WO1996015779A1 - Sequences virales specifiques associees a la maladie de kaposi et leurs utilisations - Google Patents

Sequences virales specifiques associees a la maladie de kaposi et leurs utilisations Download PDF

Info

Publication number
WO1996015779A1
WO1996015779A1 PCT/US1995/015138 US9515138W WO9615779A1 WO 1996015779 A1 WO1996015779 A1 WO 1996015779A1 US 9515138 W US9515138 W US 9515138W WO 9615779 A1 WO9615779 A1 WO 9615779A1
Authority
WO
WIPO (PCT)
Prior art keywords
kaposi
sarcoma
dna
subject
nucleic acid
Prior art date
Application number
PCT/US1995/015138
Other languages
English (en)
Inventor
Yuan Chang
Patrick S. Moore
Original Assignee
The Trustees Of Columbia University In The City Of New York
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
Priority claimed from US08/343,101 external-priority patent/US5830759A/en
Priority claimed from US08/420,235 external-priority patent/US5801042A/en
Application filed by The Trustees Of Columbia University In The City Of New York filed Critical The Trustees Of Columbia University In The City Of New York
Priority to AU43670/96A priority Critical patent/AU4367096A/en
Publication of WO1996015779A1 publication Critical patent/WO1996015779A1/fr
Priority to US08/757,669 priority patent/US6183751B1/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16411Rhadinovirus, e.g. human herpesvirus 8
    • C12N2710/16422New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Definitions

  • KS Kaposi's sarcoma
  • AIDS immunodeficiency syndrome
  • KS hemophiliac AIDS patients to develop KS
  • KS may be associated with specific sexual practices among gay men with AIDS [6, 15, 55, 83] .
  • KS is uncommon among adult AIDS patients infected through heterosexual or parenteral HIV transmission, or among pediatric AIDS patients infected through vertical HIV transmission [77] .
  • Agents previously suspected of causing KS include cytomega1ovirus, hepatitis B virus, human papillomavirus, Epstein-Barr virus, human herpesvirus 6, human immunodeficiency virus (HIV), and Mycoplasma penetrans [18, 23, 85, 91, 92] .
  • Non- infectious environmental agents such as nitrite inhalants
  • KS tumorigenesis also have been proposed to play a role in KS tumorigenesis [33] .
  • Extensive investigations however, have not demonstrated an etiologic association between any of these agents and AIDS-KS [37, 44, 46, 90] .
  • This invention provides an isolated DNA molecule which is at least 30 nucleotides in length and which uniquely defines a herpesvirus associated with Kaposi's sarcoma. This invention provides an isolated herpesvirus associated with Kaposi's sarcoma.
  • This invention provides a method of vaccinating a subject for KS, prophylaxis diagnosing or treating a subject with KS and detecting expression of a DNA virus associated with Kaposi's sarcoma in a cell.
  • FIG. 1 Agarose gel electrophoresis of RDA products from AIDS-KS tissue and uninvolved tissue.
  • RDA was performed on DNA extracted from KS skin tissue and uninvolved normal skin tissue obtained at autopsy from a homosexual man with AIDS-KS.
  • Lane 1 shows the initial PCR amplified genomic representation of the AIDS-KS DNA after Bam HI digestion.
  • Lanes 2-4 show that subsequent cycles of ligation, amplification, hybridization and digestion of the RDA products resulted in amplification of discrete bands at 380, 450, 540 and 680 bp.
  • RDA of the extracted AIDS-KS DNA performed against itself resulted in a single band at 540 bp (lane 5) .
  • Bands at 380 bp and 680 bp correspond to KS330Bam and KS627Bam respectively after removal of 28 bp priming sequences. Bands at 450 and 540 bp hybridized nonspecifically to both KS and non-KS human DNA. Lane M is a molecular weight marker.
  • Figures 3A-3F Nucleotide sequences of the DNA herpesvirus associated with KS (KSHV) .
  • Figure 4A shows the agarose gel of the amplification products from 19 KS DNA samples (lanes 1-19) and Figure 4B shows specific hybridization of the PCR products to a 32 P end- labelled 25 bp internal oligonucleotide ( Figure 3B) after transfer of the gel to a nitrocellulose filter.
  • Negative samples in lanes 3 and 15 respectively lacked microscopically detectable KS in the sample or did not amplify the constitutive p53 exon 6, suggesting that these samples were negative for technical reasons.
  • An additional 8 AIDS-KS samples were amplified and all were positive for KS330 234 .
  • Lane 20 is a negative control and Lane M is a molecular weight marker.
  • KS330Bam and KS627Bam failed to hybridize to the same fragments in the digests indicating that the two sequences are separated from each other by one or more intervening Bam HI restriction fragments. Digestion with Pvu II and hybridization to KS330Bam resulted in two distinct banding patterns (lanes 1 and 2 vs. lane 3) suggesting variation between KS samples.
  • Figure 8 A schematic diagram of the orientation of KSHV open reading frames identified on the KS5 20,710 bp DNA fragment .
  • Homologs to each open reading frame from a corresponding region of the herpesvirus saimiri (HSVSA) genome are present in an identical orientation, except for the region corresponding to the ORF 28 of HSVSA (middle schematic section) .
  • the shading for each open reading frame corresponds to the approximate % amino acid identity for the KSHV ORF compared to this homolog in HSVSA.
  • homologs that are present in this section of DNA include homologs to thy idine kinase (ORF21) , gH glycoprotein (ORF22) , major capsid protein (ORF25) and the VP23 protein (ORF26) which contains the original KS330Bam sequence derived by representational difference analysis.
  • M is molecular weight marker.
  • the antigen is a doublet between ca. 210 kD and 240 kD.
  • the 220 kD band is absent from the Western blots using patient sera without KS.
  • Figure 11 In this figure, 0.5 ml aliquots of the gradient have been fractionated (fractions 1-62) with the 30% gradient fraction being at fraction No. 1 and the 10% gradient fraction being at fraction No. 62. Each fraction has been dot hybridized to a nitrocellulose membrane and then a 32 P-labeled KSHV DNA fragment, KS631Bam has been hybridized to the membrane using standard techniques . The figure shows that the major solubilized fraction of the KSHV genome bands (i.e. is isolated) in fractions 42 through 48 of the gradient with a high concentration of the genome being present in fraction 44. A second band of solubilized KSHV DNA occurs in fractions 26 through 32.
  • KS5 open reading frames compared to translation products of herpesvirus saimiri (HSV) , equine herpesvirus 2 (EHV2) and Epstein-Barr virus (EBV) .
  • HSV herpesvirus saimiri
  • EHV2 equine herpesvirus 2
  • EBV Epstein-Barr virus
  • KS5 a 20.7 kb lambda phage clone insert derived from a human genomic library prepared from an AIDS-KS lesion. Seventeen partial and complete open reading frames (ORFs) are identified with arrows denoting reading frame orientations. Comparable regions of the Epstein- Barr virus (EBV) and herpesvirus saimiri (HVS) genomes are shown for comparison. Levels of amino acid similarity between KSHV ORFs are indicated by shading of EBV and HVS ORFs (black, over 70% similarity; dark gray, 55-70% similarity; light gray, 40-54% similarity; white, no detectable homology) . Domains of conserved herpesvirus sequence blocks and locations of restriction endonuclease sites used in subcloning are shown beneath the KSHV map (B, Bam HI site;
  • Figures 15A-15B Phylogenetic trees of KSHV based on comparison of aligned amino acid sequences between herpesviruses for the MCP gene and for a concatenated nine-gene set.
  • the comparison of MCP sequences ( Figure 15A) was obtained by the neighbor-joining method and is shown in unrooted form with branch lengths proportional to divergence (mean number of substitution events per site) between the nodes bounding each branch. Comparable results were obtained by maximum parsimony analysis. The number of times out of 100 bootstrap samplings the division indicated by each internal branch was obtained are shown next to each branch; bootstrap values below 75 are not shown.
  • Figure 15B is a phylogenetic tree of gammaherpesvirus sequences based on a nine-gene set CS1 (see text) and demonstrates that KSHV is most closely related to the gamma-2 herpesvirus sublineage, genus Rhadinovirus .
  • the CS1 amino acid sequence was used to infer a tree by the Protml maximum likelihood method; comparable results, not shown were obtained with the neighbor-joining and maximum parsimony methods.
  • the bootstrap value for the central branch is marked.
  • the root must lie between EBV and the other three species. Abbreviations for virus species used in the sequence comparisons are 1)
  • Alphaherpesvirinae HSV1 and HSV2, herpes simplex virus types 1 and 2; EHV1, equine herpesvirus 1; PRV, pseudorabies virus; and VZV, varicella-zoster virus, 2) Betaherpesvirinae: HCMV, human cytomegalovirus; HHV6 and HHV7, human herpesviruses 6 and 7, and 3) Gammaherpesvirinae: HVS, herpesvirus saimiri; EHV2, equine herpesvirus 2; EBV, Epstein-Barr virus; and Kaposi's sarcoma-associated herpesvirus.
  • Figures 16A-16B HSV1 and HSV2, herpes simplex virus types 1 and 2; EHV1, equine herpesvirus 1; PRV, pseudorabies virus; and VZV, varicella-zoster virus, 2) Betaherpesvirinae: HCMV, human cytomegal
  • KS631Bam hybridizes to a band at 270 kb as well as to a diffuse band at the origin.
  • the EBV termini sequence hybridizes to a 150-160 kb band consistent with the linear form of the genome.
  • Both KS631Bam (dark arrow) and an EBV terminal sequence hybridize to high molecular weight bands immediately below the origin indicating possible concatemeric or circular DNA. The high molecular weight KS631Bam hybridizing band reproduces poorly but is visible on the original autoradiographs.
  • Figures 18A-18C In si tu hybridization with an ORF26 oligomer to
  • RCC-1 a Raji cell line derived by cultivation of Raji with BCBL-1 in communicating chambers separated by a 0.45 ⁇ filter, shows rare cells with positive hybridization to the KSHV
  • nucleic acids refers to either DNA or RNA.
  • Nucleic acid sequence or “polynucleotide sequence” refers to a single- or double-stranded polymer of deoxyribonucleotide or ribonucleotide bases read from the 5' to the 3' end. It includes both self-replicating plasmids, infectious polymers of DNA or RNA and nonfunctional DNA or RNA.
  • nucleic acid sequence “homologous to” or “complementary to” it is meant a nucleic acid that selectively hybridizes, duplexes or binds to viral DNA sequences encoding proteins or portions thereof when the DNA sequences encoding the viral protein are present in a human genomic or cDNA library.
  • a DNA sequence which is homologous to a target sequence can include sequences which are shorter or longer than the target sequence so long as they meet the functional test set forth. Hybridization conditions are specified along with the source of the CDNA library.
  • the hybridization is done in a Southern blot protocol using a 0.2XSSC, 0.1% SDS, 65°C wash.
  • SSC refers to a citrate-saline solution of 0.15 M sodium chloride and 20 Mm sodium citrate. Solutions are often expressed as multiples or -
  • 6XSSC refers to a solution having a sodium chloride and sodium citrate concentration of 6 times this amount or 0.9 M sodium chloride and 120 mM sodium citrate.
  • 0.2XSSC refers to a solution 0.2 times the SSC concentration or 0.03 M sodium chloride and 4 mM sodium citrate.
  • nucleic acid molecule encoding refers to a nucleic acid molecule which directs the expression of a specific protein or peptide.
  • the nucleic acid sequences include both the DNA strand sequence that is transcribed into RNA and the RNA sequence that is translated into protein.
  • the nucleic acid molecule include both the full length nucleic acid sequences as well as non-full length sequences derived from the full length protein. It being further understood that the sequence includes the degenerate codons of the native sequence or sequences which may be introduced to provide codon preference in a specific host cell.
  • expression cassette refers to nucleotide sequences which are capable of affecting expression of a structural gene in hosts compatible with such sequences.
  • Such cassettes include at least promoters and optionally, transcription termination signals. Additional factors necessary or helpful in effecting expression may also be used as described herein.
  • operably linked refers to linkage of a promoter upstream from a DNA sequence such that the promoter mediates transcription of the DNA sequence.
  • vector refers to viral expression systems, autonomous self-replicating circular DNA (plasmids) , and includes both expression and nonexpression plasmids. Where a recombinant microorganism or cell culture is described as hosting an “expression vector, " this includes both extrachromosomal circular DNA and DNA that has been incorporated into the host chromosome(s) . Where a vector is being maintained by a host cell, the vector may either be stably replicated by the cells during mitosis as an autonomous structure, or is incorporated within the host' s genome.
  • plasmid refers to an autonomous circular DNA molecule capable of replication in a cell, and includes both the expression and nonexpression types. Where a recombinant microorganism or cell culture is described as hosting an "expression plasmid", this includes latent viral DNA integrated into the host chromosome (s) . Where a plasmid is being maintained by a host cell, the plasmid is either being stably replicated by the cells during mitosis as an autonomous structure or is incorporated within the host's genome.
  • recombinant protein or “recombinantly produced protein” refers to a peptide or protein produced using non-native cells that do not have an endogenous copy of DNA able to express the protein.
  • the cells produce the protein because they have been genetically altered by the introduction of the appropriate nucleic acid sequence.
  • the recombinant protein will not be found in association with proteins and other subcellular components normally associated with the cells producing the protein.
  • a “reference sequence” is a defined sequence used as a basis for a sequence comparison; a reference sequence may be a subset of a larger sequence, for example, as a segment of a full-length cDNA or gene sequence given in a sequence listing or may comprise a complete cDNA or gene sequence.
  • Optimal alignment of sequences for aligning a comparison window may be conducted by the local homology algorithm of Smith and Waterman (1981) Adv. Appl . Math . 2:482, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol . Biol . 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc. Natl . Acad. Sci . (USA) 85:2444, or by computerized implementations of these algorithms
  • the terms "substantial identity” or “substantial sequence identity” mean that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap which share at least 90 percent sequence identity, preferably at least 95 percent sequence identity, more preferably at least 99 percent sequence identity or more.
  • Percentage amino acid identity or “percentage amino acid sequence identity” refers to a comparison of the amino acids of two polypeptides which, when optimally aligned, have approximately the designated percentage of the same amino acids.
  • 95% amino acid identity refers to a comparison of the amino acids of two polypeptides which when optimally aligned have 95% amino acid identity.
  • residue positions which are not identical differ by conservative amino acid substitutions. For example, the substitution of amino acids having similar chemical properties such as charge or polarity are not likely to effect the properties of a protein. Examples include glutamine for asparagine or glutamic acid for aspartic acid.
  • substantially purified when referring to a herpesvirus peptide or protein, means a chemical composition which is essentially free of other cellular components. It is preferably in a homogeneous state although it can be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography.
  • a protein which is the predominant species present in a preparation is substantially purified. Generally, a substantially purified or isolated protein will comprise more than 80% of all macromolecular species present in the preparation. Preferably, the protein is purified to represent greater than 90% of all macromolecular species present . More preferably the protein is purified to greater than 95%, and most preferably the protein is purified to essential homogeneity, wherein other macromolecular species are not detected by conventional techniques.
  • the specified antibodies bind to the herpesvirus antigens and do not bind in a significant amount to other antigens present in the sample.
  • Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity for a particular protein.
  • antibodies raised to the human herpesvirus immunogen described herein can be selected to obtain antibodies specifically immunoreactive with the herpesvirus proteins and not with other proteins. These antibodies recognize proteins homologous to the human herpesvirus protein.
  • a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein.
  • solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow and Lane [32] for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
  • Bio sample refers to any sample obtained from a living organism or from an organism that has died. Examples of biological samples include body fluids and tissue specimens.
  • KS Kaposis's Sarcoma
  • This invention provides an isolated DNA molecule which is at least 30 nucleotides in length and which uniquely defines a herpesvirus associated with Kaposi's sarcoma.
  • the isolated DNA molecule comprises at least a portion of the nucleic acid sequence as shown in Figure 3A (SEQ ID NO: 1) .
  • the isolated DNA molecule is a 330 base pair (bp) sequence.
  • the isolated DNA molecule is a 12-50 bp sequence.
  • the isolated DNA molecule is a 30- 37 bp sequence.
  • the isolated DNA molecule is genomic DNA. In another embodiment the isolated DNA molecule is cDNA. In another embodiment a RNA is derived form the isolated nucleic acid molecule or is capable of hybridizing with the isolated DNA molecule. As used herein "genomic" means both coding and non- coding regions of the isolated nucleic acid molecule.
  • the DNA molecule above may be associated with lymphoproliferative diseases including, but not limited to: Hodgkin's disease, non-Hodgkin' s lymphoma, lymphatic leukemia, lymphosarcoma, splenomegaly, reticular cell sarcoma, Sezary's syndrome, mycosis fungoides, central nervous system lymphoma, AIDS related central nervous system lymphoma, post- transplant lymphoproliferative disorders, and Burkitt's lymphoma.
  • a lymphoproliferative disorder is characterized as being the uncontrolled clonal or polyclonal expansion of lymphocytes involving lymph nodes, lymphoid tissue and other organs.
  • This invention provides an isolated nucleic acid molecule encoding an ORF20 (SEQ ID NOs: 22 and 23) , ORF21 (SEQ ID NOs:14 and 15), ORF22 (SEQ ID NOs: 16 and 17), ORF23 (SEQ ID NOs: 18 and 19) , ORF24 (SEQ ID NOs : 20 and 21), ORF25 (SEQ ID NOs: 2 and 3), ORF26 (SEQ ID NOs:
  • ORF29B (SEQ ID NOs :4 and 5)
  • ORF30 (SEQ ID NOs:6 and
  • ORF31 SEQ ID NOs: 8 and 9
  • ORF32 SEQ ID NOs:32 and 33
  • ORF33 SEQ ID NOs: 10 and 11
  • ORF34 SEQ ID NOs: 34 and 35
  • ORF35 SEQ ID NOs :12 AND 13
  • This invention provides an isolated polypeptide encoded by ORF20 (SEQ ID NOs: 22 and 23) , ORF21 (SEQ ID NOs:14 and 15), ORF22 (SEQ ID NOs:16 and 17) , ORF23
  • ORF27 SEQ ID NOs:26 and 27
  • ORF28 SEQ ID NOs:28
  • ORF29A SEQ ID NOs:30 and 31
  • ORF29B SEQ ID NOs:30 and 31
  • TK is encoded by ORF 21; glycoprotein H (gH) by ORF 22; major capsid protein (MCP) by ORF 25; virion polypeptide (VP23) by ORF 26; and minor capsid protein by ORF 27.
  • GH glycoprotein H
  • MCP major capsid protein
  • VP23 virion polypeptide
  • ORF 27 minor capsid protein
  • This invention provides for a replicable vector comprising the isolated DNA molecule of the DNA virus.
  • the vector includes, but is not limited to: a plasmid, cosmid, ⁇ phage or yeast artificial chromosome (YAC) which contains at least a portion of the isolated nucleic acid molecule.
  • YAC yeast artificial chromosome
  • insert and vector DNA can both be exposed to a restriction enzyme to create complementary ends on both molecules which base pair with each other and are then ligated together with DNA ligase.
  • linkers can be ligated to the insert DNA which correspond to a restriction site in the vector DNA, which is then digested with the restriction enzyme which cuts at that site.
  • Other means are also available and known to an ordinary skilled practitioner.
  • a bacterial expression vector includes a promoter such as the lac promoter and for transcription initiation the Shine-Dalgarno sequence and the start codon AUG.
  • a eukaryotic expression vector includes a heterologous or homologous promoter for RNA polymerase II, a downstream polyadenylation signal, the start codon AUG, and a termination codon for detachment of the ribosome.
  • Such vectors may be obtained commercially or assembled from the sequences described by methods well-known in the art, for example the methods described above for constructing vectors in general.
  • the host cell may contain the isolated DNA molecule artificially introduced into the host cell.
  • the host cell may be a eukaryotic or bacterial cell (such as E.coli) , yeast cells, fungal cells, insect cells and animal cells.
  • Suitable animal cells include, but are not limited to Vero cells, HeLa cells, Cos cells, CV1 cells and various primary mammalian cells.
  • herpesvirus associated with Kaposi's sarcoma.
  • the herpesvirus comprises at least a portion of a nucleotide sequence as shown in Figures 3A (SEQ ID NO: 1) .
  • the herpesvirus may be a DNA virus .
  • the herpesvirus may be a Herpesviridae.
  • the herpesvirus may be a gammaherpesvirinae.
  • the classification of the herpesvirus may vary based on the phenotypic or molecular characteristics which are known to those skilled in the art.
  • This invention provides an isolated DNA virus wherein the viral DNA is about 270 kb in size, wherein the viral DNA encodes a thymidine kinase, and wherein the viral DNA is capable of selectively hybridizing to a nucleic acid probe selected from the group consisting Of SEQ ID NOs: 38-40.
  • the KS-associated human herpesvirus of the invention is associated with KS and is involved in the etiology of the disease.
  • the taxonomic classification of the virus has not yet been made and will be based on phenotypic or molecular characteristics known to those of skill in the art.
  • the novel KS-associated virus is a DNA virus that appears to be related to the Herpesviridae family and the gammaherpesvirinae subfamily, on the basis of nucleic acid homology.
  • the human herpesvirus of the invention is not limited to the virus having the specific DNA sequences described herein.
  • the KS-associated human herpesvirus DNA shows substantial sequence identity, as defined above, to the viral DNA sequences described herein.
  • DNA from the human herpesvirus typically selectively hybridizes to one or more of the following three nucleic acid probes :
  • GAAATTACCC ACGAGATCGC TTCCCTGCAC ACCGCACTTG GCTACTCATC AGTCATCGCC CCGGCCCACG TGGCCGCCAT AACTACAGAC ATGGGAGTAC ATTGTCAGGA CCTCTTTATG ATTTTCCCAG GGGACGCGTA TCAGGACCGC CAGCTGCATG ACTATATCAA AATGAAAGCG GGCGTGCAAA CCGGCTCACC GGGAAACAGA ATGGATCACG TGGGATACAC TGCTGGGGTT CCTCGCTGCG AGAACCTGCC CGGTTTGAGT CATGGTCAGC TGGCAACCTG CGAGATAATT CCCACGCCGG TCACATCTGA CGTTGCCT
  • Probe 3 SEQ ID NO: 40
  • Hybridization of a viral DNA to the nucleic acid probes listed above is determined by using standard nucleic acid hybridization techniques as described herein.
  • PCR amplification of a viral genome can be carried out using the following three sets of PCR primers:
  • oligonucleotide primers as listed above, complementary to the two 3' borders of the DNA region to be amplified are synthesized. The polymerase chain reaction is then carried out using the two primers. See PCR Protocols : A Guide to Methods and Applications [74] .
  • the PCR-amplified regions of a viral DNA can be tested for their ability to hybridize to the three specific nucleic acid probes listed above.
  • hybridization of a viral DNA to the above nucleic acid probes can be performed by a Southern blot procedure without viral DNA amplification and under stringent hybridization conditions as described herein.
  • Oligonucleotides for use as probes or PCR primers are chemically synthesized according to the solid phase phosphoramidite triester method first described by Beaucage and Carruthers [19] using an automated synthesizer, as described in Needham-VanDevanter [69] . Purification of oligonucleotides is by either native acrylamide gel electrophoresis or by anion-exchange HPLC as described in Pearson, J.D. and Regnier, F.E. [75A] . The sequence of the synthetic oligonucleotide can be verified using the chemical degradation method of Maxam, A.M. and Gilbert, W. [63] .
  • the human herpesvirus can be propagated in vi tro .
  • standard techniques for growing herpes viruses are described in Ablashi, D.V. [1] . Briefly, PHA stimulated cord blood mononuclear cells, macrophage, neuronal, or glial cell lines are cocultivated with cerebrospinal fluid, plasma, peripheral blood leukocytes, or tissue extracts containing viral infected cells or purified virus. The recipient cells are treated with 5 ⁇ g/ml polybrene for 2 hours at 37° C prior to infection. Infected cells are observed by demonstrating morphological changes, as well as being positive for antigens from the human herpesvirus by using monoclonal antibodies immunoreactive with the human herpes virus in an immunofluorescence assay.
  • the virus is either harvested directly from the culture fluid by direct centrifugation, or the infected cells are harvested, homogenized or Iysed and the virus is separated from cellular debris and purified by standard methods of isopycnic sucrose density gradient centrifugation.
  • KSHV Kaposi's sarcoma
  • the KS associated herpesvirus may be isolated from the cell DNA in the following manner.
  • An infected cell line (BHL-6 RCC-1) , which can be Iysed using standard methods such as hyposomatic shocking and Dounce homogenization, is first pelleted at 2000xg for 10 minutes, the supernatant is removed and centrifuged again at 10,000xg for 15 minutes to remove nuclei and organelles. The supernatant is filtered through a 0.45 ⁇ filter and centrifuged again at 100,000xg for 1 hour to pellet the virus. The virus can then be washed and centrifuged again at 100,000xg for 1 hour.
  • the DNA is tested for the presence of the KSHV by Southern blotting and PCR using the specific probes as described hereinafter.
  • Fresh lymphoma tissue containing viable infected cells is simultaneously filtered to form a single cell suspension by standard techniques [49, 66] .
  • the cells are separated by standard Ficoll-Plaque centrifugation and lymphocyte layer is removed.
  • the lymphocytes are then placed at >lxl0 6 cells/ml into standard lymphocyte tissue culture medium, such as RMP 1640 supplemented with 10% fetal calf serum.
  • Immortalized lymphocytes containing the KSHV virus are indefinitely grown in the culture media while nonimmortilized cells die during course of prolonged cultivation.
  • the virus may be propagated in a new cell line by removing media supernatant containing the virus from a continuously infected cell line at a concentration of >lxl0 6 cells/ml.
  • the media is centrifuged at 2000xg for 10 minutes and filtered through a 0.45 ⁇ filter to remove cells.
  • the media is applied in a 1:1 volume with cells growing at >lxl0 6 cells/ml for 48 hours. The cells are washed and pelleted and placed in fresh culture medium, and tested after 14 days of growth.
  • RCC-1 and RCC-1 2F5 were deposited on October 19, 1994 under ATCC Accession No. CRL 11734 and CRL 11735, respectively, pursuant to the Budapest Treaty on the International Deposit of Microorganisms for the Purposes of Patent Procedure with the Patent Culture Depository of the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland 20852 U.S.A. BHL-6 was deposited on November 18, 1994 under ATCC Accession No. CRL 11762 pursuant to the Budapest Treaty on the International Deposit of Microorganisms for the Purposes of Patent Procedure with the Patent Culture Depository of the American Type Culture Collection, 12301 Parklawn Drive, Rockville, Maryland 20852 U.S.A. C. Immunolo ⁇ ical Identity of the Virus
  • KS-associated human herpesvirus can also be described immunologically.
  • KS-associated human herpesviruses are selectively immunoreactive to antisera generated against a defined immunogen such as the viral major capsid protein depicted in Seq. ID No. 12, herein. Immunoreactivity is determined in an immunoassay using a polyclonal antiserum which was raised to the protein which is encoded by the amino acid sequence or nucleic acid sequence of SEQ ID NOs : 18-20. This antiserum is selected to have low crossreactivity against other herpes viruses and any such crossreactivity is removed by immunoabsorbtion prior to use in the immunoassay.
  • the protein which is encoded by the amino acid sequence or nucleic acid of SEQ ID NOs: 18-20 is isolated as described herein.
  • recombinant protein can be produced in a mammalian cell line.
  • An inbred strain of mice such as balb/c is immunized with the protein which is encoded by the amino acid sequence or nucleic acid of SEQ ID NOs: 2- 37 using a standard adjuvant, such as Freund's adjuvant, and a standard mouse immunization protocol (see [32] , supra) .
  • a synthetic peptide derived from the sequences disclosed herein and conjugated to a carrier protein can be used an immunogen.
  • Polyclonal sera are collected and titered against the immunogen protein in an immunoassay, for example, a solid phase immunoassay with the immunogen immobilized on a solid support.
  • Polyclonal antisera with a titer of 10 4 or greater are selected and tested for their cross reactivity against other viruses of the gammaherpesvirinae subfamily, particularly human herpes virus types 1-7, by using a standard immunoassay as described in [32] , supra .
  • These other gammaherpesvirinae virus can be isolated by standard techniques for isolation herpes viruses as described herein.
  • the ability of the above viruses to compete with the binding of the antisera to the immunogen protein is determined.
  • the percent crossreactivity for other viruses is calculated, using standard calculations. Those antisera with less than 10% crossreactivity with each of the other viruses listed above is selected and pooled. The cross-reacting antibodies are then removed from the pooled antisera by immunoabsorption with the above-listed viruses.
  • the immunoabsorbed and pooled antisera are then used in a competitive binding immunoassay procedure as described above to compare an unknown virus preparation to the specific KS herpesvirus preparation described herein and containing the nucleic acid sequence described in SEQ ID NOs: 2-37.
  • the immunogen protein which is encoded by the amino acid sequence or nucleic acid of SEQ ID NOs : 2-37 is the labeled antigen and the virus preparations are each assayed at a wide range of concentrations. The amount of each virus preparation required to inhibit 50% of the binding of the antisera to the labeled immunogen protein is determined.
  • viruses that specifically bind to an antibody generated to an immunogen consisting of the protein of SEQ ID NOs: 2-37 are those virus where the amount of virus needed to inhibit 50% of the binding to the protein does not exceed an established amount. This amount is no more than 10 times the amount of the virus that is needed for 50% inhibition for the KS- associated herpesvirus containing the DNA sequence of SEQ ID NO: 1.
  • the KS-associated herpesviruses of the invention can be defined by immunological comparison to the specific strain of the KS-associated herpesvirus for which nucleic acid sequences are provided herein.
  • nucleic acid molecule of at least 14 nucleotides capable of specifically hybridizing with the isolated DNA molecule.
  • the molecule is DNA.
  • the molecule is RNA.
  • nucleic acid molecule may be 14-20 nucleotides in length.
  • nucleic acid molecule may be 16 nucleotides in length.
  • This invention provides, a nucleic acid molecule of at least 14 nucleotides capable of specifically hybridizing with a nucleic acid molecule which is complementary to the isolated DNA molecule.
  • the molecule is DNA.
  • the molecule is RNA.
  • the nucleic acid molecule of at least 14 nucleotides may hybridize with moderate stringency to at least a portion of a nucleic acid molecule with a sequence shown in Figures 3A-3F (SEQ ID NOs: 1, 10-17, and 38- 40) .
  • High stringent hybridization conditions are selected at about 5° C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH.
  • Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe.
  • stringent conditions will be those in which the salt concentration is at least about 0.02 molar at pH 7 and the temperature is at least about 60°C.
  • the combination of parameters is more important than the absolute measure of any one.
  • Example high stringency may be attained for example by overnight hybridization at about 68°C in a 6x SSC solution, washing at room temperature with 6x SSC solution, followed by washing at about 68°C in a 6x SSC in a 0.6x SSX solution.
  • Hybridization with moderate stringency may be attained for example by: 1) filter pre-hybridizing and hybridizing with a solution of 3x sodium chloride, sodium citrate (SSC) , 50% formamide, 0.1M Tris buffer at Ph 7.5, 5x Denhardt's solution; 2.) pre- hybridization at 37°C for 4 hours; 3) hybridization at 37°C with amount of labelled probe equal to 3,000,000 cpm total for 16 hours; 4) wash in 2x SSC and 0.1% SDS solution; 5) wash 4x for 1 minute each at room temperature at 4x at 60°C for 30 minutes each; and 6) dry and expose to film.
  • SSC sodium citrate
  • selectively hybridizing to refers to a nucleic acid probe that hybridizes, duplexes or binds only to a particular target DNA or RNA sequence when the target sequences are present in a preparation of total cellular DNA or RNA.
  • selectively hybridizing it is meant that a probe binds to a given target in a manner that is detectable in a different manner from non-target sequence under high stringency conditions of hybridization, in a different "Complementary" or “target” nucleic acid sequences refer to those nucleic acid sequences which selectively hybridize to a nucleic acid probe.
  • Proper annealing conditions depend, for example, upon a probe's length, base composition, and the number of mismatches and their position on the probe, and must often be determined empirically.
  • nucleic acid probe design and annealing conditions see, for example, Sa brook et al . , [81] or Ausubel, F., et al . , [8] .
  • Nucleic acid probe technology is well known to those skilled in the art who readily appreciate that such probes may vary greatly in length and may be labeled with a detectable label, such as a radioisotope or fluorescent dye, to facilitate detection of the probe.
  • DNA probe molecules may be produced by insertion of a DNA molecule having the full-length or a fragment of the isolated nucleic acid molecule of the DNA virus into suitable vectors, such as plasmids or bacteriophages, followed by transforming into suitable bacterial host cells, replication in the transformed bacterial host cells and harvesting of the DNA probes, using methods well known in the art.
  • probes may be generated chemically from DNA synthesizers.
  • DNA virus nucleic acid rearrangements/mutations may be detected by Southern blotting, single stranded conformational polymorphism gel electrophoresis (SSCP) , PCR or other DNA based techniques, or for RNA species by Northern blotting, PCR or other RNA-based techniques.
  • SSCP single stranded conformational polymorphism gel electrophoresis
  • RNA probes may be generated by inserting the full length or a fragment of the isolated nucleic acid molecule of the DNA virus downstream of a bacteriophage promoter such as T3, T7 or SP6. Large amounts of RNA probe may be produced by incubating the labeled nucleotides with a linearized isolated nucleic acid molecule of the DNA virus or its fragment where it contains an upstream promoter in the presence of the appropriate RNA polymerase.
  • nucleic acid probes may be DNA or RNA fragments.
  • DNA fragments can be prepared, for example, by digesting plasmid DNA, or by use of PCR, or synthesized by either the phosphoramidite method described by Beaucage and Carruthers, [19] , or by the triester method according to Matteucci, et al . , [62] , both incorporated herein by reference.
  • a double stranded fragment may then be obtained, if desired, by annealing the chemically synthesized single strands together under appropriate conditions or by synthesizing the complementary strand using DNA polymerase with an appropriate primer sequence.
  • nucleic acid probe where a specific sequence for a nucleic acid probe is given, it is understood that the complementary strand is also identified and included. The complementary strand will work equally well in situations where the target is a double-stranded nucleic acid. It is also understood that when a specific sequence is identified for use a nucleic probe, a subsequence of the listed sequence which is 25 basepairs or more in length is also encompassed for use as a probe.
  • the DNA molecules of the subject invention also include DNA molecules coding for polypeptide analogs, fragments or derivatives of antigenic polypeptides which differ from naturally-occurring forms in terms of the identity or location of one or more amino acid residues (deletion analogs containing less than all of the residues specified for the protein, substitution analogs wherein one or more residues specified are replaced by other residues and addition analogs where in one or more amino acid residues is added to a terminal or medial portion of the polypeptides) and which share some or all properties of naturally- occurring forms.
  • These molecules include: the incorporation of codons "preferred" for expression by selected non-mammalian hosts; the provision of sites for cleavage by restriction endonuclease enzymes; and the provision of additional initial, terminal or intermediate DNA sequences that facilitate construction of readily expressed vectors.
  • This invention provides for an isolated DNA molecule which encodes at least a portion of a Kaposi's sarcoma associated herpesvirus: virion polypeptide 23, major capsid protein, capsid proteins, thymidine kinase, or tegument protein.
  • This invention also provides a method of producing a polypeptide encoded by isolated DNA molecule, which comprises growing the above host vector system under suitable conditions permitting production of the polypeptide and recovering the polypeptide so produced.
  • This invention provides an isolated peptide encoded by the isolated DNA molecule associated with Kaposi's sarcoma.
  • the peptide may be a polypeptide.
  • this invention provides a host cell which expresses the polypeptide of isolated DNA molecule.
  • the isolated peptide or polypeptide is encoded by at least a portion of an isolated DNA molecule. In another embodiment the isolated peptide or polypeptide is encoded by at least a portion of a nucleic acid molecule with a sequence as set forth in (SEQ ID NOs: 2-37) .
  • the isolated peptide or polypeptide encoded by the isolated DNA molecule may be linked to a second nucleic acid molecule to form a fusion protein by expression in a suitable host cell.
  • the second nucleic acid molecule encodes beta- galactosidase.
  • Other nucleic acid molecules which are used to form a fusion protein are known to those skilled in the art.
  • This invention provides an antibody which specifically binds to the peptide or polypeptide encoded by the isolated DNA molecule.
  • the antibody is a monoclonal antibody. In another embodiment the antibody is a polyclonal antibody.
  • the antibody or DNA molecule may be labelled with a detectable marker including, but not limited to: a radioactive label, or a colorimetric, a luminescent, or a fluorescent marker, or gold.
  • Radioactive labels include, but are not limited to: 3 H, 14 C, 2 P, 33 P; 35 S, 36 CI, 51 Cr, 57 Co, 59 Co, 59 Fe?° Y 1 , 25 _ 31 I, atffcl Re.
  • Fluorescent markers include but are not limited to: fluorescein, rhodamine and auramine .
  • Colorimetric markers include, but are not limited to: biotin, and digoxigenin. Methods of producing the polyclonal or monoclonal antibody are known to those of ordinary skill in the art.
  • the antibody or nucleic acid molecule complex may be detected by a second antibody which may be linked to an enzyme, such as alkaline phosphatase or horseradish peroxidase.
  • an enzyme such as alkaline phosphatase or horseradish peroxidase.
  • Other enzymes which may be employed are well known to one of ordinary skill in the art.
  • This invention provides a method to select specific regions on the polypeptide encoded by the isolated DNA molecule of the DNA virus to generate antibodies.
  • the protein sequence may be determined from the cDNA sequence.
  • Amino acid sequences may be analyzed by methods well known to those skilled in the art to determine whether they produce hydrophobic or hydrophilic regions in the proteins which they build.
  • hydrophobic regions are well known to form the part of the protein that is inserted into the lipid bilayer of the cell membrane, while hydrophilic regions are located on the cell surface, in an aqueous environment. Usually, the hydrophilic regions will be more immunogenic than the hydrophobic regions.
  • hydrophilic amino acid sequences may be selected and used to generate antibodies specific to polypeptide encoded by the isolated nucleic acid molecule encoding the DNA virus.
  • the selected peptides may be prepared using commercially available machines.
  • DNA such as a cDNA or a fragment thereof, may be cloned and expressed and the resulting polypeptide recovered and used as an immunogen.
  • Polyclonal antibodies against these peptides may be produced by immunizing animals using the selected peptides.
  • Monoclonal antibodies are prepared using hybridoma technology by fusing antibody producing B cells from immunized animals with myeloma cells and selecting the resulting hybridoma cell line producing the desired antibody.
  • monoclonal antibodies may be produced by in vitro techniques known to a person of ordinary skill in the art. These antibodies are useful to detect the expression of polypeptide encoded by the isolated DNA molecule of the DNA virus in living animals, in humans, or in biological tissues or fluids isolated from animals or humans .
  • the antibodies raised against the viral strain or peptides may be detectably labelled, utilizing conventional labelling techniques well-known to the art.
  • the antibodies may be radiolabelled using, for example, radioactive isotopes such as 3 H, 125 I, 131 I, and 35 S.
  • the antibodies may also be labelled using fluorescent labels, enzyme labels, free radical labels, or bacteriophage labels, using techniques known in the art.
  • fluorescent labels include fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, alophycocyanin, and Texas Red.
  • enzymes may be coupled to other molecules by covalent links, the possibility also exists that they might be used as labels for the production of tracer materials.
  • Suitable enzymes include alkaline phosphatase, beta-galactosidase, glucose-6 -phosphate dehydrogenase, maleate dehydrogenase, and peroxidase.
  • Two principal types of enzyme immunoassay are the enzyme-linked immunosorbent assay (ELISA) , and the homogeneous enzyme immunoassay, also known as enzyme-multiplied immunoassay (EMIT, Syva Corporation, Palo Alto, CA) .
  • ELISA enzyme-linked immunosorbent assay
  • EMIT enzyme-multiplied immunoassay
  • separation may be achieved, for example, by the use of antibodies coupled to a solid phase.
  • the EMIT system depends on deactivation of the enzyme in the tracer-antibody complex; the activity can thus be measured without the need for a separation step.
  • chemiluminescent compounds may be used as labels.
  • Typical chemiluminescent compounds include luminol, isoluminol, aromatic acridinium esters, imidazoles, acridinium salts, and oxalate esters.
  • bioluminescent compounds may be utilized for labelling, the bioluminescent compounds including luciferin, luciferase, and aequorin.
  • the antibody may be employed to identify and quantify immunologic counterparts (antibody or antigenic polypeptide) utilizing techniques well-known to the art.
  • RIA radioimmunoassay
  • antibodies to the human herpesvirus can be used to detect the agent in the sample.
  • the sequence being targeted is expressed in transfected cells, preferably bacterial cells, and purified.
  • the product is injected into a mammal capable of producing antibodies.
  • Either monoclonal or polyclonal antibodies (as well as any recombinant antibodies) specific for the gene product can be used in various immunoassays.
  • assays include competitive immunoassays, radioimmunoassays, Western blots, ELISA, indirect immunofluorescent assays and the like. For competitive immunoassays, see Harlow and Lane [32] at pages 567-573 and 584-589.
  • Monoclonal antibodies or recombinant antibodies may be obtained by various techniques familiar to those skilled in the art. Briefly, spleen cells or other lymphocytes from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell (see, Kohler and Milstein [50] , incorporated herein by reference) . Alternative methods of immortalization include transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods well known in the art.
  • Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host . New techniques using recombinant phage antibody expression systems can also be used to generate monoclonal antibodies. See for example: McCafferty, J et al . [64] ; Hoogenboom, H.R. et al . [39] ; and Marks, J.D. et al . [60] .
  • Such peptides may be produced by expressing the specific sequence in a recombinantly engineered cell such as bacteria, yeast, filamentous fungal, insect
  • herpes virus protein especially employing baculoviral vectors, and mammalian cells.
  • baculoviral vectors especially employing baculoviral vectors
  • mammalian cells especially employing baculoviral vectors
  • Those of skill in the art are knowledgeable in the numerous expression systems available for expression of herpes virus protein.
  • the expression of natural or synthetic nucleic acids encoding viral protein will typically be achieved by operably linking the desired sequence or portion thereof to a promoter (which is either constitutive or inducible) , and incorporated into an expression vector.
  • the vectors are suitable for replication or integration in either prokaryotes or eukaryotes.
  • Typical cloning vectors contain antibiotic resistance markers, genes for selection of transformants, inducible or regulatable promoter regions, and translation terminators that are useful for the expression of viral genes.
  • Suitable eukaryote hosts may include plant cells, insect cells, mammalian cells, yeast, and filamentous f ngi. Methods for characterizing naturally processed peptides bound to MHC (major histocompatibility complex) I molecules have been developed.
  • peptides described herein produced by recombinant technology may be purified by standard techniques well known to those of skill in the art.
  • Recombinantly produced viral sequences can be directly expressed or expressed as a fusion protein.
  • the protein is then purified by a combination of cell lysis (e.g., sonication) and affinity chromatography. For fusion products, subsequent digestion of the fusion protein with an appropriate proteolytic enzyme releases the desired peptide.
  • the proteins may be purified to substantial purity by standard techniques well known in the art, including selective precipitation with such substances as ammonium sulfate, column chromatography, immunopurification methods, and others. See, for instance, Scopes, R. [84] , incorporated herein by reference.
  • This invention further embraces diagnostic kits for detecting the presence of a KS agent in biological samples, such as serum or solid tissue samples, comprising a container containing antibodies to the human herpesvirus, and instructional material for performing the test.
  • diagnostic kits for detecting the presence of a KS agent in biological samples comprising a container containing antibodies to the human herpesvirus, and instructional material for performing the test.
  • inactivated viral particles or peptides or viral proteins derived from the human herpesvirus may be used in a diagnostic kit to detect for antibodies specific to the KS associated human herpesvirus.
  • Diagnostic kits for detecting the presence of a KS agent in tissue samples comprising a container containing a nucleic acid sequence specific for the human herpesvirus and instructional material for detecting the KS-associated herpesvirus are also included.
  • a container containing nucleic acid primers to any one of such sequences is optionally included as are antibodies to the human herpesvirus as described herein.
  • Antibodies reactive with antigens of the human herpesvirus can also be measured by a variety of immunoassay methods that are similar to the procedures described above for measurement of antigens.
  • immunoassay methods that are similar to the procedures described above for measurement of antigens.
  • immunoassays to measure antibodies reactive with antigens of the KS-associated human herpesvirus can be either competitive or noncompetitive binding assays.
  • competitive binding assays the sample analyte competes with a labeled analyte for specific binding sites on a capture agent bound to a solid surface.
  • the capture agent is a purified recombinant human herpesvirus protein produced as described above.
  • Other sources of human herpesvirus proteins, including isolated or partially purified naturally occurring protein may also be used.
  • Noncompetitive assays are typically sandwich assays, in which the sample analyte is bound between two analyte-specific binding reagents.
  • binding agents One of the binding agents is used as a capture agent and is bound to a solid surface.
  • the second binding agent is labelled and is used to measure or detect the resultant complex by visual or instrument means.
  • a number of combinations of capture agent and labelled binding agent can be used.
  • a variety of different immunoassay formats, separation techniques and labels can be also be used similar to those described above for the measurement of the human herpesvirus antigens.
  • HI Hemagglutination Inhibition
  • CF Complement Fixation
  • Serological methods can be also be useful when one wishes to detect antibody to a specific variant. For example, one may wish to see how well a vaccine recipient has responded to the new variant. Alternatively, one may take serum from a patient to see which variant the patient responds to the best.
  • This invention provides an antagonist capable of blocking the expression of the peptide or polypeptide encoded by the isolated DNA molecule.
  • the antagonist is capable of hybridizing with a double stranded DNA molecule.
  • the antagonist is a triplex oligonucleotide capable of hybridizing to the DNA molecule.
  • the triplex oligonucleotide is capable of binding to at least a portion of the isolated DNA molecule with a nucleotide sequence as shown in Figure 3A-3F (SEQ ID NOs: 1-37) .
  • This invention provides an antisense molecule capable of hybridizing to the isolated DNA molecule.
  • the antisense molecule is DNA.
  • the antisense molecule is RNA.
  • the antisense molecule may be DNA or RNA or variants thereof (i.e. DNA or RNA with a protein backbone) .
  • the present invention extends to the preparation of antisense nucleotides and ribozymes that may be used to interfere with the expression of the receptor recognition proteins at the translation of a specific mRNA, either by masking that MRNA with an antisense nucleic acid or cleaving it with a ribozyme.
  • Antisense nucleic acids are DNA or RNA molecules that are complementary to at least a portion of a specific MRNA molecule. In the cell, they hybridize to that MRNA, forming a double stranded molecule. The cell does not translate an MRNA in this double-stranded form. Therefore, antisense nucleic acids interfere with the expression of MRNA into protein. Oligomers of about fifteen nucleotides and molecules that hybridize to the AUG initiation codon are particularly efficient, since they are easy to synthesize and are likely to pose fewer problems than larger molecules upon introduction to cells.
  • This invention provides a transgenic nonhuman mammal which comprises at least a portion of the isolated DNA molecule introduced into the mammal at an embryonic stage.
  • Methods of producing a transgenic nonhuman mammal are known to those skilled in the art.
  • This invention provides a cell line containing the isolated KS associated herpesvirus of the subject invention.
  • the isolated DNA molecule is artificially introduced into the cell.
  • Cell lines include, but are not limited to: fibrobiasts, such as HFF, NIH/3T3 ; Epithelial cells, such as 5637; lymphocytes, such as FCB; T-cells, such as CCRF-CEM (ATCC CCL 119) ; B-cells, such as BJAB and Raji (ATCC CCL 86) ; and myeloid cells such as K562
  • the isolated KS associated herpesvirus is introduced into a RCC-1 cell line.
  • This invention provides a method of diagnosing Kaposi's sarcoma in a subject which comprises: (a) obtaining a nucleic acid molecule from a tumor lesion of the subject; (b) contacting the nucleic acid molecule with a labelled nucleic acid molecule of at least 15 nucleotides capable of specifically hybridizing with the isolated DNA, under hybridizing conditions; and (c) determining the presence of the nucleic acid molecule hybridized, the presence of which is indicative of Kaposi's sarcoma in the subject, thereby diagnosing Kaposi's sarcoma in the subject.
  • the DNA molecule from the tumor lesion is amplified before step (b) .
  • PCR is employed to amplify the nucleic acid molecule. Methods of amplifying nucleic acid molecules are known to those skilled in the art.
  • DNA sample obtained by the above described method may be cleaved by restriction enzyme.
  • restriction enzymes to cleave DNA and the conditions to perform such cleavage are well-known in the art.
  • a size fractionation may be employed which is effected by a polyacrylamide gel.
  • the size fractionation is effected by an agarose gel.
  • transferring the DNA fragments into a solid matrix may be employed before a hybridization step.
  • solid matrix is nitrocellulose paper.
  • This invention provides a method of diagnosing Kaposi's sarcoma in a subject which comprises: (a) obtaining a nucleic acid molecule from a suitable bodily fluid of the subject; (b) contacting the nucleic acid molecule with a labelled nucleic acid molecules of at least 15 nucleotides capable of specifically hybridizing with the isolated DNA, under hybridizing conditions; and (c) determining the presence of the nucleic acid molecule hybridized, the presence of which is indicative of Kaposi's sarcoma in the subject, thereby diagnosing Kaposi's sarcoma in the subject.
  • This invention provides a method of diagnosing a DNA virus in a subject, which comprises (a) obtaining a suitable bodily fluid sample from the subject, (b) contacting the suitable bodily fluid of the subject to a support having already bound thereto a Kaposi's sarcoma antibody, so as to bind the Kaposi's sarcoma antibody to a specific Kaposi's sarcoma antigen, (c) removing unbound bodily fluid from the support, and
  • This invention provides a method of diagnosing Kaposi's sarcoma in a subject, which comprises (a) obtaining a suitable bodily fluid sample from the subject, (b) contacting the suitable bodily fluid of the subject to a support having already bound thereto a Kaposi's sarcoma antigen, so as to bind Kaposi's sarcoma antigen to a specific Kaposi's sarcoma antibody, (c) removing unbound bodily fluid from the support, and (d) determining the level of the Kaposi's sarcoma antigen bound by the Kaposi's sarcoma antibody, thereby diagnosing Kaposi's sarcoma.
  • This invention provides a method of detecting expression of a DNA virus associated with Kaposi's sarcoma in a cell which comprises obtaining total cDNA obtained from the cell, contacting the cDNA so obtained with a labelled DNA molecule under hybridizing conditions, determining the presence of cDNA hybridized to the molecule, and thereby detecting the expression of the DNA virus.
  • mRNA is obtained from the cell to detect expression of the DNA virus.
  • the suitable bodily fluid sample is any bodily fluid sample which would contain Kaposi's sarcoma antibody, antigen or fragments thereof.
  • a suitable bodily fluid includes, but is not limited to: serum, plasma, cerebrospinal fluid, lymphocytes, urine, transudates, or exudates.
  • the suitable bodily fluid sample is serum or plasma.
  • the bodily fluid sample may be cells from bone marrow, or a supernatant from a cell culture.
  • Methods of obtaining a suitable bodily fluid sample from a subject are known to those skilled in the art.
  • Methods of determining the level of antibody or antigen include, but are not limited to: ELISA, IFA, and Western blotting. Other methods are known to those skilled in the art.
  • a subject infected with a DNA virus associated with Kaposi's sarcoma may be diagnosed with the above described methods.
  • the detection of the human herpesvirus and the detection of virus-associated KS are essentially identical processes.
  • the basic principle is to detect the virus using specific ligands that bind to the virus but not to other proteins or nucleic acids in a normal human cell or its environs.
  • the ligands can either be nucleic acid or antibodies.
  • the ligands can be naturally occurring or genetically or physically modified such as nucleic acids with non-natural or antibody derivatives, i.e., Fab or chimeric antibodies.
  • Serological tests for detection of antibodies to the virus may also be performed by using protein antigens obtained from the human herpesvirus, and described herein. Samples can be taken from patients with KS or from patients at risk for KS, such as AIDS patients.
  • the samples are taken from blood (cells, serum and/or plasma) or from solid tissue samples such as skin lesions.
  • blood cells, serum and/or plasma
  • solid tissue samples such as skin lesions.
  • the most accurate diagnosis for KS will occur if elevated titers of the virus are detected in the blood or in involved lesions .
  • KS may also be indicated if antibodies to the virus are detected and if other diagnostic factors for KS is present.
  • the diagnostic assays of the invention can be nucleic acid assays such as nucleic acid hybridization assays and assays which detect amplification of specific nucleic acid to detect for a nucleic acid sequence of the human herpesvirus described herein.
  • primers are designed to target a specific portion of the nucleic acid of the herpesvirus.
  • the primers set forth in SEQ ID NOs: 38-40 may be used to target detection of regions of the herpesvirus genome encoding ORF 25 homologue - ORF 32 homologue. From the information provided herein, those of skill in the art will be able to select appropriate specific primers.
  • Target specific probes may be used in the nucleic acid hybridization diagnostic assays for KS.
  • the probes are specific for or complementary to the target of interest. For precise allelic differentiations, the probes should be about 14 nucleotides long and preferably about 20-30 nucleotides.
  • nucleic acid probes are about 50 to about 1000 nucleotides, most preferably about 200 to about 400 nucleotides.
  • a sequence is "specific" for a target organism of interest if it includes a nucleic acid sequence which when detected is determinative of the presence of the organism in the presence of a heterogeneous population of proteins and other biologies.
  • a specific nucleic acid probe is targeted to that portion of the sequence which is determinative of the organism and will not hybridize to other sequences especially those of the host where a pathogen is being detected.
  • the specific nucleic acid probe can be RNA or DNA polynucleotide or oligonucleotide, or their analogs.
  • the probes may be single or double stranded nucleotides.
  • the probes of the invention may be synthesized enzymatically, using methods well known in the art (e.g., nick translation, primer extension, reverse transcription, the polymerase chain reaction, and others) or chemically (e.g., by methods such as the phosphoramidite method described by Beaucage and
  • the probe must be of sufficient length to be able to form a stable duplex with its target nucleic acid in the sample, i . e . , at least about 14 nucleotides, and may be longer (e.g., at least about 50 or 100 bases in length) . Often the probe will be more than about 100 bases in length. For example, when probe is prepared by nick-translation of DNA in the presence of labeled nucleotides the average probe length may be about 100- 600 bases.
  • the probe will be capable of specific hybridization to a specific KS-associated herpes virus nucleic acid.
  • specific hybridization occurs when a probe hybridizes to a target nucleic acid, as evidenced by a detectable signal, under conditions in which the probe does not hybridize to other nucleic acids (e . g. , animal cell or other bacterial nucleic acids) present in the sample.
  • nucleic acids e . g. , animal cell or other bacterial nucleic acids
  • a variety of factors including the length and base composition of the probe, the extent of base mismatching between the probe and the target nucleic acid, the presence of salt and organic solvents, probe concentration, and the temperature affect hybridization, and optimal hybridization conditions must often be determined empirically.
  • nucleic acid probe design and annealing conditions see, for example, [81] , supra , Ausubel, F., et al . [8] [hereinafter referred to as Sambrook] , Methods in Enzymology [67] or Hybridization wi th Nucleic Acid Probes [42] all of which are incorporated herein by reference.
  • the probe will have considerable sequence identity with the target nucleic acid. Although the extent of the sequence identity required for specific hybridization will depend on the length of the probe and the hybridization conditions, the probe will usually have at least 70% identity to the target nucleic acid, more usually at least 80% identity, still more usually at least 90% identity and most usually at least 95% or 100% identity.
  • a probe can be identified as capable of hybridizing specifically to its target nucleic acid by hybridizing the probe to a sample treated according the protocol of this invention where the sample contains both target virus and animal cells (e.g., nerve cells) .
  • a probe is specific if the probe's characteristic signal is associated with the herpesvirus DNA in the sample and not generally with the DNA of the host cells and non-biological materials ( e . g. , substrate) in a sample.
  • Southern to include detergents (e . g. , sodium dodecyl sulfate), chelating agents (e . g. , EDTA) or other reagents (e . g. , buffers, Denhardt' s solution, dextran sulfate) in the hybridization or wash solutions.
  • detergents e . g. , sodium dodecyl sulfate
  • chelating agents e . g. , EDTA
  • other reagents e . g. , buffers, Denhardt' s solution, dextran sulfate
  • a convenient method for determining whether a probe is specific for a KS-associated viral nucleic acid utilizes a Southern blot (or Dot blot) using DNA prepared from one or more KS-associated human herpesviruses of the invention. Briefly, to identify a target specific probe DNA is isolated from the virus. Test DNA either viral or cellular is transferred to a solid (e.g., charged nylon) matrix. The probes are labelled following conventional methods. Following denaturation and/or prehybridization steps known in the art, the probe is hybridized to the immobilized DNAs under stringent conditions.
  • Stringent hybridization conditions will depend on the probe used and can be estimated from the calculated T_ (melting temperature) of the hybridized probe (see, e.g., Sambrook for a description of calculation of the T m ) .
  • T_ melting temperature
  • RNA probes an example of stringent hybridization conditions is hybridization in a solution containing denatured probe and 5x SSC at 65°C for 8-24 hours followed by washes in 0. lx SSC, 0.1% SDS (sodium dodecyl sulfate) at 50-65°C.
  • the temperature and salt concentration are chosen so that the post hybridization wash occurs at a temperature that is about 5°C below the T M of the hybrid.
  • the temperature may be selected that is 5°C below the T M or conversely, for a particular temperature, the salt concentration is chosen to provide a T M for the hybrid that is 5°C warmer than the wash temperature.
  • a preferred method for detecting the KS-associated herpesvirus is the use of PCR and/or dot blot hybridization.
  • the presence or absence of an KS agent for detection or prognosis, or risk assessment for KS includes Southern transfers, solution hybridization or non-radioactive detection systems, all of which are well known to those of skill in the art.
  • Hybridization is carried out using probes. Visualization of the hybridized portions allows the qualitative determination of the presence or absence of the causal agent.
  • RNA or reverse transcriptase PCR can be detected by methods described above. This procedure is also well known in the art. See [81] incorporated by reference herein.
  • An alternative means for determining the presence of the human herpesvirus is in si tu hybridization, or more recently, in situ polymerase chain reaction.
  • In situ PCR is described in Neuvo et al . [71] , Intracellular localization of polymerase chain reaction (PCR) -amplified Hepatitis C cDNA; Bagasra et al . [10] , Detection of Human Immunodeficiency virus type 1 provirus in mononuclear cells by in situ polymerase chain reaction; and Heniford et al . [35] , Variation in cellular EGF receptor mRNA expression demonstrated by in situ reverse transcriptase polymerase chain reaction.
  • si tu hybridization assays are well known and are generally described in Methods Enzymol . [67] incorporated by reference herein.
  • a solid support typically a glass slide.
  • the cells are then contacted with a hybridization solution at a moderate temperature to permit annealing of target-specific probes that are labelled.
  • the probes are preferably labelled with radioisotopes or fluorescent reporters.
  • In-situ hybridization is a sensitive localization method which is not dependent on expression of antigens or native vs. denatured conditions.
  • Oligonucleotide (oligo) probes are relatively homogeneous reagents and successful hybridization conditions in tissue sections is readily transferable from one probe to another.
  • Commercially synthesized oligonucleotide probes are prepared against the identified genes. These probes are chosen for length (45-65 mers) , high G-C content (50-70%) and are screened for uniqueness against other viral sequences in GenBank.
  • Oligonucleotides are 3' end-labeled with [ ⁇ - 35 S] dATP to specific activities in the range of 1 x 10 10 dpm/ug using terminal deoxynucleotidyl transferase. Unincorporated labeled nucleotides are removed from the oligo probe by centrifugation through a Sephadex G-25 column or by elution from a Waters Sep Pak C-18 column.
  • KS tissue embedded in OCT compound and snap frozen in freezing isopentane cooled with dry ice is cut at 6 ⁇ m intervals and thawed onto 3-aminopropyltriethoxysilane treated slides and allowed to air dry.
  • the slides are then be fixed in 4% freshly prepared paraformaldehyde, rinsed in water.
  • Formalin-fixed, paraffin embedded KS tissues cut at 6 ⁇ m and baked onto glass slides can also be used.
  • the sections are then deparaffinized in xylenes and rehydrated through graded alcohols .
  • sections are dehydrated through graded alcohols containing 0.3M ammonium acetate and air dried.
  • the slides are dipped in Kodak NTB2 emulsion, exposed for days to weeks, developed, and counterstained with hematoxylin and eoxin.
  • Alternative immunohistochemical protocols may be employed which are known to those skilled in the art.
  • This invention provides a method of treating a subject with Kaposi's sarcoma, comprising administering to the subject an effective amount of the antisense molecule capable of hybridizing to the isolated DNA molecule under conditions such that the antisense molecule selectively enters a tumor cell of the subject, so as to treat the subject.
  • This invention provides a method for treating a subject with Kaposi's sarcoma (KS) comprising administering to the subject having a human herpesvirus-associated KS a pharmaceutically effective amount of an antiviral agent in a pharmaceutically acceptable carrier, wherein the agent is effective to treat the subject with KS-associated human herpes virus.
  • KS Kaposi's sarcoma
  • this invention provides a method of prophylaxis or treatment for Kaposi's sarcoma (KS) by - administering to a patient at risk for KS, an antibody that binds to the human herpesvirus in a pharmaceutically acceptable carrier.
  • the antiviral drug is used to treat a subject with the DNA herpesvirus of the subject invention.
  • KS herpesvirus-induced KS.
  • Snoeck et al . [88] found additive or synergistic effects against CMV when combining antiherpes drugs (e . g. , combinations of zidovudine [3 ' -azido-3 ' - deoxythymidine, AZT] with HPMPC, ganciclovir, foscarnet or acyclovir or of HPMPC with other antivirals) .
  • U.S. Patent Nos. 5,164,395 and 5,021,437 describe the use of a ribonucleotide reductase inhibitor (an acetylpyridine derivative) for treatment of herpes infections, including the use of the acetylpyridine derivative in combination with acyclovir.
  • U.S. Patent No. 5,137,724 (Balzari et al . [11]) describes the use of thymilydate synthase inhibitors (e . g. , 5-fluoro-uracil and 5- fluro-2' -deoxyuridine) in combination with compounds having viral thymidine kinase inhibiting activity.
  • antiviral agents have application here for treatment, such as interferons, nucleoside analogues, ribavirin, amantadine, and pyrophosphate analogues of phosphonoacetic acid
  • Antiviral agents include agents or compositions that directly bind to viral products and interfere with disease progress; and, excludes agents that do not impact directly on viral multiplication or viral titer. Antiviral agents do not include immunoregulatory agents that do not directly affect viral titer or bind to viral products. Antiviral agents are effective if they inactivate the virus, otherwise inhibit its infectivity or multiplication, or alleviate the symptoms of KS.
  • antiherpesvirus agents that will be useful for treating virus-induced KS can be grouped into broad classes based on their presumed modes of action. These classes include agents that act (i) by inhibition of viral DNA polymerase, (ii) by targeting other viral enzymes and proteins, (iii) by miscellaneous or incompletely understood mechanisms, or (iv) by binding a target nucleic acid (i.e., inhibitory nucleic acid therapeutics) . Antiviral agents may also be used in combination (i.e., together or sequentially) to achieve synergistic or additive effects or other benefits.
  • antiviral agents Although it is convenient to group antiviral agents by their supposed mechanism of action, the applicants do not intend to be bound by any particular mechanism of antiviral action. Moreover, it will be understood by those of skill that an agent may act on more than one target in a virus or virus-infected cell or through more than one mechanism.
  • nucleoside analogs believed to act through inhibition of viral DNA replication, especially through inhibition of viral DNA polymerase. These nucleoside analogs act as alternative substrates for the viral DNA polymerase or as competitive inhibitors of DNA polymerase substrates. Usually these agents are preferentially phosphorylated by viral thymidine kinase (TK) , if one is present, and/or have higher affinity for viral DNA polymerase than for the cellular DNA polymerases, resulting in selective antiviral activity. Where a nucleoside analogue is incorporated into the viral DNA, viral activity or reproduction may be affected in a variety of ways.
  • TK thymidine kinase
  • the analogue may act as a chain terminator, cause increased lability (e. g. , susceptibility to breakage) of analogue-containing DNA, and/or impair the ability of the substituted DNA to act as template for transcription or replication (see, e . g. , Balzarini et al . [11]) .
  • acyclovir is triphosphorylated to its active form, with the first phosphorylation being carried out by the herpes virus thymidine kinase, when present.
  • Other examples are the reported conversion of the compound HOE 602 to ganciclovir in a three-step metabolic pathway (Winkler et al . [95]) and the phosphorylation of ganciclovir to its active form by, e . g. , a CMV nucleotide kinase.
  • Anti-herpesvirus medications suitable for treating viral induced KS include, but are not limited to, nucleoside analogs including acyclic nucleoside p h o s p h o n a t e a n a l o g s ( e . g . , phosphonylmethoxyalkylpurines and -pyrimidines) , and cyclic nucleoside analogs.
  • vidarabine (9-/_-D-arabinofuranosyladenine; adenine arabinoside, ara-A, Vira-A, Parke-Davis) ; 1- ⁇ -D- arabinofuranosyluracil (ara- ⁇ ) ; 1- / S-D- arabinofuranosyl-cytosine (ara-C) ; HPMPC [(S)-l-[3- hydroxy-2- (phosphonylmethoxy)propyl] cytosine (e.g., GS 504 Gilead Science)] and its cyclic form (cHPMPC) ;
  • ganciclovir [(9- [1,3- dihydroxy-2 propoxymethyl] -guanine) e.g., Cymevene, Cytovene (Syntex) , DHPG (Stals et al . [89]] ; isopropylether derivatives of ganciclovir (see, e.g., Winkelmann et al .
  • Triciribine and triciribine monophosphate are potent inhibitors against herpes viruses. (Ickes et al . [43] , incorporated by reference herein) , HIV-1 and HIV-2 (Kucera et al . [51], incorporated by reference herein) and are additional nucleoside analogs that may be used to treat KS.
  • An exemplary protocol for these agents is an intravenous injection of about 0.35 mg/meter 2
  • Acyclovir and ganciclovir are of interest because of their accepted use in clinical settings.
  • Acyclovir an acyclic analogue of guanine, is phosphorylated by a herpesvirus thymidine kinase and undergoes further phosphorylation to be incorporated as a chain terminator by the viral DNA polymerase during viral replication.
  • herpesviruses Herpes simplex Types 1 and 2, Varicella- Zoster, Cytomegalovirus, and Epstein-Barr Virus
  • disease such as herpes encephalitis, neonatal herpesvirus infections, chickenpox in immunocompromised hosts, herpes zoster recurrences, CMV retinitis, EBV infections, chronic fatigue syndrome, and hairy leukoplakia in AIDS patients.
  • Exemplary intravenous dosages or oral dosages are 250 mg/kg/m 2 body surface area, every 8 hours for 7 days, or maintenance doses of 200-400 mg IV or orally twice a day to suppress recurrence.
  • Ganciclovir has been shown to be more active than acyclovir against some herpesviruses. See, e . g. , Oren and Soble [73] .
  • Treatment protocols for ganciclovir are 5 mg/kg twice a day IV or 2.5 mg/kg three times a day for 10-14 days. Maintenance doses are 5-6 mg/kg for 5-7 days.
  • HPMPC HPMPC is reported to be more active than either acyclovir or ganciclovir in the chemotherapy and prophylaxis of various HSV-1, HSV-2, TK- HSV, VZV or CMV infections in animal models ( [22] , supra) .
  • Nucleoside analogs such as BVaraU are potent inhibitors of HSV-1, EBV, and VZV that have greater activity than acyclovir in animal models of encephalitis.
  • FIAC fluroidoarbinosyl cytosine
  • FEAU fluroethyl and iodo compounds
  • HPMPA (S) -1- ( [3-hydroxy-2- phosphorylmethoxy] propyl) adenine
  • Cladribine (2- chlorodeoxyadenosine) is another nucleoside analogue known as a highly specific antilymphocyte agent (i.e., a immunosuppressive drug) .
  • 5-thien-2-yl- 2' -deoxyuridine derivatives e . g. , BTDU [5-5(5- bromothien-2-yl) -2' -deoxyuridine] and CTDU [b- (5- chlorothien-2-yl) -2' -deoxyuridine] ; and OXT-A [9- (2- deoxy-2-hydroxymethyl-j ⁇ -D-erythro-oxetanosyl) adenine] and OXT-G [9- (2-deoxy-2-hydroxymethyl-S-D-erythro- oxetanosyl) guanine] .
  • OXT-G is believed to act by inhibiting viral DNA synthesis its mechanism of action has not yet been elucidated.
  • Additional antiviral purine derivatives useful in treating herpesvirus infections are disclosed in US Pat. 5,108,994 (assigned to Beecham Group P.L.C.) .
  • 6- Methoxypurine arabinoside is a potent inhibitor of varicella-zoster virus, and will be useful for treatment of KS.
  • thymidine analogs e.g., idoxuridine (5-ido- 2' -deoxyuridine)
  • triflurothymidine have antiherpes viral activity, but due to their systemic toxicity, are largely used for topical herpesviral infections, including HSV stromal keratitis and uveitis, and are not preferred here unless other options are ruled out.
  • Foscarnet sodium trisodium phosphonoformate, PFA, Foscavir (Astra)
  • PAA phosphonoacetic acid
  • Foscarnet is an inorganic pyrophosphate analogue that acts by competitively blocking the pyrophosphate-binding site of DNA polymerase. These agents which block DNA polymerase directly without processing by viral thymidine kinase. Foscarnet is reported to be less toxic than PAA.
  • the antiherpes-virus agents described above are believed to act through inhibition of viral DNA polymerase.
  • viral replication requires not only the replication of the viral nucleic acid but also the production of viral proteins and other essential components.
  • the present invention contemplates treatment of KS by the inhibition of viral proliferation by targeting viral proteins other than DNA polymerase (e.g., by inhibition of their synthesis or activity, or destruction of viral proteins after their synthesis) .
  • agents that inhibit a viral serine protease e.g., such as one important in development of the viral capsid will be useful in treatment of viral induced KS.
  • viral enzyme targets include: OMP decarboxylase inhibitors (a target of, e . g. , parazofurin) , CTP synthetase inhibitors (targets of, e.g., cyclopentenylcytosine) , IMP dehydrogenase, ribonucleotide reductase (a target of, e.g., carboxyl- containing N-alkyldipeptides as described in U.S. Patent No. 5,110,799 (Tolman et al . , Merck)) , thymidine kinase (a target of, e . g.
  • Kutapressin is a liver derivative available from Schwarz Parma of Milwaukee, Wisconsin in an injectable form of 25 mg/ml.
  • the recommended dosage for herpesviruses is from 200 to 25 mg/ml per day for an average adult of 150 pounds.
  • Poly(I) Poly(C 12 U) an accepted antiviral drug known as Ampligen from HEM Pharmaceuticals of Rockville, MD has been shown to inhibit herpesviruses and is another antiviral agent suitable for treating KS.
  • Intravenous injection is the preferred route of administration. Dosages from about 100 to 600 mg/m 2 are administered two to three times weekly to adults averaging 150 pounds. It is best to administer at least 200 mg/m 2 per week.
  • antiviral agents reported to show activity against herpes viruses e.g., varicella zoster and herpes simplex
  • herpesvirus-induced KS include mappicine ketone (SmithKline Beecham) ; Compounds A, 79296 and A, 73209 (Abbott) for varicella zoster, and Compound 882C87 (Burroughs Wellcome) [see, The Pink Sheet 55(20) May 17, 1993] .
  • Interferon is known inhibit replication of herpes viruses. See [73] , supra . Interferon has known toxicity problems and it is expected that second -
  • herpes virus-induced KS may be treated by administering a herpesvirus reactivating agent to induce reactivation of the latent virus.
  • a herpesvirus reactivating agent to induce reactivation of the latent virus.
  • the reactivation is combined with simultaneous or sequential administration of an anti-herpesvirus agent. Controlled reactivation over a short period of time or reactivation in the presence of an antiviral agent is believed to minimize the adverse effects of certain herpesvirus infections (e.g., as discussed in PCT Application WO 93/04683) .
  • Reactivating agents include agents such as estrogen, phorbol esters, forskolin and /3-adrenergic blocking agents.
  • ganciclovir is an example of a antiviral guanine acyclic nucleotide of the type described in US Patent Nos. 4,355,032 and 4,603,219.
  • Acyclovir is an example of a class of antiviral purine d e r i v a t i v e s , i n c l u d i n g 9 - ( 2 - hydroxyethylmethyl) adenine, of the type described in U.S. Pat. Nos. 4,287,188, 4,294,831 and 4,199,574.
  • Brivudin is an example of an antiviral deoxyuridine derivative of the type described in US Patent No. 4,424,211.
  • Vidarabine is an example of an antiviral purine nucleoside of the type described in British Pat. 1,159,290.
  • Brovavir is an example of an antiviral deoxyuridine derivative of the type described in US Patent Nos. 4,542,210 and 4,386,076.
  • BHCG is an example of an antiviral carbocyclic nucleoside analogue of the type described in US Patent Nos. 5,153,352, 5,034,394 and 5,126,345.
  • HPMPC is an example of an antiviral phosphonyl methoxyalkyl derivative with of the type described in US Patent No. 5,142,051.
  • CDG Carbocyclic 2' -deoxyguanosine
  • CDG Carbocyclic 2' -deoxyguanosine
  • Trifluridine and its corresponding ribonucleoside is described in US Patent No. 3,201,387.
  • thymidine kinase inhibitors useful for treating HSV infections and for inhibiting herpes thymidine kinase.
  • inhibitory nucleic acid therapeutics which can inhibit the activity of herpesviruses in patients with KS.
  • Inhibitory nucleic acids may be single-stranded nucleic acids, which can specifically bind to a complementary nucleic acid sequence. By binding to the appropriate target sequence, an RNA-RNA, a DNA-DNA, or RNA-DNA duplex or triplex is formed. These nucleic acids are often termed “antisense” because they are usually complementary to the sense or coding strand of the gene, although recently approaches for use of "sense” nucleic acids have also been developed.
  • the term “inhibitory nucleic acids” as used herein, refers to both "sense” and “antisense” nucleic acids.
  • the inhibitory nucleic acid By binding to the target nucleic acid, the inhibitory nucleic acid can inhibit the function of the target nucleic acid. This could, for example, be a result of blocking DNA transcription, processing or poly(A) addition to mRNA, DNA replication, translation, or promoting inhibitory mechanisms of the cells, such as promoting RNA degradation. Inhibitory nucleic acid methods therefore encompass a number of different approaches to altering expression of herpesvirus genes. These different types of inhibitory nucleic acid technology are described in Helene, C. and Toulme, J. [34] , which is hereby incorporated by reference and is referred to hereinafter as "Helene and Toulme . "
  • inhibitory nucleic acid therapy approaches can be classified into those that target DNA sequences, those that target RNA sequences (including pre-mRNA and mRNA) , those that target proteins (sense strand approaches) , and those that cause cleavage or chemical modification of the target nucleic acids.
  • Nucleic acids can be designed to bind to the major groove of the duplex DNA to form a triple helical or "triplex" structure.
  • inhibitory nucleic acids are designed to bind to regions of single stranded DNA resulting from the opening of the duplex DNA during replication or transcription. See Helene and Toulme. More commonly, inhibitory nucleic acids are designed to bind to mRNA or mRNA precursors . Inhibitory nucleic acids are used to prevent maturation of pre- mRNA. Inhibitory nucleic acids may be designed to interfere with RNA processing, splicing or translation.
  • the inhibitory nucleic acids can be targeted to mRNA.
  • the inhibitory nucleic acids are designed to specifically block translation of the encoded protein.
  • the inhibitory nucleic acid can be used to selectively suppress certain cellular functions by inhibition of translation of mRNA encoding critical proteins.
  • an inhibitory nucleic acid complementary to regions of c-myc mRNA inhibits c-myc protein expression in a human promyelocytic leukemia cell line, HL60, which overexpresses the c-myc proto- oncogene. See Wickstrom E.L., et al. [93] and Harel-Bellan, A., et al . [31A] .
  • inhibitory nucleic acids targeting mRNA have been shown to work by several different mechanisms to inhibit translation of the encoded protein(s) .
  • the inhibitory nucleic acids introduced into the cell can also encompass the "sense" strand of the gene or mRNA to trap or compete for the enzymes or binding proteins involved in mRNA translation. See Helene and Toulme.
  • inhibitory nucleic acids can be used to induce chemical inactivation or cleavage of the target genes or mRNA. Chemical inactivation can occur by the induction of crosslinks between the inhibitory nucleic acid and the target nucleic acid within the cell. Other chemical modifications of the target nucleic acids induced by appropriately derivatized inhibitory nucleic acids may also be used.
  • Cleavage, and therefore inactivation, of the target nucleic acids may be effected by attaching a substituent to the inhibitory nucleic acid which can be activated to induce cleavage reactions.
  • the substituent can be one that affects either chemical, or enzymatic cleavage.
  • cleavage can be induced by the use of ribozymes or catalytic RNA.
  • the inhibitory nucleic acids would comprise either naturally occurring RNA (ribozymes) or synthetic nucleic acids with catalytic activity.
  • inhibitory nucleic acids to specific cells of the immune system by conjugation with targeting moieties binding receptors on the surface of these cells can be used for all of the above forms of inhibitory nucleic acid therapy.
  • This invention encompasses all of the forms of inhibitory nucleic acid therapy as described above and as described in Helene and Toulme.
  • This invention relates to the targeting of inhibitory nucleic acids to sequences the human herpesvirus of the invention for use in treating KS.
  • An example of an antiherpes virus inhibitory nucleic acid is ISIS 2922 (ISIS Pharmaceuticals) which has activity against CMV [see, Biotechnology News 14(14) p. 5] .
  • a problem associated with inhibitory nucleic acid therapy is the effective delivery of the inhibitory nucleic acid to the target cell in vivo and the subsequent internalization of the inhibitory nucleic acid by that cell. This can be accomplished by linking the inhibitory nucleic acid to a targeting moiety to form a conjugate that binds to a specific receptor on the surface of the target infected cell, and which is internalized after binding.
  • the subjects to be treated or whose tissue may be used herein may be a mammal, or more specifically a human, horse, pig, rabbit, dog, monkey, or rodent. In the preferred embodiment the subject is a human.
  • compositions are administered in a manner compatible with the dosage formulation, and in a therapeutically effective amount.
  • Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner and are peculiar to each subject.
  • Suitable regimes for initial administration and booster shots are also variable, but are typified by an initial administration followed by repeated doses at one or more hour intervals by a subsequent injection or other administration.
  • administration means a method of administering to a subject. Such methods are well known to those skilled in the art and include, but are not limited to, administration topically, parenterally, orally, intravenously, intramuscularly, subcutaneously or by aerosol. Administration of the agent may be effected continuously or intermittently such that the therapeutic agent in the patient is effective to treat a subject with Kaposi's sarcoma or a subject infected with a DNA virus associated with Kaposi's sarcoma.
  • the antiviral compositions for treating herpesvirus- induced KS are preferably administered to human patients via oral, intravenous or parenteral administrations and other systemic forms. Those of skill in the art will understand appropriate administration protocol for the individual compositions to be employed by the physician.
  • compositions of this invention may be in the dosage form of solid, semi-solid, or liquid such as, e.g., suspensions, aerosols or the like.
  • the compositions are administered in unit dosage forms suitable for single administration of precise dosage amounts.
  • the compositions may also include, depending on the formulation desired, pharmaceutically-acceptable, non- toxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration.
  • diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological saline, Ringer's solution, dextrose solution, and Hank's solution.
  • composition or formulation may also include other carriers, adjuvants; or nontoxic, nontherapeutic, nonimmunogenic stabilizers and the like.
  • Effective amounts of such diluent or carrier are those amounts which are effective to obtain a pharmaceutically acceptable formulation in terms of solubility of components, or biological activity, etc.
  • immunosuppressive therapies that can modulate the immunologic dysfunction that arises from the presence of viral infected tissue.
  • agents that block the immunological attack of the viral infected cells will ameliorate the symptoms of KS and/or reduce the disease progress.
  • Such therapies include antibodies that specifically block the targeting of viral infected cells.
  • agents include antibodies which bind to cytokines that upregulate the immune system to target viral infected cells.
  • the antibody may be administered to a patient either singly or in a cocktail containing two or more antibodies, other therapeutic agents, compositions, or the like, including, but not limited to, immuno- • suppressive agents, potentiators and side-effect re ⁇ lieving agents.
  • immuno- suppressive agents useful in suppressing allergic re ⁇ actions of a host.
  • Immunosuppressive agents of inter ⁇ est include prednisone, prednisolone, DECADRON (Merck, Sharp & Dohme, West Point, PA) , cyclophosphamide, cyclosporine, 6-mercaptopurine, methotrexate, azathioprine and i.v. gamma globulin or their combination.
  • Potentiators of interest include monensin, ammonium chloride and chloroquine. All of these agents are administered in generally accepted efficacious dose ranges such as those disclosed in the Physician Desk Reference, 41st Ed. (1987) , Publisher Edward R. Barnhart, New Jersey.
  • Immune globulin from persons previously infected with human herpesviruses or related viruses can be obtained using standard techniques. Appropriate titers of antibodies are known for this therapy and are readily applied to the treatment of KS. Immune globulin can be administered via parenteral injection or by intrathecal shunt. In brief, immune globulin preparations may be obtained from individual donors who are screened for antibodies to the KS-associated human herpesvirus, and plasmas from high-titered donors are pooled. Alternatively, plasmas from donors are pooled and then tested for antibodies to the human herpesvirus of the invention; high-titered pools are then selected for use in KS patients.
  • Antibodies may be formulated into an injectable preparation.
  • Parenteral formulations are known and are suitable for use in the invention, preferably for i.m. or i.v. administration.
  • the formulations containing therapeutically effective amounts of antibodies or immunotoxins are either sterile liquid solutions, liquid suspensions or lyophilized versions and optionally contain stabilizers or excipients.
  • Lyophilized compositions are reconstituted with suitable diluents, e.g., water for injection, saline, 0.3% glycine and the like, at a level of about from .01 mg/kg of host body weight to 10 mg/kg where appropriate.
  • the pharmaceutical compositions containing the antibodies or immunotoxins will be administered in a therapeutically effective dose in a range of from about .01 mg/kg to about 5 mg/kg of the treated mammal.
  • a preferred therapeutically effective dose of the pharmaceutical composition containing antibody or immunotoxin will be in a range of from about 0.01 mg/kg to about 0.5 mg/kg body weight of the treated mammal administered over several days to two weeks by daily intravenous infusion, each given over a one hour period, in a sequential patient dose-escalation regimen.
  • Antibody may be administered systemically by injection i.m., subcutaneously or intraperitoneally or directly into KS lesions.
  • the dose will be dependent upon the properties of the antibody or immunotoxin employed, e.g., its activity and biological half-life, the concentration of antibody in the formulation, the site and rate of dosage, the clinical tolerance of the patient involved, the disease afflicting the patient and the like as is well within the skill of the physician.
  • the antibody of the present invention may be administered in solution.
  • the pH of the solution should be in the range of pH 5 to 9.5, preferably pH 6.5 to 7.5.
  • the antibody or derivatives thereof should be in a solution having a suitable pharmaceutically acceptable buffer such as phosphate, tris (hydroxymethyl) aminomethane-HCl or citrate and the like. Buffer concentrations should be in the range of 1 to 100 mM.
  • the solution of antibody may also contain a salt, such as sodium chloride or potassium chloride in a concentration of 50 to 150 mM.
  • a stabilizing agent such as an albumin, a globulin, a gelatin, a protamine or a salt of protamine may also be included and may be added to a solution containing antibody or immunotoxin or to the composition from which the solution is prepared.
  • Antibody or immunotoxin may also be administered via microspheres, liposomes or other microparticulate delivery systems placed in certain tissues including blood.
  • the dosages of compounds used in accordance with the invention vary depending on the class of compound and the condition being treated.
  • the age, weight, and clinical condition of the recipient patient; and the experience and judgment of the clinician or practitioner administering the therapy are among the factors affecting the selected dosage.
  • the dosage of an immunoglobulin can range from about 0.1 milligram per kilogram of body weight per day to about 10 mg/kg per day for polyclonal antibodies and about 5% to about 20% of that amount for monoclonal antibodies.
  • the immunoglobulin can be administered once daily as an intravenous infusion.
  • the dosage is repeated daily until either a therapeutic result is achieved or until side effects warrant discontinuation of therapy.
  • the dose should be sufficient to treat or ameliorate symptoms or signs of KS without producing unacceptable toxicity to the patient.
  • An effective amount of the compound is that which provides either subjective relief of a symptom(s) or an objectively identifiable improvement as noted by the clinician or other qualified observer.
  • the dosing range varies with the compound used, the route of administration and the potency of the particular compound.
  • This invention provides a method of vaccinating a subject against Kaposi's sarcoma, comprising administering to the subject an effective amount of the peptide or polypeptide encoded by the isolated DNA molecule, and a suitable acceptable carrier, thereby vaccinating the subject.
  • naked DNA is administering to the subject in an effective amount to vaccinate a subject against Kaposi's sarcoma.
  • This invention provides a method of immunizing a subject against a disease caused by the DNA herpesvirus associated with Kaposi's sarcoma which comprises administering to the subject an effective immunizing dose of the isolated herpesvirus vaccine.
  • the invention also provides substances suitable for use as vaccines for the prevention of KS and methods for administering them.
  • the vaccines are directed against the human herpesvirus of the invention, and most preferably comprise antigen obtained from the KS- associated human herpesvirus.
  • Vaccines can be made recombinantly.
  • a vaccine will include from about 1 to about 50 micrograms of antigen or antigenic protein or peptide. More preferably, the amount of protein is from about 15 to about 45 micrograms.
  • the vaccine is formulated so that a dose includes about 0.5 milliliters.
  • the vaccine may be administered by any route known in the art. Preferably, the route is parenteral. More preferably, it is subcutaneous or intramuscular.
  • an antigen can be conjugated to a suitable carrier, usually a protein molecule.
  • a suitable carrier usually a protein molecule.
  • This procedure has several facets. It can allow multiple copies of an antigen, such as a peptide, to be conjugated to a single larger carrier molecule.
  • the carrier may possess properties which facilitate transport, binding, absorption or transfer of the antigen.
  • Suitable carriers are the tetanus toxoid, the diphtheria toxoid, serum albumin and lamprey, or keyhole limpet, hemocyanin because they provide the resultant conjugate with minimum genetic restriction.
  • Conjugates including these universal carriers can function as T cell clone activators in individuals having very different gene sets.
  • the conjugation between a peptide and a carrier can be accomplished using one of the methods known in the art. Specifically, the conjugation can use bifunctional cross-linkers as binding agents as detailed, for example, by Means and Feeney, "A recent review of protein modification techniques, " Bioconjugate Chem. 1:2-12 (1990) .
  • Vaccines against a number of the Herpesviruses have been successfully developed. Vaccines against a number of the Herpesviruses have been successfully developed. Vaccines against
  • Varicella-Zoster Virus using a live attenuated Oka strain is effective in preventing herpes zoster in the elderly, and in preventing chickenpox in both immunocompromised and normal children (Hardy, I., et al . [30] ; Hardy, I. et al . [31] ; Levin, M.J. et al .
  • Herpes simplex Types 1 and 2 are also commercially available with some success in protection against primary disease, but have been less successful in preventing the establishment of latent infection in sensory ganglia (Roizman, B. [78] ; Skinner, G.R. et al . [87]) .
  • Vaccines against the human herpesvirus can be made by isolating extracellular viral particles from infected cell cultures, inactivating the virus with formaldehyde followed by ultracentrifugation to concentrate the viral particles and remove the formaldehyde, and immunizing individuals with 2 or 3 doses containing 1 x 10 9 virus particles (Skinner, G.R. et al . [86]) .
  • envelope glycoproteins can be expressed in E. coli or transfected into stable mammalian cell lines, the proteins can be purified and used for vaccination (Lasky, L.A. [53] ) .
  • MHC - binding peptides from cells infected with the human herpesvirus can be identified for vaccine candidates per the methodology of [61] . supra .
  • the antigen may be combined or mixed with various solutions and other compounds as is known in the art.
  • it may be administered in water, saline or buffered vehicles with or without various adjuvants or immunodiluting agents.
  • adjuvants or agents include aluminum hydroxide, aluminum phosphate, aluminum potassium sulfate (alum) , beryllium sulfate, silica, kaolin, carbon, water-in- oil emulsions, oil-in-water emulsions, muramyl dipeptide, bacterial endotoxin, lipid X, Corynebacteriu parvum (Propionibacterium acnes) , Bordetella pertussis, polyribonucleotides, sodium alginate, lanolin, lysolecithin, vitamin A, saponin, liposomes, levamisole, DEAE-dextran, blocked copolymers or other synthetic adjuvants.
  • Such adjuvants are available commercially from various sources, for example, Merck Adjuvant 65 (Merck and Company, Inc., Rahway, N.J.) or Freund' s Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, Michigan) .
  • Other suitable adjuvants are Amphigen (oil-in-water) , Alhydrogel (aluminum hydroxide) , or a mixture of Amphigen and Alhydrogel. Only aluminum is approved for human use.
  • the proportion of antigen and adjuvant can be varied over a broad range so long as both are present in effective amounts.
  • aluminum hydroxide can be present in an amount of about 0.5% of the vaccine mixture (A1 2 0 3 basis) .
  • the amount of the antigen can range from about 0.1 ⁇ g to about 100 ⁇ g protein per patient.
  • a preferable range is from about 1 ⁇ g to about 50 ⁇ g per dose.
  • a more preferred range is about 15 ⁇ g to about 45 ⁇ g.
  • a suitable dose size is about 0.5 ml.
  • a dose for intramuscular injection for example, would comprise 0.5 ml containing 45 ⁇ g of antigen in admixture with 0.5% aluminum hydroxide.
  • the vaccine may be incorporated into a sterile container which is then sealed and stored at
  • -a low temperature for example 4°C, or it may be freeze-dried. Lyophilization permits long-term storage in a stabilized form.
  • the vaccines may be administered by any conventional method for the administration of vaccines including oral and parenteral (e . g. , subcutaneous or intramuscular) injection. Intramuscular administration is preferred.
  • the treatment may consist of a single dose of vaccine or a plurality of doses over a period of time. It is preferred that the dose be given to a human patient within the first 8 months of life.
  • the antigen of the invention can be combined with appropriate doses of compounds including influenza antigens, such as influenza type A antigens. Also, the antigen could be a component of a recombinant vaccine which could be adaptable for oral administration.
  • Vaccines of the invention may be combined with other vaccines for other diseases to produce multivalent vaccines.
  • a pharmaceutically effective amount of the antigen can be employed with a pharmaceutically acceptable carrier such as a protein or diluent useful for the vaccination of mammals, particularly humans.
  • Other vaccines may be prepared according to methods well-known to those skilled in the art.
  • the epitopes are typically segments of amino acids which are a small portion of the whole protein.
  • Such derivatives may include peptide fragments, amino acid substitutions, amino acid deletions and amino acid additions of the amino acid sequence for the viral proteins from the human herpesvirus.
  • Therapeutic, intravenous, polyclonal or monoclonal antibodies can been used as a mode of passive immunotherapy of herpesviral diseases including perinatal varicella and CMV.
  • Immune globulin from persons previously infected with the human herpesvirus and bearing a suitably high titer of antibodies against the virus can be given in combination with antiviral agents (e.g. ganciclovir) , or in combination with other modes of immunotherapy that are currently 9 -
  • Antibodies to human herpesvirus can be administered to the patient as described herein. Antibodies specific for an epitope expressed on cells infected with the human herpesvirus are preferred and can be obtained as described above.
  • a polypeptide, analog or active fragment can be formulated into the therapeutic composition as neutralized pharmaceutically acceptable salt forms.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide or antibody molecule) and which 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 from 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, 2-ethylamino ethanol, histidine, procaine, and the like.
  • This invention provides a method for monitoring the therapeutic efficacy of treatment for Kaposi's sarcoma, which comprises determining in a first sample from a subject with Kaposi's sarcoma the presence of the isolated DNA molecule, administering to the subject a therapeutic amount of an agent such that the agent is contacted to the cell in a sample, determining after a suitable period of time the amount of the isolated DNA molecule in the second sample from the treated subject, and comparing the amount of isolated DNA molecule determined in the first sample with the amount determined in the second sample, a difference indicating the effectiveness of the agent, thereby monitoring the therapeutic efficacy of treatment for Kaposi's sarcoma.
  • amount is viral load or copy number. Methods of determining viral load or copy number are known to those skilled in the art.
  • KS drug screening assays which determine whether or not a drug has activity against the virus described herein are contemplated in this invention.
  • Such assays comprise incubating a compound to be evaluated for use in KS treatment with cells which express the KS associated human herpesvirus proteins or peptides and determining therefrom the effect of the compound on the activity of such agent.
  • in vitro assays in which the virus is maintained in suitable cell culture are preferred, though in vivo animal models would also be effective.
  • In vitro assays include infecting peripheral blood leukocytes or susceptible T cell lines such as MT-4 with the agent of interest in the presence of varying concentrations of compounds targeted against viral replication, including nucleoside analogs, chain terminators, antisense oligonucleotides and random polypeptides (Asada, H. et al . [1] ; Kikuta et al . [48] both incorporated by reference herein) .
  • Infected cultures and their supernatants can be assayed for the total amount of virus including the presence of the viral genome by quantitative PCR, by dot blot assays, or by using immunologic methods.
  • a culture of susceptible cells could be infected with the human herpesvirus in the presence of various concentrations of drug, fixed on slides after a period of days, and examined for viral antigen by indirect immunofluorescence with monoclonal antibodies to viral peptides ( [48] , supra .
  • chemically adhered MT-4 cell monolayers can be used for an infectious agent assay using indirect immunofluorescent antibody staining to search for focus reduction (Higashi, K. et al . [36] , incorporated by reference herein) .
  • purified enzymes isolated from the human herpesvirus can be used as targets for rational drug design to determine the effect of the potential drug on enzyme activity, such as thymidine phosphotransferase or DNA polymerase.
  • enzyme activity such as thymidine phosphotransferase or DNA polymerase.
  • the genes for these two enzymes are provided herein.
  • a measure of enzyme activity indicates effect on the agent itself.
  • This invention provides an assay for screening anti-KS chemotherapeutics.
  • Infected cells can be incubated in the presence of a chemical agent that is a potential chemotherapeutic against KS (e.g. acyclo-guanosine) .
  • a chemical agent that is a potential chemotherapeutic against KS (e.g. acyclo-guanosine) .
  • the level of virus in the cells is then determined after several days by IFA for antigens or Southern blotting for viral genome or Northern blotting for MRNA and compared to control cells .
  • This assay can quickly screen large numbers of chemical compounds that may be useful against KS.
  • this invention provides an assay system that is employed to identify drugs or other molecules capable of binding to the DNA molecule or proteins, either in the cytoplasm or in the nucleus, thereby inhibiting or potentiating transcriptional activity.
  • an assay system that is employed to identify drugs or other molecules capable of binding to the DNA molecule or proteins, either in the cytoplasm or in the nucleus, thereby inhibiting or potentiating transcriptional activity.
  • Such assay would be useful in the development of drugs that would be specific against particular cellular activity, or that would potentiate such activity, in time or in level of activity.
  • representational difference analysis To search for foreign DNA sequences belonging to an infectious agent in AIDS-KS, representational difference analysis (RDA) was employed to identify and characterize unique DNA sequences in KS tissue that are either absent or present in low copy number in non-diseased tissue obtained from the same patient [58] .
  • This method can detect adenovirus genome added in single copy to human DNA but has not been used to identify previously uncultured infectious agents.
  • RDA is performed by making simplified "representations" of genomes from diseased and normal tissues from the same individual through PCR amplification of short restriction fragments. The DNA representation from the diseased tissue is then ligated to a priming sequence and hybridized to an excess of unligated, normal tissue DNA representation.
  • DNA (10 ⁇ g) extracted from both the KS lesion and unaffected tissue were separately digested to completion with Bam HI (20 units/ ⁇ g) at 37° C for 2 hours and 2 ⁇ g of digestion fragments were ligated to
  • NBaml2 and NBam24 priming sequences [primer sequences described in 58] . Thirty cycles of PCR amplification were performed to amplify "representations" of both genomes. After construction of the genomic representations, KS tester amplicons between 150 and 1500 bp were isolated from an agarose gel and NBam priming sequences were removed by digestion with Bam HI . To search for unique DNA sequences not found in non-KS driver DNA, a second set of priming sequences (JBaml2 and JBam24) was ligated onto only the KS tester DNA amplicons ( Figure 1, lane 1) .
  • ligated KS lesion amplicons were hybridized to 20 ⁇ g of unligated, normal tissue representational amplicons. An aliquot of the hybridization product was then subjected to 10 cycles of PCR amplification using JBam24 , followed by mung bean nuclease digestion. An aliquot of the mung bean-treated difference product was then subjected to 15 more cycles of PCR with the JBam24 primer ( Figure 1, lane
  • KS- associated bands (designated KS330Bam, KS390Bam, KS480Bam, KS627Bam after digestion of the two flanking 28 bp ligated priming sequences with Bam HI) were gel purified and cloned by insertion into the pCRII vector. PCR products were cloned in the pCRII vector using the TA cloning system (Invitrogen Corporation, San Diego, CA) . Experiment 2: Determination of the specificity of AIDS-KS unique sequences.
  • Control tissues used for comparison to the KS lesions included 56 lymphomas from patients with and without AIDS, 19 hyperplastic lymph nodes from patients with and without AIDS, 5 vascular tumors from nonAIDS patients and 13 tissues infected with opportunistic infections that commonly occur in AIDS patients. Control DNA was also extracted from a consecutive series of 49 surgical biopsy specimens from patients without AIDS. Additional clinical and demographic information on the specimens was not collected to preserve patient confidentiality.
  • the tissues listed in Table 1, were collected from diagnostic biopsies and autopsies between 1983 and 1993 and stored at -70°C. Each tissue sample was from a different patient, except as noted in Table 1. Most of the 27 KS specimens were from lymph nodes dissected under surgical conditions which diminishes possible contamination with normal skin flora. All specimens were digested with Bam HI prior to hybridization. KS390Bam and KS480Bam hybridized nonspecifically to both KS and non-KS tissues and were not further characterized.
  • Tissues included skin, appendix, kidney, prostate, hernia sac, lung, fibrous tissue, gallbladder, colon, foreskin, thyroid, small bowel, adenoid, vein, axillary tissue, lipo a, heart, mouth, hemorrhoid, pseudoaneurysm and fistula track. Tissues were collected from a consecutive series of biopsies on patients without AIDS but with unknown HIV serostatus.
  • sequence copy number in the AIDS-KS tissues was estimated by simultaneous hybridization with KS330Bam and a 440 bp probe for the constant region of the T cell receptor ⁇ gene [76] .
  • Samples in lanes 5 and 6 of Figures 2A-2B showed similar intensities for the two probes indicating an average copy number of approximately two KS330Bam sequences per cell, while remaining tissues had weaker hybridization signals for the KS330Bam probe.
  • KS330Bam and KS627Bam six clones for each insert were sequenced.
  • the Sequenase version 2.0 (United States Biochemical, Cleveland, OH) system was used and sequencing was performed according to manufacturer's instructions. Nucleotides sequences were confirmed with an Applied Biosystems 373A Sequencer in the DNA Sequencing Facilities at Columbia University.
  • KS330Bam is a 330 bp sequence with 51% G:C content ( Figure 3B) and KS627Bam is a 627 bp sequence with a
  • KS330Bam has 54% nucleotide identity to the BDLFl open reading frame
  • KS330Bam is 51% identical by amino acid homology to a portion of the ORF26 open reading frame encoding the capsid protein VP23 (NCBI g.i. 60348, bp 46024 - 46935) of herpesvirus saimiri [2] , a gammaherpesvirus which causes fulminant lymphoma in New world monkeys.
  • This fragment also has a 39% identical amino acid sequence to the theoretical protein encoded by the homologous open reading frame BDLFl in EBV (NCBI g.i. 59140, bp 132403 -133307) [9] .
  • amino acid sequence encoded by KS627Bam is homologous with weaker identity (31%) to the tegument protein, gpl40 (ORF 29, NCBI g.i. 60396, bpl08782-112681) of herpesvirus saimiri.
  • Sequence data from KS330Bam was used to construct PCR primers to amplify a 234bp fragment designated KS330 234
  • Each PCR reaction used 0.1 ⁇ g of genomic DNA, 50 pmoles of each primer, 1 unit of Taq polymerase, 100 ⁇ M of each deoxynucleotide triphosphate, 50 mM KC1, lOmM Tris-HCl
  • KS330 234 was found in all 25 amplifiable tissues with microscopically detectable AIDS-KS, but rarely occurred in non-KS tissues, including tissues from AIDS patients.
  • KS330Bam and KS627Bam are portions of a larger genome and to determine the proximity of the two sequences to each other.
  • samples of KS DNA were digested with Pvu II restriction enzymes.
  • Digested genomic DNA from three AIDS-KS samples were hybridized to KS330Bam and KS627Bam by Southern blotting ( Figure 5) .
  • These sequences hybridized to various sized fragments of the digested KS DNA indicating that both sequences are fragments of larger genomes.
  • Differences in the KS330Bam hybridization pattern to Pvu II digests of the three AIDS-KS specimens indicate that polymorphisms may occur in the larger genome.
  • Individual fragments from the digests failed to simultaneously hybridize with both KS330Bam and KS627Bam, demonstrating that these two Bam HI restriction fragments are not adjacent to one another.
  • KS330Bam and KS627Bam are heritable polymorphic DNA markers for KS, these sequences should be uniformly detected at non-KS tissue sites in patients with AIDS- KS.
  • KS330Bam and KS627Bam are sequences specific for an exogenous infectious agent, it is likely that some tissues are uninfected and lack detectable KS330Bam and KS627Bam sequences.
  • DNA extracted from multiple uninvolved tissues from three patients with AIDS-KS were hybridized to 32 P-labelled KS330Bam and KS627Bam probes as well as analyzed by PCR using the KS330 234 primers (Table 2) .
  • KS lesions from patients A, B and C, and uninvolved skin and muscle from patient A were positive for KS330Bam and KS627Bam, but muscle and brain tissue from patient B and muscle, brain, colon, heart and hilar lymph node tissues from patient C were negative for these sequences.
  • Uninvolved stomach lining adjacent to the KS lesion in patient C was positive by PCR, but negative by Southern blotting which suggests the presence of the sequences in this tissue at levels below the detection threshold for Southern blotting.
  • Table 2 Differential detection of KS330Bam, KS627Bam and KS330 234 sequences in KS-involved and non-involved tissues from three patients with AIDS-KS.
  • KS330Bam and KS627Bam are genomic fragments of a novel infectious agent associated with AIDS-KS.
  • a genomic library from a KS lesion was made and a phage clone with a 20 kb insert containing the KS330Bam sequence was identified.
  • the 20 kb clone digested with PvuII (which cuts in the middle of the KS330Bam sequence) produced 1.1 kb and 3 kb fragments that hybridized to KS330Bam.
  • the 1.1 kb subcloned insert and -900 bp from the 3 kb subcloned insert resulting in 9404 bp of contiguous sequence was entirely sequenced. This sequence contains partial and complete open reading frames homologous to regions in gamma herpesviruses.
  • the KS330Bam sequence is an internal portion of an 918 bp ORF with 55-56% nucleotide identity to the ORF26 and BDLFl genes of HSVSA and EBV respectively.
  • the EBV and HSVSA translated amino acid sequences for these ORFs demonstrate extensive homology with the amino acid sequence encoded by the KS-associated 918 bp ORF ( Figure 6) .
  • the VP23 protein is a late structural protein involved in capsid construction.
  • Reverse transcriptase (RT) -PCR of mRNA from a KS lesion is positive for transcribed KS330Bam mRNA and that indicates that this ORF is transcribed in KS lesions.
  • fragments obtained from Pvu II digest of the 21 Kb phage insert described above fragments obtained from a BamHI/NotI digest were also subcloned into pBluescript (Stratagene, La Jolla, CA) . The termini of these subcloned fragments were sequenced and were also found to be homologous to nucleic acid sequence EBV and HSVSA genes. These homologs have been used to develop a preliminary map of subcloned fragments ( Figure 9) . Thus, sequencing has revealed that the KS agent maintains co-linear homology to gamma herpesviruses over the length of the 21 Kb phage insert .
  • Regions flanking KS330Bam were sequenced and characterized by directional walking. This was performed by the following strategy: 1) KS genomic libraries were made and screened using the KS330Bam fragment as a hybridization probe, 2) DNA inserts from phage clones positive for the KS330Bam probe were isolated and digested with suitable restriction enzyme (s) , 3) the digested fragments were subcloned into pBluescript (Stratagene, La Jolla, CA) , and 4) the subclones were sequenced. Using this strategy, the major capsid protein (MCP) ORF homolog was the first important gene locus identified. Using sequenced unique 3' and 5' end-fragments from positive phage clones as probes, and following the strategy above a KS genomic library are screened by standard methods for additional contiguous sequences.
  • MCP major capsid protein
  • restriction fragments are subcloned into phagemid pBluescript KS+, pBluescript KS-, pBS+, or pBS- (Stratagene) or into plasmid pUC18 or pUC19.
  • Recombinant DNA was purified through CsCl density gradients or by anion-exchange chromatography (Qiagen) .
  • Nucleotide sequenced by standard screening methods of cloned fragments of KSHV were done by direct sequencing of double- stranded DNA using oligonucleotide primers synthesized commercially to "walk” along the fragments by the dideoxy-nucleotide chain termination method. Junctions between clones are confirmed by sequencing overlapping clones.
  • Targeted homologous genes in regions flanking KS330Bam include, but are not limited to: 11-10 homolog, thymidine kinase (TK) , g85, g35, gH, capsid proteins and MCP.
  • TK is an early protein of the herpesviruses functionally linked to DNA replication and a target enzyme for anti-herpesviral nucleosides.
  • TK is encoded by the EBV BXLF1 ORF located -9700 bp rightward of BDLFl and by the HSVSA ORF 21 -9200 bp rightward of the ORF 26. A subcloned fragment of KS5 was identified with strong homology to the EBV and HSVSA TK open reading frames.
  • g85 is a late glycoprotein involved in membrane fusion homologous to gH in HSV1.
  • this protein is encoded by BLXF2 ORF located -7600 bp rightward of BDLFl, and in HSVSA it is encoded by ORF 22 located -7100 bp rightward of ORF26.
  • g35 is a late EBV glycoprotein found in virion and plasma membrane. It is encoded by BDLF3 ORF which is 1300 bp leftward of BDLFl in EBV. There is no BDLF3 homolog in HSVSA. A subcloned fragment has already been identified with strong homology to the EBV gp35 open reading frame.
  • MCP Major capsid protein
  • Antibodies are generated against the MCP during natural infection with most herpesviruses .
  • the terminal 1026 bp of this major capsid gene homolog in KSHV have been sequenced.
  • Targeted homologous genes/loci in regions flanking KS627Bam include, but are not limited to: terminal reiterated repeats, LMPI, EBERs and Ori P.
  • Terminal reiterated sequences are present in all herpesviruses. In EBV, tandomly reiterated 0.5 Kb long terminal repeats flank the ends of the linear genome and become joined in the circular form. The terminal repeat region is immediately adjacent to BNRF1 in EBV and ORF 75 in HSVSA. Since the number of terminal repeats varies between viral strains, identification of terminal repeat regions may allow typing and clonality studies of KSHV in KS legions. Sequencing through the terminal repeat region may determine whether this virus is integrated into human genome in KS.
  • LMPI is an latent protein important in the transforming effects of EBV in Burkitt's lymphoma. This gene is encoded by the EBV BNRF1 ORF located -2000 bp rightward of tegument protein ORF BNRF1 in the circularized genome. There is no LMPI homolog in HSVSA.
  • EBERs are the most abundant RNA in latently EBV infected cells and Ori-P is the origin of replication for latent EBV genome. This region is located between -4000-9000 bp leftward of the BNRF1 ORF in EBV; there are no corresponding regions in HSVSA.
  • the data indicates that the KS agent is a new human herpesvirus related to gamma herpesviruses EBV and
  • HSVSA HSVSA.
  • the results are not due to contamination or to incidental co-infection with a known herpesvirus since the sequences are distinct from all sequenced herpesviral genomes (including EBV, CMV, HHV6 and HSVSA) and are associated specifically with KS in three separate comparative studies. Furthermore, PCR testing of KS DNA with primers specific for EBV-1 and EBV-2 failed to demonstrate these viral genomes in these tissues.
  • KSHV is homologous to EBV regions, the sequence does not match any other known sequence and thus provides evidence for a new viral genome, related to but distinct from known members of the herpesvirus family.
  • Virus-containing cells are coated to a microscope slide.
  • the slides are treated with organic fixatives, dried and then incubated with patient sera. Antibodies in the sera bind to the cells, and then excess nonspecific antibodies are washed off.
  • An antihuman immunoglobulin linked to a fluorochrome, such as fluorescein, is then incubated with the slides, and then excess fluorescent immunoglobulin is washed off.
  • the slides are then examined under a microscope and if the cells fluoresce, then this indicates that the sera contains antibodies directed against the antigens present in the cells, such as the virus.
  • BCBL-1 which is a naturally transformed EBV infected (nonproducing) B cell line, using 4 KS patient sera and 4 control sera (from AIDS patients without KS) . Initially, both sets of sera showed similar levels of antibody binding.
  • sera at 1:25 dilution were pre- adsorbed using 3xl0 6 1% paraformaldehyde-fixed Raji cells per ml of sera.
  • BCBL1 cells were fixed with ethanol/acetone, incubated with dilutions of patient sera, washed and incubated with fluorescein-conjugated goat anti-human IgG. Indirect immunofluorescent staining was determined.
  • Table 3 shows that unabsorbed case and control sera have similar end-point dilution indirect immunofluorescence assay (IFA) titers against the BCBL1 cell line. After Raji adsorption, case sera have four-fold higher IFA titers against BCBL1 cells than control sera. Results indicated that pre- adsorption against paraformaldehyde-fixed Raji cells reduces fluorescent antibody binding in control sera but do not eliminate antibody binding to KS case sera. These results indicate that subjects with KS have specific antibodies directed against the KS agent that can be detected in serological assays such as IFA, Western blot and Enzyme immunoassays (Table 3) .
  • IFA end-point dilution indirect immunofluorescence assay
  • Control autopsy-confirmed female, AIDS patient, no KS
  • Virus-containing cells or purified virus (or a portion of the virus, such as a fusion protein) is electrophoresed on a polyacrylamide gel to separate the protein antigens by molecular weight.
  • the proteins are blotted onto a nitrocellulose or nylon membrane, then the membrane is incubated in patient sera.
  • Antibodies directed against specific antigens are developed by incubating with a anti-human immunoglobulin attached to a reporter enzyme, such as a peroxidase. After developing the membrane, each antigen reacting against antibodies in patient sera shows up as a band on the membrane at the corresponding molecular weight region.
  • Enzyme immunoassay (“EIA or ELISA")
  • Virus-containing cells or purified virus (or a portion of the virus, such as a fusion protein) is coated to the bottom of a 96-well plate by various means (generally incubating in alkaline carbonate buffer) .
  • the plates are washed, then the wells are incubated with patient sera. Antibodies in the sera directed against specific antigens stick on the plate.
  • the wells are washed again to remove nonspecific antibody, then they are incubated with a antihuman immunoglobulin attached to a reporter enzyme, such as a peroxidase.
  • the plate is washed again to remove nonspecific antibody and then developed.
  • Wells containing antigen that is specifically recognized by antibodies in the patients sera change color and can be detected by an ELISA plate reader (a spectrophotomer) .
  • BCBL 1 from lymphomatous tissues belonging to a rare infiltrating, anaplastic body cavity lymphoma occurring in AIDS patients has been placed in continuous cell culture and shown to be continuously infected with the KS agent. This cell line is also naturally infected with Epstein-Barr Virus (EBV) .
  • EBV Epstein-Barr Virus
  • the BCBL cell line was used as an antigen substrate to detect specific KS antibodies in persons infected with the putative virus by Western-blotting.
  • Three lymphoid B cell lines were used as controls. These included the EBV genome positive cell line P3H3, the EBV genome defective cell line Raji and the EBV genome negative cell line Bjab.
  • the proteins in the supernatant was then fractionated by sodium, dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) under reducing conditions with a separation gel of 15% and a stacking gel of 5% (3) .
  • Prestained protein standards were included: myosin, 200 kDa; /3-galactosidase, 118 kDA; BSA, 78 kDa; ovalbumin, 47.1 kDa; carbonic anhydrase, 31.4 kDa; soybean trypsin inhibitor, 25.5 kDa, lysozyme, 18.8 kDa and aprotinin, 8.3 kDa (Bio-Rad) .
  • the membranes were subsequently incubated with human sera at dilution 1:200 in 1% skim milk overnight at room temperature, washed 3 times with a solution containing TBS, 0.2% Triton X-100 and 0.05% skim milk and then 2 times with TBS. The membranes were then incubated for 2 h at room temperature with alkaline phosphatase conjugated goat anti-mouse IgG + IgM + IgA (Sigma) diluted at 1:5000 in 1% skim milk. After repeating the washing, the membranes were stained with nitroblue tetranolium chloride and 5-bromo-4-chloro-3- indolylphosphate p-toluidine salt (Gibco BRL) .
  • Two bands of approximately 226 kDa and 234 kDa were identified to be specifically present on the Wester- blot of BCBL cell lysate in 5 sera from AIDS gay man patients infected with KS. These 2 bands were absent from the lysates of P3H3, Raji and Bjab cell lysates. 5 sera from AIDS gay man patients without KS and 2 sera from AIDS woman patients without KS as well as 1 sera from nasopharyncel carcinoma patient were not able to detect these 2 bands in BCBL 1, P3H3 , Raji and Bjab cell lysates. In a blinded experiment, using the 226 kDa and 234 kDa markers, 15 out of 16 sera from KS patients were correctly identified. In total, the 226 kDa and 234 kDa markers were detected in 20 out of 21 sera from KS patients.
  • the antigen is enriched in the nuclei fraction of BCBL1.
  • Enriched antigen with low background can be obtained by preparing nucleic from BCBC as the starting antigen preparation using standard, widely available protocols. For example, 500-750ml of BCBL at 5X10 5 cells/ml can be pelleted at low speed. The pellet is placed in 10 mM NaCl, 10 mM Tris pH 7.8, 1.5 mM MgCl 2 (equi volume) + 1.0% NP-40 on ice for 20 min to lyse cells. The lysate is then spun at 1500 rpm for 10 min. to pellet nucleic. The pellet is used as the starting fraction for the antigen preparation for the Western blot. This will reduce cross- reactive cytoplasmic antigens.
  • BCBLl cells were co-cultivated with Raji cell lines separated by a 0.45 ⁇ tissue filter insert. Approximately, 1-2 x 10 6 BCBLl and 2x1(5 Raji cells were co-cultivated for 2-20 days in supplemented RPMI alone, in 10 ⁇ g/ml 5' -bromodeoxyuridine (BUdR) and 0.6 ⁇ g/ml 5' -flourodeoxyuridine or 20 ng/ml 12-0- tetradecanoylphorbol-13-acetate (TPA) . After 2,8,12 or 20 days co-cultivation, Raji cells were removed, washed and placed in supplemented RPMI 1640 media.
  • BdR 5' -bromodeoxyuridine
  • TPA 12-0- tetradecanoylphorbol-13-acetate
  • RCC1 Raji culture co-cultivated with BCBLl in 20 ng/ml TPA for 2 days survived and has been kept in continuous suspension culture for >10 weeks.
  • This cell line designated RCC1 (Raji Co-Culture, No. 1) remains PCR positive for the KS330 234 sequence after multiple passages.
  • This cell line is identical to its parental Raji cell line by flow cytometry using EMA, Bl, B4 and BerH2 lymphocyte-flow cytometry (approximately 2%) .
  • RCC1 periodically undergo rapid cytolysis suggestive of lytic reproduction of the agent.
  • RCC1 is a Raji cell line newly infected with KSHV.
  • Crude virus preparations are made from either the supernatant or low speed pelleted cell fraction of BCBLl cultures. Approximately 650ml or more of log phase cells should be used (>5X10 6 cells/ml) .
  • the cell free supernatant is spun at 10,000 rpm in a GSA rotor for 10 min to remove debris.
  • PEG-8000 is added to 7%, dissolved and placed on ice for >2.5 hours.
  • the PEG- supernatant is then spun at 10,000 xg for 30 min.
  • supernatant is poured off and the pellet is dried and scraped together from the centrifuge bottles.
  • the pellet is then resuspended in a small volume (1-2 ml) of virus buffer (VB, 0.1 M NaCl, 0.01 M Tris, pH 7.5) . This procedure will precipitate both naked genome and whole virion.
  • the virion are then isolated by centrifugation at 25,000 rpm in a 10-50% sucrose gradient made with VB.
  • One ml fractions of the gradient are then obtained by standard techniques
  • each fraction is then tested by dot blotting using specific hybridizing primer sequences to determine the gradient fraction containing the purified virus (preparation of the fraction maybe needed in order to detect the presence of the virus, such as standard DNA extraction) .
  • the pellet of cells is washed and pelleted in PBS, then Iysed using hypotonic shock and/or repeated cycles of freezing and thawing in a small volume ( ⁇ 3 ml) .
  • Nuclei and other cytoplasmic debris are removed by centrifugation at 10,000g for 10 min, filtration through a 0.45 m filter and then repeat centrifugation at 10,000g for 10 min.
  • This crude preparation contains viral genome and soluble cell components.
  • the genome preparation can then be gently chloroform- phenol extracted to remove associated proteins or can be placed in neutral DNA buffer (1 M NaCl, 50 mM Tris, 10 mM EDTA, pH 7.2-7.6) with 2% sodium dodecylsulfate (SDS) and 1% sarcosyl .
  • the genome is then banded by centrifugation through 10-30% sucrose gradient in neutral DNA buffer containing 0.15% sarcosyl at 20,000 rpm in a SW 27.1 rotor for 12 hours (for 40,000 rpm for 2-3 hours in an SW41 rotor) . The band is detected as described above.
  • the supernatant is loaded on a 10-30% sucrose gradient in 1.0 M NaCl, ImM EDTA, 50mM Tris-HCl, pH 7.5.
  • the gradients are centrifuged at 20,000 rpm on a SW 27.1 rotor for 12 hours.
  • 0.5 ml aliquots of the gradient have been fractionated (fractions 1-62) with the 30% gradient fraction being at fraction No. 1 and the 10% gradient fraction being at fraction No. 62.
  • Each fraction has been dot hybridized to a nitrocellulose membrane and then a 32 P-labeled KSHV DNA fragment, KS631Bam has been hybridized to the membrane using standard techniques.
  • Figure 11 shows that the major solubilized fraction of the KSHV genome bands (i.e. is isolated) in fractions 42 through 48 of the gradient with a high concentration of the genome being present in fraction 44.
  • a second band of solubilized KSHV DNA occurs in fractions 26 through 32.
  • DNA is extracted using standard techniques from the RCC-1 or RCC-1 2F5 cell line [27, 49, 66] .
  • the DNA is tested for the presence of the KSHV by Southern blotting and PCR using the specific probes as described hereinafter.
  • Fresh lymphoma tissue containing viable infected cells is simultaneously filtered to form a single cell suspension by standard techniques [49, 66] .
  • the cells are separated by standard Ficoll-Plaque centrifugation and lymphocyte layer is removed.
  • the lymphocytes are then placed at >lxl0 6 cells/ml into standard lymphocyte tissue culture medium, such as RMP 1640 supplemented with 10% fetal calf serum.
  • Immortalized lymphocytes containing the KSHV virus are indefinitely grown in the culture media while nonimmortilized cells die during course of prolonged cultivation.
  • the virus may be propagated in a new cell line by removing media supernatant containing the virus from a continuously infected cell line at a concentration of >lxl0 6 cells/ml.
  • the media is centrifuged at 2000xg for 10 minutes and filtered through a 0.45 ⁇ filter to remove cells.
  • the media is applied in a 1:1 volume with cells growing at >lxl0 6 cells/ml for 48 hours.
  • the cells are washed and pelleted and placed in fresh culture medium, and tested after 14 days of growth.
  • the herpesvirus may be isolated from the cell DNA in the following manner.
  • An infected cell line which can be Iysed using standard methods such as hyposmotic shocking and Dounce homogenization, is first pelleted at 2000xg for 10 minutes, the supernatant is removed and centrifuged again at 10,000xg for 15 minutes to remove nuclei and organelles. The supernatant is filtered through a 0.45 ⁇ filter and centrifuged again at 100,000xg for 1 hour to pellet the virus. The virus can then be washed and centrifuged again at 100,000xg for 1 hour.
  • KS5 lambda phage
  • BamHI and Not I Boehringer-Mannheim, Indianapolis IN
  • five fragments were gel isolated and subcloned into Bluescript II KS (Stratagene, La Jolla CA) .
  • the entire sequence was determined by bidirectional sequencing at a seven fold average redundancy by primer walking and nested deletions .
  • DNA sequence data were compiled and aligned using ALIGN (IBI-Kodak, Rochester NY) and analyzed using the Wisconsin Sequence Analysis Package Version 8-UNIX
  • Protein site motifs were identified using Motif (Genetics Computer Group, Madison WI) .
  • Herpesvirus Gene Sequence Comparisons Complete genomic sequences of three gammaherpesviruses were available: Epstein-Barr virus (EBV) , a herpesvirus of humans [4] ; herpesvirus saimiri (HVS) , a herpesvirus of the New World monkey Saimiri sciureus [1] ; and equine herpesvirus 2 (EHV2 [49] ) . Additional thymidine kinase gene sequences were obtained for alcelaphine herpesvirus 1 (AHV1 [22] ) and bovine herpesvirus 4 (BHV4 [31] ) .
  • EBV Epstein-Barr virus
  • HVS herpesvirus saimiri
  • EHV2 equine herpesvirus 2
  • Additional thymidine kinase gene sequences were obtained for alcelaphine herpesvirus 1 (AHV1 [22] ) and bovine herpesvirus 4
  • positions in alignments that contained inserted gaps in one or more sequences were removed before use for tree construction.
  • Phylogenetic inference programs were from the Phylip set, version 3.5c [14] and from the GCG set [16] .
  • Trees were built with the maximum parsimony (MP) , neighbor joining (NJ) methods.
  • MP parsimony
  • NJ neighbor joining
  • For the NJ method which utilizes estimates of pairwise distances between sequences, distances were estimated as mean numbers of substitution events per site with Protdist using the PAM 250 substitution probability matrix of Schwartz & Dayhoff [46] .
  • Bootstrap analysis [15] was carried out for MP and NJ trees, with 100 sub-replicates of each alignment, and consensus trees obtained with the program Consense.
  • Protml was used to infer trees by the maximum likelihood (ML) method.
  • Protml was obtained form J. Adachi, Department of Statistical Science, The graduate University for Advanced Study, Tokyo 106, Japan. Because of computational constraints, Protml was used only with the 4-species CS1 alignment.
  • TPA 12-0-tetradecanoylphorbol-13- acetate
  • PBS phosphate-buffered saline
  • KS631Bam, EBV terminal repeat and beta-actin sequences were random-primer labeled with 32 P [13] .
  • T cell line Molt-3 (a gift from Dr. Jodi Black, Centers for Disease Control and Prevention)
  • Raji cells (American Type Culture
  • RCC-1 cells were cultured in RPMI 1640 with 10% FCS.
  • Owl monkey kidney cells (American Type Culture Collection, Rockville MD) were cultured in MEM with 10% FCS and 1% nonessential amino acids (Gibco-BRL, Gaithersburg MD) .
  • 2xl0 6 Raji cells were cultivated with 1.4xl0 6 BCBL-1 cells in the presence of 20 ng/ml TPA for 2 days in chambers separated by Falcon 0.45 ⁇ g filter tissue culture inserts to prevent contamination of Raji with BCBL-1.
  • RCC-1 were obtained by dilution in 96 well plates to obtain RCC-1.
  • FCBL Fetal cord blood lymphocytes
  • Adherent recipient cells were washed with sterile Hank's Buffered Salt Solution (HBSS, Gibco-BRL, Gaithersburg MD) and overlaid with 5 ml of BCBL-1 media supernatant. After incubation with BCBL-1 media supernatant, cells were washed three times with sterile HBSS, and suspended in fresh media. Cells were subsequently rewashed three times every other day for six days and grown for at least two weeks prior to DNA extraction and testing. PCR to detect KSHV infection was performed using nested and unnested primers from ORF 26 and ORF 25 as previously described [10, 35] .
  • HBSS Hank's Buffered Salt Solution
  • AIDS-KS sera were obtained from ongoing cohort studies (provided by Drs. Scott Holmberg, Thomas Spira and Harold Jaffe, Centers for Disease Control, and Prevention, and Isaac Weisfuse, New York City Department of Health) . Sera from AIDS-KS patients were drawn between 1 and 31 months after initial KS diagnosis, sera from intravenous drug user and homosexual/bisexual controls were drawn after non-KS AIDS diagnosis, and sera from HIV-infected hemophiliac controls were drawn at various times after HIV infection.
  • Immunofluorescence assays were performed using an equal volume mixture of goat anti-human IgG-FITC conjugate (Molecular Probes, Eugene OR) and goat anti-human IgM-FITC conjugate (Sigma Chemical Co., St. Louis MO) diluted 1:100 and serial dilutions of patient sera. End-point titers were read blindly and specific immunoglobulin binding was assessed by the presence or absence of a specular fluorescence pattern in the nuclei of the plated cells.
  • PBS phosphate-buffer saline
  • KS330Bam and KS631Bam are genomic fragments from a new and previously uncharacterized herpesvirus
  • KS5 lambda phage clone
  • the KS5 insert was subcloned after Notl/BamHI digestion into five subfragments and both strands of each fragment were sequenced by primer walking or nested deletion with a 7-fold average redundancy.
  • the KS5 sequence is 20,705 bp in length and has a G+C content of 54.0%.
  • the observed/expected CpG dinucleotide ratio is 0.92 indicating no overall CpG suppression in this region.
  • ORF Open reading frame analysis identified 15 complete ORFs with coding regions ranging from 231 bp to 4128 bp in length, and two incomplete ORFs at the termini of the KS5 clone which were 135 and 552 bp in length ( Figure 12) .
  • the coding probability of each ORF was analyzed using GRAIL 2 and CodonPreference which identified 17 regions having excellent to good protein coding probabilities. Each region is within an ORF encoding a homolog to a known herpesvirus gene with the exception of one ORF located at the genome position corresponding to ORF28 in herpesvirus saimiri
  • HVS histone deficiency virus
  • MCP conserved herpesvirus major capsid
  • gH glycoprotein H
  • TK thymidine kinase
  • ORF29a/ORF29b putative DNA packaging protein
  • the KS5 sequence spans a region which includes three of the seven conserved herpesvirus gene blocks ( Figure 14) [10] .
  • ORFs present in these blocks include genes which encode herpesvirus virion structural proteins and enzymes involved in DNA metabolism and replication. Amino acid identities between KS5 ORFs and HVS ORFs range from 30% to 60%, with the conserved MCP ORF25 and ORF29b genes having the highest percentage amino acid identity to homologs in other gammaherpesviruses.
  • KSHV ORF28 which has no detectable sequence homology to HVS or EBV genes, has positional homology to HVS ORF28 and EBV BDLF3.
  • ORF28 lies at the junction of two gene blocks ( Figure 14) ; these junctions tend to exhibit greater sequence divergence than intrablock regions among herpesviral genomes [17] .
  • Two ORFs were identified with sequence homology to the putative spliced protein packaging genes of HVS (ORF29a/ORF29b) and herpes simplex virus type 1 (UL15) .
  • the KS330Bam sequence is located within KSHV ORF26, whose HSV-1 counterpart, VP23 , is a minor virion structural component .
  • KSHV TK homolog contains a proline-rich domain at its amino terminus (nt 20343-
  • TK bovine herpesvirus 4 TK
  • KS5 translated amino acid sequences were searched against the PROSITE Dictionary of Protein Sites and Patterns (Dr. Amos Bairoch, University of Geneva, Switzerland) using the computer program Motifs.
  • sequence motif matches were identified among KSHV hypothetical protein sequences. These matches included: (i) a cytochrome c family heme-binding motif in ORF33 (CVHCHG; aa 209-214) and ORF34 (CLLCHI; aa 257-261) , (ii) an immunoglobulin and major histocompatibility complex protein signature in ORF25
  • FIG. 1 (FICQAKH; aa 1024-1030) , (iii) a mitochondrial energy transfer protein motif in ORF26 (PDDITRMRV; aa 260- 268) , and (iv) the purine nucleotide binding site identified in ORF21.
  • the purine binding motif is the only motif with obvious functional significance.
  • a cytosine-specific methylase motif present in HVS ORF27 is not present in KSHV 0RF27. This motif may play a role in the ethylation of episomal DNA in cells persistently infected with HVS [1] .
  • KSHV was most closely associated with HVS. Similar results were obtained for single-gene alignments of TK and UL15/ORF29 sets but with lower bootstrap scores so that among gamma-2 herpesvirus members branching orders for EHV2, HVS and KSHV were not resolved.
  • CS1 combined gammaherpesvirus gene set
  • the total length of CS1 was 4247 residues after removal of positions containing gaps introduced by the alignment process in one or more of the sequences.
  • the CS1 alignment was analyzed by the ML method, giving the tree shown in Figure 15B and by the MP and NJ methods used with the aligned herpesvirus MCP sequences. All three methods identified KSHV and HVS as sister groups, confirming that KSHV belongs in the gamma-2 sublineage with HVS as its closest known relative.
  • HVS and EHV2 lineages may have been contemporary with divergence of the primate and ungulate host lineages [33] .
  • the results for the CS1 set suggest that HVS and KSHV represent a lineage of primate herpesviruses and, based on the distance between KSHV and HVS relative to the position of EHV2, divergence between HVS and KSHV lines is ancient.
  • HMW high density polymorphism
  • Figure 16A arrow
  • EBV terminal repeat sequence [40] yielding a 150-160 kb band
  • Figure 16B The HMW EBV band may correspond to either circular or concatemeric EBV DNA [24] .
  • the phorbol ester TPA induces replication-competent EBV to enter a lytic replication cycle [49] .
  • TPA induces replication of KSHV and EBV in BCBL-1 cells
  • these cells were incubated with varying concentrations of TPA for 48 h ( Figure 17) .
  • Maximum stimulation of EBV occurred at 20 ng/ml TPA which resulted in an eight-fold increase in hybridizing EBV genome. Only a 1.3-1.4 fold increase in KSHV genome abundance occurred after 20-80 ng/ml TPA incubation for 48 h.
  • BCBL-1 cells Prior to determining that the agent was likely to be a member of Herpesviridae by sequence analysis, BCBL-1 cells were cultured with Raji cells, a nonlytic EBV transformed B cell line, in chambers separated by a 0.45 ⁇ tissue culture filter. Recipient Raji cells generally demonstrated rapid cytolysis suggesting transmission of a cytotoxic component from the BCBL-1 cell line.
  • Raji line cultured in 10 ng/ml TPA for 2 days underwent an initial period of cytolysis before recovery and resumption of logarithmic growth.
  • This cell line (RCC-1) is a monoculture derived from
  • RCC-1 has remained positive for the KS330 233 PCR product for > 6 months in continuous culture (approximately 70 passages) , but KSHV was not detectable by dot or
  • IFA Indirect immunofluorescence antibody assays
  • BCBL-1 instead of BHL-6 cells, by pre-adsorbing with
  • EBV-infected nonproducer Raji cells instead of P3H3 and by using sera from a homosexual male KS patient without HIV infection, in complete remission for KS for 9 months (BHL-6 titer 1:450, P3H3 titer 1:150) .
  • KS5 has a 54.0%
  • the CpG dinucleotide frequency in herpesviruses varies from global CpG suppression among gammaherpesviruses to local CpG suppression in the betaherpesviruses, which may result from deamination of 5' -methylcytosine residues at CpG sites resulting in TpG substitutions
  • the 20,705 bp KS5 fragment has 17 protein-coding regions, 15 of which are complete ORFs with appropriately located TATA and polyadenylation signals, and two incomplete ORFs located at the phage insert termini. Sixteen of these ORFs correspond by sequence and collinear positional homology to 15 previously identified herpesviral genes including the highly conserved spliced gene. The conserved positional and sequence homology for KSHV genes in this region are consistent with the possibility that the biological behavior of the virus is similar to that of other gammaherpesviruses.
  • KS5 thymidine kinase-like gene on KS5 implies that the agent is potentially susceptible to TK-activated DNA polymerase inhibitors and like other herpesviruses possesses viral genes involved in nucleotide metabolism and DNA replication [41] .
  • the presence of major capsid protein and glycoprotein H gene homologs suggest that replication competent virus would produce a capsid structure similar to other herpesviruses.
  • KSHV belongs to the gamma-2 sublineage of the Gammaherpesvirinae subfamily, and is thus the first human gamma-2 herpesvirus identified. Its closest known relative based on available sequence comparisons is HVS, a squirrel monkey gamma-2 herpesvirus that causes fulminant polyclonal T cell lymphoproliferative disorders in some New World monkey species. Data for the gamma-2 sublineage are sparse: only three viruses (KSHV, HVS and EHV2) can at present be placed on the 9 PC-7US95/15138
  • KSHV and HVS appear to represent a lineage of primate gamma-2 viruses.
  • McGeoch et al. [33] proposed that lines of gamma-2 herpesviruses may have originated by cospeciation with the ancestors of their host species. Extrapolation of this view to KSHV and HVS suggests that these viruses diverged at an ancient time, possibly contemporaneously with the divergence of the Old World and New World primate host lineages.
  • Gammaherpesviruses are distinguished as a subfamily by their lymphotrophism [41] and this grouping is supported by phylogenetic analysis based on sequence data [33] .
  • the biologic behavior of KSHV is consistent with its phylogenetic designation in that KSHV can be found in in vitro lymphocyte cultures and in in vivo samples of lymphocytes [3] .
  • This band appears to be a linear form of the genome because other "high molecular weight" bands are present for both EBV and KSHV in BCBL-1 which may represent circular forms of their genomes.
  • the linear form of the EBV genome, associated with replicating and packaged DNA [41] migrates substantially faster than the closed circular form associated with latent viral replication [24] .
  • the 270 kb band appears to be a linear form, it is also consistent with a replicating dimer plasmid since the genome size of HVS is approximately 135 kb. The true size of the genome may only be resolved by ongoing mapping and sequencing studies .
  • the EBV strain infecting Raji for example, is an BALF-2 deficient mutant [19] ; virus replication is not inducibile with TPA and its genome is maintained only as a latent circular form [23, 33] .
  • the EBV strain coinfecting BCBL-1 does not appear to be replication deficient because TPA induces eight-fold increases in DNA content and has an apparent linear form on CHEF electrophoresis.
  • KSHV replication is only marginally induced by comparable TPA treatment indicating either insensitivity to TPA induction or that the genome has undergone loss of genetic elements required for TPA induction. Additional experiments, however, indicate that KSHV DNA can be pelleted by high speed centrifugation of filtered organelie-free, DNase I-protected BCBL-1 cell extracts, which is consistent with KSHV encapsidation.
  • KSHV DNA Transmission of KSHV DNA from BCBL-1 to a variety of recipient cell lines is possible and KSHV DNA can be maintained at low levels in recipient cells for up to 70 passages.
  • detection of virus genome in recipient cell lines by PCR may be due to physical association of KSHV DNA fragments rather than true infection. This appears to be unlikely given evidence for specific nuclear localization of the ORF26 sequence in RCC-1. If transmission of infectious virus from BCBL-1 occurs, it is apparent that the viral genome declines in abundance with subsequent passages of recipient cells. This is consistent with studies of spindle cell lines derived from KS lesions. Spindle cell cultures generally have PCR detectable KSHV genome when first explanted, but rapidly lose viral genome after initial passages and established spindle cell cultures generally do not have detectable KSHV sequences [3] .
  • HLA self-human histocompatibility leukocyte antigens
  • KS Patient Enrollment Cases and controls were selected from ongoing cohort studies based on the availability of clinical information and appropriate PBMC samples. 21 homosexual or bisexual men with AIDS who developed KS during their participation in prospective cohort studies were identified [14-16] . Fourteen of these patients had paired PBMC samples collected after KS diagnosis (median +4 months) and at least four months prior to KS diagnosis (median -13 months) , while the remaining 7 had paired PBMC taken at the study visit immediately prior to KS diagnosis (median -3 months) and at entry into their cohort study (median -51 months prior to KS diagnosis) .
  • DNA Extraction and Analyses DNA from 10 6 -10 7 PBMC in each specimen was extracted and quantitated by spectrophotometry. Samples were prepared in physically isolated laboratories from the laboratory where polymerase chain reaction (PCR) analyses were performed. All samples were tested for amplifiability using primers specific for either the HLA-DQ locus (GH26/GH27) or b-globin [18] . PCR detection of KSHV DNA was performed as previously described [7] with the following nested primer sets: No. 1 outer 5'- AGCACTCGCAGGGCAGTACG-3' , 5' -GACTCTTCGCTGATGAACTGG-3' ; No.
  • the outer primer set was amplified for 35 cycles at 94° C for 30 seconds, 60° C for 1 minute and 72° C for 1 minute with a 5 minute final extension cycle at 72° C.
  • One to three ml of the PCR product was added to the inner PCR reaction mixture and amplified for 25 additional cycles with a 5 minute final extension cycle.
  • Primary determination of sample positivity was made with primer set No. 1 and confirmed with either primer sets 2 or 3 which amplify nonoverlapping regions of the KSHV hypothetical major capsid gene. Sampling two portions of the KSHV genome decreased the likelihood of intraexperimental PCR contamination.
  • KSHV Positivitv of Case and Control PBMC Samples Paired PBMC samples were available from each KS patient and homosexual/bisexual control patient; a single sample was available from each hemophilic control patient.
  • the number of KSHV positive control PBMC specimens from both homosexual/bisexual (second visit) and hemophilic patient controls was significantly lower. Only 2 of 19 (11%) hemophilic PBMC samples were positive (odds ratio 11.3, 95 % confidence interval 1.8 to 118) and only 2 of 23 (9%) PBMC samples from homosexual/bisexual men who did not develop KS were positive (odds ratio 14.0, 95% confidence interval 2.3 to 144) . If all KS patient PBMC samples taken immediately prior to or after diagnosis were truly infected, the PCR assay was at least 57% sensitive in detecting KSHV infection among PBMC samples.
  • FIG. 20A-20B The figure shows that 7 of the paired KS patient samples were positive at both visits, 5 KS patients and 2 control patients converted from negative to positive and two KS patients and one control patient reverted from positive to negative between visits. The remaining 7 KS patients and 20 control patients were negative at both visits.
  • the median length of time between the first sample and the KS diagnosis was 19 months.
  • Three of the 6 KS patients that were negative at both visits had their last PBMC sample drawn 2-3 months prior to onset of illness. It is unknown whether these patients became infected between their last study visit and the KS diagnosis date.
  • KSHV preferentially infects CD19+ B cells by PBMC subset examination of three patients [19] .
  • Other gammaherpesviruses such as Epstein-Barr virus (EBV) and herpesvirus saimiri are also lymphotrophic herpesviruses and can cause lymphoproliferative disorders in primates [11, 20] .
  • EBV Epstein-Barr virus
  • herpesvirus saimiri are also lymphotrophic herpesviruses and can cause lymphoproliferative disorders in primates [11, 20] .
  • KSHV like most human herpesviruses, is a ubiquitous infection of adults
  • EBV for example, is detectable by PCR in CD19+
  • the findings are in contrast to PCR detection of KSHV DNA in all 10 PBMC samples from KS patients by Ambroziak et al. [19] . It is possible that the assay was not sensitive enough to detect virus in all samples since it was required that each positive sample to be repeatedly positive by two independent primers in blinded PCR assays. This appears unlikely, however, given the sensitivity of the PCR nested primer sets.
  • the 7 KS patients who were persistently negative on both paired samples may represent an aviremic or low viral load subpopulation of KS patients.
  • the PCR conditions test a DNA amount equivalent to approximately 2xl0 3 lymphocytes; an average viral load less than 1 copy per 2xl0 3 cells may be negative in the assay.
  • the study was designed to answer the fundamental question of whether or not infection with KSHV precedes development of the KS phenotype.
  • the findings indicate that there is a strong antecedent association between KSHV infection and KS. This temporal relationship is an absolute requirement for establishing that KSHV is central to the causal pathway for developing KS. This study contributes additional evidence for a possible causal role for this virus in the development of KS.
  • Roizman B The family Herpesviridae. In: Roizman B, Whitley RJ, Lopez C, eds. The Human
  • Archival KS biopsy specimens were selected from approximately equal numbers of HIV- associated and endemic HIV-negative KS patients enrolled in an ongoing case-control study of cancer and HIV infection at Makerere University, Kampala Kenya. Control tissues were consecutive archival biopsies from patients with various malignancies enrolled in the same study, chosen without prior knowledge of HIV serostatus. All patients were tested for HIV antibody (measured by Cambridge Bioscience Recombigen Elisa assay) .
  • Tissue preparation Each sample examined was from an individual patient. Approximately ten tissue sections were cut (10 micron) from each paraffin block using a cleaned knife blade for each specimen. Tissue sections were deparaffinized by extracting the sections twice with 1 ml xylene for 15 min. followed by two extractions with 100% ethanol for 15 min. The remaining pellet was then resuspended and incubated overnight at 50° C in 0.5 ml of lysis buffer (25 mM KC1, 10 mM Tris-HCl, pH 8.3, 1.4 mM MgCl2, 0.01% gelatin, 1 mg/ml proteinase K) .
  • lysis buffer 25 mM KC1, 10 mM Tris-HCl, pH 8.3, 1.4 mM MgCl2, 0.01% gelatin, 1 mg/ml proteinase K
  • DNA was extracted with phenol/chloroform, ethanol precipitated and resuspended in 10 mM Tris-HCl, 0.1 mM EDTA, pH 8.3.
  • PCR Amplification 0.2-0.4 ug of DNA was used in PCR reactions with KS330 233 primers as previously described [7] .
  • the samples which were negative were retested by nested PCR amplification, which is approximately 10 2 - 10 3 fold more sensitive in detecting KS330 233 sequence than the previously published KS330 233 primer set [7] . These samples were tested twice and samples showing discordant results were retested a third time.
  • Tumors examined in the control group included carcinomas of the breast, ovaries, rectum, stomach, and colon, fibrosarcoma, lymphocytic lymphomas, Hodgkin's lymphomas, choriocarcinoma and anaplastic carcinoma of unknown primary site.
  • the median age of AIDS-KS patients was 29 years (range 3-50) compared to 36 years (range 3-79) for endemic KS patients and 38 years (range 21-73) for cancer controls.
  • KS lesions 39 of 44 (89%) were positive for KS330 233 PCR product, including KS tissues from 22 of 24 (92%) HIV seropositive and 17 of 20 (85%) HIV seronegative patients.
  • 3 of 22 (14%) nonKS cancer control tissues were positive, including 1 of 7 (14%) HIV seropositive and 2 of 15 (13%) HIV seronegative control patients ( Figure 19) .
  • These control patients included a 73 year old HIV seronegative male and a 29 year old HIV seronegative female with breast carcinomas, and a 36 year old HIV seropositive female with ovarian carcinoma.
  • the odds ratios for detecting the sequences in tissues from HIV seropositive and HIV seronegative cases and controls was 66 (95% confidence interval (95% C.I.) 3.8-3161) and 36.8 (95% C.I. 4.3-428) respectively.
  • the overall weighted Mantel-Haenzel odds ratio stratified by HIV serostatus was 49.2 (95% C.I. 9.1-335) .
  • KS tissues from four HIV seropositive children (ages 3, 5, 6, and 7 years) and four HIV seronegative children (ages 3, 4, 4, and 12 years) were all positive for KS330 233 .
  • KSHV DNA sequences are found not only in AIDS-KS [5] , classical KS [6] and transplant KS [7] but also in African KS from both HIV seropositive and seronegative patients. Despite differences in clinical and epidemiological features, KSHV DNA sequences are present in all major clinical subtypes of KS from widely dispersed geographic settings.
  • KSHV infection in over 200 cancer control tissues with the exception of an unusual AIDS-associated, body-cavity-based lymphoma [9] .
  • DNA-based detection of KSHV infection is rare in most nonKS cancer tissues from developed countries.
  • KSHV infection has been reported in post-transplant skin tumors, although well-controlled studies are needed to confirm that these findings are not due to PCR contamination [10] . Since the rate of HIV-negative KS is much more frequent in Kenya than the United States, detection of KSHV in control tissues from cancer patients in the study may reflect a relatively high prevalence infection in the general Kenyan population.
  • MOLECULE TYPE DNA (genomic)
  • HYPOTHETICAL N
  • AAACCTCGTA CATATACGAC GTGCCCACCG TCCCGACCAG CAAGCCGTGG CATTTAATGC 600
  • AAATATACTC ATGCCAAAAC AAGTTTTCGC TCCCCTTCCG GACGAACGCC ACCGCTATCC 1380
  • GCTGCACCTC GATAGCTTAA ATTTAATTCC GGCGATTAAC TGTTCAAAGA TTACAGCCGA 3780
  • CAGACTGTTT TCCATCCTTT ATTAGACGGT CAATAAAGCG TAGATTTTTA AAAGGTTTCC 4320 TGTGCATTCT TTTTGTATGG GCATATACTT GGCAAGAAAT CCGAGCACCT CAGAAAGTGG 4380
  • CAGAAAGCAT TTCAGCGTAC CCATTGCGAA GAGAAAGTGC AGCATGTCCC CACTGATGTT 6300
  • CTCATCAGTC ATCGCCCCGG CCCACGTGGC CGCCATAACT ACAGACATGG GAGTACATTG 11100

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Virology (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention porte sur une molécule d'ADN isolée d'une longueur d'au moins 30 nucléotides et qui définit spécifiquement un herpesvirus associé à la maladie de Kaposi; elle porte également sur un herpesvirus isolé associé à la maladie de Kaposi; elle porte de plus sur un anticorps spécifique au peptide et sur des molécules oligonucléotidiques antisens et triplex. L'invention a en outre trait à une méthode de vaccination contre la maladie de Kaposi, de diagnostic et de traitement prophylactique d'un patient atteint de la maladie de Kaposi, et de détection, dans une cellule de l'expression d'un virus à ADN associé à ladite maladie.
PCT/US1995/015138 1994-08-18 1995-11-21 Sequences virales specifiques associees a la maladie de kaposi et leurs utilisations WO1996015779A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU43670/96A AU4367096A (en) 1994-11-21 1995-11-21 Unique associated kaposi's sarcoma virus sequences and uses thereof
US08/757,669 US6183751B1 (en) 1994-08-18 1996-11-29 Unique associated Kaposi's Sarcoma virus sequences and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US08/343,101 US5830759A (en) 1994-08-18 1994-11-21 Unique associated Kaposi's sarcoma virus sequences and uses thereof
US08/343,101 1994-11-21
US08/420,235 US5801042A (en) 1994-08-18 1995-04-11 Unique associated Kaposi's sarcoma virus sequences and uses thereof
US08/420,235 1995-04-11

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/010194 Continuation-In-Part WO1996006159A1 (fr) 1994-08-18 1995-08-11 Sequences uniques associees de virus de la maladie de kaposi et utilisation desdites sequences

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US59296396A Continuation-In-Part 1994-08-18 1996-01-29

Publications (1)

Publication Number Publication Date
WO1996015779A1 true WO1996015779A1 (fr) 1996-05-30

Family

ID=26993330

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/015138 WO1996015779A1 (fr) 1994-08-18 1995-11-21 Sequences virales specifiques associees a la maladie de kaposi et leurs utilisations

Country Status (2)

Country Link
AU (1) AU4367096A (fr)
WO (1) WO1996015779A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0804547A1 (fr) * 1994-08-18 1997-11-05 The Trustees of Columbia University in the City of New York Sequences uniques associees de virus de la maladie de kaposi et utilisation desdites sequences
US5861240A (en) * 1996-02-28 1999-01-19 The Regents Of The University Of California Isolated human herpesvirus type 8 sequences and uses thereof
WO1999006582A1 (fr) * 1997-08-01 1999-02-11 Koszinowski Ulrich H Vecteur recombine contenant des sequences de genome virales infectieuses d'une taille superieure a 100 kb, son procede de production et son utilisation pour la mutagenese des sequences virales
WO1999062938A2 (fr) * 1998-05-29 1999-12-09 Octavian Schatz Nouveaux (poly)peptides representant des epitopes du virus 8 de l'herpes humain
US6114110A (en) * 1996-01-16 2000-09-05 University Of Michigan Isolation and propagation of a human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells
US6348586B1 (en) 1996-07-25 2002-02-19 The Trustees Of Columbia University In The City Of New York Unique associated Kaposi's sarcoma virus sequences and uses thereof
US7368557B2 (en) 1999-12-03 2008-05-06 Consejo Superior De Investigationes Cientificas Polynucleotides encoding porcine transmissible gastroenteritis virus
US7932066B2 (en) 1994-08-18 2011-04-26 The Trustees Of Columbia University In The City Of New York Unique associated kaposi's sarcoma virus sequences and uses thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BOVI D. ET AL: "Isolation of a Rearranged Human Transforming Gene Following Transfection of Kaposi Sarcoma DNA.", PNAS, vol. 84, no. 16, August 1987 (1987-08-01), pages 5660 - 5664 *
GALLO: "Aspects of the Molecular Pathogenesis of AIDS.", JOURNAL OF CELLULAR BIOCHEMISTRY, vol. 17E, January 1987 (1987-01-01) *
GALLO: "New approaches for Interfering with Human Immunodeficiency Virus Replication and for Kaposi's Sarcoma.", JOURNAL OF CELLULAR BIOCHEMISTRY, vol. 18B, January 1994 (1994-01-01), pages 108 *
MARSHALL E.: "GENE THERAPY'S GROWING PAINS", SCIENCE, AMERICAN ASSOCIATION FOR THE ADVANCEMENT OF SCIENCE, vol. 269, 25 August 1995 (1995-08-25), pages 1050 - 1055, XP000676418 *
MOORE P.S.: "PRIMARY CHARACTERIZATION OF A HERPESVIRUS AGENT ASSOCIATED WITH KAPOSI'S SARCOMA.", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, vol. 70, no. 1, 1996, pages 549 - 558, XP002050060 *
MOSCA ET AL: "Herpes Simplex Virus Type-1 Can Reactivate Transcription of Latent Human Immunodeficiency Virus.", NATURE, January 1987 (1987-01-01), pages 67 - 70 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0804547A4 (fr) * 1994-08-18 1999-11-03 Univ Columbia Sequences uniques associees de virus de la maladie de kaposi et utilisation desdites sequences
US7932066B2 (en) 1994-08-18 2011-04-26 The Trustees Of Columbia University In The City Of New York Unique associated kaposi's sarcoma virus sequences and uses thereof
EP0804547A1 (fr) * 1994-08-18 1997-11-05 The Trustees of Columbia University in the City of New York Sequences uniques associees de virus de la maladie de kaposi et utilisation desdites sequences
US6114110A (en) * 1996-01-16 2000-09-05 University Of Michigan Isolation and propagation of a human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells
US5861240A (en) * 1996-02-28 1999-01-19 The Regents Of The University Of California Isolated human herpesvirus type 8 sequences and uses thereof
US6348586B1 (en) 1996-07-25 2002-02-19 The Trustees Of Columbia University In The City Of New York Unique associated Kaposi's sarcoma virus sequences and uses thereof
WO1999006582A1 (fr) * 1997-08-01 1999-02-11 Koszinowski Ulrich H Vecteur recombine contenant des sequences de genome virales infectieuses d'une taille superieure a 100 kb, son procede de production et son utilisation pour la mutagenese des sequences virales
US7892822B1 (en) 1997-08-01 2011-02-22 Koszinowski Ulrich H Recombinant vector containing infectious, viral genome sequences greater than 100 kb, method for producing same and use for the mutagenesis of the viral sequences
WO1999062938A2 (fr) * 1998-05-29 1999-12-09 Octavian Schatz Nouveaux (poly)peptides representant des epitopes du virus 8 de l'herpes humain
WO1999062938A3 (fr) * 1998-05-29 2000-03-23 Octavian Schatz Nouveaux (poly)peptides representant des epitopes du virus 8 de l'herpes humain
US6669939B1 (en) 1998-05-29 2003-12-30 Biotrin International Properties Limited (Poly)peptides which represent the epitopes of the human herpes virus type 8
US7368557B2 (en) 1999-12-03 2008-05-06 Consejo Superior De Investigationes Cientificas Polynucleotides encoding porcine transmissible gastroenteritis virus
US7445928B2 (en) 1999-12-03 2008-11-04 Consejo Superior De Investigationes Cientificas Bacterial artificial chromosome construct encoding recombinant coronavirus

Also Published As

Publication number Publication date
AU4367096A (en) 1996-06-17

Similar Documents

Publication Publication Date Title
US5854398A (en) Kaposi's sarcoma-associated herpesvirus (KSHV) interleukin 6 (IL-6) and uses thereof
US5830759A (en) Unique associated Kaposi's sarcoma virus sequences and uses thereof
Cesarman et al. Kaposi's sarcoma-associated herpesvirus contains G protein-coupled receptor and cyclin D homologs which are expressed in Kaposi's sarcoma and malignant lymphoma
AU705890B2 (en) Latent transcripts and promoters of cytomegalovirus
Wang et al. Simian homologues of Epstein–Barr virus
US20110265195A1 (en) Unique associated kaposi's sarcoma virus sequences and uses thereof
US6348586B1 (en) Unique associated Kaposi's sarcoma virus sequences and uses thereof
Mercer et al. Pathogenesis of acute murine cytomegalovirus infection in resistant and susceptible strains of mice
US5801042A (en) Unique associated Kaposi's sarcoma virus sequences and uses thereof
US5849564A (en) Polypeptides from Kaposi's sarcoma-associated herpesvirus, DNA encoding same and uses thereof
AU700913B2 (en) Unique associated kaposi's sarcoma virus sequences and uses thereof
AU1757197A (en) Unique associated kaposi's sarcoma virus sequences and uses thereof
US5831064A (en) Kaposi's sarcoma-associated herpes virus (KSHV) interferon consensus sequence binding protein (ICSBP) and uses thereof
US5948676A (en) Immediate early protein from Kaposi's sarcoma-associated herpesvirus, DNA encoding same and uses thereof
WO1996015779A1 (fr) Sequences virales specifiques associees a la maladie de kaposi et leurs utilisations
US5854418A (en) Kaposi's sarcoma-associated herpesvirus (KSHV) viral macrophage inflammatory protein-1α II (vMIP-1α II) and uses thereof
US5861500A (en) Kaposi's sarcoma-associated herpesvirus (KSHV) interleukin 6 (IL-6) and uses thereof
CA2261164A1 (fr) Sequences de virus uniques associees au sarcome de kaposi et utilisations de ces dernieres
US5863787A (en) Kaposi's sarcoma-associated herpesvirus (KSHV) glycoprotein B (GB) and uses thereof
US6150093A (en) Unique associated Kaposi's sarcoma virus sequences and uses thereof
US5859225A (en) Virion protein 26 from Kaposi's sarcoma-associated herpesvirus, DNA encoding same and uses thereof
US5853734A (en) Glycoprotein L and clycoprotein M from kaposi's sarcoma associated herpesvirus, DNA encoding same and uses thereof
Taniguchi et al. Structure of transcripts and proteins encoded by U79-80 of human herpesvirus 6 and its subcellular localization in infected cells
EP0726708A1 (fr) Sclerose en plaques associee a un virus: traitements, prevention et diagnostic
D'Aquila et al. Isolation and characterization of phosphonoacetic acid-resistant mutants of human cytomegalovirus

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP MX US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA