[go: up one dir, main page]

WO2020165402A1 - Régulation positive du gd2 par inhibition d'ezh2 dans le traitement du cancer - Google Patents

Régulation positive du gd2 par inhibition d'ezh2 dans le traitement du cancer Download PDF

Info

Publication number
WO2020165402A1
WO2020165402A1 PCT/EP2020/053874 EP2020053874W WO2020165402A1 WO 2020165402 A1 WO2020165402 A1 WO 2020165402A1 EP 2020053874 W EP2020053874 W EP 2020053874W WO 2020165402 A1 WO2020165402 A1 WO 2020165402A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
car
specific
cells
ezh2 inhibitor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/EP2020/053874
Other languages
English (en)
Inventor
Claudia Roessig
Bianca ALTVATER
Sareetha KAILAYANGIRI
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Westfaelische Wilhelms Universitaet Muenster
Original Assignee
Westfaelische Wilhelms Universitaet Muenster
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Westfaelische Wilhelms Universitaet Muenster filed Critical Westfaelische Wilhelms Universitaet Muenster
Publication of WO2020165402A1 publication Critical patent/WO2020165402A1/fr
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/10Cellular immunotherapy characterised by the cell type used
    • A61K40/11T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/10Cellular immunotherapy characterised by the cell type used
    • A61K40/15Natural-killer [NK] cells; Natural-killer T [NKT] cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/30Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
    • A61K40/31Chimeric antigen receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/40Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
    • A61K40/41Vertebrate antigens
    • A61K40/42Cancer antigens
    • A61K40/4256Tumor associated carbohydrates
    • A61K40/4258Gangliosides, e.g. GM2, GD2 or GD3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • 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
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3076Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties
    • C07K16/3084Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells against structure-related tumour-associated moieties against tumour-associated gangliosides
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • 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
    • C12N2510/00Genetically modified cells

Definitions

  • the present invention relates an Enhancer of Zeste Homolog 2 (EZH2) inhibitor for use in a method of treating a solid tumor in a subject, wherein the method further comprises the administration of a G D2 -specific CAR-T cell or CAR-NK cell or a G D2 -specific antibody to the subject. Further, the present invention relates to a G D2 -specific chimeric antigen receptor (CAR-) T cell or CAR-NK cell or a G D2 -specific antibody for use in a method of treating a solid tumor in a subject, wherein (i) the method further comprises the administration of an EZH2 inhibitor to the subject; and/or (ii) the subject has been and/or is treated with an EZH2 inhibitor.
  • EZH2 Enhancer of Zeste Homolog 2
  • the present invention relates to methods for increasing the surface expression of G D2 in solid tumor cells and kits and kits-of-part comprising an EZH2 inhibitor and a G D2 -specific CAR-T cell or a G D2 -specific CAR-NK cell or a G D2 -specific antibody.
  • Chimeric antigen receptors are recombinant proteins that link antibody-derived antigen-binding domains to stimulatory T cell signaling pathways. Expressed in immune effector cells, they induce activation responses to surface-expressed tumor target antigens, resulting in antigen-specific tumor cytolysis (Eshhar et al, 1993). T cells engineered to express CARs against the B-lineage antigen CD19 are active to induce and maintain remissions in refractory B lineage leukemias (Gardner et al, 2017; Lee et al, 2015; Maude et al, 2018) and lymphomas (Neelapu et al, 2017; Schuster et al, 2018). The development of CAR T cells for solid tumors is limited by the paucity of target antigens reliably expressed at high densities on cancer cells but not normal cells.
  • Ewing sarcoma as one example of a solid tumor, is an aggressive solid mesenchymal malignancy arising in bone and soft tissues (Ladenstein et al, 2010). EwS are characterized by a specific chromosomal translocation, most commonly of chromosomes 22 and 11 [t(1 1 ,22)(q24; 12)], resulting in the aberrant chimeric transcription factor EWSR1-FLI1 (Delattre et al, 1992). The inventors and others have shown that EwS can express the disialoganglioside G D2 on the cell surface (Kailayangiri et al, 2017; Long et al, 2016).
  • G D2 expression normally characterizes immature neuroectodermal cells, with restricted and low-level tissue expression after birth on neuronal cells in the CNS, peripheral nerves and mesenchymal stroma cells (Martinez et al, 2007; Suzuki, 1965), reviewed in (Rossig et al, 2018). G D2 was first evaluated as a therapeutic target in neuroblastoma, a cancer with abundant G D2 surface expression due to its tissue origin from neuroectoderm (Ladenstein et al, 2018; Yu et al, 2010).
  • the present invention relates to an Enhancer of Zeste Homolog 2 (EZH2) inhibitor for use in a method of treating a solid tumor in a subject, wherein the method further comprises the administration of a G D2 -specific CAR-T or CAR-NK cell or a G D2 -specific antibody cell to the subject.
  • EZH2 Enhancer of Zeste Homolog 2
  • the present invention additionally relates to a G D2 -specific chimeric antigen receptor (CAR)-T cell or CAR-NK cell or a G D2 -specific antibody for use in a method of treating a solid tumor in a subject, wherein (i) the method further comprises the administration of an EZH2 inhibitor to the subject; and/or (ii) the subject has been and/or is treated with an EZH2 inhibitor.
  • CAR chimeric antigen receptor
  • the EZH2 inhibitor may be selected from the group consisting of GSK126, EPZ-6438 (tazemetostat), 3-deazaneplanocin A (DZNep), EPZ005687, EI 1 , and UNC1999.
  • the solid tumor may be selected from the group consisting of Ewing sarcoma, breast cancer, preferably an adenocarcinoma of the breast, neuroblastoma, melanoma, small cell lung carcinoma, lung carcinoma, brain tumors, retinoblastoma and osteosarcoma.
  • the solid tumor is Ewing sarcoma.
  • the solid tumor is neuroblastoma.
  • the solid tumor is osteosarcoma.
  • the solid tumor is breast cancer.
  • the solid tumor is lung cancer.
  • the CAR-T cell or the CAR-NK cell preferably comprises a chimeric antigen receptor specific (CAR) for G D2 and wherein the CAR is Q ⁇ 2-BBz or GD2-CD28 z.
  • the CAR ⁇ 02-BBz has an amino acid sequence with at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 98%, at least 99%, or even higher sequence identity to the amino acid sequence shown in SEQ ID NO: 3.
  • the CAR Q ⁇ 2-28z has an amino acid sequence with at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 98%, at least 99%, or even higher sequence identity to the amino acid sequence shown in SEQ ID NO: 4.
  • ⁇ 02-BBz or GD2-CD28C may comprise the CDR-H1 as depicted in SEQ ID NO: 15, the CDR-H2 as depicted in SEQ ID NO: 16, the CDR-H3 as depicted in SEQ ID NO: 17, the CDR-L1 as depicted in SEQ ID NO: 18, the CDR-L2 as depicted in SEQ ID NO: 19 and the CDR-L3 as depicted in SEQ ID NO: 20.
  • the G D2 -specific antibody preferably is dinutuximab or dinutuximab beta or an antibody having the same CDR sequences as is dinutuximab (produced in mouse cells) or dinutuximab beta (ch 14.18/produced in CHO cells) or having the same VH and VL regions as dinutuximab or dinutuximab beta.
  • Dinutuximab or dinutuximab beta may comprise the CDR-H1 as depicted in SEQ ID NO: 15, the CDR-H2 as depicted in SEQ ID NO: 16, the CDR-H3 as depicted in SEQ ID NO: 17, the CDR-L1 as depicted in SEQ ID NO: 18, the CDR-L2 as depicted in SEQ ID NO: 19 and the CDR-L3 as depicted in SEQ ID NO: 20.
  • the EZH2 inhibitor may be capable of increasing the surface expression of G D2 on G D2 - low and/or G D2 -negative cancer cells.
  • the EZH2 inhibitor does not increase the surface expression of G D2 in non cancer cells.
  • the EZH2 inhibitor may be administered before and/or simultaneously to administration of the CAR-T cell or CAR-NK cell or a G D2 -specific antibody to the subject.
  • the EZH2 inhibitor is for use of the invention or the G D2 -specific CAR-T cell or CAR-NK cell for use of the invention, wherein the method comprises a further anti-cancer treatment.
  • the further anti-cancer treatment is chemotherapy, radiotherapy, immunotherapy or surgery.
  • the subject may be human.
  • the present invention relates to a combination of an EZH2 inhibitor, and a G D2 -specific CAR-T cell or CAR-NK cell or a G D2 -specific antibody for use in a method of treating a solid tumor in a subject.
  • the EZH2 inhibitor is selected from the group consisting of GSK126, EPZ-6438 (tazemetostat), 3-deazaneplanocin A (DZNep), EPZ005687, EI1 , and UNC1999.
  • the present invention also relates to a method of enhancing susceptibility of a tumor or a tumor cell to G D2 -specific CAR-T cell or CAR-NK cell or GG D2 -specific antibody treatment comprising contacting the tumor or tumor cell with an EZH2 inhibitor.
  • the method of enhancing susceptibility of a tumor or a tumor cell to G D2 -specific CAR-T cell or CAR-NK cell or G D2 -specific antibody treatment comprises administering the EZH2 inhibitor to a subject.
  • the tumor or tumor cell may be in the subject.
  • the method of enhancing susceptibility of a tumor or a tumor cell to G D2 -specific CAR-T cell or CAR-NK cell or G D2 -specific antibody treatment may be an in vitro method.
  • the tumor or tumor cell is in a subject and the EZH2 inhibitor is administered to the subject or in other words, the method of enhancing susceptibility of a tumor or a tumor cell to G D2 -specific CAR-T cell or CAR-NK cell or G D2 -specific antibody treatment may be an in vivo method.
  • the present invention relates to a kit comprising an EZH2 inhibitor and a Go 2 -specific CAR-T cell or CAR-NK cell or a G D2 -specific antibody.
  • the present invention further relates to a kit-of-parts comprising an EZH2 inhibitor and a G D2 -specific CAR-T cell or CAR-NK cell or a G D2 -specific antibody.
  • (A) Go surface expression by flow cytometry in 19 EwS cell lines. According to RFI, cell lines were categorized as G D2 pos (RFI 32) or G D2 neg (RFI ⁇ 2). Representative experiments of two or three.
  • FIG. 1 Upregulation of G D 2 expression by EZH2 inhibition is reversible and limited to EwS cell lines.
  • A G D2 surface expression by flow cytometry in 8 G D2 neg EwS cell lines cultured with 4 pM GSK126 or equivalent volumes of DMSO (control) for 7 days (upper panel) and for 28 days (lower panel).
  • RD RFI after incubation with DMSO
  • RG RFI after incubation with GSK126.
  • FIG. 1 Expression of EZH2 and ganglioside synthases GD3S and GD2S in EwS cells.
  • A EZH2 gene expression in 9 G D2 neg and 10 G D2 pos EwS cell lines by quantitative RT- PCR.
  • B GD3S and GD2S gene expression in 9 G D2 neg and 10 G D2 pos EwS cell lines by quantitative RT-PCR.
  • C GD3S and GD2S gene expression in 9 G D2 neg EwS cell lines following 14 days of incubation in the presence of 4 pM GSK126 or DMSO (control) by quantitative RT-PCR.
  • D Screen shots of ChIP-seq data analysis of SK-N-MC EwS cells using UCSC genome Browser.
  • FIG. 4 High G D2 surface expression in EwS cells does not affect in vitro proliferation colony formation, chemosensitivity, and in vivo tumorigenicity.
  • A FACS selection strategy of tumor cell subpopulations with the highest (Go 2 hi, 30% or 1 %) and lowest (G D2 IOW, 30% or 1 %) G D2 surface expression, exemplified by cell line VH-64.
  • B G D2 surface expression on sorted G D2 hi and G D2 low subpopulations (30%) determined by flow cytometry every 6 days during subsequent cell culture over a period of 3 weeks. The median RFIs of G D2 expression in the two subpopulations are shown in relation to bulk cells from the same cell lines. *p ⁇ 0.02 (t-test).
  • C In vitro expansion of tumor cells from the sorted subpopulations (30%) for 6 days of standard monolayer cultures, quantified by trypan blue exclusion and cell counting.
  • D Colony formation of tumor cells from sorted subpopulations (30%) in semisolid media after 7 or 8 days of culture.
  • E Viabilities of tumor cells from the sorted subpopulations (30%) following incubation with the indicated concentrations of doxorubicin by luminometry.
  • F Tumor formation of G D2 hi and G D2 low subpopulations selected from VH-64 and MS-EwS-16 cells after subcutaneous or intravenous transplantation into NSG mice at the indicated cell numbers.
  • FIG. 5 Disruption of G D2 surface expression in EwS cells by GD3S gene editing does not affect colony formation, tumorigenicity and chemosensitivity.
  • A G D2 expression in 3 EwS cell lines following disruption of the GD3S gene by CRISPR/Cas9 genome editing (GD3S KO) or in wild-type cells (mock control) by flow cytometry.
  • B Proliferation of GD3S KO or wild-type EwS cells after 3-day in vitro culture by luminometry.
  • C Colony formation of GD3S KO or wild-type EwS cells in semisolid media.
  • D Viabilities of GD3S KO and wild-type cells following incubation with the indicated concentrations of doxorubicin by luminometry.
  • FIG. 1 Figure 6. EZH2 inhibitor pretreatment sensitizes G D 2-negative EwS cell lines to in vitro cytolysis by G D2 -specific CAR-T cells.
  • A Exemplary gating strategies for quantification of intracellular cytokine expression and CD107a expression in CAR (GD2-BBz)-transduced T cells.
  • B, C Quantification of intracellular expression of IFN-y (B) and TNF-a (C) in non- transduced (NT) or ⁇ 02-BBz transduced T cells co-incubated for 6 h with EwS cell lines pretreated with 4 mM GSK126 or DMSO alone for 14 days. Shown is one representative experiment of two.
  • FIG. 7 In vitro expansion and colony formation of Go 2 neg EwS cells cultured in the presence of GSK126.
  • A In vitro expansion rates of 9 G D2 neg EwS cell lines cultured in the presence of 4 pM GSK126 or equivalent volumes of DMSO (control) for 14 days by trypan blue exclusion and cell counting.
  • B Colony formation of tumor cells cultured in semisolid medium in the presence of 4 pM GSK126 or equivalent volumes of DMSO (control) for 14 days. Statistical analysis for both assays was done by t-test.
  • FIG. 8 Proliferation, colony formation and viability of tumor cells from G D2 pos and G D2 neg EwS cell lines.
  • A Proliferation of tumor cells from 10 G D2 pos and 9 G D2 neg EwS cell lines (defined as in Figure 1A) after 3 days of in vitro culture by luminometry. RLU, relative luminescence unit. Statistical analysis by t-test.
  • B Colony formation of tumor cells from 10 G D2 pos and 9 G D2 neg EwS cell lines in semisolid agar. Statistical analysis by t-test.
  • C Viabilities of tumor cells from 10 G D2 pos and 9 G D2 neg EwS cell lines following incubation with the indicated concentrations of doxorubicin by luminometry. Statistical analysis by Rank sum test for all concentrations.
  • FIG. 11 Pretreatment of G D2 -negative Ewing sarcoma cell lines with the EZH2 inhibitor renders the tumor cells susceptible to G D2 -specific antibody dependent cytolysis.
  • PBMCs were pre-activated with 500 U/ml IL-2 for 48 h and then analyzed for 4 h in the presence and absence of the G D2 -specific antibody Dinutuximab (100 pg/ml) at effector-to- target (E:T) ratios of 40:1 , 20: 1 and 10: 1.
  • the tumor cells RD-ES were treated with 10 mM DMSO (control) or GSK126 for 7 days prior the co-incubation. % cytolysis was quantified by luminometry.
  • Figure 12 In vitro EZH2 inhibition induces G D 2 upregulation also in lung cancer.
  • Tumor cells from the lung cancer cell line A549 were cultured in the presence of 1 mM tazemetostat dissolved in DSMO (dark grey) or DMSO (control, light grey) for 14 days, and then G D2 surface expression (PE) was quantified by flow cytometry. Isotype controls of the DMSO- treated cells (white area, black dashed lines) and of the GSK126-treated cells (white area, solid black line) were also performed.
  • G D2 is a very interesting target for tumor therapy because of its almost exclusive expression on tumor cells.
  • therapies making use of this tumor-specific expression are hampered by the inhomogeneous expression of G D2 on tumor cells.
  • G D2 -low or even -negative populations that can escape such a therapy.
  • previous tries to develop a therapy targeting G D2 -expressing tumor cells were not successful.
  • G D2 -specific CAR- T cell or CAR-NK cell therapies.
  • EZH2 enhancer of Zeste Homolog 2
  • EZH2 Enhanccer of Zeste Homolog 2
  • EZH2 a histone- lysine N-methyltransferase enzyme
  • Histone-lysine N- methyltransferase EZH2 ENX-1 or Lysine N-methyltransferase 6.
  • EZH2 participates in histone methylation and, ultimately, transcriptional repression.
  • EZH2 acts as a histone methyltransferase and silences genes involved in cell differentiation in a highly context- dependent manner by depositing repressive histone marks at histone 3 lysine 27 (H3K27me3) (Comet et al, 2016) by using the cofactor S-adenosyl-L-methionine. Methylation activity of EZH2 facilitates heterochromatin formation thereby silences gene function.
  • An exemplary EZH2 can be found in the UniProt database using the accession number Q15910-v2, whose sequence is also shown in SEQ ID NO: 11. [036]
  • An ⁇ ZH2 inhibitor” as used herein relates to an inhibitor, which reduces or inhibits the activity of EZH2.
  • Reducing or inhibiting the activity of EZH2 may relate to a reduced methylation of lysine 27 of histone H3 (H3K27).
  • the activity of EZH2 may be determined by an assay as essentially described in Example 1 and Fig. 1C.
  • the rate of lysine 27 of histone H3 trimethylation (H3K27me3) is analyzed by Western Blot using an antibody that detects the trimethylated form of H3K27.
  • the EZH2 inhibitor GSK126 almost completely blocks the trimethylation of H3K27 in comparison to a mock control.
  • An exemplary anti-H3K27me3 antibody is available from abeam as ab6002.
  • EZH2 and histone H3 are prepared in reaction buffer (20 mM Hepes pH 7.5, 10 mM MgCI2, 1 mM EGTA, 0.02% Brij35, 0.02 mg/ml BSA, 0.1 mM Na 3 V0 4 , 2 mM DTT, 1 % DMSO).
  • reaction buffer (20 mM Hepes pH 7.5, 10 mM MgCI2, 1 mM EGTA, 0.02% Brij35, 0.02 mg/ml BSA, 0.1 mM Na 3 V0 4 , 2 mM DTT, 1 % DMSO).
  • a putative EZH2 inhibitor is added to the reaction.
  • the reaction may then be started by the addition of 3 H-SAM (S-adenosyl methionine) and incubated, e.g. for 1 h at 37 °C. After washing away the unbound or not-reacted 3 H-SAM, the radioactivity of the histone H3 may be measured and compared to a control, in which no EZH2 inhibitor has been added.
  • An EZH2 inhibitor may reduce the activity of EZH2 by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98% or at least 99% compared to a mock control.
  • EZH2 inhibitors [037] Most of the EZH2 inhibitors known so far target the interaction of EZH2 with its co-factor S-adenosyl methionine (SAM), i.e. compete for SAM binding to EZH2.
  • SAM S-adenosyl methionine
  • GSK126 is a SAM- competitive EZH2 inhibitor.
  • the EZH2 inhibitors of the invention preferably are EZH2 specific and do not inhibit any other histone methylases such as EZH1. Examples of EZH2 inhibitors include, but are not limited to, GSK126, EPZ-6438 (tazemetostat), 3-deazaneplanocin A (DZNep), EPZ005687, EI 1 , and UNC1999.
  • Table 1 shows the structure of different EZH2 inhibitors:
  • G D2 expression marks in breast cancer a cell population with higher capacity to self-renew and reinitiate tumor growth (Battula et al, 2012; Liang et al, 2013).
  • GD 2 expression in EwS cells was surprisingly not associated with distinct functional properties (see also Examples 3 and 4).
  • the administration of an EZH2 inhibitor does not induce a more dangerous phenotype.
  • the inventors After having found that the administration of an EZH2 inhibitor increases the surface expression of G D2 in tumor cells, the inventors further analyzed whether this increase is limited to solid tumor cells. As shown in Fig. 2C, healthy and non-solid tumor cells do not express G D2 even after stimulation with an EZH2 inhibitor. This feature underlines the safety of increasing GD2 expression on solid tumor cells. Thus, increasing the surface expression of G D2 is safe. Accordingly, the EZH2 inhibitor preferably does not increase the surface expression of G D2 in non-cancer cells, more preferably in non-solid tumor cells.
  • a therapeutic strategy based on the surprising findings of the inventors would include the administration of an EZH2 inhibitor to a subject to increase the expression of G D2 specifically on the solid tumor.
  • the increased surface expression of G D2 on the solid tumor may then be used for targeting G D2 -specific CAR-T cells or CAR-NK cells to the solid tumor cells.
  • those solid tumor cells may then be specifically killed, thereby treating the solid tumor.
  • a G D2 -specific antibody may be administered to the subject.
  • the present invention relates to an Enhancer of Zeste Homolog 2 (EZH2) inhibitor for use in a method of treating a solid tumor in a subject, wherein the method further comprises the administration of a G D2 -specific CAR-T cell or CAR-NK cell or a G D2 -specific antibody to the subject.
  • EZH2 Enhancer of Zeste Homolog 2
  • the present invention relates to an Enhancer of Zeste Homolog 2 (EZH2) inhibitor for use in a method of treating a solid tumor in a subject, wherein the method further comprises the administration of a G D2 -specific CAR-T cell to the subject.
  • EZH2 Enhancer of Zeste Homolog 2
  • the present invention relates to an Enhancer of Zeste Homolog 2 (EZH2) inhibitor for use in a method of treating a solid tumor in a subject, wherein the method further comprises the administration of a G D2 -specific CAR-NK cell to the subject.
  • EZH2 Enhancer of Zeste Homolog 2
  • the present invention relates to an Enhancer of Zeste Homolog 2 (EZH2) inhibitor for use in a method of treating a solid tumor in a subject, wherein the method further comprises the administration of G D2 -specific antibody to the subject.
  • EZH2 Enhancer of Zeste Homolog 2
  • the present invention also relates to a G D2 -specific chimeric antigen receptor (CAR-) T cell or CAR-NK cell or a G D2 -specific antibody for use in a method of treating a solid tumor in a subject, wherein (i) the method further comprises the administration of an EZH2 inhibitor to the subject; and/or (ii) the subject has been and/or is treated with an EZH2 inhibitor.
  • CAR- G D2 -specific chimeric antigen receptor
  • the present invention also relates to a G D2 -specific chimeric antigen receptor (CAR-) T cell for use in a method of treating a solid tumor in a subject, wherein (i) the method further comprises the administration of an EZH2 inhibitor to the subject; and/or (ii) the subject has been and/or is treated with an EZH2 inhibitor.
  • CAR- G D2 -specific chimeric antigen receptor
  • the present invention also relates to a G D2 -specific chimeric antigen receptor (CAR-) NK cell for use in a method of treating a solid tumor in a subject, wherein (i) the method further comprises the administration of an EZH2 inhibitor to the subject; and/or (ii) the subject has been and/or is treated with an EZH2 inhibitor.
  • CAR- G D2 -specific chimeric antigen receptor
  • the present invention also relates to a G D2 -specific antibody for use in a method of treating a solid tumor in a subject, wherein (i) the method further comprises the administration of an EZH2 inhibitor to the subject; and/or (ii) the subject has been and/or is treated with an EZH2 inhibitor.
  • an EZH2 inhibitor it is possible to begin with the administration of an EZH2 inhibitor to the subject prior to, i.e. before, treatment with G D2 -specific CAR-T or CAR-NK cells or the G D2 - specific antibody. It is also possible to first start the treatment with G D2 -specific CAR-T or CAR- NK cells or the G D2 -specific antibody and then to support this treatment by the administration of an EZH2 inhibitor to the subject, i.e. to simultaneously administer the EZH2 inhibitor and the CAR-T or CAR-NK cell or G D2 -specific antibody to the subject.
  • the EZH2 inhibitor may be administered to the subject at any time before or simultaneously to the administration of the CAR-T cell or the CAR-NK cell or the G D2 -specific antibody to the subject.
  • a “chimeric antigen receptors” is a membrane-bound artificial receptor that is designed to bind to a target cell-specific antigen such as G D2 and then induce a signal cascade in an effector cell such as a T cell or a NK cell that expresses the CAR.
  • CARs usually may comprise an extracellular domain, transmembrane domain and intracellular domain.
  • the extracellular domain is the region of the CAR that is exposed to the extracellular fluid and comprises an antigen recognition region.
  • the antigen recognition domain is designed to bind GD 2 .
  • Examples for such an antigen recognition domain include, but are not limited to, an antibody or an antibody fragment or a proteinaceous binding molecule with antibody-like properties.
  • the antibody fragment thereof may be selected from the group consisting of an scFv, a di-scFv, a bi-scFv, a Fab, an Fc, an F(ab’)2, a pFc’, a nanobody, an affibody, a DARPin, a diabody, a camelid, an engineered T cell receptor and a monobody.
  • the antibody may be selected from the group consisting of an lgA1 , an lgA2, an IgD, an IgM, an IgE, an lgG1 , an lgG2, an lgG3, and an lgG4.
  • the CAR may comprise at least a portion of a single chain variable fragment (scFv).
  • the antibody may be human, fully human, humanized, human engineered, non-human, and/or chimeric antibody.
  • the transmembrane domain and/or the intracellular domain of the CAR may comprise at least a portion of a cytoplasmic signaling domain.
  • the intracellular domain may comprise at least a portion of a signaling molecule selected from the group comprising CD3zeta, CD28, and 4-1 BB, 2B4, ICOS, OX-40, or other.
  • the signaling molecule may comprise CD3zeta.
  • the signaling molecule may comprise CD28.
  • the signaling molecule may comprise 4-1 BB.
  • the intracellular domain may comprise at least a portion of CD3zeta.
  • the intracellular domain may comprise at least a portion of CD28.
  • the intracellular domain may comprise at least a portion of 4-1 BB.
  • the intracellular domain may comprise at least a portion of OX-40.
  • the intracellular domain may comprise at least a portion of 2B4.
  • the intracellular domain may comprise at least a portion of ICOS.
  • the intracellular domain may comprise at least a portion of a cytoplasmic signaling domain from one or more signaling molecules.
  • the intracellular domain may comprise at least a portion of two or more cytoplasmic signaling domains.
  • the two or more cytoplasmic signaling domains may be from two or more different signaling molecules.
  • the intracellular domain may comprise at least a portion of three or more cytoplasmic signaling domains.
  • the intracellular domain may comprise at least a portion of four or more cytoplasmic signaling domains.
  • the intracellular domain may comprise at least a portion of a ligand that binds to one or more signaling molecules.
  • a “G D2 -specific CAR” comprises an extracellular domain that binds G D2 , i.e. a G D2 - specific extracellular domain, preferably when G D2 is expressed on the solid tumor.
  • G D2 -specific CAR comprises an extracellular domain that binds G D2 , i.e. a G D2 - specific extracellular domain, preferably when G D2 is expressed on the solid tumor.
  • a person skilled in the art can readily determine whether an extracellular domain of a CAR binds G D2 .
  • a G D2 -specific extracellular domain of a CAR may bind G D2 with a binding affinity of less than about 1 nM, about 2 nM, about 2.5 nM, about 3 nM, about 4 nM, about 5 nM, about 6 nM, about 7 nM, about 8 nM, about 9 nM, about 10 nM, about 12nM, about 14 nM, about 16 nM, about 18 nM, about 20 nM, about 25 nM, about 50 nM, about 100 nM, about 125 nM or about 250 nM.
  • G D2 -specific CARs include, but are not limited to, ⁇ 02-BBz as depicted in the nucleic acid sequence of SEQ ID NO: 1 or the amino acid sequence of SEQ ID NO: 3, or GD2-28C, as depicted in the nucleic acid sequence of SEQ ID NO: 2 or the amino acid sequence of SEQ ID NO: 4.
  • the G D2 -specific CAR-T cells or CAR-NK cells described herein may comprise or express a G D2 -specific CAR as defined herein.
  • ⁇ 02-BBz and GD2-28 z for example, both comprise a single-chain antibody domain (scFv) of the monoclonal antibody 14.G2a.
  • a scFv derived from monoclonal antibody 3F8 or any other G D2 - specific monoclonal antibody may be used in a G D2 -specific CAR.
  • Antibody 3F8 or a scFv derived from 3F8 may comprise 3 heavy chain CDRs as depicted in SEQ ID NOs: 5-7 and 3 light chain CDRs as depicted in SEQ ID NOs: 8-10, which may be comprised in a G D2 -specific CAR comprised in a G D2 -specific CAR-T cell or CAR-NK cell.
  • Antibody 14.G2a or an scFv derived from 14.G2a may comprise the CDR-H1 as depicted in SEQ ID NO: 15, the CDR-H2 as depicted in SEQ ID NO: 16, the CDR-H3 as depicted in SEQ ID NO: 17, the CDR-L1 as depicted in SEQ ID NO: 18, the CDR-L2 as depicted in SEQ ID NO: 19 and the CDR-L3 as depicted in SEQ ID NO: 20, which may be comprised in a G D2 -specific CAR comprised in a Goa- specific CAR-T cell or CAR-NK cell.
  • Antibody 14.G2a may comprise a heavy chain as depicted in SEQ ID NO: 21 and a light chain as depicted in SEQ ID NO: 22, which may be comprised in a G D 2-specific CAR comprised in a G D 2-specific CAR-T cell or CAR-NK cell. Further G D 2-specific CARs are also described in WO 2013/040371 and WO 2016/134284, which are hereby incorporated by reference.
  • CDRs sequences of the disclosure follow the definition according to Kabat, as described in Sequences of Proteins of immunological Interest, US Department of Health and Human Services (1991), eds. Kabat et al. Other standards for defining CDRs exist as well, such as the definition according to Maass 2007 (Journal of Immunological Methods 324 (2007) 13-25).
  • Another standard for characterizing the antigen binding site is to refer to the hypervariable loops as described by Chothia (see, e.g., Chothia, et al. (1992); J. Mol. Biol. 227:799-817; and Tomlinson et al. (1995) EMBO J.
  • CDRs A further standard for defining CDRs is the definition according to AbM, used by Oxford Molecular's AbM antibody modelling software. See, generally, e.g., Protein Sequence and Structure Analysis of Antibody Variable Domains. In: Antibody Engineering Lab Manual (Ed.: Duebel, S. and Kontermann, R., Springer-Verlag, Heidelberg). It is understood that embodiments described with respect to the CDR definition of Kabat, can alternatively be implemented using similar described relationships such as with respect to Maass, AbM, or Chothia definition.
  • the CAR ⁇ 02-BBz has an amino acid sequence with at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 98%, at least 99%, or even higher sequence identity to the amino acid sequence shown in SEQ ID NO: 3.
  • the CAR ⁇ 02-BBz may comprise the CDR-H1 as depicted in SEQ ID NO: 15, the CDR-H2 as depicted in SEQ ID NO: 16, the CDR-H3 as depicted in SEQ ID NO: 17, the CDR-L1 as depicted in SEQ ID NO: 18, the CDR-L2 as depicted in SEQ ID NO: 19 and the CDR-L3 as depicted in SEQ ID NO: 20.
  • the CAR GD2-28C has an amino acid sequence with at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 98%, at least 99%, or even higher sequence identity to the amino acid sequence shown in SEQ ID NO: 4.
  • the CAR Q ⁇ 2-28z may comprise the CDR-H1 as depicted in SEQ ID NO: 15, the CDR-H2 as depicted in SEQ ID NO: 16, the CDR-H3 as depicted in SEQ ID NO: 17, the CDR-L1 as depicted in SEQ ID NO: 18, the CDR-L2 as depicted in SEQ ID NO: 19 and the CDR-L3 as depicted in SEQ ID NO: 20.
  • identity or“sequence identity” is meant a property of sequences that measures their similarity or relationship.
  • sequence identity means the percentage of pair-wise identical residues - following (homology) alignment of a sequence of a polypeptide of the invention with a sequence in question - with respect to the number of residues in the longer of these two sequences. Identity is measured by dividing the number of identical residues by the total number of residues and multiplying the product by 100.
  • the percentage of sequence identity can, for example, be determined herein using the program BLASTP, version blastp 2.2.5 (November 16, 2002; cf. Altschul, S. F. et al. (1997) Nucl. Acids Res. 25, 3389-3402).
  • the percentage of homology is based on the alignment of the entire polypeptide sequences (matrix: BLOSUM 62; gap costs: 11.1 ; cutoff value set to 10 3 ) optionally including the propeptide sequences, using the SEQ ID NO: 3 or 4 as reference in a pairwise comparison. It is calculated as the percentage of numbers of "positives" (homologous amino acids) indicated as result in the BLASTP program output divided by the total number of amino acids selected by the program for the alignment.
  • A“G D2 -specific antibody” as used herein relates to any antibody that specifically binds to GD2, preferably on the surface of a solid tumor or solid tumor cell, and stimulates the subject’s immune system to attack the solid tumor or solid tumor cell. It is understood that the Goa- specific antibody is preferably a therapeutic antibody.
  • the G D 2-specific antibody may induce an antibody-dependent cell-mediated cytotoxicity (ADCC) thereby stimulating the lysis of the solid tumor or the solid tumor cells by immune cells such as natural killer cells (see also Example 6).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • the G D 2-specific antibody may be conjugated to a radioactive moiety or may be in the form of antibody-drug conjugates (ADC) or to an immune-activating agent in the form of a bispecific T cell engager.
  • a "bispecific T cell engager" (BiTE) preferably refers to a class of artificial bispecific monoclonal antibodies that are capable of directing a host's immune system, more specifically the T cells' cytotoxic activity, against cancer cells.
  • a bispecific T cell engager is preferably a fusion protein comprising or consisting of two single-chain variable fragments (scFvs) of different antibodies, or amino acid sequences from four different genes, on a single peptide chain.
  • One of the scFvs binds to T cells, e.g. via the CD3 receptor, and the other to a tumor cell, e.g. via a tumor specific molecule such as a tumor specific antigen, which preferably is in the context of the invention G D2 .
  • exemplary G D2 -specific antibodies include, but are not limited to, dinutuximab and dinutuximab beta or antibodies having the same CDRs as dinutuximab or dinutuximab beta or antibodies having the same VH and VL region as dinutuximab or dinutuximab beta.
  • Such an antibody may have a VH and/or VL region that has an amino acid sequence with at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 92%, at least 95%, at least 97%, at least 98%, at least 99%, or even higher sequence identity with the VH and/or VL region of dinutuximab or dinutuximab beta.
  • the VH domain of dinutuximab or dinutuximab beta is depicted in SEQ ID NO: 21 and the VL domain of dinutuximab or dinutuximab beta is depicted in SEQ ID NO: 22.
  • Dinutuximab or dinutuximab beta may comprise may comprise the CDR-H1 as depicted in SEQ ID NO: 15, the CDR-H2 as depicted in SEQ ID NO: 16, the CDR-H3 as depicted in SEQ ID NO: 17, the CDR-L1 as depicted in SEQ ID NO: 18, the CDR-L2 as depicted in SEQ ID NO: 19 and the CDR-L3 as depicted in SEQ ID NO: 20.
  • Dinutuximab is described in WO 2013/189516, which is hereby incorporated by reference.
  • G D2 -specific antibodies are preferably administered by parenteral administration such as an intravenous injection.
  • G D2 relates to a disialoganglioside that is also known as ganglioside G D2 . It may be represented by the following Formula I, which shows its structure.
  • G D2 may be expressed on tumors of neuroectodermal origin, including human neuroblastoma and melanoma or on tumor cells from sarcomas and other solid cancers, with highly restricted expression on normal tissues.
  • the relatively tumor specific expression of G D2 makes it a suitable target for immunotherapy with monoclonal antibodies or with artificial T cell receptors.
  • a CAR-T cell or CAR-NK immunotherapy makes use of an effector cell, i.e. a T cell or a NK cell, which has been genetically engineered with the CAR.
  • the effector cell may be a T cell.
  • the effector cell may be a cell of a T cell lineage.
  • the effector cell may be a mature T cell.
  • the effector cell may be a cytotoxic T cell.
  • the effector cell may be a naive T cell.
  • the effector cell may be a memory stem cell T cell (TMSC).
  • the effector cell may be a central memory T cell (TCM).
  • TCM central memory T cell
  • the effector cell may be an effector T cell (TE).
  • the effector cell may be a CD4+ T cell.
  • the T cell may be a CD8+ T cell.
  • the effector cell may be a CD4+ and CD8+ cell.
  • the effector cell may be an alpha-beta T cell.
  • the effector cell may be a gamma-delta T cell.
  • the effector cell may be a natural killer T cell.
  • the effector cell may be a natural killer (NK) cell.
  • the effector cell may be a cytokine-induced killer (CIK) cell.
  • the premise of CAR-T or CAR-NK immunotherapy in general is to modify effector cells such as T cells or NK cells to recognize cancer cells in order to more effectively target and destroy them.
  • the T cells or NK cells are genetically engineered to target solid tumor cells expressing G D2 on their surface, i.e. with a G D2 -specific CAR.
  • T cells or NK cells may be obtained from people, genetically altered ex vivo to express a CAR, and the resulting CAR-T cells or CAR-NK cells may then be infused into patients to attack their tumors such as solid tumors described herein.
  • Effector cells may be derived from a patient's own blood (autologous) or derived from the T cells or NK cells of another healthy donor (allogenic). Once isolated from a person, these T cells or NK cells are genetically engineered to express a specific G D2 -specific CAR, which programs them to target G D2 that is present on the surface of solid tumors described herein.
  • Methods for producing a CAR-T cell are e.g. disclosed in WO 2017/049166, which is hereby incorporated by reference in its entirety. These methods may analogously be applied to CAR-NK cells.
  • T cells or NK cells are isolated from the subject. Ex vivo, they are genetically modified to express the CAR, e.g. by viral transduction or transfection of a vector that comprises the CAR as well as regulatory elements such as a promoter. After verification of the genetic modification, the resulting CAR-T cells or CAR-NK cells may be administered to the subject. Suitable modes of administration for CAR-T cells or CAR-NK cells include intravenous, subcutaneous, intracavitary (for example by reservoir-access device), intraperitoneal, and direct injection into a tumor mass.
  • A“solid tumor” as used herein relates to an abnormal mass of tissue that usually does not contain cysts or liquid areas.
  • solid tumors are sarcomas, carcinomas, and lymphomas.
  • a solid tumor include, but are not limited to, Ewing sarcoma, breast cancer, preferably an adenocarcinoma of the breast, neuroblastoma, melanoma, small cell lung carcinoma, lung carcinoma, brain tumors, retinoblastoma and osteosarcoma.
  • the solid tumor is a Ewing sarcoma (EwS).
  • the solid tumor is osteosarcoma.
  • the tumor is neuroblastoma.
  • a “subject” as used herein relates to any mammal, including a human.
  • the subject is an adult, an adolescent or an infant.
  • terms “individuaf or " atient" are used and are intended to be interchangeable with “subject.”
  • the subject may suffer from a disease such as a solid tumor described herein.
  • the administration of an EZH2 inhibitor increases the surface expression in solid tumors such as Ewing sarcoma (EwS) but also in osteosarcoma, breast cancer and neuroblastoma.
  • EwS Ewing sarcoma
  • the EZH2 inhibitor of the invention preferably is capable of increasing the surface expression of G D2 on G D2 -low and/or G D2 -negative cancer cells.
  • “Surface expression of G D2 ” within this context relates to the amount or level of G D2 that is expressed and can be determined on the cell surface of a cell, e.g. a cancer cell or a cell of a solid tumor.
  • an “increase” of the surface expression of G D2 may mean an increase by at least 1.1 -fold, 1.25-fold, 1.5-fold, 2-fold, 5-fold, 10-fold, 25-fold, 50-fold or even 100-fold.
  • the surface expression of G D2 may be classified by using flow cytometry. One exemplary method is described in the materials and methods section of the examples. Here, the surface expression of G D2 was determined and the RFI (relative fluorescence intensity) of each cell line or groups of cell has been calculated. G D2 -negative cells may have a RFI of 2 or lower while G D2 -positive cells may have an RFI of more than 2. RFI was calculated by dividing median fluorescence intensities of mAb-stained cells by those obtained with isotype antibodies
  • the EZH2 inhibitor of the invention preferably does not increase the surface expression of G D2 in non-cancer cells.
  • “Non-cancer cells” as used herein relate to cell present in healthy tissues of the subject. These cells are characterized by stopping cell division when no cell division or the like is needed for tissue growth, repair or function and are unlike cancer cells, which continue cell division even in instances, where cell division is not needed.
  • the EZH2 inhibitor may be administered in combination or separated from the administration of the CAR-T cells or the CAR-NK cells or the G D2 -specific antibody.
  • administration means administering of a therapeutically effective dose of the EZH2 inhibitor to a subject.
  • administration also relates to a method of incorporating the EZH2 inhibitor into tissues of an organism. Different routes of administration are possible.
  • the EZH2 inhibitor of the present invention can, for example, be administered via different ways such as any parenteral or non-parenteral (enteral or topical) route that is therapeutically effective for (preferably proteinaceous) drugs.
  • Parenteral application methods include, for example, subcutaneous, intramuscular, intracerebral, intracerebroventricular, intrathecal, intranasal, intra- atrial, intraperitoneal or intravenous injection and infusion techniques, e.g. in the form of injection solutions, infusion solutions or tinctures.
  • Non-parenteral delivery modes are, for instance, enteral delivery modes such as oral delivery, e.g. in the form of pills, tablets, capsules, solutions or suspensions, or rectally, e.g. in the form of suppositories.
  • Topical application routes include epicutaneous or inhalational applications.
  • EZH2 inhibitors and pharmaceutical compositions of the present invention can be administered in formulations containing conventional non-toxic pharmaceutically acceptable excipients or carriers, additives and vehicles as desired and described herein.
  • EZH2 inhibitors are often small molecules that are suitable for an oral administration.
  • the EZH2 inhibitor is administered orally.
  • a simultaneous parenteral administration e.g. an intravenous injection, together with the CAR-T cell or the CAR-NK cell or the Goa- specific antibody is envisioned.
  • a composition comprising the EZH2 inhibitor and the CAR-T cell or CAR-NK cell or the G D 2-specific antibody is administered to the subject, preferably by an intravenous injection.
  • Cancer treatment very often does not only rely on only one agent or medicament for treating. Very often, there is a combination of different medicaments to provide a better treatment of the tumor.
  • the treatment of the solid tumor comprising the administration of an EZH2 inhibitor and a G D2 -specific CAR-T cell or CAR-NK cell or the G D2 -specific antibody as described herein may be supported by a further anti-cancer treatment.
  • the method of treating a solid tumor of the invention may comprise a further anti-cancer treatment.
  • Exemplary further anti-cancer treatments include, but are not limited to, chemotherapy, radiotherapy, immunotherapy or surgery.
  • the present invention also relates to a combination of an EZH2 inhibitor and a G D2 - specific CAR-T cell or a combination of an EZH2 inhibitor and a Go 2 -specific CAR-NK cell or a combination of an EZH2 inhibitor and the G D2 -specific antibody for use in a method of treating a solid tumor in a subject.
  • the combination may be in the form of a pharmaceutical composition.
  • the combination or the pharmaceutical composition of the invention may further comprise a suitable buffer.
  • Suitable buffers include buffers, in which the fusion proteins or pharmaceutical compositions of the present invention can be stored or in which they can be directly administered to the subject. In the latter case, such buffer is a non-toxic, physiologically acceptable buffer.
  • All embodiments described herein within the context of the EZH2 inhibitor for use in a method of treating a solid tumor or a G D2 -specific CAR-T cell or CAR-NK cell or the G D2 -specific antibody for use in a method of treating a solid tumor also apply for the combination of an EZH2 inhibitor and a G D2 -specific CAR-T cell or a combination of an EZH2 inhibitor and a G D2 -specific CAR NK-cell or the G D2 -specific antibody.
  • Example 5 shows that the upregulation of G D2 by an EZH2 inhibitor increases the susceptibility of Ewing Sarcoma (EwS), a solid tumor, to a G D2 -specific CAR-T cell treatment.
  • EwS Ewing Sarcoma
  • This feature of EZH2 inhibitors may be used to make solid tumors more susceptible to a G D2 - specific CAR-T cell or NK cell treatment but also for G D2 -specific antibody treatments. Therefore, the present invention additionally relates to a method of enhancing susceptibility of a tumor or a tumor cell to a G D2 -specific CAR-T cell or CAR-NK cell treatment or the G D2 -specific antibody comprising contacting the tumor or tumor cell with an EZH2 inhibitor.
  • the method of enhancing susceptibility of a tumor or a tumor cell to G D2 -specific CAR-T cell or CAR-NK cell treatment or G D2 -specific antibody treatment of the invention is an in vitro method.
  • the tumor or tumor cell is in a subject and the EZH2 inhibitor is administered to the subject.
  • All embodiments described herein within the context of the EZH2 inhibitor for use in a method of treating a solid tumor or a G D2 -specific CAR-T cell or CAR-NK cell or the G D2 -specific antibody for use in a method of treating a solid tumor also apply for the method of enhancing susceptibility of a tumor or a tumor cell to a G D2 -specific CAR-T cell or CAR-NK cell or the G D2 -specific antibody treatment.
  • “Susceptibility of a tumor or a tumor cell to G D2 -specific CAR-T cell or NK cell or G D2 - specific antibody treatment” as used herein relates to the efficacy of a G D2 -specific CAR-T cell or NK cell or G D2 -specific antibody treatment against a specific tumor or tumor cell of a solid tumor as described herein.
  • a G D2 -specific CAR-T cell or CAR-NK cell therapy or a G D2 - specific antibody therapy shows a higher efficacy after treatment with one or more EZH2 inhibitor(s), e.g.
  • the solid tumor or tumor cells shows a higher susceptibility to G D2 -specific CAR-T cell or CAR-NK cell treatment or a G D2 -specific antibody treatment.
  • Assays for determining the amount of solid tumor cells killed by a CAR-T cell or CAR-NK cell treatment or the cytotoxicity of a CAR-T cell or CAR-NK cell are known to a person skilled in the art.
  • One example for such an assay is a calcein-AM release assay as described in the“Materials and Methods” section of the Examples or published in Neri et al.
  • “Higher susceptibility” of the solid tumor or tumor cell to a G D2 -specific CAR-T cell or CAR-NK cell therapy may relate to an increase of killed G D2 -treated tumor or tumor cells by at least 1.1 -fold, 1.25-fold, 1.5-fold, 2-fold, 5-fold, 10-fold, 25-fold, 50-fold or even 100-fold in comparison to untreated or mock-treated tumors or tumor cells.
  • the present invention further relates to a kit comprising an EZH2 inhibitor and a G D2 - specific CAR-T cell or CAR-NK cell or a G D2 -specific antibody.
  • the present invention further relates to a kit-of-parts comprising an EZH2 inhibitor and a G D2 -specific CAR-T cell or CAR-NK cell or a G D2 -specific antibody.
  • the kit or the kit-of-parts may further comprise a pharmaceutically acceptable vehicle.
  • the kit or the kit-of-parts may further comprise a buffer, preservative agents, coloring agents and the like.
  • less than 20 means less than the number indicated.
  • more than or greater than means more than or greater than the indicated number, e.g. more than 80% means more than or greater than the indicated number of 80%.
  • MS-EwS-1 previously described as MS-PES-1 (Kailayangiri et al, 2012)
  • MS-EwS-4 originally described as MS-PES-4 (Kailayangiri et al, 2017)
  • MS-EwS-6 originally described as MS-PES-6 (Spurny et al, 2018)
  • MS-EwS-16 originally described as DC-ES-6 (Leuchte et al, 2014)
  • MS-EwS-15 originally described as DC-ES-15 (Unland et al, 2015)
  • MS-EwS-34 were established from biopsy material of individual patients obtained at metastatic relapse, as reported previously (Kailayangiri et al, 2012; Leuchte et al, 2014; Spurny et
  • A4573, 5838, TTC-466 and TC-32 were a gift from the Children's Hospital Los Angeles.
  • A673, TC-71 , SK-ES-1 , RD-ES, and CADO-ES-1 were purchased from DSMZ (Braunschweig, Germany).
  • A204 is a rhabdoid tumor cell line purchased from DSMZ (Braunschweig, Germany).
  • Normal human fibroblasts generated from skin biopsies were obtained from Cliona Rooney (Houston, TX, USA).
  • Mesenchymal stem cells of two donors were cultured as described (Gieseke et al, 2013).
  • the B-cell precursor leukemia cell line SupB15 and the T cell leukemia cell line Jurkat were purchased from ATCC.
  • T cell cultures and B-lymphoblastoid cell lines were generated from healthy donors as described (Altvater et al, 2006). The identity of the cell lines was confirmed by short tandem repeat (STR) profiling.
  • STR short tandem repeat
  • tumor cells were cultured in collagen-coated 25 or 75 cm 2 tissue culture flasks (MS-EwS-1 , MS-EwS-4, MS- EwS-6, MS-EwS-15, MS-EwS-16, MS-EwS-34, TC-71 , TC-32, VH-64, CADO-ES-1 , SK-ES-1 , SK-N-MC, RD-ES and WE-68) or in uncoated flasks (all others) in RPMI 1640 medium (Invitrogen, Germany), supplemented with 10% heat-inactivated fetal calf serum (FCS; Thermo Fisher) and 2 mM L-glutamine (Sigma-Aldrich, Germany) and maintained at
  • EwS cell lines were sorted into G D2 low (30%) and G D2 hi (30%) subpopulations as described above and were seeded with 1x10 6 cells in 25 mm 2 culture flasks. Every six days, cells were harvested and stained with PE-conjugated G D2 antibody 14.G2a over a period of three weeks. The RFI of G D2 expression in each subpopulation was calculated in relation to control cells having undergone the sorting procedure without selection of subpopulations.
  • EwS tumor cells were harvested, counted and seeded in collagen-coated 6-well plates (Sarstedt, Germany) at 0.1 to 0.3x10 6 cells/well in a total volume of 2 ml. After 2-4 h, the EZH2 inhibitor GSK126 (Active Biochemicals, USA) or tazemetostat (Cayman Chemicals, USA) dissolved in DMSO or DMSO alone as control were added at concentration of 10 to 12 mM, 4 pM or 1 pM as indicated in the figures. After 4 days of incubation at 37 °C and 5% C02, the medium was changed and the EZH2 inhibitor was added again at the same concentrations. Every 7 days, cells were pooled, harvested, counted and analyzed for the G D2 expression described as above or lysates for western blot analysis were generated as detailed below.
  • EZH2 inhibitor or DMSO treated tumor cells were homogenized in 30-100 pi ice-cold RIPA-buffer (0.1 % DTT, Sigma-Aldrich) with fresh protease inhibitor cocktail (Roche, Germany), shortly fractured in liquid nitrogen, thawed on ice, and then clarified by spinning for 15 min at 4°C and 20,000 g. After measuring the protein concentration with Bradford reagent, 50 pg of the sample was separated by electrophoresis on an SDS/15% polyacrylamide gel and then electro blotted onto a nitrocellulose membrane (Bio-Rad).
  • Blocking was done in TBST buffer containing 5% BSA for 1 h, followed by incubation with anti-H3K27me3 antibody (Abeam, Germany) diluted 1 : 1 ,000 in TBST containing 5% BSA for 12 h at 4 °C.
  • the membrane was incubated with HRP-linked anti-mouse IgG whole Ab (GE Healthcare, Germany) at 1 :2,000 in TBST 5% BSA for 1 h at RT, followed by treatment with enhanced chemo luminescence reagent (ECL, Plus Western Blotting Detection System, GE Healthcare) and either exposed to Hyperfilm ECL film (GE Healthcare) for 1 min or directly analyzed by the Imager (ECL Chemostar, INTAS Science Imaging, Germany). Equal protein loading was determined by Ponceau staining. Equal protein loading was also determined by shortly washing the membrane in TBST buffer, followed by incubation with 7 ml RestorTM Plus stripping buffer (Thermo Scientific) for 15 min at RT.
  • the membrane was then washed twice in 20 ml TBST buffer for 10 min at RT, followed by blocking with TBST buffer with 5% nonfat dry milk for 1 h and detection with a p-actin-specific antibody (Cell Signaling, Germany) diluted 1 :5,000 in TBST with 5% BSA for 12 h at 4°C. After washing, the membrane was incubated with HRP-linked anti-rabbit antibody (GE Healthcare) 1 :2,000 in TBST 5% milk for 1 h, followed by detection as described above.
  • HRP-linked anti-rabbit antibody GE Healthcare
  • the relative luminescence units (RLUs) measured after 96 h of incubation were divided by the means of the RLUs on day 0 for each cell line. The results represent the rises of ATP levels indicating proliferation.
  • RLUs relative luminescence units
  • tumor cells were sorted into G D2 low (30%) and G D2 hi (30%) subpopulations as described above and seeded at 1x10 6 cells in 25 mm 2 culture flasks. On day six, cells were harvested, resuspended in RPMI media and counted using trypan blue staining and a microscope. Fold expansion was calculated by dividing the absolute cell numbers on day six by the seeded cell numbers.
  • EwS cells were plated in triplicate in 35 mm tissue culture dishes (Thermo Scientific, USA) in methylcellulose enriched media (1.9% methylcellulose, 15% fetal bovine serum, 0.23% bovine serum albumin, 1 % Penicillin/Streptomycin (Pen/Strep) and 82% Iscove's Modified Dulbecco's Media) in a total volume of 4 ml.
  • Cells were plated at 450 cells/ml in 1 ml volumes and incubated for 7 to 8 days at 37°C and 5% C02 to allow for anchorage-independent clonal growth. Numbers of colonies from each culture dish were calculated using an inverted microscope and a scoring grid. The mean colony numbers (colony-forming units, CFU) of the triplicate dishes were used for the graphic analyses.
  • the chemosensitivity of EwS cells was determined by quantification of the numbers of viable cells using the CellTiter Glo® Luminescent Cell Viability Assay (Promega, Mannheim, Germany). Tumor cells were seeded in opaque-walled 96-well plates at 5,000 cells per well in 50 pi of growth medium and incubated with doxorubicin at dilutions of 0.02, 0.1 , 0.2, 0.5 and 1 pmol/l at 37°C and 5% C02. After 72 h of incubation, 100 pi of CellTiter Glo® reagent were added to each well. The plates were placed on an orbital shaker for 2 min, and then incubated for 10 min at RT in the dark. The amount of metabolized ATP was quantified by determining the relative light units with GloMax Discover (Promega, Mannheim, Germany). Percent viability was determined by dividing the RLU of doxorubicin-treated cells with untreated cells x 100.
  • the biological scaffold SISmuc was prepared from porcine gut as previously described (Gottlich et al, 2016) followed by removal of mesentery and vascular tree from completely decellularized explants.
  • the matrix is registered under the trade mark BioVaSc-TERM® and after seeding with tumor cells under OncVaSc-TERM®. All explantations were in compliance with the German Animal Protection Laws ( ⁇ 4 Abs. 3 TierSchG) and the institute's animal protection officer regularly informed the responsible authorities.
  • the animals received proper attention and care in compliance with the Guide for Care and Use of Laboratory Animals published by the National Institute of Health (NIH publication no. 85e23, revised 1996) and was approved by the institutional animal protection board.
  • SISmuc For the treatment with EZH2 inhibitor, 1x10 5 EwS cells were seeded on the luminal side of the SISmuc.
  • static culture was performed for 3 days by administering 2.5 ml of cell specific media (RPMI-1640 + 10% FCS + Na-Pyruvate + Pen/Strep) to the cell crowns.
  • cell specific media RPMI-1640 + 10% FCS + Na-Pyruvate + Pen/Strep
  • the reseeded SISmuc scaffolds were placed into the chambers of customized flow bioreactors and attached to a tubing system containing 45 ml of cell specific media connected to a peristaltic pump which provides a slow media flow of 3-4 ml/min.
  • 3D tumors on the scaffolds were treated with the EZH2 inhibitor or control for 14 days, then extracted from the bioreactor and cut in half to fix one part in 4% formaldehyde for further paraffin embedding and to extract the cells from the other half for flow cytometry analysis using PBS/EDTA and Trypsin. G D2 staining was performed as described above.
  • H&E Hematoxylin and eosin
  • NSG mice NOD Scid gamma mice were used from own breeding in the central animal experimental facility Muenster (ZTE), originally purchased from Charles River (Germany) and were housed in pathogen-free rooms in type-2L (long) individually ventilated cages (Charles River) with a maximum of six animals per cage. They were allowed access to sterile food and water ad libitum. The RT is held constantly at 21 °C. Eight to 12-week old NSG mice of both genders were used for the experiments.
  • VH-64 or MS-EwS-16 cells were sorted into G D2 low (30%) and G D2 hi (30%) subpopulations as described above and were injected at 1x10 4 to 25x10 4 into the right and left flanks of NSG mice. Tumor growth was monitored at regular intervals and measured using a caliper. Mice were anesthetized with isoflurane and sacrificed when the tumor volumes reached the experimental endpoint. If no tumor was detectable, mice were sacrificed after 12 weeks. Additionally, VH-64 cells sorted into the 1 % lowest and highest G D2 - expressing cells were injected at 1x10 3 cells each into the right and left flanks of NSG mice. Tumor volume measurement was done as above.
  • VH-64 cells transduced with luciferase were sorted into the 30% lowest and highest G D2 -expressing cells and injected at 1x10 4 to 25x10 4 cells into the tail veins of NSG mice. EwS engraftment and growth were monitored weekly starting 2 weeks after transplantation by BLI using IVIS Spectrum Imaging System (PerkinElmer). Mice were injected intraperitoneally (i.p.) with D- luciferin (150 mg/kg; Synchem OHG, s039).
  • mice were anesthetized with isoflurane (2% isoflurane, 0.5 L/min oxygen), and images were acquired after 6 min in a dorsal and ventral position (f/stp: 1 , bining: 4, exposure time: automatic). Mice were sacrificed after tumor volumes reached the experimental endpoints. If no tumor was detectable, mice were sacrificed after 12 weeks.
  • isoflurane 2% isoflurane, 0.5 L/min oxygen
  • images were acquired after 6 min in a dorsal and ventral position (f/stp: 1 , bining: 4, exposure time: automatic). Mice were sacrificed after tumor volumes reached the experimental endpoints. If no tumor was detectable, mice were sacrificed after 12 weeks.
  • Primers for EZH2 (QT00054614), ST8SIA1 (G D3 synthase, QT00054159) and B4GALNT1 (GD 2 synthase, QT02564009) were purchased from Qiagen.
  • Primers for the reference gene HPRT1 forward primer 5'-TGAGGATTTGGAAAGGGTGT (SEQ ID NO: 12), reverse primer 5’-GAGCACACAGAGGGCTACAA (SEQ ID NO: 13)
  • HPRT1 forward primer 5'-TGAGGATTTGGAAAGGGTGT (SEQ ID NO: 12), reverse primer 5’-GAGCACACAGAGGGCTACAA (SEQ ID NO: 13)
  • Amplification was performed in triplicate reactions in two different runs at 95° C for 15 min, followed by 94° C for 15 sec and 40 cycles of 55° C (30 sec) and 12 C (30 sec) on a CFX96 Thermal Cycler (BioRad).
  • Cq threshold cycle
  • Cq values were determined using CFX Manager (BioRad). For the analysis the triplicates of each runs were taken together. Relative gene expression levels were calculated by 2A-AACq-method compared to HPRT1 and untreated or G D2 neg cells used as reference.
  • the sgRNA used for ST8SIA1 (NCBI Gene ID: 6489; G D3 synthase) knock-out was designed using the website http://crispr.mit.edu/.
  • the sgRNA (CACCGCCATTGAAGAAATGCGCGG (SEQ ID NO: 14)) was cloned into the BsmBI sites of the lentiviral vector lentiCRISPR_v2 (Addgene 52961) (Sanjana et al, 2014). Production of lentiviral supernatant was performed as described in Brabetz et al, 2017.
  • HEK293T cells were co-transfected with lentiCRISPR_v2 and the helper plasmids pMD2.G (Addgene plasmid #12259) and psPAX2 (Addgene plasmid #12260) containing gagpol and VSV-G sequences. Lentiviral supernatant was harvested after 48 h and used to transduce the tumor cells in 10 cm plates using 6 pg/ml Sequabrene (Sigma) over night. After puromycin selection for one week, G D3 S gene knockout was analyzed by staining for G D2 surface expression as described above.
  • the T cells were harvested and transferred to 24-well non-tissue culture-treated plates coated with Retronectin and co incubated with viral supernatant for 48 h, as described (Altvater et al, 2009).
  • 5x10 6 transduced or non-transduced T cells were then transferred to gas-permeable culture devices with 50 ml capacity (Wilson Wolf Manufacturing, 80040S) in 35 ml of culture medium for 14-16 days.
  • the G D2 -specific CAR GD2-BBZ as shown in SEQ ID NO: 1 was previously described (Kailayangiri et al, 2012). Briefly, it contains the single-chain antibody domain (scFv) of the monoclonal antibody (mAb) 14.G2a (Rossig et al, 2001), the hinge domain of human lgG1 followed by the transmembrane domain of CD28 and the signaling domains derived from 4-1 BB and CD3Z.
  • the CAR gene was codon-optimized and then sub cloned into the Agel and Xhol sites of the retroviral vector SFG (Altvater et al, 2009). Generation of stable retroviral producer cell lines and production of recombinant retrovirus for transduction of T cells was performed as described (Altvater et al, 2009).
  • T cells (3x10 5 /well) on days 13-14 after transduction were incubated with an equal number of target cells and with the EZH2 inhibitor for 14 days in a total volume of 200 pi in Eppendorf tubes in the presence of Monensin (eBioscience, Germany) (1 pl/ml) and CD107a- PE antibody (Biolegend, Germany) (100 ng/ml) for 3 h at 37°C and 5% C02.
  • the cells were washed, then incubated with CF488-conjugated G D2 anti-idiotype antibody ganglidiomab and a CD3-specific antibody for 15 min, fixed in 1 % PFA and analyzed by flow cytometry.
  • T cells incubated with medium alone or with DMSO-treated EwS cells served as controls.
  • CAR T cells The lytic activity of CAR T cells was tested using a calcein-AM release assay (Neri et al, 2001). EwS target cells treated with 4 pM GSK126 for 14 days were washed twice with PBS and then resuspended in PBS at a final concentration of 2x10 6 cells/ml and incubated with 10 pM calcein-AM (Thermo Fisher, Germany) for 30 min at 37°C with occasional shaking. After two washes in medium (RPMI, 10% FCS) cells were adjusted to 10 5 cells/ml. The test was performed in flat bottom 96-well microtiter plates (Thermo Scientific, Germany).
  • CAR-T cells at effector-to-target (E:T) cell ratios from 40: 1 to 10: 1 were seeded in triplicates together with 1x10 4 calcein AM-labeled target cells for spontaneous (only target cells in complete medium) and maximum release (only target cells in medium plus 9% Triton X-100). After 4 h at 37°C in 5% C02, the plates were centrifuged for 5 min and then 75 pi supernatant was harvested and transferred into new black-walled 96-well microtiter plates (Greiner Bio-One, Germany). Samples were measured using a GloMax® Discover multi-mode microplate reader (Promega, Germany) (Excitation 475 nm/Emission 500-550 nm).
  • Example 1 Pharmacological inhibition of EZH2 selectively upregulates surface GD2 expression in EwS cells
  • RFI relative fluorescence intensity
  • EwS cells were seeded onto a biological tissue matrix consisting of decellularized small-intestine submucosa and mucosa (SISmuc) and cultured in a dynamic bioreactor system in the presence or absence of 4 mM (SK-ES-1) or 12 mM (MS-EwS- 4) GSK126 for 14 days.
  • Immunohistochemistry analysis confirmed formation of multilayered tumor tissue on the matrix ( Figure 2D).
  • GSK126 effectively upregulated cell surface expression of G D2 on EwS cells also in the 3D model ( Figure 2D).
  • Figure 9 additionally provides evidence, that an EZH2 inhibitor such as tazemetostat can increase the surface expression of G D2 in osteosarcoma and neuroblastoma.
  • Figure 10 also shows that GSK126 enhances the surface expression of G D2 in the breast cancer cell line MDA-MB-231.
  • EZH2 inhibition selectively and reliably upregulates the ganglioside G D2 on the cell surface of EwS cells, also in a complex 3D tumor model and using chemically different pharmacological inhibitors.
  • Example 2 EZH2 modulates G D 2 expression in EwS cells by regulating expression of genes involved in G D 2 biosynthesis.
  • G D2 expression of G D2 during development is regulated through stage- and tissue- specific expression of glycosyltransferases, G D3 synthase (GD3S) and G D2 synthase (GD2S), which synthesize G D3 from its precursor G M 3 and convert G D3 to G D2 , respectively (Ngamukote et al, 2007).
  • G D3 synthase GD3S
  • G D2 synthase GD2 synthase
  • the inventors quantified transcripts of EZH2 and of GD3S and GD2S in 9 G D2 neg and 10 G D2 pos EwS cell lines by RT-PCR. EZH2 expression was not different in G D2 neg versus G D2 pos cell lines ( Figure 3A).
  • Example 3 G D2 pos and G D 2neg EwS cell lines and subpopulations of EwS cells have comparable in vitro proliferation rates, clonogenicity, tumorigenicity and chemosensitivity
  • G D2 Safe therapeutic upregulation of G D2 requires knowledge of the functional significance of G D2 expression in EwS.
  • G D2 was found to define a malignant population with a higher capacity to self-renew and reinitiate tumor growth than G D2 -negative cells (Battula et al, 2012; Liang et al, 2017). Inducing G D2 expression by epigenetic regulation could thus promote a cell population with high aggressiveness and metastatic properties.
  • the inventors compared the in vitro expansion and colony-forming capacities of EwS cell lines expressing high or low densities of G D2 identified by flow cytometry (Figure 1A).
  • G D2 hi expression within individual EwS cell lines does not confer to enhanced tumor-initiating capacities.
  • Example 4 Loss of G D 2 by genetic disruption of G D 3 synthase in EwS cells does not affect in vitro growth, clonogenicity and chemosensitivity
  • Example 5 Upregulation of surface G D 2 in EwS cells by pharmacological inhibition of EZH2 enables effective targeting by G D2 -specific CAR T cells.
  • the capacity of the tumor cells to induce antigen-specific, CAR-mediated T cell activation was assessed by quantification of the intracellular cytokines IFN-y and TNF-a and of CD107a, a marker of degranulation and prerequisite for perforin-mediated toxicity.
  • GSK126 pretreatment significantly enhanced cytolysis of initially G D2 neg EwS cells by ⁇ 02-BBz transduced T cells ( Figure 6E).
  • EZH2 inhibition sensitizes G D2 neg EwS cells to antigen-specific functional interactions with G D2 -redirected CAR T cells and to CAR T cell mediated in vitro cytolysis.
  • Example 6 Pretreatment of G D 2-negative Ewing sarcoma cell lines with the EZH2 inhibitor renders the tumor cells susceptible to G D2 -specific antibody dependent cytolysis
  • PBMCs were pre-activated with 500 U/ml IL-2 for 48 h and then analysed for 4 h in the presence and absence of the G D2 -specific antibody Dinutuximab (100pg/ml) as described herein, i.e. a light chain having an amino acid sequence as depicted in SEQ ID NO: 22 and a heavy chain having an amino acid sequence as depicted in SEQ ID NO: 21 , at effector-to-target (E:T) ratios of 40: 1 , 20: 1 and 10: 1.
  • the tumor cells RD-ES were treated with 10mM DMSO (control) or GSK126 for 7 days prior the co-incubation.
  • % cytolysis was quantified by luminometry. As apparent from Fig. 1 1 , Ewing sarcoma cells pretreated with the EZH2 inhibitor but not untreated cells from the same cell line are sensitive to Dinutuximab-dependent cytolysis.
  • Example 7 In vitro EZH2 inhibition induces G D2 upregulation also in lung cancer cells
  • Tumor cells from the lung cancer cell line A549 were cultured in the presence of 1 mM tazemetostat dissolved in DSMO (dark grey) or DMSO (control, light grey) for 14 days, and then G D2 surface expression was quantified by flow cytometry. As apparent from Fig. 12, pharmaceutical EZH2 inhibition induces a 4.7-fold induction of G D2 surface expression in a lung cancer cell line.
  • CD22-targeted CAR T cells induce remission in B- ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med 24: 20-28.
  • EWS-FLI 1 utilizes divergent chromatin remodeling mechanisms to directly activate or repress enhancer elements in Ewing sarcoma. Cancer Cell 26: 668-681.
  • Neural ganglioside GD2 identifies a subpopulation of mesenchymal stem cells in umbilical cord. Cell Physiol Biochem 23: 415-424.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Wood Science & Technology (AREA)
  • Microbiology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • General Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne un activateur de l'inhibiteur de séquence activatrice de l'homologue zeste 2 (EZH2) destiné à être utilisé dans un procédé de traitement d'une tumeur solide chez un sujet, le procédé comprenant en outre l'administration d'une cellule CAR-T spécifique du GD2 ou d'une cellule CAR-NK ou d'un anticorps ou d'un dérivé d'anticorps spécifique du GD2 au sujet. En outre, la présente invention concerne une cellule T (CAR) à récepteur antigénique chimérique spécifique du GD2 ou une cellule CAR-NK destinée à être utilisée dans un procédé de traitement d'une tumeur solide chez un sujet, (i) le procédé comprenant en outre l'administration d'un inhibiteur d'EZH2 au sujet ; et/ou (ii) le sujet a été et/ou est traité avec un inhibiteur d'EZH2. De plus, la présente invention concerne des procédés pour augmenter l'expression de surface du GD2 dans des cellules tumorales solides et des trousses et des trousses de composant comprenant un inhibiteur d'EZH 2 et une cellule CAR-T spécifique du GD2 ou une cellule CAR-NK spécifique du GD2.
PCT/EP2020/053874 2019-02-14 2020-02-14 Régulation positive du gd2 par inhibition d'ezh2 dans le traitement du cancer Ceased WO2020165402A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
LULU101129 2019-02-14
LU101129 2019-02-14

Publications (1)

Publication Number Publication Date
WO2020165402A1 true WO2020165402A1 (fr) 2020-08-20

Family

ID=66049628

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2020/053874 Ceased WO2020165402A1 (fr) 2019-02-14 2020-02-14 Régulation positive du gd2 par inhibition d'ezh2 dans le traitement du cancer

Country Status (1)

Country Link
WO (1) WO2020165402A1 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112626236A (zh) * 2021-01-14 2021-04-09 华南农业大学 与奶牛乳腺上皮细胞凋亡相关的circRNA标志物及其应用
CN114940969A (zh) * 2022-06-15 2022-08-26 朱文敏 一种用于提高间充质干细胞抗炎和免疫抑制功能的组合物
CN115252774A (zh) * 2022-07-15 2022-11-01 天津市肿瘤医院(天津医科大学肿瘤医院) DZNep联合IL-6抗体在制备抑制肝细胞癌肿瘤药物中的应用
WO2022246942A1 (fr) * 2021-05-25 2022-12-01 昭泰英基生物医药(香港)有限公司 Inducteur pour la reprogrammation d'un lymphocyte t en cellule de type nk et application de l'inducteur
WO2023205646A3 (fr) * 2022-04-18 2024-01-04 Wisconsin Alumni Research Foundation Immunothérapies combinatoires faisant intervenir des cellules car-m, car-nk, car-eos et car-n
IT202300011250A1 (it) * 2023-06-01 2024-12-01 Ospedale Pediatrico Bambino Gesù Cellule effettrici di recettore antigenico chimerico specifico per gd2 per l'uso nel trattamento di tumori solidi, eventualmente in combinazione con inibitori del potenziatore di zeste omologo 2 (ezh2)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013040371A2 (fr) 2011-09-16 2013-03-21 Baylor College Of Medicine Ciblage du microenvironnement tumoral au moyen de cellules nkt modifiées
WO2013189516A1 (fr) 2012-06-18 2013-12-27 Apeiron Biologics Ag Méthode de traitement d'un cancer positif pour gd2
WO2014077784A1 (fr) * 2012-11-19 2014-05-22 Agency For Science, Technology And Research Méthode de traitement du cancer
WO2016134284A1 (fr) 2015-02-19 2016-08-25 University Of Florida Research Foundation, Inc. Récepteurs antigéniques chimériques et leurs utilisations
WO2016201328A1 (fr) * 2015-06-10 2016-12-15 Epizyme, Inc. Inhibiteurs d'ezh2 pour traiter le lymphome
WO2017049166A1 (fr) 2015-09-17 2017-03-23 Novartis Ag Thérapie à base de cellules car-t présentant une efficacité accrue
WO2019014456A1 (fr) * 2017-07-12 2019-01-17 The Board Of Trustees Of The Leland Stanford Junior University Compositions et méthodes pour le traitement de cancers présentant une mutation h3k27m

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013040371A2 (fr) 2011-09-16 2013-03-21 Baylor College Of Medicine Ciblage du microenvironnement tumoral au moyen de cellules nkt modifiées
WO2013189516A1 (fr) 2012-06-18 2013-12-27 Apeiron Biologics Ag Méthode de traitement d'un cancer positif pour gd2
WO2014077784A1 (fr) * 2012-11-19 2014-05-22 Agency For Science, Technology And Research Méthode de traitement du cancer
WO2016134284A1 (fr) 2015-02-19 2016-08-25 University Of Florida Research Foundation, Inc. Récepteurs antigéniques chimériques et leurs utilisations
WO2016201328A1 (fr) * 2015-06-10 2016-12-15 Epizyme, Inc. Inhibiteurs d'ezh2 pour traiter le lymphome
WO2017049166A1 (fr) 2015-09-17 2017-03-23 Novartis Ag Thérapie à base de cellules car-t présentant une efficacité accrue
WO2019014456A1 (fr) * 2017-07-12 2019-01-17 The Board Of Trustees Of The Leland Stanford Junior University Compositions et méthodes pour le traitement de cancers présentant une mutation h3k27m

Non-Patent Citations (83)

* Cited by examiner, † Cited by third party
Title
"Antibody Engineering Lab Manual", SPRINGER-VERLAG, article "Protein Sequence and Structure Analysis of Antibody Variable Domains"
"NCBI", Database accession no. 6489
"Sequences of Proteins of immunological Interest", 1991, US DEPARTMENT OF HEALTH AND HUMAN SERVICES
ABIKO KMATSUMURA NHAMANISHI JHORIKAWA NMURAKAMI RYAMAGUCHI KYOSHIOKA YBABA TKONISHI IMANDAI M: "IFN-gamma from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer", BR J CANCER, vol. 112, 2015, pages 1501 - 1509, XP055411719, DOI: 10.1038/bjc.2015.101
ALTSCHUL, S. F. ET AL., NUCL. ACIDS RES., vol. 25, 1997, pages 3389 - 3402
ALTVATER BLANDMEIER SPSCHERER STEMME JSCHWEER KKAILAYANGIRI SCAMPANA DJUERGENS HPULE MROSSIG C: "2B4 (CD244) signaling by recombinant antigen-specific chimeric receptors costimulates natural killer cell activation to leukemia and neuroblastoma cells", CLIN CANCER RES, vol. 15, 2009, pages 4857 - 4866, XP055080748, DOI: 10.1158/1078-0432.CCR-08-2810
ALTVATER BPSCHERER SLANDMEIER SNIGGEMEIER VJUERGENS HVORMOOR JROSSIG C: "CD28 co-stimulation via tumour-specific chimaeric receptors induces an incomplete activation response in Epstein-Barr virus-specific effector memory T cells", CLIN EXP IMMUNOL, vol. 144, 2006, pages 447 - 457
BATTULA VLSHI YXEVANS KWWANG RYSPAETH ELJACAMO ROGUERRA RSAHIN AAMARINI FCHORTOBAGYI G ET AL.: "Ganglioside GD2 identifies breast cancer stem cells and promotes tumorigenesis", J CLIN INVEST, vol. 122, 2012, pages 2066 - 2078
BRABETZ OALIA VANGENENDT LSCHLIEMANN CBERDEL WEARTEAGA MFMIKESCH JH: "RNA-Guided CRISPR-Cas9 System-Mediated Engineering of Acute Myeloid Leukemia Mutations", MOL THER NUCLEIC ACIDS, vol. 6, 2017, pages 243 - 248
CHOTHIA ET AL., J. MOL. BIOL., vol. 227, 1992, pages 799 - 817
COMET IRIISING EMLEBLANC BHELIN K: "Maintaining cell identity: PRC2-mediated regulation of transcription and cancer", NAT REV CANCER, vol. 16, 2016, pages 803 - 810
DELATTRE OZUCMAN JPLOUGASTEL BDESMAZE CMELOT TPETER MKOVAR HJOUBERT IDE JONG PROULEAU G: "Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours", NATURE, vol. 359, 1992, pages 162 - 165
DI SHENGMENG ET AL: "Treatment of solid tumors with chimeric antigen receptor-engineered T cells: current status and future prospects", SCIENCE CHINA LIFE SCIENCES, ZHONGGUO KEXUE ZAZHISHE, CHINA, vol. 59, no. 4, 11 March 2016 (2016-03-11), pages 360 - 369, XP035939691, ISSN: 1674-7305, [retrieved on 20160311], DOI: 10.1007/S11427-016-5025-6 *
ESHHAR ZWAKS TGROSS GSCHINDLER DG: "Specific Activation and Targeting of Cytotoxic Lymphocytes Through Chimeric Single Chains Consisting of Antibody-Binding Domains and the Gamma-Subunit Or Zeta-Subunit of the Immunoglobulin and T-Cell Receptors", PROC NATL ACAD SCI USA, vol. 90, 1993, pages 720 - 724
FRY TJSHAH NNORENTAS RJSTETLER-STEVENSON MYUAN CMRAMAKRISHNA SWOLTERS PMARTIN SDELBROOK CYATES B ET AL.: "CD22-targeted CAR T cells induce remission in BALL that is naive or resistant to CD19-targeted CAR immunotherapy", NAT MED, vol. 24, 2018, pages 20 - 28
GARDNER RAFINNEY OANNESLEY CBRAKKE HSUMMERS CLEGER KBLEAKLEY MBROWN CMGEBROFF SKELLY-SPRATT KS ET AL.: "Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults", BLOOD, vol. 129, 2017, pages 3322 - 3331, XP055509718, DOI: 10.1182/blood-2017-02-769208
GIESEKE FKRUCHEN ATZARIBACHEV NBENTZIEN FDOMINICI MMULLER I: "Proinflammatory stimuli induce galectin-9 in human mesenchymal stromal cells to suppress T-cell proliferation", EUR J IMMUNOL, vol. 43, 2013, pages 2741 - 2749
GIULIA STAZI ET AL: "EZH2 inhibitors: a patent review (2014-2016)", EXPERT OPINION ON THERAPEUTIC PATENTS, 20 April 2017 (2017-04-20), pages 1 - 17, XP055378039, ISSN: 1354-3776, DOI: 10.1080/13543776.2017.1316976 *
GOSWAMI SAPOSTOLOU IZHANG JSKEPNER JANANDHAN SZHANG XXIONG LTROJER PAPARICIO ASUBUDHI SK ET AL.: "Modulation of EZH2 expression in T cells improves efficacy of anti-CTLA-4 therapy", J CLIN INVEST, vol. 128, 2018, pages 3813 - 3818
GOTTLICH CMULLER LCKUNZ MSCHMITT FWALLES HWALLES TDANDEKAR TDANDEKAR GNIETZER SL: "A Combined 3D Tissue Engineered In Vitro/In Silico Lung Tumor Model for Predicting Drug Effectiveness in Specific Mutational Backgrounds", J VIS EXP, vol. 110, pages e53885
HECZEY ALOUIS CUSAVOLDO BDAKHOVA ODURETT AGRILLEY BLIU HWU MFMEI ZGEE A ET AL.: "CAR T Cells Administered in Combination with Lymphodepletion and PD-1 Inhibition to Patients with Neuroblastoma", MOL THER, vol. 25, pages 2214 - 2224
HORIUCHI ET AL.: "Assay Development for Histone Methyltransferases", ASSAY AND DRUG DEVELOPMENT TECHNOLOGIES, vol. 11, no. 4, 2013, pages 227 - 236, XP055446822, DOI: 10.1089/adt.2012.480
HORTA ZULMARIE PEREZ ET AL: "Anti-GD2 mAbs and next-generation mAb-based agents for cancer therapy", IMMUNOTHERAPY, vol. 8, no. 9, 3 August 2016 (2016-08-03), pages 1097 - 1117, XP009516136 *
ITALIANO ASORIA JCTOULMONDE MMICHOT JMLUCCHESI CVARGA ACOINDRE JMBLAKEMORE SJCLAWSON ASUTTLE B ET AL.: "Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non- Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study", LANCET ONCOL, vol. 19, 2018, pages 649 - 659
J.S. PATTON ET AL.: "The lungs as a portal of entry for systemic drug delivery", PROC. AMER. THORACIC SOC., vol. 1, 2004, pages 338 - 344
JIN HJNAM HYBAE YKKIM SYIM IROH WYANG YSCHOI SJKIM SW: "GD2 expression is closely associated with neuronal differentiation of human umbilical cord blood-derived mesenchymal stem cells", CELL MOL LIFE SCI, vol. 67, 2010, pages 1845 - 1858, XP019837806
JONATHAN P H FISHER ET AL: "Effective combination treatment of GD2-expressing neuroblastoma and Ewing's sarcoma using anti-GD2 ch14.18/CHO antibody with V[gamma]9V[delta]2+ [gamma][delta]T cells", ONCOIMMUNOLOGY, vol. 5, no. 1, 2 January 2016 (2016-01-02), pages e1025194, XP055623752, DOI: 10.1080/2162402X.2015.1025194 *
JONES DAVID T ET AL: "Molecular characteristics and therapeutic vulnerabilities across paediatric solid tumours", NATURE REVIEWS. CANCER, NATUR PUBLISHING GROUP, LONDON, GB, vol. 19, no. 8, 12 July 2019 (2019-07-12), pages 420 - 438, XP036843830, ISSN: 1474-175X, [retrieved on 20190712], DOI: 10.1038/S41568-019-0169-X *
KAILAYANGIRI SALTVATER BMELTZER JPSCHERER SLUECKE ADIERKES CTITZE ULEUCHTE KLANDMEIER SHOTFILDER M ET AL.: "The ganglioside antigen GD2 is surface-expressed in Ewing sarcoma and allows for MHC-independent immune targeting", BR J CANCER, vol. 106, 2012, pages 1123 - 1133, XP055499992, DOI: 10.1038/bjc.2012.57
KAILAYANGIRI SALTVATER BSPURNY CJAMITZKY SSCHELHAAS SJACOBS AHWIEK CROELLECKE KHANENBERG HHARTMANN W ET AL.: "Targeting Ewing sarcoma with activated and GD2-specific chimeric antigen receptor-engineered human NK cells induces upregulation of immune-inhibitory HLA- G", ONCOIMMUNOLOGY, vol. 6, 2017, pages e1250050, XP055441726, DOI: 10.1080/2162402X.2016.1250050
KAILAYANGIRI SAREETHA ET AL: "EZH2 Inhibition in Ewing Sarcoma Upregulates GD2Expression for Targeting with Gene-Modified T Cells.", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY 08 MAY 2019, vol. 27, no. 5, 8 May 2019 (2019-05-08), pages 933 - 946, XP009516165, ISSN: 1525-0024 *
KENT WJSUGNET CWFUREY TSROSKIN KMPRINGLE THZAHLER AMHAUSSLER D: "The human genome browser at UCSC", GENOME RES, vol. 12, 2002, pages 996 - 1006, XP007901725, DOI: 10.1101/gr.229102. Article published online before print in May 2002
KRAUSE AGUO HFLATOUCHE JBTAN CCHEUNG NKSADELAIN M: "Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes", J EXP MED, vol. 188, 1998, pages 619 - 626, XP001240431, DOI: 10.1084/jem.188.4.619
KROESEN MBULL CGIELEN PRBROK ICARMANDARI IWASSINK MLOOMAN MWBOON LDEN BROK MHHOOGERBRUGGE PM ET AL.: "Anti-GD2 mAb and Vorinostat synergize in the treatment of neuroblastoma", ONCOIMMUNOLOGY, vol. 5, 2016, pages e1164919, XP055624006, DOI: 10.1080/2162402X.2016.1164919
KROOK MAHAWKINS AGPATEL RMLUCAS DRVAN NOORD RCHUGH RLAWLOR ER: "A bivalent promoter contributes to stress-induced plasticity of CXCR4 in Ewing sarcoma", ONCOTARGET, vol. 7, 2016, pages 61775 - 61788
KURMASHEVA RTSAMMONS MFAVOURS EWU JKURMASHEV DCOSMOPOULOS KKEILHACK HKLAUS CRHOUGHTON PJSMITH MA: "Initial testing (stage 1) of tazemetostat (EPZ-6438), a novel EZH2 inhibitor, by the Pediatric Preclinical Testing Program", PEDIATR BLOOD CANCER, vol. 64, 2017, pages e26218
LADENSTEIN RPOTSCHGER ULE DELEY MCWHELAN JPAULUSSEN MOBERLIN OVAN DEN BERG HDIRKSEN UHJORTH LMICHON J ET AL.: "Primary Disseminated Multifocal Ewing Sarcoma: Results of the Euro-EWING 99 Trial", J CLIN ONCO, vol. 28, 2010, pages 3284 - 3291
LADENSTEIN RPOTSCHGER UVALTEAU-COUANET DLUKSCH RCASTEL VYANIV ILAUREYS GBROCK PMICHON JMOWENS C ET AL.: "Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): a multicentre, randomised, phase 3 trial", LANCET ONCOL, vol. 19, 2018, pages 1617 - 1629
LEE DWKOCHENDERFER JNSTETLER-STEVENSON MCUI YKDELBROOK CFELDMAN SAFRY TJORENTAS RSABATINO MSHAH NN ET AL.: "T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial", LANCET, vol. 385, 2015, pages 517 - 528, XP055388598, DOI: 10.1016/S0140-6736(14)61403-3
LEUCHTE KALTVATER BHOFFSCHLAG SPOTRATZ JMELTZER JCLEMENS DLUECKE AHARDES JDIRKSEN UJUERGENS H ET AL.: "Anchorage-independent growth of Ewing sarcoma cells under serum-free conditions is not associated with stem-cell like phenotype and function", ONCOL REP, vol. 32, 2014, pages 845 - 852
LIANG YJDING YLEVERY SBLOBATON MHANDA KHAKOMORI SI: "Differential expression profiles of glycosphingolipids in human breast cancer stem cells vs. cancer non-stem cells", PROC NATL ACAD SCI USA, vol. 110, 2013, pages 4968 - 4973, XP055157581, DOI: 10.1073/pnas.1302825110
LIANG YJWANG CYWANG IACHEN YWLI LTLIN CYHO MYCHOU TLWANG YHCHIOU SP ET AL.: "Interaction of glycosphingolipids GD3 and GD2 with growth factor receptors maintains breast cancer stem cell phenotype", ONCOTARGET, vol. 8, 2017, pages 47454 - 47473
LIEBSCH LKAILAYANGIRI SBECK LALTVATER BKOCH RDIERKES CHOTFILDER MNAGELMANN NFABER CKOOIJMAN H ET AL.: "Ewing sarcoma dissemination and response to T-cell therapy in mice assessed by whole-body magnetic resonance imaging", BR J CANCER, vol. 109, 2013, pages 658 - 666, XP055499994, DOI: 10.1038/bjc.2013.356
LODE HNSCHMIDT MSEIDEL DHUEBENER NBRACKROCK DBLEEKE MREKER DBRANDT SMUELLER HPHELM C ET AL.: "Vaccination with anti-idiotype antibody ganglidiomab mediates a GD(2)-specific anti-neuroblastoma immune response", CANCER IMMUNOL IMMUNOTHER, vol. 62, 2013, pages 999 - 1010
LONG AHHIGHFILL SLCUI YSMITH JPWALKER AJRAMAKRISHNA SEI-ETRIBY RGALLI STSOKOS MGORENTAS RJ ET AL.: "Reduction of MDSCs with All-trans Retinoic Acid Improves CAR Therapy Efficacy for Sarcomas", CANCER IMMUNOL RES, vol. 4, 2016, pages 869 - 880
LOUIS CUSAVOLDO BDOTTI GPULE MYVON E, MYERS GDROSSIG CRUSSELL HVDIOUF OLIU E ET AL.: "Antitumor activity and long-term fate of chimeric antigen receptor-positive T cells in patients with neuroblastoma", BLOOD, vol. 118, 2011, pages 6050 - 6056, XP055550811, DOI: 10.1182/blood-2011-
MAASS, JOURNAL OF IMMUNOLOGICAL METHODS, vol. 324, 2007, pages 13 - 25
MARTINEZ CHOFMANN TJMARINO RDOMINICI MHORWITZ EM: "Human bone marrow mesenchymal stromal cells express the neural ganglioside GD2: a novel surface marker for the identification of MSCs", BLOOD, vol. 109, 2007, pages 4245 - 4248, XP055069966, DOI: 10.1182/blood-2006-08-039347
MAUDE SLLAETSCH TWBUECHNER JRIVES SBOYER MBITTENCOURT HBADER PVERNERIS MRSTEFANSKI HEMYERS GD ET AL.: "Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia", N ENGL J MED, vol. 378, 2018, pages 439 - 448, XP055665831, DOI: 10.1056/NEJMoa1709866
MCCABE MTOTT HMGANJI GKORENCHUK STHOMPSON CVAN ALLER GSLIU YGRAVES APDELIA PIETRA A, 3RDDIAZ E ET AL.: "EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations", NATURE, vol. 492, 2012, pages 108 - 112, XP002715572, DOI: 10.1038/nature11606
MICHIEL KROESEN ET AL: "Anti-GD2 mAb and Vorinostat synergize in the treatment of neuroblastoma", ONCOIMMUNOLOGY, vol. 5, no. 6, 2 June 2016 (2016-06-02), US, pages e1164919, XP055624006, ISSN: 2162-4011, DOI: 10.1080/2162402X.2016.1164919 *
NEELAPU SSLOCKE FLBARTLETT NLLEKAKIS LJMIKLOS DBJACOBSON CABRAUNSCHWEIG IOLUWOLE 00SIDDIQI TLIN Y ET AL.: "Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma", N ENGL J MED, vol. 377, 2017, pages 2531 - 2544, XP055547040, DOI: 10.1056/NEJMoa1707447
NERI SMARIANI EMENEGHETTI ACATTINI LFACCHINI A: "Calcein-acetyoxymethyl cytotoxicity assay: standardization of a method allowing additional analyses on recovered effector cells and supernatants", CLIN DIAGN LAB IMMUNOL, vol. 8, 2001, pages 1131 - 1135
NGAMUKOTE SYANAGISAWA MARIGA TANDO SYU RK: "Developmental changes of glycosphingolipids and expression of glycogenes in mouse brains", J NEUROCHEM, vol. 103, 2007, pages 2327 - 2341
PENG DKRYCZEK INAGARSHETH NZHAO LWEI SWANG WSUN YZHAO EVATAN LSZELIGA W ET AL.: "Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy", NATURE, vol. 527, 2015, pages 249 - 253, XP055312938, DOI: 10.1038/nature15520
PULE MASAVOLDO BMYERS GDROSSIG CRUSSELL HVDOTTI GHULS MHLIU ELGEE APMEI Z ET AL.: "Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma", NAT MED, vol. 14, 2008, pages 1264 - 1270, XP055021755, DOI: 10.1038/nm.1882
RASINI VDOMINICI MKLUBA TSIEGEL GLUSENTI GNORTHOFF HHORWITZ EMSCHAFER R: "Mesenchymal stromal/stem cells markers in the human bone marrow", CYTOTHERAPY, vol. 15, 2013, pages 292 - 306, XP055322650, DOI: 10.1016/j.jcyt.2012.11.009
RICHTER GHPLEHM SFASAN AROSSLER SUNLAND RBENNANI-BAITI IMHOTFILDER MLOWEL DVON LIMOSSBRUGGER I ET AL.: "EZH2 is a mediator of EWS/FLI1 driven tumor growth and metastasis blocking endothelial and neuro-ectodermal differentiation", PROC NATL ACAD SCI USA, vol. 106, 2009, pages 5324 - 5329
RIGGI NKNOECHEL BGILLESPIE SMRHEINBAY EBOULAY GSUVA MLROSSETTI NEBOONSENG WEOKSUZ OCOOK EB ET AL.: "EWS-FL11 utilizes divergent chromatin remodeling mechanisms to directly activate or repress enhancer elements in Ewing sarcoma", CANCER CELL, vol. 26, 2014, pages 668 - 681, XP029094872, DOI: 10.1016/j.ccell.2014.10.004
RIGGI NSUVA MLSUVA DCIRONI LPROVERO PTERCIER SJOSEPH JMSTEHLE JCBAUMER KKINDLER V ET AL.: "EWS-FLI-1 expression triggers a Ewing's sarcoma initiation program in primary human mesenchymal stem cells", CANCER RES, vol. 68, 2008, pages 2176 - 2185, XP055092255, DOI: 10.1158/0008-5472.CAN-07-1761
ROSSIG CBOLLARD CMNUCHTERN JGMERCHANT DABRENNER MK: "Targeting of GD2-positive tumor cells by human T lymphocytes engineered to express chimeric T-cell receptor genes", INT J CANCER, vol. 94, 2001, pages 228 - 236, XP055113644, DOI: 10.1002/ijc.1457
ROSSIG CKAILAYANGIRI SJAMITZKY SALTVATER B: "Carbohydrate Targets for CAR T Cells in Solid Childhood Cancers", FRONTIERS ONCOL, vol. 8, 2018, pages 513
ROTH MLINKOWSKI MTARIM JPIPERDI SSOWERS RGELLER DGILL JGORLICK R: "Ganglioside GD2 as a therapeutic target for antibody-mediated therapy in patients with osteosarcoma", CANCER, vol. 120, 2014, pages 548 - 554
S. FARAJ ET AL: "Neuroblastoma chemotherapy can be augmented by immunotargeting O-acetyl-GD2 tumor-associated ganglioside", ONCOIMMUNOLOGY, vol. 7, no. 1, 2 January 2018 (2018-01-02), US, pages e1373232, XP055623910, ISSN: 2162-4011, DOI: 10.1080/2162402X.2017.1373232 *
SANJANA NESHALEM OZHANG F: "Improved vectors and genome-wide libraries for CRISPR screening", NAT METHODS, vol. 11, 2014, pages 783 - 784, XP055611279, DOI: 10.1038/nmeth.3047
SANKAR STHEISEN ERBEARSS JMULVIHILL THOFFMAN LMSOMA VBECKERLE MCSHARMA SLESSNICK SL: "Reversible LSD1 inhibition interferes with global EWS/ETS transcriptional activity and impedes Ewing sarcoma tumor growth", CLIN CANCER RES, vol. 20, 2014, pages 4584 - 4597
SAREETHA KAILAYANGIRI ET AL: "Targeting Ewing sarcoma with activated and GD2-specific chimeric antigen receptor-engineered human NK cells induces upregulation of immune-inhibitory HLA-G", ONCOIMMUNOLOGY, vol. 6, no. 1, 2 January 2017 (2017-01-02), US, pages e1250050, XP055441726, ISSN: 2162-4011, DOI: 10.1080/2162402X.2016.1250050 *
SCHUSTER SJBISHOP MRTAM CSWALLER EKBORCHMANN PMCGUIRK JPJAGER UJAGLOWSKI SANDREADIS CWESTIN JR ET AL.: "Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B- Cell Lymphoma", N ENGL J MED, 2018
SHEFFIELD NCPIERRON GKLUGHAMMER JDATLINGER PSCHONEGGER ASCHUSTER MHADLER JSURDEZ DGUILLEMOT DLAPOUBLE E ET AL.: "DNA methylation heterogeneity defines a disease spectrum in Ewing sarcoma", NAT MED, vol. 23, 2017, pages 386 - 395
SHI BLIANG JYANG XWANG YZHAO YWU HSUN LZHANG YCHEN YLI R ET AL.: "Integration of estrogen and Wnt signaling circuits by the polycomb group protein EZH2 in breast cancer cells", MOL CELL BIOL, vol. 27, 2007, pages 5105 - 5119
SLOAN CACHAN ETDAVIDSON JMMALLADI VSSTRATTAN JSHITZ BCGABDANK INARAYANAN AKHO MLEE BT ET AL.: "ENCODE data at the ENCODE portal", NUCLEIC ACIDS RES, vol. 44, 2016, pages D726 - 732
SPURNY CKAILAYANGIRI SALTVATER BJAMITZKY SHARTMANN WWARDELMANN ERANFT ADIRKSEN UAMLER SHARDES J ET AL.: "T cell infiltration into Ewing sarcomas is associated with local expression of immune-inhibitory HLA-G", ONCOTARGET, vol. 9, 2018, pages 6536 - 6549, XP055485243, DOI: 10.18632/oncotarget.23815
STRAATHOF KF, BWALLACE,R.THOMAS,S.CHEUNG,G.COLLURA,A.GILEADI,T.BARTON,J.WRIGHT,G.INGLOTT,S.EDWARDS,D.: "A Cancer Research UK phase I trial of anti-GD2 chimeric antigen receptor (CAR) transduced T-cells (1 RG-CART) in patients with relapsed or refractory neuroblastoma", CANCER RES, vol. 78, no. 13, 2018
SUZUKI K: "The pattern of mammalian brain gangliosides. II. Evaluation of the extraction procedures, postmortem changes and the effect of formalin preservation", J NEUROCHEM, vol. 12, 1965, pages 629 - 638
SUZUKI YYANAGISAWA MARIGA TYU RK: "Histone acetylation-mediated glycosyltransferase gene regulation in mouse brain during development", J NEUROCHEM, vol. 116, 2011, pages 874 - 880
SVOBODA LKHARRIS ABAILEY NJSCHWENTNER RTOMAZOU EVON LEVETZOW CMAGNUSON BLJUNGMAN MKOVAR HLAWLOR ER: "Overexpression of HOX genes is prevalent in Ewing sarcoma and is associated with altered epigenetic regulation of developmental transcription programs", EPIGENETICS, vol. 9, 2014, pages 1613 - 1625
TAKASHINA TAICHI ET AL: "Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non-small-cell lung cancer cells", CANCER SCIENCE, vol. 107, no. 7, July 2016 (2016-07-01), pages 955 - 962, XP002794406 *
TOMAZOU EMSHEFFIELD NCSCHMIDL CSCHUSTER MSCHONEGGER ADATLINGER PKUBICEK SBOCK CKOVAR H: "Epigenome mapping reveals distinct modes of gene regulation and widespread enhancer reprogramming by the oncogenic fusion protein EWS-FLI1", CELL REPORTS, vol. 10, 2015, pages 1082 - 1095, XP055664459, DOI: 10.1016/j.celrep.2015.01.042
TOMLINSON ET AL., EMBO J., vol. 14, 1995, pages 4628 - 4638
UNLAND RCLEMENS DHEINICKE UPOTRATZ JCHOTFILDER MFULDA SWARDELMANN EFRUHWALD MCDIRKSEN U: "Suberoylanilide hydroxamic acid synergistically enhances the antitumor activity of etoposide in Ewing sarcoma cell lines", ANTICANCER DRUGS, vol. 26, 2015, pages 843 - 851
WALKER AJMAJZNER RGZHANG LWANHAINEN KLONG AHNGUYEN SMLOPOMO PVIGNY MFRY TJORENTAS RJ ET AL.: "Tumor Antigen and Receptor Densities Regulate Efficacy of a Chimeric Antigen Receptor Targeting Anaplastic Lymphoma Kinase", MOL THER, vol. 25, 2017, pages 2189 - 2201, XP055568995, DOI: 10.1016/j.ymthe.2017.06.008
XU JLIAO WGU DLIANG LLIU MDU WLIU PZHANG LLU SDONG C ET AL.: "Neural ganglioside GD2 identifies a subpopulation of mesenchymal stem cells in umbilical cord", CELL PHYSIOL BIOCHEM, vol. 23, 2009, pages 415 - 424
YU ALGILMAN ALOZKAYNAK MFLONDON WBKREISSMAN SGCHEN HXSMITH MANDERSON BVILLABLANCA JGMATTHAY KK ET AL.: "Anti-GD2 antibody with GM-CSF, interleukin-2, and isotretinoin for neuroblastoma", N ENGL J MED, vol. 363, 2010, pages 1324 - 1334, XP055045834, DOI: 10.1056/NEJMoa0911123

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112626236A (zh) * 2021-01-14 2021-04-09 华南农业大学 与奶牛乳腺上皮细胞凋亡相关的circRNA标志物及其应用
WO2022246942A1 (fr) * 2021-05-25 2022-12-01 昭泰英基生物医药(香港)有限公司 Inducteur pour la reprogrammation d'un lymphocyte t en cellule de type nk et application de l'inducteur
JP2024515334A (ja) * 2021-05-25 2024-04-08 昭泰英基生物医薬(香港)有限公司 T細胞をnk様細胞にリプログラミングする誘導剤およびその使用
WO2023205646A3 (fr) * 2022-04-18 2024-01-04 Wisconsin Alumni Research Foundation Immunothérapies combinatoires faisant intervenir des cellules car-m, car-nk, car-eos et car-n
CN114940969A (zh) * 2022-06-15 2022-08-26 朱文敏 一种用于提高间充质干细胞抗炎和免疫抑制功能的组合物
CN115252774A (zh) * 2022-07-15 2022-11-01 天津市肿瘤医院(天津医科大学肿瘤医院) DZNep联合IL-6抗体在制备抑制肝细胞癌肿瘤药物中的应用
IT202300011250A1 (it) * 2023-06-01 2024-12-01 Ospedale Pediatrico Bambino Gesù Cellule effettrici di recettore antigenico chimerico specifico per gd2 per l'uso nel trattamento di tumori solidi, eventualmente in combinazione con inibitori del potenziatore di zeste omologo 2 (ezh2)
WO2024246965A1 (fr) * 2023-06-01 2024-12-05 Ospedale Pediatrico Bambino Gesu' Cellules effectrices de récepteur antigénique chimérique spécifique à gd2 destinées à être utilisées dans le traitement de tumeurs solides, éventuellement en combinaison avec un activateur d'inhibiteurs de l'homologue 2 de zeste (ezh2)

Similar Documents

Publication Publication Date Title
Nishimoto et al. Allogeneic CD20‐targeted γδ T cells exhibit innate and adaptive antitumor activities in preclinical B‐cell lymphoma models
Kailayangiri et al. EZH2 inhibition in Ewing sarcoma upregulates GD2 expression for targeting with gene-modified T cells
US20250230217A1 (en) Methods for improving the efficacy and expansion of immune cells
WO2020165402A1 (fr) Régulation positive du gd2 par inhibition d'ezh2 dans le traitement du cancer
CN110248669B (zh) 工程化天然杀伤细胞及其用途
JP7082055B2 (ja) 抗癌治療における組み合わせ使用のためのメソテリンキメラ抗原受容体(car)およびpd-l1阻害剤に対する抗体
KR20190084053A (ko) 트립토판 대사 경로 조절인자 관련 면역 치료 방법 및 조성물
JP2024073656A (ja) 生体内での存続性及び治療活性及びその増殖のためのnkt細胞サブセット
EP3947471A1 (fr) Thérapie par lymphocytes t à récepteurs antigéniques chimériques (car) de tn-muc1
KR20230156808A (ko) Car-t 세포의 조절 방법
EP3491017A2 (fr) Procédés d'identification d'anticorps bloquant les récepteurs lilrb
TWI878270B (zh) 源自液體腫瘤之腫瘤浸潤性淋巴細胞的擴增和該腫瘤浸潤性淋巴細胞之治療用途
Riether et al. From “magic bullets” to specific cancer immunotherapy
US20180344768A1 (en) Nk cell-based therapy
US20230312671A1 (en) Grp78 targeted adoptive cell therapy
JP2020535796A (ja) strepタグ特異的キメラ受容体およびその使用
US20230019381A1 (en) Nk cell-based therapy
JP2024515189A (ja) 細胞免疫療法におけるキメラ共刺激受容体、ケモカイン受容体、及びそれらの使用
CN106062002B (zh) 抵抗骨髓增生障碍或淋巴组织增生障碍的手段和方法
JP2020533289A5 (fr)
CN119300843A (zh) 表达嵌合抗原受体的经修饰的不变自然杀伤t细胞及其用途
EP4321533A1 (fr) Utilisation d'immunothérapie cellulaire
Helmin-Basa et al. The application of the natural killer cells, macrophages and dendritic cells in treating various types of cancer
CN118265725A (zh) 包含PD-1和TGF-βRII结合域的多特异性结合部分
RU2776890C9 (ru) Клеточная терапия, основанная на улучшенных клетках-естественных киллерах

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: 20708039

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: 20708039

Country of ref document: EP

Kind code of ref document: A1