WO2012055980A1 - Antibodies for the treatment of hiv - Google Patents

Antibodies for the treatment of hiv Download PDF

Info

Publication number
WO2012055980A1
WO2012055980A1 PCT/EP2011/068905 EP2011068905W WO2012055980A1 WO 2012055980 A1 WO2012055980 A1 WO 2012055980A1 EP 2011068905 W EP2011068905 W EP 2011068905W WO 2012055980 A1 WO2012055980 A1 WO 2012055980A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
cdr
hiv
seq
cxcr4
Prior art date
Application number
PCT/EP2011/068905
Other languages
English (en)
French (fr)
Inventor
Christine Klinguer-Hamour
Original Assignee
Pierre Fabre Medicament
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US12/913,300 external-priority patent/US9090686B2/en
Priority to BR112013009984A priority Critical patent/BR112013009984A2/pt
Priority to RU2013122770/10A priority patent/RU2013122770A/ru
Priority to CA 2814908 priority patent/CA2814908A1/en
Priority to KR20137011891A priority patent/KR20140009174A/ko
Priority to AU2011322508A priority patent/AU2011322508A1/en
Application filed by Pierre Fabre Medicament filed Critical Pierre Fabre Medicament
Priority to US13/823,182 priority patent/US20130174288A1/en
Priority to CN2011800520640A priority patent/CN103180342A/zh
Priority to EP11779612.8A priority patent/EP2632953A1/en
Priority to MX2013004710A priority patent/MX2013004710A/es
Priority to JP2013535444A priority patent/JP2014504147A/ja
Publication of WO2012055980A1 publication Critical patent/WO2012055980A1/en
Priority to ZA2013/02639A priority patent/ZA201302639B/en

Links

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
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • 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

Definitions

  • the present invention relates to novel antibodies, in particular murine monoclonal antibodies, chimeric and humanized, able to bind specifically to chemokine receptors (CXCR) as well as the amino and nucleic acid sequences coding for such antibodies.
  • CXCR chemokine receptors
  • the invention relates to novel antibodies, functional fragments or derivatives, able to bind specifically to the CXCR4 and having strong activity against human immunodeficiency virus (HIV) infection.
  • the invention also comprises the use of such antibodies, functional fragments or derivatives as a medicament for the preventive and/or therapeutic treatment of HIV infection.
  • Chemokines are small, secreted peptides that control the migration of leukocytes along a chemical gradient of ligand, known as chemokine gradient, especially during immune reactions (Zlotnick A. et al, 2000). They are divided into two major subfamilies, CC and CXC, based on the position of their H 2 -terminal cysteine residues, and bind to G protein coupled receptors, whose two major sub families are designated CCR and CXCR. More than 50 human chemokines and 18 chemokine receptors have been discovered so far.
  • chemokine receptor family functions as co-receptors with the primary receptor CD4 to allow entry of various strains of HIV type 1 into the cells, the major co-receptors being CCR5 and CXCR4.
  • T-cell tropic X4 HIV-1 use CD4 and CXCR4 for the entry into the cells, whereas macrophage-tropic R5 HIV-1 use CD4 and CCR5.
  • Dual-tropic strains can use both CXCR4 and CCR5 as co-receptors.
  • CCR3, CCR2, CCR8, CXCR6, CXCR7, CX3CR1 among other chemokine receptors can function as co-receptors by more restricted subset of HIV strains.
  • CCL5 ligands for CCR5 are able to inhibit cell fusion and infection by various strains of HIV-1.
  • These findings have encouraged the development of anti-HIV therapeutics targeting chemokine receptors leading to the approval of maraviroc (CELSENTRI®), a small molecule antagonist of CCR5 in combination with other anti-HIV- 1 agents in patients infected by CCR5-tropic HIV-1.
  • maraviroc is neither used in patients infected by dual-tropic HIV-1 nor in patients infected by CXCR4-tropic HIV-1 (VIDAL 2009). So there is a clear medical need to extend this type of therapy in both X4-tropic and dual-tropic HIV infected patients by identifying CXCR4 antagonists able to inhibit X4-tropic HIV replication.
  • Chemokine receptor 4 (also known as fusin, CD 184, LESTR or HUMSTR) exists as two isoforms comprising 352 or 360 amino acids. Residue Asn l l i s glycosylated, residue Tyr21 is modified by the addition of a sulfate group and Cys 109 and 186 are bond with a disulfide bridge on the extracellular part of the receptor (Juarez J. et al, 2004).
  • This receptor is expressed by different kind of normal tissues, naive, non- memory T-cells, regulatory T cells, B-cells, neutrophils, endothelial cells, primary monocytes, dendritic cells, Natural Killer cells, CD34+ hematopoietic stem cells and at a low level in heart, colon, liver, kidneys and brain.
  • CXCR4 plays a key role in leukocytes trafficking, B cell lymphopoiesis and myelopoiesis.
  • CXCR4 The unique ligand of CXCR4 receptor described so far is the Stromal-cell- Derived Factor-1 (SDF-1) or CXCL12. SDF-1 is secreted in large amount in lymph node, bone marrow, liver, lung and to a less extent by kidneys, brain and skin. CXCR4 is also recognized by an antagonistic chemokine, the viral macrophage inflammatory protein II (vMIP-II) encoded by human herpesvirus type III.
  • SDF-1 Stromal-cell- Derived Factor-1
  • CXCL12 CXCL12.
  • SDF-1 is secreted in large amount in lymph node, bone marrow, liver, lung and to a less extent by kidneys, brain and skin.
  • CXCR4 is also recognized by an antagonistic chemokine, the viral macrophage inflammatory protein II (vMIP-II) encoded by human herpesvirus type III.
  • vMIP-II viral macrophage inflammatory protein II
  • CXCR4 receptor is the principal co-receptor for T- cell-tropic HIV-1 isolates (X4 viruses). Interfering with this receptor should inhibit X4 viruses replication in a very efficient way.
  • One of the inventive aspect of the present invention is to generate mouse monoclonal antibodies (Mabs) inhibiting HIV replication.
  • the invention encompasses a CXCR4 Mab 515H7 (or fragments thereof) able to bind to CXCR4 homodimers, and having strong activities against HIV infection.
  • the invention also encompasses a CXCR4 Mab 301aE5 (or fragments thereof) able to bind to CXCR4 homodimers, and having strong activities against HIV infection.
  • CXCR4 compound is one of the two human CXCR4 isoforms selected from the group consisting of:
  • chemokine (C-X-C motif) receptor 4 isoform b [Homo sapiens] having the sequence as depicted under the Genbank accession number P 003458 SEQ ID No. 27: MEGISIYTSDNYTEEMGSGDYDSMKEPCFREENANFNKIFLPTIYSIIFLTGIVGN GLVILVMGYQKKLRSMTDKYRLHLSVADLLFVITLPFWAVDAVANWYFGNFL CKAVHVIYTVNLYSSVLILAFISLDRYLAIVHATNSQRPRKLLAEKVVYVGVWI PALLLTIPDFIFANVSEADDRYICDRFYPNDLWVVVFQFQHIMVGLILPGIVILSC YCIIISKLSHSKGHQKRKALKTTVILILAFFACWLPYYIGISIDSFILLEIIKQGCEFE NTVHKWISITEALAFFHCCLNPILYAFLGAKFKTSAQHALTSVSRGSSLKILSKG KRGGH
  • chemokine (C-X-C motif) receptor 4 isoform a [Homo sapiens] having the sequence as depicted under the Genbank accession number NP 001008540 SEQ ID No. 28:
  • SDF-1 natural ligand stromal cell-derived factor- 1
  • CXCR2 is selected from the group consisting of:
  • the invention also comprises a method for selecting a compound having an anti- HIV activity or which can be used for the preparation of a composition for the treatment of HIV infection, characterized in that said method comprises the step of:
  • a subject of the present invention is a process for the generation and the selection of antibodies according to the invention.
  • the invention concerns a process for the selection of an anti CXCR4 antibody, or one of its functional fragments or derivatives, capable to inhibit HIV replication comprising the following steps:
  • PBMC peripheral blood mononuclear cells
  • step iii) testing the selected antibodies of step ii) and selecting antibodies capable to bind to CXCR4 homodimer, and then
  • step iv) testing the selected antibodies of step iii) and selecting antibodies capable to inhibit primary isolates X4-tropic HIV-1 replication in PBMC.
  • the invention concerns a process for the selection of an anti CXCR4 antibody, or one of its functional fragments or derivatives, capable to inhibit HIV replication comprising the following steps:
  • PBMC peripheral blood mononuclear cells
  • step iv) testing the selected antibodies of step iii) and selecting antibodies capable to inhibit primary isolates X4-tropic HIV-1 replication in PBMC and/or capable to inhibit primary isolates X4/R5-tropic HIV-1 replication in PBMC.
  • the generation of the antibodies can be realized by any method known by the man skilled in the art, such as for example, fusion of a myeloma cell with spleen cells from immunized mice or other species compatible with the selected myeloma cells [Kohler & Milstein, 1975, Nature, 256:495-497].
  • the immunized animals could include transgenic mice with human immunoglobulin loci which then directly produce human antibodies.
  • Another possible embodiment could consist in using phage display technologies to screen libraries.
  • the screening steps i) and ii) can be realized by any method or process known by the man skilled in the art.
  • ELISA ELISA
  • BIAcore immunohistochemistry
  • FACS analysis and functional screens can be mentioned ELISA, BIAcore, immunohistochemistry, western blot analysis using CXCR4 expressing cell membrane extracts or purified CXCR4, FACS analysis and functional screens.
  • a preferred process consists in a screen by FACS analysis on CXCR4 transfectant (step 1) and on at least PBMC (step 2) to be sure that the produced antibodies will be able to also recognize the native CXCR4 receptor conformation at the target cell surface. This process will be described more precisely in the following examples.
  • the screening step iii) can be realized by any method or process known by the man skilled in the art. As non limiting but preferred example, can be mentioned western blotting and/or immuno-precipitation techniques using antibodies of interest on membrane extract from CXCR4 transfected cells or PBMC.
  • the screening step iv) can be realized by any method or process known by the man skilled in the art.
  • a process consisting in screening antibodies for their ability to inhibit X4 primary HIV-1 and/or X4/R5 primary HIV-1 isolates replication in PBMC by using a protocol described by Holl et al. (J. Immunol. 2004, 173, 6274-83).
  • said step iii) consists in evaluating antibodies by BRET analysis on cells expressing CXCR4-RLuc/CXCR4-YFP and selecting antibodies capable to inhibit at least 40 %, preferably 45 %, 50 %, 55 % and most preferably 60 % of the BRET signal.
  • the BRET technology is a technology known as being representative of the protein dimerization [Angers et al., PNAS, 2000, 97:3684-89].
  • BRET Bioluminescence Resonance Energy Transfer
  • Rluc Renilla Luciferase
  • GFP Green Fluorescent Protein
  • YFP Yellow Fluorescent Protein
  • EYFP Enhanced Yellow Fluorescent Protein
  • This property is used to generate protein-protein interaction assays. Indeed, in order to study the interaction between two partners, the first one is genetically fused to the Renilla Luciferase and the second one to the yellow mutant of the GFP. Fusion proteins are generally, but not obligatory, expressed in mammalian cells. In presence of its membrane permeable substrate (coelenterazine), Rluc emits blue light. If the GFP mutant is closer than 10 nm from the Rluc, an energy transfer can occur and an additional yellow signal can be detected. The BRET signal is measured as the ratio between the light emitted by the acceptor and the light emitted by the donor. So the BRET signal will increase as the two fusion proteins are brought into proximity or if a conformational change brings Rluc and GFP mutant closer.
  • step iii) consists in evaluating antibodies by BRET analysis on cells expressing both CXCR4- RLuc/CXCR4-YFP and selecting antibodies capable to inhibit at least 40 %, of the BRET signal.
  • a subject of the invention is isolated antibodies, or one of their functional fragments or derivatives, being obtained by said process.
  • Said antibodies or one of their said fragments or derivatives are capable of binding specifically to the human CXCR4, said antibodies being also capable to induce CXCR4 homodimers conformational changes.
  • CXCR4 Mabs like, for example clone A 120, are able to inhibit HIV-1 laboratory strain (X4HIV-1 NL4- 3 ) entry into PBMC (Tanaka R. et al, J. Virol. 2001, 75, 11534-11543). Furthermore, it is also known that CXCR4 Mabs are able to inhibit HIV-1 X4 primary isolates into cell lines expressing CXCR4. On the contrary, it has never been disclosed antibody able to inhibit such virus in their natural environment, i.e. not only on laboratory viruses or cell lines. Nevertheless, it is a novel and not obvious aspect of the invention that CXCR4 Mabs are able to inhibit HIV-1 X4 primary isolates into PBMC.
  • the invention does not relate to the antibodies in natural form, that is to say they are not in their natural environment but that they have been able to be isolated or obtained by purification from natural sources, or else obtained by genetic recombination, or by chemical synthesis, and that they can then contain unnatural amino acids as will be described further on.
  • antibodies or one of their functional fragments or derivatives, said antibodies being characterized in that they comprise at least one complementary determining region CDR chosen from CDRs comprising the amino acid sequence SEQ ID Nos. 1 to 6 and 30 to 33 as defined by the IMGT numbering system.
  • the invention relates to an isolated antibody, or a functional fragment or derivative of same, comprising at least one CDR chosen among the CDRs of sequences SEQ ID Nos. 1 to 6, as defined according to the IMGT numbering system, or at least one CDR whose sequence has at least 80%, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID Nos. 1 to 6.
  • the invention relates to an isolated antibody, or a functional fragment or derivative of same, comprising at least one CDR chosen among the CDRs of sequences SEQ ID Nos. 1, 2 and 30 to 33, as defined according to the IMGT numbering system, or at least one CDR whose sequence has at least 80%>, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID Nos. 1, 2 and 30 to 33.
  • Such functional fragments will be described in detail later in the present description.
  • a “derived compound” or “derivative” of an antibody means in particular a binding protein composed of a peptide scaffold and at least one of the CDRs of the original antibody in order to preserve its ability to recognize CXCR4.
  • Such derived compounds well-known to a person skilled in the art, will be described in more detail later in the present description.
  • the invention comprises the antibodies, their derived compounds or their functional fragments, according to the present invention, notably chimeric or humanized, obtained by genetic recombination or chemical synthesis.
  • the antibody according to the invention is characterized in that it consists of a monoclonal antibody.
  • a monoclonal antibody is understood to mean an antibody arising from a nearly homogeneous antibody population. More particularly, the individual antibodies of a population are identical except for a few possible naturally-occurring mutations which can be found in minimal proportions.
  • a monoclonal antibody consists of a homogeneous antibody arising from the growth of a single cell clone (for example a hybridoma, a eukaryotic host cell transfected with a DNA molecule coding for the homogeneous antibody, a prokaryotic host cell transfected with a DNA molecule coding for the homogeneous antibody, etc.) and is generally characterized by heavy chains of one and only one class and subclass, and light chains of only one type.
  • Monoclonal antibodies are highly specific and are directed against a single antigen.
  • each monoclonal antibody is directed against a single epitope of the antigen.
  • the antibody, or its derived compounds or functional fragments is characterized in that it comprises a light chain comprising at least one CDR chosen from CDR-L1, CDR-L2 and CDR-L3 , wherein :
  • - CDR-L1 comprises the amino acid sequence SEQ ID No. 1,
  • - CDR-L2 comprises the amino acid sequence SEQ ID No. 2,
  • - CDR-L3 comprises the amino acid sequence SEQ ID No. 3.
  • the antibodies of the invention are characterized in that they comprise a light chain comprising at least one of the three CDRs of the sequences SEQ ID Nos. l, 2 or 3, or at least one sequence with at least 80%, preferably 85%, 90%, 95% and 98%) identity after optimal alignment with sequences SEQ ID Nos. l, 2 or 3.
  • the antibody of the invention is also characterized in that it comprises a light chain comprising CDR-L1, CDR-L2 and CDR-L3, wherein CDR-L1 comprises the amino acid sequence SEQ ID No. 1, CDR-L2 comprises the amino acid sequence SEQ ID No. 2 and CDR-L3 comprises the amino acid sequence SEQ ID No. 3.
  • the antibody of the invention or one of its functional fragments or derivatives, is characterized in that it comprises a light chain of sequence comprising the amino acid sequence SEQ ID No. 7, or at least one sequence with at least 80%, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID No. 7.
  • the antibody, or its derived compounds or functional fragments is characterized in that it comprises a light chain comprising at least one CDR chosen from CDR-L1, CDR-L2 and CDR-L3, wherein: - CDR-L1 comprises the amino acid sequence SEQ ID No. 1,
  • - CDR-L2 comprises the amino acid sequence SEQ ID No. 2,
  • - CDR-L3 comprises the amino acid sequence SEQ ID No. 30.
  • the antibodies of the invention are characterized in that they comprise a light chain comprising at least one of the three CDRs of the sequences SEQ ID Nos. l, 2 or 30, or at least one sequence with at least 80%, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID Nos. l, 2 or 30.
  • the antibody of the invention is also characterized in that it comprises a light chain comprising CDR-L1, CDR-L2 and CDR-L3, wherein CDR-L1 comprises the amino acid sequence SEQ ID No. 1, CDR-L2 comprises the amino acid sequence SEQ ID No. 2 and CDR-L3 comprises the amino acid sequence SEQ ID No. 30.
  • the antibody of the invention is characterized in that it comprises a light chain of sequence comprising the amino acid sequence SEQ ID No. 34, or at least one sequence with at least 80%, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID No. 34.
  • the antibodies of the invention are characterized in that they comprise a heavy chain comprising at least one CDR chosen from CDR-H1, CDR-H2 and CDR-H3, wherein:
  • - CDR-H1 comprises the amino acid sequence SEQ ID No. 4,
  • - CDR-H2 comprises the amino acid sequence SEQ ID No. 5,
  • - CDR-H3 comprises the amino acid sequence SEQ ID No. 6.
  • the antibodies of the invention are characterized in that they comprise a heavy chain comprising at least one of the three CDRs of the sequences SEQ ID Nos. 4, 5 or 6, or at least one sequence with at least 80%, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID Nos. 4, 5 or 6.
  • antibodies or one of their derived compounds or functional fragments, are characterized in that they comprise a heavy chain comprising CDR-H1, CDR-H2 and CDR-H3, wherein CDR-H1 comprises the amino acid sequence SEQ ID No. 4, CDR-H2 comprises the amino acid sequence SEQ ID No. 5 and the CDR-H3 comprises the amino acid sequence SEQ ID No. 6.
  • the antibody of the invention is characterized in that it comprises a heavy chain of sequence comprising the amino acid sequence SEQ ID No. 8, or at least one sequence with at least 80%, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID No. 8.
  • the antibodies of the invention are characterized in that they comprise a heavy chain comprising at least one CDR chosen from CDR-H1, CDR-H2 and CDR-H3, wherein:
  • - CDR-H1 comprises the amino acid sequence SEQ ID No. 31,
  • - CDR-H2 comprises the amino acid sequence SEQ ID No. 32,
  • - CDR-H3 comprises the amino acid sequence SEQ ID No. 33.
  • the antibodies of the invention are characterized in that they comprise a heavy chain comprising at least one of the three CDRs of the sequences SEQ ID Nos. 31, 32 or 33, or at least one sequence with at least 80%, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID Nos. 31, 32 or 33.
  • antibodies or one of their derived compounds or functional fragments, are characterized in that they comprise a heavy chain comprising CDR-H1, CDR-H2 and CDR-H3, wherein CDR-H1 comprises the amino acid sequence SEQ ID No. 31, CDR-H2 comprises the amino acid sequence SEQ
  • the antibody of the invention, or one of its functional fragments or derivatives is characterized in that it comprises a heavy chain of sequence comprising the amino acid sequence SEQ ID No. 35, or at least one sequence with at least 80%, preferably 85%, 90%, 95% and 98% identity after optimal alignment with sequences SEQ ID No. 35.
  • the antibody of the invention, or one of its functional fragments or derivatives is characterized in that it comprises a light chain comprising CDR-L1, CDR-L2 and CDR-L3 comprising respectively the amino acid sequence SEQ ID Nos. 1, 2 and 3; and a heavy chain comprising CDR-H1, CDR-H2 and CDR-H3 comprising respectively the amino acid sequence SEQ ID Nos. 4, 5 and 6.
  • the antibody of the invention can also be characterized in that it comprises a light chain comprising the amino acid sequence SEQ ID No. 7 and a heavy chain comprising the amino acid sequence SEQ ID No. 8.
  • the antibody of the invention is characterized in that it comprises a light chain comprising CDR-L1, CDR-L2 and CDR-L3 comprising respectively the amino acid sequence SEQ ID Nos. 1, 2 and 30; and a heavy chain comprising CDR-H1, CDR-H2 and CDR-H3 comprising respectively the amino acid sequence SEQ ID Nos. 31, 32 and 33.
  • the antibody of the invention can also be characterized in that it comprises a light chain comprising the amino acid sequence SEQ ID No. 34 and a heavy chain comprising the amino acid sequence SEQ ID No. 35.
  • polypeptides In the present description, the terms “polypeptides”, “polypeptide sequences”, “peptides” and “proteins attached to antibody compounds or to their sequences” are interchangeable.
  • the invention does not relate to antibodies in natural form, i.e., they are not taken from their natural environment but are isolated or obtained by purification from natural sources or obtained by genetic recombination or chemical synthesis and thus they can carry unnatural amino acids as will be described below.
  • complementarity-determining region means the hypervariable regions of the heavy and light chains of immunoglobulins as defined by Kabat et al. (Kabat et al, Sequences of proteins of immunological interest, 5 th Ed., U.S. Department of Health and Human Services, NIH, 1991, and later editions). There are three heavy-chain CDRs and three light-chain CDRs.
  • CDR and “CDRs” are used to indicate, depending on the case, one or more, or even all, of the regions containing the maj ority of the amino acid residues responsible for the antibody's binding affinity for the antigen or epitope it recognizes.
  • CDR regions or CDR(s) it is intended to indicate the hypervariable regions of the heavy and light chains of the immunoglobulins as defined by IMGT.
  • the IMGT unique numbering has been defined to compare the variable domains whatever the antigen receptor, the chain type, or the species [Lefranc M.-P., Immunology Today 18, 509 (1997) / Lefranc M.-P., The Immunologist, 7, 132-136 (1999) / Lefranc, M.-P., Pommie, C, Ruiz, M., Giudicelli, V., Foulquier, E., Truong, L., Thouvenin-Contet, V. and Lefranc, Dev. Comp. Immunol, 27, 55-77 (2003)].
  • cystein 23 (lst-CYS), tryptophan 41 (CONSERVED-TRP), hydrophobic amino acid 89, cystein 104 (2nd-CYS), phenylalanine or tryptophan 118 (J-PHE or J- TRP).
  • the IMGT unique numbering provides a standardized delimitation of the framework regions (FR1-IMGT: positions 1 to 26, FR2-IMGT: 39 to 55, FR3-FMGT: 66 to 104 and FR4-IMGT: 118 to 128) and of the complementarity determining regions: CDR 1 -IMGT: 27 to 38, CDR2-IMGT: 56 to 65 and CDR3-IMGT: 105 to 117. As gaps represent unoccupied positions, the CDR-IMGT lengths (shown between brackets and separated by dots, e.g. [8.8.13]) become crucial information.
  • the IMGT unique numbering is used in 2D graphical representations, designated as IMGT Colliers de Pedes [Ruiz, M.
  • CDR or CDRs are used here in order to indicate, according to the case, one of these regions or several, or even the whole, of these regions which contain the maj ority of the amino acid residues responsible for the binding by affinity of the antibody for the antigen or the epitope which it recognizes.
  • IMGT numbering system defines the CDRs according to the IMGT system as above defined
  • Kabat numbering system defines the CDRs according to the Kabat system as above defined.
  • CDR-L1 consists of SEQ ID No. 1 in the IMGT numbering systems and of SEQ ID No. 9 in the Kabat numbering system.
  • CDR-L2 consists of SEQ ID No. 2 in the IMGT numbering systems and of SEQ ID No. 10 in the Kabat numbering system.
  • the CDR- L3 consists of SEQ ID No. 3 for each of the two numbering systems.
  • the CDR-Hl consists of the SEQ ID No. 4 in the IMGT numbering system and of SEQ ID No. 11 in the kabat numbering system.
  • the CDR-H2 consists of SEQ ID No.
  • the CDR-H3 consists in the SEQ ID No. 6 in the IMGT numbering systems whereas it consists of SEQ ID No. 13 in the kabat numbering system.
  • CDR-L1 consists of SEQ ID No. 1 in the IMGT numbering systems and of SEQ ID No. 9 in the Kabat numbering system.
  • CDR-L2 consists of SEQ ID No. 2 in the IMGT numbering systems and of SEQ ID No. 36 in the Kabat numbering system.
  • the CDR-L3 consists of SEQ ID No. 30 in the IMGT numbering systems and of SEQ ID No. 37 in the Kabat numbering system.
  • the CDR-Hl consists of the SEQ ID No. 31 in the IMGT numbering system and of SEQ ID No. 38 in the Kabat numbering system.
  • the CDR-H2 consists of SEQ ID No.
  • the CDR-H3 consists in the SEQ ID No. 33 in the IMGT numbering systems whereas it consists of SEQ ID No. 40 in the Kabat numbering system.
  • the "percentage identity" between two sequences of nucleic acids or amino acids means the percentage of identical nucleotides or amino acid residues between the two sequences to be compared, obtained after optimal alignment, this percentage being purely statistical and the differences between the two sequences being distributed randomly along their length.
  • the comparison of two nucleic acid or amino acid sequences is traditionally carried out by comparing the sequences after having optimally aligned them, said comparison being able to be conducted by segment or by using an "alignment window”.
  • Optimal alignment of the sequences for comparison can be carried out, in addition to comparison by hand, by means of the local homology algorithm of Smith and Waterman (1981) [Ad. App. Math.
  • the percentage identity between two nucleic acid or amino acid sequences is determined by comparing the two optimally-aligned sequences in which the nucleic acid or amino acid sequence to compare can have additions or deletions compared to the reference sequence for optimal alignment between the two sequences. Percentage identity is calculated by determining the number of positions at which the amino acid nucleotide or residue is identical between the two sequences, preferably between the two complete sequences, dividing the number of identical positions by the total number of positions in the alignment window and multiplying the result by 100 to obtain the percentage identity between the two sequences.
  • the BLAST program "BLAST 2 sequences” (Tatusova et al, "Blast 2 sequences - a new tool for comparing protein and nucleotide sequences", FEMS Microbiol, 1999, Lett. 174:247-2 5 0 ) av a i l ab l e o n t h e s i t e http://www.ncbi.nlm.nih.gov/gorf/bl2.html, can be used with the default parameters (notably for the parameters "open gap penalty”: 5, and “extension gap penalty”: 2; the selected matrix being for example the "BLOSUM 62" matrix proposed by the program); the percentage identity between the two sequences to compare is calculated directly by the program.
  • amino acid sequence exhibiting at least 80%, preferably 85%, 90%, 95% and 98%) identity with a reference amino acid sequence
  • preferred examples include those containing the reference sequence, certain modifications, notably a deletion, addition or substitution of at least one amino acid, truncation or extension.
  • substitutions are preferred in which the substituted amino acids are replaced by "equivalent” amino acids.
  • the expression “equivalent amino acids” is meant to indicate any amino acids likely to be substituted for one of the structural amino acids without however modifying the biological activities of the corresponding antibodies and of those specific examples defined below.
  • Equivalent amino acids can be determined either on their structural homology with the amino acids for which they are substituted or on the results of comparative tests of biological activity between the various antibodies likely to be generated.
  • table 1 summarizes the possible substitutions likely to be carried out without resulting in a significant modification of the biological activity of the corresponding modified antibody; inverse substitutions are naturally possible under the same conditions.
  • the present invention relates to murine antibodies, or derived compounds or functional fragments of same.
  • the invention also relates to any compound derived from antibodies as described in the invention.
  • an antibody of the invention is characterized in that said derived compound consists of a binding protein comprising a peptide scaffold on which is grafted at least one CDR in such a way as to preserve all or part of the paratope recognition properties of the initial antibody.
  • One or more sequences among the CDR sequences described in the present invention can also be present on the various immunoglobulin protein scaffolding.
  • the protein sequence makes it possible to recreate a peptide skeleton favorable to the folding of the grafted CDRs, enabling them to preserve their paratope antigen- recognition properties.
  • the origin of such protein scaffolds can be, but is not limited to, the structures selected among: fibronectin and preferentially fibronectin type III domain 10, lipocalin, anticalin (Skerra A., J. BiotechnoL, 2001, 74(4):257-75), protein Z arising from domain B of protein A of Staphylococcus aureus, thioredoxin A or proteins with a repeated motif such as the "ankyrin repeat” (Kohl et al, PNAS, 2003, vol. 100, No. 4, 1700- 1705), the "armadillo repeat", the "leucine-rich repeat” and the "tetratricopeptide repeat”.
  • fibronectin and preferentially fibronectin type III domain 10 lipocalin
  • anticalin Skerra A., J. BiotechnoL, 2001, 74(4):257-75
  • protein Z arising from domain B of protein A of Staphylococcus aureus
  • Scaffolds derived from toxins such as, for example, toxins from scorpions, insects, plants, mollusks, etc., and the protein inhibitors of neuronal NO synthase (PIN) should also be mentioned.
  • toxins such as, for example, toxins from scorpions, insects, plants, mollusks, etc.
  • PIN protein inhibitors of neuronal NO synthase
  • such peptide scaffolds can comprise at least one of the CDRs arising from the original antibody.
  • a person skilled in the art will select at least one CDR from the heavy chain, the latter being known to be primarily responsible for the specificity of the antibody.
  • the selection of one or more relevant CDRs is obvious to a person skilled in the art, who will then choose suitable known techniques (Bes et al, FEBS letters 508, 2001, 67-74).
  • a specific aspect of the present invention relates to a method for selecting a compound derived from an antibody according to the invention, said derived compound being capable of inhibiting in vitro and/or in vivo HIV cell entry and said derived compound comprising a peptide scaffold on which is grafted at least one antibody CDR, characterized in that it comprises the following steps:
  • said at least one grafted CDR is selected among the following CDRs of sequence SEQ ID No. 1 to 6 and 30 to 33 or a sequence with at least
  • the method can include in step a) the placing in contact in vitro of a compound comprising a peptide scaffold on which is grafted at least two or three antibody CDRs.
  • the peptide scaffold is selected among the scaffolds or binding proteins whose structures were mentioned above.
  • the present invention thus relates to an antibody, or its derived compounds or functional fragments, characterized in that the peptide scaffold is selected among proteins that are a) phylogenetically well preserved, b) of robust architecture, c) with a well-known 3-D molecular organization, d) of small size and/or e) comprising regions that can be modified by deletion and/or insertion without modifying stability properties.
  • the antibody of the invention is characterized in that said peptide scaffold is selected among i) scaffolds arising from fibronectin, preferentially fibronectin type 3 domain 10, lipocalin, anticalin, protein Z arising from domain B of protein A of Staphylococcus aureus, thioredoxin A or proteins with a repeated motif such as the "ankyrin repeat” (Kohl et al, PNAS, 2003, vol. 100, No. 4, 1700-1705), the "armadillo repeat", the "leucine-rich repeat” and the "tetratricopeptide repeat” or iii) protein inhibitors of neuronal NO synthase (PIN).
  • said peptide scaffold is selected among i) scaffolds arising from fibronectin, preferentially fibronectin type 3 domain 10, lipocalin, anticalin, protein Z arising from domain B of protein A of Staphylococcus aureus, thioredoxin A or proteins with a repeated motif such as
  • Another aspect of the invention relates to the functional fragments of the antibody described above.
  • the invention targets an antibody, or its derived compounds or functional fragments, characterized in that said functional fragment is selected among the fragments Fv, Fab, (Fab') 2 , Fab', scFv, scFv-Fc and diabodies, or any fragment whose half-life has been increased such as PEGylated fragments.
  • PEGylation polyethylene glycol
  • said functional fragments will comprise or include a partial sequence of the variable heavy or light chain of the antibody from which they are derived, said partial sequence being sufficient to retain the same binding specificity as the antibody from which it arises and sufficient affinity, preferably at least equal to 1/100, more preferably at least 1/10 of that of the antibody from which it arises.
  • Such a functional fragment will contain at least five amino acids, preferably 6, 7, 8, 10, 15, 25, 50 or 100 consecutive amino acids of the sequence of the antibody from which it arises.
  • these functional fragments will be of the types Fv, scFv, Fab, F(ab') 2 , F(ab'), scFv-Fc or diabodies, which generally have the same binding specificity as the antibody from which they result.
  • fragments of the antibody of the invention can be obtained from the antibodies described above by methods such as enzyme digestion, including pepsin or papain, and/or by cleavage of the disulfide bridges by chemical reduction.
  • the antibody fragments can be also obtained by recombinant genetics techniques also known to a person skilled in the art or by peptide synthesis by means, for example, of automatic peptide synthesizers such as those sold by Applied BioSystems, etc.
  • table 2 summarizes the various amino acid sequences corresponding to the antibodies of the invention.
  • a particular important additional aspect of the antibodies obj ect of the present invention is that they do not exhibit effector functions, such as antibody-dependent cellular cytotoxicity (ADCC) and/or complement-dependant cytotoxicity (CDC).
  • ADCC antibody-dependent cellular cytotoxicity
  • CDC complement-dependant cytotoxicity
  • the antibodies of the invention have no affinity for the FcyR (I, II or III) or for the Clq, or for both of them. Structurally, this means for the man skilled in the art that the antibodies of the invention, or one of their functional fragments or derivatives, are devoid of Fc portion or their Fc portion does not present a correct glycosylation able to confer effector functions.
  • the antibodies of the invention are preferably selected form IgG4 or IgG2 isotypes, most preferably IgG4.
  • preferred fragments are fragments devoid of ADCC such as Fv, scFv (sc for single chain), Fab, F(ab') 2 , Fab', scFv-Fc fragments or diabodies, or any fragment of which the half-life time would have been increased by chemical modification, such as the addition of poly(alkylene) glycol such as poly(ethylene) glycol (“PEGylation") (pegylated fragments called Fv-PEG, scFv-PEG, Fab-PEG, F(ab') 2 -PEG or Fab'-PEG) ("PEG” for Poly(Ethylene) Glycol), or by incorporation in a liposome.
  • poly(alkylene) glycol such as poly(ethylene) glycol (“PEGylation")
  • pegylated fragments called Fv-PEG, scFv-PEG, Fab-PEG, F(ab') 2 -PEG or Fab'-PEG)
  • a preferred functional fragment of the invention derived from the antibody 515H7 is a scFv, hereinafter referred as 515H7 scFv-Ck fragment, comprises the amino acid sequence SEQ ID No. 54
  • the nucleotide sequence corresponding to said scFv comprises the sequence SEQ ID No. 55.
  • Another specific aspect of the present invention relates to chimeric antibodies, or their derived compounds or functional fragments, characterized in that said antibodies also comprise light-chain and heavy-chain constant regions derived from an antibody of a species heterologous with the mouse, notably man.
  • Yet another specific aspect of the present invention relates to humanized antibodies, or their derived compounds or functional fragments, characterized in that the constant regions of the light-chain and the heavy-chain derived from human antibody are, respectively, the lambda or kappa region and the gamma-2 or preferably gamma-4 region.
  • the antibody of the invention also comprises chimeric or humanized antibodies.
  • a chimeric antibody is one containing a natural variable region (light chain and heavy chain) derived from an antibody of a given species in combination with constant regions of the light chain and the heavy chain of an antibody of a species heterologous to said given species.
  • the antibodies, or chimeric fragments of same can be prepared by using the techniques of recombinant genetics.
  • the chimeric antibody could be produced by cloning recombinant DNA containing a promoter and a sequence coding for the variable region of a nonhuman monoclonal antibody of the invention, notably murine, and a sequence coding for the human antibody constant region.
  • a chimeric antibody according to the invention coded by one such recombinant gene could be, for example, a mouse-human chimera, the specificity of this antibody being determined by the variable region derived from the murine DNA and its isotype determined by the constant region derived from human DNA.
  • Verhoeyn et al. BioEssays, 8:74, 1988 for methods for preparing chimeric antibodies.
  • the table 3 herein-under summarizes the amino acids sequences of the various heavy and light chains of the chimeric antibody 515H7 (referred as c515H7 or C515H7) according to the invention.
  • the nucleotide sequence corresponding to said Antibody c515H7 heavy chains SEQ ID Nos. 56 to 58 and light chain SEQ ID No. 59 correspond respectively to the sequence SEQ ID Nos. 60 to 63 (heavy chains) and SEQ ID No. 64 (light chain) .
  • the heavy chain sequences are deleted from their C- terminal lysine residue (as found in the original pConPlus vector series from Lonza: pConPlusy4AK, pConPlusy4PROAK & pConPlusy2AK).
  • the G4PRO heavy chain corresponds to a human IgG4 isotype carrying a mutation in the Hinge region to avoid formation of half-antibodies.
  • This mutation is found in the parental pConPlusy4PROAK from Lonza [Angal S, King DJ, Bodmer MW, Turner A, Lawson AD, Roberts G, Pedley B, Adair JR. A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (IgG4) antibody. Mol Immunol. (1993);30(1): 105-108].
  • the invention relates to a chimeric antibody heavy chain characterized in that it comprises CDRs homologous to corresponding CDRs of an antibody derived from a different mammalian species, wherein said CDRs, according to IMGT, consist of CDR-H1, CDR-H2 and CDR-H3 comprising respectively the sequences SEQ ID Nos. 4, 5 and 6.
  • the invention relates to a chimeric antibody light chain characterized in that it comprises CDRs homologous to corresponding CDRs of an antibody derived from a different mammalian species, wherein said CDRs, according to IMGT, consist of CDR-Ll, CDR-L2 and CDR-L3 comprising respectively the sequences SEQ ID Nos. 1, 2 and 3.
  • the invention relates to a chimeric antibody, or a derived compound or functional fragment of same, characterized in that it comprises heavy and light chains each having CDRs homologous to corresponding CDRs of an antibody derived from a different mammalian species, wherein said CDRs, according to IMGT, consist of CDR-H1, CDR-H2 and CDR-H3 of the heavy chain comprising respectively the sequences SEQ ID Nos. 4, 5 and 6, and CDR-Ll, CDR-L2 and CDR-L3 of the light chain comprising respectively the sequences SEQ ID Nos. 1, 2 and 3.
  • the invention relates to a chimeric antibody, or a derived compound or functional fragment of same, comprising a heavy chain variable region of sequence consisting of SEQ ID No. 8, and a light chain variable region of sequence SEQ ID No. 7.
  • the invention relates to a chimeric antibody, or a derived compound or functional fragment of same, comprising a heavy chain of sequence selected from the group consisting of SEQ ID Nos. 56, 57 or 58, and a light chain of sequence SEQ ID No. 59.
  • the chimeric antibody c515H7 VH(G4wt) / VL-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain variable region of sequence SEQ ID No. 56, and a light chain variable region of sequence SEQ ID No. 59.
  • the chimeric antibody c515H7 VH(G4PRO) / VL- Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain variable region of sequence SEQ ID No. 57, and a light chain variable region of sequence SEQ ID No. 59.
  • the chimeric antibody c515H7 VH(G2wt) / VL-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain variable region of sequence SEQ ID No. 58, and a light chain variable region of sequence SEQ ID No. 59.
  • Humanized antibodies means an antibody that contains CDR regions derived from an antibody of nonhuman origin, the other parts of the antibody molecule being derived from one (or several) human antibodies.
  • some of the skeleton segment residues can be modified to preserve binding affinity (Jones et al, Nature, 321 :522-525, 1986; Verhoeyen et al, Science, 239: 1534-1536, 1988; Riechmann et al, Nature, 332:323-327, 1988).
  • humanized antibodies of the invention or fragments of same can be prepared by techniques known to a person skilled in the art (such as, for example, those described in the documents Singer et al, J. Immun., 150:2844-2857, 1992; Mountain et al, Biotechnol. Genet. Eng. Rev., 10: 1-142, 1992; and Bebbington et al, Bio/Technology, 10: 169-175, 1992).
  • Such humanized antibodies are preferred for their use in methods involving in vitro diagnoses or preventive and/or therapeutic treatment in vivo.
  • the invention also relates to humanized antibodies arising from the murine antibody described above.
  • constant regions of the light-chain and the heavy-chain derived from human antibody are, respectively, the lambda or kappa and the gamma-2 or preferably gamma-4 region.
  • the invention relates to a humanized antibody heavy chain characterized in that it comprises i) a framework region homologous to corresponding framework region of a human antibody heavy chain, and ii) CDRs homologous to corresponding CDRs of an antibody derived from a different mammalian species, wherein said CDRs, according to IMGT, consist of CDR-H1, CDR-H2 and CDR-H3 comprising respectively the sequences SEQ ID Nos. 4, 5 and 6.
  • the invention relates to a humanized antibody heavy chain comprising a variable region of sequence consisting of SEQ ID No. 64.
  • the invention relates to a humanized antibody heavy chain comprising the complete sequence selected from the group consisting of SEQ ID Nos. 67, 68, 69 and 95.
  • the invention relates to a humanized antibody light chain characterized in that it comprises i) a framework region homologous to corresponding framework region of a human antibody light chain, and ii) CDRs homologous to corresponding CDRs of an antibody derived from a different mammalian species, wherein said CDRs, according to IMGT, consist of CDR-L1, CDR-L2 and CDR-L3 comprising respectively the sequences SEQ ID Nos. 1, 2 and 3.
  • the invention relates to a humanized antibody light chain comprising a variable region of sequence selected from the group consisting of SEQ ID Nos. 65, 66, 82 or 83.
  • the invention relates to a humanized antibody light chain comprising the complete sequence selected from the group consisting of SEQ ID Nos. 70, 71, 84 or 85.
  • the invention relates to a humanized antibody, or a derived compound or functional fragment of same, characterized in that it comprises heavy and light chains each having i) framework regions homologous to corresponding framework regions of a human antibody, and ii) CDRs homologous to corresponding CDRs of an antibody derived from a different mammalian species, wherein said CDRs, according to IMGT, consist of CDR-H1, CDR-H2 and CDR-H3 of the heavy chain comprising respectively the sequences SEQ ID Nos. 4, 5 and 6, and CDR-L1, CDR-L2 and CDR- L3 of the light chain comprising respectively the sequences SEQ ID Nos. 1, 2 and 3.
  • the invention relates to a humanized antibody, or a derived compound or functional fragment of same, comprising a heavy chain variable region of sequence consisting of SEQ ID No. 64, and a light chain variable region of sequence selected from the group consisting of SEQ ID Nos. 65, 66, 82 or 83.
  • the invention relates to a humanized antibody, or a derived compound or functional fragment of same, comprising a heavy chain of sequence selected from the group consisting of SEQ ID Nos. 67, 68, 69, or 95 and a light chain of sequence selected from the group consisting of SEQ ID Nos. 70, 71, 84 or 85.
  • the humanized antibody Hz515H7 VH1 D76N (G4wt) / VL2-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 67, and a light chain of sequence SEQ ID No. 70.
  • the humanized antibody Hz515H7 VH1 D76N (G4PRO) / VL2-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 68, and a light chain of sequence SEQ ID No. 70.
  • the humanized antibody Hz515H7 VH1 D76N (G2wt) / VL2-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 69, and a light chain of sequence SEQ ID No. 70.
  • (G4wt) / VL2.1-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 67, and a light chain of sequence SEQ ID No. 71.
  • the humanized antibody Hz515H7 VH1 D76N (G4PRO) / VL2.1-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 68, and a light chain of sequence SEQ ID No. 71.
  • the humanized antibody Hz515H7 VH1 D76N (G2wt) / VL2.1-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ
  • the humanized antibody Hz515H7 VH1 D76N (G4wt) / VL2.2-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 67, and a light chain of sequence SEQ ID No. 84.
  • the humanized antibody in another preferred embodiment, the humanized antibody
  • Hz515H7 VH1 D76N (G4PRO) / VL2.2-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 68, and a light chain of sequence SEQ ID No. 84.
  • the humanized antibody Hz515H7 VH1 D76N (G2wt) / VL2.2-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 69, and a light chain of sequence SEQ ID No. 84.
  • the humanized antibody Hz515H7 VH1 D76N (G4wt) / VL2.3-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 67, and a light chain of sequence SEQ ID No. 85.
  • the humanized antibody Hz515H7 VH1 D76N (G4PRO) / VL2.3-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 68, and a light chain of sequence SEQ ID No. 85.
  • the humanized antibody Hz515H7 VH1 D76N (G2wt) / VL2.3-Ck, or a derived compound or functional fragment of same, according to the invention comprises a heavy chain of sequence SEQ ID No. 69, and a light chain of sequence SEQ ID No. 85.
  • the table 4 herein-under summarizes the amino acids sequences of the various heavy and light chains variable domains and full length (or complete), respectively, of the humanized antibody 515H7 according to the invention.
  • the heavy chain sequences are deleted from their C- terminal lysine residue (as found in the original pConPlus vector series from Lonza: pConPlusy4AK, pConPlusy4PROAK & pConPlusy2AK).
  • the G4PRO heavy chain corresponds to a human IgG4 isotype carrying a mutation in the Hinge region to avoid formation of half-antibodies.
  • This mutation is found in the parental pConPlusy4PROAK from Lonza [Angal S, King DJ, Bodmer MW, Turner A, Lawson AD, Roberts G, Pedley B, Adair JR. A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (IgG4) antibody. Mol Immunol. (1993);30(1): 105-108].
  • a humanized antibody designated hz515H7 IgG4 which comprises a heavy chain of the human IgG4 isotype, said heavy chain having the sequence represented by SEQ ID NO: 95.
  • VH1 is similar to the expressions "VH Variant 1", “VH variant 1", “VH Var 1" or "VH var 1).
  • a novel aspect of the present invention relates to an isolated nucleic acid characterized in that it is selected among the following nucleic acids (including any degenerate genetic code):
  • nucleic acid DNA or RNA, coding for an antibody, or one of its functional fragments or derivatives, according to the invention
  • nucleic acid comprising a DNA sequence selecting from the group of sequences consisting of SEQ ID Nos. 14 to 19 and 41 to 45;
  • nucleic acid comprising a DNA sequence selecting from the group of sequences consisting of SEQ ID Nos. 20, 21, 46 and 47;
  • nucleic acid of at least 18 nucleotides capable of hybridizing under conditions of high stringency with at least one of the CDRs of sequence SEQ ID Nos. 14 to 19 and 41 to 45.
  • nucleic acid means a precise sequence of nucleotides, modified or not, defining a fragment or a region of a nucleic acid, containing unnatural nucleotides or not, and being either a double-strand DNA, a single- strand DNA or transcription products of said DNAs.
  • the present invention does not relate to nucleotide sequences in their natural chromosomal environment, i.e., in a natural state.
  • the sequences of the present invention have been isolated and/or purified, i.e., they were sampled directly or indirectly, for example by a copy, their environment having been at least partially modified.
  • Isolated nucleic acids obtained by recombinant genetics, by means, for example, of host cells, or obtained by chemical synthesis should also be mentioned here.
  • nucleic sequences exhibiting a percentage identity of at least 80%, preferably 85%, 90%), 95%) and 98%>, after optimal alignment with a preferred sequence means nucleic sequences exhibiting, with respect to the reference nucleic sequence, certain modifications such as, in particular, a deletion, a truncation, an extension, a chimeric fusion and/or a substitution, notably punctual.
  • these are sequences which code for the same amino acid sequences as the reference sequence, this being related to the degeneration of the genetic code, or complementarity sequences that are likely to hybridize specifically with the reference sequences, preferably under highly stringent conditions, notably those defined below.
  • Hybridization under highly stringent conditions means that conditions related to temperature and ionic strength are selected in such a way that they allow hybridization to be maintained between two complementarity DNA fragments.
  • the highly stringent conditions of the hybridization step for the purpose of defining the polynucleotide fragments described above are advantageously as follows.
  • DNA-DNA or DNA-RNA hybridization is carried out in two steps: ( 1 ) prehybridization at 42°C for three hours in phosphate buffer (20 mM, pH 7.5) containing 5X SSC (IX SSC corresponds to a solution of 0.15 M NaCl + 0.015 M sodium citrate), 50% formamide, 7% sodium dodecyl sulfate (SDS), 10X Denhardt's, 5%) dextran sulfate and 1% salmon sperm DNA; (2) primary hybridization for 20 hours at a temperature depending on the length of the probe (i.e.
  • the invention also encompasses an isolated nucleic acid molecule characterized in that it is selected among the following nucleic acids: a) a nucleic acid, DNA or RNA, coding for a humanized antibody heavy chain, or for a derived compound or functional fragment of same, according to the invention;
  • nucleic acid DNA or RNA, coding for a humanized antibody light chain, or for a derived compound or functional fragment of same, according to the invention
  • nucleic acid DNA or RNA, coding for a humanized antibody, or for a derived compound or functional fragment of same, according to the invention; d) a nucleic acid complementary to a nucleic acid as defined in a), b) or c);
  • nucleic acid of at least 18 nucleotides capable of hybridizing under highly stringent conditions with at least a heavy chain comprising the nucleic acid sequences SEQ ID No. 72 or 75 to 77;
  • nucleic acid of at least 18 nucleotides capable of hybridizing under highly stringent conditions with at least a light chain comprising the nucleic acid sequences SEQ ID No. 73, 74, 86, 87 or 78, 79, 88, 89.
  • the table 6 thereafter summarizes the nucleotide sequences of the various heavy and light chains variable domains and full length (or complete), respectively, of the humanized antibody 515H7 according to the invention.
  • the table 7 herein-under summarizes the nucleotide sequences of the various heavy and light chains of the chimeric antibody 515H7 according to the invention.
  • the invention deals with an isolated nucleic acid, characterized in that it is chosen from the following nucleic acids:
  • nucleic acid DNA or RNA, coding for an antibody, or one of its functional fragments or derivatives, according to the invention
  • nucleic acid comprising a DNA sequence selecting from the group of CDRs sequences consisting of SEQ ID Nos. 14 to 19 and 41 to 45;
  • nucleic acid comprising a DNA sequence selecting from the group of heavy and light variable domains sequences consisting of SEQ ID Nos. 20, 21, 46, 47, 72, 73,
  • nucleic acid comprising a DNA sequence selecting from the group of heavy and light chains sequences consisting of SEQ ID Nos. 60 to 63, 75 to 79, 88, 89 and 94; e) a nucleic acid comprising a DNA sequence consisting of SEQ ID No. 55; f) the corresponding RNA nucleic acids of the nucleic acids as defined in b), c), d) or e); g) the complementary nucleic acids of the nucleic acids as defined in a), b), c), d) and e); and
  • nucleic acid of at least 18 nucleotides capable of hybridizing under conditions of high stringency with at least one of the CDRs of sequence SEQ ID Nos. 14 to 19 and 41 to 45.
  • the invention also relates to a vector comprising a nucleic acid as described in the invention.
  • the invention notably targets cloning and/or expression vectors that contain such a nucleotide sequence.
  • the vectors of the invention preferably contain elements which allow the expression and/or the secretion of nucleotide sequences in a given host cell.
  • the vector thus must contain a promoter, translation initiation and termination signals, as well as suitable transcription regulation regions. It must be able to be maintained in a stable manner in the host cell and may optionally have specific signals which specify secretion of the translated protein.
  • These various elements are selected and optimized by a person skilled in the art according to the host cell used.
  • the nucleotide sequences can be inserted in self-replicating vectors within the chosen host or be integrative vectors of the chosen host.
  • Such vectors are prepared by methods typically used by a person skilled in the art and the resulting clones can be introduced into a suitable host by standard methods such as lipofection, electroporation, heat shock or chemical methods.
  • the vectors are, for example, vectors of plasmid or viral origin. They are used to transform host cells in order to clone or express the nucleotide sequences of the invention.
  • the invention also comprises host cells transformed by or comprising a vector as described in the present invention.
  • the host cell can be selected among prokaryotic or eukaryotic systems such as bacterial cells, for example, but also yeast cells or animal cells, notably mammal cells. Insect or plant cells can also be used.
  • prokaryotic or eukaryotic systems such as bacterial cells, for example, but also yeast cells or animal cells, notably mammal cells. Insect or plant cells can also be used.
  • the invention also relates to animals, other than man, that have a transformed cell according to the invention.
  • Another aspect of the invention relates to a method for the production of an antibody according to the invention, or one of its functional fragments, characterized in that said method comprises the following steps:
  • the transformed cells according to the invention are of use in methods for the preparation of recombinant polypeptides according to the invention.
  • Methods for the preparation of polypeptide according to the invention in recombinant form characterized in that said methods use a vector and/or a cell transformed by a vector according to the invention, are also comprised in the present invention.
  • a cell transformed by a vector according to the invention is cultured under conditions that allow the expression of the aforesaid polypeptide and recovery of said recombinant peptide.
  • the host cell can be selected among prokaryotic or eukaryotic systems.
  • a vector according to the invention carrying such a sequence can thus be used advantageously for the production of recombinant proteins to be secreted. Indeed, the purification of these recombinant proteins of interest will be facilitated by the fact that they are present in the supernatant of the cellular culture rather than inside host cells.
  • polypeptides of the invention can also be prepared by chemical synthesis.
  • One such method of preparation is also an object of the invention.
  • a person skilled in the art knows methods for chemical synthesis, such as solid-phase techniques (see notably Steward et al, 1984, Solid phase peptides synthesis, Pierce Chem. Company, Rockford, 111, 2nd ed.) or partial solid-phase techniques, by condensation of fragments or by conventional synthesis in solution.
  • Polypeptides obtained by chemical synthesis and capable of containing corresponding unnatural amino acids are also comprised in the invention.
  • the antibodies, or the derived compounds or functional fragments of same, likely to be obtained by the method of the invention are also comprised in the present invention.
  • the present invention relates to antibodies as described above, characterized in that they are, in addition, capable of specifically binding to a human chemokine family receptor and/or capable of specifically inhibiting X4-tropic HIV replication.
  • the present invention relates to antibodies as described above, characterized in that they are, in addition, capable of specifically binding to a human chemokine family receptor and/or capable of specifically inhibiting X4/R5 -tropic HIV replication.
  • the invention relates to antibodies, or their derived compounds or functional fragments, consisting of antibody that are bispecific in the sense that they comprise a second motif capable of interacting with any receptor implicated in HIV cell entry, such as, for example, CCR5, CD4, CXCR4 (other than the antibody of the present invention, i.e. targeting another epitope) or CCR3, CCR2, CCR8, CXCR6, CXCR7, CX3CR1.
  • a second motif capable of interacting with any receptor implicated in HIV cell entry such as, for example, CCR5, CD4, CXCR4 (other than the antibody of the present invention, i.e. targeting another epitope) or CCR3, CCR2, CCR8, CXCR6, CXCR7, CX3CR1.
  • the bispecific or bifunctional antibodies constitute a second generation of monoclonal antibodies in which two different variable regions are combined in the same molecule (Hollinger and Bohlen, 1999, Cancer and metastasis, rev. 18:411-419). Their utility was demonstrated in both diagnostic and therapeutic domains relative to their capacity to target several molecules on the surface of cells; such antibodies can be obtained by chemical methods (Glennie MJ et al., 1987, J. Immunol. 139, 2367-2375; Repp R. et al, 1995, J. Hemat., 377-382) or somatic methods (Staerz U.D. and Bevan M.J., 1986, PNAS 83, 1453-1457; Suresh M R.
  • bispecific antibodies can be constructed as whole IgG, bispecific Fab'2, Fab'PEG, diabodies or bispecific scFv, but also as a tetravalent bispecific antibody in which two binding sites are present for each antigen targeted (Park et al, 2000, Mol. Immunol, 37(18): 1123-30) or the fragments of same as described above.
  • the use of such bispecific antibodies has the advantage of reducing the treatment's toxicity. Indeed, the use of a bispecific antibody makes it possible to decrease the overall quantity of circulating antibodies and, consequently, possible toxicity.
  • the bispecific antibodies are bivalent or tetravalent antibodies.
  • the present invention relates to the antibodies described above, or one of their functional fragments or derivatives, as a medicament.
  • the invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising as an active ingredient a compound consisting of an antibody of the invention, or one of its functional fragments or derivatives.
  • said antibody is supplemented by an excipient and/or a pharmaceutically acceptable carrier.
  • the invention also relates to the composition as above described as a medicament.
  • the antibody, or one of its functional fragments or derivatives inhibits HIV-1 KON primary isolate replication in PBMC with an IC 90 of at least 5 ⁇ g/ml, preferably at least 10 ⁇ g/ml.
  • the present invention also comprises the use of an antibody or the composition according to the invention for the preparation of a drug and/or medicament for the prevention or the treatment of HIV infection.
  • said HIV infection is a X4-tropic HIV infection.
  • said HIV infectious is a
  • the present invention also relates to the use of an antibody, or a functional fragment or derivative of same, preferably humanized, and/or of a composition according to the invention for the preparation of a drug for inhibiting HIV replication.
  • the present invention relates to the use of an antibody, or a functional fragment or derivative of same, preferably humanized, and/or of a composition, for the preparation of a drug for HIV disease prevention or treatment.
  • pharmaceutical vehicle means a compound, or a combination of compounds, entering a pharmaceutical composition that does not cause secondary reactions and that, for example, facilitates administration of the active compounds, increases its lifespan and/or effectiveness in the organism, increases its solubility in solution or improves its storage.
  • Such pharmaceutical carriers are well- known and will be adapted by a person skilled in the art according to the nature and the administration route of the active compounds selected.
  • such compounds will be administered by systemic route, notably by intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, intravaginal or oral route. More preferably, the composition composed of the antibody according to the invention will be administered in several doses spaced equally over time.
  • Their administration routes, dosing schedules and optimal galenic forms can be determined according to the criteria generally taken into account when establishing a treatment suited to a patient such as, for example, the patient's age or body weight, the seriousness of his general state, his tolerance for the treatment and the side effects experienced.
  • the invention also relates to a composition
  • a composition comprising, in addition, as a combination product for use in a simultaneous, separated or extended fashion, an anti- HIV antibody or an anti-HIV cell entry antibody or an anti-HIV replication antibody other that an antibody directed against CXCR4.
  • the present invention also relates to a pharmaceutical composition as described above that comprises at least a second anti- HIV compound selected among the compounds capable of specifically inhibiting HIV entry and/or replication such as anti-CCR5, anti-CD4 compounds and anti-CXCR4 compounds other than those described in this invention, or any other anti-HIV compound known to a person skilled in the art.
  • a second anti- HIV compound selected among the compounds capable of specifically inhibiting HIV entry and/or replication such as anti-CCR5, anti-CD4 compounds and anti-CXCR4 compounds other than those described in this invention, or any other anti-HIV compound known to a person skilled in the art.
  • Another embodiment complementary to the invention consists of a composition as described above comprised of, in addition, as a combination or conjugation product for simultaneous, separated or extended use, an anti-HIV compound.
  • Simultaneous use means the administration of both compounds of the composition comprised in a single dosage form.
  • Separatated use means administration, at the same time, of both compounds of the composition, comprised in distinct dosage forms.
  • Extended use means the successive administration of both compounds of the composition, each comprised in a distinct dosage form.
  • composition according to the invention considerably increases
  • the therapeutic effect of the antibody of the invention is enhanced in an unexpected way by the administration of an anti-HIV agent.
  • Another major subsequent advantage produced by a composition of the invention relates to the possibility of using lower effective doses of the active ingredient, thus making it possible to avoid or reduce the risks of the appearance of side effects, in particular the effect of the anti-HIV agent.
  • this composition makes it possible to achieve the expected therapeutic effect more quickly.
  • Therapeutic anti-HIV agent means a substance which, when it is administered to a patient, treats or prevents the replication of HIV in the patient.
  • Non-limiting examples of such agents include "antiretroviral drugs such as HIV protease inhibitors (PI), nucleoside /nucleotide HIV reverse-transcriptase inhibitors (NRTI/NtRTI) non- nucleoside HIV reverse-transcriptase inhibitors (NNRTI), HIV entry inhibitors, HIV integrase inhibitors”.
  • Such agents for example, are cited in VIDAL, on the pages devoted to compounds related to "anti-HIV compounds"; the anti-HIV compounds cited by reference to this document are cited herein as non limitative preferred anti-HIV agents.
  • HIV protease inhibitor refers to any substance that can inhibit HIV protease activity.
  • HIV protease inhibitors include, but are not limited to Saquinavir mesylate or SQV (Invirase®), Indinavir or IDV (Crixivan®), Ritonavir or RTV (Norvir®), Nelfinavir or NFV (Viracept®), Amprenavir (Agenerase®, Prozei®),
  • Lopinavir/ritonavir or LPV/r Kaletra®, Aluvia®), Atazanavir or ATV (Reyataz®, Zrivada®), Fosamprenavir or FPV (Lexiva®, Telzir®), Tipranavir or TPV (Aptivus®), Darunavir or DRV (Prezista®)
  • NRTI nucleoside or nucleotide reverse-transcriptase inhibitor
  • examples of NRTI include, but are not limited to Zidovudine or AZT,ZDV (Retro vir/combivir/trixivir®), Didanosine or ddi (Videx®), Zalcitabine ( H I V I D ® ) , S t a v u d i n e o r d 4 T ( Z e r i t ® ) , L a m i v u d i n e o r 3 T C (Epivir/combivir/epzicom/trixivir®), Abacavir or ABC (Ziagen/trixivir/epzicom®), Tenofovir disoproxil fumarate or TDF (V
  • Non-nucleoside HIV reverse-transcriptase inhibitor refers to a substance which is not a nucleoside or nucleotide analogue that blocks reverse- transcription of HIV RNA.
  • NNRTI include, but are not limited to Nevirapine or NVP (Viramune®), Efavirenz or EFV (Sustiva/atripla®, Stocrin®), Delavirdine or DLV (Rescriptor®) and Etravirine or ETR (Intelence®).
  • HIV entry inhibitor refers to a substance that block HIV cell entry. Examples of
  • HIV entry inhibitors include, but are not limited to Enfuvirtide or T20 (Fuzeon®), Maraviroc or MVC (Celsentri®, Celzentry®).
  • HIV integrase inhibitor refers to a substance that inhibits HIV integrase activity.
  • Example of integrase inhibitor includes, but is not limited to Raltegravir or RAL (Isentress®).
  • Such agents are also compounds belonging to the same classes of drugs described in VIDAL, which are currently in clinical trials such as but not limited to Vicriviroc, PRO 140, TNX-355, AMD070, Racivir, Apricitabine, Elvucitabine, Flosalvudine, Rilpivirine, Elvitegravir.
  • Such agents are also compounds belonging to other potential classes of drugs such as but not limited to maturation inhibitors (Bevirimat), glycoside analogues of ⁇ -galactosyl-ceramide, carbohydrate-binding agents, RNaseH inhibitors, HIV gene expression inhibitors, stimulators of HIV release from latent T cells (valproic acid ).
  • maturation inhibitors Bevirimat
  • glycoside analogues of ⁇ -galactosyl-ceramide carbohydrate-binding agents
  • RNaseH inhibitors RNaseH inhibitors
  • HIV gene expression inhibitors stimulators of HIV release from latent T cells
  • composition of the invention as a combination product is characterized in that said anti-HIV agent is bound chemically to said antibody for use simultaneously.
  • spacer molecules can be introduced between the two compounds to bind, such as the poly(alkylene)glycol polyethyleneglycol or the amino acids; or, in another embodiment, said anti-HIV agents' active derivatives, into which have been introduced functions capable of reacting with said antibody, can be used.
  • binding techniques are well-known to a person skilled in the art and will not be discussed in more detail in the present description.
  • said antibody of the invention forming said conjugate is selected among its functional fragments, notably fragments that have lost their Fc component, such as scFv fragments.
  • the invention also relates to a composition as a combination product or to an anti-CXCR4 Mab/anti-HIV drug conjugate, according to the invention, used as drug.
  • composition as a combination product or said conjugate will be supplemented by an excipient and/or a pharmaceutical vehicle.
  • the invention relates to the use of an antibody, or one of its functional fragments or derivatives, for the preparation of a drug for the specific targeting of a compound that is biologically active toward HIV replication.
  • the invention also relates to a method for HIV prevention or treatment, wherein said method comprises a step consisting of administering to a patient in need thereof, an antibody, or one of its antigen binding fragments or derivatives and/or a composition, according to the invention.
  • the method according to the invention comprises also a step consisting of administering to said patient an anti-CCR5 compound, such as Maraviroc.
  • CXCR4 Mabs 515H7 and 301 aE5 have strong activities against HIV-1 replication in PBMC, so such antibodies could be used in screening assays for identification of CXCR4 antagonist antiviral agents to treat HIV-1 infection.
  • cells expressing CXCR4 are incubated with Mabs 515H7 and/or 301aE5 and then molecules can be evaluated for their potential to inhibit Mabs 515H7 and/or 301aE5 binding.
  • Cells used in this type of assays can be transfected cell lines such as CHO-CXCR4, NIH3 T3 -CXCR4 or CXCR4 transfected human cell lines such as U373-MAGI-CXCR4, human cell lines expressing CXCR4 such as NALM6 or primary cells such as PBMC.
  • the method used to screen antagonists of CXCR4 inhibiting Mabs 515H7 and/or 301aE5 binding on CXCR4 expressing cells can be cell-based competitive enzyme-linked immunosorbent Assay (ELISA) as described by Zhao Q. et al.
  • step e) can be added:
  • Figures 1 A and IB show the gating strategy for CXCR4 expression on monocytes and lymphocytes.
  • Figure 1 A T cell staining with CD3-PE antibody.
  • Figure IB monocytes staining with CD14-PE antibody.
  • Figures 2 A and 2B show the binding of anti-CXCR4 Mabs 515H7 and 301 aE5 on monocytes and T lymphocytes.
  • Figures 3 A and 3B show the modulation of CXCR4 receptor dimer by SDF-1 and by anti-CXCR4 Mabs 515H7 and 301 aE5, respectively, via a bioluminescence resonance energy transfer (BRET) approach in HEK293 cells.
  • BRET bioluminescence resonance energy transfer
  • Figures 4 A and 4B and Figure 5 show the ability of anti-CXCR4 Mab 515H7 and 301aE5 to inhibit HIV-1 isolate KON (X4 virus) replication in human PBMC.
  • Figures 6 A, 6B and 6C show the inhibition of SDF-1 -induced calcium release in CHO-CXCR4 cells by Mabs 515H7 (figure 6 A), 301 aE5 (figure 6B) and c515H7 (figure 6C).
  • Figures 7 and 8 show the ability of anti-CXCR4 Mabs 515H7, c515H7 and
  • FIG. 9 shows the ability of anti-CXCR4 Mabs 515H7, c515H7 and 301aE5 to inhibit HIV-1 X4 virus primary isolates KON, MN and 92UG024 replication in human PBMC.
  • Figures 10 and 1 1 show the ability of anti-CXCR4 Mabs 515H7, c515H7 and 301aE5 to inhibit the dual HIV-1 X4/R5 virus primary isolate 89.6 replication in human PBMC.
  • Figure 12 shows the beneficial effect of combining Mab c515H7 with Maraviroc to inhibit HIV-1 primary isolate 89.6 (dual X4/R5 virus) replication in human PBMC.
  • Figure 13 shows the beneficial effect of combining Mab c515H7 with Maraviroc to inhibit HIV-1 primary isolate UG93067 (dual X4/R5 virus) replication in human PBMC.
  • Figure 14 shows the ability of anti-CXCR4 Mabs 515H7, c515H7 and 301aE5 to inhibit HIV-1 X4 virus primary isolate KON replication in PBMC.
  • Figure 15 illustrates the binding specificity of the c515H7 Mab by FACS analysis.
  • Figure 16 illustrates the effect of c515H7 Mab on CXCR4 homodimer, by bioluminescence resonance energy transfer (BRET) approach.
  • Figure 17 Amino acid sequences alignment of 515H7 heavy chain variable domain with the human germline IGHV3-49*04 and IGHJ4*01.
  • the 515H7 VH amino acid sequence is aligned with the selected human acceptor framework sequences.
  • VH Varl (VH1) sequences correspond to the humanized variants of 515H7 VH domains.
  • the single back mutation in position 76 is depicted in bold.
  • Figure 18 Amino acid sequences alignment of 515H7 light chain with the human germline IGKV4-1 *01 and IGKJl *01. The 515H7 VL amino acid sequence is aligned with the selected human acceptor framework sequences.
  • Var2.3 sequences correspond to implemented humanized variants of the humanized 515H7 VL Var2, with mutated residues in bold.
  • Var2. 1 and Var2.2 carry 4 more humanized residues whereas Var2.3 contains 5 more human residues.
  • Figure 19 Cross blocking of the biotinylated murine antibody 515H7 by the chimeric 515H7 and different variants of the humanized 515H7.
  • the activity of the humanized variants of 515H7 (hz515H7) to cross block the parental murine antibody 515H7 was evaluated by flow cytometry using CXCR4 transfected NIH3T3 cells.
  • the activity of the humanized variants was compared to the chimeric 5 15H7.
  • the cross blocking activity of the variant VHl combined with the chimeric VL (cVL) was very similar to that of the chimeric (A). No reduction in the activity of VH variant 1 (VHl, the variant with no back mutations) was determined when combined with variant 2 of VL (B).
  • Figure 20 Inhibition of the biotinylated SDF-1 binding by the chimeric 515H7 and different variants of the humanized 515H7.
  • the capacity of the humanized variant of 515H7 (hz515H7) to inhibit SDF-1 binding was evaluated by flow cytometry using the cell line RAMOS.
  • the inhibition capacity of the humanized variants was compared to the chimeric 515H7.
  • the humanized variant hz515H7 VHl D76N VL2 has a similar capacity to inhibit SDF-1 binding as the chimeric antibody.
  • the humanized antibody fragment hz515 VHl VL2 was fully active in inhibiting the binding of SDF-1 to RAMOS cells.
  • Figure 21 shows humanized 515H7 Mabs (hz515H7 VHl D76N VL2, hz515H7 VHl D76N VL2. 1 , hz515H7 VHl D76N VL2.2 and hz5 15H7 VHl D76N VL2.3) specific binding to CXCR4 on NIH3T3-CXCR4.
  • Figure 22 shows the effect of humanized 515H7 Mabs (hz515H7 VHl D76N VL2, hz5 15H7 VHl D76N VL2. 1 , hz515H7 VHl D76N VL2.2 and hz5 15H7 VHl D76N VL2.3) on CXCR4 homodimer, by bioluminescence resonance energy transfer (BRET) approach.
  • BRET bioluminescence resonance energy transfer
  • Figure 23 shows the ability of anti-CXCR4 Mab hz515H7 to inhibit X4 HIV- IIIIB -induced cytopathogenicity in MT-4 cells.
  • Figure 24 shows the ability of anti-CXCR4 Mab hz515H7 to inhibit HIV-1 X4 virus primary isolate KON replication in human PBMC.
  • Figure 25 shows the beneficial effect of combining Mab hz5 15H7 with
  • Figure 26 shows the beneficial effect of combining Mab hz5 15H7 with Maraviroc to inhibit HIV-1 primary isolate UG93067 (dual X4/R5 virus) replication in human PBMC.
  • Figure 27 shows the ability of anti-CXCR4 Mab hz515H7 IgG4 to inhibit HIV-1 X4 virus primary isolate KON replication in human PBMC.
  • Figure 28 shows the beneficial effect of combining Mab hz515H7 IgG4 with Maraviroc to inhibit HIV-1 primary isolate 89.6 (dual X4/R5 virus) replication in human PBMC.
  • mice were immunized with recombinant NIH3T3-CXCR4 cells and/or peptides corresponding to CXCR4 extracellular N-term and loops. 6-16 weeks of age mice were immunized once with the antigen in complete Freund' s adjuvant subcutaneously (s.c.) followed by 2 to 6 immunizations with antigen in incomplete Freund's adjuvant s.c. The immune response was monitored by retroorbital bleeds. The serum was screened by ELISA (as described bellow) and mice with the higher titers of anti-CXCR4 antibodies were used for fusions. Mice were boost intravenously with antigen two days before sacrifice and removal of the spleen.
  • mice producing anti-CXCR4 antibodies sera from immunized mice was tested by ELISA. Briefly, microtiter plates were coated with purified [1-41] N-terminal peptide conjugated to BSA at 5 ⁇ g equivalent peptide/mL, ⁇ incubated at 4°C overnight, then blocked with 25C ⁇ L/well of 0.5% gelatine in PBS. Dilutions of plasma from CXCR4-immunized mice were added to each well and incubated 2 hours at 37°C. The plates were washed with PBS and then incubated with a goat anti-mouse IgG antibody conjugated to HRP (Jackson Laboratories) for 1 hour at 37°C. After washing, plates were developed with TMB substrate, the reaction was stopped 5 min later by addition of 100 ⁇ 1M H 2 S0 4 . Mice that developed the highest titers of anti-CXCR4 antibodies were used for antibody generation.
  • mice isolated from a Balb/c mice that developed the highest titers of anti-CXCR4 antibodies were fused with PEG to a mouse myeloma cell line
  • Sp2/0 Cells were plated at approximately lx 10 5 /well in microtiter plates followed by two weeks incubation in selective medium containing ultra culture medium + 2 mM L- glutamine + 1 mM sodium pyruvate + lx HAT. Wells were then screened by ELISA for anti-CXCR4 monoclonal IgG antibodies. The antibody secreting hybridomas were then subcloned at least twice by limiting dilution, cultured in vitro to generate antibody for further analysis.
  • Example 2 Characterization of anti-CXCR4 Mab 515H7 and 301aE5 binding specificity (NIH3T3-CXCR4 transfectant) by FACS analysis
  • NIH3T3 and NIH3T3-hCXCR4 transfected cells were incubated with 10 ⁇ / ⁇ 1 of monoclonal antibodies 5 1 5H7 and 301 aE5. The cells were then washed with 1%BSA/PBS/0.01% NaN3. Next, Alexa-labeled secondary antibodies were added to the cells and were allowed to incubate at 4°C for 20 min. The cells were then washed again two times. Following the second wash, FACS analysis was performed.
  • Example 3 Characterization of anti-CXCR4 Mab 515H7 and 301aE5 binding to peripheral blood mononuclear cells (PBMC) by FACS analysis
  • Blood was collected as buff coat from healthy donor. 100 ⁇ of whole blood was incubated with anti-human CXCR4 antibodies (clones 515H7 and 301aE5) at the indicated concentration for 20 minutes at 4°C. Blood was washed three times in PBS- BSA l%-NaN3 0.01% and incubated with goat anti-human Alexa 488 IgG diluted 1 :500 (Invitrogen) for 20 minutes at 4°C. Cells were then washed and incubated with CD14-PE (Caltag) or CD3-PE (Caltag) for 10 minutes at 4°C and washed three times. Red blood cells were lysed with High- Yield lyse solution (Caltag) for 10 minutes at room temperature.
  • the anti-human CXCR4 clones 515H7 and 301aE5 stained both T lymphocytes ( Figure 2 A) and monocytes ( Figure 2B) indicating that 5 15H7 and 301 aE5 Mabs are able to recognize the native form of CXCR4 expressed at the cell surface of monocytes and T lymphocytes.
  • Example 4 Effect of 515H7 and 301aE5 Mabs on CXCR4 homodimer, by bioluminescence resonance energy transfer (BRET) approach
  • This functional assay allows to evaluate the conformational changes induced upon SDF-1 and/or 515H7 Mab binding to CXCR4 receptor at the level of CXCR4 homodimer.
  • Expression vectors for the investigated interaction partners were constructed as fusion proteins with the corresponding dye (Renilla reniformis luciferase, Rluc and Yellow fluorescent protein, YFP) by applying conventional molecular biology techniques.
  • HEK293 cells were transiently transfected with expression vectors coding for the corresponding BRET partners: [CXCR4/Rluc + CXCR4/YFP] to study CXCR4 homodimerization.
  • the day after, cells were distributed in poly-lysine pre-coated white 96 MW plates in complete culture medium [DMEM supplemented with 10 % FBS]. Cells were first cultivated at 37°C with C0 2 5 % in order to allow cell attachment to the plate.
  • SDF 1 (300 nM) increased by about 20 % the BRET signal resulting from the spatial proximity of the adaptor and acceptor proteins fused to CXCR4 receptor, it is likely to indicate CXCR4/CXCR4 homo-dimers formation or conformational changes of pre-existing dimers ( Figures 3 A and B).
  • 5 15H7 and 301 aE5 Mabs were able to modulate SDF-1 -induced conformational changes for CXCR4 homo-dimers (69 % inhibition of SDF-1 -induced BRET increase for 515H7 and 301 aE5, Figures 3A and B).
  • PBMC from normal donors seronegative for HIV-1 were isolated from buffy coats or cytapheresis by Ficoll-Hypaque gradient centrifugation. PBMC were activated in the presence of PHA in RPMI 1640 cell culture medium containing 25 mM HEPES, 5 ml penicillin (10000 U/ml) - streptomycin (10000 ⁇ ) 2 mM L-glutamine, supplemented with 10% heat-inactivated FCS and were used as cellular targets in a single cycle neutralization assay. HIV-1 replication in primary human PBMC was performed by analyzing intracellular staining of viral p24 antigen by FACS analysis.
  • Control consisting of uninfected PBMC in medium without Mab was introduced.
  • intracellular staining of viral p24 antigen was performed and analyzed by fl ow cytometry .
  • C ell s were fixed and permeabilized using the Cytofix/Cytoperm kit (Becton Dickinson) according to the manufacturer protocols and stained with a fluorescent anti-p24 Mab (clone KC57 - Coulter Beckman) used at a 1/160 dilution incubated for 10 min at 4°C in the dark. After washing in PBS-3% FCS medium, PBMC were diluted in PBS before flow cytometry analysis.
  • the percentage of p24-positive cells in the different samples was determined by gating 20,000 events on a living cell population. The living cell subsets were analyzed for p24 expression relative to background staining of uninfected cells. The p24 antigen-positive value was obtained after subtraction of background events in mock-infected cells. The percentage of neutralization was defined as the reduction of p24-positive cells compared with control infected wells with no Mab. The neutralizing titer was defined as the dilution of Mab that allows a 90% decrease in the percentage of infected cells. The anti-CXCR4 Mabs 515H7 and 301aE5 were compared to 12G5 Mab known as the anti-CXCR4 Mab of reference for HIV application.
  • the anti-CXCR4 Mabs 515H7 and 301aE5 are able to inhibit HIV-1 KON primary isolate replication in PBMC with an IC 9 o of 10 ⁇ g/ml (66 nM) and 150 ⁇ g/ml (1 ⁇ ), respectively, whereas 12G5 Mab failed to inhibit HIV-1 KON primary isolate replication in PBMC ( Figure 4A).
  • This functional assay was designed to monitor CXCR4 receptor signaling via stimulation of the phospholipase C pathway, inducing calcium liberation from intracellular stores from the endoplasmic reticulum.
  • CHO-K1 cells stably and constitutively expressing human CXCR4 receptor were obtained upon transfection of naive CHO-K1 cells (ATCC CCL-61 ) with a mammalian expression vector carrying the whole coding sequence of human CXCR4 receptor (RefSeq NM 003467).
  • Cells were propagated in complete culture medium [DMEM-Ham's F12 supplemented with 5 % fetal calf serum (FCS) and 500 ⁇ ⁇ ⁇ of geneticin].
  • FCS fetal calf serum
  • Cells were plated in black 96MW plates at a density of 100,000 cells/well in appropriate culture medium. Cells were starved overnight before conducting the experiments.
  • Fluorescence in each well is recorded during 0.1 second every second and for a time period of 20 sec. prior SDF-1 injection (basal signal). Then 20 ⁇ of SDF-1 are injected and data recording follows for a time period of 2 min. Each experimental condition is performed in duplicate. Values for each well are first corrected by subtracting the basal fluorescence and the fluorescence emitted by a control well without cells. Relative data are expressed as a percentage of the maximal stimulation obtained by SDF-1 (100 nM).
  • SDF 1 (100 nM) induced a rapid and strong release of intracellular calcium in recombinant CHO/CXCR4, whereas no fluorescence signal was detected in naive CHO- Kl cells.
  • the maximal intensity reached > 140 % over basal fluorescence and was observed at about 40 sec. upon stimulation by SDF-1 ( Figures 6 A, 6B and 6C).
  • Mabs 515H7 (133 nM) ( Figure 6A) and c515H7 (133nM) ( Figure 6C) yielded a strong inhibition of the SDF-1 (100 nM)-induced calcium signal.
  • Mab 301 aE5 (133 nM) ( Figure 6B) yielded a partial inhibition of the SDF-1 (100 nM)-induced calcium signal.
  • Example 7 Inhibition of HIV-1 primary isolates KON, MN and 92UG024 (X4 viruses) replication in human PBMC by anti-CXCR4 Mabs 515H7, c515H7 and 301aE5
  • This assay is performed in 36 h using primary isolates KON, MN and 92UG024 concentrated and diluted accordingly to allow the detection 2% infected CD4 T lymphocytes after 2 days of infection. Twenty-five microliters of various dilutions of Mabs 515H7, c5 15H7 and 301aE5 were incubated for 1 h at 37°C with 25 ⁇ of virus. Human PBMC (25 ⁇ ) at 20 x 10 6 cells/ml were added to the Mab/virus mixture in a 96-well plate (U-bottom, Costar 3599) and cultured for 36 h in RPMI 1640 10% FCS and 20 U/ml IL-2 (R&D Systems, Minneapolis, MN).
  • HIV-infected lymphocytes were detected by intracellular staining of viral p24 Ag .
  • Cell s were fixed and permeabilized using b oth Cytofix/Cytoperm and Perm/Wash kits (BD Biosciences) according to the manufacturer and stained with a fluorescent anti-p24 Mab (FITC- or PE-anti-p24, clone KC57; Beckman Coulter/Immunotech, Hialeah, FL) used at a 1/160 dilution in Perm/Wash solution added for 15 min at 4°C.
  • FITC- or PE-anti-p24 clone KC57
  • Beckman Coulter/Immunotech Hialeah, FL
  • PBMC peripheral blood mononuclear cells
  • DIVA flow cytometry analysis
  • the percentage of p24-positive cells in the different samples was determined by gating 20,000 events on a living cell population identified by forward- and side-scatter parameters.
  • the living cell subsets were analyzed with the live/dead solution kit (Invitrogen). The p24 Ag-positive value was obtained after subtraction of background events in mock-infected cells.
  • the percent of neutralization was defined as the reduction of p24-positive cells compared with control infected wells with no Mab.
  • the neutralizing titer was defined as the concentration of antibody (interpolated between successive dilutions performed in triplicate) that allows a 90% decrease in the percentage of infected cells.
  • Example 8 Inhibition of HIV-1 primary isolate 89.6 (dual X4/R5 virus) replication in human PBMC by anti-CXCR4 Mabs 515H7, c515H7 and 301aE5
  • This assay is performed in 36 h using primary isolate 89.6 concentrated and diluted accordingly to allow the detection 2% infected CD4 T lymphocytes after 2 days of infection.
  • HIV-infected lymphocytes were detected by intracellular staining of viral p24 Ag .
  • Cell s were fixed and permeabilized using b oth Cytofix/Cytoperm and Perm/Wash kits (BD Biosciences) according to the manufacturer and stained with a fluorescent anti-p24 Mab (FITC- or PE-anti-p24, clone KC57; Beckman Coulter/Immunotech, Hialeah, FL) used at a 1/160 dilution in Perm/Wash solution added for 15 min at 4°C.
  • FITC- or PE-anti-p24 clone KC57
  • Beckman Coulter/Immunotech Hialeah, FL
  • PBMC peripheral blood mononuclear cells
  • DIVA flow cytometry analysis
  • the percentage of p24-positive cells in the different samples was determined by gating 20,000 events on a living cell population identified by forward- and side-scatter parameters.
  • the living cell subsets were analyzed with the live/dead solution kit (Invitrogen). The p24 Ag-positive value was obtained after subtraction of background events in mock-infected cells.
  • the percent of neutralization was defined as the reduction of p24-positive cells compared with control infected wells with no Mab.
  • the neutralizing titer was defined as the concentration of antibody (interpolated between successive dilutions performed in triplicate) that allows a 90% decrease in the percentage of infected cells.
  • Example 9 Inhibition of HIV-1 primary isolates 89.6 and UG93067 (dual
  • This assay that combines serial dilutions of c515H7 Mab or Maraviroc or combination of both with serial dilutions of virus, analyzes multiple rounds of infection on PBMC (peripheral blood mononuclear cells). Briefly, quadruplicate 25- ⁇ 1 aliquots of serial dilutions (twofold) of c515H7 Mabs or Maraviroc or combination of both were each incubated with 25 ⁇ of serial dilutions of virus in prehydrated 96-well filter plates (1.25 ⁇ mpore size, Durapor Dv; Millipore, Molsheim, France).
  • a control titration of the virus (25 ⁇ of RPMI replacing the diluted c515H7 Mabs or Maraviroc) was performed on the same plate as the titrations in the presence of dilutions of the c515H7 Mabs or Maraviroc or combination of both.
  • 25 ⁇ of PHA-stimulated PBMC at a concentration of 4 x 10 6 cells/ml (pool of PHA activated PBMC from five healthy donors) was added to achieve a 75- ⁇ 1 final culture volume of RPMI, 10% fetal calf serum (FCS), and 20 IU of interleukin-2 (IL-2) per ml (R&D System).
  • Quadruplicate wells were used to determine the viral titer (50% tissue culture infective dose [TCID 50 ]) in the absence (Vo) and in the presence (V favor) of each dilution of the c5 15H7 Mabs or Maraviroc or combination of both.
  • Chimeric format of murine 515H7 Mab was designed: it corresponds to the light and heavy chain variable domains of the murine antibody of interest, genetically fused to human Ckappa and IgGl/IgG2/IgG4 constant domains.
  • the recombinant Mab were produced upon transient transfection by using the HEK293/EBNA system with a pCEP4 expression vector (InVitrogen, US).
  • nucleotide sequences corresponding to the variable domains of 515H7 Mab light and heavy chains were synthesized by global gene synthesis (Genecust, Germany). They were subcloned into a pCEP4 vector (InVitrogen, US) carrying the entire coding sequence of the constant domain of either the light [Ckappa] or the heavy [CH1-Hinge-CH2-CH3] chain of a human IgGl/IgG2/IgG4 immunoglobulin. All cloning steps were performed according to conventional molecular biology techniques as described in the Laboratory manual (Sambrook and Russel, 2001) or according to the supplier' s instructions. Each genetic construct was fully validated by nucleotide sequencing using Big Dye terminator cycle sequencing kit (Applied Biosystems, US) and analyzed using a 3100 Genetic Analyzer (Applied Biosystems, US).
  • Suspension-adapted HEK293 EBNA cells were routinely grown in 250 ml flasks in 50 ml of serum-free medium Excell 293 (SAFC Biosciences) supplemented with 6 mM glutamine on an orbital shaker (110 rpm rotation speed). Transient transfection was performed with 2.10 6 cells/ml using linear 25 kDa polyethyleneimine (PEI) (Polysciences) prepared in water at a final concentration of 1 mg/ml mixed and plasmid DNA (final concentration of 1.25 ⁇ g/ml for heavy to light chain plasmid ratio of 1 : 1).
  • PEI polyethyleneimine
  • the culture was diluted with one volume of fresh culture medium to achieve a final cell density of 10 6 cells/ml. Cultivation process was monitored on the basis of cell viability and Mab production. Typically, cultures were maintained for 4 to 5 days.
  • Mab was purified using a conventional chromatography approach on a Protein A resin (GE Healthcare, US). Mab was produced at levels suitable with functional evaluations. Productivity levels are typically ranging between 6 and 15 mg/1 of purified Mab.
  • Example 11 Characterization of anti-CXCR4 chimeric Mab c515H7 binding specificity by FACS analysis
  • NIH3T3-hCXCR4 transfected cells were incubated with a dose range of monoclonal antibody c515H7 from 0 ⁇ g/ml to 10 ⁇ g/ml. The cells were then washed with 1%BSA/PBS/0.01% NaN3. Next, Alexa-labeled secondary antibodies were added to the cells and were allowed to incubate at 4°C for 20 min. The cells were then washed again two times. Following the second wash, FACS analysis was performed. Results of this binding study are provided in Figure 15 which shows that anti-CXCR4 chimeric Mab c515H7 bound specifically to human CXCR4-NIH3T3 transfected cell line.
  • This functional assay allows to evaluate the conformational changes induced upon SDF-1 and/or c515H7 Mab binding to CXCR4 receptor at the level of CXCR4 homodimer.
  • Expression vectors for the investigated interaction partners were constructed as fusion proteins with the corresponding dye (Renilla reniformis luciferase, Rluc and Yellow fluorescent protein, YFP) by applying conventional molecular biology techniques.
  • HEK293 cells were transiently transfected with expression vectors coding for the corresponding BRET partners: [CXCR4/Rluc + CXCR4/YFP] to study CXCR4 homodimerization.
  • the day after, cells were distributed in poly-lysine pre-coated white 96 MW plates in complete culture medium [DMEM supplemented with 10 % FBS]. Cells were first cultivated at 37°C with C0 2 5 % in order to allow cell attachment to the plate.
  • Example 13 In vitro evaluation of Mab 515H7 anti-HIV-1 activity using GFP-transduced human osteosarcoma (GHOST) cells expressing CD4 and CXCR4 or CCR5.
  • GHOST GFP-transduced human osteosarcoma
  • This assay is performed in 48 h using either the X4 HIV-1 LAI virus (with ghost cells expressing CXCR4) or the R5 HIV-1 BaL virus (with ghost cells expressing CCR5).
  • 500 ⁇ of ghost cells were plated (2.5 10 5 cells/ml) for 24h in Dulbecco culture medium supplemented with 10% FCS.
  • Various dilutions of Mab 515H7 were incubated for 1 h at 37°C and then diluted HIV-1 LAI virus (1/10) and HIV-1 BaL virus (1/7) were added to the cells for 48h. Cells were submitted to trypsin and washed with PBS IX.
  • Example 14 humanization of 515H7 anti-CXCR4 murine antibody and generation of a fragment of said h515H7 - General procedure
  • Binding of humanized variants of 515H7 was determined on a NIH3T3 cell line stably transfected with human CXCR4. The binding activity was evaluated by a competition assay with the biotinylated mouse antibody. In a second attempt, humanized antibodies were evaluated for their ability to inhibit binding of biotinylated SDF-1 to RAMOS cells. RAMOS cells were chosen because of their high expression of CXCR4 and low expression of CXCR7 and SDF- 1.
  • Recombi nants antib ody fragm ents were generated by PCR u si ng oligonucleotides specific for the variable domains of the humanized antibody and subcloning these into an E. coli system. Purification of antibody fragments was done by immobilized metal ion affinity chromatography (IMAC). - Humanization of 515H7 variable domains
  • VHl VH variant 1
  • the amino acid sequence of the variable domain of VHl comprises the SEQ ID No. 90 whereas the nucleotide sequence comprises the SEQ ID No. 91.
  • the amino acid sequence of the full length VHl comprises the SEQ ID No. 92 whereas the nucleotide sequence comprises the SEQ ID No. 93.
  • This construct showed similar capacity to compete with the murine antibody as the chimeric antibody ( Figure 19 A). This indicates that the most human VH variant has the same binding capacity as chimeric. Therefore, VHl was combined with the variant 2 of VL ( Figure 19B).
  • NIH3T3, NIH3T3-hCXCR4 transfected were incubated with 0 to 10 ⁇ g/mL of humanized Mabs 515H7 (hz515H7 VHl D76N VL2, hz515H7 VHl D76N VL2. 1, hz515H7 VHl D76N VL2.2, hz515H7 VHl D76N VL2.3) for 20 min at 4°C in the dark in 100 ⁇ Facs buffer. After 3 washing in Facs buffer, cells were incubated with the secondary antibody, a goat anti-human Alexa 488 (dilution 1/500), for 20 minutes at 4°C in the dark. After 3 washing in Facs buffer, propidium iodide was added in each well and only viable cells were analyzed by Facs. At least 5000 viable cells were assessed to evaluate the mean value of fluorescence intensity for each condition.
  • This functional assay allows to evaluate the conformational changes induced upon SDF-1 and/or hz515H7 VH1 D76N VL2, hz515H7 VH1 D76N VL2.1, hz515H7 VH1 D76N VL2.2, hz515H7 VH1 D76N VL2.3 binding to CXCR4 receptor at the level of CXCR4 homodimer.
  • Expression vectors for the investigated interaction partners were constructed as fusion proteins with the corresponding dye (Renilla reniformis luciferase, Rluc and Yellow fluorescent protein, YFP) by applying conventional molecular biology techniques.
  • HEK293 cells were transiently transfected with expression vectors coding for the corresponding BRET partners: [CXCR4/Rluc + CXCR4/YFP] to study CXCR4 homodimerization.
  • the day after, cells were distributed in poly-lysine pre-coated white 96 MW plates in complete culture medium [DMEM supplemented with 10 % FBS]. Cells were first cultivated at 37°C with C0 2 5 % in order to allow cell attachment to the plate. Cells were then starved with 200 ⁇ DMEM/well overnight. Immediately prior to the BRET experiment,
  • DMEM fetal calf serum
  • BRET ratio corresponds to the ratio 530/485 nm obtained when the two BRET partners are present, corrected by the ratio 530/485 nm obtained under the same experimental conditions, when only the partner fused to Rluc is present in the assay.
  • results are expressed in milliBRET units (mBU); mBU corresponds to the BRET ratio multiplied by 1000.
  • SDF 1 100 nM
  • SDF 1 increased by about 12 % the BRET signal resulting from the spatial proximity of the donor and acceptor proteins fused to CXCR4 receptor, it is likely to indicate CXCR4/CXCR4 homo-dimers formation or conformational changes of pre-existing dimers (Figure 22).
  • the activity of hz515H7 Mab against HIV- I HIB was based on the inhibition of virus-induced cytopathogenicity in MT-4 cells.
  • Cells were infected with HIV- I IIIB isolate at 5 times the tissue culture infective dose 50 (TCID 50 ) a virus dose which decreases the number of viable cells by 90% within 5 days. After adsorption at TCID 50 .
  • infected cells were adjusted to 2 10 5 cells/ml in RPMI 1640 medium supplemented with 20% heat-inactivated fetal calf serum (FCS), 100 IU/ml penicillin, 100 ⁇ g/ml streptomycin, 2 mM glutamine and seeded in 96-well flat-bottom tissue culture plates (COSTAR 3596) ( ⁇ /well) with 100 ⁇ of the hz515H7 Mab at various concentrations.
  • FCS heat-inactivated fetal calf serum
  • COSTAR 3596 96-well flat-bottom tissue culture plates
  • hz515H7 Mab displays a marked anti-HIV-1 activity since it was able to inhibit HIV-lm B -induced cytopathogenicity in MT-4 cells.
  • Example 18 Inhibition of HIV-1 primary isolate KON (X4 virus) replication in human PBMC by anti-CXCR4 Mab hz515H7
  • This assay was performed in 36 h using primary isolate KON concentrated and diluted accordingly to allow the detection 2 % infected CD4 T lymphocytes after 2 days of infection.
  • Human PBMC 25 ⁇
  • FCS 20 U/ml IL-2
  • HIV-infected lymphocytes were detected by intracellular staining of viral p24 Ag .
  • Cell s were fixed and permeabilized using b oth Cytofix/Cytoperm and Perm/Wash kits (BD Biosciences) according to the manufacturer and stained with a fluorescent anti-p24 Mab (FITC- or PE-anti-p24, clone KC57; Beckman Coulter/Immunotech, Hialeah, FL) used at a 1/160 dilution in Perm/Wash solution added for 15 min at 4°C. After washing in PBS with 3 % FBS, PBMC were diluted in 300 ⁇ of PBS before flow cytometry analysis (LSRII; BD Biosciences) with
  • the percentage of p24-positive cells in the different samples was determined by gating 20,000 events on a living cell population identified by forward- and side-scatter parameters.
  • the living cell subsets were analyzed with the live/dead solution kit (Invitrogen).
  • the p24 Ag-positive value was obtained after subtraction of background events in mock-infected cells.
  • the percent of neutralization was defined as the reduction of p24-positive cells compared with control infected wells with no Mab.
  • the neutralizing titer was defined as the concentration of antibody (interpolated between successive dilutions performed in triplicate) that allows a decrease in the percentage of infected cells.
  • the anti-CXCR4 Mab hz515H7 is able to inhibit HIV-1 X4 KON primary isolate replication in PBMC.
  • Example 19 Inhibition of HIV-1 primary isolates 89.6 and UG93067 (dual X4/R5 viruses) replication in human PBMC by anti-CXCR4 Mab hz515H7 combined with the anti-CCR5 molecule Maraviroc
  • This assay that combines serial dilutions of hz515H7 Mab or Maraviroc or combination of both with serial dilutions of virus, analyzes multiple rounds of infection on PBMC (peripheral blood mononuclear cells). Briefly, quadruplicate 25- ⁇ 1 aliquots of serial dilutions (twofold) of hz515H7 Mab or Maraviroc or combination of both were each incubated with 25 ⁇ of serial dilutions of virus in prehydrated 96-well filter plates (1.25 ⁇ mpore size, Durapor Dv; Millipore, Molsheim, France).
  • a control titration of the virus (25 ⁇ of RPMI replacing the diluted hz515H7 Mab or Maraviroc) was performed on the same plate as the titrations in the presence of dilutions of the hz515H7 Mab or Maraviroc or combination of both.
  • 25 ⁇ of PHA-stimulated PBMC at a concentration of 4 x 10 6 cells/ml (pool of PHA activated PBMC from five healthy donors) was added to achieve a 75- ⁇ 1 final culture volume of RPMI, 10 % fetal calf serum (FCS), and 20 IU of interleukin-2 (IL-2) per ml (R&D System).
  • Quadruplicate wells were used to determine the viral titer (50% tissue culture infective dose [TCID 50 ]) in the absence (Vo) and in the presence (V favor) of each dilution of the 515H7 Mab or Maraviroc or combination of both.
  • This assay was performed in 36 h using primary isolate KON concentrated and diluted accordingly to allow the detection 2% infected CD4 T lymphocytes after 2 days of infection.
  • HIV-infected lymphocytes were detected by intracellular staining of viral p24 Ag.
  • Cells were fixed and permeabilized using b oth Cytofix/Cytoperm and Perm/Wash kits (BD Biosciences) according to the manufacturer and stained with a fluorescent anti-p24 Mab (FITC- or PE-anti-p24, clone KC57; Beckman Coulter/Immunotech, Hialeah, FL) used at a 1/160 dilution in Perm/Wash solution added for 15 min at 4°C.
  • FITC- or PE-anti-p24 clone KC57
  • Beckman Coulter/Immunotech Hialeah, FL
  • PBMC peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • DIVA DIVA software
  • the percentage of p24-positive cells in the different samples was determined by gating 20,000 events on a living cell population identified by forward- and side-scatter parameters.
  • the living cell subsets were analyzed with the live/dead solution kit (Invitrogen).
  • the p24 Ag-positive value was obtained after subtraction of background events in mock-infected cells.
  • the percent of neutralization was defined as the reduction of p24-positive cells compared with control infected wells with no Mab.
  • the neutralizing titer was defined as the concentration of antibody (interpolated between successive dilutions performed in triplicate) that allows a decrease in the percentage of infected cells.
  • the anti-CXCR4 Mab hz515H7 IgG4 is able to inhibit
  • HIV-1 X4 KON primary isolate replication in PBMC HIV-1 X4 KON primary isolate replication in PBMC.
  • Example 21 Inhibition of HIV-1 primary isolate 89.6 (dual X4/R5 virus) replication in human PBMC by anti-CXCR4 Mab hz515H7 IgG4 combined with the anti-CCR5 molecule Maraviroc
  • This assay that combines serial dilutions of hz515H7 IgG4 Mab or Maraviroc or combination of both with serial dilutions of virus, analyzes multiple rounds of infection on PBMC (peripheral blood mononuclear cells). Briefly, quadruplicate 25- ⁇ 1 aliquots of serial dilutions (twofold) of hz515H7 IgG4 Mab or Maraviroc or combination of both were each incubated with 25 ⁇ of serial dilutions of virus in prehydrated 96-well filter plates (1.25- ⁇ pore size, Durapor Dv; Millipore, Molsheim, France).
  • a control titration of the virus (25 ⁇ of RPMI replacing the diluted hz515H7 IgG4 Mab or Maraviroc) was performed on the same plate as the titrations in the presence of dilutions of the hz515H7 IgG4 Mab or Maraviroc or combination of both.
  • 25 ⁇ of PHA-stimulated PBMC at a concentration of 4 x 10 6 cells/ml (pool of PHA activated PBMC from five healthy donors) was added to achieve a 75- ⁇ 1 final culture volume of RPMI, 10% fetal calf serum (FCS), and 20 IU of interleukin-2 (IL-2) per ml (R&D System).
  • Quadruplicate wells were used to determine the viral titer (50% tissue culture infective dose [TCID 50 ]) in the absence (Vo) and in the presence (V favor) of each dilution of the hz515H7 IgG4 Mab or Maraviroc or combination of both.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Communicable Diseases (AREA)
  • AIDS & HIV (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
PCT/EP2011/068905 2009-04-29 2011-10-27 Antibodies for the treatment of hiv WO2012055980A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
JP2013535444A JP2014504147A (ja) 2010-10-27 2011-10-27 Hivの治療用抗体
RU2013122770/10A RU2013122770A (ru) 2010-10-27 2011-10-27 Антитела для лечения вич
CA 2814908 CA2814908A1 (en) 2010-10-27 2011-10-27 Antibodies for the treatment of hiv
KR20137011891A KR20140009174A (ko) 2010-10-27 2011-10-27 Hiv의 치료를 위한 항체들
AU2011322508A AU2011322508A1 (en) 2010-10-27 2011-10-27 Antibodies for the treatment of HIV
BR112013009984A BR112013009984A2 (pt) 2009-04-29 2011-10-27 anticorpos para tratamento do hiv
US13/823,182 US20130174288A1 (en) 2009-04-29 2011-10-27 Antibodies for the treatment of hiv
CN2011800520640A CN103180342A (zh) 2010-10-27 2011-10-27 用于治疗hiv的抗体
EP11779612.8A EP2632953A1 (en) 2010-10-27 2011-10-27 Antibodies for the treatment of hiv
MX2013004710A MX2013004710A (es) 2010-10-27 2011-10-27 Anticuerpos para el tratamiento de vih.
ZA2013/02639A ZA201302639B (en) 2010-10-27 2013-04-12 Antibodies for the treatment of hiv

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US12/913,300 US9090686B2 (en) 2009-04-29 2010-10-27 Antibodies for the treatment of HIV
US12/913,300 2010-10-27

Publications (1)

Publication Number Publication Date
WO2012055980A1 true WO2012055980A1 (en) 2012-05-03

Family

ID=44913256

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/068905 WO2012055980A1 (en) 2009-04-29 2011-10-27 Antibodies for the treatment of hiv

Country Status (10)

Country Link
EP (1) EP2632953A1 (ko)
JP (1) JP2014504147A (ko)
KR (1) KR20140009174A (ko)
CN (1) CN103180342A (ko)
AU (1) AU2011322508A1 (ko)
CA (1) CA2814908A1 (ko)
MX (1) MX2013004710A (ko)
RU (1) RU2013122770A (ko)
WO (1) WO2012055980A1 (ko)
ZA (1) ZA201302639B (ko)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014134547A1 (en) * 2013-02-28 2014-09-04 Therabiol, Inc. Hiv antigens and antibodies
WO2017220988A1 (en) 2016-06-20 2017-12-28 Kymab Limited Multispecific antibodies for immuno-oncology

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3077417A1 (en) * 2013-12-08 2016-10-12 Peptcell Limited Hiv antigens and antibodies and compositions, methods and uses thereof

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0451216A1 (en) 1988-12-28 1991-10-16 Protein Design Labs Inc CHIMERAL IMMUNGLOBULINE, SPECIFIC FOR P55 TAC PROTEIN OF THE IL-2 RECEPTOR.
EP0566647A1 (en) 1990-12-19 1993-10-27 Protein Design Labs, Inc. Improved humanized immunoglobulins
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US5877293A (en) 1990-07-05 1999-03-02 Celltech Therapeutics Limited CDR grafted anti-CEA antibodies and their production
US5886152A (en) 1991-12-06 1999-03-23 Sumitomo Pharmaceuticals Company, Limited Humanized B-B10
US6054297A (en) 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
WO2004059285A2 (en) * 2002-12-23 2004-07-15 Protein Design Labs, Inc. Tumor killing/tumor regression using cxcr4 antagonists
EP2172485A1 (en) * 2008-10-01 2010-04-07 Pierre Fabre Medicament Novel anti CXCR4 antibodies and their use for the treatment of cancer
EP2246364A1 (en) * 2009-04-29 2010-11-03 Pierre Fabre Médicament Anti CXCR4 antibodies for the treatment of HIV
EP2371863A1 (en) * 2010-03-30 2011-10-05 Pierre Fabre Médicament Humanized anti CXCR4 antibodies for the treatment of cancer

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2010012435A (es) * 2008-05-14 2011-05-03 Lilly Co Eli Anticuerpos anti-cxcr4.
JP2012158521A (ja) * 2009-04-17 2012-08-23 Kureha Corp 外用剤

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0682040A1 (en) 1988-12-28 1995-11-15 Protein Design Labs, Inc. Humanized immunoglobulins and their production and use
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US6180370B1 (en) 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
US5693761A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Polynucleotides encoding improved humanized immunoglobulins
EP0451216A1 (en) 1988-12-28 1991-10-16 Protein Design Labs Inc CHIMERAL IMMUNGLOBULINE, SPECIFIC FOR P55 TAC PROTEIN OF THE IL-2 RECEPTOR.
EP0939127A2 (en) 1988-12-28 1999-09-01 Protein Design Labs, Inc. Humanized immunoglobulins and their production and use
US5877293A (en) 1990-07-05 1999-03-02 Celltech Therapeutics Limited CDR grafted anti-CEA antibodies and their production
EP0566647A1 (en) 1990-12-19 1993-10-27 Protein Design Labs, Inc. Improved humanized immunoglobulins
US6054297A (en) 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
US5886152A (en) 1991-12-06 1999-03-23 Sumitomo Pharmaceuticals Company, Limited Humanized B-B10
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
WO2004059285A2 (en) * 2002-12-23 2004-07-15 Protein Design Labs, Inc. Tumor killing/tumor regression using cxcr4 antagonists
EP2172485A1 (en) * 2008-10-01 2010-04-07 Pierre Fabre Medicament Novel anti CXCR4 antibodies and their use for the treatment of cancer
EP2246364A1 (en) * 2009-04-29 2010-11-03 Pierre Fabre Médicament Anti CXCR4 antibodies for the treatment of HIV
EP2371863A1 (en) * 2010-03-30 2011-10-05 Pierre Fabre Médicament Humanized anti CXCR4 antibodies for the treatment of cancer

Non-Patent Citations (49)

* Cited by examiner, † Cited by third party
Title
ANGAL S, KING DJ, BODMER MW, TURNER A, LAWSON AD, ROBERTS G, PEDLEY B, ADAIR JR: "A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (IgG4) antibody", MOLLMMUNOL, vol. 30, no. 1, 1993, pages 105 - 108, XP023683005, DOI: doi:10.1016/0161-5890(93)90432-B
ANGAL S, KING DJ, BODMER MW, TURNER A, LAWSON AD, ROBERTS G, PEDLEY B, ADAIR JR: "A single amino acid substitution abolishes the heterogeneity of chimeric mouse/human (IgG4) antibody", MOLLMMUNOL., vol. 30, no. 1, 1993, pages 105 - 108, XP023683005, DOI: doi:10.1016/0161-5890(93)90432-B
ANGERS ET AL., PNAS, vol. 97, 2000, pages 3684 - 89
BARIBAUD FREDERIC ET AL: "Antigenically distinct conformations of CXCR4", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 75, no. 19, 1 October 2001 (2001-10-01), pages 8957 - 8967, XP002234668, ISSN: 0022-538X, DOI: 10.1128/JVI.75.19.8957-8967.2001 *
BEBBINGTON ET AL., BIO/TECHNOLOGY, vol. 10, 1992, pages 169 - 175
BES ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 343, no. 1, 2006, pages 334 - 344
BES ET AL., FEBS LETTERS, vol. 508, 2001, pages 67 - 74
CARNEC XAVIER ET AL: "Anti-CXCR4 monoclonal antibodies recognizing overlapping epitopes differ significantly in their ability to inhibit entry of human immunodeficiency virus type 1", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 79, no. 3, 1 February 2005 (2005-02-01), pages 1930 - 1933, XP002518542, ISSN: 0022-538X, DOI: 10.1128/JVI.79.3.1930-1933.2005 *
GLENNIE MJ ET AL., J. IMMUNOL., vol. 139, 1987, pages 2367 - 2375
HOLL ET AL., J. IMMUNOL., vol. 173, 2004, pages 6274 - 83
HOLLINGER, BOHLEN, CANCER AND METASTASIS, 1999, pages 411 - 419
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
JUAREZ J. ET AL., LEUKEMIA, vol. 17, 2003, pages L294 - 1300
KAAS, Q., LEFRANC, M.-P., CURRENT BIOINFORMATICS, vol. 2, 2007, pages 21 - 30
KAAS, Q., RUIZ, M., LEFRANC, M.-P.: "T cell receptor and MHC structural data", NUCL. ACIDS. RES., vol. 32, 2004, pages D208 - D210
KABAT ET AL.: "Sequences of proteins of immunological interest", 1991, U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES, NIH
KOHL ET AL., PNAS, vol. 100, no. 4, 2003, pages 1700 - 1705
KOHLER, MILSTEIN, NATURE, vol. 256, 1975, pages 495 - 497
LEFRANC M.-P., IMMUNOLOGY TODAY, vol. 18, 1997, pages 509
LEFRANC M.-P., THE IMMUNOLOGIST, vol. 7, 1999, pages 132 - 136
LEFRANC, M.-P., POMMIÉ, C., RUIZ, M., GIUDICELLI, V., FOULQUIER, E., TRUONG, L., THOUVENIN-CONTET, V., LEFRANC, DEV., COMP. IMMUNOL., vol. 27, 2003, pages 55 - 77
MERCHAND ET AL., NATURE BIOTECH., vol. 16, 1998, pages 677 - 681
MOUNTAIN ET AL., BIOTECHNOL. GENET. ENG. REV., vol. 10, 1992, pages 1 - 142
MURGA JOSE D ET AL: "Potent antiviral synergy between monoclonal antibody and small-molecule CCR5 inhibitors of human immunodeficiency virus type 1", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 50, no. 10, October 2006 (2006-10-01), pages 3289 - 3296, XP002670018, ISSN: 0066-4804 *
NEDDLEMAN, WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
NICAISE ET AL., PROTEIN SCIENCE, vol. 13, no. 7, 2004, pages 1882 - 1891
PARK ET AL., MOL. IMMUNOL., vol. 37, no. 18, 2000, pages 1123 - 30
PEARSON, LIPMAN, PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444
REEVES J D ET AL: "The second extracellular loop of CXCR4 is involved in CD4-independent entry of human immunodeficiency virus type 2", JOURNAL OF GENERAL VIROLOGY, SOCIETY FOR GENERAL MICROBIOLOGY, SPENCERS WOOD, GB, vol. 79, no. 7, 1 January 1998 (1998-01-01), pages 1793 - 1799, XP002290004, ISSN: 0022-1317 *
REPP R. ET AL., J. HEMAT., 1995, pages 377 - 382
REY-CUILLE M A ET AL: "Conserved CXCR4 usage and enhanced replicative capacity of HIV-2/287, an isolate highly pathogenic in Macaca nemestrina", AIDS, LONDON, GB, vol. 15, no. 18, 7 December 2001 (2001-12-07), pages 2349 - 2357, XP002548958, ISSN: 0269-9370, DOI: 10.1097/00002030-200112070-00002 *
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 327
RUIZ, M., LEFRANC, M.-P., IMMUNOGENETICS, vol. 53, 2002, pages 857 - 883
SAMBROOK ET AL.: "Molecular cloning: a laboratory manual", 2001, COLD SPRING HARBOR LABORATORY
See also references of EP2632953A1 *
SINGER ET AL., J. IMMUN., vol. 150, 1992, pages 2844 - 2857
SKERRA A., J. BIOTECHNOL., vol. 74, no. 4, 2001, pages 257 - 75
SKERRA A., J. MOL. RECOGN., vol. 13, 2000, pages 167 - 187
SMITH, WATERMAN, AD. APP. MATH., vol. 2, 1981, pages 482
STAERZ U.D., BEVAN M.J., PNAS, vol. 83, 1986, pages 1453 - 1457
STEWARD ET AL.: "Solid phase peptides synthesis", 1984, PIERCE CHEM. COMPANY
STRIZKI J M ET AL: "A monoclonal antibody (12G5) directed against CXCR-4 inhibits infection with the dual-tropic human immunodeficiency virus type 1 isolate HIV-1(89.6) but not the T-tropic isolate HIV-1(HxB)", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 71, no. 7, 1 July 1997 (1997-07-01), pages 5678 - 5683, XP002548957, ISSN: 0022-538X, Retrieved from the Internet <URL:http://jvi.asm.org/cgi/reprint/71/7/5678.pdf> *
SURESH M.R. ET AL., METHOD ENZYMOL., vol. 121, 1986, pages 210 - 228
TANAKA R ET AL: "Unique monoclonal antibody recognizing the third extracellular loop of CXCR4 induces lymphocyte agglutination and enhances human immunodeficiency virus type 1-mediated syncytium formation and productive infection", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 75, no. 23, 1 December 2001 (2001-12-01), pages 11534 - 11543, XP002548956, ISSN: 0022-538X, DOI: 10.1128/JVI.75.23.11534-11543.2001 *
TANAKA R. ET AL., J. VIROL., vol. 75, 2001, pages 11534 - 11543
TATUSOVA ET AL.: "Blast 2 sequences - a new tool for comparing protein and nucleotide sequences", FEMS MICROBIOL., vol. 174, 1999, pages 247 - 250
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
VERHOEYN, BIOESSAYS, vol. 8, 1988, pages 74
ZHAO Q. ET AL., AIDS RESEARCH AND HUMAN RETROVIRUSES, vol. 19, 2003, pages 947 - 955

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014134547A1 (en) * 2013-02-28 2014-09-04 Therabiol, Inc. Hiv antigens and antibodies
WO2017220988A1 (en) 2016-06-20 2017-12-28 Kymab Limited Multispecific antibodies for immuno-oncology
WO2017220989A1 (en) 2016-06-20 2017-12-28 Kymab Limited Anti-pd-l1 and il-2 cytokines
WO2017220990A1 (en) 2016-06-20 2017-12-28 Kymab Limited Anti-pd-l1 antibodies

Also Published As

Publication number Publication date
EP2632953A1 (en) 2013-09-04
CN103180342A (zh) 2013-06-26
KR20140009174A (ko) 2014-01-22
CA2814908A1 (en) 2012-05-03
MX2013004710A (es) 2013-08-29
RU2013122770A (ru) 2014-12-10
AU2011322508A1 (en) 2013-05-02
ZA201302639B (en) 2013-11-27
JP2014504147A (ja) 2014-02-20

Similar Documents

Publication Publication Date Title
AU2010243526B2 (en) Anti CXCR4 antibodies for the treatment of HIV
JP6936361B2 (ja) ヒト免疫不全ウイルス中和抗体
KR101671039B1 (ko) 항 cxcr4 항체 및 그들의 암의 치료를 위한 용도
KR20160006168A (ko) 인간화 항-cd134(ox40) 항체 및 이의 용도
KR20140069172A (ko) 항­cd 134(ox40) 및 이의 용도
JP2021505137A (ja) 悪性b細胞を特異的に認知する抗体またはその抗原結合断片、これを含むキメラ抗原受容体及びその用途
KR102165934B1 (ko) 악성 b 세포를 특이적으로 인지하는 항체 또는 그의 항원 결합 단편, 이를 포함하는 키메라 항원 수용체 및 이의 용도
WO2012055980A1 (en) Antibodies for the treatment of hiv
KR20240014058A (ko) 항-cea 항체 및 항-cd137 다중 특이적 항체 및 사용 방법
JP2019048804A (ja) 抗pd−1抗体並びにその治療及び診断のための使用
WO2021175954A1 (en) Antibodies having specificity for btnl8 and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11779612

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 13823182

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2814908

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013535444

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/004710

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2011322508

Country of ref document: AU

Date of ref document: 20111027

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20137011891

Country of ref document: KR

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2011779612

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2011779612

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2013122770

Country of ref document: RU

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013009984

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013009984

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130424