WO2004005498A1 - Chimeric gb virus b (gbv-b) - Google Patents

Chimeric gb virus b (gbv-b) Download PDF

Info

Publication number
WO2004005498A1
WO2004005498A1 PCT/US2003/021002 US0321002W WO2004005498A1 WO 2004005498 A1 WO2004005498 A1 WO 2004005498A1 US 0321002 W US0321002 W US 0321002W WO 2004005498 A1 WO2004005498 A1 WO 2004005498A1
Authority
WO
WIPO (PCT)
Prior art keywords
ntr
hcv
gbv
virus
domain
Prior art date
Application number
PCT/US2003/021002
Other languages
French (fr)
Inventor
Annette Martin
David V. Sangar
Stanley M. Lemon
Rene Rijnbrand
Original Assignee
Board Of Regents, The University Of Texas System
Institut Pasteur
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 Board Of Regents, The University Of Texas System, Institut Pasteur filed Critical Board Of Regents, The University Of Texas System
Priority to AU2003248821A priority Critical patent/AU2003248821A1/en
Priority to US10/520,153 priority patent/US7473772B2/en
Priority to EP03763178A priority patent/EP1539938A4/en
Publication of WO2004005498A1 publication Critical patent/WO2004005498A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0337Animal models for infectious diseases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5256Virus expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24111Flavivirus, e.g. yellow fever virus, dengue, JEV
    • C12N2770/24122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24211Hepacivirus, e.g. hepatitis C virus, hepatitis G virus
    • C12N2770/24221Viruses 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24211Hepacivirus, e.g. hepatitis C virus, hepatitis G virus
    • C12N2770/24222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24211Hepacivirus, e.g. hepatitis C virus, hepatitis G virus
    • C12N2770/24241Use of virus, viral particle or viral elements as a vector
    • C12N2770/24243Use 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
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/24011Flaviviridae
    • C12N2770/24211Hepacivirus, e.g. hepatitis C virus, hepatitis G virus
    • C12N2770/24261Methods of inactivation or attenuation
    • 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
    • C12N2840/00Vectors comprising a special translation-regulating system
    • C12N2840/20Vectors comprising a special translation-regulating system translation of more than one cistron
    • C12N2840/203Vectors comprising a special translation-regulating system translation of more than one cistron having an IRES

Definitions

  • the present invention relates generally to the fields of biochemistry, molecular biology, and virology. More particularly, it relates to the identification of 259 nucleotides of previously unrecognized sequence located at the 3' end of the GB virus B (GBV-B) genome.
  • GBV-B GB virus B
  • Chronic hepatitis C is a major threat to the public health. Serologic surveys suggest that as many as 3.9 million Americans are chronically infected with the responsible virus, hepatitis C virus (HCV) (Alter, 1997). These individuals are at increased risk of developing progressive hepatic fibrosis leading to cirrhosis and loss of hepatocellular function, as well as hepatocellular carcinoma. The course of chronic hepatitis C is typically lengthy, often extending over decades, with insidious clinical progression usually occurring in the absence of symptoms. Nonetheless, liver disease due to HCV results in the death of 8,000-10,000 Americans annually, and chronic hepatitis C is the most common cause of liver transplantation within the U.S. Therefore, HCV is a major public health problem. However, therapy for chronic hepatitis
  • interferon- ⁇ is approved for treatment of chronic hepatitis C (Consensus Development Panel, 1997).
  • the benefit of interferon- ⁇ results primarily from its antiviral properties and its ability to inhibit production of virus by infected hepatocytes (Neumann et al, 1998). Nonetheless, even under optimal therapeutic regimens, the majority of patients with chronic hepatitis C fail to eliminate the virus or resolve their liver disease. Treatment failures are especially common in persons infected with genotype 1 HCV, unfortunately the most prevalent genotype in the U.S. Thus, there is an urgent need to better understand the virus and develop better treatment. Unfortunately, technical difficulties in working with HCV have made it necessary to use infectious surrogate viruses in efforts to develop treatments and vaccines for HCV.
  • GBV-B is a hepatotropic flavivirus that has a unique phylogenetic relationship to human HCV and strong potential to serve as a surrogate virus in drug discovery efforts related to hepatitis C antiviral drug development.
  • GBV-B causes acute hepatitis in experimentally infected tamarins (Simons et al, 1995; Schlauder et al, 1995; Karayiannis et al, 1989) and can serve as a surrogate virus for HCV in drug discovery efforts (due to technical difficulties in working with HCV).
  • GBV-B virus is much closer in sequence and biological properties than the above- described models. It will be easier to make biologically relevant chimeras between HCV and
  • GBV-B than by using more distantly related viruses.
  • GBV-B is hepatotropic (as is HCV), whereas the viruses used in these competing technologies are not.
  • HCV hepatotropic
  • an infectious clone of GBV-B would be useful to those working on HCV treatment and prevention.
  • GBV-B as a surrogate or model for HCV has not been possible in the past, because no infectious molecular clone of GBV-B virus genome could be prepared. It is now known that this obstacle was encountered because the GBV-B genome was believed to be 259 nucleotides shorter than its actual length (Muerhoff et al, 1995; Simons et al, 1995). Others, previous to the inventors, had failed to realize that the 3' sequence of GBV-B was missing from the prior sequences. Without this 3' sequence, it is not possible to prepare an infectious GBV-B molecular clone.
  • an infectious molecular clone of GBV-B would be very useful for the development of HCV preventative and therapeutic treatments.
  • the construction of an infectious molecular clone of this virus will require the newly determined 3' sequence to be included in order for the clone to be viable.
  • the inventors have elucidated the previously unrecognized 3' terminal sequence of GBV-B (SEQ ID NO:l). This sequence has been reproducibly recovered from tamarin serum containing GBV-B RNA, in RT-PCR protocols using several different primer sets, and as a fusion with previously reported 5' GBV-B sequences.
  • the invention has utility in that the inclusion of the sequence will be necessary for construction of an infectious molecular GBV-B clone.
  • Such clones clearly have the potential to be constructed as chimeras including relevant hepatitis C virus sequences in lieu of the homologous GBV-B sequence, providing unique tools for drug discovery efforts.
  • GBV-B can be used as a model for HCV, and the GBV-B genome can be used as the acceptor molecule in the construction of chimeric viral RNAs containing sequences of both HCV and GBV-B.
  • HCV activities e.g., proteinase
  • all this work is dependent upon construction of an infectious clone of GBV-B, which is itself dependent on the incorporation of the correct 3' terminal nucleotide sequence within this clone.
  • GBV-B has unique advantages over HCV in terms of its ability to replicate and cause liver disease in tamarins, which present fewer restrictions to research than chimpanzees, the only nonhuman primate species known to be permissive for HCV.
  • An infectious molecular clone of GBV-B is expected to have utility in liver-specific gene expression or in gene therapy. This application might be enhanced by the inclusion of HCV genomic sequence in the form of a GBV-B/HCV chimera. Further, an infectious GBV-B/HCV chimera expressing HCV envelope proteins can have utility as a vaccine immunogen for hepatitis
  • RNA transcribed from this cDNA copy of the genome would be infectious when inoculated into the liver of a GBV-B permissive tamarin, giving rise to rescued GBV-B virus particles.
  • a chimeric molecule would then be constructed from this infectious GBV-B clone in which the HCV NS3 proteinase or proteinase/helicase sequence (or other relevant HCV sequences of interest in drug discovery efforts) would be placed in frame in lieu of the homologous GBV-B sequence, and this chimeric cDNA would be used to generate infectious GBV-B/HCV chimeric viruses by intrahepatic inoculation of synthetic RNA in tamarins.
  • Published studies indicate that the GBV-B and HCV proteinases have closely related substrate recognition and cleavage properties, making such chimeras highly likely to be viable. These newly generated chimeric GBV-B/HCV viruses could be used in preclinical testing of candidate HCV NS3 proteinase inliibitors.
  • the present invention encompasses an isolated polynucleotide encoding a 3' sequence of the GBV-B genome.
  • the polynucleotide may include the sequence identified as SEQ ID NO: 1. It is contemplated that the polynucleotide may be a DNA molecule or it can be an RNA molecule. It is further contemplated that expression constructs may contain a polynucleotide that has a stretch of contiguous nucleotides from SEQ ID NO:l and/or SEQ ID NO:2, for example, lengths of 50, 100, 150, 250, 500, 1000, 5000, as well as the entire length of SEQ ID NO:l or 2, are considered appropriate.
  • polynucleotides may also be contained in other constructs of the invention or be used in the methods of the invention.
  • Polynucleotides employing sequences from SEQ ID NO:l may alternatively contain sequences from SEQ LD NO:2 in the constructs and methods of the present invention. Specific mutations discussed in the Examples and/or figures are included as embodiments of the invention.
  • the invention is also understood as covering a viral expression construct that includes a polynucleotide encoding a 3' sequence of the GBV-B genome.
  • This expression construct is further understood to contain the sequence identified as SEQ ID NO:l.
  • the present invention contemplates the expression construct as a plasmid or as a virus.
  • the expression construct can express GBV-B sequences; alternatively it may express sequences from a chimeric GBV-B HCV virus.
  • the identification and isolation of a 3' sequence of GBV-B additionally provides a method of producing a virus, particularly a full-length virus, by introducing into a host cell a viral expression construct containing a polynucleotide encoding a 3' sequence of GBV-B and by culturing the host cell under conditions permitting production of a virus from the construct.
  • This method can be practiced using a prokaryotic cell as a host cell, or by using a eukaryotic cell as a host cell.
  • the eukaryotic cell can be located within an animal.
  • a method of producing virus according to the claimed invention can also be employed using a polynucleotide that contains synthetic RNA and or synthetic DNA.
  • a step can be added to the method by also isolating any virus produced from the host cell.
  • the virus can then be purified to homogeneity.
  • the present invention encompasses an oligonucleotide between about 10 and about 259 consecutive bases of SEQ ID NO:l. This oligonucleotide is contemplated to be about 15 bases in length, about 20 bases in length, about 25 bases in length, about 30 bases in length, about 35 bases in length, about 50 bases in length, about 100 bases in length, about 150 bases in length, about 200 bases in length, or about 259 bases in length.
  • Additional examples of the claimed invention include a method for identifying a compound active against a viral infection by providing a virus expressed from a viral construct containing a 3' sequence of a GBV-B virus, by contacting the virus with a candidate substance; and by comparing the infectious ability of the virus in the presence of the candidate substance with the infectious ability of the virus in a similar system in the absence of the candidate substance. It is contemplated that the invention can be practiced using GBV-B virus or a GBV- B HCV chimera. Infectious ability of the virus is comparable to infectivity of the virus. Infectivity can be evaluated by evaluating or assessing the ability of a host cell to become infected by the virus or by the ability of the virus to replicate in the cell. However, it may also be evaluated by virus production, virus infectivity, viral load, or phenotype of the cell such as signs of CPE.
  • the present invention can also be understood to provide a compound active against a viral infection identified by providing a virus expressed from a viral construct containing a 3' sequence of a GBV-B virus; contacting the virus with a candidate substance; and comparing the infectious ability of the virus in the presence of the candidate substance with the infectious ability of the virus in a similar system in the absence of the candidate substance.
  • an active compound is identified using a GBV-B virus, while in other embodiments an active compound is identified using a GBV-B/HCV chimera.
  • Compounds considered active against viral infection would include, but are not limited to, those compounds that inhibit viral infection or that expedite clearance of the virus.
  • a GBV-B polynucleotide may encode a GBV-
  • the B/HCV chimera that includes at least part of a 5' NTR sequence derived from a HCV 5' NTR.
  • the 5' NTR may comprise at least one domain, i.e., domain I, JJ, m and or IV derived from the HCV in some embodiments.
  • the GVB-B/HCV chimera may include at least domain III of the 5' NTR derived from the 5 'NTR of HCV.
  • the infectious GBV-B clone may comprise domain III of the 5' NTR of HCV, which may or may not include one or more structural or non-structural genes of HCV also inco ⁇ orated into the chimeric virus.
  • the portions of the 5' NTR of the GVB-B/HCV chimeras will generally be replaced by analogous sequences from the 5' NTR of HCV. It will be understood that the portions or parts of the 5' NTR of GBV-B that may be replaced include all or part of domain I (including sub-region la and lb of GBV-B), domain ⁇ , domain HI, domain IV, or any combination thereof. Any combination of 5' NTR domains of GBV-B may be replaced with an analogous region of HCV. In certain embodiments, the replacement of a GBV-B region may be accompanied by the deletion of the 5' NTR GBV-B domain lb region. In addition, any one, two, or three of the 5' NTR domains of GBV-B may be replaced in any combination with analogous sequences from HCV.
  • a polynucleotide encoding a GBV-B/HCV chimera including a 5' NTR domain III sequence derived from a HCV 5' NTR may be propagated in vivo, in particular, in the liver of an appropriate host.
  • Various other embodiments may include isolated polynucleotides comprising a chimeric GBV-B genome, wherein at least part, but not all of a 5' NTR sequence is derived from a HCV 5' NTR.
  • the polynucleotides may be synthetic RNA, RNA, DNA or the like.
  • Some embodiments include one or more virus, one or more hepatotropic virus, and/or one or more viral expression constructs comprising a chimeric GBV-B polynucleotides including at least a part of the 5' NTR sequence is derived from a HCV 5' NTR.
  • Methods of producing a chimeric GBV-B virus encoding at least part of a 5' NTR sequence derived from a HCV 5' NTR sequence comprising introducing into a host cell a viral expression construct comprising a chimeric GBV-B polynucleotide encoding at least part of a 5' NTR sequence derived from a HCV 5' NTR sequence and culturing said host cell under conditions permitting production of a virus from said construct are contemplated.
  • the method may use a host cell that is a eukaryotic cell and the host cell may be in an animal.
  • the method may further include the step of isolating virus from said host cell and in particular purify the virus to homogeneity.
  • methods for identifying a compound active against a viral infection comprising are contemplated.
  • the methods may include providing a virus expressed from a viral construct comprising at least part of a 5' NTR derived from a HCV 5' NTR, as described herein; contacting said virus with a candidate substance; and comparing the infectious ability of the virus in the presence of said candidate substance with the infectious ability of the virus in a similar system in the absence of said candidate substance.
  • Each of the embodiments may use or include any of the 5' NTR chimeras described herein.
  • Other embodiments of the invention may include a compound active against a viral infection identified according to the method described above.
  • additional methods include providing or administering to a subject or patient in need of a compound active against viral infection the compound active against a viral infection, particularly a flavivirus such as HCV infection.
  • the patient may have been diagnosed with HCV infection or be at risk for HCV infection, or another flavivirus infection.
  • a subject or patient may be administered a vaccine that immunizes the subject or patient with respect to a flavivirus infection, such as HCV.
  • the invention includes such methods.
  • any limitation discussed with respect to one embodiment of the invention may apply to any other embodiment of the invention.
  • any composition of the invention may be used in any method of the invention, and any method of the invention may be used to produce or to utilize any composition of the invention.
  • FIG. 1 Predicted secondary RNA structure at the 3' end of the novel 3' GBV-B sequence.
  • the structure shown is that predicted for the 3' terminal 108 nucleotides of the GBV-B genomic RNA by the MFOLD 3.0 computer program of Zucker and Turner, and has an initial free energy of -48 kcal/mole.
  • the predicted structure contains 3 stem-loops, numbered from the 3' end of the genome and labeled SL-1, SL-2, and SL-3.
  • the structure of SL-1 is highly probable, given its terminal position within the genome. Alternative foldings of SL-2 and SL-3 are possible.
  • FIG. 2 Illustration of the organization of exemplary chimeric cDNAs containing HCV 5' nontranslated (5' NTR) RNA sequences within the background of a modified (MM restriction- site containing) GBV-B genetic background.
  • FIG. 3 A and 3B In vitro translation of RLuc reporter transcripts (shown schematically in
  • FIG. 3 A in which the initiation of translation of RLuc is dependent upon the upstream viral or chimeric IRES.
  • FIG. 3B, top panel shows the SDS-PAGE gel on which the products of translation were separated.
  • FIG. 4 Shows the translational activity of RLuc reporter transcripts (normalized to FLuc control transcript) in examples of transfected primary tamarin hepatocytes.
  • FIG. 5. Shows the replication of an exemplary i ⁇ HC chimeric RNA following intrahepatic inoculation of synthetic RNA in a GBV-B naive tamarin (S. mystax).
  • FIG. 6. Shows the replication of an exemplary UI chime ⁇ c virus following intravenous inoculation of a GBV-B na ⁇ ve tamarin (S. mystax) with virus present in the serum of T16444 14 weeks after it had been inoculated with synthetic RNA.
  • FIG. 7 Shows the sequence differences between chimeric viral RNAs recovered from animals T16444 and T16451, and the parental synthetic m HC RNA.
  • FIG. 8 Replication of the GB/LU HC chimeric virus in primary cultures of tamarin hepatocytes. Viral titers were determined both in infected-cell extracts ("cytosol") and in culture supernatants ("secreted") by a quantitative real-time RT-PCR assay.
  • FIG. 9 Replication of the chimeric RNA following intrahepatic inoculation of
  • GBV-B viremia was determined in a quantitative real-time
  • FIG. 10 Mutations identified in the virus isolated at week 14 p.i. in an animal (T 16444) inoculated with the GB/IJJ chimeric RNA.
  • FIG. 11 Replication of the GB/IJI chime ⁇ c virus following intravenous inoculation m a na ⁇ ve tamarin.
  • FIG. 12 Schematic representation of the RNA transcripts of mutated cDNAs derived from the parental GB/]U HC construct and their in vivo replication capacity following intrahepatic inoculation into GBV-B na ⁇ ve tamarins.
  • FIG. 13A and 13B Replication of the GB/HI HC -m3 and GB/r ⁇ HC -m4 chimeric RNAs following intrahepatic inoculation of RNA in individual na ⁇ ve tamarins. See Fig. 2 for details relative to monitoring of the infection.
  • FIG. 14A and 14B Predicted models of the secondary structures of the 59NTRs of GBV-B (FIG. 14A) and HCV (FIG. 14B).
  • FIG. 14A Structure proposed for the GBV-B 59NTR by Honda et al. (1996 and 1999), each of which is inco ⁇ orated herein by reference.
  • Major predicted structural domains are labeled I to TV, while individual stem-loops are labeled la and are analogous to similarly labeled structures in the HCV 59NTR.
  • Base-pair interactions involving the loop sequence of stem-loop JJIf that are predicted to result in a putative RNA pseudoknot are drawn as solid lines.
  • GBV-B genome structure is very similar to hepatitis C and these viruses share approximately 25% nucleotide identity (Simons et al, 1995; Muerhoff et al, 1995). As indicated above, this makes GBV-B more closely related to HCV than any other known virus.
  • GBV-B genomic RNA is about 9.5 kb in length (Muerhoff et al, 1995) with a structured 5' noncoding region that contains an IRES that shares many structural features with the HCV IRES (Honda et al, 1996; Rijnbrand et al, 1999, each of which is inco ⁇ orated herein by reference). As in HCV, this IRES drives the cap-independent translation of a long open reading frame.
  • the polyprotein expressed from this reading frame appears to be organized identically to that of HCV, and processed to generate proteins with functions similar to those of HCV (Muerhoff et al, 1995).
  • the major serine proteinases of these viruses (NS3) have been shown to have similar cleavage specificities (Scarselli et al, 1997).
  • the genomic RNA of GBV-B has a poly(U) tract located near its 3" terminus (Simons et al, 1995; Muerhoff et al, 1995).
  • unreported sequences located at the extreme 3' end of the genome have been identified.
  • the present invention provides a nucleic acid sequence encoding a 3' sequence of the
  • a "3' sequence of the GBV-B genome” may contain a variety of different bases and yet still be functionally indistinguishable from the sequences disclosed herein. Such functionally indistinguishable sequences are likely to maintain the basic structure depicted in FIGs. 1 and 2, which may be used to guide the prediction of viable nucleotide substitutions.
  • a 3' sequence of the GBV-B genome disclosed in SEQ ID NO:l is one aspect of the present invention.
  • Nucleic acids according to the present invention may encode the 3' sequence of the GBV-B genome set forth in SEQ ID NO:l, the entire GBV-B genome, or any other fragment of a 3' sequence of the GBV-B genome set forth herein.
  • the nucleic acid may be derived from genomic RNA as cDNA, i.e., cloned directly from the genome of GBV-B.
  • cDNA may also be assembled from synthetic oligonucleotide segments.
  • a 3' sequence of the GBV-B genome may be represented by natural variants that have slightly different nucleic acid sequences but, nonetheless, maintain the same general structure (see FIGs. 1 and 2) and perform the same function in RNA replication.
  • a nucleic acid encoding a 3' sequence of the GBV-B genome refers to a nucleic acid molecule that may be isolated free of total viral nucleic acid.
  • the invention concerns nucleic acid sequences essentially as set forth in SEQ ID NO:l, SEQ LO NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ LD NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ LD NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:l l, and SEQ ID NO:12.
  • SEQ ID NO:l means that the nucleic acid sequence substantially corresponds to a portion of SEQ ID NO:l. It is contemplated that the techniques and methods described in this disclosure may apply to any of the sequences contained herein, including SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ LD NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:ll, and SEQ ID NO:12.
  • sequences that have at least about 50%, usually at least about 60%, more usually about 70%, most usually about 80%, preferably at least about 90% and most preferably about 95% of nucleotides that are identical to the nucleotides of SEQ ID NO:l will be sequences that are "as set forth in SEQ ID NO:l.” Sequences that are essentially the same as those set forth in SEQ ID NO:l may also be functionally defined as sequences that are capable of hybridizing to a nucleic acid segment containing the complement of SEQ ID NO:l under standard conditions.
  • nucleic acid segments and polynucleotides of the present invention include those encoding biologically functional equivalent 3' sequences of the GBV-B genome. Changes designed by man may be introduced through the application of site-directed mutagenesis techniques or maybe introduced randomly and screened later for the desired function, as described below.
  • the present invention concerns cDNA segments reverse transcribed from GBV-B genomic RNA (referred to as "DNA").
  • DNA GBV-B genomic RNA
  • polynucleotide refers to an RNA or DNA molecule that may be isolated free of other RNA or DNA of a particular species. "Isolated substantially away from other coding sequences” means that the 3' sequence of the GBV-B genome forms the significant part of the RNA or DNA segment and that the segment does not contain large portions of naturally-occurring coding RNA or DNA, such as large fragments or other functional genes or cDNA noncoding regions. Of course, this refers to the polynucleotide as originally isolated, and does not exclude genes or coding regions later added to the it by the hand of man.
  • the invention concerns isolated DNA segments (cDNA segments reverse transcribed from GVB-B genomic RNA) and recombinant vectors that include within their sequence a nucleic acid sequence essentially as set forth in SEQ LD NO:l.
  • the term "essentially as set forth in SEQ ID NO:l" is used in the same sense as described above.
  • nucleic acid sequences may include additional residues, such as additional 5' or 3' sequences, and still be essentially as set forth in one of the sequences disclosed herein, so long as the sequence meets the criteria set forth above, including the maintenance of biological activity.
  • the addition of terminal sequences particularly applies to nucleic acid sequences that may, for example, include additional various non-coding sequences flanking either of the 5' or 3' portions of the coding region, which are known to occur within viral genomes.
  • Sequences that are essentially the same as those set forth in SEQ ID NO:l also may be functionally defined as sequences that are capable of hybridizing to a nucleic acid segment containing the complement of SEQ ID NO:l under relatively stringent conditions. Suitable relatively stringent hybridization conditions will be well known to those of skill in the art.
  • nucleic acid segments of the present invention may be combined with other RNA or DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol.
  • nucleic acid fragments may be prepared that include a short contiguous stretch identical to or complementary to SEQ ID NO:l, such as about 15-24 or about 25-34 nucleotides and that are up to about 259 nucleotides being prefe ⁇ ed in certain cases.
  • Other stretches of contiguous sequence that may be identical or complementary to any of the sequences disclosed herein, including the SEQ ID NOS. include the following ranges of nucleotides: 50- 9,399, 100-9,000, 150-8,000, 200-7,000, 250-6,000, 300-5,000, 350-4,000, 400-3,000, 450- 2,000, 500-1000.
  • RNA and DNA segments with total lengths of about 1,000, about 500, about 200, about 100 and about 50 base pairs in length (including all intermediate lengths) are also contemplated to be useful.
  • one or more nucleic acid constructs may be prepared that include a contiguous stretch of nucleotides identical to or complementary to SEQ ID NO:l, SEQ ID NO:2, SEQ LD NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO:ll, or SEQ ID NO:12.
  • Such a stretch of nucleotides, or a nucleic acid construct may be about, or at least about, 3, about 4, about 5, about 6, about 7, about 8solv about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39 about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, about 200, about 210, about 220, about 230, about 240, about 250, about 260, about 270, about 280, about 290, about
  • intermediate lengths in these contexts means any length between the quoted ranges, such as 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, etc.; 100, 101, 102, 103, etc.; 150, 151, 152, 153, etc.; including all integers through the 200-500; 500-1,000; 1,000-2,000; ranges, up to and including sequences of about 1,001, 1,250, 1,500, and the like.
  • GBV-B polynucleotides include chimeric GBV-B poynucleotides.
  • Chimeric GBV-B polynucleotides may include GBV-B/HCV chimeras.
  • GBV- B/HCV chimeras include, but are not limited to GBV-B polynucleotides in which portions of the GBV-B virus have been replaced with or mutated to resemble analogous sequences from the HCV virus.
  • a GVB-B chimera may comprise all or part of the 5 'NTR region of a HCV virus.
  • the polynucleotide may comprise domain I, II. Ill and/or IV of the 5' NTR derived from a HCV 5 'NTR.
  • the polynucleotide may have the 5' NTR domain lb of GBV-B is deleted.
  • the polynucleotide may comprise domain II and domain III of the 5' NTR derived from a HCV 5 'NTR.
  • the polynucleotide may comprise domain II and domain IN of the 5' ⁇ TR derived from a HCV 5 ' ⁇ TR.
  • the polynucleotide may compirse domain III and domain IV of the 5' ⁇ TR derived from a HCV 5 ' ⁇ TR.
  • the polynucleotide may comprise domain II, domain III and domain IV of the 5' ⁇ TR derived from a HCV 5 ' ⁇ TR.
  • Domain lb of GBV-B may or may not be deleted form the polynucleotide.
  • the polynucleotide may be D ⁇ A or R ⁇ A.
  • the polynucleotide may further comprising at least part of a structural and/or nonstructural protein coding region of derived from HCV. It will be understood that the term "derived" indicates the origin of the sequence. Thus, the recited domains can be obtained from HCV.
  • a HCV/GBV-B chimera will comprise a virus where one or more portions of the HCV virus have been replaced with or mutated to resemble the analogous GBV-B portions of the GBV-B virus.
  • the various probes and primers designed around the disclosed nucleotide sequences of the present invention may be of any length.
  • an algorithm defining all primers can be proposed: n to n + y where n is an integer from 1 to the last number of the sequence and y is the length of the primer minus one, where n + y does not exceed the last number of the sequence.
  • the probes co ⁇ espond to bases 1 to 20, 2 to 21, 3 to 22 ... and so on.
  • the probes co ⁇ espond to bases 1 to 30, 2 to 31, 3 to 32 ... and so on.
  • the probes co ⁇ espond to bases 1 to 35, 2 to 36, 3 to 37 ... and so on.
  • RNA and DNA segments that are complementary, or essentially complementary, to the sequence set forth in SEQ ID NO:l.
  • Nucleic acid sequences that are "complementary” are those that are capable of base-pairing according to the standard Watson-Crick complementary rules.
  • the term “complementary sequences” means nucleic acid sequences that are substantially complementary, as may be assessed by the same nucleotide comparison set forth above, or as defined as being capable of hybridizing to the nucleic acid segment of SEQ ID NO. 1 under relatively stringent conditions such as those described herein. Such sequences may encode the entire 3' sequence of the GBV-B genome or functional or non-functional fragments thereof.
  • the hybridizing segments may be shorter oligonucleotides. Sequences of 17 bases long should occur only once in the human genome and, therefore, suffice to specify a unique target sequence. Although shorter oligomers are easier to make and increase in vivo accessibility, numerous other factors are involved in determining the specificity of hybridization. Both binding affinity and sequence specificity of an oligonucleotide to its complementary target increases with increasing length.
  • exemplary oligonucleotides of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225 or more base pairs will be used, although others are contemplated. Longer polynucleotides encoding 250, 500, 1000, 1212, 1500, 2000, 2500, 3000 or 3431 bases and longer are contemplated as well. Such oligonucleotides will find use, for example, as probes in Southern and Northern blots and as primers in amplification reactions.
  • hybridization conditions will be well known to those of skill in the art. In certain applications, for example, substitution of amino acids by site-directed mutagenesis, it is appreciated that lower stringency conditions are required. Under these conditions, hybridization may occur even though the sequences of probe and target strand are not perfectly complementary but are mismatched at one or more positions. Conditions may be rendered less stringent by increasing salt concentration and decreasing temperature. For example, a medium stringency condition could be provided by about 0.1 to 0.25 M NaCl at temperatures of about 37°C to about 55°C, while a low stringency condition could be provided by about 0.15 M to about 0.9 M salt, at temperatures ranging from about 20°C to about 55°C. Thus, hybridization conditions can be readily manipulated and thus will generally be a method of choice depending on the desired results.
  • hybridization may be achieved under conditions of, for example, 50 mM Tris-HCl (pH 8.3), 75 mM KCl, 3 mM MgCl 2 , 10 mM dithiothreitol, at temperatures between approximately 20°C to about 37°C.
  • Other hybridization conditions utilized could include approximately 10 mM Tris-HCl (pH 8.3), 50 mM KCl, 1.5 mM MgCl 2 , at temperatures ranging from approximately 40°C to about 72°C.
  • Formamide and SDS also may be used to alter the hybridization conditions.
  • One method of using probes and primers of the present invention is in the search for other viral sequences related to GBV-B or, more particularly, homologs of the GBV-B sequence. By varying the stringency of hybridization, and the region of the probe, different degrees of homology may be discovered.
  • probes and primers of the present invention are in site-directed, or site-specific, mutagenesis.
  • the technique provides a ready ability to prepare and test sequence variants, inco ⁇ orating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into complementary DNA.
  • Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences that encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed.
  • a primer of about 17 to 25 nucleotides in length is prefe ⁇ ed, with about 5 to 10 residues on both sides of the junction of the sequence being altered.
  • the technique typically employs a bacteriophage vector that exists in both a single stranded and double stranded form.
  • Typical vectors useful in site-directed mutagenesis include vectors such as the Ml 3 phage. These phage vectors are commercially available and their use is generally well known to those skilled in the art.
  • Double stranded plasmids are also routinely employed in site directed mutagenesis, which eliminates the step of transferring the gene of interest from a phage to a plasmid.
  • site-directed mutagenesis is performed by first obtaining a single-stranded vector, or melting of two strands of a double stranded vector which includes within its sequence a DNA sequence encoding the desired protein.
  • An oligonucleotide primer bearing the desired mutated sequence is synthetically prepared. This primer is then annealed with the single-stranded DNA preparation, taking into account the degree of mismatch when selecting hybridization conditions, and subjected to DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand.
  • DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment
  • This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected that include recombinant vectors bearing the mutated sequence arrangement.
  • appropriate cells such as E. coli cells
  • clones are selected that include recombinant vectors bearing the mutated sequence arrangement.
  • site-directed mutagenesis techniques that can also be employed for this pvurpose. These include procedures marketed in kit form that are readily available to one of ordinary skill in the art.
  • sequence variants of the selected gene using site-directed mutagenesis is provided as a means of producing potentially useful species and is not meant to be limiting, as there are other ways in which sequence variants of genes may be obtained.
  • recombinant vectors encoding the desired gene may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants.
  • Antisense Constructs hi certain embodiments of the invention, the use of antisense constructs of the 3' sequence of the GBV-B genome is contemplated.
  • Antisense methodology takes advantage of the fact that nucleic acids tend to pair with "complementary" sequences.
  • complementary it is meant that polynucleotides are those which are capable of base-pairing according to the standard Watson-Crick complementarity rules. That is, the larger purines will base pair with the smaller pyrimidines to form combinations of guanine paired with cytosine (G:C) and adenine paired with either thymine (A:T) in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA. Inclusion of less common bases such as inosine, 5-methylcytosine, 6-methyladenine, hypoxanthine and others in hybridizing sequences does not interfere with pairing.
  • Antisense polynucleotides when introduced into a target cell, specifically bind to their target polynucleotide and interfere with transcription, RNA processing, transport, translation and/or stability.
  • Antisense RNA constructs, or DNA encoding such antisense RNAs may be employed to inhibit gene transcription or translation or both within a host cell, either in vitro or in vivo, such as within a host animal, including a human subject.
  • Antisense constructs could be used to block early steps in the replication of GBV-B and related viruses, by annealing to 3' terminal sequences and blocking their role in negative-strand initiation.
  • complementary or “antisense” means polynucleotide sequences that are substantially complementary over their entire length and have very few base mismatches. For example, sequences of 15 bases in length may be termed complementary when they have complementary nucleotides at 13 or 14 positions. Naturally, sequences which are completely complementary will be sequences which are entirely complementary throughout their entire length and have no base mismatches. Other sequences with lower degrees of homology also are contemplated. For example, an antisense construct which has limited regions of high homology but also contains a non-homologous region (e.g., ribozyme; see below) could be designed. These molecules, though having less than 50% homology, would bind to target sequences under appropriate conditions.
  • the present invention utilizes amplification techniques in a number of its embodiments.
  • Nucleic acids used as a template for amplification are isolated from cells contained in the biological sample, according to standard methodologies (Sambrook et al, 1989).
  • the nucleic acid may be genomic DNA or RNA or fractionated or whole cell RNA. Where RNA is used, it may be desired to convert the RNA to a complementary DNA using reverse transcriptase (RT).
  • RT reverse transcriptase
  • the RNA is genomic RNA and is used directly as the template for amplification.
  • genomic RNA is first converted to a complementary DNA sequence (cDNA) and this product is amplified according to protocols described below.
  • primers that selectively hybridize to nucleic acids co ⁇ esponding to GBV-B sequences are contacted with the isolated nucleic acid under conditions that permit selective hybridization.
  • primer as defined herein, is meant to encompass any nucleic acid that is capable of priming the synthesis of a nascent nucleic acid in a template-dependent process.
  • primers are oligonucleotides from ten to twenty base pairs in length, but longer sequences can be employed.
  • Primers may be provided in double-stranded or single-stranded form, although the single-stranded form is prefe ⁇ ed.
  • the nucleic acid:primer complex is contacted with one or more enzymes that facilitate template-dependent nucleic acid synthesis. Multiple rounds of amplification, also refe ⁇ ed to as "cycles,” are conducted until a sufficient amount of amplification product is produced.
  • the amplification product is detected.
  • the detection may be performed by visual means.
  • the detection may involve indirect identification of the product via chemiluminescence, radioactive scintigraphy of inco ⁇ orated radiolabel or fluorescent label or even via a system using electrical or thermal impulse signals.
  • a number of template dependent processes are available to amplify the marker sequences present in a given template sample.
  • One of the best known amplification methods is the polymerase chain reaction (refe ⁇ ed to as PCRTM) which is described in detail in U.S. Patent Nos. 4,683,195, 4,683,202 and 4,800,159, and each inco ⁇ orated herein by reference in entirety.
  • primer sequences are prepared that are complementary to regions on opposite complementary strands of the marker sequence.
  • An excess of deoxynucleoside triphosphates are added to a reaction mixture along with a DNA polymerase, e.g., Taq polymerase. If the marker sequence is present in a sample, the primers will bind to the marker and the polymerase will cause the primers to be extended along the marker sequence by adding on nucleotides. By raising and lowering the temperature of the reaction mixture, the extended primers will dissociate from the marker to form reaction products, excess primers will bind to the marker and to the reaction products and the process is repeated.
  • a DNA polymerase e.g., Taq polymerase
  • a reverse transcriptase PCRTM amplification procedure may be performed in order to quantify the amount of mRNA amplified.
  • Methods of reverse transcribing RNA into cDNA are well known and described in Sambrook et al, 1989.
  • Alternative methods for reverse transcription utilize thermostable, RNA-dependent DNA polymerases. These methods are described in WO 90/07641, filed December 21, 1990, inco ⁇ orated herein by reference.
  • Polymerase chain reaction methodologies are well known in the art.
  • Another method for amplification is the ligase chain reaction ("LCR”), disclosed in EPA
  • Qbeta Replicase described in PCT Application No. PCT/US 87/00880, inco ⁇ orated herein by reference, also may be used as still another amplification method in the present invention.
  • SDA Strand Displacement Amplification
  • RCR Repair Chain Reaction
  • the other two bases can be added as biotinylated derivatives for easy detection.
  • a similar approach is used in SDA.
  • Target specific sequences also can be detected using a cyclic probe reaction (CPR).
  • CPR cyclic probe reaction
  • a probe having 3' and 5' sequences of non-specific DNA and a middle sequence of specific RNA is hybridized to DNA that is present in a sample.
  • the reaction is treated with RNase H, and the products of the probe identified as distinctive products that are released after digestion.
  • the original template is annealed to another cycling probe and the reaction is repeated.
  • primers are used in a PCR-like, template- and enzyme-dependent synthesis.
  • the primers may be modified by labeling with a capture moiety (e.g., biotin) and/or a detector moiety (e.g., enzyme), hi the latter application, an excess of labeled probes are added to a sample.
  • a capture moiety e.g., biotin
  • a detector moiety e.g., enzyme
  • nucleic acid amplification procedures include transcription-based amplification systems (TAS), including nucleic acid sequence based amplification (NASBA) and 3SR Gingeras et al, PCT Application WO 88/10315, inco ⁇ orated herein by reference.
  • TAS transcription-based amplification systems
  • NASBA nucleic acid sequence based amplification
  • 3SR Gingeras et al PCT Application WO 88/10315, inco ⁇ orated herein by reference.
  • Davey et al, EPA No. 329 822 disclose a nucleic acid amplification process involving cyclically synthesizing single-stranded RNA ("ssRNA”), ssDNA, and double-stranded DNA (dsDNA), which may be used in accordance with the present invention.
  • ssRNA single-stranded RNA
  • dsDNA double-stranded DNA
  • Miller et al, PCT Application WO 89/06700 disclose a nucleic acid sequence amplification scheme based on the hybridization of a promoter/primer sequence to a target ssDNA followed by transcription of many RNA copies of the sequence. This scheme is not cyclic, i.e., new templates are not produced from the resultant RNA transcripts.
  • Other amplification methods include "RACE” and “one-sided PCR” (Frohman, 1990 inco ⁇ orated by reference).
  • Methods based on ligation of two (or more) oligonucleotides in the presence of nucleic acid having the sequence of the resulting "di-oligonucleotide", thereby amplifying the di-oligonucleotide also maybe used in the amplification step of the present invention.
  • amplification products are separated by agarose, agarose-acrylamide or polyacrylamide gel electrophoresis using standard methods. See Sambrook et al, 1989.
  • chromatographic techniques may be employed to effect separation. There are many kinds of chromatography that may be used in the present invention: adso ⁇ tion, partition, ion-exchange and molecular sieve, and many specialized techniques for using them including column, paper, thin-layer and gas chromatography. Amplification products must be visualized in order to confirm amplification of the marker sequences.
  • One typical visualization method involves staining of a gel with ethidium bromide and visualization under UV light.
  • the amplification products are integrally labeled with radio- or fluorometrically-labeled nucleotides, the amplification products can then be exposed to x-ray film or visualized under the appropriate stimulating spectra, following separation.
  • visualization is achieved indirectly.
  • a labeled nucleic acid probe is brought into contact with the amplified marker sequence.
  • the probe preferably is conjugated to a chromophore but may be radiolabeled.
  • the probe is conjugated to a binding partner, such as an antibody or biotin, and the other member of the binding pair carries a detectable moiety.
  • detection is by Southern blotting and hybridization with a labeled probe.
  • the techniques involved in Southern blotting are well known to those of skill in the art and can be found in many standard books on molecular protocols. See Sambrook et al, 1989. Briefly, amplification products are separated by gel electrophoresis. The gel is then contacted with a membrane, such as nitrocellulose or nylon, permitting transfer of the nucleic acid and non-covalent binding. Subsequently, the membrane is incubated with a chromophore-conjugated probe that is capable of hybridizing with a target amplification product. Detection is by exposure of the membrane to x-ray film or ion-emitting detection devices.
  • an expression construct that encodes a 3' sequence of GBV-B is utilized.
  • expression construct is meant to include any type of genetic construct containing a nucleic acid coding for a gene product in which part or all of the nucleic acid encoding sequence is capable of being transcribed. The transcript may be translated into a protein, but it need not be. Expression includes both transcription of a gene and translation of mRNA into a gene product. Expression may also include only transcription of the nucleic acid encoding a gene of interest. hi some constructs, the nucleic acid encoding a gene product is under transcriptional control of promoter and/or enhancer.
  • promoter will be used here to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II. Much of the thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 bp of nucleic acids, and containing one or more recognition sites for transcriptional activator or repressor proteins.
  • At least one module in each promoter functions to position the start site for RNA synthesis.
  • the best known example of this is the TATA box, but in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.
  • promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well.
  • the spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline.
  • Enhancers were originally detected as genetic elements that increased transcription from a promoter located at a distant position on the same molecule of DNA.
  • enhancers The basic distinction between enhancers and promoters is operational. An enhancer region as a whole must be able to stimulate transcription at a distance; this need not be true of a promoter region or its component elements. On the other hand, a promoter must have one or more elements that direct initiation of RNA synthesis at a particular site and in a particular orientation, whereas enhancers lack these specificities. Promoters and enhancers are often overlapping and contiguous, often seeming to have a very similar modular organization.
  • host cell refers to a prokaryotic or eukaryotic cell, and it includes any transformable organisms that is capable of replicating a vector or virus and/or expressing viral proteins.
  • a host cell can, and has been, used as a recipient for vectors, including viral vectors.
  • a host cell may be "transfected” or “transformed,” which refers to a process by which exogenous nucleic acid is transfe ⁇ ed or introduced into the host cell.
  • a transformed cell includes the primary subject cell and its progeny.
  • a "permissive cell” refers to a cell that supports the replication of a given virus and consequently undergoes cell lysis. In the context of the present invention, such a virus would include HCV, GBV-B, or other hepatitis viruses. In a “nonpermissive cell,” productive infection does not result, but the cell may become stably transformed.
  • methods employ permissive cells that are a cell line derived from liver cells (liver cell line).
  • Host cells may be derived from prokaryotes or eukaryotes, depending upon whether the desired result is replication of the vector or expression of part or all of the vector-encoded nucleic acid sequences.
  • Numerous cell lines and cultures are available for use as a host cell, and they can be obtained through the American Type Culture Collection (ATCC), which is an organization that serves as an archive for living cultures and genetic materials (www.atcc.org).
  • ATCC American Type Culture Collection
  • An appropriate host can be determined by one of skill in the art based on the vector backbone and the desired result.
  • a plasmid or cosmid for example, can be introduced into a prokaryote host cell for replication of many vectors.
  • Bacterial cells used as host cells for vector replication and or expression include DH5 ⁇ , JM109, and KC8, as well as a number of commercially available bacterial hosts such as SURE ® Competent Cells and SOLOPACKTM Gold Cells (STRATAGENE ® , La Jolla).
  • bacterial cells such as E. coli LE392 could be used as host cells for phage viruses.
  • eukaryotic host cells for replication and/or expression of a vector examples include HeLa, NIH3T3, Jurkat, 293, Cos, CHO, Saos, and PC12. Many host cells from various cell types and organisms are available and would be known to one of skill in the art.
  • a viral vector or virus or virus particle may be used in conjunction with either a eukaryotic or prokaryotic host cell, particularly one that is permissive for replication or expression of the vector. It is contemplated that the present invention includes vectors composed of viral sequences, viruses, and viral particles in the methods of the present invention, and that they may be used interchangeably in these methods, depending on their utility.
  • Some vectors may employ control sequences that allow it to be replicated and/or expressed in both prokaryotic and eukaryotic cells.
  • control sequences that allow it to be replicated and/or expressed in both prokaryotic and eukaryotic cells.
  • One of skill in the art would further understand the conditions under which to incubate all of the above described host cells to maintain them and to permit replication of a vector. Also understood and known are techniques and conditions that would allow large-scale production of vectors, as well as production of the nucleic acids encoded by vectors and their cognate polypeptides, proteins, or peptides.
  • compositions of the present invention comprise an effective amount of GBV-B clone as a therapeutic dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium.
  • pharmaceutically acceptable carrier refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and abso ⁇ tion delaying agents and the like.
  • the biological material should be extensively dialyzed to remove undesired small molecular weight molecules and/or lyophilized for more ready formulation into a desired vehicle, where appropriate.
  • the active compounds will then generally be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, intralesional, or even intraperitoneal routes.
  • parenteral administration e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, intralesional, or even intraperitoneal routes.
  • the preparation of an aqueous composition that contains GVB-B nucleic acid sequences as an active component or ingredient will be known to those of skill in the art in light of the present disclosure.
  • compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for using to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions of the active compounds as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • a GBV-B clone of the present invention can be formulated into a composition in a neutral or salt form.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like, i terms of using peptide therapeutics as active ingredients, the technology of U.S. Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,792; and 4,578,770, each inco ⁇ orated herein by reference, may be used.
  • inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides
  • organic bases as isopropylamine, trimethylamine, histidine, procaine and the like, i terms of using peptide therapeutics as active ingredients, the technology of U.S. Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,
  • the carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged abso ⁇ tion of the injectable compositions can be brought about by the use in the compositions of agents delaying abso ⁇ tion, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by inco ⁇ orating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by inco ⁇ orating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the prefe ⁇ ed methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the preparation of more, or highly, concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small area.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like also can be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035- 1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • other pharmaceutically acceptable forms include, e.g., tablets or other solids for oral administration; liposomal formulations; time release capsules; and any other form cu ⁇ ently used, including cremes.
  • Oral formulations include such nonnally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders.
  • oral pharmaceutical compositions will comprise an inert diluent or assimilable edible carrier, or they may be enclosed in hard or soft shell gelatin capsule, or they may be compressed into tablets, or they may be inco ⁇ orated directly with the food of the diet.
  • the active compounds may be inco ⁇ orated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 0.1% of active compound.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 75% of the weight of the unit, or preferably between 25-60%.
  • the amount of active compounds in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • the tablets, troches, pills, capsules and the like may also contain the following: a binder, as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring.
  • a binder as gum tragacanth, acacia, cornstarch, or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • the dosage unit form When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules maybe coated with shellac, sugar or both.
  • a syrup of elixir may contain the active compounds sucrose as a sweetening agent methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor.
  • the inventors elucidated the previously unrecognized 3' terminal sequence of GBV-B (SEQ ID NO 1). This sequence was reproducibly recovered from tamarin serum containing GBV-B RNA, in RT-PCR nucleic acid amplification procedures using several different primer sets, and as a fusion with previously reported 5' GBV-B sequences.
  • AATTCGGCCCTGCAGGCCACAACAGTC-3' which was phosphorylated at the 5 ⁇ end and chemically blocked at the 3 ⁇ end, was ligated to the RNA essentially using the method described by Kolykhalov et al. (Behrens et al, 1996).
  • the RNA was initially dissolved in DMSO and the following additions were made: Tris-Cl, pH 7.5 (10 mM), MgCl 2 (10 mM), DTT (5 mM), hexamine cobalt chloride (1 mM), 10 pmol oligo and 8 U T4 ligase.
  • the final concentration of DMSO was 30% in a final volume of 10 ⁇ L.
  • the ligation reaction was incubated for 4 or 20 hours at 19° C. 1 ⁇ L of the ligation reaction was used directly to make cDNA, using a primer complementary to the ligated oligonucleotide and the Superscript 2 system, in a final volume of 15 ⁇ L. 1 ⁇ L of cDNA was amplified using the Advantage cDNA system (Clontech) and two additional oligonucleotide primers. These primers included one that was complementary to the ligated oligonucleotide (i.e., "negative sense") and a positive-sense primer located near the 3" end of the reported GBV-B sequence. A product approximately 290 bases in length was obtained, and this was gel purified and directly sequenced.
  • Amplification was then done by PCR using the primer previously utilized for cDNA synthesis and a positive-sense primer mapping within the previously published GBV-B sequence, hi the initial studies, although a product was readily detected, DNA sequencing showed that this product was missing all of the sequence distal to the poly-U tract. Carrying out the cDNA synthesis in the presence of DMSO circumvented this problem.
  • a cDNA product of approximately 290 bases was obtained. This was sequenced and shown to consist of the 5" primer, 20 bases of the published GBV-B sequence, and 259 bases of the novel sequence obtained in the preceding studies and containing the sequence of the 3" primer. The sequence of the 3 ⁇ end of GBV-B is shown in SEQ ID NO: 1 (FIG. 1).
  • the possible secondary RNA structure for this region is shown in FIG. 2, as predicted by a computer-based RNA folding program.
  • the presence of a predicted hai ⁇ in structure at the extreme 3' end of this novel sequence is consistent with its location at the 3' terminus of the viral RNA.
  • the GBV-B cDNA (synthesis described above) was used as a template for PCR amplification of the 3' 1553 nucleotides (nts) of the GBV-B genome. This PCR amplification product was gel purified and cloned into plasmid DNA using the "Perfectly Blunt Cloning Kit" (Novagene).
  • RNA transcribed from this cDNA copy of the genome will be infectious when inoculated into the liver of a GBV-B permissive tamarin, giving rise to rescued GBV-B virus particles.
  • a 1:1000 dilution of GBV-B infectious tamarin serum was obtained. This material was used as a source of viral RNA for the amplification of GBV-B nucleic acid sequences by reverse- transcription polymerase chain reaction. For amplification of previously reported segments of the GBV-B genome, 250 ⁇ L of the diluted serum was extracted with Trizol using the manufacturer's instructions.
  • RNA pellet was dissolved in 10 ⁇ L of a 100 mM DTT buffer containing 5% RNasin.
  • This material was converted into cDNA using Superscript 2 reverse transcriptase and oligonucleotide primers designed to be complementary to the reported GBV-B RNA sequence and to contain unique restriction sites.
  • This cDNA was amplified using the Advantage cDNA kit (Clontech) employing the cDNA primer (negative sensiv as the downstream primer and a similar positive-sense upstream primer, again containing a unique restriction site.
  • the published sequence of GBV-B allowed for the selection of primers in convenient areas of the genome containing unique restriction sites.
  • nucleotides (nts) 1-1988 using an upstream primer containing a T7 RNA polymerase promoter and a BamHl site upstream of nt 1, and a downstream primer containing a unique EcolRl site (nt 1978); (2) nts 1968-5337, using a downstream primer containing a unique Clal site at position 5o27; (3) nts 5317- 7837, using a downstream primer containing a Sail site at nt 7847; and, (4) nts 7837-9143, using a downstream primer containing an added Xhol site.
  • RT-PCR products generated in these reactions were cloned into plasmid DNA after gel purification, using the "Perfectly Blunt Cloning Kit" (Novagene).
  • Ten bacterial colonies from each of the four RT-PCR products were analyzed for insert size by restriction endonuclease digestion using EcoRI, the sites for this enzyme being located on either side of the insert in the resulting plasmids.
  • 9 of 10 colonies contained plasmids with the co ⁇ ect size insert.
  • the EcoRI -Clal amplicon generated only 1/10 colonies with a co ⁇ ect size insert.
  • the 5' region of the cloned sequence revealed a relatively long nontranslated region co ⁇ esponding to the published sequence of the GBV-B 5 'NTR, which includes an IRES. This region was followed by a long open reading frame. Near the 3' end of the genome a poly-U tract was identified; however, this was shorter than the published 3' homopolymeric poly-U region.
  • GenBank database accesion U22304, "Hepatitis GB virus B polypeptide complete genome"
  • an infectious full-length clone can be produced by the following protocol.
  • a plasmid will be made containing a cassette including the 5' and 3 ⁇ ends of the virus flanked by appropriate restriction sites. These constructs have been shown to efficiently translate reporter genes, with transcription taking place via a T7 promoter placed immediately upstream of the 5 'NTR (e.g., see Rijnbrand et al, 1999). The major portion of the GBV-B genome would then be amplified by long range RT-PCR. This method is now well established for hepatitis C virus and other flaviviruses (Teller et al, 1996), and it has been used successfully also to amplify rhinovirus RNA.
  • this technique uses "Superscript” reverse transcriptase to synthesize cDNA and a mixture of "KlenTaq 1", and "Deep Vent” polymerases to amplify this cDNA .
  • Primers that can be used will contain restriction sites to allow cloning of the RT-PCR products into the cassette vector.
  • extensive restriction analysis will enable us to determine which clones contain inserts that are of full length and which have a high probability of being co ⁇ ect.
  • Apparent full-length clones will be analyzed further by coupled transcription-translation using the Promega "TnT” system, with the addition of microsomal membranes to allow the cleavage of the structural proteins by cellular signalase enzymes.
  • Clones which appear co ⁇ ect by restriction analysis and which produce GBV-B proteins, in particular the protein coded for by the extreme 3' end of the genome, NS5B, will be selected, and RNA will be transcribed from these clones using the Ambion MegaScript system. "Co ⁇ ect" looking clones (>10) can be injected directly into a tamarin liver at several sites. A successful infection will be determined as described below. If a positive signal is detected the entire genome will be amplified and sequenced to determine which plasmid the virus originated from.
  • RNA will be injected under direct visualization following a limited incision and exposure of the liver.
  • RT-PCR-detectable viral RNA or cDNA has not been detected in serum samples collected within days of this procedure in the absence of viral replication (Kolykhalov et al, 1997; Yanagi et al, 1998; Beard et al, 1999).
  • RNA in serum collected subsequently from these tamarins will be strong evidence for the replication competence of the synthetic RNA.
  • Serum will be collected weekly for six weeks, then every other week for an additional 6 weeks from inoculated animals.
  • alanine aminotransferase (ALT) levels will be measured as an indicator of liver injury and to assess liver histology in punch biopsies taken at the time of ALT elevation.
  • Maximum viremia and an acute phase ALT response is expected to occur around 14-28 days post-inoculation of infectious RNA (Simons et al, 1995; Schlauder et al, 1995; Karayiannis et al, 1989).
  • Transfections will be considered to have failed to give rise to infectious virus if RNA is not detected in the serum within 12 weeks of inoculation. Successfully infected animals will be followed with twice weekly bleeds until resolution of the viremia, or for 6 months, whichever is longest.
  • the GBV-B genome can be used as the acceptor molecule in the construction of chimeric viral RNAs containing sequences of both HCV and GBV-B. Such constructs will allow one to investigate the mechanisms for the different biological properties of these viruses and to discover and investigate potential inhibitors of specific HCV activities (e.g., proteinase) required for HCV replication.
  • HCV activities e.g., proteinase
  • Different classes of chimeric viruses are contemplated. These include: (a) replacement of the GBV-B IRES with that of HCV; and (b) replacement of the NS3 major serine proteinase and helicase, and (c) the replacement of the NS5B RNA-dependent RNA polymerase with the homologous proteins of HCV.
  • chimeric constructs described in the following sections will be made by PCR mutagenesis, using high fidelity polymerases and oligonucleotide primers designed to include the specific fusions of GBV-B and HCV sequences (Landt et al, 1990).
  • First round PCR reactions will create the desired fusion, and generate a new "primer” to be used in a second PCR reaction spanning the region to a convenient unique restriction site.
  • PCR cycles will be kept to the minimum number necessary for successful amplification, and all segments of viral sequence that are amplified by PCR will be subjected to DNA sequencing to exclude the presence of unwanted PCR-introduced e ⁇ ors. Sequencing will be accomplished at UTMB's core Recombinant DNA Laboratory.
  • Amplified segments will be kept to the minimum by the exchange of cloned cDNA segments spanning convenient restriction sites in subgenomic clones, and where necessary PCR artifacts can be co ⁇ ected by site-directed mutagenesis (QuickChange mutagenesis kit, Stratagene).
  • RNA virus chimeras A number of viable positive-strand RNA virus chimeras have been constructed previously in which LRES elements have been swapped between different viruses. Most of these chimeras have involved the exchange of IRES elements between picornaviruses. Others have been successful in constructing viable pohovirus chimeras containing the HCV IRES in place of the native pohovirus LRES (Zhao et al, 1999; Lu and Wimmer, 1996). A similar rhinovirus 14 chimera containing the HCV LRES has been constructed, although its replication phenotype is not as robust as the pohovirus chimera described by Lu and Wimmer (Lu and Wimmer, 1996).
  • Frolov et al (Frolov et al, 1998) recently reported chimeric flaviviruses in which the HCV LRES was inserted into the genetic background of a pestivirus, bovine viral diarrhea virus (BVDV) in lieu of the homologous BVDV sequence. Although these viable chimeric polioviruses and pestiviruses replicate in cell cultures, they are poor su ⁇ ogates for HCV in animal models as neither virus is hepatotropic or causes liver disease. Importantly, Frolov et al.
  • BVDV bovine viral diarrhea virus
  • Frolov et al demonstrated quite convincingly that the requirement for cis-acting replication signals at the 5' terminus of the pestivirus genome was limited to a short tetranucleotide sequence. This requirement presumably reflects the need for the complement of this sequence at the 3' end of the negative strand during initiation of positive-strand RNA synthesis.
  • the work of Frolov et al shows that the IRES of BVDV does not contain necessary replication signals, or that if these are present within the BVDV IRES they can be complemented with similar signals in either the HCV or encephalomyocarditis virus (EMCV, a picornavirus) IRES sequence.
  • EMCV encephalomyocarditis virus
  • HCV-H77C infectious HCV clone
  • pCV-H77C contains the sequence of the genotype la Hutchinson strain virus (Yanagi et al, 1998), whose infectivity in a chimpanzee following intrahepatic inoculation with synthetic RNA transcribed from pCV-H77C has been confirmed.
  • This GB/C:IRES construct will retain two upstream hai ⁇ ins within the GBV-B sequence (stem-loops la and lb), and it is thus analogous to the viable "BVDV+HCVdelB2B3Hl" chimera of Frolov et al (Frolov et al, 1998).
  • a second chimera can be constructed in which the entire HCV 5' nontranslated RNA will be inserted in lieu of nts 62-446 of the GBV-B genome ("GB/C:5'NTR"). This construct will add to the inserted HCV sequence the most 5' stem-loop from HCV (stem-loop I).
  • RNA derived from these constructs will be assessed and compared to the translational activity of the wild-type GBV-B and HCV RNAs. These studies will be carried out in a cell-free translation assay utilizing rabbit reticulocyte lysates (Rijnbrand et al, 1999). Synthetic RNA will be produced by runoff T7 RNA polymerase transcription using as template Clal-digested plasmid DNA (BamHI digestion in the case of the genome HCV construct) (T7 Megascript kit, Ambion).
  • RNA at least 50% full-length as determined by agarose gel electrophoresis
  • microsomal membranes Promega.
  • S- Methionine-labelled translation products will be separated by SDS-PAGE, and the quantity of El protein produced from each RNA determined by Phosphorlmager analysis (Molecular Dynamics). Comparisons of the activity of the HCV IRES in the background of GBV-B and HCV will take into account differences in the methionine content of the El proteins of these viruses.
  • GB/C:5'NTR may generate viremia and liver injury more closely resembling that observed with wild-type GBV-B infection (Frolov et al. , 1998).
  • Chimeric flaviviruses containing the HCV NS3 serine proteinase/helicase within the GBV-B background are also contemplated within the present invention.
  • the construction of chimeric flaviviruses containing specific heterologous functional polyprotein domains poses a number of special problems. Unlike the situation with the IRES, where the relevant RNA segments appear to have a unique function restricted to cap-independent translation initiation and interact with host cell macromolecules, viral proteins often have multiple functions and may form specific macromolecular complexes with other viral proteins that are essential for virus replication (Lindenbach and Rice, 1999). Furthermore, such chimeric polyproteins must be amenable to efficient processing by the viral proteinases (NS2/NS3 or NS3).
  • NS3 contains the major serine proteinase that is responsible for most cleavage events in the processing of the nonstructural proteins, i.e., those that occur at the NS3/4A, 4A/4B, 4B/5A and 5A/5B junctions.
  • the active proteinase domain of HCV is located within the amino terminal third of the NS3 protein (residues 1-181), which shares 31% amino acid identity with the analogous segment of the GBV-B polyprotein (GBV-B vs HCV-BK) (Muerhoff et al, 1995). Importantly, the active site of this proteinase appears to be particularly well conserved in GBV-B.
  • the GBV-B proteinase maintains the residues that are responsible for catalysis and zinc binding in the HCV enzyme (Muerhoff et al, 1995), and unlike the NS3 proteinases of some other flaviviruses preserves the Phe- 154 residue that determines in part the S ⁇ specificity pocket of the enzyme and the preference of the HCV proteinase for substrates with a cysteine residue at the PI position (Scarselli et al, 1997). Thus, it is not su ⁇ rising that the relevant proteolytic cleavage sites within the GBV-B polyprotein that are predicted from alignments with the HCV polyprotein all possess a Cys residue at this position.
  • HCV NS3 proteinase functions as a noncovalent assembly of the NS3 proteinase domain and the amino terminal portion of NS4A, a proteinase accessory factor.
  • the details of this association are well known, and have been studied at the crystallographic level (Kim et al, 1996).
  • the N-terminal domain of the folded proteinase contains eight ⁇ strands, including one contributed directly by the NS4A peptide backbone. X-ray studies have shown that this a ⁇ ay of ⁇ strands gives rise to a much more ordered N-terminus. Thus, the presence of the NS4A strand seems likely to contribute to the structure of the substrate-binding pocket.
  • NS3 proteinase of GBV-B also requires a similar interaction with NS4A of that virus for complete activity, or, if so, whether the NS4A of GBV-B could substitute for NS4A of HCV in forming the fully active NS3 proteinase of HCV.
  • the predicted GBV-B NS4A molecule is 54 amino acid residues in length (Simons, et al, 1995; Muerhoff et al, 1995), just as in HCV.
  • the level of amino acid homology between the NS4A molecules is not especially high, and the potential interaction with either NS3 molecule cannot be predicted from this sequence on the basis of available knowledge.
  • chimeras will be created in which not only the NS3 proteinase domain of GBV-B is replaced, but also the relevant NS4A segment as well, with homologous segments of the HCV polyprotein.
  • the interaction of the HCV NS3 and NS4A domains represents a unique target for antiviral drug design, and it would be beneficial to have this specific interaction present in any virus to be used as a su ⁇ ogate for HCV in the evaluation of candidate antiviral inhibitors of HCV proteinase in vivo.
  • the NS3 proteinase chimeras that can be made include "GB/C:NS3 P ", which will contain the sequence encoding the first 181 amino acid residues of the HCV NS3 molecule in lieu of that encoding the first 181 residues of GBV-B NS3, and "GB/C:NS3 P 4A", which will include the same NS3 substitution as well as the HCV sequence encoding the amino-terminal segment of NS4A that forms the interaction with NS3.
  • the precise NS4A sequence to be included in the latter chimera will be based on the modeling studies, which may also suggest more effective fusions of the NS3 proteinase domain of HCV with the downstream NS3 helicase domain of GBV-B.
  • HCV cDNA for these studies will be the infectious HCV clone, pCV- H77C, which contains the sequence of the genotype la Hutchinson strain virus (Yanagi et al, 1998).
  • pCV- H77C which contains the sequence of the genotype la Hutchinson strain virus (Yanagi et al, 1998).
  • RNA helicase activity In addition to serine proteinase activity located within the amino-third of NS3, the downstream carboxy-terminal two-thirds of the molecule contains an RNA helicase activity. These two functional domains appear to be separated by a flexible spacer, within which the fusion of HCV proteinase or helicase domain sequences with GBV-B sequence will be placed.
  • the exact role of the helicase in the HCV life-cycle is not known, but it is almost certainly required for dsRNA strand-separation during some phase of viral RNA synthesis.
  • the helicase domains of GBV-B and HCV are remarkably well conserved, with some regions within the helicase showing as much as 55% amino acid identity (Muerhoff et al, 1995).
  • the GBV-B helicase is more closely related to the HCV helicase than all other flaviviral NS3 helicases, and it preserves many residues found within the conserved helicase motifs of HCV.
  • the HCV NS3 helicase may be capable of functioning when placed within the polyprotein of GBV-B, and such a chimeric virus may be capable of replication.
  • Residues 182-620 of the GBV-B NS3 molecule will be substituted with the analogous segment of HCV (FIG. 5, "GB/C:NS3 h ").
  • a chimera will also be made in which the entire NS3 and amino terminal NS4 protein sequences of GBV-B is replaced with the homologous HCV sequences ("GB/C:NS3-4A").
  • the latter construct will thus represent a dual proteinase-helicase chimera.
  • the HCV cDNA will be derived from pCV-H77C (Yanagi et al, 1998).
  • RNAs produced in vitro from these clones will characterized in vitro. This evaluation will be restricted to a documentation of the proper processing of the expressed polyprotein (i.e., NS2/NS3 and NS3 proteinase functions), since there are no relevant assays that can determine whether the helicase or RNA-dependent RNA polymerase activities in these polyproteins are sufficient for virus replication.
  • the proteolytic processing of the polyprotein is important, however, as it may be altered either by inclusion of the heterologous HCV NS3 proteinase in lieu of the natural GBV-B protease, or by a change in the folding of the polyprotein induced by inclusion of HCV sequence anywhere within the polyprotein.
  • TnT cell-free coupled transcription/translation assays
  • Template DNAs will be digested with Sal I, which restricts the cDNA within the NS5B coding region.
  • 35 S-methionine-labelled translation products will be separated by SDS- PAGE, and the mature NS3 protein identified by its apparent molecular mass.
  • the NS3 and NS5B proteins will be identified by immunoblot analysis using rabbit antisera to the GBV-B NS3 and NS5B proteins.
  • a mature ⁇ 68 kDa NS3 protein will provide proof of both the cw-active NS2/NS3 cleavage and the NS3-mediated cleavage of NS3/NS4A.
  • identification of a mature, processed NS5B molecule will provide further support for the activity of the NS3 proteinase.
  • Controls for these studies will be the wild-type GBV-B polyprotein expressed in similar fashion from the full-length GBV-B clone. If necessary to more clearly demonstrate the processing of the nonstructural proteins in these constructs, subclones representing the nonstructural region of the chimeric sequences could be produced.
  • Synthetic RNAs produced from each of the chimeric GBV-B/HCV constructs described in the preceding section will be tested for their ability to induce infection and cause liver disease in susceptible tamarins. These studies will be carried out using the approach described above.
  • the HCV NS5B molecule contains an RNA-dependent RNA polymerase that plays a central role in replication of the virus. Although this molecule represents a prime target for drug discovery efforts, it has proven difficult to express NS5B in a form that retains enzymatic activity specific for HCV RNA as a substrate. Thus, relatively little is known of the functional activity of the HCV replicase, including structure-function relationships of NS5B. Despite this, the NS5B proteins of GBV-B and HCV appear to be functionally closely related, as they share as much as 43% amino acid identity (Muerhoff et al, 1995). A more important question may be whether an RNA dependent RNA polymerase can act on foreign substrates.
  • a chimeric genome-length virus can be created in which the NS5B coding sequence of HCV (amino acids 2422-3014, 593 residues) is inserted within the background of GBV-B in lieu of its native RNA-dependent RNA polymerase (amino acids 2274-2864, 591 residues).
  • This chimeric virus would be valuable for animal studies of candidate antiviral inhibitors of HCV RNA synthesis.
  • This NS5B chimera would be evaluated to determine that there was proper proteolytic processing of the polyprotein. This would be accomplished by expression of the chimeric polyprotein in a coupled translation-transcription reaction, followed by immunoblot analysis for the mature NS5B protein, as described for the NS3 and NS3-4A chimeras in the preceding section. If these results confirm that the GB/C:5B chimeric polyprotein is processed with release of NS5B, studies in tamarins would progress to determine whether synthetic RNA transcribed from the clone is infectious and capable of causing liver disease in intrahepatically inoculated animals. These studies would be carried out as described above.
  • a chimeric molecule can be constructed from an infectious GBV-B clone in which the
  • HCV NS3 proteinase or proteinase helicase sequence would be placed in frame in lieu of the homologous GBV-B sequence, and this chimeric cDNA would be used to generate infectious GBV-B HCV chimeric viruses by intrahepatic inoculation of synthetic RNA in tamarins.
  • GBV-B and HCV proteinases have closely related substrate recognition and cleavage properties, likely making such chimeras viable and capable of initiating viral replication in appropriate cell types.
  • a chimeric virus containing HCV structural proteins could answer many fundamental questions concerning the structure and interaction of these proteins in HCV. They would also be useful in addressing the nature of the immune response to HCV structural proteins in infected primates (Farci et al, 1992). More to the point of this application, the availability of such chimeric viruses would allow studies of candidate HCV vaccines to be carried out in the tamarin model. This would be a major advance, because at present such studies are limited to chimpanzees (Choo et al, 1994).
  • the resulting virus would be an exceptionally valuable resource for future studies as it would contain all of the nonstructural replication elements, as well as the 5' and 3' nontranslated regions, of HCV. Such a virus would allow the tamarin model to be used to address many unresolved issues in HCV biology and pathogenesis.
  • EXAMPLE 16 Construction and Evaluation of Structural Protein Chimeras hi designing structural protein chimeras it is important to note that the two envelope proteins of HCV, El and E2, form noncovalent heterodimeric complexes that are likely to be important in the assembly of infectious virus particles. This is not known to be the case with the envelope proteins of GBV-B, but it is likely given similarities in the sizes and hydropathy profiles of these proteins (Simons et al, 1995; Muerhoff et al, 1995). Accordingly, the El and E2 proteins will be replaced as a unit, and chimeras containing only one of these proteins from the heterologous virus will generally not be produced.
  • a chimera will be created where the El and E2 regions of GBV-B virus are replaced with those of HCV, "GB/C:El-2".
  • the source of HCV cDNA for these constructions will be pCV-H77C (Yanagi et al, 1998).
  • a chimera will also be made in which the core protein, in addition to the envelope proteins, is replaced with the homologous proteins of HCV ("GB/C:Co-E2").
  • Additional chimeras will be made to determine whether tamarins can be infected with chimeras containing the GBV-B structural proteins within the genetic background of HCV. These will include "C/GB:El-2" and "C/GB:Co-E2".
  • the backbone for these chimeras will be pCV-H77C, the infectious genotype la cDNA clone developed in the Purcell laboratory at NIALO (Yanagi et al, 1998).
  • GBV-B The specific amino acid sequences of GBV-B to be replaced with the homologous segments of HCV have been determined by alignments of the GBV-B and HCV sequences, coupled with the location of signalase cleavage sites predicted to be present within the amino terminal third of the GBV-B polyprotein using the computer algorithm of Von Heijne. These predicted signalase cleavages lie between residues 156/157 (core/El), aa 348-349 (E1/E2) and 732/733 (E2 NS2) in the GBV-B sequence.
  • the chimera GB/C:El-2 will contain sequence encoding HCV aa 192-809 in lieu of that encoding aa 157-732 in GBV-B, while the insertion in the GB/C:Co-E2 chimera will extend from the initiator AUG codon (aa 1) to residue 809 in HCV, and will be spliced into GBV-B in lieu of the segment encoding aa 1-732 in the GBV-B clone.
  • the complementary chimeras to be constructed within the background of HCV will involve exchanges of the same segments of the genomes.
  • this virus will be passaged in GBV-B na ⁇ ve tamarins to further characterize the nature of the infection induced by the chimera. This will be accomplished by taking a pool of the 3 highest titer GBV-B RNA-containing serum specimens from the animal that was successfully transfected with RNA, and inoculating 1 mL of a 1:100 dilution of this pool intravenously into two susceptible animals. These animals will be monitored for infection and liver disease. These animals will be followed until resolution of the viremia and appearance of antibodies detectable in immunoblots with GST-NS3 protein expressed in E. coli, or for at least 6 months should an animal sustain a chronic infection.
  • RT-PCR amplification of chimeric segments of the genome may be employed to determine whether the altered phenotype results from mutations within the heterologous portion of the genome.
  • GBV-B and/or GBV-B/HCV chimeras can be used as a model for HCV. Such studies will allow one to investigate the mechanisms for the different biological properties of these viruses and to discover and investigate potential inhibitors of specific HCV activities (e.g., proteinase) required for HCV replication.
  • HCV activities e.g., proteinase
  • GBV-B/HCV Chimeras to Test Candidate HCV NS3 Proteinase Inhibitors or Other Inhibitors of HCV GBV-B/HCV viruses can be used in preclinical testing of candidate HCV NS3 proteinase inhibitors or other inhibitors of HCV.
  • the term “candidate substance” refers to any molecule that is capable of modulating HCV NS3 proteinase activity or any other activity related to HCV infection.
  • the candidate substance may be a protein or fragment thereof, a small molecule inhibitor, or even a nucleic acid molecule. It may prove to be the case that the most useful pharmacological compounds for identification through application of the screening assay will be compounds that are structurally related to other known modulators of HCV NS3 proteinase activity.
  • the active compounds may include fragments or parts of naturally-occurring compounds or may be only found as active combinations of known compounds that are otherwise inactive. However, prior to testing of such compounds in humans or animal models, it will be necessary to test a variety of candidates to determine which ones have potential.
  • the active compounds may include fragments or parts of naturally-occurring compounds or may be found as active combinations of known compounds that are otherwise inactive.
  • the present invention provides screening assays to identify agents that are capable of inhibiting proteinase activity in a cell infected with chimeric GBV-B HCV viruses containing the HCV proteinase. It is proposed that compounds isolated from natural sources, such as animals, bacteria, fungi, plant sources, including leaves and bark, and marine samples may be assayed as candidates for the presence of potentially useful pharmaceutical agents. It will be understood that the pharmaceutical agents to be screened could also be derived or synthesized from chemical compositions or man-made compounds.
  • candidate substance identified by the present invention may be polypeptide, polynucleotide, small molecule inhibitors or any other compounds that may be designed through rational drug design starting from known inhibitors of proteinases or from structural studies of the HCV proteinase.
  • the candidate screening assays are simple to set up and perform.
  • a candidate substance in assaying for a candidate substance, after obtaining a chimeric GBV-B HCV virus with infectious properties, a candidate substance can be incubated with cells infected with the virus, under conditions that would allow measurable changes in infection by the virus to occur.
  • the ability of the candidate inhibitory substance to reduce, abolish, or otherwise diminish viral infection may be determined.
  • Effective amounts in certain circumstances are those amounts effective to reproducibly reduce infection by the virus in comparison to the normal infection level. Compounds that achieve significant appropriate changes in activity will be used.
  • Candidate compounds can be administered by any of a wide variety of routes, such as intravenously, intraperitoneally, intramuscularly, orally, or any other route typically employed. It will, of course, be understood that all the screening methods of the present invention are useful in themselves notwithstanding the fact that effective candidates may not be found. The invention provides methods of screening for such candidates, not solely methods of finding them. 2. In vitro Assays
  • the invention encompasses in vitro screening of candidate substances. Using a cell line that can propagate GBV-B in culture, in vitro screening can be used such that GBV-B or HCV virus production or some indicator of viremia is monitored in the presence of candidate compounds. A comparison between the absence and presence of the candidate can identify compounds with possible preventative and therapeutic value.
  • the present invention also encompasses the use of various animal models to test for the ability of candidate substances to inhibit infection by HCV. This form of testing may be done in tamarins.
  • the assays previously described could be extended to whole animal studies in which the chimeric virus could be used to infect a GBV-B permissive primate, such as a tamarin. One would then look for suppression of viral replication in the animal, and a possible impact on liver disease related to replication of the infectious chimeric virus.
  • the advantage of this in vivo assay over present available assays utilizing HCV infection in chimpanzees is the reduced cost and greater availability of GBV-B permissive nonhuman primate species.
  • Treatment of animals with test compounds will involve the administration of the compound, in an appropriate form, to the animal.
  • Administration will be by any route that could be utilized for clinical or non-clinical pu ⁇ oses, including but not limited to oral, nasal, buccal, rectal, vaginal or topical.
  • administration may be by intratracheal instillation, bronchial instillation, intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection.
  • systemic intravenous injection, intraperitoneal injection, and oral administration Determining the effectiveness of a compound in vivo may involve a variety of different criteria. Such criteria include, but are not limited to, survival, reduction of rate of infection, a ⁇ est or slowing of infection, elimination of infection, increased activity level, improvement in liver function, and improved food intake.
  • Infectious GBV-B/HCV chimeras expressing HCV envelope proteins will have utility as a vaccine immunogen for hepatitis C.
  • Such clones clearly have the potential to be constructed as chimeras including relevant hepatitis C virus sequences in lieu of the homologous GBV-B sequence, providing unique tools for drug discovery efforts.
  • Chimeric viruses containing the envelope proteins of hepatitis C virus would confer the antigenic characteristics of hepatitis C virus on the chimera.
  • These chimeras may have the ability to replicate in chimpanzees (and thus humans) by virtue of the fact that the chimeric envelope is now able to interact with the human hepatocyte cell surface, a necessary first step in virus replication. Therefore, the chimeric virus, while able to infect and replicate in humans, may not cause much or any disease-the reasoning here is that the genetic backbone of the chimera that encodes the nonstructural proteins of GBV-B has not evolved for replication in human cells and thus may not replicate well.
  • the chimera may have limited replication ability, cause no disease, but still elicit immunity to the surface envelope proteins of HCV and thus have potential as a hepatitis C vaccine. These chimeras can be tested for their ability to promote immunity to HCV through an immune response.
  • the serum sample was diluted with 100 ⁇ l of fetal calf serum and extracted using the Trizol system (GIBCO/BRL). The pellet was dissolved in 10 mM dithiothreotol containing 20 u/mL RNasin (Promega).
  • Primer 1 5'CGGGATCCCGTAATACGACTCACTATAGACCACAAACACTCCAGT TTG - 3' (SEQ ID NO:3)
  • Primer 2 5'-GTGGAATTCACAGCGTCATA-3' (SEQ ID NO:4) (Places a T7 RNA polymerase promoter sequence immediately upstream of the GBV-B sequence; the amplified segment extends from the 5' terminus of the viral genome to the unique EcoRI site at position
  • Primer 3 5'-TGTGAATTCCACTCTCCTACC-3' (SEQ ID NO:5)
  • Primer 4 5'-TTATCGATTGCAGCAACCATG-3' (SEQ ID NO:6) (Overlapping EcoRI site at position 1978 and the unique Clal site at 5327.)
  • Primer 5 5'-CATGGTTGCTGCAATCGATAAGCTGAAGAGTACAATAAC-3' (S ⁇ Q ID
  • Primer 7 5'-CTGTCATGGGAGATGCGTAC-3' (S ⁇ Q ID NO:9)
  • Primer 8 5'-CGAGCTCGAGCACATCGCGGGGTCGTTAAGCCCGGGGTCTCC-3' (S ⁇ Q
  • Primer 10 5'-CCGACTCGAGAATTCGGCCCTGCAGGCCACAACAGTCTCGCGAG
  • TTGGTAC-3' (SEQ ID NO: 12) (Overlaps product of primers 7 and 8 and extends to the novel 3' terminal sequence; includes an XJio I site at the 3' terminus of the GBV-B sequence to aid subsequent manipulations.)
  • RT-PCR products were gel-purified using the QIAquick Gel Extraction Kit (Qiagen) and ligated into a plasmid vector with the PstBlue-1 Perfectly Blunt Cloning Kit (Novagene).
  • the cloned overlapping cDNA fragments were assembled into the pACNR1180 plasmid using the unique restriction sites noted in the primer descriptions.
  • the final, fully assembled plasmid bearing a 9.4 kb cDNA GBV-B insertion was subjected to DNA sequencing to confirm the validity of the construction and the absence of any mutations or e ⁇ ors in the sequence that may have been introduced during the assembly process.
  • the sequence of the GBV-B infectious cDNA is shown in SEQ LD NO:2. Plasmid DNA containing the GBV-B cDNA was purified by isopycnic centrifugation on a CsCl gradient and isolated by ethanol precipitation.
  • the plasmid DNA was linearized at the 3' terminus of the cDNA insert by digestion I. Two ⁇ g of the linearized DNA was used as template in an in vitro transcription reaction using the Ambion T7 MEGAscript Kit according to the manufacturer's instructions. The integrity of the resultant synthetic RNA product was confirmed by nondenaturing agarose gel electrophoresis.
  • RNA transcribed from the GBV-B cDNA clone was diluted in phosphate buffered saline and injected under direct visualization at laparotomy into the liver of a healthy tamarin without a prior history of GBV-B infection (Saguinus oedipus). Serum samples were collected from the animal weekly beginning at 2 weeks post-inoculation and tested for the presence of GBV-B RNA by real-time quantitative RT-PCR (TaqMan® assay). Results are shown in Table 2.
  • a genome-length molecular clone of GBV-B was assembled from a series of overlapping cDNA fragments produced by RT-PCR from viral RNA isolated in a GBV- B-infected tamarin serum. This clone inco ⁇ orates the novel GBV-B 3 'NTR sequence, as described herein.
  • the cDNA has been placed downstream of a T7 RNA polymerase promoter in the context of the bacterial vector pACNR1180.
  • a C ⁇ T substitution was introduced at nucleotide (nt) position 496 of the GBV-B cDNA, so that a unique M restriction site was generated in the core-coding sequence without modifying the amino acid sequence.
  • the resulting plasmid containing the full-length GBV-B cDNA with the engineered Mlul site at nt position 491 will be refe ⁇ ed to as pGBV-B/Mlu.
  • Chimeric GBV-B / HCV 5'NTRs were generated by substituting domains of the GBV-B 5'NTR by HCV counte ⁇ arts using a gene fusion PCR strategy (Landt et al, 1990) and HCV sequences derived from Hutchinson strain of genotype la, Genbank accession numbers AF011751 (SEQ LD NO:13), AF011752, and AF011753, each of which is inco ⁇ orated herein by reference (Inchauspe et al, 1991).
  • the invention is not limited to a particular strain of HCV and sequences found in other HCV sequences may also be used, such as Genbank accession numbers AF009606, M67463, AF290978, AF387806, AF271632, M62321, AF387807, AF387805, AF387808, D10749, AF511948, AF177040, AF177038, AF177039, AF177037, AF511949,or M32084, each of which is inco ⁇ orated herein by reference.
  • Chimeric 5'NTRs were amplified using overlapping PCR fragments spanning the desired HCV region framed by appropriate GBV- B regions, so that the BamHI restriction site located upstream of the T7 promoter sequence and the Mlul restriction site (nt 491) were used to substitute the parent GBV-B fragment within the molecular infectious clone.
  • the sequence of all PCR-amplified fragments within the resulting, chimeric pGB/I- ⁇ -JJI HC , pGB/ ⁇ Ib/II-III HC , pGB/ ⁇ -JH HC , and pGB/m HC plasmids (FIG. 2) were confirmed.
  • RNAs transcribed from the plasmids shown in FIG. 2 some of which contain chimeric IRES viral sequences, translation initiation activities were determined in vitro in rabbit reticulocyte lysates and ex vivo in primary tamarin hepatocytes.
  • the Mlul-Xhol fragment spanning almost the entire GBV-B coding sequence and the 3' NTR of the cDNA in pGBV-B/Mlu, was replaced by a fragment encoding Renilla luciferase (RLuc) followed by the GBV-B 3' NTR.
  • RNAs representing I-H-IEF, ⁇ lb/ ⁇ -i ⁇ HC , and ⁇ -Ifl HC all of which contain the complete HCV IRES element, translated with about 2.5-fold higher efficiency as compared to GBV-B/Mlu RNA which contains the intact GBV-B LRES (FIG. 2).
  • the translational activity observed for i ⁇ HC was similar to that observed for the GBV-B/Mlu RNA. This is remarkable as this RNA contains a chimeric IRES in which upstream and downstream sequences flanking domain IJJ of the HCV LRES are derived from the GBV-B LRES.
  • RNA transcript encoding firefly luciferase (FLuc) under translational control of the EMCV IRES was cotransfected at 1:19th the abundance of the RLuc reporter constructs.
  • the RNAs were inoculated into the liver under direct visualization following exposure by laparotomy. Inoculated animals were followed as described in the preceding sections of this document, with periodic testing of serum ALT, antibody to the NS3 protein of GBV-B, GBV-B genome, and with liver biopsy to assess pathologic changes in the liver. All tamarins were housed at the Southwest Regional Primate Research Center at the Southwest Foundation for Biomedical Research. Animals were cared for by members of the Department of Laboratory Animal Medicine at the Southwest Foundation for Biomedical Research in accordance with the Guide for the Care and Use of laboratory Animals. All protocols were approved by the Institutional Animal Care and Use Committee.
  • the cDNA product was quantified in a 5' exonuclease PCR (TaqMan®) assay using a primer-probe combination that recognized sequences in the GBV-B NS5A gene.
  • the primers NS5ARPp and NS5AFPp (5'-CCAGTTCCGGGCAAGAACT-3', position 6844-6862)(SEQ ID NO: 15) and probe (nts 6913-6938) were obtained from Fisher and Perkin Elmer, respectively.
  • a synthetic RNA derived from the infectious clone was used as the standard for quantitation of the RNA.
  • viremia reappeared in tamarin T16444, rapidly reaching a titer in excess of 10 genome copies per ml, and subsequently persisting for 20 weeks (terminating 32 weeks following inoculation of the RNA) (FIG. 5).
  • the course of the infection in this animal from week 12 on was similar to what is seen in infection of na ⁇ ve animals with the wild-type infectious clone, with the exception that there was no detectable evidence of antibodies to the NS3 protein.
  • the long delay to the appearance of the secondary viremia in this animal suggests a requirement for the accumulation of adaptive mutations that are compensatory to changes in the replication capacity of the RNA related to the presence of the chimeric 5'NTR sequence.
  • results shown in FIG. 5 indicate the replication competence of the in chimeric RNA (see FIG.' 2) following intrahepatic inoculation of synthetic RNA in a GBV-B naive tamarin (S. mystax).
  • the presence of the chimeric 5'NTR sequence was confirmed in the RNA replicating in this animal by RT-PCR amplification of RNA extracted from the serum, followed by direct DNA sequencing of the amplified cDNA.
  • This second animal rapidly developed evidence of GBV-B infection, with viremia detected as early as 1 week after infection, and with a peak viremia of greater than 10 8 genome copies/ml by 4 weeks postinfection.
  • the rapid appearance of the viremia and early robust replication of the virus in this second animal suggested that the chimeric virus had indeed undergone adaptation and that it no longer required a lengthy incubation period to induce viremia.
  • Random or GBV-B specific primers were used to prepare cDNA using MMLV reverse transcriptase from the Advantage RT-for-PCR kit (Clontech). PCR amplification of 1-2 kb fragments was carried out using the Advantage 2 PCR enzyme system (Clontech) utilizing GBV-B specific primers. Amplimers were purified through a silica gel membrane (QiaQuick PCR purification kit, Qiagen) and subjected to direct sequencing on an ABI 373XL instrument. The 3' terminus of the viral genome was ligated to a 27-mer oligonucleotide as described in the previous section for parent GBV-B.
  • RNA was then subjected to reverse transcription using a primer complementary to the ligated primer, and the cDNA was subsequently amplified using the same 3' primer and a 5' primer specific for a sequence in the 3 'NTR (nts 9092-9111).
  • a semi-nested PCR was performed with an internal 5' primer (nts 9097-9114).
  • the 5' terminus of the viral genome was subjected to reverse transcription using a 3' primer complementary to the domain Ifl of the HCV 5'NTR sequence (nts 209-229 of the HCV cDNA).
  • GB/ni HC chimeric virus replicates robustly in small primates (see FIG. 11) and were therefore used for antiviral validation studies in the small primate model.
  • GB/L ⁇ HC chimeric virus were also demonstrated to replicate well in primary cultures of tamarin hepatocytes (FIG.
  • tamarin hepatocytes Primary tamarin hepatocytes (Saguinus mystax) were prepared as described previously (Beanies et al, 2000). Wild-type (wt) GBV-B and chimeric virus GB/nj HC inocula were derived from tamarins that had been infected with the co ⁇ esponding viruses. 2.5 x 10 cells were seeded into each well of 6-well plates, and subsequently infected with virus (wt or chimera) at a multiplicity of infection (m.o.i.) of approximately 0.6 genome equivalents (ge)/cell. One hour after infection, cells were thoroughly washed 5 times and fresh medium was added. The media was replaced with fresh media every 24 hours thereafter.
  • m.o.i. multiplicity of infection
  • GBV-B RNA in culture supernatants and cell extracts was measured quantitatively at the indicated times (FIG. 8) by a realtime RT- PCR (TaqMan®) assay targeting GBV-B core protein coding sequence (Beames et al, 2000). Results showed that there is synthesis and secretion of newly fo ⁇ ned viral particles in the culture supernatant after infection with both the wild-type and chimeric viruses. The chimeric IRES virus replicated efficiently in these cultures, although to titers that were about 10-fold less than the wild-type GBV-B titers.
  • GB/ffl chimeric virus makes possible the validation of candidate antiviral compounds targeting domain in of the HCV IRES in the context of a complete viral replication cycle in primary hepatocytes in ex vivo culture. This is a significant advantage over the only other system for cell culture validation of antiviral compounds, which consists of a cell culture system wherein only the effect on viral RNA replication can be monitored.
  • the GB/LTI chimeric virus also allows such studies in the small primate model: tamarins, marmosets, or owl monkeys.
  • Virus recovered from T 16444 that contains the chimeric 5'NTR as well as all of these mutations, replicated efficiently when passaged in a second tamarin (T16451, FIG. 11).
  • the RNAs were inoculated into the liver under direct visualization following exposure by laparotomy. Inoculated animals were followed as described above, with periodic testing of serum ALT, antibody to the NS3 protein of GBV-B, and the presence of GBV-B RNA in serum.
  • the tamarins were housed at the Southwest Regional Primate Research Center at the Southwest Foundation for Biomedical Research. They were cared for by members of the Department of Laboratory Animal Medicine of the Southwest Foundation for Biomedical Research in accordance with the Guide for the Care and Use of laboratory Animals. All protocols were approved by the Institutional Animal Care and Use Committee (IACUC).
  • the cDNA product was quantified in a 5' exonuclease PCR (TaqMan®) assay using a primer-probe combination that recognized sequence in the GBV-B NS5A coding region.
  • the primers NS5ARPp and NS5AFPp (5'-CCAGTTCCGGGCAAGAA CT-3' (SEQ LD NO: 15), position 6844-6862) and probe (nts 6913-6938) were obtained from Fisher and Perkin-Elmer, respectively.
  • a synthetic RNA derived from the infectious clone was used as the standard for quantitation of the RNA.
  • RNAs containing only the NS5A mutation failed to replicate.
  • a chimeric RNA containing the NS5A mutation together with both 3TSTTR mutations (GB/UI -m2) replicated in a robust fashion at week 4 post- inoculation, reaching viremia levels in excess of 10 7 genome equivalents/ml (FIG. 12).
  • GBV-B (C9052U and C9065U) are involved in an efficient-replication phenotype of the domain m chimeric virus. These mutations appear to be useful for efficient replication both in vivo in intrahepatically inoculated animals and ex vivo in cultured primary hepatocytes.
  • compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of prefe ⁇ ed embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents, which are both chemically and physiologically related, may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

Abstract

The present invention relates generally to the fields of biochemistry, molecular biology, and virology. More particularly, it relates to the identification of GB virus B (GBV-B)/HCV chimeras. The invention involves nucleic acid constructs and compositions encoding GBV-B/HCV chimera, including at least part of a 5’ NTR derived from a HCV 5’ NTR. This construct, and chimeric versions of it, may be employed to study GBV-B and related hepatitis family members, such as hepatitis C virus. The invention thus includes methods of preparing GBV-B/HCV chimeric sequences, constructs, and viruses, as well as methods of employing these compositions.

Description

DESCRIPTION
CHIMERIC GB VIRUS B (GBV-B)
BACKGROUND OF THE INVENTION
The present application claims priority to U.S. Patent Application Serial Number 10/189,359 filed on July 3, 2002. The entire text of the above-referenced disclosures is herein incoφorated by reference.
A. Field of the Invention
The present invention relates generally to the fields of biochemistry, molecular biology, and virology. More particularly, it relates to the identification of 259 nucleotides of previously unrecognized sequence located at the 3' end of the GB virus B (GBV-B) genome.
B. Description of Related Art
Chronic hepatitis C is a major threat to the public health. Serologic surveys suggest that as many as 3.9 million Americans are chronically infected with the responsible virus, hepatitis C virus (HCV) (Alter, 1997). These individuals are at increased risk of developing progressive hepatic fibrosis leading to cirrhosis and loss of hepatocellular function, as well as hepatocellular carcinoma. The course of chronic hepatitis C is typically lengthy, often extending over decades, with insidious clinical progression usually occurring in the absence of symptoms. Nonetheless, liver disease due to HCV results in the death of 8,000-10,000 Americans annually, and chronic hepatitis C is the most common cause of liver transplantation within the U.S. Therefore, HCV is a major public health problem. However, therapy for chronic hepatitis
C is problematic. Recombinant interferon-α is approved for treatment of chronic hepatitis C (Consensus Development Panel, 1997). The benefit of interferon-α results primarily from its antiviral properties and its ability to inhibit production of virus by infected hepatocytes (Neumann et al, 1998). Nonetheless, even under optimal therapeutic regimens, the majority of patients with chronic hepatitis C fail to eliminate the virus or resolve their liver disease. Treatment failures are especially common in persons infected with genotype 1 HCV, unfortunately the most prevalent genotype in the U.S. Thus, there is an urgent need to better understand the virus and develop better treatment. Unfortunately, technical difficulties in working with HCV have made it necessary to use infectious surrogate viruses in efforts to develop treatments and vaccines for HCV.
Scientists' efforts to better understand HCV and to develop new drugs for treatment of hepatitis C have been stymied by two overwhelming technical deficiencies: first, the nonexistence of a high permissive cell line that supports replication of the virus and second, the absence of a permissive animal species other than chimpanzees, which are endangered and therefore available on a limited basis.
Presently, those who are working on HCV treatment and prevention are employing an infectious chimeric virus of sindbis and HCV and/or an infectious clone of pestiviruses as surrogate virus models in HCV drug discovery efforts, due to the above technical difficulties of working with HCV. Alternatively, they are using isolated proteins or RNA segments of HCV for biochemical and structural studies. This approach precludes functional studies of virus replication and its inhibition.
GBV-B is a hepatotropic flavivirus that has a unique phylogenetic relationship to human HCV and strong potential to serve as a surrogate virus in drug discovery efforts related to hepatitis C antiviral drug development. GBV-B causes acute hepatitis in experimentally infected tamarins (Simons et al, 1995; Schlauder et al, 1995; Karayiannis et al, 1989) and can serve as a surrogate virus for HCV in drug discovery efforts (due to technical difficulties in working with HCV). GBV-B virus is much closer in sequence and biological properties than the above- described models. It will be easier to make biologically relevant chimeras between HCV and
GBV-B than by using more distantly related viruses. GBV-B is hepatotropic (as is HCV), whereas the viruses used in these competing technologies are not. In view of the above, an infectious clone of GBV-B would be useful to those working on HCV treatment and prevention.
Unfortunately, the use of GBV-B as a surrogate or model for HCV has not been possible in the past, because no infectious molecular clone of GBV-B virus genome could be prepared. It is now known that this obstacle was encountered because the GBV-B genome was believed to be 259 nucleotides shorter than its actual length (Muerhoff et al, 1995; Simons et al, 1995). Others, previous to the inventors, had failed to realize that the 3' sequence of GBV-B was missing from the prior sequences. Without this 3' sequence, it is not possible to prepare an infectious GBV-B molecular clone. SUMMARY OF THE INVENTION
As discussed above, an infectious molecular clone of GBV-B would be very useful for the development of HCV preventative and therapeutic treatments. The construction of an infectious molecular clone of this virus will require the newly determined 3' sequence to be included in order for the clone to be viable. The inventors have elucidated the previously unrecognized 3' terminal sequence of GBV-B (SEQ ID NO:l). This sequence has been reproducibly recovered from tamarin serum containing GBV-B RNA, in RT-PCR protocols using several different primer sets, and as a fusion with previously reported 5' GBV-B sequences.
The newly identified 3' sequence is not included in published reports of the GBV-B sequence, nor described in patents relating to the original identification of the viral sequence (see
U.S. Patent No. 5,807,670 and references therein).
The invention has utility in that the inclusion of the sequence will be necessary for construction of an infectious molecular GBV-B clone. Such clones clearly have the potential to be constructed as chimeras including relevant hepatitis C virus sequences in lieu of the homologous GBV-B sequence, providing unique tools for drug discovery efforts. A full-length molecular clone of GBV-was constructed, as described in later sections of this specification.
GBV-B can be used as a model for HCV, and the GBV-B genome can be used as the acceptor molecule in the construction of chimeric viral RNAs containing sequences of both HCV and GBV-B. Such studies will allow one to investigate the mechanisms for the different biological properties of these viruses and to discover and investigate potential inhibitors of specific HCV activities (e.g., proteinase) required for HCV replication. However, all this work is dependent upon construction of an infectious clone of GBV-B, which is itself dependent on the incorporation of the correct 3' terminal nucleotide sequence within this clone. GBV-B has unique advantages over HCV in terms of its ability to replicate and cause liver disease in tamarins, which present fewer restrictions to research than chimpanzees, the only nonhuman primate species known to be permissive for HCV.
An infectious molecular clone of GBV-B is expected to have utility in liver-specific gene expression or in gene therapy. This application might be enhanced by the inclusion of HCV genomic sequence in the form of a GBV-B/HCV chimera. Further, an infectious GBV-B/HCV chimera expressing HCV envelope proteins can have utility as a vaccine immunogen for hepatitis
C.
A full-length cDNA copy of the GBV-B genome was constructed to contain the newly identified 3' terminal sequences. RNA transcribed from this cDNA copy of the genome would be infectious when inoculated into the liver of a GBV-B permissive tamarin, giving rise to rescued GBV-B virus particles. A chimeric molecule would then be constructed from this infectious GBV-B clone in which the HCV NS3 proteinase or proteinase/helicase sequence (or other relevant HCV sequences of interest in drug discovery efforts) would be placed in frame in lieu of the homologous GBV-B sequence, and this chimeric cDNA would be used to generate infectious GBV-B/HCV chimeric viruses by intrahepatic inoculation of synthetic RNA in tamarins. Published studies indicate that the GBV-B and HCV proteinases have closely related substrate recognition and cleavage properties, making such chimeras highly likely to be viable. These newly generated chimeric GBV-B/HCV viruses could be used in preclinical testing of candidate HCV NS3 proteinase inliibitors.
Therefore, the present invention encompasses an isolated polynucleotide encoding a 3' sequence of the GBV-B genome. The polynucleotide may include the sequence identified as SEQ ID NO: 1. It is contemplated that the polynucleotide may be a DNA molecule or it can be an RNA molecule. It is further contemplated that expression constructs may contain a polynucleotide that has a stretch of contiguous nucleotides from SEQ ID NO:l and/or SEQ ID NO:2, for example, lengths of 50, 100, 150, 250, 500, 1000, 5000, as well as the entire length of SEQ ID NO:l or 2, are considered appropriate. Such polynucleotides may also be contained in other constructs of the invention or be used in the methods of the invention. Polynucleotides employing sequences from SEQ ID NO:l may alternatively contain sequences from SEQ LD NO:2 in the constructs and methods of the present invention. Specific mutations discussed in the Examples and/or figures are included as embodiments of the invention.
The invention is also understood as covering a viral expression construct that includes a polynucleotide encoding a 3' sequence of the GBV-B genome. This expression construct is further understood to contain the sequence identified as SEQ ID NO:l. The present invention contemplates the expression construct as a plasmid or as a virus. Furthermore, the expression construct can express GBV-B sequences; alternatively it may express sequences from a chimeric GBV-B HCV virus.
The identification and isolation of a 3' sequence of GBV-B additionally provides a method of producing a virus, particularly a full-length virus, by introducing into a host cell a viral expression construct containing a polynucleotide encoding a 3' sequence of GBV-B and by culturing the host cell under conditions permitting production of a virus from the construct. This method can be practiced using a prokaryotic cell as a host cell, or by using a eukaryotic cell as a host cell. Furthermore, the eukaryotic cell can be located within an animal. A method of producing virus according to the claimed invention can also be employed using a polynucleotide that contains synthetic RNA and or synthetic DNA. Moreover, a step can be added to the method by also isolating any virus produced from the host cell. The virus can then be purified to homogeneity. hi further embodiments, the present invention encompasses an oligonucleotide between about 10 and about 259 consecutive bases of SEQ ID NO:l. This oligonucleotide is contemplated to be about 15 bases in length, about 20 bases in length, about 25 bases in length, about 30 bases in length, about 35 bases in length, about 50 bases in length, about 100 bases in length, about 150 bases in length, about 200 bases in length, or about 259 bases in length. Additional examples of the claimed invention include a method for identifying a compound active against a viral infection by providing a virus expressed from a viral construct containing a 3' sequence of a GBV-B virus, by contacting the virus with a candidate substance; and by comparing the infectious ability of the virus in the presence of the candidate substance with the infectious ability of the virus in a similar system in the absence of the candidate substance. It is contemplated that the invention can be practiced using GBV-B virus or a GBV- B HCV chimera. Infectious ability of the virus is comparable to infectivity of the virus. Infectivity can be evaluated by evaluating or assessing the ability of a host cell to become infected by the virus or by the ability of the virus to replicate in the cell. However, it may also be evaluated by virus production, virus infectivity, viral load, or phenotype of the cell such as signs of CPE.
The present invention can also be understood to provide a compound active against a viral infection identified by providing a virus expressed from a viral construct containing a 3' sequence of a GBV-B virus; contacting the virus with a candidate substance; and comparing the infectious ability of the virus in the presence of the candidate substance with the infectious ability of the virus in a similar system in the absence of the candidate substance. In some embodiments an active compound is identified using a GBV-B virus, while in other embodiments an active compound is identified using a GBV-B/HCV chimera. Compounds considered active against viral infection would include, but are not limited to, those compounds that inhibit viral infection or that expedite clearance of the virus. In various embodiments of the invention, a GBV-B polynucleotide may encode a GBV-
B/HCV chimera that includes at least part of a 5' NTR sequence derived from a HCV 5' NTR. The 5' NTR may comprise at least one domain, i.e., domain I, JJ, m and or IV derived from the HCV in some embodiments. In certain embodiments, the GVB-B/HCV chimera may include at least domain III of the 5' NTR derived from the 5 'NTR of HCV. In yet other embodiments the infectious GBV-B clone may comprise domain III of the 5' NTR of HCV, which may or may not include one or more structural or non-structural genes of HCV also incoφorated into the chimeric virus. The portions of the 5' NTR of the GVB-B/HCV chimeras will generally be replaced by analogous sequences from the 5' NTR of HCV. It will be understood that the portions or parts of the 5' NTR of GBV-B that may be replaced include all or part of domain I (including sub-region la and lb of GBV-B), domain π, domain HI, domain IV, or any combination thereof. Any combination of 5' NTR domains of GBV-B may be replaced with an analogous region of HCV. In certain embodiments, the replacement of a GBV-B region may be accompanied by the deletion of the 5' NTR GBV-B domain lb region. In addition, any one, two, or three of the 5' NTR domains of GBV-B may be replaced in any combination with analogous sequences from HCV.
In further embodiments of the invention, a polynucleotide encoding a GBV-B/HCV chimera including a 5' NTR domain III sequence derived from a HCV 5' NTR may be propagated in vivo, in particular, in the liver of an appropriate host.
Various other embodiments may include isolated polynucleotides comprising a chimeric GBV-B genome, wherein at least part, but not all of a 5' NTR sequence is derived from a HCV 5' NTR. The polynucleotides may be synthetic RNA, RNA, DNA or the like. Some embodiments include one or more virus, one or more hepatotropic virus, and/or one or more viral expression constructs comprising a chimeric GBV-B polynucleotides including at least a part of the 5' NTR sequence is derived from a HCV 5' NTR.
Methods of producing a chimeric GBV-B virus encoding at least part of a 5' NTR sequence derived from a HCV 5' NTR sequence comprising introducing into a host cell a viral expression construct comprising a chimeric GBV-B polynucleotide encoding at least part of a 5' NTR sequence derived from a HCV 5' NTR sequence and culturing said host cell under conditions permitting production of a virus from said construct are contemplated. The method may use a host cell that is a eukaryotic cell and the host cell may be in an animal. The method may further include the step of isolating virus from said host cell and in particular purify the virus to homogeneity.
In addition, methods for identifying a compound active against a viral infection comprising are contemplated. The methods may include providing a virus expressed from a viral construct comprising at least part of a 5' NTR derived from a HCV 5' NTR, as described herein; contacting said virus with a candidate substance; and comparing the infectious ability of the virus in the presence of said candidate substance with the infectious ability of the virus in a similar system in the absence of said candidate substance. Each of the embodiments may use or include any of the 5' NTR chimeras described herein. Other embodiments of the invention may include a compound active against a viral infection identified according to the method described above.
Furthermore, additional methods include providing or administering to a subject or patient in need of a compound active against viral infection the compound active against a viral infection, particularly a flavivirus such as HCV infection.. The patient may have been diagnosed with HCV infection or be at risk for HCV infection, or another flavivirus infection. Alternatively, a subject or patient may be administered a vaccine that immunizes the subject or patient with respect to a flavivirus infection, such as HCV. The invention includes such methods.
It is specifically contemplated that any limitation discussed with respect to one embodiment of the invention may apply to any other embodiment of the invention. Furthermore, any composition of the invention may be used in any method of the invention, and any method of the invention may be used to produce or to utilize any composition of the invention.
The use of the term "or" in the claims is used to mean "and/or" unless explicitly indicated to refer to alternatives only or the alternative are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and "and/or." Throughout this application, the term "about" is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value.
As used herein the specification, "a" or "an" may mean one or more, unless clearly indicated otherwise. As used herein in the claim(s), when used in conjunction with the word "comprising," the words "a" or "an" may mean one or more than one. As used herein "another" may mean at least a second or more.
Other objects, features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. BRIEF DESCRIPTION OF THE DRAWINGS
The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present invention. The invention may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.
FIG. 1. Predicted secondary RNA structure at the 3' end of the novel 3' GBV-B sequence. The structure shown is that predicted for the 3' terminal 108 nucleotides of the GBV-B genomic RNA by the MFOLD 3.0 computer program of Zucker and Turner, and has an initial free energy of -48 kcal/mole. The predicted structure contains 3 stem-loops, numbered from the 3' end of the genome and labeled SL-1, SL-2, and SL-3. The structure of SL-1 is highly probable, given its terminal position within the genome. Alternative foldings of SL-2 and SL-3 are possible.
FIG. 2. Illustration of the organization of exemplary chimeric cDNAs containing HCV 5' nontranslated (5' NTR) RNA sequences within the background of a modified (MM restriction- site containing) GBV-B genetic background.
FIG. 3 A and 3B. In vitro translation of RLuc reporter transcripts (shown schematically in
FIG. 3 A) in which the initiation of translation of RLuc is dependent upon the upstream viral or chimeric IRES. FIG. 3B, top panel, shows the SDS-PAGE gel on which the products of translation were separated. FIG. 3B, bottom panel, are the results of the Phosphorlmager analysis. Mock = no RNA transcript in the translation mix; and m = size markers.
FIG. 4. Shows the translational activity of RLuc reporter transcripts (normalized to FLuc control transcript) in examples of transfected primary tamarin hepatocytes.
FIG. 5. Shows the replication of an exemplary iπHC chimeric RNA following intrahepatic inoculation of synthetic RNA in a GBV-B naive tamarin (S. mystax). FIG. 6. Shows the replication of an exemplary UI chimeπc virus following intravenous inoculation of a GBV-B naϊve tamarin (S. mystax) with virus present in the serum of T16444 14 weeks after it had been inoculated with synthetic RNA.
FIG. 7. Shows the sequence differences between chimeric viral RNAs recovered from animals T16444 and T16451, and the parental synthetic mHC RNA. FIG. 8 Replication of the GB/LUHC chimeric virus in primary cultures of tamarin hepatocytes. Viral titers were determined both in infected-cell extracts ("cytosol") and in culture supernatants ("secreted") by a quantitative real-time RT-PCR assay. FIG. 9 . Replication of the
Figure imgf000010_0001
chimeric RNA following intrahepatic inoculation of
RNA in a GBV-B naive tamarin. GBV-B viremia was determined in a quantitative real-time
TaqMan® RT-PCR assay targeting GBV-B NS5A sequence (see text for details). Serum ALT activities were monitored on a biweekly basis. Antibodies to NS3 were assayed by an ELISA using recombinant GBV-B NS3 as antigen.
FIG. 10 Mutations identified in the virus isolated at week 14 p.i. in an animal (T 16444) inoculated with the GB/IJJ chimeric RNA.
FIG. 11 Replication of the GB/IJI chimeπc virus following intravenous inoculation m a naϊve tamarin. FIG. 12 Schematic representation of the RNA transcripts of mutated cDNAs derived from the parental GB/]UHC construct and their in vivo replication capacity following intrahepatic inoculation into GBV-B naϊve tamarins.
FIG. 13A and 13B. Replication of the GB/HIHC-m3 and GB/rπHC-m4 chimeric RNAs following intrahepatic inoculation of RNA in individual naϊve tamarins. See Fig. 2 for details relative to monitoring of the infection.
FIG. 14A and 14B Predicted models of the secondary structures of the 59NTRs of GBV-B (FIG. 14A) and HCV (FIG. 14B). (FIG. 14A) Structure proposed for the GBV-B 59NTR by Honda et al. (1996 and 1999), each of which is incoφorated herein by reference. Major predicted structural domains are labeled I to TV, while individual stem-loops are labeled la and are analogous to similarly labeled structures in the HCV 59NTR. Base-pair interactions involving the loop sequence of stem-loop JJIf that are predicted to result in a putative RNA pseudoknot are drawn as solid lines. Lightly shaded boxes represent AUG triplets located within the 59NTR, while the solid black box represents the polyprotein translation initiation site. The open box indicates a helical segment at the base of domain It, and open circles represent individual nucleotides that were subjected to site-directed mutagenesis in the studies described here. Unpaired bases within domain LT that are conserved in domain π of the HCV structure are shown in boldface type. The 59 and 39 limits of the GBV-B IRES, as determined in this study, are indicated by the arrows. (FIG. 14B) Structure proposed for the HCV 59NTR (Deinhardt et al, 1967, Honda et al, 1996 and 1999, each of which is incoφorated herein by reference). There are numerous similarities with the GBV-B structure but no predicted stem-loop structures analogous to the lb, Hb3 and he stem-loops of GBV-B. DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
A. GVB-B Virus
The GBV-B genome structure is very similar to hepatitis C and these viruses share approximately 25% nucleotide identity (Simons et al, 1995; Muerhoff et al, 1995). As indicated above, this makes GBV-B more closely related to HCV than any other known virus. GBV-B genomic RNA is about 9.5 kb in length (Muerhoff et al, 1995) with a structured 5' noncoding region that contains an IRES that shares many structural features with the HCV IRES (Honda et al, 1996; Rijnbrand et al, 1999, each of which is incoφorated herein by reference). As in HCV, this IRES drives the cap-independent translation of a long open reading frame. The polyprotein expressed from this reading frame appears to be organized identically to that of HCV, and processed to generate proteins with functions similar to those of HCV (Muerhoff et al, 1995). In fact, the major serine proteinases of these viruses (NS3) have been shown to have similar cleavage specificities (Scarselli et al, 1997). Finally, like HCV and distinct from the pestiviruses, the genomic RNA of GBV-B has a poly(U) tract located near its 3" terminus (Simons et al, 1995; Muerhoff et al, 1995). In addition, unreported sequences located at the extreme 3' end of the genome have been identified. This work indicates that the GBV-B RNA, like that of HCV (HCV (Tanaka et al, 1995; Kolykhalov et al, 1996), terminates in a lengthy run of heterogeneous bases (310 nts in GBV-B) possessing a readily apparent secondary structure
B. Nucleic Acids The present invention provides a nucleic acid sequence encoding a 3' sequence of the
GBV-B genome (SEQ LD NO:l).
It should be clear that the present invention is not limited to the specific nucleic acids disclosed herein. As discussed below, a "3' sequence of the GBV-B genome" may contain a variety of different bases and yet still be functionally indistinguishable from the sequences disclosed herein. Such functionally indistinguishable sequences are likely to maintain the basic structure depicted in FIGs. 1 and 2, which may be used to guide the prediction of viable nucleotide substitutions.
1. Polynucleotides Encoding the 3' Sequence of the GBV-B Genome
A 3' sequence of the GBV-B genome disclosed in SEQ ID NO:l is one aspect of the present invention. Nucleic acids according to the present invention may encode the 3' sequence of the GBV-B genome set forth in SEQ ID NO:l, the entire GBV-B genome, or any other fragment of a 3' sequence of the GBV-B genome set forth herein. The nucleic acid may be derived from genomic RNA as cDNA, i.e., cloned directly from the genome of GBV-B. cDNA may also be assembled from synthetic oligonucleotide segments.
It also is contemplated that a 3' sequence of the GBV-B genome may be represented by natural variants that have slightly different nucleic acid sequences but, nonetheless, maintain the same general structure (see FIGs. 1 and 2) and perform the same function in RNA replication.
As used in this application, the term "a nucleic acid encoding a 3' sequence of the GBV-B genome" refers to a nucleic acid molecule that may be isolated free of total viral nucleic acid. In preferred embodiments, the invention concerns nucleic acid sequences essentially as set forth in SEQ ID NO:l, SEQ LO NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ LD NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ LD NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:l l, and SEQ ID NO:12. The term "as set forth in SEQ ID NO:l" means that the nucleic acid sequence substantially corresponds to a portion of SEQ ID NO:l. It is contemplated that the techniques and methods described in this disclosure may apply to any of the sequences contained herein, including SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ LD NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:ll, and SEQ ID NO:12.
Allowing for the degeneracy of the genetic code, sequences that have at least about 50%, usually at least about 60%, more usually about 70%, most usually about 80%, preferably at least about 90% and most preferably about 95% of nucleotides that are identical to the nucleotides of SEQ ID NO:l will be sequences that are "as set forth in SEQ ID NO:l." Sequences that are essentially the same as those set forth in SEQ ID NO:l may also be functionally defined as sequences that are capable of hybridizing to a nucleic acid segment containing the complement of SEQ ID NO:l under standard conditions.
The nucleic acid segments and polynucleotides of the present invention include those encoding biologically functional equivalent 3' sequences of the GBV-B genome. Changes designed by man may be introduced through the application of site-directed mutagenesis techniques or maybe introduced randomly and screened later for the desired function, as described below.
3' sequence of the GBV-B genome sequences also are provided. Each of the foregoing is included within all aspects of the following description. The present invention concerns cDNA segments reverse transcribed from GBV-B genomic RNA (referred to as "DNA"). As used herein, the term "polynucleotide" refers to an RNA or DNA molecule that may be isolated free of other RNA or DNA of a particular species. "Isolated substantially away from other coding sequences" means that the 3' sequence of the GBV-B genome forms the significant part of the RNA or DNA segment and that the segment does not contain large portions of naturally-occurring coding RNA or DNA, such as large fragments or other functional genes or cDNA noncoding regions. Of course, this refers to the polynucleotide as originally isolated, and does not exclude genes or coding regions later added to the it by the hand of man.
In certain other embodiments, the invention concerns isolated DNA segments (cDNA segments reverse transcribed from GVB-B genomic RNA) and recombinant vectors that include within their sequence a nucleic acid sequence essentially as set forth in SEQ LD NO:l. The term "essentially as set forth in SEQ ID NO:l" is used in the same sense as described above.
It also will be understood that nucleic acid sequences may include additional residues, such as additional 5' or 3' sequences, and still be essentially as set forth in one of the sequences disclosed herein, so long as the sequence meets the criteria set forth above, including the maintenance of biological activity. The addition of terminal sequences particularly applies to nucleic acid sequences that may, for example, include additional various non-coding sequences flanking either of the 5' or 3' portions of the coding region, which are known to occur within viral genomes.
Sequences that are essentially the same as those set forth in SEQ ID NO:l also may be functionally defined as sequences that are capable of hybridizing to a nucleic acid segment containing the complement of SEQ ID NO:l under relatively stringent conditions. Suitable relatively stringent hybridization conditions will be well known to those of skill in the art.
The nucleic acid segments of the present invention, regardless of the length of the coding sequence itself, may be combined with other RNA or DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol.
For example, nucleic acid fragments may be prepared that include a short contiguous stretch identical to or complementary to SEQ ID NO:l, such as about 15-24 or about 25-34 nucleotides and that are up to about 259 nucleotides being prefeπed in certain cases. Other stretches of contiguous sequence that may be identical or complementary to any of the sequences disclosed herein, including the SEQ ID NOS. include the following ranges of nucleotides: 50- 9,399, 100-9,000, 150-8,000, 200-7,000, 250-6,000, 300-5,000, 350-4,000, 400-3,000, 450- 2,000, 500-1000. RNA and DNA segments with total lengths of about 1,000, about 500, about 200, about 100 and about 50 base pairs in length (including all intermediate lengths) are also contemplated to be useful. In a non-limiting example, one or more nucleic acid constructs may be prepared that include a contiguous stretch of nucleotides identical to or complementary to SEQ ID NO:l, SEQ ID NO:2, SEQ LD NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO:ll, or SEQ ID NO:12. Such a stretch of nucleotides, or a nucleic acid construct, may be about, or at least about, 3, about 4, about 5, about 6, about 7, about 8„ about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39 about 40, about 45, about 50, about 55, about 60, about 65, about 70, about 75, about 80, about 85, about 90, about 95, about 100, about 105, about 110, about 115, about 120, about 125, about 130, about 135, about 140, about 145, about 150, about 155, about 160, about 165, about 170, about 175, about 180, about 185, about 190, about 195, about 200, about 210, about 220, about 230, about 240, about 250, about 260, about 270, about 280, about 290, about 300, about 310, about 320, about 330, about 340, about 350, about 360, about 370, about 380, about 390, about 400, about 410, about 420, about 430, about 440, about 450, about 460, about 470, about 480, about 490, about 500, about 510, about 520, about 530, about 540, about 550, about 560, about 570, about 580, about 590, about 600, about 610, about 618, about 650, about 700, about 750, about 1,000, about 2,000, about 3,000, about 4,000, about 5,000, about 6,000, about 7,000, about 8,000, about 9,000, about 9,100, about 9,200, about 9,300, about 9,399, about 9,400, about 9,500, about 9,600, about 9,700, about 9,800, about 9,900, about 10,000, about 15,000, about 20,000, about 30,000, about 50,000, about 100,000, about 250,000, about 500,000, about 750,000, to about 1,000,000 nucleotides in length, as well as constructs of greater size, up to and including chromosomal sizes (including all intermediate lengths and intermediate ranges), given the advent of nucleic acids constructs such as a yeast artificial chromosome are known to those of ordinary skill in the art. It will be readily understood that "intermediate lengths," in these contexts means any length between the quoted ranges, such as 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, etc.; 100, 101, 102, 103, etc.; 150, 151, 152, 153, etc.; including all integers through the 200-500; 500-1,000; 1,000-2,000; ranges, up to and including sequences of about 1,001, 1,250, 1,500, and the like.
In certain embodiments, GBV-B polynucleotides include chimeric GBV-B poynucleotides. Chimeric GBV-B polynucleotides may include GBV-B/HCV chimeras. GBV- B/HCV chimeras include, but are not limited to GBV-B polynucleotides in which portions of the GBV-B virus have been replaced with or mutated to resemble analogous sequences from the HCV virus. A GVB-B chimera may comprise all or part of the 5 'NTR region of a HCV virus. The polynucleotide may comprise domain I, II. Ill and/or IV of the 5' NTR derived from a HCV 5 'NTR. The polynucleotide may have the 5' NTR domain lb of GBV-B is deleted. The polynucleotide may comprise domain II and domain III of the 5' NTR derived from a HCV 5 'NTR. The polynucleotide may comprise domain II and domain IN of the 5' ΝTR derived from a HCV 5 'ΝTR. The polynucleotide may compirse domain III and domain IV of the 5' ΝTR derived from a HCV 5 'ΝTR. The polynucleotide may comprise domain II, domain III and domain IV of the 5' ΝTR derived from a HCV 5 'ΝTR. Domain lb of GBV-B may or may not be deleted form the polynucleotide. The polynucleotide may be DΝA or RΝA. The polynucleotide may further comprising at least part of a structural and/or nonstructural protein coding region of derived from HCV. It will be understood that the term "derived" indicates the origin of the sequence. Thus, the recited domains can be obtained from HCV.
In other embodiments a HCV/GBV-B chimera is contemplated. A HCV/GB V-B chimera will comprise a virus where one or more portions of the HCV virus have been replaced with or mutated to resemble the analogous GBV-B portions of the GBV-B virus.
The various probes and primers designed around the disclosed nucleotide sequences of the present invention may be of any length. By assigning numeric values to a sequence, for example, the first residue is 1, the second residue is 2, etc., an algorithm defining all primers can be proposed: n to n + y where n is an integer from 1 to the last number of the sequence and y is the length of the primer minus one, where n + y does not exceed the last number of the sequence. Thus, for a 20-mer, the probes coπespond to bases 1 to 20, 2 to 21, 3 to 22 ... and so on. For a 30-mer, the probes coπespond to bases 1 to 30, 2 to 31, 3 to 32 ... and so on. For a 35-mer, the probes coπespond to bases 1 to 35, 2 to 36, 3 to 37 ... and so on.
2. Oligonucleotide Probes and Primers
Naturally, the present invention also encompasses RNA and DNA segments that are complementary, or essentially complementary, to the sequence set forth in SEQ ID NO:l. Nucleic acid sequences that are "complementary" are those that are capable of base-pairing according to the standard Watson-Crick complementary rules. As used herein, the term "complementary sequences" means nucleic acid sequences that are substantially complementary, as may be assessed by the same nucleotide comparison set forth above, or as defined as being capable of hybridizing to the nucleic acid segment of SEQ ID NO. 1 under relatively stringent conditions such as those described herein. Such sequences may encode the entire 3' sequence of the GBV-B genome or functional or non-functional fragments thereof.
Alternatively, the hybridizing segments may be shorter oligonucleotides. Sequences of 17 bases long should occur only once in the human genome and, therefore, suffice to specify a unique target sequence. Although shorter oligomers are easier to make and increase in vivo accessibility, numerous other factors are involved in determining the specificity of hybridization. Both binding affinity and sequence specificity of an oligonucleotide to its complementary target increases with increasing length. It is contemplated that exemplary oligonucleotides of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 125, 150, 175, 200, 225 or more base pairs will be used, although others are contemplated. Longer polynucleotides encoding 250, 500, 1000, 1212, 1500, 2000, 2500, 3000 or 3431 bases and longer are contemplated as well. Such oligonucleotides will find use, for example, as probes in Southern and Northern blots and as primers in amplification reactions.
Suitable hybridization conditions will be well known to those of skill in the art. In certain applications, for example, substitution of amino acids by site-directed mutagenesis, it is appreciated that lower stringency conditions are required. Under these conditions, hybridization may occur even though the sequences of probe and target strand are not perfectly complementary but are mismatched at one or more positions. Conditions may be rendered less stringent by increasing salt concentration and decreasing temperature. For example, a medium stringency condition could be provided by about 0.1 to 0.25 M NaCl at temperatures of about 37°C to about 55°C, while a low stringency condition could be provided by about 0.15 M to about 0.9 M salt, at temperatures ranging from about 20°C to about 55°C. Thus, hybridization conditions can be readily manipulated and thus will generally be a method of choice depending on the desired results.
In other embodiments, hybridization may be achieved under conditions of, for example, 50 mM Tris-HCl (pH 8.3), 75 mM KCl, 3 mM MgCl2, 10 mM dithiothreitol, at temperatures between approximately 20°C to about 37°C. Other hybridization conditions utilized could include approximately 10 mM Tris-HCl (pH 8.3), 50 mM KCl, 1.5 mM MgCl2, at temperatures ranging from approximately 40°C to about 72°C. Formamide and SDS also may be used to alter the hybridization conditions.
One method of using probes and primers of the present invention is in the search for other viral sequences related to GBV-B or, more particularly, homologs of the GBV-B sequence. By varying the stringency of hybridization, and the region of the probe, different degrees of homology may be discovered.
Another way of exploiting probes and primers of the present invention is in site-directed, or site-specific, mutagenesis. The technique provides a ready ability to prepare and test sequence variants, incoφorating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into complementary DNA. Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences that encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. Typically, a primer of about 17 to 25 nucleotides in length is prefeπed, with about 5 to 10 residues on both sides of the junction of the sequence being altered.
The technique typically employs a bacteriophage vector that exists in both a single stranded and double stranded form. Typical vectors useful in site-directed mutagenesis include vectors such as the Ml 3 phage. These phage vectors are commercially available and their use is generally well known to those skilled in the art. Double stranded plasmids are also routinely employed in site directed mutagenesis, which eliminates the step of transferring the gene of interest from a phage to a plasmid.
In general, site-directed mutagenesis is performed by first obtaining a single-stranded vector, or melting of two strands of a double stranded vector which includes within its sequence a DNA sequence encoding the desired protein. An oligonucleotide primer bearing the desired mutated sequence is synthetically prepared. This primer is then annealed with the single-stranded DNA preparation, taking into account the degree of mismatch when selecting hybridization conditions, and subjected to DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand. Thus, a heteroduplex is formed wherein one strand encodes the original non-mutated sequence and the second strand bears the desired mutation. This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected that include recombinant vectors bearing the mutated sequence arrangement. There are newer and simpler site-directed mutagenesis techniques that can also be employed for this pvurpose. These include procedures marketed in kit form that are readily available to one of ordinary skill in the art.
The preparation of sequence variants of the selected gene using site-directed mutagenesis is provided as a means of producing potentially useful species and is not meant to be limiting, as there are other ways in which sequence variants of genes may be obtained. For example, recombinant vectors encoding the desired gene may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants.
3. Antisense Constructs hi certain embodiments of the invention, the use of antisense constructs of the 3' sequence of the GBV-B genome is contemplated.
Antisense methodology takes advantage of the fact that nucleic acids tend to pair with "complementary" sequences. By complementary, it is meant that polynucleotides are those which are capable of base-pairing according to the standard Watson-Crick complementarity rules. That is, the larger purines will base pair with the smaller pyrimidines to form combinations of guanine paired with cytosine (G:C) and adenine paired with either thymine (A:T) in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA. Inclusion of less common bases such as inosine, 5-methylcytosine, 6-methyladenine, hypoxanthine and others in hybridizing sequences does not interfere with pairing.
Targeting double-stranded (ds) DNA with polynucleotides leads to triple-helix formation; targeting RNA will lead to double-helix formation. Antisense polynucleotides, when introduced into a target cell, specifically bind to their target polynucleotide and interfere with transcription, RNA processing, transport, translation and/or stability. Antisense RNA constructs, or DNA encoding such antisense RNAs, may be employed to inhibit gene transcription or translation or both within a host cell, either in vitro or in vivo, such as within a host animal, including a human subject.
Antisense constructs could be used to block early steps in the replication of GBV-B and related viruses, by annealing to 3' terminal sequences and blocking their role in negative-strand initiation.
As stated above, "complementary" or "antisense" means polynucleotide sequences that are substantially complementary over their entire length and have very few base mismatches. For example, sequences of 15 bases in length may be termed complementary when they have complementary nucleotides at 13 or 14 positions. Naturally, sequences which are completely complementary will be sequences which are entirely complementary throughout their entire length and have no base mismatches. Other sequences with lower degrees of homology also are contemplated. For example, an antisense construct which has limited regions of high homology but also contains a non-homologous region (e.g., ribozyme; see below) could be designed. These molecules, though having less than 50% homology, would bind to target sequences under appropriate conditions.
4. Amplification and PCR™
The present invention utilizes amplification techniques in a number of its embodiments. Nucleic acids used as a template for amplification are isolated from cells contained in the biological sample, according to standard methodologies (Sambrook et al, 1989). The nucleic acid may be genomic DNA or RNA or fractionated or whole cell RNA. Where RNA is used, it may be desired to convert the RNA to a complementary DNA using reverse transcriptase (RT). hi one embodiment, the RNA is genomic RNA and is used directly as the template for amplification. In others, genomic RNA is first converted to a complementary DNA sequence (cDNA) and this product is amplified according to protocols described below. Pairs of primers that selectively hybridize to nucleic acids coπesponding to GBV-B sequences are contacted with the isolated nucleic acid under conditions that permit selective hybridization. The term "primer," as defined herein, is meant to encompass any nucleic acid that is capable of priming the synthesis of a nascent nucleic acid in a template-dependent process. Typically, primers are oligonucleotides from ten to twenty base pairs in length, but longer sequences can be employed. Primers may be provided in double-stranded or single-stranded form, although the single-stranded form is prefeπed.
Once hybridized, the nucleic acid:primer complex is contacted with one or more enzymes that facilitate template-dependent nucleic acid synthesis. Multiple rounds of amplification, also refeπed to as "cycles," are conducted until a sufficient amount of amplification product is produced.
Next, the amplification product is detected. In certain applications, the detection may be performed by visual means. Alternatively, the detection may involve indirect identification of the product via chemiluminescence, radioactive scintigraphy of incoφorated radiolabel or fluorescent label or even via a system using electrical or thermal impulse signals. A number of template dependent processes are available to amplify the marker sequences present in a given template sample. One of the best known amplification methods is the polymerase chain reaction (refeπed to as PCR™) which is described in detail in U.S. Patent Nos. 4,683,195, 4,683,202 and 4,800,159, and each incoφorated herein by reference in entirety. Briefly, in PCR™, two or more primer sequences are prepared that are complementary to regions on opposite complementary strands of the marker sequence. An excess of deoxynucleoside triphosphates are added to a reaction mixture along with a DNA polymerase, e.g., Taq polymerase. If the marker sequence is present in a sample, the primers will bind to the marker and the polymerase will cause the primers to be extended along the marker sequence by adding on nucleotides. By raising and lowering the temperature of the reaction mixture, the extended primers will dissociate from the marker to form reaction products, excess primers will bind to the marker and to the reaction products and the process is repeated.
A reverse transcriptase PCR™ amplification procedure may be performed in order to quantify the amount of mRNA amplified. Methods of reverse transcribing RNA into cDNA are well known and described in Sambrook et al, 1989. Alternative methods for reverse transcription utilize thermostable, RNA-dependent DNA polymerases. These methods are described in WO 90/07641, filed December 21, 1990, incoφorated herein by reference. Polymerase chain reaction methodologies are well known in the art. Another method for amplification is the ligase chain reaction ("LCR"), disclosed in EPA
No. 320 308, incoφorated herein by reference in its entirety. U.S. Patent 4,883,750 describes a method similar to LCR for binding probe pairs to a target sequence.
Qbeta Replicase, described in PCT Application No. PCT/US 87/00880, incoφorated herein by reference, also may be used as still another amplification method in the present invention.
An isothermal amplification method, in which restriction endonucleases and ligases are used to achieve the amplification of target molecules that contain nucleotide 5'-[alpha-thio]-triphosphates in one strand of a restriction site also may be useful in the amplification of nucleic acids in the present invention. Strand Displacement Amplification (SDA) is another method of carrying out isothermal amplification of nucleic acids which involves multiple rounds of strand displacement and synthesis, i.e., nick translation. A similar method, called Repair Chain Reaction (RCR), involves annealing several probes throughout a region targeted for amplification, followed by a repair reaction in which only two of the four bases are present. The other two bases can be added as biotinylated derivatives for easy detection. A similar approach is used in SDA. Target specific sequences also can be detected using a cyclic probe reaction (CPR). hi CPR, a probe having 3' and 5' sequences of non-specific DNA and a middle sequence of specific RNA is hybridized to DNA that is present in a sample. Upon hybridization, the reaction is treated with RNase H, and the products of the probe identified as distinctive products that are released after digestion. The original template is annealed to another cycling probe and the reaction is repeated.
Still another amplification methods described in GB Application No. 2 202 328, and in PCT Application No. PCT/US89/01025, each of which is incoφorated herein by reference in its entirety, may be used in accordance with the present invention, hi the former application, "modified" primers are used in a PCR-like, template- and enzyme-dependent synthesis. The primers may be modified by labeling with a capture moiety (e.g., biotin) and/or a detector moiety (e.g., enzyme), hi the latter application, an excess of labeled probes are added to a sample. In the presence of the target sequence, the probe binds and is cleaved catalytically. After cleavage, the target sequence is released intact to be bound by excess probe. Cleavage of the labeled probe signals the presence of the target sequence.
Other nucleic acid amplification procedures include transcription-based amplification systems (TAS), including nucleic acid sequence based amplification (NASBA) and 3SR Gingeras et al, PCT Application WO 88/10315, incoφorated herein by reference. Davey et al, EPA No. 329 822 (incoφorated herein by reference in its entirety) disclose a nucleic acid amplification process involving cyclically synthesizing single-stranded RNA ("ssRNA"), ssDNA, and double-stranded DNA (dsDNA), which may be used in accordance with the present invention.
Miller et al, PCT Application WO 89/06700 (incoφorated herein by reference in its entirety) disclose a nucleic acid sequence amplification scheme based on the hybridization of a promoter/primer sequence to a target ssDNA followed by transcription of many RNA copies of the sequence. This scheme is not cyclic, i.e., new templates are not produced from the resultant RNA transcripts. Other amplification methods include "RACE" and "one-sided PCR" (Frohman, 1990 incoφorated by reference). Methods based on ligation of two (or more) oligonucleotides in the presence of nucleic acid having the sequence of the resulting "di-oligonucleotide", thereby amplifying the di-oligonucleotide, also maybe used in the amplification step of the present invention.
Following any amplification, it may be desirable to separate the amplification product from the template and the excess primer for the puφose of determining whether specific amplification has occuπed. In one embodiment, amplification products are separated by agarose, agarose-acrylamide or polyacrylamide gel electrophoresis using standard methods. See Sambrook et al, 1989. Alternatively, chromatographic techniques may be employed to effect separation. There are many kinds of chromatography that may be used in the present invention: adsoφtion, partition, ion-exchange and molecular sieve, and many specialized techniques for using them including column, paper, thin-layer and gas chromatography. Amplification products must be visualized in order to confirm amplification of the marker sequences. One typical visualization method involves staining of a gel with ethidium bromide and visualization under UV light. Alternatively, if the amplification products are integrally labeled with radio- or fluorometrically-labeled nucleotides, the amplification products can then be exposed to x-ray film or visualized under the appropriate stimulating spectra, following separation. hi one embodiment, visualization is achieved indirectly. Following separation of amplification products, a labeled nucleic acid probe is brought into contact with the amplified marker sequence. The probe preferably is conjugated to a chromophore but may be radiolabeled. hi another embodiment, the probe is conjugated to a binding partner, such as an antibody or biotin, and the other member of the binding pair carries a detectable moiety.
In one embodiment, detection is by Southern blotting and hybridization with a labeled probe. The techniques involved in Southern blotting are well known to those of skill in the art and can be found in many standard books on molecular protocols. See Sambrook et al, 1989. Briefly, amplification products are separated by gel electrophoresis. The gel is then contacted with a membrane, such as nitrocellulose or nylon, permitting transfer of the nucleic acid and non-covalent binding. Subsequently, the membrane is incubated with a chromophore-conjugated probe that is capable of hybridizing with a target amplification product. Detection is by exposure of the membrane to x-ray film or ion-emitting detection devices.
One example of the foregoing is described in U.S. Patent 5,279,721, incoφorated by reference herein, which discloses an apparatus and method for the automated electrophoresis and transfer of nucleic acids. The apparatus permits electrophoresis and blotting without external manipulation of the gel and is ideally suited to carrying out methods according to the present invention.
5. Expression Constructs In some embodiments of the present invention, an expression construct that encodes a 3' sequence of GBV-B is utilized. The term "expression construct" is meant to include any type of genetic construct containing a nucleic acid coding for a gene product in which part or all of the nucleic acid encoding sequence is capable of being transcribed. The transcript may be translated into a protein, but it need not be. Expression includes both transcription of a gene and translation of mRNA into a gene product. Expression may also include only transcription of the nucleic acid encoding a gene of interest. hi some constructs, the nucleic acid encoding a gene product is under transcriptional control of promoter and/or enhancer. The term promoter will be used here to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II. Much of the thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 bp of nucleic acids, and containing one or more recognition sites for transcriptional activator or repressor proteins.
At least one module in each promoter functions to position the start site for RNA synthesis. The best known example of this is the TATA box, but in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.
Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 bp upstream of the start site, although a number of promoters have recently been shown to contain functional elements downstream of the start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between promoter elements can be increased to 50 bp apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either co-operatively or independently to activate transcription. Enhancers were originally detected as genetic elements that increased transcription from a promoter located at a distant position on the same molecule of DNA. This ability to act over a large distance had little precedent in classic studies of prokaryotic transcriptional regulation. Subsequent work showed that regions of nucleic acids with enhancer activity are organized much like promoters. That is, they are composed of many individual elements, each of which binds to one or more transcriptional proteins.
The basic distinction between enhancers and promoters is operational. An enhancer region as a whole must be able to stimulate transcription at a distance; this need not be true of a promoter region or its component elements. On the other hand, a promoter must have one or more elements that direct initiation of RNA synthesis at a particular site and in a particular orientation, whereas enhancers lack these specificities. Promoters and enhancers are often overlapping and contiguous, often seeming to have a very similar modular organization.
6. Host Cells and Permissive Cells As used herein, the terms "cell," "cell line," and "cell culture" may be used interchangeably. All of these terms also include their progeny, which is any and all subsequent generations. It is understood that all progeny may not be identical due to deliberate or inadvertent mutations. In the context of the present invention, "host cell" refers to a prokaryotic or eukaryotic cell, and it includes any transformable organisms that is capable of replicating a vector or virus and/or expressing viral proteins. A host cell can, and has been, used as a recipient for vectors, including viral vectors. A host cell may be "transfected" or "transformed," which refers to a process by which exogenous nucleic acid is transfeπed or introduced into the host cell. A transformed cell includes the primary subject cell and its progeny. A "permissive cell" refers to a cell that supports the replication of a given virus and consequently undergoes cell lysis. In the context of the present invention, such a virus would include HCV, GBV-B, or other hepatitis viruses. In a "nonpermissive cell," productive infection does not result, but the cell may become stably transformed. In some embodiments, methods employ permissive cells that are a cell line derived from liver cells (liver cell line).
Host cells may be derived from prokaryotes or eukaryotes, depending upon whether the desired result is replication of the vector or expression of part or all of the vector-encoded nucleic acid sequences. Numerous cell lines and cultures are available for use as a host cell, and they can be obtained through the American Type Culture Collection (ATCC), which is an organization that serves as an archive for living cultures and genetic materials (www.atcc.org). An appropriate host can be determined by one of skill in the art based on the vector backbone and the desired result. A plasmid or cosmid, for example, can be introduced into a prokaryote host cell for replication of many vectors. Bacterial cells used as host cells for vector replication and or expression include DH5α, JM109, and KC8, as well as a number of commercially available bacterial hosts such as SURE® Competent Cells and SOLOPACK™ Gold Cells (STRATAGENE®, La Jolla). Alternatively, bacterial cells such as E. coli LE392 could be used as host cells for phage viruses.
Examples of eukaryotic host cells for replication and/or expression of a vector include HeLa, NIH3T3, Jurkat, 293, Cos, CHO, Saos, and PC12. Many host cells from various cell types and organisms are available and would be known to one of skill in the art. Similarly, a viral vector or virus or virus particle may be used in conjunction with either a eukaryotic or prokaryotic host cell, particularly one that is permissive for replication or expression of the vector. It is contemplated that the present invention includes vectors composed of viral sequences, viruses, and viral particles in the methods of the present invention, and that they may be used interchangeably in these methods, depending on their utility.
Some vectors may employ control sequences that allow it to be replicated and/or expressed in both prokaryotic and eukaryotic cells. One of skill in the art would further understand the conditions under which to incubate all of the above described host cells to maintain them and to permit replication of a vector. Also understood and known are techniques and conditions that would allow large-scale production of vectors, as well as production of the nucleic acids encoded by vectors and their cognate polypeptides, proteins, or peptides.
7. Pharmaceutical Compositions
The present invention encompasses the use of a 3 ' sequence of GBV-B in the production of or use as a vaccine to combat HCV infection. Compositions of the present invention comprise an effective amount of GBV-B clone as a therapeutic dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. The phrases "pharmaceutically or pharcnacologically acceptable" refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absoφtion delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incoφorated into the compositions. For human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.
The biological material should be extensively dialyzed to remove undesired small molecular weight molecules and/or lyophilized for more ready formulation into a desired vehicle, where appropriate. The active compounds will then generally be formulated for parenteral administration, e.g., formulated for injection via the intravenous, intramuscular, sub-cutaneous, intralesional, or even intraperitoneal routes. The preparation of an aqueous composition that contains GVB-B nucleic acid sequences as an active component or ingredient will be known to those of skill in the art in light of the present disclosure. Typically, such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for using to prepare solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. hi all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
Solutions of the active compounds as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
A GBV-B clone of the present invention can be formulated into a composition in a neutral or salt form. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like, i terms of using peptide therapeutics as active ingredients, the technology of U.S. Patents 4,608,251; 4,601,903; 4,599,231; 4,599,230; 4,596,792; and 4,578,770, each incoφorated herein by reference, may be used.
The carrier also can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absoφtion of the injectable compositions can be brought about by the use in the compositions of agents delaying absoφtion, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incoφorating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incoφorating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the prefeπed methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The preparation of more, or highly, concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small area.
Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like also can be employed.
For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035- 1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
In addition to the compounds formulated for parenteral administration, such as intravenous or intramuscular injection, other pharmaceutically acceptable forms include, e.g., tablets or other solids for oral administration; liposomal formulations; time release capsules; and any other form cuπently used, including cremes. Oral formulations include such nonnally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders. In certain defined embodiments, oral pharmaceutical compositions will comprise an inert diluent or assimilable edible carrier, or they may be enclosed in hard or soft shell gelatin capsule, or they may be compressed into tablets, or they may be incoφorated directly with the food of the diet. For oral therapeutic administration, the active compounds may be incoφorated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 75% of the weight of the unit, or preferably between 25-60%. The amount of active compounds in such therapeutically useful compositions is such that a suitable dosage will be obtained. The tablets, troches, pills, capsules and the like may also contain the following: a binder, as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules maybe coated with shellac, sugar or both. A syrup of elixir may contain the active compounds sucrose as a sweetening agent methyl and propylparabens as preservatives, a dye and flavoring, such as cherry or orange flavor.
C. Examples
The following examples are included to demonstrate prefeπed embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventor to function well in the practice of the invention, and thus can be considered to constitute prefeπed modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention. EXAMPLE 1 Elucidation of a 3' Sequence of the GBV-B Genome
The inventors elucidated the previously unrecognized 3' terminal sequence of GBV-B (SEQ ID NO 1). This sequence was reproducibly recovered from tamarin serum containing GBV-B RNA, in RT-PCR nucleic acid amplification procedures using several different primer sets, and as a fusion with previously reported 5' GBV-B sequences.
There is information in the published literature reporting the putative sequences of the 5' and 3' termini of the GBV-B genome. The nucleic acid sequences of these termini were reportedly determined by ligating the ends of the viral RNA together, amplifying the sequence in the region of the resulting junction by reverse-transcription polymerase chain reaction (RT-PCR), and sequencing of the cDNA amplification product across the junction. However, the inventors believed these results required confirmation. Of particular concern was the fact that the 3Λ terminus appeared to be shorter than the equivalent region of other viruses in the family Flaviviridae (especially within the genus Hepacivirus) and that the reported 3' sequence lacked a defined RNA haiφin structure such as those present in these related viruses. Additional novel sequences at the 3' end of the GBV-B genome were investigated using a serum sample collected from a tamarin that was experimentally infected with virus. Amplification was used to determine the sequence of the 3" end.
First, serum (50 μL) known to contain GBV-B RNA by RT-PCR assay was extracted with Trizol, and the RNA was washed and dried. A synthetic oligonucleotide was then ligated to the 3' end of the viral RNA. The oligonucleotide, 5'-
AATTCGGCCCTGCAGGCCACAACAGTC-3', which was phosphorylated at the 5Λ end and chemically blocked at the 3Λ end, was ligated to the RNA essentially using the method described by Kolykhalov et al. (Behrens et al, 1996). The RNA was initially dissolved in DMSO and the following additions were made: Tris-Cl, pH 7.5 (10 mM), MgCl2 (10 mM), DTT (5 mM), hexamine cobalt chloride (1 mM), 10 pmol oligo and 8 U T4 ligase. The final concentration of DMSO was 30% in a final volume of 10 μL. The ligation reaction was incubated for 4 or 20 hours at 19° C. 1 μL of the ligation reaction was used directly to make cDNA, using a primer complementary to the ligated oligonucleotide and the Superscript 2 system, in a final volume of 15 μL. 1 μL of cDNA was amplified using the Advantage cDNA system (Clontech) and two additional oligonucleotide primers. These primers included one that was complementary to the ligated oligonucleotide (i.e., "negative sense") and a positive-sense primer located near the 3" end of the reported GBV-B sequence. A product approximately 290 bases in length was obtained, and this was gel purified and directly sequenced. Sequencing was done in both directions using the oligonucleotide primers employed for the amplification; 259 bases that had not been previously reported were identified as fused to the sequence that had been previously described as the 3" teπninus of the viral genome. To ensure that this novel 3' sequence from viral RNA could be reproducibly amplified, an additional 10 μL of infected tamarin serum was extracted using Trizol. cDNA was prepared by reverse transcription using an oligonucleotide primer complementary to the penultimate 3' 25 bases of the novel sequence. Amplification was then done by PCR using the primer previously utilized for cDNA synthesis and a positive-sense primer mapping within the previously published GBV-B sequence, hi the initial studies, although a product was readily detected, DNA sequencing showed that this product was missing all of the sequence distal to the poly-U tract. Carrying out the cDNA synthesis in the presence of DMSO circumvented this problem. A cDNA product of approximately 290 bases was obtained. This was sequenced and shown to consist of the 5" primer, 20 bases of the published GBV-B sequence, and 259 bases of the novel sequence obtained in the preceding studies and containing the sequence of the 3" primer. The sequence of the 3λ end of GBV-B is shown in SEQ ID NO: 1 (FIG. 1). The possible secondary RNA structure for this region is shown in FIG. 2, as predicted by a computer-based RNA folding program. The presence of a predicted haiφin structure at the extreme 3' end of this novel sequence is consistent with its location at the 3' terminus of the viral RNA. The GBV-B cDNA (synthesis described above) was used as a template for PCR amplification of the 3' 1553 nucleotides (nts) of the GBV-B genome. This PCR amplification product was gel purified and cloned into plasmid DNA using the "Perfectly Blunt Cloning Kit" (Novagene).
EXAMPLE 2 Construction of an Infectious GBV-B Clone
The elucidation of a 3' sequence of the GBV-B genome will allow those of skill in the art to construct and validate an infectious molecular clone of GBV-B. This will be done using the following procedures.
A full-length cDNA copy of the GBV-B genome containing the newly identified 3' terminal sequences was constructed. RNA transcribed from this cDNA copy of the genome will be infectious when inoculated into the liver of a GBV-B permissive tamarin, giving rise to rescued GBV-B virus particles. A 1:1000 dilution of GBV-B infectious tamarin serum was obtained. This material was used as a source of viral RNA for the amplification of GBV-B nucleic acid sequences by reverse- transcription polymerase chain reaction. For amplification of previously reported segments of the GBV-B genome, 250 μL of the diluted serum was extracted with Trizol using the manufacturer's instructions. The final RNA pellet was dissolved in 10 μL of a 100 mM DTT buffer containing 5% RNasin. This material was converted into cDNA using Superscript 2 reverse transcriptase and oligonucleotide primers designed to be complementary to the reported GBV-B RNA sequence and to contain unique restriction sites. This cDNA was amplified using the Advantage cDNA kit (Clontech) employing the cDNA primer (negative sensiv as the downstream primer and a similar positive-sense upstream primer, again containing a unique restriction site. The published sequence of GBV-B allowed for the selection of primers in convenient areas of the genome containing unique restriction sites. Using this general strategy, the inventors amplified segments of the reported GBV-B genome representing: (1) nucleotides (nts) 1-1988, using an upstream primer containing a T7 RNA polymerase promoter and a BamHl site upstream of nt 1, and a downstream primer containing a unique EcolRl site (nt 1978); (2) nts 1968-5337, using a downstream primer containing a unique Clal site at position 5o27; (3) nts 5317- 7837, using a downstream primer containing a Sail site at nt 7847; and, (4) nts 7837-9143, using a downstream primer containing an added Xhol site. It was found necessary to use different PCR conditions for each primer set. The RT-PCR products generated in these reactions were cloned into plasmid DNA after gel purification, using the "Perfectly Blunt Cloning Kit" (Novagene). Ten bacterial colonies from each of the four RT-PCR products were analyzed for insert size by restriction endonuclease digestion using EcoRI, the sites for this enzyme being located on either side of the insert in the resulting plasmids. For three of the RT-PCR amplicons, 9 of 10 colonies contained plasmids with the coπect size insert. The EcoRI -Clal amplicon generated only 1/10 colonies with a coπect size insert. Thus, 30 additional colonies were examined, yielding two more clones with insert of the coπect size. For each of these plasmids, simple restriction patterns were obtained using two restriction enzymes. As these appeared to be coπect, the plasmid DNAs were subjected to sequencing using an ABI automatic sequencer.
EXAMPLE 3
Nucleotide sequence of the cloned GBV-B cDNA
The 5' region of the cloned sequence revealed a relatively long nontranslated region coπesponding to the published sequence of the GBV-B 5 'NTR, which includes an IRES. This region was followed by a long open reading frame. Near the 3' end of the genome a poly-U tract was identified; however, this was shorter than the published 3' homopolymeric poly-U region. The sequence from these clones was compared with those in the GenBank database (Accession U22304, "Hepatitis GB virus B polypeptide complete genome"). Twenty-two nucleotide differences were identified, of which 14 gave rise to amino acid changes (Table 1). In order to determine whether these changes were genuine or RT-PCR artifacts, which could have been introduced due to the very small amount of material from which these sequences were amplified, segments of the genome containing these changes were reamplified using a serum sample from an independently infected tamarin. Of the 14 changes noted in the original cDNA clones, 12 were not present in these newly amplified sequences and thus were probably RT-PCR artifacts (Table 2.). A particularly interesting difference from the published GenBank sequence, however, which was present in both the original clones as well as a repeat amplification, was a two- nucleotide substitution that obliterated the Sal I site present in the published sequence.
Table 1. Differences in the amino acid sequences of GBV-B cDNA clones and the GBV-B sequence reported by Simons et al. (1995).
Figure imgf000032_0001
EXAMPLE 4 Construction of a full-length GBV-B cDNA clone.
The four GBV-B cDNA inserts described above were cloned into Bluescript ks+ using unique restriction sites. Since the unique Sail site that was reported to be present in the published GBV-B sequence (nt position 7847) was absent in these cDNA clones, this restriction site was created by engineering two silent nucleotide changes using the "Quick Change" mutagenesis system (Stratagene). Although the most 5" clones (nucleotides 1-7847) could be readily constructed, attempts to add the remaining 3Λ clones were unsuccessful due to rearrangements and deletions. This problem was overcome by use of pACNR1180, a plasmid that had been used to construct an infectious clone of yellow fever virus. Finally, the most 3' 771 nucleotides of GBV-B were excised from the plasmid containing the novel, previously unreported 3' sequence, and inserted into the truncated assembled GBV-B cDNA construct to complete the 3' end. The 3' terminus of this full-length cDNA was then subjected to DNA sequencing to confirm its integrity. Extensive restriction digests indicated that this construct had the characteristics of a full-length c DNA copy of GBV-B virus. Because there is not yet an understanding of which cultured cells (if any) might be permissive for GBV-B replication, the infectivity of the synthetic GBV-B RNA will be assessed by injecting the RNA directly into the liver of a susceptible tamarin.
Alternatively, an infectious full-length clone can be produced by the following protocol. A plasmid will be made containing a cassette including the 5' and 3Λ ends of the virus flanked by appropriate restriction sites. These constructs have been shown to efficiently translate reporter genes, with transcription taking place via a T7 promoter placed immediately upstream of the 5 'NTR (e.g., see Rijnbrand et al, 1999). The major portion of the GBV-B genome would then be amplified by long range RT-PCR. This method is now well established for hepatitis C virus and other flaviviruses (Teller et al, 1996), and it has been used successfully also to amplify rhinovirus RNA. Briefly this technique uses "Superscript" reverse transcriptase to synthesize cDNA and a mixture of "KlenTaq 1", and "Deep Vent" polymerases to amplify this cDNA . Primers that can be used will contain restriction sites to allow cloning of the RT-PCR products into the cassette vector. After being transformed into suitable competent bacteria, extensive restriction analysis will enable us to determine which clones contain inserts that are of full length and which have a high probability of being coπect. Apparent full-length clones will be analyzed further by coupled transcription-translation using the Promega "TnT" system, with the addition of microsomal membranes to allow the cleavage of the structural proteins by cellular signalase enzymes. Clones which appear coπect by restriction analysis and which produce GBV-B proteins, in particular the protein coded for by the extreme 3' end of the genome, NS5B, will be selected, and RNA will be transcribed from these clones using the Ambion MegaScript system. "Coπect" looking clones (>10) can be injected directly into a tamarin liver at several sites. A successful infection will be determined as described below. If a positive signal is detected the entire genome will be amplified and sequenced to determine which plasmid the virus originated from.
EXAMPLE 5 Rescue of Infectious GVB-B Infectious GBV-B will be rescued from synthetic genome-length RNA following its injection into the liver of tamarins (Saquinus sp.). past studies, HAV from synthetic RNA in owl monkeys has been recovered (Aotus trivirgatus) (Shaffer et al, 1995), and more recently, the recovery of virus from a chimpanzee injected intrahepatically with RNA transcribed from a full-length genotype lb HCV cDNA clone was reported (Beard et al, 1999). RNA will be prepared for these studies using the T7 MegaScript kit (Ambion) and a total of 10 μg of plasmid DNA as template. An aliquot of the reaction products will be utilized to ensure the integrity of the RNA by electrophoresis in agarose-formaldehyde gels. The remainder of the transcription reaction mix will be frozen at -80C° until its injection, without further purification, into the liver of a tamarin. Because of the small size of the tamarin, the RNA will be injected under direct visualization following a limited incision and exposure of the liver. Under similar conditions, in other primate species, RT-PCR-detectable viral RNA or cDNA has not been detected in serum samples collected within days of this procedure in the absence of viral replication (Kolykhalov et al, 1997; Yanagi et al, 1998; Beard et al, 1999). Thus, the appearance of RNA in serum collected subsequently from these tamarins will be strong evidence for the replication competence of the synthetic RNA. Serum will be collected weekly for six weeks, then every other week for an additional 6 weeks from inoculated animals. In addition to RT-PCR for detection of viral RNA (see FIG. 2B), alanine aminotransferase (ALT) levels will be measured as an indicator of liver injury and to assess liver histology in punch biopsies taken at the time of ALT elevation. Maximum viremia and an acute phase ALT response is expected to occur around 14-28 days post-inoculation of infectious RNA (Simons et al, 1995; Schlauder et al, 1995; Karayiannis et al, 1989). Transfections will be considered to have failed to give rise to infectious virus if RNA is not detected in the serum within 12 weeks of inoculation. Successfully infected animals will be followed with twice weekly bleeds until resolution of the viremia, or for 6 months, whichever is longest.
EXAMPLE 6 Construction of GBV-B/HCV Chimeras
The GBV-B genome can be used as the acceptor molecule in the construction of chimeric viral RNAs containing sequences of both HCV and GBV-B. Such constructs will allow one to investigate the mechanisms for the different biological properties of these viruses and to discover and investigate potential inhibitors of specific HCV activities (e.g., proteinase) required for HCV replication. Different classes of chimeric viruses are contemplated. These include: (a) replacement of the GBV-B IRES with that of HCV; and (b) replacement of the NS3 major serine proteinase and helicase, and (c) the replacement of the NS5B RNA-dependent RNA polymerase with the homologous proteins of HCV.
The chimeric constructs described in the following sections will be made by PCR mutagenesis, using high fidelity polymerases and oligonucleotide primers designed to include the specific fusions of GBV-B and HCV sequences (Landt et al, 1990). First round PCR reactions will create the desired fusion, and generate a new "primer" to be used in a second PCR reaction spanning the region to a convenient unique restriction site. PCR cycles will be kept to the minimum number necessary for successful amplification, and all segments of viral sequence that are amplified by PCR will be subjected to DNA sequencing to exclude the presence of unwanted PCR-introduced eπors. Sequencing will be accomplished at UTMB's core Recombinant DNA Laboratory. Amplified segments will be kept to the minimum by the exchange of cloned cDNA segments spanning convenient restriction sites in subgenomic clones, and where necessary PCR artifacts can be coπected by site-directed mutagenesis (QuickChange mutagenesis kit, Stratagene).
A number of viable positive-strand RNA virus chimeras have been constructed previously in which LRES elements have been swapped between different viruses. Most of these chimeras have involved the exchange of IRES elements between picornaviruses. Others have been successful in constructing viable pohovirus chimeras containing the HCV IRES in place of the native pohovirus LRES (Zhao et al, 1999; Lu and Wimmer, 1996). A similar rhinovirus 14 chimera containing the HCV LRES has been constructed, although its replication phenotype is not as robust as the pohovirus chimera described by Lu and Wimmer (Lu and Wimmer, 1996). More importantly, Frolov et al (Frolov et al, 1998) recently reported chimeric flaviviruses in which the HCV LRES was inserted into the genetic background of a pestivirus, bovine viral diarrhea virus (BVDV) in lieu of the homologous BVDV sequence. Although these viable chimeric polioviruses and pestiviruses replicate in cell cultures, they are poor suπogates for HCV in animal models as neither virus is hepatotropic or causes liver disease. Importantly, Frolov et al. (Frolov et al, 1998) demonstrated quite convincingly that the requirement for cis-acting replication signals at the 5' terminus of the pestivirus genome was limited to a short tetranucleotide sequence. This requirement presumably reflects the need for the complement of this sequence at the 3' end of the negative strand during initiation of positive-strand RNA synthesis. The work of Frolov et al shows that the IRES of BVDV does not contain necessary replication signals, or that if these are present within the BVDV IRES they can be complemented with similar signals in either the HCV or encephalomyocarditis virus (EMCV, a picornavirus) IRES sequence. Since GBV-B and HCV are more closely related to each other than BVDV and HCV, these studies provide strong support for the viability of chimeras containing the HCV IRES in the background of GBV-B. Construction of a viable LRES chimera will be enhanced by studies that have documented the sequence requirements and secondary structures of the IRES elements of both HCV and GBV-B (see Lemon and Honda, 1997; Honda et al, 1996; Rijnbrand et al, 1999). To a considerable extent, the work of Frolov et al. (Frolov et al, 1998) was guided by studies of the HCV IRES structure. More recently, these studies have been extended to include a detailed mutational analysis of the GBV-B IRES. The results of these studies indicate that the functional IRES of GBV-B extends from the 5' end of structural domain II (nt 62) to the initiator AUG codon (nt 446). This segment of the full-length GBV-B clone will be replaced with HCV sequence extending from 5' end of the analogous domain II within the HCV IRES (nt 42) to the initiator codon at the 5' end of the HCV open reading frame (nt 341) to construct the candidate chimera, "GB/C:IRES". The source of HCV cDNA for these studies will be the infectious HCV clone, pCV-H77C, which contains the sequence of the genotype la Hutchinson strain virus (Yanagi et al, 1998), whose infectivity in a chimpanzee following intrahepatic inoculation with synthetic RNA transcribed from pCV-H77C has been confirmed.
This GB/C:IRES construct will retain two upstream haiφins within the GBV-B sequence (stem-loops la and lb), and it is thus analogous to the viable "BVDV+HCVdelB2B3Hl" chimera of Frolov et al (Frolov et al, 1998). A second chimera can be constructed in which the entire HCV 5' nontranslated RNA will be inserted in lieu of nts 62-446 of the GBV-B genome ("GB/C:5'NTR"). This construct will add to the inserted HCV sequence the most 5' stem-loop from HCV (stem-loop I). A similar insertion was shown to substantially increase the replication capacity of BVDV+HCVdelB2B3Hl by Frolov et al. (Frolov et al, 1998), providing a replication phenotype similar to wild-type BVDV in cell culture.
It is important to point out that there is strong evidence from multiple lines of investigation indicating that it will not be necessary to include coding sequence in these IRES chimeras. This is the case even though Reynolds et al. (Reynolds et al, 1995) have argued that the HCV IRES extends past the initiator codon, and into the core-coding region of that virus. Although Lu and Wimmer (Lu and Wimmer, 1996) found it necessary to include HCV core sequence to obtain a viable chimeric pohovirus, the BVDV chimeras reported by Frolov et al. (Frolov et al, 1998) did not contain any HCV coding sequence. This discrepancy may be explained by the observation that the only downstream requirement for full activity of both the GBV-B and HCV IRES elements is the presence of an unstructured RNA segment (Honda et al, 1996; Rijnbrand et al, 1999). Presumably, this facilitates interaction of the viral RNA with the 40S ribosome subunit in the early steps of cap-independent translation (Honda et al, 1996). The 5' GBV-B coding sequence fulfills this criterion (Rijnbrand et al, 1999).
EXAMPLE 7 In vitro Characterization of the Translational Activity of IRES Chimeras
The fidelity of the genome-length chimeric constructs will be confirmed by sequencing any DNA segments that have been subjected to PCR during the construction process, as well as confirming sequence at the junction sites. In addition, the translational activity of synthetic RNA derived from these constructs will be assessed and compared to the translational activity of the wild-type GBV-B and HCV RNAs. These studies will be carried out in a cell-free translation assay utilizing rabbit reticulocyte lysates (Rijnbrand et al, 1999). Synthetic RNA will be produced by runoff T7 RNA polymerase transcription using as template Clal-digested plasmid DNA (BamHI digestion in the case of the genome HCV construct) (T7 Megascript kit, Ambion). 3H-UTP will be added to the reaction mix to allow for quantification of the RNA product. Reticulocyte lysates (Promega) will be programmed for translation by the addition of RNA (at least 50% full-length as determined by agarose gel electrophoresis) at 20, 40 and 80 μg/ml, and translation reactions will be supplemented with microsomal membranes (Promega). S- Methionine-labelled translation products will be separated by SDS-PAGE, and the quantity of El protein produced from each RNA determined by Phosphorlmager analysis (Molecular Dynamics). Comparisons of the activity of the HCV IRES in the background of GBV-B and HCV will take into account differences in the methionine content of the El proteins of these viruses. Based on previous studies of both the GBV-B and HCV IRES elements (Honda et al, 1996; Rijnbrand et al, 1999), it is expected that these studies will confirm that the HCV IRES will retain nearly full activity when placed within the GBV-B background.
EXAMPLE 8 In vivo Characterization of IRES Chimeras
Synthetic RNAs produced from each of the two chimeric GBV-B/HCV constructs
(GB/C:IRES and GB/C:5'NTR) will be tested for their ability to induce infection and cause liver disease in susceptible tamarins. These studies will be carried out as described in Example 2.
GB/C:5'NTR may generate viremia and liver injury more closely resembling that observed with wild-type GBV-B infection (Frolov et al. , 1998).
EXAMPLE 9 Chimeric Flaviviruses Containing the HCV NS3 Serine Proteinase/Helicase within the
GBV-B Background
Chimeric flaviviruses containing the HCV NS3 serine proteinase/helicase within the GBV-B background are also contemplated within the present invention. The construction of chimeric flaviviruses containing specific heterologous functional polyprotein domains, however, poses a number of special problems. Unlike the situation with the IRES, where the relevant RNA segments appear to have a unique function restricted to cap-independent translation initiation and interact with host cell macromolecules, viral proteins often have multiple functions and may form specific macromolecular complexes with other viral proteins that are essential for virus replication (Lindenbach and Rice, 1999). Furthermore, such chimeric polyproteins must be amenable to efficient processing by the viral proteinases (NS2/NS3 or NS3). This requires knowledge of the proteinase cleavage specificities as well as specific sites of proteolytic cleavage. Although to date there have been no published studies of the processing of the GBV-B polyprotein, the relatively close relationship between GBV-B and HCV, about 30% overall amino acid identity within the polyprotein (Muerhoff et al, 1995), allows good computer predictions of the alignments of these proteins. The crystallographic structures of both the proteinase and helicase domains of the HCV NS3 protein have been solved (Yao et al, 1997). Thus, both linear alignments and models of the 3D structure of the NS3 proteins of these viruses can provide guidelines for designing specific chimeric fusions that are likely to preserve function. EXAMPLE 10 NS3 Proteinase-Domain Chimeras
In HCV, NS3 contains the major serine proteinase that is responsible for most cleavage events in the processing of the nonstructural proteins, i.e., those that occur at the NS3/4A, 4A/4B, 4B/5A and 5A/5B junctions. The active proteinase domain of HCV is located within the amino terminal third of the NS3 protein (residues 1-181), which shares 31% amino acid identity with the analogous segment of the GBV-B polyprotein (GBV-B vs HCV-BK) (Muerhoff et al, 1995). Importantly, the active site of this proteinase appears to be particularly well conserved in GBV-B. The GBV-B proteinase maintains the residues that are responsible for catalysis and zinc binding in the HCV enzyme (Muerhoff et al, 1995), and unlike the NS3 proteinases of some other flaviviruses preserves the Phe- 154 residue that determines in part the S\ specificity pocket of the enzyme and the preference of the HCV proteinase for substrates with a cysteine residue at the PI position (Scarselli et al, 1997). Thus, it is not suφrising that the relevant proteolytic cleavage sites within the GBV-B polyprotein that are predicted from alignments with the HCV polyprotein all possess a Cys residue at this position. Of greatest significance for the proposed studies, however, is the work of Scarselli et al (Scarselli et al, 1997) who demonstrated that the GBV-B NS3 proteinase is able to effectively process the polyprotein of HCV in studies carried out in vitro. Using synthetic peptide substrates, these investigators demonstrated that the enzymatic activities of the GBV-B proteinase (residues 1-181) had kinetic parameters similar to the HCV proteinase on NS4A/4B and NS4B/4A substrates HCV. They did not possess reagents allowing a determination of whether the HCV proteinase is able to cleave a GBV-B substrate, but their results indicate that these viral proteinases share important functional properties. Therefore, these similarities suggest that the HCV proteinase could function in lieu of the GBV-B proteinase if used to replace this segment of an infectious GBV-B clone. In addition, studies with sindbis/HCV chimeras have shown that the HCV proteinase can cleave within the framework of a sindbis polyprotein (Filocamo et al, 1997).
In considering the design of these NS3 proteinase chimeras, there are two additional important considerations. First, in HCV, the cleavage between NS2 and NS3 occurs in cis, as the result of a zinc-dependent metalloproteinase that spans the NS2/NS3 junction (Hijikata et al, 1993). As only the NS3 sequences will initially be exchanged, the viability of the resulting chimeras will be dependent upon preservation of the c/s-active cleavage across a chimeric NS2 NS3 proteinase domain. The alignment of GBV-B and HCV sequences shows that residues in HCV that have been shown by Grakoui et al, 1993, to be essential for the NS2/NS3 cleavage are conserved in GBV-B (Muerhoff et al, 1995). Additional chimeras that will include the relevant carboxyl-terminal portion of NS2 can also be created.
A second important consideration is that the mature HCV NS3 proteinase functions as a noncovalent assembly of the NS3 proteinase domain and the amino terminal portion of NS4A, a proteinase accessory factor. The details of this association are well known, and have been studied at the crystallographic level (Kim et al, 1996). The N-terminal domain of the folded proteinase contains eight β strands, including one contributed directly by the NS4A peptide backbone. X-ray studies have shown that this aπay of β strands gives rise to a much more ordered N-terminus. Thus, the presence of the NS4A strand seems likely to contribute to the structure of the substrate-binding pocket. It is not known whether the NS3 proteinase of GBV-B also requires a similar interaction with NS4A of that virus for complete activity, or, if so, whether the NS4A of GBV-B could substitute for NS4A of HCV in forming the fully active NS3 proteinase of HCV. The predicted GBV-B NS4A molecule is 54 amino acid residues in length (Simons, et al, 1995; Muerhoff et al, 1995), just as in HCV. However, the level of amino acid homology between the NS4A molecules is not especially high, and the potential interaction with either NS3 molecule cannot be predicted from this sequence on the basis of available knowledge. To overcome this potential problem, chimeras will be created in which not only the NS3 proteinase domain of GBV-B is replaced, but also the relevant NS4A segment as well, with homologous segments of the HCV polyprotein. The interaction of the HCV NS3 and NS4A domains represents a unique target for antiviral drug design, and it would be beneficial to have this specific interaction present in any virus to be used as a suπogate for HCV in the evaluation of candidate antiviral inhibitors of HCV proteinase in vivo.
The NS3 proteinase chimeras that can be made include "GB/C:NS3P", which will contain the sequence encoding the first 181 amino acid residues of the HCV NS3 molecule in lieu of that encoding the first 181 residues of GBV-B NS3, and "GB/C:NS3P4A", which will include the same NS3 substitution as well as the HCV sequence encoding the amino-terminal segment of NS4A that forms the interaction with NS3. The precise NS4A sequence to be included in the latter chimera will be based on the modeling studies, which may also suggest more effective fusions of the NS3 proteinase domain of HCV with the downstream NS3 helicase domain of GBV-B. The source of HCV cDNA for these studies will be the infectious HCV clone, pCV- H77C, which contains the sequence of the genotype la Hutchinson strain virus (Yanagi et al, 1998). EXAMPLE 11 NS3 Helicase Domain Chimeras
In addition to serine proteinase activity located within the amino-third of NS3, the downstream carboxy-terminal two-thirds of the molecule contains an RNA helicase activity. These two functional domains appear to be separated by a flexible spacer, within which the fusion of HCV proteinase or helicase domain sequences with GBV-B sequence will be placed. The exact role of the helicase in the HCV life-cycle is not known, but it is almost certainly required for dsRNA strand-separation during some phase of viral RNA synthesis. The helicase domains of GBV-B and HCV are remarkably well conserved, with some regions within the helicase showing as much as 55% amino acid identity (Muerhoff et al, 1995). The GBV-B helicase is more closely related to the HCV helicase than all other flaviviral NS3 helicases, and it preserves many residues found within the conserved helicase motifs of HCV. Thus the HCV NS3 helicase may be capable of functioning when placed within the polyprotein of GBV-B, and such a chimeric virus may be capable of replication. Residues 182-620 of the GBV-B NS3 molecule will be substituted with the analogous segment of HCV (FIG. 5, "GB/C:NS3h"). A chimera will also be made in which the entire NS3 and amino terminal NS4 protein sequences of GBV-B is replaced with the homologous HCV sequences ("GB/C:NS3-4A"). The latter construct will thus represent a dual proteinase-helicase chimera. As with the proteinase chimeras, the HCV cDNA will be derived from pCV-H77C (Yanagi et al, 1998).
EXAMPLE 12
In vitro Characterization of NS3 and NS3-NS4A Chimera
Prior to being evaluated for infectivity in susceptible tamarins, RNAs produced in vitro from these clones will characterized in vitro. This evaluation will be restricted to a documentation of the proper processing of the expressed polyprotein (i.e., NS2/NS3 and NS3 proteinase functions), since there are no relevant assays that can determine whether the helicase or RNA-dependent RNA polymerase activities in these polyproteins are sufficient for virus replication. The proteolytic processing of the polyprotein is important, however, as it may be altered either by inclusion of the heterologous HCV NS3 proteinase in lieu of the natural GBV-B protease, or by a change in the folding of the polyprotein induced by inclusion of HCV sequence anywhere within the polyprotein. These studies will be carried out in cell-free coupled transcription/translation assays ("TnT" system, Promega) supplemented with microsomal membranes. Template DNAs will be digested with Sal I, which restricts the cDNA within the NS5B coding region. 35S-methionine-labelled translation products will be separated by SDS- PAGE, and the mature NS3 protein identified by its apparent molecular mass. The NS3 and NS5B proteins will be identified by immunoblot analysis using rabbit antisera to the GBV-B NS3 and NS5B proteins. Generation of a mature ~68 kDa NS3 protein will provide proof of both the cw-active NS2/NS3 cleavage and the NS3-mediated cleavage of NS3/NS4A. Similarly, identification of a mature, processed NS5B molecule will provide further support for the activity of the NS3 proteinase. Controls for these studies will be the wild-type GBV-B polyprotein expressed in similar fashion from the full-length GBV-B clone. If necessary to more clearly demonstrate the processing of the nonstructural proteins in these constructs, subclones representing the nonstructural region of the chimeric sequences could be produced.
EXAMPLE 13
In vivo Characterization of NS3 and NS3-4A Chimeras
Synthetic RNAs produced from each of the chimeric GBV-B/HCV constructs described in the preceding section will be tested for their ability to induce infection and cause liver disease in susceptible tamarins. These studies will be carried out using the approach described above.
EXAMPLE 14
Chimeric Flaviviruses Containing the HCV NS5B RNA Dependent RNA Replicase within the GBV-B Background
The HCV NS5B molecule contains an RNA-dependent RNA polymerase that plays a central role in replication of the virus. Although this molecule represents a prime target for drug discovery efforts, it has proven difficult to express NS5B in a form that retains enzymatic activity specific for HCV RNA as a substrate. Thus, relatively little is known of the functional activity of the HCV replicase, including structure-function relationships of NS5B. Despite this, the NS5B proteins of GBV-B and HCV appear to be functionally closely related, as they share as much as 43% amino acid identity (Muerhoff et al, 1995). A more important question may be whether an RNA dependent RNA polymerase can act on foreign substrates. However, published work has shown that in vitro purified HCV polymerase has very little specificity for its template, using hepatitis C or globin message with equal fidelity (Behrens et al, 1996; Al et al, 1998). This finding is very similar to that obtained with picornaviral polymerases, where it has been known for many years that in vitro the enzyme exhibits very little specificity. It has always been considered highly likely that this situation would not pertain in vivo where it was thought that the interaction of viral or cellular factors with the 3Λ end of the genome would generate template specificity. However, recent reports have shown that the removal of the entire 3Λ untranslated sequence (leaving, however, the poly(A) region present) from both the pohovirus and rhinovirus genome does not completely abrogate the infectivity of the virus (Todd et al, 1997). Furthermore, virus, which was recovered after the initial transfections, was shown to have recovered much of the infectivity of the original virus (Todd et al, 1997). The mechanism for this recovery of infectivity is at present unknown, but these results suggest that the HCV polymerase may be able to function to replicate infectious GBV-B/HCV NS5B chimeras.
Thus a chimeric genome-length virus can be created in which the NS5B coding sequence of HCV (amino acids 2422-3014, 593 residues) is inserted within the background of GBV-B in lieu of its native RNA-dependent RNA polymerase (amino acids 2274-2864, 591 residues). This chimeric virus would be valuable for animal studies of candidate antiviral inhibitors of HCV RNA synthesis.
This NS5B chimera would be evaluated to determine that there was proper proteolytic processing of the polyprotein. This would be accomplished by expression of the chimeric polyprotein in a coupled translation-transcription reaction, followed by immunoblot analysis for the mature NS5B protein, as described for the NS3 and NS3-4A chimeras in the preceding section. If these results confirm that the GB/C:5B chimeric polyprotein is processed with release of NS5B, studies in tamarins would progress to determine whether synthetic RNA transcribed from the clone is infectious and capable of causing liver disease in intrahepatically inoculated animals. These studies would be carried out as described above. A chimeric molecule can be constructed from an infectious GBV-B clone in which the
HCV NS3 proteinase or proteinase helicase sequence would be placed in frame in lieu of the homologous GBV-B sequence, and this chimeric cDNA would be used to generate infectious GBV-B HCV chimeric viruses by intrahepatic inoculation of synthetic RNA in tamarins. Published studies indicate that the GBV-B and HCV proteinases have closely related substrate recognition and cleavage properties, likely making such chimeras viable and capable of initiating viral replication in appropriate cell types.
EXAMPLE 15
Chimeric Viruses Containing HCV Structural Proteins within a GBV-B Genetic
Background, and GBV-B Structural Proteins within an HCV Background It is well documented that the structural proteins of one flavivirus may in some cases be substituted for those from another member of the family. Such chimeric viruses have been recovered from viruses as distantly related to each other as dengue virus and tick-borne encephalitis virus (Pletnev et al, 1992). More recently, the prM and E proteins of Japanese encephalitis virus have been used to replace the equivalent proteins in a vaccine strain of yellow fever virus to produce a JE/YF chimera (Chambers et al, 1999). These observations suggest that chimeras in which the structural proteins of HCV have been used to replace the homologous proteins of GBV-B may well be viable and capable of replication. The isolation of a chimeric virus containing HCV structural proteins, but having the growth characteristics of GBV-B virus, could answer many fundamental questions concerning the structure and interaction of these proteins in HCV. They would also be useful in addressing the nature of the immune response to HCV structural proteins in infected primates (Farci et al, 1992). More to the point of this application, the availability of such chimeric viruses would allow studies of candidate HCV vaccines to be carried out in the tamarin model. This would be a major advance, because at present such studies are limited to chimpanzees (Choo et al, 1994).
The basis for the difference in the host ranges of HCV and GBV-B is completely unknown. Among many other possibilities, it is conceivable that the host range is dependent upon the availability of a specific receptor(s). If this were the case, host range might be dependent upon the envelope proteins that must interact with the putative cellular receptor. Thus, a chimeric virus containing the envelope proteins of HCV within the genetic background of GBV-B might be noninfectious in tamarins (but potentially infectious in chimpanzees). Thus, a finding that both structural protein chimeras are noninfectious in the tamarin, may require the construction of complementary chimeras in which the relevant GBV-B structural proteins will be inserted into the background of an infectious HCV clone. If inclusion of the GBV-B envelope proteins within the backbone of HCV confers on the resulting chimera the ability to replicate in tamarins, it will confirm an important role for the structural proteins in defining the different host ranges of these viruses. More importantly, the resulting virus would be an exceptionally valuable resource for future studies as it would contain all of the nonstructural replication elements, as well as the 5' and 3' nontranslated regions, of HCV. Such a virus would allow the tamarin model to be used to address many unresolved issues in HCV biology and pathogenesis.
EXAMPLE 16 Construction and Evaluation of Structural Protein Chimeras hi designing structural protein chimeras, it is important to note that the two envelope proteins of HCV, El and E2, form noncovalent heterodimeric complexes that are likely to be important in the assembly of infectious virus particles. This is not known to be the case with the envelope proteins of GBV-B, but it is likely given similarities in the sizes and hydropathy profiles of these proteins (Simons et al, 1995; Muerhoff et al, 1995). Accordingly, the El and E2 proteins will be replaced as a unit, and chimeras containing only one of these proteins from the heterologous virus will generally not be produced. First, a chimera will be created where the El and E2 regions of GBV-B virus are replaced with those of HCV, "GB/C:El-2". The source of HCV cDNA for these constructions will be pCV-H77C (Yanagi et al, 1998). A chimera will also be made in which the core protein, in addition to the envelope proteins, is replaced with the homologous proteins of HCV ("GB/C:Co-E2"). Additional chimeras will be made to determine whether tamarins can be infected with chimeras containing the GBV-B structural proteins within the genetic background of HCV. These will include "C/GB:El-2" and "C/GB:Co-E2". The backbone for these chimeras will be pCV-H77C, the infectious genotype la cDNA clone developed in the Purcell laboratory at NIALO (Yanagi et al, 1998).
The specific amino acid sequences of GBV-B to be replaced with the homologous segments of HCV have been determined by alignments of the GBV-B and HCV sequences, coupled with the location of signalase cleavage sites predicted to be present within the amino terminal third of the GBV-B polyprotein using the computer algorithm of Von Heijne. These predicted signalase cleavages lie between residues 156/157 (core/El), aa 348-349 (E1/E2) and 732/733 (E2 NS2) in the GBV-B sequence. Thus, the chimera GB/C:El-2 will contain sequence encoding HCV aa 192-809 in lieu of that encoding aa 157-732 in GBV-B, while the insertion in the GB/C:Co-E2 chimera will extend from the initiator AUG codon (aa 1) to residue 809 in HCV, and will be spliced into GBV-B in lieu of the segment encoding aa 1-732 in the GBV-B clone. The complementary chimeras to be constructed within the background of HCV will involve exchanges of the same segments of the genomes.
EXAMPLE 17 Characterization of Structural Protein Chimeras
Prior to being evaluated for infectivity in tamarins, the processing of these chimeric polyproteins will be examined in coupled transcription/translation reactions supplemented with microsomal membranes, as described in the preceding sections for the proteinase and other proposed chimeras. If these results confirm that the polyprotein is processed as expected, with production of glycosylated El and E2 proteins from each of the chimeras (and seen in similar studies with HCV proteins), studies would proceed in tamarins as previously described. The results of these studies may provide novel information on the basis of the host range differences that exist between HCV and GBV-B. If these results suggest that the envelope proteins play a critical role in determining host range, additional studies could be carried out with these chimeras in chimpanzees (which are permissive for HCV but apparently not for GBV-B). EXAMPLE 18 Further Characterization of Rescued Chimeric Viruses
Where infection with chimeric viruses is induced in animals that are injected within the liver with synthetic RNA, this virus will be passaged in GBV-B naϊve tamarins to further characterize the nature of the infection induced by the chimera. This will be accomplished by taking a pool of the 3 highest titer GBV-B RNA-containing serum specimens from the animal that was successfully transfected with RNA, and inoculating 1 mL of a 1:100 dilution of this pool intravenously into two susceptible animals. These animals will be monitored for infection and liver disease. These animals will be followed until resolution of the viremia and appearance of antibodies detectable in immunoblots with GST-NS3 protein expressed in E. coli, or for at least 6 months should an animal sustain a chronic infection. RT-PCR amplification of chimeric segments of the genome may be employed to determine whether the altered phenotype results from mutations within the heterologous portion of the genome.
EXAMPLE 19
Use of GBV-B as Model for HCV
GBV-B and/or GBV-B/HCV chimeras can be used as a model for HCV. Such studies will allow one to investigate the mechanisms for the different biological properties of these viruses and to discover and investigate potential inhibitors of specific HCV activities (e.g., proteinase) required for HCV replication.
EXAMPLE 20
Use of GBV-B/HCV Chimeras to Test Candidate HCV NS3 Proteinase Inhibitors or Other Inhibitors of HCV GBV-B/HCV viruses can be used in preclinical testing of candidate HCV NS3 proteinase inhibitors or other inhibitors of HCV.
1. Candidate Substances
As used herein the term "candidate substance" refers to any molecule that is capable of modulating HCV NS3 proteinase activity or any other activity related to HCV infection. The candidate substance may be a protein or fragment thereof, a small molecule inhibitor, or even a nucleic acid molecule. It may prove to be the case that the most useful pharmacological compounds for identification through application of the screening assay will be compounds that are structurally related to other known modulators of HCV NS3 proteinase activity. The active compounds may include fragments or parts of naturally-occurring compounds or may be only found as active combinations of known compounds that are otherwise inactive. However, prior to testing of such compounds in humans or animal models, it will be necessary to test a variety of candidates to determine which ones have potential. Accordingly, the active compounds may include fragments or parts of naturally-occurring compounds or may be found as active combinations of known compounds that are otherwise inactive. As such, the present invention provides screening assays to identify agents that are capable of inhibiting proteinase activity in a cell infected with chimeric GBV-B HCV viruses containing the HCV proteinase. It is proposed that compounds isolated from natural sources, such as animals, bacteria, fungi, plant sources, including leaves and bark, and marine samples may be assayed as candidates for the presence of potentially useful pharmaceutical agents. It will be understood that the pharmaceutical agents to be screened could also be derived or synthesized from chemical compositions or man-made compounds. Thus, it is understood that the candidate substance identified by the present invention may be polypeptide, polynucleotide, small molecule inhibitors or any other compounds that may be designed through rational drug design starting from known inhibitors of proteinases or from structural studies of the HCV proteinase.
The candidate screening assays are simple to set up and perform. Thus, in assaying for a candidate substance, after obtaining a chimeric GBV-B HCV virus with infectious properties, a candidate substance can be incubated with cells infected with the virus, under conditions that would allow measurable changes in infection by the virus to occur. In this fashion, one can measure the ability of the candidate substance to prevent or inhibit viral replication, in relationship to the replication ability of the virus in the absence of the candidate substance. In this fashion, the ability of the candidate inhibitory substance to reduce, abolish, or otherwise diminish viral infection may be determined. "Effective amounts" in certain circumstances are those amounts effective to reproducibly reduce infection by the virus in comparison to the normal infection level. Compounds that achieve significant appropriate changes in activity will be used. Candidate compounds can be administered by any of a wide variety of routes, such as intravenously, intraperitoneally, intramuscularly, orally, or any other route typically employed. It will, of course, be understood that all the screening methods of the present invention are useful in themselves notwithstanding the fact that effective candidates may not be found. The invention provides methods of screening for such candidates, not solely methods of finding them. 2. In vitro Assays
In one particular embodiment, the invention encompasses in vitro screening of candidate substances. Using a cell line that can propagate GBV-B in culture, in vitro screening can be used such that GBV-B or HCV virus production or some indicator of viremia is monitored in the presence of candidate compounds. A comparison between the absence and presence of the candidate can identify compounds with possible preventative and therapeutic value.
3. In vivo Assays
The present invention also encompasses the use of various animal models to test for the ability of candidate substances to inhibit infection by HCV. This form of testing may be done in tamarins.
The assays previously described could be extended to whole animal studies in which the chimeric virus could be used to infect a GBV-B permissive primate, such as a tamarin. One would then look for suppression of viral replication in the animal, and a possible impact on liver disease related to replication of the infectious chimeric virus. The advantage of this in vivo assay over present available assays utilizing HCV infection in chimpanzees is the reduced cost and greater availability of GBV-B permissive nonhuman primate species.
Treatment of animals with test compounds will involve the administration of the compound, in an appropriate form, to the animal. Administration will be by any route that could be utilized for clinical or non-clinical puφoses, including but not limited to oral, nasal, buccal, rectal, vaginal or topical. Alternatively, administration may be by intratracheal instillation, bronchial instillation, intradermal, subcutaneous, intramuscular, intraperitoneal or intravenous injection. Specifically contemplated are systemic intravenous injection, intraperitoneal injection, and oral administration. Determining the effectiveness of a compound in vivo may involve a variety of different criteria. Such criteria include, but are not limited to, survival, reduction of rate of infection, aπest or slowing of infection, elimination of infection, increased activity level, improvement in liver function, and improved food intake.
EXAMPLE 21
Use of Infectious GBV-B/HCV Chimeras as Vaccines
Infectious GBV-B/HCV chimeras expressing HCV envelope proteins will have utility as a vaccine immunogen for hepatitis C. Such clones clearly have the potential to be constructed as chimeras including relevant hepatitis C virus sequences in lieu of the homologous GBV-B sequence, providing unique tools for drug discovery efforts.
Chimeric viruses containing the envelope proteins of hepatitis C virus would confer the antigenic characteristics of hepatitis C virus on the chimera. These chimeras may have the ability to replicate in chimpanzees (and thus humans) by virtue of the fact that the chimeric envelope is now able to interact with the human hepatocyte cell surface, a necessary first step in virus replication. Therefore, the chimeric virus, while able to infect and replicate in humans, may not cause much or any disease-the reasoning here is that the genetic backbone of the chimera that encodes the nonstructural proteins of GBV-B has not evolved for replication in human cells and thus may not replicate well. Thus, the chimera may have limited replication ability, cause no disease, but still elicit immunity to the surface envelope proteins of HCV and thus have potential as a hepatitis C vaccine. These chimeras can be tested for their ability to promote immunity to HCV through an immune response.
EXAMPLE 22 Construction of a GBV-B Infectious Clone
A genome-length cDNA copy of the complete GB virus B (GBV-B) genome sequence, including the novel 3 ' terminal sequence, was assembled from fragments amplified by reverse transcription-polymerase chain reaction (RT-PCR) from viral RNA present in a 0.2 μl aliquot of infectious serum (GB agent pool mystrax 666, 8/93) supplied by Dr. Jens Bukh of the National Institutes of Health. The serum sample was diluted with 100 μl of fetal calf serum and extracted using the Trizol system (GIBCO/BRL). The pellet was dissolved in 10 mM dithiothreotol containing 20 u/mL RNasin (Promega). The selection of primers for cDNA synthesis and PCR amplification was based on the published sequence of GBV-B (Simons et al, 1995). RT/PCR was carried out using Superscript Reverse Transcriptase (GIBCO BRL) and the Advantage cDNA PCR Kit (Clontech). Four subgenomic regions were amplified covering the entire published GBV-B genome sequence. A fifth subgenomic region included the novel 3' terminal sequence that was identified in our laboratory. The oligonucleotide primer sets (listed 5'-»3') used for amplification of the individual regions included:
Primer 1: 5'CGGGATCCCGTAATACGACTCACTATAGACCACAAACACTCCAGT TTG - 3' (SEQ ID NO:3)
Primer 2: 5'-GTGGAATTCACAGCGTCATA-3' (SEQ ID NO:4) (Places a T7 RNA polymerase promoter sequence immediately upstream of the GBV-B sequence; the amplified segment extends from the 5' terminus of the viral genome to the unique EcoRI site at position
1978.)
Primer 3: 5'-TGTGAATTCCACTCTCCTACC-3' (SEQ ID NO:5)
Primer 4: 5'-TTATCGATTGCAGCAACCATG-3' (SEQ ID NO:6) (Overlapping EcoRI site at position 1978 and the unique Clal site at 5327.)
Primer 5: 5'-CATGGTTGCTGCAATCGATAAGCTGAAGAGTACAATAAC-3' (SΕQ ID
NO:7)
Primer 6: 5'-GACAACAGACGCTTGACACG-3' (SΕQ ID NO:8) (Overlapping the Cla I site at
5327 and a unique Sal I site at 7847 in the published sequence: however, the Sal I site was not present in the amplified GBV-B sequence, and was thus introduced into the cDNA using the
QuikChange Site-directed Mutagenesis Kit (Stratagene).)
Primer 7: 5'-CTGTCATGGGAGATGCGTAC-3' (SΕQ ID NO:9)
Primer 8: 5'-CGAGCTCGAGCACATCGCGGGGTCGTTAAGCCCGGGGTCTCC-3' (SΕQ
LD NO:10) (Overlapping the Sal I site at 7847 and the published 3' end of the genome.) Primer 9: 5'-GACAACAGACGCTTGACACG-3" (SΕQ ID NO:ll)
Primer 10: 5'-CCGACTCGAGAATTCGGCCCTGCAGGCCACAACAGTCTCGCGAG
TTTTTAATTCCAAGCGGGGGTTGCCCTCCGCTTGGAACAAAAACCGGGGTGCAGCCC
TTGGTAC-3' (SEQ ID NO: 12) (Overlaps product of primers 7 and 8 and extends to the novel 3' terminal sequence; includes an XJio I site at the 3' terminus of the GBV-B sequence to aid subsequent manipulations.)
RT-PCR products were gel-purified using the QIAquick Gel Extraction Kit (Qiagen) and ligated into a plasmid vector with the PstBlue-1 Perfectly Blunt Cloning Kit (Novagene).
Bacterial colonies were screened for plasmid DNAs with the coπect insert size. Those harboring an insert of the approximate coπect size were subjected to diagnostic restriction analysis. cDNA inserts generating the appropriate restriction patterns were sequenced in both directions using an
ABI DNA sequencer, and those with sequences closest to the published GBV-B sequence
(Simons et al, 1995) were selected for subsequent assembly into the full-length clone.
The cloned overlapping cDNA fragments were assembled into the pACNR1180 plasmid using the unique restriction sites noted in the primer descriptions. The final, fully assembled plasmid bearing a 9.4 kb cDNA GBV-B insertion was subjected to DNA sequencing to confirm the validity of the construction and the absence of any mutations or eπors in the sequence that may have been introduced during the assembly process. The sequence of the GBV-B infectious cDNA is shown in SEQ LD NO:2. Plasmid DNA containing the GBV-B cDNA was purified by isopycnic centrifugation on a CsCl gradient and isolated by ethanol precipitation. The plasmid DNA was linearized at the 3' terminus of the cDNA insert by digestion I. Two μg of the linearized DNA was used as template in an in vitro transcription reaction using the Ambion T7 MEGAscript Kit according to the manufacturer's instructions. The integrity of the resultant synthetic RNA product was confirmed by nondenaturing agarose gel electrophoresis.
Approximately 60 μg of RNA transcribed from the GBV-B cDNA clone was diluted in phosphate buffered saline and injected under direct visualization at laparotomy into the liver of a healthy tamarin without a prior history of GBV-B infection (Saguinus oedipus). Serum samples were collected from the animal weekly beginning at 2 weeks post-inoculation and tested for the presence of GBV-B RNA by real-time quantitative RT-PCR (TaqMan® assay). Results are shown in Table 2.
Table 2. Serum samples collected from the animal weekly beginning at 2 weeks post-inoculation and tested for the presence of GBV-B RNA by real-time quantitative RT-PCR.
Figure imgf000051_0001
The presence of high and increasing titers of viral RNA in the serum of this ammal between 2-4 weeks postinoculation confirms the infectivity of the full-length cDNA clone containing the novel 3' terminal sequence. The presence of viremia demonstrated that viral replication had ensued following the delivery of the synthetic viral RNA to hepatocytes.
EXAMPLE 23
Construction of a Chimeric Virus Containing the Domain III of the 5' NTR of HCV (IRES)
Within the Genetic Background of GBV-B.
1. Construction of plasmids containing chimeric cDNAs.
As described herein, a genome-length molecular clone of GBV-B was assembled from a series of overlapping cDNA fragments produced by RT-PCR from viral RNA isolated in a GBV- B-infected tamarin serum. This clone incoφorates the novel GBV-B 3 'NTR sequence, as described herein. The cDNA has been placed downstream of a T7 RNA polymerase promoter in the context of the bacterial vector pACNR1180. To facilitate further cloning steps, a C → T substitution was introduced at nucleotide (nt) position 496 of the GBV-B cDNA, so that a unique M restriction site was generated in the core-coding sequence without modifying the amino acid sequence. The resulting plasmid containing the full-length GBV-B cDNA with the engineered Mlul site at nt position 491 will be refeπed to as pGBV-B/Mlu. Chimeric GBV-B / HCV 5'NTRs were generated by substituting domains of the GBV-B 5'NTR by HCV counteφarts using a gene fusion PCR strategy (Landt et al, 1990) and HCV sequences derived from Hutchinson strain of genotype la, Genbank accession numbers AF011751 (SEQ LD NO:13), AF011752, and AF011753, each of which is incoφorated herein by reference (Inchauspe et al, 1991). The invention is not limited to a particular strain of HCV and sequences found in other HCV sequences may also be used, such as Genbank accession numbers AF009606, M67463, AF290978, AF387806, AF271632, M62321, AF387807, AF387805, AF387808, D10749, AF511948, AF177040, AF177038, AF177039, AF177037, AF511949,or M32084, each of which is incoφorated herein by reference. Chimeric 5'NTRs were amplified using overlapping PCR fragments spanning the desired HCV region framed by appropriate GBV- B regions, so that the BamHI restriction site located upstream of the T7 promoter sequence and the Mlul restriction site (nt 491) were used to substitute the parent GBV-B fragment within the molecular infectious clone. The sequence of all PCR-amplified fragments within the resulting, chimeric pGB/I-π-JJIHC, pGB/ΔIb/II-IIIHC, pGB/π-JHHC, and pGB/mHC plasmids (FIG. 2) were confirmed.
2. In vitro and ex vivo analysis of translation efficiency of chimeric IRES elements.
To assess the translational efficiencies of RNAs transcribed from the plasmids shown in FIG. 2, some of which contain chimeric IRES viral sequences, translation initiation activities were determined in vitro in rabbit reticulocyte lysates and ex vivo in primary tamarin hepatocytes. To facilitate quantitation ex vivo, the Mlul-Xhol fragment (nts 491-9398), spanning almost the entire GBV-B coding sequence and the 3' NTR of the cDNA in pGBV-B/Mlu, was replaced by a fragment encoding Renilla luciferase (RLuc) followed by the GBV-B 3' NTR. This substitution was carried out downstream of the chimeric sequences, generating plasmids pI-H-HI -Luc, pΔIMI-IIιHC-Luc, pπ-mHC-Luc, and piπHC-Luc that were used in translation studies.
These plasmids were linearized at the Xho I restriction site and used as templates for in vitro transcription using T7 RNA polymerase (Megascript kit, Ambion). In vitro translations were performed in Flexi Rabbit Reticulocyte lysates as described by the supplier (Promega) with
125mM KCl in the presence of 35S-Methionine. The RNA transcript concentration was
12.5ng/μl, well below the saturation point. Translation products were analyzed by 8% SDS- PAGE and quantitated by Phosphor nager analysis (Molecular Dynamics). Exemplary results are shown in FIG. 3.
Reporter RNAs representing I-H-IEF, Δlb/π-iπHC, and π-IflHC, all of which contain the complete HCV IRES element, translated with about 2.5-fold higher efficiency as compared to GBV-B/Mlu RNA which contains the intact GBV-B LRES (FIG. 2). The translational activity observed for iπHC was similar to that observed for the GBV-B/Mlu RNA. This is remarkable as this RNA contains a chimeric IRES in which upstream and downstream sequences flanking domain IJJ of the HCV LRES are derived from the GBV-B LRES. These data suggest that the fusion of HCV and GBV-B 5 'NTR sequences did not adversely affect translational activity of the internal ribosome entry site (IRES) when assayed in the cell-free translation system.
To assess the translational activity of these chimeric reporter RNA transcripts in living tamarin cells, in vitro transcribed RNA was mixed with the lipid-based compound DMRLE (Life Technologies) and transfected into primary tamarin hepatocytes (S. oedipus) that were prepared as described previously (Beames et al, 2000, incoφorated herein by reference). As an internal control, an RNA transcript encoding firefly luciferase (FLuc) under translational control of the EMCV IRES was cotransfected at 1:19th the abundance of the RLuc reporter constructs.
Translational activity was quantitated by determimng the levels of luciferase activity 24 hours following transfection (FIG. 4). In this system, which may represent more closely the conditions present during GBV-B infection of the tamarin, the translational activity of each of the RNA transcripts containing the complete HCV IRES was lower than that observed with the GBV-B/Mlu control. Thus, the GBV-B IRES may be more active in tamarin cells (the normal host species for GBV-B) than the HCV IRES. However, the chimeric transcript LπHC which contains domain m of the HCV IRES in lieu of its GBV-B counteφart had translational activity comparable to that of GBV-B/Mlu. Although the data shown in FIG. 3 and FIG. 4 indicate that there may be variation in the relative activities of these chimeric RNAs in different translation systems (in tamarin hepatocyte cultures and in a cell free translation system prepared from rabbit reticulocytes), these results may indicate that each of the chimeric IRES elements in these RNAs retains substantial translation-initiation activity. EXAMPLE 24 Infectivity of Chimeric Viral cDNAs Containing HCV 5'NTR Sequences within the Genetic
Background of GBV-B. 1. Inoculation of synthetic RNA into GBV-B susceptible tamarins. After linearization of the plasmid DNAs shown in FIG. 2 by digestion with Xhol, genome-length RNA transcripts containing the chimeric 5'NTR sequences were transcribed from the plasmids using the T7 MegaScript kit (Ambion). An aliquot of the reaction products was tested by agarose gel electrophoresis to ensure RNA integrity and to approximate the quantity of RNA. The remaining RNA was frozen at -80°C until inoculation, without further purification, into the liver of susceptible tamarins. Individual animals (S. mystax) received a dose of approximately 100-200 μg of RNA transcribed from only one of the plasmids shown in FIG. 2. The RNAs were inoculated into the liver under direct visualization following exposure by laparotomy. Inoculated animals were followed as described in the preceding sections of this document, with periodic testing of serum ALT, antibody to the NS3 protein of GBV-B, GBV-B genome, and with liver biopsy to assess pathologic changes in the liver. All tamarins were housed at the Southwest Regional Primate Research Center at the Southwest Foundation for Biomedical Research. Animals were cared for by members of the Department of Laboratory Animal Medicine at the Southwest Foundation for Biomedical Research in accordance with the Guide for the Care and Use of laboratory Animals. All protocols were approved by the Institutional Animal Care and Use Committee.
The primary indicator of successful infection was detection of sustained viremia in samples of serum collected over a period of weeks. Viremia was monitored by detection of GBV-B RNA using a sensitive and specific real-time RT-PCR assay. RNA was isolated from virus present in tamarin serum using the QiaAmp viral RNA extraction kit (Qiagen) and amplified in a one-step RT-PCR reaction utilizing TaqMan® EZ core reagents (Perkin Elmer). First-strand cDNA synthesis was carried out using the primer NS5ARPp (5'- GAAGGAGGGAGGTTTGAAGGA-3*, position 6949-6969)(SEQ ID NO: 14). The cDNA product was quantified in a 5' exonuclease PCR (TaqMan®) assay using a primer-probe combination that recognized sequences in the GBV-B NS5A gene. The primers NS5ARPp and NS5AFPp (5'-CCAGTTCCGGGCAAGAACT-3', position 6844-6862)(SEQ ID NO: 15) and probe (nts 6913-6938) were obtained from Fisher and Perkin Elmer, respectively. A synthetic RNA derived from the infectious clone was used as the standard for quantitation of the RNA. There was no evidence for replication of the I-II-iπ , π-HI , and ΔIb/11-iπ chimeras following their inoculation into the liver of individual GBV-B naive tamarins during the 22-week follow up period. In contrast, RT-PCR suggested the possible presence of a low level of viral RNA in the serum of the animal injected with iπHC chimera 1-2 weeks postinoculation, followed by disappearance of any sign of viremia. However, 12 weeks after inoculation with the iπHC RNA, viremia reappeared in tamarin T16444, rapidly reaching a titer in excess of 10 genome copies per ml, and subsequently persisting for 20 weeks (terminating 32 weeks following inoculation of the RNA) (FIG. 5). The course of the infection in this animal from week 12 on was similar to what is seen in infection of naϊve animals with the wild-type infectious clone, with the exception that there was no detectable evidence of antibodies to the NS3 protein. The long delay to the appearance of the secondary viremia in this animal suggests a requirement for the accumulation of adaptive mutations that are compensatory to changes in the replication capacity of the RNA related to the presence of the chimeric 5'NTR sequence.
The results shown in FIG. 5 indicate the replication competence of the in chimeric RNA (see FIG.' 2) following intrahepatic inoculation of synthetic RNA in a GBV-B naive tamarin (S. mystax). The presence of the chimeric 5'NTR sequence was confirmed in the RNA replicating in this animal by RT-PCR amplification of RNA extracted from the serum, followed by direct DNA sequencing of the amplified cDNA.
To prove that inoculation of T 16444 with synthetic fflHC chimeric RNA had led to the rescue and replication of virus containing the chimeric 5'NTR sequence, serum collected 14 weeks after the inoculation of this animal with RNA was used to inoculate a second GBV-B naive animal (T 16451) by intravenous injection. This is a stringent test for the presence of virus, since nonpackaged viral RNA that might be released from the liver into the serum would be expected to be highly degraded and no longer infectious. Only viral RNA packaged into viral particles would be expected to transmit the infection. This second animal rapidly developed evidence of GBV-B infection, with viremia detected as early as 1 week after infection, and with a peak viremia of greater than 108 genome copies/ml by 4 weeks postinfection. The rapid appearance of the viremia and early robust replication of the virus in this second animal suggested that the chimeric virus had indeed undergone adaptation and that it no longer required a lengthy incubation period to induce viremia.
These results confirm that the synthetic chimeric iπHC RNA replicated and produced infectious virus particles, capable of transmitting the infection to a second animal. Liver biopsy in this animal confirmed the presence of hepatic inflammation, while serial determinations of serum ALT activities demonstrated the typical profile of acute liver injury that normally accompanies wild-type GBV-B or HCV infections. The demonstration of hepatitis in association with infection with the chimeric IJJHC virus enhances its potential utility as a suπogate virus for evaluation of antiviral agents directed against the IRES or RNA replication signals residing within domain III of the HCV 5'NTR.
EXAMPLE 25 Sequence Analysis of Recovered Chimeric IIIHC Virus
The lengthy delay prior to the appearance of high level viremia in T 16444 coupled with the early robust replication of virus collected late in the course of infection in T 16444 in a second animal suggests that the chimeric iπHC virus had undergone adaptation to the presence of the chimeric 5'NTR. To determine the nature of the mutations contributing to this adaptation, viral RNA from both animals was amplified by RT-PCR and subjected to sequencing and comparison with the parent synthetic mHC RNA. Viral RNA was isolated from serum obtained at week 14 from tamarin T 16444, and week
4 or 8 from T16451 using Qiamp Viral RNA extraction kit (Qiagen). Random or GBV-B specific primers were used to prepare cDNA using MMLV reverse transcriptase from the Advantage RT-for-PCR kit (Clontech). PCR amplification of 1-2 kb fragments was carried out using the Advantage 2 PCR enzyme system (Clontech) utilizing GBV-B specific primers. Amplimers were purified through a silica gel membrane (QiaQuick PCR purification kit, Qiagen) and subjected to direct sequencing on an ABI 373XL instrument. The 3' terminus of the viral genome was ligated to a 27-mer oligonucleotide as described in the previous section for parent GBV-B. This RNA was then subjected to reverse transcription using a primer complementary to the ligated primer, and the cDNA was subsequently amplified using the same 3' primer and a 5' primer specific for a sequence in the 3 'NTR (nts 9092-9111). A semi-nested PCR was performed with an internal 5' primer (nts 9097-9114). The 5' terminus of the viral genome was subjected to reverse transcription using a 3' primer complementary to the domain Ifl of the HCV 5'NTR sequence (nts 209-229 of the HCV cDNA). The resulting cDNA was ligated to the 27-mer oligonucleotide and amplified using a primer complementary to the ligated primer and the same 3' primer used for RT. A semi-nested PCR was then performed using an internal 3' primer (nts 137-156 of the HCV cDNA). Results are shown in FIG. 7. EXAMPLE 26 Use of the Chimeric IRES Virus GB/IIIHC to Study the Efficacy of Potential Antiviral Candidates Targeting Domain III of the HCV IRES (IIIHC)
GB/niHC chimeric virus replicates robustly in small primates (see FIG. 11) and were therefore used for antiviral validation studies in the small primate model. GB/LπHC chimeric virus were also demonstrated to replicate well in primary cultures of tamarin hepatocytes (FIG.
8).
Primary tamarin hepatocytes (Saguinus mystax) were prepared as described previously (Beanies et al, 2000). Wild-type (wt) GBV-B and chimeric virus GB/njHC inocula were derived from tamarins that had been infected with the coπesponding viruses. 2.5 x 10 cells were seeded into each well of 6-well plates, and subsequently infected with virus (wt or chimera) at a multiplicity of infection (m.o.i.) of approximately 0.6 genome equivalents (ge)/cell. One hour after infection, cells were thoroughly washed 5 times and fresh medium was added. The media was replaced with fresh media every 24 hours thereafter. GBV-B RNA in culture supernatants and cell extracts was measured quantitatively at the indicated times (FIG. 8) by a realtime RT- PCR (TaqMan®) assay targeting GBV-B core protein coding sequence (Beames et al, 2000). Results showed that there is synthesis and secretion of newly foπned viral particles in the culture supernatant after infection with both the wild-type and chimeric viruses. The chimeric IRES virus replicated efficiently in these cultures, although to titers that were about 10-fold less than the wild-type GBV-B titers.
These data show that the GB/ffl chimeric virus makes possible the validation of candidate antiviral compounds targeting domain in of the HCV IRES in the context of a complete viral replication cycle in primary hepatocytes in ex vivo culture. This is a significant advantage over the only other system for cell culture validation of antiviral compounds, which consists of a cell culture system wherein only the effect on viral RNA replication can be monitored. The GB/LTI chimeric virus also allows such studies in the small primate model: tamarins, marmosets, or owl monkeys.
EXAMPLE 27 Determination of the Mutations Contributing to Efficient Replication of the IIIHC Chimeric Virus.
Six mutations were identified in the viral RNA isolated at week 14 post-inoculation (p.i.) from a tamarin (T16444) that was inoculated with the domain HI chimeric RNA (iπHC) (FIG. 9). Two mutations were located in the 3'NTR, two were silent substitutions, in the El and NS5B coding regions, respectively, and two were substitutions leading to amino acid changes in the E2 and NS5B sequences, respectively (FIG.10). The fact that high titers of virus appeared in the serum only many weeks after RNA inoculation of tamarin T 16444, and that this virus contained these mutations, suggests that the mutations (either singly or collectively) may be required for robust replication of the chimeric virus.
Virus recovered from T 16444, that contains the chimeric 5'NTR as well as all of these mutations, replicated efficiently when passaged in a second tamarin (T16451, FIG. 11).
To assess the mutations role m replication of III chimera, these mutations were iteratively introduced into the cDNA of the initial IHHC chimera. Four plasmid DNAs were thus constructed (FIG. 12). After linearization of the plasmid DNAs by digestion wit XhoI, genome- length RNA transcripts containing the chimeric 5'NTR and the indicated mutations elsewhere in the genome were transcribed from the cognate plasmids using the T7 MegaScript kit™ (Ambion). An aliquot of the reaction products was tested by agarose gel electrophoresis to ensure RNA integrity and to approximate the quantity of RNA. The remaining RNA was frozen at -80°C until inoculation, without further purification, into the liver of susceptible GBV-B naϊve tamarins.
Individual animals (S. mystax) received a dose of approximately 100 μg of RNA transcribed from one of the plasmids shown in FIG. 12. The RNAs were inoculated into the liver under direct visualization following exposure by laparotomy. Inoculated animals were followed as described above, with periodic testing of serum ALT, antibody to the NS3 protein of GBV-B, and the presence of GBV-B RNA in serum. The tamarins were housed at the Southwest Regional Primate Research Center at the Southwest Foundation for Biomedical Research. They were cared for by members of the Department of Laboratory Animal Medicine of the Southwest Foundation for Biomedical Research in accordance with the Guide for the Care and Use of laboratory Animals. All protocols were approved by the Institutional Animal Care and Use Committee (IACUC).
The primary indicator of successful infection was detection of sustained viremia in sequential serum samples collected over a period of weeks. Viremia was monitored by detection of GBV-B RNA using a sensitive and specific real-time RT-PCR assay. RNA was isolated from virus present in tamarin serum using the QIAamp® viral RNA extraction kit (Qiagen) and amplified in a one-step RT-PCR reaction utilizing TaqMan® EZ core reagents (Perkin Elmer). First-strand cDNA synthesis was carried out using the primer NS5ARPp (5 - GAAGGAGGGAGGTTTGAAGGA-3' (SEQ ID NO: 14), position 6949-6969). The cDNA product was quantified in a 5' exonuclease PCR (TaqMan®) assay using a primer-probe combination that recognized sequence in the GBV-B NS5A coding region. The primers NS5ARPp and NS5AFPp (5'-CCAGTTCCGGGCAAGAA CT-3' (SEQ LD NO: 15), position 6844-6862) and probe (nts 6913-6938) were obtained from Fisher and Perkin-Elmer, respectively. A synthetic RNA derived from the infectious clone was used as the standard for quantitation of the RNA.
RNAs containing only the NS5A mutation (GB/πi -ml) failed to replicate. On the other hand, in one of two inoculated animals, a chimeric RNA containing the NS5A mutation together with both 3TSTTR mutations (GB/UI -m2) replicated in a robust fashion at week 4 post- inoculation, reaching viremia levels in excess of 107 genome equivalents/ml (FIG. 12). Consistent with this latter result, the introduction of the E2 mutation into the mutated chimeric RNA containing the NS5A and 3 'NTR mutations (GB/IIIHC-m3), or a modified chimeric RNA containing all 6 mutations (GB/iπHC-m4) (FIG. 12), resulted in rapid rescue of virus and the presence of substantial viremia within 2 weeks of intrahepatic inoculation of the RNA (FIG. 13). These data indicate that one or both of the two mutations in the 3' NTR sequence of
GBV-B (C9052U and C9065U) are involved in an efficient-replication phenotype of the domain m chimeric virus. These mutations appear to be useful for efficient replication both in vivo in intrahepatically inoculated animals and ex vivo in cultured primary hepatocytes.
These data are strongly suggestive of a diret interaction between the 3 'NTR in the region of nucleotides 9052 and 9065 and the 5'NTR in the region of domain III of the IRES. Such circularizing interactions of the termini of genomic RNA are well known to occur in other members of the Flaviviridae family and are essential for viral replication. However, they have not been previously documented to occur in hepaciviruses.
All of the compositions and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of prefeπed embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents, which are both chemically and physiologically related, may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
REFERENCES
The following references, to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incoφorated herein by reference.
U.S. Patent 4,578,770
U.S. Patent 4,596,792
U.S. Patent 4,599,230
U.S. Patent 4,599,231 U.S. Patent 4,601,903
U.S. Patent 4, 608,251
U.S. Patent 4,683,195
U.S. Patent 4,683,202
U.S. Patent 4,800,159 U.S. Patent 4,883,750
U.S. Patent 5,279,721
U.S. Patent 5,807,670
EPA No. 320,308
EPA No. 329,822 GB Application No. 2,202,328
PCT/US87/00880
PCT/US89/01025
WO 88/10315
WO 89/06700 WO 90/07641
P et al, Virus Res., 53:141-149, 1998.
Alter etal, Hepatology, 26:625-655, 1997.
Beames et al, J. Virol, 74:11764-11772, 2000.
Beard et al, Hepatology, 1999. Behrens et al, EMBO. J., 15:12-22, 1996.
Chambers et al, J. Virol, 73:3095-3101, 1999.
Choo et al, Proc. Natl Acad. Sci. USA, 91:1294-1298, 1994.
Consensus Development Panel. National Institutes of Health Consensus Development Conference Panel statement: Management of hepatitis C. Hepatology, 26:Supplement
1[3], 2S-10S, 1997.
Deinhardt et al, J. ExpMed. 125:637-688, 1967.
Farci et al, Science, 258:135-140. 1992.
Farci et al, J. Infectious Diseases, 165:1006-1011, 1992. Filocamo et al, J. Virol, 71 1417-27, 1997.
Frohman, PCR Protocols: A Guide To Methods And Applications, Academic Press, New York, 1990.
Frolov et al, RNA, 4:1418-1435, 1998.
Grakoui et α/., J. Virol, 67(5):2832-2843, 1993. Grakoui et al, J. Virol, 67(3):1385-95. 1993.
Hijikata et α/., J Virol, 67:4665-4675, 1993.
Honda et al, RNA, 2:955-956, 1996.
Honda et al, J. Virol, 73:1165-1174, 1999. hichauspe et al, Proc. Natl. Acad. Sci. USA, 88:10292-10296, 1991. Karayiannis et al, Hepatology, 9:186-192, 1989.
Kim et al, Cell, 87:343-355, 1996.
Kolykhalov et al, Science, 277:570-574, 1997.
Kolykhalov et α/., J Virol, 70:3363-3371, 1996.
Landt et al, Gene, 96:125-128, 1990. ' Lemon and Honda, Seminars in Virology, 8:274-288, 1997.
Lindenbach and Rice, J. Virol, 73:4611-4621, 1999.
Lu and Wimmer, Proc, Natl, Acad., Sci, USA, 93:1412-1417, 1996.
Muerhoffet α/., J. Virol, 69:5621-5630, 1995.
Neumann et al, Science, 282:103-107, 1998. Plernev et al, Proc. Natl. Acad. Sci. USA, 89:10532-10536, 1992.
Remington's Pharmaceutical Sciences, 15th Edition, pages 1035-1038 and 1570-1580.
Reynolds et al, EMBO. J., 14:6010-6020, 1995. Rijnbrand et al, Mutational analysis of the GB virus B internal ribosome entry site. J Virol,
74(2):773-783, 1999. Sambrook, et al, Molecular Cloning, A Laboratory Manual, 2nd ed., Cold Spring Harbor
Laboratory, Cold Spring, Harbor, N.Y., 1989. Scarselli et al, J. Virol, 71 4985-4989, 1997. Schlauder et al, J. Med. Virol, 46:81-90, 1995. Shaffer et al, J. Virol, 69:600-6604, 1995. Simons, et al, Proc. Natl. Acad. Sci, 92:3401-3405, 1995. Tanaka et al, Biochem. Biophys. Res. Comm., 215:744-749, 1995. Teller et al, J. Clin. Micro., 34:3085-3091, 1996. Todd et α/., J. Virol, 71:8868-74, 1997. Yanagi et al, Proc. Natl. Acad. Sci. USA, 16:8738-8743, 1998. Yao et al, Nature Structural Biology, 4: 463-467, 1997. Zhao et al., J. Virol, 73:1546-1554, 1999.

Claims

1. An isolated polynucleotide comprising a chimeric GBV-B polynucleotide encoding a virus.
2. The polynucleotide of claim 1, comprising a chimeric GBV-B genome, wherein at least part, but not all of a 5' NTR sequence is from a HCV 5' NTR.
3. The polynucleotide of claim 2, wherein at least one, but not all of domain I, LT, m, or TV of the 5' NTR is from a HCV 5' NTR.
4. The polynucleotide of claim 2, wherein domain I of the 5' NTR is from a HCV 5'NTR.
5. The polynucleotide of claim 2, wherein domain LT of the 5' NTR is from a HCV 5'NTR.
6. The polynucleotide of claim 2, wherein domain III of the 5' NTR is from a HCV 5'NTR.
7. The polynucleotide of claim 6, wherein the 5 ' NTR domain lb of GBV-B is deleted.
8. The polynucleotide of claim 2, wherein domain IV of the 5' NTR is from a HCV 5'NTR.
9. The polynucleotide of claim 2, wherein domain I and domain II of the 5' NTR is from a HCV 5'NTR.
10. The polynucleotide of claim 2, wherein domain I and domain III of the 5' NTR is from a HCV 5'NTR.
11. The polynucleotide of claim 2, wherein domain I and domain IV of the 5' NTR is from a HCV 5'NTR.
12. The polynucleotide of claim 2, wherein domain II and domain III of the 5' NTR is from a HCV 5'NTR.
13. The polynucleotide of claim 2, wherein domain LT and domain IV of the 5' NTR is from a HCV 5'NTR.
14. The polynucleotide of claim 2, wherein domain III and domain IV of the 5' NTR is from a HCV 5'NTR.
15. The polynucleotide of claim 2, wherein domain II, domain III and domain IV of the 5 ' NTR is from a HCV 5'NTR.
16. The polynucleotide of claim 15, wherein the 5 ' NTR domain lb of GBV-B is deleted.
17. The polynucleotide of claim 2, wherein said polynucleotide is DNA.
18. The polynucleotide of claim 2, wherein said polynucleotide is RNA.
19. The polynucleotide of claim 1 , further comprising at least part of a structural protein coding region of HCV.
20. The polynucleotide of claim 1 , further comprising at least part of a non-structural protein coding region of HCV.
21. A viral expression construct comprising a chimeric GBV-B polynucleotide, wherein at least a part of the 5' NTR sequence is from a HCV 5' NTR.
22. The viral expression construct of claim 21, wherein domain III of the 5' NTR is from a HCV 5' NTR.
23. The viral expression construct of claim 21 , furtl er comprising a deletion of the GBV-B 5 ' NTR domain lb region.
24. The viral expression construct of claim 21, wherein said construct is a plasmid.
25. The viral expression construct of claim 21, wherein said construct is a virus.
26. The viral expression construct of claim 21 , further defined as a construct for the expression of a chimeric GBV-B/HCV virus.
27. A method for identifying a compound active against a viral infection comprising: providing a virus expressed from a viral construct comprising a chimeric GBV-B/HCV virus; contacting the virus with a candidate substance; and comparing infectivity of the virus in the presence of the candidate substance with the infectivity of the virus in the absence of the candidate substance.
28. The method of claim 27, wherein the chimeric virus comprises at least part of a 5 ' NTR sequence from a HCV 5' NTR.
29. The method of claim 28, wherein the chimeric virus comprises domain III of the 5' NTR
Figure imgf000066_0001
30. The method of claim 28, wherein the chimeric virus comprises a deletion of domain lb of GBV-B.
31. A compound active against a viral infection identified according to a method comprising: providing a virus expressed from a viral construct comprising a GBV-B/HCV chimeric virus; contacting the virus with a candidate substance; and comparing the infectivity of the virus in the presence of said candidate substance with the infectivity of the virus in the absence of said candidate substance.
32. The compound of claim 31 , wherein the compound is identified by providing a viral construct comprising at least part of a 5' NTR sequence from a HCV 5' NTR.
33. The compound of claim 32, wherein the compound is identified by providing a viral construct comprising domain III of the 5' NTR is from a HCV 5'NTR.
34. The compound of claim 32, wherein the compound is identified by providing a viral construct lacks domain lb of GBV-B.
35. A hepatotropic virus comprising a chimeric GBV-B polynucleotide, wherein at least a part of the 5' NTR sequence is from a HCV 5' NTR.
36. The hepatotropic virus of claim 35, wherein domain III of the 5' NTR is from a HCV 5' NTR.
37. The hepatotropic virus of claim 35, further comprising a deletion of the GBV-B 5' NTR domain lb region.
38. The hepatotropic virus of claim 36, wherein the virus propagates in vivo.
39. The hepatotropic virus of claim 35, further defined as a construct for the expression of a chimeric GBV-B/HCV virus.
40. A method of producing a virus comprising: introducing into a host cell a viral expression construct comprising a chimeric GBV-B polynucleotide encoding at least part of a 5' NTR sequence from a HCV 5' NTR sequence; and culturing the host cell under conditions permitting production of a virus from the construct.
41. The method of claim 40, wherein said polynucleotide comprises at least a 5 ' NTR domain
Figure imgf000067_0001
42. The method of claim 40, wherein said polynucleotide comprises at least a 5 ' NTR domain
Figure imgf000067_0002
43. The method of claim 40, wherein said polynucleotide comprises at least a 5' NTR domain
Figure imgf000067_0003
44. The method of claim 43, wherein said polynucleotide comprises a deletion of 5' NTR domain lb of GBV-B.
45. The method of claim 40, wherein said polynucleotide comprises at least a 5' NTR domain I and domain II from a HCV 5 ' NTR.
46. The method of claim 40, wherein said polynucleotide comprises at least a 5 ' NTR domain
I and domain IV from a HCV 5' NTR.
47. The method of claim 40, wherein said polynucleotide comprises at least a 5' NTR domain I, domain II and domain III from a HCV 5' NTR.
48. The method of claim 40, wherein said polynucleotide comprises a 5' NTR from a HCV 5' NTR.
49. The method of claim 40, wherein said polynucleotide comprises at least a 5' NTR domain
II and domain III from a HCV 5' NTR.
50. The method of claim 40, wherein said polynucleotide comprises at least a 5' NTR domain III and domain IV from a HCV 5 ' NTR.
51. The method of claim 40, wherein said host cell is a eukaryotic cell.
52. The method of claim 51, wherein said host cell is in an animal.
53. The method of claim 40, wherein said polynucleotide comprises synthetic RNA.
54. The method of claim 40, further comprising the step of isolating virus from said host cell.
55. The method of claim 54, wherein the isolated virus is purified to homogeneity.
56. A method for identifying a compound active against a viral infection comprising: providing a chimeric virus expressed from a viral construct comprising at least part of a
5' NTR from a HCV 5' NTR; contacting the virus with a candidate substance; and comparing the infectivity of the virus in the presence of the candidate substance with the infectivity of the virus in the absence of the candidate substance.
57. The method of claim 56, wherein the viral construct comprises at least a 5' NTR domain III from a HCV 5' NTR.
58. The method of claim 56, wherein the viral construct lacks a 5' NTR domain lb of GBV- B.
PCT/US2003/021002 2002-07-03 2003-07-02 Chimeric gb virus b (gbv-b) WO2004005498A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU2003248821A AU2003248821A1 (en) 2002-07-03 2003-07-02 Chimeric gb virus b (gbv-b)
US10/520,153 US7473772B2 (en) 2002-07-03 2003-07-02 Chimeric GB virus B (GBV-B)
EP03763178A EP1539938A4 (en) 2002-07-03 2003-07-02 Chimeric gb virus b (gbv-b)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10/189,359 US7141405B2 (en) 1999-06-04 2002-07-03 Chimeric GB virus B (GBV-B)
US10/189,359 2002-07-03

Publications (1)

Publication Number Publication Date
WO2004005498A1 true WO2004005498A1 (en) 2004-01-15

Family

ID=30114028

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/021002 WO2004005498A1 (en) 2002-07-03 2003-07-02 Chimeric gb virus b (gbv-b)

Country Status (4)

Country Link
US (2) US7141405B2 (en)
EP (1) EP1539938A4 (en)
AU (1) AU2003248821A1 (en)
WO (1) WO2004005498A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010008010A1 (en) * 2008-07-15 2010-01-21 株式会社先端生命科学研究所 Hcv/gbv-b chimeric virus
CN102154226A (en) * 2011-01-10 2011-08-17 南方医科大学 Chimeric virus of complete envelope protein of HCV (hepatitis C virus) and GB virus B

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7129342B1 (en) * 1999-06-04 2006-10-31 The United States Of America As Represented By The Department Of Health And Human Services Infectious cDNA clone of GB virus B and uses thereof
WO2006036879A2 (en) * 2004-09-27 2006-04-06 Board Of Regents, The University Of Texas System Chimeric gb virus b (gbv-b)
US20100129902A1 (en) * 2008-11-24 2010-05-27 Erhard Ralf Schoenbrunner Replication Stable and RNase Resistant Chimeras of Pestivirus with Insertion in 3' Nontranslated Region (3'NTR)
WO2016061210A2 (en) * 2014-10-14 2016-04-21 The Trustees Of Columbia University In The City Of New York Novel viruses found in rattus norvegicus and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000075337A1 (en) * 1999-06-04 2000-12-14 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services INFECTIOUS cDNA CLONE OF GB VIRUS B AND USES THEREOF
US20010034019A1 (en) * 1999-12-22 2001-10-25 Zhi Hong Chimeric HCV/GBV-B viruses

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5807670A (en) 1995-08-14 1998-09-15 Abbott Laboratories Detection of hepatitis GB virus genotypes
DE19853338B4 (en) * 1998-11-19 2008-04-17 Volkswagen Ag Arrangement with a front pillar for a body frame of a motor vehicle
GB9912432D0 (en) * 1999-05-27 1999-07-28 Angeletti P Ist Richerche Bio Novel gbv sequence

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000075337A1 (en) * 1999-06-04 2000-12-14 The Government Of The United States Of America As Represented By The Secretary, Department Of Health And Human Services INFECTIOUS cDNA CLONE OF GB VIRUS B AND USES THEREOF
US20010034019A1 (en) * 1999-12-22 2001-10-25 Zhi Hong Chimeric HCV/GBV-B viruses

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
FROLOV ET AL.: "Cis-acting RNA elements required for replication of bovine viral diarrhea virus-hepatitis C virus 5' nontranslated region chimeras", RNA, vol. 4, 1998, pages 1418 - 1435, XP002919368 *
LEMON ET AL.: "Internal ribosome entry sites within the RNA genomes of hepatitis C virus and other flaviviruses", SEMINARS IN VIROLOGY, vol. 8, 1997, pages 274 - 288, XP002973504 *
RIJNBRAND ET AL.: "Mutational analysis of the GB virus B internal ribosome entry site", JOURNAL OF VIROLOGY, vol. 74, no. 2, January 2000 (2000-01-01), pages 773 - 783, XP002973505 *
See also references of EP1539938A4 *
ZHAO ET AL.: "Poliovirus/hepatitis C virus (Internal ribosome entry site-core) chimeric viruses: improved growth properties through modification of a proteolytic cleavage site and requirement for core RNA sequences but not for core-related polypeptides", JOURNAL OF VIROLOGY, vol. 73, no. 2, February 1999 (1999-02-01), pages 1546 - 1554, XP001066303 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010008010A1 (en) * 2008-07-15 2010-01-21 株式会社先端生命科学研究所 Hcv/gbv-b chimeric virus
US8741607B2 (en) 2008-07-15 2014-06-03 Advanced Life Science Institute, Inc. HCV/GBV-B chimeric virus
CN102154226A (en) * 2011-01-10 2011-08-17 南方医科大学 Chimeric virus of complete envelope protein of HCV (hepatitis C virus) and GB virus B
CN102154226B (en) * 2011-01-10 2012-10-17 南方医科大学 Chimeric virus of complete envelope protein of HCV (hepatitis C virus) and GB virus B

Also Published As

Publication number Publication date
US7141405B2 (en) 2006-11-28
US7473772B2 (en) 2009-01-06
AU2003248821A8 (en) 2004-01-23
US20060160067A1 (en) 2006-07-20
EP1539938A1 (en) 2005-06-15
US20040039187A1 (en) 2004-02-26
EP1539938A4 (en) 2007-11-28
AU2003248821A1 (en) 2004-01-23

Similar Documents

Publication Publication Date Title
Khromykh et al. cis-and trans-acting elements in flavivirus RNA replication
Gu et al. The RNA helicase and nucleotide triphosphatase activities of the bovine viral diarrhea virus NS3 protein are essential for viral replication
Tilgner et al. Structure and function of the 3′ terminal six nucleotides of the West Nile virus genome in viral replication
Appel et al. Efficient rescue of hepatitis C virus RNA replication by trans-complementation with nonstructural protein 5A
EP0856051B1 (en) Novel 3' terminal sequence of hepatitis c virus genome and diagnostic and therapeutic uses thereof
Westaway et al. Replication and gene function in Kunjin virus
Grassmann et al. Assignment of the multifunctional NS3 protein of bovine viral diarrhea virus during RNA replication: an in vivo and in vitro study
US7935676B2 (en) Nucleic acid construct containing a nucleic acid derived from the genome of hepatitis C virus (HCV) of genotype 2A, and a cell having such nucleic acid construct introduced therein
CA2295552A1 (en) Cloned genomes of infectious hepatitis c viruses and uses thereof
Cheney et al. Mutations in NS5B polymerase of hepatitis C virus: impacts on in vitro enzymatic activity and viral RNA replication in the subgenomic replicon cell culture
Tellinghuisen et al. The NS5A protein of bovine viral diarrhea virus contains an essential zinc-binding site similar to that of the hepatitis C virus NS5A protein
JP2003533232A (en) HCV mutant
EP1801116A1 (en) HCV replicon shuttle vectors
Nam et al. In vivo analysis of the 3′ untranslated region of GB virus B after in vitro mutagenesis of an infectious cDNA clone: persistent infection in a transfected tamarin
Shoji et al. Proteolytic activity of NS3 serine proteinase of hepatitis C virus efficiently expressed in Escherichia coli
US7473772B2 (en) Chimeric GB virus B (GBV-B)
US20060105365A1 (en) Chimeric GB virus B (GBV-B)
US7476499B2 (en) Methods of identifying anti-viral agents
EP1185665B1 (en) INFECTIOUS cDNA CLONE OF GB VIRUS B AND USES THEREOF
WO2000075338A2 (en) CLONED GENONE OF INFECTIOUS HEPATITIS C VIRUS OF GENOTYPE 2a AND USES THEREOF
AU2008333339A1 (en) HCV NS3 protease replicon shuttle vectors
JP4573776B2 (en) Nucleic acid and gene derived from a novel HCV strain, and replicon-replicating cells using the gene
US7244585B1 (en) 3′ Sequence of the GB virus B (GBV-B) genome
CN104126008B (en) Nucleic acid construct including nucleic acid derived from genotype 3[alpha] HCV genome
Couto et al. Animal models for HCV study

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2003763178

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003763178

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2006160067

Country of ref document: US

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 10520153

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

WWP Wipo information: published in national office

Ref document number: 10520153

Country of ref document: US