WO1997026373A1 - Isolation and propagation of a human herpesvirus derived from aids-associated kaposi's sarcoma cells - Google Patents

Isolation and propagation of a human herpesvirus derived from aids-associated kaposi's sarcoma cells Download PDF

Info

Publication number
WO1997026373A1
WO1997026373A1 PCT/US1997/000019 US9700019W WO9726373A1 WO 1997026373 A1 WO1997026373 A1 WO 1997026373A1 US 9700019 W US9700019 W US 9700019W WO 9726373 A1 WO9726373 A1 WO 9726373A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
cells
hhv
infected
culture
Prior art date
Application number
PCT/US1997/000019
Other languages
French (fr)
Inventor
Gary J. Nabel
Brian J. Nickoloff
Original Assignee
The University Of Michigan
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The University Of Michigan filed Critical The University Of Michigan
Publication of WO1997026373A1 publication Critical patent/WO1997026373A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5091Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing the pathological state of an organism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/245Herpetoviridae, e.g. herpes simplex virus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5064Endothelial cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • G01N33/56994Herpetoviridae, e.g. cytomegalovirus, Epstein-Barr virus
    • 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
    • C12N2710/16421Viruses as such, e.g. new isolates, mutants or their genomic sequences
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/16011Herpesviridae
    • C12N2710/16411Rhadinovirus, e.g. human herpesvirus 8
    • C12N2710/16434Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • the present invention relates to the isolation and propagation of a novel human herpesvirus derived from AIDS- associated Kaposi' s Sarcoma cells, diagnostic methods using the same, methods for screening for anti-viral drugs using the same and vaccines against Kaposi' s Sarcoma.
  • KS Kaposi' s sarcoma
  • HAV-l human immunodeficiency virus -1
  • AIDS-related KS also known as epidemic KS
  • other forms of KS including classical (Mediterranean) , African (endemic) , and iatrogenic immunosuppressive-drug associated with organ transplantation [reviewed in (4,5)].
  • AIDS-KS lesions can involve incer-nal organs, particularly the lungs and gastrointestinal tract, resulting in severe and potentially fatal hemorrhagic disease (6) .
  • KS is characterized histologically by a proliferation of spindle-shaped tumor cells mixed wich endothelial cells forming blood vessels (angiogenesis), fibroblasts, dermal dendrocytes, and an inflammatory cell infiltrate (7) .
  • the origin of the spindle-shaped KS tumor cells has been difficult to define with certainty, but is thought to derive from either endothelial or vascular smooth muscle cells (8-11) .
  • a variety of immune or inflammatory cytokines and oncogenes or viral proteins have been found m association with these lesions (11- 15) . While the etiologic agent causing KS is unclear, epidemiological data has suggested that an infectious agent could potentially spread the disease through sexual contact (16) .
  • herpesvirus-like DNA sequences were homologous to, but distinct from, minor cagsid and tegument proteins of Epstein-Barr virus (EBV) and herpesvirus saimiri (17) , and defined a putative new member of the ga maherpesvirus family which is currently referred to as KS-as ⁇ ociated herpesvirus, KSHV, or human herpesvirus 8, HHV-8.
  • EBV Epstein-Barr virus
  • KSHV herpes ⁇ ociated herpesvirus
  • HHV-8 human herpesvirus 8
  • this line contained a second herpesvirus, ⁇ BV, which did not allow definitive isolation or identification of the KS-associated virus. In addition, it did not provide a means to replicate or propagate the virus which was carried in these cells but could not be amplified.
  • the ability now to propagate and clone this virus allows specific diagnostic tests to be developed, by allowing the definition of the full range of the DNAs, RNAs and proteins in the native virus, by permitting the generation of antibodies to diagnose the virus, and by providing a means to establish its epidemiologic association with the disease. It also establishes a method to generate a prototype virus which may be useful for the development of a vaccine to prevent this disease.
  • one object of this invention is to provide a method for cultivating KS tumor cells that contain HHV-8 virus in a nonrepiicative state.
  • a second object cf the present invention is to provide an intact human herpesvirus derived from AIDS-associated Kaposi' s Sarcoma cells (HHV-8) .
  • a third object of the present invention is to provide methods for propagating human herpesvirus derived from AIDS- associated Kaposi' s Sarcoma cells in vi tro .
  • a fourth object of the present invention is to provide diagnostic methods for detecting human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells in a patient.
  • a fifth object of the present invention is to provide methods for screening for antiviral drugs using cells infected with human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells.
  • the present inventors have now achieved these and other objects by isolating human herpesvirus and discovering methods for propagating the same.
  • the present inventors utilized growth conditions that allow multiple passages of primary KS cell lines.
  • KSHV, or HHV-8 DNA sequences were detected in early passages of these tumor cells which were nearly identical to the previously described herpes-like DNA sequences (17,19) .
  • By co-culture with the human embryonal kidney epithelial cell line, 293 cells cell-free viral lysate was isolated which induced a cytopathic effect on 293 cells. Characterization of the virus was performed, and propagation by serial passage was confirmed.
  • Figure 1 Phase contrast and transmission electron microscopy of isolated KS cells. (Magnification: (A) xio,- (B) x20, inset x70,000) .
  • Figure 2 Detection of viral DNA sequences in 293 cell initially co-cultured with KS cells and subsequently serial passage of virus.
  • Figure 3 293 cells from control (uninfected) or those exposed to cell-free viral lysates (infected) for the indicated number of days in vi tro before analysis by phase contrast microscopy. (Magnification: xlO) .
  • FIG. 5 Light and electron microscopic analysis of 293 cells before (panel A) and 5 days after (panels B anc C) exposure to lysate. Electron microscopic analysis of 293 cells 2 days after infection (D) , with inset showing a nuclear virus- like particle. Arrows indicate multinucleated giant cells or show an intact virion within the nucleus of an infected 293 cell. (Magnification: A, x 20, B, x 20, C x 20, D x 29,200, D, inset x 62,800) . DESCRIPTION CF THE PREFERRED EMBODIMENTS Cultivation cf Kaposi's Sarcoma Cells that Contain HHV-8 Virus in a Nonreplicative State
  • the method of cultivating KS cells according to the present invention comprises the steps cf : isolating a ' ⁇ S ceil from a KS lesion cr tissue taken from a patient biopsy; and culturing said KS cell on a media coated with endothelial cell attachment factor.
  • KS ceils can be isolated from tissue taken from a patient biopsy or, preferably, from KS lesions of the skin from HIV- positive patients. These cells can be plated on tissue culture dishes coated with microvascular endothelial cell attachment factor and naintained in RPMI media supplemented with L-glutamine, heparin, appropriate antibiotics (preferably penicillin, streptomycin and gentamicin) , serum (preferably heat inactivated fetal bovine serum) and growth factors (preferably endothelial cell growth supplement) . Suitable serum concentrations are 0-20%. Cells can suitably be grown at a temperature range of from 35 to 40°C (preferably 37-38°C) in a C0 2 incubator (preferably in an atmosphere containing 2-5% CO.) . Cells are allowed to reach confluency. KS cells can be removed from confluent tissue cultures using conventional procedures such as exposure to trypsin and EDTA.
  • the present method differs from conventional methods which did not incorporate endothelial cell attachment factor or endothelial cell growth factor (See Nakamura et al, Science 112.:430-433 (1988) ) .
  • Intact Human- Herpesvirus 8 ⁇ HV-3 See Nakamura et al, Science 112.:430-433 (1988) .
  • HHV-8 can be isolated from KS cells.
  • HHV- 9 can be isolated from co-cultures of KS ceils and cells of epithelial or umbilical endothelial origin. Suitable epiteiial and umbilical endothelial cells are disclosed below. Any ratio of KS cells to cells of renal epithelial or umbilical endothelial origin can be used. Preferably 1:10 KS cells to cells of renal epithelial or umbilical endothelial origin can be used. Cells are co-plated onto media, optionally in the presence of growth factors such as TNF-a and allowed to reach confluency.
  • DNA can be isolated from the above tissue culture cells as previously described (28) . Briefly, the cells are washed in phosphate-buffered saline and gently lysed at room temperature. The solution in then centrifuged and the resulting supernatant treated with an enzyme such as pronase. The DNA is then extracted, precipitated, washed and dried. Isolated DNA is then amplified.
  • the DNA is then ligated into a suitable expression vector such as lambda gtlO or gt 11, Bluescript or SuperCosl (Stratagene) .
  • a suitable expression vector such as lambda gtlO or gt 11, Bluescript or SuperCosl (Stratagene) .
  • Human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells can be propagated in cells of epithelial or umbilical endothelial origin. Unlike previous methods, the method of the present invention propagates cells infected with HHV- ⁇ in the absence of HTLV-II conditioned media.
  • the method of the present invention comprises: contacting a culture of epithelial cells or umbilical endothelial cord cells with cells or cell lysate of a human herpesvirus derived from Kaposi's sarcoma cell to form an infected ceil culture, incubating the infected cell culture for at least 1 day in the presence of endothelial cell attachment factor, and passaging the infected cell culture.
  • Suitable epithelial cells can be derived from adult or, preferably embryonic sources. Suitable epithelial and umbilical endothelial cells -can be derived from primates. or, preferably, human sources. Preferred epithelial cells are renal epithelial cells. For example, 293 cells, which are human embryonal kidney epithelial cells can be used. 293 cells are available from the American Type Culture Collection. Embryonic epithelial or umbilical endothelial cells can be infected with human herpesvirus using known methods 3 to 24 hours after seeding. Suitable inoculum includes virally infected KS cells or viral lysates. The concentration of. inoculum is preferably at least 10"' .
  • the infected cell culture can be incubated at 35-60 ⁇ C in relevant media in a humidified atmosphere and appropriate 0-. and CO, concentrations.
  • the virus can be serial passages using methods known in the art. Briefly, cells infected with the virus are lysed and cell lysates are contacted with uninfected cells . Passaging is conducted after at least three days of infection. Infected cells are characterized by the presence of multinucleated giant cells, nuclear molding and dissolution of chromatin with residual chromatin marginating along the nuclear membrane. Within 5 days after infection, virtually all of the cells undergo necrosis with condensation of chromatin and other morphological changes consistent with apoptosis.
  • HHV-8 can be propated less efficiently using methods known in the art.
  • Virus can suitably be harvested from cell culture when the maximal cytopathic effect is observed. Observation of cytopathic effect in cell cultures can be endpoint determined up to 5 days; this can suitably be done by visual evaluation of morphological changes on the monolayer or by fluorescent antibody staining techniques.
  • Suitable harvest methods include: agitation, aspiration, scraping, or freeze thawing accompanied by aspiration.
  • harvested viruses are stored at -70°C.
  • the activity of the virus can be suitably measured quantitatively by preparing dilutions of the sample and determining the highest dilution (endpoint) at which activity- is still detectable.
  • the preferred method is the Reed Muench method which permits interpretation of the 50% endpoint from data derived from a quantal response.
  • the formula can be applied similarly to rates of infection in any host system.
  • the unit cf infectivity used c express the results are mean embryo infective dose, EID s:, and mean tissue culture infective dose, TCID,;.
  • Vaccines can be produced from the harvested virus and viral lysates using techniques known m the art.
  • Antibodies to the intact human herpesvirus can be generated using methods known in the art (see for example, chapter 18 of Sambrook et al, "Molecular Cloning: A Laboratory Manual", 2nd ed. , Cold Spring Harbor Laboratory Press, NY, 1989) .
  • Antibodies, preferably polyclonal, against the intact virus can be used in Western blot tests to determine the presence or absence of human herpesvirus shed from Kaposi's sarcoma cells in a biological sample such as blood, semen or urine.
  • Western blot analysis can be conduct using methods well known in the art .
  • Cells infected with human herpesvirus derived from Kaposi's sarcoma cells can be used to screen for antiviral activity.
  • the method of the present invention comprises, contacting an antiviral drug candidate with a newly infected cell (within 2 to 24 hours following infecting) , culturing the infected cell and determining the effect of the antiviral drug candidate .
  • the effect cf the antiviral drug can be determined by comparing cytopathicity of . ntreated infected cells with treated infected cells. If treated infected cells persist for at least 7 days, preferably at least 10 days, the drug has antiviral activity against human herpesvirus derived from Kaposi's sarcoma cells.
  • screening can be performed using Elisa, RIA or other methods of immune detection known in the art to detect decreased production of specific viral gene products in the presence of the antiviral drug. These tests could screen directly for antigens in immunoprecipitate.
  • the immune response cf the patient to an antigen can be screened using a western blot assay.
  • KS cells Human KS cells were isolated independently from KS lesions of the skin from five different HIV-positive patients (designated KS#l-8) through mechanical disruption and enzymatic digestion of biopsies as previously described (29) . In contrast to previous reports which used an HTLV-II conditioned media, we have developed alternative media and culture conditions for these experiments.
  • the ceils were plated cn tissue culture dishes (Corning, Corning, NY) coated with microvascular endothelial cell attachment factor ( Cell Systems, Kirkland, 'WA) and were maintained in RPMI 1640 and 20% heat inactivated fetal bovine serum (FBS) supplemented with 10% utridoma HU (Boehringer Mannheim Biochemicals, Indianapolis, IN) , 2 mM L-glutamine, 100 units/ml penicillin, 100 mg/ml streptomycin, 50 mg/ml gentamicin, 50 mg/ml endothelial cell growth supplement (ECGS, ICN Biochemicals, Aurora, OH) and 16 units/ml bovine heparin.
  • FBS heat inactivated fetal bovine serum
  • Isolated KS tumor cells were characterized by immunohistochemistry as stated in the text (29,30) .
  • Human umbilical vein endothelial cells (HUVECs) were isolated by collagenase treatment of freshly obtained human umbilical cords and plated on gelatin-coated tissue culture dishes (31) .
  • RPMI-1640 and 20% heat inactivated FBS supplemented with 2 mM L-glutamine, 100 units/ml penicillin, 100 mg/ml streptomycin, 50 mg/ml gentamicin, 50 mg/ml ECGS (Collaborative Research, Bedford, MA) and 16 units/ml bovine heparin was used to maintain the cells.
  • HUVECs were characterized by a cobblestone appearance and specific staining for von Willebrand factor.
  • the transformed human embryonal kidney cell line, 293, was provided by Dr. Garry Nolan, and the cells maintained in Dulbecco's modified Eagle's media containing 10% heat inactivated FBS, 2 mM L-glutamine, 100 units/ml penicillin, and 100 mg/ml streptomycin.
  • KS cells were removed from confluent tissue culture dishes using brief exposure to 0.03% trypsin and 0.01% ethylenediaminetetraacetic acid ⁇ DTA) .
  • the ceils were pelleted and resuspended in 50% giuteraldehyde and C .
  • IM cacodylate buffer 50% giuteraldehyde and C .
  • the confluent monolayers of cells were fixed with giuteraldehyde and cacodylate buffer and removed from the plate by gently scrapping with a rubber policeman.
  • the fixed cells were embedded, sectioned and stained for electron microscopy by the departmental electron microscopy core facility.
  • KS tumor cells in confluent areas demonstrated loss of contact inhibition by forming large spherules on the dish (Figure IA) adjacent to the epithelial appearing microvascular endctheliai ceils and, on subsequent passage, only the spindle- shaped cell continued to proliferate ( Figure IB) .
  • Figure IA large spherules on the dish
  • Figure IB A representative herpes-like virus particle detected in the cytoplasm of passage 2 KS cells is shown ( Figure IB, inset) .
  • DNA was isolated from tissue culture cells as previously described (28) .
  • tissue culture cells For isolation of DNA from paraffin-embedded sections, the tissue from 50 five-micron sections was dewaxed twice with xylene, washed twice with ethanol and dried before addition of the digestion buffer.
  • Hirt supernatants were prepared as previously described (32,33) . Briefly, the cells were washed five times in phosphate-buffered saline (pH 7.0) and gently lysed for 15 minutes at room temperature in lysing solution 'O.DIM Tris ?H 3.:) , 3.6% SDS, :.01M ⁇ DTA ) .
  • Sodium chloride was added for a final concentration of IM and the mixture was incubated at 4°C overnight.
  • the solution was centrifuged at 17,000 xg for one hour at 4°C, and the resulting supernatant treated with 1 mg/ml pronase at 37°C for 3-4 hours.
  • the DMA was extracted three times with equal volumes of buffer saturated phenol and precipitated with 2 volumes of isopropanol and 1/10 volume of 3M sodium acetate at -20°C.
  • the precipitated DNA was washed successively with cold ethanol at 70%, 90% and 100%.
  • the DNA was dried and resuspended in TE buffer.
  • Isolated DNA was amplified by PCR using primers designed from the specific herpesvirus-like DNA sequence (17) .
  • the primers were 5' -AGC CGA AAG GAT TCC ACC ATT GTG CTC-3' (SEQ ID NO.:l) and 5' -TCC GTG TTG TCT ACG TCC AGA CGA TAT-3' (SEQ ID NO.:2) .
  • PCR reaction mixtures 50 ml volume) were set up containing 0.
  • DNA was amplified as follows. 94°C for 3 minutes (1 cycle), 94°C for 30 seconds, 72°C for 2 minute (40 cycles) , 72°C for 10 minutes (1 cycle) .
  • PCR products were electrophoresed through a 2% agarose gel containing ethidium bromide.
  • DNA from isolated KS cells was amplified using PCR as described. 5 ml of the PCR product was electrophoresed through a 2% agarose gel containing ethidium bromide to demonstrate successful amplification. The PCR product was then ligated into a pCRII Vector according to the manufacturer's instructions (TA Cloning Kit, Invitrogen, San Diego, CA) .. Sequencing was performed using dideoxy (Sanger) sequencing and the Sequenase kit (United States Biochemical, Cleveland, OH) . Sequencing of PCR products was performed at least two times to confirm the results. Isolation cf -Virus From Co-Cultures
  • KS cells were plated in 6-well dishes coated with microvascular endothelial cell attachment factor and allowed to grow 50 - 70% confluence.
  • Primary KS cells 2xl0 5 cells, passage 2-3) were removed from primary culture and incubated with 293 cells (2x10*) , in 35 mm 6-well Costar plates in the presence or absence of TNF-a (200 IU/ml) .
  • Cell lysates were prepared by three cycles of freeze-thawing after 3-5 days and incubated at different dilutions with 293 ceils in the absence of TNF-a. For serial propagation, half of the cell culture was used to prepare lysates at day 3 post-infection, while the remainder of the cell culture was maintained to confirm subsequent CPE.
  • PCR positivity was confirmed in the cell lysate.
  • Virus was propagated serially for at least 5 passages. Estimation of titer was made by incubation of lysates at progressive dilutions (10 * --10 "10 ) with 293 or other target cells.
  • Viral DNA was detected by PCR in Hirt supernatants of 293 cells at the indicated times after infection ( Figure 2A) and with DNA isolated from the indicated enriched subcellular fractions ( Figure 2B) .
  • the arrow denotes the specific 233 bp DNA fragment. (+) represents a positive control sample from KS lesion, (-) represents uninfected 293 cell DNA.
  • Figure 3 is a phase contrast microscopy (Magnification: xlO) of a cell.
  • cell-free virus lysates were derived by serial challenge of 293 cells and analyzed for the presence of the indicated herpesvirus sequences by PCR. Positive control samples (+) , (lanes 1,4,7,10,13), negative (-) uninfected 293 ceil DNA controls flanes 2,5,3,1 ,1-1) , and infected 233 cell DNA from ceil-free lysate (L) preparations ' lanes 3,6,9,12,15) were included. Positive control samples for HSV-1 and -2 (92 bp fragment; and KHV-6 223 bp fragment ) were derived from tissue samples of patients with documented infections. The HHV-3 positive control INA was derived from paraffin sections of pulmonary KS in an HIV-7 patient.
  • EBV positive control DNA was isolated from M16B cells, chronically infected with EBV (115 bp fragment) .
  • CMV positive controls (908 bp fragment) were derived from a control plasmid, VCL 1012, which contained this regulatory element (37) .
  • Figure 5 is Light and electron microscopic analysis of infected 293 cells. Representative light microscopic appearance of 293 cells before (panel A) and 5 days after (panel B) exposure to lysate. Electron microscopic analysis of 293 cells 2 days after infection (C) . Note that compared to uninfected cells which display uniformly viable cells with typical epithelial differentiation, the infected cells are undergoing degeneration with formation of multinucleated giant cells and chromatin alteration typical cf herpes group viral infection.
  • KS cell lines grew rapidly after initiation of culture. Initially, macroscopic clusters of cells were observed which appear to contain both epithelioid-appearmg endothelial cells and spindle-shaped mesenchymal cells ⁇ Fig. IA) . Subsequently, after the first passage, spindle-shaped cells characteristic of KS were observed, whose doubling times ranged from 24-48 hours ( Fig. IB) .
  • KS spindle cell line was positive for factor Xllla and VCAM-1 by immunohistochemical staining, and negative for factor XIIIs, E-selectin, PECAM-1 (CD31), factor VIII, and CD34. Expression of these markers was not altered by I?N-g or TNF-a stimulation.
  • PCR was used to determine whether the proliferating KS cells contained recently described herpesvirus-like DNA sequences (17) . 75% (6 of 8 isolates) of early passage, isolated KS tumor cells were positive for this DNA sequence using PCR (Table 1) .
  • DNA was isolated from the indicated cell lines, and tissue samples were analyzed by PCR. Positive or negative indicates the presence or absence of relevant size fragment by ethidium bromide staining on agarose gels. Positive control reactions for each virus type were performed using an equal concentration (1 mg) of DNA as in the test reactions.
  • the signal was present in the cell lines until passage 2 or 3. and was then no longer detectable.
  • the 233 bp PCR product was also found in DNA isolated from two separate paraffin embedded tissue samples obtained from a patient with pulmonary KS, as previously described in other patients (17,18). No evidence of KS-associated herpesvirus DNA sequences was detected in samples from isolated HUVECs, 293 cells, or DNA isolated from paraffin-embedded tissue from a psoriasis patient (Table 1). To determine if other herpesviruses might be present in these samples. PCR analysis was performed to detect HHV-6. HSV- 1 and -2. CMV. EBV. DNA derived from these isolated KS cells were negative for each of these viral sequences (Table 1). Analysis of the 233 bp PCR product revealed a sequence identical to the herpesvirus-li e DNA sequence originally
  • KS cells were co-cultured with a variety of other cell types for two to seven days.
  • free lysates were prepared and tested for the presence of viral DNA by PCR.
  • HUVEC none • +•/- +/-
  • CPE cytopathic effect
  • + + J - denotes consistent and readily visible 233 bp product by ethidium bromide staining; - indicates weaker positivity; - - indicates inconsistent detection.
  • + + + + indicates > 80% nonviable cells by day 5, 4- by day 10-1 1. + /- inconsistent effects.
  • CPE cytopathic effect
  • TNF-a enhanced viral replication during the primary KS/293 cell co-culture, it did not enhance CPE or viral titers during serial passage on 293 cells and was not used further.
  • No PCR signal was detected in uninfected cells, but a signal was readily detected in infected 293 cells within 1 day after infection.
  • PCR signals of viral DNA were maximal three days after viral challenge, and diminished on day five ( Fig. 2A), when > 80 of cells showed severe cytopathic effects (Fig. 3), possibly due to nonspecific degradation.
  • cell lysates were prepared on day 3-4, prior to the generation of CPE.-
  • the resultant cell lysates (day 3) conferred CPE and a positive PCR signal on uninfected 293 cells and was maintained consistently with titers 3 10 ⁇ 7ml for at least 5 serial passages.
  • KS cells were analyzed using transmission electron microscopy to visualize cells in greater detail and to detect the potential presence of virus.
  • evidence of particles was found in the cytoplasm whose size and shape was consistent with that of a herpes virion (Fig. IB, inset); however, particles were infrequent, and the mo ⁇ hology was not sufficient to identify the particle definitively as a herpesvirus.
  • Additional studies were undertaken to characterize the virus particle propagated on 293 cells. Cells were examined daily after exposure to cell-free viral lysates. Virus panicles were observed in the nucleus of infected cells as early as two days after infection.

Abstract

This present invention demonstrates the presence of a replication-competent herpesvirus associated with AIDS-associated Kaposi's sarcoma cells, vaccines derived therefrom, methods for diagnosing for Kaposi sarcoma, and methods for screening for antiviral drugs effective against human herpesvirus.

Description

TITLE OF THE INVENTION
Isolation and Propagation of a Human Herpesvirus Derived from AIDS-Associated Kaposi' s Sarcoma Cells
This work was supported in part by grants from the National Institutes of Health.
BACKGROUND OF THE INVENTION Field of the Invention
The present invention relates to the isolation and propagation of a novel human herpesvirus derived from AIDS- associated Kaposi' s Sarcoma cells, diagnostic methods using the same, methods for screening for anti-viral drugs using the same and vaccines against Kaposi' s Sarcoma.
Discussion of the Background
Kaposi' s sarcoma (KS) was originally described in the late 1800's as a rare and relatively benign neoplasm of elderly men of Jewish or Mediterranean descent. Today, KS is recognized as the most common malignancy in AIDS patients, affecting approximately 20% of human immunodeficiency virus -1 (HIV-l) - positive patients (1-3) . AIDS-related KS (also known as epidemic KS) is clinically more aggressive than other forms of KS, including classical (Mediterranean) , African (endemic) , and iatrogenic immunosuppressive-drug associated with organ transplantation [reviewed in (4,5)]. While often presenting in the skin of HIV-l positive patients, AIDS-KS lesions can involve incer-nal organs, particularly the lungs and gastrointestinal tract, resulting in severe and potentially fatal hemorrhagic disease (6) .
KS is characterized histologically by a proliferation of spindle-shaped tumor cells mixed wich endothelial cells forming blood vessels (angiogenesis), fibroblasts, dermal dendrocytes, and an inflammatory cell infiltrate (7) . The origin of the spindle-shaped KS tumor cells has been difficult to define with certainty, but is thought to derive from either endothelial or vascular smooth muscle cells (8-11) . In addition, a variety of immune or inflammatory cytokines and oncogenes or viral proteins have been found m association with these lesions (11- 15) . While the etiologic agent causing KS is unclear, epidemiological data has suggested that an infectious agent could potentially spread the disease through sexual contact (16) . Although several viruses, including cytomegalovirus, hepatitis B virus, and human papillomavirus were found in KS patients, no single agent was found consistently in all patient lesions until recently when Chang, Moore, and colleagues demonstrated that over 90% of AIDS-KS tissue samples were positive for herpesvirus-like DNA sequences (17) . These DNA sequences were homologous to, but distinct from, minor cagsid and tegument proteins of Epstein-Barr virus (EBV) and herpesvirus saimiri (17) , and defined a putative new member of the ga maherpesvirus family which is currently referred to as KS-asεociated herpesvirus, KSHV, or human herpesvirus 8, HHV-8. Since then, several different laboratories have demonstrated the presence of this viral sequence m tissue from KS patients with classic, African endemic and AIDS-associated KS (18-20) as well as patients with body-cavity-based lymphomas (21,22) . It has remained uncertain, however, whether this new herpesvirus was replication-competent or represented a replication-defective, adventitious virus present in KS tissue. To date, although the presence of this virus has been noted in B lymphoma cells which also contain EBV, it has not been possible to demonstrate viral replication in vitro.
Since the publication of a DNA sequence associated with Kaposi sarcoma, several reports have either supported (18- 20,23-25) or refuted '26,27) the notion that this novel DNA virus was important in KS. To date, it had not been possible to propagate this putative virus or even to maintain it in cell culture in the absence of other known herpesviruses, making it difficult to characterize its transmissibility, host cell range, and to develop anti-viral and diagnostic reagents.
It is therefore desirable to isolate intact human herpesvirus from Kaposi' s sarcoma' cells and to propagate the same in vi tro . The earliest studies in this field defined a novel viral DNA sequence associated with Kaposi's sarcoma lesions (Chang et al. , Science (1994) ££:1865-1869) . Although the sequence suggested that it was a member of the gamma herpesvirus family, it has not been possible to address the question of whether this represents a true replicating virus or a helper virus associated with another pathogen that might cause the disease. More recently, the presence of the virus DNA sequence was noted in a B cell lymphoma in a persistent form (22) . Unfortunately, this line contained a second herpesvirus, ΞBV, which did not allow definitive isolation or identification of the KS-associated virus. In addition, it did not provide a means to replicate or propagate the virus which was carried in these cells but could not be amplified. The ability now to propagate and clone this virus allows specific diagnostic tests to be developed, by allowing the definition of the full range of the DNAs, RNAs and proteins in the native virus, by permitting the generation of antibodies to diagnose the virus, and by providing a means to establish its epidemiologic association with the disease. It also establishes a method to generate a prototype virus which may be useful for the development of a vaccine to prevent this disease. This isolation of this virus will also make it possible to fulfill Koch's postulates in animal models, showing that it can transmit the genetic information which causes the disease. Finally, it provides a critical assay system for the replication of the virus and allows for large scale screening of antiviral compounds that inhibit viral replication in vitro. Without the ability to replicate the virus, these goals could not be achieved. The present invention provides a method to establish and propagate replication-competent virus which will make these goals possible and likely to succeed. It thus, represent a major advance in the field which was not obvious and widely recognized as needed. SUMMARY ? THE INVENTION Accordingly, one object of this invention is to provide a method for cultivating KS tumor cells that contain HHV-8 virus in a nonrepiicative state.
A second object cf the present invention is to provide an intact human herpesvirus derived from AIDS-associated Kaposi' s Sarcoma cells (HHV-8) .
A third object of the present invention is to provide methods for propagating human herpesvirus derived from AIDS- associated Kaposi' s Sarcoma cells in vi tro .
A fourth object of the present invention is to provide diagnostic methods for detecting human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells in a patient.
A fifth object of the present invention is to provide methods for screening for antiviral drugs using cells infected with human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells.
The present inventors have now achieved these and other objects by isolating human herpesvirus and discovering methods for propagating the same. The present inventors utilized growth conditions that allow multiple passages of primary KS cell lines. KSHV, or HHV-8, DNA sequences were detected in early passages of these tumor cells which were nearly identical to the previously described herpes-like DNA sequences (17,19) . By co-culture with the human embryonal kidney epithelial cell line, 293 cells, cell-free viral lysate was isolated which induced a cytopathic effect on 293 cells. Characterization of the virus was performed, and propagation by serial passage was confirmed. BRIEF DESCRIPTION OF THE DRAWINGS A more complete appreciation of the invention and many of the attendant advantages thereof will be readily obtained as the same becomes better understood by reference to the following detailed description when considered in connection with the accompanying drawings, wherein:
Figure 1. Phase contrast and transmission electron microscopy of isolated KS cells. (Magnification: (A) xio,- (B) x20, inset x70,000) .
Figure 2. Detection of viral DNA sequences in 293 cell initially co-cultured with KS cells and subsequently serial passage of virus.
Figure 3. 293 cells from control (uninfected) or those exposed to cell-free viral lysates (infected) for the indicated number of days in vi tro before analysis by phase contrast microscopy. (Magnification: xlO) .
Figure 4. Presence of HHV-8 and absence of other herpesvirus sequences in viral isolates.
Figure 5. Light and electron microscopic analysis of 293 cells before (panel A) and 5 days after (panels B anc C) exposure to lysate. Electron microscopic analysis of 293 cells 2 days after infection (D) , with inset showing a nuclear virus- like particle. Arrows indicate multinucleated giant cells or show an intact virion within the nucleus of an infected 293 cell. (Magnification: A, x 20, B, x 20, C x 20, D x 29,200, D, inset x 62,800) . DESCRIPTION CF THE PREFERRED EMBODIMENTS Cultivation cf Kaposi's Sarcoma Cells that Contain HHV-8 Virus in a Nonreplicative State
The method of cultivating KS cells according to the present invention comprises the steps cf : isolating a '^S ceil from a KS lesion cr tissue taken from a patient biopsy; and culturing said KS cell on a media coated with endothelial cell attachment factor.
KS ceils can be isolated from tissue taken from a patient biopsy or, preferably, from KS lesions of the skin from HIV- positive patients. These cells can be plated on tissue culture dishes coated with microvascular endothelial cell attachment factor and naintained in RPMI media supplemented with L-glutamine, heparin, appropriate antibiotics (preferably penicillin, streptomycin and gentamicin) , serum (preferably heat inactivated fetal bovine serum) and growth factors (preferably endothelial cell growth supplement) . Suitable serum concentrations are 0-20%. Cells can suitably be grown at a temperature range of from 35 to 40°C (preferably 37-38°C) in a C02 incubator (preferably in an atmosphere containing 2-5% CO.) . Cells are allowed to reach confluency. KS cells can be removed from confluent tissue cultures using conventional procedures such as exposure to trypsin and EDTA.
The present method differs from conventional methods which did not incorporate endothelial cell attachment factor or endothelial cell growth factor (See Nakamura et al, Science 112.:430-433 (1988) ) . Intact Human- Herpesvirus 8 ΗHV-3)
HHV-8 can be isolated from KS cells. Alternatively, HHV- 9 can be isolated from co-cultures of KS ceils and cells of epithelial or umbilical endothelial origin. Suitable epiteiial and umbilical endothelial cells are disclosed below. Any ratio of KS cells to cells of renal epithelial or umbilical endothelial origin can be used. Preferably 1:10 KS cells to cells of renal epithelial or umbilical endothelial origin can be used. Cells are co-plated onto media, optionally in the presence of growth factors such as TNF-a and allowed to reach confluency.
DNA can be isolated from the above tissue culture cells as previously described (28) . Briefly, the cells are washed in phosphate-buffered saline and gently lysed at room temperature. The solution in then centrifuged and the resulting supernatant treated with an enzyme such as pronase. The DNA is then extracted, precipitated, washed and dried. Isolated DNA is then amplified.
The DNA is then ligated into a suitable expression vector such as lambda gtlO or gt 11, Bluescript or SuperCosl (Stratagene) .
293 cells infected with HHV-8 have been deposited in accordance with the Budapest Treaty with the American Type Culture Collection, 12301 Parklawn Drive, Rockville MD 20852 on January 5, 1996 under accession no. .
Novel Methods for Propagating Human Herpesvirus
Human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells can be propagated in cells of epithelial or umbilical endothelial origin. Unlike previous methods, the method of the present invention propagates cells infected with HHV-Θ in the absence of HTLV-II conditioned media. The method of the present invention comprises: contacting a culture of epithelial cells or umbilical endothelial cord cells with cells or cell lysate of a human herpesvirus derived from Kaposi's sarcoma cell to form an infected ceil culture, incubating the infected cell culture for at least 1 day in the presence of endothelial cell attachment factor, and passaging the infected cell culture.
Suitable epithelial cells can be derived from adult or, preferably embryonic sources. Suitable epithelial and umbilical endothelial cells -can be derived from primates. or, preferably, human sources. Preferred epithelial cells are renal epithelial cells. For example, 293 cells, which are human embryonal kidney epithelial cells can be used. 293 cells are available from the American Type Culture Collection. Embryonic epithelial or umbilical endothelial cells can be infected with human herpesvirus using known methods 3 to 24 hours after seeding. Suitable inoculum includes virally infected KS cells or viral lysates. The concentration of. inoculum is preferably at least 10"' .
The infected cell culture can be incubated at 35-60βC in relevant media in a humidified atmosphere and appropriate 0-. and CO, concentrations.
The virus can be serial passages using methods known in the art. Briefly, cells infected with the virus are lysed and cell lysates are contacted with uninfected cells . Passaging is conducted after at least three days of infection. Infected cells are characterized by the presence of multinucleated giant cells, nuclear molding and dissolution of chromatin with residual chromatin marginating along the nuclear membrane. Within 5 days after infection, virtually all of the cells undergo necrosis with condensation of chromatin and other morphological changes consistent with apoptosis.
Alternatively, HHV-8 can be propated less efficiently using methods known in the art.
Vaccines derived from Human herpesvirus
Virus can suitably be harvested from cell culture when the maximal cytopathic effect is observed. Observation of cytopathic effect in cell cultures can be endpoint determined up to 5 days; this can suitably be done by visual evaluation of morphological changes on the monolayer or by fluorescent antibody staining techniques.
Suitable harvest methods include: agitation, aspiration, scraping, or freeze thawing accompanied by aspiration. Preferably, harvested viruses are stored at -70°C.
The activity of the virus can be suitably measured quantitatively by preparing dilutions of the sample and determining the highest dilution (endpoint) at which activity- is still detectable. The preferred method is the Reed Muench method which permits interpretation of the 50% endpoint from data derived from a quantal response. The formula can be applied similarly to rates of infection in any host system. The unit cf infectivity used c express the results are mean embryo infective dose, EIDs:, and mean tissue culture infective dose, TCID,;.
Vaccines can be produced from the harvested virus and viral lysates using techniques known m the art.
Diagnostic Methods for Detecting Kaposi's Sarcoma Cells
Antibodies to the intact human herpesvirus can be generated using methods known in the art (see for example, chapter 18 of Sambrook et al, "Molecular Cloning: A Laboratory Manual", 2nd ed. , Cold Spring Harbor Laboratory Press, NY, 1989) .
Antibodies, preferably polyclonal, against the intact virus can be used in Western blot tests to determine the presence or absence of human herpesvirus shed from Kaposi's sarcoma cells in a biological sample such as blood, semen or urine. Western blot analysis can be conduct using methods well known in the art .
Methods for Screening for Antiviral Drugs
Cells infected with human herpesvirus derived from Kaposi's sarcoma cells can be used to screen for antiviral activity. The method of the present invention comprises, contacting an antiviral drug candidate with a newly infected cell (within 2 to 24 hours following infecting) , culturing the infected cell and determining the effect of the antiviral drug candidate .
- 1 The effect cf the antiviral drug can be determined by comparing cytopathicity of. ntreated infected cells with treated infected cells. If treated infected cells persist for at least 7 days, preferably at least 10 days, the drug has antiviral activity against human herpesvirus derived from Kaposi's sarcoma cells.
Alternatively, screening can be performed using Elisa, RIA or other methods of immune detection known in the art to detect decreased production of specific viral gene products in the presence of the antiviral drug. These tests could screen directly for antigens in immunoprecipitate. Alternatively, the immune response cf the patient to an antigen can be screened using a western blot assay.
EJ£ £j-S_ Having generally described this invention, a further understanding can be obtained by reference to certain specific examples which are provided herein for purposes of illustration only and are not intended tc be limiting unless otherwise specified.
Cell Cu ture
Human KS cells were isolated independently from KS lesions of the skin from five different HIV-positive patients (designated KS#l-8) through mechanical disruption and enzymatic digestion of biopsies as previously described (29) . In contrast to previous reports which used an HTLV-II conditioned media, we have developed alternative media and culture conditions for these experiments. The ceils were plated cn tissue culture dishes (Corning, Corning, NY) coated with microvascular endothelial cell attachment factor (Cell Systems, Kirkland, 'WA) and were maintained in RPMI 1640 and 20% heat inactivated fetal bovine serum (FBS) supplemented with 10% utridoma HU (Boehringer Mannheim Biochemicals, Indianapolis, IN) , 2 mM L-glutamine, 100 units/ml penicillin, 100 mg/ml streptomycin, 50 mg/ml gentamicin, 50 mg/ml endothelial cell growth supplement (ECGS, ICN Biochemicals, Aurora, OH) and 16 units/ml bovine heparin. Isolated KS tumor cells were characterized by immunohistochemistry as stated in the text (29,30) . Human umbilical vein endothelial cells (HUVECs) were isolated by collagenase treatment of freshly obtained human umbilical cords and plated on gelatin-coated tissue culture dishes (31) . RPMI-1640 and 20% heat inactivated FBS supplemented with 2 mM L-glutamine, 100 units/ml penicillin, 100 mg/ml streptomycin, 50 mg/ml gentamicin, 50 mg/ml ECGS (Collaborative Research, Bedford, MA) and 16 units/ml bovine heparin was used to maintain the cells. HUVECs were characterized by a cobblestone appearance and specific staining for von Willebrand factor. The transformed human embryonal kidney cell line, 293, was provided by Dr. Garry Nolan, and the cells maintained in Dulbecco's modified Eagle's media containing 10% heat inactivated FBS, 2 mM L-glutamine, 100 units/ml penicillin, and 100 mg/ml streptomycin.
Electron microscopy
KS cells were removed from confluent tissue culture dishes using brief exposure to 0.03% trypsin and 0.01% ethylenediaminetetraacetic acid ΞDTA) . The ceils were pelleted and resuspended in 50% giuteraldehyde and C . IM cacodylate buffer. Alternatively, the confluent monolayers of cells were fixed with giuteraldehyde and cacodylate buffer and removed from the plate by gently scrapping with a rubber policeman. The fixed cells were embedded, sectioned and stained for electron microscopy by the departmental electron microscopy core facility.
KS tumor cells in confluent areas demonstrated loss of contact inhibition by forming large spherules on the dish (Figure IA) adjacent to the epithelial appearing microvascular endctheliai ceils and, on subsequent passage, only the spindle- shaped cell continued to proliferate (Figure IB) . It should be noted that despite several years and much experience in growing KS cells using previously published methodology, no such prolific population of tumor cells has previously been seen. A representative herpes-like virus particle detected in the cytoplasm of passage 2 KS cells is shown (Figure IB, inset) .
DNA isolation and preparation of Hirt supernatants
DNA was isolated from tissue culture cells as previously described (28) . For isolation of DNA from paraffin-embedded sections, the tissue from 50 five-micron sections was dewaxed twice with xylene, washed twice with ethanol and dried before addition of the digestion buffer. To selectively isolate low molecular weight DNA, Hirt supernatants were prepared as previously described (32,33) . Briefly, the cells were washed five times in phosphate-buffered saline (pH 7.0) and gently lysed for 15 minutes at room temperature in lysing solution 'O.DIM Tris ?H 3.:) , 3.6% SDS, :.01M ΞDTA) . Sodium chloride was added for a final concentration of IM and the mixture was incubated at 4°C overnight. The solution was centrifuged at 17,000 xg for one hour at 4°C, and the resulting supernatant treated with 1 mg/ml pronase at 37°C for 3-4 hours. The DMA was extracted three times with equal volumes of buffer saturated phenol and precipitated with 2 volumes of isopropanol and 1/10 volume of 3M sodium acetate at -20°C. The precipitated DNA was washed successively with cold ethanol at 70%, 90% and 100%. The DNA was dried and resuspended in TE buffer.
Polymerase chain reaction
Isolated DNA was amplified by PCR using primers designed from the specific herpesvirus-like DNA sequence (17) . The primers were 5' -AGC CGA AAG GAT TCC ACC ATT GTG CTC-3' (SEQ ID NO.:l) and 5' -TCC GTG TTG TCT ACG TCC AGA CGA TAT-3' (SEQ ID NO.:2) . PCR reaction mixtures (50 ml volume) were set up containing 0. lmg and lmg of isolated DNA, 50 pmol of each primer, 200 mM of each deoxynucleotide triphosphate, .5 mM magnesium chloride, 50 mM KCl, 20 mM Tris-HCl (pH 8.4) and 2.5 units of Taq DNA polymerase. DNA was amplified as follows. 94°C for 3 minutes (1 cycle), 94°C for 30 seconds, 72°C for 2 minute (40 cycles) , 72°C for 10 minutes (1 cycle) . PCR products were electrophoresed through a 2% agarose gel containing ethidium bromide. Successful PCR amplification with the herpesvirus-like DNA primers results in a single band at approximately 233 base pairs which is positioned at base pairs 987-1219 in the sequence published by Chang et al (17) . Positive control DNA was isolated from a biopsy of pulmonary KS. PCR for human herpesvirus 6 (HHV-6) , EBV, herpes simplex virus 1 and 2 (HSV 1 and 2) was performed using previously described primers and amplification conditions '34-36) . CMV was detected using the following primers derived from the intron of the immediate early -1 gene under standard conditions: (sense) 5' CCA AGC TTC CAC GCT GTT TTG ACC TCC ATA GA 3' (SEQ ID NO. : 3) ; (antisense) 5' CCA AGC TTC TGT CAG CTA TTA TGT CTG GTG GC 3' (SEQ ID N0.:4), generating a 908 bp product. Positive controls for these viruses were obtained from paraffin embedded tissue samples or from the EBV transformed B-cell line, M16B. For critical samples, the DNA was isolated and PCR reactions were performed independently by two investigators (KEF and JF) in separate laboratories.
DNA cloning and sequencing
DNA from isolated KS cells was amplified using PCR as described. 5 ml of the PCR product was electrophoresed through a 2% agarose gel containing ethidium bromide to demonstrate successful amplification. The PCR product was then ligated into a pCRII Vector according to the manufacturer's instructions (TA Cloning Kit, Invitrogen, San Diego, CA) .. Sequencing was performed using dideoxy (Sanger) sequencing and the Sequenase kit (United States Biochemical, Cleveland, OH) . Sequencing of PCR products was performed at least two times to confirm the results. Isolation cf -Virus From Co-Cultures
Early passage KS cells were plated in 6-well dishes coated with microvascular endothelial cell attachment factor and allowed to grow 50 - 70% confluence. Primary KS cells 2xl05 cells, passage 2-3) were removed from primary culture and incubated with 293 cells (2x10*) , in 35 mm 6-well Costar plates in the presence or absence of TNF-a (200 IU/ml) . Cell lysates were prepared by three cycles of freeze-thawing after 3-5 days and incubated at different dilutions with 293 ceils in the absence of TNF-a. For serial propagation, half of the cell culture was used to prepare lysates at day 3 post-infection, while the remainder of the cell culture was maintained to confirm subsequent CPE. PCR positivity was confirmed in the cell lysate. Virus was propagated serially for at least 5 passages. Estimation of titer was made by incubation of lysates at progressive dilutions (10*--10"10) with 293 or other target cells.
Viral DNA was detected by PCR in Hirt supernatants of 293 cells at the indicated times after infection (Figure 2A) and with DNA isolated from the indicated enriched subcellular fractions (Figure 2B) . The arrow denotes the specific 233 bp DNA fragment. (+) represents a positive control sample from KS lesion, (-) represents uninfected 293 cell DNA.
Figure 3 is a phase contrast microscopy (Magnification: xlO) of a cell.
In Figure 4, cell-free virus lysates were derived by serial challenge of 293 cells and analyzed for the presence of the indicated herpesvirus sequences by PCR. Positive control samples (+) , (lanes 1,4,7,10,13), negative (-) uninfected 293 ceil DNA controls flanes 2,5,3,1 ,1-1) , and infected 233 cell DNA from ceil-free lysate (L) preparations 'lanes 3,6,9,12,15) were included. Positive control samples for HSV-1 and -2 (92 bp fragment; and KHV-6 223 bp fragment) were derived from tissue samples of patients with documented infections. The HHV-3 positive control INA was derived from paraffin sections of pulmonary KS in an HIV-7 patient. EBV positive control DNA was isolated from M16B cells, chronically infected with EBV (115 bp fragment) . CMV positive controls (908 bp fragment) were derived from a control plasmid, VCL 1012, which contained this regulatory element (37) .
Figure 5 is Light and electron microscopic analysis of infected 293 cells. Representative light microscopic appearance of 293 cells before (panel A) and 5 days after (panel B) exposure to lysate. Electron microscopic analysis of 293 cells 2 days after infection (C) . Note that compared to uninfected cells which display uniformly viable cells with typical epithelial differentiation, the infected cells are undergoing degeneration with formation of multinucleated giant cells and chromatin alteration typical cf herpes group viral infection.
RESULTS Culture and characterization of isolated KS cells
Cell culture conditions were developed which did not require the presence of HTLV-II conditioned media as used in previous studies. Under these conditions, KS cell lines grew rapidly after initiation of culture. Initially, macroscopic clusters of cells were observed which appear to contain both epithelioid-appearmg endothelial cells and spindle-shaped mesenchymal cells ^Fig. IA) . Subsequently, after the first passage, spindle-shaped cells characteristic of KS were observed, whose doubling times ranged from 24-48 hours (Fig. IB) . Each KS spindle cell line was positive for factor Xllla and VCAM-1 by immunohistochemical staining, and negative for factor XIIIs, E-selectin, PECAM-1 (CD31), factor VIII, and CD34. Expression of these markers was not altered by I?N-g or TNF-a stimulation. These results are consistent with previously published data on the phenotype of isolated KS cells using different culture conditions (9,29).
Detection of herpesvirus-like DNA in KS cells
PCR was used to determine whether the proliferating KS cells contained recently described herpesvirus-like DNA sequences (17) . 75% (6 of 8 isolates) of early passage, isolated KS tumor cells were positive for this DNA sequence using PCR (Table 1) .
. aoi e
Probe
Sample KS herpesvirus HHV •6 HSV 1 &2 EBV -A EBV-B CMV
Isolated KS cells
KS- l Negative Negative Negative Negative Negative Negative
KS-2 Positive Negative Negativ e Negative Negative Negative
KS-3 Positive Negative Negative Negative Negative Negative
KS-4 Positive Negative Negative Negative Negative Negative
KS-5 Posim e Negative Negative Negative Negative Negative
KS-6 Positive Negative Negative Negative Negative Negative
KS-7 Negative Negative Negative Negative Negative Negative
KS-8 Positive ND~ ND ND ND Negative
Patient Samples
Pulmonary KS A Posime Positive Negative Negative Negative Negative
Pulmonary KS B Positive Positive Negative Negative Negative Negative
Psoπasis Negative Negative ND ND ND Negative
Controls
HUVECs Negative Negative Negative Negative Negative Negative
293 Negative Negative Negative Negative Negative Negative
DNA was isolated from the indicated cell lines, and tissue samples were analyzed by PCR. Positive or negative indicates the presence or absence of relevant size fragment by ethidium bromide staining on agarose gels. Positive control reactions for each virus type were performed using an equal concentration (1 mg) of DNA as in the test reactions.
*ND indicates not determined.
The signal was present in the cell lines until passage 2 or 3. and was then no longer detectable. The 233 bp PCR product was also found in DNA isolated from two separate paraffin embedded tissue samples obtained from a patient with pulmonary KS, as previously described in other patients (17,18). No evidence of KS-associated herpesvirus DNA sequences was detected in samples from isolated HUVECs, 293 cells, or DNA isolated from paraffin-embedded tissue from a psoriasis patient (Table 1). To determine if other herpesviruses might be present in these samples. PCR analysis was performed to detect HHV-6. HSV- 1 and -2. CMV. EBV. DNA derived from these isolated KS cells were negative for each of these viral sequences (Table 1). Analysis of the 233 bp PCR product revealed a sequence identical to the herpesvirus-li e DNA sequence originally
reported by Chang and colleagues (17), with the exception of a single bp change (C to T) at codon 47, a substitution reported subsequently in seven KS patients ( 19).
Isolation of lvtic virus
KS cells were co-cultured with a variety of other cell types for two to seven days. Cell-
free lysates were prepared and tested for the presence of viral DNA by PCR.
Consistently strong PCR signals were found on day two in DNA isolated from co-cultures
with 293 cells while DNA from co-cultures with other cell types were largely negative
(Table 2A). Table 2A
Sample Description PCR Positivitv
YPE swine ECs
YSM swine smooth muscle cells
HSM human smooth muscle cells
HUVEC human umbilical vein ECs +/-
HDMEC human microvascular ECs
H9 human T-leukerrua
CEM human T-leukerrua
Jurkat human T-leukemia T-4 human T-leukemia
HL-60 human myelomonocytic leukemia
U937 human promonocytic leukemia
HELA human cervical carcinoma
293 human embryonal kidney cell
CaSki HPV-16 positive epithelial cell
M16B human B-cell infected with EBV
In addition, the PCR signal in 293 cells was detected as the KS cells were lost from the culture (days 4-5). These findings suggested that the positive PCR signals were likely due to viral replication rather than persistence of DNA sequences from the primary KS cell line. In addition, greater cytopathicity was detected when KS and 293 co-cultures were incubated with TNF-a (Table 2B; see also Fig. 3). Table 2B
B. Cell Activation
Cell Stimulus PCR Positivitv CPE
293 none - -
IL- 1 IL-6
TNF-a + + + + -
PMA LPS
HUVEC none +•/- +/-
IL-1 IL-6 TNF-a PMA LPS
PCR analysis of the indicated cell lines atter co-cuiture with KS ceil lines (KS-3 - KS-6) for KS herpesvirus (A). Determination of PCR posttivity and cytopathic effect (CPE) vvas performed with the indicated stimulus in 293 cells and HUVEC (B). For PCR, + + J- denotes consistent and readily visible 233 bp product by ethidium bromide staining; - indicates weaker positivity; - - indicates inconsistent detection. For CPE, + + + indicates > 80% nonviable cells by day 5, 4- by day 10-1 1. + /- inconsistent effects.
The cytopathic effect (CPE) was manifested by > 80% cell death, with the presence of multi-nucleated giant cells, prominent chromatin condensation, and nuclear molding noted in the culture. This CPE was not observed after incubation of 293 with TNF-a in the absence of KS cells (data not shown). Finally, although a PCR signal had been observed on occasion in HUVEC, no consistent cytopathic effect was observed in these cells. We therefore focused our efforts on 293 cells as a host cell for viral replication.
Propagation of virus bv serial passage
To propagate the virus, cell-free lysates were prepared at different times after infection and incubated with 293 cells in the presence or absence of TNF-a. Although TNF-a enhanced viral replication during the primary KS/293 cell co-culture, it did not enhance CPE or viral titers during serial passage on 293 cells and was not used further. No PCR signal was detected in uninfected cells, but a signal was readily detected in infected 293 cells within 1 day after infection. PCR signals of viral DNA were maximal three days after viral challenge, and diminished on day five ( Fig. 2A), when > 80 of cells showed severe cytopathic effects (Fig. 3), possibly due to nonspecific degradation. The signal was detected more readily in the nuclear fraction, with a weaker signal present in the cytoplasm (Fig. 2B). Hirt supernatants of low molecular weight DNA from the nucleus were also found to contain the Kaposi's herpesvirus sequence by PCR (Fig. 2A). No PCR signal was detected from other known herpesviruses in serially passaged viral isolates, despite their ready detection in positive control tissues or cell lines infected with the relevant viruses (Fig. 4). To serially passage the virus, cell lysates were prepared on day 3-4, prior to the generation of CPE.- The resultant cell lysates (day 3) conferred CPE and a positive PCR signal on uninfected 293 cells and was maintained consistently with titers 310<7ml for at least 5 serial passages.
Virus filtration, heat, inactivation. and detection. of viral DNA bv endpoint dilution analysis
Cell-free lysates from the primary TNF-a stimulated co-cultures were prepared and incubated with fresh 293 cells in the absence of other cell types. Transfer of this cytopathic activity and detection of PCR signal after serial passage was maintained after filtration with a 0.4 mM filter, but was lost after heat inactivation of viral lysates (Table
3). Table 3
CPE day post treatment
Treatment 1 -ι _j 4 5 cell free viral lysates - - _
~" "*~ cell free viral lysates: heat activated - - - - - cell free viral lysates: filtered" - - + + + -"- + -T determination of cytopathic erfect iCPE) in 293 cells was performed after exposure to lysate subjected to the indicated treatments, and seventy was graded as follows- - . -r .
+ + , + + +
293 cells were incubated w ith increasing dilutions of viral lysates. and cells were examined for CPE and viral sequences by PCR. As noted previously, CPE was observed at a lysate dilution of 10° but not 10"* Infection of 293 cells and the development of CPE also correlated with the appearance of a detectable PCR signal for HHV-8 Viral DNA was also detectable by Southern blot hybridization during propagation. Quantitation of the Southern blot signal suggested a relatively low copy number of virus per cell (1-10 copier per genome).
Light microscopic analysis
To characterize the effects of viral infection further and document the presence of the virus after serial propagation, additional microscopic analyses were performed. Light microscopic analysis of the cytopathic effect was performed in one micron-thick plastic embedded toluidine-blue stained sections of 293 cells. Uninfected 293 ceils revealed viable appearing epithelial cells with round nuclei containing evenly distributed chromatin and prominent nucleoli. In contrast, after exposure to the infected cell lysate. 293 cells, beginning as an early as day 2 demonstrated significant cytopathic changes. Infected 293 cells were characterized by the presence ot multinuclea.ed giant cells, nuclear molding, and dissolution of chromatin with residual chromatin marginating along the nuclear membrane ( Figure 5. A vs B.O By day 5. virtually all of the cells were undergoing necrosis with condensation of chromatin and other morphological changes consistent with apoptosis (Figures 3 and 5 B.C)
Electron microscopy of KS cells and infected 293 cells
Initially, cultured KS cells were analyzed using transmission electron microscopy to visualize cells in greater detail and to detect the potential presence of virus. In these early passage cells, evidence of particles was found in the cytoplasm whose size and shape was consistent with that of a herpes virion (Fig. IB, inset); however, particles were infrequent, and the moφhology was not sufficient to identify the particle definitively as a herpesvirus. Additional studies were undertaken to characterize the virus particle propagated on 293 cells. Cells were examined daily after exposure to cell-free viral lysates. Virus panicles were observed in the nucleus of infected cells as early as two days after infection.
REFERENCES
1. Beral, V., Peterman, T.A., Berkelman, R.L., and Jaffe, H.W. Kaposi's sarcoma among persons with AIDS: a sexually transmitted infection? Lancet, 335: 123-128, 1990.
2. Safai, B., Johnson, K.G., Myskowski, P.L., Koziner, B. , Yang, S.Y., Cunningham-Rundles, S., Godbold, J.H.. and Dupont. B. The natural history of Kaposi's sarcoma in the acquired immunodeficiency syndrome. Ann. Intern. Med. 103: 744-750, 1985.
3. Beral, V , Jaffe, H. , and Weiss, R. Cancer surveys: cancer. HIV, and AIDS. Eur. J. Cancer. 27: 1057-1058, 1991. 4 Stem. M . Spencer. D.. Kuten. A. , and Bezwoda. W AIDS-related Kaposi's sarcoma: a review Isr. J. Med. Sci. 30: 298-305. 1994
5. Finesmith. T.H. and Shrum. J.P. Kaposf s sarcoma. Int. J. Dermatol 33. 755- 762. 1994
6. Ioachim. H.L.. Adsay, V . Giancotti. F.R.. Dorsett. B . and Melamed. J Kaposi's sarcoma of internal organs Cancer. 75: 1376-1385. 1995.
7. Nickoloff. B.J., Griffiths. C.E.M., Gallo, R.C, Salahuddin. S.Z., and Nakamura, S. Factor Xllla-expressing dermal dendrocytes in AIDS-associated cutaneous Kaposi's sarcomas. Science, 243: 1736-1737, 1989.
8. Rutgers. J.F. , Wieczorek, R.. Bonetti. F. , Kaplan, K.E., Posenett, D.N. , Friedman-Kien, A.E. , and Knowles, D.M. The expression of endothelial cell surface antigen by AIDS-associated Kaposi's sarcoma: evidence for a vascular endothelial cell origin. Am. J. Pathol. 122: 493-499, 1986.
9. Naidu, Y.M.. Rosen. E.M.. Zitnick. R.. Goldberg, I.. Park. M.. Naujokas. M.. Polveπni. P.J. , and Nickoloff, B.J. Role of scatter factor in the pathogenesis of AIDS- related Kaposi's sarcoma. Proc. Natl. Acad. Sci. USA, 91 : 5281-5285, 1994.
10. Browning, P.J., Sechler, J.M., Kaplan, M. , Washington, R.H., Gendelman, R.. Yarchoan, R., Ensoli, B., and Gallo, R.C. Identification and culture of Kaposi's sarcoma-like spindle cells from the peripheral blood of human immunodeficiency virus- 1- infected individuals and normal controls. Blood, 84: 2711-2720, 1994.
11. Samaniego, F. , Markham, P.D. , Gallo, R.C, and Ensoli, B. Inflammatory cytokines induce AIDS-Kaposi's sarcoma-derived spindle cells to produce and release basic fibroblast growth factor and enhance Kaposi's sarcoma-like lesion formation in nude mice. J. Immunol. 154: 3582-3592, 1995.
12. Ensoli, B.. Baπllari, G. , and Gallo. R.C. Cytokines and growth factors in the pathogenesis of AIDS-associated Kaposi's sarcoma. Immunol. Rev. 127: 147-155, 1992.
13. Barillari, G. , Buonaguro, L., Fiorelli, V. , Hoffman. J., Michaels, F. , Gallo, R.C, and Ensoli. B. Effects of cytokines from activated immune cells on vascular cell growth and HIV-l gene expression. J. Immunol. 149: 3727-3734, 1992.
14. Ensoli, B., Gendelman, R., Markham, P., Fiorelli, V. , Colombini, S. , Raffeld, M., Cafaro, A., Chang, H.K., Brady, J.N., and Gallo, R.C. Synergy between basic fibroblast growth factor and HIV-l Tat protein in induction of Kaposi's sarcoma. Nature, 371 : 674-680, 1994.
15. Nair. B.C., DeVico. A.L., Nakamura, S.. Copeland. T.D., Chen. Y. , Patel. A., O'Neil, T. , Oroszlan, S. , Gallo, R.C, and Samgadharan, M.G. Identification of a major growth factor for AIDS-Kaposi's sarcoma cells as oncostatin M. Science, 255: 1430- 1432, 1992. 16. Berai. V- Epidemiology of Kaposi's sarcoma. Cancer Surv 10: 5-22. 1991.
17. Chang, Y. , Cesarman. E.. Pessin, M.S , Lee, F. , Culpepper. J.. Knowles, D.M . and Moore, P S . Identification of heφesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma. Science. 266 1865-1869, 1994
18. Huang, Y.Q. , Li. J.J.. Kaplan. M.H.. Poiesz. B.. Katabira, E. , Zhang, W C, Feinder. D . and Friedman-Kien. A.E. Human heφesvirus-like nucleic acid in various forms of Kaposi's sarcoma. Lancet, 345: 759-761. 1995.
19. Moore, P.S. and Chang, Y. Detection of heφes-like virus DNA sequences in Kaposi's sarcoma in patients with and without HIV infection. N. Eng J. Med. 332: 1 181- 1185, 1995.
20. Ambroziak, J.A. , Blackburn, D.J., Herndier. B.G., Glogau, R.G.. Gullet. J.H.. MacDonald, A.R.. Lennette. E.T., and Levy, J.A. Heφes-llike sequences in HIV- infected and uninfected Kaposi's sarcoma patients. Science, 268: 582-583, 1995.
21. Cesarman. E. , Chang, Y . Moore. P.S.. Said, J.W , and Knowles, D.W Kaposi's sarcoma-associated heφesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas. N. Eng. J. Med. 332: 1186-1191, 1995.
22. Cesarman, E. , Moore, P.S., Raα, P.H., Inghirami, G., Knowles, D.M., and Chang, Y. In vitro establishment and characterization of two acquired immunodeficiency syndrome-related lymphoma cell lines (BC-1 and BC-2) containing Kaposi's sarcoma- associated heφesvirus-like (KSHV) DNA sequences. Blood, 86: 2708-2714, 1995.
23. Lebbe, C, de Cremoux, P., Rybojad, M., Costa da Cunha, C, Morel, P. , and Calvo, F. Kaposi's sarcoma and new heφesvirus. Lancet, 345: 11801995.
24. Dupm, N. , Grandadam. M. , Calvez, V. , .Goπn, I. , Aubin, J.T. , Havard, S., Lamy, F., Leibowitch, M.. Huraux, J.M. , Escande, J.P. , and Agut, H. Heφesvirus-like DNA sequences in patients with Mediterranean Kaposi's sarcoma. Lancet. 345: 761-762. 1995.
25. Whitby, D. , Howard, M.R., Tenant-Flowers, M. , Brink, N.S., Copas. A., Boshoff, C , Hatzioannou, T., Suggett, F.E., Aldam, D.M. , Denton, A.S., Miller; R.F. , Weller, I.V., Weiss, R.A., Tedder, R.S., and Schulz, T.F. Detection of Kaposi sarcoma associated heφesvirus in peripheral blood of HlV-infected individuals and progression to Kaposi's sarcoma. Lancet. 346: 799-802, 1995.
26. Levy, J.A. A new human heφesvirus: KSHV or HHV8? Lancet, 346: 7861995.
27. Cohen, J. Controversy: is KS really caused by new herpesvirus? Science, 268: 1847-1848, 1995. 28. Coen. D M. Quantitation of rare DNAs by PCR. In: F.M. Ausubel. R. Brent. R.E. Kingston. D.D. Moore. J.G. Seidman. J.A. Smith and K. Struhl (eds. ). Current Protocols in Molecular Biology, pp." 15.3.1-15.3.8, New York: Greene Publishing Associates and Wiley-Interscience. 1992.
29. Huang, Y Q. , Friedman-Kien. A.E.. Li. J.J.. and Nickoloff. B J. Cultured Kaposi's sarcoma cell lines express Factor Xllla. CD14. and VCAM-1. but not Factor VIII or ELAM-1 Arch. Dermatol. 129: 1291-1296. 1993
30. Yang, J.. Xu. Y. , Zhu. C, Hagan. M.K. , Lawiey, T. , and Offermaπn. M.K. Regulation of adhesion molecule expression in Kaposi's sarcoma cells. J. Immunol. 152: 361-373 , 1994
31. Jaffe, E.A. Culture and identification of large vessel endothelial cells. In: E.A. Jaffe (ed.), Biology of Endothelial Cells, pp. 1-3, the Hague: Martinus Nijoff. 1984.
32. Eizuru, Y. , Inagawa, S., and Minamishima, Y. Application of "Hirt supernatant" DNA to the molecular epidemiology of cytomegalovirus infections. J. Clin. Microbiol. 20: 1012-1014. 1984
33. Pater, M.M. , Hyman, R.W. , and Rapp, F. Isolation of heφes simplex virus DNA from the "Hirt supernatant" . Virol. 75: 481-483, 1976.
34. Sa'Adu, A., Thomson, B.J., Bountiff, L., and Webster, A.D. Lymphotropic viruses in "common variable" immunodeficiency-PCR analysis of lymphocyte DNA for HIV-l and HHV-6. Clin. Exp. Immunol. 91 : 50-53, 1993.
35. Cao, M., Xiao, X., Egbert, B., Darragh, T.M. , and Yen, T.S. Rapid detection of cutaneous heφes simplex virus infection with the polymerase chain reaction. J. Invest. Dermatol. 92: 391-392, 1989.
36. Apolloni, A. and Sculley, T.B. Detection of A-type and B-type Epstein-Barr virus in throat washings and lymphocytes. Virol. 202: 978-981 , 1994.
37. Manthoφe, M., Cornefert-Jensen, F., Hartikka, J., Feigner, J. , Rundell, A., Margalith, M., and Dwarki, V. Gene therapy by intramuscular injection of plasmid DNA: studies on firefly luciferase gene expression in mice. Hum. Gene Ther. 4: 419- 431, 1993.
Having now fully described the invention, it will be apparent to one of ordinary skill in the art that many changes and modifications can be made thereto without departing from the spirit or scope of the invention as set forth herein.

Claims

CLAIM:
1 A method of cultivating KS cells according to the present invention comprises
the steps ol isolating a KS cell from a KS lesion or from tissue taken from a patient biopsy,
and culturing said KS cell on a medium coated with endothelial cell attachment factor
2. The method of Claim 1. wherein said KS cell is taken from a KS lesion derived from a patient with AIDS
3. The method of Claim 1. wherein said medium is RPMI medium supplemented with antibiotics, serum and growth factors.
4 An isolated intact human heφesvirus 8 (HHV-8)
5. An isolated intact human heφesvirus 8 (HHV-8) obtained by the steps of. contacting a culture of epithelial cells or umbilical endothelial cord cells with a cell or cell lysate of HHV-8 to form an infected cell culture, incubating the infected cell culture for at least 1 day, and isolating a HHV-8 virus from said infect cell culture.
6. A method for propagating a cell infected with human heφesvirus 8 (HHV-8) comprising: contacting a culture of epithelial cells or umbilical endothelial cord cells with a cell or cell lysate of a HHV-8 to form an infected cell culture, incubating the infected cell culture for at least 1 day, and passaging the infected cell culture.
7 The method of Claim 6, wherem said epithelial cell is a embryonic epithelial cell.
8. The method of Claim 7 wherein said embryonic epithelial cell is a human embryonic kidney epithelial cell.
9 The method of Claim 7. wherem said human embryonic kidney epithelial cell is a 293 cell.
10. The method of Claim 6. wherein the infected cell culture is incubated for at least 3 days prior to passaging.
11. A method of propagating primary KS cells infected with human heφesvirus 8 (HHV-8) comprising: contacting a culture of epithelial cells or umbilical endothelial cord cells with a cell or cell lysate of a HHV-8 to form an infected cell culture, incubating the infected cell culture for at least 1 day, and passaging the infected cell culture.
12. A vaccine comprising intact human heφesvirus 8 (HHV-8) and a pharmaceutically acceptable carrier or adjuvant.
13. A method of detecting the presence of human heφesvirus 8 (HHV-8) in a biological fluid comprising: contacting said biological fluid from said patient with an antibody specific HHV-8. and detecting the presence of complexes of antibody and human heφesvirus.
14. The method of Claim 13, wherein said biological fluid is blood, semen or urine.
15 A method of detecting the presence of antibodies selective for human heφesvirus 8 (HHV-8) in a biological fluid comprising: contacting said biological fluid from said patient with HHV-8 v iral antigen so as to form complexes between said viral antigen and any antibodies selective for HHV-8. detecting the presence of said complexes of antibody and viral antigen
16. A method of screening for antiviral activity against human heφesvirus 8 (HHV-8), comprising: cultivating a cell line infected with HHV-8 for at least 1 day, contacting said infected cell culture with an antiviral drug candidate to form an exposed cell culture, culturing the exposed cell in the presence of the antiviral drug candidate, and determining the effect of the antiviral drug candidate.
PCT/US1997/000019 1996-01-16 1997-01-16 Isolation and propagation of a human herpesvirus derived from aids-associated kaposi's sarcoma cells WO1997026373A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/585,588 US6114110A (en) 1996-01-16 1996-01-16 Isolation and propagation of a human herpesvirus derived from AIDS-associated Kaposi's sarcoma cells
US08/585,588 1996-01-16

Publications (1)

Publication Number Publication Date
WO1997026373A1 true WO1997026373A1 (en) 1997-07-24

Family

ID=24342102

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/000019 WO1997026373A1 (en) 1996-01-16 1997-01-16 Isolation and propagation of a human herpesvirus derived from aids-associated kaposi's sarcoma cells

Country Status (2)

Country Link
US (1) US6114110A (en)
WO (1) WO1997026373A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999061909A2 (en) * 1998-05-26 1999-12-02 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods and compositions for the detection of human herpesvirus
CN113278580A (en) * 2021-04-02 2021-08-20 肇庆大华农生物药品有限公司 Grass carp skin tissue cell line and application thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5131907A (en) * 1986-04-04 1992-07-21 Thomas Jefferson University Method of treating a synthetic naturally occurring surface with a collagen laminate to support microvascular endothelial cell growth, and the surface itself

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5834599A (en) * 1987-05-29 1998-11-10 Tanox Biosystems, Inc. Immunoconjugates which neutralize HIV-1 infection
US5830759A (en) * 1994-08-18 1998-11-03 The Trustees Of Columbia University In The City Of New York Unique associated Kaposi's sarcoma virus sequences and uses thereof
US5801042A (en) * 1994-08-18 1998-09-01 The Trustees Of Columbia University In The City Of New York Unique associated Kaposi's sarcoma virus sequences and uses thereof
WO1996015779A1 (en) * 1994-11-21 1996-05-30 The Trustees Of Columbia University In The City Of New York Unique associated kaposi's sarcoma virus sequences and uses thereof
US5723125A (en) * 1995-12-28 1998-03-03 Tanox Biosystems, Inc. Hybrid with interferon-alpha and an immunoglobulin Fc linked through a non-immunogenic peptide
US5882644A (en) * 1996-03-22 1999-03-16 Protein Design Labs, Inc. Monoclonal antibodies specific for the platelet derived growth factor β receptor and methods of use thereof
US5853734A (en) * 1996-11-13 1998-12-29 The Trustees Of Columbia University In The City Of New York Glycoprotein L and clycoprotein M from kaposi's sarcoma associated herpesvirus, DNA encoding same and uses thereof
US5863787A (en) * 1996-07-25 1999-01-26 The Trustees Of Columbia University In The City Of New York Kaposi's sarcoma-associated herpesvirus (KSHV) glycoprotein B (GB) and uses thereof
US5861500A (en) * 1996-07-25 1999-01-19 The Trustees Of Columbia University In The City Of New York Kaposi's sarcoma-associated herpesvirus (KSHV) interleukin 6 (IL-6) and uses thereof
US5859225A (en) * 1996-09-05 1999-01-12 The Trustees Of Columbia University In The City Of New York Virion protein 26 from Kaposi's sarcoma-associated herpesvirus, DNA encoding same and uses thereof
US5854398A (en) * 1996-07-25 1998-12-29 The Trustees Of Columbia University In The City Of New York Kaposi's sarcoma-associated herpesvirus (KSHV) interleukin 6 (IL-6) and uses thereof
US5831064A (en) * 1996-07-25 1998-11-03 The Trustees Of Columbia University In The City Of New York Kaposi's sarcoma-associated herpes virus (KSHV) interferon consensus sequence binding protein (ICSBP) and uses thereof
US5854418A (en) * 1996-07-25 1998-12-29 The Trustees Of Columbia University In The City Of New York Kaposi's sarcoma-associated herpesvirus (KSHV) viral macrophage inflammatory protein-1α II (vMIP-1α II) and uses thereof
US5849564A (en) * 1996-11-29 1998-12-15 The Trustees Of Columbia University In The City Of New York Polypeptides from Kaposi's sarcoma-associated herpesvirus, DNA encoding same and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5131907A (en) * 1986-04-04 1992-07-21 Thomas Jefferson University Method of treating a synthetic naturally occurring surface with a collagen laminate to support microvascular endothelial cell growth, and the surface itself

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BLOOD, 01 October 1995, Vol. 86, No. 7, CASARMAN et al., "In Vitro Establishment and Characterization of Two Acquired immunodeficiency Syndrome-Related Lymphoma Cell Lines (BC-1 abd BC-2) Containing Kaposi's Sarcoma-Associated Herpesvirus-Like (KSHV) DNA Sequences", pages 2708-2714. *
NATURE MEDICINE, December 1995, Vol. 1, No. 12, BOSHOFF et al., "Kaposi's Sarcoma-Associated Herpesvirus Infects Endothelial and Spindle Cells", pages 1274-1278. *
SCIENCE, 16 December 1994, Vol. 266, CHANG et al., "Identification of Herpesvirus-Like DNA Sequences in AIDS-Associated Kaposi's Sarcoma", pages 1865-1869. *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999061909A2 (en) * 1998-05-26 1999-12-02 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods and compositions for the detection of human herpesvirus
WO1999061909A3 (en) * 1998-05-26 2000-03-23 Us Health Methods and compositions for the detection of human herpesvirus
CN113278580A (en) * 2021-04-02 2021-08-20 肇庆大华农生物药品有限公司 Grass carp skin tissue cell line and application thereof

Also Published As

Publication number Publication date
US6114110A (en) 2000-09-05

Similar Documents

Publication Publication Date Title
Monaco et al. JC virus infection of hematopoietic progenitor cells, primary B lymphocytes, and tonsillar stromal cells: implications for viral latency
Kojaoghlanian et al. The impact of adenovirus infection on the immunocompromised host
Söderberg-Nauclér et al. Interferon-gamma and tumor necrosis factor-alpha specifically induce formation of cytomegalovirus-permissive monocyte-derived macrophages that are refractory to the antiviral activity of these cytokines.
Vieira et al. Use of the red fluorescent protein as a marker of Kaposi's sarcoma-associated herpesvirus lytic gene expression
Minton et al. Human cytomegalovirus infection of the monocyte/macrophage lineage in bone marrow
Ablashi et al. Utilization of human hematopoietic cell lines for the propagation and characterization of HBLV (human herpesvirus 6)
Benditt et al. Viruses in the etiology of atherosclerosis.
Vieira et al. Transmissible Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) in saliva of men with a history of Kaposi's sarcoma
Campadelli-Fiume et al. Human herpesvirus 6: An emerging pathogen.
Flamand et al. Activation of the Epstein-Barr virus replicative cycle by human herpesvirus 6
Luppi et al. The new lymphotropic herpesviruses (HHV-6, HHV-7, HHV-8) and hepatitis C virus (HCV) in human lymphoproliferative diseases: an overview
Wu et al. Chronic infection of human umbilical vein endothelial cells by human herpesvirus-6.
Barcy et al. γδ+ T cells involvement in viral immune control of chronic human herpesvirus 8 infection
Okano Haematological associations of Epstein–Barr virus infection
Black et al. Biologic properties of human herpesvirus 7 strain SB
Heieren et al. Cytomegalovirus replicates efficiently in human kidney mesangial cells.
Tedeschi et al. Laboratory diagnosis of human herpesvirus 8 infection in humans
Yamanishi Human herpesvirus 6
US6114110A (en) Isolation and propagation of a human herpesvirus derived from AIDS-associated Kaposi&#39;s sarcoma cells
Westmoreland et al. Comparative pathobiology of Kaposi sarcoma-associated herpesvirus and related primate rhadinoviruses
JP2002513274A (en) Unique sequence of Kaposi&#39;s sarcoma-associated virus and its use
Stöckl et al. Potential of in situ hybridization for early diagnosis of productive cytomegalovirus infection
Purushothaman et al. Human DNA tumor viruses and oncogenesis
Ron et al. Spontaneous curing of a minute virus of mice carrier state by selection of cells with an intracellular block of viral replication
Salahuddin et al. Human herpes virus-6 (HHV-6) in diseases

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97523141

Format of ref document f/p: F

NENP Non-entry into the national phase

Ref country code: CA

122 Ep: pct application non-entry in european phase