WO2013175237A1 - Composition comprenant un agent de ligature cd2 et un agent de ligature nkg2d - Google Patents

Composition comprenant un agent de ligature cd2 et un agent de ligature nkg2d Download PDF

Info

Publication number
WO2013175237A1
WO2013175237A1 PCT/GB2013/051387 GB2013051387W WO2013175237A1 WO 2013175237 A1 WO2013175237 A1 WO 2013175237A1 GB 2013051387 W GB2013051387 W GB 2013051387W WO 2013175237 A1 WO2013175237 A1 WO 2013175237A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
ligation agent
nkg2d
cells
ligation
Prior art date
Application number
PCT/GB2013/051387
Other languages
English (en)
Inventor
Mark Lowdell
Original Assignee
Ucl Business Plc
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 GBGB1209097.3A external-priority patent/GB201209097D0/en
Priority claimed from GB201304762A external-priority patent/GB201304762D0/en
Application filed by Ucl Business Plc filed Critical Ucl Business Plc
Publication of WO2013175237A1 publication Critical patent/WO2013175237A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4613Natural-killer cells [NK or NK-T]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464411Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464429Molecules with a "CD" designation not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70596Molecules with a "CD"-designation not provided for elsewhere
    • 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
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2806Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD2
    • 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
    • C07K16/2851Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the lectin superfamily, e.g. CD23, CD72
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0646Natural killers cells [NK], NKT cells
    • 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
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/53CD2
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/599Cell markers; Cell surface determinants with CD designations not provided for elsewhere

Definitions

  • the present invention relates to a composition for priming a human Natural Killer (NK) cell. Following priming, the NK cell may have the capacity to lyse an NK-resistant cancer cell. BACKGROUND TO THE INVENTION
  • HSCT hematopoietic stem cell transplantation
  • NK cells Natural Killer (NK) cells are a subset of peripheral blood lymphocytes which can spontaneously lyse certain tumour cells.
  • the use of NK cells in adoptive tumour immunotherapy has been proposed, and there has been interest in the in vitro or ex vivo stimulation of NK cells to increase their capacity to lyse tumour cells.
  • NK cells require at least two activating signals before commitment to cytokine secretion and/or target cell lysis. It has been shown that these two signals can be divided into discrete "priming” and “triggering” events, with the priming signal being provided either by an activating cytokine, such as IL-2, or conjugation to a tumour cell expressing an appropriate intensity and combination of signals (North et al (2007) J. Immunol. 178:85-94).
  • WO 2006/097743 describes a method for activating an NK cell by contacting the NK cell in vitro with a preparation of the tumour cell line CTV-1 , which primes resting NK cells but fails to trigger lysis.
  • rNK resting human NK
  • ATCP activating tumour cell preparation
  • Membrane preparations have the advantage over preparations comprising intact tumour cells as they avoid the risk of transferring potentially malignant tumour cells to the patient.
  • Figure 1 Investigating the optimum concentrations of the anti-CD2 and anti-NKG2D antibodies for bead coating.
  • CD2-ligation is one of the mechanisms whereby resting human N K (rN K) cells are primed to activate.
  • CTV-1 causes CD2 ligation through expression of CD15, which can bind CD2 (Sabry et al (201 1 ) J . Immunol. 187:6227- 6234).
  • CD15 alone is insufficient to prime rNK since normal human myeloid cells which constitutively express CD15 do not prime rNK.
  • the present inventors have now found that it is possible to prime rNK cells by co- ligating both CD2 and NKG2D.
  • the invention provides a human natural killer (NK) cell-priming composition which comprises (i) a CD2 ligation agent; and (ii) an N KG2D ligation agent.
  • NK human natural killer
  • the CD2 ligation agent may comprise the CD2 ligand from CD15.
  • the CD2 ligation agent may be CD15 with its associated carbohydrate structure.
  • the CD2 ligation agent may be an anti-CD2 antibody.
  • the NKG2D ligation agent may be an anti-NKG2D antibody.
  • the N KG2D ligation agent may comprise the N KG2D binding site from an N KG2D ligand, such as MICA, MICB or ULBPs.
  • the NKG2D ligation agent may comprise all or a part of MICA, MICB or a ULBP.
  • the invention provides a human NK-cell priming substrate or vesicle which comprises a CD2 ligation agent and an N KG2 D l igation agent as defined in connection with the first aspect of the invention.
  • the substrate may, for example, be a two-dimensional surface (e.g. filter, plate, well, flask, bag, or the like) or a three-dimensional surface (e.g., bead, nanoparticle or the like), coated with a CD2 ligation agent and an NKG2D ligation agent.
  • the vesicle may be a liposome which expresses a CD2 ligation agent and an NKG2D ligation agent at the surface.
  • the liposome may also contain one or more cytokines involved in NK function/survival//proliferation, such that the cytokine(s) are released when NK cell priming occurs.
  • the cytokine(s) may, for example, be IL2, IL12 and/or IL15.
  • the present invention provides a method for activating a human Natural Killer (NK) cell, which comprises the step of contacting the NK cell in vitro with:
  • a human NK-cell priming substrate or vesicle according to the second aspect of the invention.
  • the present invention provides an activated NK cell produced by a method according to the third aspect of the invention.
  • the activated NK cell may maintain its activated state following removal of the NK-cell priming composition, substrate or vesicle.
  • the activated NK cell may maintain its activated state following cryopreservation.
  • the present invention provides a pharmaceutical composition comprising a plurality of activated NK cells according to the fourth aspect of the invention for treating a subject in need of same.
  • the present invention provides the use of a pharmaceutical composition according to the fifth aspect of the invention in the manufacture of a medicament for the treatment of cancer.
  • the present invention provides a method for treating cancer or infection which comprises the step of administering a pharmaceutical composition according to the fifth aspect of the invention to a subject.
  • the invention provides a human NK-cell priming composition according to the first aspect of the invention, or an NK-cell priming substrate or vesicle according to the second aspect of the invention for use in the treatment of cancer or infection.
  • the present invention provides a method for activating a human NK cell in vivo which comprises the step of administering a NK-cell priming composition according to the first aspect of the invention, or an NK-cell priming substrate or vesicle according to the second aspect of the invention to a subject.
  • the present invention provides a method for treating cancer or infection in a subject in need of same which comprises which comprises the step of administering an NK-cell priming composition according to the first aspect of the invention, or an NK-cell priming substrate or vesicle according to the second aspect of the invention to the subject.
  • the invention provides a kit for preparing a composition according to the first aspect of the invention, a substrate or vesicle according to the second aspect of the invention and/or for use in a method according to the third aspect of the invention which kit comprises (i) a CD2 ligation agent; and (ii) an NKG2D ligation agent.
  • the present invention provides a method for making a substrate according to the second aspect of the invention which comprises the step of attaching a CD2 ligation agent and an NKG2D ligation agent to a substrate.
  • the present invention provides a method of making a vesicle according to the present invention which comprises the step of incorporating or causing the expression of a CD2 ligation agent and an NKG2D ligation agent at the vesicle surface. Because the method of the present invention causes activation of an NK cell via ligation of two cell surface receptors by ligation agents, the method is entirely "synthetic" and does not rely on the presence of an activating tumour cell or cell membrane preparation thereof.
  • the invention replaces CTV-1 with an artifical NK priming reagent using a solid phase presentation system (such as a filter, plate, well, flask, bag or other two dimensional surface, beads, nanoparticles or liposomes).
  • the method of the invention therefore overcomes the disadvantages associates with the tumour cell activation method, as outlined in the previous section.
  • NK CELL H uman N K cells are a subset of peripheral blood lymphocytes defined by the expression of CD56 or CD16 and the absence of the T cell receptor (CD3). They recognise and kill transformed cell lines without priming, in an MHC-unrestricted fashion. NK cells represent the predominant lymphoid cell in the peripheral blood for many months after clinical allogeneic or autologous stem cell transplant and they have a primary role in immunity to pathogens during this period (Reittie et al (1989) Blood 73: 1351-1358; Lowdell et al (1998) Bone Marrow Transplant 21 : 679-686). The role of NK cells in engraftment, graft-versus-host disease, anti-leukemia activity and post- transplant infection is reviewed in Lowdell (2003) Transfusion Medicine 13:399-404.
  • Human NK cells mediate the lysis of tumour cells and virus-infected cells via natural cytotoxicity and antibody-dependent cellular cytotoxicity (ADCC).
  • Human NK are controlled by positive and negative cytolytic signals.
  • Negative (inhibitory) signals are transduced by C-lectin domain containing receptors CD94/NKG2A and by some Killer Immunoglobulin-like Receptors (KIRs).
  • KIRs Killer Immunoglobulin-like Receptors
  • the regulation of NK lysis by inhibitory signals is known as the "missing self" hypothesis in which specific HLA-class I alleles expressed on the target cell surface ligate inhibitory receptors on NK cells.
  • the down-regulation of HLA molecules on tumor cells and some virally infected cells e.g.
  • KIRs Killer Immunoglobulin-like Receptors
  • NKG2 the lectin family
  • KIRs have up to 4 intracellular domains which contain ITIMs and the best characterized are KIR2DL1 , KIR2DL2 and KIR2DL3 which are known to bind HLA-C molecules.
  • KIR2DL2 and KIR2DL3 bind the group 1 HLA-C alleles whilst KIR2DL1 binds to group 2 alleles.
  • Certain leukemia/lymphoma cells express both group 1 and 2 HLA-C alleles and are known to be resistant to NK-mediated cell lysis
  • ADCC is mediated via CD16 and a number triggering receptors involved in natural cytotoxicity have been identified, including CD2, CD38, CD69, NKRP-1 , CD40, B7-2, NK-TR, NKp46, N Kp30 and N Kp44.
  • KIR2DS1 , KIR2DS2 and KIR2DS4 are known to bind to HLA-C; their extracellular domains being identical to their related inhibitory KIRs.
  • the activatory KIRs lack the ITIMs and instead associate with DAP12 leading to NK cell activation. The mechanism of control of expression of inhibitory versus activatory KIRs remains unknown.
  • the NK cells activated by the method of the present invention may be autologous or allogeneic NK cells.
  • NK cells are cells derived from the patient.
  • Allogeneic NK cells are derived from another individual, having non-identical gene at one or more loci. If the NK cells are derived from an identical twin, they may be termed "syngeneic".
  • Donor NK cells may be HLA-KI R matched or mismatched. The present inventors have shown that the degree of matching between the NK cells and target tumour cells is of no significance.
  • NK-sensitive tumours provide both priming and triggering signals, leading to lysis.
  • NK-resistant tumours evade lysis, mostly by failure to prime.
  • tumour cells such as the tumour cell line CTV-1 , which have the capacity to prime tumour cells but fail to trigger lysis. It has previously been shown that such cells can be used to prime or activate NK cells, such that it can go on to lyse a target cell which is resistant to lysis by an equivalent unstimulated NK cell.
  • NK cells are used synonymously in this document to mean rendering a resting NK into a state such that can lyse a target cell which is normally resistant to NK cell lysis.
  • the target cell may be an NK resistant tumour cell.
  • Raji and Daudi cell lines are useful models for NK-resistant tumours.
  • the composition of the present invention is capable of priming an NK cell in the same way as treatment with an activating tumour cell preparation of, for example, CTV-1 cells.
  • Primed or activated NK cells have a characteristic phenotype, which distinguishes them from resting NK cells.
  • Tumour-mediated NK priming occurs by a different mechanism than IL-2 stimulation.
  • Tumour-mediated NK priming involves activation of the ⁇ 3 ⁇ - ⁇ _ ⁇ -8 ⁇ 3 ⁇ 5 pathway (see below) whereas IL-2 is known to activate NK cells via MAPK1/extracellular signal-related protein kinase.
  • IL-2 is known to activate NK cells via MAPK1/extracellular signal-related protein kinase.
  • upregulation of CD69 and IFNy synthesis is observed within 4 hours, whereas it takes a minimum of 48 hours following activation by IL-2 (Sabry et al (201 1) as above).
  • Upregulation of CD25 expression following IL-2 stimulation is also significantly slower than that induced by CTV-1 , and the proportion of activating NK cells is smaller.
  • Tumour-mediated NK activation is also stable after removal of the priming signal. Whereas IL-2 activated NK cells rapidly return to the non-activated state after removal of IL-2, CTV-1 activated NK cells retain their capacity for Raji-cell lysis even after many months of cryopreservation.
  • incubation of NK cells with the subject ligation agents as described herein causes rapid upregulation of CD69 on the NK cells.
  • the I L-2 receptor, CD25 is also upregulated.
  • the subject ligation agents produce an activated NK cell population that is CD69+ and/or CD25+.
  • contact with appropriate ligation agents may also result in the transfer of CD15 to the activated NK cells (e.g., activated NK cells gain CD15), and the reduction of CD16 expression from the NK cell after activation.
  • the activating agents contemplated for use herein may further produce an activated NK cell population that is also CD15+ and/or CD16low.
  • composition of the present invention comprises at least two ligation agents: a CD2 ligation agent, and an NKG2D ligation agent.
  • a ligation agent is an entity which binds and activates a receptor.
  • a receptor is a cell-associated protein that binds to a bioactive molecule (the "ligand") and mediates the effect of the ligand on the cell. Binding of ligand to receptor results in a change in the receptor (and, in some cases, receptor multimerization, i. e. , association of identical or different receptor subunits) that causes interactions between the effector domain (s) of the receptor and other molecule (s) in the cell. These interactions in turn lead to alterations in the metabolism of the cell.
  • the ligation agent may be a natural or synthetic ligand for the receptor. The ligation agent may trigger the same intracellular effect as the natural ligand.
  • the ligation agent may be a binding agent such as an antibody.
  • the ligation agent may be an agonist for the receptor, or an analog or derivative thereof including, e.g., a fusion of the agonist with another protein or compound.
  • the ligation agent may be an antibody or an antigen-binding fragment thereof, including, e.g., a monoclonal or polyclonal antibody, a tetrabody, a nanobody, a chimeric antibody, a deimmunized antibody, a humanized antibody or a human antibody.
  • the antigen binding fragment is selected from the group consisting of F(ab)2, F(ab')2, Fab, Fab', Fd, Fv, single-chain Fv, and disulfide-linked Fvs (dsFv).
  • the term "antibody” includes antibody-like molecules with alternative scaffolds such as DARPins and other repeat protein scaffolds and domain antibodies (d(Ab)s).
  • the ligation agents can be modified, e.g., by the covalent attachment of any type of molecule as long as such covalent attachment permits the agonist or antibody to retain its activation of the receptor.
  • suitable derivatives and analogs of the ligation agents include those that have been further modified, e.g., by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a substrate or other protein, etc. Any of numerous chemical modifications can be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, etc.
  • the analog or derivative can contain one or more unnatural amino acids, or have a modification (e.g., substitutions, deletions or additions) in amino acid residues that interact with Fc receptors.
  • CD2 is a cell adhesion molecule found on the surface of T cells and NK cells. It acts as a costimulatory molecule.
  • CD2 on the NK cell binds to a ligand within CD15 on the tumour cell (Sabry et al (201 1) J. Immunol. 187:6227-6234). Blockade of CD15 on tumour cells has been shown to significantly inhibit priming of NK cells. Also, NK-resistant Raji cells become susceptible to NK lysis following transfection and expression of CD15.
  • the CD2 ligation agent may comprise the CD2L site of CD15.
  • the ligand for CD2 of CD15 is a carbohydrate structure which is closely associated with, yet distinct from CD15 (Warren et al (1996) J. Immunol. 156:2866-2873).
  • the carbohydrate structure is Gal ⁇ 1-4 GlcNAc a1-3Fuc.
  • the ligation agent may comprise this carbohydrate structure optionally in association with all or a part of CD15.
  • Various anti-human CD2 antibodies are known, such as clone RPA-2.10 (Bryceson et al (2006) Blood 107:159-166), and OKT11 (Sabry et al (201 1 as above).
  • CD2 ligation activates the ⁇ 3 ⁇ - ⁇ _ ⁇ -8 ⁇ 3 ⁇ 5 pathway.
  • the cytoplasmic ⁇ 16/ ⁇ 3 ⁇ complex interacts with the intracellular domain of CD2, leading to the phosphorylation of linker for activation of T cells (LAT).
  • LAT T cells
  • Ligation of CD2 leads to phosphorylation of Stat5 and upregulation of CD25 and CD69.
  • the CD2 ligation agent may cause activation of the ⁇ 3 ⁇ - ⁇ _ ⁇ -8 ⁇ 3 ⁇ 5 pathway. This may be detected by monitoring phosphorylation of LAT and/or Stat5; by examining the levels of CD25 and/or CD69; or by investigating the production of IFNy.
  • NKG2D LIGATION AGENT NKG2D is an activating receptor found on the surface of NK cells and CD8+ T cells.
  • NKG2D consists of two disulphide-linked type II transmembrane proteins with short intracellular proteins which are incapable of transducing signals. NKG2D therefore requires an adaptor protein in order to transduce signals. Two adaptor proteins exist, DAP10 and DAP12, which associate as homodimers to the receptor. The entire receptor complex therefore appears as a hexamer.
  • DAP10 the signalling subunit, carries a phosphatidylinositol-3-kinase binding motif.
  • Ligands for NKG2D are induced during times of cellular stress, either as a result of infection or genomic stress such as in cancer, which renders the cell susceptible to NK cell mediated lysis.
  • ligands for NKG2D include MICA, MICB, ULBP1 , ULBP2, ULBP3 and ULBP4-6.
  • the NKG2D ligation agent used in the composition of the present invention may comprise all or part of a NKG2D ligand.
  • the ligand may also comprise a portion of an antibody, for example the Fc portion, giving MICA-Fc and ULBP-Fc.
  • the NKG2D ligation agent used in the composition of the present invention may comprise all or part of an antibody.
  • Commercial anti-human NKG2D antibodies are known, such as clone 149810 from R&D systems, Minneapolis, and 1 D1 1 from eBioscience.
  • the second aspect of the present invention provides a substrate which comprises a CD2 ligation agent and an NKG2D ligation agent.
  • the ligation agents may be attached to the surface of the substrate.
  • the substrate may be used to activate rN K cells, by bringing the rN K cells into contact with the substrate such that ligation of CD2 and NKG2D on the NK cells occurs.
  • the resulting preparation should comprise activated NK cells in a relatively pure form (i.e. without the CD2 and NKG2D ligation agents).
  • the ligation agents are antibodies
  • substrates coated with a "second-layer” antibody i.e. an antibody reactive with the ligation agent
  • the substrate may present a two dimensional surface, such as a filter, plate, well, flask, roller bottle, capillary, bag and the like; or a three-dimensional surface, such as a bead or microparticle. Magnetic/paramagnetic beads or particles may be used to aid separation.
  • the substrate material employed in the present invention may be of any suitable material and may be a porous or a non-porous support.
  • the substrate is a solid support, particle or bead.
  • the substrate is comprised of a cross-linked carbohydrate material, such as agarose, agar, cellulose, dextran, chitosan, konjac, carrageenan, gellan, alginate etc.
  • the substrate may easily be prepared according to standard methods, or is a commercially available product, such as DYNABEADSTM (Life Technologies, California, USA) or SEPHADEXTM or SEPHAROSETM FF (Amersham Biosciences AB, Uppsala, Sweden).
  • the substrate is comprised of cross-linked synthetic polymers, such as styrene or styrene derivatives, divinylbenzene, acrylamides, acrylate esters, methacrylate esters, vinyl esters, vinyl amides, or may be an inorganic material such as glass or silica.
  • the polymer is selected from the group consisting of polystyrene, polypropylene, polyvinyltoluene, polyacrylamide, polyacrylonitrile and polycarbonate.
  • Solid supports of such polymers are easily produced according to standard methods, see e.g. "Styrene based polymer supports developed by suspension polymerization” (Arshady, R., Chimica e L'lndustria, (1988), 70(9), 70-75).
  • the invention also provides a method for making a substrate according to the second aspect of the invention which comprises the step of attaching a CD2 ligation agent and an NKG2D ligation agent to a substrate.
  • the agents may be directly attached, or attached via another entity, such as an antibody or linker.
  • the invention also provides a vesicle which comprises a CD2 ligation agent and an NKG2D ligation agent.
  • the ligation agents are displayed on the surface of the vesicle such that they can interact with CD2 and NKG2D on the surface of rNK cells.
  • the vesicle may be a membranous vesicle.
  • a liposome is an artificial vesicle composed of a lipid bilayer.
  • the vesicle may comprise one or more cytokines.
  • the cytokine(s) may be associated with NK cell activation, survival and/or proliferation.
  • the cytokine(s) are available to the NK cell when NK cell priming occurs.
  • the vesicle may, for example, comprise IL2, IL12 or IL15.
  • the cytokines may be released from the vesicle at the site of NK cell activation or taken up by the NK cell within the vesicle.
  • the vesicle may be a cell-like structure but may not be a continuously growing tumour cell line. Where the vesicle is a cell, the CD2 ligation agent and an NKG2D ligation agent should not be endogenous to the cell.
  • the cell may be a non-tumorous cell or a primary tumour within or isolated from a patient induced to express these agents artificially (i.e. by recombinant means).
  • the invention also provides a method of making a vesicle according to the present invention which comprises the step of incorporating or causing the expression of a CD2 ligation agent and an NKG2D ligation agent at the vesicle surface.
  • the CD2 ligation agent and NKG2D ligation agent may be, comprise, or be attached to an entity which is capable of being included in the membrane.
  • the agent(s) may be attached to a transmembrane protein.
  • Methods for including membrane proteins in the lipid bilayer of liposomes include: methods involving the use of an organic solvent, methods involving the use of the mechanical means, methods involving the use of detergents and direct incorporation of the protein into the preformed liposomes.
  • the present invention also provides a method for activating a human Natural Killer (NK) cell, which comprises the step of contacting the NK cell in vitro with:
  • NK cells may be activated by the one of the above methods alone, or in combination with another NK cell activation technique.
  • US7435596 describes the expression of chimeric receptors on NK cells to enhance their capacity to kill target cells.
  • the NK cells of the present invention may express a chimeric receptor comprising an anti-CD19 receptor and a signalling domain.
  • the signalling domain may, for example be ⁇ 3 ⁇ or DAP10.
  • the chimeric receptor may also comprise the costimulatory molecule 4-1 BB.
  • the resting NK cell may be autologous or allogeneic.
  • Allogenic NK cells may be obtained from peripheral blood from a donor individual. Allogeneic peripheral blood mononuclear cells may be collected by standard techniques (e.g. conventional apheresis). To minimize the possibility of graft versus host disease and immune mediated aplasia, allogeneic cells may be depleted of T cells. For example, the cell preparation may be depleted of CD3+ T-cells using microbeads conjugated with monoclonal mouse anti-human CD3 antibody and a cell selection device (such as the Miltenyi Biotec CliniMACS® cell selection device).
  • a cell selection device such as the Miltenyi Biotec CliniMACS® cell selection device.
  • NK cells produced by such "negative selection” procedures alone do not have a high degree of purity and may be contaminated with T and B cells.
  • the product may be depleted for CD3+ cells (for example using CD3 FITC and anti-FITC beads).
  • the NK cell preparation may comprise at least 80%, at least 90%, at least 95% or at least 98% CD56+ cells.
  • the N K cell preparation may comprise less than 15%, less than 10%, less than 5% or less than 3% CD3+ cells.
  • the present invention also provides an NK cell activated by a method according to the present invention.
  • the present invention also provides a pharmaceutical composition comprising NK cells activated by a method of the invention.
  • composition may comprise or consist essentially of autologous and/or allogeneic NK cells.
  • Allogeneic NK cells may be HLA mismatched.
  • the composition may also comprise the CD2 ligation agent and/or NKG2D ligation agent; or a substrate or vesicle comprising the CD2 ligation agent and/or NKG2D ligation agent.
  • the CD2 ligation/ NKG2D ligation may be the only activation the NK cells receive, or there may be further activation steps.
  • the N K cells may or may not also be non- specifically activated by I L-2 (for example by incubation of the cells in medium supplemented with IL-2). Alternatively, the cells may be activated in the absence of IL-2, but IL-2 may be used for the ex vivo expansion of stimulated cells.
  • composition of the present invention may be used in medicine.
  • the composition may be used to treat or prevent cancer or infection in a subject.
  • composition comprising activated NK cells may be manufacture of a medicament for the treatment of cancer or infection.
  • the composition may be administered to the subject by any suitable method known in the art, for example, intravenous infusion.
  • the present invention also provides a method for treating a subject in need of same, which comprises the following steps:
  • the method may be for treating a disease in the subject.
  • the disease may be cancer or an infection.
  • the composition may be used to treat a subject in need of same.
  • the procedure is low-risk and particularly suitable for cancer patients for whom intensive cancer treatments are precluded (for example, elderly patients). It also provides an alternative for patients (with, for example, lymphoma, myeloma or AML) who lack a suitable donor for allogeneic stem cell transplantation.
  • the patient may receive some pre-treatment, for example, to de-bulk the tumour and /or immunosuppress the patient. This may be achieved, for example, by chemotherapy, radiotherapy or a combination thereof.
  • the activated NK cell composition may be used to treat or prevent a disease or medical condition.
  • the disease may be a cancer.
  • Some more common cancers include leukaemia (acute and chronic), bladder cancer, bone cancer (osteosarcoma), Bowel (colorectal cancer), brain cancer, breast cancer, cervical cancer, oesophageal cancer, Hodgkin's lymphoma, kidney cancer, liver cancer, lung cancer, mesothelioma, multiple myeloma, non-Hodgkin's lymphoma, ovarian cancer, pancreatic cancer, penile cancer, prostate cancer, skin cancer (melanoma and non-melanoma) soft tissue carcinoma, gastric cancer, testicular cancer, thyroid cancer and endometrial cancer.
  • the activated NK cell composition produced by the method of the present invention may be useful to treat any cancer which is accessible to NK cells.
  • the cancer may be a haematological malignancy, such as leukaemia (AML); myeloma; Lymphoma.
  • AML leukaemia
  • myeloma myeloma
  • Lymphoma a malignancy
  • Myeloma is an incurable and fatal malignancy. NK activity against myeloma plasma cells is documented in vitro and enhanced NK activity against autologous myeloma cells has been shown to correlate with response to treatment with Thalidomide derivatives. Myeloma patients are generally young and fit enough to undergo autologous haematopoietic stem cell transplantation and could readily undergo a less invasive procedure such as the one provided by the present invention.
  • PTLD Post-transplant lymphoproliferative disease
  • T cell immunotherapy is currently under trial but with little success.
  • Therapy using NK cells activated according to the present invention therapy would be easy and safe in this group of patients.
  • the composition may be used to treat solid tumours such as breast cancer.
  • NK-resistant tumours Normal NK cells can spontaeously lyse some human tumours, but many other tumours are NK- resistant. "NK-resistant” as used herein, therefore, indicates tumour cells resistant to lysis by normal N K cells which have not been stimulated with a ATCP or by the method according to the present invention.
  • NK-mediated lysis is controlled by expression of specific MHC class I molecules on the target cell surface, particularly HLA-C.
  • HLA-C MHC class I molecules
  • Some leukemia/lymphoma-derived cell lines, such as Raji and Daudi express both types of HLA-C allele, making them useful models for NK-resistant tumour cells in vivo.
  • Common infections that may be treated with a cellular composition comprising the activated NK cells as described herein include viral infections such as, e.g., hepatitis type A virus, hepatitis type B virus, hepatitis type C virus, etc.; parvoviruses, such as adeno-associated virus and cytomegalovirus; papovaviruses such as papilloma virus, polyoma viruses, and SV40; adenoviruses; herpes viruses such as herpes simplex type I (HSV-I), herpes simplex type II (HSV-II), and Epstein-Barr virus; poxviruses, such as variola (smallpox) and vaccinia virus; RNA viruses, including but not limited to human immunodeficiency virus type I (HIV-I), human immunodeficiency virus type II (HIV-II), human T-cell lymphotropic virus type I (HTLV-I), and human T-cell lymphotropic virus
  • the present invention also provides a kit for preparing a NK cell activating composition according to the present invention.
  • the present invention also provides a kit for use in a method for activating an NK cell according to the method of present invention.
  • the kit may comprise (i) a CD2 ligation agent; and (ii) an NKG2D ligation agent or a precursor thereof.
  • a CD2 ligation agent for example, where the CD2 ligation agent and/or NKG2D ligation agent is/are a protein, the kit may comprise a protein-encoding gene.
  • the present invention also provides a method for preparing a composition according to the present invention, which comprises the step of combining (i) a CD2 ligation agent; and (ii) an NKG2D ligation agent.
  • the present invention also provides the use of the kit in a method for preparing a substrate according to the present invention, which method comprises the step of attaching or immobilising (i) a CD2 ligation agent; and (ii) an NKG2D ligation agent on a substrate.
  • the present invention also provides the use of a kit in a method for preparing a vesicle according to the present invention, which method comprises the step of causing (i) a CD2 ligation agent; and (ii) an NKG2D ligation agent to be expressed or present on the surface of a vesicle.
  • the kit may also comprise instructions for use.
  • Example 1 Preparation of activating microbeads The optimal concentrations of each mAb to label 3-D microbeads is determined by co-incubating aliquots of 5x10 5 Dynal goat anti-sheep 450uM beads with increasing amounts of
  • the cognate ligand(s) for NKG2D may be MICA, MICB or ULBP.
  • CTV-1 cells are immunophenotyped for MICA, MICB and ULBPs to determine which are expressed and their relative densities compared to each other and to CD15. Blocking of CTV-1 -mediated NK priming is tested with saturating concentrations of anti-MICA, anti-MICB and available mAbs to ULBPs to determine the most important NKG2D ligand for NK priming induced by CTV-1.
  • rNK cells Resting human NK (rNK) cells are directly isolated from healthy volunteer donors by immunomagnetic selection with anti-CD56 PE and anti-PE microbeads (Miltenyi Biotec UK) and resuspended in culture medium supplemented with clinical-grade FBS (10%) at 10 6 /ml.
  • the results from Example 1 above is used to provide an appropriate ratio of anti-CD2 and anti-NKGD2 coated beads.
  • a 2-D surface such as a 96 well ELISA plate is coated with an appropriate concentration of each mAb selected following the information provided in Example 1 , and the activation experiment outlined in Example 3 is repeated.
  • This allows a) easier analysis of the temporal dynamics of NK priming by co-ligation of CD2/NKG2D; b) a scale-up production process for commercialisation as there is no bead removal step.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Mycology (AREA)
  • Veterinary Medicine (AREA)
  • Biomedical Technology (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Toxicology (AREA)
  • Wood Science & Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Oncology (AREA)
  • General Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

La présente invention concerne une composition d'activation de cellules tueuses naturelles (NK), qui comprend (i) un agent de ligature CD2; et (ii) un agent de ligature NKG2D. L'invention concerne également un substrat ou une vésicule d'activation de cellules NK (telle qu'un liposome) qui comprend un agent de ligature CD2 et un agent de ligature NKG2D. L'invention concerne également un procédé d'activation d'une cellule NK qui comprend l'étape de mise en contact de la cellule NK in vitro avec une telle composition d'activation de cellules NK ou un tel substrat ou une telle vésicule d'activation de cellules NK.
PCT/GB2013/051387 2012-05-24 2013-05-24 Composition comprenant un agent de ligature cd2 et un agent de ligature nkg2d WO2013175237A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GBGB1209097.3A GB201209097D0 (en) 2012-05-24 2012-05-24 Composition
GB1209097.3 2012-05-24
GB1304762.6 2013-03-15
GB201304762A GB201304762D0 (en) 2013-03-15 2013-03-15 Composition

Publications (1)

Publication Number Publication Date
WO2013175237A1 true WO2013175237A1 (fr) 2013-11-28

Family

ID=48534448

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2013/051387 WO2013175237A1 (fr) 2012-05-24 2013-05-24 Composition comprenant un agent de ligature cd2 et un agent de ligature nkg2d

Country Status (1)

Country Link
WO (1) WO2013175237A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014005072A1 (fr) 2012-06-28 2014-01-03 University Of Central Florida Research Foundation, Inc. Procédés et compositions pour des cellules tueuses naturelles
CN105296422A (zh) * 2015-11-26 2016-02-03 广州赛莱拉干细胞科技股份有限公司 一种nk细胞培养组合物及其培养方法
CN105296423A (zh) * 2015-11-26 2016-02-03 广州赛莱拉干细胞科技股份有限公司 一种cd3ak细胞培养组合物及其培养方法
WO2016185224A1 (fr) * 2015-05-21 2016-11-24 Ucl Business Plc Composition d'activation de cellules tueuses naturelles
CN107326008A (zh) * 2017-08-09 2017-11-07 上海莱馥医疗科技有限公司 一种从外周血中高效高纯度扩增自然杀伤细胞的方法
US10758567B2 (en) 2015-09-16 2020-09-01 Immune Ventures LLC In vivo priming of natural killer cells
WO2020219928A1 (fr) * 2019-04-25 2020-10-29 Actinium Pharmaceuticals, Inc. Compositions et méthodes d'immunodéplétion pour le traitement de maladies hématologiques malignes et non malignes
CN113502267A (zh) * 2021-08-24 2021-10-15 羽铂精制生物技术(成都)有限公司 一种用于外周血中nk细胞扩增的培养基及方法

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056873A1 (fr) * 2002-12-20 2004-07-08 Medinnova Ges Med Innovationen Accroissement de la reponse immunitaire par des substances qui influencent la fonction de cellules k naturelles
WO2008058728A1 (fr) * 2006-11-14 2008-05-22 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts Compositions et procédés pour une immunothérapie
WO2011004201A1 (fr) * 2009-07-10 2011-01-13 Mark Lowdell Compositions conservées de cellules tueuses naturelles (nk) activées et leurs procédés d'utilisation

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004056873A1 (fr) * 2002-12-20 2004-07-08 Medinnova Ges Med Innovationen Accroissement de la reponse immunitaire par des substances qui influencent la fonction de cellules k naturelles
WO2008058728A1 (fr) * 2006-11-14 2008-05-22 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts Compositions et procédés pour une immunothérapie
WO2011004201A1 (fr) * 2009-07-10 2011-01-13 Mark Lowdell Compositions conservées de cellules tueuses naturelles (nk) activées et leurs procédés d'utilisation

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
KIM TAE-JIN ET AL: "Suppression of human anti-porcine natural killer cell xenogeneic responses by combinations of monoclonal antibodies specific to CD2 and NKG2D and extracellular signal-regulated kinase kinase inhibitor", IMMUNOLOGY, vol. 130, no. 4, August 2010 (2010-08-01), pages 545 - 555, XP002699086, ISSN: 0019-2805 *
SABRY MAY ET AL: "Leukemic Priming of Resting NK Cells Is Killer Ig-like Receptor Independent but Requires CD15-Mediated CD2 Ligation and Natural Cytotoxicity Receptors", JOURNAL OF IMMUNOLOGY, vol. 187, no. 12, December 2011 (2011-12-01), pages 6227 - 6234, XP002699085, ISSN: 0022-1767 *
WHITESIDE THERESA L ET AL: "Role of Human Natural Killer Cells in Health and Disease", CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY, AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 1, no. 2, 1 March 1994 (1994-03-01), pages 125 - 133, XP009169548, ISSN: 1071-412X *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10463715B2 (en) 2012-06-28 2019-11-05 University Of Central Florida Research Foundation, Inc. Methods and compositions for natural killer cells
EP2866834A4 (fr) * 2012-06-28 2015-12-09 Univ Central Florida Res Found Procédés et compositions pour des cellules tueuses naturelles
US11617781B2 (en) 2012-06-28 2023-04-04 University Of Central Florida Research Foundation, Inc. Methods and compositions for natural killer cells
WO2014005072A1 (fr) 2012-06-28 2014-01-03 University Of Central Florida Research Foundation, Inc. Procédés et compositions pour des cellules tueuses naturelles
US9623082B2 (en) 2012-06-28 2017-04-18 University Of Central Florida Research Foundation, Inc. Methods and compositions for natural killer cells
US10874715B2 (en) 2012-06-28 2020-12-29 University Of Central Florida Research Foundation, Inc. Methods and compositions for natural killer cells
WO2016185224A1 (fr) * 2015-05-21 2016-11-24 Ucl Business Plc Composition d'activation de cellules tueuses naturelles
US10758567B2 (en) 2015-09-16 2020-09-01 Immune Ventures LLC In vivo priming of natural killer cells
CN105296423A (zh) * 2015-11-26 2016-02-03 广州赛莱拉干细胞科技股份有限公司 一种cd3ak细胞培养组合物及其培养方法
CN105296422B (zh) * 2015-11-26 2018-09-11 广州赛莱拉干细胞科技股份有限公司 一种nk细胞培养组合物及其培养方法
CN105296422A (zh) * 2015-11-26 2016-02-03 广州赛莱拉干细胞科技股份有限公司 一种nk细胞培养组合物及其培养方法
CN107326008B (zh) * 2017-08-09 2019-10-22 上海莱馥医疗科技有限公司 一种从外周血中高效高纯度扩增自然杀伤细胞的方法
CN107326008A (zh) * 2017-08-09 2017-11-07 上海莱馥医疗科技有限公司 一种从外周血中高效高纯度扩增自然杀伤细胞的方法
WO2020219928A1 (fr) * 2019-04-25 2020-10-29 Actinium Pharmaceuticals, Inc. Compositions et méthodes d'immunodéplétion pour le traitement de maladies hématologiques malignes et non malignes
CN113502267A (zh) * 2021-08-24 2021-10-15 羽铂精制生物技术(成都)有限公司 一种用于外周血中nk细胞扩增的培养基及方法

Similar Documents

Publication Publication Date Title
US20210252055A1 (en) Modified gamma delta t cells and uses thereof
US20240115609A1 (en) METHODS FOR EXPANDING AND ACTIVATING yo T CELLS FOR THE TREATMENT OF CANCER AND RELATED MALIGNANCIES
JP2023037006A (ja) Cart細胞における遺伝子発現の改変およびその使用
WO2013175237A1 (fr) Composition comprenant un agent de ligature cd2 et un agent de ligature nkg2d
Dianat‐Moghadam et al. Natural killer cell–based immunotherapy: from transplantation toward targeting cancer stem cells
KR20180082493A (ko) Traf-유도 도메인을 함유하는 키메라 수용체 및 관련 조성물 및 방법
KR20170101206A (ko) 조작된 감마 델타 t 세포
Kannan et al. Natural killer cells in malignant hematology: A primer for the non-immunologist
US20220064255A1 (en) Anti-tcr antibody molecules and uses thereof
EP3786287A1 (fr) Population de cellules cd3 négatives exprimant un récepteur de chimiokine et une molécule d'adhérence cellulaire, et application ainsi que procédé de fabrication associés
EP3898946B1 (fr) Cellules t invariantes associées aux muqueuses (mait) exprimant des récepteurs de l'antigène chimérique
CN110603320A (zh) 高活性nk细胞及其应用
CA3160997A1 (fr) Molecules d'anticorps anti-tcr et leurs utilisations
Yu et al. Revolution of CAR engineering for next-generation immunotherapy in solid tumors
US20180142209A1 (en) Natural killer cell priming composition
Tarazona et al. Expression of NK-associated receptors on cytotoxic T cells from melanoma patients: a two-edged sword?
EP4173640A1 (fr) Procédé de stabilisation de liaison d'un anticorps sur une cellule nk et utilisation associée
Hörster New strategies to improve Natural Killer cells for “off-the-shelf” allogeneic immunotherapy
Özkazanç Ünsal Development of a novel in vitro screening method using genetically modified NK-92 cells against various tumor cells
Nowakowska Establishment of a good manufacturing practice-compliant procedure for expansion of therapeutic doses of genetically modified, CAR expressing NK-92 cells for the treatment of ErbB2-positive malignancies
EA045258B1 (ru) СПОСОБЫ АКТИВАЦИИ, МОДИФИКАЦИИ И ЭКСПАНСИИ γδ T-КЛЕТОК ДЛЯ ЛЕЧЕНИЯ РАКА И СВЯЗАННЫХ С НИМ ЗЛОКАЧЕСТВЕННЫХ ЗАБОЛЕВАНИЙ
EA043265B1 (ru) Модифицированные гамма-дельта-t-клетки и их применение
EA040572B1 (ru) Изменение экспрессии гена в cart-клетках и их применения

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: 13725465

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13725465

Country of ref document: EP

Kind code of ref document: A1