WO1997026329A1 - Monoclonal antibody to herpes simplex virus and cell line producing same - Google Patents

Monoclonal antibody to herpes simplex virus and cell line producing same Download PDF

Info

Publication number
WO1997026329A1
WO1997026329A1 PCT/US1996/000641 US9600641W WO9726329A1 WO 1997026329 A1 WO1997026329 A1 WO 1997026329A1 US 9600641 W US9600641 W US 9600641W WO 9726329 A1 WO9726329 A1 WO 9726329A1
Authority
WO
WIPO (PCT)
Prior art keywords
hsv
antibody
cell line
cell
human
Prior art date
Application number
PCT/US1996/000641
Other languages
French (fr)
Inventor
Elisabeth Harfeldt
Philip Lake
Barbara Nottage
Lars G. Ostberg
Original Assignee
Sandoz Pharmaceuticals Corporation
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 Sandoz Pharmaceuticals Corporation filed Critical Sandoz Pharmaceuticals Corporation
Priority to EP96902712A priority Critical patent/EP0876478A4/en
Priority to AU47016/96A priority patent/AU4701696A/en
Publication of WO1997026329A1 publication Critical patent/WO1997026329A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/081Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from DNA viruses
    • C07K16/085Herpetoviridae, e.g. pseudorabies virus, Epstein-Barr virus
    • C07K16/087Herpes simplex virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/734Complement-dependent cytotoxicity [CDC]

Definitions

  • This invention relates to monoclonal antibodies specific for the herpes simplex virus, to cell lines which produce these antibodies, methods of producing the cell lines, and methods of using the antibodies in therapy.
  • HSV-l and HSV-2 are common human pathogens which generally produce mild and self- limiting diseases such as recurrent oral lesions, e .g. , "cold sores" or more severe recurrent genital lesions.
  • HSV viruses may also produce still more serious and even life-threatening infections. The most frequent of these serious diseases are: (1) ocular infections, which, if untreated, may lead to blindness; and
  • glycoproteins which are antigenic have been identified on the virion envelope and include those designated gB, gC, gD, gE, gG, gl, gH, gJ, gK, gL and gM. These glycoproteins appear on both HSV-l and HSV-2 with varying degrees of structural and antigenic variability. Monoclonal antibodies, many of them murine, have been made which are specific to various of these glycoproteins. See, for example, Balachandran et al . , 1982, Infection and Immunity vol. 37 (3) .1132-1137; Rector et al . , 1982, Infection and Immunity vol. 38 (1):168-174; and Dix et al .
  • This invention provides human monoclonal antibodies which are able to bind to the glycoprotein gD antigen of HSV-l and HSV-2 and are able to neutralize HSV-l and HSV-2 with substantially equivalent potency.
  • This invention also provides for a virus-free cell line which produces human monoclonal antibodies which are able to bind to the glycoprotein D antigen of HSV-l and HSV-2.
  • the cell line is a xenogeneic hybridoma, i.e., a cell which results from the fusion of cells from different species.
  • a further aspect of this invention is a cell supernatant which comprises a human monoclonal antibody (Mab) able to bind to the glycoprotein gD antigen of HSV-l and HSV-2 and is also able to neutralize HSV-l and HSV-2 with substantially equivalent potency, the supernatant being substantially free from any other antibody.
  • a human monoclonal antibody Mob
  • Another aspect of this invention is use of the aforementioned antibodies in vivo to prevent or lessen the severity of HSV-l and/or HSV-2 symptoms by administering to a mammal in need of such treatment a symptom-lessening or symptom-preventing amount of a human monoclonal antibody which is able to bind to the glycoprotein gD antigen and neutralize the virus.
  • the hybridomas of this invention can be made by fusing an immortalized cell to another cell which is chosen for its ability to produce antibodies to the desired antigen. See, e .g. , K ⁇ hler and Milstein, 1975, Nature 256:495-497.
  • K ⁇ hler and Milstein 1975, Nature 256:495-497.
  • problems in developing hybridomas which stably produce human monoclonal antibodies using the K ⁇ hler-Milstein technique It was discovered by ⁇ stberg that xenogeneic triomas, made by using a xenogeneic hybridoma fusion partner which has lost its ability to produce immunoglobulins as the immortalizing cell, are more stable and produce antibodies efficiently. This procedure is described in detail in U.S. Patent 4,634,664 and in Hybridoma , 1983, vol.
  • the hybridoma is a xenogeneic trio a which produces human monoclonal antibodies against the glycoprotein gD antigen of HSV-l and HSV-2.
  • a parent rodent immortalizing cell such as a murine myeloma cell, e .g. , SP-2
  • immortalizing xenogeneic hybridoma cells which, when fused to cells capable of producing an antibody against Herpes Simplex virus 1 and 2 (HSV-l and 2) , provide a novel cell lines capable of generating human antibody effective against such virus in the human.
  • SPAZ 4 a xenogeneic hybridoma referred to as SPAZ 4, prepared from cell line SP-2 obtainable, e . g. , from the NIGMS Human Genetic Mutant Cell Repository Ref. CM35669A (see U.S. DHHS 1982 Catalog of Cell Lines) .
  • SP-2 cell line is drug resistant and is fused with normal human peripheral lymphocytes by conventional techniques. A large number of hybrids is obtained and, after approximately five weeks, five clones are selected which show fast growth and no antibody production. These cells are selected for resistance to 8- azaguanine and with three of these lines it is possible to obtain mutants which are resistant to 20 ⁇ g/mL of 8- azaguanine. These cells are at the same time sensitive to
  • hypoxanthine-Aminopterin-Thymidine (HAT) medium which showed that they had lost their ability to produce hypoxanthine phosphoribosyl transferase.
  • SPAZ 4 Hypoxanthine-Aminopterin-Thymidine
  • Two methodologies were used to make the monoclonal antibodies of this invention. Both use the cell line SPAZ 4.
  • Cell line SPAZ 4 may be fused with spleen cells obtained from an individual whose spleen is sensitized with HSV-l or HSV-2 antigen. It was found in the first methodology that both hybridoma-like cells and lymphoblastoid cells (spontaneously Epstein-Barr Virus (EBV) transformed) were obtained after such fusion. Contrary to what was hoped, cloning of the hybridoma- like cells failed to generate the fused cells desired.
  • EBV Epstein-Barr Virus
  • a lymphoblastoid cell is re ⁇ fused to SPAZ 4 and resulting cell line having hybridoma-like morphology, designated SB IV-6-2 produces antibodies which neutralize HSV-l and HSV-2 in vitro , in some cases at concentrations as low as 50 ng/mL.
  • SB IV-6-2 when tested in mice injected with HSV-2 is effective in protecting such mice at a dosage of 100 ⁇ g/animal (approximately 4 mg/kg) .
  • HSV 863 also known as 64-683
  • the HSV 863 cell line is extremely stable over time; it has been in culture over 12 months.
  • One of the surprising traits of this cell line is that it can produce a high yield of antibody.
  • FIGURES Figure 1 Sequences of the HSV 863 antibody light chain (A) and heavy chain (B) variable region cDNAs (SEQ. ID. Nos. 3 and 1) and the translated amino acid sequences (1-letter code) (SEQ. ID. Nos. 4 and 2) .
  • the first amino acid of the mature light chain and of the mature heavy chain is double underlined, and the three complementarity determining regions (CDRs) in each chain are underlined.
  • the preferred monoclonal antibody of this invention is produced by cell line HSV 863 and is also designated HSV 863. It has been surprisingly found that the potency of HSV 863 substantially exceeds that of any other known anti-HSV preparation.
  • Monoclonal antibody HSV 863 belongs to the IgGl class. It has lambda light chains. It was also found to bind to Staphylococcus aureus protein A (SpA) . This property is particularly advantageous because binding to SpA provides an efficient, specific and economical method to purify antibodies using a single step technique.
  • Mab HSV 863 was shown to be specific for HSV viruses. It reacted with 15 independent isolates of HSV-l and HSV-2, but no cross-reactivity was observed with uninfected Vero cells or with varicella-zoster virus. Further and surprisingly, HSV 863 was found to neutralize both HSV-l and HSV-2 substantially equally, and with a surprising potency. It is believed that HSV 863 is the most potent HSV neutralizing monoclonal antibody of any origin to date. The monoclonal antibodies of this invention are particularly well suited for clinical and diagnostic use, but especially for clinical use.
  • HSV 863 The preferred monoclonal antibody of this invention, HSV 863, has been shown to protect mice from both HSV-l and HSV-2 infection.
  • the antibodies can be administered prior to infection or even after infection.
  • HSV-l the antibody was strongly protective up to 24 hours after infection and was protective, albeit to a lesser degree, for 48 hours thereafter.
  • HSV 863, and other preferred antibodies of the invention are fully human antibodies, which comprises natural combinations of heavy and light chains. These antibodies differ, for example, from many of the antibodies produced from phage combinatorial libraries of heavy and light chains, in which the individual heavy and light chains may not be naturally associated.
  • Antibody HSV 863 was also shown to be effective in lessening the symptoms of HSV cutaneous disease, when administered after infection.
  • an important aspect of this invention is the use of the antibodies of the present invention for both their prophylactic and therapeutic benefits in mammals, and especially in humans.
  • the HSV diseases particularly targeted include primary or reactivation disease in immunocompromised patients (e .g. , bone marrow and other transplanted patients, cancer therapy patients, and AIDS patients). For these patients, the probability of HSV disease often exceeds 50% and the HSV infection can be severe and even life-threatening.
  • acyclovir (9-[ (2-hydroxyethety1-methyl] guanine) , may be used as prophylaxis in some circumstances, but not in all cases.
  • transplacentally-derived antibody affects the outcome of infection after human neonatal exposure to HSV.
  • the antibody appears to completely neutralize virus in some infants and prolongs the incubation period and modifies the infection in others (Yeager et al . , Infection and Immunity, 29:532-538, 1980; Sullender et al. J. Inf . Dis . 155:28-37, 1987).
  • An estimate of the dose of commercial immune globulin which would be required to achieve prophylaxis of newborn infants at risk concluded that at least 100 mL (approximately 5 g) of immune globulin would be needed.
  • the benefits include but are not limited to decreasing latent viral infection and symptoms and sequelae of HSV disease; extending, the incubation time of infection which would create an extended time for clinicians to definitively diagnose the HSV infection; and providing a more slowly evolving illness in which other therapies (e .g. , acyclovir) are more effective.
  • acyclovir e.g. , acyclovir
  • Using the monoclonal antibodies of the present invention either as a replacement for, or administered along with acyclovir, is especially valuable for patients with impaired renal function, those with ocular or CNS disease, and those having or at risk to develop acyclovir- resistant variants.
  • acyclovir-resistant virus strains See, e .g. , Schinazi et ai.
  • the antibodies of the present invention are particularly useful in providing pre- or post-exposure prophylactic cover to neonates at risk in acquiring HSV disease since morbidity and sequelae are severe in this patient group despite currently available therapies. Animal studies have shown that neonates can be protected even when antibody administration is delayed 24 hours or more after infection. Since HSV disease evolves more slowly in man, including infants, the opportunity to administer beneficial antibody is extended for a considerably longer time. Administration of the antibody decreases the attack rate (proportion of patients who become infected relative to those at risk) and reduces the severity and rate of progression and dissemination of disease.
  • the antibodies of this invention are also useful in the treatment of ocular HSV infections (250,000-500,000 per year in the U.S.A.). Again, in primary disease, the antibodies can ameliorate symptoms (in possible synergy with acyclovir) and interfere with establishment of latency. This would offer great value since a large proportion of patients later reactivate latent ocular HSV infections and some progress to blindness.
  • the antibodies of this invention are thus suitable for parenteral therapy for prophylaxis of HSV in a variety of patients and for many clinical presentations of HSV disease. They are especially suited in instances of low index of suspicion, where drug therapy may be contraindicated or undesirable, since they have no mechanistic toxicity and do not possess any inherent toxicity or side effects.
  • Antibody HSV 863 is the most potent neutralizing antibody yet to be reported and shows an extremely broad reactivity with all HSV-l and HSV-2 isolates. As an IgGl antibody, it can mediate immunological effects via several potential mechanisms: steric interference upon binding to virus, complement activation through classical or alternate routes, inhibition of cell to cell spread of virus, opsonization mediated by Fc receptors on mononuclear and polymorphonuclear phagocytes and antibody-dependent cellular cytotoxicity (ADCC) . Mab HSV 863 can thus independently or in collaboration with serum complement and cellular elements, direct immune effector responses to free virions or to viral antigens expressed on newly infected cells.
  • ADCC antibody-dependent cellular cytotoxicity
  • HSV 863 will be highly effective as a sole treatment.
  • Another aspect of this invention is the antibody fragments, e .g. , Fabs which retain the ability to bind to the HSV antigen.
  • the antibodies of the invention can be used to manufacture pharmaceutical compositions.
  • the antibodies and pharmaceutical compositions containing the antibodies are useful for parenteral administration, i.e., subcutaneously, intramuscularly or intravenously.
  • the antibodies and pharmaceutical compositions of the invention can also be administered, typically for local application, by gavage or lavage, intraperitoneal injection, ophthalmic ointment, topical ointment, intravaginal , intranasal or intrabursal injection.
  • compositions for parenteral administration commonly comprise a solution of the immunoglobulin or a cocktail thereof dissolved in an acceptable carrier, preferably an aqueous carrier.
  • an acceptable carrier preferably an aqueous carrier.
  • aqueous carriers can be used, e . g. , water, buffered water, phosphate buffered saline (PBS), 0.4% saline, 0.3% giycine, human albumin solution and the like. These solutions are sterile and generally free of particulate matter.
  • the compositions can contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example sodium citrate, sodium acetate, sodium chloride, potassium chloride, calcium chloride and sodium lactate.
  • concentration of antibody in these formulations can vary widely, i .e . , from less than about 0.005%, usually at least about 1% to as much as 15 or 20% by weight and is selected primarily based on fluid volumes and vis
  • a typical pharmaceutical composition for injection contains 1 L sterile buffered water, and 1-70 mg of immunoglobulin.
  • a typical composition for intravenous in- fusion contains 250 mL of sterile Ringer's solution, and 150 mg of antibody.
  • Actual methods for preparing parenterally administrable compositions are described in more detail in, for example. Remington ' s Pharmaceutical Science (15th ed. , Mack Publishing company, Easton, Pennsylvania, 1980) , which is incorporated herein by reference in its entirety for all purposes. Compositions suitable for lavage or other routes are selected according to the particular use intended. The compositions containing the present antibodies or a cocktail thereof can be administered for prophylactic and/or therapeutic treatments.
  • compositions are administered to a patient already infected with HSV-l or -2, in an amount sufficient to cure or at least partially arrest the disease and its complications.
  • An amount adequate to accomplish this is defined as a "therapeutically effective dose.”
  • Amounts effective for this use depend upon the severity of the disease and the general state of the patient's own immune system, but generally range from about 1 to about 200 mg of antibody per dose, with dosages of from 5 to 70 mg per patient being more commonly used.
  • Dosing schedules vary with the disease state and status of the patient, and range from a single bolus dosage or continuous infusion to multiple administrations per day (e .g. , every 4-6 hours) , or as indicated by the treating physician and the patient's condition.
  • compositions containing the present antibodies or a cocktail thereof are administered to a patient not already infected with HSV-l and/or HSV-2, who is at risk of such infection (e.g., the sexual partner or fetus of an infected person) to enhance the patient's resistance.
  • a patient not already infected with HSV-l and/or HSV-2 who is at risk of such infection (e.g., the sexual partner or fetus of an infected person) to enhance the patient's resistance.
  • Such an amount is defined to be a "prophylactically effective dose.”
  • the precise amounts again depend upon the patient's state of health and general level of immunity, but generally range from 1 to 70 mg per dose.
  • Single or multiple administrations of the compositions can be carried out with dose levels and pattern being selected by the treating physician.
  • the invention is further illustrated by reference to the following, non-limiting examples.
  • Example 1 Production of Cell Line SB-IV-6-2 Spleen cells from spleen donor SB IV were sensitized with a dilution of soluble HSV-2 antigen. Approximately 3xl0 8 cells were cultivated in 150 milliliters of RPMI-1640 and 5% human serum. After days of culture, 3xl0 7 viable cells were recovered and fused to a similar number of SPAZ-4 cells (passage 58) . When the supematants of the cultures were tested (after three weeks) , some cultures were positive in an ELISA test to detect anti-HSV antibodies. One of these cultures was designated SB IV-6-2, and it was consistently positive in ELISA tests both on HSV-2 as well as on HSV-l. It also showed neutralizing capacity against HSV-2 virus.
  • the culture contained both hybridoma-like cells and lymphoblastoid cells and the supernatant contains antibodies of both the IgG and IgM class.
  • the antibodies also had light chains of both kappa and lambda types.
  • the selection procedure was the standard HAT- treatment but with an addition of 2xlO ⁇ 7 M ouabain to suppress the growth of unfused lymphoblastoid cells. This fusion yielded several cultures with antibodies against HSV and with cells of hybridoma-like morphology. The best growing of these cell lines, which was still designated. SB IV-6-2 (deposited subject to the Budapest Treaty with the ATCC, Rockville, MD on February 3, 1987, and designated HB 9316), has been cloned and expanded and been found to produce an IgG, kappa-light chain antibody, that binds to the antigens of both HSV-l and HSV-2 and neutralizes both viruses in vitro. The neutralizing capacity was found to be substantially stronger against HSV-2 and was detected down to concentrations of 50 ng/mL.
  • the antigen containing cells were harvested, washed in phosphate buffered saline (PBS) , diluted in PBS (one 180 cm 2 flask per 10 mL) then sonicated, heat- inactivated 56 ⁇ C, 30 min., and then frozen at -80°C until used.
  • PBS phosphate buffered saline
  • SPAZ-4 mouse x mouse hybridoma.
  • SPAZ-4 cells are known in the art and can be produced according to methods described in US 4,634,664, which is hereby incorporated by reference.
  • the SBIII x SPAZ-4 fused cells were grown in Dulbecco's modified minimal essential medium containing supplementary pyruvate, oxaloacetic acid, non-essential amino acids, 20% fetal calf serum (from a selected batch) , hypoxanthine, a inopterin, and thymidine according to standard protocols.
  • HSV 863 cultures A high concentration of HSV 863 cells (IO 7 per mL) was cultured directly into serum-free media DMEM/F12 without any supplements. While most cells die, the surviving fraction grew well at high densities in serum free medium and produced high levels of antibody. Cells were subcultured at approximately 14 day intervals by feeding with 50% volume additions on days 7 and 14.
  • HSV 863 cells proliferate and produce antibody in stationary cultures, but these cells will die in 3-5 days if placed in rotating bottles. If, however, the cells are cultures in 1% serum, rotation does not adversely affect cells.
  • Antibody can be harvested from "exhausted" supematants by culturing cells at 37 ⁇ C and allowing the cultures to grow without further addition of medium. After 5- 10 days, the cell death is complete and the supernatant is clarified by centrifugation. Serologic tests using ELISA methods and a standard of pure HSV 863 antibody showed that cells grown in DMEM with 5% fetal calf serum (FCS) produced exhausted supematants containing approximately 300 ⁇ g per mL monoclonal antibody and cells grown in DMEM with 0% FCS produced in excess of 60 ⁇ g per mL. This represents one of the highest ranges reported for monoclonal antibody production, regardless of origin. The genetic stability of antibody production by HSV
  • the immunoglobulin class of antibody HSV 863 was determined using ELISA methods. The antibody was captured on antigen-coated plates and the assay was developed with subclass specific, peroxidase-conjugated anti-human lg (Tago) . Antibody HSV 863 was clearly identified as an IgGl antibody.
  • HSV 863 was tested with anti K or ⁇ light chain reagents (Tago) .
  • the .antibody was clearly identified as having ⁇ light chains.
  • HSV 863 was captured on antigen, and the ELISA was developed with peroxidase- conjugated SpA. A strong stain identified HSV 863 as an SpA- binding immunoglobulin.
  • HSV 863 behaved as a highly basic protein.
  • the antibody was used to immunoprecipitate proteins from biosynthetically labelled cells, mock-infected or infected with HSV-l or HSV-2.
  • the culture media were adjusted to supply 14 C-glucosamine or 35 S-methionine to the infected cells.
  • the cells were dissolved in detergent buffer and the radiolabelled solutions were exposed to antibody HSV 863 or to irrelevant antibody 53-2-4.
  • the complexes of antigen plus antibody were captured on insoluble beads containing SpA and were dissolved, denatured and reduced in a sample buffer prior to PAGE analysis and autoradiography.
  • Antibody HSV 863 precipitated an antigen of HSV-l of 55 kD and an antigen of HSV-2 of 50-55 kD.
  • the HSV antigen was glycosylated since 14 C-glucosamine was incorporated into the molecule. Control, uninfected Vero cells, and antibody 53-2-4 do not display this prominent antigen.
  • the molecules precipitated by HSV 863 were identified as HSV-l gD using methods described in Eisenberg et al . , J. Virol . 36:428-435 (1980), which is hereby incorporated by reference.
  • As anti-gD Mab can be divided into groups on the basis of numerous biological and biochemical characteristics (see Eisenberg et al . , J. Virol .
  • HSV 863 binds to the gD glycoprotein, which is a highly conserved type-common epitope essential for infectivity of the virus.
  • HSV 863 recognizes isolates of HSV-l and HSV-2 which are present in the human population.
  • antibody HSV 863 was tested for its ability to react with HSV viruses of independent origin by ELISA assay. In addition, it was tested for reactivity with antigens of uninfected cells and antigens of cells infected with the distantly related varicella-zoster virus (VZV) . Results show that HSV 863 reacted with 15 independent isolates of HSV-l and 15 independent isolates of HSV-2 but not at all with Vero or VZV antigens. Thus, the antigen recognized by HSV 863 was shared by all herpes simplex virus isolates tested to date, but was not expressed by uninfected cells or cells infected by a different virus.
  • Antibody HSV 863 was purified and tested in a virus neutralization assay using HSV-l and HSV-2.
  • the microneutral- ization assay was designed so that 10 to 20 plaque forming units (p.f.u.) were present in each well in the presence of the antibody. After incubation for 30 min. at 37°C, the virus was transferred to susceptible Vero cells and incubated for 48 hours to allow plaques to become evident. Results showed that high concentrations of HSV 863 totally abolished the appearance of plaques. Doubling dilutions of antibody revealed a 50% inhibition of p.f.u. at an antibody concentration of about 0.05-0.35 ⁇ g/mL depending on the strain, with an average of 0.2 ⁇ g/mL, which defines the IC 50 .
  • HSV 863 To determine the ability of HSV 863 to inhibit HSV-induced cell-cell fusion, the mAb was tested in fusion assays as follows.
  • the syncytial HSV-l variant HFEMsyn ('113) was added to freshly confluent Vero cell monolayers in 24-well plates at an MOI of 20 (200 ⁇ L/well in DMEM-2% FCS) .
  • MOI 200 ⁇ L/well in DMEM-2% FCS
  • HSV 863 was found to be highly effective at preventing syncytium formation with an approximate IC 50 of 10 ⁇ g/mL and some efficacy still evident at concentrations as low as 2.5 ⁇ g/mL.
  • concentration required to inhibit syncytium formation is greater than that required to neutralize free virus.
  • HSV 863 is effective at inhibiting the intercellular spread of syncytial and non-syncytial HSV strains, albeit at higher concentration than necessary to neutralize virus or mediate ADCC activity in vitro.
  • HSV 863 Since ADCC is implicated as one of the pivotal mechanisms of antibody-mediated defense in HSV infection, HSV 863 was tested for this activity in vitro using a 51 Cr-release microcytotoxicity assay. There was a marked dose-dependent increase in ADCC-mediated specific release of 51 Cr in the presence of HSV-863 as compared to the irrelevant mAb control or the medium-alone control. Maximum ADCC efficacy was achieved with concentrations of HSV 863 as low as l ⁇ g/mL with HSV-l infected target cells and 5 ⁇ g/mL with HSV-2 infected target cells. These results, therefore, demonstrate that even at low concentrations, HSV 863 is highly effective at mediating ADCC destruction of both HSV-l and HSV-2 infected cells.
  • HSV 863 human immunoglobulin-1 (Mclntyre) and HSV-2 (clinical isolate 1811) .
  • Virus samples ( ⁇ 10 7 pfu/mL) were incubated for 30 min at 37 ⁇ C with an equal volume of HSV 863 (final concentration of 1 mg/mL) in DMEM-2% FCS.
  • the virus-antibody mixture was then added (200 ⁇ L/well) to freshly confluent Vero cell monolayers in 24-well plates. The monolayers were incubated for 48 h at 37°C, fixed, stained, and checked for plaques which would not bind/stain with HSV 863 in the presence of peroxidase-conjugated goat antibody specific for the heavy chain of human IgG.
  • HSV 863 To generate or select for neutralization-resistant variants to HSV 863, greater numbers of virus PFU, cultured in the presence of low concentrations HSV 863, were repeatedly passaged over several weeks. Following neutralization with range of HSV 863 concentrations, a minimum IO 8 PFU of HSV-l (Mclntyre) and 10 7 PFU of HSV-2 (clinical isolate 1811) were passaged 16 and 15 times respectively. For each passage, the virus samples (a minimum of 10 7 HSV-l pfu/well or IO 6 HSV-2 pfu/well) were mixed in duplicate with doubling dilutions of HSV 863 (20- 0.6 ⁇ g/mL .
  • HSV 863 Even after 16 passages ( ⁇ 10 8 pfu/passage) in HSV 863, there were still no detectable MAR variants in the HSV-l (Mclntyre) population. However, following 13 passages (> IO 7 pfu/passage) in HSV 863, the resultant HSV-2 (1811) population demonstrated an increased resistance to the mAb. The HSV-2 populations resulting from 13 to 15 passages in HSV 863 required 10 to 30-fold more HSV 863 to achieve complete neutralization as compared to the parental strain. These data indicate that increased resistance to HSV 863 neutralization is difficult to achieve in vitro.
  • HSV 863 Protection from HSV lethality in vivo
  • Mouse models of HSV-l and HSV-2 disease were developed to assess the ability of HSV 863 to protect young animals from HSV infection. Mice were chosen since only a small amount of antibody is required for therapeutic dose levels and since it is possible to test large number of animals in dose-response analyses. Virulent forms of HSV-l (strain Patton) and HSV-2 (333) were grown for use. Preliminary experiments established the appropriate doses for LD 50 in 3-week-old mice using HSV-l for i.p. inoculation and HSV-2 for footpad inoculation. Protection by HSV 863 was tested by dosing mice with 10xLD 50 and administering purified HSV 863 in phosphate buffered saline intraperitoneally.
  • Purified antibody retains full specific functional activit y : RSV-2 rpt gt ⁇ Q ⁇ in VJvo
  • groups of 15 mice were administered either purified or unpurified HSV 863 antibody 24 hours prior to inoculation with HSV-2 (l ⁇ xLD 50 ) in the footpad.
  • Antibody was administered at 0, 1, 10, 100 or 1000 ⁇ g per animal.
  • the percent survival after 21 days is given in Table I, below.
  • the purified antibody activity was indistinguishable from that of the crude supernatant, and the purification procedure which involved a brief exposure to pH 3.5 had no detectable effect on antibody specific activity.
  • Antibody HSV 863 wa ⁇ administered i.p., 24h prior to injection with IOXLD JO of HSV-2 strain 333 in the footpad. Values are shown for survival at 21 days. This result demonstrates that immunoglobulin purification does not reduce the biological efficacy of antibody HSV 863.
  • HSV 863 antibody efficacy of the HSV 863 antibody was shown in mice inoculated intranasally with HSV-2. Mice received a single 0.5 mL does i.p. of antibody (450 or 900 ⁇ g) after HSV-2 inoculation at the time indicated in the table. Protection was determined by measuring percentage mortality and mean day of death (MDD) in groups of 15 mice treated with HSV 863 antibody versus placebo. Also shown in the table are data for a neutralizing antibody (HuFd79D) specific for the gB protein of HSV-2. TABLE II
  • NS Not statistically significant when compared to the appropriate placebo-treated group.
  • Table II shows that HSV 863 treatment resulted in statistically significant reduction in mortality at both the 450 ⁇ g and 900 ⁇ g dosages and was effective up to 96 hr postinoculation at the higher dosage.
  • HuFdl38 which is specific to the gD glycoprotein and has a similar in vitro neutralizing power to the HSV 863 antibody. Mice received 0.5 mL i.p. antibody 24 h after HSV-2 infection.
  • Antibody HSV 863 was administered i.p., 24h prior to injection with 10xLD 50 neurovirulent HSV-l strain Patton intraperitoneally. Values are shown for survival at 14 days.
  • HSV 863 is protective whether administered prior to or subsequent to infection for both HSV-l and HSV-2 infections.
  • mice were treated with human immunoglobulin (SANDOGLOBULIN®) or with HSV 863.
  • the specific activities of the two immunoglobulin preparations were compared in Tables VI and VII below.
  • Antibody HSV 863 and Sandoglobulin® were administered i . p. , 24h prior to injection with IOXLD J - of HSV-l strain Patton intraperitoneally. Values are shown for survival at 14 days .
  • Antibody HSV 863 and Sandoglobulin® were administered i . p. , 24h prior 1 to injection with 10xLD M of HSV-2 strain 333 in the footpa . Values 1 are shown for survival at 21 days.
  • the results show that for HSV-l, human immunoglobulin is protective at 300 ⁇ g per animal while HSV 863 protects at a dose of 15 ⁇ g, (a potency factor of about 20 fold) .
  • human immunoglobulin is needed at about 15,000 ⁇ g per animal while HSV 863 is effective at 25 ⁇ g (a potency factor of approximately 600) .
  • HSV 863 is much more potent than gamma globulin in the prophylaxis models of HSV infection for both HSV-l and HSV-2.
  • the large difference in dose of human immunoglobulins effective for HSV-l vs. HSV-2 prophylaxis may be explained by the known difference in titers of antibodies to those respective viruses which is seen in the population (many fewer people are seropositive for HSV-2 than for HSV-l) .
  • HSV 863 Inhibits HSV Lesion Formation bv Virus Discharge from Neurons
  • HSV 863 A model of skin lesion formation by HSV discharged form sensory neurons was adapted to test the ability of HSV 863 to inhibit the process of viral lesion recrudescence.
  • Adult BALB/c mice were infected by scarification on the lateral thorax with HSV-l. After 5-7 days, severe band-like confluent lesions form unilaterally along the entire dermatome. This process is known to involve the centripetal ascent of virions to the dorsal root ganglia and centrifugal descent of virus within sensory afferents of the dermatome.
  • Administration of HSV 863 3 days after inoculation of virus substantially decreased the frequency and severity of lesion formation and prevents mortality as shown in Table IX, below.
  • Table IX Table IX
  • mice 15-20 animals per group, were treated with a single dose of 300 ⁇ g of HSV 863, or control antibody, 24 h before or after virus challenge by IO 4 p.f.u. of neurovirulent HSV-l into the right hind footpad. After 7-9 weeks the animals were sacrificed and their dorsal root ganglia removed. From each mouse 6 thoracicolu bar (T13 to L- 5) ganglia, each ipsilateral and contralateral to the site of HSV injection, were removed and individually explanted in tissue culture medium into microtiter plates. After 3 days in culture, 8x10* Vero cells were added to each well as an indicator cell.
  • An alternative method of digestion was to use one- tenth volume of 0.077M dithiothreitol (instead of mercaptoethanol) and 25 units of insoluble papain (papain attached to beaded agarose; one unit hydrolyzes 1.0 mole of N- ⁇ -benzoyl-L-arginineethyl ester (BAEE) per min at pH 7.0 at 30 ⁇ C) per 67 mg HSV 863 IgG.
  • the solution was rocked constantly at 37°C for 2 hr. Iodoacetamide was added as previously described except the concentration of the stock solution was only 0.16M.
  • This method can also be used with concentrated cell supernatant that has been dialyzed against the phosphate buffer mentioned above if the antibody protein from the fetal calf serum is approximately the same or lower in concentration than the HSV 863 antibody.
  • a method of isolating Fab fragments from the digestion mixture is to dialyze the digested antibody against phosphate-buffered saline and remove the Fc fragments by passage through a Protein A column equilibrated in phosphate- buffered saline.
  • trioma cell lines The genes encoding the heavy and light chains of antibodies secreted by trioma cell lines are cloned according to methods described in Sambrook et al., Molecular Cloning: A Laboratory Manual , (2nd ed. , Cold Spring Harbor, NY, 1989; Berger & Ki mel, Methods in Enzymology, Volume 152, Guide to Molecular Cloning Techniques , (Academic Press, San Diego, CA, 1987); Co et al. , J. Immunol . , 148:1149 (1992) (each of which is incorporated by reference in its entirety for all purposes) .
  • genes encoding heavy and light chains are cloned from a trioma's cDNA produced by reverse transcription of the trioma's mRNA.
  • Cloning is accomplished by conventional techniques including the use of PCR primers that hybridize to an enzymatically inserted sequence (e .g. , a G tail) or to the sequences flanking or overlapping the genes, or segments of genes, to be cloned.
  • the cDNA and predicted amino acid sequences of the heavy and light chain variable domains from the HSV 863 antibody are shown in Fig. 1. The provision of amino acid sequences for the
  • HSV 863 antibody can be exploited to provide alternate means for generating antibodies having the same or similar binding specificity. That is, antibodies that bind to the same epitope as the HSV 863 antibody or an epitope sufficiently proximal to that bound by the HSV 863 antibody to compete with the HSV 863 antibody for binding to the HSV-l and/or -2 glycoprotein. Such antibodies can, of course, also be produced using the same procedure as noted for the HSV 863 antibody, employing the HSV 863 antibody as a reference for selection of appropriate binding specificity. However, generation of variants from sequence data has the advantage of avoiding the need for immunization and immortalization.
  • Competition is determined by an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody (e.g., HSV 863) to an antigenic determinant on an HSV gD protein.
  • a reference antibody e.g., HSV 863
  • Numerous types of competitive binding assays are known for example: solid phase direct or indirect radioimmunoassay (RIA) , solid phase direct or indirect enzyme immunoassay (EIA) , sandwich competition assay and ELISA (see Stahli et al.. Methods in Enzymology 9:242-253 (1983)); Harlow and Lane, "Antibodies, A Laboratory Manual," Cold Spring Harbor Press (1988)). Usually the test immunoglobulin is present in excess.
  • a competing antibody when present in excess, it will inhibit specific binding of a reference antibody to gD protein by at least 10, 25, 50 or 75%.
  • Many of the amino acids in the disclosed HSV 863 sequences can undergo non-critical substitutions, additions or deletions without loss of binding specificity or effector functions, or intolerable reduction of binding affinity (i.e., below about 10 7 M "1 ) .
  • the binding affinity of most antibodies is within a range of about 10 7 M" 1 or IO 8 M "1 to about 10 9 M” 1 .
  • antibody chains incorporating such alterations exhibit substantial sequence identity to the reference antibody chain from which they were derived.
  • the mature light chain of antibodies derived from the HSV 863 antibody usually shows substantial sequence identity (i.e., at least 75, 85% or 95%) to the sequence of the mature light chain of the HSV 863 antibody shown in Fig. 1 (panel A) .
  • the mature heavy chains of derivatives typically show substantial sequence identity to the sequence of the mature heavy chain of the HSV 863 antibody shown in Fig. 1 (panel B) .
  • Sequence identity comparisons are performed between two polypeptide sequences when optimally aligned, such as by the programs BLAZE (Intelligenetics) GAP or BESTFIT using default gap weights.
  • antibodies comprise two identical pairs of heavy and light chains.
  • bifunctional antibodies can be produced that have one heavy/light chain pair specific for the gD antigen and the other heavy/light chain pair specific for a second antigen.
  • Conservative.substitutions as defined by, e.g. , Co, WO 94/12215 (incorporated by reference in its entirety for all purposes)
  • amino acids interacting closely with the CDR regions see Queen et al., WO 90/07861 (incorporated by reference in its entirety for all purposes) are likely to have little effect on the functional properties of antibodies, such as binding affinity, kinetics and specificity.
  • derivatives harboring such mutations are not preferred in comparison with the original HSV 863 antibody.
  • a mutated antibody is selected having the same specificity and increased affinity or faster binding kinetics compared with the HSV 863 antibody.
  • the affinity of the mutated antibody is within a factor of 2, 5, 10 or 50 of the HSV 863 antibody.
  • Phage-display technology offers powerful techniques for selecting such antibodies. See, e .g. , Dower et al., WO 91/17271; McCafferty et al., WO
  • variable segments of human antibodies produced as described supra are typically linked to at least a portion of an antibody constant region (Fc) , typically that of a human antibody.
  • Fc antibody constant region
  • Human constant region DNA sequences can be isolated in accordance with well-known procedures from a variety of human cells, but preferably immortalized B-cells (see Kabat et al. , Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD (1987) and (1991)), and WO 87/02671) (each of which is incorporated by reference in its entirety for all purposes) .
  • the antibody contains both light chain and heavy chain constant regions.
  • the heavy chain constant region usually includes CHI, hinge, CH2, CH3, and sometimes CH4 regions.
  • the present antibodies include antibodies having all types of constant regions, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgGl, IgG2, IgG3 and IgG4.
  • the constant domain is usually a complement-fixing constant domain and the class is typically IgGl, IgG3, IgM.
  • the constant domain may be of the IgG2 or IgG4 class.
  • the human antibody can comprise sequences from more than one class or isotype.
  • DNA segments encoding heavy or light chains of the antibodies described supra.
  • the DNA segments encode at least one CDR regions and usually all three CDR regions from the heavy or light chain of an antibody.
  • a DNA segment encodes all or substantially all of the variable region of a heavy or light chain and is thereby capable of exhibiting antigen binding capacity.
  • a nucleic acid encodes an entire heavy or light chain. Because of the degeneracy of the code, a variety of nucleic acid sequences will encode each immunoglobulin amino acid sequence.
  • the desired nucleic acid sequences can be produced by de novo solid-phase DNA synthesis or by PCR mutagenesis of an earlier prepared variant of the desired polynucleotide.
  • Preferred nucleic acids include those encoding mature light chain variable regions having substantial sequence identity to the mature light chain variable region of the HSV 863 antibody shown in Fig. 1 (Panel A) (SEQ. ID. No. 4) . Preferred nucleic acids also include those encoding mature heavy chain variable regions having substantial sequence identity to the mature heavy chain variable region of the HSV 863 antibody shown in Fig. 1 (panel B) (SEQ. ID. No. 2).
  • ATC CTG TAT CTG CAA ATG AAC AGC CTG AGA GCC GAG GAC ACG GCT GTG 336 lie Leu Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val 100 105 110
  • MOLECULE TYPE protein

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Virology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Human monoclonal antibodies effective for the diagnosis and treatment of Herpes Simplex Virus 1 and 2 have been prepared from a cell line obtained by fusing a xenogeneic hybridoma designated SPAZ 4 with spleen cells of a patient immune to Herpes Simplex Virus.

Description

MONOCLONAL ANTIBODY TO HERPES SIMPLEX VIRUS AND CELL LINE PRODUCING SAME
This invention relates to monoclonal antibodies specific for the herpes simplex virus, to cell lines which produce these antibodies, methods of producing the cell lines, and methods of using the antibodies in therapy.
BACKGROUND OF THE INVENTION The two herpes simplex viruses, HSV-l and HSV-2 are common human pathogens which generally produce mild and self- limiting diseases such as recurrent oral lesions, e .g. , "cold sores" or more severe recurrent genital lesions. However, under certain circumstances HSV viruses may also produce still more serious and even life-threatening infections. The most frequent of these serious diseases are: (1) ocular infections, which, if untreated, may lead to blindness; and
(2) serious/potentially fatal infections in immunocompromised individuals such as AIDS patients, cancer patients, transplant recipients, and newborn infants.
Numerous glycoproteins which are antigenic have been identified on the virion envelope and include those designated gB, gC, gD, gE, gG, gl, gH, gJ, gK, gL and gM. These glycoproteins appear on both HSV-l and HSV-2 with varying degrees of structural and antigenic variability. Monoclonal antibodies, many of them murine, have been made which are specific to various of these glycoproteins. See, for example, Balachandran et al . , 1982, Infection and Immunity vol. 37 (3) .1132-1137; Rector et al . , 1982, Infection and Immunity vol. 38 (1):168-174; and Dix et al . , 1981, Infection and Immunity vol. 34 (1):192-199. There are a few reports in the literature of human antibodies against HSV. For instance, Erlich et al., 1986, Review of Infectious Diseases vol. 8 (Supp. 4) :S439-S445 discusses human immunoglobulin G which shows activity against HSV. Seigneurin et al . , 1983, Science vol. 221:173-175 alleges to have isolated a human monoclonal antibody which binds to glycoprotein gD, and can neutralize, albeit at varying potencies, both HSV-l and HSV-2. This cell line was obtained from Epstein-Barr virus infected bone marrow.
SUMMARY OF THE INVENTION This invention provides human monoclonal antibodies which are able to bind to the glycoprotein gD antigen of HSV-l and HSV-2 and are able to neutralize HSV-l and HSV-2 with substantially equivalent potency. This invention also provides for a virus-free cell line which produces human monoclonal antibodies which are able to bind to the glycoprotein D antigen of HSV-l and HSV-2. In one embodiment of this aspect of the invention, the cell line is a xenogeneic hybridoma, i.e., a cell which results from the fusion of cells from different species.
A further aspect of this invention is a cell supernatant which comprises a human monoclonal antibody (Mab) able to bind to the glycoprotein gD antigen of HSV-l and HSV-2 and is also able to neutralize HSV-l and HSV-2 with substantially equivalent potency, the supernatant being substantially free from any other antibody.
Another aspect of this invention is use of the aforementioned antibodies in vivo to prevent or lessen the severity of HSV-l and/or HSV-2 symptoms by administering to a mammal in need of such treatment a symptom-lessening or symptom-preventing amount of a human monoclonal antibody which is able to bind to the glycoprotein gD antigen and neutralize the virus.
The hybridomas of this invention can be made by fusing an immortalized cell to another cell which is chosen for its ability to produce antibodies to the desired antigen. See, e .g. , Kδhler and Milstein, 1975, Nature 256:495-497. However, there have been problems in developing hybridomas which stably produce human monoclonal antibodies using the Kδhler-Milstein technique. It was discovered by δstberg that xenogeneic triomas, made by using a xenogeneic hybridoma fusion partner which has lost its ability to produce immunoglobulins as the immortalizing cell, are more stable and produce antibodies efficiently. This procedure is described in detail in U.S. Patent 4,634,664 and in Hybridoma , 1983, vol. 2(4):361 which are hereby incorporated by reference. In a preferred embodiment of this invention, the hybridoma is a xenogeneic trio a which produces human monoclonal antibodies against the glycoprotein gD antigen of HSV-l and HSV-2. More particularly, it has now been found that cell lines comprising a parent rodent immortalizing cell, such as a murine myeloma cell, e .g. , SP-2, when fused to a human partner cell results in immortalizing xenogeneic hybridoma cells, which, when fused to cells capable of producing an antibody against Herpes Simplex virus 1 and 2 (HSV-l and 2) , provide a novel cell lines capable of generating human antibody effective against such virus in the human.
The publications earlier referred to describe the preparation of a xenogeneic hybridoma referred to as SPAZ 4, prepared from cell line SP-2 obtainable, e . g. , from the NIGMS Human Genetic Mutant Cell Repository Ref. CM35669A (see U.S. DHHS 1982 Catalog of Cell Lines) . This SP-2 cell line is drug resistant and is fused with normal human peripheral lymphocytes by conventional techniques. A large number of hybrids is obtained and, after approximately five weeks, five clones are selected which show fast growth and no antibody production. These cells are selected for resistance to 8- azaguanine and with three of these lines it is possible to obtain mutants which are resistant to 20 μg/mL of 8- azaguanine. These cells are at the same time sensitive to
Hypoxanthine-Aminopterin-Thymidine (HAT) medium which showed that they had lost their ability to produce hypoxanthine phosphoribosyl transferase. One of these cell lines is SPAZ 4. Two methodologies were used to make the monoclonal antibodies of this invention. Both use the cell line SPAZ 4.
Cell line SPAZ 4 may be fused with spleen cells obtained from an individual whose spleen is sensitized with HSV-l or HSV-2 antigen. It was found in the first methodology that both hybridoma-like cells and lymphoblastoid cells (spontaneously Epstein-Barr Virus (EBV) transformed) were obtained after such fusion. Contrary to what was hoped, cloning of the hybridoma- like cells failed to generate the fused cells desired.
Instead, after great difficulty, a lymphoblastoid cell is re¬ fused to SPAZ 4 and resulting cell line having hybridoma-like morphology, designated SB IV-6-2 produces antibodies which neutralize HSV-l and HSV-2 in vitro , in some cases at concentrations as low as 50 ng/mL.
It has been found that SB IV-6-2 when tested in mice injected with HSV-2 is effective in protecting such mice at a dosage of 100 μg/animal (approximately 4 mg/kg) .
The second methodology involved fusion of SPAZ 4 cells with spleen cells responding in vitro to heat inactivated HSV-l antigens. One cell line obtained in this way is designated HSV 863 (also known as 64-683) . The HSV 863 cell line is extremely stable over time; it has been in culture over 12 months. One of the surprising traits of this cell line is that it can produce a high yield of antibody.
This is particularly advantageous for scale up production of commercial quantities of antibody.
BRIEF DESCRIPTION OF THE FIGURES Figure 1. Sequences of the HSV 863 antibody light chain (A) and heavy chain (B) variable region cDNAs (SEQ. ID. Nos. 3 and 1) and the translated amino acid sequences (1-letter code) (SEQ. ID. Nos. 4 and 2) . The first amino acid of the mature light chain and of the mature heavy chain is double underlined, and the three complementarity determining regions (CDRs) in each chain are underlined.
DESCRIPTION OF THE PREFERRED EMBODIMENT The preferred monoclonal antibody of this invention is produced by cell line HSV 863 and is also designated HSV 863. It has been surprisingly found that the potency of HSV 863 substantially exceeds that of any other known anti-HSV preparation. Monoclonal antibody HSV 863 belongs to the IgGl class. It has lambda light chains. It was also found to bind to Staphylococcus aureus protein A (SpA) . This property is particularly advantageous because binding to SpA provides an efficient, specific and economical method to purify antibodies using a single step technique.
Mab HSV 863 was shown to be specific for HSV viruses. It reacted with 15 independent isolates of HSV-l and HSV-2, but no cross-reactivity was observed with uninfected Vero cells or with varicella-zoster virus. Further and surprisingly, HSV 863 was found to neutralize both HSV-l and HSV-2 substantially equally, and with a surprising potency. It is believed that HSV 863 is the most potent HSV neutralizing monoclonal antibody of any origin to date. The monoclonal antibodies of this invention are particularly well suited for clinical and diagnostic use, but especially for clinical use.
The preferred monoclonal antibody of this invention, HSV 863, has been shown to protect mice from both HSV-l and HSV-2 infection. The antibodies can be administered prior to infection or even after infection. For HSV-l, the antibody was strongly protective up to 24 hours after infection and was protective, albeit to a lesser degree, for 48 hours thereafter. HSV 863, and other preferred antibodies of the invention are fully human antibodies, which comprises natural combinations of heavy and light chains. These antibodies differ, for example, from many of the antibodies produced from phage combinatorial libraries of heavy and light chains, in which the individual heavy and light chains may not be naturally associated.
For HSV-2 infections, the antibody was strongly protective even at 48 hours post-infection. Antibody HSV 863 was also shown to be effective in lessening the symptoms of HSV cutaneous disease, when administered after infection.
Even better results are expected in humans due to the longer half-life and lack of immunogenicity of human antibodies in humans. Thus, an important aspect of this invention is the use of the antibodies of the present invention for both their prophylactic and therapeutic benefits in mammals, and especially in humans. The HSV diseases particularly targeted include primary or reactivation disease in immunocompromised patients (e .g. , bone marrow and other transplanted patients, cancer therapy patients, and AIDS patients). For these patients, the probability of HSV disease often exceeds 50% and the HSV infection can be severe and even life-threatening. Presently acyclovir (9-[ (2-hydroxyethety1-methyl] guanine) , may be used as prophylaxis in some circumstances, but not in all cases. Published data suggests that transplacentally-derived antibody affects the outcome of infection after human neonatal exposure to HSV. The antibody appears to completely neutralize virus in some infants and prolongs the incubation period and modifies the infection in others (Yeager et al . , Infection and Immunity, 29:532-538, 1980; Sullender et al. J. Inf . Dis . 155:28-37, 1987). An estimate of the dose of commercial immune globulin which would be required to achieve prophylaxis of newborn infants at risk concluded that at least 100 mL (approximately 5 g) of immune globulin would be needed. This high dose of conventional immune globulin containing only in part functionally active anti-HSV antibodies, is difficult or impossible to administer, and side-effects may be severe. However, a preparation of monoclonal antibody, in which all molecules are functionally active could achieve efficacy at much lower and more manageable doses. This improved efficacy can be seen in neonates in which prophylactic antibody results in complete absence of disease or in greatly reduced severity or modified duration of incubation of disease and reduced latent infection. Improved efficacy can also be observed in neonates, children or adults in which monoclonal antibody administered therapeutically (i.e., after virus infection is evident) reduces latent virus infection and lessens the severity of HSV disease.
The benefits include but are not limited to decreasing latent viral infection and symptoms and sequelae of HSV disease; extending, the incubation time of infection which would create an extended time for clinicians to definitively diagnose the HSV infection; and providing a more slowly evolving illness in which other therapies (e .g. , acyclovir) are more effective. Using the monoclonal antibodies of the present invention, either as a replacement for, or administered along with acyclovir, is especially valuable for patients with impaired renal function, those with ocular or CNS disease, and those having or at risk to develop acyclovir- resistant variants. (Recently, there have been reports of acyclovir-resistant virus strains. See, e .g. , Schinazi et ai. (1986) J. Antimicrobial Chemotherapy 18 (Supp B) :127-134; Chatis et al . (1989) New Engl . J. Med. 320(5) :297-300; and Erlich et al. (1989) New Engl . J. Med . 320(5) :293-296.) From animal studies, it can be concluded that the antibodies of this invention are safe even at high doses and can provide prophylactic cover with a half-life of 2-3 weeks. An additional result of monoclonal antibody therapy is the reduction of colonization and latency by HSV primary infections; this is especially relevant to pediatric patients and genital infections.
The antibodies of the present invention are particularly useful in providing pre- or post-exposure prophylactic cover to neonates at risk in acquiring HSV disease since morbidity and sequelae are severe in this patient group despite currently available therapies. Animal studies have shown that neonates can be protected even when antibody administration is delayed 24 hours or more after infection. Since HSV disease evolves more slowly in man, including infants, the opportunity to administer beneficial antibody is extended for a considerably longer time. Administration of the antibody decreases the attack rate (proportion of patients who become infected relative to those at risk) and reduces the severity and rate of progression and dissemination of disease. This can provide critical time to establish a definitive diagnosis of HSV (by virus culture) and to initiate acyclovir and additional monoclonal antibody therapy as well as to provide additional time for infant immunological maturation. It is in the immediate perinatal period (0 to 6 weeks) when infants are especially vulnerable to the devastating form of HSV disease.
The antibodies of this invention are also useful in the treatment of ocular HSV infections (250,000-500,000 per year in the U.S.A.). Again, in primary disease, the antibodies can ameliorate symptoms (in possible synergy with acyclovir) and interfere with establishment of latency. This would offer great value since a large proportion of patients later reactivate latent ocular HSV infections and some progress to blindness.
The antibodies of this invention are thus suitable for parenteral therapy for prophylaxis of HSV in a variety of patients and for many clinical presentations of HSV disease. They are especially suited in instances of low index of suspicion, where drug therapy may be contraindicated or undesirable, since they have no mechanistic toxicity and do not possess any inherent toxicity or side effects.
Antibody HSV 863 is the most potent neutralizing antibody yet to be reported and shows an extremely broad reactivity with all HSV-l and HSV-2 isolates. As an IgGl antibody, it can mediate immunological effects via several potential mechanisms: steric interference upon binding to virus, complement activation through classical or alternate routes, inhibition of cell to cell spread of virus, opsonization mediated by Fc receptors on mononuclear and polymorphonuclear phagocytes and antibody-dependent cellular cytotoxicity (ADCC) . Mab HSV 863 can thus independently or in collaboration with serum complement and cellular elements, direct immune effector responses to free virions or to viral antigens expressed on newly infected cells. These multiple mechanisms likely account for the high in vivo biological activity of HSV 863, and indicate HSV 863 will be highly effective as a sole treatment. Another aspect of this invention is the antibody fragments, e .g. , Fabs which retain the ability to bind to the HSV antigen. The antibodies of the invention can be used to manufacture pharmaceutical compositions. The antibodies and pharmaceutical compositions containing the antibodies are useful for parenteral administration, i.e., subcutaneously, intramuscularly or intravenously. The antibodies and pharmaceutical compositions of the invention can also be administered, typically for local application, by gavage or lavage, intraperitoneal injection, ophthalmic ointment, topical ointment, intravaginal , intranasal or intrabursal injection. The compositions for parenteral administration commonly comprise a solution of the immunoglobulin or a cocktail thereof dissolved in an acceptable carrier, preferably an aqueous carrier. A variety of aqueous carriers can be used, e . g. , water, buffered water, phosphate buffered saline (PBS), 0.4% saline, 0.3% giycine, human albumin solution and the like. These solutions are sterile and generally free of particulate matter. The compositions can contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example sodium citrate, sodium acetate, sodium chloride, potassium chloride, calcium chloride and sodium lactate. The concentration of antibody in these formulations can vary widely, i .e . , from less than about 0.005%, usually at least about 1% to as much as 15 or 20% by weight and is selected primarily based on fluid volumes and viscosities.
Thus, a typical pharmaceutical composition for injection contains 1 L sterile buffered water, and 1-70 mg of immunoglobulin. A typical composition for intravenous in- fusion contains 250 mL of sterile Ringer's solution, and 150 mg of antibody. Actual methods for preparing parenterally administrable compositions are described in more detail in, for example. Remington ' s Pharmaceutical Science (15th ed. , Mack Publishing company, Easton, Pennsylvania, 1980) , which is incorporated herein by reference in its entirety for all purposes. Compositions suitable for lavage or other routes are selected according to the particular use intended. The compositions containing the present antibodies or a cocktail thereof can be administered for prophylactic and/or therapeutic treatments. In therapeutic applications, compositions are administered to a patient already infected with HSV-l or -2, in an amount sufficient to cure or at least partially arrest the disease and its complications. An amount adequate to accomplish this is defined as a "therapeutically effective dose." Amounts effective for this use depend upon the severity of the disease and the general state of the patient's own immune system, but generally range from about 1 to about 200 mg of antibody per dose, with dosages of from 5 to 70 mg per patient being more commonly used. Dosing schedules vary with the disease state and status of the patient, and range from a single bolus dosage or continuous infusion to multiple administrations per day (e .g. , every 4-6 hours) , or as indicated by the treating physician and the patient's condition.
In prophylactic applications, compositions containing the present antibodies or a cocktail thereof are administered to a patient not already infected with HSV-l and/or HSV-2, who is at risk of such infection (e.g., the sexual partner or fetus of an infected person) to enhance the patient's resistance. Such an amount is defined to be a "prophylactically effective dose." In this use, the precise amounts again depend upon the patient's state of health and general level of immunity, but generally range from 1 to 70 mg per dose. Single or multiple administrations of the compositions can be carried out with dose levels and pattern being selected by the treating physician. The invention is further illustrated by reference to the following, non-limiting examples.
Example 1 Production of Cell Line SB-IV-6-2 Spleen cells from spleen donor SB IV were sensitized with a dilution of soluble HSV-2 antigen. Approximately 3xl08 cells were cultivated in 150 milliliters of RPMI-1640 and 5% human serum. After days of culture, 3xl07 viable cells were recovered and fused to a similar number of SPAZ-4 cells (passage 58) . When the supematants of the cultures were tested (after three weeks) , some cultures were positive in an ELISA test to detect anti-HSV antibodies. One of these cultures was designated SB IV-6-2, and it was consistently positive in ELISA tests both on HSV-2 as well as on HSV-l. It also showed neutralizing capacity against HSV-2 virus. The culture, however, contained both hybridoma-like cells and lymphoblastoid cells and the supernatant contains antibodies of both the IgG and IgM class. The antibodies also had light chains of both kappa and lambda types. After performing some unsuccessful experiments to clone the positive cells, it was subjectively estimated that the larger the proportion of lymphoblastoid cells in the culture the higher the antibody concentration. Addition of ouabain exterminated the lymphoblastoid cells and the antibody disappeared. The lymphoblastoid cells were grown in a larger scale and approximately 6 months after the original fusion, the lymphoblastoid cells were fused to the SPAZ-4 cell line (passage 116) . The selection procedure was the standard HAT- treatment but with an addition of 2xlO~7M ouabain to suppress the growth of unfused lymphoblastoid cells. This fusion yielded several cultures with antibodies against HSV and with cells of hybridoma-like morphology. The best growing of these cell lines, which was still designated. SB IV-6-2 (deposited subject to the Budapest Treaty with the ATCC, Rockville, MD on February 3, 1987, and designated HB 9316), has been cloned and expanded and been found to produce an IgG, kappa-light chain antibody, that binds to the antigens of both HSV-l and HSV-2 and neutralizes both viruses in vitro. The neutralizing capacity was found to be substantially stronger against HSV-2 and was detected down to concentrations of 50 ng/mL.
Ex-am le 2 Use of SB IV-6-2 Antibody In Vivo
An in vivo protection study was performed by pre- treating three-week old Balb/c mice with antibody from SB IV- 6-2 and control cells 24 hours prior to footpad injection of 10 LD50-doses of HSV-2.virus. Two control groups were used, one receiving only saline and one receiving a human anti- cytomegalovirus antibody. There were 12 animals in each group; in the saline treated group all animals succumbed by day 21. In the group receiving control antibody, one animal was still alive at day 26 and in the antibody treated group five animals were still alive at day 26. The dose of antibody given was 100 μg per animal. This experimental model involved an infection in the central nervous system since the virus has a capacity of retrograde transport inside nerves. The result was statistically significant for comparison of the treated group to the saline group and the treated group to the combined saline and control antibody groups.
Exam le 3
Production of Cell Line HSV 863 A. Production of antibodv-producinσ cells
Vero cells were infected with HSV-l (Mclntyre) at an m.o.i. = 1 for 48-72 hours. The antigen containing cells were harvested, washed in phosphate buffered saline (PBS) , diluted in PBS (one 180 cm2 flask per 10 mL) then sonicated, heat- inactivated 56βC, 30 min., and then frozen at -80°C until used.
B. Fusion
After 7 days of culture, in the presence of antigen in RPMI-1640 medium containing 10% human plasma, SBIII human spleen cells were fused with SPAZ-4 (mouse x mouse) hybridoma. SPAZ-4 cells are known in the art and can be produced according to methods described in US 4,634,664, which is hereby incorporated by reference. The SBIII x SPAZ-4 fused cells were grown in Dulbecco's modified minimal essential medium containing supplementary pyruvate, oxaloacetic acid, non-essential amino acids, 20% fetal calf serum (from a selected batch) , hypoxanthine, a inopterin, and thymidine according to standard protocols. In addition, ouabain (10"7M) was added to eradicate any unfused human B cells (which may secrete abundant HSV antibody and can be misleading in ELISA assays) . The cells were cultured for two weeks with occasional medium changes, after which supematants were tested for HSV antibody production with ELISA assays as described below.
C. Screen for antibody production
HSV antigen was prepared by infecting Vero cells with HSV-l (Mclntyre) at m.o.i. = 10. After culturing for 48 hr. cells were washed in PBS with centrifugation at lOOg for 10 min. , and disrupted in 10 mL Vestergaard buffer per
180 cm2 flask of cells. The detergent-disrupted lysates were absorbed to plastic wells and exposed to supematants of the fused cells from Step V, supra . Antibody which binds to the HSV-l was revealed with a peroxidase-conjugated goat antibody to human H and L chains (Tago, Burlingame, Ca.) The antigen- binding supematants were identified and their respective cells were expanded in culture.
After 1-2 weeks in 2 mL wells, supematants were re- tested for antigen binding and also for lack of binding to Vero host cell lysates. This test established preliminary evidence for specificity. The cells were then cloned four times at a seeding density of 30, 3, and 0.3 cells per well in 96 wells, and samples of live cells were stored frozen at each stage. From one cell fusion, 30 cultures tested "positive" in the screening ELISA, and one of these, identified as HSV 863 which had an intermediate score in the screening ELISA, was found to be stable for antibody production in continuous culture.
Example 4 Optimization of HSV 863 cultures A high concentration of HSV 863 cells (IO7 per mL) was cultured directly into serum-free media DMEM/F12 without any supplements. While most cells die, the surviving fraction grew well at high densities in serum free medium and produced high levels of antibody. Cells were subcultured at approximately 14 day intervals by feeding with 50% volume additions on days 7 and 14.
In serum-free conditions, HSV 863 cells proliferate and produce antibody in stationary cultures, but these cells will die in 3-5 days if placed in rotating bottles. If, however, the cells are cultures in 1% serum, rotation does not adversely affect cells.
Antibody can be harvested from "exhausted" supematants by culturing cells at 37βC and allowing the cultures to grow without further addition of medium. After 5- 10 days, the cell death is complete and the supernatant is clarified by centrifugation. Serologic tests using ELISA methods and a standard of pure HSV 863 antibody showed that cells grown in DMEM with 5% fetal calf serum (FCS) produced exhausted supematants containing approximately 300 μg per mL monoclonal antibody and cells grown in DMEM with 0% FCS produced in excess of 60 μg per mL. This represents one of the highest ranges reported for monoclonal antibody production, regardless of origin. The genetic stability of antibody production by HSV
863 cells was demonstrated by testing antibody production of individual clones isolated from cells which are grown in protein-free media for over six months. 48 of 49 growing colonies produced HSV 863 antibody.
Example 5 immunochemical Characterization A. Antibody class/subclass: IσGl
The immunoglobulin class of antibody HSV 863 was determined using ELISA methods. The antibody was captured on antigen-coated plates and the assay was developed with subclass specific, peroxidase-conjugated anti-human lg (Tago) . Antibody HSV 863 was clearly identified as an IgGl antibody.
B. Liσht Chain Type: λ
Using ELISA methods similar to those described in Step A, HSV 863 was tested with anti K or λ light chain reagents (Tago) . The .antibody was clearly identified as having λ light chains.
C. Binding to Staphγlococcus aureus Protein A fSpAi Antibodies were tested for the ability to bind to SpA by ELISA methods. Antibody HSV 863 was captured on antigen, and the ELISA was developed with peroxidase- conjugated SpA. A strong stain identified HSV 863 as an SpA- binding immunoglobulin.
D. Polvacrvlamide Gel Electrophoresis (PAGE)
To assess purity and homogeneity of HSV 863 obtained from SpA purification, PAGE was performed. The antibody displayed two bands of approximately 50-55 kD and 25-30 kD. Thus it was concluded that the antibody was homogeneous and highly pure after elution from SpA.
E. Isoelectric Focusing (IEFi
A sample of antibody HSV 863 was applied to a gel. HSV 863 behaved as a highly basic protein.
F.
Figure imgf000017_0001
To determine the molecular weight and state of glycosylation of the HSV antigen which was recognized by HSV 863, the antibody was used to immunoprecipitate proteins from biosynthetically labelled cells, mock-infected or infected with HSV-l or HSV-2. The culture media were adjusted to supply 14C-glucosamine or 35S-methionine to the infected cells. The cells were dissolved in detergent buffer and the radiolabelled solutions were exposed to antibody HSV 863 or to irrelevant antibody 53-2-4. The complexes of antigen plus antibody were captured on insoluble beads containing SpA and were dissolved, denatured and reduced in a sample buffer prior to PAGE analysis and autoradiography. Antibody HSV 863 precipitated an antigen of HSV-l of 55 kD and an antigen of HSV-2 of 50-55 kD. The HSV antigen was glycosylated since 14C-glucosamine was incorporated into the molecule. Control, uninfected Vero cells, and antibody 53-2-4 do not display this prominent antigen. The molecules precipitated by HSV 863 were identified as HSV-l gD using methods described in Eisenberg et al . , J. Virol . 36:428-435 (1980), which is hereby incorporated by reference. As anti-gD Mab can be divided into groups on the basis of numerous biological and biochemical characteristics (see Eisenberg et al . , J. Virol . 53:634-644 (1985)), the binding of HSV 863 to the gD glycoprotein was further characterized by competitive binding in ELISA with a variety of murine Mab of known classification. This work showed that 84-863 binds to the lb epitope of HSV, which is a highly conserved type-common epitope essential for infectivity of the virus.
G. Reactivity ?f HSV 9$3 To determine if HSV 863 recognizes isolates of HSV-l and HSV-2 which are present in the human population, antibody HSV 863 was tested for its ability to react with HSV viruses of independent origin by ELISA assay. In addition, it was tested for reactivity with antigens of uninfected cells and antigens of cells infected with the distantly related varicella-zoster virus (VZV) . Results show that HSV 863 reacted with 15 independent isolates of HSV-l and 15 independent isolates of HSV-2 but not at all with Vero or VZV antigens. Thus, the antigen recognized by HSV 863 was shared by all herpes simplex virus isolates tested to date, but was not expressed by uninfected cells or cells infected by a different virus.
Example 6 Biological Characterization
A. HSV neutralization bv HSV 863
Antibody HSV 863 was purified and tested in a virus neutralization assay using HSV-l and HSV-2. The microneutral- ization assay was designed so that 10 to 20 plaque forming units (p.f.u.) were present in each well in the presence of the antibody. After incubation for 30 min. at 37°C, the virus was transferred to susceptible Vero cells and incubated for 48 hours to allow plaques to become evident. Results showed that high concentrations of HSV 863 totally abolished the appearance of plaques. Doubling dilutions of antibody revealed a 50% inhibition of p.f.u. at an antibody concentration of about 0.05-0.35 μg/mL depending on the strain, with an average of 0.2 μg/mL, which defines the IC50. Virtually identical results were obtained for HSV-l and HSV-2 using HSV 863. The in vitro neutralizing potency is the same in the presence or absence of complement, and it is 128 and 256 times higher against HSV-l and HSV-2, respectively, per microgram of protein, than that of pooled polyspecific human gamma globulin (Sandoglobulin®) .
B. Fusion Assays
To determine the ability of HSV 863 to inhibit HSV-induced cell-cell fusion, the mAb was tested in fusion assays as follows. The syncytial HSV-l variant HFEMsyn ('113) was added to freshly confluent Vero cell monolayers in 24-well plates at an MOI of 20 (200 μL/well in DMEM-2% FCS) . Following a 1 h incubation at 37βC, the contents of the wells were aspirated, and 500 μL/well of appropriate antibody dilutions were added to duplicate wells. The plates were incubated for an additional 7 h at 37βC, stained with a 1% crystal violet-10% formaldehyde solution, and the wells evaluated for syncytium formation. At concentrations of 20 μg/mL, HSV 863 was found to be highly effective at preventing syncytium formation with an approximate IC50 of 10 μg/mL and some efficacy still evident at concentrations as low as 2.5 μg/mL. Thus, HSV 863 can neutralize virus after adsorption as well as before absorption. However, the concentration required to inhibit syncytium formation is greater than that required to neutralize free virus.
C. Plague Inhibition Assays Freshly confluent Vero cell monolayers in 24-well plates were infected with approximately 30 pfu of HSV-l (Mclntyre) or HSV-2(333) in 200 μL of DMEM-2% FCS. After adsorption for 2 h at 37βC in 5% C02, an appropriate dilution of antibody was added per well. The plates were again incubated for an additional 48 h, and the monolayers were then fixed and stained with a 1% crystal violet-10% formaldehyde solution. To quantitate plaque area, a standardized image of each sample was obtained using a Wild-Heerburgg stereo microscope with a Panasonic WV-CD50 video camera. Images were stored for future analysis on disk in a Zeiss IBAS image analysis system. Fifteen plaques per sample were isolated and each plaque area measured in μm2. There was a marked dose-dependent decrease in mean plaque size in the presence of HSV 863 as compared with the irrelevant mAb control, indicating inhibition of intercellular spread of virus. Maximum decrease of plaque size was achieved at concentrations of HSV 863 as low as 50 μg/mL for both HSV-l (IC50 « 30.4 μg/mL) and HSV-2 (IC50 « 21.3 μg/mL). Thus, HSV 863 is effective at inhibiting the intercellular spread of syncytial and non-syncytial HSV strains, albeit at higher concentration than necessary to neutralize virus or mediate ADCC activity in vitro.
D. ADCC
Since ADCC is implicated as one of the pivotal mechanisms of antibody-mediated defense in HSV infection, HSV 863 was tested for this activity in vitro using a 51Cr-release microcytotoxicity assay. There was a marked dose-dependent increase in ADCC-mediated specific release of 51Cr in the presence of HSV-863 as compared to the irrelevant mAb control or the medium-alone control. Maximum ADCC efficacy was achieved with concentrations of HSV 863 as low as l μg/mL with HSV-l infected target cells and 5 μg/mL with HSV-2 infected target cells. These results, therefore, demonstrate that even at low concentrations, HSV 863 is highly effective at mediating ADCC destruction of both HSV-l and HSV-2 infected cells.
E. Half-life gtqflj g in rheg-ug mc-pHeY?
Two healthy male rhesus monkeys (Macaca mulatta) weighing 7.8 and 8.4 kg were injected once i.v. with HSV 863 at a 0.5 mg/kg body weight dose. Serum samples were obtained prior to injection with HSV 863 and at regular weekly intervals thereafter for 7 weeks. The half-life of SDZ 264- 863 was calculated to be 17.0 days in one monkey and 20.1 days in the other monkey (average 18.6 days).
F. Monoclonal Antibody Resistant ( AR) Variants
A staining protocol was used in attempts to detect the frequency with which pre-existing (non-binding) MAR variants are encountered in a total of 109 pfu of HSV-l
(Mclntyre) and HSV-2 (clinical isolate 1811) . Virus samples (~107 pfu/mL) were incubated for 30 min at 37βC with an equal volume of HSV 863 (final concentration of 1 mg/mL) in DMEM-2% FCS. The virus-antibody mixture was then added (200 μL/well) to freshly confluent Vero cell monolayers in 24-well plates. The monolayers were incubated for 48 h at 37°C, fixed, stained, and checked for plaques which would not bind/stain with HSV 863 in the presence of peroxidase-conjugated goat antibody specific for the heavy chain of human IgG. Although the clinical HSV-2 strain demonstrated a higher rate of neutralization initial-escape plaques (3.3xl0~4 versus l.lxlO"7 for the laboratory HSV-l strain), all plaques bound HSV 863 and even 3 days post-infection appeared to be neutralized and contained, unlike the controls which demonstrated secondary infection and poor monolayer integrity. Therefore, non-binding HSV 863 MAR variants could not be detected in a total of IO9 PFU of HSV assayed, implying that the freguency of non-binding HSV-863 MAR variants may be less than IO"9. To generate or select for neutralization-resistant variants to HSV 863, greater numbers of virus PFU, cultured in the presence of low concentrations HSV 863, were repeatedly passaged over several weeks. Following neutralization with range of HSV 863 concentrations, a minimum IO8 PFU of HSV-l (Mclntyre) and 107 PFU of HSV-2 (clinical isolate 1811) were passaged 16 and 15 times respectively. For each passage, the virus samples (a minimum of 107 HSV-l pfu/well or IO6 HSV-2 pfu/well) were mixed in duplicate with doubling dilutions of HSV 863 (20- 0.6 μg/mL.final concentration), incubated for 2 h at 37°C, and added to freshly confluent Vero cell monolayers in 6-well plates. The monolayers were incubated for 2-3 days at 37°C to allow for proliferation of virus which had escaped neutralization of HSV-863. The monolayers were aspirated, scraped into DMEM-2% FCS, pooled, sonicated to release cell-free virus, and again passaged in HSV 863.
Even after 16 passages (≥108 pfu/passage) in HSV 863, there were still no detectable MAR variants in the HSV-l (Mclntyre) population. However, following 13 passages (> IO7 pfu/passage) in HSV 863, the resultant HSV-2 (1811) population demonstrated an increased resistance to the mAb. The HSV-2 populations resulting from 13 to 15 passages in HSV 863 required 10 to 30-fold more HSV 863 to achieve complete neutralization as compared to the parental strain. These data indicate that increased resistance to HSV 863 neutralization is difficult to achieve in vitro.
G. Protection from HSV lethality in vivo Mouse models of HSV-l and HSV-2 disease were developed to assess the ability of HSV 863 to protect young animals from HSV infection. Mice were chosen since only a small amount of antibody is required for therapeutic dose levels and since it is possible to test large number of animals in dose-response analyses. Virulent forms of HSV-l (strain Patton) and HSV-2 (333) were grown for use. Preliminary experiments established the appropriate doses for LD50 in 3-week-old mice using HSV-l for i.p. inoculation and HSV-2 for footpad inoculation. Protection by HSV 863 was tested by dosing mice with 10xLD50 and administering purified HSV 863 in phosphate buffered saline intraperitoneally.
Purified antibody retains full specific functional activity: RSV-2 rpt gtΪQη in VJvo In one experiment, groups of 15 mice were administered either purified or unpurified HSV 863 antibody 24 hours prior to inoculation with HSV-2 (lθxLD50) in the footpad. Antibody was administered at 0, 1, 10, 100 or 1000 μg per animal. The percent survival after 21 days is given in Table I, below. As is evident from this table, the purified antibody activity was indistinguishable from that of the crude supernatant, and the purification procedure which involved a brief exposure to pH 3.5 had no detectable effect on antibody specific activity.
TAB! E I
PROTECTION OF YC(UNO MICE Fl IOM LETHAL HSV-2 INFECTION
BY HUMAN MONOCLONAL ANTIBODY HSV 863
PURIFIED ANTIBODY CRUDE SUPERNATANT
Dose of Antibody N % Survival Dose of Antibody N % Survival
1000 15 100 1000 15 100
300 15 93.3 300 14 85.7
100 15 60.0 100 15 46.7
30 15 33.3 30 15 30.0
10 15 33.3 10 14 28.6
3.0 15 13.3 3.0 15 20.0
1.0 15 0 1.0 15 6.7
0.3 15 0 0.3 15 0
PBS 15 13.3
Antibody HSV 863 waβ administered i.p., 24h prior to injection with IOXLDJO of HSV-2 strain 333 in the footpad. Values are shown for survival at 21 days. This result demonstrates that immunoglobulin purification does not reduce the biological efficacy of antibody HSV 863.
In an additional experiment, efficacy of the HSV 863 antibody was shown in mice inoculated intranasally with HSV-2. Mice received a single 0.5 mL does i.p. of antibody (450 or 900 μg) after HSV-2 inoculation at the time indicated in the table. Protection was determined by measuring percentage mortality and mean day of death (MDD) in groups of 15 mice treated with HSV 863 antibody versus placebo. Also shown in the table are data for a neutralizing antibody (HuFd79D) specific for the gB protein of HSV-2. TABLE II
EFFECT OF TREATMENT WITH HSV ANTIBODY ON THE MORTALITY 1 OF MICE INOCULATED INTRANASALLY WITH HSV-2
Mortality
Treatment Number Percent P-Value MDD P-Value
Control 14/15 93 7.6 Placebo 15/15 100 NS 8.2 NS
HuFd79D
900 μg +24h 0/15 0 <0.001 NS 900 μg +48h 2/15 13 <0.001 10.0 NS 900 μg +72h 6/15 40 <0.001 9.8 NS 900 μg +96h 7/15 47 <0.01 8.7 NS
450 μg +24h 4/15 27 <0.001 10.3 0.06 450 μg +48h 7/15 47 <0.01 11.1 <0.01 450 μg +72h 5/15 33 <0.001 10.4 <0.05 450 μg +96h 12/15 80 NS 10.4 <0.05
HSV 863
900 μg +24h 4/15 27 <0.001 13.8 NS 900 μg +48h 9/15 60 <0.05 12.2 <0.01 900 μg +72h 8/15 53 <0.01 10.3 <0.05 900 μg +96h 8/15 53 <0.01 9.3 NS
450 μg +24h 4/15 27 <0.001 12.8 0.01 450 μg +48h 6/15 40 <0.001 13.3 0.001 450 μg +72h 6/15 40 <0.001 9.0 NS 450 μg +96h 14/15 93 NS 9.9 0.05
NS = Not statistically significant when compared to the appropriate placebo-treated group.
Table II shows that HSV 863 treatment resulted in statistically significant reduction in mortality at both the 450 μg and 900 μg dosages and was effective up to 96 hr postinoculation at the higher dosage.
The in vivo efficacy of the HSV 863 antibody is not shared by all antibodies to the gD glycoprotein of HSV viruses, even those having strong neutralizing power in vitro . A similar experiment was performed on humanized antibody
HuFdl38, which is specific to the gD glycoprotein and has a similar in vitro neutralizing power to the HSV 863 antibody. Mice received 0.5 mL i.p. antibody 24 h after HSV-2 infection.
Figure imgf000025_0001
The Table shows that HuFdl38 did not significantly reduce mortality. The different behavior of HuFdl38 and HSV 863 probably arises through binding to different epitopes within the gD glycoprotein. These data suggest that although several epitopes within the gD glycoprotein may elicit antibodies with strong in vitro neutralizing power, only a limited number of such epitopes (e.g., the one bound by HSV 863) elicit antibodies that are effective in vivo.
H. Protection from HSV-l
In a procedure similar to that described in Step B, above, groups of 13 young mice were treated with various doses of antibody HSV 863 and challenged with HSV-l intraperitoneally after 24 hours. Survival at 14 days is shown in Table IV, below, ED50 is 17 μg per animal. At high dosages, antibody HSV 863 provided complete protection. TABLE IV
PROTECTION OF YOUNG MICE FROM LETHAL HSV-l INFECTION BY HUMAN MONOCLONAL ANTIBODY HSV 863
Dose of Antibody (μg per animal) N % Survival
1000 14 100
300 13 92.3
100 13 76.9
30 13 69.2
10 13 23.1
1 12 8.3
PBS 11 0
Antibody HSV 863 was administered i.p., 24h prior to injection with 10xLD50 neurovirulent HSV-l strain Patton intraperitoneally. Values are shown for survival at 14 days.
I. Timing of HSV 863 Administration for Protection
Iii Vjv
Two experiments were performed to analyze whether dosing with antibody was needed prior to virus challenge in order to be protective (prophylaxis) , or if the antibody could be administered during or even after infection is established. As shown below in Tables V and VI, administration of 300 μg HSV 863 antibody intraperitoneally prior to, or at the time of, infection was highly effective to protect groups of 14 or 15 mice from infection with HSV-l or HSV-2. In the case of HSV-2 (which is spread slowly by predominantly neural routes) antibody was strongly protective even at 48 hours after infection. For HSV-l (which spreads more rapidly through hematogenous routes) , antibody was strongly protective to 24 hours after infection and showed weaker effects thereafter. Thus, HSV 863 is protective whether administered prior to or subsequent to infection for both HSV-l and HSV-2 infections.
Figure imgf000027_0001
Figure imgf000028_0001
J. In vivo Potency of HSV 863
To assess the relative potency of antibody HSV 863, mice were treated with human immunoglobulin (SANDOGLOBULIN®) or with HSV 863. The specific activities of the two immunoglobulin preparations were compared in Tables VI and VII below. 27
TABLE VII
PROTECTION OF YOUNG MICE FROM LETHAL HSV-l INFECTION
BY SANDOGLOBULIN® OR HUMAN MAB HSV 863
Ab 24h PRECEDING VIRUS
HUMAN MAB HSV 863 SANDOGLOBULIN®
Dose of Antibody Dose of Antibody (μg per animal ) N % Survival (μg per animal ) N % Survival
300 14 85.7 100, 000 15 100
100 15 86.7 30, 000 15 100
30 15 53.3 10, 000 15 100
10 15 46.7 3 , 000 15 86.7
3 15 26.7 1, 000 14 78.6
300 15 33.3
100 15 33.3
PBS 15 0
Antibody HSV 863 and Sandoglobulin® were administered i . p. , 24h prior to injection with IOXLDJ- of HSV-l strain Patton intraperitoneally. Values are shown for survival at 14 days .
TABLE VIII
PROTECTION OF YOUNG MICE FROM LETHAL HSV-2 INFECTION
BY SANDOGLOBULIN® OR HUMAN MAB HSV 863 1
Ab 24h PRECEDING VIRUS
HUMAN MAB HSV 863 SANDOGLOBULIN®
Dose of Antibody Dose of Antibody ( μg per animalj N % Survival (μg per animal] N % Survival
300 15 93.3 100, 000 15 73.3
100 15 73.3 30, 000 15 66.7
30 15 53.3 10,000 15 40.0
10 15 26.6 3 , 000 15 26.7
3 14 7.1 1 , 000 15 26.7
300 15 26. 7
100 15 0
PBS 15 20.0
Antibody HSV 863 and Sandoglobulin® were administered i . p. , 24h prior 1 to injection with 10xLDM of HSV-2 strain 333 in the footpa . Values 1 are shown for survival at 21 days. The results show that for HSV-l, human immunoglobulin is protective at 300 μg per animal while HSV 863 protects at a dose of 15 μg, (a potency factor of about 20 fold) . For HSV-2, human immunoglobulin is needed at about 15,000 μg per animal while HSV 863 is effective at 25 μg (a potency factor of approximately 600) . Thus, HSV 863 is much more potent than gamma globulin in the prophylaxis models of HSV infection for both HSV-l and HSV-2. The large difference in dose of human immunoglobulins effective for HSV-l vs. HSV-2 prophylaxis may be explained by the known difference in titers of antibodies to those respective viruses which is seen in the population (many fewer people are seropositive for HSV-2 than for HSV-l) .
K. HSV 863 Inhibits HSV Lesion Formation bv Virus Discharge from Neurons
A model of skin lesion formation by HSV discharged form sensory neurons was adapted to test the ability of HSV 863 to inhibit the process of viral lesion recrudescence. Adult BALB/c mice were infected by scarification on the lateral thorax with HSV-l. After 5-7 days, severe band-like confluent lesions form unilaterally along the entire dermatome. This process is known to involve the centripetal ascent of virions to the dorsal root ganglia and centrifugal descent of virus within sensory afferents of the dermatome. Administration of HSV 863 3 days after inoculation of virus substantially decreased the frequency and severity of lesion formation and prevents mortality as shown in Table IX, below. Thus, antibody HSV 863 can act at the time of discharge of virus from neurons to reduce the severity of lesion formation. Since the half-life of human antibody in the mouse is short, these results are unexpectedly favorable. TABLE IX
Figure imgf000031_0001
L. HSV 863 Inhibits Establishment of Latent HSV Infection in a Murine Ascending Myelitis Model
Young Balb/c mice, 15-20 animals per group, were treated with a single dose of 300 μg of HSV 863, or control antibody, 24 h before or after virus challenge by IO4 p.f.u. of neurovirulent HSV-l into the right hind footpad. After 7-9 weeks the animals were sacrificed and their dorsal root ganglia removed. From each mouse 6 thoracicolu bar (T13 to L- 5) ganglia, each ipsilateral and contralateral to the site of HSV injection, were removed and individually explanted in tissue culture medium into microtiter plates. After 3 days in culture, 8x10* Vero cells were added to each well as an indicator cell. The monolayers were observed for 3-4 weeks for cytopathic effects due to HSV. To ensure detectability of infection, supematants were transferred weekly onto fresh Vero cell monolayers. The results (Table X) show that HSV 863 can very potently suppress the establishment of latency, especially when given before virus challenge. These data may actually underestimate the efficacy of HSV 863, since there was about 25% mortality rate in the control groups, so that viable virus could not be isolated. TABLE Z
HUMAN MAB HSV 863 ACTION
ON ESTABLISHMENT OF VIRAL LATENCY
Treatment Isolated Infected Infected
Treatment day ganglia ganglia %
PBS -1 193 72 37
Control mAb -1 145 46 32
HSV 863 -1 163 2 1
HSV 863 1 180 21 11
Example 7 Preparation of HSV 863 Fabs Antibody HSV 863 (0.5 to 1.5 mg/mL) was dialyzed at
4°C against 0.04M Na2HP04, 0.01M NaH2P04, 0.15M NaCl, ImM EDTA, and adjusted to pH 7.3 with HCI. One microliter of mercaptoethanol is added per mL of solution. Mercuripapain, 1/300 the mass of the IgG, was added and a stream of nitrogen was directed on top of the solution for a few seconds. The solution was sealed under parafilm and incubated for 90 min. at 37°C. The solution was then put on ice and one-tenth volume of 0.25M iodoacetamide is added. Incubation was for 0.5 hr on ice in the dark. An alternative method of digestion was to use one- tenth volume of 0.077M dithiothreitol (instead of mercaptoethanol) and 25 units of insoluble papain (papain attached to beaded agarose; one unit hydrolyzes 1.0 mole of N-α-benzoyl-L-arginineethyl ester (BAEE) per min at pH 7.0 at 30βC) per 67 mg HSV 863 IgG. The solution was rocked constantly at 37°C for 2 hr. Iodoacetamide was added as previously described except the concentration of the stock solution was only 0.16M. This method can also be used with concentrated cell supernatant that has been dialyzed against the phosphate buffer mentioned above if the antibody protein from the fetal calf serum is approximately the same or lower in concentration than the HSV 863 antibody.
A method of isolating Fab fragments from the digestion mixture is to dialyze the digested antibody against phosphate-buffered saline and remove the Fc fragments by passage through a Protein A column equilibrated in phosphate- buffered saline.
Example 8
Nudeotide and Amino Acid Sequence of HSV 863 Variable ppmajns
The genes encoding the heavy and light chains of antibodies secreted by trioma cell lines are cloned according to methods described in Sambrook et al., Molecular Cloning: A Laboratory Manual , (2nd ed. , Cold Spring Harbor, NY, 1989; Berger & Ki mel, Methods in Enzymology, Volume 152, Guide to Molecular Cloning Techniques , (Academic Press, San Diego, CA, 1987); Co et al. , J. Immunol . , 148:1149 (1992) (each of which is incorporated by reference in its entirety for all purposes) . For example, genes encoding heavy and light chains are cloned from a trioma's cDNA produced by reverse transcription of the trioma's mRNA. Cloning is accomplished by conventional techniques including the use of PCR primers that hybridize to an enzymatically inserted sequence (e .g. , a G tail) or to the sequences flanking or overlapping the genes, or segments of genes, to be cloned. The cDNA and predicted amino acid sequences of the heavy and light chain variable domains from the HSV 863 antibody are shown in Fig. 1. The provision of amino acid sequences for the
HSV 863 antibody can be exploited to provide alternate means for generating antibodies having the same or similar binding specificity. That is, antibodies that bind to the same epitope as the HSV 863 antibody or an epitope sufficiently proximal to that bound by the HSV 863 antibody to compete with the HSV 863 antibody for binding to the HSV-l and/or -2 glycoprotein. Such antibodies can, of course, also be produced using the same procedure as noted for the HSV 863 antibody, employing the HSV 863 antibody as a reference for selection of appropriate binding specificity. However, generation of variants from sequence data has the advantage of avoiding the need for immunization and immortalization. Competition is determined by an assay in which the immunoglobulin under test inhibits specific binding of a reference antibody (e.g., HSV 863) to an antigenic determinant on an HSV gD protein. Numerous types of competitive binding assays are known for example: solid phase direct or indirect radioimmunoassay (RIA) , solid phase direct or indirect enzyme immunoassay (EIA) , sandwich competition assay and ELISA (see Stahli et al.. Methods in Enzymology 9:242-253 (1983)); Harlow and Lane, "Antibodies, A Laboratory Manual," Cold Spring Harbor Press (1988)). Usually the test immunoglobulin is present in excess. Usually, when a competing antibody is present in excess, it will inhibit specific binding of a reference antibody to gD protein by at least 10, 25, 50 or 75%. Many of the amino acids in the disclosed HSV 863 sequences can undergo non-critical substitutions, additions or deletions without loss of binding specificity or effector functions, or intolerable reduction of binding affinity (i.e., below about 107 M"1) . Thus, the binding affinity of most antibodies is within a range of about 107 M"1 or IO8 M"1 to about 109 M"1. Usually, antibody chains incorporating such alterations exhibit substantial sequence identity to the reference antibody chain from which they were derived. For example, the mature light chain of antibodies derived from the HSV 863 antibody usually shows substantial sequence identity (i.e., at least 75, 85% or 95%) to the sequence of the mature light chain of the HSV 863 antibody shown in Fig. 1 (panel A) . Similarly, the mature heavy chains of derivatives typically show substantial sequence identity to the sequence of the mature heavy chain of the HSV 863 antibody shown in Fig. 1 (panel B) . Sequence identity comparisons are performed between two polypeptide sequences when optimally aligned, such as by the programs BLAZE (Intelligenetics) GAP or BESTFIT using default gap weights. Usually, antibodies comprise two identical pairs of heavy and light chains. However, bifunctional antibodies can be produced that have one heavy/light chain pair specific for the gD antigen and the other heavy/light chain pair specific for a second antigen. Conservative.substitutions (as defined by, e.g. , Co, WO 94/12215 (incorporated by reference in its entirety for all purposes)) at positions other than the CDR regions and amino acids interacting closely with the CDR regions (see Queen et al., WO 90/07861 (incorporated by reference in its entirety for all purposes) are likely to have little effect on the functional properties of antibodies, such as binding affinity, kinetics and specificity. However, in general, derivatives harboring such mutations are not preferred in comparison with the original HSV 863 antibody. Mutations within the CDR regions and at positions closely interacting with these regions, particularly nonconservative mutations, generally result in greater changes in the functional properties of an antibody. Occasionally, a mutated antibody is selected having the same specificity and increased affinity or faster binding kinetics compared with the HSV 863 antibody. Usually, the affinity of the mutated antibody is within a factor of 2, 5, 10 or 50 of the HSV 863 antibody. Phage-display technology offers powerful techniques for selecting such antibodies. See, e .g. , Dower et al., WO 91/17271; McCafferty et al., WO
92/01047; Huse, WO 92/06204 (each of which is incorporated by reference in its entirety for all purposes) .
The variable segments of human antibodies produced as described supra are typically linked to at least a portion of an antibody constant region (Fc) , typically that of a human antibody. Human constant region DNA sequences can be isolated in accordance with well-known procedures from a variety of human cells, but preferably immortalized B-cells (see Kabat et al. , Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD (1987) and (1991)), and WO 87/02671) (each of which is incorporated by reference in its entirety for all purposes) . Ordinarily, the antibody contains both light chain and heavy chain constant regions. The heavy chain constant region usually includes CHI, hinge, CH2, CH3, and sometimes CH4 regions. The present antibodies include antibodies having all types of constant regions, including IgM, IgG, IgD, IgA and IgE, and any isotype, including IgGl, IgG2, IgG3 and IgG4. When it is desired that the human antibody exhibit cytotoxic activity, the constant domain is usually a complement-fixing constant domain and the class is typically IgGl, IgG3, IgM. When such cytotoxic activity is not desirable, the constant domain may be of the IgG2 or IgG4 class. The human antibody can comprise sequences from more than one class or isotype.
Also provided in isolated form are DNA segments encoding heavy or light chains of the antibodies described supra. The DNA segments encode at least one CDR regions and usually all three CDR regions from the heavy or light chain of an antibody. Frequently, a DNA segment encodes all or substantially all of the variable region of a heavy or light chain and is thereby capable of exhibiting antigen binding capacity. Often a nucleic acid encodes an entire heavy or light chain. Because of the degeneracy of the code, a variety of nucleic acid sequences will encode each immunoglobulin amino acid sequence. The desired nucleic acid sequences can be produced by de novo solid-phase DNA synthesis or by PCR mutagenesis of an earlier prepared variant of the desired polynucleotide. Preferred nucleic acids include those encoding mature light chain variable regions having substantial sequence identity to the mature light chain variable region of the HSV 863 antibody shown in Fig. 1 (Panel A) (SEQ. ID. No. 4) . Preferred nucleic acids also include those encoding mature heavy chain variable regions having substantial sequence identity to the mature heavy chain variable region of the HSV 863 antibody shown in Fig. 1 (panel B) (SEQ. ID. No. 2).
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: HARFELDT, Elisabeth LAKE, Philip NOTTAGE, Barbara OSTBERG, Lars G.
(ii) TITLE OF INVENTION: MONOCLONAL ANTIBODY TO HERPES SIMPLEX VIRUS AND CELL LINE PRODUCING THE SAME
(iii) NUMBER OF SEQUENCES: 4
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Townsend and Townsend and Crew
(B) STREET: 379 Lytton Avenue
(C) CITY: Palo Alto
(D) STATE: California
(E) COUNTRY: US
(F) ZIP: 94301
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.25
(viii) ATTORNE /AGENT INFORMATION:
(A) NAME: Liebeβchuetz, Joe
(B) REGISTRATION NUMBER: 37,505
(C) REFERENCE/DOCKET NUMBER: 11823-005240
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: (415) 326-2400
(B) TELEFAX: (415) 326-2422
(2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 426 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..426
(D) OTHER INFORMATION: /product= "HSV863 heavy chain variable region"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
ATG GAG TTT GGG CTG AGC TGG GTT TTC CTC GTT GCT CTT TTA AGA GGT 48 Met Glu Phe Gly Leu Ser Trp Val Phe Leu Val Ala Leu Leu Arg Gly 1 5 10 15
GTC CAG TGT CAG GTG CAG CTG GTG GAG TCG GGG GGA GGC GTG GTC CAG 96 Val Gin Cys Gin Val Gin Leu Val Glu Ser Gly Gly Gly Val Val Gin 20 25 30 CCT GGG AGG TCC CTG AGA CTC. TCC TGT GCA GCG TCT GGA TTC ACC TTC 144 Pro Gly Arg Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe 35 40 45
AGT AGC CAT GTC ATG CAT TGG GTC CGC CAG GCT CCA GGC AAG GGG CTG 192 Ser Ser His Val Met His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu 50 55 60
CAG TGG CTG GCA GTT ACA TGG TAC GAT GGA AGT AAC AAA GCC TAT GGA 240 Gin Trp Leu Ala Val Thr Trp Tyr Asp Gly Ser Asn Lys Ala Tyr Gly 65 70 75 80
GAG TCC GTG AAG GGC CGA TTC ATC ATC TCC AGA GAC AAT TCC AAG AAT 288 Glu Ser Val Lys Gly Arg Phe Ile Ile Ser Arg Asp Asn Ser Lys Aβn 85 90 95
ATC CTG TAT CTG CAA ATG AAC AGC CTG AGA GCC GAG GAC ACG GCT GTG 336 lie Leu Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val 100 105 110
TAT TAC TGT GCG AGA GAG GGC TAC GGA AGG GGG CAC TAC TTC TAC GGT 384 Tyr Tyr Cys Ala Arg Glu Gly Tyr Gly Arg Gly His Tyr Phe Tyr Gly 115 120 125
CTG GAC GTC TGG GGC CGA GGG ACC ACG GTC ACC GTC TTC TCA 426
Leu Asp Val Trp Gly Arg Gly Thr Thr Val Thr Val Phe Ser 130 135 140
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 142 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Met Glu Phe Gly Leu Ser Trp Val Phe Leu Val Ala Leu Leu Arg Gly 1 5 10 15
Val Gin Cys Gin Val Gin Leu Val Glu Ser Gly Gly Gly Val Val Gin 20 25 30
Pro Gly Arg Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe 35 40 45
Ser Ser His Val Met His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu 50 55 60
Gin Trp Leu Ala Val Thr Trp Tyr Asp Gly Ser Asn Lys Ala Tyr Gly 65 70 75 80
Glu Ser Val Lys Gly Arg Phe Ile lie Ser Arg Asp Asn Ser Lys Asn 85 90 95
Ile Leu Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val 100 105 110
Tyr Tyr Cys Ala Arg Glu Gly Tyr Gly Arg Gly His Tyr Phe Tyr Gly 115 120 125
Leu Asp Val Trp Gly Arg Gly Thr Thr Val Thr Val Phe Ser 130 135 140 (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 393 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..393
(D) OTHER INFORMATION: /product= "HSV863 light chain variable region"
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
ATG GCC TGG TCT CCT CTC CTC CTC ACT CTC CTC GCT CAC TGC ACA GGG 48 Met Ala Trp Ser Pro Leu Leu Leu Thr Leu Leu Ala His Cys Thr Gly 1 5 10 15
TCC TGG GCC CAG TCT GTG CTG ACG CAG CCG CCC TCA GTG TCT GGG GCC 96
Ser Trp Ala Gin Ser Val Leu Thr Gin Pro Pro Ser Val Ser Gly Ala 20 25 30
CCA GGG GAG GGG GTC ACC CTC TCC TGC ACT GGG AGC CGC TCC AAC ATC 144 Pro Gly Glu Gly Val Thr Leu Ser Cys Thr Gly Ser Arg Ser Asn lie 35 40 45
GGG GCA GGT TAT GAT GTA CAC TGG TAC CAG CAC CTT CCA GGA ACA GCC 192 Gly Ala Gly Tyr Asp Val His Trp Tyr Gin His Leu Pro Gly Thr Ala 50 55 60
CCC AAA CTC CTC ATC TAT GGT GAC AAC AAT CGG CCC TCA GGG GTC CCT 240 Pro Lys Leu Leu Ile Tyr Gly Asp Asn Asn Arg Pro Ser Gly Val Pro 65 70 75 80
GAC CGA TTC TCT GGC TCC AAG TCT GGC ACC TCA GCC TCC CTG GCC ATC 288 Asp Arg Phe Ser Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile 85 90 95
ACT GGG CTC CAG GCT GAA GAT GAG GCT GAT TAT TAC TGC CAG TCG TAT 336 Thr Gly Leu Gin Ala Glu Asp Glu Ala Asp Tyr Tyr Cys Gin Ser Tyr 100 105 110
GAC AGC GGC CTG AGT GGG TCG ATA TTC GGC GGA GGG ACC AAG CTG ACC 384 Asp Ser Gly Leu Ser Gly Ser Ile Phe Gly Gly Gly Thr Lys Leu Thr 115 120 125
GTC CTA GGT 393
Val Leu Gly 130
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 131 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
Met Ala Trp Ser Pro Leu Leu Leu Thr Leu Leu Ala His Cys Thr Gly 1 5 10 15
Ser Trp Ala Gin Ser Val Leu Thr Gin Pro Pro Ser Val Ser Gly Ala 20 25 30
Pro Gly Glu Gly Val Thr Leu Ser Cys Thr Gly Ser Arg Ser Asn Ile 35 40 45
Gly Ala Gly Tyr Asp Val His Trp Tyr Gin His Leu Pro Gly Thr Ala 50 55 60
Pro Lys Leu Leu Ile Tyr Gly Asp Asn Asn Arg Pro Ser Gly Val Pro 65 70 75 80
Asp Arg Phe Ser Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile 85 90 95
Thr Gly Leu Gin Ala Glu Asp Glu Ala Asp Tyr Tyr Cys Gin Ser Tyr 100 105 110
Asp Ser Gly Leu Ser Gly Ser Ile Phe Gly Gly Gly Thr Lys Leu Thr 115 120 125
Val Leu Gly 130

Claims

WHAT IS CLAIMED IS: 1. A hybridoma cell line which produces a human monoclonal antibody which competes with the HSV 863 antibody for binding to the glycoprotein gD antigen of Herpes Simplex Virus-1 (HSV-l) and Herpes Simplex Virus-2 (HSV-2) .
2. A cell line according to claim 1 which is a xenogeneic hybridoma cell line comprising a xenogeneic immortalizing cell fused to a human cell producing an antibody able to bind to the glycoprotein gD of HSV-l and HSV-2.
3. A cell line according to claim 2 wherein the xenogeneic immortalizing cell comprises a hybridoma derived from a parent immortalizing cell and a lymphoid partner cell.
4. A cell line according to claim 3 wherein the parent immortalizing cell is a murine myeloma.
5. A cell line according to claim 4 which is a [human x (human x mouse) ] cell line.
6. A cell line according to claim 5 which is able to grow in serum free medium.
7. A cell line according to claim 7 which is designated HSV 863.
8. A method of producing the hybridoma cell line of claim 1 which comprises making a xenogeneic hybridoma cell line drug resistant, fusing the latter cell to a human antibody producing cell, and selecting a desired hybrid.
9. A cell supernatant comprising a human monoclonal antibody which competes with the HSV 863 antibody for binding to the glycoprotein gD antigen of Herpes Simplex Virus-1 (HSV-l) and Herpes Simplex Virus-2 (HSV-2) and is also able to neutralize HSV-l and HSV-2 with substantially equivalent potency, the supernatant being substantially free from any other antibodies.
10. A human monoclonal antibody substantially free from other protein, which competes with the HSV 863 antibody for binding to the glycoprotein gD antigen of Herpes Simplex Virus-1 (HSV-l) and Herpes Simplex Virus-2 (HSV-2) and is also able to neutralize HSV-l and HSV-2 with substantially equivalent potency.
11. An antibody according to claim 14 which has lambda light chains.
12. An antibody according to claim 11 which is of the IgGl type.
13. An antibody according to claim 13 which is further reactive with Staphylococcus aureus protein A.
14. An antibody according to claim 10 comprising a mature light chain variable region having at least 85% sequence identity to the mature light chain variable sequence of SEQ. ID. No. 4 and a mature heavy chain variable region having at least 85% sequence identity to the mature heavy chain variable sequence of SEQ. ID. No. 2.
15. The antibody of claim 14, wherein the mature light chain variable region comprises the three CDR regions of the sequence of SEQ. ID. No. 4, and the heavy chain variable region comprises the three CDR regions of the sequence of SEQ. ID. No. 2.
16. The antibody according to claim 15 which is HSV 863.
17. An antibody produced by the cell line designated ATCC HB 9316.
18. A method of preventing or lessening the severity of HSV-l or HSV-2 symptoms by administering to a patient a symptom-lessening or preventing amount of a human monoclonal antibody which competes with the HSV 863 antibody for binding to the glycoprotein gD antigen of HSV-l and HSV-2.
19. The method according to claim 18 wherein the step of administering the antibody occurs at a time prior to infection.
20. The method according to claim 18, wherein the antibody is HSV 863.
21. A Fab fragment of the human monoclonal antibody of claim 10.
22. A fragment according to claim 21 which is prepared from monoclonal antibody HSV 863.
PCT/US1996/000641 1994-09-13 1996-01-16 Monoclonal antibody to herpes simplex virus and cell line producing same WO1997026329A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP96902712A EP0876478A4 (en) 1994-09-13 1996-01-16 Monoclonal antibody to herpes simplex virus and cell line producing same
AU47016/96A AU4701696A (en) 1996-01-16 1996-01-16 Monoclonal antibody to herpes simplex virus and cell line producing same

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US08/305,683 US5646041A (en) 1987-02-12 1994-09-13 Monoclonal antibody to herpes simplex virus and cell line producing same

Publications (1)

Publication Number Publication Date
WO1997026329A1 true WO1997026329A1 (en) 1997-07-24

Family

ID=23181868

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/000641 WO1997026329A1 (en) 1994-09-13 1996-01-16 Monoclonal antibody to herpes simplex virus and cell line producing same

Country Status (3)

Country Link
US (1) US5646041A (en)
EP (1) EP0876478A4 (en)
WO (1) WO1997026329A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2248825A1 (en) * 2009-05-04 2010-11-10 Ribovax Biotechnologies SA Antigen binding fragments of an antibody for use in treating and diagnosing ocular diseases
WO2011038933A2 (en) 2009-10-01 2011-04-07 Universität Duisburg-Essen Anti-hsv antibody
US8252906B2 (en) 2009-01-05 2012-08-28 Dcb-Usa Llc Anti-herpes simplex virus antibodies and methods of use thereof
WO2012139106A3 (en) * 2011-04-08 2013-06-06 Duke University Herpes simplex virus
CN105925537A (en) * 2016-04-21 2016-09-07 中国科学院武汉病毒研究所 Anti-HSV (herpes simplex virus) glycoprotein gD monoclonal antibody and hybridoma cell for generating antibody
JP2018505181A (en) * 2015-01-28 2018-02-22 ポリケム・ソシエテ・アノニム Human monoclonal antibody having strong neutralizing activity against HSV-1 and HSV-2

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2015225499B2 (en) * 2014-03-03 2020-05-14 Albert Einstein College Of Medicine, Inc. Recombinant herpes simplex virus 2 (HSV-2) vaccine vectors
WO2024117057A1 (en) * 2022-11-28 2024-06-06 マルホ株式会社 Antibody or antigen-binding fragment thereof, and immunological measurement device using same

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH652145A5 (en) * 1982-01-22 1985-10-31 Sandoz Ag METHOD FOR IN VITRO PRODUCTION OF HYBRID OMEN WHAT human monoclonal antibodies GENERATE.
US4745182A (en) * 1984-08-24 1988-05-17 University Patents, Inc. Herpes virus specific immunological materials and methods
WO1986002092A1 (en) * 1984-09-28 1986-04-10 Teijin Limited Mouse human hybridoma producing antivirus human antibody, process for its preparation, and antivirus human monoclonal antibody
US4757018A (en) * 1985-02-11 1988-07-12 Hazleton Biotechnologies, Inc. Myeloma cell lines and uses thereof
CH670098B (en) * 1985-05-23 1989-05-12
WO1995018634A1 (en) * 1994-01-04 1995-07-13 The Scripps Research Institute Human monoclonal antibodies to herpes simplex virus and methods therefor

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
JOURNAL OF VIROLOGY, Volume 65, Number 11, issued November 1991, STAATS et al., "Anti-Glycoprotein D Monoclonal Antibody Protects Against Herpes Simplex Virus Type 1-Induced Diseases in Mice Functionally Depleted of Selected T-Cell sets or Asialo GM1+ Cells", pages 6008-14. *
JOURNAL OF VIROLOGY, Volume 70, Number 2, issued February 1996, SU et al., "Protective Antibody Therapy is Associated with Reduced Chemokine Transcripts in Herpes Simplex Type 1 Corneal Infection", pages 1277-81. *
PROC. NATL. ACAD. SCI. U.S.A., Volume 88, Number 7, issued 01 April 1991, CO et al., "Humanized Antibodies for Antiviral Therapy", pages 2869-73. *
PROC. NATL. ACAD. SCI. U.S.A., Volume 92, Number 14, issued 03 July 1995, SANNA et al., "Directed Selection of Recombinant Human Monoclonal Antibodies to Herpes Simplex Virus Glycoproteins from Phage Display Libraries", pages 6439-43. *
See also references of EP0876478A4 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8252906B2 (en) 2009-01-05 2012-08-28 Dcb-Usa Llc Anti-herpes simplex virus antibodies and methods of use thereof
US8431118B2 (en) 2009-01-05 2013-04-30 Dcb-Usa Llc Anti-herpes simplex virus antibodies and methods of use thereof
WO2010128053A1 (en) * 2009-05-04 2010-11-11 Ribovax Biotechnologies Sa Antigen binding fragments of an antibody for use in treating or diagnosing ocular diseases
EP2248825A1 (en) * 2009-05-04 2010-11-10 Ribovax Biotechnologies SA Antigen binding fragments of an antibody for use in treating and diagnosing ocular diseases
US8747856B2 (en) 2009-05-04 2014-06-10 Ribovax Biotechnologies S.A. Antigen binding fragments of an antibody for use in treating or diagnosing ocular diseases
US9657088B2 (en) 2009-10-01 2017-05-23 Heidelberg Immunotherapeutics Gmbh Anti-HSV antibody
WO2011038933A2 (en) 2009-10-01 2011-04-07 Universität Duisburg-Essen Anti-hsv antibody
EP2308895A1 (en) 2009-10-01 2011-04-13 Universität Duisburg-Essen Anti-HSV antibody
US8889137B2 (en) 2009-10-01 2014-11-18 Universität Duisberg Essen Anti-HSV antibody
EP3539983A1 (en) 2009-10-01 2019-09-18 Heidelberg ImmunoTherapeutics GmbH Anti-hsv antibody
WO2012139106A3 (en) * 2011-04-08 2013-06-06 Duke University Herpes simplex virus
JP2018505181A (en) * 2015-01-28 2018-02-22 ポリケム・ソシエテ・アノニム Human monoclonal antibody having strong neutralizing activity against HSV-1 and HSV-2
US10626167B2 (en) 2015-01-28 2020-04-21 Polichem S.A. Human monoclonal antibodies endowed with strong neutralizing activity against HSV-1 and HSV-2
CN105925537B (en) * 2016-04-21 2019-09-06 中国科学院武汉病毒研究所 A kind of monoclonal antibody of the glycoprotein gd of anti-HSV and the hybridoma for generating the antibody
CN105925537A (en) * 2016-04-21 2016-09-07 中国科学院武汉病毒研究所 Anti-HSV (herpes simplex virus) glycoprotein gD monoclonal antibody and hybridoma cell for generating antibody

Also Published As

Publication number Publication date
EP0876478A1 (en) 1998-11-11
EP0876478A4 (en) 2002-12-18
US5646041A (en) 1997-07-08

Similar Documents

Publication Publication Date Title
AU654827B2 (en) Novel antibodies for treatment and prevention of infection in animals and man
US9657088B2 (en) Anti-HSV antibody
CA2218515C (en) Monoclonal antibodies against hiv-1 and vaccines made thereof
AU681633B2 (en) Anti-HIV monoclonal antibody
KR100304333B1 (en) Recombinant humanized anti-human immunodeficiency virus antibody
EP0295803B1 (en) Monoclonal antibodies
Hamilton et al. A humanized antibody against human cytomegalovirus (CMV) gpUL75 (gH) for prophylaxis or treatment of CMV infections
WO1997026329A1 (en) Monoclonal antibody to herpes simplex virus and cell line producing same
AU695584B2 (en) Human monoclonal antibodies to cytomegalovirus
US5506132A (en) Human antibodies against varicella-zoster virus
AU4701696A (en) Monoclonal antibody to herpes simplex virus and cell line producing same
CA2242669A1 (en) Monoclonal antibody to herpes simplex virus and cell line producing same
WO1991005876A1 (en) Human cytomegalovirus-specific monoclonal antibody cocktail
WO1991016448A1 (en) Human monoclonal antibody against glycoprotein gp iii of varicella zoster virus
Casadevall et al. Antibody-based therapies for infectious diseases: renaissance for an abandoned arsenal?
JPH0411897A (en) Antipyocyanic exotoxin a human monoclonal antibody
NZ250415A (en) Monoclonal antibody directed against rsv epitope 417-438, its use and production

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AZ BY KG KZ RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE

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

Ref document number: 2242669

Country of ref document: CA

Ref country code: CA

Ref document number: 2242669

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1996902712

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996902712

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97525944

Format of ref document f/p: F

WWW Wipo information: withdrawn in national office

Ref document number: 1996902712

Country of ref document: EP