compounds effecting PKR degradation (e.g. poliovirus 2A pro ), compounds inhibiting activation of PKR, e.g. through hiding viral dsRNA (e.g. vaccinia virus E3/E3L; reovirus sigma3; influenza virus NS 1, herpes simplex virus type 1 (HSV-1) US 1 1), compounds blocking dimerization (e.g.
  • decoy dsRNA e.g. adenovirus VAI RNA; Epstein-Barr virus EBER; HIV TAR
  • compounds effecting PKR degradation e.g. poliovirus 2A pro
  • compounds inhibiting activation of PKR e.g. through hiding viral
  • Vaccinia virus E3 is a 25 kDa dsRNA- binding protein (encoded by gene E3L) that binds and sequesters dsRNA to prevent the activation of PKR and OAS. E3 can bind directly to PKR and inhibits its activity, resulting in reduced phosphorylation of eIF2-alpha.
  • Vaccinia virus gene K3L encodes a 10.5 kDa homolog of the eIF2-alpha subunit that acts as a non-phosphorylable pseudosubstrate of PKR and competitively inhibits phosphorylation of eIF2-alpha.
  • Vaccinia virus C7/C7L inhibits phosphorylation of eIF2-alpha.
  • the ICP34.5 protein from HSV-1 functions as a regulatory subunit of the cellular PPl phosphatase, directing it to dephosphorylate eIF2-alpha, thereby terminating the PKR-induced signal.
  • the murine cytomegalovirus (MCMV) proteins ml 42 and ml 43 have been characterized as dsRNA binding proteins that inhibit PKR activation, phosphorylation of the translation initiation factor eIF2, and a subsequent protein synthesis shutoff.
  • a decoy RNA is pseudosubstrate RNA that has similar structure to the RNA substrate of an enzyme, in order to make the enzyme bind to the pseudosubstrate rather than to the real substrate, thus blocking the activity of the enzyme.
  • E3 preferably relates to a protein comprising the amino acid sequence according to SEQ ID NO: 26 of the sequence listing or a variant of said amino acid sequence.
  • E3 relates to a protein comprising an amino acid sequence encoded by the nucleic acid sequence according to SEQ ID NO: 25.
  • E3 includes any variants, in particular mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present.
  • K3 preferably relates to a protein comprising the amino acid sequence according to SEQ ID NO: 28 of the sequence listing or a variant of said amino acid sequence.
  • the term “ 3” relates to a protein comprising an amino acid sequence encoded by the nucleic acid sequence according to SEQ ID NO: 27.
  • the term “ 3” includes any variants, in particular mutants, splice variants, conformations, isoforms, allelic variants, species variants and species homologs, in particular those which are naturally present.
  • RNA-dependent protein kinase relates to measures that result in a lower degree of homodimerization of PKR, in a lower degree of autophosphorylation of PKR and/or in a lower degree of phosphorylation of targets which are kinase substrates of PKR such as eIF2-alpha compared to the normal situation, in particular the normal situation in a cell, wherein the activity of PKR is not reduced/has not been reduced by man.
  • said term includes all measures that result in a lower degree of autophosphorylation of PKR and/or in a lower degree of phosphorylation of targets which are kinase substrates of PKR.
  • reducing the activity of RNA-dependent protein kinase (PKR) in a cell comprises treating the cell with an inhibitor of expression and/or activity of PKR.
  • inhibitor expression and/or activity includes a complete or essentially complete inhibition of expression and/or activity and a reduction in expression and/or activity.
  • said PKR inhibitor is directed at the PKR protein and preferably is specific for PKR.
  • PKR can be inhibited in various ways, e.g. through inhibiting PKR autophosphorylation and/or dimerization, providing a PKR pseudo-activator, or providing a PKR pseudo-substrate.
  • the PKR inhibitor may be an agent which is involved in a viral defense mechanism as discussed above.
  • vaccinia virus E3L encodes a dsRNA binding protein that inhibits PKR in virus-infected cells, presumably by sequestering dsRNA activators.
  • K3 also encoded by vaccinia virus, functions as a pseudosubstrate inhibitor by binding to PKR.
  • providing vaccinia virus E3L may result in inhibition of PKR.
  • Providing adenovirus VAI RNA, HIV Tat or Epstein-Barr virus EBER1 RNA may result in PKR pseudo-activation.
  • all viral factors, i.e. virally derived inhibitors, blocking PKR activity such as those described herein may be used for reducing the activity of PKR.
  • the PKR inhibitor is a chemical inhibitor.
  • the PKR inhibitor is an inhibitor of RNA-induced PKR autophosphorylation.
  • the PKR inhibitor is an ATP-binding site directed inhibitor of PKR.
  • the PKR inhibitor is 6,8-dihydro-8-(lH-imidazol-5-ylmethylene)-7H- pyrrolo[2,3-g]benzothiazol-7-one. In one embodiment, the PKR inhibitor has the following formula:
  • the PKR inhibitor is 2-aminopurine. In one embodiment, the PKR inhibitor has the following formula:
  • an inhibitor as disclosed above is used in a concentration of at least 0.5 ⁇ or higher, at least 1 ⁇ or higher or at least 2 ⁇ or higher and preferably in a concentration up to 5 ⁇ , up to 4 ⁇ , up to 3 ⁇ or up to 2 ⁇ .
  • the inhibitor of activity of PKR is an antibody that specifically binds to PKR. Binding of the antibody to PKR can interfere with the function of PKR, e.g. by inhibiting binding activity or catalytic activity.
  • PKR activity of PKR in a cell by treating the cell with one or more virally derived inhibitors such as vaccinia virus E3 and/or K3 as well as treating the cell with one or more chemical PKR inhibitors such as 6,8-dihydro-8-(lH- imidazol-5-ylmethylene)-7H-pyrrolo[2,3-g]benzothiazol-7-one and/or 2-aminopurine.
  • virally derived inhibitors such as vaccinia virus E3 and/or K3
  • chemical PKR inhibitors such as 6,8-dihydro-8-(lH- imidazol-5-ylmethylene)-7H-pyrrolo[2,3-g]benzothiazol-7-one and/or 2-aminopurine.
  • the OAS-dependent pathway may be inhibited by an agent inhibiting or reducing the activity or activation of OAS and/or RNaseL.
  • vaccinia virus E3 is a 25 kDa dsRNA-binding protein (encoded by gene E3L) that binds and sequesters dsRNA to prevent the activation of OAS.
  • the terms "reducing the activity of OAS” preferably relates to measures that result in a lower degree of production of 2',5'-oligoadenlylates and thus, activation of RNaseL.
  • reducing the activity of OAS and/or RNaseL in a cell comprises treating the cell with an inhibitor of expression and/or activity of OAS and/or RNaseL.
  • the phrase "inhibit expression and/or activity” includes a complete or essentially complete inhibition of expression and/or activity and a reduction in expression and/or activity.
  • inhibition of expression of P R, OAS or RNaseL in the following referred to as "target protein” may take place by inhibiting the production of or reducing the level of transcript, i.e. mRNA, coding for the target protein, e.g. by inhibiting transcription or inducing degradation of transcript, and/or by inhibiting the production of the target protein, e.g. by inhibiting translation of transcript coding for the target protein.
  • said inhibitor is specific for a nucleic acid encoding the target protein.
  • the inhibitor of expression of the target protein is an inhibitory nucleic acid (e.g., antisense molecule, ribozyme, iRNA, siRNA or a DNA encoding the same) selectively hybridizing to and being specific for nucleic acid encoding the target protein, thereby inhibiting (e.g., reducing) transcription and/or translation thereof.
  • an inhibitory nucleic acid e.g., antisense molecule, ribozyme, iRNA, siRNA or a DNA encoding the same
  • Inhibitory nucleic acids of this invention include oligonucleotides having sequences in the antisense orientation relative to the target nucleic acids. Suitable inhibitory oligonucleotides typically vary in length from five to several hundred nucleotides, more typically about 20-70 nucleotides in length or shorter, even more typically about 10-30 nucleotides in length. These inhibitory oligonucleotides may be applied, either in vitro or in vivo, as free (naked) nucleic acids or in protected forms, e.g., encapsulated in liposomes. The use of liposomal or other protected forms may be advantageous as it may enhance in vivo stability and thus facilitate delivery to target sites.
  • the target nucleic acid may be used to design ribozymes that target the cleavage of the corresponding mRNAs in cells.
  • these ribozymes may be administered in free (naked) form or by the use of delivery systems that enhance stability and/or targeting, e.g., liposomes.
  • the target nucleic acid may be used to design siRNAs that can inhibit (e.g., reduce) expression of the nucleic acid.
  • the siRNAs may be administered in free (naked) form or by the use of delivery systems that enhance stability and/or targeting, e.g., liposomes. They may also be administered in the form of their precursors or encoding DNAs.
  • siRNA preferably comprises a sense RNA strand and an antisense RNA strand, wherein the sense and antisense RNA strands form an RNA duplex, and wherein the sense RNA strand comprises a nucleotide sequence substantially identical to a target sequence of about 19 to about 25 contiguous nucleotides in a target nucleic acid, preferably mRNA coding for P R.
  • nucleic acids encoding the peptides or proteins can be provided instead of the peptides or proteins mentioned above for (i) preventing engagement of IFN receptor by extracellular IFN and (ii) inhibiting intracellular IFN signalling.
  • nucleic acids encoding the peptides or proteins can be provided.
  • the phrase "provided in the form of a nucleic acid" as used herein is to account for this possibility.
  • cells may be transfected with nucleic acid, in particular RNA, encoding the peptides or proteins and the nucleic acid may be expressed in the cells so as to produce the peptides or proteins.
  • cells are treated to (i) prevent engagement of IFN receptor by extracellular IFN and/or (ii) inhibit intracellular IFN signalling prior to, simultaneously with and/or following introduction of RNA encoding the peptide or protein to be expressed, e.g. one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics, or the first introduction (e.g. in case of repeated transfections) of RNA.
  • cells are treated to (i) prevent engagement of IFN receptor by extracellular IFN and/or (ii) inhibit intracellular IFN signalling following, preferably immediately following introduction of RNA or the first introduction (e.g. in case of repeated transfections) of RNA.
  • cells are treated to (i) prevent engagement of IFN receptor by extracellular IFN and/or (ii) inhibit intracellular IFN signalling for at least 24 h, at least 48 h, at least 72 h, at least 96 h, at least 120 h or even longer. Most preferably, cells are treated to (i) prevent engagement of IFN receptor by extracellular IFN and/or (ii) inhibit intracellular IFN signalling for the entire period of time in which expression of RNA is desired, such as permanently, optionally with repeated transfection of RNA.
  • the cell can be an isolated cell or it can form part of an organ, a tissue and/or an organism.
  • Antisense molecules or “antisense nucleic acids” may be used for regulating, in particular reducing, expression of a nucleic acid.
  • the term “antisense molecule” or “antisense nucleic acid” refers according to the invention to an oligonucleotide which is an oligoribonucleotide, oligodeoxyribonucleotide, modified oligoribonucleotide or modified oligo- deoxyribonucleotide and which hybridizes under physiological conditions to DNA comprising a particular gene or to mRNA of said gene, thereby inhibiting transcription of said gene and/or translation of said mRNA.
  • an "antisense molecule” also comprises a construct which contains a nucleic acid or a part thereof in reverse orientation with respect to its natural promoter.
  • An antisense transcript of a nucleic acid or of a part thereof may form a duplex with naturally occurring mRNA and thus prevent accumulation of or translation of the mRNA.
  • Another possibility is the use of ribozymes for inactivating a nucleic acid.
  • the antisense oligonucleotide hybridizes with an N-terminal or 5' upstream site such as a translation initiation site, transcription initiation site or promoter site. In further embodiments, the antisense oligonucleotide hybridizes with a 3'-untranslated region or mRNA splicing site.
  • small interfering RNA or "siRNA” as used herein is meant an RNA molecule, preferably greater than 10 nucleotides in length, more preferably greater than 15 nucleotides in length, and most preferably 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length that is used to identify a target gene or mRNA to be degraded. A range of 19-25 nucleotides is the most preferred size for siRNAs.
  • the siRNA can also comprise a 3'-overhang.
  • a "3 - overhang” refers to at least one unpaired nucleotide extending from the 3'-end of an RNA strand.
  • the siRNA comprises at least one 3'-overhang of from 1 to about 6 nucleotides (which includes ribonucleotides or deoxynucleotides) in length, preferably from 1 to about 5 nucleotides in length, more preferably from 1 to about 4 nucleotides in length, and particularly preferably from about 2 to about 4 nucleotides in length.
  • each strand of the siRNA of the invention can comprise 3'-overhangs of dideoxythymidylic acid ("TT") or diuridylic acid ("uu").
  • TT dideoxythymidylic acid
  • uu diuridylic acid
  • the 3'-overhangs can be also stabilized against degradation.
  • the overhangs are stabilized by including purine nucleotides, such as adenosine or guanosine nucleotides.
  • substitution of pyrimidine nucleotides by modified analogues e.g., substitution of uridine nucleotides in the 3'- overhangs with 2'-deoxythymidine, is tolerated and does not affect the efficiency of RNAi degradation.
  • the absence of a 2'-hydroxyl in the 2'-deoxythymidine significantly enhances the nuclease resistance of the 3'-overhang in tissue culture medium.
  • the sense and antisense strands of the siRNA can comprise two complementary, single- stranded RNA molecules or can comprise a single molecule in which two complementary portions are base-paired and are covalently linked by a single-stranded "hairpin" area. That is, the sense region and antisense region can be covalently connected via a linker molecule.
  • the linker molecule can be a polynucleotide or non-nucleotide linker. Without wishing to be bound by any theory, it is believed that the hairpin area of the latter type of siRNA molecule is cleaved intracellularly by the "Dicer" protein (or its equivalent) to form a siRNA of two individual base-paired RNA molecules.
  • target mRNA refers to an RNA molecule that is a target for downregulation.
  • siRNA can be expressed from pol III expression vectors without a change in targeting site, as expression of RNAs from pol III promoters is only believed to be efficient when the first transcribed nucleotide is a purine.
  • siRNA according to the invention can be targeted to any stretch of approximately 19-25 contiguous nucleotides in any of the target mRNA sequences (the "target sequence”). Techniques for selecting target sequences for siRNA are given, for example, in Tuschl T. et al., "The siRNA User Guide", revised Oct. 1 1, 2002, the entire disclosure of which is herein incorporated by reference.
  • the siRNA User Guide is available on the world wide web at a website maintained by Dr. Thomas Tuschl, Laboratory of RNA Molecular Biology, Rockefeller University, New York, USA, and can be found by accessing the website of the Rockefeller University and searching with the keyword "siRNA".
  • the sense strand of the present siRNA comprises a nucleotide sequence substantially identical to any contiguous stretch of about 19 to about 25 nucleotides in the target mRNA.
  • a target sequence on the target mRNA can be selected from a given cDNA sequence corresponding to the target mRNA, preferably beginning 50 to 100 nt downstream (i.e., in the 3'-direction) from the start codon.
  • the target sequence can, however, be located in the 5'- or 3 '-untranslated regions, or in the region nearby the start codon.
  • siRNA can be obtained using a number of techniques known to those of skill in the art. For example, siRNA can be chemically synthesized or recombinantly produced using methods known in the art, such as the Drosophila in vitro system described in U.S. published application 2002/0086356 of Tuschl et al., the entire disclosure of which is herein incorporated by reference.
  • siRNA is chemically synthesized using appropriately protected ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer.
  • siRNA can be synthesized as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions.
  • siRNA can also be expressed from recombinant circular or linear DNA plasmids using any suitable promoter.
  • suitable promoters for expressing siRNA of the invention from a plasmid include, for example, the U6 or HI RNA pol III promoter sequences and the cytomegalovirus promoter.
  • the recombinant plasmids of the invention can also comprise inducible or regulatable promoters for expression of the siRNA in a particular tissue or in a particular intracellular environment.
  • siRNA expressed from recombinant plasmids can either be isolated from cultured cell expression systems by standard techniques, or can be expressed intracellularly.
  • the use of recombinant plasmids to deliver siRNA to cells in vivo is within the skill in the art.
  • siRNA can be expressed from a recombinant plasmid either as two separate, complementary RNA molecules, or as a single RNA molecule with two complementary regions.
  • plasmids suitable for expressing siRNA Selection of plasmids suitable for expressing siRNA, methods for inserting nucleic acid sequences for expressing the siRNA into the plasmid, and methods of delivering the recombinant plasmid to the cells of interest are within the skill in the art.
  • cell or "host cell” preferably relates to an intact cell, i.e. a cell with an intact membrane that has not released its normal intracellular components such as enzymes, organelles, or genetic material.
  • An intact cell preferably is a viable cell, i.e. a living cell capable of carrying out its normal metabolic functions.
  • said term relates according to the invention to any cell which can be transformed or transfected with an exogenous nucleic acid.
  • the term "cell” includes according to the invention prokaryotic cells (e.g., E. coli) or eukaryotic cells. Mammalian cells are particularly preferred, such as cells from humans, mice, hamsters, pigs, goats, and primates.
  • the cell is a somatic cell as described herein. In one embodiment, the cell is a cell having a barrier function.
  • the cell is a fibroblast such as a fibroblast described herein, a keratinocyte, an epithelial cell, or an endothelial cell such as an endothelial cell of the heart, an endothelial cell of the lung, or an umbilical vein endothelial cell.
  • the cell is a human cell.
  • a fibroblast is a type of cell that synthesizes the extracellular matrix and collagen and plays a critical role in wound healing.
  • the main function of fibroblasts is to maintain the structural integrity of connective tissues by continuously secreting precursors of the extracellular matrix.
  • Fibroblasts are the most common cells of connective tissue in animals. Fibroblasts are morphologically heterogeneous with diverse appearances depending on their location and activity.
  • Keratinocytes are the predominant cell type in the epidermis, the outermost layer of the human skin. The primary function of keratinocytes is the formation of the keratin layer that protects the skin and the underlying tissue from environmental damage such as heat, UV radiation and water loss.
  • Epithelium is a tissue composed of cells that line the cavities and surfaces of structures throughout the body. Many glands are also formed from epithelial tissue. It lies on top of connective tissue, and the two layers are separated by a basement membrane. In humans, epithelium is classified as a primary body tissue, the other ones being connective tissue, muscle tissue and nervous tissue. Functions of epithelial cells include secretion, selective absorption, protection, transcellular transport and detection of sensation.
  • the endothelium is the thin layer of cells that lines the interior surface of blood vessels, forming an interface between circulating blood in the lumen and the rest of the vessel wall. These cells are called endothelial cells. Endothelial cells line the entire circulatory system, from the heart to the smallest capillary. Endothelial tissue is a specialized type of epithelium tissue.
  • the term "peptide” comprises oligo- and polypeptides and refers to substances comprising two or more, preferably 3 or more, preferably 4 or more, preferably 6 or more, preferably 8 or more, preferably 10 or more, preferably 13 or more, preferably 16 more, preferably 21 or more and up to preferably 8, 10, 20, 30, 40 or 50, in particular 100 amino acids joined covalently by peptide bonds.
  • the term “protein” refers to large peptides, preferably to peptides with more than 100 amino acid residues, but in general the terms “peptides” and “proteins” are synonyms and are used interchangeably herein.
  • the present invention also includes "variants" of the peptides, proteins, or amino acid sequences described herein.
  • variants of an amino acid sequence comprise amino acid insertion variants, amino acid addition variants, amino acid deletion variants and/or amino acid substitution variants.
  • Amino acid insertion variants comprise insertions of single or two or more amino acids in a particular amino acid sequence.
  • amino acid sequence variants having an insertion one or more amino acid residues are inserted into a particular site in an amino acid sequence, although random insertion with appropriate screening of the resulting product is also possible.
  • Amino acid addition variants comprise amino- and/or carboxy-terminal fusions of one or more amino acids, such as 1, 2, 3, 5, 10, 20, 30, 50, or more amino acids.
  • Amino acid deletion variants are characterized by the removal of one or more amino acids from the sequence, such as by removal of 1, 2, 3, 5, 10, 20, 30, 50, or more amino acids.
  • the deletions may be in any position of the protein.
  • Amino acid deletion variants that comprise the deletion at the N-terminal and/or C-terminal end of the protein are also called N-terminal and/or C-terminal truncation variants.
  • Amino acid substitution variants are characterized by at least one residue in the sequence being removed and another residue being inserted in its place. Preference is given to the modifications being in positions in the amino acid sequence which are not conserved between homologous proteins or peptides and/or to replacing amino acids with other ones having similar properties.
  • amino acid changes in protein variants are conservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids.
  • a conservative amino acid change involves substitution of one of a family of amino acids which are related in their side chains.
  • Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate), basic (lysine, arginine, histidine), non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and uncharged polar (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine) amino acids. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids.
  • the degree of similarity, preferably identity between a given amino acid sequence and an amino acid sequence which is a variant of said given amino acid sequence will be at least about 60%, 65%, 70%, 80%, 81%, 82%, 83%, 84%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • the degree of similarity or identity is given preferably for an amino acid region which is at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or about 100% of the entire length of the reference amino acid sequence.
  • the degree of similarity or identity is given preferably for at least about 20, at least about 40, at least about 60, at least about 80, at least about 100, at least about 120, at least about 140, at least about 160, at least about 180, or about 200 amino acids, preferably continuous amino acids.
  • the degree of similarity or identity is given for the entire length of the reference amino acid sequence.
  • the alignment for determining sequence similarity, preferably sequence identity can be done with art known tools, preferably using the best sequence alignment, for example, using Align, using standard settings, preferably EMBOSS: :needle, Matrix: Blosum62, Gap Open 10.0, Gap Extend 0.5.
  • Sequence similarity indicates the percentage of amino acids that either are identical or that represent conservative amino acid substitutions.
  • Sequence identity indicates the percentage of amino acids or nucleotides that are identical between the sequences.
  • the term “percentage identity” is intended to denote a percentage of amino acid residues which are identical between the two sequences to be compared, obtained after the best alignment, this percentage being purely statistical and the differences between the two sequences being distributed randomly and over their entire length. Sequence comparisons between two amino acid sequences are conventionally carried out by comparing these sequences after having aligned them optimally, said comparison being carried out by segment or by "window of comparison" in order to identify and compare local regions of sequence similarity.
  • the optimal alignment of the sequences for comparison may be produced, besides manually, by means of the local homology algorithm of Smith and Waterman, 1981 , Ads App. Math. 2, 482, by means of the local homology algorithm of Neddleman and Wunsch, 1970, J. Mol. Biol. 48, 443, by means of the similarity search method of Pearson and Lipman, 1988, Proc. Natl Acad. Sci. USA 85, 2444, or by means of computer programs which use these algorithms (GAP, BESTFIT, FASTA, BLAST P, BLAST N and TFASTA in Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Drive, Madison, Wis.).
  • the percentage identity is calculated by determining the number of identical positions between the two sequences being compared, dividing this number by the number of positions compared and multiplying the result obtained by 100 so as to obtain the percentage identity between these two sequences.
  • Homologous amino acid sequences exhibit according to the invention at least 40%, in particular at least 50%, at least 60%, at least 70%, at least 80%, at least 90% and preferably at least 95%, at least 98 or at least 99% identity of the amino acid residues.
  • the amino acid sequence variants described herein may readily be prepared by the skilled person, for example, by recombinant DNA manipulation. The manipulation of DNA sequences for preparing peptides or proteins having substitutions, additions, insertions or deletions, is described in detail in Sambrook et al. (1989), for example. Furthermore, the peptides and amino acid variants described herein may be readily prepared with the aid of known peptide synthesis techniques such as, for example, by solid phase synthesis and similar methods.
  • the invention includes derivatives of the peptides or proteins described herein which are comprised by the terms “peptide” and "protein".
  • “derivatives” of proteins and peptides are modified forms of proteins and peptides. Such modifications include any chemical modification and comprise single or multiple substitutions, deletions and/or additions of any molecules associated with the protein or peptide, such as carbohydrates, lipids and/or proteins or peptides.
  • derivatives of proteins or peptides include those modified analogs resulting from glycosylation, acetylation, phosphorylation, amidation, palmitoylation, myristoylation, isoprenylation, lipidation, alkylation, derivatization, introduction of protective/blocking groups, proteolytic cleavage or binding to an antibody or to another cellular ligand.
  • the term “derivative” also extends to all functional chemical equivalents of said proteins and peptides.
  • a modified peptide has increased stability and/or increased immunogenicity.
  • a variant of a peptide or protein preferably has a functional property of the peptide or protein from which it has been derived. Such functional properties are described herein for OCT4, SOX2, NANOG, LIN28, LF4 and c-MYC, respectively.
  • a variant of a peptide or protein has the same property in reprogramming an animal differentiated cell as the peptide or protein from which it has been derived.
  • the variant induces or enhances reprogramming of an animal differentiated cell.
  • the peptide or protein encoded by the RNA is a factor allowing the reprogramming of somatic cells to cells having stem cell characteristics.
  • the peptide or protein comprises one or more antigens and/or one or more antigen peptides.
  • said RNA is capable of expressing said peptide or protein, in particular if introduced into a cell.
  • a "stem cell” is a cell with the ability to self-renew, to remain undifferentiated, and to become differentiated.
  • a stem cell can divide without limit, for at least the lifetime of the animal in which it naturally resides.
  • a stem cell is not terminally differentiated; it is not at the end stage of a differentiation pathway. When a stem cell divides, each daughter cell can either remain a stem cell or embark on a course that leads toward terminal differentiation.
  • Totipotent stem cells are cells having totipotential differentiation properties and being capable of developing into a complete organism. This property is possessed by cells up to the 8-cell stage after fertilization of the oocyte by the sperm. When these cells are isolated and transplanted into the uterus, they can develop into a complete organism.
  • Pluripotent stem cells are cells capable of developing into various cells and tissues derived from the ectodermal, mesodermal and endodermal layers. Pluripotent stem cells which are derived from the inner cell mass located inside of blastocysts, generated 4-5 days after fertilization are called “embryonic stem cells” and can differentiate into various other tissue cells but cannot form new living organisms.
  • Multipotent stem cells are stem cells differentiating normally into only cell types specific to their tissue and organ of origin. Multipotent stem cells are involved not only in the growth and development of various tissues and organs during the fetal, neonatal and adult periods but also in the maintenance of adult tissue homeostasis and the function of inducing regeneration upon tissue damage. Tissue-specific multipotent cells are collectively called "adult stem cells”.
  • An "embryonic stem cell” or “ESC” is a stem cell that is present in or isolated from an embryo. It can be pluripotent, having the capacity to differentiate into each and every cell present in the organism, or multipotent, with the ability to differentiate into more than one cell type.
  • embryo refers to an animal in the early stages of it development. These stages are characterized by implantation and gastrulation, where the three germ layers are defined and established and by differentiation of the germs layers into the respective organs and organ systems. The three germ layers are the endoderm, ectoderm and mesoderm.
  • a "blastocyst” is an embryo at an early stage of development in which the fertilized ovum has undergone cleavage, and a spherical layer of cells surrounding a fluid-filled cavity is forming, or has formed. This spherical layer of cells is the trophectoderm. Inside the trophectoderm is a cluster of cells termed the inner cell mass (ICM). The trophectoderm is the precursor of the placenta, and the ICM is the precursor of the embryo.
  • ICM inner cell mass
  • An adult stem cell also called a somatic stem cell, is a stem cell found in an adult.
  • An adult stem cell is found in a differentiated tissue, can renew itself, and can differentiate, with some limitations, to yield specialized cell types of its tissue of origin. Examples include mesenchymal stem cells, hematopoietic stem cells, and neural stem cells.
  • a “differentiated cell” is a mature cell that has undergone progressive developmental changes to a more specialized form or function.
  • Cell differentiation is the process a cell undergoes as it matures to an overtly specialized cell type. Differentiated cells have distinct characteristics, perform specific functions, and are less likely to divide than their less differentiated counterparts.
  • An "undifferentiated" cell typically has a nonspecific appearance, may perform multiple, non-specific activities, and may perform poorly, if at all, in functions typically performed by differentiated cells.
  • Somatic cell refers to any and all differentiated cells and does not include stem cells, germ cells, or gametes.
  • stem cells germ cells, or gametes.
  • sematic cell refers to a terminally differentiated cell.
  • transmitted refers to cells which are considered to be permanently committed to a specific function. Committed cells are also referred to as “terminally differentiated cells”.
  • differentiation refers to the adaptation of cells for a particular form or function. In cells, differentiation leads to a more committed cell.
  • de-differentiation refers to loss of specialization in form or function. In cells, de-differentiation leads to a less committed cell.
  • reprogramming refers to the resetting of the genetic program of a cell.
  • a reprogrammed cell preferably exhibits pluripotency.
  • de-differentiated and reprogrammed or similar terms are used interchangeably herein to denote somatic cell-derived cells having stem cell characteristics. However, said terms are not intended to limit the subject-matter disclosed herein by mechanistic or functional considerations.
  • RNA inducing the development of stem cell characteristics or "RNA capable of expressing one or more factors allowing the reprogramming of the somatic cells to cells having stem cell characteristics” refers to RNA which when introduced into a somatic cell induces the cell to de-differentiate.
  • germ cell refers to a reproductive cell such as a spermatocyte or an oocyte, or a cell that will develop into a reproductive cell.
  • pluripotent refers to cells that can give rise to any cell type except the cells of the placenta or other supporting cells of the uterus.
  • Cell having stem cell characteristics are used herein to designate cells which, although they are derived from differentiated somatic non-stem cells, exhibit one or more features typical for stem cells, in particular embryonic stem cells.
  • Such features include an embryonic stem cell morphology such as compact colonies, high nucleus to cytoplasm ratio and prominent nucleoli, normal karyotypes, expression of telomerase activity, expression of cell surface markers that are characteristic for embryonic stem cells, and/or expression of genes that are characteristic for embryonic stem cells.
  • the cell surface markers that are characteristic for embryonic stem cells are, for example, selected from the group consisting of stage-specific embryonic antigen- 3 (SSEA-3), SSEA-4, tumor-related antigen- 1-60 (TRA-1-60), TRA-1 -81, and TRA-2-49/6E.
  • the genes that are characteristic for embryonic stem cells are selected, for example, from the group consisting of endogenous OCT4, endogenous NANOG, growth and differentiation factor 3 (GDF3), reduced expression 1 (REX1), fibroblast growth factor 4 (FGF4), embryonic cell-specific gene 1 (ESG1), developmental pluripotency-associated 2 (DPPA2), DPPA4, and telomerase reverse transcriptase (TERT).
  • the one or more features typical for stem cells include pluripotency.
  • the stem cell characteristics comprise an embryonic stem cell morphology, wherein said embryonic stem cell morphology preferably comprises morphological ciriteria selected from the group consisting of compact colonies, high nucleus to cytoplasm ratio and prominent nucleoli.
  • the cells having stem cell characteristics have normal karyotypes, express telomerase activity, express cell surface markers that are characteristic for embryonic stem cells and/or express genes that are characteristic for embryonic stem cells.
  • the cell surface markers that are characteristic for embryonic stem cells may be selected from the group consisting of stage-specific embryonic antigen-3 (SSEA-3), SSEA-4, rumor-related antigen- 1-60 (TRA-1 -60), TRA-1-81, and TRA- 2-49/6E and the genes that are characteristic for embryonic stem cells may be selected from the group consisting of endogenous OCT4, endogenous NANOG, growth and differentiation factor 3 (GDF3), reduced expression 1 (REX1), fibroblast growth factor 4 (FGF4), embryonic cell-specific gene 1 (ESG1 ), developmental pluripotency-associated 2 (DPPA2), DPPA4, and telomerase reverse transcriptase (TERT).
  • SSEA-3 stage-specific embryonic antigen-3
  • SSEA-4 rumor-related antigen- 1-60
  • TRA-1-81 TRA-1-81
  • the genes that are characteristic for embryonic stem cells may be selected from the group consisting of endogenous OCT4, endogenous NAN
  • the cells having stem cell characteristics are de-differentiated and/or reprogrammed somatic cells.
  • the cells having stem cell characteristics exhibit the essential characteristics of embryonic stem cells such as a pluripotent state.
  • the cells having stem cell characteristics have the developmental potential to differentiate into advanced derivatives of all three primary germ layers.
  • the primary germ layer is endoderm and the advanced derivative is gut-like epithelial tissue.
  • the primary germ layer is mesoderm and the advanced derivative is striated muscle and/or cartilage.
  • the primary germ layer is ectoderm and the advanced derivative is neural tissue and/or epidermal tissue.
  • the cells having stem cell characteristics have the developmental potential to differentiate into neuronal cells and/or cardiac cells.
  • the somatic cells are embryonic stem cell derived somatic cells with a mesenchymal phenotype.
  • the somatic cells are fibroblasts such as fetal fibroblasts or postnatal fibroblasts or keratinocytes, preferably hair follicle derived keratinocytes.
  • the fibroblasts are lung fibroblasts, foreskin fibroblasts or dermal fibroblasts.
  • the fibroblasts are fibroblasts as deposited at the American Type Culture Collection (ATCC) under Catalog No. CCL-186, as deposited at the American Type Culture Collection (ATCC) under Catalog No.
  • the fibroblasts are adult human dermal fibroblasts.
  • the somatic cells are human cells. According to the present invention, the somatic cells may be genetically modified.
  • factor when used in conjunction with the expression thereof by RNA includes proteins and peptides as well as derivatives and variants thereof.
  • factor comprises OCT4, SOX2, NANOG, LIN28, .LF4 and c-MYC.
  • the factors can be of any animal species; e.g., mammals and rodents.
  • mammals include but are not limited to human and non-human primates.
  • Primates include but are not limited to humans, chimpanzees, baboons, cynomolgus monkeys, and any other New or Old World monkeys.
  • Rodents include but are not limited to mouse, rat, guinea pig, hamster and gerbil.
  • one or more factors capable of allowing the reprogramming of somatic cells to cells having stem cell characteristics comprise an assembly of factors selected from the group consisting of (i) OCT4 and SOX2, (ii) OCT4, SOX2, and one or both of NANOG and LIN28, (iii) OCT4, SOX2 and one or both of LF4 and c-MYC.
  • said one or more factors capable of being expressed by the RNA comprise OCT4, SOX2, NANOG and LIN28 or OCT4, SOX2, LF4 and c-MYC.
  • the RNA is introduced into said animal differentiated somatic cell by electroporation or microinjection.
  • the method of the invention further comprises allowing the development of cells having stem cell characteristics, e.g. by culturing the somatic cell under embryonic stem cell culture conditions, preferably conditions suitable for maintaining pluripotent stem cells in an undifferentiated state.
  • OCT4 is a transcription factor of the eukaryotic POU transcription factors and an indicator of pluripotency of embryonic stem cells. It is a maternally expressed Octomer binding protein. It has been observed to be present in oocytes, the inner cell mass of blastocytes and also in the primordial germ cell.
  • the gene POU5F1 encodes the OCT4 protein. Synonyms to the gene name include OCT3, OCT4, OTF3 and MGC22487. The presence of OCT4 at specific concentrations is necessary for embryonic stem cells to remain undifferentiated.
  • OCT4 protein or simply “OCT4" relates to human OCT4 and preferably comprises an amino acid sequence encoded by the nucleic acid according to SEQ ID NO: 1, preferably the amino acid sequence according to SEQ ID NO: 2.
  • SEQ ID NO: 1 preferably the amino acid sequence according to SEQ ID NO: 2.
  • Sox2 is a member of the Sox (SRY-related HMG box) gene family that encode transcription factors with a single HMG DNA-binding domain. SOX2 has been found to control neural progenitor cells by inhibiting their ability to differentiate. The repression of the factor results in delamination from the ventricular zone, which is followed by an exit from the cell cycle. These cells also begin to lose their progenitor character through the loss of progenitor and early neuronal differentiation markers.
  • SOX2 protein or simply “SOX2” relates to human SOX2 and preferably comprises an amino acid sequence encoded by the nucleic acid according to SEQ ID NO: 3, preferably the amino acid sequence according to SEQ ID NO: 4.
  • SEQ ID NO: 3 preferably the amino acid sequence according to SEQ ID NO: 4.
  • NANOG is a NK-2 type homeodomain gene, and has been proposed to play a key role in maintaining stem cell pluripotency presumably by regulating the expression of genes critical to embryonic stem cell renewal and differentiation.
  • NANOG behaves as a transcription activator with two unusually strong activation domains embedded in its C terminus. Reduction of NANOG expression induces differentiation of embryonic stem cells.
  • "NANOG protein" or simply "NANOG” relates to human NANOG and preferably comprises an amino acid sequence encoded by the nucleic acid according to SEQ ID NO: 5, preferably the amino acid sequence according to SEQ ID NO: 6.
  • SEQ ID NO: 5 preferably the amino acid sequence according to SEQ ID NO: 6
  • LIN28 is a conserved cytoplasmic protein with an unusual pairing of RNA-binding motifs: a cold shock domain and a pair of retroviral-type CCHC zinc fingers. In mammals, it is abundant in diverse types of undifferentiated cells. In pluripotent mammalian cells, LIN28 is observed in RNase-sensitive complexes with Poly(A)-Binding Protein, and in polysomal fractions of sucrose gradients, suggesting it is associated with translating mRNAs.
  • LIN28 protein or simply “LIN28” relates to human LIN28 and preferably comprises an amino acid sequence encoded by the nucleic acid according to SEQ ID NO: 7, preferably the amino acid sequence according to SEQ ID NO: 8.
  • SEQ ID NO: 7 preferably the amino acid sequence according to SEQ ID NO: 8.
  • cDNA sequence of LIN28 as described above would be equivalent to LIN28 mRNA, and can be used for the generation of RNA capable of expressing LIN28.
  • rueppel-like factor (KLF4) is a zinc-finger transcription factor, which is strongly expressed in postmitotic epithelial cells of different tissues, e.g. the colon, the stomach and the skin. .LF4 is essential for the terminal differentiation of these cells and involved in the cell cycle regulation.
  • LF4 protein or simply " LF4" relates to human LF4 and preferably comprises an amino acid sequence encoded by the nucleic acid according to SEQ ID NO: 9, preferably the amino acid sequence according to SEQ ID NO: 10.
  • SEQ ID NO: 9 preferably the amino acid sequence according to SEQ ID NO: 10.
  • cDNA sequence of LF4 as described above would be equivalent to LF4 mRNA, and can be used for the generation of RNA capable of expressing KLF4.
  • M YC (cMYC) is a protooncogene, which is overexpressed in a wide range of human cancers. When it is specifically-mutated, or overexpressed, it increases cell proliferation and functions as an oncogene.
  • MYC gene encodes for a transcription factor that regulates expression of 15% of all genes through binding on Enhancer Box sequences (E-boxes) and recruiting histone acetyltransferases (HATs).
  • E-boxes Enhancer Box sequences
  • HATs histone acetyltransferases
  • MYC belongs to MYC family of transcription factors, which also includes N-MYC and L-MYC genes.
  • MYC-family transcription factors contain the bHLH/LZ (basic Helix-Loop-Helix Leucine Zipper) domain
  • cMYC protein or simply “cMYC” relates to human cMYC and preferably comprises an amino acid sequence encoded by the nucleic acid according to SEQ ID NO: 1 1 , preferably the amino acid sequence according to SEQ ID NO: 12.
  • SEQ ID NO: 1 1 preferably the amino acid sequence according to SEQ ID NO: 12.
  • a reference herein to specific factors such as OCT4, SOX2, NANOG, LI 28, KLF4 or c- MYC or to specific sequences thereof is to be understood so as to also include all variants of these specific factors or the specific sequences thereof as described herein.
  • miRNA relates to 21-23-nucleotide-long noncoding RNAs found in eukaryotic cells that, by inducing degradation and/or preventing translation of target mRNAs, modulate a plethora of cell functions, including those related to ESC self- renewal/differentiation and cell cycle progression.
  • miRNAs are post-transcriptional regulators that bind to complementary sequences on target messenger RNA transcripts (mRNAs), usually resulting in translational repression or target degradation and gene silencing. It has been found that miRNAs in the right combination are capable of inducing direct cellular reprogramming of somatic cells to cells having stem cell characteristics in vitro. For example, it has been observed that miRNA cluster 302-367 enhances somatic cell reprogramming.
  • the step of allowing the development of cells having stem cell characteristics used in the methods for providing cells having stem cell characteristics described herein comprises culturing the somatic cells under embryonic stem cell culture conditions, preferbly conditions suitable for maintaining pluripotent stem cells in an undifferentiated state.
  • cells are cultivated in the presence of one or more DNA methyltransferase inhibitors and/or one or more histone deacetylase inhibitors.
  • Preferred compounds are selected from the group consisting of 5'-azacytidine (5'-azaC), suberoylanilide hydroxamic acid (SAHA), dexamethasone, trichostatin A (TSA), sodium butyrate (NaBu), Scriptaid and valproic acid (VPA).
  • SAHA suberoylanilide hydroxamic acid
  • TSA trichostatin A
  • NaBu sodium butyrate
  • VP A valproic acid
  • cells are cultivated in the presence of valproic acid (VP A), preferably in a concentration of between 0.5 and 10 mM, more preferably between 1 and 5 mM, most preferably in a concentration of about 2 mM.
  • the methods of the present invention can be used to effect de-differentiation of any type of somatic cell.
  • Cells that may be used include cells that can be de-differentiated or reprogrammed by the methods of the present invention, in particular cells that are fully or partially differentiated, more preferably terminally differentiated.
  • the somatic cell is a diploid cell derived from pre-embryonic, embryonic, fetal, and post-natal multi-cellular organisms.
  • fibroblasts such as fetal and neonatal fibroblasts or adult fibroblasts
  • keratinocytes in particular primary keratinocytes, more preferably keratinocytes derived from hair, adipose cells, epithelial cells, epidermal cells, chondrocytes, cumulus cells, neural cells, glial cells, astrocytes, cardiac cells, esophageal cells, muscle cells, melanocytes, hematopoietic cells, osteocytes, macrophages, monocytes, and mononuclear cells.
  • fibroblasts such as fetal and neonatal fibroblasts or adult fibroblasts
  • keratinocytes in particular primary keratinocytes, more preferably keratinocytes derived from hair, adipose cells, epithelial cells, epidermal cells, chondrocytes, cumulus cells, neural cells, glial cells, astrocytes, cardiac cells,
  • the cells with which the methods of the invention can be used can be of any animal species; e.g., mammals and rodents.
  • mammalian cells that can be de-differentiated and re-differentiated by the present invention include but are not limited to human and non-human primate cells.
  • Primate cells with which the invention may be performed include but are not limited to cells of humans, chimpanzees, baboons, cynomolgus monkeys, and any other New or Old World monkeys.
  • Rodent cells with which the invention may be performed include but are not limited to mouse, rat, guinea pig, hamster and gerbil cells.
  • De-differentiated cells prepared according to the present invention are expected to display many of the same requirements as pluripotent stem cells and can be expanded and maintained under conditions used for embryonic stem cells, e.g. ES cell medium or any medium that supports growth of the embryonic cells.
  • Embryonic stem cells retain their pluripotency in vitro when maintained on inactivated fetal fibroblasts such as irradiated mouse embryonic fibroblasts or human fibroblasts (e.g., human foreskin fibroblasts, human skin fibroblasts, human endometrial fibroblasts, human oviductal fibroblasts) in culture.
  • the human feeder cells may be autologous feeder cells derived from the same culture of reprogrammed cells by direct differentiation.
  • human embryonic stem cells can successfully be propagated on Matrigel in a medium conditioned by mouse fetal fibroblasts. Human stem cells can be grown in culture for extended period of time and remain undifferentiated under specific culture conditions.
  • the cell culture conditions may include contacting the cells with factors that can inhibit differentiation or otherwise potentiate de-differentiation of cells, e.g., prevent the differentiation of cells into non-ES cells, trophectoderm or other cell types.
  • De-differentiated cells prepared according to the present invention can be evaluated by methods including monitoring changes in the cells' phenotype and characterizing their gene and protein expression. Gene expression can be determined by RT-PCR, and translation products can be determined by immunocytochemistry and Western blotting.
  • dedifferentiated cells can be characterized to determine the pattern of gene expression and whether the reprogrammed cells display a pattern of gene expression similar to the expression pattern expected of undifferentiated, pluripotent control cells such as embryonic stem cells using techniques well known in the art including transcriptomics.
  • the expression of the following genes of de-differentiated cells can be assessed in this respect: OCT4, NANOG, growth and differentiation factor 3 (GDF3), reduced expression 1 (REX1), fibroblast growth factor 4 (FGF4), embryonic cell-specific gene 1 (ESG1), developmental pluripotency-associated 2 (DPPA2), DPPA4, telomerase reverse transcriptase (TERT), embryonic antigen-3 (SSEA-3), SSEA-4, tumor-related antigen-1-60 (TRA-1-60), TRA-1-81, and TRA-2-49/6E.
  • the undifferentiated or embryonic stem cells to which the reprogrammed cells may be compared may be from the same species as the differentiated somatic cells. Alternatively, the undifferentiated or embryonic stem cells to which the reprogrammed cells may be compared may be from a different species as the differentiated somatic cells.
  • a similarity in gene expression pattern exists between a reprogrammed cell and an undifferentiated cell, e.g., embryonic stem cell, if certain genes specifically expressed in an undifferentiated cell are also expressed in the reprogrammed cell. For example, certain genes, e.g., telomerase, that are typically undetectable in differentiated somatic cells may be used to monitor the extent of reprogramming. Likewise, for certain genes, the absence of expression may be used to assess the extent of reprogramming.
  • Self-renewing capacity marked by induction of telomerase activity, is another characteristic of stem cells that can be monitored in de-differentiated cells.
  • aryotypic analysis may be performed by means of chromosome spreads from mitotic cells, spectral karyotyping, assays of telomere length, total genomic hybridization, or other techniques well known in the art.
  • RNA encoding appropriate factors is incorporated into one or more somatic cells, e.g. by electroporation.
  • cells are preferably cultured using conditions that support maintenance of de-differentiated cells (i.e. stem cell culture conditions).
  • the de-differentiated cells can then be expanded and induced to re-differentiate into different type of somatic cells that are needed for cell therapy.
  • De-differentiated cells obtained according to the present invention can be induced to differentiate into one or more desired somatic cell types in vitro or in vivo.
  • the de-differentiated cells obtained according to the present invention may give rise to cells from any of three embryonic germ layers, i.e., endoderm, mesoderm, and ectoderm.
  • the de-differentiated cells may differentiate into skeletal muscle, skeleton, dermis of skin, connective tissue, urogenital system, heart, blood (lymph cells), and spleen (mesoderm); stomach, colon, liver, pancreas, urinary bladder; lining of urethra, epithelial parts of trachea, lungs, pharynx, thyroid, parathyroid, intestine (endoderm); or central nervous system, retina and lens, cranial and sensory, ganglia and nerves, pigment cells, head connective tissue, epidermis, hair, mammary glands (ectoderm).
  • the dedifferentiated cells obtained according to the present invention can be re-differentiated in vitro or in vivo using techniques known in the art.
  • the reprogrammed cells resulting from the methods of this invention are used to produce differentiated progeny.
  • the present invention provides a method for producing differentiated cells, comprising: (i) obtaining reprogrammed cells using the methods of this invention; and (ii) inducing differentiation of the reprogrammed cells to produce differentiated cells. Step (ii) can be performed in vivo or in vitro.
  • differentiation can be induced through the presence of appropriate differentiation factors which can either be added or are present in situ, e.g.
  • the differentiated cells can be used to derive cells, tissues and/or organs which are advantageously used in the area of cell, tissue, and/or organ transplantation. If desired, genetic modifications can be introduced, for example, into somatic cells prior to reprogramming.
  • the differentiated cells of the present invention preferably do not possess the pluripotency of an embryonic stem cell, or an embryonic germ cell, and are, in essence, tissue-specific partially or fully differentiated cells.
  • One advantage of the methods of the present invention is that the reprogrammed cells obtained by the present invention can be differentiated without prior selection or purification or establishment of a cell line. Accordingly in certain embodiments, a heterogeneous population of cells comprising reprogrammed cells are differentiated into a desired cell type. In one embodiment, a mixture of cells obtained from the methods of the present invention is exposed to one or more differentiation factors and cultured in vitro.
  • Methods of differentiating reprogrammed cells obtained by the methods disclosed herein may comprise a step of permeabilization of the reprogrammed cell.
  • cells generated by the reprogramming techniques described herein, or alternatively a heterogeneous mixture of cells comprising reprogrammed cells may be permeabilized before exposure to one or more differentiation factors or cell extract or other preparation comprising differentiation factors.
  • differentiated cells may be obtained by culturing undifferentiated reprogrammed cells in the presence of at least one differentiation factor and selecting differentiated cells from the culture.
  • Selection of differentiated cells may be based on phenotype, such as the expression of certain cell markers present on differentiated cells, or by functional assays (e.g., the ability to perform one or more functions of a particular differentiated cell type).
  • the cells reprogrammed according to the present invention are genetically modified through the addition, deletion, or modification of their DNA sequence(s).
  • Reprogrammed or de-differentiated cells prepared according to the present invention or cells derived from the reprogrammed or de-differentiated cells are useful in research and in therapy.
  • Reprogrammed pluripotent cells may be differentiated into any of the cells in the body including, without limitation, skin, cartilage, bone skeletal muscle, cardiac muscle, renal, hepatic, blood and blood forming, vascular precursor and vascular endothelial, pancreatic beta, neurons, glia, retinal, neuronal, intestinal, lung, and liver cells.
  • the reprogrammed cells are useful for regenerative/reparative therapy and may be transplanted into a patient in need thereof.
  • the cells are autologous with the patient.
  • the reprogrammed cells provided in accordance with the present invention may be used, for example, in therapeutic strategies in the treatment of cardiac, neurological, endocrinological, vascular, retinal, dermatological, muscular-skeletal disorders, and other diseases.
  • the reprogrammed cells of the present invention can be used to replenish cells in animals whose natural cells have been depleted due to age or ablation therapy such as cancer radiotherapy and chemotherapy.
  • the reprogrammed cells of the present invention are useful in organ regeneration and tissue repair.
  • reprogrammed cells can be used to reinvigorate damaged muscle tissue including dystrophic muscles and muscles damaged by ischemic events such as myocardial infarcts.
  • the reprogrammed cells disclosed herein can be used to ameliorate scarring in animals, including humans, following a traumatic injury or surgery.
  • the reprogrammed cells of the present invention are administered systemically, such as intravenously, and migrate to the site of the freshly traumatized tissue recruited by circulating cytokines secreted by the damaged cells.
  • the reprogrammed cells can be administered locally to a treatment site in need or repair or regeneration.
  • the RNA used in the present invention encodes a peptide or protein which is of therapeutic value.
  • Cells containing the RNA can, for example, be manipulated in vitro to express the RNA and thus, the peptide or protein, using the methods of the invention. The cells expressing the peptide or protein can subsequently be introduced into a patient.
  • the RNA used in the present invention encodes a peptide or protein comprising an immunogen, antigen or antigen peptide.
  • the peptide or protein is processed after expression to provide said immunogen, antigen or antigen peptide.
  • the peptide or protein itself is the immunogen, antigen or antigen peptide.
  • Cells expressing such peptide or protein comprising an immunogen, antigen or antigen peptide can be used, for example, in immunotherapy to elicit an immune response against the immunogen, antigen or antigen peptide in a patient.
  • An "antigen" according to the invention covers any substance that will elicit an immune response.
  • an "antigen” relates to any substance that reacts specifically with antibodies or T-lymphocytes (T-cells).
  • the term "antigen” comprises any molecule which comprises at least one epitope.
  • an antigen in the context of the present invention is a molecule which, optionally after processing, induces an immune reaction, which is preferably specific for the antigen.
  • any suitable antigen may be used, which is a candidate for an immune reaction, wherein the immune reaction may be both a humoral as well as a cellular immune reaction.
  • the antigen is preferably presented by a cell, preferably by an antigen presenting cell, in the context of MHC molecules, which results in an immune reaction against the antigen.
  • An antigen is preferably a product which corresponds to or is derived from a naturally occurring antigen.
  • Naturally occurring antigens may include or may be derived from allergens, viruses, bacteria, fungi, parasites and other infectious agents and pathogens or an antigen may also be a tumor antigen.
  • an antigen may correspond to a naturally occurring product, for example, a viral protein, or a part thereof.
  • the antigen is a tumor antigen, i.e., a part of a tumor cell which may be derived from the cytoplasm, the cell surface or the cell nucleus, in particular those which primarily occur intracellularly or as surface antigens of tumor cells.
  • tumor antigens include the carcinoembryonal antigen, a 1 -fetoprotein, isoferritin, and fetal sulphoglycoprotein, a2-H-ferroprotein and ⁇ -fetoprotein, as well as various virus tumor antigens.
  • a tumor antigen preferably comprises any antigen which is characteristic for tumors or cancers as well as for tumor or cancer cells with respect to type and/or expression level.
  • the antigen is a virus antigen such as viral ribonucleoprotein or coat protein.
  • the antigen should be presented by MHC molecules which results in modulation, in particular activation of cells of the immune system, preferably CD4 + and CD8 + lymphocytes, in particular via the modulation of the activity of a T-cell receptor.
  • the antigen is a tumor antigen and the present invention involves the stimulation of an anti-tumor CTL response against tumor cells expressing such tumor antigen and preferably presenting such tumor antigen with class I MHC.
  • immunogenicity relates to the relative effectivity of an antigen to induce an immune reaction.
  • pathogenic microorganisms relates to pathogenic microorganisms and comprises viruses, bacteria, fungi, unicellular organisms, and parasites.
  • pathogenic viruses are human immunodeficiency virus (HIV), cytomegalovirus (CMV), herpes virus (HSV), hepatitis A- virus (HAV), HBV, HCV, papilloma virus, and human T-lympho trophic virus (HTLV).
  • Unicellular organisms comprise plasmodia trypanosomes, amoeba, etc.
  • antigens examples include p53, ART-4, BAGE, ss- catenin/m, Bcr-abL CAMEL, CAP-1, CASP-8, CDC27/m, CD 4/m, CEA, CLAUDIN-12, c- MYC, CT, Cyp-B, DAM, ELF2M, ETV6-AML1, G250, GAGE, GnT-V, Gapl OO, HAGE, HER-2/neu, HPV-E7, HPV-E6, HAST-2, hTERT (or hTRT), LAGE, LDLR/FUT, MAGE-A, preferably MAGE-A 1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A5, MAGE-A6, MAGE- A7, MAGE-A8, MAGE-A9, MAGE-A 10, MAGE-A 1 1, or MAGE-A 12, MAGE-B, MAGE- C, MART- 1 /Melan
  • the invention also makes use of peptides comprising amino acid sequences derived from antigens, also termed "antigen peptides" herein.
  • antigen peptide or “antigen peptide derived from an antigen” is meant an oligopeptide or polypeptide comprising an amino acid sequence substantially corresponding to the amino acid sequence of a fragment or peptide of an antigen.
  • An antigen peptide may be of any length.
  • the antigen peptides are capable of stimulating an immune response, preferably a cellular response against the antigen or cells characterized by expression of the antigen and preferably by presentation of the antigen.
  • an antigen peptide is capable of stimulating a cellular response against a cell characterized by presentation of an antigen with class I MHC and preferably is capable of stimulating an antigen-responsive CTL.
  • the antigen peptides according to the invention are MHC class I and/or class II presented peptides or can be processed to produce MHC class I and/or class II presented peptides.
  • the antigen peptides comprise an amino acid sequence substantially corresponding to the amino acid sequence of a fragment of an antigen.
  • said fragment of an antigen is an MHC class I and/or class II presented peptide.
  • an antigen peptide according to the invention comprises an amino acid sequence substantially corresponding to the amino acid sequence of such fragment and is processed to produce such fragment, i.e., an MHC class I and/or class II presented peptide derived from an antigen.
  • an antigen peptide is to be presented directly, i.e., without processing, in particular without cleavage, it has a length which is suitable for binding to an MHC molecule, in particular a class I MHC molecule, and preferably is 7-20 amino acids in length, more preferably 7-12 amino acids in length, more preferably 8-1 1 amino acids in length, in particular 9 or 10 amino acids in length.
  • the sequence of an antigen peptide which is to be presented directly is derived from the amino acid sequence of an antigen, i.e., its sequence substantially corresponds and is preferably completely identical to a fragment of an antigen.
  • the peptide produced by processing has a length which is suitable for binding to an MHC molecule, in particular a class I MHC molecule, and preferably is 7-20 amino acids in length, more preferably 7-12 amino acids in length, more preferably 8-1 1 amino acids in length, in particular 9 or 10 amino acids in length.
  • the sequence of the peptide which is to be presented following processing is derived from the amino acid sequence of an antigen, i.e., its sequence substantially corresponds and is preferably completely identical to a fragment of an antigen.
  • an antigen peptide according to the invention in one embodiment comprises a sequence of 7-20 amino acids in length, more preferably 7-12 amino acids in length, more preferably 8-1 1 amino acids in length, in particular 9 or 10 amino acids in length which substantially corresponds and is preferably completely identical to a fragment of an antigen and following processing of the antigen peptide makes up the presented peptide.
  • the antigen peptide may also comprise a sequence which substantially corresponds and preferably is completely identical to a fragment of an antigen which is even longer than the above stated sequence.
  • an antigen peptide may comprise the entire sequence of an antigen.
  • Peptides having amino acid sequences substantially corresponding to a sequence of a peptide which is presented by the class I MHC may differ at one or more residues that are not essential for TCR recognition of the peptide as presented by the class I MHC, or for peptide binding to MHC. Such substantially corresponding peptides are also capable of stimulating an antigen-responsive CTL.
  • Peptides having amino acid sequences differing from a presented peptide at residues that do not affect TCR recognition but improve the stability of binding to MHC may improve the immunogenicity of the antigen peptide, and may be referred to herein as "optimized peptide".
  • Antigen processing refers to the degradation of an antigen into fragments (e.g., the degradation of a protein into peptides) and the association of one or more of these fragments (e.g., via binding) with MHC molecules for presentation by "antigen presenting cells” to specific T-cells.
  • Antigen presenting cells are cells which present peptide fragments of protein antigens in association with MHC molecules on their cell surface. Some APCs may activate antigen-specific T-cells.
  • immunotherapy relates to a treatment involving activation of a specific immune reaction.
  • in vivo relates to the situation in a subject.
  • subject and “individual” are used interchangeably and relate to mammals.
  • mammals in the context of the present invention are humans, non-human primates, domesticated animals such as dogs, cats, sheep, cattle, goats, pigs, horses etc., laboratory animals such as mice, rats, rabbits, guinea pigs, etc. as well as animals in captivity such as animals of zoos.
  • animal as used herein also includes humans.
  • subject may also include a patient, i.e., an animal, preferably a human having a disease.
  • composition for administration is generally administered in pharmaceutically compatible amounts and in pharmaceutically compatible preparations.
  • pharmaceutically compatible refers to a nontoxic material which does not interact with the action of the active component of the pharmaceutical composition. Preparations of this kind may usually contain salts, buffer substances, preservatives, excipients and carriers and are administered in a manner known to the skilled person.
  • the present invention is described in detail by the figures and examples below, which are used only for illustration purposes and are not meant to be limiting. Owing to the description and the examples, further embodiments which are likewise included in the invention are accessible to the skilled worker.
  • Example 1 Repetitive transfer of IVT-RNA (Reprogramming-TF) Reprogramming of somatic cells into induced pluripotent stem cells (iPS) requires the continuous expression of reprogramming transcription factors (rTF). To avoid the risk of genomic integration which arises when the rTF are delivered virally into the cell, rTF can be efficiently delivered as mRNA by electroporation or lipofection without accompanied modifications of the host genome. Nevertheless the delivery has to be performed repetitively to assure constant expression of the rTF.
  • rTF reprogramming transcription factors
  • CCD1079Sk fibroblasts were electroporated as indicated in the side panel of Fig. 2 A either with 15 ⁇ g or 5 ⁇ g of each in vitro transcribed (IVT)-RNA encoding the transcription factors OCT4 (O), SOX2 (S), LF4 (K) and cMYC (M) and cultivated in human embryonic stem (ES) cell medium. Electroporations were performed in 4 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts.
  • CCD1079Sk fibroblasts were electroporated as indicated in the side panel of Fig. 2B with ⁇ g of each IVT-RNA encoding the transcription factors OSKM and cultivated in human ES cell medium. Electroporations were performed in 4 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts. At the indicated time points remaining cells were counted and survival rate in relation to the starting cells was calculated. Repetitive IVT-RNA transfer is accompanied with massive cell death and successful reprogramming is therefore not achievable.
  • CCD1079Sk fibroblasts were electroporated with 1 ⁇ g IVT RNA encoding for firefly luciferase (Luc) and 5 ⁇ g IVT RNA encoding for green fluorescent protein (GFP). Electroporations were performed in 2 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts. 24h post electroporation, cells were pelleted, total RNA was isolated and mRNA-expression of Interferon (IFN)-a and -b was quantified by qRT-PCR. It was observed that electroporation of IVT-RNA is followed by an induction of IFNa and b 24h thereafter; cf. Fig. 2C.
  • IFN Interferon
  • CCD1079Sk fibroblasts were electroporated with 33,4 ⁇ g IVT RNA encoding reprogramming mixture (29,5 ⁇ rTF (OS M NANOG (N) LIN28 (L) ( 1 : 1 : 1 : 1 : 1 : 1 )), ⁇ ,3 ⁇ SV40 largeT antigen (lgT), l ,3 ⁇ g HTLV E6 and l ,25 ⁇ g GFP). Electroporations were performed in 4 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts.
  • IFN-response genes are induced 48h post electroporation of IVT-RNA; cf. Fig. 2D.
  • the IFN-response has originally evolved as part of the host innate immune response to viral infections. Viral nucleic acids are efficiently recognized by sensor molecules which leads to antiviral activities including apoptosis, cytoskeletal remodeling, RNA degradation and a halt in protein translation. These mechanisms obviously hinder RNA-based gene transfer.
  • CCD1079Sk fibroblasts were electroporated once with the amounts of IVT-RNA encoding the reporter genes Luc, GFP or the Protein Kinase R (PKR) wild type indicated in Fig. 2E. Electroporations were performed in 4 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts. Cells were lysed 24h post electroporation and expression and phosphorylation status of the PKR target eukaryotic initiation factor 2a (eIF2a) was monitored by Western Blotting using specific antibodies. One of the major player in the IFN response is the PKR which upon activation phosphorylates its target eIF2a leading to an inhibition of translation.
  • eIF2a eukaryotic initiation factor 2a
  • eIF2a is phosphorylated 24h after electroporation of IVT- RNA identifying activation of PKR as one of the prominent obstacles in RNA-based reprogramming.
  • Repetitive RNA-based gene transfer is accompanied with an induction of the IFN-response which hinders the continuous expression of rTF and therefore successful RNA-based reprogramming.
  • Example 2 Use of E3, K3 and B18R in R A-based gene transfer
  • IVT-RNAs coding for the viral escape proteins E3, 3 and B 18R were added to a mixture of unmodified IVT-RNA (Luciferase/GFP), lipofected into human foreskin fibroblasts (HFF) and translation of the reporter gene GFP and IFN-response to the RNA was analyzed. Furthermore the survival of the cells after repetitive lipofections was assessed by an Cell Proliferation Kit II (Roche).
  • CCD1079Sk fibroblasts were plated into 6 wells (100,000 cells/well) and lipofected the next day using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • the IVT-RNA mixtures was thereby composed of 0 ⁇ g GFP with 0 ⁇ g of each B18R, E3 or K3 (as indicated in Fig. 3A,B).
  • IVT- RNA encoding for Luc was used to sum up the mixtures to 1.4 ⁇ Lipofections were performed according to the manufacturers instructions and cells were harvested 48h post transfection. 20% of the cells were used for analysis of GFP expression by FACS (Fig.
  • FIG. 3A Fig. 3 A
  • IFNb and the IFN-response gene OAS 1 are clearly induced by lipofection of IVT-RNA (Luc/GFP). This induction can be reduced in the case of IFNb by E3/K3 alone, but only the combination of all 3 viral escape proteins is able to reduce significantly both IFNb and OAS 1 -induction by IVT- RNA 48h post lipofection.
  • expression of the reporter gene GFP is enhanced by addition of E3 and K3.
  • B18R has no effect on the translation of GFP.
  • CCD1079Sk fibroblasts were plated into 6 wells (100.000 cells/well) and lipofected the next four consecutive days using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • the IVT-RNA mixture was thereby composed of 0 ⁇ g GFP with 0 ⁇ g of each B18R, E3 or K3 (as indicated).
  • IVT-RNA encoding for Luc was used to sum up the mixture to 1 ⁇ g total IVT- RNA.
  • l ⁇ g modified (mod.) IVT-RNA encoding for Luc (0 ⁇ g) and GFP (0 ⁇ g) was used.
  • RNAs were composed of 100% pseudouridine (psi) and 100% 5- methylcytidine (5mC) instead of uridine and cytidine which display less immunstimulatory characteristics. Lipofections were performed according to the manufacturers instructions. 24h after the last lipofection, cell viability was assayed using the Cell Proliferation Kit II (Roche) and normalized to the mock transfected cells. As shown in Fig. 3C, daily lipofection of unmodified IVT-RNA (Luc/GFP, 4 times) is accompanied with massive loss in cell viability. The combination of all 3 viral escape proteins is able to overcome this obstacle and enhances survival of the cells even more than the use of modified IVT-RNA (100% psi and 5mC). It is concluded that repetitive RNA-based gene transfer is possible when the combination of E3, 3 and B18R coded by IVT-RNA is added. Proof of concept was achieved.
  • Example 3 Use of E3, K3 and B18R in RNA-based gene transfer for reprogramming
  • CCD1079Sk fibroblasts were plated into 6 wells (80,000 cells/well) and lipofected the next four consecutive days using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • the IVT-RNA mixtures were thereby composed of 0 ⁇ g unmodified GFP or 0 ⁇ g OSKMNL (1 : 1 : 1 : 1 : 1 : 1) either unmodified or modified and either with 0 ⁇ g of each B18R, E3 and K3 unmodified or modified. If necessary IVT-RNA encoding for Luc was used to sum up the mixture to 1 ⁇ g total IVT-RNA.
  • RNAs were composed of 100% psi and 100% 5mC instead of uridine and cytidine which display less immunstimulatory characteristics. Lipofections were performed according to the manufacturers instructions. 24h after the last lipofection, cell viability was assayed using the Cell Proliferation Kit II (Roche) with normalization to mock transfected cells (Fig. 4A) and by microscopy (Fig. 4B). After that, cells were pelleted, total RNA was isolated and mRNA-expression of IFNb and OAS 1 was quantified by qRT-PCR (Fig. 4C).
  • CCD1079Sk fibroblasts were plated into 6 wells (100,000 cells/well) and lipofected the next three consecutive days using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • the IVT-RNA mixtures was thereby composed of 0 ⁇ g GFP with 0 ⁇ g OCT4 or SOX2 or NANOG either unmodified or modified and 0 ⁇ g of each B18R, E3 and K3 (EKB) either unmodified or modified.
  • Modified RNAs were composed of 100% psi and 100% 5mC instead of uridine and cytidine which display less immunstimulatory characteristics. Lipofections were performed according to the manufacturers instructions.
  • OSN intracellular expression of OSN was monitored by FACS analysis using the human pluripotent stem cell transcription factor analysis kit (BD 560589). Expression levels of NANOG, OCT4 and SOX2 were higher in the presence of EKB when applicated unmodified; cf. Fig. 5.
  • rTF-mixture Repetitive lipofection of rTF-mixture was achieved by addition of EKB to the reprogramming mixture leading to a better survival and higher reduction of IFN-response after daily lipofections. Furthermore higher expression levels of rTF were achieved in the presence of E B. In the next experiments these mixture was used for long time lipofections to achieve reprogramming of HFFs. To further enhance reprogramming the microRNAs of cluster 302/367 were added to the reprogramming mixture as well. These miRNAs are thought to be mainly involved in the cellular maintenance of self-renewal and pluripotency, and could lead to reprogramming when expressed by lentiviral vectors (Anokye-Danso et al., 201 1).
  • HFF fibroblasts (System Bioscience) were plated into 6 wells (100,000 cells/well) and lipofected 5 times a week (Monday to Friday) for two weeks using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT-RNA (Fig. 6A).
  • the IVT-RNA mixtures were thereby composed of 0 ⁇ g unmodified GFP or 0 ⁇ g OSKMNL (1 : 1 : 1 : 1 :1 : 1) either unmodified or modified with either 0 ⁇ g of each B18R, E3 and K.3 (EKB) either unmodified or modified and 0 ⁇ g of a miRNA mixture composed of miRNAs 302a-d and 367 [0.4 ⁇ each].
  • RNAs were composed of 100% psi and 100% 5mC instead of uridine and cytidine which display less immunstimulatory characteristics.
  • Lipofections in stem cell media were performed according to the manufacturers instructions. On day 6 and day 13, cells were pelleted, total RNA was isolated and mRNA-expression of the human ES-marker TERT, DPPA4, GDF3, LIN28 (endogenous) and REXl was quantified by qRT-PCR. Colony growth was observed by microscopy and for further analysis, colonies were stained for the ES surface marker TRA-1-60 using the StainAlive TRA- 1-60 antibody (Stemgent) following the manufacturers instructions.
  • Example 6 Reprogramming of HFF using rTF and microR A in the presence of EKB (splitting 1:8)
  • EKB and miRNA-mixture 302/367 cells were splitted 1 :4.
  • cells were splitted 1 :8 to avoid a dense growing of the cells.
  • HFF fibroblasts (System Bioscience) were plated into 6 wells (100,000 cells/well) and lipofected 5 times a week (Monday to Friday) for two weeks using 6 ⁇ 1 RNAiMAX (Invitrogen) and 1 ⁇ g IVT-RNA (Fig. 7A).
  • the IVT-RNA mixtures were thereby composed of 0 ⁇ g OSKMNL (1 : 1 :1 : 1 : 1 : 1) either unmodified or modified with either 0 ⁇ g of each B18R, E3 and K3 (EKB) unmodified or modified and 0 ⁇ g of a miRNA mixture composed of miRNAs 302a-d and 367 [0.4 ⁇ each].
  • RNAs were composed of 100% psi and 100% 5mC instead of uridine and cytidine which display less immunstimulatory characteristics.
  • Lipofections in stem cell media were performed according to the manufacturers instructions. On day 5 and day 12, cells were pelleted, total RNA was isolated and mRNA-expression of the human ES-marker TERT, DPPA4, GDF3, LIN28 (endogenous) and REX1 was quantified by qRT-PCR.
  • Colony growth was observed by microscopy and for further analysis, colonies were stained for the ES surface marker TRA- 1-60 using the StainAlive TRA-1-60 antibody (Stemgent) or for the activity of alkaline phosphatase (Vector Red staining kit) following the manufacturers instructions.
  • HFF fibroblasts (System Bioscience) were plated into 6 wells (100,000 cells/well) and lipofected the next four consecutive days using 6 ⁇ 1 RNAiMAX (Invitrogen) and l ⁇ g IVT.
  • the IVT-RNA mixtures were thereby composed of 0 ⁇ g unmodified OSKMNL ( 1 : 1 : 1 : 1 : 1 : 1) with variable amounts of unmodfied B18R, E3 and K3 as indicated.
  • IVT-RNA encoding for Luc was used to sum up the mixture to l ⁇ g total IVT-RNA. According to the reprogramming experiments 0 ⁇ g of a miRNA mixture composed of miRNAs 302a-d and 367 [0.4 ⁇ each] was added to the samples.
  • RNAs were composed of 100% psi and 5mC instead of uridine and cytidine which display less immunstimulatory characteristics. Lipofections were performed according to the manufacturers instructions. 24h after the last lipofection, cell viability was assayed using the Cell Proliferation Kit II (Roche). After that, cells were pelleted, total RNA was isolated and mRNA-expression of IFNb and OAS 1 was quantified by qRT-PCR. As shown in Fig.
  • EKB can be reduced to 0.025-0.05 ⁇ g of each IVT-RNA.
  • CCD 1079SK fibroblasts were electroporated with IVT RNA encoding Luc (1 ⁇ g), GFP (5 ⁇ g) and 3 ⁇ g of E3 or K3 or both as indicated. Electroporations were performed in 2 mm gap cuvettes using optimized parameters for CCD1079Sk fibroblasts. 10000 cells/well were plated in duplicates into 96-well-plates. Luciferase activity was measured at the time points indicated in Fig. 9A after electroporation using the Bright Glo Luciferase Assay System (Promega). Mean values of the duplicates are given.
  • Fig. 9A the translation of Luc was enhanced with E3 or K3 alone as much as with both of them together.
  • IFNb and the IFN-response gene OAS 1 are clearly induced by electroporation of IVT-RNA (Luc/GFP). These inductions cannot be reduced neither by E3 or K3 nor by the combination of both 24h post electroporation; cf. Fig. 9B.

Abstract

La présente invention concerne l'expression d'ARN dans des cellules et, en particulier, l'amélioration de la viabilité de cellules dans lesquelles l'ARN doit être exprimé. Spécifiquement, la présente invention concerne des procédés pour l'expression d'ARN dans des cellules, comprenant les étapes consistant à prévenir l'engagement du récepteur IFN par IFN extracellulaire et inhiber la signalisation d'IFN intracellulaire dans les cellules. Ainsi, la prévention de l'engagement de récepteur IFN par IFN extracellulaire et l'inhibition de la signalisation d'IFN intracellulaire dans les cellules permet le transfert répétitif d'ARN dans les cellules.
PCT/EP2012/004673 2012-11-09 2012-11-09 Procédé pour l'expression d'arn cellulaire WO2014071963A1 (fr)

Priority Applications (10)

Application Number Priority Date Filing Date Title
PCT/EP2012/004673 WO2014071963A1 (fr) 2012-11-09 2012-11-09 Procédé pour l'expression d'arn cellulaire
AU2013343864A AU2013343864B2 (en) 2012-11-09 2013-11-07 Method for cellular RNA expression
ES13792854.5T ES2676470T3 (es) 2012-11-09 2013-11-07 Método para la expresión de ARN en células
TR2018/09547T TR201809547T4 (tr) 2012-11-09 2013-11-07 Hücresel RNA ifadesine yönelik yöntem.
EP13792854.5A EP2917350B1 (fr) 2012-11-09 2013-11-07 Méthode pour l'expression cellulaire de rna
PCT/EP2013/003362 WO2014072061A1 (fr) 2012-11-09 2013-11-07 Procédé pour l'expression d'arn cellulaire
CA2890529A CA2890529C (fr) 2012-11-09 2013-11-07 Methode de modification de l'expression d'arn cellulaire comprenant des recepteurs d'interferon (ifn) et la signalisation
JP2015541037A JP6353846B2 (ja) 2012-11-09 2013-11-07 細胞のrna発現方法
US14/706,228 US10207009B2 (en) 2012-11-09 2015-05-07 Method for cellular RNA expression
US16/245,353 US10729784B2 (en) 2012-11-09 2019-01-11 Method for cellular RNA expression

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/EP2012/004673 WO2014071963A1 (fr) 2012-11-09 2012-11-09 Procédé pour l'expression d'arn cellulaire

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EPPCT/EP2013/002234 Continuation 2012-11-09 2013-07-29

Publications (1)

Publication Number Publication Date
WO2014071963A1 true WO2014071963A1 (fr) 2014-05-15

Family

ID=47191691

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/004673 WO2014071963A1 (fr) 2012-11-09 2012-11-09 Procédé pour l'expression d'arn cellulaire

Country Status (1)

Country Link
WO (1) WO2014071963A1 (fr)

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
EP2861612A4 (fr) * 2012-06-13 2016-02-17 Stemgent Inc Méthodes de préparation de cellules souches pluripotentes
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
WO2017127750A1 (fr) 2016-01-22 2017-07-27 Modernatx, Inc. Acides ribonucléiques messagers pour la production de polypeptides de liaison intracellulaires et leurs procédés d'utilisation
WO2017162461A1 (fr) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Arn à réplication trans
WO2017180917A2 (fr) 2016-04-13 2017-10-19 Modernatx, Inc. Compositions lipidiques et leurs utilisations pour l'administration intratumorale de polynucléotides
WO2017201350A1 (fr) 2016-05-18 2017-11-23 Modernatx, Inc. Polynucléotides codant pour l'interleukine 12 (il-12) et leurs utilisations
WO2018011406A1 (fr) 2016-07-15 2018-01-18 Biontech Rna Pharmaceuticals Gmbh Formulation d'administration d'arn.
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
WO2018231990A2 (fr) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucléotides codant pour la méthylmalonyl-coa mutase
WO2019053012A1 (fr) 2017-09-13 2019-03-21 Biontech Rna Pharmaceuticals Gmbh Réplicon d'arn pour la reprogrammation de cellules somatiques
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2019137999A1 (fr) 2018-01-11 2019-07-18 Biontech Rna Pharmaceuticals Gmbh Formulation d'administration d'arn.
WO2019148101A1 (fr) 2018-01-29 2019-08-01 Modernatx, Inc. Vaccins à base d'arn contre le vrs
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US10493143B2 (en) 2015-10-22 2019-12-03 Modernatx, Inc. Sexually transmitted disease vaccines
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
EP3701959A1 (fr) 2016-03-21 2020-09-02 BioNTech RNA Pharmaceuticals GmbH Réplicon d'arn pour une expression génique polyvalente et efficace
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
WO2021154763A1 (fr) 2020-01-28 2021-08-05 Modernatx, Inc. Vaccins à arn contre le coronavirus
WO2021159040A2 (fr) 2020-02-07 2021-08-12 Modernatx, Inc. Vaccins à domaine arnm anti sars-cov-2
WO2021159130A2 (fr) 2020-05-15 2021-08-12 Modernatx, Inc. Vaccins a arn de coronavirus et procédés d'utilisation
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
WO2021222304A1 (fr) 2020-04-27 2021-11-04 Modernatx, Inc. Vaccins à arn contre le sars-cov-2
WO2021245090A1 (fr) 2020-06-04 2021-12-09 BioNTech SE Réplicon d'arn pour obtenir une expression génique versatile et efficace
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
WO2022067010A1 (fr) 2020-09-25 2022-03-31 Modernatx, Inc. Vaccins à base de protéine de spicule de coronavirus à substitution multiproline
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
WO2022197624A1 (fr) 2021-03-15 2022-09-22 Modernatx, Inc. Utilisation thérapeutique de vaccins à domaine arnm de sars-cov-2
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
WO2023025404A1 (fr) 2021-08-24 2023-03-02 BioNTech SE Technologies de transcription in vitro
WO2023066875A1 (fr) 2021-10-18 2023-04-27 BioNTech SE Arn réplicable modifié et compositions associées et leur utilisation
WO2023069498A1 (fr) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Composition de vaccin à base d'arnm
WO2023066874A1 (fr) 2021-10-18 2023-04-27 BioNTech SE Procédés pour déterminer les mutations permettant d'augmenter la fonction de l'arn modifié réplicable et compositions connexes et leur utilisation
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
WO2023092069A1 (fr) 2021-11-18 2023-05-25 Modernatx, Inc. Vaccins à domaine arnm de sars-cov-2 et méthodes d'utilisation
WO2023096858A1 (fr) 2021-11-23 2023-06-01 Senda Biosciences, Inc. Composition lipidique dérivée de bactéries et son utilisation
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
WO2024015890A1 (fr) 2022-07-13 2024-01-18 Modernatx, Inc. Vaccins à arnm de norovirus
WO2024056856A1 (fr) 2022-09-15 2024-03-21 BioNTech SE Systèmes et compositions comportant des vecteurs d'arn trans-amplifiants avec miarn

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020086356A1 (en) 2000-03-30 2002-07-04 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020086356A1 (en) 2000-03-30 2002-07-04 Whitehead Institute For Biomedical Research RNA sequence-specific mediators of RNA interference

Non-Patent Citations (18)

* Cited by examiner, † Cited by third party
Title
CHEN YIN ET AL: "Rhinovirus induces airway epithelial gene expression through double-stranded RNA and IFN-dependent pathways", AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY, vol. 34, no. 2, 1 February 2006 (2006-02-01), AMERICAN LUNG ASSOCIATION, NEW YORK, NY, US, pages 192 - 203, XP002487498, ISSN: 1044-1549, DOI: 10.1165/RCMB.2004-0417OC *
DAISY A. ROBINTON ET AL: "The promise of induced pluripotent stem cells in research and therapy", NATURE, vol. 481, no. 7381, 18 January 2012 (2012-01-18), pages 295 - 305, XP055050160, ISSN: 0028-0836, DOI: 10.1038/nature10761 *
DR. THOMAS TUSCHL: "The siRNA User Guide", LABORATORY OF RNA MOLECULAR BIOLOGY
GARCÍA-SASTRE ADOLFO ET AL: "Type 1 interferons and the virus-host relationship: a lesson in détente .", SCIENCE, vol. 312, no. 5775, 12 May 2006 (2006-05-12), (NEW YORK, N.Y.), pages 879 - 882, XP008159457, ISSN: 1095-9203 *
GEERTRUI TAVERNIER ET AL: "Activation of pluripotency-associated genes in mouse embryonic fibroblasts by non-viral transfection with invitro-derived mRNAs encoding Oct4, Sox2, Klf4 and cMyc", BIOMATERIALS, vol. 33, no. 2, 23 September 2011 (2011-09-23), ELSEVIER SCIENCE PUBLISHERS BV., BARKING, GB, pages 412 - 417, XP028101920, ISSN: 0142-9612, [retrieved on 20110926], DOI: 10.1016/J.BIOMATERIALS.2011.09.062 *
H.G.W. LEUENBERGER, B. NAGEL, AND H. KOLBL,: "Helvetica Chimica Acta", 1995, article "A multilingual glossary of biotechnological terms: (IUPAC Recommendations"
J. SAMBROOK ET AL.: "Molecular Cloning: A Laboratory Manual, 2nd Edition,", 1989, COLD SPRING HARBOR LABORATORY PRESS
JORDAN R PLEWS ET AL: "Activation of pluripotency genes in human fibroblast cells by a novel mRNA based approach", PLOS ONE, vol. 5, no. 12, E14397, 1 December 2010 (2010-12-01), PUBLIC LIBRARY OF SCIENCE, US, pages 1 - 10, XP002635012, ISSN: 1932-6203, DOI: 10.1371/JOURNAL.PONE.0014397 *
KATHARINA DREWS ET AL: "The cytotoxic and immunogenic hurdles associated with non-viral mRNA-mediated reprogramming of human fibroblasts", BIOMATERIALS, vol. 33, no. 16, 9 February 2012 (2012-02-09), ELSEVIER SCIENCE PUBLISHERS BV., BARKING, GB, pages 4059 - 4068, XP028474938, ISSN: 0142-9612, [retrieved on 20120214], DOI: 10.1016/J.BIOMATERIALS.2012.02.025 *
KUHN ANDREAS N ET AL: "mRNA as a versatile tool for exogenous protein expression.", CURRENT GENE THERAPY, vol. 12, no. 5, 1 October 2012 (2012-10-01), pages 347 - 361, XP008159357, ISSN: 1875-5631 *
LUIGI WARREN ET AL: "Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA", CELL STEM CELL, vol. 7, no. 5, 5 November 2010 (2010-11-05), CELL PRESS, US, pages 618 - 630, XP002640639, ISSN: 1934-5909, [retrieved on 20100930], DOI: 10.1016/J.STEM.2010.08.012 *
MATTHEW ANGEL ET AL: "Innate Immune Suppression Enables Frequent Transfection with RNA Encoding Reprogramming Proteins", PLOS ONE, vol. 5, no. 7, E11756, 23 July 2010 (2010-07-23), pages 1 - 7, XP055047047, DOI: 10.1371/journal.pone.0011756 *
NEDDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
NISHIKAWA SHIN-ICHI ET AL: "The promise of human induced pluripotent stem cells for research and therapy", NATURE REVIEWS MOLECULAR CELL BIOLOGY, vol. 9, no. 9, 1 September 2008 (2008-09-01), NATURE PUBLISHING, GB, pages 725 - 729, XP002520703, ISSN: 1471-0072, DOI: 10.1038/NRM2466 *
PEARSON; LIPMAN, PROC. NATL ACAD. SCI. USA, vol. 85, 1988, pages 2444
SMITH; WATERMAN, ADS APP. MATH., vol. 2, 1981, pages 482
TUSCHL T. ET AL.: "The siRNA User Guide", 11 October 2002
YAKUBOV EDUARD ET AL: "Reprogramming of human fibroblasts to pluripotent stem cells using mRNA of four transcription factors.", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 394, no. 1, 26 March 2010 (2010-03-26), pages 189 - 193, XP008159426, ISSN: 1090-2104 *

Cited By (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
EP2861612A4 (fr) * 2012-06-13 2016-02-17 Stemgent Inc Méthodes de préparation de cellules souches pluripotentes
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US10022435B2 (en) 2014-04-23 2018-07-17 Modernatx, Inc. Nucleic acid vaccines
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11007260B2 (en) 2015-07-21 2021-05-18 Modernatx, Inc. Infectious disease vaccines
US10702597B2 (en) 2015-07-21 2020-07-07 Modernatx, Inc. CHIKV RNA vaccines
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US11643441B1 (en) 2015-10-22 2023-05-09 Modernatx, Inc. Nucleic acid vaccines for varicella zoster virus (VZV)
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
US10493143B2 (en) 2015-10-22 2019-12-03 Modernatx, Inc. Sexually transmitted disease vaccines
US11278611B2 (en) 2015-10-22 2022-03-22 Modernatx, Inc. Zika virus RNA vaccines
WO2017127750A1 (fr) 2016-01-22 2017-07-27 Modernatx, Inc. Acides ribonucléiques messagers pour la production de polypeptides de liaison intracellulaires et leurs procédés d'utilisation
EP3964584A1 (fr) 2016-03-21 2022-03-09 BioNTech SE Arn transrépliquant
WO2017162461A1 (fr) 2016-03-21 2017-09-28 Biontech Rna Pharmaceuticals Gmbh Arn à réplication trans
EP3701959A1 (fr) 2016-03-21 2020-09-02 BioNTech RNA Pharmaceuticals GmbH Réplicon d'arn pour une expression génique polyvalente et efficace
WO2017180917A2 (fr) 2016-04-13 2017-10-19 Modernatx, Inc. Compositions lipidiques et leurs utilisations pour l'administration intratumorale de polynucléotides
WO2017201350A1 (fr) 2016-05-18 2017-11-23 Modernatx, Inc. Polynucléotides codant pour l'interleukine 12 (il-12) et leurs utilisations
EP3881827A1 (fr) 2016-07-15 2021-09-22 BioNTech RNA Pharmaceuticals GmbH Formulation d'administration d'arn
WO2018011406A1 (fr) 2016-07-15 2018-01-18 Biontech Rna Pharmaceuticals Gmbh Formulation d'administration d'arn.
US11696946B2 (en) 2016-11-11 2023-07-11 Modernatx, Inc. Influenza vaccine
US10925958B2 (en) 2016-11-11 2021-02-23 Modernatx, Inc. Influenza vaccine
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US11752206B2 (en) 2017-03-15 2023-09-12 Modernatx, Inc. Herpes simplex virus vaccine
US11576961B2 (en) 2017-03-15 2023-02-14 Modernatx, Inc. Broad spectrum influenza virus vaccine
US11464848B2 (en) 2017-03-15 2022-10-11 Modernatx, Inc. Respiratory syncytial virus vaccine
US11045540B2 (en) 2017-03-15 2021-06-29 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
US11918644B2 (en) 2017-03-15 2024-03-05 Modernatx, Inc. Varicella zoster virus (VZV) vaccine
WO2018231990A2 (fr) 2017-06-14 2018-12-20 Modernatx, Inc. Polynucléotides codant pour la méthylmalonyl-coa mutase
WO2019053012A1 (fr) 2017-09-13 2019-03-21 Biontech Rna Pharmaceuticals Gmbh Réplicon d'arn pour la reprogrammation de cellules somatiques
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
WO2019137999A1 (fr) 2018-01-11 2019-07-18 Biontech Rna Pharmaceuticals Gmbh Formulation d'administration d'arn.
WO2019148101A1 (fr) 2018-01-29 2019-08-01 Modernatx, Inc. Vaccins à base d'arn contre le vrs
US11911453B2 (en) 2018-01-29 2024-02-27 Modernatx, Inc. RSV RNA vaccines
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
WO2021154763A1 (fr) 2020-01-28 2021-08-05 Modernatx, Inc. Vaccins à arn contre le coronavirus
WO2021159040A2 (fr) 2020-02-07 2021-08-12 Modernatx, Inc. Vaccins à domaine arnm anti sars-cov-2
WO2021222304A1 (fr) 2020-04-27 2021-11-04 Modernatx, Inc. Vaccins à arn contre le sars-cov-2
WO2021159130A2 (fr) 2020-05-15 2021-08-12 Modernatx, Inc. Vaccins a arn de coronavirus et procédés d'utilisation
WO2021245090A1 (fr) 2020-06-04 2021-12-09 BioNTech SE Réplicon d'arn pour obtenir une expression génique versatile et efficace
WO2022067010A1 (fr) 2020-09-25 2022-03-31 Modernatx, Inc. Vaccins à base de protéine de spicule de coronavirus à substitution multiproline
WO2022197624A1 (fr) 2021-03-15 2022-09-22 Modernatx, Inc. Utilisation thérapeutique de vaccins à domaine arnm de sars-cov-2
WO2023025404A1 (fr) 2021-08-24 2023-03-02 BioNTech SE Technologies de transcription in vitro
WO2023066875A1 (fr) 2021-10-18 2023-04-27 BioNTech SE Arn réplicable modifié et compositions associées et leur utilisation
WO2023066874A1 (fr) 2021-10-18 2023-04-27 BioNTech SE Procédés pour déterminer les mutations permettant d'augmenter la fonction de l'arn modifié réplicable et compositions connexes et leur utilisation
WO2023069498A1 (fr) 2021-10-22 2023-04-27 Senda Biosciences, Inc. Composition de vaccin à base d'arnm
WO2023092069A1 (fr) 2021-11-18 2023-05-25 Modernatx, Inc. Vaccins à domaine arnm de sars-cov-2 et méthodes d'utilisation
WO2023096858A1 (fr) 2021-11-23 2023-06-01 Senda Biosciences, Inc. Composition lipidique dérivée de bactéries et son utilisation
WO2024015890A1 (fr) 2022-07-13 2024-01-18 Modernatx, Inc. Vaccins à arnm de norovirus
WO2024056856A1 (fr) 2022-09-15 2024-03-21 BioNTech SE Systèmes et compositions comportant des vecteurs d'arn trans-amplifiants avec miarn

Similar Documents

Publication Publication Date Title
US10729784B2 (en) Method for cellular RNA expression
CA2819522C (fr) Procede pour l'expression cellulaire d'arn comprenant la reduction de l'activite de la proteine kinase dependante de l'arn
WO2014071963A1 (fr) Procédé pour l'expression d'arn cellulaire
ES2532125T3 (es) Uso de ARN para reprogramar células somáticas
JP7424968B2 (ja) 細胞におけるrna発現を増強する方法
EP2917350B1 (fr) Méthode pour l'expression cellulaire de rna
JP2020534823A (ja) 体細胞を再プログラムするためのrnaレプリコン
EP2646557B1 (fr) Procédé pour l'expression cellulaire d'arn
RU2784654C2 (ru) Способ повышения экспрессии рнк в клетке

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: 12787651

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC, FORM 1205A DATED 28-08-2015

122 Ep: pct application non-entry in european phase

Ref document number: 12787651

Country of ref document: EP

Kind code of ref document: A1