WO2024098038A2 - Polynucleotide construct and related viral vectors and methods - Google Patents
Polynucleotide construct and related viral vectors and methods Download PDFInfo
- Publication number
- WO2024098038A2 WO2024098038A2 PCT/US2023/078748 US2023078748W WO2024098038A2 WO 2024098038 A2 WO2024098038 A2 WO 2024098038A2 US 2023078748 W US2023078748 W US 2023078748W WO 2024098038 A2 WO2024098038 A2 WO 2024098038A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- seq
- nucleotide sequence
- amino acid
- acid sequence
- cell
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/30—Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/11—T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/30—Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
- A61K40/31—Chimeric antigen receptors [CAR]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/30—Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
- A61K40/35—Cytokines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
- A61K40/4202—Receptors, cell surface antigens or cell surface determinants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/40—Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
- A61K40/41—Vertebrate antigens
- A61K40/42—Cancer antigens
- A61K40/4202—Receptors, cell surface antigens or cell surface determinants
- A61K40/421—Immunoglobulin superfamily
- A61K40/4211—CD19 or B4
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70528—CD58
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/70532—B7 molecules, e.g. CD80, CD86
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/715—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
- C07K14/7155—Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for interleukins [IL]
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/44—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/10—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterized by the structure of the chimeric antigen receptor [CAR]
- A61K2239/17—Hinge-spacer domain
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/10—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterized by the structure of the chimeric antigen receptor [CAR]
- A61K2239/22—Intracellular domain
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/10—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterized by the structure of the chimeric antigen receptor [CAR]
- A61K2239/23—On/off switch
- A61K2239/24—Dimerizable CARs; CARs with adapter
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K2239/00—Indexing codes associated with cellular immunotherapy of group A61K40/00
- A61K2239/31—Indexing codes associated with cellular immunotherapy of group A61K40/00 characterized by the route of administration
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/60—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
- C07K2317/62—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
- C07K2317/622—Single chain antibody (scFv)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/02—Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/03—Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/33—Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/50—Fusion polypeptide containing protease site
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/70—Fusion polypeptide containing domain for protein-protein interaction
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2740/00—Reverse transcribing RNA viruses
- C12N2740/00011—Details
- C12N2740/10011—Retroviridae
- C12N2740/16011—Human Immunodeficiency Virus, HIV
- C12N2740/16041—Use of virus, viral particle or viral elements as a vector
- C12N2740/16043—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2800/00—Nucleic acids vectors
- C12N2800/22—Vectors comprising a coding region that has been codon optimised for expression in a respective host
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2830/00—Vector systems having a special element relevant for transcription
- C12N2830/20—Vector systems having a special element relevant for transcription transcription of more than one cistron
Definitions
- the present disclosure provides polycistronic constructs for coexpression of a synthetic cytokine receptor complex and a chimeric antigen receptor system, as well as vectors, such as viral vectors comprising same, cells comprising same, and methods of using same.
- Chimeric antigen receptor (CAR) T cell therapies have demonstrated limited efficacy against solid tumors, in part due to challenges overcoming solid tumor heterogeneity and CAR T cell exhaustion associated with the immunosuppressive tumor microenvironment (TME).
- TEE immunosuppressive tumor microenvironment
- CAR-expressing cells to subjects in a manner that can provide controllable persistence for therapeutic efficacy of the CAR in the treatment of various diseases, including cancer.
- a polycistronic construct containing in 5’ to 3’ order (a) a first expression cassette including a nucleotide sequence encoding FRB, (b) a second expression cassette including a nucleotide sequence encoding a synthetic cytokine gamma chain polypeptide, (c) a third expression cassette including a nucleotide sequence encoding a synthetic cytokine beta chain polypeptide, and (d) a fourth expression cassette including a nucleotide sequence encoding a chimeric antigen receptor (CAR), wherein each of the expression cassettes are separated by a cleavage site sequence.
- a first expression cassette including a nucleotide sequence encoding FRB
- a second expression cassette including a nucleotide sequence encoding a synthetic cytokine gamma chain polypeptide
- a third expression cassette including a nucleotide sequence encoding a synthetic cytokine beta chain polypeptide
- the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:3, 13, or 50. In some of any embodiments, the nucleotide sequence encoding the FRB includes the nucleotide sequence of SEQ ID Nos:3, 13, or 50. In some of any embodiments, the FRB includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some of any embodiments, the FRB includes the amino acid sequence of SEQ ID NOs:4, 14, or 51.
- the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID No: 15. In some of any embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide includes the nucleotide sequence of SEQ ID No: 15.
- the synthetic cytokine gamma chain polypeptide includes interleukin 2 receptor subunit y (IL2RG).
- IL2RG interleukin 2 receptor subunit y
- the IL2RG includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID No: 16.
- the IL2RG includes the amino acid sequence of SEQ ID No: 16.
- the second expression cassette further includes a nucleotide sequence encoding FRB.
- the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB includes the nucleotide sequence of SEQ ID NO: 13.
- the FRB includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 14.
- the FRB includes the amino acid sequence of SEQ ID NO: 14.
- the second expression cassette is codon optimized.
- the second expression cassette includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 11. In some of any embodiments, the second expression cassette includes the nucleotide sequence of SEQ ID NO:11. In some of any embodiments, the second expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 12. In some of any embodiments, the second expression cassette encodes an amino acid sequence including the sequence of SEQ ID NO: 12.
- the second expression cassette further includes a nucleotide sequence encoding FKBP12.
- the nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:21 or 55.
- the nucleotide sequence encoding the FKBP12 includes the nucleotide sequence of SEQ ID Nos:21 or 55.
- the FKBP12 includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:22. In some of any embodiments, the FKBP12 includes the amino acid sequence of SEQ ID NO: 22.
- the second expression cassette includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56. In some of any embodiments, the second expression cassette includes the nucleotide sequence of SEQ ID NOs: 53 or 56. In some of any embodiments, the second expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some of any embodiments, the second expression cassette encodes an amino acid sequence including the sequence of SEQ ID NOs: 54, 57, or 128.
- the synthetic cytokine beta chain polypeptide includes interleukin 2 receptor subunit P (IL2RB).
- the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:23 or 61.
- the nucleotide encoding the synthetic cytokine beta chain polypeptide includes the nucleotide sequence of SEQ ID Nos:23 or 61.
- the IL2RB includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID Nos:24 or 62. In some of any embodiments, the IL2RB includes the amino acid sequence of SEQ ID Nos: 24 or 62.
- the third expression cassette further includes a nucleotide sequence encoding FKBP12.
- the nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 21.
- the nucleotide sequence encoding the FKBP12 includes the nucleotide sequence of SEQ ID NO: 21.
- the FKBP12 includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:22.
- the FKBP12 includes the amino acid sequence of SEQ ID NO: 22.
- the nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 55. In some of any embodiments, the nucleotide sequence encoding the FKBP12 includes the nucleotide sequence of SEQ ID NO: 55.
- the third expression cassette is codon optimized.
- the third expression cassette further contains a nucleotide sequence encoding FRB.
- the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB includes the nucleotide sequence of SEQ ID NO: 13.
- the FRB includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 14.
- the FRB includes the amino acid sequence of SEQ ID NO: 14.
- the third expression cassette includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 19. In some of any embodiments, the third expression cassette includes the nucleotide sequence of SEQ ID NO: 19. In some of any embodiments, the third expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 20. In some of any embodiments, the third expression cassette encodes an amino acid sequence including the sequence of SEQ ID NO: 20.
- the third expression cassette includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 59. In some of any embodiments, the third expression cassette includes the nucleotide sequence of SEQ ID NO: 59. In some of any embodiments, the third expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some of any embodiments, the third expression cassette encodes an amino acid sequence including the sequence of SEQ ID NOs: 60 or 129.
- the CAR includes an extracellular antigen binding domain, a transmembrane domain and an endodomain containing a costimulatory signaling domain and a primary activation signaling domain such as a CD3zeta signaling domain.
- the extracellular antigen binding domain and transmembrane domain are separated by a spacer sequence, such as containing a hinge domain.
- the extracellular antigen binding domain comprises an scFv.
- the CAR includes an scFv domain.
- the scFv domain includes anti-fluorescein isothiocyanate (FITC) E2.
- the scFv domain includes a light chain variable domain (VL), a linker, and a heavy chain variable domain (VH).
- VL light chain variable domain
- VH heavy chain variable domain
- the scFv VL includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:30 or 65.
- the scFv VL includes the nucleotide sequence of SEQ ID Nos:30 or 65.
- the scFv VL includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:31. In some of any embodiments, the scFv VL includes the amino acid sequence of SEQ ID NO:31. [0024] In some of any embodiments, the scFv VH includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:34 or 67.
- the scFv VH includes the nucleotide sequence of SEQ ID Nos:34 or 67. In some of any embodiments, the scFv VH includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:35. In some of any embodiments, the scFv VH includes the amino acid sequence of SEQ ID NO:35.
- the scFv linker includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:32 or 66. In some of any embodiments, the scFv linker includes the nucleotide sequence of SEQ ID Nos:32 or 66. In some of any embodiments, the scFv linker includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:33. In some of any embodiments, the scFv linker includes the amino acid sequence of SEQ ID NO:33.
- the scFv includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:28 or 64. In some of any embodiments, the scFv includes the nucleotide sequence of SEQ ID Nos:28 or 64. In some embodiments, the scFv includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:29. In some of any embodiments, the scFv includes the amino acid sequence of SEQ ID NO:29.
- the CAR includes a hinge domain.
- the hinge domain includes a short hinge or a medium hinge domain.
- the hinge domain includes a CD8 or an IgG.
- the CD 8 hinge includes CD 8 a hinge.
- the CD8a hinge includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:38 or 114. In some of any embodiments, the CD8a hinge includes the nucleotide sequence of SEQ ID NO: 38. In some of any embodiments, the CD8a hinge includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs:39 or 115. In some of any embodiments, the CD8a hinge includes the amino acid sequence of SEQ ID NOs: 39 or 115.
- the CAR contains a transmembrane domain.
- the transmembrane domain includes a CD8 or a CD28.
- the CD8 transmembrane domain includes CD8a transmembrane domain.
- the transmembrane domain includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO:40. In some of any embodiments, the transmembrane domain includes the nucleotide sequence of SEQ ID NO:40. In some of any embodiments, the transmembrane domain includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:41. In some of any embodiments, the transmembrane domain includes the amino acid sequence of SEQ ID NO:41.
- the CAR includes an endodomain.
- the endodomain includes a costimulatory molecule signaling domain.
- the endodomain includes a signaling domain of 4- IBB, CD3 and/or CD28.
- the 4- IBB endodomain includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:42 or 69.
- the 4-1BB endodomain includes the nucleotide sequence of SEQ ID Nos: 42 or 69.
- the 4-1BB endodomain includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:43.
- the 4-1BB endodomain includes the amino acid sequence of SEQ ID NO:43.
- the CD3 ⁇ endodomain includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos: 46, 70, 100 or 118. In some of any embodiments, the CD3 ⁇ endodomain includes the nucleotide sequence of SEQ ID Nos: 46, 70, 100 or 118. In some of any embodiments, the CD3 ⁇ endodomain includes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:47.
- the CD3 ⁇ endodomain includes the amino acid sequence of SEQ ID NO:47.
- the fourth expression cassette includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos: 26, 63, 71, or 82. In some of any embodiments, the fourth expression cassette includes the nucleotide sequence of SEQ ID Nos: 26, 63, 71, or 82.
- the fourth expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127. In some of any embodiments, the fourth expression cassette encodes the amino acid sequence of SEQ ID NOs: 27, 72 or 127.
- each of the cleavage site sequences comprises a 2A cleavable linker sequence.
- each nucleotide encoding a 2A cleavable linker sequences is different.
- the 2A cleavable linker is independently a T2A, P2A, E2A or F2A cleavage site.
- the 2A cleavable linker is independently a P2A or a T2A.
- At least one 2A cleavable linker is a P2A and the nucleotide sequence encoding the P2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NOs: 17, 25, 52, or 58.
- nucleotide sequence encoding the P2A cleavable linker is set forth in SEQ ID NOs: 17, 25, 52, or 58.
- the P2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 18.
- the P2A cleavable linker comprises the sequence set forth in SEQ ID NO: 18.
- At least one 2A cleavable linker is a T2A and the nucleotide sequence encoding the T2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:9.
- the nucleotide sequence encoding the T2A cleavable linker is set forth in SEQ ID NO:9.
- the T2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10.
- the T2A cleavable linker comprising the sequence set forth in SEQ ID NO: 10.
- At least one of the cleavage site sequences comprises a furin cleavage site sequence.
- the furin cleavage site sequence is located between the first expression cassette and the second expression cassette.
- the nucleotide sequence encoding the furin cleavage site sequence comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:7.
- the nucleotide sequence encoding the furin cleavage site sequence comprises the sequence set forth in SEQ ID NO: 7.
- the furin cleavage site sequence comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 8.
- the furin cleavage site sequence comprises the amino acid sequence of SEQ ID NO: 8.
- the cleavage site sequence comprises a furin cleavage site sequence and a T2A cleavage sequence (furinT2A).
- the nucleotide sequence encoding the cleavage site sequence is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 5.
- the nucleotide sequence encoding the cleavage site sequence comprises the nucleotide sequence of SEQ ID NO: 5.
- the cleavage site sequence comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 6.
- the cleavage site sequence comprises the amino acid sequence of SEQ ID NO:6.
- the first expression cassette and second expression cassette are separated by a furinT2A
- the second expression cassette and third expression cassette are separated by a P2A
- the third expression cassette and fourth expression cassette are separated by a P2A.
- the construct includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO:1. In some of any embodiments, the construct includes the nucleotide sequence of SEQ ID NO:1.
- the construct encodes a polypeptide including an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:2. In some of any embodiments, the construct encodes a polypeptide including the amino acid sequence of SEQ ID NO: 2.
- the construct includes a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO:48. In some of any embodiments, the construct includes the nucleotide sequence of SEQ ID NO:48.
- the construct encodes a polypeptide including an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:49. In some of any embodiments, the construct encodes a polypeptide including the amino acid sequence of SEQ ID NO:49.
- a viral vector containing any one of the polycistronic constructs disclosed herein.
- the viral vector is a lentiviral vector.
- the viral vector further contains one or more surface T cell activating agents.
- the one or more surface T cell activating agents include CD58, anti-CD3, or CD80.
- a cell containing any one of the viral vectors disclosed herein includes a stem cell or a progenitor cell.
- the stem cell includes an induced pluripotent stem cell (iPSC).
- the progenitor cell includes a peripheral blood mononuclear cell (PBMC). In some of any embodiments, the cell includes a T cell. In some of any embodiments, the cell includes a cytotoxic innate lymphocyte (CIL) cell. In some of any embodiments, the cell includes a natural killer (NK) cell.
- PBMC peripheral blood mononuclear cell
- CIL cytotoxic innate lymphocyte
- NK natural killer
- a method of transducing a cell including contacting a target cell with any of the polycistronic constructs disclosed herein. In some aspects, provided herein is a method of transducing a cell comprising contacting a target cell with any one of the viral vectors disclosed herein.
- the target cell includes a stem cell. In some embodiments, the stem cell includes an induced pluripotent stem cell (iPSC).
- the target cell includes a progenitor cell.
- the progenitor cell includes a peripheral blood mononuclear cell (PBMC).
- PBMC peripheral blood mononuclear cell
- the target cell includes a T cell.
- the T cell includes a CD4+ or CD 8+ T cell.
- the method further includes contacting the target cell with a (i) a guide RNA (gRNA) targeting a target site in an endogenous gene, and (ii) an RNA-guided endonuclease, thereby inserting the nucleotide sequence into the endogenous gene.
- a guide RNA gRNA
- gRNA guide RNA
- provided herein is a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a target cell.
- a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a target cell comprising contacting the target cell with any one of the viral vectors disclosed herein.
- the target cell includes a stem cell.
- the stem cell includes an induced pluripotent stem cell (iPSC).
- the target cell includes a progenitor cell.
- the progenitor cell includes a peripheral blood mononuclear cell (PBMC).
- PBMC peripheral blood mononuclear cell
- the target cell includes a T cell.
- the T cell includes a CD4+ or CD8+ T cell.
- the method is performed ex vivo or in vitro.
- the method is performed in vivo.
- a method of transducing a T cell comprising contacting the T cell with a viral vector comprising one or more T cell activating agents and any one of the polycistronic constructs disclosed herein, wherein the one or more T cell activating agents bind a receptor on the T cell.
- a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a T cell comprising contacting the T cell with a viral vector comprising one or more T cell activating agents and any one of the polycistronic constructs disclosed herein, wherein the one or more T cell activating agents bind a receptor on the T cell.
- a method of delivering a payload to a T cell comprising contacting the T cell with a viral vector comprising one or more T cell activating agents and any one of the polycistronic constructs disclosed herein, wherein the one or more T cell activating agents bind a receptor on the T cell.
- the T cell comprises a a CD4+ or CD8+ T cell.
- the method is performed ex vivo or in vitro.
- the method is performed in vivo.
- the one or more T cell activating agents comprise CD58, anti-CD3, or CD80.
- the viral vector comprises a lentiviral vector.
- provided herein is a cell produced by any one of the methods disclosed herein.
- a method of administering to a subject any one of the cells disclosed herein is a method of administering to a subject any one of the cells disclosed herein.
- the chimeric antigen receptor is able to be targeted to an antigen associated with a disease or condition in the subject and the subject has a disease or condition that is able to be treated by the chimeric antigen receptor.
- the disease or condition is a cancer.
- the method treats a disease or condition in the subject.
- the disease or condition is treatable by the chimeric antigen receptor (CAR) encoded by the polycistronic construct.
- the CAR is an anti-FITC CAR and the CAR is targeted to a cell of the disease or condition by administering a bifunctional ligand comprising FITC and a ligand that specifically binds a molecule expressed on the cell of the disease or condition.
- the disease or condition is a cancer.
- the cancer is a solid tumor.
- the cell is a cancer cell.
- the chimeric antigen receptor is able to be targeted to an antigen associated with a disease or condition in the subject and the subject has a disease or condition that is able to be treated by the chimeric antigen receptor.
- the disease or condition is a cancer.
- the CAR is a CAR that targets a ligand that is able to bind to a an antigen on the surface of a cell associated with a disease or condition.
- the CAR of any of the provided embodiments is an anti-FITC CAR directed against FITC and the ligand is a bifunctional ligand composed of FITC and a binding molecule that is able to bind to a surface molecule or receptor on the target cell.
- the method further including administering the bifunctional ligand to tag a cancer cell in the subject, wherein the bifunctional ligand specifically binds a molecule expressed on a tumor.
- the bifunctional ligand is FITC-folate.
- the cancer is an osteosarcoma.
- the bifunctional ligand comprises a fluorescein isothiocyanate (FITC) moiety and the chimeric antigen receptor (CAR) encoded by the polycistronic construct is an anti-FITC CAR.
- FITC fluorescein isothiocyanate
- CAR chimeric antigen receptor
- the method further including administering a non- physiological ligand to the subject.
- the non-physiological ligand is able to bind to the synthetic cytokine receptor and induce gamma cytokine signaling in the cell.
- the nonphysiological ligand binds to a synthetic cytokine receptor composed of the synthetic gamma chain polypeptide and the synthetic cytokine beta chain polypeptide encoded by the polycistronic construct.
- the non-physiological ligand includes rapamycin or a rapamycin analog.
- binding of the non-physiological ligand to the synthetic cytokine receptor stimulates an intracellular cytokine signal in cells transduced to express the synthetic cytokine receptor. In some of any embodiments, binding of the non-physiological ligand to the synthetic cytokine receptor promotes proliferation of cells transduced to express the synthetic cytokine receptor.
- FIG. 1A shows lentivirus particles surface-engineered to engage and activate T cells to deliver a payload containing free FKBP12-rapamycin binding (FRB), rapamycin activated cytokine receptor (RACR), and a chimeric antigen receptor (CAR) that binds a tumor tag (TagCAR).
- FRB free FKBP12-rapamycin binding
- RACR rapamycin activated cytokine receptor
- CAR chimeric antigen receptor
- Rapamycin, FRB and RACR drive expansion of transduced TagCAR T cells while also inhibiting tumor proliferation and immune responses against the lentivirus particles.
- FIG. IB shows how the TagCAR T system targets tumor cells.
- Bispecific tumor tags comprise a universal tag antigen on one end and a swappable ligand on the other end that engages a tumor- or tumor microenvironment-associated antigen.
- T cells express TagCAR, the TagCAR can bind the universal tag (e.g., FITC-Folate).
- FIGS. 2A-2B show 8 polycistronic constructs.
- the polynucleotide of the TagCAR encodes a CAR with the following components in N-terminal to C-terminal order: an scFv (e.g. anti-FITC E2), a hinge (spacer), a transmembrane domain, and an endodomain with a costimulatory signaling domain and a CD3zeta signaling domain (Z).
- an scFv e.g. anti-FITC E2
- a hinge spacer
- Z CD3zeta signaling domain
- constructs differed in the hinge (spacer) domain, either IgG4 hinge (IgG4H) or CD8alpha hinge (CD8H); transmembrane domain, either CD28 TM or CD8 TM; and costimulatory domain, either 4 IBB costimulatory domain or CD28 costimulatory domain.
- the constructs also differed in the placement of the TagCAR, being present either as the front or end of the construct transgene sequence.
- the individual polynucleotide components of the construct were separated by a 2A cleavage site sequence.
- FIGS. 3A-3B show day 3 activation and day 7 transduction of CD8+ and CD4+T cell populations cultured with the polynucleotide constructs in FIGS. 2A-2B at a multiplicity of infection (MOI) of 2 or 10.
- MOI multiplicity of infection
- FIG. 3C shows FRB expression in T cells when the FRB is placed first or second in the transgenes as depicted in FIGS. 2A-2B.
- FIG. 4 shows the percentage of TagCAR+ T cells generated when transduced with Construct D.2 or Construct C.2, and cultured in IL-2 alone, rapamycin alone, or both.
- FIGS. 5A-5B show a tumor cell killing assay with PBMCs transduced with the constructs disclosed in FIGS. 2A-2B and treated with IL-2 only or IL-2 and rapamycin (rapa).
- FIG. 6A shows CD 19 chimeric antigen receptor (CAR) and TagCAR expression in PBMCs transduced with a lentivirus encoding Construct V or Construct C.2 polynucleotide.
- FIG. 6B shows the number of CD19-CAR+ (left panel) or FITC-Folate+ (right panel) PBMCs across 11 days. PBMCs were either transduced with lentivirus encoding Construct V or Construct C.2 polynucleotide.
- FIG. 7A shows the orientation of polynucleotide constructs from N- to C-terminal. In Construct C.2, the FRB is encoded next to IL2Rp. In the Construct C.2U, the FRB is encoded next to IL2Ry.
- FIG. 7B shows the percentage of CD25 T cells 3 days after transfection with lentivirus encoding Construct V or Construct C.2 polynucleotide, in 5 donors.
- FIG. 7C shows the percentage of TagCAR T cells on day 7.
- PBMCs were transduced with a lentivirus encoding Construct C.2 or Construct C.2U polynucleotide and stained on day 7.
- FIG. 7D shows TagCAR MFI in T cells on day 7.
- PBMCs were transduced with a lentivirus encoding Construct C.2 or Construct C.2U polynucleotide.
- FIG. 7E shows an immunoblot depicting expression of FRBJL2RP and FRB:IL2Ry in T cells.
- PBMCs were transduced with a lentivirus encoding Construct C.2 or Construct C.2U polynucleotide.
- a rabbit pAb against FRB was used to detect FRB.
- Cells were collected on day 8 for western blot.
- FIG. 7F shows an immunoblot depicting expression of FKBP12:IL2RP and FKBP12:IL2Ry in T cells.
- PBMCs transduced with a lentivirus encoding Construct C.2 or Construct C.2U polynucleotide.
- a mouse mAb against FKBP12 was used to detect FKBP12. Cells were collected on day 8 for western blot.
- FIG. 8A show total Tag-CAR+ T cells transduced with Construct C.2U or Construct C.2 across 14 days in 1 donor.
- Tag-CAR+ T cells were treated with either IL-2 (250 U/mL) or rapamycin (lOnM).
- FIG. 8B show total Tag-CAR+ T cells transduced with Construct C.2U or Construct C.2 across 12 days in 1 donor.
- Tag-CAR+ T cells were treated with either IL-2 (250 U/mL), rapamycin (lOnM), rapamycin and IL-2, or AP21967 (50nM).
- FIG. 9A shows day 3 activation and day 7 transduction of CD8+ and CD4+T cell populations cultured with Construct C.2 at a multiplicity of infection (MOI) of 2 or 10.
- FIG. 9B shows representative flow cytometry plots of TagCAR expressing CD8+ T cells transduced with Construct C.2 on day 7 post-transduction.
- FIG. 10A shows a timeline of an in vitro assay method for measuring T cell activation and TagCAR T cell abundance.
- PBMCs are transduced with the lentivirus particles.
- IL-2 or IL-2 and rapamycin are added to measure T cell activation.
- TagCAR T cell abundance was measured.
- FIG. 10B shows enrichment (left) and expansion (right) of TagCAR T cells across 2 weeks. Enrichment was measured by flow cytometry. Expansion was measured by flow cytometry with counting beads.
- FIG. 11A shows a graph depicting tumor cell growth after incubating breast carcinoma cells (MDA-MB-231 or MDA) with rapamycin, TagCAR T cells and FITC-Folate, or TagCAR T cells, FITC-Folate and rapamycin.
- Breast carcinoma cells that have not been incubated with TagCAR T cells, FITC-folate or rapamycin are shown by arrow.
- Tumor cells were reintroduced (i.e., tumor cell re-challenge) at 72 hours, 144 hours, and 216 hours.
- FIG. 11B shows a graph depicting T cell proliferation after incubating breast carcinoma cells (MDA-MB-231 or MDA) with rapamycin, TagCAR T cells and FITC-Folate, or TagCAR T cells, FITC-Folate and rapamycin (rapa).
- Breast carcinoma cells that have not been incubated TagCAR T cells, FITC-folate or rapamycin are shown by arrow.
- Tumor cells were reintroduced (i.e., tumor cell re-challenge) at 72 hours, 144 hours, and 216 hours.
- FIG. 12A shows a timeline of an in vivo mouse model of breast cancer.
- NSG MHCI/II DKO mice were injected with FRa+ MDA-MB-231 cells.
- DO mice were infused with ex vivo generated TagCAR T cells.
- Mice were injected with FITC-Folate subcutaneously two times per week for four weeks. Blood was collected weekly for flow cytometry.
- FIG. 12B shows a graph depicting tumor volume in mice across 4 weeks.
- Mice were injected with: (i) 10e6 T cells that have not been transduced with lentivirus particles (Mock T cells) with FITC-Folate; (ii) 10e6 TagCAR T cells without FITC-Folate; (iii) 5e6 TagCAR T cells with FITC-Folate; and (iv) 10e6 TagCAR T cells with FITC-Folate. Tumor volume was measured using calipers.
- FIG. 13A shows a timeline of an in vivo mouse model of breast cancer.
- NSG MHCI/II DKO mice were injected with FRa+ MDA-MB-231 cells.
- DO mice were humanized with PBMCs and administered lentivirus particles.
- Mice were injected with FITC-Folate subcutaneously two times per week for 7 weeks. Blood was collected weekly for flow cytometry. Tumor volume was measured using calipers.
- FIG. 13B shows circulating TagCAR T cell detection by flow cytometry.
- the left panel depicts the percentage of TagCAR+ per CD3+ T cells on day 7.
- the right panel depicts the total number of CD3+/TagCAR+ T cells per pL of blood on day 7.
- Mice were injected with: (i) no vector and FITC-Folate; (ii) 100e6 transfecting units (TU) of TagCAR vector and FITC-Folate; (iii) 25e6 TU TagCAR vector and FITC-Folate; and (iv) 100e6 TU TagCAR vector and FITC-Folate.
- FIG. 13C shows a graph depicting tumor volume in mice across 7 weeks. Tumor volume was measured using calipers.
- FIG. 14A shows a graph depicting tumor volume in mice across 25 days. Mice were injected with: (i) FITC-Folate alone; (ii) 5.0e6 transfecting units (TU) of TagCAR vector without FITC-Folate; (iii) 0.2e6 TU of TagCAR vector with FITC-Folate; (iv) 1.0e6 TU TagCAR vector with FITC-Folate; and (v) 5.0e6 TU of TagCAR vector with FITC-Folate. Tumor volume was measured using calipers.
- FIG. 14B shows circulating CD3 TagCAR T cells per p L of blood as detected by flow cytometry.
- Mice were injected with: (i) FITC-Folate alone (triangle); (ii) 5.0e6 TU TagCAR vector alone; (iii) 0.2e6 TU TagCAR vector with FITC-Folate; (iv) 1.0e6 TU TagCAR vector with FITC- Folate; and (v) 5.0e6 TU TagCAR vector with FITC-Folate.
- the disclosure relates generally to a polynucleotide construct comprising a contiguous polynucleotide sequence encoding at least two synthetic receptors and methods for uses thereof.
- the polynucleotide construct is a polycistronic construct encoding a synthetic cytokine receptor, a synthetic chimeric antigen receptor (CAR), and a freely diffusible FRB, in which the cytokine receptor is responsive to rapamycin binding.
- FRB reduces the inhibitory effects of rapamycin on mTOR in cells engineered to express the polycistronic constructs provided herein. Expression of the freely diffusible FRB can promote consistent activation and proliferation of engineered cells.
- the disclosure also provides that the 5’ to 3’ order of the polycistronic construct is important for the expression of polypeptides encoded by the construct.
- the 5’ to 3’ order of the polynucleotide constructs improve expression of encoded polypeptides.
- a polycistronic construct provided herein comprises a nucleotide encoding FRB at the 5’ end, which improves FRB expression and correlates with increased protection against rapamycin-mediated immunosuppression.
- the CAR expression - even when positioned at the 3’ end of the construct - was sufficiently high to mediate antigen-directed killing by cells in which the polynucleotide construct was expressed.
- a polycistronic construct comprising four expression cassettes separate by cleavage site sequences.
- the four expression cassettes comprise, in 5’ to 3’ order, a first expression a first expression cassette comprising a nucleotide sequence encoding FRB, a second expression cassette comprising a nucleotide sequence encoding a synthetic cytokine gamma chain polypeptide, a third expression cassette comprising a nucleotide sequence encoding a synthetic cytokine beta chain polypeptide, and a fourth expression cassette comprising a nucleotide sequence encoding a CAR.
- a viral vector comprising any one of the polycistronic constructs disclosed herein.
- a cell comprising any of the viral vectors disclosed herein.
- provided herein is a method of transducing a cell comprising contacting a target cell with any one of the viral vectors disclosed herein.
- a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a target cell comprising contacting the target cell with any one of the viral vectors disclosed herein.
- provided herein is a cell produced by any of the methods disclosed herein.
- provided herein is a method of administering to a subject any of the cells disclosed herein. In some aspects, provided herein is a method of administering to a subject any of the viral vectors disclosed herein.
- polycistronic constructs encoding one or more separate proteins.
- the polycistronic constructs comprise one, two, three, or four expression cassettes each encoding a separate protein.
- the polycistronic constructs comprise four expression cassettes each encoding a separate protein.
- the expression cassettes are separated by cleavable linkers.
- the polycistronic constructs provided herein comprise a nucleotide sequence encoding an FRB. In some embodiments, the polycistronic constructs provided herein comprise a nucleotide sequence encoding a chimeric antigen receptor (CAR). In some embodiments, the polycistronic constructs provided herein comprise a nucleotide sequence encoding a synthetic cytokine polypeptide. In some embodiments, the synthetic cytokine polypeptide comprises a synthetic cytokine gamma chain polypeptide and a synthetic cytokine beta chain polypeptide. In some embodiments, the synthetic cytokine gamma chain comprises interleukin 2 receptor subunit y (IL2RG).
- IL2RG interleukin 2 receptor subunit y
- the synthetic cytokine gamma chain further comprises FRB.
- the synthetic cytokine beta chain comprises interleukin 2 receptor subunit P (IL2RB).
- the synthetic cytokine gamma chain comprises further FKBP12.
- the synthetic cytokine gamma chain comprises interleukin 2 receptor subunit y (IL2RG).
- the synthetic cytokine gamma chain further comprises FKBP12.
- the synthetic cytokine beta chain comprises interleukin 2 receptor subunit P (IL2RB).
- the synthetic cytokine beta chain further comprises FRB.
- the polycistronic construct provided herein comprises nucleotide sequences encoding an FRB, a synthetic cytokine polypeptide, and a CAR.
- the polycistronic construct comprises in 5’ to 3’ order a nucleotide sequence encoding FRB, a nucleotide sequence encoding a synthetic cytokine polypeptide, and a nucleotide sequence encoding a CAR.
- the nucleotide sequence encoding the synthetic cytokine polypeptide comprises in 5’ to 3’ order a first nucleotide sequence encoding FRB:IL2RG and a second nucleotide sequence encoding FKBP12:IL2RB. In some embodiments, the nucleotide sequence encoding the synthetic cytokine polypeptide comprises in 5’ to 3’ order a first nucleotide sequence encoding FKBP12:IL2RG and a second nucleotide sequence encoding FRB:IL2RB.
- a polycistronic construct comprising in 5’ to 3’ order (a) a first expression cassette comprising a nucleotide sequence encoding FRB, (b) a second expression cassette comprising a nucleotide sequence encoding a synthetic cytokine gamma chain polypeptide, (c) a third expression cassette comprising a nucleotide sequence encoding a synthetic cytokine beta chain polypeptide, and (d) a fourth expression cassette comprising a nucleotide sequence encoding a chimeric antigen receptor (CAR), wherein each of the expression cassettes are separated by a nucleotide sequence encoding a cleavage site sequence.
- CAR chimeric antigen receptor
- an expression cassette of the polycistronic construct encodes an FRB domain.
- the FRB domain is an approximately 270 base pair (bp) domain derived from the mTOR protein kinase. It may be expressed in the cytosol as a freely diffusible soluble protein.
- the first expression cassette in the polycistronic construct comprises a nucleotide sequence encoding an FRB.
- FRB when expressed, it is a freely diffusible soluble protein.
- the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50. In some embodiments, the nucleotide sequence encoding the FRB is at least 80% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50. In some embodiments, the nucleotide sequence encoding the FRB is at least 85% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50.
- the nucleotide sequence encoding the FRB is at least 90% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50. In some embodiments, the nucleotide sequence encoding the FRB is at least 95% identical to the nucleotide sequence of SEQ ID N0s:3, 13, or 50. In some embodiments, the nucleotide sequence encoding the FRB is at least 96% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50. In some embodiments, the nucleotide sequence encoding the FRB is at least 97% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50.
- the nucleotide sequence encoding the FRB is at least 98% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50. In some embodiments, the nucleotide sequence encoding the FRB is at least 99% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50. In some embodiments, the nucleotide sequence encoding the FRB is at least 100% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50. In some embodiments, the nucleotide sequence encoding the FRB comprises the nucleotide sequence of SEQ ID Nos: 3, 13, or
- the nucleotide sequence encoding the FRB consists the nucleotide sequence of SEQ ID Nos: 3, 13, or 50.
- the FRB comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some embodiments, the FRB comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some embodiments, the FRB comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NOs:4, 14, or
- the FRB comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some embodiments, the FRB comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some embodiments, the FRB comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some embodiments, the FRB comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some embodiments, the FRB comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51.
- the FRB comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some embodiments, the FRB comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NOs:4, 14, or 51. In some embodiments, the FRB comprises the amino acid sequence of SEQ ID NOs: 4, 14, or 51. In some embodiments, the FRB consists of the amino acid sequence of SEQ ID NOs: 4, 14, or 51. [0134] In some embodiments, synthetic cytokine receptor complex comprises a cytosolic polypeptide that binds to the ligand or a complex comprising the ligand.
- the cytosolic FRB confers resistance to the immunosuppressive effect of the non-physiological ligand (e.g., rapamycin or rapalog).
- the non-physiological ligand e.g., rapamycin or rapalog.
- an expression cassette of the polycistronic construct encodes a synthetic cytokine receptor.
- the synthetic cytokine receptors of the present disclosure comprise a synthetic gamma chain and a synthetic beta chain, each comprising a dimerization domain.
- the dimerization domains controllable dimerize in the present of a non-physiological ligand, thereby activating signaling the synthetic cytokine receptor.
- the synthetic cytokine receptor can include transmembrane receptor proteins that include the synthetic gamma chain polypeptide and the synthetic beta chain polypeptide, such as provided as a first transmembrane receptor and a second transmembrane receptor.
- the synthetic gamma chain polypeptide comprises a first dimerization domain, a first transmembrane domain, and an interleukin-2 receptor subunit gamma (IL-2RG) intracellular domain.
- the dimerization domain may be extracellular (N-terminal to the transmembrane domain) or intracellular (C-terminal to the transmembrane domain and N- or C-terminal to the IL-2G intracellular domain).
- the synthetic beta chain polypeptide comprises a second dimerization domain, a second transmembrane domain, and an intracellular domain selected from an interleukin-2 receptor subunit beta (IL-2RB) intracellular domain, an interleukin-7 receptor subunit beta (IL-7RB) intracellular domain, or an interleukin-21 receptor subunit beta (IL-21RB) intracellular domain.
- the dimerization domain may be extracellular (N-terminal to the transmembrane domain) or intracellular (C-terminal to the transmembrane domain and N- or C-terminal to the IL-2RB or IL-7RB intracellular domain).
- the polycistronic construct provided herein comprises one or more nucleotide sequences encoding a synthetic cytokine receptor.
- the one or more nucleotide sequences correspond to one or more expression cassettes.
- the polynucleotide construct provided herein comprises one expression cassette encoding IL2RG chain of the synthetic cytokine receptor and a second expression cassette encoding IL2RB chain of the synthetic cytokine receptor.
- the synthetic gamma chain polypeptide is encoded by a nucleic acid sequence that encodes a signal peptide.
- the synthetic beta chain polypeptide is encoded by a nucleic acid sequence that encodes a signal peptide.
- a signal peptide A skilled artisan is readily familiar with signal peptides that can provide a signal to transport a nascent protein in the cells. Any of a variety of signal peptides can be employed.
- the intracellular signaling domain of the first transmembrane receptor protein comprises an interleukin-2 receptor subunit gamma (IL2Rg) domain.
- IL2Rg interleukin-2 receptor subunit gamma
- the synthetic cytokine receptor comprises a first transmembrane receptor protein comprising an IL-2RG intracellular domain, a first dimerization domain, a second transmembrane receptor protein comprising an IL-2RB intracellular domain, and a second dimerization domain.
- the synthetic beta chain comprises an interleukin-2 receptor subunit beta (IL2RB) intracellular domain.
- IL2RB is also known as IL15RB or CD122.
- IL2RB can also mean IL15RB. That is, the terms are used interchangeably in the present disclosure.
- the synthetic cytokine receptor comprises a first transmembrane receptor protein comprising an IL-2RG intracellular domain, a first dimerization domain, a second transmembrane receptor protein comprising an IL-7RB intracellular domain, and a second dimerization domain.
- the synthetic beta chain comprises an interleukin-7 receptor subunit beta (IL7RB) intracellular domain.
- IL7RB interleukin-7 receptor subunit beta
- the synthetic cytokine receptor comprises a first transmembrane receptor protein comprising an IL-2RG intracellular domain, a first dimerization domain, a second transmembrane receptor protein comprising an IL-21RB intracellular domain, and a second dimerization domain.
- the synthetic beta chain comprises an interleukin-21 receptor subunit beta (IL21RB) intracellular domain.
- IL21RB interleukin-21 receptor subunit beta
- the dimerization domains may be heterodimerization domains, including but not limited to FK506-Binding Protein of size 12 kD (FKBP) and a FKBP12-rapamycin binding (FRB) domain, which are known in the art to dimerize in the presence of rapamycin or a rapalog.
- FKBP FK506-Binding Protein of size 12 kD
- FKBP12-rapamycin binding domain FKBP12-rapamycin binding domain
- the first dimerization domain and the second dimerization domain may be a FK506-Binding Protein of size 12 kD (FKBP) and a calcineurin domain, which are known in the art to dimerize in the presence of FK506 or an analogue thereof.
- FKBP FK506-Binding Protein of size 12 kD
- calcineurin domain which are known in the art to dimerize in the presence of FK506 or an analogue thereof.
- the dimerization domains are homodimerization domains selected from: i) FK506-Binding Protein of size 12 kD (FKBP); ii) ii) cyclophili A (CypA); or iii) iii) gyrase B (CyrB); with the corresponding non-physiological ligands being, respectively.
- FKBP FK506-Binding Protein of size 12 kD
- CypA cyclophili A
- CyrB gyrase B
- the dimerization domains are homodimerization domains selected from: i) FK506-Binding Protein of size 12 kD (FKBP); ii) ii) cyclophili A (CypA); or iii) iii) gyrase B (CyrB); with the corresponding non-physiological ligands being, respectively.
- the first and second dimerization domains of the transmembrane receptor proteins are a FKBP domain and a cyclophilin domain.
- the first and second dimerization domains of the transmembrane receptor proteins are a FKBP domain and a bacterial dihydrofolate reductase (DHFR) domain.
- DHFR bacterial dihydrofolate reductase
- the first and second dimerization domains of the transmembrane receptor proteins are a calcineurin domain and a cyclophilin domain.
- the first and second dimerization domains of the transmembrane receptor proteins are PYRl-like 1 (PYL1) and abscisic acid insensitive 1 (ABI1).
- the transmembrane domain is the sequence of the synthetic cytokine receptor that spans the membrane.
- the transmembrane domain may comprise a hydrophobic alpha helix.
- the transmembrane domain is derived from a human protein.
- the TM domain and the intracellular signaling domain are from the same cytokine receptor.
- the synthetic gamma chain polypeptide contains an IL-2RG TM domain and an IL-2RG intracellular domain.
- the synthetic beta chain polypeptide contains an IL-2RB TM domain and an IL-2RB intracellular domain.
- the synthetic beta chain polypeptide contains an IL-7RB TM domain and an IL- 7RB intracellular domain.
- the synthetic beta chain polypeptide contains an IL-21RB TM domain and an IL-21RB intracellular domain.
- one or more additional contiguous amino acids of the ectodomain directly adjacent to the TM domain of the cytokine receptor also can be included as part of the polypeptide sequence of a chain of the synthetic cytokine receptor.
- 1-20 contiguous amino acids of the ectodomain adjacent to the TM domain of the cytokine receptor is included as part of the polypeptide sequence of a chain of the synthetic cytokine receptor.
- the portion of the ectodomain may be a contiguous sequence of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 amino acids directly adjacent (e.g. N-terminal to) the TM sequence.
- the synthetic cytokine receptor is able to be bound by the non- physiological ligand rapamycin or a rapamycin analog. In some embodiments, the synthetic cytokine receptor is responsive to the non-physiological ligand rapamycin or a rapamycin analog, in which binding of the non-physiological ligand to the dimerization domains of the synthetic cytokine receptor induces cytokine receptor-mediated signaling in the cell, such as via the JAK/STAT pathway.
- the synthetic cytokine receptors of the present disclosure comprise a synthetic gamma chain and a synthetic beta chain, each comprising a dimerization domain.
- the dimerization domains controllable dimerize in the present of a non-physiological ligand, thereby activating signaling the synthetic cytokine receptor.
- the synthetic gamma chain polypeptide comprises a first dimerization domain, a first transmembrane domain, and an interleukin-2 receptor subunit gamma (IL-2RG) intracellular domain.
- the dimerization domain may be extracellular (N-terminal to the transmembrane domain) or intracellular (C-terminal to the transmembrane domain and N- or C-terminal to the IL-2G intracellular domain.
- the synthetic gamma chain polypeptide comprises an FRB:IL2RG fusion protein.
- the synthetic gamma chain polypeptide comprises an FKBP12:IL2RG fusion protein.
- the synthetic beta chain polypeptide comprises a second dimerization domain, a second transmembrane domain, and an intracellular domain selected from an interleukin-2 receptor subunit beta (IL-2RB) intracellular domain, an interleukin-7 receptor subunit beta (IL-7RB) intracellular domain, or an interleukin-21 receptor subunit beta (IL-21RB) intracellular domain.
- the dimerization domain may be extracellular (N-terminal to the transmembrane domain) or intracellular (C-terminal to the transmembrane domain and N- or C-terminal to the IL-2RB or IL- 7RB intracellular domain).
- the synthetic beta chain polypeptide comprises an FKBP12JL2RB fusion protein.
- the synthetic beta chain polypeptide comprises an FRBJL2RB fusion protein.
- the second expression cassette comprises a nucleotide sequence encoding a synthetic cytokine gamma chain polypeptide, wherein the synthetic cytokine gamma chain polypeptide is an FRBJL2RG fusion protein.
- the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 15.
- the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 80% identical to the nucleotide sequence of SEQ ID NO: 15.
- the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 85% identical to the nucleotide sequence of SEQ ID NO: 15. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 90% identical to the nucleotide sequence of SEQ ID NO: 15. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 95% identical to the nucleotide sequence of SEQ ID NO: 15.
- the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 96% identical to the nucleotide sequence of SEQ ID NO: 15. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 97% identical to the nucleotide sequence of SEQ ID NO: 15. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 98% identical to the nucleotide sequence of SEQ ID NO: 15.
- the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 99% identical to the nucleotide sequence of SEQ ID NO: 15. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 100% identical to the nucleotide sequence of SEQ ID NO: 15. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide comprises the nucleotide sequence of SEQ ID NO: 15. In some embodiments, the nucleotide encoding the synthetic cytokine gamma chain polypeptide consists of the nucleotide sequence of SEQ ID NO: 15.
- the synthetic cytokine gamma chain polypeptide comprises interleukin 2 receptor subunit y (IL2RG).
- the IL2RG comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 16.
- the IL2RG comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 16.
- the IL2RG comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 16.
- the IL2RG comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 16.
- the IL2RG comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 16. In some embodiments, the IL2RG comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 16. In some embodiments, the IL2RG comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 16. In some embodiments, the IL2RG comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 16. In some embodiments, the IL2RG comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 16. In some embodiments, the IL2RG comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 16. In some embodiments, the IL2RG comprises the amino acid sequence of SEQ ID NO: 16. In some embodiments, the IL2RG consists of the amino acid sequence of SEQ ID NO: 16.
- the second expression cassette further comprises a nucleotide sequence encoding FRB.
- the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB is at least 80% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB is at least 85% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB is at least 90% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 95% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 96% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 97% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB is at least 98% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 99% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 100% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB comprises the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB consists of the nucleotide sequence of SEQ ID NO: 13.
- the FRB comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 14.
- the FRB comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB consists of the amino acid sequence of SEQ ID NO: 14.
- the second expression cassette is codon optimized.
- the second expression cassette comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO:11. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 11. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 11. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO:11.
- the second expression cassette comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 11. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO:11. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 11. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 11. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO:11. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO:11.
- the second expression cassette comprises the nucleotide sequence of SEQ ID NO:11. In some embodiments, the second expression cassette consists of the nucleotide sequence of SEQ ID NO: 11.
- the second expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 12.
- the second expression cassette encodes an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence comprising the sequence of SEQ ID NO: 12. In some embodiments, the second expression cassette encodes an amino acid sequence consists of the sequence of SEQ ID NO: 12.
- the second expression cassette comprises a nucleotide encoding a synthetic cytokine gamma chain polypeptide, wherein the synthetic cytokine gamma chain polypeptide is an FKBP12:IL2RG fusion protein.
- the second expression cassette comprises a nucleotide sequence encoding FKBP12.
- the nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:21 or 55.
- the nucleotide sequence encoding the FKBP12 is at least 80% identical to the nucleotide sequence of SEQ ID NOs:21 or 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 85% identical to the nucleotide sequence of SEQ ID NOs:21 or 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 90% identical to the nucleotide sequence of SEQ ID NOs:21 or 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 95% identical to the nucleotide sequence of SEQ ID NOs:21 or 55.
- the nucleotide sequence encoding the FKBP12 is at least 96% identical to the nucleotide sequence of SEQ ID NOs:21 or 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 97% identical to the nucleotide sequence of SEQ ID NOs:21 or 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 98% identical to the nucleotide sequence of SEQ ID NOs:21 or 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 99% identical to the nucleotide sequence of SEQ ID N0s:21 or 55.
- the nucleotide sequence encoding the FKBP12 is at least 100% identical to the nucleotide sequence of SEQ ID NOs:21 or 55. In some embodiments, the nucleotide sequence encoding the FKBP12 comprises the nucleotide sequence of SEQ ID NOs:21 or 55. In some embodiments, the nucleotide sequence encoding the FKBP12 consists of the nucleotide sequence of SEQ ID NOs:21 or 55.
- the FKBP12 comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:22.
- the FKBP12 comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:22. In some embodiments, the FKBP12 comprises the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 consists of the amino acid sequence of SEQ ID NO: 22.
- the second expression cassette comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56.
- the second expression cassette comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56. In some embodiments, the second expression cassette comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56.
- the second expression cassette comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs: 53 or 56. In some embodiments, the second expression cassette comprises a nucleotide sequence comprising the nucleotide sequence of SEQ ID NOs: 53 or 56. In some embodiments, the second expression cassette consists of a nucleotide sequence comprising the nucleotide sequence of SEQ ID NOs: 53 or 56
- the second expression cassette encodes an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some embodiments, the second expression cassette encodes an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some embodiments, the second expression cassette encodes an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some embodiments, the second expression cassette encodes an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128.
- the second expression cassette encodes an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some embodiments, the second expression cassette encodes an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some embodiments, the second expression cassette encodes an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some embodiments, the second expression cassette encodes an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128.
- the second expression cassette encodes an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some embodiments, the second expression cassette encodes an amino acid sequence comprising the amino acid sequence of SEQ ID NOs: 54, 57, or 128. In some embodiments, the second expression cassette encodes an amino acid sequence consisting of the amino acid sequence of SEQ ID NOs: 54, 57, or 128.
- the third expression cassette comprises a nucleotide encoding a synthetic cytokine beta chain polypeptide, wherein the synthetic cytokine beta chain polypeptide is an FKBP12:IL2RB fusion protein.
- the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:23 or 61.
- the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 80% identical to the nucleotide sequence of SEQ ID NOs:23 or 61.
- the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 85% identical to the nucleotide sequence of SEQ ID NOs:23 or 61. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 90% identical to the nucleotide sequence of SEQ ID NOs:23 or 61. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 95% identical to the nucleotide sequence of SEQ ID NOs:23 or 61.
- the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 96% identical to the nucleotide sequence of SEQ ID NOs:23 or 61. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 97 % identical to the nucleotide sequence of SEQ ID NOs:23 or 61. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 98% identical to the nucleotide sequence of SEQ ID NOs:23 or 61.
- the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 99% identical to the nucleotide sequence of SEQ ID NOs:23 or 61. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 100% identical to the nucleotide sequence of SEQ ID NOs:23 or 61. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide comprises the nucleotide sequence of SEQ ID NOs: 23 or 61. In some embodiments, the nucleotide encoding the synthetic cytokine beta chain polypeptide consists of the nucleotide sequence of SEQ ID NOs:23 or 61.
- the synthetic cytokine beta chain polypeptide comprises interleukin 2 receptor subunit P (IL2RB).
- IL2RB interleukin 2 receptor subunit P
- the IL2RB comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NOs:24 or 62.
- the IL2RB comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NOs:24 or 62.
- the IL2RB comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB comprises the amino acid sequence of SEQ ID NOs:24 or 62. In some embodiments, the IL2RB consists of the amino acid sequence of SEQ ID NOs:24 or 62.
- the third expression cassette further comprises a nucleotide sequence encoding FKBP12.
- the nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 80% identical to the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 85% identical to the nucleotide sequence of SEQ ID NO: 21.
- the nucleotide sequence encoding the FKBP12 is at least 90% identical to the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 95% identical to the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 96% identical to the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 97% identical to the nucleotide sequence of SEQ ID NO: 21.
- the nucleotide sequence encoding the FKBP12 is at least 98% identical to the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 99% identical to the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 100% identical to the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 comprises the nucleotide sequence of SEQ ID NO: 21. In some embodiments, the nucleotide sequence encoding the FKBP12 consists of the nucleotide sequence of SEQ ID NO: 21.
- the nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 80% identical to the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 85% identical to the nucleotide sequence of SEQ ID NO: 55.
- the nucleotide sequence encoding the FKBP12 is at least 90% identical to the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 95% identical to the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 96% identical to the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 97% identical to the nucleotide sequence of SEQ ID NO: 55.
- the nucleotide sequence encoding the FKBP12 is at least 98% identical to the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 99% identical to the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 is at least 100% identical to the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 comprises the nucleotide sequence of SEQ ID NO: 55. In some embodiments, the nucleotide sequence encoding the FKBP12 consists of the nucleotide sequence of SEQ ID NO: 55.
- the FKBP12 comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 22.
- the FKBP12 comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO :22. In some embodiments, the FKBP12 comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 comprises the amino acid sequence of SEQ ID NO: 22. In some embodiments, the FKBP12 consists of the amino acid sequence of SEQ ID NO: 22.
- the third expression cassette is codon optimized.
- the third expression cassette comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO: 19.
- the third expression cassette comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO: 19.
- the third expression cassette comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette comprises the nucleotide sequence of SEQ ID NO: 19. In some embodiments, the third expression cassette consists of the nucleotide sequence of SEQ ID NO: 19.
- the third expression cassette comprises a nucleotide encoding a synthetic cytokine beta chain polypeptide, wherein the synthetic cytokine beta chain polypeptide is an FRB:IL2RB fusion protein.
- the third expression cassette comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 59.
- the third expression cassette comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 59.
- the third expression cassette comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO: 59.
- the third expression cassette comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 59. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO: 59. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 59. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 59.
- the third expression cassette comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO: 59. In some embodiments, the third expression cassette comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 59. In some embodiments, the third expression cassette comprises the nucleotide sequence of SEQ ID NO: 59. In some embodiments, the third expression cassette consists of the nucleotide sequence of SEQ ID NO: 59.
- the third expression cassette encodes an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NOs: 60 or 129.
- the third expression cassette encodes an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence comprising the sequence of SEQ ID NOs: 60 or 129. In some embodiments, the third expression cassette encodes an amino acid sequence consisting of the sequence of SEQ ID NOs: 60 or 129.
- the third expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:60. In some embodiments, the third expression cassette encodes an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 60. In some embodiments, the third expression cassette encodes an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:60. In some embodiments, the third expression cassette encodes an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:60. In some embodiments, the third expression cassette encodes an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:60.
- the third expression cassette encodes an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:60. In some embodiments, the third expression cassette encodes an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO:60. In some embodiments, the third expression cassette encodes an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:60. In some embodiments, the third expression cassette encodes an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:60. In some embodiments, the third expression cassette encodes an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:60. In some embodiments, the third expression cassette encodes an amino acid sequence comprising the sequence of SEQ ID NO:60.
- the third expression cassette encodes an amino acid sequence consisting the sequence of SEQ ID NO: 60.
- the third expression cassette further comprises a nucleotide sequence encoding FRB.
- the nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB is at least 80% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB is at least 85% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 90% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 95% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 96% identical to the nucleotide sequence of SEQ ID NO: 13.
- the nucleotide sequence encoding the FRB is at least 97% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 98% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 99% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB is at least 100% identical to the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB comprises the nucleotide sequence of SEQ ID NO: 13. In some embodiments, the nucleotide sequence encoding the FRB consists of the nucleotide sequence of SEQ ID NO: 13.
- the FRB comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 14.
- the FRB comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB comprises the amino acid sequence of SEQ ID NO: 14. In some embodiments, the FRB consists of the amino acid sequence of SEQ ID NO: 14.
- an expression cassette of the polycistronic construct encodes a chimeric antigen receptor.
- the CAR construct contains an extracellular binding portion, a transmembrane domain and an intracellular signaling domain.
- the intracellular signaling domain contains a costimulatory signaling domain and/or an activation signaling domain.
- the CAR construct contains an extracellular binding portion, a transmembrane domain and an intracellular signaling domain comprising a costimulatory signaling domain.
- the CAR construct contains an extracellular binding portion, a transmembrane domain and an intracellular signaling domain comprising an activation signaling domain.
- the CAR construct contains an extracellular binding portion, a transmembrane domain and an intracellular signaling domain comprising a costimulatory signaling domain and an activation signaling domain.
- the binding portion of the CAR can be, for example, a single chain fragment variable region (scFv) of an antibody, a Fab, Fv, Fc, or (Fab’)2 fragment, and the like.
- scFv single chain fragment variable region
- a costimulatory signaling domain serves to enhance the proliferation and survival of the lymphocytes upon binding of the CAR to a targeted moiety.
- the identity of the costimulatory signaling domain is limited only in that it has the ability to enhance cellular proliferation and survival activation upon binding of the targeted moiety by the CAR.
- Suitable costimulatory signaling domains include, but are not limited to: CD28 (see, e.g., Alvarez- Vallina, L. et al., Eur J Immunol. 1996. 26(10):2304-9); CD 137 (4- IBB), a member of the tumor necrosis factor (TNF) receptor family (see, e.g., Imai, C.
- sequence variants of these costimulatory signaling domains can be used, where the variants have the same or similar activity as the domain on which they are modeled. In various embodiments, such variants have at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, at least about 99%, or at least about 99.5% sequence identity to the amino acid sequence of the domain from which they are derived.
- the CAR constructs comprise two costimulatory signaling domains. While the particular combinations include all possible variations of the four noted domains, specific examples include: 1) CD28+CD137 (4-1BB) and 2) CD28+CD134 (0X40).
- the activation signaling domain serves to activate cells upon binding of the CAR to a targeted moiety.
- the identity of the activation signaling domain is limited only in that it has the ability to induce activation of the selected cell upon binding of the targeted moiety by the CAR.
- Suitable activation signaling domains include the CD3 ⁇ chain and Fc receptor y.
- the signaling domain is a signaling domain of NKG2C or NKp44.
- sequence variants of these noted activation signaling domains can be used without adversely impacting the invention, where the variants have the same or similar activity as the domain on which they are modeled. Such variants may have at least about 80%, at least about 90%, at least about 95%. at least about 97%, at least about 98%, at least about 99%, or at least about 99.5% sequence identity to the amino acid sequence of the domain from which they are derived.
- the CARs may include additional elements, such a signal peptide to ensure proper export of the fusion protein to the cells surface, a transmembrane domain to ensure the fusion protein is maintained as an integral membrane protein, and a hinge domain that imparts flexibility to the recognition region and allows strong binding to the targeted moiety.
- nucleotide sequence that encodes a CAR comprising an extracellular domain, optionally a hinge domain, a transmembrane domain, and an intracellular signaling domain.
- the intracellular signaling domain comprises a costimulatory domain and an activation signaling domain.
- the costimulatory and activation signaling domains are a single domain, for example a single intracellular domain that provides both costimulation and activation signals to a cell.
- the intracellular signaling domain comprises either a costimulatory domain or an activation signaling domain.
- the CAR comprises an extracellular domain, a CD8a hinge, a CD8a transmembrane domain, a 4- IBB costimulatory domain, and a CD3zeta signaling domain.
- a nucleotide sequence encodes an extracellular domain, an CD28 hinge domain, a CD28 transmembrane domain, a CD28 co-stimulatory domain, and a CD3zeta signaling domain.
- the nucleotide sequence encodes an extracellular domain, an IgG4 hinge domain, a CD28 transmembrane domain, a 4- IBB co-stimulatory domain, and a CD3zeta signaling domain.
- the nucleotide sequence encodes a CAR comprising an extracellular domain, a CD8a hinge, a CD28 transmembrane domain, a 4- IBB costimulatory domain, and a CD3zeta signaling domain.
- SCFV-CD16TM-2B4IC-CD3 ⁇ S see, e.g., Ei Y, Hermanson DL, Moriarity BS Kaufman DS, Cell Stem Cell. 2018; 23: 181-192
- scFv-NKp44iM-DAP10ic-CD3 ⁇ s see, e.g., Li Y, Hermanson DL, Moriarity BS Kaufman DS, Cell Stem Cell. 2018; 23: 181-192
- SCFV-NKG2DTM-CD3 ⁇ S see, e.g., Li Y, Hermanson DL, Moriarity BS Kaufman DS, Cell Stem Cell. 2018; 23: 181-192).
- the binding portion of the CAR can be directed to any antigen that is desired to be targeted, such as due to its overexpression on cells or association with a disease or conditions like cancer.
- the binding portion of the CAR is specific to a tumor antigen.
- Tumor antigens are well known in the art and include, for example, a glioma-associated antigen, carcinoembryonic antigen (CEA), EGFRvIII, IL-l lRa, IL-13Ra, EGFR, FAP, B7H3, Kit, CA LX, CS-1, MUC1, BCMA, bcr-abl, HER2, P-human chorionic gonadotropin, alphafetoprotein (AFP), ALK, CD19, CD123, cyclin Bl, lectin-reactive AFP, Fos-related antigen 1, ADRB3, thyroglobulin, EphA2, RAGE-1, RU1, RU2, SSX2, AKAP-4, LCK, OY-TES1, PAXS, SART3, CLL-1, fucosyl GM1,
- tumor antigens include the following: Differentiation antigens such as tyrosinase, TRP- 1, TRP-2 and tumor- specific multilineage antigens such as MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, pi 5; overexpressed embryonic antigens such as CEA; overexpressed oncogenes and mutated tumor-suppressor genes such as p53, Ras, HER-2/neu; unique tumor antigens resulting from chromosomal translocations; such as BCR-ABL, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR; and viral antigens, such as the Epstein Barr virus antigens EBVA and the human papillomavirus (HPV) antigens E6 and E7.
- Differentiation antigens such as tyrosinase, TRP- 1, TRP-2 and tumor- specific multilineage antigens such as MAGE-1, MAGE-3, BAGE, G
- the CAR comprises binding domains that target two or more antigens as disclosed herein, in any combination.
- the CAR comprises binding domains that target two or more antigens on the same target protein, for example two epitopes in BCMA.
- a skilled artisan is readily familiar with CARs against diverse tumor antigens. Any one of such CARs can be employed as the CAR.
- CARs have been incorporated into products approved by the FDA and include, but are not limited to, anti-CD19 and anti-BCMA CAR T cells such as tisagenlecleucel (Kymriah), axicabtagene ciloleucel (Yescarta), brexucabtagene autoleucel (Tecartus), lisocabtagene maraleucel (Breyanzi), idecabtagene vicleucel (Abecma), or ciltacabtagene autoleucel (Carvykti). It is within the level of a skilled artisan to generate similar constructs for specific targeting of a desired tumor antigen.
- the binding portion of the CAR can be directed to a universal antigen to target a wide variety of tumors without the need to prepare separate CAR constructs.
- the targeted moiety recognized by the CAR may also remain constant.
- a ligand may be administered to the subject to allow interaction with target cells and interaction with the binding portion of the CAR. It is only the ligand portion of the small conjugate molecule that needs to be altered to allow the system to target cancer cells of different identity. Exemplary CAR systems are described in the section below.
- the CAR is an anti-CD19 CAR and the extracellular binding domain of the CD 19 CAR is specific to CD 19, for example, human CD 19.
- the extracellular domain of the CD 19 CAR comprises an scFv derived from the FMC63 monoclonal antibody (FMC63), which comprises the heavy chain variable region (VH) and the light chain variable region (VE) of FMC63 connected by a linker.
- FMC63 and the derived scFv have been described in Nicholson et al., Mol. Immun. 34(16-17): 1157-1165 (1997) and PCT Application Publication No. WO2018/213337, the entire contents of each of which are incorporated by reference herein.
- An exemplary anti-CD19 CAR is shown in Table 1 with its different portions including the extracellular domain.
- the CAR is an anti-CD20 CAR and the extracellular binding domain of the CD20 CAR is specific to CD20, for example, human CD20.
- the extracellular binding domain of the CD20 CAR is derived from an antibody specific to CD20, including, for example, Eeul6, IF5, 1.5.3, rituximab, obinutuzumab, ibritumomab, ofatumumab, tositumumab, odronextamab, veltuzumab, ublituximab, and ocrelizumab.
- the extracellular binding domain of the CD20 CAR can comprise or consist of the VH, the VL, and/or one or more CDRs of any of the antibodies.
- An exemplary anti-CD20 CAR is shown in Table 2 and Table 3 with its different portions including its extracellular domain.
- CARs are generated by fusing a polynucleotide encoding a VL, VH, or scFv to the 5' end of a polynucleotide encoding transmembrane and intracellular domains, and transducing cells with that polynucleotide as well as with the corresponding VH or VL, if needed.
- VL/VH pairs and scFvs for innumerable haptens are known in the art or can be generated by conventional methods routinely. Accordingly, the present disclosure contemplates using any known hapten-binding domain.
- the CAR is an anti-FITC CAR and the ligand is composed of a fluorescein or fluorescein isothiocyanate (FITC) moiety conjugated to an agent that binds to a desired target cell (such as a cancer cell).
- FITC fluorescein or fluorescein isothiocyanate
- Exemplary ligands are described below.
- the ligand is FITC-folate.
- the CAR comprises an scFv domain.
- the scFv domain comprises anti-fluorescein isothiocyanate (FITC) E2.
- FITC fluorescein isothiocyanate
- the scFv domain comprises a light chain variable domain (VL), a linker, and a heavy chain variable domain (VH).
- the scFv VL comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs:30 or 65.
- the scFv VL comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs:30 or 65.
- the scFv VL comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL comprises the nucleotide sequence of SEQ ID NOs:30 or 65. In some embodiments, the scFv VL consists of the nucleotide sequence of SEQ ID NOs:30 or 65.
- the scFv VL comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:31.
- the scFv VL comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:31.
- the scFv VL comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL comprises the amino acid sequence of SEQ ID NO:31. In some embodiments, the scFv VL consists the amino acid sequence of SEQ ID NO:31.
- the scFv VH comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs:34 or 67.
- the scFv VH comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs:34 or 67.
- the scFv VH comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH comprises the nucleotide sequence of SEQ ID NOs:34 or 67. In some embodiments, the scFv VH consists of the nucleotide sequence of SEQ ID NOs:34 or 67.
- the scFv VH comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:35.
- the scFv VH comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:35.
- the scFv VH comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH comprises the amino acid sequence of SEQ ID NO:35. In some embodiments, the scFv VH consists the amino acid sequence of SEQ ID NO: 35.
- the scFv linker comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs:32 or 66.
- the scFv linker comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs:32 or 66.
- the scFv linker comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker comprises the nucleotide sequence of SEQ ID NOs:32 or 66. In some embodiments, the scFv linker consists the nucleotide sequence of SEQ ID NOs:32 or 66.
- the scFv linker comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:33.
- the scFv linker comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:33.
- the scFv linker comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker comprises the amino acid sequence of SEQ ID NO:33. In some embodiments, the scFv linker consists the amino acid sequence of SEQ ID NO: 33.
- the scFv comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs:28 or 64.
- the scFv comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs:28 or 64.
- the scFv comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv comprises the nucleotide sequence of SEQ ID NOs:28 or 64. In some embodiments, the scFv consists of the nucleotide sequence of SEQ ID NOs:28 or 64.
- the scFv comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:29.
- the scFv comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:29.
- the scFv comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv comprises the amino acid sequence of SEQ ID NO:29. In some embodiments, the scFv consists of the amino acid sequence of SEQ ID NO: 29.
- a fluorescein or fluorescein isothiocyanate (FITC) moiety may be conjugated to an agent that binds to a desired target cell (such as a cancer cell), and thereby a CAR expressing an anti-fluorescein/FITC chimeric antigen receptor may be selectively targeted to the target cell labeled by the conjugate.
- a fluorescein or fluorescein isothiocyanate (FITC) moiety may be conjugated to an agent that binds to a desired target cell (such as a cancer cell), and thereby a CAR expressing an anti-fluorescein/FITC chimeric antigen receptor may be selectively targeted to the target cell labeled by the conjugate.
- a desired target cell such as a cancer cell
- haptens recognized by CARs may be used in place of fluorescein/FITC.
- the CAR may be generated using various scFv sequences known in the art, or scFv sequences generated by conventional and routine methods. Further illustrative scFv sequences for fluorescein/FITC and for other haptens are provided in, for example, WO 2021/076788, the disclosure of which is incorporated by reference herein.
- the disclosure provides an illustration of this conjugate molecule/CAR system.
- the CAR system of the disclosure utilizes conjugate molecules as the bridge between CAR-expressing cells and targeted cancer cells.
- the conjugate molecules are conjugates comprising a hapten and a cell-targeting moiety, such as any suitable tumor cell-specific ligand.
- Illustrative haptens that can be recognized and bound by CARs include small molecular weight organic molecules such as DNP (2,4-dinitrophenol), TNP (2,4,6-trinitrophenol), biotin, and digoxigenin, along with fluorescein and derivatives thereof, including FITC (fluorescein isothiocyanate), NHS-fluorescein, and pentafluorophenyl ester (PFP) and tetrafluorophenyl ester (TFP) derivatives, a knottin, a centyrin, and a DARPin.
- Suitable cell-targeting moiety that may themselves act as a hapten for a CAR include knottins (see Kolmar H. et al., The FEES Journal. 2008. 275(11):26684-90), centyrins, and DARPins (see Reichert, J.M. MAbs 2009. 1(3): 190-209).
- a DUPA derivative can be the ligand of the small molecule ligand linked to a targeting moiety, and DUPA derivatives are described in WO 2015/057852, incorporated herein by reference.
- the cell-targeting moiety is CCK2R ligand, a ligand bound by CCK2R-positive cancer cells (e.g., cancers of the thyroid, lung, pancreas, ovary, brain, stomach, gastrointestinal stroma, and colon; see Wayua. C. et al., Molecular Pharmaceutics . 2013. ePublication).
- CCK2R ligand a ligand bound by CCK2R-positive cancer cells (e.g., cancers of the thyroid, lung, pancreas, ovary, brain, stomach, gastrointestinal stroma, and colon; see Wayua. C. et al., Molecular Pharmaceutics . 2013. ePublication).
- the cell-targeting moiety is folate, folic acid, or an analogue thereof, a ligand bound by the folate receptor on cells of cancers that include cancers of the ovary, cervix, endometrium, lung, kidney, brain, breast, colon, and head and neck cancers; see Sega, E.I. et al., Cancer Metastasis Rev. 2008. 27(4):655-64).
- the cell-targeting moiety is an NK-1R ligand.
- Receptors for NK- 1R the ligand are found, for example, on cancers of the colon and pancreas.
- the NK-1R ligand may be synthesized according to the method disclosed in Int’l Patent Appl. No. PCT/US2015/044229, incorporated herein by reference.
- the cell-targeting moiety may be a peptide ligand, for example, the ligand may be a peptide ligand that is the endogenous ligand for the NK1 receptor.
- the small conjugate molecule ligand may be a regulatory peptide that belongs to the family of tachykinins which target tachykinin receptors. Such regulatory peptides include Substance P (SP), neurokinin A (substance K), and neurokinin B (neuromedin K), (see Hennig et al., International Journal of Cancer: 61, 786-792).
- the cell-targeting moiety is a CAIX ligand. Receptors for the CAIX ligand found, for example, on renal, ovarian, vulvar, and breast cancers. The CAIX ligand may also be referred to herein as CA9.
- the cell-targeting moiety is a ligand of gamma glutamyl transpeptidase.
- the transpeptidase is overexpressed, for example, in ovarian cancer, colon cancer, liver cancer, astrocytic gliomas, melanomas, and leukemias.
- the cell-targeting moiety is a CCK2R ligand.
- Receptors for the CCK2R ligand found on cancers of the thyroid, lung, pancreas, ovary, brain, stomach, gastrointestinal stroma, and colon, among others.
- the cell-targeting moiety is a PSMA ligand.
- the cell-targeting moiety is a FAP ligand.
- the cell-targeting moiety may have a mass of less than about 10,000 Daltons, less than about 9000 Daltons, less than about 8,000 Daltons, less than about 7000 Daltons, less than about 6000 Daltons, less than about 5000 Daltons, less than about 4500 Daltons, less than about 4000 Daltons, less than about 3500 Daltons, less than about 3000 Daltons, less than about 2500 Daltons, less than about 2000 Daltons, less than about 1500 Daltons, less than about 1000 Daltons, or less than about 500 Daltons.
- the small molecule ligand may have a mass of about 1 to about 10,000 Daltons, about 1 to about 9000 Daltons, about 1 to about 8,000 Daltons, about 1 to about 7000 Daltons, about 1 to about 6000 Daltons, about 1 to about 5000
- Daltons about 1 to about 4500 Daltons, about 1 to about 4000 Daltons, about 1 to about 3500
- Daltons about 1 to about 3000 Daltons, about 1 to about 2500 Daltons, about 1 to about 2000
- Daltons about 1 to about 1500 Daltons, about 1 to about 1000 Daltons, or about 1 to about 500
- the linkage in a conjugate described herein can be a direct linkage (e.g., a reaction between the isothiocyanate group of FITC and a free amine group of a small molecule ligand) or the linkage can be through an intermediary linker.
- an intermediary linker can be any biocompatible linker known in the art, such as a divalent linker.
- the divalent linker can comprise about 1 to about 30 carbon atoms. In another illustrative embodiment, the divalent linker can comprise about 2 to about 20 carbon atoms.
- linkers lengths that are suitable include, but are not limited to, linkers having 2, 3, 4, 5, 6, 7, 8, 9. 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37. 38, 39 or 40, or more atoms.
- the hapten and the cell-targeting moiety can be directly conjugated through such means as reaction between the isothiocyanate group of FITC and free amine group of small ligands (e.g., folate, DUPA, and CCK2R ligand).
- small ligands e.g., folate, DUPA, and CCK2R ligand.
- suitable linking domains include: 1) polyethylene glycol (PEG); 2) polyproline; 3) hydrophilic amino acids; 4) sugars; 5) unnatural peptidoglycans; 6) polyvinylpyrrolidone; 7) Pluronic F-127.
- Linker lengths that are suitable include, but are not limited to, linkers having 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39 or 40, or more atoms.
- the linker may be a divalent linker that may include one or more spacers.
- An illustrative conjugate of the disclosure is FITC-Folate
- An illustrative conjugate of the disclosure is FITC-CA9
- Illustrative conjugates of the disclosure include the following molecules: FITC-(PEG)i2-
- FITC-(PEG) 2 o-Folate Folate, FITC-(PEG) 2 o-Folate, FITC-(PEG)io8-Folate, FITC-DUPA, FITC-(PEG)I 2 -DUPA, FITC- CCK2R ligand, FITC-(PEG)I 2 -CCK2R ligand, FITC-(PEG)n-NKlR ligand and FITC-(PEG) 2 - CA9.
- the affinity at which the ligands and cancer cell receptors bind can vary, and in some cases low affinity binding may be preferable (such as about 1 pM), the binding affinity of the ligands and cancer cell receptors will generally be at least about 100 pM, 1 nM, 10 nM, or 100 nM, preferably at least about 1 pM or 10 pM, even more preferably at least about 100 pM.
- the CAR comprises a hinge domain.
- the hinge domain comprises a short hinge or a medium hinge domain.
- the hinge domain comprises a CD8 or an IgG.
- the CD8 hinge comprises CD8a hinge.
- the IgG hinge comprises an IgG4 hinge.
- the IgG4 hinge is modified.
- the IgG hinge comprises an IgGl hinge.
- the hinge domain comprises a PD1 hinge.
- the hinge domain comprises a CD28 hinge.
- the CD8a hinge comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:38 or 114. In some embodiments, the CD8a hinge comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs:38 or 114. In some embodiments, the CD8a hinge comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs:38 or 114.
- the CD8a hinge comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs:38 or 114. In some embodiments, the CD8a hinge comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID Nos:38 or 114. In some embodiments, the CD8a hinge comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs:38 or 114. In some embodiments, the CD8a hinge comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs:38 or 114.
- the CD8a hinge comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs:38 or 114. In some embodiments, the CD8a hinge comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs:38 or 114. In some embodiments, the CD8a hinge comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs:38 or 114. In some embodiments, the CD8a hinge comprises the nucleotide sequence of SEQ ID NOs: 38 or 114. In some embodiments, the CD8a hinge consists of the nucleotide sequence of SEQ ID NOs: 38 or 114.
- the CD8a hinge comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID Nos:39 or 115. In some embodiments, the CD8a hinge comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NOs:39 or 115. In some embodiments, the CD8a hinge comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NOs:39 or 115. In some embodiments, the CD8a hinge comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID Nos:39 or 115.
- the CD8a hinge comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NOs:39 or 115. In some embodiments, the CD8a hinge comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NOs:39 or 115. In some embodiments, the CD8a hinge comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID Nos:39 or 115. In some embodiments, the CD8a hinge comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID Nos:39 or 115. In some embodiments, the CD8a hinge comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NOs:39 or 115.
- the CD8a hinge comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NOs:39 or 115. In some embodiments, the CD8a hinge comprises the amino acid sequence of SEQ ID Nos: 39 or 115. In some embodiments, the CD8a hinge consists of the amino acid sequence of SEQ ID NOs: 39 or 115.
- the CD8 hinge comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD8 hinge comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD 8 hinge comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD8 hinge comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO: 123.
- the CD 8 hinge comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD8 hinge comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD 8 hinge comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD8 hinge comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 123.
- the CD 8 hinge comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD8 hinge comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD 8 hinge comprises the nucleotide sequence of SEQ ID NO: 123. In some embodiments, the CD8 hinge consists of the nucleotide sequence of SEQ ID NO: 123.
- the modified IgG4 hinge comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 119. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:119. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 119. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:119.
- the modified IgG4 hinge comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 119. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 119. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO:119. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 119. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:119.
- the modified IgG4 hinge comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:119. In some embodiments, the modified IgG4 hinge comprises the amino acid sequence of SEQ ID NO: 119. In some embodiments, the modified IgG4 hinge consists of the amino acid sequence of SEQ ID NO: 119.
- the modified IgG4 hinge comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 120.
- the modified IgG4 hinge comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 120.
- the modified IgG4 hinge comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge comprises the amino acid sequence of SEQ ID NO: 120. In some embodiments, the modified IgG4 hinge consists of the amino acid sequence of SEQ ID NO: 120.
- the IgGl hinge comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 122.
- the IgGl hinge comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 122.
- the IgGl hinge comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge comprises the amino acid sequence of SEQ ID NO: 122. In some embodiments, the IgGl hinge consists of the amino acid sequence of SEQ ID NO: 122.
- the PD1 hinge comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 121.
- the PD1 hinge comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 121.
- the PD1 hinge comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge comprises the amino acid sequence of SEQ ID NO: 121. In some embodiments, the PD1 hinge consists of the amino acid sequence of SEQ ID NO: 121.
- the CD28 hinge comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 124.
- the CD28 hinge comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge comprises the amino acid sequence of SEQ ID NO: 124. In some embodiments, the CD28 hinge consists of the amino acid sequence of SEQ ID NO: 124.
- the CAR comprises a transmembrane domain.
- the transmembrane domain comprises a CD8 or a CD28.
- the transmembrane domain comprises a CD8 domain.
- the transmembrane domain comprises a CD28 domain.
- the CD8 transmembrane domain comprises CD8a transmembrane domain.
- the transmembrane domain comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO:40.
- the transmembrane domain comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO:40.
- the transmembrane domain comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain comprises the nucleotide sequence of SEQ ID NO:40. In some embodiments, the transmembrane domain consists of the nucleotide sequence of SEQ ID NO:40.
- the transmembrane domain comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:41.
- the transmembrane domain comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:41.
- the transmembrane domain comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain comprises the amino acid sequence of SEQ ID NO:41. In some embodiments, the transmembrane domain consists of the amino acid sequence of SEQ ID NO:41. d. Intracellular domain (i.e. endodomain)
- the CAR comprises an endodomain.
- the endodomain comprises a costimulatory molecule.
- the endodomain comprises 4-1BB, CD3 ⁇ , and/or CD28.
- the endodomain comprises 4-1BB.
- the endodomain comprises CD3Q
- the endodomain comprises CD28.
- the 4- IBB endodomain comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:42 or 69.
- the 4-1BB endodomain comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs:42 or 69.
- the 4-1BB endodomain comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs:42 or 69.
- the 4- IBB endodomain comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs:42 or 69. In some embodiments, the 4-1BB endodomain comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs:42 or 69. In some embodiments, the 4- IBB endodomain comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs:42 or 69. In some embodiments, the 4- IBB endodomain comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs:42 or 69.
- the 4- IBB endodomain comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs:42 or 69. In some embodiments, the 4-1BB endodomain comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs:42 or 69. In some embodiments, the 4-1BB endodomain comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs:42 or 69. In some embodiments, the 4- IBB endodomain comprises the nucleotide sequence of SEQ ID NOs: 42 or 69. In some embodiments, the 4-1BB endodomain consists the nucleotide sequence of SEQ ID NOs: 42 or 69.
- the 4- IBB endodomain comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:43.
- the 4-1BB endodomain comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:43.
- the 4- 1BB endodomain comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:43.
- the 4-1BB endodomain comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:43.
- the 4-1BB endodomain comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 43. In some embodiments, the 4- IBB endodomain comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:43. In some embodiments, the 4- IBB endodomain comprises an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO:43. In some embodiments, the 4-1BB endodomain comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:43. In some embodiments, the 4- IBB endodomain comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:43.
- the 4- 1BB endodomain comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:43.
- the 4- IBB endodomain comprises the amino acid sequence of SEQ ID NO:43.
- the 4-1BB endodomain consists of the amino acid sequence of SEQ ID NO: 43.
- the CD3 ⁇ endodomain comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118. In some embodiments, the CD3 ⁇ endodomain comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118. In some embodiments, the CD3 ⁇ endodomain comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118.
- the CD3 ⁇ endodomain comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118. In some embodiments, the CD3 ⁇ endodomain comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118. In some embodiments, the CD3 ⁇ endodomain comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118.
- the CD3 ⁇ endodomain comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118. In some embodiments, the CD3 ⁇ endodomain comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118. In some embodiments, the CD3 ⁇ endodomain comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118.
- the CD3 ⁇ endodomain comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118. In some embodiments, the CD3 ⁇ endodomain comprises the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118. In some embodiments, the CD3 ⁇ endodomain consists the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118.
- the CD3 ⁇ endodomain comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain comprises an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain comprises an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:47.
- the CD3 ⁇ endodomain comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain comprises an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain comprises an amino acid sequence at least 97 % identical to the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain comprises an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain comprises an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:47.
- the CD3 ⁇ endodomain comprises an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain comprises the amino acid sequence of SEQ ID NO:47. In some embodiments, the CD3 ⁇ endodomain consists of the amino acid sequence of SEQ ID NO:47.
- the CAR is an anti-CD19 CAR
- the polycistronic vector comprises a fourth expression cassette that contains a nucleotide sequence encoding a CD19 CAR.
- the CD19 CAR may comprise a signal peptide, an extracellular binding domain that specifically binds CD 19, a hinge domain, a transmembrane domain, an intracellular costimulatory domain, and an intracellular activation signaling domain.
- the fourth expression cassette encodes an anti-CD19 CAR with features set forth in Table 1.
- the CAR is an anti-CD20 CAR
- the polycistronic vector comprises a fourth expression cassette that contains a nucleotide sequence encoding a CD20 CAR.
- the CD20 CAR may comprise a signal peptide, an extracellular binding domain that specifically binds CD20, a hinge domain, a transmembrane domain, an intracellular costimulatory domain, and an intracellular activation signaling domain.
- the fourth expression cassette encodes an anti-CD20 CAR with features set forth in Table 2 (anti-CD20 CAR with flag) or Table 3 (anti-CD20 CAR without flag).
- the fourth expression cassette encodes a CAR with features set forth in Table 4.
- the CAR is an anti-FITC CAR.
- the fourth expression cassette comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82. In some embodiments, the fourth expression cassette comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82. In some embodiments, the fourth expression cassette comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82.
- the fourth expression cassette comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82. In some embodiments, the fourth expression cassette comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82. In some embodiments, the fourth expression cassette comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82.
- the fourth expression cassette comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82. In some embodiments, the fourth expression cassette comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82. In some embodiments, the fourth expression cassette comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82.
- the fourth expression cassette comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82. In some embodiments, the fourth expression cassette comprises the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82. In some embodiments, the fourth expression cassette consists of the nucleotide sequence of SEQ ID NOs: 26, 63, 71, or 82.1n some embodiments, the fourth expression cassette encodes an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127.
- the fourth expression cassette encodes an amino acid sequence at least 80% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127. In some embodiments, the fourth expression cassette encodes an amino acid sequence at least 85% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127. In some embodiments, the fourth expression cassette encodes an amino acid sequence at least 90% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127. In some embodiments, the fourth expression cassette encodes an amino acid sequence at least 95% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127.
- the fourth expression cassette encodes an amino acid sequence at least 96% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127. In some embodiments, the fourth expression cassette encodes an amino acid sequence at least 97% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127. In some embodiments, the fourth expression cassette encodes an amino acid sequence at least 98% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127. In some embodiments, the fourth expression cassette encodes an amino acid sequence at least 99% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127.
- the fourth expression cassette encodes an amino acid sequence at least 100% identical to the nucleotide sequence of SEQ ID NOs: 27, 72 or 127. In some embodiments, the fourth expression cassette encodes an amino acid sequence comprising the sequence of SEQ ID NOs: 27, 72 or 127. In some embodiments, the fourth expression cassette encodes an amino acid sequence consisting of the sequence of SEQ ID NOs: 27, 72 or 127. [0257] In further embodiments, an illustrative nucleotide sequence encoding a CAR may comprise SEQ ID NO: 71, and an illustrative CAR amino acid sequence may comprise SEQ ID NO:72.
- An illustrative nucleotide insert may comprise SEQ ID NO:73.
- the CAR may be encoded by a nucleic acid sequence that encodes a signal peptide to signal transport of the CAR in the cell. It is understood that typically the signal peptide is removed from the protein.
- An illustrative CAR amino acid sequence without a signal peptide may comprise SEQ
- An illustrative CAR amino acid sequence signal peptide may comprise SEQ ID NO:75.
- CAR-expressing cells comprising the nucleic acid of SEQ ID NO:71 or 73 are provided.
- a chimeric antigen receptor polypeptide comprising SEQ ID NO:72 is contemplated.
- a chimeric antigen receptor polypeptide comprising SEQ ID NO:74 is contemplated.
- a vector is contemplated comprising SEQ ID NO: 71 or 73.
- a lentiviral vector is contemplated comprising SEQ ID NO: 71 or 73.
- SEQ ID NO: 72 can comprise or consist of human or humanized amino acid sequences.
- SEQ ID NO: 74 can comprise or consist of human or humanized amino acid sequences.
- variant nucleic acid sequences or amino acid sequences having at least about 80%, at least about 90%, at least about 95%, at least about 97%, at least about 98%, at least about 99%, or at least about 99.5% sequence identity to SEQ ID NO: 71, SEQ ID NO: 72, SEQ ID NO: 73 or SEQ ID NO: 74 are contemplated.
- the binding affinity of the CARs to the targeted ligand will generally be at least about 100 nM, 1 pM, or 10 pM, preferably at least about 100 pM, 1 fM or 10 fM, even more preferably at least about 100 fM.
- linkers comprise sites for cleavage, making them cleavable linkers.
- Cleavage sites can be used in the design of polycistronic constructs to achieve coexpression of multiple genes.
- the cleavage sites comprise self-cleaving sites.
- the self-cleaving site comprises a 2A site.
- 2A peptides are a class of 18-22 amino acid-long peptides that can induce ribosomal skipping during translation resulting in loss of a peptide bond between glycine and proline residues, which allows a proteolytic enzyme to recognize the 2A site.
- the most commonly used 2A peptides in molecular biology include T2A, P2A, E2A, and F2A.
- the polycistronic constructs provided herein comprise one or more cleavable linkers.
- the one or more cleavable linkers separating the expression cassettes are the same.
- the cleavable linkers separating the expression cassettes are different.
- the one or more cleavable linkers separating the expression cassettes comprise one or more cleavage sites.
- the one or more cleavage sites are the same.
- the one or more cleavage sites are different.
- the cleavable linker in addition to a 2A site, may also comprise another cleavage site.
- the additional cleavage site comprises a furin site. There are three known furin sites, including FC1, FC2 and FC3.
- the polycistronic construct provided herein comprises a T2A, P2A, E2A, or F2A cleavage site in the cleavable linker.
- the polycistronic construct comprises a T2A cleavage site in a cleavable linker.
- the polycistronic construct comprises a P2A cleavage site in a cleavable linker.
- the polycistronic construct comprises a furin cleavage site in a cleavable linker.
- the polycistronic construct comprises a T2A cleavage site and a furin cleavage site in a cleavable linker.
- the polycistronic construct provided herein comprises at least one, at least two or at least three 2A cleavable linker sequences.
- the polycistronic construct herein comprises a T2A cleavage site and a P2A, E2A or F2A cleavage site.
- the polycistronic construct herein comprises a P2A cleavage site and a T2A, E2A or F2A cleavage site.
- the polycistronic construct herein comprises a E2A cleavage site and a P2A, T2A or F2A cleavage site.
- the polycistronic construct herein comprises a F2A cleavage site and a P2A, E2A or T2A cleavage site.
- the polycistronic construct provided herein comprises a 2A cleavable linker sequence.
- each nucleotide sequence encoding the 2A cleavable linker sequences is different.
- the 2A cleavable linker is independently a T2A, P2A, E2A or F2A cleavage site.
- the 2A cleavable linker is independently a P2A or a T2A.
- the 2A cleavable linker is a P2A and the nucleotide sequence encoding the P2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NOs: 17, 25, 52, or 58.
- the nucleotide sequence encoding the P2A cleavable linker is set forth in SEQ ID NOs: 17, 25, 52, or 58.
- the P2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 18. In some embodiments, the P2A cleavable linker comprises the sequence set forth in SEQ ID NO: 18.
- At least one 2A cleavable linker is a T2A and the nucleotide sequence encoding the T2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:9.
- the nucleotide sequence encoding the T2A cleavable linker is set forth in SEQ ID NO:9.
- the T2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 10.
- the T2A cleavable linker comprising the sequence set forth in SEQ ID NO: 10.
- At least one of the cleavage site sequences comprises a furin cleavage site sequence.
- the furin cleavage site sequence is located between the first expression cassette and the second expression cassette.
- the nucleotide sequence encoding the furin cleavage site sequence comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO:7.
- the nucleotide sequence encoding the furin cleavage site sequence comprises the sequence set forth in SEQ ID NO: 7.
- the furin cleavage site sequence comprises an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 8. In some embodiments, the furin cleavage site sequence comprises the amino acid sequence of SEQ ID NO: 8.
- the cleavage site sequence comprises a furin cleavage site sequence and a T2A cleavage sequence (furinT2A).
- the nucleotide sequence encoding the cleavage site sequence is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 5.
- the nucleotide sequence encoding the cleavage site sequence comprises the nucleotide sequence of SEQ ID NO: 5.
- the cleavage site sequence comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 6. In some embodiments, the cleavage site sequence comprises the amino acid sequence of SEQ ID NO:6.
- the first expression cassette and second expression cassette are separated by a furinT2A
- the second expression cassette and third expression cassette are separated by a P2A
- the third expression cassette and fourth expression cassette are separated by a P2A.
- the polycistronic constructs provided herein comprise the features set forth in Table 5, Table 6 or Table 7. [0278] In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO:2.
- the construct encodes a polypeptide comprising an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO:2.
- the construct encodes a polypeptide comprising an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide comprising the amino acid sequence of SEQ ID NO:2. In some embodiments, the construct encodes a polypeptide consisting of the amino acid sequence of SEQ ID NO:2.
- the construct comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO: 125.
- the construct comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO: 125.
- the construct comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct comprises the nucleotide sequence of SEQ ID NO: 125. In some embodiments, the construct consists of the nucleotide sequence of SEQ ID NO: 125.
- the construct comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO:1.
- the construct comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 1.
- the construct comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 1.
- the construct comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO: 1.
- the construct comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 1. In some embodiments, the construct comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO: 1. In some embodiments, the construct comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 1. In some embodiments, the construct comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 1. In some embodiments, the construct comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO: 1.
- the construct comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 1. In some embodiments, the construct comprises the nucleotide sequence of SEQ ID NO: 1. In some embodiments, the construct consists of the nucleotide sequence of SEQ ID NO:1.
- the construct encodes a polypeptide comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:48. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 48. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 48. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 48.
- the construct encodes a polypeptide comprising an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 48. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 48. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 48. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 48. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 48.
- the construct encodes a polypeptide comprising an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 48. In some embodiments, the construct encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 48. In some embodiments, the construct encodes a polypeptide consisting of the amino acid sequence of SEQ ID NO: 48.
- the construct comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO: 126.
- the construct comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO: 126.
- the construct comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct comprises the nucleotide sequence of SEQ ID NO: 126. In some embodiments, the construct consists of the nucleotide sequence of SEQ ID NO: 126.
- the construct comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO:48.
- the construct comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO:48.
- the construct comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct comprises the nucleotide sequence of SEQ ID NO:48. In some embodiments, the construct consists of the nucleotide sequence of SEQ ID NO:48.
- the construct encodes a polypeptide comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 85% identical to the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 90% identical to the amino acid sequence of SEQ ID NO: 103.
- the construct encodes a polypeptide comprising an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 96% identical to the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 97% identical to the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 98% identical to the amino acid sequence of SEQ ID NO: 103.
- the construct encodes a polypeptide comprising an amino acid sequence at least 99% identical to the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide comprising an amino acid sequence at least 100% identical to the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 103. In some embodiments, the construct encodes a polypeptide consisting of the amino acid sequence of SEQ ID NO: 103.
- the construct comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO: 102.
- the construct comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO: 102.
- the construct comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct comprises the nucleotide sequence of SEQ ID NO: 102. In some embodiments, the construct consists of the nucleotide sequence of SEQ ID NO: 102.
- the construct comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct comprises a nucleotide sequence at least 80% identical to the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct comprises a nucleotide sequence at least 85% identical to the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct comprises a nucleotide sequence at least 90% identical to the nucleotide sequence of SEQ ID NO: 101.
- the construct comprises a nucleotide sequence at least 95% identical to the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct comprises a nucleotide sequence at least 96% identical to the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct comprises a nucleotide sequence at least 97% identical to the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct comprises a nucleotide sequence at least 98% identical to the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct comprises a nucleotide sequence at least 99% identical to the nucleotide sequence of SEQ ID NO: 101.
- the construct comprises a nucleotide sequence at least 100% identical to the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct comprises the nucleotide sequence of SEQ ID NO: 101. In some embodiments, the construct consists of the nucleotide sequence of SEQ ID NO:101.
- the polycistronic constructs can be inserted into a nucleic acid vector.
- nucleic acid vector is intended to mean any nucleic acid that functions to carry, harbor or express a nucleic acid of interest.
- Nucleic acid vectors can have specialized functions such as expression, packaging, pseudotyping, transduction or sequencing, for example.
- Nucleic acid vectors also can have, for example, manipulatory functions such as a cloning or shuttle vector.
- the structure of the vector can include any desired form that is feasible to make and desirable for a particular use. Such forms include, for example, circular forms such as plasmids and phagemids, as well as linear or branched forms.
- a nucleic acid vector can be composed of, for example, DNA or RNA, as well as contain partially or fully, nucleotide derivatives, analogs and mimetics. Such nucleic acid vectors can be obtained from natural sources, produced recombinantly or chemically synthesized.
- Non-limiting examples of vector systems of the present disclosure include a retrovirus, a lentivirus, a foamy virus, and a Sleeping Beauty transposon.
- Retroviruses include lentiviruses, gamma-retroviruses, and alpha-retroviruses, each of which may be used to deliver polynucleotides to cells using methods known in the art.
- Lentiviruses are complex retroviruses, which, in addition to the common retroviral genes gag, pol, and env, contain other genes with regulatory or structural function. The higher complexity enables the virus to modulate its life cycle, as in the course of latent infection.
- Some examples of lentivirus include the Human Immunodeficiency Viruses (HIV-1 and HIV-2) and the Simian Immunodeficiency Virus (SIV).
- Retroviral vectors have been generated by multiply attenuating the HIV virulence genes, for example, the genes env, vif, vpr, vpu and nef are deleted, making the vector biologically safe.
- Illustrative lentiviral vectors include those described in Naldini et al. (1996) Science 272:263-7; Zufferey et al. (1998) J. Virol. 72:9873-9880; Dull et al. (1998) J. Virol. 72:8463-8471; U.S. Pat. No. 6,013,516; and U.S. Pat. No. 5,994,136, which are each incorporated herein by reference in their entireties.
- these vectors are configured to carry the essential sequences for selection of cells containing the vector, for incorporating foreign nucleic acid into a lentiviral particle, and for transfer of the nucleic acid into a target cell.
- a commonly used lentiviral vector system is the so-called third-generation system.
- Third-generation lentiviral vector systems include four plasmids.
- the “transfer plasmid” encodes the polynucleotide sequence that is delivered by the lentiviral vector system to the target cell.
- the transfer plasmid generally has one or more transgene sequences of interest flanked by long terminal repeat (LTR) sequences, which facilitate integration of the transfer plasmid sequences into the host genome.
- LTR long terminal repeat
- transfer plasmids are generally designed to make the resulting vector replication incompetent.
- the transfer plasmid lacks gene elements necessary for generation of infective particles in the host cell.
- the transfer plasmid may be designed with a deletion of the 3' LTR, rendering the virus “self-inactivating” (SIN). See Dull et al. (1998) J. Virol. 72:8463-71; Miyoshi et al. (1998) J. Virol. 72:8150-57.
- the viral particle may also comprise a 3' untranslated region (UTR) and a 5' UTR.
- the UTRs comprise retroviral regulatory elements that support packaging, reverse transcription and integration of a proviral genome into a cell following contact of the cell by the retroviral particle.
- Third-generation systems also generally include two “packaging plasmids” and an “envelope plasmid.”
- the “envelope plasmid” generally encodes an Env gene operatively linked to a promoter.
- the Env gene is VSV-G and the promoter is the CMV promoter.
- the third-generation system uses two packaging plasmids, one encoding gag and pol and the other encoding rev as a further safety feature; an improvement over the single packaging plasmid of so-called second-generation systems. Although safer, the third-generation system can be more cumbersome to use and result in lower viral titers due to the addition of an additional plasmid.
- Illustrative packing plasmids include, without limitation, pMD2.G, pRSV-rev, pMDLG-pRRE, and pRRL-GOI.
- the packaging cell line is a cell line whose cells are capable of producing infectious retroviral particles when the transfer plasmid, packaging plasmid(s), and envelope plasmid are introduced into the cells.
- Various methods of introducing the plasmids into the cells may be used, including transfection or electroporation.
- a packaging cell line is adapted for high-efficiency packaging of a retroviral vector system into retroviral particles.
- Retroviral vector or “lentiviral vector” is intended to mean a nucleic acid that encodes a retroviral or lentiviral cis nucleic acid sequence required for genome packaging and one or more polynucleotide sequence to be delivered into the target cell.
- Retroviral particles and lentiviral particles generally include an RNA genome (derived from the transfer plasmid), a lipid-bilayer envelope in which the Env protein is embedded, and other accessory proteins including integrase, protease, and matrix protein.
- the terms “retroviral particle” and “lentiviral particle” refers a viral particle that includes an envelope, has one or more characteristics of a lentivirus, and is capable of invading a target host cell. Such characteristics include, for example, infecting non-dividing host cells, transducing non-dividing host cells, infecting or transducing host immune cells, containing a retroviral or lentiviral virion including one or more of the gag structural polypeptides, containing a retroviral or lentiviral envelope including one or more of the env encoded glycoproteins, containing a genome including one or more retrovirus or lentivirus cis-acting sequences functioning in replication, proviral integration or transcription, containing a genome encoding a retroviral or lentiviral protease, reverse transcriptase or integrase, or containing a genome encoding regulatory activities such as Tat or Rev.
- the transfer plasmids may comprise a cPPT sequence, as
- the efficiency of the system is an important concern in vector engineering.
- the efficiency of a retroviral or lentiviral vector system may be assessed in various ways known in the art, including measurement of vector copy number (VCN) or vector genomes (vg) such as by quantitative polymerase chain reaction (qPCR), or titer of the virus in infectious units per milliliter (lU/mL).
- VCN vector copy number
- vg vector genomes
- qPCR quantitative polymerase chain reaction
- titer of the virus in infectious units per milliliter titer may be assessed using a functional assay performed on the cultured tumor cell line HT1080 as described in Humbert et al. Development of third-generation Cocal Envelope Producer Cell Lines for Robust Retroviral Gene Transfer into Hematopoietic Stem Cells and T-cells. Molecular Therapy 24: 1237-1246 (2016).
- the retroviral particles and/or lentiviral particles of the disclosure comprise a polynucleotide comprising a sequence encoding a receptor that specifically binds to the gating adaptor.
- a sequence encoding a receptor that specifically binds to the gating adaptor is operatively linked to a promoter.
- Illustrative promoters include, without limitation, a cytomegalovirus (CMV) promoter, a CAG promoter, an SV40 promoter, an SV40/CD43 promoter, and a MND promoter.
- the retroviral particles comprise transduction enhancers. In some embodiments, the retroviral particles comprise tagging proteins.
- each of the retroviral particles comprises a polynucleotide comprising, in 5' to 3' order: (i) a 5' long terminal repeat (LTR) or untranslated region (UTR), (ii) a promoter, (iii) a sequence encoding a receptor that specifically binds to a ligand, and (iv) a 3' LTR or UTR.
- LTR 5' long terminal repeat
- UTR untranslated region
- the retroviral particles comprise a cell surface receptor that binds to a surface marker on a target host cell, allowing host cell transduction.
- the cell surface receptor is a T cell surface receptor.
- the viral vector may comprise a heterologous viral envelope glycoprotein giving a pseudotyped viral vector.
- the viral envelope glycoprotein may be derived from RD114 or one of its variants, VSV-G, Gibbon-ape leukaemia virus (GALV), or is the Amphotropic envelope, Measles envelope or baboon retroviral envelope glycoprotein.
- the cell-surface receptor is a VSV G protein from the Cocal strain or a functional variant thereof.
- fusion glycoproteins can be used to pseudotype lentiviral vectors. While the most commonly used example is the envelope glycoprotein from vesicular stomatitis virus (VSVG), many other viral proteins have also been used for pseudotyping of lentiviral vectors. See Joglekar et al. Human Gene Therapy Methods 28:291-301 (2017). The present disclosure contemplates substitution of various fusion glycoproteins. Notably, some fusion glycoproteins result in higher vector efficiency.
- VSVG vesicular stomatitis virus
- pseudotyping a fusion glycoprotein or functional variant thereof facilitates targeted transduction of specific cell types, including, but not limited to, innate lymphoid cells, cytotoxic innate lymphoid cells, or NK cells.
- the fusion glycoprotein or functional variant thereof is/are full-length polypeptide(s), functional fragment(s), homolog(s), or functional variant(s) of Human immunodeficiency virus (HIV) gpl60, Murine leukemia virus (MLV) gp70, Gibbon ape leukemia virus (GALV) gp70, Feline leukemia virus (RD114) gp70, Amphotropic retrovirus (Ampho) gp70, 10A1 MLV (10A1) gp70, Ecotropic retrovirus (Eco) gp70, Baboon ape leukemia virus (BaEV) gp70, Measles virus (MV) H and F, Nipah virus (NiV) H and
- the fusion glycoprotein or functional variant thereof is a full- length polypeptide, functional fragment, homolog, or functional variant of the G protein of Vesicular Stomatitis Alagoas Virus (VSAV), Carajas Vesiculovirus (CJSV), Chandipura Vesiculovirus (CHPV), Cocal Vesiculovirus (COCV), Vesicular Stomatitis Indiana Virus (VSIV), Isfahan Vesiculovirus (ISFV), Maraba Vesiculovirus (MARAV), Vesicular Stomatitis New Jersey virus (VSNJV), Bas-Congo Virus (BASV).
- the fusion glycoprotein or functional variant thereof is the Cocal virus G protein.
- the fusion glycoprotein or functional variant thereof is a full- length polypeptide, functional fragment, homolog, or functional variant of the G protein of Vesicular Stomatitis Alagoas Virus (VSAV), Carajas Vesiculovirus (CJSV), Chandipura Vesiculovirus (CHPV), Cocal Vesiculovirus (COCV), Vesicular Stomatitis Indiana Virus (VSIV), Isfahan Vesiculovirus (ISFV), Maraba Vesiculovirus (MARAV), Vesicular Stomatitis New Jersey virus (VSNJV), Bas-Congo Virus (BASV).
- the fusion glycoprotein or functional variant thereof is the Cocal virus G protein.
- the disclosure further provides various retroviral vectors, including but not limited to gamma-retroviral vectors, alpha-retroviral vectors, and lentiviral vectors.
- the vector may be a viral vector, a retroviral vector, a lentiviral vector, a gamma-retroviral vector.
- the viral vector comprises a VSV G-protein or functional variant thereof.
- the viral vector comprises a Cocal G-protein or functional variant thereof.
- a viral vector comprising any of the polycistronic constructs provided herein.
- the viral vector is a lentiviral vector.
- the viral vector further comprises one or more surface T cell activating agents.
- the one or more surface T cell activating agents comprise CD58, anti-CD3, or CD80.
- a virus particle encapsulating the polycistronic constructs disclosed herein.
- any of the polycistronic constructs can be provided as a payload in the generation of a viral particle.
- viral particles such as lentiviral vectors, incorporating any of the provided polycistronic constructs for delivery of components of the rapamycin activated cytokine receptor (RACR) system, including FRB, synthetic cytokine receptor and CAR, to a target cell.
- the virus particles can be engineered to express one or more surface T cell activating agents.
- the one or more surface T cell activating agents comprise a T cell surface receptor.
- the T cell surface receptor comprises CD58, anti-CD3, or CD80.
- a viral particle is a tool that allows or facilitates the transfer of an entity from one environment to another.
- some viral particles used in recombinant DNA techniques allow entities, such as a segment of DNA, to be transferred into a host cell.
- vectors used in recombinant DNA techniques include but are not limited to, plasmids, chromosomes, artificial chromosomes or viruses.
- expression vector means a construct capable of in vivo or in vitro/ex vivo expression.
- the virus particle comprises a retroviral particle.
- the disclosure provides a method for preparing a viral formulation.
- the virus is a retrovirus. A large number of different retroviruses have been identified.
- retrovirus examples include but are not limited to: murine leukemia virus (MLV), human immunodeficiency virus (HIV), human T-cell leukemia virus (HTLV), mouse mammary tumor virus (MMTV), Rous sarcoma virus (RSV), Fujinami sarcoma virus (FuSV), Moloney murine leukemia virus (Mo-MEV), FBR murine osteosarcoma virus (FBR MSV), Moloney murine sarcoma virus (Mo-MSV), Abelson murine leukemia virus (A-MEV), Avian myelocytomatosis virus-29 (MC29), and Avian erythroblastosis virus (AEV).
- MMV murine leukemia virus
- HMV human immunodeficiency virus
- HTLV human T-cell leukemia virus
- MMTV mouse mammary tumor virus
- RSV Rous sarcoma virus
- Fujinami sarcoma virus Fujinami sarcoma virus
- Retroviruses include lentiviruses, gamma-retroviruses, and alpha-retroviruses, each of which may be used to deliver polynucleotides to cells using methods known in the art.
- Lentiviruses are complex retroviruses, which, in addition to the common retroviral genes gag, pol, and env, contain other genes with regulatory or structural function. The higher complexity enables the virus to modulate its life cycle, as in the course of latent infection.
- Some examples of lentivirus include the Human Immunodeficiency Viruses (HIV-1 and HIV-2) and the Simian Immunodeficiency Virus (SIV).
- Retroviral vectors have been generated by multiply attenuating the HIV virulence genes, for example, the genes env, vif, vpr, vpu and nef are deleted, making the vector biologically safe.
- a lentiviral vector of the disclosure may be derived from or may be derivable from any suitable lentivirus.
- a recombinant retroviral vector particle is capable of transducing a recipient cell with a nucleotide of interest (NOI). Once within the cell, the RNA genome from the vector particle is reverse transcribed into DNA and integrated into the DNA of the recipient cell.
- NOI nucleotide of interest
- the RNA genome from the vector particle is reverse transcribed into DNA and integrated into the DNA of the recipient cell.
- at least part of one or more protein coding regions essential for replication may be removed from the virus. This makes the viral vector replication defective.
- Portions of the viral genome may also be replaced by an NOI in order to generate a vector comprising an NOI which is capable of transducing a target non-dividing host cell and/or integrating its genome into a host genome.
- Illustrative lentiviral vectors include those described in Naldini et al. (1996) Science 272:263-7; Zufferey et al. (1998) J. Virol. 72:9873-9880; Dull et al. (1998) J. Virol. 72:8463-8471; U.S. Pat. No. 6,013,516; and U.S. Pat. No. 5,994,136, which are each incorporated herein by reference in their entireties.
- these vectors are configured to carry the essential sequences for selection of cells containing the vector, for incorporating foreign nucleic acid into a lentiviral particle, and for transfer of the nucleic acid into a target cell.
- a commonly used lentiviral vector system is the so-called third-generation system.
- Third-generation lentiviral vector systems include four plasmids.
- the “transfer plasmid” encodes the polynucleotide sequence that is delivered by the lentiviral vector system to the target cell.
- the transfer plasmid generally has one or more transgene sequences of interest flanked by long terminal repeat (LTR) sequences, which facilitate integration of the transfer plasmid sequences into the host genome.
- LTR long terminal repeat
- transfer plasmids are generally designed to make the resulting vector replication incompetent.
- the transfer plasmid lacks gene elements necessary for generation of infective particles in the host cell.
- the transfer plasmid may be designed with a deletion of the 3' LTR, rendering the virus “self-inactivating” (SIN). See Dull et al. (1998) J. Virol. 72:8463-71; Miyoshi et al. (1998) J. Virol. 72:8150-57.
- the viral particle may also comprise a 3' untranslated region (UTR) and a 5' UTR.
- the UTRs comprise retroviral regulatory elements that support packaging, reverse transcription and integration of a proviral genome into a cell following contact of the cell by the retroviral particle.
- Third-generation systems also generally include two “packaging plasmids” and an “envelope plasmid.”
- the “envelope plasmid” generally encodes an Env gene operatively linked to a promoter.
- the Env gene is VSV-G and the promoter is the CMV promoter.
- the third-generation system uses two packaging plasmids, one encoding gag and pol and the other encoding rev as a further safety feature; an improvement over the single packaging plasmid of so-called second-generation systems. Although safer, the third-generation system can be more cumbersome to use and result in lower viral titers due to the addition of an additional plasmid.
- Illustrative packing plasmids include, without limitation, pMD2.G, pRSV-rev, pMDLG-pRRE, and pRRL-GOI.
- the packaging cell line is a cell line whose cells are capable of producing infectious retroviral particles when the transfer plasmid, packaging plasmid(s), and envelope plasmid are introduced into the cells.
- Various methods of introducing the plasmids into the cells may be used, including transfection or electroporation.
- a packaging cell line is adapted for high-efficiency packaging of a retroviral vector system into retroviral particles.
- Retroviral vector or “lentiviral vector” is intended to mean a nucleic acid that encodes a retroviral or lentiviral cis nucleic acid sequence required for genome packaging and one or more polynucleotide sequence to be delivered into the target cell.
- Retroviral particles and lentiviral particles generally include an RNA genome (derived from the transfer plasmid), a lipid-bilayer envelope in which the Env protein is embedded, and other accessory proteins including integrase, protease, and matrix protein.
- the terms “retroviral particle” and “lentiviral particle” refers a viral particle that includes an envelope, has one or more characteristics of a lentivirus, and is capable of invading a target host cell. Such characteristics include, for example, infecting non-dividing host cells, transducing non-dividing host cells, infecting or transducing host immune cells, containing a retroviral or lentiviral virion including one or more of the gag structural polypeptides, containing a retroviral or lentiviral envelope including one or more of the env encoded glycoproteins, containing a genome including one or more retrovirus or lentivirus cis-acting sequences functioning in replication, proviral integration or transcription, containing a genome encoding a retroviral or lentiviral protease, reverse transcriptase or integrase, or containing a genome encoding regulatory activities such as Tat or Rev.
- the transfer plasmids may comprise a cPPT sequence, as
- the efficiency of the system is an important concern in vector engineering.
- the efficiency of a retroviral or lentiviral vector system may be assessed in various ways known in the art, including measurement of vector copy number (VCN) or vector genomes (vg) such as by quantitative polymerase chain reaction (qPCR), or titer of the virus in infectious units per milliliter (lU/mL).
- VCN vector copy number
- vg vector genomes
- qPCR quantitative polymerase chain reaction
- titer of the virus in infectious units per milliliter titer may be assessed using a functional assay performed on the cultured tumor cell line HT1080 as described in Humbert et al. Development of third-generation Cocal Envelope Producer Cell Lines for Robust Retroviral Gene Transfer into Hematopoietic Stem Cells and T-cells. Molecular Therapy 24: 1237-1246 (2016).
- the retroviral particles and/or lentiviral particles of the disclosure comprise a polynucleotide comprising a sequence encoding a receptor that specifically binds to the gating adaptor.
- a sequence encoding a receptor that specifically binds to the gating adaptor is operatively linked to a promoter.
- Illustrative promoters include, without limitation, a cytomegalovirus (CMV) promoter, a CAG promoter, an SV40 promoter, an SV40/CD43 promoter, and a MND promoter.
- the retroviral particles comprise transduction enhancers. In some embodiments, the retroviral particles comprise tagging proteins.
- each of the retroviral particles comprises a polynucleotide comprising, in 5' to 3' order: (i) a 5' long terminal repeat (LTR) or untranslated region (UTR), (ii) a promoter, (iii) a sequence encoding a receptor that specifically binds to a ligand, and (iv) a 3' LTR or UTR.
- LTR 5' long terminal repeat
- UTR untranslated region
- the retroviral particles comprise a cell surface receptor that binds to a surface marker on a target host cell, allowing host cell transduction.
- the cell surface receptor is a T cell surface receptor.
- the viral vector may comprise a heterologous viral envelope glycoprotein giving a pseudotyped viral vector.
- the viral envelope glycoprotein may be derived from RD114 or one of its variants, VSV-G, Gibbon-ape leukaemia virus (GALV), or is the Amphotropic envelope, Measles envelope or baboon retroviral envelope glycoprotein.
- the cell-surface receptor is a VSV G protein from the Cocal strain or a functional variant thereof.
- the viral envelope comprises a viral envelope protein.
- a viral envelope protein is a VSV-G envelope protein, a measles virus envelope protein, a nipha virus envelope protein, or a cocal virus G protein.
- the viral particle comprises a modified VSV G protein that lacks LDLR binding affinity.
- these mutations comprise mutations at positions 47 (for example, K47Q) and/or 354 (for example, R354A).
- the viral envelope protein is a protein from the Cocal strain (Cocal glycoprotein).
- the protein is a Cocal envelope protein containing a mutation at position 354 (R354).
- the protein is a Cocal envelope protein containing a mutation at position 47 (K47).
- the protein is a Cocal envelope variant containing a R354Q mutation.
- the protein is a Cocal envelope variant containing a K47Q mutation. In some embodiments, this variant may be referred to as “blinded” Cocal envelope.
- Illustrative Cocal envelope variants are provided in, e.g., US 2020/0216502 Al, which is incorporated herein by reference in its entirety.
- fusion glycoproteins can be used to pseudotype lentiviral vectors. While the most commonly used example is the envelope glycoprotein from vesicular stomatitis virus (VSVG), many other viral proteins have also been used for pseudotyping of lentiviral vectors. See Joglekar et al. Human Gene Therapy Methods 28:291-301 (2017). The present disclosure contemplates substitution of various fusion glycoproteins. Notably, some fusion glycoproteins result in higher vector efficiency.
- VSVG vesicular stomatitis virus
- pseudotyping a fusion glycoprotein or functional variant thereof facilitates targeted transduction of specific cell types, including, but not limited to, innate lymphoid cells or NK-cells.
- the fusion glycoprotein or functional variant thereof is/are full-length polypeptide(s), functional fragment(s), homolog(s), or functional variant(s) of Human immunodeficiency virus (HIV) gpl60, Murine leukemia virus (MLV) gp70, Gibbon ape leukemia virus (GALV) gp70, Feline leukemia virus (RD114) gp70, Amphotropic retrovirus (Ampho) gp70, 10A1 MLV (10A1) gp70, Ecotropic retrovirus (Eco) gp70, Baboon ape leukemia virus (BaEV) gp70, Measles virus (MV) H and F, Nipah virus (NiV) H and F, Rabies virus (RabV
- the fusion glycoprotein or functional variant thereof is a full- length polypeptide, functional fragment, homolog, or functional variant of the G protein of Vesicular Stomatitis Alagoas Virus (VSAV), Carajas Vesiculovirus (CJSV), Chandipura Vesiculovirus (CHPV), Cocal Vesiculovirus (COCV), Vesicular Stomatitis Indiana Virus (VSIV), Isfahan Vesiculovirus (ISFV), Maraba Vesiculovirus (MARAV), Vesicular Stomatitis New Jersey virus (VSNJV), Bas-Congo Virus (BASV).
- the fusion glycoprotein or functional variant thereof is the Cocal virus G protein.
- the fusion glycoprotein or functional variant thereof is a full- length polypeptide, functional fragment, homolog, or functional variant of the G protein of Vesicular Stomatitis Alagoas Virus (VSAV), Carajas Vesiculovirus (CJSV), Chandipura Vesiculovirus (CHPV), Cocal Vesiculovirus (COCV), Vesicular Stomatitis Indiana Virus (VSIV), Isfahan Vesiculovirus (ISFV), Maraba Vesiculovirus (MARAV), Vesicular Stomatitis New Jersey virus (VSNJV), Bas-Congo Virus (BASV).
- the fusion glycoprotein or functional variant thereof is the Cocal virus G protein.
- the disclosure further provides various retroviral vectors, including but not limited to gamma-retroviral vectors, alpha-retroviral vectors, and lentiviral vectors.
- the vector may be a viral vector, a retroviral vector, a lentiviral vector, a gamma-retroviral vector.
- the viral vector comprises a VSV G-protein or functional variant thereof.
- the viral vector comprises a Cocal G-protein or functional variant thereof.
- the virus particles encapsulating nucleotide vectors provided herein can comprise engineered viral envelopes.
- the viral envelope comprises a transduction enhancer.
- the viral envelope comprises at an immune cell-activating protein.
- the viral envelope comprises a costimulation molecule.
- the viral envelope comprises an immune cellactivating protein, and a co-stimulation molecule.
- the viral envelope comprises one or more transduction enhancers.
- the transduction enhancers include T cell activation receptors, NK cell activation receptors, and/or co-stimulatory molecules.
- one or more transduction enhancers comprise one or more of anti-CD3scFv, CD86, CD80, and/or CD58.
- the transduction enhancers comprise at least an anti-CD3 scFv, and CD58.
- the transduction enhancers comprise at least an anti-CD3 scFv, and CD80.
- the transduction enhancers comprise at least an anti-CD3 scFv, and CD86.
- the transduction enhancers comprise at least an anti-CD3 scFv, a CD80, and CD58. In some embodiments, the transduction enhancers comprise at least an anti-CD3 scFv, a CD86, and CD58.
- the viral particle comprises a cell surface receptor that binds to a ligand on a target host cell, allowing host cell transduction.
- the cell surface receptor is a T cell surface receptor.
- the viral particle comprises a heterologous viral envelope glycoprotein yielding a pseudotyped viral particle.
- the viral envelope glycoprotein may be derived from RD114 or one of its variants, VSV-G, Gibbon-ape leukemia virus (GALV), or is the Amphotropic envelope, Measles envelope or baboon retroviral envelope glycoprotein.
- the viral envelope glycoprotein is a VSV G protein from the Cocal strain (Cocal glycoprotein) or a functional variant thereof.
- the viral envelope comprises more than one polypeptide on the surface.
- the more than one polypeptide binds to a target immune cells and replicates an immunological synapse.
- the viral envelope comprises an immune cell-activating protein, a co-stimulatory molecule, and an adhesion molecule, wherein the immune cell-activating protein, co-stimulatory molecule, and adhesion molecule each bind a target immune cell.
- the transduction enhancer comprises a mitogenic stimulus, which is incorporated into a retroviral or lentiviral capsid, such that the virus both activates and transduces T cells. This removes the need to add vector and mitogen.
- the transduction enhancer comprises a mitogenic transmembrane protein and/or one or more costimulatory molecules, which get(s) incorporated into the retrovirus when it buds from the producer/packaging cell membrane.
- the transduction enhancers are expressed as separate cell surface molecules on the producer cell rather than being part of the viral envelope glycoprotein.
- the viral vector described herein comprises a mitogenic transduction enhancer in the viral envelope.
- the mitogenic transduction enhancer is derived from the host cell during retroviral vector production.
- the mitogenic transduction enhancer is made by the packaging cell and expressed at the cell surface. When the nascent retroviral vector buds from the host cell membrane, the mitogenic transduction enhancer may be incorporated in the viral envelope as part of the packaging cell-derived lipid bilayer.
- the mitogenic enhancer is an antibody or fragment thereof.
- the mitogenic enhancer is a single domain antibody, for example, a camelid antibody.
- the mitogenic enhancer is an scFv.
- the mitogenic enhancer is a nanobody.
- the transduction enhancer is host-cell derived.
- hostcell derived indicates that the mitogenic transduction enhancer is derived from the host cell as described above and is not produced as a fusion or chimera from one of the viral genes, such as gag, which encodes the main structural proteins; or env, which encodes the envelope protein.
- Envelope proteins are formed by two subunits, the transmembrane (TM) that anchors the protein into the lipid membrane and the surface (SU) which binds to the cellular receptors.
- the packaging-cell derived mitogenic transduction enhancer of the present invention does not comprise the surface envelope subunit (SU).
- the mitogenic transduction enhancer has the structure: M-S-TM, in which M is a mitogenic domain; S is an optional spacer domain and TM is a transmembrane domain.
- the mitogenic domain is the part of the mitogenic transduction enhancer which causes T-cell activation. It may bind or otherwise interact, directly or indirectly, with a T cell, leading to T cell activation. In some embodiments, the mitogenic domain binds a T cell surface antigen, such as CD3, CD28, CD 134 and CD 137.
- CD3 is a T-cell co-receptor. It is a protein complex composed of four distinct chains. In mammals, the complex contains a CD3y chain, a CD35 chain, and two CD3e chains. These chains associate with the T-cell receptor (TCR) and the z-chain to generate an activation signal in T lymphocytes. The TCR, z-chain, and CD3 molecules together comprise the TCR complex. In some embodiments, the mitogenic domain binds to a CD3 e chain.
- the mitogenic domain comprises all or part of an antibody or other molecule which specifically binds a T-cell surface antigen.
- the antibody activates the TCR or CD28.
- the antibody binds the TCR, CD3 or CD28. Examples of such antibodies include: 0KT3, 15E8 and TGN1412.
- Other suitable antibodies include:
- Anti-CD28 CD28.2, 10F3
- Anti-CD3/TCR UCHT1 , YTH12.5, TR66
- the mitogenic domain comprises the binding domain from OKT3, 15E8, TGN1412, CD28.2, 10F3, UCHT1, YTH12.5 or TR66.
- the mitogenic domain comprises all or part of a co-stimulatory molecule such as OX40E and 41BBE.
- the mitogenic domain may comprise the binding domain from OX40E or 41BBE.
- OKT3 also known as Muromonab-CD3 is a monoclonal antibody targeted at the CD3e chain. It is clinically used to reduce acute rejection in patients with organ transplants. It was the first monoclonal antibody to be approved for clinical use in humans.
- the viral envelope comprises an immune cell-activating protein.
- the immune cell-activating protein specifically binds a receptor on an immune cell.
- the immune cell-activating protein provides signal one for T cell activation.
- the immune cell-activating protein specifically binds CD2, CD3, CD28H, EFA-1, DNAM-1, CD27, ICOS, EIGHT, GITR, CD30, SEAM, Ey-9, CD84, Eyl08, NKG2D, NKp46, NKp44, NKp30, CD244, or NKp80. In some embodiments, the immune cellactivating protein specifically binds CD3y, CD35, or CD3s.
- the immune cell-activating protein specifically binds CD3y, CD35, CD3s, CD9, CD5, CD22, CD33, CD37, CD64, CD45, CD28H, EFA-1, DNAM-1, CD27, ICOS, LIGHT, GITR, CD30, SLAM, Ly-9, CD84, Lyl08, CD 16, CD56, NKG2D, NKp46, NKp44, NKp30, CD244, NKp80, TCRa chain, TCRP chain, TCRy chain, or TCR5 chain.
- the immune cell- activating protein specifically binds CD3y, CD35, or CD3s.
- the immune cell-activating protein specifically binds CD3.
- the immune cell-activating protein is an antibody or antigen binding fragment thereof that specifically binds a receptor on an immune cell.
- the immune cell-activating protein is an antibody or antigen binding fragment thereof that specifically binds CD28, CD2, CD3, CD28H, LFA-1, 0X40, 4-1BB, CD40L, DNAM- 1, CD27, ICOS, LIGHT, GITR, CD30, SLAM, Ly-9, CD84, Lyl08, NKG2D, NKp46, NKp44, NKp30, CD244, or NKp80.
- the immune cell-activating protein is an antibody or antigen binding fragment thereof that specifically binds CD28, CD2, CD3y, CD35, CD3s, CD4, CD8, CD9, CD5, CD22, CD33, CD37, CD64, CD45, CD28H, LFA-1, 0X40, 4-1BB, CD40L, DNAM-1, CD27, ICOS, LIGHT, GITR, CD30, SLAM, Ly-9, CD84, Lyl08, CD16, CD56, NKG2D, NKp46, NKp44, NKp30, CD244, NKp80, TCRa chain, TCR0 chain, TCRy chain, or TCR5 chain, some embodiments, the immune cell-activating protein is an antibody or antigen binding fragment thereof that specifically binds CD3y, CD35, or CD3s. In some embodiments, immune cell-activating protein is an antibody or antigen binding fragment thereof that specifically binds CD3.
- Antibodies targeting the polypeptides described herein are known to those of skill in the art. Methods for generating antibodies are known to those of skill in the art.
- the viral envelope comprises an anti-CD3s antibody, or antigenbinding fragment thereof.
- the anti-CD3s antibody, or antigen -binding fragment thereof is coupled to a transmembrane domain.
- An illustrative anti-CD3s antibody is 0KT3.
- 0KT3, also known as Muromonab-CD3, is a monoclonal antibody targeted at the CD3s chain.
- the viral envelope comprises a single chain Fv fragment (scFv) of an anti-CD3 antibody.
- scFv single chain Fv fragment
- the viral envelope comprises at least one co-stimulatory molecule.
- the co-stimulatory molecule specifically binds a receptor on an immune cell.
- the co-stimulatory provides signal two for cell activation.
- costimulatory molecule refers to a molecule capable of generating a costimulatory signal to T cells.
- Lymphocytes such as T cells and natural killer (NK) cells, typically require several signals and interactions with antigen presenting cells (APCs) for optimal priming to gain full effector functions.
- APCs antigen presenting cells
- T cells these include signaling through the T cell receptor (TCR), costimulatory molecules (such as CD28 and CD2), cytokines, as well as various adhesion molecules necessary to allow sufficient time for proper synapse formation and signal transduction.
- TCR T cell receptor
- costimulatory molecules such as CD28 and CD2
- cytokines as well as various adhesion molecules necessary to allow sufficient time for proper synapse formation and signal transduction.
- NK cells require similar types of stimulation but may rely on different activating receptors, such as NKG2D, NKp46, and DNAM-1.
- costimulation in addition to TCR stimulation, is especially important for effective priming and many studies have shown that TCR stimulation alone can lead to functional anergy and unresponsiveness.
- Costimulatory signals augment T and NK cell function by enhancing cell metabolism, cytokine production, differentiation, and long-term persistence. Costimulation is an important factor for cell proliferation, differentiation and survival.
- costimulatory molecules include, but are not limited to, CD45, CD2, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD28, CD37, CD64, CD80, CD86, CD134, CD 137, and CD 154.
- the costimulatory molecule includes, but is not limited to, binding agents, such as scFvs, antibodies, single-domain antibodies, antibody fragments, nanobodies that bind to any of the costimulatory molecules described herein.
- these binding agents may include anti-CD28, anti-CD2, anti-CD45, anti-CD4, anti-CD5, anti-CD8, anti-CD9, anti-CD16, anti-CD22, anti-CD33, anti-CD37, anti-CD64, anti-CD80, anti-CD86, antiCD 137, anti-CD154, anti-CD28H, anti-LFA-1, anti-OX40, anti-4- IBB, anti-CD40L, anti-DNAM-1, anti-CD27, anti-ICOS, anti-LIGHT, anti-GITR, anti-CD30, anti-SLAM, anti-Ly-9, anti-CD84, anti- Lyl08, anti-NKG2D, anti-NKp46, anti-NKp44, anti-NKp30
- the co-stimulation molecule is a ligand for CD28.
- CD28 is one of the proteins expressed on T cells that provide co-stimulatory signals required for T cell activation and survival. T cell stimulation through CD28 in addition to the T-cell receptor (TCR) can provide a potent signal for the production of various interleukins (IL-6 in particular).
- the co-stimulation molecule is an antibody, or fragment thereof, that binds to CD28. Examples of such antibodies include: 15E8 and TGN1412. Other suitable antibodies include: CD28.2 and 10F3.
- the co-stimulation molecule is CD86.
- CD86 also known as B7-2, is a ligand for CD28.
- the ligand for CD28 is CD86.
- the co-stimulation molecule is CD80.
- CD80 is an additional ligand for CD28.
- the ligand for CD28 is CD80.
- the ligand for CD28 is an anti- CD28 antibody or an anti-CD28 scFv.
- the anti-CD28 antibody or an anti- CD28 scFv is coupled to a transmembrane domain for display on the surface of the viral envelope.
- the co-stimulation molecule is a CD86 polypeptide comprising the amino acid sequence of SEQ ID NO: 76. In some embodiments, the co-stimulation molecule is a CD86 polypeptide comprising an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 76.
- the co-stimulation molecule is a CD80 polypeptide comprising the amino acid sequence of SEQ ID NO: 77. In some embodiments, the co-stimulation molecule is a CD80 polypeptide comprising an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 77.
- the CD86 polypeptide is encoded by the nucleotide sequence of SEQ ID NO: 78. In some embodiments, the CD86 polypeptide is encoded by a nucleotide sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 78.
- the CD80 polypeptide is encoded by the nucleotide sequence of SEQ ID NO: 79. In some embodiments, the CD80 polypeptide is encoded by a nucleotide sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 79.
- CD 134 also known as 0X40, is a member of the TNFR- superfamily of receptors which is expressed on activated T cells. 0X40 may promote cell division and survival. 0X40 is a secondary costimulatory molecule, expressed after 24 to 72 hours following activation; its ligand, OX40L, is also not expressed on resting antigen presenting cells, but is following their activation.
- the viral particle comprises a ligand for 0X40, or functional fragment thereof, coupled to its native transmembrane domain or a heterologous transmembrane domain.
- CD 134 also known as 0X40, is a member of the TNFR- superfamily of receptors which is expressed on activated T cells. 0X40 may promote cell division and survival. 0X40 is a secondary costimulatory molecule, expressed after 24 to 72 hours following activation; its ligand, OX40L, is also not expressed on resting antigen presenting cells, but is following their activation.
- the viral particle comprises a ligand for 0X40, or functional fragment thereof, coupled to its native transmembrane domain or a heterologous transmembrane domain.
- CD 137 also known as 4- IBB, is a member of the tumor necrosis factor (TNF) receptor family. CD 137 is expressed on activated T cells. In addition, CD 137 expression is found on dendritic cells, follicular dendritic cells, natural killer cells, granulocytes and cells of blood vessel walls at sites of inflammation. The best characterized activity of CD 137 is its costimulatory activity for activated T cells. Crosslinking of CD 137 enhances T cell proliferation, IL-2 secretion survival and cytolytic activity.
- the viral particle comprises a ligand for 4- IBB, or functional fragment thereof, coupled to its native transmembrane domain or a heterologous transmembrane domain.
- 4-1 BBL is a cytokine that belongs to the tumor necrosis factor (TNF) ligand family. This transmembrane cytokine is a bidirectional signal transducer that acts as a ligand for 4- IBB, which is a costimulatory receptor molecule in T lymphocytes. 4-1BBL has been shown to reactivate anergic T lymphocytes in addition to promoting T lymphocyte proliferation.
- TNF tumor necrosis factor
- Viral particles comprising one or more activation or co-stimulation molecule(s) may be made by engineering the packaging cell line by methods provided by WO 2016/139463; or by expression of the T-cell activation or co-stimulation molecule(s) from a polycistronic helper vector as described in IntT Pat. Pub. No. WO 2020/106992 Al, both of which are incorporated herein by reference in their entireties.
- the viral particle comprises an adhesion molecule.
- adhesion molecule refers to a subset of cell surface molecules involved in the binding of cells with other cells. Adhesion cells may help to form more stable interactions, such as an immunological synapse, between immune cells.
- the immunological synapse is a stable adhesive junction between a polarized immune effector cell and an antigen-bearing cell.
- the adhesion molecule may provide a costimulatory signal to the target cell.
- adhesion molecules include, but are not limited to, CD58, HHLA2, ICAM-1, OX40L, 4-1BBL, CD40, CD155, CD70, HVEM, GITRL, ICOSL, CD30L, SLAM, Ly-9, CD84, Lyl08, MICA, MICB, ULBP1, ULBP2, ULBP3, ULBP4, ULBP5, ULBP6, and B7-H6.
- the adhesion molecule includes, but is not limited to, binding agents, such as scFvs, antibodies, single-domain antibodies, antibody fragments, and nanobodies that bind to any of the adhesion or costimulatory molecules described herein.
- these binding agents may include anti-CD28, anti-CD2, anti-CD28H, anti-LFA-1, anti-OX40, anti-4-lBB, anti-CD40L, anti-DNAM-1, anti-CD27, anti-ICOS, anti-LIGHT, anti-GITR, anti-CD30, anti-SLAM, anti-Ly-9, anti-CD84, anti-LylO8, anti-NKG2D, anti-NKp46, anti-NKp44, anti-NKp30, anti-CD244, anti- NKp80, anti-TCRa chain, anti-TCRP chain, anti-TCRy chain, and anti-TCR8 chain agents.
- the adhesion molecule binds to CD2.
- CD2 is also known as T11, LFA-2, and the erythrocyte rosette receptor and is a surface protein expressed on T lymphocytes and NK cells.
- CD2 is a natural ligand for CD58.
- engagement of CD2 provides a costimulatory signal that may enhance activation and effector functions.
- the lentiviral particle comprises a molecule that binds to CD2.
- the lentiviral particle comprises an antibody, single domain antibody, antibody fragment, and/or nanobody specific for CD2.
- the lentiviral particle comprises CD58, or a functional portion thereof that binds to CD2.
- the adhesion molecule is CD58.
- the costimulation molecule is a CD58 polypeptide comprising the amino acid sequence of SEQ ID NO: 80.
- the co-stimulation molecule is a CD58 polypeptide comprising an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 80.
- the CD58 polypeptide is encoded by the nucleotide sequence of SEQ ID NO: 81. In some embodiments, the CD58 polypeptide is encoded by a nucleotide sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identical to SEQ ID NO: 81.
- the viral particle comprises at least one non-viral protein. In some embodiments, the viral particle comprises at least one non-viral protein in addition to those described supra.
- the viral particle comprises a targeting ligand.
- the viral particle comprises CD 19, or a functional fragment thereof, coupled to its native transmembrane domain or a heterologous transmembrane domain.
- CD19 acts as a ligand for blinatumomab, thus providing an adapter for coupling the particle to T- cells via the anti-CD3 moiety of blinatumomab.
- another type of particle surface ligand can serve to couple an appropriately surface engineered lentiviral particle to a T-cell using a multispecific antibody comprising a binding moiety for the particle surface ligand.
- the multispecific antibody is a bispecific antibody, for example, a Bispecific T-cell engager (BiTE).
- the non-viral protein is a cytokine.
- the cytokine may be selected from the group consisting of IL-2, IL-7, IL-12, IL-15, IL-18, IL-21, and any combination thereof.
- the non-viral protein used is a soluble protein (such as an scEv or a cytokine) it may be tethered to the surface of the viral particle by fusion to a transmembrane domain, such as the transmembrane domain of CD8. Alternatively, it may be indirectly tethered to the lentiviral particle by use of a transmembrane protein engineered to bind the soluble protein. Eurther inclusion of one or more cytoplasmic residues may increase the stability of the fusion protein.
- the mitogenic transduction enhancer and/or cytokine- based transduction enhancer may comprise a “spacer sequence” to connect the antigen-binding domain with the transmembrane domain.
- a flexible spacer allows the antigen-binding domain to orient in different directions to facilitate binding.
- the term “coupled to” refers to a chemical linkage, a direct C- terminal to N-terminal fusion of two protein; chemical linkage to a non-peptide space; chemical linkage to a polypeptide space; and C-terminal to N-terminal fusion of two protein via peptide bonds to a polypeptide spacer, e.g., a spacer sequence.
- the spacer sequence may, for example, comprise an IgGl Ec region, an IgGl hinge or a human CD8 stalk or the mouse CD8 stalk.
- the spacer may alternatively comprise an alternative linker sequence which has similar length and/or domain spacing properties as an IgGl Ec region, an IgGl hinge or a CD8 stalk.
- a human IgGl spacer may be altered to remove Ec binding motifs.
- the spacer sequence may be derived from a human protein.
- the spacer sequence comprises a CD8 derived hinge.
- the spacer sequence comprises a ‘short’ hinge.
- the short hinge is described as hinge region comprising fewer nucleotides relative to CAR hinge regions known in the art.
- the transmembrane domain is the sequence of the mitogenic transduction enhancer and/or cytokine-based transduction enhancer that spans the membrane.
- the transmembrane domain may comprise a hydrophobic alpha helix.
- the transmembrane domain may be derived from CD28. In some embodiments, the transmembrane domain is derived from a human protein.
- the viral particle of the present invention may comprise a cytokine-based transduction enhancer in the viral envelope.
- the cytokine-based transduction enhancer is derived from the host cell during viral particle production.
- the cytokinebased transduction enhancer is made by the host cell and expressed at the cell surface. When the nascent viral particle buds from the host cell membrane, the cytokine-based transduction enhancer may be incorporated in the viral envelope as part of the packaging cell-derived lipid bilayer.
- the cytokine -based transduction enhancer may comprise a cytokine domain and a transmembrane domain. It may have the structure C-S-TM, where C is the cytokine domain, S is an optional spacer domain (e.g., a spacer sequence) and TM is the transmembrane domain.
- C is the cytokine domain
- S is an optional spacer domain (e.g., a spacer sequence)
- TM is the transmembrane domain.
- the spacer domain and transmembrane domains are as defined above.
- the cytokine domain may comprise a T-cell activating cytokine, such as from IL2, IL7 and IL15, or a functional fragment thereof.
- a “functional fragment” of a cytokine is a fragment of a polypeptide that retains the capacity to bind its particular receptor and activate T- cells.
- IL2 is one of the factors secreted by T cells to regulate the growth and differentiation of T cells and certain B cells.
- IL2 is a lymphokine that induces the proliferation of responsive T cells. It is secreted as a single glycosylated polypeptide, and cleavage of a signal sequence is required for its activity.
- Solution NMR suggests that the structure of IL2 comprises a bundle of 4 helices (termed A-D), flanked by 2 shorter helices and several poorly defined loops. Residues in helix A, and in the loop region between helices A and B, are important for receptor binding.
- provided herein are methods of using the polycistronic constructs or nucleotide vectors disclosed herein.
- the provided methods deliver the polycistronic constructs to cells for expression of the provided systems including a cytosolic FRB, synthetic cytokine receptor and CAR.
- the polycistronic constructs are contained within a viral vector and the viral vector is used to transduce target cells.
- the provided methods are carried out ex vivo or in vitro to engineer target cells with the polycistronic construct.
- the engineered cells are administered to a subject.
- the provided methods are carried out in vivo and a viral vector containing the polycistronic construct is introduced to a subject for in vivo targeted delivery of the polycistronic vector to a target cell, such as a T cell.
- the target cell comprises a stem cell.
- the stem cell comprises an induced pluripotent stem cell (iPSC).
- iPSC induced pluripotent stem cell
- the target cell comprises a progenitor cell.
- the progenitor cell comprises a peripheral blood mononuclear cell (PBMC).
- PBMC peripheral blood mononuclear cell
- the target cell comprises a T cell.
- the T cell comprises a CD4+ or CD8+ T cell.
- the method further comprises contacting the target cell with a (i) a guide RNA (gRNA) targeting a target site in an endogenous gene, and (ii) an RNA-guided endonuclease, thereby inserting the nucleotide sequence into the endogenous gene.
- a guide RNA gRNA
- gRNA guide RNA
- the polynucleotides described herein may be delivered to cells in vivo.
- a polynucleotide encoding the elements of the polycistronic construct disclosed herein is administered to a subject directly via administration of a particle comprising the polynucleotide.
- the particle is a viral particle.
- the viral particle comprises an anti-CD3 scFv and a cocal glycoprotein and is capable of delivering the polynucleotide to cells in vivo.
- the polynucleotide encodes a cytosolic FRB, a synthetic cytokine receptor, and a chimeric antigen receptor (CAR).
- the polynucleotides disclosed herein may be administered to the subject which allows the production of the various construct components (e.g. FRB, synthetic cytokine receptor, and CAR) in vivo.
- the administration of such polynucleotide generates similar effect in vivo as direct administration of ex vivo engineered cells expressing the FRB, synthetic cytokine receptor, and CAR.
- the administration of such polynucleotide improves the in vivo transduction efficiency of a particle.
- the polynucleotide is an mRNA.
- the polynucleotides described herein may also be delivered to cells ex vivo. Viral particles described herein may be used ex vivo, either in convention cell manufacturing processes or in an extracorporeal or bedside process as described, e.g., in IntT Pat. Pub. No.
- the disclosure provides an ex vivo method of transducing target cells, comprising contacting the target cells with the particle according to the present disclosure.
- the particles described herein may be used to transduce cells that have not been previously activated.
- the particles described herein may be useful for transducing cells that have not been previously contacted with cell activation beads or activation reagents (e.g. Dynabeads or other reagents comprising anti-CD3 and/or anti-CD28 antibodies or binding fragments thereof).
- the disclosure provides a method of delivering a nucleic acid to a cell in an ex vivo CAR T manufacturing process.
- Such methods typically involve the isolation of PBMCs from a patient via leukapheresis. These cells are washed and optionally further purified via one or more selection steps to isolate particular T cell populations of interest. In some aspects, these might include CD4+ and/or CD8+ T cells.
- the washed and/or purified cells may be optionally activated and then transduced using a lentiviral vector.
- the activation step may comprise contacting the cells with an exogenous activation agent such as anti-CD3 and anti-CD28 antibodies bound to a substrate or using unbound antibodies.
- Illustrative activation agents include anti-CD3 and anti- CD28-presenting beads and/or soluble polymers. After transduction, the cells may be optionally further washed and cultured until harvest.
- Methods of manufacturing engineered cell therapies, including CAR T cells are known in the art (see e.g., Abou-el-Enein, M. et al. Blood Cancer Discov (2021), Vol 2(5): 408-422; Arcangeli, S. et al. Front. Immunol (19 Jun 2020), Vol. 11 (1217) 1-13; Ghassemi, S. et al. Nat Biomed Eng (Feb 2022), Vol 6(2): 118-128; Vormittag, P. et al.
- the disclosure provides a method of delivering a nucleic acid to a cell in an ex-vivo closed-loop manufacturing process.
- an ex-vivo manufacturing process is an extracorporeal process.
- the lentiviral vectors disclosed herein permit delivery of a nucleic acid to a target cell during a closed-loop process. Exemplary methods of closed-loop and/or extracorporeal processes are disclosed in US Patent Publication No. 2021/0244871 and WO2022072885, both of which are incorporated herein in their entirety.
- the lentiviral vectors as disclosed herein may be used to transduce cells ex vivo.
- cells are obtained from a subject, washed, incubated and/or contacted with lentiviral particles, optionally washed again, and infused into the subject in a closed-loop system.
- the lentiviral particles as disclosed herein are useful even without prior activation of the cells and are capable of binding to the cells in a short incubation and/or contacting step.
- the incubation and/or contacting step is approximately or less than one hour.
- the incubation and/or contacting step is approximately or less than two hours, approximately or less than three hours, approximately or less than four hours, or approximately or less than five hours.
- the incubation and/or contacting step is less than 12 hours or less than 24 hours.
- a nucleic acid is delivered to a cell by transduction with a lentiviral vector such that the nucleic acid enters the cell ex- vivo.
- a nucleic acid is delivered to a cell by contacting the lentiviral vector to the surface of the cell.
- the nucleic acid may enter the cell ex-vivo or in vivo after the cells (complexed with the lentiviral vector) are infused back into the subject.
- the lentiviral vectors as disclosed herein eliminate the need for an ex-vivo activation step.
- the isolated cells could be transduced directly after leukapheresis, washing, or selection.
- the surface engineering described herein enables the lentiviral particles disclosed herein to activate and transduce cells in a single step.
- the lentiviral particles disclosed herein may enable a short or truncated manufacturing process, reducing the time spent in ex-vivo manufacturing by eliminating one or more unit operations (e.g. activation prior to transduction) and/or reducing the amount of time necessary in post-transduction cell culture.
- a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a target cell include contacting a target cell with a particle, such as a viral vector, containing any of the polycistronic constructs provided herein.
- the contacting is carried out ex vivo or in vitro.
- the contacting is carried out in vivo in a subject by administering the polynucleotide constructs or particles, such as viral vectors, containing the same to a subject.
- the target cell comprises a stem cell.
- the stem cell comprises an induced pluripotent stem cell (iPSC).
- iPSC induced pluripotent stem cell
- the target cell comprises a progenitor cell.
- the progenitor cell comprises a peripheral blood mononuclear cell (PBMC).
- PBMC peripheral blood mononuclear cell
- the target cell comprises a T cell.
- the T cell comprises a CD4+ or CD8+ T cell.
- the method is performed ex vivo or in vitro.
- the method is performed in vivo.
- provided herein is a cell produced by any of the methods provided herein.
- the disease or condition is a tumor or cancer.
- the cells or pharmaceutical composition thereof is administered in an effective amount to effect treatment of the disease or disorder. Uses include uses of the cells or pharmaceutical compositions thereof in such methods and treatments, and in the preparation of a medicament in order to carry out such therapeutic methods. In some embodiments, the methods thereby treat the disease or condition or disorder in the subject.
- engineered cells that contain any of the provided polycistronic constructs that encode a cytosolic FRB, a synthetic cytokine receptor, and a chimeric antigen receptor (CAR) may be administered to a subject to treat a disease or condition.
- particles, such as viral vectors e.g. lentiviral vectors
- viral vectors e.g. lentiviral vectors
- the disclosed cells or particles may be administered in a number of ways depending upon whether local or systemic treatment is desired.
- parenteral administration may be topical, parenteral, or enteral.
- the compositions of the disclosure are typically suitable for parenteral administration.
- parenteral administration of a pharmaceutical composition includes any route of administration characterized by physical breaching of a tissue of a subject and administration of the pharmaceutical composition through the breach in the tissue, thus generally resulting in the direct administration into the blood stream, into muscle, or into an internal organ.
- Parenteral administration thus includes, but is not limited to, administration of a pharmaceutical composition by injection of the composition, by application of the composition through a surgical incision, by application of the composition through a tissue- penetrating non-surgical wound, and the like.
- parenteral administration is contemplated to include, but is not limited to, subcutaneous, intraperitoneal, intramuscular, intrastemal, intravenous, intraarterial, intrathecal, intraventricular, intraurethral, intracranial, intratumoral, intrasynovial injection or infusions; and kidney dialytic infusion techniques.
- parenteral administration of the compositions of the present disclosure comprises intravenous administration.
- the viral particle is administered by intraperitoneal injection of the viral particle.
- the viral particle is administered by intra-nodal injection - that is, the viral particle may be administered via injection into a lymph node, such as an inguinal lymph node.
- the viral particle is administered by injection of the viral particle into tumor sites (i.e. intratumoral). In some embodiments, the viral particle is administered subcutaneously. In some embodiments, the viral particle is administered systemically. In some embodiments, the viral particle is administered intravenously. In some embodiments, the viral particle is administered intraarterially. In some embodiments, the viral particle is a lentiviral particle.
- Formulations of a pharmaceutical composition suitable for parenteral administration typically generally comprise the active ingredient combined with a pharmaceutically acceptable carrier, such as sterile water or sterile isotonic saline. Such formulations may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable formulations may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi-dose containers containing a preservative. Formulations for parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and the like. Such formulations may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents.
- the active ingredient is provided in dry (i.e. powder or granular) form for reconstitution with a suitable vehicle (e.g. sterile pyrogen-free water) prior to parenteral administration of the reconstituted composition.
- a suitable vehicle e.g. sterile pyrogen-free water
- Parenteral formulations also include aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
- Illustrative parenteral administration forms include solutions or suspensions in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
- Other parentally-administrable formulations which are useful include those which comprise the active ingredient in microcrystalline form, or in a liposomal preparation.
- Formulations for parenteral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
- the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
- these particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
- the solution intended for subcutaneous administration includes hyaluronidase.
- compositions of the present invention may additionally contain other adjunct components conventionally found in pharmaceutical compositions.
- the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
- additional materials useful in physically formulating various dosage forms of the compositions of the present invention such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers.
- such materials when added, should not unduly interfere with the biological activities of the components of the compositions of the present invention.
- the formulations can be sterilized and, if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the nucleic acid(s) of the formulation.
- auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the nucleic acid(s) of the formulation.
- the present polycistronic constructs may be administered in amounts effective to treat or prevent the disease or condition, such as a therapeutically effective or prophylactically effective amount.
- Therapeutic or prophylactic efficacy in some embodiments is monitored by periodic assessment of treated subjects. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until a desired suppression of disease symptoms occurs.
- other dosage regimens may be useful and can be determined.
- the desired dosage can be delivered by a single bolus administration of the composition, by multiple bolus administrations of the composition, or by continuous infusion administration of the composition.
- a subject in the context of infusing differentiated cells or transgenic differentiated cells according to the disclosure, is administered the range of about one million to about 100 billion cells, such as, e.g., 1 million to about 50 billion cells (e.g., about 5 million cells, about 25 million cells, about 500 million cells, about 1 billion cells, about 5 billion cells, about 20 billion cells, about 30 billion cells, about 40 billion cells, or a range defined by any two of the foregoing values), such as about 10 million to about 100 billion cells (e.g., about 20 million cells, about 30 million cells, about 40 million cells, about 60 million cells, about 70 million cells, about 80 million cells, about 90 million cells, about 10 billion cells, about 25 billion cells, about 50 billion cells, about 75 billion cells, about 90 billion cells, or a range defined by any two of the foregoing values), and in some cases about 100 million cells to about 50 billion cells (e.g., about 120 million cells, about 250 million cells, about 350 million cells, about 450 million cells,
- provided herein is a method of administering to a subject any of the cells provided herein.
- provided herein is a method of administering to a subject any of the viral vectors provided herein.
- the CAR encoded by the provided polycistronic construct is targeted to an antigen associated with a disease or condition and the methods include administering the cells (e.g. by adoptive cell therapy) or viral vector, such as lentiviral vectors, to a subject that has or is suspected of having the disease or condition.
- the methods include administering the cells (e.g. by adoptive cell therapy) or viral vector, such as lentiviral vectors, to a subject that has or is suspected of having the disease or condition.
- methods for treating a subject suffering from cancer including the step of administering a provided cell engineered with any of the provided polycistronic constructs of the disclosure to the subject, wherein the cancer is treated in the subject.
- a provide viral vector such as lentiviral particle
- a provide viral vector such as lentiviral particle
- the cancer is a solid tumor, such as a melanoma, non-small cell lung cancer, or breast cancer.
- the methods of the present disclosure may include treating any cancer, including, without limitation, acute granulocytic leukemia, acute lymphocytic leukemia, acute myelogenous leukemia, adenocarcinoma, adenosarcoma, adrenal cancer, adrenocortical carcinoma, anal cancer, anaplastic astrocytoma, angiosarcoma, appendix cancer, astrocytoma, basal cell carcinoma, B-cell lymphoma, bile duct cancer, bladder cancer, bone cancer, bone marrow cancer, bowel cancer, brain cancer, brain stem glioma, brain tumor, breast cancer, carcinoid tumors, cervical cancer, cholangiocarcinoma, chondrosarcoma, chronic lymphocytic leukemia, chronic myelogenous leukemia, colon cancer, colore
- the CAR encoded by the provided polycistronic construct is a CAR that targets a ligand that is able to bind to an antigen on the surface of a cell associated with a disease or condition.
- the CAR of any of the provided embodiments is an anti-FITC CAR directed against FITC and the ligand is a bifunctional ligand composed of FITC and a binding molecule that is able to bind to a surface molecule or receptor on the target cell.
- the method further includes administering the bifunctional ligand to tag a cancer cell in the subject, wherein the bifunctional ligand specifically binds a molecule expressed on a tumor.
- the bifunctional ligand is FITC-folate.
- the cancer is an osteosarcoma.
- the method further comprises administering a bifunctional ligand to tag a cancer cell in the subject, wherein the bifunctional ligand specifically binds a molecule expressed on a tumor.
- the bifunctional ligand comprises FITC-folate.
- the method further including administering a non- physiological ligand to the subject.
- the non-physiological ligand is able to bind to the synthetic cytokine receptor and induce gamma cytokine signaling in the cell.
- the nonphysiological ligand includes rapamycin or a rapamycin analog.
- the system comprises a non-physiological ligand.
- Illustrative small molecules useful as ligands include, without limitation: rapamycin, fluorescein, fluorescein isothiocyanate (FITC), 4-[(6-methylpyrazin-2-yl) oxy] benzoic acid (aMPOB), folate, rhodamine, acetazol amide, and a CA9 ligand.
- the synthetic cytokine receptor is activated by a ligand.
- the ligand is a non-physiological ligand.
- the non-physiological ligand is a rapalog.
- the non-physiological ligand is rapamycin.
- the non-physiological ligand is AP21967.
- the non-physiological ligand is FK506. [0416] In some embodiments, the non-physiological ligand is FK1012. In some embodiments, the non-physiological ligand is AP1510. In some embodiments, the non-physiological ligand is AP1903. In some embodiments, the non-physiological ligand is AP20187. In some embodiments, the non-physiological ligand is cyclosporin-A (CsA). In some embodiments, the non-physiological ligand is coumermycin.
- CsA cyclosporin-A
- the synthetic cytokine receptor complex activated by folate, fluorescein, aMPOB, acetazolamide, a CA9 ligand, tacrolimus, rapamycin, a rapalog (a rapamycin analog), CD28 ligand, poly(his) tag, Strep-tag, FLAG-tag, VS-tag, Myc-tag, HA-tag, NE-tag, biotin, digoxigenin, dinitrophenol, or a derivative thereof.
- the non-physiological ligand may be an inorganic or organic compound that is less than 1000 Daltons.
- the ligand may be rapamycin or a rapamycin analog (rapalog).
- the rapalog comprises variants of rapamycin having one or more of the following modifications relative to rapamycin: demethylation, elimination or replacement of the methoxy at C7, C42 and/or C29; elimination, derivatization or replacement of the hydroxy at C13, C43 and/or C28; reduction, elimination or derivatization of the ketone at Cl 4, C24 and/or C30; replacement of the 6-membered pipecolate ring with a 5-membered prolyl ring; and alternative substitution on the cyclohexyl ring or replacement of the cyclohexyl ring with a substituted cyclopentyl ring.
- the rapalog is everolimus, novolimus, pimecrolimus, ridaforolimus, tacrolimus, temsirolimus, umirolimus, zotarolimus, Temsirolimus (CCI-779), C20- methallylrapamycin, C16-(S)-3-methylindolerapamycin, C16-(S)-3-methylindolerapamycin (C16- iRap), AP21967 (A/C Heterodimerizer, Takara Bio®), sodium mycophenolic acid, benidipine hydrochloride, rapamine, AP23573 (Ridaforolimus), AP1903 (Rimiducid), or metabolites, derivatives, and/or combinations thereof.
- the ligand comprises FK1012 (a semisynthetic dimer of FK506), tacrolimus (FK506), FKCsA (a composite of FK506 and cyclosporine), rapamycin, coumermycin, gibberellin, HaXS dimerizer (chemical dimerizers of HaloTag and SNAP-tag), TMP-HTag (trimethoprim haloenzyme protein dimerizer), or ABT-737 or functional derivatives thereof.
- FK1012 a semisynthetic dimer of FK506
- tacrolimus FK506
- FKCsA a composite of FK506 and cyclosporine
- rapamycin rapamycin
- coumermycin gibberellin
- HaXS dimerizer chemical dimerizers of HaloTag and SNAP-tag
- TMP-HTag trimethoprim haloenzyme protein dimerizer
- the non-physiological ligand is present or provided in an amount from 0 nM to 1000 nM such as, e.g., 0.05 nM, 0.1 nM, 0.5.
- nM 1.0 nM, 5.0 nM, 10.0 nM, 15.0 nM, 20.0 nM, 25.0 nM, 30.0 nM, 35.0 nM, 40.0 nM, 45.0 nM, 50.0 nM, 55.0 nM, 60.0 nM, 65.0 nM, 70.0 nM, 75.0 nM, 80.0 nM, 90.0 nM, 95.0 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, or 1000 nM, or an amount that is within a range defined by any two of the aforementioned amounts.
- the non-physiological ligand is AP21967 and is present or provided at 10 nM. In some embodiments, the non-physiological ligand is AP21967 and is present or provided at 20 nM. In some embodiments, the non-physiological ligand is AP21967 and is present or provided at 50 nM. In some embodiments, the non-physiological ligand is AP21967 and is present or provided at 100 nM.
- the non-physiological ligand is rapamycin and is present or provided at 1 nM. In some embodiments, the non-physiological ligand is rapamycin and is present or provided at 10 nM. In some embodiments, the non-physiological ligand is rapamycin and is present or provided at 20 nM. In some embodiments, the non-physiological ligand is rapamycin and is present or provided at 50 nM.
- the non-physiological ligand is a rapalog and is present or provided at 1 nM. In some embodiments, the non-physiological ligand is a rapalog and is present or provided at 10 nM. In some embodiments, the non-physiological ligand is a rapalog and is present or provided at 20 nM. In some embodiments, the non-physiological ligand is a rapalog and is present or provided at 50 nM. In some embodiments, the non-physiological ligand is a rapalog and is present or provided at 100 nM.
- the non-physiological ligand is present or provided at 1 nM.
- the non-physiological ligand is present or provided at 10 nM.
- the non-physiological ligand is present or provided at 100 nM.
- the non-physiological ligand is present or provided at 1000 nM.
- an optionally substituted group means that the group is unsubstituted or is substituted.
- composition refers to any mixture of two or more products, substances, or compounds, including cells or antibodies. It may be a solution, a suspension, liquid, powder, a paste, aqueous, non-aqueous or any combination thereof.
- the preparation is generally in such form as to permit the biological activity of the active ingredient (e.g. antibody) to be effective.
- a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
- a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
- combination refers to any association between or among two or more items.
- the combination can be two or more separate items, such as two compositions or two collections, can be a mixture thereof, such as a single mixture of the two or more items, or any variation thereof.
- the elements of a combination are generally functionally associated or related.
- a kit is a packaged combination that optionally includes other elements, such as additional agents and instructions for use of the combination or elements thereof, for a purpose including, but not limited to, therapeutic uses.
- Subject refers to the recipient of polycistronic construct or other agent.
- the term includes mammal, such as primate, mouse, rat, dog, cat, cow, horse, goat, camel, sheep or a pig, preferably a human.
- Treat,” “treating” or “treatment” as used herein refers to any type of action or administration that imparts a benefit to a subject that has a disease or disorder, including improvement in the condition of the patient (z.e., improvement, reduction, or amelioration of one or more symptoms, and partial or complete response to treatment).
- an “individual” or a “subject” is a mammal.
- a “mammal” for purposes of treatment includes humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, rabbits, cattle, pigs, hamsters, gerbils, mice, ferrets, rats, cats, etc.
- the individual or subject is human.
- Polynucleotide refers to a biopolymer composed of two or more nucleotide monomers covalently bonded through ester linkages between the phosphoryl group of one nucleotide and the hydroxyl group of the sugar component of the next nucleotide in a chain.
- DNA and RNA are non-limiting examples of polynucleotides.
- Polypeptide refers to a polymer consisting of amino acid residues chained together by peptide bonds, forming part of (or the whole of) a protein.
- Nucleic acids may comprise DNA or RNA. They may be single-stranded or doublestranded. They may also be polynucleotides which include within them synthetic or modified nucleotides.
- oligonucleotides A number of different types of modification to oligonucleotides are known in the art. These include methylphosphonate and phosphorothioate backbones, addition of acridine or polylysine chains at the 3' and/or 5' ends of the molecule.
- polynucleotides may be modified by any method available in the art. Such modifications may be carried out in order to enhance the in vivo activity or life span of polynucleotides of interest.
- variant means a polynucleotide or polypeptide having at least one substitution, insertion, or deletion in its sequence compared to a reference polynucleotide or polypeptide.
- a “functional variant” is a variant that retains one or functions of the reference polynucleotide or polypeptide.
- sequence identity in relation to polynucleotides or polypeptide sequences, refers to the extent to which two optimally aligned polynucleotides or polypeptide sequences match at each position in the alignment across the full length of the reference sequence.
- the “percent identity” is the number of matched positions in the optimal alignment, divided by length of the reference sequence plus the sum of the lengths of any gaps in the reference sequence in the alignment.
- the optimal alignment is the alignment that results in the maximum percent identity. Alignment of sequences to determine percent identity can be accomplished by a number of well-known methods, including for example by using mathematical algorithms, such as, for example, those in the BLAST suite or Clustal Omega sequence analysis programs.
- sequence identity in the claims refers to sequence identity as calculated by BLAST version 2.12.0 using default parameters. And, unless noted otherwise, the alignment is an alignment of all or a portion of the polynucleotide or polypeptide sequences of interest across the full length of the reference sequence.
- small molecule refers to a low molecular weight ( ⁇ 1000 Daltons), organic compound. Small molecules may bind specific biological macromolecules and can have a variety of biological functions or applications including, but not limited to, serving as cell signaling molecules, drugs, secondary metabolites, or various other modes of action.
- analog in relation to a small molecule refers to a compound having a structure and/or function similar to that of another compound but differing from it in respect to a certain component. The analog may differ in one or more atoms, functional groups, or substructures, which are replaced with other atoms, groups, or substructures. Despite a high structural and/or functional similarity, analogs can have different physical, chemical, physiochemical, biochemical, or pharmacological properties.
- rapalog is an art-recognized group of analogs of rapamycin analog that share structural and functional similarity to rapamycin. Certain rapalogs are known to share some but not all functional attributes of rapamycin. For example, some rapalogs are suitable for uses as a non- physiological ligand because they promote dimerization but have substantially no immunosuppressive activity (e.g., AP21967, AP23102, or iRAP).
- An illustrative rapalog of the disclosure is AP23102
- cell population refers to mixture of cells suspended in solution, attached to a substrate, or stored in a container.
- the characteristics of a cell population as a whole can be studied with bulk measurements of sample volumes having a plurality of cells. Flow cytometry methods may be employed to reduce problems with background fluorescence which are encountered in bulk cell population measurements.
- the term “engineered” refers to a cell that has been stably transduced with a heterologous polynucleotide or subjected to gene editing to introduce, delete, or modify polynucleotides in the cell, or cells transiently transduced with a polynucleotide in a manner that causes a stable phenotypic change in the cell.
- a polycistronic construct comprising in 5’ to 3’ order (a) a first expression cassette comprising a nucleotide sequence encoding FRB, (b) a second expression cassette comprising a nucleotide sequence encoding a synthetic cytokine gamma chain polypeptide, (c) a third expression cassette comprising a nucleotide sequence encoding a synthetic cytokine beta chain polypeptide, and (d) a fourth expression cassette comprising a nucleotide sequence encoding a chimeric antigen receptor (CAR), wherein each of the expression cassettes are separated by a nucleotide sequence encoding a cleavage site sequence.
- CAR chimeric antigen receptor
- nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:3, 13, or 50.
- nucleotide encoding the synthetic cytokine gamma chain polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 15.
- nucleotide encoding the synthetic cytokine gamma chain polypeptide comprises the nucleotide sequence of SEQ ID NO: 15.
- IL2RG interleukin 2 receptor subunit y
- nucleotide sequence encoding the FRB is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 13.
- polycistronic construct of embodiment 11 or embodiment 12, wherein the nucleotide sequence encoding the FRB comprises the nucleotide sequence of SEQ ID NO: 13.
- nucleotide sequence encoding the FKBP12 is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:21 or 55.
- nucleotide sequence encoding the FKBP12 comprises the nucleotide sequence of SEQ ID NOs:21 or 55.
- nucleotide encoding the synthetic cytokine beta chain polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:23 or 61.
- IL2RB interleukin 2 receptor subunit P
- polycistronic construct of embodiment 31 or embodiment 32, wherein the nucleotide sequence encoding the FKBP12 comprises the nucleotide sequence of SEQ ID NO: 21.
- polycistronic construct of embodiment 48 wherein the scFv VL comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:30 or 65.
- polycistronic construct of embodiment 48 or embodiment 49, wherein the scFv VL comprises the nucleotide sequence of SEQ ID NOs:30 or 65.
- scFv linker comprises the nucleotide sequence of SEQ ID NOs:32 or 66.
- scFv linker comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:33.
- polycistronic construct of embodiment 68, wherein the CD8a hinge comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID Nos:38 or 114.
- polycistronic construct of embodiment 68 or embodiment 69, wherein the CD8a hinge comprises the nucleotide sequence of SEQ ID Nos: 38 or 114.
- transmembrane domain comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO:40.
- polycistronic construct of embodiment 82, wherein the 4- IBB endodomain comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs:42 or 69.
- polycistronic construct of embodiment 82 or embodiment 83, wherein the 4- IBB endodomain comprises the nucleotide sequence of SEQ ID NOs: 42 or 69.
- CD3 ⁇ endodomain comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NOs: 46, 70, 100 or 118.
- each of the cleavage site sequences comprises a 2A cleavable linker sequence.
- T2A cleavable linker comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NOTO.
- nucleotide sequence encoding the furin cleavage site sequence comprises a sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NOT.
- polycistronic construct of any one of embodiments 107 to 109, wherein the nucleotide sequence encoding the furin cleavage site sequence comprises the sequence set forth in SEQ ID NO: 7.
- polycistronic construct of any one of embodiments 106 to 111, wherein the furin cleavage site sequence comprises the amino acid sequence of SEQ ID NO: 8.
- cleavage site sequence comprises a furin cleavage site sequence and a T2A cleavage sequence (furinT2A).
- nucleotide sequence encoding the cleavage site sequence is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 5.
- polycistronic construct of any one of embodiments 1 to 114, wherein the nucleotide sequence encoding the cleavage site sequence comprises the nucleotide sequence of SEQ ID NO: 5.
- cleavage site sequence comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 6.
- polycistronic construct of any one of embodiments 1 to 118 wherein the construct comprises a nucleotide sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the nucleotide sequence of SEQ ID NO: 1.
- polycistronic construct of any one of embodiments 1 to 120, wherein the construct encodes a polypeptide comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO:2.
- polycistronic construct of any one of embodiments 1 to 121, wherein the construct encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 2.
- polycistronic construct of any one of embodiments 1 to 118, 123, or 124, wherein the polycistronic construct encodes a polypeptide comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to the amino acid sequence of SEQ ID NO: 49.
- polycistronic construct of any one of embodiments 1 to 118 or 123 to 125, wherein the construct encodes a polypeptide comprising the amino acid sequence of SEQ ID NO: 49.
- a viral vector comprising the polycistronic construct of any one of embodiments 1 to 126.
- the viral vector of embodiment 129, wherein the one or more surface T cell activating agents comprise CD58, anti-CD3, or CD80.
- a cell comprising the viral vector of any one of embodiments 127 to 130.
- the cell of embodiment 131, wherein the cell comprises a stem cell or a progenitor cell.
- the cell of embodiment 132, wherein the stem cell comprises an induced pluripotent stem cell (iPSC).
- iPSC induced pluripotent stem cell
- PBMC peripheral blood mononuclear cell
- 136 The cell of embodiment 131, wherein the cell comprises a cytotoxic innate lymphocyte (CIL) cell.
- CIL cytotoxic innate lymphocyte
- NK natural killer
- a method of transducing a cell comprising contacting a target cell with any of the polycistronic constructs of any of embodiments 1 to 126.
- a method of transducing a cell comprising contacting a target cell with the viral vector of any one of embodiments 127-130.
- the stem cell comprises an induced pluripotent stem cell (iPSC).
- iPSC induced pluripotent stem cell
- PBMC peripheral blood mononuclear cell
- T cell comprises a CD4+ or CD8+ T cell.
- a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a target cell is a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a target cell.
- a method of expressing a a chimeric antigen receptor and/or a synthetic cytokine receptor in a target cell comprising contacting the target cell with the viral vector of any one of embodiments 127-130.
- stem cell comprises an induced pluripotent stem cell (iPSC).
- iPSC induced pluripotent stem cell
- PBMC peripheral blood mononuclear cell
- T cell comprises a CD4+ or CD8+ T cell.
- a method of transducing a T cell comprising contacting the T cell with a viral vector comprising one or more T cell activating agents and a polycistronic construct of any one of embodiments 1-126, wherein the one or more T cell activating agents bind a receptor on the T cell.
- a method of expressing a chimeric antigen receptor and/or a synthetic cytokine receptor in a T cell comprising contacting the T cell with a viral vector comprising one or more T cell activating agents and a polycistronic construct of any one of embodiments 1-126, wherein the one or more T cell activating agents bind a receptor on the T cell.
- a method of delivering a payload to a T cell comprising contacting the T cell with a viral vector comprising one or more T cell activating agents and a polycistronic construct of any one of embodiments 1-126, wherein the one or more T cell activating agents bind a receptor on the T cell.
- T cell comprises a CD4+ or CD8+ T cell.
- bifunctional ligand comprises a fluorescein isothiocyanate (FITC) moiety and the chimeric antigen receptor (CAR) encoded by the polycistronic construct is an anti-FITC CAR.
- FITC fluorescein isothiocyanate
- CAR chimeric antigen receptor
- any one of embodiments 167 to 176 further comprising administering to the subject a non-physiological ligand, optionally wherein the non-physiological ligand binds to a synthetic cytokine receptor composed of the synthetic gamma chain polypeptide and the synthetic cytokine beta chain polypeptide encoded by the polycistronic construct.
- This example describes the generation of polycistronic constructs and their evaluation for RACR and CAR expression levels in order to elucidate the impact of the vector orientation on TagCAR activity.
- polynucleotide of the TagCAR encoded a CAR with the following components in N-terminal to C-terminal order: an scFv (e.g. anti-FITC E2), a hinge (spacer), a transmembrane domain, and an endodomain with a costimulatory signaling domain and a CD3zeta signaling domain (Z).
- an scFv e.g. anti-FITC E2
- hinge spacer
- transmembrane domain e.g. anti-FITC E2
- Z CD3zeta signaling domain
- constructs differed in the hinge (spacer) domain, either IgG4 hinge (IgG4H) or CD8alpha hinge (CD8H); transmembrane domain, either CD28 TM or CD8 TM; and costimulatory domain, either 41BB costimulatory domain or CD28 costimulatory domain.
- the constructs also differed in the placement of the TagCAR, being present either as the front or end of the construct transgene sequence.
- the individual polynucleotide components of the construct were separated by a 2A cleavage site sequence.
- lentiviral vector production 293T producer cells were transfected with plasmids expressing viral vector proteins (gag/pol, rev) and a transfer plasmid encoding each of the above payload constructs.
- the lentiviral vectors also were engineered to encode CD58, anti-CD3, and CD80 surface proteins to mediate T cell activation.
- the cell culture was centrifuged to pellet the cells and the supernatant containing crude virus was collected.
- PBMCs from 3 donors were seeded in a 24-well plate at 2xl0 6 cells/mL in media containing 50 U/mL of IL-2 and transduced with lentivirus at an MOI of 2 or 10.
- virus was washed out and cells were stained by flow cytometry for CD25 as a marker of T cell activation and also were split into separate plates and exposed to either 50 U/mL IL-2 + either 10 nM rapamycin or no rapamycin.
- cells were stained to assess CAR expression, lysed for western blot, or were used for co-culture killing and expansion assays.
- T cell activation (FIG. 3A) and T cell transduction (FIG. 3B) increased with particle dose, with higher activation and transduction occurring with MOI 10 (light bars) than MOI 2 (dark bars).
- Results in FIG. 3A also showed that T cell activation (as determined by % of CD25+ of CD8+ or CD4+ T cells) was similar across all lentiviral vector preparations.
- FIG. 3B showed that T cell transduction (as determined by % CAR expression of CD8+ and CD4+ T cells) also was similar across all lentiviral vector preparations.
- the results indicate surprisingly that the orientation of the components in the polynucleotide constructs do not impact the percentage of activated CD8+ and CD4+ T cells or the percentage of transduced CD8+ and CD4+ TagCAR T cells.
- lenti viral vectors with Construct C.2 and Construct D.2 oriented transgenes were further analyzed following transduction of T cells.
- PBMCs from 3 donors were seeded in a 24-well plate at 2xl0 6 cells/mL in media containing 50 U/mL of IL-2 and transduced with lentivirus at an MOI of 10.
- virus was washed out and cells were split into separate plates and exposed to either 50 U/mL IL-2, 10 nM rapamycin or 50 U/mL IL-2 +10 nM rapamycin.
- 11 and 14 cells were stained to assess CAR expression.
- CAR T cells transduced with either transgene were enriched and expanded with rapamycin.
- Transduced T cells from Day 10 after transduction were assayed in a killing assay.
- 6xl0 3 FRa+ MDA-MB-231 targets cells that were labeled with mCherry were plated in a 96-well plate in folic acid-free media.
- transduced CAR-T cells were added to target cells at a 1:1 or 1:4 effector to target (E:T) ratio.
- Folate-Fluorescein (FITC- folate) was added at 0, 0.1 or 10 nM to each well.
- supernatant was collected and used for analysis of cytokines.
- FIG. 5A IgG4 hinge
- FIG. 5A IgG4 hinge
- CAR T cells with a CD8alpha hinge exhibited superior activity at eliminating FRa+ target cells at low doses of the Folate-Fluorescein across all donors, regardless of prior expansion in rapamycin. More specifically, tumor object area at 96hrs decreased when co-cultured with cells transduced with the following constructs (top to bottom in FIG.
- PBMCs from 3 donors were seeded in a 24-well plate at IxlO 6 cells/mL in medium containing 250 U/mL of IL2 and transduced with lentivirus encoding the above constructs.
- cells were counted and stained for CD25 to confirm T cell activation by the lentiviral vector or cells were transferred to separate plates for stimulation with IL2 and rapamycin.
- cells were either counted again and stained to assess CAR expression, lysed for western blot, or stimulated with IL2 (250 U/mL) and rapamycin (lOnM).
- days 11, 14, and 18, cells were counted, stained to assess CAR expression or stimulated with IL2 (250 U/mL) and rapamycin (lOnM).
- PBMCs transduced with lentiviral vector containing Construct V or Construct C.2 both expressed CAR (FIG. 6A). Consistent with the results in Example 1, enhanced ERB and RACR expression was observed by Western Blot in PBMCs transduced with Construct C.2 compared to Construct V (data not shown). The enhanced expression of FRB correlated with enhanced proliferation of PBMCs transduced with Construct C.2 compared to Construct V in the presence of rapamycin in transduced cells from each donor. (FIG. 6B). These data demonstrate that cells containing Construct C.2 responded better to rapamycin stimulation compared to cells containing Construct V.
- a polynucleotide construct was generated by rearranging the orientation of the polynucleotide compared to Construct C.2. Specifically, the FRB and FKBP12 were swapped to encode a synthetic gamma chain polypeptide composed of FRB fused to!L2Ry and a synthetic beta chain polypeptide composed of FKBP12 sequence fused to IL2Rp. Further, a furin T2A linker was included in place of P2A between the first and second transgenes of the construct, namely between FRB and FRB:IL2Ry. Finally, the polynucleotide encoding the synthetic cytokine receptor (RACR) components was codon optimized. The resultant optimized polynucleotide construct is shown as Construct C.2U in FIG. 7A and SEQ ID NO: 1.
- PBMCs from five different donors were seeded in a 24-well plate at IxlO 6 cells/mL and transduced with lentivirus containing construct C.2 polynucleotide or Construct C.2U polynucleotide.
- cells were stained for CD25.
- cells were either stained to assess expression of the RACR-TagCAR (FITC-CAR).
- FITC-CAR FITC-CAR
- T cells transduced with lentivirus containing polynucleotide Construct C.2U exhibited higher detectable expression by Western Blot of FRB (FIG. 7E) and RACR (FRB-IL2Ry and FKBP12-IL2RP) (FIG. 7F).
- FIG. 8A activation and transduction of gated T cell populations was assessed by flow cytometry on day 3 and day 7 post-transduction (FIG. 8A). Following transduction, the percentage of CD8+ and CD4+ T cells as a percentage of CD25+ cells on day 3 of activation increased (FIG. 8A, left graph). Also, the percentage RACR-TagCAR+ CD4+ or CD8+ T cells increased following transduction at an MOI 10 (FIG. 8A, right graph). Representative flow plots on day 7 post-transduction show an increased percentage of CD8+ T cells express RACR- TagCAR, particularly at MOI 10 (FIG. 8B).
- PBMCs at 1 x 10 6 cells/mL in 24-well plates were transduced with lentivirus containing polynucleotide Construct C.2 or C.2U on day 0.
- cells were stimulated with IL2 (250 U/mL) or rapamycin (lOnM) and stained for CD25.
- IL2 250 U/mL
- rapamycin rapamycin
- CD25 CD25
- cells were stimulated with IL2 only, rapamycin only, AP21967 (50nM) only, or rapamycin and IL2.
- T cells transduced with Construct C.2U transgene lentiviral vector demonstrated enhanced CAR T cell expansion in the presence of IL2 or rapamycin beginning on day 3 compared to T cells transduced with Construct C.2 transgene lentiviral vector. Results from one representative donor is shown in FIG. 9A.
- Construct C.2U transgene transduced T cells also demonstrated enhanced CAR T cell expansion in the presence of rapamycin or rapamycin analog AP21967 as shown in FIG. 9B.
- This example demonstrates characterization (e.g., activation, transduction, cytotoxicity, etc.) of primary human T cells transduced with Construct C.2U transgene lentiviral vector described in Example 3.
- PBMCs from 3 healthy donors were transduced in vitro with lentivirus vectors containing Construct C.2U transgene at a multiplicity of infection (MOI) 2 or 10 in the absence of T cell activating reagents.
- MOI multiplicity of infection
- rapamycin-mediated RACR activation of RACR-TagCAR T cells was assessed.
- PBMCs from 2 healthy donors were cultured with Construct C.2U transgene lentiviral vector at MOI 10.
- PBMCs were split into two co-cultures. One co-culture contained 50U/mL of IL-2 and the other co-culture contained 50U/mL of IL-2 and 10 nM rapamycin, which were added on day 3 of co-culture, and enrichment and expansion of T cells was measured on days 7, 11 and 14 by flow cytometry (FIG. 10A).
- Rapamycin-mediated RACR activation of T cells resulted in increased enrichment of RACR-TagCAR expressing T cells across days 11 and 14 (FIG. 10B, left graph). Rapamycin-mediated RACR activation of T cells also resulted in increased expansion of RACR- TagCAR expressing T cells across days 11 and 14 (FIG. 10B, right graph).
- T cell anti-tumor activity and persistence was assessed using a co-culture approach with folate receptor (FR)-expressing MDA-MB-231 breast carcinoma cells and titrated doses of FITC- Folate.
- T cells transduced with Construct C.2U transgene lentiviral vector.
- rapamycin-expanded T cells were co-cultured with mCherry+FRa+MDA-MB-231 tumor cells in the presence of 0.1 nM FITC-Folate or 0.1 nM FITC-Folate andlOnM rapamycin.
- Breast carcinoma cells were quantified over time using the Incucyte® Live-Cell Analysis System. At time points indicated with the arrows in FIG.
- breast carcinoma cells were reintroduced (tumor re-challenge).
- RACR-TagCAR T cells repeatedly killed FRa+ MDA-MB-231 breast carcinoma cells in the presence of FITC-Folate with improved activity upon rapamycin treatment (FIG. 11A). Further, RACR-TagCAR T cell proliferation persisted across the course of the experiment and with each tumor re-challenge (FIG. 11B).
- RACR-TagCAR T cells were tested for their ability to control tumors in vivo in a mouse model receiving ex vivo transduced RACR-TagCAR T cells (FIG. 12A).
- NSG MHCI/II DKO mice were injected with FRa+ MDA-MB-231 breast carcinoma cells. Once tumors reached a size of 100 mm 3 , mice received an i.v. injection of 5e6 or 10e6 RACR-TagCAR T cells that were transduced ex vivo with CD58/CD3/CD80 surface-engineered lentiviral particles containing Construct C.2 transgene encoding FRB -RACR-TagCAR.
- mice were then dosed twice per week with FITC-Folate. Efficacy was measured by tumor regression using digital calipers and plotted as shown in FIG. 12B. Administration of both 5e6 and 10e6 RACR- TagCAR T cell numbers rapidly cleared FRa+ solid tumors in mice (FIG. 12B).
- RACR-TagCAR lentivirus particles were tested for their ability to control tumors in vivo in a mouse model. Mice were administered CD58/CD3/CD80 surface-engineered lentiviral particles containing Construct C.2U transgene encoding FRB -RACR-TagCAR vectors (FIG. 13A). Female NSG MHCI/IIDKO mice were engrafted with FRa+MDA-MB-231 tumors in the flank and tumors were allowed to grow until 100-200 mm 3 in size.
- mice were humanized with healthy human donor PBMCs (25e6 or 100e6) followed by administration of CD58/CD3/CD80 surface-engineered lentiviral particles containing Construct C.2U transgene encoding the FRB-RACR-TagCAR vectors.
- the lentivirus particles were administered at a dose of 100e6 transfecting units (TU) of TagCAR vector or 25e6 TU TagCAR vector.
- Mice were treated with FITC-Folate twice per week and efficacy was determined by assessing tumor regression and circulating RACR-TagCAR T cells in blood by flow cytometry from weekly blood draws.
- RACR- TagCAR T cells were detected in blood as early as day 7 (FIG. 13B).
- Administration of the lentiviral vector particles also controlled FRa+ solid tumors in mice as evidenced by a reduction in tumor volume (FIG. 13C).
- mice were administered CD58/CD3/CD80 surface-engineered lentiviral particles containing Construct C.2U at doses of 0.2e6 TU TagCAR vector, 1.0e6 TU TagCAR vector or 5.0e6 TU TagCAR vector. Efficacy was determined by monitoring tumor regression and circulating RACR-TagCAR T cells as described above. There was dose-dependent control of solid tumors, and mice that received 5.0e6 TU of lentiviral particles had the greatest tumor reduction by Day 25 (FIG. 14A).
- TagCAR T cells can be generated in vivo with administration of the lentiviral particles and mediate tumor control and regression with FITC-folate administration.
- the studies herein and above establish the utility of a TagCAR T cell system for ex vivo or in vivo delivery to T cells and administration of a tumor tag, which can provide utility in the context of additional antigen-targeting tumor tags.
- the present invention is not intended to be limited in scope to the particular disclosed embodiments, which are provided, for example, to illustrate various aspects of the invention. Various modifications to the compositions and methods described will become apparent from the description and teachings herein. Such variations may be practiced without departing from the true scope and spirit of the disclosure and are intended to fall within the scope of the present disclosure.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Genetics & Genomics (AREA)
- Zoology (AREA)
- Molecular Biology (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Medicinal Chemistry (AREA)
- Cell Biology (AREA)
- Toxicology (AREA)
- Gastroenterology & Hepatology (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Wood Science & Technology (AREA)
- Biomedical Technology (AREA)
- Biotechnology (AREA)
- General Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Virology (AREA)
- Physics & Mathematics (AREA)
- Plant Pathology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Medicinal Preparation (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
Description
Claims
Priority Applications (7)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| JP2025526210A JP2025538152A (en) | 2022-11-04 | 2023-11-03 | Polynucleotide constructs and related viral vectors and methods |
| KR1020257017821A KR20250120449A (en) | 2022-11-04 | 2023-11-03 | Polynucleotide constructs and related viral vectors and methods |
| EP23822165.9A EP4611799A2 (en) | 2022-11-04 | 2023-11-03 | Polynucleotide construct and related viral vectors and methods |
| IL320495A IL320495A (en) | 2022-11-04 | 2023-11-03 | Polynucleotide construct and related viral vectors and methods |
| AU2023371650A AU2023371650A1 (en) | 2022-11-04 | 2023-11-03 | Polynucleotide construct and related viral vectors and methods |
| CN202380090428.7A CN120475987A (en) | 2022-11-04 | 2023-11-03 | Polynucleotide constructs and related viral vectors and methods |
| MX2025005028A MX2025005028A (en) | 2022-11-04 | 2025-04-30 | Polynucleotide construct and related viral vectors and methods |
Applications Claiming Priority (6)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202263422920P | 2022-11-04 | 2022-11-04 | |
| US63/422,920 | 2022-11-04 | ||
| US202363449289P | 2023-03-01 | 2023-03-01 | |
| US63/449,289 | 2023-03-01 | ||
| US202363466714P | 2023-05-15 | 2023-05-15 | |
| US63/466,714 | 2023-05-15 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2024098038A2 true WO2024098038A2 (en) | 2024-05-10 |
| WO2024098038A3 WO2024098038A3 (en) | 2024-09-06 |
Family
ID=89190518
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2023/078748 Ceased WO2024098038A2 (en) | 2022-11-04 | 2023-11-03 | Polynucleotide construct and related viral vectors and methods |
Country Status (9)
| Country | Link |
|---|---|
| EP (1) | EP4611799A2 (en) |
| JP (1) | JP2025538152A (en) |
| KR (1) | KR20250120449A (en) |
| CN (1) | CN120475987A (en) |
| AU (1) | AU2023371650A1 (en) |
| IL (1) | IL320495A (en) |
| MX (1) | MX2025005028A (en) |
| TW (1) | TW202434621A (en) |
| WO (1) | WO2024098038A2 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2024238153A1 (en) * | 2023-05-15 | 2024-11-21 | Umoja Biopharma, Inc. | Lentiviral delivery of anti-cd20 chimeric antigen receptors |
Citations (19)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US5994136A (en) | 1997-12-12 | 1999-11-30 | Cell Genesys, Inc. | Method and means for producing high titer, safe, recombinant lentivirus vectors |
| US6013516A (en) | 1995-10-06 | 2000-01-11 | The Salk Institute For Biological Studies | Vector and method of use for nucleic acid delivery to non-dividing cells |
| US8093042B2 (en) | 1999-10-12 | 2012-01-10 | Institut Pasteur | Lentiviral triplex DNA, and vectors and recombinant cells containing lentiviral triplex DNA |
| WO2015057852A1 (en) | 2013-10-15 | 2015-04-23 | The California Institute For Biomedical Research | Chimeric antigen receptor t cell switches and uses thereof |
| US20160122782A1 (en) | 2014-11-05 | 2016-05-05 | Juno Therapeutics, Inc. | Methods for transduction and cell processing |
| WO2016139463A1 (en) | 2015-03-02 | 2016-09-09 | Ucl Business Plc | Retroviral and lentiviral vectors |
| US20170037369A1 (en) | 2014-04-23 | 2017-02-09 | Juno Therapeutics, Inc. | Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy |
| US20170290900A1 (en) | 2012-12-20 | 2017-10-12 | Purdue Research Foundation | Chimeric antigen receptor-expressing t cells as anti-cancer therapeutics |
| WO2018213337A1 (en) | 2017-05-15 | 2018-11-22 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Bicistronic chimeric antigen receptors and their uses |
| US20190091308A1 (en) | 2016-04-08 | 2019-03-28 | Purdue Research Foundation | Methods and compositions for car t cell therapy |
| US20190269727A1 (en) | 2015-12-28 | 2019-09-05 | Novartis Ag | Methods of making chimeric antigen receptor-expressing cells |
| US20200023009A1 (en) | 2017-02-28 | 2020-01-23 | Endocyte, Inc. | Compositions and methods for car t cell therapy |
| US20200123224A1 (en) | 2016-12-13 | 2020-04-23 | Seattle Children's Hospital (dba Seattle Children's Research Institute) | Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo |
| WO2020106992A1 (en) | 2018-11-21 | 2020-05-28 | Umoja Biopharma, Inc. | Multicistronic vector for surface engineering lentiviral particles |
| US20200216502A1 (en) | 2017-09-22 | 2020-07-09 | Centre National De La Recherche Scientifique (Cnrs) | Mutated Glycoprotein of Vesicular Stomatitis Virus |
| WO2021076788A2 (en) | 2019-10-16 | 2021-04-22 | Umoja Biopharma, Inc. | Retroviral vector for univeral receptor therapy |
| US20210163893A1 (en) | 2018-08-09 | 2021-06-03 | Juno Therapeutics, Inc. | Processes for generating engineered cells and compositions thereof |
| US20210244871A1 (en) | 2018-05-11 | 2021-08-12 | Lupagen, Inc. | Systems and methods for closed loop, real-time modifications of patient cells |
| WO2022072885A1 (en) | 2020-10-02 | 2022-04-07 | Lupagen, Inc. | Closed loop, bedside cell purification systems and methods |
Family Cites Families (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CA3206009A1 (en) * | 2021-01-27 | 2022-08-04 | Umoja Biopharma, Inc. | Lentivirus for generating cells expressing anti-cd19 chimeric antigen receptor |
| KR20240133796A (en) * | 2021-12-17 | 2024-09-04 | 우모자 바이오파마 인코포레이티드 | Cytotoxic innate lymphoid cells and uses thereof |
-
2023
- 2023-11-03 TW TW112142462A patent/TW202434621A/en unknown
- 2023-11-03 CN CN202380090428.7A patent/CN120475987A/en active Pending
- 2023-11-03 WO PCT/US2023/078748 patent/WO2024098038A2/en not_active Ceased
- 2023-11-03 IL IL320495A patent/IL320495A/en unknown
- 2023-11-03 JP JP2025526210A patent/JP2025538152A/en active Pending
- 2023-11-03 EP EP23822165.9A patent/EP4611799A2/en active Pending
- 2023-11-03 KR KR1020257017821A patent/KR20250120449A/en active Pending
- 2023-11-03 AU AU2023371650A patent/AU2023371650A1/en active Pending
-
2025
- 2025-04-30 MX MX2025005028A patent/MX2025005028A/en unknown
Patent Citations (19)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US6013516A (en) | 1995-10-06 | 2000-01-11 | The Salk Institute For Biological Studies | Vector and method of use for nucleic acid delivery to non-dividing cells |
| US5994136A (en) | 1997-12-12 | 1999-11-30 | Cell Genesys, Inc. | Method and means for producing high titer, safe, recombinant lentivirus vectors |
| US8093042B2 (en) | 1999-10-12 | 2012-01-10 | Institut Pasteur | Lentiviral triplex DNA, and vectors and recombinant cells containing lentiviral triplex DNA |
| US20170290900A1 (en) | 2012-12-20 | 2017-10-12 | Purdue Research Foundation | Chimeric antigen receptor-expressing t cells as anti-cancer therapeutics |
| WO2015057852A1 (en) | 2013-10-15 | 2015-04-23 | The California Institute For Biomedical Research | Chimeric antigen receptor t cell switches and uses thereof |
| US20170037369A1 (en) | 2014-04-23 | 2017-02-09 | Juno Therapeutics, Inc. | Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy |
| US20160122782A1 (en) | 2014-11-05 | 2016-05-05 | Juno Therapeutics, Inc. | Methods for transduction and cell processing |
| WO2016139463A1 (en) | 2015-03-02 | 2016-09-09 | Ucl Business Plc | Retroviral and lentiviral vectors |
| US20190269727A1 (en) | 2015-12-28 | 2019-09-05 | Novartis Ag | Methods of making chimeric antigen receptor-expressing cells |
| US20190091308A1 (en) | 2016-04-08 | 2019-03-28 | Purdue Research Foundation | Methods and compositions for car t cell therapy |
| US20200123224A1 (en) | 2016-12-13 | 2020-04-23 | Seattle Children's Hospital (dba Seattle Children's Research Institute) | Methods of exogenous drug activation of chemical-induced signaling complexes expressed in engineered cells in vitro and in vivo |
| US20200023009A1 (en) | 2017-02-28 | 2020-01-23 | Endocyte, Inc. | Compositions and methods for car t cell therapy |
| WO2018213337A1 (en) | 2017-05-15 | 2018-11-22 | The United States Of America, As Represented By The Secretary, Department Of Health And Human Services | Bicistronic chimeric antigen receptors and their uses |
| US20200216502A1 (en) | 2017-09-22 | 2020-07-09 | Centre National De La Recherche Scientifique (Cnrs) | Mutated Glycoprotein of Vesicular Stomatitis Virus |
| US20210244871A1 (en) | 2018-05-11 | 2021-08-12 | Lupagen, Inc. | Systems and methods for closed loop, real-time modifications of patient cells |
| US20210163893A1 (en) | 2018-08-09 | 2021-06-03 | Juno Therapeutics, Inc. | Processes for generating engineered cells and compositions thereof |
| WO2020106992A1 (en) | 2018-11-21 | 2020-05-28 | Umoja Biopharma, Inc. | Multicistronic vector for surface engineering lentiviral particles |
| WO2021076788A2 (en) | 2019-10-16 | 2021-04-22 | Umoja Biopharma, Inc. | Retroviral vector for univeral receptor therapy |
| WO2022072885A1 (en) | 2020-10-02 | 2022-04-07 | Lupagen, Inc. | Closed loop, bedside cell purification systems and methods |
Non-Patent Citations (31)
| Title |
|---|
| ABOU-EL-ENEIN, M. ET AL., BLOOD CANCER DISCOV, vol. 2, no. 5, 2021, pages 408 - 422 |
| ALTVATER BLANDMEIER SPSCHERER S ET AL., CLIN CANCER RES., vol. 15, 2009, pages 4857 - 4866 |
| ALVAREZ-VALLINA, L. ET AL., EUR J IMMUNOL., vol. 26, no. 10, 1996, pages 2304 - 9 |
| ARCANGELI, S. ET AL., FRONT. IMMUNOL, vol. 11, no. 1217, 19 June 2020 (2020-06-19), pages 1 - 13 |
| CHU JDENG YBENSON DM ET AL., LEUKEMIA, vol. 28, 2014, pages 917 - 927 |
| COFFIN ET AL.: "Retroviruses", 1997, COLD SPRING HARBOR LABORATORY PRESS, pages: 758 - 763 |
| GAERERTS ET AL.: "Comparison of retroviral vector titration methods", BMC BIOTECHNOL., vol. 6, 2006, pages 34, XP021017061, DOI: 10.1186/1472-6750-6-34 |
| GHASSEMI, S. ET AL., NAT BIOMED ENG, vol. 6, no. 2, February 2022 (2022-02-01), pages 118 - 128 |
| HAN JCHU JKEUNG CW ET AL., SCI REP., vol. 5, 2015, pages 11483 |
| HENNIG ET AL., INTERNATIONAL JOURNAL OF CANCER, vol. 61, pages 786 - 792 |
| HUMBERT ET AL.: "Development of third-generation Cocal Envelope Producer Cell Lines for Robust Retroviral Gene Transfer into Hematopoietic Stem Cells and T-cells", MOLECULAR THERAPY, vol. 24, 2016, pages 1237 - 1246, XP055659688, DOI: 10.1038/mt.2016.70 |
| IMAI, C. ET AL., LEUKEMIA, vol. 18, 2004, pages 676 - 84 |
| JOGLEKAR ET AL., HUMAN GENE THERAPY METHODS, vol. 28, 2017, pages 291 - 301 |
| KLOSS SOBERSCHMIDT OMORGAN M ET AL., HUM GENE THER., vol. 28, 2017, pages 897 - 913 |
| KOLMAR H. ET AL., THE FEES JOURNAL, vol. 275, no. 11, 2008, pages 26684 - 90 |
| KRUSCHINSKI AMOOSMANN APOSCHKE I ET AL., PROC NATL ACAD SCI U S A, vol. 105, 2008, pages 17481 - 17486 |
| KULARATNE, S.A. ET AL., MOL PHARM., vol. 6, no. 3, 2009, pages 780 - 9 |
| LATZA, U. ET AL., EUR. J. IMMUNOL., vol. 24, 1994, pages 677 |
| LI YHERMANSON DLMORIARITY BSKAUFMAN DS, CELL STEM CELL, vol. 23, 2018, pages 181 - 192 |
| LI YHERMANSON DLMORIARITY BSKAUFMAN DS, CELL STEM CELL., vol. 23, 2018, pages 181 - 192 |
| LIU ETONG YDOTTI G ET AL., LEUKEMIA., vol. 32, 2018, pages 520 - 531 |
| MULLER NMICHEN STIETZE S ET AL., J IMMUNOTHER., vol. 38, 2015, pages 197 - 210 |
| NALDINI ET AL., SCIENCE, vol. 272, 1996, pages 263 - 7 |
| NALDINI ET AL., SCIENCE, vol. 272, pages 263 - 7 |
| NICHOLSON ET AL., MOL. IMMUN., vol. 34, no. 16-17, 1997, pages 1157 - 1165 |
| REICHERT, J.M., MABS, vol. 1, no. 3, 2009, pages 190 - 209 |
| SEGA, E.I. ET AL., CANCER METASTASIS REV., vol. 27, no. 4, 2008, pages 655 - 64 |
| VORMITTAG, P. ET AL., CURR OPIN BIOTECHNOL, vol. 54, October 2018 (2018-10-01), pages 164 - 181 |
| WAYUA. C. ET AL., MOLECULAR PHARMACEUTICS, 2013 |
| XU YLIU QZHONG M ET AL., JHEMATOL ONCOL., vol. 12, 2019, pages 49 |
| ZUFFEREY ET AL., J. VIROL., vol. 72, 1998, pages 8150 - 8471 |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2024238153A1 (en) * | 2023-05-15 | 2024-11-21 | Umoja Biopharma, Inc. | Lentiviral delivery of anti-cd20 chimeric antigen receptors |
Also Published As
| Publication number | Publication date |
|---|---|
| KR20250120449A (en) | 2025-08-08 |
| JP2025538152A (en) | 2025-11-26 |
| EP4611799A2 (en) | 2025-09-10 |
| IL320495A (en) | 2025-06-01 |
| WO2024098038A3 (en) | 2024-09-06 |
| AU2023371650A1 (en) | 2025-05-29 |
| TW202434621A (en) | 2024-09-01 |
| CN120475987A (en) | 2025-08-12 |
| MX2025005028A (en) | 2025-07-01 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US20250122532A1 (en) | Viral vectors and packaging cell lines | |
| CN109952309B (en) | Constitutive active cytokine receptors for cell therapy | |
| US10457729B2 (en) | Compositions and methods for treating cancer | |
| US12187808B2 (en) | GD2-based chimeric antigen receptor and application thereof | |
| US20250051801A1 (en) | Viral particle with surface stimulating molecules | |
| KR20190101979A (en) | Synthetic immune receptors and methods of use thereof | |
| TW202242121A (en) | Use of cd8-targeted viral vectors | |
| BR112019018288A2 (en) | METHODS AND COMPOSITIONS FOR TRANSDUCING AND EXPANDING LYMPHOCYTES AND REGULATING THE SAME ACTIVITY | |
| CN116745427A (en) | Vector systems for delivery of various polynucleotides and their uses | |
| TW202321457A (en) | Use of cd4-targeted viral vectors | |
| US20230272039A1 (en) | Gated adapter targeting receptor | |
| TW202434735A (en) | Particles displaying adhesion-molecule fusions | |
| WO2024098038A2 (en) | Polynucleotide construct and related viral vectors and methods | |
| WO2024213153A1 (en) | Vector and use thereof | |
| KR20250121979A (en) | Particles displaying adhesive molecule fusions | |
| WO2025176853A1 (en) | Cd4- or cd8-targeted retroviral vector particles for generation of cells expressing a bispecific chimeric antigen receptor | |
| WO2025021173A1 (en) | Viral vector and use of tcr subunit | |
| CN120464576A (en) | A controllable cell transformation scalpel |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| WWE | Wipo information: entry into national phase |
Ref document number: 320495 Country of ref document: IL |
|
| WWE | Wipo information: entry into national phase |
Ref document number: MX/A/2025/005028 Country of ref document: MX |
|
| ENP | Entry into the national phase |
Ref document number: 2025526210 Country of ref document: JP Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2025526210 Country of ref document: JP |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 821393 Country of ref document: NZ Ref document number: AU2023371650 Country of ref document: AU |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 202517051415 Country of ref document: IN |
|
| ENP | Entry into the national phase |
Ref document number: 2023371650 Country of ref document: AU Date of ref document: 20231103 Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 11202502741U Country of ref document: SG |
|
| WWP | Wipo information: published in national office |
Ref document number: 821393 Country of ref document: NZ Ref document number: 11202502741U Country of ref document: SG |
|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 23822165 Country of ref document: EP Kind code of ref document: A2 |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2023822165 Country of ref document: EP Ref document number: 2025115053 Country of ref document: RU |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| ENP | Entry into the national phase |
Ref document number: 2023822165 Country of ref document: EP Effective date: 20250604 |
|
| REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112025008662 Country of ref document: BR |
|
| WWP | Wipo information: published in national office |
Ref document number: MX/A/2025/005028 Country of ref document: MX |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 202380090428.7 Country of ref document: CN |
|
| WWP | Wipo information: published in national office |
Ref document number: 1020257017821 Country of ref document: KR |
|
| WWP | Wipo information: published in national office |
Ref document number: 202380090428.7 Country of ref document: CN |
|
| WWP | Wipo information: published in national office |
Ref document number: 202517051415 Country of ref document: IN |
|
| WWP | Wipo information: published in national office |
Ref document number: 2025115053 Country of ref document: RU |
|
| WWP | Wipo information: published in national office |
Ref document number: 2023822165 Country of ref document: EP |