WO2025088378A2 - Compositions et méthodes pour traiter le cancer à l'aide de vecteurs viraux chimériques - Google Patents
Compositions et méthodes pour traiter le cancer à l'aide de vecteurs viraux chimériques Download PDFInfo
- Publication number
- WO2025088378A2 WO2025088378A2 PCT/IB2024/000596 IB2024000596W WO2025088378A2 WO 2025088378 A2 WO2025088378 A2 WO 2025088378A2 IB 2024000596 W IB2024000596 W IB 2024000596W WO 2025088378 A2 WO2025088378 A2 WO 2025088378A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- adenoviral vector
- composition
- recombinant adenoviral
- epitope
- protein
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K35/00—Medicinal preparations containing materials or reaction products thereof with undetermined constitution
- A61K35/66—Microorganisms or materials therefrom
- A61K35/76—Viruses; Subviral particles; Bacteriophages
- A61K35/761—Adenovirus
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/12—Viral antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/51—Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
- A61K2039/525—Virus
- A61K2039/5256—Virus expressing foreign proteins
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/57—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
- A61K2039/572—Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/58—Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
- A61K2039/585—Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
-
- 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
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/10011—Adenoviridae
- C12N2710/10311—Mastadenovirus, e.g. human or simian adenoviruses
- C12N2710/10322—New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2710/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
- C12N2710/00011—Details
- C12N2710/10011—Adenoviridae
- C12N2710/10311—Mastadenovirus, e.g. human or simian adenoviruses
- C12N2710/10341—Use of virus, viral particle or viral elements as a vector
- C12N2710/10343—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
- C12N2810/00—Vectors comprising a targeting moiety
- C12N2810/50—Vectors comprising as targeting moiety peptide derived from defined protein
- C12N2810/60—Vectors comprising as targeting moiety peptide derived from defined protein from viruses
- C12N2810/6009—Vectors comprising as targeting moiety peptide derived from defined protein from viruses dsDNA viruses
- C12N2810/6018—Adenoviridae
Definitions
- Viral vectors are critical tools for delivering genetic material to cells and have been instrumental in furthering methods of gene therapy and vaccines.
- Adenoviral vectors are useful for transducing multiple tissue types and can deliver larger payloads to target cells compared to other viral vectors.
- Chimeric (e.g., hybrid) viral vectors may demonstrate greater stability in vivo and enhanced long-term transgene expression.
- compositions and methods described herein can be used for enhanced efficiency and long-term expression of genetic payload, e.g., a single cancer-specific CD8+ and/or CD4+ epitope.
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) a deletion or a functional deletion in the El gene; (ii) a deletion or a functional deletion in the E3 gene; and (iii) a sequence encoding a single CD8+ T-cell epitope inserted in the El region; wherein the recombinant adenoviral vector comprises a nucleotide sequence comprising nucleic acid sequences of at least two adenovirus serotypes.
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) a deletion or a functional deletion in an El gene; (ii) a deletion or a functional deletion in an E3 gene; and (iii) a sequence encoding a single CD8+ T-cell epitope inserted in a region of the El gene wherein the recombinant adenoviral vector comprises a nucleotide sequence comprising nucleic acid sequences of at least two adenovirus serotypes.
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an adenovirus serotype 3 (Ad3) penton protein; (iii) an Ad3 hexon protein; and (iv) an Ad3 fiber protein comprising a tail portion, a shaft portion, and a knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad3 Ad3 hexon protein
- Ad3 fiber protein comprising a tail portion, a shaft portion, and a knob portion
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) an Ad5 hexon protein; and (iv) an adenovirus serotype 3 (Ad3) fiber protein comprising a tail portion, a shaft portion, and a knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad5 penton protein e.g., Ad5 hexon protein
- Ad3 adenovirus serotype 3
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) an Ad5 hexon protein; and (iv) a chimeric fiber protein comprising an Ad5 tail portion, an adenovirus serotype 3 (Ad3) shaft portion, and an Ad3 knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T- cell epitope.
- Ad5 adenovirus serotype 5
- Ad3 Ad3
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) an Ad5 hexon protein; and (iv) a chimeric fiber protein comprising an Ad5 tail portion, an adenovirus serotype 35 (Ad35) shaft portion, and an Ad35 knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad35 Ad35
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) a chimeric hexon protein wherein in the chimeric hexon protein hypervariable region sequences HVR1 to HVR7 of Ad5 have been replaced with those of adenovirus serotype 48 (Ad48); and (iii) a deletion or a functional deletion in an El gene; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad48 adenovirus serotype 48
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) a chimeric hexon protein wherein in the chimeric hexon protein hypervariable region sequences HVR1 to HVR7 of Ad5 have been replaced with those of adenovirus serotype 48 (Ad48); and (iv) an Ad5 fiber protein comprising a tail portion, a shaft portion, and a knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad5 penton protein a chimeric hexon protein wherein in the chimeric
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) a chimeric hexon protein wherein in the chimeric hexon protein hypervariable region sequences HVR1 to HVR7 of Ad5 have been replaced with those of adenovirus serotype 48 (Ad48); and (iv) an Ad3 fiber protein comprising a tail portion, a shaft portion, and a knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad5 penton protein a chimeric hexon protein wherein in the chimeric
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) a chimeric hexon protein wherein in the chimeric hexon protein hypervariable region sequences HVR1 to HVR7 of Ad5 have been replaced with those of adenovirus serotype 48 (Ad48); and (iv) a chimeric fiber protein comprising an Ad5 tail portion, an adenovirus serotype 3 (Ad3) shaft portion, and an Ad3 knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad5 penton protein a
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) a chimeric hexon protein wherein in the chimeric hexon protein hypervariable region sequences HVR1 to HVR7 of Ad5 have been replaced with those of adenovirus serotype 48 (Ad48); and (iv) a chimeric fiber protein comprising an Ad5 tail portion, an adenovirus serotype 35 (Ad35) shaft portion, and an Ad35 knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad5 penton protein
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) an adenovirus serotype 3 (Ad3) hexon protein; and (iv) an Ad3 fiber protein comprising a tail portion, a shaft portion, and a knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad3 Ad3 fiber protein
- a composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) an adenovirus serotype 3 (Ad3) hexon protein; and (iv) a chimeric fiber protein comprising an Ad5 tail portion, an Ad3 shaft portion, and an Ad3 knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- Ad5 adenovirus serotype 5
- Ad3 Ad3 hexon protein
- a chimeric fiber protein comprising an Ad5 tail portion, an Ad3 shaft portion, and an Ad3 knob portion
- the recombinant adenoviral vector further comprises
- composition comprising a recombinant adenoviral vector, wherein the recombinant adenoviral vector comprises: (i) an adenovirus serotype 5 (Ad5) nucleic acid backbone; (ii) an Ad5 penton protein; (iii) an adenovirus serotype 3 (Ad3) hexon protein; and (iv) a chimeric fiber protein comprising an Ad5 tail portion, an adenovirus serotype 35 (Ad35) shaft portion, and an Ad35 knob portion; wherein the recombinant adenoviral vector further comprises a transgene insertion at an El gene locus, and wherein the transgene comprises a sequence encoding a single CD8+ T-cell epitope.
- the adenovirus serotype 3 comprises a simian adenovirus serotype 3 (SAd3).
- composition comprising a recombinant adenoviral vector, wherein a backbone of the recombinant adenoviral vector is an adenovirus serotype 5 (Ad5) nucleic acid backbone, wherein the recombinant adenoviral vector comprises a deletion of an El gene at an El gene locus, wherein the vector further comprises a transgene insertion at the El gene locus, and wherein the transgene insertion comprises a sequence encoding a single cancerspecific CD8+ T cell epitope.
- Ad5 adenovirus serotype 5
- the recombinant adenoviral vector is a recombinant chimeric adenovirus.
- the recombinant adenoviral vector comprises a nucleotide sequence comprising nucleic acid sequences of at least three adenovirus serotypes.
- the recombinant adenoviral vector comprises a sequence encoding a capsid comprising: (i) a hexon protein; (ii) a penton protein; and (iii) a fiber protein comprising (A) a tail portion, (B) a shaft portion, and (C) a knob portion; and wherein the shaft portion is from an adenovirus serotype 3 (Ad3) fiber protein or an adenovirus serotype 35 (Ad35) fiber protein, and/or the knob portion is from an Ad3 fiber protein or an Ad35 fiber protein.
- a capsid comprising: (i) a hexon protein; (ii) a penton protein; and (iii) a fiber protein comprising (A) a tail portion, (B) a shaft portion, and (C) a knob portion; and wherein the shaft portion is from an adenovirus serotype 3 (Ad3) fiber protein or an adenovirus serotype 35 (Ad
- the transgene insertion comprises at least one recombination site.
- at least one of the recombination sites is an att site or a lox site.
- at least one of the recombination sites is an att site.
- the composition described herein comprises a first recombination site and a second recombination site.
- the first recombination site and the second recombination site do not recombine with each other.
- the recombinant adenoviral vector is notan oncolytic adenoviral vector.
- the recombinant adenoviral vector does not comprise an arginine- glycine-aspartate (RGD) peptide.
- the Ad3 fiberprotein comprises a simian Ad3 (SAd3)fiberprotein.
- the shaft portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 65.
- the shaft portion comprises an amino acid sequence as set forth in SEQ ID NO: 65.
- the knob portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 66.
- the knob portion comprisesan amino acid sequence as set forth in SEQ ID NO: 66.
- the tail portion is from an adenovirus serotype 3 (Ad3) fiber protein.
- Ad3 adenovirus serotype 3
- the tail portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 64.
- the tail portion comprises an amino acid sequence as set forth in SEQ ID NO: 64.
- the penton protein is from an adenovirus serotype 3 (Ad3) penton protein.
- the Ad3 penton protein is a simian Ad3 (SAd3) penton protein.
- the hexon protein is from an adenovirus serotype 3 (Ad3) hexon protein.
- the Ad3 hexon protein is a simian Ad3 (SAd3) hexon protein.
- the Ad3 fiber protein is a simian Ad3 (SAd3) fiber protein.
- the shaft portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 53.
- the shaft portion comprises an amino acid sequence as set forth in SEQ ID NO: 53.
- the knob portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 54.
- the knob portion comprises an amino acid sequence as set forth in SEQ ID NO: 54.
- recombinant adenoviral vector is a replication-deficient adenoviral vector.
- the recombinant adenoviral vector comprises a deletion of an E3 gene at an E3 gene locus.
- the transgene insertion further comprises a CMV promoter.
- composition comprising a recombinant adenoviral vector, wherein a backbone of the recombinant adenoviral vector is an adenovirus serotype 5 (Ad5) nucleic acid backbone, wherein the recombinant adenoviral vector comprises a sequence encoding a capsid comprising: (i) a hexon protein, (ii) a penton protein, and (iii) a fiber protein comprising (A) a tail portion, (B) a shaft portion, and (C) a knob portion; and wherein the shaft portion is from an adenovirus serotype 3 (Ad3) fiber protein or an adenovirus serotype 35 (Ad35) fiber protein, and/ortheknob portionisfrom an Ad3 fiberproteinoran Ad35 fiberprotein; and wherein the recombinant adenoviral vector further comprises a transgene insertion, and wherein the transgene
- the transgene insertion is at the El gene locus.
- the transgene insertion comprises at least one recombination site.
- at least one of the recombination sites is an att site or a lox site.
- at least one of the recombination sites is an att site.
- the composition described herein comprises a first recombination site and a second recombination site.
- the first recombination site and the second recombination site do not recombine with each other.
- the recombinant adenoviral vector is notan oncolytic adenoviral vector.
- the recombinant adenoviral vector does not comprise an arginine- glycine-aspartate (RGD) peptide.
- the Ad3 fiber protein comprises a simian Ad3 (SAd3) fiber protein.
- the shaft portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 65.
- the shaft portion comprises an amino acid sequence as set forth in SEQ ID NO: 65.
- the knob portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 66.
- the knob portion comprises an amino acid sequence as set forth in SEQ ID NO: 66.
- the tail portion is from an adenovirus serotype 3 (Ad3) fiber protein.
- Ad3 adenovirus serotype 3
- the tail portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 64.
- the tail portion comprises an amino acid sequence as set forth in SEQ ID NO: 64.
- the penton protein is from an adenovirus serotype 3 (Ad3) penton protein.
- the Ad3 penton protein is a simian Ad3 (SAd3) penton protein.
- the hexon protein is from an adenovirus serotype 3 (Ad3) hexon protein.
- the Ad3 hexon protein is a simian Ad3 (SAd3) hexon protein.
- the shaft portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 53.
- the shaft portion comprises an amino acid sequence as set forth in SEQ ID NO: 53.
- the knob portion comprises an amino acid sequence with 97-99% sequence identity to an amino acid sequence as set forth in SEQ ID NO: 54.
- the knob portion comprises an amino acid sequence as set forth in SEQ ID NO: 54.
- recombinant adenoviral vector is a replication-deficient adenoviral vector.
- the recombinant adenoviral vector comprises a deletion of an E3 gene at an E3 gene locus.
- the transgene insertion further comprises a CMV promoter.
- composition comprising a recombinant adenoviral vector, wherein a backbone of the recombinant adenoviral vector is an adenovirus serotype 5 (Ad5) nucleic acid backbone, wherein the recombinant adenoviral vector comprises a sequence encoding a capsid comprising a chimeric hexon protein, wherein the chimeric hexonprotein comprise seven hypervariable region sequences HVR1 to HVR7, wherein the seven hypervariable region sequences HVR1 to HVR7 of Ad5 have been replaced by the seven hypervariable region sequences HVR1 to HVR7 of adenovirus serotype 48 (Ad48), wherein the sequences between the HVR sequences of the chimeric hexon protein are from Ad5, wherein the recombinant adenoviral vector further comprises a transgene insertion, and wherein the transgene insertion comprises a sequence
- the recombinant adenoviral vector comprises a deletion of an El gene at an El gene locus.
- the transgene insertion is at the El gene locus.
- the transgene insertion comprises at least one recombination site.
- at least one of the recombination sites is an att site or a lox site.
- at least one of the recombination sites is an att site.
- the composition described herein comprises a first recombination site and a second recombination site.
- the first recombination site and the second recombination site do not recombine with each other.
- the recombinant adenoviral vector is not an oncolytic adenoviral vector.
- the recombinant adenoviral vector does not comprise an arginine- glycine-aspartate (RGD) peptide.
- recombinant adenoviral vector is a replication-deficient adenoviral vector.
- the recombinant adenoviral vector comprises a deletion of an E3 gene at an E3 gene locus.
- the transgene insertion further comprises a CMV promoter.
- the chimeric hexon protein comprises a HVR1 sequence of SEQ ID NO: 55, a HVR2 sequence of SEQ ID NO: 56, a HVR3 sequence of SEQ ID NO: 57, a HVR4 sequence of SEQ ID NO: 58, a HVR5 sequence of SEQ ID NO: 59, a HVR6 sequence of SEQ ID NO: 60, and a HVR7 sequence of SEQ ID NO: 61.
- composition comprising a recomb inant adenoviral vector, wherein a backbone of the recombinant adenoviral vector is an adenovirus serotype 5 (Ad5) nucleic acid backbone, wherein the recombinant adenoviral vector comprises a sequence encoding a capsid comprising: (i) a hexon protein, (ii) a penton protein, and (iii) a fiber protein comprising (A) a tail portion, (B) a shaft portion, and (C) a knob portion; and wherein the knob portion is from a simian adenovirus serotype 3 (SAd3) having an amino acid sequence which is at least 98% identical to the amino acid sequence of SEQ ID NO. : 66.
- SAd3 simian adenovirus serotype 3
- the recombinant adenoviral vector further comprises a transgene insertion, and wherein the transgene insertion comprises a sequence encoding a single cancerspecific CD8+ T cell epitope.
- the shaft portion is from a simian adenovirus serotype 3 (SAd3) having an amino acid sequence which is at least 98% identical to the amino acid sequence of SEQ ID NO.: 65.
- SAd3 simian adenovirus serotype 3
- the tail portion is from a simian adenovirus serotype 3 (SAd3) having an amino acid sequence which is at least 98% identical to the amino acid sequence of SEQ ID NO.: 64.
- SAd3 simian adenovirus serotype 3
- the recombinant adenoviral vector comprises a deletion of an El gene at an El gene locus.
- the transgene insertion comprises at least one recombination site.
- at least one of the recombination sites is an att site or a lox site.
- at least one of the recombination sites is an att site.
- the composition described herein comprises a first recombination site and a second recombination site.
- the first recombination site and the second recombination site do not recombine with each other.
- the recombinant adenoviral vector is not an oncolytic adenoviral vector.
- the recombinant adenoviral vector does not comprise an arginine- glycine-aspartate (RGD) peptide.
- recombinant adenoviral vector is a replication-deficient adenoviral vector.
- the recombinant adenoviral vector comprises a deletion of an E3 gene at an E3 gene locus.
- the transgene insertion further comprises a CMV promoter.
- composition comprising a recombinant adenoviral vector, wherein a backbone of the recombinant adenoviral vector is an adenovirus serotype 5 (Ad5) nucleic acid backbone, wherein the recombinant adenoviral vector comprises a sequence encoding a capsid comprising: (i) a hexon protein, (ii) a penton protein, and (iii) a fiber protein comprising (A) a tail portion, (B) a shaft portion, and (C) a knob portion; and wherein the hexon protein is from a simian adenovirus serotype 3 (SAd3) having an amino acid sequence which is at least 98% identical to the amino acid sequence of SEQ ID NO.: 68.
- SAd3 simian adenovirus serotype 3
- the recombinant adenoviral vector further comprises a transgene insertion, and wherein the transgene insertion comprises a sequence encoding a single cancerspecific CD8+ T cell epitope.
- the penton portion is from a simian adenovirus serotype 3 (SAd3) having an amino acid sequence which is at least 98% identical to the amino acid sequence of SEQ ID NO.: 67.
- SAd3 simian adenovirus serotype 3
- composition comprising a recombinant adenoviral vector, wherein a backbone of the recombinant adenoviral vector is an adenovirus serotype 5 (Ad5) nucleic acid backbone, wherein the recombinant adenoviral vector comprises a deletion of an El gene at an El gene locus, wherein the vector further comprises a transgene insertion at the El gene locus, wherein the transgene insertion comprises a sequence encoding a single cancerspecific CD8+ T cell epitope, and wherein the single cancer-specific CD8+ T cell epitope comprises a sequence as set forth in SEQ ID NO: 4.
- Ad5 adenovirus serotype 5
- composition comprising a recombinant chimeric adenoviral vector, wherein the recombinant chimeric adenoviral vector comprises a transgene insertion comprising a sequence encoding a single cancer-specific CD8+T cell epitope as setforth in SEQ ID NO: 4, and wherein the recombinant chimeric adenoviral vector comprises a functional deletion of a gene selected from the group consisting of El, E2, E3, and E4.
- the recombinant chimeric adenoviral vector comprises at least two adenovirus serotypes selected from the group consisting of adenovirus serotype 5 (Ad5), adenovirus serotype 3 (Ad3), adenovirus serotype 35 (Ad35), and adenovirus serotype 48 (Ad48).
- the adenovirus serotype 3 comprises a human adenovirus serotype 3 (HuAd3).
- the adenovirus serotype 3 comprises a simian adenovirus serotype 3 (SAd3).
- the recombinant chimeric adenoviral vector comprises a functional deletion of two or more genes selected from the group consisting of El, E2, E3, and E4.
- a pharmaceutical composition comprising a recombinant adenoviral vector described herein and a pharmaceutically acceptable carrier, diluent, adjuvant, or excipient.
- provided herein is a method of inducing an immune response in a mammal, comprising administering a therapeutically effective amount of the recombinant adenoviral vector described herein or the pharmaceutical composition described herein to a subject in need thereof.
- a method of treating or preventing a cancer comprising administering a therapeutically effective amount of the recombinant adenoviral vector described herein or the pharmaceutical composition described herein to a subject in need thereof.
- the cancer is selected from an esophageal cancer, colorectal cancer, prostate cancer, liver cancer, renal cancer, lung cancer, bladder cancer, breast cancer, pancreatic cancer, brain cancer, hepatocellular cancer, lymphoma, leukemia, gastric cancer, cervical cancer, ovarian cancer, thyroid cancer, melanoma, carcinoma, head and neck cancer, glioblastoma, skin cancer, nasopharyngeal cancer, Epstein Barr driven cancers, Human Papilloma virus driven cancers and soft tissue sarcoma.
- an esophageal cancer colorectal cancer
- prostate cancer liver cancer
- renal cancer lung cancer
- bladder cancer breast cancer
- pancreatic cancer brain cancer
- hepatocellular cancer lymphoma
- leukemia gastric cancer
- cervical cancer cervical cancer
- ovarian cancer thyroid cancer
- melanoma carcinoma
- head and neck cancer glioblastoma
- Epstein Barr driven cancers Epstein Barr driven cancers
- the vector or pharmaceutical composition is administered intravenously or intramuscularly.
- the vector or pharmaceutical composition is administered as a single dose. [0123] In some embodiments, the vector or pharmaceutical composition is administered as multiple doses.
- the vector or pharmaceutical composition is administered in two doses comprising a first dose and a second dose.
- the first dose is a prime dose.
- the first dose elicits a measurable immune response in a subject as compared to the immune response in the subject in the absence of administration of the first dose.
- the seconddose is abooster dose. In some embodiments, the second dose elicits a measurable immune response in a subject as compared to the immune response in the subject in the absence of administration of the second dose.
- the second dose is administered 24 weeks after the first dose.
- the second dose elicit an immune response that is at least 2 fold greater than the immune response elicited by the first dose.
- the vector or pharmaceutical composition is administered as an additional booster dose.
- the additional booster dose is administered annually after administering the first booster dose.
- the vector or pharmaceutical composition is administered prophylactically to the subject.
- the vector or pharmaceutical composition is administered in combination with a second therapeutic agent.
- the second therapeutic agent is selected from an immune checkpoint inhibitor, a chemotherapeutic agent, a small molecule inhibitor, and radiotherapy. [0135] In some embodiments, the second therapeutic agent is a vaccine.
- the immune check point inhibitor is an inhibitor of an immune checkpoint protein selected from the group consisting of CTLA-4, PD-1 , PD-L1 , PD-L2, TIM3, LAG -3, B7-H3, B7-H4, B7-H6, A2aR, BTLA, GAL9 and IDO.
- a pharmaceutical composition comprising at least a first composition described herein and a second composition described herein.
- the recombinant viral vector comprises one or more of a promoter, a translation initiating sequence, a start codon, a T cell epitope codon-optimized sequence, a stop codon, and a polyadenylation sequence, wherein the promoter is selected from a CMV promoter, an RSV promoter, and an EFla promoter.
- provided herein is a method of treating or preventing an infectious disease comprising administering a therapeutically effective amount of the recombinant adenoviral vector described herein or the pharmaceutical composition described herein to a subject in need thereof.
- a method of treating or preventing an autoimmune disease comprising administering a therapeutically effective amount of the recombinant adenoviral vector described herein or the pharmaceutical composition described herein to a subject in need thereof.
- FIG. 1 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises a SAd3 penton protein, a SAd3 hexon protein, and a SAd3 fiber protein (version 1).
- FIG. 2 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises a SAd3 fiber protein (version 2).
- FIG. 3 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises a tail portion of an Ad5 fiber protein, a shaft portion of a SAd3 fiber protein, and a knob portion of a SAd3 fiber protein (version 3).
- FIG. 4 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises a tail portion of an Ad5 fiber protein, a shaft portion of a Ad35 fiber protein, and a knob portion of a Ad35 fiber protein (version 4).
- FIG. 5 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises hypervariable loops 1-7 (HVR1-HVR7) of Ad48 (version 5).
- FIG. 6 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises hypervariable loops 1-7 (HVR1-HVR7) of Ad48 and a SAd3 fiber protein (version 6).
- FIG. 7 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises hypervariable loops 1 -7 (HVR1 -HVR7) of Ad48, a tail portion of an Ad5 fiber protein, a shaft portion of a SAd3 fiber protein, and a knob portion of a SAd3 fiber protein (version 7).
- FIG. 8 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises hypervariable loops 1 -7 (HVR1 -HVR7) of Ad48, a tail portion of an Ad5 fiber protein, a shaft portion of a Ad35 fiber protein, and a knob portion of a Ad35 fiber protein (version 8).
- FIG. 9 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises a SAd3 hexon protein, and a SAd3 fiber protein (version 9).
- FIG. 10 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises a SAd3 hexon protein, a tail portion of an Ad5 fiber protein, a shaft portion of a SAd3 fiber protein, and a knob portion of a SAd3 fiber protein (version 10).
- FIG. 11 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises a SAd3 hexon protein, a tail portion of an Ad5 fiber protein, a shaft portion of a Ad35 fiber protein, and a knob portion of a Ad35 fiber protein (version 11).
- FIG. 12 shows an exemplary configuration of a recombinant adenoviral vector construct.
- the construct comprises a SAd3 hexon protein (version 12).
- FIG. 13 shows assessment of tropism of selected chimeric viral vectors disclosed herein.
- Human embryonic kidney 293 cells infected with version 1-GFP (top left), version 2-GFP (top second from left), version 3 -GFP (top middle), version4-GFP (top second from right), and version 5-GFP (top right) chimeric vectors express GFP at day 2 or day 7 post-infection.
- Wild type Adhu5- GFP vector and uninfected cells were used as positive and negative cells respectively.
- FIGs. 14A-14AP show the recombinant vectors retained their tropism for human cells.
- GFP protein coding sequence was cloned into version2, version 3 , version 4, and version 5 vectors and they were added to A549 (ECACC 86012804) adenocarcinomic human alveolar basal epithelial cell line (FIGs. 14B-14E), A375 (ECACC 88113005)Human malignant melanoma cell line (FIGs. 14H-14K), Ca Ski (ECACC 87020501) Human epithelial cell line (FIGs. 14N-14Q), DMS 273 (ECACC 95062830) Human lung small cell carcinoma cell line (FIGs.
- FIGs. 14T-14W NCI-H322 (ECACC 95111734) Human non-small cell carcinoma cell line (FIGs. 14Z-14AC), SHP-77 (ECACC 98110201) small cell lung carcinoma cell line (FIGs. 14AF-14AI), andHPF-c Primary human pulmonary fibroblasts (FIGs. 14AL-14AO).
- infected cells expressing GFP were detected by fluorescence microscopy.
- Wild type Adhu5-GFP vector FIGS. 14F, 14L, 14R, 14X, 14AD, 14AJ, and 14AP
- uninfected cells vector FIGS. 14A, 14G, 14M, 14S, 14Y, 14AE, and 14AK
- FIGs. 15A-15BT show the recombinant vectors retained their tropism for human cells.
- GFP protein coding sequence was cloned into version 2, version 3, version 4, version 5, version 6, version 7, and version 8 vectors and they were added to HEK293 cell line (FIGs. 15A-15I), Ca Ski (ECACC 87020501) human epithelial cell line cell line (FIGs. 15J-15R), DMS 273 (ECACC 95062830) human lung small cell carcinoma cell line (FIGs. 15S-15AA), NCI-H322 (ECACC 95111734) human non-small cell carcinoma cell line (FIGs.
- FIGs. 15AB-15AJ SHP-77 (ECACC 98110201) small cell lung carcinoma cell line (FIGs. 15AK-15AS), A375 (ECACC 88113005) human malignant melanoma cell line (FIGs. 15AT-15BB), A549 (ECACC 86012804) adenocarcinomic human alveolar basal epithelial cell line (FIGs. 15BC-15BK), and HPF-c Primary human pulmonary fibroblasts (FIGs. 15BL-15BT). At day 2 or day 5 post-infection, infected cells expressing GFP were detected by fluorescence microscopy. Wild type Adhu5-GFP vector (FIGs.
- 15B, 15K, 15T, 15AC,15AL, 15AU, 15BD,and 15BM)anduninfected cells vector (FIGs. 15A, 15J, 15S, 15AB, 15AK, 15AT, 15BC, and 15BL) were used as positive and negative cells respectively.
- FIGs. 16A-16E show a longitudinal flow cytometry analysis of circulating AH1- dextramer positive cytotoxic T-cells (CD8+ T cells).
- FIG. 16A shows the experimental timeline for the longitudinal study. Mice were vaccinated with the chimeric vectors version 2, version 4, and version 8 encoding the AH-1 epitope in minigene form. Wild type Adhu5 vector was used as control. Blood sampling occurred at multiple time points following initial treatment on Day 0. Tumor implantation occurred atDay 54.
- FIG. 16B shows percentage of AHl-dextramer+CD8+ cells following vaccination.
- FIG. 16C shows percentage of AHl-dextramer+ CD8+ CD44+ CD62L- cells following vaccination.
- FIG. 16D shows percentage of AHl-dextramer+ CD8+ CX3CR1+ cells following vaccination.
- FIG. 16E shows percentage of AHl-dextramer+ CD8+ Klrgl+ cells following vaccination.
- FIGs. 17A-17C show a flow cytometry analysis of circulating AHI -dextramer positive cytotoxic T-cells (CD8). Surface markers of T cell exhaustion were measured at various time points. Mice were vaccinated with the chimeric vectors version 2, version 4, and version 8 encoding the AH-1 epitope in minigene form. Vaccination and phenotype analysis followed a similar timeline as show in FIG. 16A.
- FIG. 17A shows a longitudinal analysis by serial blood draw of AHl-dextramer+ CD8+ PD-1 + cells.
- FIG. 17B shows measures of AHl-dextramer+ CD8+ Tim3+ cells post-tumor challenge.
- FIG. 17C shows measures of AHl-dextramer+ CD8+ Lag3+ cells post-tumor challenge.
- FIGs. 18A-18D show a longitudinal flow cytometry analysis of circulating AH1- dextramer positive cytotoxic T-cells (CD8+ T cells). Mice were vaccinated with the chimeric vectors version 3 and version 6 encodingthe AH-1 epitope in minigene form. Wild type Adhu5 vector was used as control. Vaccination and phenotype analysis followed a similar timeline as show in FIG. 16A.
- FIG. 18A shows percentage of AHl-dextramer+ CD8+ cells following vaccination.
- FIG. 18B shows percentage of AHl-dextramer+ CD8+ CD44+ CD62L- cells following vaccination.
- FIG. 18C shows percentage of AHl-dextramer+CD8+ CX3CR1+ cells following vaccination.
- FIG. 18D shows percentage of AHl-dextramer+ CD8+ Klrgl+ cells following vaccination.
- FIGs. 19A-19C show a flow cytometry analysis of circulating AHI -dextramer positive cytotoxic T-cells (CD8). Surface markers of T cell exhaustion were measured at various time points. Mice were vaccinated with the chimeric vectors version 3 and version 6 encoding the AH- 1 epitope in minigene form.
- FIG. 19A shows a longitudinal analysis by serial blood draw of AH1- dextramer+ CD8+ PD- 1 + cells.
- FIG. 19B shows measures of AH 1 -dextramer+ CD8+ Tim3 + cells post-tumor challenge.
- FIG. 19C shows measures of AHl-dextramer+ CD8+ Lag3+ cells posttumor challenge.
- FIG. 20 shows volume growth curves of mice bearing subcutaneous CT26 tumors treated with novel vector constructs of the AHI adenoviral vaccine. Mice were vaccinated with the chimeric vectors version 2, version 4, and version 8 encoding the AH-1 epitope in minigene form and challenged with tumors 54 days post-vac cination. Mice vaccinated with chimeric vectors version 2, version 4, and version 8 showed significantly decreased tumor volume compared to naive mice. ** p ⁇ 0.01.
- FIG. 21 shows survival curves of mice bearing subcutaneous CT26 tumors treated with novel vector constructs version 2, 4, and 8 of the AHI adenoviral vaccine. At Day 62, mice that received vector construct versions 2 and 4 showed 100% survival across the cohort.
- FIG. 22 shows volume growth curves of mice bearing subcutaneous tumors treated with novel vector constructs of the AH-1 adenoviral vaccine.
- Mice were vaccinated with the chimeric vectors version 3 and 6 encoding the AH-1 epitope in minigene form and challenged with tumors 42 days post-vaccination.
- Mice vaccinated with chimeric vectors version 3 and version 6 showed significantly decreasedtumorvolume comparedto naive mice or mice vaccinated with Ad5 vector backbone control.
- FIG. 23 shows survival curves of mice bearing subcutaneous CT26 tumors treated with novel vector constructs version 3 and 6 of the AHI adenoviral vaccine. At Day 20, mice that received AdHu5 and vector construct versions 3 and 6 showed 100% survival across the cohort.
- Ranges throughout this disclosure, various aspects of the present disclosure can be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the present disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values withinthat range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 2.7, 3, 4, 5, 5.3, and 6.
- a range such as 95-99% identity includes something with 95%, 96%, 97%, 98% or 99% identity, and includes subranges such as 96-99%, 96-98%, 96-97%, 97- 99%, 97-98% and 98-99% identity. This applies regardless of the breadth of the range.
- determining As used herein, the terms “determining,” “measuring,” “evaluating,” “assessing” “assaying,” and “analyzing” are often used interchangeably herein to refer to forms of measurement.
- the terms include determining if an element is present or not (for example, detection). These terms can include quantitative, qualitative or quantitative and qualitative determinations. Assessing can be relative or absolute. “Detecting the presence of’ can include determining the amount of something present in addition to determining whether it is present er absent depending on the context.
- Cancer and “tumor” are used interchangeably herein. These terms refer to the presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Cancer cells are often in the form of a tumor, but such cells can exist alone within an animal, or can be a non-tumorigenic cancer cell, such as a leukemia cell. These terms include a hematologic cancer, a solid tumor, a softtissue tumor, or a metastatic lesion. “Cancer” includes premalignant, as well as malignant cancers. The term ’’Cancer” may include smoldering neoplasms, dysplasia and metaplasia as well.
- Cancer cells can spread locally or through the bloodstream and lymphatic system to other parts of the body.
- various cancers include but are not limited to colorectal cancer, prostate cancer, esophageal cancer, liver cancer, renal cancer, lung cancer, breast cancer, breast cancer, pancreatic cancer, brain cancer, hepatocellular cancer, lymphoma, leukemia, gastric cancer, cervical cancer, ovarian cancer, thyroid cancer, melanoma, carcinoma, head and neck cancer, glioblastoma, skin cancer, nasopharyngeal cancer, Epstein Barr driven cancers, Human Papilloma virus driven cancers and soft tissue sarcoma.
- the cancer described herein can be a stage I cancer, a stage II cancer, a stage III cancer, or a stage IV cancer.
- tumor refers to the physiological condition in mammals characterized by deregulated cell growth.
- the term “individual,” “patient,” or “subject” refers to individuals diagnosed with, suspected of being afflicted with, or at-risk of developing at least one disease for which the described compositions and methods are useful for treating.
- the individual is a human.
- the individual is a mammal.
- the mammal is a non-human primate (e.g., rhesus or other types of macaques), mouse, rat, rabbit, dog, cat, horse, cow, sheep, pig, goat, llama, alpaca, or yak.
- the individual is a human.
- the term “linker” as used herein refers to a molecule linking two other molecules or moieties.
- the linker can be an amino acid sequence in the case of a linker joining two fusion proteins.
- the linker can also be a nucleotide sequence in the case of joining two nucleotide sequences together.
- a linker may have various lengths, depending on the application of a linker or the sequences or molecules being linked by a linker.
- treatment or “treating” are used in reference to a pharmaceutical or other intervention regimen used for obtaining beneficial or desired results in the recipient.
- beneficial or desired results includebut are not limited to a therapeutic b enefit and/or a prophylactic benefit.
- Atherapeuticbenefit may referto eradication or amelioration of symptoms or of an underlying disorder being treated.
- a therapeutic benefit can be achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the subject, notwithstanding that the subjectmay still be afflicted with the underlying disorder.
- Aprophylactic effect includes delaying preventing, or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
- a subject at risk of developing a particular disease, or to a subject reporting one or more of the physiological symptoms of a disease may undergo treatment, even though a diagnosis of this disease may not have been made.
- Skilled artisans will recognize that given a population of potential individuals for treatment not all will respond or respond equally to the treatment. Such individuals are considered treated if administered the compositions including the pharmaceutical compositions described herein.
- the term “effective amount”, “therapeutically effective amount” or “pharmaceutically effective amount” refers to an amount that is sufficient to effect treatment, as defined below, when administered to a subject (e.g. , a mammal, such as a human) in need of such treatment.
- the therapeutically or pharmaceutically effective amount will vary dependinguponthe subject and disease condition being treated, the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
- a “therapeutically effective amount” or a “pharmaceutically effective amount” of AAV2-derived capsid as described herein is an amount sufficient to generate an immune response in a subject (e.g , a human).
- the immune response is sufficientto raise AAV capsid neutralizing antibodies against the relevant capsid(s) in the subject.
- sequence identity refers to the subunit sequence identity between two polymeric molecules, for example, between two polynucleotide or polypeptide sequences.
- An analysis of sequence identity beginsby aligningtwo sequences. Identical sequences (100% sequence identity) have the same nucleotide or amino acid at each position of the alignment.
- Percent sequence identity is determined by comparing the number of positions that are identical to the total number of subunits in the sequence alignment. Percent sequence identity can be determined over a fraction of the sequences or over the whole of the sequences. Percent sequence identity can be determined using a sequence comparison algorithm. Test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then aligns the sequences and calculates the percent sequence identity based on the designated program parameters.
- Sequence identity is typically measured using sequence analysis software. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. AppL Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol. 48 :443 (1970), by the search for similarity method of Pearson & Lipman, Proc. Nat'l. Acad. Sci.
- NS'd 85 :2444( l 988) by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), orby visual inspection (see generally Ausubel et al., Current Protocols in Molecular Biology).
- GAP Garnier et al., Current Protocols in Molecular Biology
- Such software matches identical or similar sequences by assigning degrees of homology to various substitutions, deletions, and/or other modifications.
- a BLAST program may be used, with a probability score between e-3 and e-100 indicating a closely related sequence.
- BLAST algorithm One example of algorithm that is suitable for determiningpercent sequenceidentity and sequence similarity is the BLAST algorithm, which is described in Altschul et al., J. Mol. Biol. 215 :403 (1990).
- Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information (publicly accessible through the National Institutes of Health NCBI internet server).
- default program parameters can be used to perform the sequence comparison, although customized parameters can also be used.
- W wordlength
- E expectation
- BLOSUM62 scoring matrix see Henikoff & Henikoff, Proc. Natl. Acad Set USA 89: 10915 (1989)).
- sequence identities are determined by Needleman- Wunsch alignment of two sequences with Gap Costs set to Existence: 11 Extension: 1 where percent identity is calculated by dividing the number of identities by the length of the alignment.
- Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
- the term “antigen presenting cell” or “APC” refers to an immune system cell such as an accessory cell (e.g., a B-cell, a dendritic cell, and the like) that displays a foreign antigen complexed with major histocompatibility complexes (MHC’s) on its surface.
- T cells may recognize these complexes usingtheirT cell receptors (TCRs).
- APCs process antigens and present them to T cells.
- the term “encoding” refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (e.g., rRNA, tRNA, and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
- a gene, cDNA, or RNA encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system.
- Both the coding strand the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referredto as encodingthe protein or other product of that gene or cDNA.
- nucleotide sequence encoding an amino acid sequence includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence.
- the phrase nucleotide sequence that encodes a protein or an RNA may also include introns to the extent that the nucleotide sequence encoding the protein may in some version contain one or more introns.
- T cell and its grammatical equivalents as used herein can refer to a T cell from any origin.
- a T cell can be a primary T cell, e.g., an autologous T cell, an allogenic T cell, a cell line, etc.
- the T cell can also be human or non-human in origin.
- CD8 refers to the Cluster of Differentiation 8 protein, a transmembrane glycoprotein that serves as a co-receptor for the T-cell receptor (TCR).
- effector T cell includes T helper (e.g., CD4+) cells and cytotoxic (e.g., CD8+) T cells.
- CD4+ effector T cells contribute to the development of several immunologic processes, including maturation of B cells into plasma cells and memory B cells, and activation of cytotoxic T cells and macrophages.
- CD8+ effector T cells destroy virus-infected cells and tumor cells.
- nucleic acid or “polynucleotide” refers to deoxyribonucleic acids (DNA) or ribonucleic acids (RNA) and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides.
- nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, and complementary sequences as well as the sequence explicitly indicated.
- degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)).
- peptide As used herein, the terms “peptide,” “polypeptide,” and “protein” are used interchangeably, and refer to a compound comprised of amino acid residues covalently linked by peptide bonds.
- a protein or peptide must contain at least two amino acids, and no limitation is placed on the maximum number of amino acids that can comprise a protein’s or peptide’s sequence.
- Polypeptides include any peptide orprotein comprisingtwo or more amino acidsjoined to each other by peptide bonds.
- polypeptides include, for example, biologically active fragments, substantially homologous polypeptides, oligopeptides, homodimers, heterodimers, variants of polypeptides, modified polypeptides, derivatives, analogs, fusion proteins, among others.
- a polypeptide includes a natural peptide, a recombinant peptide, or a combination thereof.
- promoter refers to a DNA sequence recognized by the transcription machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a polynucleotide sequence.
- promoter/regulatory sequence refers to a nucleic acid sequence which is required for expression of a gene product operably linked to the promoter/regulatory sequence. In some instances, this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product.
- the promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue specific manner.
- vector refers to a nucleic acid sequence capable of transporting into a cell another nucleic acid to which the vector sequence has been linked.
- vectors are known in the art including, but not limited to, linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses.
- viral vectors include, but are not limited to, adenoviral vectors, adeno-associated virus vectors (AAVs), retroviral vectors, lentiviral vectors, and the like.
- a vector of the present disclosure can be adenoviral and comprises the nucleotide sequence encoding a single cancer specific CD8+ T cell epitope containing a gene construct in a form suitable for expression by a cell (e.g., linked to a transcriptional control element).
- the term “functional deletion” or “functionally deleted” may refer to an amount of a genetic region that is removed (e.g., mutated or modified). In some embodiments, the genetic region may notbe able to produce a functional genetic product. A functionally deleted region may be entirely removed. A functionally deleted region may be partially removed.
- epitope refers to a part of an antigen that is recognized by the immune system which may be a short protein sequence.
- a “cancer specific CD8+ T cell epitope” refers to an epitope thatmay be presented by an antigen presenting cell bound to an MHC molecule which are then recognized by the T-cell receptor (TCR).
- CD8+ T cells express the CD8 coreceptor, which binds to MHC I, and recognize peptides presented by MHC I molecules.
- HLAbinding affinity or “MHC binding affinity” means affinity of binding between a specific antigen and a specific MHC allele.
- HLA binding rank score or “MHC binding rank score” means the predicted rank score.
- the predicted rank score is a percentage rank position of a query sequence (antigen) in relation to a distribution of prediction scores for the MHC in question, estimated from a set of random natural peptides. A rank score below the value of 2% indicates binding between a specific antigen and a specific MHC allele.
- recombinant polypeptides or polynucleotides encoding the recombinant polypeptide
- a T cell epitope that is capable of binding to one or more MHC molecules, presented by the one or more MHC molecules, being immunogenic and/or capable of activating T cells to become cytotoxic and compositions thereof.
- the present disclosure provides a vector.
- the vector may b e a viral vector.
- the vector may be a non-viral vector.
- a viral vector can be an adenovirus vector, an adeno-associated virus (AAV) vector (e.g., AAV type 5 and type 2), an alphavirus vectors (e.g., Venezuelan equine encephalitis virus (VEE), a Sindbis virus (SIN), semliki forest virus (SFV), and VEE-SIN chimeras), a flaviviral vector, a herpesvirus vector (e.g. vectors derived from cytomegaloviruses, like rhesus cytomegalovirus (RhCMV), an arena virus vector (e.g.
- the viral vector can be an oncolytic viral vector. In some embodiments, the viral vector can be an oncolytic adenoviral vector. In some embodiments, the viral vector may not be an oncolytic adenoviral vector.
- the viral vector may be an adenoviral vector.
- An adenovirus particle can comprise a protein capsid surrounding a core which contains a payload (e.g., genetic material).
- the capsid can be composed of three main proteins: hexon, penton, and fiber proteins.
- a viral vector may comprise hexon and penton proteins capsomers. Hexons are the largest and most abundant structural proteins of the adenovirus capsid particle. Hexon proteins organize as trimers, comprising three hexon monomers. Hexon monomers comprise seven flexible loops, termed hypervariable loops (HVRs) which can be located on the surface of the hexon and virion.
- HVRs hypervariable loops
- HVRs may interact with other proteins, receptors, cellular structures, and/or cells.
- the penton and fiber may form the penton complex at each virion vertex of the adenovirus particle.
- a fiber may comprise a knob, a shaft, and a tail.
- the knob, shaft, and/or tail of a fiber protein of an adenoviral vector may be replaced or substituted.
- the sequence of a hexon, penton, and/or fiber protein can vary between different adenovirus serotypes.
- the vector may be a chimeric vector or a chimeric adenoviral vector.
- chimeric adenovirus or “chimeric adenoviral vector” may refer to an adenovirus with a nucleic acid sequence comprising nucleic acid sequences from at least two different adenoviral serotypes.
- a chimeric nucleic acid may refer to a nucleic acid that has been modified wherein a heterologous nucleic acid has been introduced to the native (e.g., non-chimeric) form of the nucleic acid.
- the adenoviral vector can be or derived from a human adenovirus vector or a simian adenovirus.
- the 57 human adenoviruses characterized havebeen organized into seven major subgroups as shown in Table 1.
- the adenoviral vector may be an adenoviral vector of subgroup A, subgroup B, subgroup C, subgroup D, subgroup E, subgroup F, or subgroup G.
- the adenoviral vector described herein may be a chimeric adenoviral vector comprising two or more adenoviral subgroups.
- the adenoviral vector described herein may be a chimeric adenoviral vector comprising two or more adenoviral serotypes from the same adenoviral subgroup.
- the adenoviral vector described herein may be a chimeric adenoviral vector comprising three or more adenoviral subgroups. In some embodiments, the adenoviral vector described herein may be a chimeric adenoviral vector comprising three or more adenoviral serotypes from the same adenoviral subgroup. In some embodiments, the adenoviral vector may be a serotype vector derived from a non-human primate.
- Exemplary serotypes include SAdV26, SAdV27, SAdV28, SAdV29, SAdV30, SAdV31, SAdV32, SAdV33, SAdV34, SAdV35, SAdV36, SAdV37, SAdV38, SAdV39, SAdV40, SAdV41, SAdV42, SAdV43, SAdV44, SAdV45, SAdV46, SAdV47, SAdV48, SAdV49, SAdV50, SAdV51, SAdV52, SAdV53, SAdV54, SAdV55, or a combination thereof.
- the adenoviral vector is a human serotype 5 (AdHu5).
- the vector may be an animal derived adenoviral vector for example canine, or simian.
- the simian-derived adenoviral vector may be rhesus monkey, chimpanzee, bonobo, gorilla,, macaque, grivet, golden snubnosed monkey, yellow baboon, or cynomolgus monkey.
- the adenoviral vector may be a rare serotypevector derived from anon-human primate. Vectors derived from chimpanzee may be suitable for the vector for the present disclosure, examples include but are not limited to ChAd63, ChAd3, ChAdY25.
- the adenoviral vector may be an adenoviral vector of a simian adenovirus.
- the adenoviral vector maybe a simian serotype 1 (SAdVl), SAdV2, SAdV3, SAdV4, SAdV5, SAdV6, SAdV7, SAdV8, SAdV9, SAdVIO, SAdVl 1, SAdV12, SAdV13, SAdV14, SAdVl 5, SAdVl 6, SAdVl 7, SAdVl 8, SAdVl 9, SAdV20, SAdV21, SAdV22, SAdV23, SAdV24, or SAdV25, or a combination thereof.
- SAdVl simian serotype 1
- SAdV2 SAdV2, SAdV3, SAdV4, SAdV5, SAdV6, SAdV7, SAdV8, SAdV9, SAdVIO, SAdVl 1, SAdV12, SAdV13, SAdV14
- the adenoviral vector may comprise at least one simian adenovirus serotype, at least two simian adenovirus serotypes, at least three simian adenovirus serotypes, at least four simian adenovirus serotypes, at least five simian adenovirus serotypes, at least six simian adenovirus serotypes, at least seven simian adenovirus serotypes, or at least eight simian adenovirus serotypes.
- the adenoviral vector described herein may comprise a combination of human adenovirus serotype and simian adenovirus serotype. The classifications of simian adenovirus species are shown in
- the vector is an adenoviral vector, e.g., an adenoviral vector comprising a polynucleotide sequence encoding a single epitope, e.g., a single T cell epitope, e.g, a single cancer-specific CD8+ epitope.
- the vector is capable of inducing an inflating memory CD8+ T cell response.
- the adenoviral vector comprises a polynucleotide sequence encoding a single epitope, e.g., a single T cell epitope, e.g, a single cancer-specific CD8+ epitope, and the vector does not comprise any additional cancer specific CD8+ T cell epitopes.
- the adenoviral vector comprises a polynucleotide sequence encoding multiple epitopes, e.g., multiple T cell epitopes, e.g., multiple cancer-specific CD8+ epitopes.
- the vector is or comprises an AAV vector.
- the AAV vector may be an AAV1, AAV2, AAV5 or any combination thereof.
- the AAV vector may be a recombinant AAV vector (rAAV).
- the AAV vector is selected for delivery to a particular cell, tissue, or organ, e.g., AAV serotypes 1, 2, 5 or a hybrid capsid AA VI, AAV2, AAV5 or any combination thereoffor targeting brain or neuronal cells; AAV4 for targeting cardiac tissue, and AAV8 for delivery to the liver.
- the vector further comprises a promoter.
- the vector is an in vitro transcribed vector.
- the vector may comprise a strong promoter, examples include but are not limited to a CMV promoter, an RSV promoter, an EFla promoter, a simian virus 40 (SV40) early promoter, a mouse mammary tumor virus (MMTV), a human immunodeficiency virus (HIV) long terminal repeat (LTR) promoter, a MoMuLV promoter, an avian leukemia virus promoter, an Epstein-Barr virus immediate early promoter, a Rous sarcoma virus (RSV) promoter, as well as human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the elongation factor- la promoter, the hemoglobin promoter, and the creatine kinase promoter.
- the vector comprises a CMV promoter.
- the vector may comprise an inducible promoter.
- inducible promoters include, but are not limited to a metallothionine promoter, a glucocorticoid promoter, a progesterone promoter, and a tetracycline-regulated promoter.
- a vector disclosed herein can comprise additional promoter elements, e.g., enhancers and/or silencers.
- the vector does not comprise an arginine-glycine-aspartate (RGD) peptide.
- the vector comprises an arginine-glycine-aspartate (RGD) peptide.
- the vector may comprise a TATA box.
- the vector comprises a translation initiation sequence, for example a Kozak sequence.
- a Kozak sequence has the consensus sequence (gcc)gccRccAUGG, a suitable Kozak sequence is provided in SEQ ID NO. : 3.
- the vector comprises a termination sequence and/or a polyadenylation sequence.
- a suitable polyadenylation sequence is provided in SEQ ID NO.: 5.
- the viral vector comprises the vector back bone, a promoter region and a nucleotide sequence encoding a single epitope, e.g., a single T cell epitope, e.g., a single cancer-specific CD8+ and/or CD4+ epitope, e.g., a single cancer-specific CD8+ epitope.
- a single epitope e.g., a single T cell epitope, e.g., a single cancer-specific CD8+ and/or CD4+ epitope, e.g., a single cancer-specific CD8+ epitope.
- the ITR sequences flank the encoded the epitope, e.g., the T cell epitope, e.g., the cancer-specific CD8+ and/or CD4+ epitope, e.g., the cancer-specific CD8+ epitope.
- helper plasmids in order to produce the viral vector comprising the cancer specific CD8+ T cell epitope, helper plasmids may be used.
- a helper plasmid or plasmids may be used to provide genes required for viral vector replication or packaging.
- helper plasmid encodes E2A, E4 and VA adenoviral proteins and or encodes the rep and cap genes of AAV.
- an AAV vector disclosed herein may have the following elements from a 5’ to 3’ direction: (i) 5’ITR, (ii) an att site or a RE site, (iii) a CMV promoter, (iv) a translation initiation sequence, for example a Kozak sequence, (v) a start codon sequence, (vi) a cancer specific CD8+ T cell epitope, e.g., a cancer specific CD8+T cell epitope disclosed herein; e.g., a codon optimized cancer specific CD8+ T cell epitope disclosed herein, (vii) a stop codon sequence, (viii) a BGH poly A sequence, (ix) an att site or a RE site, and (x) 3 ’ ITR.
- a vector may be an adenoviral vector or an adeno-associated vector of the present invention may reduce or eliminate viral DNA replication and/or viral gene product production when infected into the target cells as compared with first-generation El -deleted vectors. While not wishing to be held to any particular theory of the invention, it appears that reduced viral replication advantageously results in prolonged transgene expression following transduction with the deleted adenovirus vectors of the invention. Reduced viral specific activities following vector transduction and transgene expression may also result in reduced host immune responses and cytotoxic effects on the host cell. These, in turn, may result in a longer duration of transgene expression.
- the vector may be modified to reduce the immunogenicity and improve biosafety of the vector.
- the vector may be replication-deficient.
- the vector may lack the El and E3 proteins.
- the present disclosure provides a recombinant (e.g., engineered) adenoviral vector, wherein the backbone of the recombinant adenoviral vector is an adenovirus serotype 5 (Ad5) nucleic acid backbone.
- the recombinant adenoviral vector may comprise a deletion or a functional deletion of an El gene.
- the recombinant adenoviral vector may comprise a deletion or a functional deletion of an E3 gene.
- the recombinant adenoviral vector may comprise a deletion ora functional deletion of the El andE3 genes.
- the recombinant adenoviral vector may comprise a transgene inserted at the locus of the deleted or functionally deleted El gene.
- the recombinant adenoviral vector may comprise a transgene inserted at the locus of the deleted or functionally deleted E3 gene.
- the transgene insertion may comprise a single cancer-specific CD8+ T cell epitope.
- the transgene insertion may comprise a single cancer-specific CD4+ T cell epitope.
- the transgene insertion may comprise more than one cancer-specific CD4+ T cell epitope.
- the transgene insertion may comprise a cancer-specific CD8+ T cell epitope and a cancer-specific CD4+ T cell epitope.
- a nucleic acid sequence encoding the single cancer-specific CD8+ T cell epitope may be inserted in the El region and the recombinant adenoviral vector may comprise a nucleic acid sequence of a first adenovirus serotype and a nucleic acid sequence of a second adenovirus serotype.
- a nucleic acid sequence encoding the single cancer-specific CD8+ T cell epitope may be inserted in the El region and the recombinant adenoviral vector may comprise a nucleic acid sequence of a first adenovirus serotype, a nucleic acid sequence of a second adenovirus serotype, and a nucleic acid sequence of a third adenovirus serotype.
- a nucleic acid sequence encoding the single cancer-specific CD8+ T cell epitope may be inserted in the El region and the recombinant adenoviral vector may comprise a nucleic acid sequence of a first adenovirus serotype, a nucleic acid sequence of a second adenovirus serotype, a nucleic acid sequence of a third adenovirus serotype, and a nucleic acid sequence of a fourth adenovirus serotype.
- the recombinant adenoviral vector may be a recombinant chimeric adenoviral vector (e.g., a recombinant chimeric adenovirus).
- the recombinant chimeric adenoviral vector may comprise a nucleic acid sequence comprising nucleic acid sequences of at least two adenovirus serotypes, at least three adenovirus serotypes, at least four adenovirus serotypes, at least five adenovirus serotypes, at least six adenovirus serotypes, at least seven adenovirus serotypes, or at least eight adenovirus serotypes.
- a chimeric adenovirus may have a nucleic acid sequence encoding a capsid.
- the capsid may comprise a hexon protein, a penton protein, and/or a fiber protein.
- the fiber protein may comprise a tail portion, a shaft portion, and/or a knob portion.
- the recombinant chimeric adenoviral vector may comprise a first adenovirus serotype backbone and a hexon protein, a penton protein, and/or a fiber protein from a second adenovirus serotype.
- the recombinant chimeric adenoviral vector may comprise a hexon protein and a penton protein from a first adenovirus serotype and a fiber protein from a second adenovirus serotype.
- the recombinant chimeric adenoviral vector may comprise a hexon protein, a penton protein, and a tail portion of a fiber protein from a first adenovirus serotype and a shaft portion and a knob portion of a fiber protein from a second adenovirus serotype.
- the recombinant chimeric adenoviral vector may comprise a penton protein and a fiber protein from a first adenovirus serotype and hexon HVRs from a second adenovirus serotype.
- the recombinantchimeric adenoviral vector may comprise a penton protein from a first adenovirus serotype, hexon HVRs from a second adenovirus serotype, and a fiber protein from a third adenovirus serotype.
- the recombinant chimeric adenoviral vector may comprise a penton protein and a tail portion of a fiber protein from a first adenovirus serotype, hexon HVRs from a second adenovirus serotype, and a shaft portion and a knob portion of a fiber protein from a third adenovirus serotype.
- the recombinant chimeric adenoviral vector may comprise a penton protein from a first adenovirus serotype and a hexon protein and a fiber protein from a second adenovirus serotype. In some embodiments, the recombinant chimeric adenoviral vector may comprise a penton protein and a tail portion of a fiber protein from a first adenovirus serotype and a hexon protein from a second adenovirus serotype and a shaft portion and a knob portion of a fiber protein from the second adenovirus serotype or a third adenovirus serotype.
- the recombinant chimeric adenoviral vector may comprise a penton protein and a fiber protein from a first adenovirus serotype and a hexon protein from a second adenovirus serotype.
- the recombinantadenoviral vector has abackboneof an adenovirus serotype 5 (Ad5).
- the adenovirus serotype 5 is a human adenovirus serotype 5 (AdHu5).
- the recombinant adenoviral vector comprises at least one adenovirus serotype, atleasttwo adenovirus serotypes, orthree adenovirus serotypes.
- the recombinant adenoviral vector comprises an Ad5 backbone and/or a penton protein from an adenovirus serotype 3 (Ad3).
- the adenovirus serotype 3 is a human adenovirus serotype 3 (AdHu3) or a simian adenovirus serotype 3 (SAd3).
- the recombinant adenoviral vector comprises an AdHu5 backbone and an AdHu5 penton protein.
- the recombinant adenoviral vector comprises an AdHu5 backbone and an Ad3 hexon protein, e.g., a SAd3 hexon protein.
- the recombinant adenoviral vector comprises an AdHu5 backbone and an AdHu5 hexon protein.
- the recombinant adenoviral vector comprises an AdHu5 backbone and a chimeric hexon protein comprising an AdHu5 hexon protein with HVRs (e.g., HVR1-HVR7) from adenovirus serotype 48.
- HVRs e.g., HVR1-HVR7
- the recombinant adenoviral vector comprises an AdHu5 backbone and an Ad3 fiber protein, e.g., a SAd3 fiber protein.
- the recombinant adenoviral vector comprises an AdHu5 backbone and an AdHu5 fiber protein.
- the recombinant adenoviral vector comprises an AdHu5 backbone and a fiber protein with a tail portion, a shaft portion, and/or a knob portion from Ad3, e.g., SAd3.
- the recombinant adenoviral vector comprises an AdHu5 backbone and a fiber protein with a tail portion, a shaft portion, and/or a knob portion from adenovirus serotype 35 (Ad35).
- a polynucleotide may encode a recombinant adenoviral vector or a portion of a recombinant adenoviral vector described herein.
- a polynucleotide disclosed herein comprises a nucleic acid sequence as shown in Table 2.
- the recombinant adenoviral vector comprises a backbone of a subgroup C adenovirus and a hex on protein, a penton protein, and/or a fiber protein of a subgroup A, B, C, D, E, F, or G adenovirus.
- the backbone of the recombinant adenoviral vector may be from a subgroup A, B, C, D, E, F, or G adenovirus.
- the hexon protein of the recombinant adenoviral vector may comprise an adenovirus serotype 12, 18, or 31 .
- the hexon protein of the recombinant adenoviral vector may comprise an adenovirus serotype 3, 7, 11, 14, 16, 21, 34, 35, or 55.
- the hexon protein of the recombinant adenoviral vector may comprise an adenovirus serotype 1, 2, 5, or 6.
- the hexon protein of the recombinant adenoviral vector may comprise an adenovirus serotype 8, 9, 10, 13, 15, 17, 19, 20, 22-30, 32, 33, 36, 37, 38, 39, 42-49, 51, 53, 54, or 56.
- the hexon protein of the recombinant adenoviral vector may comprise an adenovirus serotype 4, 40, 41 , or 52.
- the penton protein of the recombinant adenoviral vector may comprise an adenovirus serotype 12, 18, or 31.
- the penton protein of the recombinant adenoviral vector may comprise an adenovirus serotype 3, 7, 11, 14, 16, 21, 34, 35, or 55.
- the penton protein of the recombinant adenoviral vector may comprise an adenovirus serotype 1, 2, 5, or 6.
- the penton protein of the recombinant adenoviral vector may comprise an adenovirus serotype 8, 9, 10, 13, 15, 17, 19, 20, 22-30, 32, 33, 36, 37, 38, 39, 42-49, 51, 53, 54, or 56.
- the penton protein of the recombinantadenoviralvector may comprise an adenovirus serotype 4, 40, or 41 .
- the fiber protein or a portion of the fiber protein (e.g., the tail portion, the shaft portion, or the knob portion) of the recombinant adenoviral vector may comprise an adenovirus serotype 12, 18, or 31 .
- the fiber protein or a portion of the fiber protein (e.g., the tail portion, the shaftportion, orthe knob portion) of the recombinant adenoviral vector may comprise an adenovirus serotype 3, 7, 11, 14, 16, 21, 34, 35, or 55.
- the fiber protein or a portion of the fiber protein (e.g., the tail portion, the shaft portion, or the knob portion) of the recombinant adenoviral vector may comprise an adenovirus serotype 1, 2, 5, or 6.
- the fiber protein or a portion of the fiber protein (e.g., the tail portion, the shaft portion, or the knob portion) of the recombinant adenoviral vector may comprise an adenovirus serotype 8, 9, 10, 13, 15, 17, 19, 20, 22-30, 32, 33, 36, 37, 38, 39, 42-49, 51, 53, 54, or 56.
- the fiber protein or a portion of the fiber protein (e.g., the tail portion, the shaft portion, or the knob portion) of the recombinant adenoviral vector may comprise an adenovirus serotype 4, 40, or 41 .
- the recombinant adenoviral vector may comprise a backbone of human Ad5 and a hexon protein, penton protein, fiber protein, and/or portion of a fiber protein of a simian adenovirus species.
- a hexon protein, penton protein, fiber protein, and/or portion of a fiber protein may be from a simian serotype 1 (SAdVl), SAdV2, SAdV3, SAdV4, SAdV5, SAdV6, SAdV7, SAdV8, SAdV9, SAdVIO, SAdVl 1, SAdV12, SAdV13, SAdV14, SAdV15, SAdV16, SAdV17, SAdV18, SAdV19, SAdV20, SAdV21, SAdV22, SAdV23, SAdV24, SAdV25, SAdV26, SAdV27, SAdV28, SAdV29, SAdV30, SAdV31, SAdV32, SAdV33, SAdV34, SAdV35, SAdV36, SAdV37, SAdV38, SAdV39, SAdV40, SAdV41, SAdV42, SAdV43, SAdV
- a fiber protein described herein may comprise a human adenovirus, a simian adenovirus, or a combination thereof.
- a tail portion of a fiber protein is from a simian adenovirus species.
- a tail portion of a fiber protein is from a human adenovirus species.
- a shaft portion of a fiber protein is from a simian adenovirus species.
- a shaft portion of a fiber protein is from a human adenovirus species.
- a knob portion of a fiber protein is from a simian adenovirus species.
- a knob portion of a fiber protein is from a human adenovirus species.
- the recombinant adenoviral vector may comprise a penton protein from an Ad5 or SAd3 serotype.
- the recombinant adenoviral vector may comprise a hexon protein from an Ad5, SAd3, or Ad48 serotype.
- the recombinant adenoviral vector may comprise a portion of a hexon protein and/or at least one HVR from an Ad5, SAd3, or Ad48 serotype.
- the recombinant adenoviral vector may comprise a fiber protein from an Ad5, SAd3, or Ad35 serotype.
- the recombinant adenoviral vector may comprise a fiber protein from a combination of Ad5, SAd3, or Ad35 serotypes.
- the recombinant adenoviral vector may comprise a tail portion of a fiber protein from an Ad5, SAd3, orAd35 serotype.
- Therecombinantadenoviralvector maycomprisea shaftportion of a fiber protein from an Ad5, SAd3, or Ad35 serotype.
- the recombinant adenoviral vector may comprise a knob portion of a fiber protein from an Ad5, SAd3, or Ad35 serotype.
- the recombinant adenoviral vector comprises an AdHu5 backbone, a SAd3 penton protein, a SAd3 hexon protein, and a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding a SAd3 penton protein, a sequence encoding a SAd3 hexon protein, and a sequence encoding a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequence comprising an AdHu5 nucleic acid backbone, a SAd3 penton protein (e.g., penton region), a SAd3 hexon protein (e.g., hexon region), and a SAd3 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises a SAd3 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 67; a SAd3 hexon protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 68; and a tail portion, shaft portion, and knob portion of a SAd3 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 68
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, an AdHu5 hexon protein, and a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding a AdHu5 penton protein, a sequence encoding a AdHu5 hexon protein, and a sequence encoding a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequence comprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), an AdHu5 hexon protein (e.g., hexon region), and a SAd3 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted atthe El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises an AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 62; an AdHu5 hexon protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- a SAd3 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 64, SEQ ID NO.: 65, and SEQ ID NO. : 66, respectively.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, an AdHu5 hexon protein, a tail portion of an AdHu5 fiber protein, a shaft portion of a SAd3 fiber protein, and a knob portion of a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding a AdHu5 penton protein, a sequence encoding a AdHu5 hexon protein, a sequence encoding a tail portion of an AdHu5 fiber protein, a sequence encoding a shaft portion of a SAd3 fiber protein, and a sequence encoding a knob portion of a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequence comprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), an AdHu5 hexonprotein (e.g., hexonregion), and a tail portion of an AdHu5 fiber protein (e.g., fiber region), a shaft portion of a SAd3 fiber protein (e.g., fiber region), and a knob portion of a SAd3 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises an AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- an AdHu5 hexon protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 63; a tail portion of a AdHu5 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 50; and a shaft portion and knob portion of a SAd3 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 65 and SEQ ID NO.: 66, respectively
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, an AdHu5 hexon protein, a tail portion of an AdHu5 fiber protein, a shaft portion of a AdHu35 fiber protein, and a knob portion of a AdHu35 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding a AdHu5 penton protein, a sequence encoding a AdHu5 hexon protein, a sequence encoding a tail portion of an AdHu5 fiber protein, a sequence encoding a shaft portion of a AdHu35 fiber protein, and a sequence encoding a knob portion of a AdHu35 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequencecomprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), an AdHu5 hexon protein (e.g., hexon region), and a tail portion of an AdHu5 fiber protein (e.g., fiber region), a shaft portion of a AdHu35 fiber protein (e.g., fiber region), and a knob portion of a AdHu35 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 generegionsand atransgenemaybeinserted attheEl genelocus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises an AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 62; an AdHu5 hexon protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- a tail portion of a AdHu5 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 50; and a shaft portion and knob portion of a AdHu35 fiber protein comprising an amino acid sequence with atleastabout90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 70 and SEQ ID NO.: 71, respectively.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, an AdHu5 hexon protein with HVR1 , HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 of Ad48, and an AdHu5 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding an AdHu5 penton protein, a sequence encoding an AdHu5 hexon protein with HVR1 , HVR2, HVR3, HVR4, HVR5, HVR6, and HVR7 sequences of Ad48, and a sequence encoding an AdHu5 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequencecomprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), a AdHu5 hexon protein (e.g., hexon region) with HVR1, HVR2, HVR3, HVR4, HVR5, HVR6, andHVR7 of Ad48, and an AdHu5 fiberprotein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises an AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, an AdHu5 hexon protein with HVR1 , HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 of Ad48, and a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding an AdHu5 penton protein, a sequence encoding an AdHu5 hexon protein with HVR1 , HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 sequences of Ad48, and a sequence encoding a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequencecomprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), a AdHu5 hexon protein (e.g., hexon region) with HVR1, HVR2, HVR3, HVR4, HVR5, HVR6, and HVR7 of Ad48, and a SAd3 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises an AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, an AdHu5 hexon protein with HVR1, HVR2, HVR3, HVR4, HVR5, HVR6, and HVR7 of Ad48, a tail portion of an AdHu5 fiber protein, a shaft portion of a SAd3 fiber protein, and a knob portion of a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding an AdHu5 penton protein, a sequence encoding an AdHu5 hexon protein with HVR1 , HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 sequences of Ad48, a sequence encoding a tail portion of an AdHu5 fiber protein, a sequence encoding a shaft portion of a SAd3 fiber protein, and a sequence encoding a knob portion of a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequence comprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), a AdHu5 hexon protein (e.g., hexon region) with HVR1, HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 of Ad48, a tail portion of an AdHu5 fiber protein (e.g., fiber region), a shaft portion of a SAd3 fiber protein (e.g., fiber region), and a knob portion of a SAd3 fiber protein (e.g., fiber region).
- AdHu5 nucleic acid backbone e.g., an AdHu5 penton protein (e.g., penton region), a AdHu5 hexon protein (e.g., hexon region) with HVR1, HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 of Ad48, a tail portion of an AdHu5 fiber protein (e.
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises an AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, an AdHu5 hexon protein with HVR1, HVR2, HVR3, HVR4, HVR5, HVR6, andHVR7 of Ad48, atail portion of an AdHu5 fiberprotein, a shaftportion of an AdHu35 fiber protein, and a knob portion of an AdHu35 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding an AdHu5 penton protein, a sequence encoding an AdHu5 hexon protein with HVR1 , HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 sequences of Ad48, a sequence encoding a tail portion of an AdHu5 fiber protein, a sequence encoding a shaft portion of an AdHu35 fiber protein, and a sequence encoding a knob portion of an AdHu35 protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequence comprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), a AdHu5 hexon protein (e.g., hexon region) with HVR1, HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 of Ad48, a tail portion of an AdHu5 fiber protein (e.g., fiber region), a shaft portion of an AdHu35 fiber protein (e.g., fiber region), and a knob portion of an AdHu35 fiber protein (e.g., fiber region).
- AdHu5 nucleic acid backbone e.g., an AdHu5 penton protein (e.g., penton region), a AdHu5 hexon protein (e.g., hexon region) with HVR1, HVR2, HVR3 , HVR4, HVR5, HVR6, and HVR7 of Ad48, a tail portion of an AdHu5 fiber protein (e.g.
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises an AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- AdHu35 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 70 and SEQ ID NO.: 71, respectively.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, a SAd3 hexon protein, and a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding an AdHu5 penton protein, a sequence encoding a SAd3 hexon protein, and a sequence encoding a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequence comprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), a SAd3 hexon protein (e.g., hexon region), and a SAd3 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises a AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 62; a SAd3 hexon protein comprising an amino acid coding sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- a SAd3 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 64, SEQ ID NO.: 65, and SEQ ID NO. : 66, respectively.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, a SAd3 hexon protein, a tail portion of an AdHu5 fiber protein, a shaft portion of a SAd3 fiber protein, and a knob portion of a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding a AdHu5 penton protein, a sequence encoding a SAd3 hexon protein, a sequence encoding a tail portion of an AdHu5 fiber protein, a sequence encoding a shaft portion of a SAd3 fiber protein, and a sequence encoding a knob portion of a SAd3 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequence comprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), a SAd3 hexon protein (e.g., hexon region), and a tail portion of an AdHu5 fiber protein (e.g., fiber region), a shaft portion of a SAd3 fiber protein (e.g., fiber region), and a knob portion of a SAd3 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises a AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 62; a SAd3 hexon protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- AdHu5 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 50; and a shaft portion and knob portion of a SAd3 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 65 and SEQ ID NO.: 66, respectively.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, a SAd3 hexon protein, a tail portion of an AdHu5 fiber protein, a shaft portion of an AdHu35 fiber protein, and a knob portion of an AdHu35 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding a AdHu5 penton protein, a sequence encoding a SAd3 hexon protein, a sequence encoding a tail portion of an AdHu5 fiber protein, a sequence encoding a shaft portion of an AdHu35 fiber protein, and a sequence encoding a knob portion of an AdHu35 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequencecomprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), a SAd3 hexon protein (e.g., hexon region), and a tail portion of an AdHu5 fiber protein (e.g., fiber region), a shaft portion of an AdHu35 fiber protein (e.g., fiber region), and a knob portion of an AdHu35 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted atthe El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises a AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 62; a SAd3 hexon protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- AdHu5 fiber protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 50; and a shaft portion and knob portion of a AdHu35 fiber protein comprising an amino acid sequence with atleastabout90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO. : 70 and SEQ ID NO.: 71, respectively.
- the recombinant adenoviral vector comprises an AdHu5 backbone, an AdHu5 penton protein, a SAd3 hexon protein, and an AdHu5 fiber protein.
- the recombinant adenoviral vector comprises a modified genome comprising a nucleic acid sequence with an AdHu5 nucleic acid backbone, a sequence encoding an AdHu5 penton protein, a sequence encoding a SAd3 hexon protein, and a sequence encoding an AdHu5 fiber protein.
- the recombinant adenoviral vector comprises a modified genome.
- the modified genome may comprise an amino acid coding sequence comprising an AdHu5 nucleic acid backbone, an AdHu5 penton protein (e.g., penton region), a SAd3 hexon protein, and an AdHu5 fiber protein (e.g., fiber region).
- the recombinant adenoviral vector can comprise a deletion or functional deletion in the El gene and/or E3 gene regions and a transgene may be inserted at the El gene locus.
- the transgene may comprise a single CD8+ T cell epitope.
- the recombinant adenoviral vector comprises a AdHu5 penton protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.: 62; a SAd3 hexon protein comprising an amino acid sequence with at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, or 100% sequence identity to a sequence as set forth in SEQ ID NO.
- the transgene insertion comprises at least one recombination site, at leasttwo recombination sites, atleastthree recombination sites atleastfour recombination sites, or at least five recombination sites.
- the transgene insertion comprises at most five recombination sites, at most four recombination sites, at most three recombination sites, at most two recombination sites, or at most one recombination site.
- the recombination site may be an att site. In some embodiments, the recombination site may be a lox site.
- a recombinant adenoviral vector as described herein or a component of a recombinant adenoviral vector described herein may comprise an amino acid sequence as shown in Table 3.
- the recombinant adenoviral vector comprises a shaft portion comprising an amino acid sequence with at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at leastabout95%, at least about 96%, atleastabout97%, atleastabout98%, at least about 98.5%, at least about 99%, at least about 99.5%, or at least about 99.9% sequence identity to an amino acid sequence as set forth in SEQ ID NO. : 65.
- the recombinant adenoviral vector comprises a shaft portion comprising an amino acid sequence with at most about 99.9%, at most about 99.5%, at most about 99%, at most about 98.5%, at most about 98%, at most about 97%, atmost about96%, atmostabout95%, atmostabout94%, atmostabout93%, atmost about 92%, at most about 91%, or at most about 90% sequence identity to an amino acid sequence as set forth in SEQ ID NO.: 65.
- the recombinant adenoviral vector comprises a shaft portion comprising an amino acid sequence from about 85% to about 99% sequence identity to an amino acid sequence as set forth in SEQ ID NO. : 65.
- the recombinant adenoviral vector comprises a shaft portion comprising an amino acid sequence from about 85% to about 87.5%, about 85% to about 90%, about 85% to about 91%, about 85% to about 92%, about 85% to about 93%, about 85% to about 94%, about 85% to about 95%, about 85% to about 96%, about 85% to about 97%, about 85% to about 98%, about 85% to about 99%, about 87.5% to about 90%, about 87.5% to about 91%, about 87.5% to about 92%, about 87.5% to about 93%, about 87.5% to about 94%, about 87.5% to about 95%, about 87.5% to about 96%, about 87.5% to about 97%, about 87.5% to about 98%, about 87.5% to about 99%, about 90% to about 91%, about 90% to about 92%, about 90% to about 93%, about 90% to about 94%, about 90% to about 95%, about 90% to about 95%,
- the recombinant adenoviral vector comprises a sequence encoding a knob portion from an SAd3 fiber protein.
- the recombinant adenoviral vector comprises a knob portion comprising an amino acid sequence with at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, atleast about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 98.5%, at least about 99%, at least about 99.5%, or atleast about 99.9% sequence identity to an amino acid sequence as set forth in SEQ ID NO.: 66.
- the recombinant adenoviral vector comprises a knob portion comprising an amino acid sequence with at most about 99.9%, at most about 99.5%, at most about 99%, atmost about 98.5%, at most about 98%, at most about 97%, atmost about 96%, atmost about95%, atmostabout94%, atmostabout93%, at most about 92%, atmost about 91%, or at most about 90% sequence identity to an amino acid sequence as set forth in SEQ ID NO.: 66.
- the recombinant adenoviral vector comprises a knob portion comprising an amino acid sequence from about 85% to about 99% sequence identity to an amino acid sequence as set forth in SEQ ID NO. : 66.
- the recombinant adenoviral vector comprises a knob portion comprising an amino acid sequence from about 85% to about 87.5%, about 85%to about 90%, about 85% to about 91%, about 85% to about 92%, about 85% to about 93%, about 85% to about 94%, about 85% to about 95%, about 85% to about 96%, about 85 % to ab out 97% , ab out 85 % to ab out 98 % , ab out 85 % to ab out 99 % , ab out 87.5% to ab out 90%, about 87.5% to about 91%, about 87.5% to about 92%, about 87.5% to about 93%, about 87.5% to about 94%, about 87.5% to about 95%, about 87.5% to about 96%, about 87.5% to about 97%, about 87.5% to about 98%, about 87.5% to about 99%, about 90% to about 91%, about 90% to about 92%, about 90% to about 93%, about 85% to about
- the recombinant adenoviral vector comprises a sequence encoding a tail portion from an SAd3 fiber protein.
- the recombinant adenoviral vector comprises a tail portion comprising an amino acid sequence with at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, atleast about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 98.5%, at least about 99%, at least about 99.5%, or atleast about 99.9% sequence identity to an amino acid sequence as set forth in SEQ ID NO.: 64.
- the recombinant adenoviral vector comprises a tail portion comprising an amino acid sequence with at most about 99.9%, at most about 99.5%, at most about 99%, at most about 98.5%, at most about 98%, at most about 97%, at most about 96%, atmost about95%, atmostabout94%, atmostabout93%, at most about 92%, atmost about 91%, or at most about 90% sequence identity to an amino acid sequence as set forth in SEQ ID NO.: 64.
- the recombinant adenoviral vector comprises a tail portion comprising an amino acid sequence from about 85% to about 99% sequence identity to an amino acid sequence as set forth in SEQ ID NO. : 64.
- the recombinant adenoviral vector comprises a tail portion comprising an amino acid sequence from about 85% to about 87.5%, about 85%to about 90%, about 85% to about 91%, about 85% to about 92%, about 85% to about 93%, about 85% to about 94%, about 85% to about 95%, about 85% to about 96%, about 85 % to ab out 97% , ab out 85 % to ab out 98 % , ab out 85 % to ab out 99 % , ab out 87.5% to ab out 90%, about 87.5% to about 91%, about 87.5% to about 92%, about 87.5% to about 93%, about 87.5% to about 94%, about 87.5% to about 95%, about 87.5% to about 96%, about 87.5% to about 97%, about 87.5% to about 98%, about 87.5% to about 99%, about 90% to about 91%, about 90% to about 92%, about 90% to about 93%, about 85% to about
- the recombinant adenoviral vector comprises a tail portion comprising an amino acid sequence as set forth in SEQ ID NO.: 64. [0256] In some embodiments, the recombinant adenoviral vector comprises a sequence encoding a shaft portion from an Ad35 fiber protein.
- the recombinant adenoviral vector comprises a shaft portion comprising an amino acid sequence with at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, atleast about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 98.5%, at least about 99%, at least about 99.5%, or atleast about 99.9% sequence identity to an amino acid sequence as set forth in SEQ ID NO. : 53.
- the recombinant adenoviral vector comprises a shaft portion comprising an amino acid sequence with at most about 99.9%, at most about 99.5%, at most about 99%, atmost about 98.5%, at most about 98%, at most about 97%, atmost about 96%, atmost about95%, atmostabout94%, atmostabout93%, at most about 92%, atmost about 91%, or at most about 90% sequence identity to an amino acid sequence as set forth in SEQ ID NO. : 53.
- the recombinant adenoviral vector comprises a shaft portion comprising an amino acid sequence from about 85% to about 99% sequence identity to an amino acid sequence as set forth in SEQ ID NO. : 53.
- the recombinant adenoviral vector comprises a shaft portion comprising an amino acid sequence from about 85% to about 87.5%, about 85%to about 90%, about 85% to about 91%, about 85% to about 92%, about 85% to about 93%, about 85% to about 94%, about 85% to about 95%, about 85% to about 96%, about 85 % to ab out 97% , ab out 85 % to ab out 98 % , ab out 85 % to ab out 99 % , ab out 87.5% to ab out 90%, about 87.5% to about 91%, about 87.5% to about 92%, about 87.5% to about 93%, about 87.5% to about 94%, about 87.5% to about 95%, about 87.5% to about 96%, about 87.5% to about 97%, about 87.5% to about 98%, about 87.5% to about 99%, about 90% to about 91%, about 90% to about 92%, about 90% to about 93%, about 85% to about
- the recombinant adenoviral vector comprises a sequence encoding a knob portion from an Ad35 fiber protein.
- the recombinant adenoviral vector comprises a knob portion comprising an amino acid sequence with at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, atleast about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 98.5%, at least about 99%, at least about 99.5%, or atleast about 99.9% sequence identity to an amino acid sequence as set forth in SEQ ID NO.: 54.
- the recombinant adenoviral vector comprises a knob portion comprising an amino acid sequence with at most about 99.9%, at most about 99.5%, at most about 99%, at most about 98.5%, at most about 98%, at most about 97%, at most about 96%, atmost about95%, atmostabout94%, atmostabout93%, atmostabout92%, atmost about 91%, or at most about 90% sequence identity to an amino acid sequence as set forth in SEQ ID NO.: 54.
- the recombinant adenoviral vector comprises a knob portion comprising an amino acid sequence from about 85% to about 99% sequence identity to an amino acid sequence as set forth in SEQ ID NO. : 54.
- the recombinant adenoviral vector comprises a knob portion comprising an amino acid sequence from about 85% to about 87.5%, about 85% to about 90%, about 85% to about 91%, about 85% to about 92%, about 85% to about 93%, about 85% to about 94%, about 85% to about 95%, about 85% to about 96%, about 85 % to ab out 97% , ab out 85 % to ab out 98 % , ab out 85 % to ab out 99 % , ab out 87.5% to ab out 90%, about 87.5% to about 91%, about 87.5% to about 92%, about 87.5% to about 93%, about 87.5% to about 94%, about 87.5% to about 95%, about 87.5% to about 96%, about 87.5% to about 97%, about 87.5% to about 98%, about 87.5% to about 99%, about 90% to about 91%, about 90% to about 92%, about 90% to about 93%, about 85% to about
- the disclosure provides a host cell, comprising one or more recombinant polypeptides, polynucleotides, vectors and/or the compositions described herein.
- Host cells are genetically engineered (transduced or transformed or transfected) with the vectors of this disclosure which can be, for example, a cloning vector or an expression vector.
- the vector can be, for example, in the form of a plasmid, a viral particle, a phage, etc.
- the host cell genetically engineered (transduced or transformed or transfected) with one or more vectors, e.g., one or more vectors disclosed herein.
- the host cell may be usedto produce an adenoviral stock.
- the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the polynucleotides.
- the culture conditions such as temperature, pH and the like, are those previously used with the host cell selected for expression and will be apparent to a person skilled in the arts.
- Suitable host cells for expression of a polypeptide include prokaryotes, yeast, insect or higher eukaryotic cells under the control of appropriate promoters.
- Prokaryotes include gram negative or gram positive organisms, forexampleE. coli orbacilli.
- Higher eukaryotic cells include established cell lines of mammalian origin. Cell-free translation systems could also be employed.
- Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are well known in the art (see Pouwels et al. , Cloning Vectors : A Lab oratory Manual, Elsevier, N.Y., 1985).
- the host cell can be mammalian for example human or mouse.
- the recombinant adenoviral vectors described herein may have a greater yield than a control vector system.
- a chimeric vector described herein comprising more than one adenovirus serotype may have a greater yield than a vector comprising one adenovirus serotype.
- a yield of viral vector (measured in infectious units, lU/ml) may be quantified from a cell lysate.
- a yield of viral vector may be at least about 1.0x10 3 IU/ml, atleastabout5.0xl0 3 IU/ml, atleast about 1.0x10 4 IU/ml, atleast about 5.0xl0 4 IU/ml, atleast about l .OxlO 5 lU/ml, at least about 5.0x10 5 lU/ml, at least about l .OxlO 6 lU/ml, at least about 5.0x10 6 lU/ml, atleast about 1 .0x10 7 lU/ml, at least about 5.0x10 7 lU/ml, at least about 1.0x10 8 IU/ml, atleastabout5.0xl 0 8 IU/ml, atleast about 1.0x10 9 IU/ml, atleast about 5.
- Ox 10 9 IU/ml atleast about 1.
- a yield of viral vector may be at most about l .Oxl O 15 lU/ml, 1 .0xl 0 14 IU/ml, l .OxlO 13 lU/ml, l .OxlO 12 lU/ml, 1 .Oxl O 11 lU/ml, l .Oxl O 10 lU/ml, at most about 5.
- OxlO 9 lU/ml at most about 1 .OxlO 9 lU/ml, at most about 5.
- Ox lO 7 lU/ml at most about 1.0xl0 7 IU/ml, at most about 5.0xl 0 6 IU/ml, at most about l .OxlO 6 lU/ml, at most about 5.
- OxlO 5 lU/ml at most about 1 .OxlO 5 lU/ml, at most about 5.
- Oxl O 4 lU/ml at most about 1 .Oxl O 4 lU/ml, at most about 5.
- a chimeric recombinant adenoviral vector described herein may have a lx, 1.5x, 2x, 2.5x, 3x, 3.5x, 4x, 4.5x, 5x, 6x, 7x, 8x, 9x, or l Ox greater yield than an adenoviral vector comprising one adenovirus serotype.
- the recombinant adenoviral vector described herein comprises a sequence with at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 98.5%, at least about 99%, at least about 99.5%, or at least about 99.9% sequence identity to a nucleotide sequence as set forth in SEQ ID NOs. : 177-18 l or 388-394.
- the recombinant adenoviral vector described herein comprises a sequence with at most about 99.9%, at most about 99.5%, at most about 99%, at most about 98.5%, at most about 98%, at most about 97%, at most about 96%, at mostabout95%, atmost about94%, atmost about93%, atmost about92%, atmostabout91%, at mostabout90%, atmost about85%, atmost about80%, or atmost about75% sequenceidentity to a nucleotide sequence as setforth in SEQ ID NOs. : 177-181 or 388-394.
- the recombinant adenoviral vector described herein comprises a sequence from about 70% to about 99% sequence identity to a nucleotide sequence as set forth in SEQ ID NOs. : 177-181 or 388-394.
- the recombinant adenoviral vector described herein comprises a sequence from about 70% to about 75%, about 70% to about 80%, about 70% to about 85%, about 70% to about 90%, about 70% to about 92%, about 70% to about 94%, about 70% to about 95%, about 70% to about 96%, about 70% to about 97%, about 70% to about 98%, about 70% to about 99 % , ab out 75 % to ab out 80 %, ab out 75 % to ab out 85% , ab out 75% to ab out 90 % , ab out 75% to about 92%, about 75% to about 94%, about 75% to about 95%, about 75% to about 96%, about 75% to about 97%, about 75% to about 98%, about 75% to about 99%, about 80% to about 85%, about 80% to about 90%, about 80% to about 92%, about 80% to about 94%, about 80% to about 95%, about 80% to about 96%, about 80% to about 97%, about 80% to about 99%,
- a recombinant adenoviral vector described herein may comprise a nucleotide sequence having at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 98.5%, at least about 99%, at least about 99.5%, or at least about 99.9% sequence identity to a sequence as set forth in Table 8.
- a recombinant adenoviral vector described herein may comprise a combination of serotypes as shown in Table 10.
- the recombinant viral vector describedherein comprises a single epitope, e.g., a single T cell epitope, e.g., a single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or a CD4+ epitope, e.g., the single cancerspecific CD8+ epitope is an immunogenic epitope, e.g., it elicits an immune response.
- binding of the single epitope, e.g., the single T cell epitope, e.g., the single cancerspecific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope to the MHC complex initiates a subsequent immune response, e.g., an inflating memory T cell response, e.g, an inflating memory CD8+ and/or CD4+ T cell response, e.g., an inflating memory CD8+ T cell response.
- a subsequent immune response e.g., an inflating memory T cell response, e.g, an inflating memory CD8+ and/or CD4+ T cell response, e.g., an inflating memory CD8+ T cell response.
- the inflating memory T cell response e.g., the inflating memory CD8+ and/or CD4+ T cell response, e.g., the inflating memory CD8+ T cell response targets cancer cells.
- the recombinant polypeptide comprising the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope binds an HLA type encoding an MHC of a patient’s cancer cells.
- the recombinant polypeptide comprising the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or epitope, e.g., the single cancerspecific CD8+ epitope does not bind to an HLA type encoding an MHC of a patient’ s non-cancer cells.
- the recombinant polypeptide comprising the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope does not cause T cell exhaustion while binding to the HLA type encoding the MHC of the patient’s cancer cells.
- the recombinant polypeptide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in any of the SEQ ID NOs. : 72-106.
- the recombinant polypeptide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence with at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 98.5%, at least about 99%, at least about 99.5%, or atleast about 99.9% sequence identity to an amino acid sequence as set forth in any of the SEQ ID NOs.
- the recombinant polypeptide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence with at most about 99.9%, at most about 99.5%, at most about 99%, at most about 98.5%, at most about 98%, at most about 97%, at most about 96%, at most about 95%, at most about 94%, at most about 93%, at most about 92%, at most about 91%, or at most about 90% sequence identity to an amino acid sequence as set forth in any of the SEQ ID NOs.: 72-106.
- the recombinant polypeptide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence from about 85% to about 99% sequence identity to an amino acid sequence as set forth in any of the SEQ ID NOs.: 72-106.
- the recombinant polypeptide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence from about 85% to about 87.5%, about 85% to about 90%, about 85% to about 91%, about 85% to about 92%, about 85% to about 93%, about 85% to about 94%, about 85% to about 95%, about 85% to about 96%, about 85% to about 97%, about 85% to about 98%, about 85% to about 99%, about 87.5% to about 90%, about 87.5% to about 91%, about 87.5% to about 92%, about 87.5% to about 93%, about 87.5% to about 94%, about 87.5% to about 95%, about 87.5% to about 96%, about 87.5% to about 97%, about
- the recombinant adenoviral vectors described herein may comprise a recombinant polypeptide (e.g., a single cancer-specific CD8+ epitope) as set forth in an amino acid sequences selected from SEQ ID NOs.: 72-106.
- a recombinant polypeptide e.g., a single cancer-specific CD8+ epitope
- the recombinant polypeptide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence with at least 1 , at least 2, or at least 3 amino acid mutations compared to an amino acid sequence as set forth in any of the SEQ ID NOs. : 72-106.
- a single epitope e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence with at least 1 , at least 2, or at least 3 amino acid mutations compared to an amino acid sequence as set forth in any of the SEQ ID NOs. : 72-106.
- the recombinant polypeptide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence with at most 3 , at most 2, or at most 1 amino acid mutation(s) compared to an amino acid sequence as set forth in any of the SEQ ID NOs. : 72-106.
- a single epitope e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence with at most 3 , at most 2, or at most 1 amino acid mutation(s) compared to an amino acid sequence as set forth in any of the SEQ ID NOs. : 72-106.
- the recombinantpolypeptide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence selected from
- the recombinant polypeptide comprises more than one, e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 30 epitopes, e.g., T cell epitopes, e.g., cancer-specific CD8+ and/or CD4+ epitopes, e.g., cancer-specific CD8+ epitopes.
- the recombinant polypeptide comprises at least two epitopes, e.g., at least T cell epitopes, e.g., at least cancerspecific CD8+ and/or CD4+ epitopes, e.g., at least cancer-specific CD8+ epitopes.
- the epitopes are identical. In some embodiments, the epitopes are different. In a non-limiting example, in some embodiments, a recombinant polypeptide disclosed herein may comprise three epitopes e.g., T cell epitopes, e.g., cancer-specific CD8+ and/or CD4+ epitopes, e.g., cancer-specific CD8+ epitopes of which two are identical and one different.
- T cell epitopes e.g., cancer-specific CD8+ and/or CD4+ epitopes, e.g., cancer-specific CD8+ epitopes of which two are identical and one different.
- the first epitope e.g., the first T cell epitope, e.g., the first cancer-specific CD8+ and/or CD4+ epitope, e.g., the first cancer-specific CD8+ epitope comprises an amino acid sequence setforth in any of SEQ ID NOs. : 72-106.
- the second epitope e.g, the second T cell epitope, e.g., the second cancer-specific CD8+ and/or CD4+ epitope, e.g., the second cancer-specific CD8+ epitope comprises an amino acid sequence setforth in any of SEQ ID NOs.: 72-106.
- the third epitope e.g., the third T cell epitope, e.g., the third cancer-specific CD8+ and/or CD4+ epitope, e.g., the third cancer-specific CD8+ epitope comprises an amino acid sequence set forth in any of SEQ ID NOs.: 72-106.
- the fourth epitope e.g., the fourth T cell epitope, e.g., the fourth cancer-specific CD8+ and/or CD4+ epitope, e.g., the fourth cancer-specific CD8+ epitope comprises an amino acid sequence setforth in any of SEQ ID NOs. : 72-106.
- the fifth epitope e.g., the fifth T cell epitope, e.g., the fifth cancer-specific CD8+ and/or CD4+ epitope, e.g., the fifth cancer-specific CD8+ epitope comprises an amino acid sequence set forth in any of SEQ ID NOs.: 72-106.
- the sixth epitope e. g., the sixth T cell epitope, e.g., the sixth cancer-specific CD8+ and/or CD4+ epitope, e.g., the sixth cancer-specific CD8+ epitope comprises an amino acid sequence set forth in any of SEQ ID NOs.: 72-106.
- the seventh T cell epitope e.g., the seventh cancer-specific CD8+ and/or CD4+ epitope, e.g., the seventh cancer-specific CD8+ epitope comprises an amino acid sequence set forth in any of SEQ ID NOs. : 72-106.
- the eighth epitope e.g., the eighth T cell epitope, e.g., the eighth cancer-specific CD8+ and/or CD4+ epitope, e.g., the eighth cancer-specific CD8+ epitope comprises an amino acid sequence set forth in any of SEQ ID NOs.: 72-106.
- the ninth epitope e.g., the ninth T cell epitope, e.g, the ninth cancer-specific CD8+ and/or CD4+ epitope, e.g., the ninth cancer-specific CD8+ epitope comprises an amino acid sequence set forth in any of SEQ ID NOs.: 72-106.
- the tenth epitope e.g., the tenth T cell epitope, e.g., the tenth cancer-specific CD8+ and/or CD4+ epitope, e.g., the tenth cancer-specific CD8+ epitope comprises an amino acid sequence set forth in any of SEQ ID NOs.: 72-106.
- the epitope e.g., the T cell epitope, e.g., the cancer-specific CD8+ and/or CD4+ epitope, e.g., the cancer-specific CD8+ epitope is derived from a tumor associated antigen (TAA).
- TAA tumor associated antigen
- a TAA is an antigenic product produced by a cancer and it provides a biomarker for targeted identification of a tumor.
- TAAs can be broadly categorized into aberrantly expressed self-antigens, mutated self-antigens and tumor specific antigens. As such, the TAA may be upregulated or over-expressed in the cancer cell.
- the TAA may be mutated within the cancer cell.
- the TAA may be specific for the cancer cell and only expressed within the cancer cell, this may also be referred to as a tumor specific antigen.
- the TAA may be overexpressed antigens that have lower expression levels in normal tissues and high expression levels in tumors.
- the TAA is associated with aggressive tumor behavior and/or show cancer-restricted expression. In some embodiments, the TAA enhances a CD8+ T cell immune response.
- the tumor-associated antigen is a tumor specific antigen.
- the single epitope e.g., the single T cell epitope, e.g., the single cancerspecific CD8+ epitope is a cancertestis antigen, e.g., a cancer testis antigen thatis NY-ESO-1 or a member of theMAGE-A family.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ epitope is an endogenous retroviral protein.
- the TAA is associated with aggressive tumor behavior and/or show cancer-restricted expression.
- the TAA enhances a CD4+ T cell immune response.
- the tumor-associated antigen is a tumor specific antigen.
- the single epitope e.g., the single T cell epitope, e.g., the single cancerspecific CD4+ epitope is a cancertestis antigen, e.g., a cancer testis antigen that is NY-ESO-1 or a member of theMAGE-A family.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD4+ epitope is an endogenous retroviral protein.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is mutated in a cancer cell.
- the cancer specific the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is overexpressed in a cancer cell.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+epitope is or comprises a non-codingtumor specific epitope.
- non-coding tumor specific epitope refers to a peptide found on a cancer cell, wherein the peptide is derived from a nucleotide sequence that is epigenetically suppressed in healthy cells. These peptide sequences are aberrantly expressed within tumor cells.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is not a cryptic epitope.
- a “cryptic epitope” refers to refers to an epitope which is not immunogenic in immunocompetent individuals.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a cryptic epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope can be a viral epitope optionally associated with a virally driven cancer.
- the virally driven cancer may be a cancer caused by HPV (human papilloma virus), HTLV (human T-lymphotropic virus), or EBV (Epstein Barr virus).
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ epitope is or is derived from a tumor associated antigen selected from the group comprising: TRP-1, CEA, TAG-72, 9D7, Ep-CAM, EphA3, telomerase, mesothelin, SAP- 1 Melan-A/MART-1, tyrosinase, CLPP, cyclin-Al, cyclin-Bl MAGE-A1, MAGE-CI, MAGE- 02, SSX2, XAGElb/GAGED2a, CD45, glypican-3, IGF2B3, kallikrein-4, KIF20A, lengsin, meloe, MUC5AC, survivin, PRAME, SSX-2, NY-ESO-1/LAGE1, gp70, MC1R, TRP-1/-2, p- catenin,
- a tumor associated antigen selected
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD4+ epitope is or is derived from a tumor associated antigen selected from the group comprising: EBNA2, TARP, EBV LMP1, STEAP, WT1, HTLV-1 env, HER2/neu, MAGE-A3, BRAF-V600E, NY-ESO-1, or gplOO.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is an epitope associated with colorectal cancer, prostate cancer, esophageal cancer, liver cancer, renal cancer, lung cancer, bladder cancer, breast cancer, pancreatic cancer, brain cancer, hepatocellular cancer, glioblastoma, lymphoma, leukemia, gastric cancer, cervical cancer, ovarian cancer, thyroid cancer, melanoma, carcinoma, head and neck cancer, skin cancer, nasopharyngeal cancer, Epstein Barr driven cancers, Human Papilloma virus driven cancers or soft tissue sarcoma.
- the cancer is a primary cancer.
- the cancer is a relapsed and/or a refractory cancer.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is an esophageal cancer-specific CD8+ T cell epitope.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a colorectal cancer-specific CD8+ T cell epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a prostate cancerspecific CD8+ T cell epitope.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a liver cancer-specific CD8+ T cell epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a renal cancer-specific CD8+ T cell epitope.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancerspecific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a breast cancer-specific CD8+ T cell epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancerspecific CD8+ epitope is a bladder cancer-specific CD8+ T cell epitope.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a pancreatic cancer-specific CD8+ T cell epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a lung cancer-specific CD8+ T cell epitope.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a brain cancer-specific CD8+ T cell epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a hepatocellular cancer-specific CD8+ T cell epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancerspecific CD8+ epitope is a gastric cancer-specific CD8+ T cell epitope.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a cervical cancer-specific CD8+ T cell epitope.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is an ovarian cancer-specific CD8+ T cell epitope.
- the single epitope e.g, the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a thyroid cancer-specific CD8+ T cell epitope.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a CD8+T cell epitope specific for melanoma.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a CD8+ T cell epitope specific for lymphoma.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a CD8+ T cell epitope specific for leukemia.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope specific for carcinoma.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancerspecific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a CD8+ T cell epitope specific for head and neck cancer.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a CD8+ T cell epitope specific for glioblastoma.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a CD8+T cell epitope specific for skin cancer.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is a CD8+ T cell epitope specific for soft tissue sarcoma.
- the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+epitope is a CD8+ T cell epitope specific for nasopharyngeal cancer.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope may be an epitope for a benign tumor.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope may be an epitope for a pre-malignant tumor.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+epitope may be an epitope of smolderingneoplasms.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope maybe an epitope for dysplasia.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope may be an epitope for metaplasia.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope may be a private epitope.
- the term “private epitope” refers to an epitope which is found exclusively on a single antigen in a cancer of a single person.
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope may be a public epitope.
- the term “public epitope” refers to an epitope that is found on a cancer of two or more people.
- the recombinant polypeptide comprising the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is capable of inducing an epitope-specific T cell response.
- the recombinant polypeptide comprising the single epitope, e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancerspecific CD8+ epitope is capable of inducing an inflating memory CD8+ T cell response.
- the recombinant polypeptide comprising the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope
- APCs antigen presenting cells
- the single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope may be a neoepitope.
- the term “neoepitope” refers to epitopes which have arisen through mutations within the tumor cells, in particular somatic or passenger mutations may lead to the production of a neoepitope.
- single epitope e.g., the single T cell epitope, e.g., the single cancer-specific CD8+ and/or CD4+ epitope, e.g., the single cancer-specific CD8+ epitope is not a neoepitope.
- the recombinant vectors described herein comprise a polynucleotide encoding a recombinant polypeptide comprising a single epitope, e.g., a single T cell epitope, e.g., a single cancer-specific CD8+ and/or CD4+ epitope, e.g., a single cancerspecific CD8+ epitope described herein.
- a single epitope e.g., a single T cell epitope, e.g., a single cancer-specific CD8+ and/or CD4+ epitope, e.g., a single cancerspecific CD8+ epitope described herein.
- a polynucleotide encodes a recombinant polypeptide comprising a single epitope, e.g., a single T cell epitope, e.g., a single cancer-specific CD8+ and/or CD4+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in any of SEQ ID NOs.: 72-106.
- Polynucleotides includes, for example, genomic DNA, cDNA, RNA, e.g., mRNA, and DNA-RNA hybrid molecules. Polynucleotides can be naturally occurring, recombinant, or synthetic. In addition, polynucleotides can be single-stranded, double-stranded or triple-stranded. In certain embodiments, polynucleotides can be modified. In the case of a double-stranded polymer, “nucleic acid” can refer to either or both strands of the molecule. In some embodiments, the polynucleotide is provided as DNA.
- the polynucleotide is provided as RNA, e.g., an mRNA.
- a polynucleotide disclosed herein comprises a nucleic acid sequence as set forth in any of SEQ ID NOs. : 107-141.
- a polynucleotide disclosed herein comprises a nucleic acid sequence as shown in Table 5.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in any of the SEQ ID NOs. : 107- 141.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an nucleotide sequence with at least about 90%, at least about 91 %, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about97%, atleastabout98%, atleastabout98.5%, atleastabout99%, atleastabout99.5%, or at least about 99.9% sequence identity to an nucleotide sequence as set forth in any of the SEQ ID NOs.: 107-141.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an nucleotide sequence with at most about 99.9%, at most about 99.5%, at most about 99%, at most about 98.5%, at most about 98%, at most about 97%, atmost about96%, atmostabout95%, atmostabout94%, atmostabout93%, atmostabout 92%, at most about 91%, or at most about 90% sequence identity to an nucleotide sequence as set forth in any of the SEQ ID NOs.: 107-141.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an nucleotide sequence from about 85% to about 99% sequence identity to an nucleotide sequence as set forth in any of the SEQ ID NOs.: 107-141.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an nucleotide sequence from about 85% to about 87.5%, about 85%to about 90%, about 85% to about 91%, about 85% to about 92%, about 85% to about 93%, about 85% to about 94%, about 85% to about 95%, about 85% to about 96%, about 85 % to ab out 97% , ab out 85 % to ab out 98 % , ab out 85 % to ab out 99 % , ab out 87.5% to ab out 90%, about 87.5% to about 91%, about 87.5% to about 92%, about 87.5% to about 93%, about 87.5% to about 94%, about 87.5% to about 95%, about 87.5% to about
- the polynucleotides disclosed herein can be codon optimized, usage bias hasbeen reported for numerous organisms, from viruses to eukaryotes. Since the genetic code is degenerate (i.e., each amino acid can be coded by on average three different codons), the DNA sequence can be modifiedby synonymous nucleotide substitutions without altering the amino acid sequence of the encoded protein. Such synonymous codon optimization has been performed for the purpose of optimizing expression in a desired host, as described in the scientific literature and in patent documents.
- the nucleic acid described herein can be modified to improve cloning efficiency.
- the nucleic acids described herein are subjected to codon optimization to increase the efficiency of gene expression, e.g., SEQ ID NOs. : 107-141. are subjected to codon optimization.
- the polypeptide disclosed herein are encoded by polynucleotides whose sequence has been codon optimized for expression in a mammalian cell, e.g., expressional in a human cell. Exemplary codon optimized sequences are disclosed in Table 6.
- polypeptides encoded by a codon optimized nucleotide have increased expression in a mammalian cell, e.g., a human cell compared to a polypeptide that is encoded by a non-codon optimized polynucleotide.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 72.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancerspecific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 74.
- a recombinant adenoviralvectordescribedherein can comprise a single epitope, e.g., a single T cell epitope, e.g, the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 75.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 76.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 77.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 78.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 79.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancerspecific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 80.
- a recombinant adenoviralvectordescribedherein can comprise a single epitope, e.g., a single T cell epitope, e.g, the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 81.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 82.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 83.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 84.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 85.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancerspecific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 86.
- a recombinant adenoviralvectordescribedherein can comprise a single epitope, e.g., a single T cell epitope, e.g, the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 87.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 88.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 89.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 90.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 91.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancerspecific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 92.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 94.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 95.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 96.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 97.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancerspecific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 98.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g, the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 99.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 100.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 101.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 102.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 103.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO. : 104.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in SEQ ID NO.: 105.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an amino acid sequence as set forth in any of the SEQ ID NOs. : 72- 106.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an nucleotide sequence with at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about97%, atleastabout98%, atleastabout98.5%, atleastabout99%, atleastabout99.5%, or at least about 99.9% sequence identity to an nucleotide sequence as set forth in any of the SEQ ID NOs.: 142-176.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an nucleotide sequence with at most about 99.9%, at most about 99.5%, at most about 99%, at most about 98.5%, at most about 98%, at most about 97%, atmost about96%, atmostabout95%, atmostabout94%, atmostabout93%, atmost about 92%, at most about 91%, or at most about 90% sequence identity to an nucleotide sequence as set forth in any of the SEQ ID NOs.: 142-176.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an nucleotide sequence from about 85% to about 99% sequence identity to an nucleotide sequence as set forth in any of the SEQ ID NOs.: 142-176.
- the polynucleotide comprises a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope comprising an nucleotide sequence from about 85% to about 87.5%, about 85%to about 90%, about 85% to about 91%, about 85% to about 92%, about 85% to about 93%, about 85% to about 94%, about 85% to about 95%, about 85% to about 96%, about 85 % to ab out 97% , ab out 85 % to ab out 98 % , ab out 85 % to ab out 99 % , ab out 87.5% to ab out 90%, about 87.5% to about 91%, about 87.5% to about 92%, about 87.5% to about 93%, about 87.5% to about 94%, about 87.5% to about 95%, about 87.5% to about
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 107 or 142.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 108 or 143.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 109 or 144.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 110 or 145.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancerspecific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: I l l or 146.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as setforth in SEQ ID NO. : 112 or 147.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 113 or 148.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as setforth in SEQ ID NO. : 114 or 149.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 115 or 150.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancerspecific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 116 or 151.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as setforthin SEQ ID NO. : 117 or 152.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 118 or 153.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 119 or 154.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as setforthin SEQ ID NO.: 120 or 155.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancerspecific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 121 or 156.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 122 or 157.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 123 or 158.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 124 or 159.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as setforthin SEQ ID NO. : 125 or 160.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancerspecific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 126 or 161.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 127 or 162.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 128 or 163.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 129 or 164.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 130 or 165.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancerspecific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 131 or 166.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 132 or 167.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 133 or 168.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 134 or 169.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 135 or 170.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer- specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 136 or 171.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 137 or 172.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 138 or 173.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO. : 139 or 174.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancer-specific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 140 or 175.
- a recombinant adenoviral vector described herein can comprise a single epitope, e.g., a single T cell epitope, e.g., the single cancer-specific CD4+ and/or CD8+ epitope, e.g., a single cancerspecific CD8+ epitope encoded by a polynucleotide having a sequence as set forth in SEQ ID NO.: 141 or 176.
- a polynucleotide disclosed herein comprises multiple smaller and discrete nucleotide sequences that are typically heterologous and exhibit different and measurable function(s), a promoter sequence, a translation initiating sequence, a start codon, a polyadenylation sequence, a stop codon, a nucleotide sequence encoding an epitope, e.g., a T cell epitope, e.g., a cancer-specific CD8+ and/or CD4+ epitope, e.g., a cancer-specific CD8+ epitope, e.g., an epitope comprising an amino acid sequence as set forth in any of SEQ ID NOs.
- the polynucleotide is codon optimized, e.g., codon optimized for optimal expression in a mammalian cell, e.g., a human cell.
- the polynucleotide e.g., the RNA, e.g., the mRNA encoding the epitope, e.g., the T cell epitope, e.g., the cancer-specific CD8+ and/or CD4+ epitope, e.g., the cancer-specific CD8+ epitope is introduced into cells.
- in vitro transcribed RNA can be introduced to a cell as a form of transient transfection.
- the polynucleotide can include some or all of the 5’ and/or 3’ untranslated regions (UTRs).
- the polynucleotide can include exons and introns.
- the DNA to be used forPCR is a human nucleic acid sequence.
- theDNAto be used for PCR is a human nucleic acid sequence including the 5’ and 3 ’ UTRs.
- the DNA can alternatively be an artificial DNA sequence that is not normally expressed in a naturally occurring organism.
- An exemplary artificial DNA sequence is one that contains portions of genes that are ligated together to form an open reading frame that encodes a fusion protein. The portions of DNA that are ligated together can be from a single organism or from more than one organism.
- the 5’ UTR can contain the Kozak sequence of the endogenous nucleic acid.
- a consensus Kozak sequence can be redesigned by adding the 5 ’ UTR sequence.
- Kozak sequences can increase the efficiency of translation of some RNA transcripts.
- the 5 ’ UTR can be 5 ’UTR of an RNA virus whose RNA genome is stable in cells.
- various nucleotide analogues can be used in the 3 ’ or 5 ’ UTR to impede exonuclease degradation of the mRNA.
- administration of one or more epitopes may demonstrate a level of immunogenicity.
- a vector described herein e.g., a chimeric vector
- comprising an epitope may be capable of inducing production of CD8+ T cells characterized by markers.
- the markers maybe markers indicative of memory inflation.
- the vector described herein (e.g., a chimeric vector) comprising an epitope maybe capable of inducing production of T cells (e.g., CD8+ T cells) positive for CD44, CX3CR1, KLRG-1, or any combination thereof.
- administration of a vector described herein comprising an epitope can be capable of inducing production of CD8+/CX3CR1+/KLRG- 1+/CD44+ T cells, CD8+/CX3CR1+/KLRG-1+/CD62L- T cells, CD8+/CX3CR1+/KLRG- 1+/CD44+/CD62L- T cells, CD8+/CX3CR1+/KLRG-1+/CD44+/CD62L-/CD27-/CD127- T cells, CD8+/CX3CRl+/KLRG-l+/CD44+/CD62L-/CD27(low)/CD127- T cells,
- CD8+/CX3CR1+/KLRG-1+/CD44+/CD62L-/CD27- /CD127(low) T cells or
- administration of a vector described herein comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which at least about 50%, at least about 55%, at least about 60%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or greater than about 95% of the T cells are CD44+/CD62L- T cells.
- a population of induced CD44+/CD62L- T cells may be maintained over a period of time.
- the period of time may be at least about 1 day, at least about 2 days, at least about 3 days, at least about 4 days, at least about 5 days, at least about 6 days, at least about 7 days, at least about 8 days, at least about 9 days, at least about 10 days, at least about 15 days, at least about 20 days, at least about 25 days, at least about 30 days, at least about 40 days, at least about 50 days, at least about 60 days, at least about 70 days, at least about 80 days, at least about 90 days, at least about 100 days, or greater than about 100 days.
- Maintenance of the T cell population may refer to a population that does not fluctuate (e.g., increase and/or decrease) more than 70% or 80% from a starting level.
- administration of a vector described herein comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or greater than about 95% of the T cells are CX3CR1+ T cells.
- a population of induced CX3CR1+ T cells may be maintained over a period of time.
- the period of time may be at least about 1 day, at least about 2 days, at least about 3 days, at least about 4 days, at least about 5 days, at least about 6 days, at least about 7 days, at least about 8 days, at least about 9 days, at least about 10 days, at least about 15 days, at least about 20 days, atleast about 25 days, atleast about 30 days, at least about 40 days, at least about 50 days, at least about 60 days, at least about 70 days, at least about 80 days, at least about 90 days, at least about 100 days, or greater than about 100 days.
- Maintenance of the T cell population may refer to a population that does not fluctuate (e.g., increase and/or decrease) more than 70% or 80% from a starting level.
- administration of a vector described herein comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or greater than about95% of the T cells areKLRG-l+T cells.
- a population ofinduced KLRG-1+ T cells may be maintained over a period of time.
- the period of time may be at least about 1 day, at least about 2 days, at least about 3 days, at least about 4 days, at least about 5 days, at least about 6 days, at least about 7 days, at least about 8 days, at least about 9 days, at least about 10 days, at least about 15 days, at least about 20 days, at least about 25 days, at least about 30 days, at least about 40 days, at least about 50 days, at least about 60 days, at least about 70 days, at least about 80 days, at least about 90 days, at least about 100 days, or greater than about 100 days.
- Maintenance of the T cell population may refer to a population that does not fluctuate (e.g., increase and/or decrease) more than 70% or 80% from a starting level.
- the inflating memory CD8+ T cells induced by the vector described herein may lack one or more markers of T cell exhaustion.
- T cell exhaustion can occur from excessive TCR (T cell receptor) stimulation.
- Markers of T cell exhaustion can includebut are notlimited to upregulation of markers such as PD-1, Tim-3, or Lag-3.
- the inflating memory CD8+ T cells may lack or demonstrate low expression of markers selected from the group consisting of, but notlimited to, PD-1, Tim-3, and/or Lag-3.
- the exhaustion markers may be present prior to, concurrently with, or subsequent to tumor challenge.
- administration of a vector described herein comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which atleast about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, atleast about 50%, or greater than about 50% of the T cells are PD-1 + T cells.
- T cells e.g., CD8+ T cells
- administration of a vector described herein comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which at most about 50%, at most about 40%, at most about 35%, at most about 30%, at most about 25%, at most about 20%, at most about 10%, at most about 5%, at most about 4%, at most about 3%, at most about 2%, at most about 1%, or less than about 1% of the T cells are PD-1+ T cells.
- T cells e.g., CD8+ T cells
- administration of Malawi ⁇ ество comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which atleast about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, or greater than about 50% of the T cells are Tim-3+ T cells.
- T cells e.g., CD8+ T cells
- T cells e.g., CD8+ T cells
- administration of a vector described herein comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which at most about 50%, at most about 40%, at most about 35%, at most about 30%, at most about 25%, at most about 20%, at most about 10%, at most about 5%, at most about 4%, at most about 3%, at most about 2%, at most about 1%, or less than about 1% ofthe T cells are Tim-3+ T cells.
- T cells e.g., CD8+ T cells
- administration of a vector described herein comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which atleast about 1%, at least about 2%, at least about 3%, at least about 4%, at least about 5%, at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, atleast about 50%, or greater than about 50% of the T cells are Lag3+ T cells.
- T cells e.g., CD8+ T cells
- administration of a vector described herein comprising an epitope can be capable of inducing a population of T cells (e.g., CD8+ T cells) in which at most about 50%, at most about 40%, at most about 35%, at most about 30%, at most about 25%, at most about 20%, at most about 10%, at most about 5%, at most about 4%, at most about 3%, at most about 2%, atmost about 1%, or less than about 1% of the T cells are Lag3+ T cells.
- T cells e.g., CD8+ T cells
- a subject administered a vector described herein (e.g., a chimeric vector) comprising an epitope may demonstrate a tumor volume of atmost about 200 mm 3 , at most about 150 mm 3 , at most about 100 mm 3 , at most about 75 mm 3 , at most about 50 mm 3 , at most about 40 mm 3 , at most about 30 mm 3 , at most about 20 mm 3 , at most about 10 mm 3 , at most about 5 mm 3 , at most about 4 mm 3 , at most about 3 mm 3 , at most about 2 mm 3 , at most about 1 mm 3 , or less than about 1 mm 3 .
- administration of a chimeric vector may result in decreased tumor volume in a subject, compared to a tumor volume resulting from administration of no vector (e.g., a naive subject) or administration of a vector that does not comprise a single cancer-specific epitope.
- a subjectwith atumor administered a vector or composition described herein may have a tumor volume of at least about 2x, at least about 3x, at least about 4x, at least about 5x, at least about 1 Ox, at least about 20x, at least about 30x, at least about 40x, at least about 5 Ox, at least about lOOx, atleast about 150x, at least about200x, or greaterthan about200x less than a tumor volume of a subject administered no vector or administered a vector that does not comprise a single cancer-specific epitope.
- a vector described herein (e.g., a chimeric vector) comprising an epitope can be capable of extending survival in a subject expressing a tumor.
- the vectors described herein may provide a greater percentage of survival in a subject expressing a tumor compared to a subject not administered a vector comprising a single cancer-specific epitope.
- the vectors described herein may provide comparable or greater anti-tumor activity than a non-chimeric vector (e.g, a vector of only one serotype).
- a chimeric vector described herein can provide an extended survival period in a subject expressing a tumor and the survival period may be any number of days.
- a subject administered a chimeric vector described herein may survive for any number of days longer than a subject administered no vector and/or a subject administered a vector comprising an epitope other than a single cancer-specific epitope (e.g., a viral epitope or non-cancer related epitope).
- a subject administered a chimeric vector described herein may survive for at least about 1 day longer, at least about 2 days longer, at least about 3 days longer, at least about 4 days longer, at least about 5 days longer, at least about 6 days longer, at least about?
- a subject administered a chimeric vector described herein may survive for at most about 50 days longer, at most about 40 days longer, at most about 35 days longer, at most about 30 days longer, at most about 25 days longer, at most about 20 days longer, at most about 15 days longer, at most about 10 days longer, at most about 9 days longer, at most about 8 days longer, at most about 7 days longer, at most about 6 days longer, at most about 5 days longer, at most about 4 days longer, at most about 3 days longer, at most about 2 days longer, at most about 1 day longer, or less than about 1 day longer than a subject administered no vector or a subject administered a vector comprising an epitope other than a single cancer-specific epitope (e.g., a viral epitope or non- cancer related epitope).
- an epitope other than a single cancer-specific epitope e.g., a viral epitope or non- cancer related epitope.
- a subject administered a chimeric vector described herein may survive from about 1 day longer to about 50 days longer than a subject administered no vector or a subject administered a vector comprising an epitope other than a single cancer-specific epitope (e.g., a viral epitope or non-cancer related epitope).
- an epitope other than a single cancer-specific epitope e.g., a viral epitope or non-cancer related epitope.
- a subject administered a chimeric vector described herein may survive from about 1 day longer to about 2 days longer, about 1 day longer to about 3 days longer, about 1 day longer to about 4 days longer, about 1 day longer to about 5 days longer, about 1 day longer to about 8 days longer, about 1 day longer to about 10 days longer, about 1 day longer to about 15 days longer, about 1 day longer to about 20 dayslonger, about 1 day longer to about 30 days longer, about 1 day longer to about40 days longer, about 1 day longer to about 50 days longer, about 2 days longer to about 3 days longer, about 2 days longer to about 4 days longer, about 2 days longer to about 5 days longer, about 2 days longer to about 8 days longer, about 2 days longer to about 10 days longer, about 2 days longer to about 15 days longer, about 2 days longer to about 20 days longer, about 2 days longer to about 30 days longer, about 2 days longer to about 40 days longer, about 2 days longer to about 50 days longer, about 3 days longer to about 4 days longer, about 3 days longer to about 5 days longer, about 3 days longer to about 8 days
- the disclosure provides a pharmaceutical composition comprising the recombinant adenoviral vectors described herein.
- the disclosure provides a pharmaceutical composition comprising one or more polynucleotides, vectors, cells, or compositions disclosed herein and optionally further comprising a pharmaceutically acceptable carrier, excipient or additive.
- pharmaceutical composition refers to a composition formulated for pharmaceutical use.
- the pharmaceutical compositions may comprise an immunologically effective amount of one or more polynucleotides, vectors, cells, or compositions disclosed herein, and optionally one or more other components which are pharmaceutically acceptable.
- the pharmaceutical composition comprises at least one recombinant adenoviral vector, at least two recombinant adenoviral vectors, at least three recombinant adenoviral vectors, at least four recombinant adenoviral vectors, at least five recombinant adenoviral vectors, or more.
- the recombinant adenoviral vector may be a chimeric recombinant adenoviral vector.
- the pharmaceutical composition comprises the same chimeric recombinant adenoviral vector.
- the pharmaceutical composition comprises different chimeric recombinant adenoviral vectors.
- a pharmaceutical composition disclosed herein can contain at least one, at least 2, at least 3, at least, 4, at least 5, or 6 epitopes, e.g., T cell epitopes, e.g., cancerspecific CD8+ and/or CD4+ epitopes, e.g., cancer specific CD8+ T cell epitopes.
- epitopes e.g., T cell epitopes, e.g., cancerspecific CD8+ and/or CD4+ epitopes, e.g., cancer specific CD8+ T cell epitopes.
- a pharmaceutical composition disclosed herein can contain 1, 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, or 30 epitopes, e.g, T cell epitopes, e.g., cancer-specific CD8+ and/or CD4+ epitopes, e.g., cancer specific CD8+ T cell epitopes.
- the polynucleotides, vectors, cells, or compositions can be formulated as separate compositions that are given at the same time or different times, or the polynucleotides, vectors, cells, or compositions can be given as a single composition.
- the pharmaceutical composition comprises additional agents, e.g, for specific delivery, increasing half-life, or other therapeutic compounds.
- the pharmaceutical composition may comprise one or more of dimethylsulfoxide (DMSO), dextrose, water, succinate, poly I: poly C, poly-L-lysine, carb oxy methylcellulose, and/or chloride.
- a pharmaceutically acceptable carrier comprises any vehicle, such as a liquid or solid filler, diluent, excipient, manufacturing aid (e.g., lubricant, talc magnesium, calcium or zinc stearate, or steric acid), or solvent encapsulating material, involved in carrying or transporting the compound from one site (e.g., the delivery site) of the body, to another site (e.g, organ, tissue or portion of the body).
- a pharmaceutically acceptable carrier is “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the tissue of the subject (e.g., physiologically compatible, sterile, physiologic pH, etc.)
- Some examples of materials which can serve as pharmaceutically -acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, methylcellulose, ethyl cellulose, microcrystalline cellulose and cellulose acetate; (4) powdered tragacanthin; (5) malt; (6) gelatin; (7) lubricating agents, such as magnesium stearate, sodium lauryl sulfate and talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, com oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol (PEG); (12) esters,
- wetting agents, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservative and antioxidants can also be present in the formulation.
- excipient carrier
- pharmaceutically acceptable carrier or the like are used interchangeably herein.
- the carrier may comprise Tris, histidine, sucrose, NaCl, MgCl 2 , a polysorbate (e.g., polysorbate 80), EDTA, and/or ethanol.
- the carrier may comprise a mixture of Tris, histidine, sucrose, NaCl, MgCl 2 , a polysorbate (e.g., polysorbate 80), EDTA, and/or ethanol.
- a carrier for a pharmaceutical compositiondescribedherein may comprise a formulation buffer known in the art (see U.S. Patent No. 7,456,009).
- a carrier may be diluted in saline or PBS prior to injection.
- compositions can comprise one or more pH buffering compounds to maintain the pH of the formulation at a predetermined level that reflects physiological pH, such as in the range of about 5.0 to about 8.0.
- the pH buffering compound usedin the aqueous liquid formulation can be an amino acid or mixture of amino acids, such as histidine or a mixture of amino acids such as histidine and glycine.
- the pH buffering compound is preferably an agent which maintains the pH of the formulation at a predetermined level, such as in the range of about 5.0 to about 8.0, and which does not chelate calcium ions.
- Illustrative examples of such pH buffering compounds include, but are not limited to, imidazole and acetate ions.
- compositions can also contain one or more osmotic modulating agents, i.e., a compound that modulates the osmotic properties (e.g., tonicity, osmolality, and/or osmotic pressure) of the formulation to a level that is acceptable to the blood stream and blood cells of recipient individuals.
- osmotic modulating agent can be an agent that does not chelate calcium ions.
- the osmotic modulating agent can be any compound known or available to those skilled in the art that modulates the osmotic properties of the formulation.
- osmotic modulating agents include, but are not limited to, salts, such as sodium chloride and sodium acetate; sugars, such as sucrose, dextrose, and mannitol; amino acids, such as glycine; and mixtures of one or more of these agents and/or types of agents.
- the osmotic modulating agent(s) may be present in any concentration sufficient to modulate the osmotic properties of the formulation.
- the use of a cocktail of vectors encoding different epitopes may result in a stronger immune response, further there may be a synergistic effect which enhances the immune response.
- the immunogenic composition comprises a cocktail of vectors encoding different epitopes, e.g., cancer-specific CD8+ epitopes, e.g., cancer-specific CD8+ epitopes.
- compositions comprising one or more recombinant polypeptides, polynucleotides, vectors, cells, or compositions disclosed herein may be used in vitro, ex-vivo, or in vivo.
- the pharmaceutical composition is formulated for delivery to a subject, e.g., for inducing an immune response.
- the pharmaceutical composition described herein is administered to a subjectby injection, by means of a catheter, by means of a suppository, or by means of an implant, the implant being of a porous, non-porous, or gelatinous material, including a membrane, such as a sialastic membrane, or a fiber.
- the pharmaceutical composition described herein is delivered in a controlled release system.
- a pump can be used.
- polymeric materials can be used.
- the pharmaceutical composition is formulated in accordance with routine procedures as a composition adapted for intravenous or subcutaneous administration to a subject, e.g., a human.
- pharmaceutical composition for administration by injection are solutions in sterile isotonic use as solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
- the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachets indicating the quantity of active agent.
- the pharmaceutical is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
- an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
- a pharmaceutical composition for systemic administration can be a liquid, e.g., sterile saline, viral sterile saline, lactated Ringer's or Hank's solution.
- the pharmaceutical composition can be in solid forms and re-dissolved or suspended immediately prior to use. Lyophilized forms are also contemplated.
- the pharmaceutical composition can be contained within a lipid particle or vesicle, such as a liposome or microcrystal, which is also suitable for parenteral administration.
- the particles can be of any suitable structure, such as unilamellar or plurilamellar, so long as compositions are contained therein.
- SPLP stabilized plasmid-lipid particles
- DOPE fusogenic lipid dioleoylphosphatidylethanolamine
- PEG polyethyleneglycol
- Positively charged lipids such as N-[l-(2,3- dioleoyloxi)propyl]-N,N,N-trimethyl-amoniummethylsulfate, or “DOTAP,” are particularly preferred for such particles and vesicles.
- the pharmaceutical composition described herein can be administered or packaged as a unit dose, for example, in reference to a pharmaceutical composition to physically discrete units suitable as unitary dosage for the subject, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect in association with the required diluent, i.e., carrier, or vehicle.
- the pharmaceutical composition can be provided as a pharmaceutical kit comprising (a) a container containing a compound of the invention in lyophilized form and (b) a second container containing a pharmaceutically acceptable diluent (e.g., sterile used for reconstitution or dilution of the lyophilized compound of the invention.
- a pharmaceutically acceptable diluent e.g., sterile used for reconstitution or dilution of the lyophilized compound of the invention.
- Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by
- Formulation of the pharmaceutical compositions described herein can be prepared by any method known or hereafter developed in the art of pharmacology.
- preparatory methods include the step of bringing the active ingredient(s) into association with an excipient and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit.
- compositions can additionally comprise a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants, and the like, as suited to the particular dosage form desired.
- a pharmaceutically acceptable excipient includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants, and the like, as suited to the particular dosage form desired.
- a formulation of the pharmaceutical compositions described herein may comprise aqueous and non-aqueous, isotonic sterile injection solutions. These solutions may contain antioxidants, buffers, bacterio stats, and solutes that render the formulation isotonic with the blood of the subject, patient, or other recipient.
- the aqueous and nonaqueous sterile suspensions may comprise suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
- the pharmaceutical compositions described herein may be presented in single dose or multi-dose containers (e.g., vials and/or ampules). The pharmaceutical compositions may be lyophilized or the pharmaceutical compositions may not be lyophilized. Additional solutions may be prepared from sterile powders, granules, and/or tablets.
- a formulation for intravenous administration of the recombinant adenoviral vectors, recombinant polypeptides, polynucleotides, cells, or compositions of the disclosure may be in the form of a sterile injectable aqueous or non-aqueous (e.g., oleaginous) solution or suspension.
- the sterile injectable preparation may also be in a sterile injectable solution or suspension in a nontoxic parenterally-accep table diluent or solvent, for example, a solution in 1 ,3 -butanediol.
- acceptable vehicles and solvents that may be employed are water, phosphate buffer solution, Ringer's solution and isotonic sodium chloride solution.
- sterile, fixed oils may be employed as a solvent or suspending medium.
- any bland fixed oil may be employed, including synthetic mono- or diglycerides.
- fatty acids such as oleic acid may be used in the preparation of the intravenous formulation of the disclosure.
- the composition or pharmaceutical composition comprises at least two vectors as described herein.
- the vectors are identical, e.g., both adenoviral vectors.
- the vectors are different, e.g., an adenoviral vector and an AAV.
- the vectors may be provided as separate medicaments for administration at the same time or at different times to a subject in need thereof.
- the vectors are the same chimeric vector. In some embodiments, the vectors are different chimeric vectors.
- the vectors may be provided as separate medicaments for administration at different times. When administered separately and at different times, the composition comprising either vector may be administered first. In some embodiments, the compositions can be administered on the same day or on different days. In some embodiments, the compositions can be administered using the same schedule or at different schedules during the treatment cycle. [0326] Alternatively, wherein the composition comprises at least two vectors as described herein, the administration of the vectors may be performed simultaneously. Wherein simultaneous administration is used, the vectors may be formulated as separate pharmaceutical compositions. In some embodiments, each of the at least two vectors may be formulated as a single pharmaceutical composition.
- At least three vectors maybe formulated as a single pharmaceutical composition.
- atleastfourvectors may be formulated as a single pharmaceutical composition.
- the at least five vectors may be formulated as a single pharmaceutical composition.
- at least six vectors may be formulated as a single pharmaceutical composition.
- at least seven vectors may be formulated as a single pharmaceutical composition.
- at least eight vectors may be formulated as a single pharmaceutical composition.
- the composition comprises all vectors that are different.
- the composition comprises atleast two identical vectors.
- the composition comprises atleast three identical vectors.
- the composition comprises at least four identical vectors.
- the composition comprises at least five identical vectors.
- the composition comprises at least six identical vectors.
- the recombinant polypeptides, polynucleotides, vectors, cells, compositions, pharmaceutical compositions, or formulations disclosed herein induce an inflating memory T cell response, e.g., an inflating memory CD8+ T cell response in the subject.
- the inflating memory T cell response e.g., an inflating memory CD8+ T cell response, targets cancer cells.
- recombinant polypeptides, polynucleotides, vectors, cells, compositions, pharmaceutical compositions, or formulations disclosed herein may comprise of one or a variety of epitopes (e.g., one or a variety of epitopes described herein) targeted to an HLA type encoding an MHC of a subject’s cancer cells.
- this immunotherapy prevents T cell exhaustion while specifically targeting engineered T cells to the patient’s specific cancer HLA type to ensure an effective immune response.
- the recombinant adenoviral vectors described herein may be used for treating a cancer.
- the polynucleotides, recombinant polypeptides, cells e.g., APCs, T cells
- composition, pharmaceutical compositions, or formulations described herein can be used for treating a cancer.
- the cancer can be selected from colorectal cancer, prostate cancer, esophageal cancer, liver cancer, renal cancer, lung cancer, bladder cancer, breast cancer, pancreatic cancer, brain cancer, hepatocellular cancer, lymphoma, leukemia, gastric cancer, cervical cancer, ovarian cancer, thyroid cancer, melanoma, carcinoma, head and neck cancer, skin cancer, nasopharyngeal cancer, Epstein Barr driven cancers, Human Papilloma vims driven cancers or soft tissue sarcoma.
- the composition can be used for treating esophageal cancer.
- the cancer is a primary cancer.
- the cancer is a relapsed and/or a refractory cancer.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- composition, pharmaceutical compositions, or formulations described herein can be used for preventing formation of a secondary tumor.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- composition, pharmaceutical compositions, or formulations described herein can be used for preventing metastasis.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- compositions, pharmaceutical compositions, or formulations described herein may be used for treating a benign tumor.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- compositions, pharmaceutical compositions, or formulations described herein may be used for treating a pre-malignant tumor.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- compositions, pharmaceutical compositions, or formulations described herein may be used for treating smoldering neoplasms.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- compositions, pharmaceutical compositions, or formulations described herein may be used for treating dysplasia.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- compositions, pharmaceutical compositions, or formulations described herein may be used for treating metaplasia.
- the recombinant adenoviral vectors described herein may be used for treating an infectious disease.
- the polynucleotides, recombinant polypeptides, cells e.g., APCs, T cells
- composition, pharmaceutical compositions, or formulations described herein can be used for treating and/or preventing an infectious disease.
- infectious disease can be selected from chickenpox, E.
- coli diphtheria, giardiasis, infectious mononucleosis, influenza, meningitis, malaria, shingles (e.g., herpes zoster), West Nile virus, pertussis, tuberculosis, tetanus, chlamydia, gonorrhea, salmonella, Lyme disease, measles, mumps, poliomyelitis, pneumonia, HIV/AIDS, and rubella.
- shingles e.g., herpes zoster
- West Nile virus e.g., pertussis, tuberculosis, tetanus, chlamydia, gonorrhea, salmonella, Lyme disease, measles, mumps, poliomyelitis, pneumonia, HIV/AIDS, and rubella.
- the recombinant adenoviral vectors described herein may be used for treating an autoimmune disease.
- the polynucleotides, recombinant polypeptides, cells e.g., APCs, T cells
- composition, pharmaceutical compositions, or formulations described herein can be used for treating and/or preventing an autoimmune disease.
- the autoimmune disease can be selected from amyotrophic lateral sclerosis (ALS), coeliac disease (CD), ankylosing spondylitis (AS), primary Sjogren’s syndrome (PSS), systemic lupus erythematosus (SLE), type 1 diabetes, autoimmune hepatitis (e.g., type 1 or type 2), multiple sclerosis, Guillain-Barre syndrome, chronic inflammatory demyelinating polyneuropathy (CIDP), transverse myelitis, Devic’ s disease (neuromyelitis optica), paraneoplastic cerebellar degeneration (PCD), CREST syndrome (calcinosis, Raynaud phenomenon, esophageal dysmotility, sclerodactyly, and telangiectasia), dermatitis herpetiformis, dermatomyositis, rheumatoid arthritis (RA), psoriatic arthritis, relapsing polychondritis, chronic rheuma
- Parenteral administration includes, for example, intravenous, intramuscular, intraarterial, intraperitoneal, intranasal, rectal, intravesical, intradermal, topical, or subcutaneous administration.
- the vector or composition is administered intravenously or intramuscularly.
- Compositions can take the form of one or more dosage units.
- multiple recombinant adenoviral vectors may be administered. In some embodiments, 1, 2, 3, 4, 5, or more recombinant adenoviral vectors may be administered. The recombinant adenoviral vectors administered may comprise the same adenovirus serotypes. The recombinant adenoviral vectors administered may comprise different adenovirus serotypes. [0334] In specific embodiments, it may be desirable to administer the recombinant polypeptides, polynucleotides, vectors, cells (e.g., APCs, T cells), compositions, pharmaceutical compositions, or formulations described herein locally to the area in need of treatment such at as the site of a tumor.
- APCs e.g., APCs, T cells
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- compositions pharmaceutical compositions, or formulations described herein by intravenous injection or infusion.
- the amount of the recombinant polypeptides, polynucleotides, vectors, or cells (e.g., APCs, T cells) of the present disclosure that is effective/active in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition and can be determined by standard clinical techniques.
- in vitro or in vivo assays can optionally be employed to help identify optimal dosage ranges.
- the precise dose to be employed in the compositions will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances.
- compositions, pharmaceutical compositions, or formulations described herein comprise an effective amount of the recombinant polypeptides, polynucleotides, vectors, or cells (e.g., APCs, T cells) according to the present disclosure such that a suitable dosage will be obtained.
- the correct dosage of the compounds will vary according to the particular formulation, the mode of administration, and its particular site, host and the disease beingtreated. Other factors like age, body weight, sex, diet, time of administration, rate of excretion, condition of the host, drug combinations, reaction sensitivities and severity of the disease shall be taken into account. Administration can be carried out continuously or periodically.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- compositions, pharmaceutical compositions, or formulations described herein may be administered as a single dose.
- the dose may be provided in a prophylactic setting or a therapeutic setting.
- the single dose may be provided as a single dose unit further comprising one or more additional active ingredients, pharmaceutically acceptable carrier, diluent, excipient or adjuvant.
- compositions, pharmaceutical compositions, or formulations described herein may be administered as multiple doses. Wherein multiple doses are administered, one or more may be administered prophylactically or one or more may be administered therapeutically. Where multiple doses are administered, one or more may be administered prophylactically and one or more may be administered therapeutically.
- a single dose of the recombinant adenoviral vectors described herein is administered.
- multiple doses, (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) of the recombinant adenoviral vectors described herein are administered.
- the first dose is administered 1 day, 2 day, 3 days, 4 days, 5 days, 6 days, 7 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, or 15 weeks after the diagnostic biopsy.
- the second dose is administered after 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, or 24 weeks after the first dose.
- the second dose is administered after 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, 24 weeks, 25 weeks, 26 weeks, 27 weeks, or 28 weeks after the diagnostic biopsy.
- the third dose is administered after 1 month, 2 months, 3 months, 4 months, 5 months, 6 months or longer after the first dose.
- the first and the second dose comprise identical recombinant adenoviral vectors described herein. In some embodiments, the first and the second dose comprise different recomb inant adenoviral vectors described herein. In some embodiments, the firstand the second dose comprise different recombinant adenoviral vectors wherein the recombinant adenoviral vectors comprise different epitopes, e.g., T cell epitopes, e.g., cancer-specific CD8+ and/or CD4+epitopes, e.g., cancer-specific CD8+ epitopes.
- epitopes e.g., T cell epitopes, e.g., cancer-specific CD8+ and/or CD4+epitopes, e.g., cancer-specific CD8+ epitopes.
- a sample e.g., tumor sample
- a sample is analyzed for presence of T cell epitopes, e.g., cancer-specific CD8+ and/or CD4+T cell epitopes, e.g., cancer-specific CD8+T cell epitopes after administration of the first dose.
- the composition of the first dose and the second dose is identical.
- the composition of the second dose is altered based on the analysis result, e.g., to specifically target the T cell epitopes, e.g., cancer-specific CD8+ and/or CD4+ T cell epitopes, e.g., cancer-specific CD8+ T cell epitopes present in the subject sample (e.g., tumor sample).
- the T cell epitopes e.g., cancer-specific CD8+ and/or CD4+ T cell epitopes, e.g., cancer-specific CD8+ T cell epitopes present in the subject sample (e.g., tumor sample).
- the subject is administered a second therapeutic agent before administering the first dose.
- the second therapeutic agent is a chemotherapeutic agent.
- the second therapeutic agent is an immunotherapeutic agent.
- the subject is administered a second therapy before administering the first dose.
- the second therapy can be one or more of radiotherapy, chemotherapy, and/orimmunotherapy.
- the secondtherapy can be oncolytic viruses and cellular therapies derived from allogenic or autologous immune cells.
- a second therapeutic agent can comprise fluorouracil, folinic acid (e.g., leucovorin), oxaliplatin, cisplatin, docetaxel, or a combination thereof.
- a second therapeutic agent can comprise fluorouracil, folinic acid (e.g., leucovorin), oxaliplatin, and docetaxel.
- the second therapeutic agent can comprise fluorouracil and oxaliplatin.
- the second therapeutic agent can comprise fluorouracil and cisplatin.
- the second therapeutic agent is a combination of multiple (e.g, 1, 2, 3, or 4) chemotherapeutic agents.
- the second therapeutic agent is selected from one or more of fluorouracil, leucovorin, oxaliplatin, and docetaxel.
- the second therapeutic agent can optionally comprise leucovorin.
- the second therapeutic agent is administered 1 day, 2 day, 3 days, 4 days, 5 days, 6 days, 7 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks before the first dose.
- the second therapeutic agent is administered 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, 24 weeks, 25 weeks, 26 weeks, 27 weeks, or 28 weeks before the second dose.
- the subject is administered a second therapeutic agent after administering the first dose.
- the second therapeutic agent is a chemotherapeutic agent.
- the second therapeutic agent is an immunotherapeutic agent.
- the subject is administered a second therapy before administering the first dose.
- the second therapy can be one or more of radiotherapy, chemotherapy, and/or immunotherapy.
- a second therapeutic agent can comprise fluorouracil, folinic acid (e.g., leucovorin), oxaliplatin, cisplatin, docetaxel, or a combination thereof.
- a second therapeutic agent can comprise fluorouracil, folinic acid (e.g., leucovorin), oxaliplatin, and docetaxel.
- the second therapeutic agent can comprise fluorouracil and oxaliplatin.
- the second therapeutic agent can comprise fluorouracil and cisplatin.
- the second therapeutic agent is a combination of multiple (e.g., 1, 2, 3, or 4) chemotherapeutic agents.
- the second therapeutic agent is selected from one or more of fluorouracil, leucovorin, oxaliplatin, and docetaxel.
- the second therapeutic agent can optionally comprise leucovorin.
- a second dose of the second therapeutic agent therapeutic agent is administered.
- the second dose of the second therapeutic agent is administered after administering the first dose.
- the second dose of the second therapeutic agent therapeutic agent is administered before administering the second dose.
- the second dose of the second therapeutic agent is administered after administering the second dose.
- the second dose of the second therapeutic agent is administered after 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, or 24 weeks after the first dose.
- the second dose of the second therapeutic agent therapeutic agent is administered 1 week, 2 week, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks before the second dose.
- the second dose of the second therapeutic agent therapeutic agent is administered 1 week, 2 week, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, or 10 weeks after the second dose.
- the first dose of the second therapeutic agent is administered at once.
- the second dose of the second therapeutic agent is administered at once.
- the second dose of the second therapeutic agent administration is spread over 1 days, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, or 8 weeks.
- the recombinant adenoviral vectors described herein can be administered in a prime-boost regime.
- the first dose is administered to prime the immune response to a target antigen (prime dose).
- the second dose is administered after the first dose has been administered to prime the immune response to the target antigen.
- the boost dose elicits an equal or greater measurable immune response compared to the response elicited by administration of the prime dose.
- the prime dose elicits an immune response to the target antigen
- the boost dose elicits an equal or greater measurable immune response compared to the response elicited by administration of the prime dose.
- an “effective amount” of a “prime dose” refers to the amount of target antigen (e.g., T cell epitope, e.g., a cancer-specific CD8+ epitope described herein) which elicits a measurable immune response in a subject as compared to the immune response in the mammalian subject in the absence of administration of the antigen.
- An “effective amount” of a “boost dose” refers to the amount of antigen (e.g., T cell epitope, e.g., a cancerspecific CD8+ epitope described herein) which elicits an immune response to the target antigen upon administration to a subject which previously has been administered a prime dose of the target antigen.
- the prime dose is administered intramuscularly. In some embodiments, the prime dose is administeredintravenously . In some embodiments, the boost dose is administered intramuscularly. In some embodiments, the boost dose is administered intravenously. In some embodiments, the prime dose is administered intramuscularly, and the boost dose is administered intramuscularly. In some embodiments, the prime dose is administered intravenously, and the boost dose is administered intravenously. In some embodiments, the prime dose is administered intravenously, and the boost dose is administered intramuscularly. In some embodiments, prime dose is administered intramuscularly, and the boost dose is administered intravenously.
- the prime dose comprises at least 1, at least 2, at least 3, at least 4, at least 5, or no more than 6 vectors. In some embodiments, the prime dose comprises 2 xlO 10 viral particles per dose. In some embodiments, the prime dose comprises a first vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, a second vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, a third vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, a fourth vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, a fifth vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, and/or a sixth vector at a concentration of 1 x
- the prime dose comprises a first vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, a second vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, a third vector at a concentration of 1 xl 0 11 , 2 xl 0 11 , or 3 xl 0 11 viral particle, a fourth vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, a fifth vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, and/or a sixth vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, wherein the total concentration of viral particle in the dose is no more than 3 xlO 11 viral particle per dose.
- the boost dose comprises at least 1, at least 2, at least 3, at least 4, at least 5, or no more than 6 vectors. In some embodiments, the boost dose comprises 2 x 10 10 viral particles per dose. In some embodiments, the boost dose comprises a firstvector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, a second vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, a third vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, a fourth vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, a fifth vector at a concentration of 1 xlO 10 , 5 xlO 10 , or 1 xlO 11 viral particles, and/or a sixth vector at a concentration of 1 x
- the boost dose comprises a first vector at a concentration of 1 xl0 11 , 2 xl0 11 , or3 xlO 11 viral particle, a second vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, a third vector at a concentration of l xl0 11 , 2 xl0 11 , or 3 xl0 n viral particle, a fourth vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, a fifth vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, and/or a sixth vector at a concentration of 1 xlO 11 , 2 xlO 11 , or 3 xlO 11 viral particle, wherein the total concentration of viral particle in the dose is no more than 3 xlO 11 viral particle per dose.
- the prime dose is administered 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, or 20 weeks after the diagnostic biopsy.
- the boost dose is administered after 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, or 24 weeks after the prime dose.
- the boost dose is administered 4 weeks, 5 weeks, 6 weeks, 7 weeks, 8 weeks, 9 weeks, 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, 21 weeks, 22 weeks, 23 weeks, 24 weeks, 25 weeks, 26 weeks, 27 weeks, or 28 weeks after the diagnostic biopsy.
- a second boost dose is administered after 1 month, 2 month, 3 month, 4 month, 5 month, 6 months or longer after the prime dose.
- a second boost dose is administered after 1 month, 2 month, 3 month, 4 month, 5 month, 6 months or longer after the first boost dose.
- a sample e.g., tumor sample
- a sample from the subject is analyzed for presence of T cell epitopes, e.g., cancer-specific CD8+ T cell epitopes after administration of the prime dose.
- the composition of the prime dose and the boost dose is identical.
- the composition of the boost dose is altered based on the analysis result, e.g., to specifically target the T cell epitopes, e.g., cancer-specific CD8+ T cell epitopes present in the subject sample (e.g., tumor sample).
- Administration of the boost dose can be initiated when a population of antigen-specific immune cells has expanded (increased in number) to at least20% the maximal number of antigenspecific immune cells that is eventually attained; to at least 30%; to at least 40%; to at least 50%; to at least 60%; to at least 70%; to at least 80%; to at least 90%; to at least 95%; to at least 99% the maximal number of antigen-specific immune cells that is eventually attained.
- the boost dose can be initiated when the population of antigen-specific cells has contracted to under 90% the maximal number of antigen-specific cells; under 80%; under 70%; under 60%; under 50%; under 40%; under 30%; under 20%; under 10%; under 5%; under 1 .0%; under 0.5%; under 0.1%; under 0.05%; or under 0.01% the maximal number of antigen-specific immune cells.
- the antigen-specific cells can be identified as specific for an empty vector, or specific for a cancer-specific CD8+ T cell epitope expressed by a polynucleotide contained in the vector.
- the boost dose can enhance the prime dose immune response by at least two-fold, at times between about three- and five-fold or five-fold to ten-fold, or from tenfold to 100-fold or greater.
- the prime dose and boost dose have a synergistic effect on the immune response.
- the enhanced immune response will include a T-cell response, e.g., a CD8+ T-cell response.
- the prime dose and boost dose will break a subject’s tolerogenic state towards a target cancer-specific CD8+ T cell epitope disclosed herein.
- the vector e.g., the viral vector in the therapeutic composition can be administered in a dose, or dosages, where each dose comprises between IxlO 3 and 7.5 x 10 13 viral particles per 70 kg body weight; and so on.
- the vector e.g., the viral vector in the therapeutic composition can be administered in a dose, or dosages, where each dose comprises between IxlO 2 and 5x10 12 viral particles per 70 kg body weight; between IxlO 3 and 5x10 11 viral particles per 70 kg body weight; between IxlO 4 and 5xl0 n viral particles per 70 kgbody weight; between IxlO 5 and 5xl0 n viral particles per 70 kgbody weight; between IxlO 6 and 5x10 11 viral particles per 70 kgbody weight; between IxlO 7 and 5xl0 n viral particles per 70 kgbody weight; between IxlO 8 and 5xl0 n viral particles per 70 kg body weight; between 1x10 9 and 5x10 11 viral particles per 70 kg body weight; between 1x10 10 and 5x10 11 viral particles per 70 kg body weight; between 1x10 11 and 5x10 11 viral particles per 70 kg body weight; 2xl0 2 and 5x10 12 viral particles per 70 kg body weight; between
- the recombinant adenoviral vectors described herein can be used in combination with existingtherapies.
- the recombinant adenoviral vectors described herein is used in combination with an additional therapy or therapeutic agent (e.g., existing), for example an anti-cancer therapy, e.g., a neoadjuvant therapy.
- an additional therapy or therapeutic agent e.g., existing
- an anti-cancer therapy e.g., a neoadjuvant therapy.
- the disclosure also relates to a combination therapy comprising administration of the recombinant adenoviral vectors described herein and an anti-cancer therapy.
- the anti-cancer therapy may include one or more of therapeutic agent (e.g., a combination of different chemotherapeutic agents, antibodies), radiotherapies (y-rays, X-rays, and/or the directed delivery of radioisotopes, microwaves, UV radiation and the like), hormone therapy (steroidal, peptide, and the like), gene therapies (e.g., antisense, retroviral therapy and the like), viral therapy, RNA therapy, bone marrow transplantation, nanotherapy, and/or oncolytic drugs or viruses.
- therapeutic agent e.g., a combination of different chemotherapeutic agents, antibodies
- radiotherapies y-rays, X-rays, and/or the directed delivery of radioisotopes, microwaves, UV radiation and the like
- hormone therapy steroidal, peptide, and the like
- gene therapies e.g., antisense, retroviral therapy and the like
- viral therapy e.g., RNA therapy, bone marrow transplantation, nanotherapy
- Exemplary therapeutic agents include but are not limited to antibodies, antibody-drug conjugates, immunogenic vaccines, checkpoint inhibitors (e.g., immune checkpoint inhibitors, e.g., PD-1 antibodies), antineoplastic agents, immunogenic agents, attenuated cancerous cells, cellular therapies derived from allogenic or autologous immune cells, hormones, tumor antigens, antigen presenting cells such as dendritic cells pulsed with tumor-derived antigen or nucleic acids, immune stimulating cytokines (e.g., IL- 2, IFNa2, GM-CSF), targeted small molecules and biological molecules (such as components of signal transduction pathways, e.g., modulators of tyrosine kinases and inhibitors of receptor tyrosine kinases, and agents that bind to tumor- specific antigens, including EGFR antagonists), an anti-inflammatory agent, a cytotoxic agent, a radiotoxic agent, an immunosuppressive agent and cells transfected with a gene encoding an immune stimulating
- the recombinant adenoviral vectors described herein may be administered in combination with an anti-cancer therapy as described herein. In some embodiments, the recombinant adenoviral vectors described herein may be administered in combination with cisplatin or histone deacetylase inhibitors. In some embodiments, the recombinant adenoviral vectors described herein is used in combination with surgery. In some embodiments, the recombinant adenoviral vectors describedherein is administered before, during or after surgery.
- the recombinant adenoviral vectors described herein are administered in combination with anti-diarrheal agents, anti-emetic agents, analgesics, opioids and/or non-steroidal anti-inflammatory agents.
- the recombinant adenoviral vectors described herein may be administered at the same time or at a different time as the other therapy, e.g., simultaneously, separately or sequentially.
- the recombinant adenoviral vectors described herein may be administered with a vaccine.
- the recombinant adenoviral vectors and the additional therapy or therapeutic agent may be provided as separate medicaments for administration at the same time or at different times.
- recombinant adenoviral vectors and the additional therapy or therapeutic agent are provided as separate medicaments for administration at different times.
- either the recombinant adenoviral vectors as described herein, or the additional therapy or therapeutic agent may be administered first.
- both can be administered on the same day or on different days, and they can be administered using the same schedule or at different schedules during the treatment cycle.
- the recombinant polypeptides, polynucleotides, vectors, cells e.g., APCs, T cells
- compositions, pharmaceutical compositions, or formulations described herein may be administered prior to an additional therapy (e.g., chemotherapy, radiotherapy, and/or immunotherapy).
- additional therapy e.g., chemotherapy, radiotherapy, and/or immunotherapy.
- the recombinant polypeptides, polynucleotides, vectors, cells (e.g., APCs, T cells), compositions, pharmaceutical compositions, or formulations described herein may be administered after an additional therapy (e.g., chemotherapy, radiotherapy, and/or immunotherapy).
- compositions, pharmaceutical compositions, or formulations described herein may be administered prior to and subsequent to an additional therapy (e.g., chemotherapy, radiotherapy, and/or immunotherapy).
- additional therapy e.g., chemotherapy, radiotherapy, and/or immunotherapy.
- the administration of the additional therapy or therapeutic agent may be performed simultaneously with the administration of the recombinant adenoviral vectors as described herein. Wherein simultaneous administration is used, the recombinant adenoviral vectors described herein, and the additional therapy or therapeutic agent may be formulated as separate pharmaceutical compositions. In some embodiments, the recombinant adenoviral vectors described herein, and the additional therapy or therapeutic agent may be formulated as a single pharmaceutical composition.
- the recombinant adenoviral vectors described herein can be used in combination with an immunomodulatory agent.
- the immunomodulatory agent may be administered simultaneously, sequentially or separately with the immunomodulatory agent.
- the immunomodulatory agent may be an immune checkpoint inhibitor.
- immune checkpoint inhibitors include but are not limited to inhibitors of an immune checkpoint protein selected from the group consisting of CTLA-4, PD-1, PD-L1, PD-L2, TIM-3, LAG-3, B7-H3, B7-H4, B7-H6, A2aR, BTLA, GAL9 and IDO.
- a vector described herein can result in enhanced tumor control when administered in combination with a checkpoint inhibitor such as an anti-PD- L1 therapy. This has been shown effective in tumor models which are known to be unresponsive to standard checkpoint inhibitor therapy.
- the recombinant adenoviral vectors described herein may be used in combination with a check point inhibitor for the treatment of checkpoint inhibitor unresponsive tumors.
- the present disclosure provides methods for preventing relapse of cancer in a subject, the methods comprising administering to a subject in need thereof recombinant adenoviral vectors described herein.
- the present disclosure also provides methods for preventing relapse of cancer in a subject, the methods comprising administering to a subject in need thereof an immunogenic composition comprising one or more of (i) a recombinant adenoviral vector, (ii) a cell, (iii) a composition, (iv) a pharmaceutical composition, or (v) a formulation as described herein.
- the present disclosure provides methods for preventing metastasis of cancer in a subject, the methods comprising administering to a subj ect in need thereof recombinant adenoviral vectors described herein.
- the present disclosure also provides methods for preventingmetastasis of cancer in a subject, the methods comprising administering to a subject in need thereof an immunogenic composition comprising one or more of (i) a recombinant adenoviral vector, (ii) a cell, (iii) a composition, (iv) a pharmaceutical composition, or (v) a formulation as described herein.
- the recombinant adenoviral vectors and the immunomodulatory agent may be provided as separate medicaments for administration at the same time or at different times.
- the vector or composition of the present disclosure and the immunomodulatory agent are provided as separate medicaments for administration at different times.
- either the vector or the immunomodulatory agent may be administered first.
- both can be administered on the same day or on different days, and they can be administered using the same schedule or at different schedules during the treatment cycle.
- the administration of the immunomodulatory agent may be performed simultaneously with the administration of the vector or immunogenic composition as described herein. Wherein simultaneous administration is used, the vector or immunogenic composition and the immunomodulatory agent may be formulated as separate pharmaceutical compositions. In some embodiments, the vector or immunogenic composition and the immunomodulatory agent may be formulated as a single pharmaceutical composition.
- the recombinant adenoviral vectors described herein can be administered prophylactically or therapeutically.
- prophylactically refers to administration intended to have a protective effect against disease.
- therapeutically refers to administration intended to have a curative effect.
- composition, pharmaceutical composition, or formulation comprises at least two vectors and wherein the vectors comprise recombinant polypeptide comprising different epitopes as disclosed herein or polynucleotides encoding different epitopes as described above, there may be synergy between the vectors.
- the immunogenic composition comprises at least three vectors and wherein the vectors comprise recombinant polypeptide comprising different epitopes as disclosed herein or polynucleotides encoding different epitopes as described above, there may be synergy between the vectors.
- the immunogenic composition comprises at least four vectors and wherein the vectors comprise recombinant polypeptide comprising different epitopes as disclosed herein or polynucleotides encoding different epitopes as described above, there may be synergy between the vectors.
- the immunogenic composition comprises at least five vectors and wherein the vectors comprise recombinant polypeptide comprising different epitopes as disclosed herein or polynucleotides encoding different epitopes as described above, there may be synergy between the vectors.
- the immunogenic composition comprises at least six vectors and wherein the vectors comprise recombinant polypeptide comprising different epitopes as disclosed herein or polynucleotides encoding different epitopes as described above, there may be synergy between the vectors.
- each of the vectors may be administered at a sub -optimal dose.
- sub -optimal dose refers to a dose level that it is not intended to fully remove or eradicate the tumor, but nevertheless results in some tumor cells or tissue becoming necrotic.
- the skilled person will be able to determine an appropriate dose required in order to achieve this, depending on factors such as, age of the patient, status of the disease and size and location of tumor or metastases.
- kits for producingthe composition described herein comprises a recombinant adenoviral vector as described herein.
- the kit may comprise at least one recombinant adenoviral vector, at least two recombinant adenoviral vectors, at least three recombinant adenoviral vectors, at least four recombinant adenoviral vectors, at least five recombinant adenoviral vectors, or more.
- the kit comprises a chimeric recombinant adenoviral vector as described herein.
- the kit comprises 1, 2, 3, 4, 5 or more chimeric recombinant adenoviral vectors as described herein.
- the chimeric recombinant adenoviral vectors may comprise different adenoviral serotypes.
- the chimeric recombinant adenoviral vectors may comprise the same adenoviral serotypes.
- the kit comprises a recombinant polypeptide, a polynucleotide, a vector, a cell (e.g., APCs, T cells), a population of cells, a composition, a pharmaceutical composition, or a formulation described herein.
- the kit comprises a recombinant polypeptide comprising one or more cancer-specific epitopes comprising GVYDGREHTV andKLVELEHTL and SPSSASLAL and KPRPDVTNEL and KEFAFLEHSL and HELGFKVVL or any combination thereof.
- the kit comprises a recombinant polypeptide comprising one or more cancerspecific epitopes comprising GVYDGREHTV and KLVELEHTL and SPSSASLAL and KPRPDVTNEL and KEFAFLEHSL, HELGFKVVL, LKPDHIQR, SLFGARPGR, KTEVHGRLK, NLRPPTQEL, RVSLPKLGYK, LRPPTQEL, and RPASPRPAP or any combination thereof.
- cancerspecific epitopes comprising GVYDGREHTV and KLVELEHTL and SPSSASLAL and KPRPDVTNEL and KEFAFLEHSL, HELGFKVVL, LKPDHIQR, SLFGARPGR, KTEVHGRLK, NLRPPTQEL, RVSLPKLGYK, LRPPTQEL, and RPASPRPAP or any combination thereof.
- the kit comprises a recombinant polypeptide comprising one or more cancer-specific epitopes comprising GVYDGREHTV, KLVELEHTL, SPSSASLAL, KPRPDVTNEL, KEFAFLEHSL, HELGFKVVL, LKPDHIQR, SLFGARPGR, KTEVHGRLK, NLRPPTQEL, RVSLPKLGYK, LRPPTQEL, RPASPRPAP, IATKIALQM, SLYQTIRLK, KTPLHTLLK, RLHSFTLRQK, RVFTSSLKTK, RVLAKGLAK, GPRPSPTRSV, GQHLHLETF, SPRSPSPSL, VPQEAVRAPL, IYPFINSH, SPSSASLTL, LNKVKTSL, NLKTHLRL, FRGVFVHRY, TSGPVTEKY, and VVAAHLAGA or any combination thereof.
- the kit comprises a vector, composition, or formulation as described
- a kit described herein includes one or more additional active ingredients, pharmaceutically acceptable carrier, diluent, excipient or adjuvant, and optionally instructions for use.
- the additional active agent may include checkpoint inhibitors, antineoplastic agents, immunogenic agents, attenuated cancerous cells, tumor antigens, antigen presenting cells such as dendritic cells pulsed with tumor-derived antigen or nucleic acids, immune stimulating cytokines (e.g., IL-2, IFNa2, GM-CSF), targeted small molecules and biological molecules (such as components of signal transduction pathways, e.g.
- modulators of tyrosine kinases and inhibitors of receptor tyrosine kinases, and agents that bind to tumor- specific antigens including EGFR antagonists
- an anti-inflammatory agent including a cytotoxic agent, a radiotoxic agent, or an immunosuppressive agent and cells transfected with a gene encoding an immune stimulating cytokine (e.g., GM-CSF).
- a cytotoxic agent e.g., GM-CSF
- an immunosuppressive agent e.g., GM-CSF
- the pharmaceutical acceptable carrier, diluent, excipient or adjuvant may include; sterile diluents such as water, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or digy Icerides, polyethylene glycols, glycerin, or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; and agents for the adjustment of tonicity such as sodium chloride or dextrose.
- sterile diluents such as water, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or digy Icerides, polyethylene glycols, glycerin, or other solvents
- antibacterial agents such as benzyl alcohol or methyl paraben
- agents for the adjustment of tonicity such as sodium chloride or dextrose.
- kits are compartmentalized for ease of use and can include one or more containers with reagents. In certain embodiments, all of the kit components are packaged together. Alternatively, one or more individual components of the kit can be provided in a separate package from the other kits components. The kits can also include instructions for using the kit components.
- EXAMPLE 1 Infection of 293 cells by chimeric vectors
- EXAMPLE 2 Tropism of the recombinant vectors to human cell lines
- the GFP protein coding sequence was cloned into the chimeric vector systems.
- the vectors were then added to a range of different human cell lines. These included A549 (an adenocarcinomic human alveolar basal epithelial cell line), A375 (a Human malignant melanoma cell line), Ca Ski (a human epithelial cell line), DMS273 (a human lung small cell carcinoma cell line), NCI-H322 (a human non-small cell carcinoma cell line), and SHP-77 (a small cell lung carcinoma cell line).
- infected cells expressing GFP were detected by fluorescence microscopy (FIGs. 14A-14AP) in samples infected with vector versions 2, 3, 4, or 5. The results indicated the ability of the chimeric vectors to infect and enter human cells. Tropism for vector versions 2-8 is also shown in FIGs. 15A-15BT.
- EXAMPLE 3 Analysis of immunogenicity and T cell exhaustion following vaccination with chimeric vectors
- FIG. 16A A longitudinal flow cytometry analysis of circulating AHI -dextramer positive cytotoxic T-cells (CD8).
- FIG. 16A A schematic representation of the experimental timeline is shown in FIG. 16A.
- the experimental One group comprised chimeric vector versions 2, 4, and 8 (FIGs. 16B- 16E and FIGs. 17A-17C) and a second group comprised chimeric vector versions 3 and 6 (FIGs.
- mice were purchased from Charles River at six to seven weeks of age. Mice were housed within a pathogen-free environment with a twelve- hour light cycle at the Biomedical Sciences Building at the University of Oxford. Prior to commencing any in vivo studies, mice were given seven days acclimatization after delivery.
- mice were humanly euthanized by an approved Schedule One method (in line with UK Animals [Scientific Procedures] Act 1986 [ASP A]) and tissues immediately retrieved for processing.
- mice were administered the adenoviral vaccine. Mice were placed into predetermined treatment groups and received either a version of active adenoviral vaccine, or a vector control. For each treatment group, each mouse received from 5x10 7 to 5x10 8 infectious units (LU) of adenovirus. Each adenoviral vaccine, for each novel vector, consisted of virus suspended in Phosphate-Buffered Saline (PBS) (Gibco, 11503387). Mice were monitored for any adverse reactions for thirty minutes post-vaccination, then observed six hours later.
- PBS Phosphate-Buffered Saline
- Dextramers were constructed and provided by Immundex, Denmark (JG03294). Whole blood was stained with 10 pl of dextramer for 20 minutes at 37 °C. Blood was then subsequently stained with monoclonal antibody (mAB) and live/dead near-IR (Life Technologies Ltd, L34975) at 4 °C for 20 minutes. Red blood cells were lysed and remaining cells lysed using BD FACS lysis buffer (BD BioSciences, 349202).
- each chimeric vector showed greater percentage of AHl-dextramer+ CD8+ cells compared to that of the vector control (FIG. 16B). Cells were also analyzed for markers of memory inflation phenotype. The chimeric vectors showed similar levels of AHl-dextramer+ CD8+ CD44+ CD62L- as compared to that of the wildtype Ad5 control (FIG. 16C). The chimeric vectors also showed similar levels of AHl-dextramer+CD8+CX3CRl+cellsas compared to that of the wildtype Ad5 control (FIG.
- Flow cytometry analysis was also performed to examine evidence of T cell exhaustion following vaccination with chimeric vector versions 3 and 6. All versions displayed low levels of T cell exhaustion and the resistance to exhaustion was maintained even after tumor challenge. A longitudinal analysis was performed by serial blood draw to measure the relative percentage of AHl-dextramer+ CD8+ PD-1+ cells. All versions displayed low levels of PD-1 (FIG. 19A). Levels of Tim3 and Lag3 were also measured at Day 40 pre-tumor challenge. Mice vaccinated with chimeric vector versions 3 and 6 showedlow levels of AHl-dextramer+ CD8+ Tim3+ cells (FIG. 19B) and AHl-dextramer+ CD8+ Lag3+ cells (FIG.
- Chimeric vector version 3 tended to show lower levels of T cell exhaustion markers than those of chimeric vector version 6. These results indicated that vaccination with chimeric vector versions 3 and 6 led to immunogenicity, strong memory inflation phenotypes, and low T cell exhaustion in tested mice.
- EXAMPLE 4 Analysis of anti-tumor activity using chimeric vectors
- mice from Example 3 After 50 days following vaccination, mice from Example 3 then received a tumor challenge. Prior to injection, Colon Tumor 26 (CT26) cells were resuspended in PBS. Mice received a subcutaneous injection of 2x10 5 CT26 cells into the flank. The resulting volume of the CT26-derieved tumors, determined from 0.5 x (length x width 2 ) as measured using calipers, was monitored every two days. For the mouse survival curves, mice were euthanized by an approved Schedule One method as tumors began to approach a volume of 1200mm 3 .
- CT26 Colon Tumor 26
- mice vaccinated with chimeric vector versions 2, 4, or 8 showed significantly decreased tumor volume comparedto thatof naive-untreated mice (FIG.20). Mice vaccinated with chimeric vectors also showed substantially lower tumor volumes compared to mice treated with vector control. Notably, the low tumor volumes were maintained for 40 days post-tumor implantation. These results demonstrate the chimeric vectors have beneficial anti-tumor activity.
- mice were euthanized as tumors exceeded a geometric mean of 10 mm 2 . As shown in FIG. 21, all micethatreceivedvaccinationwith chimeric vector 2, 4, or 8 showed 100% survival, which was significantly higher than the percent survival of naive mice or those mice that received I8V- minigene vector. At day 62, mice that received the chimeric vector version 2 or 4 showed 100% survival across the cohort. These results demonstrated that the chimeric vectors administered provided advantageous survival in mice with CT26 tumors compared to negative control groups (naive mice and mice that received 18 V minigene construct) as well as the AdHu5 group.
- mice vaccinated with chimeric vector versions 3 or 6 also showed significantly decreased tumor volume comparedto thatof naive-untreated mice (FIG.22). Mice vaccinated with chimeric vector 3 or 6 also showed substantially lower tumor volumes compared to mice treated with vector control. The low tumor volumes in treated mice were maintained for 12 days post-tumor implantation. These results provided further support that the chimeric vectors had beneficial antitumor activity.
- mice were euthanized as tumors exceeded a geometric mean of 10 mm 2 .
- all mice that received vaccination with chimeric vector version 3 or 6 showed 100% survival over 20 days, which was higher than the percent survival of mice that received 18 V-minigene vector.
- mice that received the AdHu5, or chimeric vector version 3 or 6 showed 100% survival across the cohort.
- these results demonstrated that the chimeric vectors administered provided advantageous survival in mice implanted with CT26 tumors compared to negative control groups (naive mice and mice that received I8V minigene construct).
- NGS reads were passed through a standard RefSeq pipeline, with confirmation that reads covered one hundred percent of the plasmid sequence. Average NGS read depth across each construct ranged from 1450 to 3170. Targeted Sanger sequencing was performed to confirm the sequence accuracy of highly or tandem repeated areas, such as in the ITRs.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Virology (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Biotechnology (AREA)
- Epidemiology (AREA)
- General Engineering & Computer Science (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Molecular Biology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Mycology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biomedical Technology (AREA)
- Physics & Mathematics (AREA)
- Biophysics (AREA)
- Plant Pathology (AREA)
- Immunology (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Biochemistry (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
L'invention concerne des compositions et des méthodes pour traiter le cancer chez un sujet humain comprenant l'administration d'un vecteur chimérique recombinant. Le vecteur chimérique recombinant comprend un épitope de lymphocyte T CD8+ spécifique du cancer qui peut induire, en réponse, une inflation des lymphocytes T CD8 + et/ou CD4+ mémoire chez le sujet.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363593110P | 2023-10-25 | 2023-10-25 | |
| US63/593,110 | 2023-10-25 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2025088378A2 true WO2025088378A2 (fr) | 2025-05-01 |
| WO2025088378A3 WO2025088378A3 (fr) | 2025-06-19 |
Family
ID=93924707
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/IB2024/000596 Pending WO2025088378A2 (fr) | 2023-10-25 | 2024-10-25 | Compositions et méthodes pour traiter le cancer à l'aide de vecteurs viraux chimériques |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025088378A2 (fr) |
Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US7456009B2 (en) | 2000-03-07 | 2008-11-25 | Merck & Co., Inc. | Adenovirus formulations |
Family Cites Families (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US7741099B2 (en) * | 2004-10-13 | 2010-06-22 | Beth Israel Deaconess Medical Center Inc. | Adenoviral vectors and uses thereof |
| WO2010014567A2 (fr) * | 2008-08-01 | 2010-02-04 | Merck & Co., Inc. | Polynucléotides variants pp65, ie1 et ie2 du cmvh et utilisations associées |
| EP3773710A4 (fr) * | 2018-04-04 | 2022-03-16 | Altimmune Inc | Combinaisons de compositions vaccinales induisant des lymphocytes t et leurs utilisations |
| MX2022004484A (es) * | 2019-10-16 | 2022-07-19 | Cancer Research Tech Ltd | Vector para el tratamiento de cancer. |
| WO2024209270A2 (fr) * | 2023-04-07 | 2024-10-10 | Infinitopes Limited | Méthodes et compositions pour le traitement du cancer à l'aide de polypeptides recombinants |
-
2024
- 2024-10-25 WO PCT/IB2024/000596 patent/WO2025088378A2/fr active Pending
Patent Citations (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US7456009B2 (en) | 2000-03-07 | 2008-11-25 | Merck & Co., Inc. | Adenovirus formulations |
Non-Patent Citations (10)
| Title |
|---|
| ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 |
| AUSUBEL ET AL., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY |
| BATZER ET AL., NUCLEIC ACID RES., vol. 19, 1991, pages 5081 |
| HENIKOFFHENIKOFF, PROC. NATL. ACAD. SCI. USA, vol. 89, 1989, pages 10915 |
| NEEDLEMANWUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443 |
| OHTSUKA ET AL., J. BIOL. CHEM., vol. 260, 1985, pages 2605 - 2608 |
| PEARSONLIPMAN, PROC. NAT'L. ACAD. SCI. USA, vol. 85, 1988, pages 2444 |
| POUWELS ET AL.: "Cloning Vectors: A Lab oratory Manual", 1985, ELSEVIER |
| ROSSOLINI ET AL., MOL. CELL. PROBES, vol. 8, 1994, pages 91 - 98 |
| SMITHWATERMAN, ADV. APPL. MATH., vol. 2, 1981, pages 482 |
Also Published As
| Publication number | Publication date |
|---|---|
| WO2025088378A3 (fr) | 2025-06-19 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US12116396B2 (en) | LAMP constructs | |
| JP7657282B2 (ja) | 癌抗原を含む改善されたlamp構築物 | |
| KR20220141849A (ko) | 다발성 골수종에서 발현된 신생항원 및 이의 용도 | |
| WO2025088378A2 (fr) | Compositions et méthodes pour traiter le cancer à l'aide de vecteurs viraux chimériques | |
| WO2024209270A2 (fr) | Méthodes et compositions pour le traitement du cancer à l'aide de polypeptides recombinants | |
| US20250352639A1 (en) | Bicistronic LAMP Constructs Comprising Immune Response Enhancing Genes and Methods of Use Thereof | |
| JP2022502066A (ja) | 癌治療のための修飾b5r遺伝子を有する腫瘍溶解性ワクシニアウイルス | |
| US20240165175A1 (en) | Muc16 promoter containing virus | |
| EP4452305A1 (fr) | Virus mva recombinants pour administration intrapéritonéale pour le traitement du cancer | |
| CN115515623A (zh) | 在卵巢癌中表达的新抗原及其用途 | |
| JP2022551732A (ja) | がん抗原を含む改良lamp構築物 | |
| WO2025149656A1 (fr) | Traitement et prévention du cancer pulmonaire | |
| HK40123095A (en) | Improved lamp constructs | |
| HK40085962A (en) | Neoantigens expressed in ovarian cancer and their uses | |
| EA048996B1 (ru) | Вектор для лечения рака | |
| HK40014081B (en) | Improved lamp constructs | |
| HK40014081A (en) | Improved lamp constructs |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24827126 Country of ref document: EP Kind code of ref document: A2 |