US20090311219A1 - Oncolytic Adenoviruses for Cancer Treatment - Google Patents

Oncolytic Adenoviruses for Cancer Treatment Download PDF

Info

Publication number
US20090311219A1
US20090311219A1 US12/184,881 US18488108A US2009311219A1 US 20090311219 A1 US20090311219 A1 US 20090311219A1 US 18488108 A US18488108 A US 18488108A US 2009311219 A1 US2009311219 A1 US 2009311219A1
Authority
US
United States
Prior art keywords
promoter
adenovirus
sequence
oncolytic
tumor
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.)
Abandoned
Application number
US12/184,881
Other languages
English (en)
Inventor
Ramon Alemany Bonastre
Juan Jose Rojas Exposito
Manel Maria Cascallo Piqueras
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
DNAtrix Inc
Original Assignee
DNAtrix Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by DNAtrix Inc filed Critical DNAtrix Inc
Assigned to DNATRIX, INC. reassignment DNATRIX, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: INSTITUT CATALA D'ONCOLOGIA
Publication of US20090311219A1 publication Critical patent/US20090311219A1/en
Priority to US14/327,840 priority Critical patent/US20150071881A1/en
Priority to US15/144,637 priority patent/US10016470B2/en
Priority to US16/028,037 priority patent/US20190183946A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • C12N15/861Adenoviral vectors
    • C12N15/8613Chimaeric vector systems comprising heterologous sequences for production of another viral vector
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10021Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10032Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10041Use of virus, viral particle or viral elements as a vector
    • C12N2710/10043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10321Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10332Use of virus as therapeutic agent, other than vaccine, e.g. as cytolytic agent
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10341Use of virus, viral particle or viral elements as a vector
    • C12N2710/10343Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2820/00Vectors comprising a special origin of replication system
    • C12N2820/007Vectors comprising a special origin of replication system tissue or cell-specific

Definitions

  • the field of the invention is related in general terms to the field of tumor biology.
  • the invention refers to selective-replication adenoviruses in tumors, known as oncolytic adenoviruses, and their use to inhibit cancer.
  • the tumor cell dies as a result of the cytopathic effect caused by the internal replication of the virus more than because of the effect of a therapeutic gene.
  • Preferential replication in a tumor cell is called oncotropism and the lysis of the tumor is called oncolysis.
  • Viruses that are replicated selectively in tumors are called oncolytic viruses.
  • Cancer virotherapy significantly predates gene therapy.
  • the first observations of tumor cure with viruses date from early in the last century. Already in 1912, De Pace observed tumor regressions after inoculating the rabies virus in cervical carcinomata 2 . Since then, many types of virus have been injected in tumors to treat them 3 .
  • viruses that present a natural oncotropism for example the autonomous parvovirus, the vesicular-stomatitis virus 5 and the reovirus 6 .
  • Other viruses can be manipulated genetically for selective replication in tumors.
  • the herpes simplex virus has been made oncotropic on selecting the gene of ribonucleotide reductase, a dispensable enzyme activity in cells in active proliferation such as tumor cells 7 .
  • the adenovirus in view of its low pathogenicity and high capacity to infect tumor cells, has been the virus used most in both virotherapy and gene therapy for cancer.
  • Ad5 human adenovirus
  • Ad5 infects epithelial cells, which during a natural infection are the cells of the bronchial epithelium. It enters the cells by means of interaction of the fiber, a viral protein that extends like an antenna from the twelve vertices of the capsid, with a cell protein involved in intercellular adhesion called Coxsackie-Adenovirus Receptor (CAR).
  • CAR Coxsackie-Adenovirus Receptor
  • E1A early 1A
  • E2F is released to activate the transcription of other viral genes such as E2, E3 and E4 and cell genes that activate the cell cycle.
  • E1B bonds with p53 to activate the cell cycle and prevent the apoptosis of the infected cell.
  • E2 codifies for replication proteins of the virus, E3 for proteins that inhibit the antiviral immune response and E4 for proteins that transport viral RNA. The expression of these early genes leads to the replication of the viral DNA and once replicated, activates the promoter that regulates the expression of the late or structural genes that form the capsid.
  • oncolytic adenoviruses Methods have been used to construct oncolytic adenoviruses: the selection of viral functions that are not necessary in tumor cells and the replacement of viral promoters with tumor-selective promoters 1 .
  • the gene to be selected or regular gene belongs preferably to the E1 region, and in particular, affects E1a because it controls the expression of other viral genes.
  • the protein E1b-55K has, for example, been eliminated. This protein inactivates p53 to induce in the infected cell the entry in phase S of the cell cycle and to prevent cell apoptosis.
  • E1b-55K A mutated adenovirus in E1b-55K known as Onyx-015 has been used to treat tumors defective in p53 although with little clinical success owing to its low propagation capacity or oncolytic potency.
  • Another mutation performed in the adenoviral genome to achieve selective replication in tumors affects the CR2 field of E1a.
  • This E1a field mediates the bonding to proteins of the Retinoblastoma (Rb) family.
  • pRb proteins block the transition of the Go/G1 phase to the S phase of the cell cycle, forming a complex transcription inhibitor along with E2F.
  • E2F transcription factor of the pRb-E2F complex When E1a bonds with a pRb, the E2F transcription factor of the pRb-E2F complex is released and E2F acts as a transcriptional activator of the genes responsible for moving on to the S phase and viral genes such as E2.
  • the release of E2F is thus a key step in the replication of the adenovirus.
  • tumor cells the cell cycle is out of control because pRb is absent or inactivated by hyperphosphorylation and E2F is free. In these cells, the inactivation of pRb by E1a is now not necessary.
  • an adenovirus with a mutation in E1a called Delta-24 that prevents its bonding with pRb can be propagated normally in cells with inactive pRb 9,10 .
  • the E1a promoter has been replaced by various promoters such as the alpha-fetoprotein promoter, a prostatic-specific antigen (PSA), kallikrein, mucine 1 and osteocalcin 11-15 .
  • PSA prostatic-specific antigen
  • kallikrein kallikrein
  • mucine 1 mucine 1
  • osteocalcin 11-15 a major problem has been identified in the use of cell promoters in the viral context: the existence of viral sequences that interfere with the proper regulation of the promoter and reduce selectivity 16,17 . It has been attempted to correct this loss of selectivity by regulating other viral genes as well as E1a, such as E1b, E2 and E4 18,19 .
  • the regulation of various viral genes can be done with a different promoter for each viral gene, for example the E2F1 promoter for E1a and the telomerase promoter for E4.
  • the two promoters must be expressed at high levels to allow viral replication such that oncolytic potency can remain reduced in many tumor cells 20 .
  • two viral genes can be regular with the same promoter, for example in the oncolytic adenovirus Onyx 411, in which E1a and E4 are regulated by the E2F1 promoter 21 .
  • E1a and E4 are regulated by the E2F1 promoter 21 .
  • E2F1 promoter 2021,29,30 A particularly interesting promoter used in the design of oncolytic adenoviruses is the E2F1 promoter 20,21,29,30 .
  • This promoter presents two E2F bonding sites.
  • the family of E2F transcription factors regulates the transcription of genes that allow entry to the S phase of the cell cycle. These factors serve as activators when they are released and as repressors when they bond with the pRb retinoblastoma protein 31 .
  • the bonding of pRb to E2F is regulated by phosphorylation of pRb such that the phosphorylation of pRb prevents its bonding with E2F.
  • Tumors present alterations in the signal-translation routes that result in the hyperphosphorylation of pRb and an increase in free E2F.
  • genes are expressed that respond to E2F such as the E2F1 gene.
  • pRb is not phosphorylated and remains bonded to E2F, forming a complex that acts as a transcriptional repressor.
  • oncolytic adenoviruses however, the simple regulation of E1a with the E2F1 promoter results in a low level of selective replication in tumors, of the order of 10 times 20 .
  • the regulation of other viral genes in addition to E1a is a possible solution to this low selectivity, but presents the problems described in the paragraph above.
  • OAS403 is an oncolytic adenovirus with E1a regulated with the promoter of E2F1 and E4 regulated with the promoter of telomerase, which furthermore includes a polyadenylation signal to eliminate transcription from the ITR (inverted terminal repetition) and in which the packaging signal has been relocated to the extreme right of the genome to reduce interference with the E1a promoter 20 .
  • ITR inverted terminal repetition
  • packaging signal has been relocated to the extreme right of the genome to reduce interference with the E1a promoter 20 .
  • the packaging signal and sequences adjacent to E4 change position in the genome 22 . It has moreover been described that even minor modifications of the E4 region cause genomic instability, and so strategies based on modification of the E4 region have been abandoned 22 .
  • E2F1 promoter Another problem found with the E2F1 promoter apart from its selectivity is the lack of potency.
  • an oncolytic adenovirus with E1a regulated by the E2F1 promoter loses its lytic capacity with regard to the salvage adenovirus as shown by Ryan et al. 20 and in the examples presented in this invention.
  • This invention describes the use of appropriate DNA sequences to achieve the correct functioning of a genome promoter of an oncolytic adenovirus. With these sequences, an oncolytic adenovirus is designed that presents greater selectivity and anti-tumor potency.
  • the use of the elements described in this invention allows the attainment of a high tumor selectivity and oncolytic capacity using only a tumor-specific promoter.
  • the use of a single promoter reduces the problems of genomic instability associated with the repetition of the same promoter in the adenoviral genome.
  • the regulation of only E1a avoiding the regulation of other viral genes, allows the correct temporal regulation of adenoviral genes and prevents the genomic instability associated with modification of the E4 region.
  • This invention refers to an oncolytic adenovirus for cancer treatment that contains a human DNA sequence isolating a promoter that confers selective expression on an adenoviral gene.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • adenovirus contains a sequence that optimizes the protein translation of an adenoviral gene regulated by a promoter that confers tumor selectivity.
  • this sequence is the Kozak sequence.
  • Another object of the invention is an oncolytic adenovirus for cancer treatment that contains a human DNA sequence isolating a promoter of selective expression that regulates an adenoviral gene and a sequence that optimizes the protein translation of the same adenoviral gene.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • Another object of this invention is an adenovirus that contains a human DNA sequence isolating a promoter of selective expression that regulates an adenoviral gene and a sequence that optimizes the protein translation of the same adenoviral gene and that also presents mutations in one or more genes of the E1a, E1b and E4 group to achieve selective replication in tumors.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • Yet another object of this invention is an oncolytic adenovirus that contains a human DNA sequence isolating a promoter of selective expression that regulates an adenoviral gene and a sequence that optimizes the protein translation of the same adenoviral gene and modifications in its capsid to increase its infectivity or to direct it to a receptor present in a tumor cell.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • Yet another object of this invention is an oncolytic adenovirus that contains a human DNA sequence isolating a promoter of selective expression that regulates an adenoviral gene and a sequence that optimizes the protein translation of the same adenoviral gene and that said adenovirus, in turn, contains other genes commonly used in the field of cancer gene therapy as prodrug activators, tumor suppressors or immunostimulators.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • Yet another object of this invention is an oncolytic adenovirus that contains a human DNA sequence isolating a promoter of selective expression that regulates an adenoviral gene and a sequence that optimizes the protein translation of the same adenoviral gene
  • the adenovirus is a human adenovirus derived from a serotype between 1 and 50.
  • the adenovirus is a human adenovirus serotype 5.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • Yet another object of this invention is an oncolytic adenovirus that contains a human DNA sequence isolating the promoter of the modified human E2F1 gene by the addition of sites for bonding to E2F to regulate the expression of an adenoviral gene and a sequence that optimizes the protein translation of the same gene.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • Another object of this invention is a pharmaceutical composition that includes an effective quantity of an oncolytic adenovirus that contains a human DNA sequence isolating a promoter of selective expression that regulates an adenoviral gene and a sequence that optimizes the protein translation of the same adenoviral gene and one or more pharmaceutically acceptable carriers and excipients.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • Another object of this invention is the use of an oncolytic adenovirus that contains a human DNA sequence isolating a promoter of selective expression that regulates an adenoviral gene and a sequence that optimizes the protein translation of the same adenoviral gene for the preparation of a drug for the treatment or prevention of cancer or a premalignant condition thereof.
  • the human DNA sequence is a sequence derived from the locus of myotonic dystrophy.
  • the adenovirus of this invention may optionally be combined with other methods of cancer treatment such as chemotherapy or radiotherapy.
  • This invention describes an oncolytic adenovirus that contains a human DNA sequence, in particular a sequence derived from the locus of myotonic dystrophy, as a sequence isolating a promoter of selective expression that regulates an adenoviral gene and, in turn, contains a sequence that optimizes the protein translation of the same adenoviral gene, as well as the use of said oncolytic adenovirus for the treatment or prevention of cancer or a premalignant condition thereof.
  • Previously, the use of isolating sequences derived from B-globin of chickens in adenoviral vectors has been described 26,27 .
  • the isolators described previously are not of human origin and have not been used in a context of oncolytic adenoviruses.
  • the locus of myotonic dystrophy is located in the human chromosome 13 in the position 19q13.3. This locus contains two bonding sites for the CTCF protein and a variable number according to each individual of CTG repetitions that jointly function as a potent isolator of the effect of enhancers or activators on promoters 32 .
  • its activity had never been analyzed in a viral genome. Its activity in a viral genome is not obvious, as its activity has been demonstrated only in the context of a cell chromosome in which the associated histones can play a role in its functioning.
  • Its human origin offers a superior alternative to the use of the HS4 sequence of chickens as the transfer of sequences of non-human origin can have biosafety implications.
  • this invention describes the use of an optimized sequence for protein translation to increase the levels produced of the adenoviral protein regulated below the tumor-specific promoter.
  • the regulation of the expression of a viral gene with a tumor-selective promoter presents the disadvantage that the level of expression is usually lower than the level of expression observed in Ad5. This lower expression results in lower replicative potency of the oncolytic adenovirus. Insertion of the Kozak sequence at the beginning of translation of the gene regulated by the selective promoter is capable of restoring the levels of expression of the gene regulated.
  • This invention also describes the strategy of increasing the number of binding sites at E2F in the sequence of the human promoter E2F1 to better control the expression of E1a in an oncolytic adenovirus.
  • This increase in binding sites at E2F produces greater expression of E1a in tumor cells and reduced expression of E1a in normal cells, resulting in an increase in tumor selectivity of adenoviral replication.
  • the invention is directed towards the need to find better treatments for cancer, including, but not limited to, cancer of the pancreas, colon and lung.
  • Cancer treatment with the oncolytic adenovirus that contains the human DNA sequence and the sequence that optimizes protein translation can be performed by direct injection inside the tumor or by systemic intravenous injection in patients suffering from cancer using standard methods in the field of gene therapy and virotherapy with adenoviruses.
  • FIG. 1 Structure of adenovirus expounded in this invention.
  • the arrows indicate the modifications most representative of each virus regarding parental versions.
  • Adwt is the wild virus with no modification. It shows inverted terminal regions (ITR) for its replication and a packaging signal ( ⁇ ) together with ITR on the left.
  • ITR inverted terminal regions
  • packaging signal
  • the virus AdwtRGD coincides with Adwt but also contains the tripeptide sequence RGD (Arginine-Glycine-Aspartic Acid) in the sequence of the viral fiber. This sequence serves to bind 5 integrins that are overexpressed in the membrane of tumor cells. This virus is used as a positive control of replication.
  • the virus Ad- ⁇ 24RGD is similar to AdwtRGD but has a deletion of 24 nucleotides, corresponding to 8 amino acids, in the binding site of E1a to pRB. Said deletion prevents the separation of complex pRB-E2F present in quiescent normal cells so that said virus is preferentially replicated in cells in division or tumor cells. This virus is used to compare the level of selectivity of the viruses described in this invention.
  • the Ad-TLRGD virus is a virus similar to AdwtRGD but with the E1 region replaced by luciferase genes and green fluorescence protein (GFP) genes. This virus because it lacks the E1 region cannot be replicated and is used as a negative control.
  • the ICOVIR viruses are derived from Ad- ⁇ 24RGD by the substitution of the E1a promoter by a selective activation promoter in tumors, promoter E2F1.
  • ICOVIR1 is similar to Ad- ⁇ 24RGD but contains said substitution.
  • This virus is used as a control of E1A expression, controlled by promoter E2F1 in the absence of insulating sequences of the promoter.
  • ICOVIR2 is similar to ICOVIR1 but contains a sequence of the myotonic dystrophy locus at promoter E2F1.
  • ICOVIR5 also contains the Kozak sequence in the beginning of translation of E1a in order to optimize its translation and thus increase the levels of expression of E1a in tumor cells.
  • ICOVIR7 also has two additional binding sites to E2F in the E2F1 promoter.
  • the ICOVIR2, 5 and 7 viruses serve to demonstrate the object of this invention: the best gene regulation when the DM insulating sequence is used.
  • FIG. 2 Diagram of the functioning of oncolytic adenoviruses containing the DM sequence of the myotonic dystrophy insulating locus at promoter E2F1 that regulates E1a.
  • the oncolytic viruses ICOVIR2, 5 and 7 contain promoter E2F1 insulated with the DM sequence.
  • the first codon of the E1a sequence is preceded by the Kozak sequence (CCACC) to optimize protein translation.
  • CCACC Kozak sequence
  • the promoter of E2F1 is modified by the insertion of additional binding sites to E2F to increase its potency and selectivity.
  • complex pRB E2F acts as a repressor of the promoter of E2F1 through the action of histone deacetylases (HDAC) and E1a is not expressed.
  • HDAC histone deacetylases
  • pRB is hyperphosphorylated or absent and E2F is free. In this manner it acts as a transcriptional activator of E1a.
  • the Kozak sequence preceding E1a allows a correct level of expression of E1a.
  • the insulating DM avoids the interference of the ITR and adenoviral packaging signal in the modified E2F1 promoter.
  • FIG. 3 demonstration of the effect on the expression of E1a resulting from the insertion of a DM insulating sequence in front of the E2F1 promoter.
  • Endothelial cells from human umbilical cord (HUVEC), human fibroblasts and human hepatocytes are used as controls of non-tumor cells.
  • the cell lines NP-9 (adenocarcinoma of the pancreas), A-549 (adenocarcinoma of the lung), FaDu (head and neck tumors), SCC25 (head and neck tumors), SKMel-28 (melanoma) and 1.36.1.5 (melanoma) are used as a model of the human tumor cell.
  • Adwt and AdwtRGD positive controls of non-selective expression of E1a
  • Ad- ⁇ 24RGD non-selective expression of E1a-D24
  • ICOVIR1 E1a controlled by promoter E2F1
  • ICOVIR2 E1a controlled by promoter E2F1 insulated with the DM sequence, object of this invention
  • O represents a cellular extract of uninfected cells.
  • virus Ad-TLRGD which has region E1 replaced by luciferase genes and green fluorescence protein (GFP) genes. This negative control shows no expression of E1a.
  • ICOVIR1 and ICOVIR2 tumor cells are capable of expressing E1a, but in FaDu, SCC25 and SKMel-28, the expression of E1a in cells infected with ICOVIR1 or ICOVIR2 is lower than that obtained with the adenovirus where E1a is not regulated by E2F1 (virus Adwt, AdwtRGD and Ad- ⁇ 24RGD).
  • E2F1 virus Adwt, AdwtRGD and Ad- ⁇ 24RGD.
  • the promoter of E2F1 insulated or not with DM, does not have the power required to allow a level of expression of E1a in tumor cells comparable to wild adenovirus.
  • this invention solves this problem with insertion of the Kozak sequence in E1a (in ICOVIR5) and modification of the promoter E2F1 (in ICOVIR7).
  • FIG. 4 The DM sequence allows for an increase in antitumor selectivity of an oncolytic adenovirus with E1a regulated with the promoter of E2F1.
  • the amount of virus in the cell extract was determined by infection of a monolayer of HEK293 cells and subsequent staining with the monoclonal antibody 2Hx-2 (ATCC) that recognizes the adenoviral hexon and a mouse anti-IgG secondary antibody, Alexa 488 (Molecular Probes, Eugene, Oreg.).
  • the monolayer was observed under fluorescence microscope and every fluorescent cell was quantified as a transduction unit (TU). Thus, the number of units per milliliter of viral extract was determined. The bars show said number of units of transduction per milliliter of viral extract.
  • ICOVIR2 The presence of the DM insulating sequence in ICOVIR2 results in a lower viral replication in normal fibroblasts and HUVEC compared with ICOVIR1 which has the non-insulated promoter E2F1.
  • NP-9 adenocarcinoma of the pancreas
  • A-549 adenocarcinoma of the lung
  • FaDu head and neck tumors
  • SCC25 head and neck tumors
  • SKMel-28 melanoma
  • 1.36.1.5 (melanoma).
  • this invention describes the method to preserve the selectivity provided by promoter E2F1 insulated with DM by increasing the replicative capacity via the insertion of the Kozak sequence in E1a and modification of promoter E2F1.
  • FIG. 5 Effect of inserting the Kozak sequence to increase the power of the promoter insulated with DM.
  • Human fibroblasts were infected with AdwtRGD (positive control of infectivity increased by the RGD sequence and non-selective expression of E1a) and oncolytic viruses Ad- ⁇ 24RGD (non-selective expression of E1a- ⁇ A24), and ICOVIR5 (E1a preceded by the Kozak sequence and controlled by promoter E2F1 insulated with the DM sequence). After 24 hours the cells were read and E1a was detected by Western blot. The band corresponding to E1a in fibroblasts infected with ICOVIR5 is less intense than that for fibroblasts infected with the control virus.
  • FIG. 6 In vitro oncolytic effectiveness of adenoviruses containing E1a regulated with the promoter of E2F1 insulated with the DM sequence and the Kozak sequence to optimize translation of E1a.
  • Cells from the melanoma tumor line SKMel28 or head and neck tumor FaDu were cultured in wells from a plate of 96 wells (3000 cells per well) and were infected with increasing concentrations of AdwtRGD (positive control of infectivity increased by the RGD sequence and non-selective expression of E1a), ICOVIR2 (E1a controlled by the promoter E2F1 insulated with the DM sequence), or ICOVIR5 (E1a preceded by the Kozak sequence and controlled by the promoter E2F1 insulated with the DM sequence).
  • AdwtRGD positive control of infectivity increased by the RGD sequence and non-selective expression of E1a
  • ICOVIR2 E1a controlled by the promoter E2F1 insulated with the DM sequence
  • ICOVIR5 E1a preceded by the Kozak sequence and controlled by the promoter E2F1 insulated with the DM sequence.
  • the X-axis shows the concentration of viral particles
  • CPE cytopathic effect
  • FIG. 7 Effect of modification of E2F1 promoter to increase its power when it is insulated with the DM sequence.
  • Cells of the melanoma tumor line 1.36.1.5. were infected with Ad-TLRGD (non-replicative negative control of virus for lack of E1a), AdwtRGD (positive control of infectivity increased by the RGD sequence and non-selective expression of E1a), and oncolytic viruses ICOVIR2 (E1a controlled by the E2F1 promoter insulated with the DM sequence), ICOVIR5 (E1a preceded by the Kozak sequence and controlled by the promoter E2F1 insulated with the DM sequence) and ICOVIR7 (E1a preceded by the Kozak sequence and controlled by a promoter E2F1 modified by two additional binding sites to E2F and insulated with the DM sequence).
  • Ad-TLRGD non-replicative negative control of virus for lack of E1a
  • AdwtRGD positive control of infectivity increased by the RGD sequence and non-selective expression of E1a
  • oncolytic viruses ICOVIR2 (
  • the amount of virus in the cell extract was determined by infection of a monolayer of HEK293 cells and subsequent staining with the monoclonal antibody 2Hx-2 (ATCC) that recognizes the adenoviral hexon and a mouse anti-IgG secondary antibody, Alexa 488 (Molecular Probes, Eugene, Oreg.). The monolayer was observed under fluorescence microscope and every fluorescent cell was quantified as a transduction unit (TU).
  • ATCC monoclonal antibody 2Hx-2
  • Alexa 488 Molecular Probes, Eugene, Oreg.
  • the number of transduction units per milliliter (TU/ml) of viral extract was determined.
  • AdwtRGD is used, in which E1a is not regulated.
  • ICOVIR7 is capable of propagating with the same power as the control AdwtRGD.
  • FIG. 8 An adenovirus containing E1a regulated with promoter E2F1 insulated with the DM sequence and Kozak sequence at the beginning of translation of E1a can be used to treat tumors.
  • the top of the figure shows an experiment in vivo with athymic mice of the BALB/c strain containing NP9 tumors.
  • the tumors were injected with PBS ( ⁇ ) or 109 viral particles of ICOVIR-2 ( ⁇ ) or AdwtRGD ( ⁇ ).
  • the graph shows the evolution of tumor volume.
  • ICOVIR2 can inhibit tumor growth.
  • FIG. 9 Demonstration in vivo of the reduction of toxicity after intravenous injection of adenovirus containing regulated E1a with the promoter of E2F1 insulated with the DM sequence and the Kozak sequence to optimize translation of E1a.
  • the toxicity in vivo of an adenovirus containing the Kozak sequence in E1a and a promoter E2F1 insulated by DM was compared with that of the wild virus Adwt and the oncolytic virus Ad- ⁇ 24RGD expressing E1a under its natural promoter.
  • the viruses were administered intravenously at different doses (10 10 , 5 ⁇ 10 10 and 10 11 ) in immunocompetent Balb/c mice. For 3 days post-injection, parameters associated with toxicity were evaluated. A shows the number of deaths with respect to the number of animals treated. This mortality includes animals sacrificed for having a weight loss equal to or greater than 20%.
  • B represents the percentage variation in body weight for each group of animals treated with the control vehicle (PBS) or different viruses at the doses indicated.
  • C shows the international units (IU) of serum transaminases aspartate aminotransferase (AST) and alanine-aminotransferase (ALT) per liter of blood plasma detected after the intravenous injection of the control vehicle or the viruses indicated at the doses indicated.
  • D shows the number of platelets per milliliter of blood detected after intravenous injection of the control vehicle or the viruses indicated at the doses indicated. For each of these parameters the toxicity associated with the administration of ICOVIR 5 is very low even at the highest dose.
  • FIG. 10 Demonstration in vivo of reduction in the expression of E1a in non-tumor tissue and toxicity after intravenous injection of adenovirus containing E1a regulated with the promoter of E2F1 insulated with the DM sequence and the Kozak sequence to optimize the translation of E1a.
  • This invention describes the use in cancer treatment of adenoviruses that contain E1a regulated with the E2 ⁇ l promoter isolated with the MD sequence, the Kozak sequence to optimize E1a translation and the addition of sites for bonding to E2F in the E2F1 promoter.
  • the treatment is based on the selective replication of these viruses in tumors that have an altered retinoblastoma route.
  • the retinoblastoma route is the set of protein interactions that occur from the cell membrane up to the nucleus to regulate the level of phosphorylation of the protein of retinoblastoma pRb. Cancer is characterized by an alteration of this route such that the pRb protein is hyperphosphorylated or lost. This pRb alteration causes a loss of pRb bonding to the E2F transcription factor and an increase in free E2F in the nucleus of the tumor cells. This transcription factor bonds to the promoters with specific E2F bonding sites, as an E2F1 promoter, to increase its expression.
  • the selective-replication mechanism in tumors of adenoviruses containing E1a regulated with the E2F1 promoter isolated with the MD sequence, the Kozak sequence at the start of E1a translation and the addition of sites for bonding to E2F in the E2F1 promoter is based on the idea that the presence of free E2F in the tumors activates the expression of the E2F1 promoter in this virus and is indicated in FIG. 2 of this invention.
  • the presence of the MD sequence enables correct activation of the promoter.
  • the presence of the Kozak sequence enables synthesis of a quantity of E1a sufficient for maintaining the appropriate replicative and lytic capacity of the oncolytic virus.
  • the presence of additional sites for bonding to E2F in the E2 ⁇ l promoter enables an increase in the level of expression of E1a to maintain the appropriate replicative and lytic capacity of the oncolytic virus.
  • the DM insulating human sequence derived from the locus of myotonic dystrophy is represented by SEQ. ID 1 (from position 368 to 1096 of sequence 1).
  • the DM sequence is characterized in that it contains two binding sites to factor CTCF and a variable number of repetitions of sequence CGT which function together as a powerful insulator against transcriptional interference 32 .
  • the DM sequence acts to insulate the effect of enhancers, located in the sequence of adenovirus packaging next to the promoter of E1a.
  • the promoter of E1a is replaced by a selective promoter of tumors such as, for example, the promoter E2F1 and, to insulate this promoter from the enhancers present in the sequence of adenoviral packaging, the DM sequence is inserted between said sequence of packaging and promoter E2F1.
  • the sequence of the promoter of E2F1 is shown in SEQ. ID 1 (from position 1283 until position 1564 of sequence 1). This promoter is characterized by having two binding sites to E2F organized in imperfect palindromes and four binding sites to Sp1 34 .
  • the sequence of promoter E2F is modified by the insertion of binding sites to E2F in addition to those that already exist in the wild human promoter (from position 1321 until position 1447 of SEQ.
  • adenoviral genome 41 There are several ways to manipulate the adenoviral genome.
  • the methods of construction of genetically modified adenoviruses are well established in the field of gene therapy and virotherapy with adenovirus 36-41 .
  • the most commonly used method is based on first building the genetic modification desired into a plasmid that contains the adenoviral region to be modified, and then performing a homologous recombination in bacteria with a plasmid that contains the first of the viral genome 41 . This process can be as follows:
  • adenoviruses with the expression of a viral gene regulated by the selective promoter isolated with the MD sequence and potentiated with the Kozak sequence can contain modifications of their capsid to increase their inefficacy or be directed to receptors present in the tumor cell.
  • the proteins of the adenoviral capsid have been genetically modified to include ligands that increase inefficacy or direct the virus to a receptor in the tumor cell 47-53 . Directing the adenovirus to the tumor can also be achieved with bifunctional ligands that bond to the virus on one side and to the tumor receptor on the other 53-56 .
  • the capsid can be covered with polymers such as polyethylene glycol 57-60 .
  • polymers such as polyethylene glycol 57-60 .
  • Another feature of this invention is adenoviruses that contain E1a regulated with the E2F1 promoter isolated with the MD sequence, the Kozak sequence at the start of E1a translation and the addition of sites for bonding to E2F in the E2F1 promoter, but which are derived from other serotypes of adenoviruses other than Ad5.
  • Another feature of this invention refers to adenoviruses that contain E1a regulated with the E2F1 promoter isolated with the MD sequence, the Kozak sequence at the start of E1a translation and the addition of sites for bonding to E2F in the E2F1 promoter and that, in turn, contain other genes for increasing their cytotoxicity to tumor cells such as the gene of thymidine kinase, cytosine deaminase, proapoptotic genes, immunostimulators or tumor suppressors.
  • the adenoviruses described in this invention are propagated following standard methods in the fields of adenovirology and adenoviral vectors 36,37 .
  • the preferred propagation method is by infection of a cell line permitting the replication of adenoviruses that contain E1a regulated with the E2F1 promoter isolated with the MD sequence, the Kozak sequence at the start of E1a translation and the addition of sites for bonding to E2F in the E2F1 promoter.
  • the line of pulmonary adenocarcinoma A549 is an example of this line. Propagation is performed, for example, as follows: The A549 cells are grown on plastic plates for cell cultivation and infected using 50 viral particles per cell.
  • the cells are collected and stored in tubes. After centrifugation at 1,000 rpm for 5 minutes, the cell precipitate is frozen and thawed three times to break the cells. The resulting cell extract is centrifuged at 1,000 rpm for 5 minutes and the supernatant with viruses is loaded above a gradient of caesium chloride and centrifuged for 1 hour at 35,000 rpm. The virus band in the gradient is reloaded above another gradient of caesium chloride and centrifuged for 16 hours at 35,000 rpm. The virus band is collected and dialyzed with PBS-10% glycerol. The dialyzed virus is aliquoted and stored at ⁇ 80° C. The number of particles and plate-forming units is quantified following standard protocols 39 .
  • a saline phosphate buffer with glycerol at 10% is a standard formulation for storing adenoviruses.
  • new formulations have been described that improve the stability of the virus 61,62 .
  • This invention describes the use of adenoviruses that contain E1a regulated with the E2F1 promoter isolated with the MD sequence, the Kozak sequence at the start of E1a translation and the addition of sites for bonding to E2F in the E2F1 promoter for the treatment of cancer.
  • the treatment is based on the selective replication of these viruses in cells with an active RB route.
  • the protocols for using the viruses described in this invention in the treatment of cancer follow the same procedures as those used in the fields of virotherapy with adenoviruses and gene therapy with adenoviruses.
  • adenoviruses with selective-replication methods other than that proposed in this invention have been used to treat cancer 9,37,63-68 .
  • the purified adenovirus in any of the forms described above is added to the culture medium for the infection of tumoral cells.
  • the adenovirus can be administered locoregionally by injection in the tumor or in a body cavity where the tumor is located, or even systematically by injection into the bloodstream.
  • adenovirus replications can be administered loco-regionally by injection in the tumor or in a body cavity where the tumor is located, or systemically by injection in the bloodstream.
  • the treatment of tumors with the adenoviruses described that are the subject of this invention can be combined with other methods of treatment such as chemotherapy or radiotherapy.
  • An adenovirus was constructed with E1a regulated with the E2F1 promoter isolated with the MD sequence as follows: To generate ICOVIR-1 (Ad-E2F- ⁇ 24RGD), the human E2F1 promoter was obtained by PCR of mononuclear cells of human peripheral blood using oligonucleotides stretching from the pair of bases ⁇ 218 to +51 of the E2F-1 promoter (position +1 indicates the start of transcription). The oligonucleotides contained KpnI and HindIII restriction targets for cloning in the plasmid pGL3 (Promega, Southampton, UK). The resulting plasmid was called pGL3-E2F.
  • pE2F- ⁇ 24 by recombination with a plasmid containing the 5,766 pairs of base from the extreme left of the adenoviral genome except nucleotides (nt) 122 and 129 of E1a (derived from pXC1- ⁇ 24 with a HindIII site between nt 348 and nt 522 of the Ad5 genome 9 ).
  • pE2F- ⁇ 24 was recombined with pShuttle 41 to obtain pShuttle-E2F- ⁇ 24.
  • This plasmid was linearized with Pme1 and recombined with pVK503 (which contains the Ad5 sequence with the fiber modified with RDG 69 ) to generate the plasmid pAd-E2F- ⁇ 24RGD or pICOVIR-1.
  • the combination of the E2F1 promoter and other modifications described in this invention with the E1a mutation called ⁇ 24 and the insertion of the peptide RGD in the fiber was done to demonstrate that the modifications presented in this invention increases the oncolytic potency and selectivity of a virus known as selective towards Rb and powerful in the field of oncolysis (adenovirus Ad- ⁇ 24RGD 70 ).
  • the mutation ⁇ 24 and the insertion of peptide RGD are modifications described above in the field of virotherapy of cancer. In particular, they have been described together in reference 70 of this invention.
  • This reference describes the use of the RGD peptide.
  • This peptide is a tripeptide formed by the amino acids Arginine, Glycine and Aspartic Acid, which are bound to the integrins. Since the integrins are over-expressed in tumor cells, tripeptide RGD serves to increase the infectivity of the virus in tumor cells and is used for this purpose.
  • the virus ICOVIR1 was generated by digestion with PacI of this plasmid and transfection in HEK293 cells.
  • ICOVIR-2 Ad-DM-E2F- ⁇ 24RGD
  • the DM-1 insulating sequence was obtained from PCR of human peripheral mononuclear blood cells using oligonucleotides that amplify from nt 13006 to nt 13474 of locus DM1 (sequence published in GenBank with number L08835). Oligonucleotides of the PCR were designed to incorporate flanking sites Xho I. DM-1 was subcloned in Xho1 of pShuttle-E2F- ⁇ 24 described above to obtain pShuttle-DM-E2F- ⁇ 24.
  • the correct orientation of the DM1 fragment was verified by restriction with BamH1, Hind111, Xho1 and Sma1.
  • pShuttle-DM-E2F- ⁇ 24 is recombined with pVK503 to generate plCOVIR2.
  • the virus ICOVIR2 was generated by digestion with PacI from this plasmid and transfection in HEK293 cells.
  • ICOVIR1 and ICOVIR2 spread in the A549 line and were purified by methods described in gene therapy and virotherapy 36 .
  • the correct structure of the genomes of ICOVIR-1 and ICOVIR-2 was verified by restriction with Kpn1 and HinIII, respectively.
  • DM-1 region, promoter E2F, mutation E1A- ⁇ 24 and the region of the fiber containing RGD were sequenced.
  • the oligonucleotides used for these sequencings are: DM1-Up (5′-GGGCAGATGGAGGGCCTTTTATTC-3′), E2F-Up (5′-GTGTTACTCATAGCGCGTAA-3′), ⁇ 24-down (5′-CCTCCGGTGATAATGACAAG-3′) and FiberUp (5′-CAAACGCTGTTGGATTTATG-3′). The sequences obtained are shown in SEQ. ID 1.
  • the cells were lysed in a lysis buffer (400 mM NaCl, 1 mM EDTA, 5 mM NaF, 10% glycerol, 1 mM sodium orthovanadate, 0.5% Nonidet P-40, 100 ⁇ g/ml phenylmethylsulfonyl fluoride, 1 ⁇ g/ml leupeptin and 10 ⁇ g/ml aproptinin in 10 mM Tris-HCl (pH 7.4) for 1 hour at 4° C.
  • a lysis buffer 400 mM NaCl, 1 mM EDTA, 5 mM NaF, 10% glycerol, 1 mM sodium orthovanadate, 0.5% Nonidet P-40, 100 ⁇ g/ml phenylmethylsulfonyl fluoride, 1 ⁇ g/ml leupeptin and 10 ⁇ g/ml aproptinin in 10 mM Tris-HCl (pH 7.4) for 1 hour at 4°
  • the lysate was centrifuged at 14,000 rpm, and the supernatant with proteins was separated by electrophoresis in 10% SDS-PAGE (25 ⁇ g/track, determined by Bradford, BioRad, CA, USA) and transferred to nitrocellulose (Schleicher and Schuell, Dassel, Germany).
  • the membrane was blocked with 5% skimmed milk, 0.05% Tween 20 and 0.9% NaCl in 50 mM Tris (pH 7.5), and incubated for 16 hours at 4° C. with a polyclonal antibody anti-adenovirus-2-E1a (clone 13 S-5, Santa Cruz Biotechnology Inc., Santa Cruz, Calif., USA).
  • E1a was detected with a secondary anti-rabbit IgG antibody (DAKO A/S) joined with peroxydase and Amersham's Enhanced Chemiluminescence protocol (Amersham, Arlington Heights, Ill., USA). The result is shown in FIG. 3 of this invention. It is shown that the presence of the E2F1 promoter (ICOVIR1) is capable of reducing the expression of E1a in normal cells. But the MD sequence confers greater control of the expression of E1a by the E2F promoter (ICOVIR2).
  • both ICOVIR1 and ICOVIR2 are capable of expressing E1a, but it is important to note that in some tumor lines such as FaDu, SCC25 and SKMel-28, the expression of E1a is less than that obtained with the salvage adenovirus and the oncolytic AdD24RGD in which E1a is not regulated by E2F1. This indicates that the E2F1 promoter, whether isolated or not with MD, does not have the necessary potency to enable a level of expression of E1a in tumor cells comparable to the salvage adenovirus.
  • the cells were infected with ICOVIR1 and ICOVIR2 as described in the previous paragraph. Five days after infection, the cells and their culture media were collected and submitted to three cycles of freezing-thawing to release the virus. The quantity of the virus in the cell extract was determined by infection in HEK293 and anti-hexon staining using the monoclonal antibody 2Hx-2 (ATCC) and a secondary antibody, Alexa 488 anti-IgG of a rat (Molecular Probes, Eugene, Oreg.). The result is shown in FIG. 4 .
  • ICOVIR1 The presence of the E2F1 promoter in ICOVIR1 reduces viral replication in normal cells (fibroblasts and HUVEC). However, the isolating sequence in ICOVIR2 results in lower viral replication. In certain tumor-cell lines such as A549, ICOVIR1 and ICOVIR2 show a level of replication similar to the salvage adenovirus Adwt, but in the majority of tumor lines, its replicative capacity is less than that of Adwt.
  • the Kozak Sequence Enables an Increase in the Expression of E1a an Oncolytic Adenovirus in which the Expression of E1a is Regulated with the E2F1 Promoter Isolated with the MD Sequence
  • An oncolytic adenovirus was constructed with E1a regulated with the E2F1 promoter isolated with the MD sequence and with the Kozak sequence to increase its translation.
  • E1a a fragment of DNA containing the MD sequence, the E2F1 promoter and E1a was isolated from the pShuttle-MD-E2F-D24 described in example 1 by restriction with Kpn1 and subcloned in pGEM3Z (Promega), obtaining the plasmid pGEM-E2F-d24.
  • This plasmid was used to replace the start of E1a translation using oligonucleotides with the Kozak sequence obtaining pGEM-E24-KD24.
  • the Kpn1 fragment thus modified was recloned in Kpn1 from pShuttle-DM-E2F-D24 to obtain pShuttle-DM-E2F-KD24. Finally, pShuttle-DM-E2F-KD24 was recombined with pVK503 to obtain pICOVIR5.
  • the virus ICOVIR5 was generated by digestion with PacI of this plasmid and transfection to HEK293 cells. ICOVIR5 was propagated in the A549 line and purified by methods described in gene therapy and virotherapy 36 . Its structure is presented in FIG. 1 of this invention. The correct sequence of the promoter and E1a was checked by restriction and sequencing. The sequence obtained is shown in SEQ.
  • E1a is expressed conditionally in tumor cells when its expression is regulated with the E2F1 promoter isolated with the MD sequence and in addition, its translation is optimized with the Kozak sequence
  • the expression of E1a was analyzed as described in example 1. In this case, it was included in oncolytic adenovirus ICOVIR5, which is distinguished from ICOVIR2 by the fact that it contains the Kozak sequence in the start of E1a translation. The results are shown in FIG. 5 of this invention.
  • ICOVIR5 does not express E1a by presenting the E2F promoter isolated with MD.
  • the level of expression of E1a is higher in ICOVIR5 than in ICOVIR2, which demonstrates the effect of the Kozak sequence to increase the potency of the promoter isolated with MD.
  • the Kozak Sequence Enables an Increase in the Oncolytic Potency of an Adenovirus in which the Expression of E1a is Regulated with the E2F1 Promoter Isolated with the MD Sequence
  • An oncolytic adenovirus was constructed with E1a regulated with an E2F1 promoter modified by the insertion of four sites for bonding to E2F.
  • E1a regulated with an E2F1 promoter modified by the insertion of four sites for bonding to E2F.
  • pGEM-E2FKE1ad24 described in example 2
  • BsiWI linked two copies of oligonucleotides with the palindromic sequence of bonding to E2F and that had extremes compatible with BsiWI.
  • the promoter thus modified was subcloned in Kpn1 of pShuttle-MD-E2F-D24 to obtain pShMDE2FBsiE2F2KE1ad24.
  • the plasmid pICOVIR7 was obtained.
  • the virus ICOVIR7 was generated by digestion in the A549 line and purified by methods described in gene therapy and virotherapy 36 . Its structure is presented in FIG. 1 of this invention.
  • the correct sequence of the promoter and E1a was checked by restriction and sequencing. The sequence obtained is shown in SEQ. ID 3.
  • E1a in the tumor line 1.36.1.5 of melanoma by western blot as described in example 1.
  • the oncolytic adenovirus ICOVIR7 is distinguished from ICOVIR5 by having the modified E2F1 promoter.
  • the results are shown in FIG. 7 of this invention.
  • the level of expression of E1a is greater in ICOVIR7, which demonstrates the potentiating role of the two additional sites for bonding to E2F in ICOVIR7.
  • the addition of E1a is greater in ICOVIR5 than in ICOVIR2, which demonstrates once again the effect of the Kozak sequence in increasing the potency of the promoter isolated with MD.
  • FIG. 8 shows the tumor volume compared with the start of treatment (day 0). The results are presented as a mean ⁇ SD. The existence of significant differences between results was calculated using a Mann-Whitney non-parametric study of data not paired. The growth curves were compared using a variance analysis. The results were considered significant if p ⁇ 0.05. The calculations were made with the statistics package SPSS(SPSS Inc., Chicago, Ill.). There is a significant difference between tumor size on days 16 and 21.
  • ICOVIR5 Tumors of the cell line of human melanoma SKMel-28 (1.10 7 cells/tumor) were planted in Balb C nu/nu athymic rats, and once established, were treated by administration in the tail vein with PBS, with a single injection on day 0 of ICOVIR-5 of 2.5.10 10 viral particles (vp), or 1.10 11 vp, or with an injection of 3.10 10 vp and another of 1.10 11 one hour apart. The results are shown in the lower part of FIG. 8 of this invention. All regimes of treatment with ICOVIR-5 showed oncolytic activity that results in a suppression of tumor growth that is significantly different from the control group (PBS), p ⁇ 0.05.
  • PBS control group
  • the toxicity in vivo of an adenovirus that contains the Kozak sequence in E1a and an E2F1 promoter isolated by MD was compared with that of a salvage virus and the oncolytic virus AdD24RGD that expresses E1a below the salvage promoter.
  • the viruses were administered intravenously at different doses and at 5 days post-injection, we assessed parameters related to toxicity, such as animal survival, body weight, level of serum transaminases, and blood count. The results are shown in FIG. 9 of this invention.
  • the lethal-dose 50 value (LD 50 ) for AdwtRGD or Ad ⁇ 24RGD in immunocompetent Balb/C rats is located in 5.10 10 viral particles (vp)/rat on day 5 post-injection, while the double of this dose (1.10 11 vp/rat) is lethal for only 10% of rats (LD 10 ) injected with ICOVIR-5.
  • the rats injected with 5.10 10 vp of AdwtRGD or Ad ⁇ 24RGD on day 5 post-injection experienced significant weight loss, while the weight of the rats injected with ICOVIR-5 increased.
  • the analysis of the expression of the adenoviral protein E1A in the rats' livers by immunodetection in frozen sections obtained on day 5 post-injection shows that the presence of an isolated version of the E2F-1 promoter in ICOVIR-5 is effective in restricting the expression of viral proteins, even when the dose administered is increased ( FIG. 10 ).
  • the histological assessment by staining with hematoxylin/eosin of sections in paraffin of the livers on day 3 post-injection also confirmed the low toxicity of ICOVIR-5 ( FIG. 10 ).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Virology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Plant Pathology (AREA)
  • Molecular Biology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US12/184,881 2006-01-02 2008-08-01 Oncolytic Adenoviruses for Cancer Treatment Abandoned US20090311219A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/327,840 US20150071881A1 (en) 2006-02-01 2014-07-10 Oncolytic Adenoviruses for Cancer Treatment
US15/144,637 US10016470B2 (en) 2006-02-01 2016-05-02 Oncolytic adenoviruses for cancer treatment
US16/028,037 US20190183946A1 (en) 2006-02-01 2018-07-05 Oncolytic adenoviruses for cancer treatment

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
ESP200600216 2006-01-02
ES200600216A ES2304281B1 (es) 2006-02-01 2006-02-01 Adenovirus oncoliticos para el tratamiento del cancer.
PCT/ES2007/000050 WO2007088229A1 (es) 2006-02-01 2007-01-31 Adenovirus oncolíticos para el tratamiento del cáncer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/ES2007/000050 Continuation WO2007088229A1 (es) 2006-01-02 2007-01-31 Adenovirus oncolíticos para el tratamiento del cáncer

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/327,840 Continuation US20150071881A1 (en) 2006-02-01 2014-07-10 Oncolytic Adenoviruses for Cancer Treatment

Publications (1)

Publication Number Publication Date
US20090311219A1 true US20090311219A1 (en) 2009-12-17

Family

ID=38327139

Family Applications (4)

Application Number Title Priority Date Filing Date
US12/184,881 Abandoned US20090311219A1 (en) 2006-01-02 2008-08-01 Oncolytic Adenoviruses for Cancer Treatment
US14/327,840 Abandoned US20150071881A1 (en) 2006-02-01 2014-07-10 Oncolytic Adenoviruses for Cancer Treatment
US15/144,637 Expired - Fee Related US10016470B2 (en) 2006-02-01 2016-05-02 Oncolytic adenoviruses for cancer treatment
US16/028,037 Abandoned US20190183946A1 (en) 2006-02-01 2018-07-05 Oncolytic adenoviruses for cancer treatment

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/327,840 Abandoned US20150071881A1 (en) 2006-02-01 2014-07-10 Oncolytic Adenoviruses for Cancer Treatment
US15/144,637 Expired - Fee Related US10016470B2 (en) 2006-02-01 2016-05-02 Oncolytic adenoviruses for cancer treatment
US16/028,037 Abandoned US20190183946A1 (en) 2006-02-01 2018-07-05 Oncolytic adenoviruses for cancer treatment

Country Status (8)

Country Link
US (4) US20090311219A1 (zh)
EP (1) EP1990418B1 (zh)
JP (1) JP5075839B2 (zh)
CN (1) CN101484583A (zh)
AU (1) AU2007211434A1 (zh)
CA (1) CA2640528C (zh)
ES (1) ES2304281B1 (zh)
WO (1) WO2007088229A1 (zh)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140227226A1 (en) * 1999-05-12 2014-08-14 The Uab Research Foundation Infectivity-enhanced conditionally-replicative adenovirus and uses thereof
US20150374766A1 (en) * 2013-03-14 2015-12-31 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US10849945B2 (en) 2015-04-30 2020-12-01 Psioxus Therapeutics Limited Oncolytic adenovirus encoding a B7 protein or active fragment
US11130968B2 (en) 2016-02-23 2021-09-28 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
US11155622B2 (en) 2015-12-17 2021-10-26 Psioxus Therapeutics Limited Virus encoding an anti-TCR-complex antibody or fragment
US11401529B2 (en) 2016-02-23 2022-08-02 Salk Institute For Biological Studies Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics
US11439678B2 (en) 2013-10-25 2022-09-13 Psioxus Therapeutics Limited Oncolytic adenoviruses armed with heterologous genes
US11813337B2 (en) 2016-12-12 2023-11-14 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof
US11840702B2 (en) 2017-08-28 2023-12-12 Akamis Bio Limited Adenovirus armed with bispecific T cell activator

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2304281B1 (es) * 2006-02-01 2009-08-12 Dnatrix Inc. Adenovirus oncoliticos para el tratamiento del cancer.
US8138318B2 (en) * 2007-09-13 2012-03-20 Abbott Laboratories Hepatitis B pre-S2 nucleic acid
WO2010097419A1 (en) 2009-02-25 2010-09-02 Fundació Privada Centre De Regulació Genòmica (Crg) Conditionally replicating adenovirus effective in the treatment of tumors
ES2355882B1 (es) * 2009-03-24 2012-02-13 INSTITUT CATALÀ D`ONCOLOGIA (Titular al 50%) Combinación de adenovirus oncolítico y un bloqueador de canal de calcio y su uso para el tratamiento del cáncer.
FI123955B (en) 2011-11-25 2014-01-15 Oncos Therapeutics Ltd Oncolytic adenovirus
CN114317461A (zh) * 2013-11-22 2022-04-12 德那翠丝有限公司 表达免疫细胞刺激受体激动剂的腺病毒
CN108289920A (zh) * 2015-10-12 2018-07-17 汉阳大学校产学协力团 用于基因转移和基因治疗的腺病毒复合物
HUE053236T2 (hu) * 2016-09-12 2021-06-28 Targovax Oy Adenovírus és ellenõrzõpont-gátlók kombinálása rák kezelésére
WO2020047345A1 (en) 2018-08-31 2020-03-05 Yale University Compositions and methods of using cell-penetrating antibodies in combination with immune checkpoint modulators
CN110684743A (zh) * 2019-07-16 2020-01-14 伍泽堂 特异性杀伤肿瘤细胞的病毒和肿瘤治疗药物
CN114423860A (zh) 2019-08-05 2022-04-29 迈索布拉斯特国际有限公司 包含病毒载体的细胞组合物及治疗方法
AU2021324483A1 (en) 2020-08-10 2023-04-13 Mesoblast International Sàrl Cellular compositions and methods of treatment
WO2024081736A2 (en) 2022-10-11 2024-04-18 Yale University Compositions and methods of using cell-penetrating antibodies

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6936450B2 (en) * 2000-04-12 2005-08-30 Compugen Ltd. Variants of protein kinases
US20060147420A1 (en) * 2004-03-10 2006-07-06 Juan Fueyo Oncolytic adenovirus armed with therapeutic genes
US20060188990A1 (en) * 2002-04-19 2006-08-24 Schering Aktiengesellschaft Novel prostate tumor-specific promoter
US20070292396A1 (en) * 2006-03-02 2007-12-20 Juan Fueyo Combination therapy with oncolytic adenovirus
US20090312401A1 (en) * 2006-04-28 2009-12-17 Osvaldo Luis Podhajcer Isolated dna fragment of the sparc human promoter and its use for driving the expression of an heterologous gene in tumor cells
US20100113569A1 (en) * 2006-04-28 2010-05-06 Inis Biotech Llc Isolated dna fragment of the human a33 promoter and its use to control the expression of a heterologous gene in tumor cells

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001049868A1 (en) * 1999-12-31 2001-07-12 Korea Research Institute Of Bioscience And Biotechnology Cancer cell-specific gene expression system
US7109029B2 (en) * 2001-02-23 2006-09-19 Cell Genesys, Inc. Vector constructs
WO2003070958A1 (en) * 2002-02-19 2003-08-28 The Hospital For Sick Children Retroviral gene therapy vectors including insulator elements to provide high levels of gene expression
ES2304281B1 (es) * 2006-02-01 2009-08-12 Dnatrix Inc. Adenovirus oncoliticos para el tratamiento del cancer.

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6936450B2 (en) * 2000-04-12 2005-08-30 Compugen Ltd. Variants of protein kinases
US20060188990A1 (en) * 2002-04-19 2006-08-24 Schering Aktiengesellschaft Novel prostate tumor-specific promoter
US20060147420A1 (en) * 2004-03-10 2006-07-06 Juan Fueyo Oncolytic adenovirus armed with therapeutic genes
US20070292396A1 (en) * 2006-03-02 2007-12-20 Juan Fueyo Combination therapy with oncolytic adenovirus
US20090312401A1 (en) * 2006-04-28 2009-12-17 Osvaldo Luis Podhajcer Isolated dna fragment of the sparc human promoter and its use for driving the expression of an heterologous gene in tumor cells
US20100113569A1 (en) * 2006-04-28 2010-05-06 Inis Biotech Llc Isolated dna fragment of the human a33 promoter and its use to control the expression of a heterologous gene in tumor cells

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Duque et al, Direct Comparison of the Insulating Properties of Two Genetic Elements in an Adenoviral Vector Containing Two Different Expression Cassettes, HUMAN GENE THERAPY 15:995-1002 (October 2004) *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10391183B2 (en) * 1999-05-12 2019-08-27 The Uab Research Foundation Infectivity-enhanced conditionally-replicative adenovirus and uses thereof
US20140227226A1 (en) * 1999-05-12 2014-08-14 The Uab Research Foundation Infectivity-enhanced conditionally-replicative adenovirus and uses thereof
US20150374766A1 (en) * 2013-03-14 2015-12-31 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US11077156B2 (en) * 2013-03-14 2021-08-03 Salk Institute For Biological Studies Oncolytic adenovirus compositions
US11439678B2 (en) 2013-10-25 2022-09-13 Psioxus Therapeutics Limited Oncolytic adenoviruses armed with heterologous genes
US11938159B2 (en) 2013-10-25 2024-03-26 Akamis Bio Limited Oncolytic adenoviruses armed with heterologous genes
US10849945B2 (en) 2015-04-30 2020-12-01 Psioxus Therapeutics Limited Oncolytic adenovirus encoding a B7 protein or active fragment
US11000559B2 (en) 2015-04-30 2021-05-11 Psioxus Therapeutics Limited Oncolytic adenovirus encoding a B7 protein
US11155622B2 (en) 2015-12-17 2021-10-26 Psioxus Therapeutics Limited Virus encoding an anti-TCR-complex antibody or fragment
US11970536B2 (en) 2015-12-17 2024-04-30 Akamis Bio Limited Group B adenovirus encoding an anti-TCR-complex antibody or fragment
US11401529B2 (en) 2016-02-23 2022-08-02 Salk Institute For Biological Studies Exogenous gene expression in recombinant adenovirus for minimal impact on viral kinetics
US11130968B2 (en) 2016-02-23 2021-09-28 Salk Institute For Biological Studies High throughput assay for measuring adenovirus replication kinetics
US11813337B2 (en) 2016-12-12 2023-11-14 Salk Institute For Biological Studies Tumor-targeting synthetic adenoviruses and uses thereof
US11840702B2 (en) 2017-08-28 2023-12-12 Akamis Bio Limited Adenovirus armed with bispecific T cell activator

Also Published As

Publication number Publication date
ES2304281A1 (es) 2008-10-01
US20150071881A1 (en) 2015-03-12
US20160354420A1 (en) 2016-12-08
AU2007211434A1 (en) 2007-08-09
US10016470B2 (en) 2018-07-10
ES2304281B1 (es) 2009-08-12
CN101484583A (zh) 2009-07-15
US20190183946A1 (en) 2019-06-20
WO2007088229A1 (es) 2007-08-09
CA2640528C (en) 2015-02-24
CA2640528A1 (en) 2007-08-09
EP1990418B1 (en) 2012-08-29
JP5075839B2 (ja) 2012-11-21
EP1990418A1 (en) 2008-11-12
EP1990418A4 (en) 2011-02-23
JP2009525036A (ja) 2009-07-09

Similar Documents

Publication Publication Date Title
US10016470B2 (en) Oncolytic adenoviruses for cancer treatment
EP1180932B1 (en) Infectivity-enhanced conditionally-replicative adenovirus and uses thereof
US10391183B2 (en) Infectivity-enhanced conditionally-replicative adenovirus and uses thereof
JP5746823B2 (ja) E3−19kタンパク質の小胞体保持ドメインに突然変異を有するアデノウイルス及び癌治療におけるその用途
Wu et al. Cancer gene therapy by adenovirus-mediated gene transfer
US20050036989A1 (en) Subgroup B adenoviral vectors for treating disease
US9175309B2 (en) Recombinant adenovirus with enhanced therapeutic effect and pharmaceutical composition comprising said recombinant adenovirus
RU2814581C1 (ru) Генетический вектор Ad6/3-hTERT-GMCSF, содержащий геномные последовательности рекомбинантного аденовируса 6 серотипа, промотор теломеразы человека, ген гранулоцитарно-макрофагального колониестимулирующего фактора человека, а также ген белка файбер со встройкой домена fiber knob аденовируса 3 серотипа, обладающий повышенной трансдукцией в опухолевые клетки
CA2627638A1 (en) Conditionally replicating viruses and methods for cancer virotherapy
Alemany Conditionally replicating adenoviruses for cancer treatment
Coughlan Transductional retargeting of human adenovirus type 5 to ανβ6 integrin for cancer gene therapy
Särkioja Adenoviral gene therapy for non-small cell lung cancer
Tanaka et al. Recent developments in cancer gene therapy with adenovirus vectors

Legal Events

Date Code Title Description
AS Assignment

Owner name: DNATRIX, INC., TEXAS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INSTITUT CATALA D'ONCOLOGIA;REEL/FRAME:022803/0798

Effective date: 20070301

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION