US20060029932A1 - Method for preventing HIV-1 infection of CD4+ cells - Google Patents

Method for preventing HIV-1 infection of CD4+ cells Download PDF

Info

Publication number
US20060029932A1
US20060029932A1 US11/175,815 US17581505A US2006029932A1 US 20060029932 A1 US20060029932 A1 US 20060029932A1 US 17581505 A US17581505 A US 17581505A US 2006029932 A1 US2006029932 A1 US 2006029932A1
Authority
US
United States
Prior art keywords
cells
hiv
chemokine
fusion
chemokines
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/175,815
Inventor
Graham Allaway
Virginia Litwin
Paul Maddon
William Olson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Progenics Pharmaceuticals Inc
Original Assignee
Progenics Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Progenics Pharmaceuticals Inc filed Critical Progenics Pharmaceuticals Inc
Priority to US11/175,815 priority Critical patent/US20060029932A1/en
Publication of US20060029932A1 publication Critical patent/US20060029932A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56983Viruses
    • G01N33/56988HIV or HTLV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses
    • G01N2333/15Retroviridae, e.g. bovine leukaemia virus, feline leukaemia virus, feline leukaemia virus, human T-cell leukaemia-lymphoma virus
    • G01N2333/155Lentiviridae, e.g. visna-maedi virus, equine infectious virus, FIV, SIV
    • G01N2333/16HIV-1, HIV-2
    • G01N2333/162HIV-1, HIV-2 env, e.g. gp160, gp110/120, gp41, V3, peptid T, DC4-Binding site
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70514CD4
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/715Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/20Screening for compounds of potential therapeutic value cell-free systems

Definitions

  • Chemokines are a family of related soluble proteins of molecular weight between 8 and 10 KDa, secreted by lymphocytes and other cells, which bind receptors on target cell surfaces resulting in the activation and mobilization of leukocytes, for example in the inflammatory process. Recently, Cocchi et al. demonstrated that the chemokines RANTES, MIP-1 ⁇ and MIP-1 ⁇ are factors produced by CD8 + T lymphocytes which inhibit infection by macrophage-tropic primary isolates of HIV-1, but not infection by laboratory-adapted strains of the virus (1).
  • chemokines are members of the C-C group of chemokines, so named because they have adjacent cysteine residues, unlike the C-X-C group which has a single amino acid separating these residues (2). While Cocchi et al. found that expression of HIV-1 RNA was suppressed by treatment with the chemokines, they did not identify the site of action of these molecules.
  • RET resonance energy transfer
  • fusion accessory molecules are found on a subset of human CD4 + cells and are required for infection by HIV-1 isolates with particular tropisms.
  • macrophage-tropic primary strains of HIV-1 such as HIV-1 JR-FL may have different requirements for accessory molecules compared with laboratory-adapted T lymphotropic strains such as HIV-1 LAI . This phenomenon may explain differences in tropism between HIV-1 strains.
  • the current invention comprises a series of new therapeutics for HIV-1 infection. It was demonstrated for the first time that chemokines act at the fusion step of HIV-1 entry and specifically inhibit membrane fusion mediated by the envelope glycoprotein of primary macrophage-tropic primary viral isolates, not laboratory-adapted T lymphotroohic strains of the virus. Primary macrophage-tropic isolates of the virus are of particular importance since they are the strains usually involved in virus transmission, and may have particular importance in the pathogenesis of HIV-1 infection.
  • RET resonance energy transfer
  • This invention provides a method for inhibiting fusion of HIV-1 to CD4 + cells which comprises contacting CD4 ⁇ cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4 ⁇ cells is inhibited.
  • This invention also provides a method for inhibiting HIV- 1 infection of CD4 + cells which comprises contacting CD4 ⁇ cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4 + cells is inhibited, thereby inhibiting the HIV-1 infection.
  • This invention further provides non-chemokine agents capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4 + cells.
  • agent which is capable of binding to fusin and inhibiting infection.
  • the agent is an oligopeptide.
  • the agent is an polypeptide.
  • the agent is an antibody or a portion of an antibody.
  • the agent is a nonypeptidyl agent.
  • compositions comprising an amount of the above non-chemokine agents or agents capable of binding to fusin effective to inhibit fusion of HIV-1 to CD4 + cells and a pharmaceutically acceptable carrier.
  • This invention provides a composition of matter capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4 ⁇ cells comprising a non-chemokine agent linked to a ligand capable of binding to a cell surface receptor of the CD4 + cells other than the chemokine receptor such that the binding of the non-chemokine agent to the chemokine receptor does not prevent the binding of the ligand to the other receptor.
  • This invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an amount of the above-described composition of matter effective to inhibit fusion of HIV-1 to CD4 ⁇ cells and a pharmaceutically acceptable carrier.
  • This invention provides a composition of matter capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4 + cells comprising a non-chemokine agent linked to a compound capable of increasing the in vivo half-life of the non-chemokine agent.
  • This invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an amount of a composition of matter comprising a non-chemokine agent linked to a compound capable of increasing the in vivo half-life of the non-chemokine agent effective to inhibit fusion of HIV-1 to CD4 + cells and a pharmaceutically acceptable carrier.
  • This invention provide methods for reducing the likelihood of HIV-1 infection in a subject comprising administering an above-described pharmaceutical composition to the subject.
  • This invention also provides methods for treating HIV-1 infection in a subject comprising administering an above-described pharmaceutical composition to the subject.
  • This invention also provides methods for determining whether a non-chemokine agent is capable of inhibiting the fusion of HIV-1 to a CD4 ⁇ cell which comprise: (a) contacting (i) a CD4 + cell which is labeled with a first dye and (ii) a cell expressing the HIV-1 envelope glycoprotein on its surface which is labeled with a second dye, in the presence of an excess of the agent under conditions permitting the fusion of the CD4 + cell to the cell expressing the HIV-1 envelope glycoprotein on its surface in the absence of the agent, the first and second dyes being selected so as to allow resonance energy transfer between the dyes; (b) exposing the product of step (a) to conditions which would result in resonance energy transfer if fusion has occurred; and (c) determining whether there is a reduction of resonance energy transfer, when compared with the resonance energy transfer in the absence of the agent, a decrease in transfer indicating that the agent is capable of inhibiting fusion of HIV-1 to CD4 + cells.
  • FIG. 1 Membrane fusion mediated by the HIV- 1 JR-FL envelope glycoprotein is inhibited by RANTES, MIP-1 ⁇ and MIP-1 ⁇ .
  • FIG. 2 CD4:HIV-1 gp120 binding in the presence of human chemokines.
  • FIG. 3 Specificity. time course and stage of ⁇ -chemokine inhibition of HIV-1 replication.
  • FIG. 4 HIV-1 env-mediated membrane fusion of cells transiently expressing C-C CKR-5.
  • FIG. 5 Membrane fusion mediated by the HIV LAI envelope glycoprotein is inhibited by SDF-1.
  • This invention provides a method for inhibiting fusion of HIV-1 to CD4 + cells which comprises contacting CD4 ⁇ cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4 ⁇ cells is inhibited.
  • This invention also provides a method for inhibiting HIV-1 infection of CD4 ⁇ cells which comprises contacting CD4 + cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4 ⁇ cells is inhibited, thereby inhibiting the HIV-1 infection.
  • a chemokine means RANTES, MIP-1- ⁇ , MIP-1- ⁇ or another chemokine which blocks HIV-1 infection.
  • a chemokine receptor means a receptor capable of binding RANTES, MIP-1- ⁇ , MIP-1- ⁇ or another chemokine which blocks HIV-1 infection.
  • the receptor “fusin” is also named CXCR4 and the chemokine receptor C-C CKR5 is also named CCR5.
  • the HIV-1 used in this application unless specified will mean clinical or primary or field isolates or HIV-1 viruses which maintain their clinical characteristics.
  • the HIV-1 clinical isolates may be passaged in primary peripheral blood mononuclear cells.
  • the HIV-1 clinical isolates may be macrophage-trophic.
  • the non-chemokine agents of this invention are capable of binding to chemokine receptors and inhibiting fusion of HIV-1 to CD4 ⁇ cells.
  • the non-chemokine agents include, but are not limited to, chemokine fragments and chemokine derivatives and analogues, but do not include naturally occurring chemokines.
  • the non-chemokine agents include multimeric forms of the chemokine fragments and chemokine derivatives and analogues or fusion molecules which contain chemokine fragments, derivatives and analogues linked to other molecules.
  • the non-chemokine agent is an oligopeptide. In another embodiment, the non-chemokine agent is a polypeptide. In still another embodiment, the non-chemokine agent is an antibody or a portion thereof. Antibodies against the chemokine receptor may easily be generated by routine experiments. It is also within the level of ordinary skill to synthesize fragments of the antibody capable of binding to the chemokine receptor. In a further embodiment, the non-chemokine agent is a nonpeptidyl agent.
  • Non-chemokine agents which are purely peptidyl in composition can be either chemically synthesized by solid-phase methods (Merrifield, 1966) or produced using recombinant technology in either prokaryotic or eukaryotic systems.
  • the synthetic and recombinant methods are well known in the art.
  • Non-chemokine agents which contain biotin or other nonpeptidyl groups can be prepared by chemical modification of synthetic or recombinant chemokines or non-chemokine agents.
  • One chemical modification method involves periodate oxidation of the 2-amino alcohol present on chemokines or non-chemokine agents possessing serine or threonine as their N-terminal amino acid (Geophegan and Stroh, 1992).
  • the resulting aldehyde group can be used to link peptidyl or non-peptidyl groups to the oxidized chemokine or non-chemokine agent by reductive amination, hydrazine, or other chemistries well known to those skilled in the art.
  • a N-terminus of a protein should mean the terminus of the protein after it has been processed.
  • the N-terminus of a secretory protein should be the terminus after the cleavage of a signal peptide.
  • This invention provides a method of identifying these non-chemokine agents.
  • One way of identifying such agents, including non-peptidyl agents, that bind to a chemokine receptor and inhibit fusion of HIV-1 to CD4 + cells is to use the following assay: 1) Incubate soluble CD4 with biotinylated gp120 from HIV- 1 JR-FL or HIV-1 LAI ; 2) Incubate this complex with CCR5 or CXCR4-expressing cells (for HIV-1 JR-FL or HIV- 1 LAI gp120s, respectively) that do not express CD4, in the presence of absence of a candidate inhibitor; 3) Wash and then incubate with streptavidin-phycoerythrin; and 4) Wash and then measure the amount of bound gp120 using a flow cytometer or fluorometer and calculate the degree of inhibition of binding by the inhibitor.
  • This invention further provides the non-chemokine agents identified by the above methods.
  • the non-chemokine is a polypeptide.
  • this polypeptide is a fragment of the chemokine RANTES (Gong et al., 1996).
  • the polypeptide may also comprise the RANTES sequence with deletion of the N-terminal amino acids of said sequence. The deletion may be the first eight N-terminal amino acids of the RANTES sequence (SEQ ID NO:5).
  • the polypeptide may comprise the MIP-1 ⁇ sequence with deletion of the N-terminal amino acids of said sequence.
  • the deletion may be the first seven, eight, nine or ten N-terminal amino acids of the MIP-1 ⁇ sequence.
  • the polypeptide comprises the MIP-1 ⁇ sequence with the N-terminal sequence modified by addition of an amino acid or oligopeptide.
  • the polypeptide comprises the MIP-1 ⁇ sequence with the N-terminal sequence modified by removing the N-terminal alanine and replaced it by serine or threonine and additional amino acid or oligopeptide or nonpeptidyl moiety.
  • the additional amino acid is methionine.
  • fusin a cofactor for HIV-1 fusion and entry was identified and designated “fusin” (Feng et al., 1996).
  • This invention provides an agent which is capable of binding to fusin and inhibiting infection.
  • the agent is an oligopeptide.
  • the agent is an polypeptide.
  • the polypeptide comprises SD-1 with deletion of the N-terminal amino acids of said sequence.
  • the deletion may be the first six, seven, eight, or nine N-terminal amino acids of the SDF-1 sequence.
  • This invention also provides the above non-chemokine agent, wherein the polypeptide comprises SDF-1 sequence with the N-terminal sequence modified to produce antagonistic effect to SDF-1.
  • the polypeptide comprises SDF-1 sequence with the N-terminal sequence modified to produce antagonistic effect to SDF-1.
  • One modification is to replace the N-terminal glycine of SDF-1 by serine and derivatized with biotin.
  • Another modification is to replace the N-terminal glycine of SDF-1 by serine and derivatized with methionine.
  • a further modification is to add the N-terminus of SDF-1 with a methionine before the terminal glycine.
  • the agent is an antibody or a portion of an antibody.
  • the agent is a nonpeptidyl agent.
  • the agents capable of binding to fusin may be identified by screening different compounds for their capability to bind to fusin in vitro.
  • yeast cells having a pheromone system are engineered to express a heterologous surrogate of a yeast pheromone system protein.
  • the surrogate incorporates fusin and under some conditions performs in the pheromone system of the yeast cell a function naturally performed by the corresponding yeast pheromone system protein.
  • Such yeast cells are also engineered to express a library of peptides whereby a yeast cell containing a peptide which binds fusin exhibits modulation of the interaction of surrogate yeas pheromone system protein with the yeast pheromone system and this modulation is a selectable or screenable event. Similar approaches may be used to identify agents capable of binding to both fusin and the chemokine receptor C-C CKR-5.
  • This invention also provides pharmaceutical compositions comprising an amount of such non-chemokine agents or agents capable of binding to fusin effective to inhibit fusion of HIV-1 to CD4 + cells and a pharmaceutically acceptable carrier.
  • Such pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions.
  • non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate.
  • Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, chelating agents, inert gases and the like.
  • This invention provides a composition of matter capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4 + cells comprising a non-chemokine agent linked to a ligand capable of binding to a cell surface receptor of the CD4 + cells other than the chemokine receptor such that the binding of the non-chemokine agent to the chemokine receptor does not prevent the binding of the ligand to the other receptor.
  • the cell surface receptor is CD4.
  • the ligand is an antibody or a portion of an antibody.
  • This invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an amount of an above-described composition of matter effective to inhibit fusion of HIV-1 to CD4 ⁇ cells and a pharmaceutically acceptable carrier.
  • This invention provides a composition of matter capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4 + cells comprising a non-chemokine agent linked to a compound capable of increasing the in vivo half-life of the non-chemokine agent.
  • the compound is polyethylene glycol.
  • This invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an amount of a composition of matter comprising a non-chemokine agent linked to a compound capable of increasing the in vivo half-life of the non-chemokine agent effective to inhibit fusion of HIV-1 to CD4 + cells and a pharmaceutically acceptable carrier.
  • This invention provide methods for reducing likelihood of HIV-1 infection in a subject comprising administering the above-described pharmaceutical compositions to the subject.
  • This invention also provides methods for treating HIV-1 infection in a subject comprising administering the above-described pharmaceutical compositions to the subject.
  • This invention also provides methods for determining whether a non-chemokine agent is capable of inhibiting the fusion of HIV-1 to a CD4 + cell which comprise: (a) contacting (i) a CD4 + cell which is labeled with a first dye and (ii) a cell expressing the HIV-1 envelope glycoprotein on its surface which is labeled with a second dye, in the presence of an excess of the agent under conditions permitting the fusion of the CD4 + cell to the cell expressing the HIV-1 envelope glycoprotein on its surface in the absence of the agent, the first and second dyes being selected so as to allow resonance energy transfer between the dyes; (b) exposing the product of step (a) to conditions which would result in resonance energy transfer if fusion has occurred; and (c) determining whether there is a reduction of resonance energy transfer, when compared with the resonance energy transfer in the absence of the agent, a decrease in transfer indicating that the agent is capable of inhibiting fusion of HIV-1 to CD4 + cells.
  • HIV-1 only fuses with appropriate CD4 + cells.
  • laboratory-adapted T lymphotropic HIV-1 strains fuse with most CD4 + human cells.
  • Clinical HIV-1 isolates do not fuse with most transformed CD4 + human cell lines but do fuse with human primary CD4 + cells such as CD4 + T lymphocytes and macrophages. Routine experiments may be easily performed to determine whether the CD4 + cell is appropriate for the above fusion assay.
  • HIV-1 membrane fusion is monitored by a resonance energy transfer assay.
  • the assay was described in the International Application Number, PCT/US94/14561, filed Dec. 16, 1994 with International Publication Number WO 95/16789. This assay is further elaborated in a United States co-pending application Ser. No. 08/475,515, filed Jun. 7, 1995. The contents of these applications are hereby incorporated by reference into this application.
  • the non-chemokine agent is an oligopeptide. In another embodiment, the non-chemokine agent is a polypeptide. In still another embodiment, the agent is an antibody or a portion thereof. In a further embodiment, the non-chemokine agent is a nonpeptidyl agent.
  • the CD4 ⁇ cell is a PM1 cell.
  • the cell expressing the HIV-1 envelope glycoprotein is a HeLa cell expressing HIV- 1 JR-FL gp120/gp41.
  • the chemokines RANTES, MIP-1 ⁇ and MIP-1 ⁇ were obtained from R & D systems (Minneapolis, Minn.). They were tested in the RET assay for ability to inhibit fusion between HeLa-env JR-FL cells (expressing gp120/gp41 from the macrophage tropic isolate HIV- 1 JR-FL ) and PM1 cells, or for inhibition of fusion between HeLa-env LAI cells (expressing gp120/gp41 from the laboratory-adapted strain HIV-1 LAI ) and various CD4 + T lymphocyte cell lines. As shown in FIG. 1 , all three chemokines inhibited fusion mediated by the macrophage tropic virus envelope glycoprotein, but not that mediated by the laboratory-adapted strain envelope glycoprotein.
  • chemokines The ability of the chemokines to block the interaction between CD4 and HIV-1 gp120 which occurs at virus attachment was then tested. It was found that the chemokines did not inhibit this interaction ( FIG. 2 ), demonstrating that their blockade of HIV-1 envelope glycoprotein-mediated membrane fusion occurs at the membrane fusion event itself, rather than the initial CD4-gp120 interaction which precedes fusion.
  • the non-chemokines include chemokine fragments and chemokine derivatives that are tested in the RET assay to determine which are active in inhibiting HIV-1 membrane fusion. Particular attention is focused on fragments or derivatives that inhibit HIV-1 fusion but do not activate leukocyte responses. These non-chemokines include:
  • N-terminal derivatives of the chemokines Addition of residues to the N-terminus of chemokines inhibits the function of these proteins without significantly reducing their ability to bind chemokine receptors.
  • Met-RANTES RANTES with an N-terminal methionine
  • the mechanism of antagonism appears to be competition for receptor binding (9).
  • Similar results were found using other derivatives of the N terminus of RANTES (9) and also by N-terminal modification of other chemokines, such as IL-8 (a member of the C-X-C chemokines) (10).
  • the current invention includes Met-RANTES and other chemokines derivatised by the addition of methionine, or other residues, to the N-terminus so that they inhibit fusion mediated by the envelope glycoprotein of HIV-1 JR-FL , and inhibit infection by many isolates of HIV-1, yet do not activate the inflammatory response.
  • Chemokines with N-terminal amino acids deleted have been generated by deleting amino acids in the N-terminal region. For example, deletion of up to 8 amino acids at the N-terminus of the chemokine MCP-1 (a member of the C-C chemokine group), ablated the bioactivity of the protein while allowing it to retain chemokine receptor binding and the ability to inhibit activity of native MCP-1 (11,12).
  • the current invention includes N-terminal deletants of RANTES, MIP-1 ⁇ and MIP-1 ⁇ , lacking the biological activity of the native proteins, which inhibit HIV-1 fusion and HIV-1 infection.
  • peptides A series of overlapping peptides (e.g. o 20-67 residues) from all regions of RANTES, MIP-1 ⁇ and MIP-1 ⁇ are screened by the same approaches to identify peptides which inhibit HIV-1 fusion most potently without activating leukocytes. Activation of leukocyte responses is measured following routine procedures (9, 10, 11, 12).
  • Chemokine receptors required for HIV-1 fusion are cloned by the following strategy.
  • First a cDNA library is made in a mammalian expression vector (e.g. pcDNA3.1 from Invitrogen Corp. San Diego, Calif.) using mRNA prepared from the PM1 cell line or CD4 + T-lymphocytes or macrophages. Degenerate oligonucleotide probes are used to identify members of the cDNA library encoding members of the chemokine receptor family, for example following previously published methods (2).
  • the vectors containing chemokine receptor cDNAs are then individually expressed in one of several mammalian cell lines which express human CD4 but do not fuse with HeLa-env JR-FL , cells (e.g.
  • HeLa-CD4, CHO-CD4 or COS-CD4) or HeLa-env LAI cells e.g. CHO-CD4 or COS-CD4.
  • clones which gain the ability to fuse with HeLa-env JR-FL or HeLa-env LAI are identified and the coding sequences recovered, for example by PCR amplification, following procedures well known to those skilled in the art. DNA sequencing is then performed to determine whether the cDNA recovered encodes a known chemokine receptor.
  • monoclonal and polyclonal antibodies are prepared and tested for ability to inhibit infection by a panel of HIV-1 isolates.
  • N-syncytium-inducing HIV-1 isolates in CD4 ⁇ T-cells are inhibited by the C-C ⁇ -chemokines MIP-1 ⁇ , MIP-1 ⁇ and RANTES (1,2), but T-cell line-adapted (TCLA) or syncytium-inducing (SI) primary strains are insensitive (2,3).
  • the ⁇ -chemokines are small (8 kDa), related proteins active on cells of the lymphoid and monocyte lineage (4-8).
  • Fusin is not known to be a ⁇ -chemokine receptor (7-9).
  • ⁇ -chemokines inhibit HIV-1 replication
  • NL4/3 ⁇ env which also carries the luciferase reporter gene
  • envelope glycoproteins expressed in trans was used (10,11).
  • Various env-complemented viruses were tested in PM1 cells, a variant of HUT-78 that has the unique ability to support replication of primary and TCLA HIV-1 strains, allowing comparison of envelope glycoprotein functions against a common cellular background (2,12).
  • MIP-1 ⁇ , MIP-1 ⁇ and RANTES are most active against HIV-1 in combination (2,3), and strongly inhibited infection of PM1 cells by complemented viruses whose envelopes are derived from the NSI primary strains ADA and BaL (Table 1a).
  • PM1 cells were cultured as described by Lusso et al (12). Ficoll/hypaque-isolated PBMC from laboratory workers (LW) stimulated with PHA for 72 h before depletion of CD8+ Lymphocytes with anti-CD8 immunomagnetic beads (DYNAL, Great Neck, N.Y.). CD4+ Lymphocytes were maintained in culture medium containing interleukin-2 (110 U/ml; Hofmann LaRoche, Nutley, N.J.), as described previously (3).
  • interleukin-2 110 U/ml; Hofmann LaRoche, Nutley, N.J.
  • Target cells (1-2 ⁇ 10 5 ) were infected with supernatants (10-50 ng of HIV-1 p24) from 293-cells co-transfected with an NL4/3 ⁇ env-luciferase vector and a HIV-1 env-expressing vector (10,11).
  • ⁇ -Chemokines R & D Systems, Minneapolis
  • the ⁇ -chemokine concentration range was selected based on prior studies (2,3). After 2 h, the cells were washed twice with PBS, resuspended in ⁇ -chemokine-containing media and maintained for 48-96 h.
  • Luciferase activity in cell lysates was measured as described previously (10,11). The values indicated represent luciferase activity (cpm)/ng p24/mg protein, expressed relative to that in virus-control cultures lacking ⁇ -chemokines (100%), and are the means of duplicate or sextuplicate determinations. nd, not done. R/M ⁇ /M ⁇ , RANTES+MIP-1 ⁇ +MIP-1 ⁇ .
  • RANTES and MIP-1 ⁇ were strongly active when added individually, while other ⁇ -chemokines—MIP-1 ⁇ , MCP-1, MCP-2 and MCP-3 (refs. 13-15)—were weaker inhibitors (Table 1a).
  • MIP-1 ⁇ , MIP-1 ⁇ and RANTES, in combination did not inhibit infection of PM1 cells by the TCLA strains NL4/3 and HxB2, or by the amphotropic murine leukemia virus (MuLV-Ampho) pseudotype (Table 1a).
  • MIP-1 ⁇ , MIP-1 ⁇ and RANTES did not inhibit infection of PM1 cells by the TCLA strains NL4/3 and HxB2, or by the amphotropic murine leukemia virus (MuLV-Ampho) pseudotype (Table 1a).
  • MuLV-Ampho amphotropic murine leukemia virus
  • the env-complementation assay was used to assess HIV-1 entry into CD4+ T-cells from two control individuals (LW4 and LW5).
  • MIP-1 ⁇ , MIP-1 ⁇ and RANTES strongly inhibited infection by the NSI primary strain JR-FL infection of LW4's and LW5's CD4 + T-cells, and weakly reduced HxB2 infection of LW cells (Table 1b), suggesting that there may be some overlap in receptor usage on activated CD4 + T-cells by different virus strains.
  • BaL env-mediated replication in normal PBL was also inhibited by MIP-1 ⁇ , MIP-1 ⁇ and RANTES, albeit with significant inter-donor variation in sensitivity (data not shown).
  • a PCR-based assay was next used to detect HIV-1 early DNA reverse transcripts in PM1 cells after 10 h of infection; reverse transcription of ADA, but not of NL4/3, could not be detected in the presence of MIP-1 ⁇ and RANTES ( FIG. 3 b ).
  • ⁇ -chemokines requires their presence during at least one of the early stages of HIV-1 replication: virus attachment, fusion and early reverse transcription.
  • Cell-cell membrane fusion is also induced by the gp120-CD4 interaction, and can be monitored directly by resonance energy transfer (RET) between fluorescent dyes incorporated into cell membranes (17).
  • RET resonance energy transfer
  • OKT4a completely inhibits membrane fusion of PM1 cells with HeLa cells expressing the envelope glycoproteins of either JR-FL (HeLa-JR-FL, the same cell line referred to above as HeLa-env JR-FL ) or BRU (HeLa-BRU, the same cell line referred to above as HeLa-env LAI ), confirming the specificity of the process (17).
  • MIP-1 ⁇ (and to a lesser extent, MIP-1 ⁇ ) strongly Inhibited membrane fusion of HeLa-JR-FL cells with PM1 cells, whereas fusion between PM1 cells and HeLa-BRU cells was insensitive to these ⁇ -chemokines ( FIG. 1 and Table 2a).
  • CD4 + target cells mitogen-activated CD4 + lymphocytes or PM1 cells
  • octadecyl rhodamine Molecular Probes, Eugene, Oreg.
  • HeLa-JR-FL cells HeLa-BRU cells (or control HeLa cells, not shown) were labeled with octadecyl fluorescein (Molecular Probes), overnight at 37° C.
  • Equal numbers of labeled target cells and env-expressing cells were mixed in 96-well plates and ⁇ -chemokines (or CD4 MAb OKT4a) were added at the final concentrations (ng/ml) indicated in parentheses in the first column. Fluorescence emission values were determined 4 h after cell mixing (17).
  • RET resonance energy transfer
  • the % RET value is defined by a calculation described elsewhere (17), and each is the mean of triplicate determinations. These values were, for HeLa-JR-FL and HeLa-BRU cells respectively: PM1 cells 11.5%, 10.5%; LW5 CD4 + cells, 6.0%, 10.5%; R/M ⁇ /M ⁇ , RANTES+MIP-1 ⁇ +MIP-1 ⁇ .
  • C-C CKR-1 and, especially, C-C CKR-5 were identified as the most likely candidates, based on tissue expression patterns and their abilities to bind MIP-1 ⁇ , MIP-1 ⁇ and RANTES (4, 7, 8, 15, 22).
  • C-C CKR-1, C-C CKR-5 and LESTR are each expressed at the mRNA level in PM1 cells and primary macrophages (data not shown). These and other ⁇ -chemokine receptors were therefore PCR-amplified, cloned and expressed.
  • C-C CKR-5 in HeLa-CD4 (human), COS-CD4 (simian) and 3T3-CD4 (murine) cells rendered each of them readily infectible by the primary, NSI strains ADA and BaL in the env-complementation assay of HIV-1 entry (Table 3) TABLE 3 C—C CKR-5 expression permits infection of CD4-expressing cells by primary, NSI HIV-1 strains R/M ⁇ /M ⁇ pcDNA3.1 LESTR CKR-1 CKR-2a CKR-3 CKR-4 CKR-5 CKR-5 COS-CD4 ADA 798 456 600 816 516 534 153000 3210 BaL 660 378 600 636 516 618 58800 756 HxB2 5800 96700 5240 5070 5470 5620 4850 5000 HeLa-CD4 ADA 678 558 4500 912 558 600 310000 6336 BaL 630 738 1800 654 516 636 104000 750 Hx
  • Chemokine receptor genes C-C CKR-1, C-C CKR-2a, C-C CKR-3, C-C CKR-4 and C-C CKR-5 have no introns (4-8,15,22) and were isolated by PCR performed directly on a human genomic DNA pool derived from the PBMC of seven healthy donors. Oligonucleotides overlapping the ATG and the stop codons and containing BamHI and Xhol restriction sites for directional cloning into the pcDNA3.1 expression vector (Invitrogen Inc.) were used.
  • LESTR also known as fusin or HUMSTR (4,9,24) was cloned by PCR performed directly on cDNA derived from PM1 cells, using sequences derived from the NIH database. Listed below are the 5′ and 3′ primer pairs used in first (5-1 and 3-1) and second (5-2 and 3-2) round PCR amplification of the CKR genes directly from human genomic DNA, and of LESTR from PM1 cDNA. Only a single set of primers was used to amplify CKR-5.
  • the human CD4-expressing cell lines HeLa-CD4 (P42), 3T3-CD4 (sc6) and COS-CD4 (Z28T1) (23) were transfected with the different pcDNA3.1-CKR constructs by the calcium phosphate method, then infected 48 h later with different reporter viruses (200 ng of HIV-1 p24/10 6 cells) in the presence or absence of ⁇ -chemokines (400 ng/ml each of RANTES, MIP-1 ⁇ and MIP-1 ⁇ ). Luciferase activity in cell lysates was measured 48 h later (10,11).
  • ⁇ -Chemokine blocking data is only shown for C-C CKR-5, as infection mediated by the other C-C CKR genes was too weak for inhibition to be quantifiable.
  • PCR-based assays of HIV-1 entry a low level of entry of NL4/3 and ADA into C-C CKR-1 expressing cells (data not shown) was consistently observed.
  • LESTR nor C-C CKR-1, -2a, -3 or -4 could substitute for C-C CKR-5 in this assay.
  • C-C CKR-5 The second receptor function of C-C CKR-5 was confirmed in assays of env-mediated membrane fusion.
  • C-C CKR-5 was transiently expressed in COS and HeLa cell lines that permanently expressed human CD4, both cell lines fused strongly with HeLa cells expressing the JR-FL envelope glycoproteins, whereas no fusion occurred when control plasmids were used (data not shown).
  • Expression of LESTR instead of C-C CKR-5 did not permit either COS-CD4 or HeLa-CD4 cells to fuse with HeLa-JR-FL cells, but did allow fusion between COS-CD4 cells and HeLa-BRU cells (data not shown).
  • the fusion capacity of ⁇ -chemokine receptors was also tested in the RET assay.
  • the extent of fusion between HeLa-JR-FL cells and C-C CKR-5-expressing HeLa-CD4 cells was greater than the constitutive level of fusion between HeLa-BRU cells and HeLa-CD4 cells ( FIG. 4 )
  • the fusion-conferring function of C-C CKR-5 for primary, NSI HIV-1 strains has therefore been confirmed in two independent fusion assays.
  • the chemokine SDF-1 (stromal cell-derived factor 1) is the natural ligand for Fusin/CXCR4 and blocks infection by laboratory-adapted strains of HIV-1 (Ref. 1 and 2).
  • SDF-1 exists as at least two forms, SDF-1 ⁇ and SDF-1 ⁇ based on variable splicing of the SDF-1 gene (Ref. 1 and 3)
  • this chemokine specifically inhibits membrane fusion mediated by gp120/gp41 form the laboratory-adapted strain HIV LAI but not by gp120/gp41 from the macrophage-tropic isolate HIV- 1 JR-FL as shown in FIG. 5 .

Abstract

This invention provides methods for inhibiting fusion of HIV-1 to CD4 cells which comprise contacting CD4 cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4+ cells is inhibited. This invention also provides methods for inhibiting HIV-1 infection of CD4+ cells which comprise contacting CD4+ cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4+ cells is inhibited, thereby inhibiting the HIV-1 infection. This invention provides non-chemokine agents capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4+ cells. This invention also provides pharmaceutical compositions comprising an amount of the non-chemokine agent capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4+ cells effective to prevent fusion of HIV-1 to CD4 + cells and a pharmaceutically acceptable carrier.

Description

  • This application claims priority of U.S. Provisional Application No. 60/019,715, filed Jun. 14, 1996, and U.S. Provisional Application No. 60/014,532, filed Apr. 2, 1996, the content of which are incorporated by reference into this application.
  • Throughout this application, various references are referred to within parentheses. Disclosures of these publications in their entireties are hereby incorporated by reference into this application to more fully describe the state of the art to which this invention pertains. Full bibliographic citation for these references may be found at the end of each series of experiments.
  • BACKGROUND OF THE INVENTION
  • Chemokines are a family of related soluble proteins of molecular weight between 8 and 10 KDa, secreted by lymphocytes and other cells, which bind receptors on target cell surfaces resulting in the activation and mobilization of leukocytes, for example in the inflammatory process. Recently, Cocchi et al. demonstrated that the chemokines RANTES, MIP-1α and MIP-1β are factors produced by CD8+ T lymphocytes which inhibit infection by macrophage-tropic primary isolates of HIV-1, but not infection by laboratory-adapted strains of the virus (1). These chemokines are members of the C-C group of chemokines, so named because they have adjacent cysteine residues, unlike the C-X-C group which has a single amino acid separating these residues (2). While Cocchi et al. found that expression of HIV-1 RNA was suppressed by treatment with the chemokines, they did not identify the site of action of these molecules.
  • A resonance energy transfer (RET) assay of HIV-1 envelope glycoprotein-mediated membrane fusion was used to determine whether fusion mediated by the envelope glycoprotein from the primary macrophage-tropic isolate of HIV-1JR-FL would be specifically inhibited by chemokines, when compared with fusion mediated by the envelope glycoprotein from the laboratory-adapted T lymphotropic strain HIV-1LAI. As described below, it was demonstrated that this is indeed the case. This demonstrates that some chemokine receptors are fusion accessory molecules required for HIV-1 infection. Previous studies have indicated that unidentified cell surface molecules are required for virus entry in addition to the HIV-1 receptor, CD4. While CD4 is required for HIV-1 attachment, the accessory molecules are required for the membrane fusion step of entry. These accessory molecules are generally expressed only on human cells, so HIV-1 does not infect non-human CD4+ cells (3-6). Moreover it is possible to complement non-human CD4+ cells by fusing them (using polyethylene glycol) with CD4 human cells, resulting in a heterokaryon which is a competent target for HIV-1 envelope-mediated membrane fusion (7,8). These studies have been performed using laboratory-adapted T lymphotropic strains of the virus.
  • In some cases, it appears that fusion accessory molecules are found on a subset of human CD4+ cells and are required for infection by HIV-1 isolates with particular tropisms. For example, macrophage-tropic primary strains of HIV-1 such as HIV-1JR-FL may have different requirements for accessory molecules compared with laboratory-adapted T lymphotropic strains such as HIV-1LAI. This phenomenon may explain differences in tropism between HIV-1 strains.
  • The current invention comprises a series of new therapeutics for HIV-1 infection. It was demonstrated for the first time that chemokines act at the fusion step of HIV-1 entry and specifically inhibit membrane fusion mediated by the envelope glycoprotein of primary macrophage-tropic primary viral isolates, not laboratory-adapted T lymphotroohic strains of the virus. Primary macrophage-tropic isolates of the virus are of particular importance since they are the strains usually involved in virus transmission, and may have particular importance in the pathogenesis of HIV-1 infection.
  • These results were obtained using a resonance energy transfer (RET) assay of HIV-1 envelope-mediated membrane fusion. Moreover, this assay is used to identify non-chemokines, including fragments of chemokines and modified chemokines, that inhibit HIV-1 envelope glycoprotein-mediated membrane fusion and thereby neutralize the virus, yet do not induce an inflammatory response.
  • SUMMARY OF THE INVENTION
  • This invention provides a method for inhibiting fusion of HIV-1 to CD4+ cells which comprises contacting CD4 cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4 cells is inhibited.
  • This invention also provides a method for inhibiting HIV-1 infection of CD4+ cells which comprises contacting CD4 cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4+ cells is inhibited, thereby inhibiting the HIV-1 infection.
  • This invention further provides non-chemokine agents capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4+ cells.
  • This invention provides an agent which is capable of binding to fusin and inhibiting infection. In an embodiment, the agent is an oligopeptide. In another embodiment, the agent is an polypeptide. In still another embodiment, the agent is an antibody or a portion of an antibody. In a separate embodiment, the agent is a nonypeptidyl agent.
  • In addition, this invention provides pharmaceutical compositions comprising an amount of the above non-chemokine agents or agents capable of binding to fusin effective to inhibit fusion of HIV-1 to CD4+ cells and a pharmaceutically acceptable carrier.
  • This invention provides a composition of matter capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4 cells comprising a non-chemokine agent linked to a ligand capable of binding to a cell surface receptor of the CD4+ cells other than the chemokine receptor such that the binding of the non-chemokine agent to the chemokine receptor does not prevent the binding of the ligand to the other receptor.
  • This invention also provides a pharmaceutical composition comprising an amount of the above-described composition of matter effective to inhibit fusion of HIV-1 to CD4 cells and a pharmaceutically acceptable carrier.
  • This invention provides a composition of matter capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4+ cells comprising a non-chemokine agent linked to a compound capable of increasing the in vivo half-life of the non-chemokine agent.
  • This invention also provides a pharmaceutical composition comprising an amount of a composition of matter comprising a non-chemokine agent linked to a compound capable of increasing the in vivo half-life of the non-chemokine agent effective to inhibit fusion of HIV-1 to CD4+ cells and a pharmaceutically acceptable carrier.
  • This invention provide methods for reducing the likelihood of HIV-1 infection in a subject comprising administering an above-described pharmaceutical composition to the subject. This invention also provides methods for treating HIV-1 infection in a subject comprising administering an above-described pharmaceutical composition to the subject.
  • This invention also provides methods for determining whether a non-chemokine agent is capable of inhibiting the fusion of HIV-1 to a CD4 cell which comprise: (a) contacting (i) a CD4+ cell which is labeled with a first dye and (ii) a cell expressing the HIV-1 envelope glycoprotein on its surface which is labeled with a second dye, in the presence of an excess of the agent under conditions permitting the fusion of the CD4+ cell to the cell expressing the HIV-1 envelope glycoprotein on its surface in the absence of the agent, the first and second dyes being selected so as to allow resonance energy transfer between the dyes; (b) exposing the product of step (a) to conditions which would result in resonance energy transfer if fusion has occurred; and (c) determining whether there is a reduction of resonance energy transfer, when compared with the resonance energy transfer in the absence of the agent, a decrease in transfer indicating that the agent is capable of inhibiting fusion of HIV-1 to CD4+ cells.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. Membrane fusion mediated by the HIV-1 JR-FL envelope glycoprotein is inhibited by RANTES, MIP-1α and MIP-1β.
      • FRET resulting from the fusion of PM1 cells and HeLa-envJR-FL (▪) or HeLa-envLAI(♦) was measured in the presence and absence of recombinant human chemokines at a range of concentrations: RANTES (80-2.5 ng/ml), MIP-1α (400-12.5 ng/ml) and MIP-1β (200-6.25 ng/ml), as indicated. Chemokines were added simultaneously with the cells at the initiation of a four hour incubation. Data are representative of more than three independent experiments which were run in duplicate. The percent inhibition of RET is defined as follows:
        % Inhibition=100.[(Max RET−Min RET)−(Exp RET−Min RET)]/(Max RET−Min RET)
      • where Max RET is the % RET value obtained at four hours with HeLa-env cells and CD4-expressing cells in the absence of an inhibitory compound; Exp RET is the FRET value obtained for the same cell combination in the presence of an inhibitory compound and Min RET is the background FRET value obtained using HeLa cells in place of HeLa envelope-expressing cells.
  • FIG. 2. CD4:HIV-1 gp120 binding in the presence of human chemokines.
      • The binding of soluble human CD4 to HIV-1LAI and HIV-1JR-FL gp120 was determined in an ELISA assay in the presence and absence of the monoclonal antibody OKT4A or recombinant human chemokines at a range of concentrations, identical to those used in the RET inhibition studies of FIG. 1: OKT4A (62-0.3 nM), RANTES (10.3-0.3 nM), MIP-1a (53.3-2.9 nM), and MIP-1β (25.6-0.8 nM). Inhibitors were added simultaneously with biotinylated HIV-1 gp120 to soluble CD4 coated microtiter plates (Dynatech Laboratories, Inc., Chantilly, Va.). Following a two hour incubation at room temperature and extensive washing, an incubation with streptavidin-horseradish peroxidase was performed for one hour at room temperature. Following additional washes, substrate was added and the OD at 492 nm determined in an ELISA plate reader. Data are representative of two independent experiments which were run in quadruplicate.
  • FIG. 3. Specificity. time course and stage of β-chemokine inhibition of HIV-1 replication.
      • (a) PM1 cells (1×106) were preincubated with RANTES+MIP-1α+MIP-1β (R/Mα/Mβ; 100 ng/ml of each) for 24 h (−24 h) or 2 h (−2 h), then washed twice with phosphate buffered saline (PBS). HIV-1 (BaL env-complemented) virus (50 ng of p24; see legend to Table 1) was added for 2 h, then the cells were washed and incubated for 48 h before measurement of luciferase activity in cell lysates as described previously (10,11). Alternatively, virus and R/Mα/Mβ were added simultaneously to cells, and at the indicated time points (1 h, 3 h, etc) the cells were washed twice in PBS, resuspended in culture medium and incubated for 48 h prior to luciferase assay. Time 0 represents the positive control, to which no β-chemokines were added. +2 h represents the mixture of virus with cells for 2 h prior to washing twice in PBS, addition of R/Mα/Mβ and continuation of the culture for a further 48 h before luciferase assay.
      • (b) PM1 cells (1×106) were infected with HIV-1 (500 pg p24) grown in CEM cells (NL4/3; lanes 1-4) or macrophages (ADA; lanes 5-8), in the presence of 500 ng/ml of RANTES (lanes 1 and 5) or MIP-1β (lanes 2 and 6), or with no β-chemokine (lanes 4 and 8). Lanes 3 and 7 are negative controls (no virus). All viral stocks used for the PCR assay were treated with DNAse for 30 min at 37° C., and tested for DNA contamination before use. After 2 h, the cells were washed and resuspended in medium containing the same S-chemokines for a further 8 h. DNA was then extracted from infected cells using a DNA/RNA isolation kit (US Biochemicals). First round nested PCR was performed with primers: U3+, 5′-CAAGGCTACTTCCCTGATTGGCAGAACTACACACCAGG-3′ (SEQ ID NO:1) preGag, 5′-AGCAAGCCGAGTCCTGCGTCGAGAG-3′ (SEQ ID NO:2) and the second round with primers: LTR-test, 5′-GGGACTTTCCGCTGGGGACTTTC 3′ (SEQ ID NO:3) LRC2, 5′-CCTGTTCGGGCGCCACTGCTAGAGATTTTCCAC 3′ (SEQ ID NO:4) in a Perkin Elmer 2400 cycler with the following amplification cycles: 94° C. for 5 min, 35 cycles of 94° C. for 30 s, 55° C. for 30 s, 72° C. for 30 s, 72° C. for 7 min. M indicates 1 kb DNA ladder; 1, 10, 100, 1000 indicate number of reference plasmid (pAD8) copies. The assay can detect 100 copies of reverse transcripts.
  • FIG. 4: HIV-1 env-mediated membrane fusion of cells transiently expressing C-C CKR-5.
      • Membrane fusion mediated by β-chemokine receptors expressed in HeLa cells was demonstrated as follows: Cells were transfected with control plasmid pcDNA3.1 or plasmid pcDNA3.1-CKR constructs using lipofectin (Gibco BRL). The pcDNA3.1 plasmid carries a T7-polymerase promoter and transient expression of β-chemokine receptors was boosted by infecting cells with 1×107 pfu of vaccinia encoding the T7-polymerase (vFT7.3) 4 h post-lipofection (9). Cells were then cultured overnight in R18-containing media and were tested for their ability to fuse with HeLa-JR-FL cells (filled columns) or HeLa-BRU cells (hatched column) in the RET assay. The % RET with control HeLa cells was between 3% and 4% irrespective of the transfected plasmid.
  • FIG. 5 Membrane fusion mediated by the HIVLAI envelope glycoprotein is inhibited by SDF-1.
      • % RET resulting from the fusion of PM1 cells and HeLa-envJR-FL or HeLa-envLAI cells (as indicated on the graph) was measured in the presence of recombinant SDF-1α (Gryphon Science, San Francisco) at the indicated concentrations. Experimental method as described in the legend to FIG. 1.
    DETAILED DESCRIPTION OF THE INVENTION
  • This invention provides a method for inhibiting fusion of HIV-1 to CD4+ cells which comprises contacting CD4 cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4 cells is inhibited.
  • This invention also provides a method for inhibiting HIV-1 infection of CD4 cells which comprises contacting CD4+ cells with a non-chemokine agent capable of binding to a chemokine receptor in an amount and under conditions such that fusion of HIV-1 to the CD4 cells is inhibited, thereby inhibiting the HIV-1 infection.
  • In this invention, a chemokine means RANTES, MIP-1-α, MIP-1-β or another chemokine which blocks HIV-1 infection. A chemokine receptor means a receptor capable of binding RANTES, MIP-1-α, MIP-1-β or another chemokine which blocks HIV-1 infection.
  • Throughout this application, the receptor “fusin” is also named CXCR4 and the chemokine receptor C-C CKR5 is also named CCR5.
  • The HIV-1 used in this application unless specified will mean clinical or primary or field isolates or HIV-1 viruses which maintain their clinical characteristics. The HIV-1 clinical isolates may be passaged in primary peripheral blood mononuclear cells. The HIV-1 clinical isolates may be macrophage-trophic.
  • The non-chemokine agents of this invention are capable of binding to chemokine receptors and inhibiting fusion of HIV-1 to CD4 cells. The non-chemokine agents include, but are not limited to, chemokine fragments and chemokine derivatives and analogues, but do not include naturally occurring chemokines. The non-chemokine agents include multimeric forms of the chemokine fragments and chemokine derivatives and analogues or fusion molecules which contain chemokine fragments, derivatives and analogues linked to other molecules.
  • In an embodiment of this invention, the non-chemokine agent is an oligopeptide. In another embodiment, the non-chemokine agent is a polypeptide. In still another embodiment, the non-chemokine agent is an antibody or a portion thereof. Antibodies against the chemokine receptor may easily be generated by routine experiments. It is also within the level of ordinary skill to synthesize fragments of the antibody capable of binding to the chemokine receptor. In a further embodiment, the non-chemokine agent is a nonpeptidyl agent.
  • Non-chemokine agents which are purely peptidyl in composition can be either chemically synthesized by solid-phase methods (Merrifield, 1966) or produced using recombinant technology in either prokaryotic or eukaryotic systems. The synthetic and recombinant methods are well known in the art.
  • Non-chemokine agents which contain biotin or other nonpeptidyl groups can be prepared by chemical modification of synthetic or recombinant chemokines or non-chemokine agents. One chemical modification method involves periodate oxidation of the 2-amino alcohol present on chemokines or non-chemokine agents possessing serine or threonine as their N-terminal amino acid (Geophegan and Stroh, 1992). The resulting aldehyde group can be used to link peptidyl or non-peptidyl groups to the oxidized chemokine or non-chemokine agent by reductive amination, hydrazine, or other chemistries well known to those skilled in the art.
  • As used herein, a N-terminus of a protein should mean the terminus of the protein after it has been processed. In case of a secretory protein which contains a cleavable signal sequence, the N-terminus of a secretory protein should be the terminus after the cleavage of a signal peptide.
  • This invention provides a method of identifying these non-chemokine agents. One way of identifying such agents, including non-peptidyl agents, that bind to a chemokine receptor and inhibit fusion of HIV-1 to CD4+ cells is to use the following assay: 1) Incubate soluble CD4 with biotinylated gp120 from HIV-1 JR-FL or HIV-1LAI; 2) Incubate this complex with CCR5 or CXCR4-expressing cells (for HIV-1JR-FL or HIV-1 LAI gp120s, respectively) that do not express CD4, in the presence of absence of a candidate inhibitor; 3) Wash and then incubate with streptavidin-phycoerythrin; and 4) Wash and then measure the amount of bound gp120 using a flow cytometer or fluorometer and calculate the degree of inhibition of binding by the inhibitor.
  • Alternative methods to detect bound gp120 can also be used in place of the biotinylated gp120-streptavidin-phycoerythrin method described above. For example, peroxidase-conjugated gp120 could be used in place of the biotinylated gp120 and binding detected using an appropriate colorimetric substrate for peroxidase, with a spectrometric readout.
  • This invention further provides the non-chemokine agents identified by the above methods.
  • This invention provides a non-chemokine agent capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4+ cells. In an embodiment, the non-chemokine is a polypeptide. In a further embodiment, this polypeptide is a fragment of the chemokine RANTES (Gong et al., 1996). In a still further embodiment, the polypeptide may also comprise the RANTES sequence with deletion of the N-terminal amino acids of said sequence. The deletion may be the first eight N-terminal amino acids of the RANTES sequence (SEQ ID NO:5).
  • In a separate embodiment, the polypeptide may comprise the MIP-1β sequence with deletion of the N-terminal amino acids of said sequence. The deletion may be the first seven, eight, nine or ten N-terminal amino acids of the MIP-1β sequence.
  • In another embodiment of non-chemokine agent, the polypeptide comprises the MIP-1β sequence with the N-terminal sequence modified by addition of an amino acid or oligopeptide. In a separate embodiment, the polypeptide comprises the MIP-1β sequence with the N-terminal sequence modified by removing the N-terminal alanine and replaced it by serine or threonine and additional amino acid or oligopeptide or nonpeptidyl moiety. In a further embodiment, the additional amino acid is methionine.
  • As described infra in the section of Experimental Details, a cofactor for HIV-1 fusion and entry was identified and designated “fusin” (Feng et al., 1996). This invention provides an agent which is capable of binding to fusin and inhibiting infection. In an embodiment, the agent is an oligopeptide. In another embodiment, the agent is an polypeptide.
  • In a further embodiment, the polypeptide comprises SD-1 with deletion of the N-terminal amino acids of said sequence. The deletion may be the first six, seven, eight, or nine N-terminal amino acids of the SDF-1 sequence.
  • This invention also provides the above non-chemokine agent, wherein the polypeptide comprises SDF-1 sequence with the N-terminal sequence modified to produce antagonistic effect to SDF-1. One modification is to replace the N-terminal glycine of SDF-1 by serine and derivatized with biotin. Another modification is to replace the N-terminal glycine of SDF-1 by serine and derivatized with methionine. A further modification is to add the N-terminus of SDF-1 with a methionine before the terminal glycine.
  • In still another embodiment, the agent is an antibody or a portion of an antibody. In a separate embodiment, the agent is a nonpeptidyl agent.
  • The agents capable of binding to fusin may be identified by screening different compounds for their capability to bind to fusin in vitro.
  • A suitable method has been described by Fowlkes, et al. (1994), international application number: PCT/US94/03143, international publication number: WO 94/23025, the content of which is incorporated by reference into this application. Briefly, yeast cells having a pheromone system are engineered to express a heterologous surrogate of a yeast pheromone system protein. The surrogate incorporates fusin and under some conditions performs in the pheromone system of the yeast cell a function naturally performed by the corresponding yeast pheromone system protein. Such yeast cells are also engineered to express a library of peptides whereby a yeast cell containing a peptide which binds fusin exhibits modulation of the interaction of surrogate yeas pheromone system protein with the yeast pheromone system and this modulation is a selectable or screenable event. Similar approaches may be used to identify agents capable of binding to both fusin and the chemokine receptor C-C CKR-5.
  • This invention also provides pharmaceutical compositions comprising an amount of such non-chemokine agents or agents capable of binding to fusin effective to inhibit fusion of HIV-1 to CD4+ cells and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are well known to those skilled in the art. Such pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, saline and buffered media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, chelating agents, inert gases and the like.
  • This invention provides a composition of matter capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4+ cells comprising a non-chemokine agent linked to a ligand capable of binding to a cell surface receptor of the CD4+ cells other than the chemokine receptor such that the binding of the non-chemokine agent to the chemokine receptor does not prevent the binding of the ligand to the other receptor. In an embodiment, the cell surface receptor is CD4. In another embodiment, the ligand is an antibody or a portion of an antibody.
  • This invention also provides a pharmaceutical composition comprising an amount of an above-described composition of matter effective to inhibit fusion of HIV-1 to CD4 cells and a pharmaceutically acceptable carrier.
  • This invention provides a composition of matter capable of binding to the chemokine receptor and inhibiting fusion of HIV-1 to CD4+ cells comprising a non-chemokine agent linked to a compound capable of increasing the in vivo half-life of the non-chemokine agent. In an embodiment, the compound is polyethylene glycol.
  • This invention also provides a pharmaceutical composition comprising an amount of a composition of matter comprising a non-chemokine agent linked to a compound capable of increasing the in vivo half-life of the non-chemokine agent effective to inhibit fusion of HIV-1 to CD4+ cells and a pharmaceutically acceptable carrier.
  • This invention provide methods for reducing likelihood of HIV-1 infection in a subject comprising administering the above-described pharmaceutical compositions to the subject. This invention also provides methods for treating HIV-1 infection in a subject comprising administering the above-described pharmaceutical compositions to the subject.
  • This invention also provides methods for determining whether a non-chemokine agent is capable of inhibiting the fusion of HIV-1 to a CD4+ cell which comprise: (a) contacting (i) a CD4+ cell which is labeled with a first dye and (ii) a cell expressing the HIV-1 envelope glycoprotein on its surface which is labeled with a second dye, in the presence of an excess of the agent under conditions permitting the fusion of the CD4+ cell to the cell expressing the HIV-1 envelope glycoprotein on its surface in the absence of the agent, the first and second dyes being selected so as to allow resonance energy transfer between the dyes; (b) exposing the product of step (a) to conditions which would result in resonance energy transfer if fusion has occurred; and (c) determining whether there is a reduction of resonance energy transfer, when compared with the resonance energy transfer in the absence of the agent, a decrease in transfer indicating that the agent is capable of inhibiting fusion of HIV-1 to CD4+ cells.
  • HIV-1 only fuses with appropriate CD4+ cells. For example, laboratory-adapted T lymphotropic HIV-1 strains fuse with most CD4+ human cells. Clinical HIV-1 isolates do not fuse with most transformed CD4+ human cell lines but do fuse with human primary CD4+ cells such as CD4+ T lymphocytes and macrophages. Routine experiments may be easily performed to determine whether the CD4+ cell is appropriate for the above fusion assay.
  • As described in this invention, HIV-1 membrane fusion is monitored by a resonance energy transfer assay. The assay was described in the International Application Number, PCT/US94/14561, filed Dec. 16, 1994 with International Publication Number WO 95/16789. This assay is further elaborated in a United States co-pending application Ser. No. 08/475,515, filed Jun. 7, 1995. The contents of these applications are hereby incorporated by reference into this application.
  • In an embodiment of the above method, the non-chemokine agent is an oligopeptide. In another embodiment, the non-chemokine agent is a polypeptide. In still another embodiment, the agent is an antibody or a portion thereof. In a further embodiment, the non-chemokine agent is a nonpeptidyl agent.
  • In a separate embodiment, the CD4 cell is a PM1 cell. In another embodiment, the cell expressing the HIV-1 envelope glycoprotein is a HeLa cell expressing HIV-1 JR-FL gp120/gp41.
  • This invention will be better understood by reference to the Experimental Details which follow, but those skilled in the art will readily appreciate that the specific experiments detailed are only illustrative of the invention as described more fully in the claims which follow thereafter.
  • Experimental Details
  • First Series of Experiments
  • 1) Chemokines Inhibit Fusion Mediated by the Envelope Glycoprotein from a Macrophage-Tropic Primary Isolate of HIV-1 but not from a Laboratory-Adapted-T-Lymphotrophic Strain of the Virus
  • The chemokines RANTES, MIP-1α and MIP-1β were obtained from R & D systems (Minneapolis, Minn.). They were tested in the RET assay for ability to inhibit fusion between HeLa-envJR-FL cells (expressing gp120/gp41 from the macrophage tropic isolate HIV-1 JR-FL) and PM1 cells, or for inhibition of fusion between HeLa-envLAI cells (expressing gp120/gp41 from the laboratory-adapted strain HIV-1LAI) and various CD4+ T lymphocyte cell lines. As shown in FIG. 1, all three chemokines inhibited fusion mediated by the macrophage tropic virus envelope glycoprotein, but not that mediated by the laboratory-adapted strain envelope glycoprotein.
  • The ability of the chemokines to block the interaction between CD4 and HIV-1 gp120 which occurs at virus attachment was then tested. It was found that the chemokines did not inhibit this interaction (FIG. 2), demonstrating that their blockade of HIV-1 envelope glycoprotein-mediated membrane fusion occurs at the membrane fusion event itself, rather than the initial CD4-gp120 interaction which precedes fusion.
  • 2) Non-Chemokine Peptides and Derivatives that Inhibit HIV-1 Fusion
  • The non-chemokines include chemokine fragments and chemokine derivatives that are tested in the RET assay to determine which are active in inhibiting HIV-1 membrane fusion. Particular attention is focused on fragments or derivatives that inhibit HIV-1 fusion but do not activate leukocyte responses. These non-chemokines include:
  • a) N-terminal derivatives of the chemokines. Addition of residues to the N-terminus of chemokines inhibits the function of these proteins without significantly reducing their ability to bind chemokine receptors. For example, Met-RANTES (RANTES with an N-terminal methionine) has been shown to be a powerful antagonist of native RANTES and is unable to induce chemotaxis or calcium mobilization in certain systems. The mechanism of antagonism appears to be competition for receptor binding (9). Similar results were found using other derivatives of the N terminus of RANTES (9) and also by N-terminal modification of other chemokines, such as IL-8 (a member of the C-X-C chemokines) (10). The current invention includes Met-RANTES and other chemokines derivatised by the addition of methionine, or other residues, to the N-terminus so that they inhibit fusion mediated by the envelope glycoprotein of HIV-1JR-FL, and inhibit infection by many isolates of HIV-1, yet do not activate the inflammatory response.
  • b) Chemokines with N-terminal amino acids deleted: Chemokine antagonists have been generated by deleting amino acids in the N-terminal region. For example, deletion of up to 8 amino acids at the N-terminus of the chemokine MCP-1 (a member of the C-C chemokine group), ablated the bioactivity of the protein while allowing it to retain chemokine receptor binding and the ability to inhibit activity of native MCP-1 (11,12).
  • The current invention includes N-terminal deletants of RANTES, MIP-1α and MIP-1β, lacking the biological activity of the native proteins, which inhibit HIV-1 fusion and HIV-1 infection.
  • c) Other peptides: A series of overlapping peptides (e.g. o 20-67 residues) from all regions of RANTES, MIP-1α and MIP-1β are screened by the same approaches to identify peptides which inhibit HIV-1 fusion most potently without activating leukocytes. Activation of leukocyte responses is measured following routine procedures (9, 10, 11, 12).
  • 3) Cloning the Chemokine Receptors
  • Chemokine receptors required for HIV-1 fusion are cloned by the following strategy. First a cDNA library is made in a mammalian expression vector (e.g. pcDNA3.1 from Invitrogen Corp. San Diego, Calif.) using mRNA prepared from the PM1 cell line or CD4+ T-lymphocytes or macrophages. Degenerate oligonucleotide probes are used to identify members of the cDNA library encoding members of the chemokine receptor family, for example following previously published methods (2). The vectors containing chemokine receptor cDNAs are then individually expressed in one of several mammalian cell lines which express human CD4 but do not fuse with HeLa-envJR-FL, cells (e.g. HeLa-CD4, CHO-CD4 or COS-CD4) or HeLa-envLAI cells (e.g. CHO-CD4 or COS-CD4). Following analysis in the RET assay, clones which gain the ability to fuse with HeLa-envJR-FL or HeLa-envLAI are identified and the coding sequences recovered, for example by PCR amplification, following procedures well known to those skilled in the art. DNA sequencing is then performed to determine whether the cDNA recovered encodes a known chemokine receptor. Following expression of the receptor, monoclonal and polyclonal antibodies are prepared and tested for ability to inhibit infection by a panel of HIV-1 isolates.
  • REFERENCES OF THE FIRST SERIES OF EXPERIMENTS
    • 1. Cocchi, F., DeVico, A. L., Garzino-Demo, A., Arya, S. K., Gallo, R. C., Lusso, P. 1995. Science. 270:181-1815.
    • 2. Raport, C. J., Schweickart, V. L., Chantry, D., Eddy Jr., R. L., Shows, T. B., Godiska, R., Gray, P. W. 1996. Journal of Leukocyte Biology. 59: 18-23.
    • 3. Maddon P J., Dalgleish A G., McDougal J S., Clapham P R., Weiss R A., Axel R. 1986. Cell. 47:333-348.
    • 4. Ashorn P A., Berger E A., Moss B. 1990. J. Virol. 64:2149-2156.
    • 5. Clapham P R., Blanc D., Weiss R A. 1991. Virology. 181:703-715.
    • 6. Harrington R D., Geballe A P. 1993. J. Virol. 67:5939-5947.
    • 7. Broder C C., Dimitrov D S., Blumenthal R., Berger E A. 1993. Virology. 193:483-491.
    • 8. Dragic T., Charneau P., Clavel F., Alizon M. 1992. J. Virol. 66:4794-4802.
    • 9. Wells, T. N., Power, C. A., Lusti-Narasimhan, M., Hoogewerf, A. J., Cooke, R. M., Chung, C. W., Peitsch, M. C., Proudfoot, A. E. 1996. Journal of Leukocyte Biology. 59:53-60.
    • 10. Moser, B., Dewald, B., Barella, L., Schumacher, C., Baggiolini, M., Clark-Lewis, I. 1993. Journal of Biological Chemistry. 268:7125-7128.
    • 11. Gong, J. H., Clark-Lewis, I. 1995. J. Exp. Med. 181:631-640.
    • 12. Zhang, Y. J., Rutledge, B. J., Rollins, B. J. 1994. Journal of Biological Chemistry. 269:15918-15924.
    • 13. Merrifield, R. B. (1963) J. Am. Chem. Soc. 85: 2149-2154.
    • 14. Goeghegan, K. F. Stroh, J. F. (1992) Bioconjugate Chem. 3: 138-146.
      Second Series of Experiments
  • The replication of primary, non-syncytium-inducing (NSI) HIV-1 isolates in CD4 T-cells is inhibited by the C-C β-chemokines MIP-1α, MIP-1β and RANTES (1,2), but T-cell line-adapted (TCLA) or syncytium-inducing (SI) primary strains are insensitive (2,3). The β-chemokines are small (8 kDa), related proteins active on cells of the lymphoid and monocyte lineage (4-8). Their receptors are members of the 7-membrane-spanning, G-protein-linked superfamily, one or which (the LESTR orphan receptor) has been identified as the second receptor for TCLA HIV-1 strains, and is now designated fusin (9). Fusin is not known to be a β-chemokine receptor (7-9).
  • To study how β-chemokines inhibit HIV-1 replication, a virus entry assay based on single-cycle infection by an env-deficient virus, NL4/3Δenv (which also carries the luciferase reporter gene), complemented by envelope glycoproteins expressed in trans was used (10,11). Various env-complemented viruses were tested in PM1 cells, a variant of HUT-78 that has the unique ability to support replication of primary and TCLA HIV-1 strains, allowing comparison of envelope glycoprotein functions against a common cellular background (2,12). MIP-1α, MIP-1β and RANTES are most active against HIV-1 in combination (2,3), and strongly inhibited infection of PM1 cells by complemented viruses whose envelopes are derived from the NSI primary strains ADA and BaL (Table 1a).
    TABLE 1
    Inhibition of HIV-1 entry in PM1 cells
    and CD4+ T-cells by β-chemokines
    a) % luciferase activity
    PM1 cells BaL ADA NL4/3 HxB2 MuLV
    control without virus 2 2 2 5 3
    control with virus 100 100 100 100 100
    +R/Mα/Mβ (50/50/50) 2 3 92 117 100
    +RANTES (100) 1 1 nd nd nd
    +MIP-1α (100) 54 54 nd nd nd
    +MIP-1β (100) 1 6 nd nd nd
    +MCP-1 (100) 46 50 nd nd nd
    +MCP-2 (100) 28 26 nd nd nd
    +MCP-3 (100) 58 46 nd nd nd
    b) JR-FL HxB2 MuLV
    LW4 CD4+ T-cells
    control without virus 1 1 1
    control with virus 100 100 100
    +R/Mα/Mβ (200/200/200) 14 68 nd
    LW5 CD4+ T-cells
    control without virus 1 1 1
    control with virus 100 100 100
    +R/Mα/Mβ (200/200/200) 15 73 nd

    Table 1 Legend:
  • PM1 cells were cultured as described by Lusso et al (12). Ficoll/hypaque-isolated PBMC from laboratory workers (LW) stimulated with PHA for 72 h before depletion of CD8+ Lymphocytes with anti-CD8 immunomagnetic beads (DYNAL, Great Neck, N.Y.). CD4+ Lymphocytes were maintained in culture medium containing interleukin-2 (110 U/ml; Hofmann LaRoche, Nutley, N.J.), as described previously (3). Target cells (1-2×105) were infected with supernatants (10-50 ng of HIV-1 p24) from 293-cells co-transfected with an NL4/3Δenv-luciferase vector and a HIV-1 env-expressing vector (10,11). β-Chemokines (R & D Systems, Minneapolis) were added to the target cells simultaneously with virus, at the final concentrations (ng/ml) indicated in parentheses in the first column. The β-chemokine concentration range was selected based on prior studies (2,3). After 2 h, the cells were washed twice with PBS, resuspended in β-chemokine-containing media and maintained for 48-96 h. Luciferase activity in cell lysates was measured as described previously (10,11). The values indicated represent luciferase activity (cpm)/ng p24/mg protein, expressed relative to that in virus-control cultures lacking β-chemokines (100%), and are the means of duplicate or sextuplicate determinations. nd, not done. R/Mα/Mβ, RANTES+MIP-1α+MIP-1β.
  • RANTES and MIP-1β were strongly active when added individually, while other β-chemokines—MIP-1α, MCP-1, MCP-2 and MCP-3 (refs. 13-15)—were weaker inhibitors (Table 1a). However, MIP-1α, MIP-1β and RANTES, in combination, did not inhibit infection of PM1 cells by the TCLA strains NL4/3 and HxB2, or by the amphotropic murine leukemia virus (MuLV-Ampho) pseudotype (Table 1a). Thus, phenotypic characteristics of the HIV-1 envelope glycoproteins influence their sensitivity to β-chemokines in a virus entry assay.
  • The env-complementation assay was used to assess HIV-1 entry into CD4+ T-cells from two control individuals (LW4 and LW5). MIP-1α, MIP-1β and RANTES strongly inhibited infection by the NSI primary strain JR-FL infection of LW4's and LW5's CD4+ T-cells, and weakly reduced HxB2 infection of LW cells (Table 1b), suggesting that there may be some overlap in receptor usage on activated CD4+ T-cells by different virus strains. BaL env-mediated replication in normal PBL was also inhibited by MIP-1α, MIP-1β and RANTES, albeit with significant inter-donor variation in sensitivity (data not shown).
  • It was determined when β-chemokines inhibited HIV-1 replication by showing that complete inhibition of infection of PM1 cells required the continuous presence of β-chemokines for up to 5 h after addition of ADA or BaL env-complemented virus (FIG. 3 a). Pre-treatment of the cells with β-chemokines for 2 h or 24 h prior to infection had no inhibitory effect if the cells were subsequently washed before virus addition. Furthermore, adding β-chemokines 2 h after virus only minimally affected virus entry (FIG. 3 a). A PCR-based assay was next used to detect HIV-1 early DNA reverse transcripts in PM1 cells after 10 h of infection; reverse transcription of ADA, but not of NL4/3, could not be detected in the presence of MIP-1β and RANTES (FIG. 3 b). Thus, inhibition by β-chemokines requires their presence during at least one of the early stages of HIV-1 replication: virus attachment, fusion and early reverse transcription.
  • As described in part in the First Series of Experiments, these sites of action were discriminated, first by testing whether β-chemokines inhibited binding of JR-FL or BRU (LAI) gp120 to soluble CD4, or of tetrameric CD4-IgG2 binding to HeLa-JR-FL cells expressing oligomeric envelope glycoproteins (17). No inhibition by any of the β-chemokines was found in either assay, whereas the OKT4a CD4-MAb was strongly inhibitory in both (FIG. 2 and data not shown). Thus, β-chemokines inhibit a step after CD4 binding, when conformational changes in the envelope glycoproteins lead to fusion of the viral and cellular membranes (18). Cell-cell membrane fusion is also induced by the gp120-CD4 interaction, and can be monitored directly by resonance energy transfer (RET) between fluorescent dyes incorporated into cell membranes (17). In the RET assay, OKT4a completely inhibits membrane fusion of PM1 cells with HeLa cells expressing the envelope glycoproteins of either JR-FL (HeLa-JR-FL, the same cell line referred to above as HeLa-envJR-FL) or BRU (HeLa-BRU, the same cell line referred to above as HeLa-envLAI), confirming the specificity of the process (17). RANTES, MIP-1β (and to a lesser extent, MIP-1α) strongly Inhibited membrane fusion of HeLa-JR-FL cells with PM1 cells, whereas fusion between PM1 cells and HeLa-BRU cells was insensitive to these β-chemokines (FIG. 1 and Table 2a).
    TABLE 2
    Effect of β-chemokines on HIV-1 envelope glycoprotein-mediated
    membrane fusion measured using the RET assay
    % Fusion
    HeLa-JR-FL HeLa-BRU
    a) PM1 cells
    no chemokines 100 100
    +R/Mα/Mβ (80/400/100) 1 95
    +RANTES (80) 8 100
    +MIP-1α (400) 39 100
    +MIP-1β (100) 13 93
    +MCP-1 (100) 99 98
    +MCP-2 (100) 72 93
    +MCP-3 (100) 98 99
    b) LW5 CD4+ cells
    no chemokines 100 100
    +R/Mα/Mβ (106/533/133) 39 100
    +RANTES (106) 65 95
    +MIP-1α (533) 72 100
    +MIP-1β (133) 44 92
    +OKT4A (3 ug/ml) 0 0

    Table 2 Legend:
  • CD4+ target cells (mitogen-activated CD4+ lymphocytes or PM1 cells) were labeled with octadecyl rhodamine (Molecular Probes, Eugene, Oreg.), and HeLa-JR-FL cells, HeLa-BRU cells (or control HeLa cells, not shown) were labeled with octadecyl fluorescein (Molecular Probes), overnight at 37° C. Equal numbers of labeled target cells and env-expressing cells were mixed in 96-well plates and β-chemokines (or CD4 MAb OKT4a) were added at the final concentrations (ng/ml) indicated in parentheses in the first column. Fluorescence emission values were determined 4 h after cell mixing (17).
  • If cell fusion occurs, the dyes are closely associated in the conjoined membrane such that excitation of fluorescein at 450 nm results in resonance energy transfer (RET) and emission by rhodamine at 590 nm. Percentage fusion is defined as equal to 100×[(Exp RET−Min RET)/(Max RET−Min RET)], where Max RET=% RET obtained when HeLa-Env and CD4 cells are mixed, Exp RET=% RET obtained when HeLa-Env and CD4+ cells are mixed in the presence of fusion-inhibitory compounds, and Min RET=% RET obtained when HeLa cells (lacking HIV-1 envelope glycoproteins) and CD4+ cells are mixed. The % RET value is defined by a calculation described elsewhere (17), and each is the mean of triplicate determinations. These values were, for HeLa-JR-FL and HeLa-BRU cells respectively: PM1 cells 11.5%, 10.5%; LW5 CD4+ cells, 6.0%, 10.5%; R/Mα/Mβ, RANTES+MIP-1α+MIP-1β.
  • Similar results were obtained with primary CD4+ T-cells from LW5 (Table 2b), although higher concentrations of β-chemokines were required to inhibit membrane fusion in the primary cells than in PM1 cells. Thus, the actions of the β-chemokines are not restricted to the PM1 cell line. The RET assay demonstrates that β-chemokines interfere with env-mediated membrane fusion.
  • The simplest explanation of these results is that the binding of certain β-chemokines to their receptor(s) prevents, directly or otherwise, the fusion of HIV-1 with CD4+ T-cells. It has been known for a decade that HIV-1 requires a second receptor for entry into CD4+ cells (19-21). This function is supplied, for TCLA strains, by fusin (9). Several receptors for MIP-1α, MIP-1β and RANTES have been identified (6,7), and β-chemokines exhibit considerable cross-reactivity in receptor usage (4-8). However, C-C CKR-1 and, especially, C-C CKR-5 were identified as the most likely candidates, based on tissue expression patterns and their abilities to bind MIP-1α, MIP-1β and RANTES (4, 7, 8, 15, 22). C-C CKR-1, C-C CKR-5 and LESTR are each expressed at the mRNA level in PM1 cells and primary macrophages (data not shown). These and other β-chemokine receptors were therefore PCR-amplified, cloned and expressed.
  • The expression of C-C CKR-5 in HeLa-CD4 (human), COS-CD4 (simian) and 3T3-CD4 (murine) cells rendered each of them readily infectible by the primary, NSI strains ADA and BaL in the env-complementation assay of HIV-1 entry (Table 3)
    TABLE 3
    C—C CKR-5 expression permits infection of CD4-expressing
    cells by primary, NSI HIV-1 strains
    R/Mα/Mβ
    pcDNA3.1 LESTR CKR-1 CKR-2a CKR-3 CKR-4 CKR-5 CKR-5
    COS-CD4 ADA 798 456 600 816 516 534 153000 3210
    BaL 660 378 600 636 516 618 58800 756
    HxB2 5800 96700 5240 5070 5470 5620 4850 5000
    HeLa-CD4 ADA 678 558 4500 912 558 600 310000 6336
    BaL 630 738 1800 654 516 636 104000 750
    HxB2 337000 nd nd nd nd nd nd 356000
    3T3-CD4 ADA 468 558 450 618 534 606 28400 1220
    BaL 606 738 660 738 534 558 11700 756
    HxB2 456 24800 618 672 732 606 618 606

    Table 3 Legend:
  • Chemokine receptor genes C-C CKR-1, C-C CKR-2a, C-C CKR-3, C-C CKR-4 and C-C CKR-5 have no introns (4-8,15,22) and were isolated by PCR performed directly on a human genomic DNA pool derived from the PBMC of seven healthy donors. Oligonucleotides overlapping the ATG and the stop codons and containing BamHI and Xhol restriction sites for directional cloning into the pcDNA3.1 expression vector (Invitrogen Inc.) were used. LESTR (also known as fusin or HUMSTR) (4,9,24) was cloned by PCR performed directly on cDNA derived from PM1 cells, using sequences derived from the NIH database. Listed below are the 5′ and 3′ primer pairs used in first (5-1 and 3-1) and second (5-2 and 3-2) round PCR amplification of the CKR genes directly from human genomic DNA, and of LESTR from PM1 cDNA. Only a single set of primers was used to amplify CKR-5.
    LESTR:
    (SEQ ID NO: 6)
    L/5-1 = AAG CTT GGA GAA CCA GCG GTT ACC ATG GAG GGG ATC;
    (SEQ ID NO: 7)
    L/5-2 = GTC TGA GTC TGA GTC AAG CTT GGA GAA CCA;
    (SEQ ID NO: 8)
    L/3-1 = CTC GAG CAT CTG TGT TAG CTG GAG TGA AAA CTT GAA GAC TC;
    (SEQ ID NO: 9)
    L/3-2 = GTC TGA GTC TGA GTC CTC GAG CAT CTG TGT;
    CKR-1:
    (SEQ ID NO: 10)
    C1/5-1 = AAG CTT CAG AGA GAA GCC GGG ATG GAA ACT CC;
    (SEQ ID NO: 11)
    C1/5-2 = GTC TGA GTC TGA GTC AAG CTT CAG AGA GAA;
    (SEQ ID NO: 12)
    C1/3-1 = CTC GAG CTG AGT CAG AAC CCA GCA GAG AGT TC;
    (SEQ ID NO: 13)
    C1/3-2 = GTC TGA GTC TGA GTC CTC GAG CTG AGT CAG;
    CKR-2a:
    (SEQ ID NO: 14)
    C2/5-1 = AAG CTT CAG TAC ATC CAC AAC ATG CTG TCC AC;
    (SEQ ID NO: 15)
    C2/5-2 = GTC TGA GTC TGA GTC AAG CTT CAG TAC ATC;
    (SEQ ID NO: 16)
    C2/3-1 = CTC GAG CCT CGT TTT ATA AAC CAG CCG AGA C;
    (SEQ ID NO: 17)
    C2/3-2 = GTC TGA GTC TGA GTC CTC GAG CCT CGT TTT;
    CKR-3:
    (SEQ ID NO: 18)
    C3/5-1 = AAG CTT CAG GGA GAA GTG AAA TGA CAA CC;
    (SEQ ID NO: 19)
    C3/5-2 = GTC TGA GTC TGA GTC AAG CTT CAG GGA GAA;
    (SEQ ID NO: 20)
    C3/3-1 = CTC GAG CAG ACC TAA AAC ACA ATA GAG AGT TCC;
    (SEQ ID NO: 21)
    C3/3-2 = GTC TGA GTC TGA GTC CTC GAG CAG ACC TAA;
    CKR-4:
    (SEQ ID NO: 22)
    C4/5-1 = AAG CTT CTG TAG AGT TAA AAA ATG AAC CCC ACG G;
    (SEQ ID NO: 23)
    C4/5-2 = GTC TGA GTC TGA GTC AAG CTT CTG TAG AGT;
    (SEQ ID NO: 24)
    C4/3-1 = CTC GAG CCA TTT CAT TTT TCT ACA GGA CAG CAT C;
    (SEQ ID NO: 25)
    C4/3-2 = GTC TGA GTC TGA GTC CTC GAG CCA TTT CAT;
    CKR-5:
    (SEQ ID NO: 26)
    C5/5-12 = GTC TGA GTC TGA GTC AAG CTT AAC AAG ATG GAT TAT CAA;
    (SEQ ID NO: 37)
    C5/3-12 = GTC TGA GTC TGA GTC CTC GAG TCC GTG TCA CAA GCC CAC.
  • The human CD4-expressing cell lines HeLa-CD4 (P42), 3T3-CD4 (sc6) and COS-CD4 (Z28T1) (23) were transfected with the different pcDNA3.1-CKR constructs by the calcium phosphate method, then infected 48 h later with different reporter viruses (200 ng of HIV-1 p24/106 cells) in the presence or absence of β-chemokines (400 ng/ml each of RANTES, MIP-1α and MIP-1β). Luciferase activity in cell lysates was measured 48 h later (10,11). β-Chemokine blocking data is only shown for C-C CKR-5, as infection mediated by the other C-C CKR genes was too weak for inhibition to be quantifiable. In PCR-based assays of HIV-1 entry, a low level of entry of NL4/3 and ADA into C-C CKR-1 expressing cells (data not shown) was consistently observed.
  • Neither LESTR nor C-C CKR-1, -2a, -3 or -4 could substitute for C-C CKR-5 in this assay. The expression of LESTR in COS-CD4 and 3T3-CD4 cells permitted HxB2 entry, and HxB2 readily entered untransfected (or control plasmid-transfected) HeLa-CD4 cells (Table 3). Entry of BAL and ADA into all three C-C CKR-5-expressing cell lines was almost completely inhibited by the combination of MIP-1α, MIP-1β and RANTES, whereas HxB2 entry into LESTR-expressing cells was insensitive to β chemokines (Table 3) These results suggest that C-C CKR-5 functions as a β-chemokine-sensitive second receptor for primary, NSI HIV-1 strains.
  • The second receptor function of C-C CKR-5 was confirmed in assays of env-mediated membrane fusion. When C-C CKR-5 was transiently expressed in COS and HeLa cell lines that permanently expressed human CD4, both cell lines fused strongly with HeLa cells expressing the JR-FL envelope glycoproteins, whereas no fusion occurred when control plasmids were used (data not shown). Expression of LESTR instead of C-C CKR-5 did not permit either COS-CD4 or HeLa-CD4 cells to fuse with HeLa-JR-FL cells, but did allow fusion between COS-CD4 cells and HeLa-BRU cells (data not shown).
  • The fusion capacity of β-chemokine receptors was also tested in the RET assay. The expression of C-C CKR-5, but not of C-C CKR-1, -2a, -3 or -4, permitted strong fusion between HeLa-CD4 cells and HeLa-JR-FL cells. The extent of fusion between HeLa-JR-FL cells and C-C CKR-5-expressing HeLa-CD4 cells was greater than the constitutive level of fusion between HeLa-BRU cells and HeLa-CD4 cells (FIG. 4) The fusion-conferring function of C-C CKR-5 for primary, NSI HIV-1 strains has therefore been confirmed in two independent fusion assays.
  • Experimental Discussion
  • Together, the above results establish that MIP-1α, MIP-1β and RANTES inhibit HIV-1 infection at the entry stage, by interfering with the virus-cell fusion reaction subsequent to CD4 binding. It was also shown that C-C CKR-5 can serve as a second receptor for entry of primary NSI strains of HIV-1 into CD4+ T-cells, and that the interaction of β-chemokines with C-C CKR-5 inhibits the HIV-1 fusion reaction.
  • REFERENCES OF THE SECOND SERIES OF EXPERIMENTS
    • 1. Levy, J. A., Mackewicz, C. E. & Barker, E. Immunol. Today 17, 217-224 (1996).
    • 2. Cocchi, F. et al. Science 270, 1811-1815 (1995).
    • 3. Paxton, W. A. et al. Nat. Med. 2, 412-417 (1996).
    • 4. Neote, K., DiGregorio, D., Mak, J. Y., Horuk, R., & Schall, T. J. Cell 72, 415-425 (1993).
    • 5. Gao, J.-L. et al. J. Exp. Med. 177, 1421-1427 (1993).
    • 6. Bacon, K. B., Premack, B. A., Gardner, P. & Schall, T. J. Science 269, 1727-1729 (1995).
    • 7. Raport, C. J. et al. J. Leukoc. Biol. 59,18-23 (1996).
    • 8. Wells, T. N. C. et al. J. Leukoc. Biol. 59, 53-60 (1996).
    • 9. Feng, Y., Broder, C. C., Kennedy, P. E. & Berger, E. A. Science 272, 872-877 (1996).
    • 10. Chen, B. K., Saksela, K., Andino, R. & Baltimore, D. J. Virol. 68, 654-660 (1994).
    • 11. Connor, R. I., Chen, B. K., Choe, S., & Landau, N. R. Virology 206, 935-944 (1995).
    • 12. Lusso, P. et al. J. Virol. 69, 3712-3720 (1995).
    • 13. Charo, l. F. et al. Proc. Natl. Acad. Sci. USA 91, 2752-2756 (1994).
    • 14. Ben-Baruch, A. et al. J. Biol. Chem. 270, 22123-22128 (1995).
    • 15. Combadiere, C et al. J. Biol. Chem. 270, 29671-29675 (1995).
    • 16. Lip, J. P., D'Andrea, A. D., Lodish, H. F. & Baltimore, D. Nature 343, 762-764 (1990).
    • 17. Litwin, V. et al. J. Virol. (submitted for publication).
    • 18. Moore, J. P., Jameson, B. A., Weiss, R. A. & Sattentau, Q. J. in Viral Fusion Mechanisms (ed Bentz, J.) 233-239 (CRC Press Inc, Boca Raton, USA, 1993).
    • 19. Maddon, P. J. et al. Cell 47, 333-348 (1986).
    • 20. Ashorn, P. A., Berger, E. A. & Moss, B. J. Virol. 64, 2149-2156 (1990).
    • 21. Clapham, P. R., Blanc, D. & Weiss, R. A. Virology 181, 703-715 (1991).
    • 22. Samson, M., Labbe, O., Mollereau, C., Vassart, G. & Parmentier, M. Biochemistry 11, 3362-3367 (1996).
    • 23. Dragic, T., Charneau, P., Clavel, F. & Alizon, M. J. Virol. 66, 4794-4802 (1992)
    • 24. Loetscher, M. et al. J. Biol. Chem. 269, 232-237 (1994).
    • 25. Moore, J. P. & Ho, D. D. AIDS 9 (suppl A), S117-S136 (1995).
    • 26. Trkola, A. & Moore, J. P. (unpublished data).
    • 27. Chaudhuri, A., et al. 1994. J. Biol. Chem. 269, 7835-7838 (1994).
    • 28. Neote, K., Mak, J. Y., Kolakowski Jr., L. F. & Schall, T. J. Blood 84, 44-52 (1994).
    • 29. Dragic, T., Picard, L. & Alizon, M. J. Virol. 69, 1013-1018 (1995).
    • 30. Puri, A., Morris, S. J., Jones, P., Ryan, M. & Blumenthal, R. Virology 219, 262-267 (1996).31
      Third Series of Experiments
  • The chemokine SDF-1 (stromal cell-derived factor 1) is the natural ligand for Fusin/CXCR4 and blocks infection by laboratory-adapted strains of HIV-1 (Ref. 1 and 2). SDF-1 exists as at least two forms, SDF-1α and SDF-1β based on variable splicing of the SDF-1 gene (Ref. 1 and 3) In the RET assay, this chemokine specifically inhibits membrane fusion mediated by gp120/gp41 form the laboratory-adapted strain HIVLAI but not by gp120/gp41 from the macrophage-tropic isolate HIV-1 JR-FL as shown in FIG. 5.
  • REFERENCES OF THE THIRD SERIES OF EXPERIMENTS
    • 1. Bleul, C. C., et al. (1996) Nature 382:829-833
    • 2. Oberlin, E., et al. (1996) Nature 382:833-835
    • 3. Shirozu, M., et al. (1995) Genomics 28:495-500

Claims (7)

1-50. (canceled)
51. An isolated antibody which binds the polypeptide of SEQ ID NO:2 when said polypeptide is expressed on the surface of a cell.
52. The antibody of claim 51, wherein said antibody is monoclonal.
53. The antibody of claim 51, wherein said antibody is humanized.
54. An antibody that specifically binds to the extracellular domain of a chemokine receptor 88C polypeptide comprising the amino acid sequence in SEQ ID NO:2,
wherein said antibody fails to cross-react with an MCP-1 receptor (CCCKR-2), and
wherein the antibody binds chemokine receptor 88C expressed on the surface of cells and inhibits human or simian human immunodeficiency (HIV or SIV) infection of said cells.
55. A monoclonal antibody according to claim 54.
56. A humanized antibody according to claim 54.
US11/175,815 1996-04-01 2005-07-05 Method for preventing HIV-1 infection of CD4+ cells Abandoned US20060029932A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/175,815 US20060029932A1 (en) 1996-04-01 2005-07-05 Method for preventing HIV-1 infection of CD4+ cells

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US1453296P 1996-04-01 1996-04-01
US1971596P 1996-06-14 1996-06-14
US08/831,823 US6344545B1 (en) 1996-06-14 1997-04-02 Method for preventing HIV-1 infection of CD4+ cells
US09/888,938 US20020155429A1 (en) 1996-04-01 2001-06-25 Method for preventing HIV-1 infection of CD4+ cells
US11/175,815 US20060029932A1 (en) 1996-04-01 2005-07-05 Method for preventing HIV-1 infection of CD4+ cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/888,938 Continuation US20020155429A1 (en) 1996-04-01 2001-06-25 Method for preventing HIV-1 infection of CD4+ cells

Publications (1)

Publication Number Publication Date
US20060029932A1 true US20060029932A1 (en) 2006-02-09

Family

ID=26692524

Family Applications (4)

Application Number Title Priority Date Filing Date
US08/831,823 Expired - Lifetime US6344545B1 (en) 1996-04-01 1997-04-02 Method for preventing HIV-1 infection of CD4+ cells
US09/888,938 Abandoned US20020155429A1 (en) 1996-04-01 2001-06-25 Method for preventing HIV-1 infection of CD4+ cells
US11/175,815 Abandoned US20060029932A1 (en) 1996-04-01 2005-07-05 Method for preventing HIV-1 infection of CD4+ cells
US11/894,677 Abandoned US20080187539A1 (en) 1996-04-01 2007-08-20 Method for preventing HIV-1 infection of CD4 cells

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/831,823 Expired - Lifetime US6344545B1 (en) 1996-04-01 1997-04-02 Method for preventing HIV-1 infection of CD4+ cells
US09/888,938 Abandoned US20020155429A1 (en) 1996-04-01 2001-06-25 Method for preventing HIV-1 infection of CD4+ cells

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/894,677 Abandoned US20080187539A1 (en) 1996-04-01 2007-08-20 Method for preventing HIV-1 infection of CD4 cells

Country Status (1)

Country Link
US (4) US6344545B1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020045161A1 (en) * 1995-06-07 2002-04-18 Progenics Pharmaceuticals, Inc. Fluorescence resonance energy transfer screening assay for the identification of HIV-1 envelope glycoprotein-medicated cell
US20020155429A1 (en) * 1996-04-01 2002-10-24 Progenics Pharmaceuticals, Inc. Method for preventing HIV-1 infection of CD4+ cells
US20060194244A1 (en) * 1996-06-14 2006-08-31 Progenics Pharmaceuticals, Inc. Uses of a chemokine receptor for inhibiting HIV-1 infection
US20060233798A1 (en) * 2001-04-06 2006-10-19 Progenics Pharmaceuticals, Inc. Methods for inhibiting HIV-1 infection
US20070020280A1 (en) * 2000-09-15 2007-01-25 Progenics Pharmaceuticals, Inc. Compositions and methods for inhibition of HIV-1 infection
US20070026441A1 (en) * 2005-07-22 2007-02-01 Olson William C Methods for reducing viral load in HIV-1-infected patients
US20070231327A1 (en) * 1998-12-16 2007-10-04 Progenics Pharmaceuticals, Inc. Anti-CCR5 antibodies
US7345153B2 (en) 1996-01-17 2008-03-18 Progenics Pharmaceuticals, Inc. Compounds capable of inhibiting HIV-1 infection
US20080107595A1 (en) * 2002-02-22 2008-05-08 Olson William C Anti-CCR5 antibody
US7858298B1 (en) 1996-04-01 2010-12-28 Progenics Pharmaceuticals Inc. Methods of inhibiting human immunodeficiency virus type 1 (HIV-1) infection through the administration of CCR5 chemokine receptor antagonists
US20110135634A1 (en) * 2007-07-19 2011-06-09 Olson William C Methods of inhibiting infection by diverse subtypes of drug-resistant hiv-1

Families Citing this family (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6025154A (en) * 1995-06-06 2000-02-15 Human Genome Sciences, Inc. Polynucleotides encoding human G-protein chemokine receptor HDGNR10
US20040151719A1 (en) * 1995-06-06 2004-08-05 Human Genome Sciences, Inc. Human G-protein chemokine receptor (CCR5) HDGNR10
US6258527B1 (en) * 1996-05-20 2001-07-10 The Aaron Diamond Aids Research Center Methods of identifying g-coupled receptors associated with macrophage-trophic HIV, and diagnostic and therapeutic uses thereof
WO1997045543A2 (en) * 1996-05-28 1997-12-04 The Government Of The United States Of America, As Represented By The Secretary Of Health And Human Services, National Institutes Of Health Cc chemokine receptor 5, antibodies thereto, transgenic animals
US6528625B1 (en) * 1996-10-28 2003-03-04 Millennium Pharmaceuticals, Inc. Anti-CCR5 antibodies and kits comprising same
US20080015348A1 (en) * 1998-12-16 2008-01-17 Progenics Pharmaceuticals, Inc. Nucleic acids encoding polypeptides of anti-CCR5 antibodies
WO2001004361A2 (en) * 1999-07-09 2001-01-18 Gen-Probe Incorporated Detection of hiv-1 by nucleic acid amplification
KR20030032916A (en) * 2000-02-09 2003-04-26 휴먼 게놈 사이언시즈, 인크. Antibodies to ccr5
AU2001247254A1 (en) * 2000-02-29 2001-09-12 Aaron Diamond Aids Research Centre Sulfated CCR5 peptides for HIV -1 infection
US7175988B2 (en) * 2001-02-09 2007-02-13 Human Genome Sciences, Inc. Human G-protein Chemokine Receptor (CCR5) HDGNR10
US20020146415A1 (en) * 2001-04-06 2002-10-10 Olson William C. Methods for inhibiting HIV-1 infection
US20030104496A1 (en) * 2001-11-30 2003-06-05 Cytokinetics, Inc. Novel motor protein of P. falciparum and methods for its use
US7393934B2 (en) * 2001-12-21 2008-07-01 Human Genome Sciences, Inc. Human G-protein chemokine receptor (CCR5) HDGNR10
EP1723178A4 (en) * 2004-03-12 2007-12-12 Human Genome Sciences Inc Human g-protein chemokine receptor (ccr5) hdgnr10
AP2007004095A0 (en) * 2005-01-07 2007-08-31 Univ Emory Cxcr4 antagonists for the treatment of hiv infection
US20080227799A1 (en) * 2006-07-11 2008-09-18 Liotta Dennis C CXCR4 Antagonists Including Heteroatoms for the Treatment of Medical Disorders
WO2008008854A2 (en) * 2006-07-11 2008-01-17 Emory University Cxcr4 antagonists including diazine and triazine structures for the treatment of medical disorders
US20080283557A1 (en) * 2007-05-17 2008-11-20 Julianne Desautels Spout for food stuff container
US20090074766A1 (en) * 2007-09-14 2009-03-19 Ketas Thomas J Methods of inhibiting HIV-2 infection
AU2009228034A1 (en) 2008-03-28 2009-10-01 Altiris Therapeutics Chemokine receptor modulators
EP2550362B1 (en) 2010-03-25 2017-01-04 Oregon Health&Science University Cmv glycoproteins and recombinant vectors
EP2691530B1 (en) 2011-06-10 2018-03-07 Oregon Health & Science University Cmv glycoproteins and recombinant vectors
EP2568289A3 (en) 2011-09-12 2013-04-03 International AIDS Vaccine Initiative Immunoselection of recombinant vesicular stomatitis virus expressing hiv-1 proteins by broadly neutralizing antibodies
US9402894B2 (en) 2011-10-27 2016-08-02 International Aids Vaccine Initiative Viral particles derived from an enveloped virus
WO2013096926A1 (en) 2011-12-22 2013-06-27 Glycomimetics, Inc. E-selectin antagonist compounds, compositions, and methods of use
EP2679596B1 (en) 2012-06-27 2017-04-12 International Aids Vaccine Initiative HIV-1 env glycoprotein variant
ES2668045T3 (en) 2012-12-07 2018-05-16 Glycomimetics, Inc. Compounds, compositions and methods that use E-selectin antagonists for the mobilization of hematopoietic cells
US20150065381A1 (en) 2013-09-05 2015-03-05 International Aids Vaccine Initiative Methods of identifying novel hiv-1 immunogens
US10058604B2 (en) 2013-10-07 2018-08-28 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
SI3227310T1 (en) 2014-12-03 2019-11-29 Glycomimetics Inc Heterobifunctional inhibitors of e-selectins and cxcr4 chemokine receptors
EP3069730A3 (en) 2015-03-20 2017-03-15 International Aids Vaccine Initiative Soluble hiv-1 envelope glycoprotein trimers
US9931394B2 (en) 2015-03-23 2018-04-03 International Aids Vaccine Initiative Soluble HIV-1 envelope glycoprotein trimers
US11291678B2 (en) 2016-03-02 2022-04-05 Glycomimetics, Inc Methods for the treatment and/or prevention of cardiovascular disease by inhibition of E-selectin
WO2018031445A1 (en) 2016-08-08 2018-02-15 Glycomimetics, Inc. Combination of t-cell checkpoint inhibitors with inhibitors of e-selectin or cxcr4, or with heterobifunctional inhibitors of both e-selectin and cxcr4
CA3037850A1 (en) 2016-10-07 2018-04-12 Glycomimetics, Inc. Highly potent multimeric e-selectin antagonists
CA3054605A1 (en) 2017-03-15 2018-09-20 Glycomimetics, Inc. Galactopyranosyl-cyclohexyl derivatives as e-selectin antagonists
WO2019108750A1 (en) 2017-11-30 2019-06-06 Glycomimetics, Inc. Methods of mobilizing marrow infiltrating lymphocytes and uses thereof
EP3732186A1 (en) 2017-12-29 2020-11-04 GlycoMimetics, Inc. Heterobifunctional inhibitors of e-selectin and galectin-3
AU2019230013A1 (en) 2018-03-05 2020-09-10 Glycomimetics, Inc. Methods for treating acute myeloid leukemia and related conditions
US11845771B2 (en) 2018-12-27 2023-12-19 Glycomimetics, Inc. Heterobifunctional inhibitors of E-selectin and galectin-3

Citations (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4886743A (en) * 1985-04-24 1989-12-12 California Institute Of Technology Diagnostic reagents based on unique sequences within the variable region of the T cell receptor and uses thereof
US5021409A (en) * 1989-12-21 1991-06-04 Johnson Matthey Plc Antiviral cyclic polyamines
US5071964A (en) * 1988-05-04 1991-12-10 Dana-Farber Cancer Institute, Inc. Protein micelles
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5126433A (en) * 1986-08-21 1992-06-30 The Trustees Of Columbia University In The City Of New York Soluble forms of the t cell surface protein cd4
US5215913A (en) * 1987-11-30 1993-06-01 Roger Williams General Hospital IgG-1 human monoclonal antibody reactive with an HIV-1 antigen and methods of use
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5440021A (en) * 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
US5449608A (en) * 1993-03-22 1995-09-12 The United States Of America As Represented By The Department Of Health And Human Services Parvovirus B19 receptor and parvovirus B19 detection
US5504003A (en) * 1994-03-08 1996-04-02 Human Genome Sciences, Inc. Macrophage inflammatory protein-3 and -4
US5603933A (en) * 1993-08-31 1997-02-18 Board Of Regents, The University Of Texas CD4 peptides for binding to viral envelope proteins
US5668149A (en) * 1990-01-26 1997-09-16 The United States Of America As Represented By The Department Of Health And Human Services Inhibition of human immunodeficiency virus-1 infectivity in human cells
US5817767A (en) * 1993-02-24 1998-10-06 Progenics Pharmaceuticals, Inc. Synergistic composition of CD4-based protein and anti-HIV-1 antibody, and methods of using same
US5854400A (en) * 1987-05-29 1998-12-29 Tanox, Inc. Monoclonal antibodies which neutralize HIV-1 infection
US5939320A (en) * 1996-05-20 1999-08-17 New York University G-coupled receptors associated with macrophage-trophic HIV, and diagnostic and therapeutic uses thereof
US5994515A (en) * 1996-06-25 1999-11-30 Trustees Of The University Of Pennsylvania Antibodies directed against cellular coreceptors for human immunodeficiency virus and methods of using the same
US6025154A (en) * 1995-06-06 2000-02-15 Human Genome Sciences, Inc. Polynucleotides encoding human G-protein chemokine receptor HDGNR10
US6100087A (en) * 1998-03-11 2000-08-08 City Of Hope Ribozymes targeted to human CCR5 mRNA
US6107019A (en) * 1996-06-14 2000-08-22 Progenics Pharmaceuticals, Inc. Method for preventing HIV-1 infection of CD4+ cells
US20010000241A1 (en) * 1995-06-06 2001-04-12 Yi Li Antibodies to human G-protein chemokine receptor HDGNR10 (CCR5 receptor)
US6258782B1 (en) * 1998-05-20 2001-07-10 Trimeris, Inc. Hybrid polypeptides with enhanced pharmacokinetic properties
US6258527B1 (en) * 1996-05-20 2001-07-10 The Aaron Diamond Aids Research Center Methods of identifying g-coupled receptors associated with macrophage-trophic HIV, and diagnostic and therapeutic uses thereof
US6261763B1 (en) * 1995-06-07 2001-07-17 Progenics Pharmaceuticals, Inc. Fluorescence resonance energy transfer screening assay for the identification of compounds that are capable of abrogating macrophage-tropic HIV-1 cell fusion
US6265184B1 (en) * 1995-12-20 2001-07-24 Icos Corporation Polynucleotides encoding chemokine receptor 88C
US6265284B1 (en) * 1998-09-21 2001-07-24 Samsung Electronics Co., Ltd. Method of manufacturing a trench isolation region in a semiconductor device
US20010046512A1 (en) * 1996-01-17 2001-11-29 Progenics Pharmaceuticals, Inc. Compounds capable of inhibiting HIV-1 infection
US6344545B1 (en) * 1996-06-14 2002-02-05 Progenics Pharmaceuticals, Inc. Method for preventing HIV-1 infection of CD4+ cells
US20020048786A1 (en) * 2000-02-09 2002-04-25 Rosen Craig A. Human G-protein Chemokine Receptor HDGNR10
US20020106374A1 (en) * 2000-09-15 2002-08-08 Olson William C. Compositions and methods for inhibition of HIV-1 infection
US20020110870A1 (en) * 1996-03-01 2002-08-15 Michel Samson Therapeutic applications for CCR5 chemokine receptors
US20020146415A1 (en) * 2001-04-06 2002-10-10 Olson William C. Methods for inhibiting HIV-1 infection
US6488375B2 (en) * 1999-10-28 2002-12-03 Ocular Sciences, Inc. Tinted contact lens and method for making same
US20030003440A1 (en) * 2001-03-14 2003-01-02 Lucia Lopalco Novel CCR5 epitope and antibodies against it
US6528625B1 (en) * 1996-10-28 2003-03-04 Millennium Pharmaceuticals, Inc. Anti-CCR5 antibodies and kits comprising same
US20030044411A1 (en) * 2001-04-06 2003-03-06 Olson William C. Methods for inhibiting HIV-1 infection
US6548636B2 (en) * 2000-02-29 2003-04-15 Progenics Pharmaceuticals, Inc. Sulfated CCR5 peptides for HIV-1 infection
US20030092632A1 (en) * 2001-02-28 2003-05-15 Tatjana Dragic Sulfated CCR5 peptides for HIV-1 infection
US20030100058A1 (en) * 2001-02-09 2003-05-29 Human Genome Sciences, Inc. Human G-protein Chemokine Receptor (CCR5) HDGNR10
US20030195348A1 (en) * 1996-05-28 2003-10-16 The United States Of America, Represented By The Secretary, CC chemokine receptor 5 DNA, new animal models and therapeutic agents for HIV infection
US20030228306A1 (en) * 2002-02-22 2003-12-11 Olson William C. Anti-CCR5 antibody
US20040062767A1 (en) * 2000-01-28 2004-04-01 Progenics Pharmaceuticals, Inc. Compositions and methods for inhibition of HIV-1 infection
US20040086528A1 (en) * 1996-06-14 2004-05-06 Progenics Pharmaceuticals, Inc. Uses of a chemokine receptor for inhibiting HIV-1 infection
US20040151719A1 (en) * 1995-06-06 2004-08-05 Human Genome Sciences, Inc. Human G-protein chemokine receptor (CCR5) HDGNR10
US20040228869A1 (en) * 1998-12-16 2004-11-18 Progenics Pharmaceuticals, Inc. Synergistic inhibition of HIV-1 fusion and attachment, compositions and antibodies thereto
US20050154193A1 (en) * 2001-12-21 2005-07-14 Viktor Roschke Human G-protein chemokine receptor (CCR5) HDGNR10

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5126432A (en) * 1988-02-12 1992-06-30 Miles Inc. Calcium channel modulating substances
DE69511245T2 (en) * 1994-02-18 1999-12-16 Immuno Ag Wien COMPOSITION AND METHOD FOR PREVENTING AND TREATING IGNITION WITH IMMUNOGLOBIN A
WO1997028258A1 (en) * 1996-01-30 1997-08-07 The National Institutes Of Health Cells expressing both human cd4 and cxcr4
US6825210B2 (en) * 2001-04-19 2004-11-30 Bristol-Myers Squibb Pharma Company Tricyclic compounds useful as HIV reverse transcriptase inhibitors
CN1805740A (en) * 2003-05-16 2006-07-19 马里兰大学生物技术研究所 Compositions for down-regulation of CCR5 expression and methods of use thereof
US20050131042A1 (en) * 2003-12-11 2005-06-16 Flentge Charles A. HIV protease inhibiting compounds
EP1723178A4 (en) * 2004-03-12 2007-12-12 Human Genome Sciences Inc Human g-protein chemokine receptor (ccr5) hdgnr10
MX2008000984A (en) * 2005-07-22 2008-04-04 Progenics Pharm Inc Methods for reducing viral load in hiv-1-infected patients.

Patent Citations (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4886743A (en) * 1985-04-24 1989-12-12 California Institute Of Technology Diagnostic reagents based on unique sequences within the variable region of the T cell receptor and uses thereof
US5225539A (en) * 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5126433A (en) * 1986-08-21 1992-06-30 The Trustees Of Columbia University In The City Of New York Soluble forms of the t cell surface protein cd4
US5091513A (en) * 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5854400A (en) * 1987-05-29 1998-12-29 Tanox, Inc. Monoclonal antibodies which neutralize HIV-1 infection
US5215913A (en) * 1987-11-30 1993-06-01 Roger Williams General Hospital IgG-1 human monoclonal antibody reactive with an HIV-1 antigen and methods of use
US5071964A (en) * 1988-05-04 1991-12-10 Dana-Farber Cancer Institute, Inc. Protein micelles
US5021409A (en) * 1989-12-21 1991-06-04 Johnson Matthey Plc Antiviral cyclic polyamines
US5668149A (en) * 1990-01-26 1997-09-16 The United States Of America As Represented By The Department Of Health And Human Services Inhibition of human immunodeficiency virus-1 infectivity in human cells
US5440021A (en) * 1991-03-29 1995-08-08 Chuntharapai; Anan Antibodies to human IL-8 type B receptor
US5817767A (en) * 1993-02-24 1998-10-06 Progenics Pharmaceuticals, Inc. Synergistic composition of CD4-based protein and anti-HIV-1 antibody, and methods of using same
US5449608A (en) * 1993-03-22 1995-09-12 The United States Of America As Represented By The Department Of Health And Human Services Parvovirus B19 receptor and parvovirus B19 detection
US5603933A (en) * 1993-08-31 1997-02-18 Board Of Regents, The University Of Texas CD4 peptides for binding to viral envelope proteins
US5504003A (en) * 1994-03-08 1996-04-02 Human Genome Sciences, Inc. Macrophage inflammatory protein-3 and -4
US20040151719A1 (en) * 1995-06-06 2004-08-05 Human Genome Sciences, Inc. Human G-protein chemokine receptor (CCR5) HDGNR10
US6025154A (en) * 1995-06-06 2000-02-15 Human Genome Sciences, Inc. Polynucleotides encoding human G-protein chemokine receptor HDGNR10
US6511826B2 (en) * 1995-06-06 2003-01-28 Human Genome Sciences, Inc. Polynucleotides encoding human G-protein chemokine receptor (CCR5) HDGNR10
US20010000241A1 (en) * 1995-06-06 2001-04-12 Yi Li Antibodies to human G-protein chemokine receptor HDGNR10 (CCR5 receptor)
US6759519B2 (en) * 1995-06-06 2004-07-06 Human Genome Sciences, Inc. Antibodies to human G-protein chemokine receptor HDGNR10 (CCR5receptor)
US20030023044A1 (en) * 1995-06-06 2003-01-30 Human Genome Sciences, Inc. Human G-Protein chemokine receptor (CCR5) HDGNR10
US6800729B2 (en) * 1995-06-06 2004-10-05 Human Genome Sciences, Inc. Human G-Protein chemokine receptor HDGNR10 (CCR5 receptor)
US6743594B1 (en) * 1995-06-06 2004-06-01 Human Genome Sciences, Inc. Methods of screening using human G-protein chemokine receptor HDGNR10 (CCR5)
US20020045161A1 (en) * 1995-06-07 2002-04-18 Progenics Pharmaceuticals, Inc. Fluorescence resonance energy transfer screening assay for the identification of HIV-1 envelope glycoprotein-medicated cell
US20060140977A1 (en) * 1995-06-07 2006-06-29 Progenics Pharmaceuticals, Inc. Methods for assaying inhibition of HIV-1 envelope glycoprotein-mediated membrane fusion
US6972126B2 (en) * 1995-06-07 2005-12-06 Progenics Pharmaceuticals, Inc. Methods for using resonance energy transfer-based assay of HIV-1 envelope glycoprotein-mediated membrane fusion, and kits for practicing same
US6261763B1 (en) * 1995-06-07 2001-07-17 Progenics Pharmaceuticals, Inc. Fluorescence resonance energy transfer screening assay for the identification of compounds that are capable of abrogating macrophage-tropic HIV-1 cell fusion
US6265184B1 (en) * 1995-12-20 2001-07-24 Icos Corporation Polynucleotides encoding chemokine receptor 88C
US6797811B1 (en) * 1995-12-20 2004-09-28 Icos Corporation Antibodies to chemokine receptor 88C
US6268477B1 (en) * 1995-12-20 2001-07-31 Icos Corporation Chemokine receptor 88-C
US20020150888A1 (en) * 1995-12-20 2002-10-17 Gray Patrick W. Methods of screening for modulators of HIV infection
US20040230037A1 (en) * 1995-12-20 2004-11-18 Icos Corporation Chemokine receptor materials and methods
US20050260565A1 (en) * 1995-12-20 2005-11-24 Icos Corporation Method of screening for modulators of HIV infection
US20010046512A1 (en) * 1996-01-17 2001-11-29 Progenics Pharmaceuticals, Inc. Compounds capable of inhibiting HIV-1 infection
US6800447B2 (en) * 1996-03-01 2004-10-05 Euroscreen S.A. Methods for identifying compounds which bind the active CCR5 chemokine receptor
US20040110127A1 (en) * 1996-03-01 2004-06-10 Euroscreen Screening methods for identifying compounds which decrease HIV entry into a cell
US6448375B1 (en) * 1996-03-01 2002-09-10 Euroscreen S.A. Active and inactive CC-chemokine receptor
US20020110870A1 (en) * 1996-03-01 2002-08-15 Michel Samson Therapeutic applications for CCR5 chemokine receptors
US20040161739A1 (en) * 1996-03-01 2004-08-19 Michel Samson HIV diagnostic methods
US6692938B2 (en) * 1996-03-01 2004-02-17 Euroscreen, S.A. Nucleic acids encoding active and inactive CCR5 chemokine receptors
US6930174B2 (en) * 1996-03-01 2005-08-16 Euroscreen, S.A. CCR5 chemokine receptor-specific antibodies
US20020155429A1 (en) * 1996-04-01 2002-10-24 Progenics Pharmaceuticals, Inc. Method for preventing HIV-1 infection of CD4+ cells
US20030096221A1 (en) * 1996-05-20 2003-05-22 Littman Dan R. Methods of identifying g-couple receptors associated with macrophage-thophic hiv, and diagnostic and therapeutic uses thereof
US5939320A (en) * 1996-05-20 1999-08-17 New York University G-coupled receptors associated with macrophage-trophic HIV, and diagnostic and therapeutic uses thereof
US6258527B1 (en) * 1996-05-20 2001-07-10 The Aaron Diamond Aids Research Center Methods of identifying g-coupled receptors associated with macrophage-trophic HIV, and diagnostic and therapeutic uses thereof
US20030195348A1 (en) * 1996-05-28 2003-10-16 The United States Of America, Represented By The Secretary, CC chemokine receptor 5 DNA, new animal models and therapeutic agents for HIV infection
US20040259785A1 (en) * 1996-05-28 2004-12-23 The Govt. Of The Usa, As Represented By The Secretary Of The Dept. Of Health & Human Services CC chemokine receptor 5 DNA, new animal models and therapeutic agents for HIV infection
US20050118677A1 (en) * 1996-05-28 2005-06-02 The Gov. Of The U.S.A, As Represented By The Secretary Of The Dept. Of Health & Human Services CC chemokine receptor 5 DNA, new animal models and therapeutic agents for HIV infection
US6107019A (en) * 1996-06-14 2000-08-22 Progenics Pharmaceuticals, Inc. Method for preventing HIV-1 infection of CD4+ cells
US20040086528A1 (en) * 1996-06-14 2004-05-06 Progenics Pharmaceuticals, Inc. Uses of a chemokine receptor for inhibiting HIV-1 infection
US6344545B1 (en) * 1996-06-14 2002-02-05 Progenics Pharmaceuticals, Inc. Method for preventing HIV-1 infection of CD4+ cells
US5994515A (en) * 1996-06-25 1999-11-30 Trustees Of The University Of Pennsylvania Antibodies directed against cellular coreceptors for human immunodeficiency virus and methods of using the same
US6528625B1 (en) * 1996-10-28 2003-03-04 Millennium Pharmaceuticals, Inc. Anti-CCR5 antibodies and kits comprising same
US20030166870A1 (en) * 1996-10-28 2003-09-04 Millennium Pharmaceuticals, Inc. Anti-CCR5 antibodies and methods of use therefor
US6100087A (en) * 1998-03-11 2000-08-08 City Of Hope Ribozymes targeted to human CCR5 mRNA
US6258782B1 (en) * 1998-05-20 2001-07-10 Trimeris, Inc. Hybrid polypeptides with enhanced pharmacokinetic properties
US6265284B1 (en) * 1998-09-21 2001-07-24 Samsung Electronics Co., Ltd. Method of manufacturing a trench isolation region in a semiconductor device
US20040228869A1 (en) * 1998-12-16 2004-11-18 Progenics Pharmaceuticals, Inc. Synergistic inhibition of HIV-1 fusion and attachment, compositions and antibodies thereto
US6488375B2 (en) * 1999-10-28 2002-12-03 Ocular Sciences, Inc. Tinted contact lens and method for making same
US20040062767A1 (en) * 2000-01-28 2004-04-01 Progenics Pharmaceuticals, Inc. Compositions and methods for inhibition of HIV-1 infection
US20030166024A1 (en) * 2000-02-09 2003-09-04 Human Genome Sciences, Inc. Human G-protein chemokine receptor (CCR5) HDGNR10
US20020048786A1 (en) * 2000-02-09 2002-04-25 Rosen Craig A. Human G-protein Chemokine Receptor HDGNR10
US20020061834A1 (en) * 2000-02-09 2002-05-23 Rosen Craig A. Human G-protein Chemokine receptor (CCR5) HDGNR10
US6908734B2 (en) * 2000-02-29 2005-06-21 Progenics Pharmaceuticals, Inc. Sulfated CCR5 peptides for HIV-1 infection
US6548636B2 (en) * 2000-02-29 2003-04-15 Progenics Pharmaceuticals, Inc. Sulfated CCR5 peptides for HIV-1 infection
US20020106374A1 (en) * 2000-09-15 2002-08-08 Olson William C. Compositions and methods for inhibition of HIV-1 infection
US20030100058A1 (en) * 2001-02-09 2003-05-29 Human Genome Sciences, Inc. Human G-protein Chemokine Receptor (CCR5) HDGNR10
US20030092632A1 (en) * 2001-02-28 2003-05-15 Tatjana Dragic Sulfated CCR5 peptides for HIV-1 infection
US20030003440A1 (en) * 2001-03-14 2003-01-02 Lucia Lopalco Novel CCR5 epitope and antibodies against it
US20020146415A1 (en) * 2001-04-06 2002-10-10 Olson William C. Methods for inhibiting HIV-1 infection
US20030044411A1 (en) * 2001-04-06 2003-03-06 Olson William C. Methods for inhibiting HIV-1 infection
US20050154193A1 (en) * 2001-12-21 2005-07-14 Viktor Roschke Human G-protein chemokine receptor (CCR5) HDGNR10
US20030228306A1 (en) * 2002-02-22 2003-12-11 Olson William C. Anti-CCR5 antibody

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060140977A1 (en) * 1995-06-07 2006-06-29 Progenics Pharmaceuticals, Inc. Methods for assaying inhibition of HIV-1 envelope glycoprotein-mediated membrane fusion
US7901685B2 (en) 1995-06-07 2011-03-08 Progenics Pharmaceuticals Inc. Methods for assaying inhibition of HIV-1 envelope glycoprotein-mediated membrane fusion
US20020045161A1 (en) * 1995-06-07 2002-04-18 Progenics Pharmaceuticals, Inc. Fluorescence resonance energy transfer screening assay for the identification of HIV-1 envelope glycoprotein-medicated cell
US7862994B2 (en) 1995-06-07 2011-01-04 Progenics Pharmaceuticals Inc. Methods for inhibiting HIV-1 envelope glycoprotein-medicated membrane fusion
US20070048820A1 (en) * 1995-06-07 2007-03-01 Progenics Pharmaceuticals, Inc. Methods for assaying inhibition of HIV-1 envelope glycoprotein-mediated membrane fusion
US7345153B2 (en) 1996-01-17 2008-03-18 Progenics Pharmaceuticals, Inc. Compounds capable of inhibiting HIV-1 infection
US7858298B1 (en) 1996-04-01 2010-12-28 Progenics Pharmaceuticals Inc. Methods of inhibiting human immunodeficiency virus type 1 (HIV-1) infection through the administration of CCR5 chemokine receptor antagonists
US20020155429A1 (en) * 1996-04-01 2002-10-24 Progenics Pharmaceuticals, Inc. Method for preventing HIV-1 infection of CD4+ cells
US20060194244A1 (en) * 1996-06-14 2006-08-31 Progenics Pharmaceuticals, Inc. Uses of a chemokine receptor for inhibiting HIV-1 infection
US20070231327A1 (en) * 1998-12-16 2007-10-04 Progenics Pharmaceuticals, Inc. Anti-CCR5 antibodies
US20070020280A1 (en) * 2000-09-15 2007-01-25 Progenics Pharmaceuticals, Inc. Compositions and methods for inhibition of HIV-1 infection
US7736649B2 (en) 2000-09-15 2010-06-15 Progenics Pharmaceuticals, Inc. Methods of inhibiting human immunodeficiency virus infection through the administration of synergistic combinations of anti-CCR5 monoclonal antibodies, fusion inhibitors, and CD4-GP120 binding inhibitors
US20060233798A1 (en) * 2001-04-06 2006-10-19 Progenics Pharmaceuticals, Inc. Methods for inhibiting HIV-1 infection
US7666419B2 (en) 2002-02-22 2010-02-23 Progenics Pharmaceuticals Inc. Anti-CCR5 antibody
US7851600B2 (en) 2002-02-22 2010-12-14 Progenics Pharmaceuticals Inc. Anti-CCR5 antibody
US20080107595A1 (en) * 2002-02-22 2008-05-08 Olson William C Anti-CCR5 antibody
US20080241135A1 (en) * 2005-07-22 2008-10-02 Progenics Pharmaceuticals, Inc. Methods for reducing viral load in HIV-1-infected patients
US20070026441A1 (en) * 2005-07-22 2007-02-01 Olson William C Methods for reducing viral load in HIV-1-infected patients
US20110200592A1 (en) * 2005-07-22 2011-08-18 Olson William C Methods For Reducing Viral Load in HIV-1 Infected Patients
US8821877B2 (en) 2005-07-22 2014-09-02 Cytodyn Inc. Methods for inhibiting HIV-1 replication involving the administration of an anti-CCR5 receptor monoclonal antibody and small molecule CCR5 receptor antagonist
US20110135634A1 (en) * 2007-07-19 2011-06-09 Olson William C Methods of inhibiting infection by diverse subtypes of drug-resistant hiv-1

Also Published As

Publication number Publication date
US20020155429A1 (en) 2002-10-24
US6344545B1 (en) 2002-02-05
US20080187539A1 (en) 2008-08-07

Similar Documents

Publication Publication Date Title
US6344545B1 (en) Method for preventing HIV-1 infection of CD4+ cells
US6107019A (en) Method for preventing HIV-1 infection of CD4+ cells
AU728512B2 (en) Method for preventing HIV-1 infection of CD4+ cells
AU735460B2 (en) Uses of a chemokine receptor for inhibiting HIV-1 infection
Dragic et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5
Frade et al. The amino-terminal domain of the CCR2 chemokine receptor acts as coreceptor for HIV-1 infection.
Willett et al. Shared usage of the chemokine receptor CXCR4 by the feline and human immunodeficiency viruses
US20060194244A1 (en) Uses of a chemokine receptor for inhibiting HIV-1 infection
Deng et al. Identification of a major co-receptor for primary isolates of HIV-1
Trkola et al. Genetic subtype-independent inhibition of human immunodeficiency virus type 1 replication by CC and CXC chemokines
US7935797B2 (en) CCR5 chemokine receptor-specific monoclonal antibodies capable of inhibiting HIV-1 cell fusion
Mills et al. Chemokine receptor-directed agents as novel anti-HIV-1 therapies
AU2004233505B2 (en) Method for preventing HIV-1 infection of CD4+ cells
AU776239B2 (en) Method for preventing HIV-1 infection of CD4+ cells
JP4116087B2 (en) Novel mouse CXC chemokine receptor
AU2004205143A1 (en) Uses of a chemokine receptor for inhibiting HIV-1 infection
Cormier et al. An overview of HIV-1 coreceptor function and its inhibitors
MXPA98010425A (en) Uses of a chemiocine receiver to inhibit theinfection with human immunodeficiency viruses
Le et al. The role of chemokine receptors in the promotion of viral infections
Combadiere et al. Identification of CX3CR1

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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