[go: up one dir, main page]

WO2025190565A1 - Récepteur de protéine 1 de mort cellulaire programmée chimérique - Google Patents

Récepteur de protéine 1 de mort cellulaire programmée chimérique

Info

Publication number
WO2025190565A1
WO2025190565A1 PCT/EP2025/052975 EP2025052975W WO2025190565A1 WO 2025190565 A1 WO2025190565 A1 WO 2025190565A1 EP 2025052975 W EP2025052975 W EP 2025052975W WO 2025190565 A1 WO2025190565 A1 WO 2025190565A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
receptor
polypeptide
chimeric
domain
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.)
Pending
Application number
PCT/EP2025/052975
Other languages
English (en)
Inventor
Mikhail STEKLOV
Friederike KNIPPING
Panagiota SOTIROPOULOU
Marleen Van Loenen
Josephine KEMNA
Alvaro HAROUN-IZQUIERDO
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.)
T Knife GmbH
Original Assignee
T Knife GmbH
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 T Knife GmbH filed Critical T Knife GmbH
Publication of WO2025190565A1 publication Critical patent/WO2025190565A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70532B7 molecules, e.g. CD80, CD86
    • 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
    • A61K40/30Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
    • A61K40/32T-cell receptors [TCR]
    • 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
    • A61K40/36Immune checkpoint inhibitors
    • 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
    • A61K40/4271Melanoma antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70578NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/03Fusion polypeptide containing a localisation/targetting motif containing a transmembrane segment

Definitions

  • the present invention relates to a chimeric transmembrane receptor comprising a polypeptide, wherein the polypeptide comprises at least one “Programmed cell death protein 1” (PD1) polypeptide region comprising a PD1 extracellular ligand binding domain, and further wherein the polypeptide comprises at least one non-PD1 polypeptide region.
  • the present invention further relates to an (isolated) nucleic acid comprising a nuclear acid sequence encoding for the chimeric transmembrane receptor, a vector comprising the nucleic acid, and to (isolated) T-cells expressing the chimeric transmembrane receptor and/or comprising the vector or nucleic acid encoding for the chimeric transmembrane receptor.
  • the invention further relates to a kit for preparing the (isolated) T-cell of the present invention, as well as to a pharmaceutical composition comprising the T-cells.
  • the invention also relates to a method for preparing a T-cell for immunotherapy, and to methods for treating a patient having a disease comprising administering the pharmaceutical composition, and/or for increasing cytotoxicity of a T- cell in adoptive cell therapy, comprising introducing the vector into the T-cell.
  • T-cells are known to be important mediators of adaptive cell-mediated immune responses.
  • Adoptive T-cell therapy (ACT) with T-cells expressing native or transgenic a£-T-cell receptors (TCRs) is a promising treatment for cancer, as TCRs cover a wide range of potential target antigens [Chandran and Klebanoff, 2019].
  • Native TCR specificities have successfully been exploited for ACT with tumor infiltrating lymphocytes (TILs) for melanoma [Dafni et al., 2019] and other tumors [Chandran and Klebanoff, 2019], or with virus-specific T-cells (VSTs) for viral-associated malignancies [Leung and Heslop, 2019].
  • TILs tumor infiltrating lymphocytes
  • VSTs virus-specific T-cells
  • Transgenic TCR-based ACT allows the genetic redirection of T-cell specificity in a highly specific and reproducible manner, and has produced promising results in melanoma and several solid tumors [Robbins et al., 2015], multiple myeloma (MM) [Rapoport et al., 2015], viral-associated malignancies [Doran et al., 2019] and acute myeloid leukemia (AML) [ Chapuis et al., 2019].
  • Another promising option in ACT is the treatment with chimeric antigen receptor (CAR)-T-cells, which has produced remarkable clinical responses with certain subsets of B cell leukemia or lymphoma [Sterner and Sterner, 2019]. Promising results have also been reported with multiple myeloma.
  • T-cell antigen recognition and subsequent T-cell activation is known to depend on the interaction between the T-cell receptor (TCR) and peptide-major histocompatibility complex (pMHC) molecules [Davis and Bjbrkman, 1988].
  • TCR T-cell receptor
  • pMHC peptide-major histocompatibility complex
  • the CD8 co-receptor plays a major role in CD8 T-cell activation
  • the CD4 co-receptor stabilizes the interaction between the TCR on CD4 T-cells and the MHC class II molecule on antigen-presenting cells (APCs).
  • APCs antigen-presenting cells
  • T-cells need to receive positive signals.
  • co-signaling molecules have a crucial role in regulating T-cell activation, subset differentiation, effector function and survival.
  • CD28 is constitutively expressed on naive CD4 and CD8 T- cells and has been shown to act as positive co-stimulatory molecule. CD28 engagement in the immunological synapse decreases the amount of antigen necessary to elicit T-cell activation [Kamphorst et al., 2015].
  • TNFRSF co-signaling receptors with co-stimulatory function include HVEM (herpesvirus entry mediator), death receptor 3 (DR3; also known as TNFRSF25), CD40 (also known as TNFRSF5) and lymphotoxin-[3 receptor (LTBR; also known as TNFRSF3) [Chen and Flies, 2013].
  • HVEM herpesvirus entry mediator
  • DR3 death receptor 3
  • CD40 also known as TNFRSF5
  • LTBR lymphotoxin-[3 receptor
  • IgSF co-signaling receptors with co-stimulatory function include - in addition to CD 28, e.g. the co-stimulatory receptor inducible T-cell co-stimulator (ICOS), CD226, CRTAM, TIM 1 , CD2, SLAM, CD 84, Ly9, and CRACC [Chen and Flies, 2013],
  • TLRs Toll like receptors
  • innate immune cells particularly antigen-presenting cells
  • TLR agonists can enhance cytokine production by activated T- cells, increase T-cell sensitivity to T-cell receptor stimulation, promote long-lived T-cell memory, and reduce the suppressive activity of regulatory T-cells.
  • ACT tumor heterogeneity
  • antigen escape T-cell trafficking
  • T-cell trafficking T-cell trafficking
  • an immunosuppressive tumor microenvironment solid tumors can effectively evade the immune response, including the promising T cell therapies, through the expression of various inhibitory molecules that can hinder the function of T cells.
  • IgSF immunoglobulin superfamily
  • TNFRSF tumor necrosis factor receptor superfamily
  • T-cell immunosuppressive tumor microenvironment - and to transmit the inhibitory effect to the T-cell.
  • IgSG for example, PD1 and T cell immunoreceptor with Ig and ITIM domains (TIGIT) and TIM-3 have been described to transmit inhibitory signals coming from solid tumors that may inhibit activation, and/or promote exhaustion of T-cells.
  • TAGIT T cell immunoreceptor with Ig and ITIM domains
  • TIM-3 have been described to transmit inhibitory signals coming from solid tumors that may inhibit activation, and/or promote exhaustion of T-cells.
  • chimeric switch receptors i.e. chimeric receptors comprising the extracellular domain of an inhibitory receptor and the cytoplasmic domain of an activating receptor
  • chimeric switch receptors i.e. chimeric receptors comprising the extracellular domain of an inhibitory receptor and the cytoplasmic domain of an activating receptor
  • PD1 switch receptors have been described, wherein the extracellular domain of PD1 has been fused to the cytoplasmic domain of ICOS or CD28, respectively [WO 2013019615A2],
  • the described PD1 switch receptors with ICOS or CD28 co-stimulatory domains, respectively could be co-expressed with a specific CAR in T-cells.
  • This object is inter alia accomplished by the chimeric transmembrane receptor, the (isolated) nucleic acid, the vectors, the (isolated) T-cells, the pharmaceutical compositions, the kits and the methods, having the features of the respective independent claims.
  • the invention provides a chimeric Programmed cell death protein 1 (PD1) receptor; comprising a polypeptide, wherein said polypeptide comprises at least one PD1 polypeptide region having at least 60% sequence identity with a polypeptide domain, a polypeptide region or a polypeptide motif of a human PD1 receptor as set forth in SEQ ID No.
  • PD1 Programmed cell death protein 1
  • said human PD1 polypeptide region comprises a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif, wherein said at least one cytoplasmic polypeptide domain, region or motif is a cytoplasmic polypeptide domain, region or motif of CD30, CD40, or Herpesvirus entry mediator (HVEM).
  • HVEM Herpesvirus entry mediator
  • the invention provides a chimeric PD1 receptor; comprising a polypeptide, wherein said polypeptide comprises at least one PD1 polypeptide region comprising a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif, wherein said at least one cytoplasmic polypeptide domain, region or motif is a cytoplasmic polypeptide domain, region or motif of CD30, CD40, or HVEM.
  • a chimeric PD1 -receptor including the costimulatory domain of CD30, CD40, or HVEM, together with a PD1 extracellular ligand binding domain, thus being engineered for use as a switch receptor, is able to turn negative signals e.g. present in a tumor microenvironment into positive signals for T-cell activation.
  • the chimeric transmembrane receptors comprising the co-stimulatory domains as herein described which are linked to the at least one PD1 polypeptide region that is still capable to bind to its natural ligand, are conveying resistance to the T-cell to the immunosuppressive tumor microenvironment.
  • the T-cells expressing the chimeric transmembrane receptor as herein provided exhibit less TCR-T exhaustion and depletion through apoptosis, and show stimulated TCR-T proliferation and functional activity.
  • the chimeric PD1 switch receptors as herein provided lack the cytoplasmic inhibitory motif/domain/region of the wildtype PD1 receptor, the binding of a natural ligand to the PD1 switch receptor, e.g. in tumor microenvironment, does no longer lead to e.g. inhibition of activation, promotion of exhaustion and/or induction of apoptosis of the T-cell expressing the chimeric PD1 switch receptor, but - instead - even co-stimulates the T-cell, thereby enhancing its cytotoxic effect.
  • the invention provides a (isolated) nucleic acid encoding for the chimeric PD1 receptor as herein provided.
  • said human PD1 polypeptide region comprises a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif of CD2, wherein the vector further comprises a nucleic acid encoding for a recombinant T-cell receptor.
  • the vector in addition to comprising the nucleic acid sequence encoding for the chimeric PD1 receptor, may further comprise the nucleic acid sequence encoding for an engineered/recombinant T-cell receptor. According to a further embodiment, the vector may additionally comprise a nucleic acid encoding for a CD8 Coreceptor, such as a wildtype CD8 Co-receptor or a chimeric CD8 Co-receptor.
  • a CD8 Coreceptor such as a wildtype CD8 Co-receptor or a chimeric CD8 Co-receptor.
  • overexpression of a chimeric human PD1 switch receptor as herein described in the T-cells as herein provided, e.g. alongside an engineered transgenic aP-T-cell receptor TCR and, in some embodiments, for example, alongside a CD8 Co-receptor, may offer several advantages and expands the therapeutic potential of this approach.
  • the optional, additional provision of the CD8 Co-receptor together with the chimeric PD1 switch receptor and e.g. together with the engineered T-cell receptor in the T-cell of the present invention may e.g. allow for the efficient incorporation of CD4 cells into TCR-T-cell therapy, such that it becomes possible to harness their unique properties to augment the antitumor immune response.
  • CD4 cells possess the ability to regulate the function of other immune cells, such as CD8 cytotoxic T-cells, dendritic cells, macrophages and B cells, by providing vital signals through the secretion of cytokines and direct cell-cell interactions.
  • This known helper function may be crucial for enhancing the persistence and potency of TCR-T-cells within the tumour microenvironment.
  • the invention provides an isolated T-cell, the T-cell comprising the vector or the nucleic acid according to the present invention.
  • the invention provides an isolated T-cell, the T-cell being introduced with, such as transfected, transduced or transformed with the vector or the nucleic acid according to the present invention.
  • the invention provides an isolated T-cell, the T-cell being treated, such as transfected, transduced or transformed to express the chimeric PD1 receptor as herein described.
  • the T-cell may be treated, such as transfected, transduced or transformed to express the chimeric PD1 receptor as herein described together with an engineered T-cell receptor.
  • the invention provides a kit comprising means to prepare the isolated and/or engineered T-cells according to the present invention. [0027] In a ninth aspect, the invention provides a pharmaceutical composition comprising the isolated T-cell according to the present invention.
  • the invention provides a method for preparing a T-cell for immunotherapy, comprising
  • the invention provides a pharmaceutical composition comprising T-cells expressing the chimeric PD1 receptor as herein described.
  • the composition may comprise T-cells expressing the chimeric PD1 receptor as herein described, and further expressing an engineered T-cell receptor.
  • the invention provides a method for treating a patient having a disease, comprising administering to the patient the pharmaceutical composition according to the present invention.
  • the invention provides a method for treating a patient having a disease, comprising introducing in vivo the nucleic acid as herein described, or the vector as herein provided into a T-cell of the patient.
  • the invention provides a method for increasing cytotoxicity of a T-cell in adoptive cell therapy, comprising - introducing a vector into the T-cell, wherein the vector comprises a nucleic acid encoding for a chimeric PD1 receptor as herein described, and, for example, further encoding an engineered T-cell receptor.
  • said human PD1 polypeptide region comprises a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif, wherein said at least one cytoplasmic polypeptide domain, region or motif is a cytoplasmic polypeptide domain, region or motif of CD30, CD40 or HVEM; or the provision of - T-cells comprising both an engineered/recombinant T-cell receptor and a chimeric PD1 receptor comprising a polypeptide, (wherein said polypeptide comprises at least one PD1 polypeptide region having at least 60% sequence identity with a polypeptide domain, a polypeptide region or a polypeptide motif of a human PD1 receptor as set forth in SEQ ID No.
  • said human PD1 polypeptide region comprises a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif of CD2; respectively.
  • the switch receptors and T-cells herein provided are capable of turning negative signals (with respect to e.g. the T-cell’s activation status and/or cytotoxic capacity) into positive signals.
  • Fig. 1 shows a graphical representation of results from a flow cytometric analysis of T-cells transduced with chimeric PD1 receptor polypeptides.
  • Fig. 2 shows a graphical representation of results from an in-vitro T-cell killing assay of T-cells transduced with an engineered T-cell receptor and a chimeric PD1 receptor polypeptide in PD-L1 -overexpressing Corl_23-A2-NLR cells. For better clarity, Fig 2 is depicted on two pages.
  • Fig. 5 shows a graphical representation of results from a an in-vitro T-cell killing assay of T-cells transduced with an engineered T-cell receptor and the chimeric PD1 receptor polypeptides shown in Fig. 4 in PD-L1 -expressing NCI-H1703 cells.
  • the invention is directed to a chimeric Programmed cell death protein 1 (PD1) receptor; comprising a polypeptide, wherein said polypeptide comprises at least one PD1 polypeptide region having at least 60% sequence identity with a polypeptide domain, a polypeptide region or a polypeptide motif of a PD1 receptor as set forth in SEQ ID No.
  • PD1 Programmed cell death protein 1
  • said human PD1 polypeptide region comprises a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif, wherein said at least one cytoplasmic polypeptide domain, region or motif is a cytoplasmic polypeptide domain, region or motif of CD30, CD40 or Herpesvirus entry mediator (HVEM).
  • HVEM Herpesvirus entry mediator
  • the invention is directed to a chimeric Programmed cell death protein 1 (PD1) receptor; comprising a polypeptide, wherein said polypeptide comprises at least one PD1 polypeptide region comprising a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 costimulatory cytoplasmic polypeptide domain, region or motif, wherein said at least one cytoplasmic polypeptide domain, region or motif is a cytoplasmic polypeptide domain, region or motif of CD30, CD40 or Herpesvirus entry mediator (HVEM).
  • PD1 Programmed cell death protein 1
  • the PD1 polypeptide region may be a human PD1 polypeptide region from a human wildtype PD1 receptor.
  • human wildtype PD1 receptor relates to a protein having an amino acid sequence according to UniProtKB database entry No. Q15116 • PDCD1_HUMAN, as set forth e.g. in SEQ ID No. 1.
  • PD1 polypeptide region refers to a polypeptide containing at least a functional portion (e.g., an extracellular ligand binding domain) of a wild-type PD1 protein or a variant thereof, such as a variant that has at least 60% sequence identity (e.g., at least 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or more) to the amino acid sequence of SEQ ID NO: 1 and that retains the ability to bind an endogenous PD1 ligand.
  • a functional portion e.g., an extracellular ligand binding domain
  • PD1 immunoglobulin variable domain refers to a portion of wild-type PD1 comprising the corresponding protein domain (i.e., the immunoglobulin variable domain, ligand binding domain, or co-stimulatory cytoplasmic polypeptide domain, respectively, of wild-type PD1) or a sequence variant thereof, such as a sequence variant recited herein.
  • non-PD1 polypeptide domains, regions, or motifs refers to a polypeptide domain, region, and motif, respectively, that is neither obtained from wild-type PD1 nor is a functional variant thereof.
  • non-PD1 polypeptide domains, regions, or motifs are those that are obtained from, e.g., CD30, CD40, HVEM, or CD2, as well as sequence variants thereof, such as sequence variants recited herein.
  • Other Examples of non-PD1 polypeptide domains, regions, or motifs are e.g. those that may be obtained from other costimulatory molecules, as well as sequence variants thereof.
  • the extracellular ligand binding domain may comprise a PD1 immunoglobulin variable (IgV) domain, wherein the PD1 IgV domain may have at least 85% sequence identity to the amino acid sequence of SEQ ID No. 2 (relating to the IgV domain comprising the amino acid sequence 35 - 145 of SEQ ID No 1 ).
  • IgV immunoglobulin variable
  • sequence identity means the percentage of pair-wise identical residues, following homology alignment of a sequence of a polypeptide and or nucleic acid of the present invention with a sequence in question, with respect to the number of residues in the longer of these two sequences.
  • the percentage of sequence homology or 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 chimeric PD1 receptor comprising both a functional extracellular PD1 receptor ligand binding domain and at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif of CD30, CD40 or HVEM, is able to turn negative signals (e.g. present in a tumor microenvironment) into positive signals for T-cell activation.
  • negative signals e.g. present in a tumor microenvironment
  • ITMS motif amino acids 247 - 251 of SEQ ID No.
  • T-cell comprising the chimeric PD1 receptor and e.g. an engineered T- cell receptor to bypass the inhibitory effects of PD1 ligands expressed by the tumor microenvironment, thus creating resistance to tumor-mediated immune suppression.
  • the chimeric PD1 receptors as herein provided may act as a molecular switch, redirecting the signaling pathways triggered by PD1 engagement with a PD1 ligand. Instead of inducing inhibition, the fusion of the PD1 receptor ligand binding domain to a co-stimulatory domain of CD30, CD40 or HVEM alters the intracellular signaling events, promoting T cell activation, persistence and enhanced anti-tumor responses.
  • a T-cell as herein provided comprising an engineered chimeric PD1 receptor together with e.g. an engineered T-cell receptor into T cell therapy, the expression of PD1 ligands by solid tumors is rendered ineffective in hindering T cell function.
  • This innovative approach empowers the T cells as herein provided to resist the immune evasion mechanisms deployed by solid tumors, enabling them to better recognize and eliminate tumor cells.
  • the extracellular ligand binding domain of the chimeric PD1 receptor may be functional in binding at least one PD1 ligand or any other protein/polypeptide having the ability of binding to the wildtype PD1 receptor ligand binding domain.
  • the at least one PD1 ligand may comprise at least one of the PD1 ligands PD-L1 and/or PD-L2.
  • the chimeric PD1 receptor polypeptide as herein provided may comprise, in addition to comprising the PD1 ligand binding domain, further domain regions /motif regions/ binding site regions from a wildtype human PD1 receptor in every conceivable combination to establish a functional chimeric PD1 receptor polypeptide.
  • “Functional” chimeric PD1 receptor in this context relates to a chimeric PD1 receptor that is capable of redirecting the signaling pathways triggered by PD1 receptor engagement with at least one PD1 ligand such that - instead of inducing inhibitory pathways and/or apoptotic pathways in the T-cell - binding of the at least one PD1 ligand promotes T-cell activation, persistence and enhanced anti-tumor responses of the T-cell expressing a chimeric PD1 receptor as herein provided.
  • an optional test for functionality of a chimeric PD1 receptor may be an in-vitro T-cell killing assay as described e.g.
  • the “ITSM” motif of wildtype PD1 may lack the “ITSM” motif of wildtype PD1 (amino acids 247-251 of SEQ ID No. 1), or may merely comprise a altered “ITIM” and/or “ITSM” motif, respectively, which no longer function(s) in promoting inhibitory/apoptotic pathways of the T-cell, e.g. due to mutations which abolish any inhibitory promoting functionality of the “ITIM” and/or “ITSM” motif(s), respectively.
  • the at least one PD1 polypeptide region has at least 70%, or at least 75%, or at least 80%, or at least 85%, or at least 90%, or at least 95%, or at least 97%, or 100% sequence identity with the functional polypeptide domain or a functional polypeptide motif of a wildtype human PD1 receptor (e.g. Seq ID No. 1).
  • the at least one PD1 polypeptide region may have one or more conservative amino acid substitutions relative to the amino acid sequence of the wildtype PD1 receptor.
  • the (human) PD1 polypeptide region comprising a PD1 extracellular ligand binding domain may have -in general- a sufficient portion of the human wildtype PD1 extracellular ligand binding domain to be functional in binding at least one PD1 ligand.
  • said at least one PD1 polypeptide region having at least 60% sequence identity with PD1 ligand binding domain of a human wildtype PD1 receptor may comprise the complete or a considerable part of the human wildtype PD1 ligand binding domain and/or all amino acids at respective amino acid positions of the wildtype PD1 receptor that are necessary and sufficient for binding of at least one ligand to the PD1 receptor.
  • the PD1 polypeptide region may comprise a complete wildtype PD1 receptor extracellular domain.
  • the expression “wildtype PD1 receptor extracellular domain” as referred to herein may relate to a polypeptide comprising amino acid sequence 24-170 of UniProtKB database entry No. Q15116 ⁇ PDCD1_HUMAN, as set forth e.g. in SEQ ID No. 1.
  • the wildtype PD1 receptor extracellular domain as referred to herein may relate to a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 3.
  • a truncated wildtype PD1 receptor extracellular domain may be used.
  • the PD1 polypeptide region may comprise amino acids 24-164 of UniProtKB database entry No. Q15116 ⁇ PDCD1 _HUMAN, as set forth e.g. in SEQ ID No. 1. It is herewith envisaged that the PD1 receptor extracellular domain as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the extracellular domain of the wildtype PD1 receptor. In particular, all amino acid substitutions that maintain the functional activity of the wildtype PD1 extracellular domain are envisaged.
  • the PD1 polypeptide region may comprise a signal sequence of the wildtype PD1 receptor.
  • a signal sequence of the PD1 receptor as referred to herein may relate to a polypeptide comprising amino acid sequence 1 - 23 of UniProtKB database entry No. Q15116 • PDCD1_HUMAN, as set forth e.g. in SEQ ID No. 1.
  • domain region As used herein are understood to relate to e.g. a region of the chimeric PD1 receptor polypeptide which is necessary and/or sufficient for a biological function of the chimeric receptor, or to a region of the chimeric PD1 receptor which is defined e.g. by a localization with respect to a cell, or to a structurally defined unit of the chimeric PD1 receptor polypeptide.
  • the chimeric PD1 receptor polypeptide may be a single-chain polypeptide.
  • the chimeric PD1 receptor polypeptide may comprise a transmembrane domain from CD30, CD40, or HVEM.
  • the chimeric PD1 receptor as herein provided may comprise - a transmembrane region from CD30 and at least one costimulatory cytoplasmic polypeptide domain, region or motif from CD30, or a transmembrane region from CD40 and at least one costimulatory cytoplasmic polypeptide domain, region or motif from CD40, or
  • transmembrane region from HVEM and at least one costimulatory cytoplasmic polypeptide domain, region or motif from HVEM.
  • the chimeric PD1 receptor polypeptide may comprise a transmembrane polypeptide region of HVEM.
  • the transmembrane domain of wildtype HVEM as referred to herein may relate to a polypeptide comprising amino acid sequence 203-223 of UniProtKB database entry No. Q92956- TNR14_HUMAN, as set forth e.g. in SEQ ID No. 4.
  • the transmembrane domain of wildtype HVEM as referred to herein may relate to a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 5.
  • the transmembrane domain as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the transmembrane domain of the wildtype HVEM receptor.
  • all amino acid substitutions that maintain the functional activity of the wildtype transmembrane domain are envisaged.
  • the transmembrane polypeptide region of the chimeric PD1 receptor may have at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least
  • the chimeric PD1 receptor polypeptide may comprise a transmembrane polypeptide region of CD40.
  • the transmembrane domain of wildtype CD40 as referred to herein may relate to a polypeptide comprising amino acid sequence 194-215 of UniProtKB database entry No. P25942 • TNR5_HUMAN, as set forth e.g. in SEQ ID No. 31.
  • the transmembrane domain of wildtype CD40 as referred to herein may relate to a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 32.
  • the transmembrane domain as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the transmembrane domain of the wildtype CD40 receptor.
  • all amino acid substitutions that maintain the functional activity of the wildtype transmembrane domain are envisaged.
  • the transmembrane polypeptide region of the chimeric PD1 receptor may have at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least
  • the chimeric PD1 receptor polypeptide region may comprise a transmembrane polypeptide region of CD30.
  • the transmembrane domain of wildtype CD30 as referred to herein may relate to a polypeptide comprising amino acid sequence 386-406 of UniProtKB database entry P28908 ⁇ TNR8_HUMAN, as set forth e.g. in SEQ ID No. 6.
  • the transmembrane domain of wildtype CD30 as referred to herein may relate to a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 7.
  • the transmembrane domain as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the transmembrane domain of the wildtype CD30 receptor.
  • all amino acid substitutions that maintain the functional activity of the wildtype transmembrane domain are envisaged.
  • the transmembrane polypeptide region of the chimeric PD1 receptor may have at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least
  • the transmembrane domain of the chimeric PD1 receptor of the T-cell as herein provided may be from other proteins which comprise a transmembrane domain.
  • the transmembrane domain may be from a further co-stimulatory molecule known in the art.
  • any transmembrane domain which is functional and allows surface detectable expression of the chimeric PD1 receptor is herewith envisaged.
  • the PD1 polypeptide region may comprise a PD1 transmembrane polypeptide domain or region.
  • the transmembrane domain of wildtype PD1 as referred to herein may relate to a polypeptide comprising amino acid sequence 171 - 191 of UniProtKB database entry No. Q15116 • PDCD1_HUMAN, as set forth e.g. in SEQ ID No. 1.
  • the transmembrane domain of wildtype PD1 as referred to herein may relate to a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 8. It is herewith envisaged that the transmembrane domain as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the transmembrane domain of the wildtype PD1 receptor. In particular, all amino acid substitutions that maintain the functional activity of the wildtype transmembrane domain are envisaged.
  • the chimeric PD1 receptor as herein provided may further comprise at least one linker region.
  • This may be e.g. a polypeptide linker region.
  • Such linker(s) may be included e.g. between functional domains/regions/motifs of the chimeric transmembrane receptor. It may be a linker region naturally occurring e.g. in wildtype PD1 receptor, or e.g. in co-stimulatory proteins, e.g. in costimulatory proteins from which the costimulatory domain of the receptor is derived.
  • polypeptide linker regions may be included between the transmembrane domain and the ligand binding domain, and/or between the transmembrane domain and the IgV domain of the chimeric PD1 receptor, and/or between the transmembrane domain and the at least one intracellular costimulatory domain, and/or between individual co-stimulatory domains (in embodiments comprising more than one co-stimulatory domain).
  • Such linker region may comprise 1-100 amino acids, or e.g. 1-80 amino acids, or e.g. 1-50 amino acids, or e.g. 5-100 amino acids.
  • a linker region of the chimeric PD1 receptor as herein provided may comprise the amino acid sequence as set forth in SEQ ID No. 9 (GGGS)n or as set forth in Seq ID No. 10 (GGGGS) n , wherein n is between 0 and 20, or wherein n is between 0 and 10, or where n is between 0 and 5, or where n is between 3 and 5.
  • each (polypeptide) linker known in the art is herewith envisaged as being potentially included in the chimeric PD1 switch receptor of the present invention.
  • the chimeric transmembrane receptor polypeptide as herein provided may comprise at least one costimulatory cytoplasmic polypeptide domain or cytoplasmic polypeptide motif of HVEM, CD40 or CD30.
  • cytoplasmic co-stimulatory domains of HVEM, CD40 or CD30 may be fused to the at least one PD1 polypeptide region comprising a functional PD1 receptor extracellular ligand binding domain in order to generate a functional chimeric PD1 switch receptor capable of redirecting the signaling pathways triggered by PD1 receptor engagement with at least one PD1 ligand such that - instead of inducing inhibitory and/or apoptotic pathways in the cell - binding of the at least one PD1 ligand promotes T-cell activation, persistence and enhanced anti-tumor responses of a T-cell expressing the chimeric transmembrane receptors as herein provided, for example when the T-cell at the same time comprises/expresses e.g. an engineered T-cell receptor.
  • the chimeric PD1 receptor as herein provided may comprise e.g. at least one complete cytoplasmic domain of HVEM, CD40, or CD30.
  • the at least one cytoplasmic polypeptide domain or cytoplasmic polypeptide motif selected from the group consisting of HVEM, CD40, and CD30 may have an amino acid sequence having at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least 79%, or at least 80%, or at least 81%, or at least 82%, or at least 83%, or at least 84%, or at least 85%, or at least 86%, or at least 87%, or at least 88%, or at least 89% or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96% or at least 97%, or at least 98%, or at least 99%, or 100% sequence identity with the respective functional polypeptide domain or a functional polypeptide motif of a wildtype
  • the chimeric PD1 receptor as herein provided may be able to sustain or enhance cytotoxicity and/or cytokine secretion of a T-cell upon binding a PD1 ligand.
  • the chimeric PD1 receptor as herein provided may be capable of increasing resistance of T-cells to PD1 ligand expressing cancer cells.
  • the chimeric PD1 receptor as herein provided may comprise any functional combination of co-stimulatory cytoplasmic polypeptide domain(s) motif(s) and/or region(s) of the cytoplasmic polypeptide domain, region or motif selected from the group consisting of HVEM, CD40 and CD30.
  • the chimeric PD1 receptor as herein provided may comprise at least one cytoplasmic polypeptide domain, region or motif of HVEM.
  • a cytoplasmic polypeptide region of HVEM may comprise the complete cytoplasmic domain of wildtype human HVEM.
  • the cytoplasmic polypeptide region of HVEM included in the chimeric PD1 receptor may comprise at least one functional, co-stimulatory motif/domain/region of the complete wildtype human HVEM cytoplasmic domain.
  • the expression “wildtype human HVEM cytoplasmic domain” as referred to herein may relate to a polypeptide comprising amino acid sequence 224-283 of UniProtKB database entry No.
  • the cytoplasmic polypeptide domain, region or motif of wildtype HVEM as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the amino acid sequence as set forth in SEQ ID No. 11.
  • all amino acid substitutions that maintain the functional activity of the cytoplasmic polypeptide domain, region or motif of wildtype HVEM are envisaged.
  • the polypeptide of the chimeric transmembrane receptor may have an amino acid sequence with at least 85% identity to the amino acids as set forth in SEQ ID NO: 12.
  • the chimeric PD1 receptor as herein provided may comprise at least one cytoplasmic polypeptide domain, region or motif of CD40.
  • a cytoplasmic polypeptide region of CD40 may comprise the complete cytoplasmic domain of wildtype human CD40.
  • the cytoplasmic polypeptide region of CD40 included in the chimeric PD1 receptor may comprise at least one functional, co-stimulatory motif/domain/region of the complete wildtype human CD40 cytoplasmic domain.
  • the expression “wildtype human CD40 cytoplasmic domain” as referred to herein may relate to a polypeptide comprising amino acid sequence 216-277 of UniProtKB database entry No.
  • the cytoplasmic polypeptide domain, region or motif of wildtype CD40 as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the amino acid sequence as set forth in SEQ ID No. 33.
  • all amino acid substitutions that maintain the functional activity of the cytoplasmic polypeptide domain, region or motif of wildtype CD40 are envisaged.
  • the polypeptide of the chimeric transmembrane receptor may have an amino acid sequence with at least 85% identity to the amino acids as set forth in SEQ ID NO: 28.
  • the at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif further may comprise - in addition to the complete cytoplasmic domain of wildtype human CD40 - at least one further cytoplasmic motif of CD40.
  • the at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif may further comprise at least one further TRAF6 binding motif of CD40, and/or at least one further TRAF1/2/3 binding motif of CD40.
  • TRAF6 binding motifs and TRAF 1/2/3 binding motifs have been described in the art (e.g. Park HH, Front. Immunol. 9:1999, 2018).
  • suitable methods known in the art such as e.g. yeast two hybrid assays or surface plasmon resonance assay for determining binding of TRAF 6 and/or TRAF1 , TRAF2 and/or TRAF3 to a specific binding motif.
  • CD40 TRAF 6 binding motif as referred to herein may relate to any polypeptide region of CD40 which is functional in binding TRAF 6.
  • CD40 TRAF 1/2/3 binding motif as referred to herein may relate to any polypeptide region of CD40 which is functional in binding TRAF 1, TRAF 2 and/or TRAF3.
  • CD40 TRAF6 binding motif as referred to herein may relate to a specific polypeptide sequence of human wildtype CD40 which is necessary and sufficient for binding to TRAF6.
  • the “TRAF6” binding motif of wildtype CD40 as referred to herein may relate to a polypeptide comprising amino acid sequence 225 to 243 of UniProtKB database entry No. P25942 ⁇ TNR5_HUMAN, as set forth e.g. in SEQ ID No. 31.
  • the “TRAF6” binding motif of wildtype CD40 as referred to herein may relate to a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 37.
  • the “TRAF6” binding motif of wildtype CD40 as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the “TRAF6” motif of wildtype CD40.
  • the “TRAF6” binding motif of wildtype CD40 as included in the chimeric PD1 receptor as herein provided may comprise a polypeptide region of CD40 including amino acid position 237 of SEQ ID No 31 (wildtype CD40) , and further wherein, at said position, the TRAF6 binding motif may be mutated, optionally wherein said mutation may consist of an exchange of an asparagine (N) to an aspartic acid (D).
  • the “TRAF6” binding motif of wildtype CD40 as included in the chimeric PD1 receptor as herein provided may comprise a polypeptide region of CD40 including amino acid position 229 of SEQ ID No 31 (wildtype CD40) , and further wherein, at said position, the TRAF6 binding motif may be mutated, optionally wherein said mutation may consist of an exchange of an proline (P) to an alanine (A).
  • P proline
  • A alanine
  • the “TRAF6” motif of wildtype CD40 of the chimeric PD1 receptor may have at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least 79%, or at least 80%, or at least 81%, or at least 82%, or at least 83%, or at least 84%, or at least 85%, or at least 86%, or at least 87%, or at least 88%, or at least 89% or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96% or at least 97%, or at least 98%, or at least 99%, or 100% sequence identity with the amino acid sequence as set forth in SEQ ID NO: 37.
  • CD40 TRAF1/2/3 binding motif as referred to herein may relate to a specific polypeptide sequence of human wildtype CD40 which is necessary and sufficient for binding to TRAF1, TRAF2, and/or TRAF3.
  • the “TRAF1/2/3 binding motif” of wildtype CD40 as referred to herein may relate to a polypeptide comprising amino acid sequence 244 to 259 of UniProtKB database entry No. P25942 • TNR5_HUMAN, as set forth e.g. in SEQ ID No. 31.
  • the “TRAF1/2/3” motif of wildtype CD40 as referred to herein may relate to a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 38.
  • the “TRAF1/2/3” motif of wildtype CD40 as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the “TRAF1/2/3” motif of wildtype CD40.
  • all amino acid substitutions that maintain the functional activity of the “TRAF123” motif of wildtype CD40 are envisaged.
  • the “TRAF1/2/3” motif of wildtype CD40 of the chimeric PD1 receptor may have at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least
  • the at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif may further comprise - in addition to the complete cytoplasmic domain of wildtype human CD40 - at least one further TRAF6 motif of CD40, and at least one further TRAF 1/2/3 binding motif of CD40.
  • the non-PD1 costimulatory cytoplasmic polypeptide domain may comprise a complete CD40 cytoplasmic domain, a further CD40 TRAF6 binding motif, and a further CD40 TRAF1/2/3 binding motif.
  • the chimeric transmembrane receptor may comprise a polypeptide linker between the at least two non-PD1 co-stimulatory cytoplasmic polypeptide domains, regions or motifs.
  • the polypeptide linker may consist of a polypeptide having the amino acid sequence as set forth in SEQ ID No 10 or 39.
  • the polypeptide of the chimeric PD1 receptor may comprise or consist of a polypeptide having an amino acid sequence with at least 70% identity to a polypeptide having the amino acid sequence as set forth in SEQ ID NO: 35.
  • the chimeric PD1 receptor may have at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least 79%, or at least 80%, or at least 81%, or at least 82%, or at least 83%, or at least 84%, or at least 85%, or at least 86%, or at least 87%, or at least 88%, or at least 89% or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96% or at least 97%, or at least 98%, or at least 99%, or 100% sequence identity with the amino acid sequence as set forth in SEQ ID NO: 35.
  • the chimeric PD1 receptor as herein provided may comprise at least one cytoplasmic polypeptide domain, region or motif of CD30.
  • a cytoplasmic polypeptide region of CD30 may comprise the complete cytoplasmic domain of wildtype human CD30.
  • the cytoplasmic polypeptide region of CD30 included in the chimeric PD1 receptor may comprise at least one functional, co-stimulatory motif/domain/region of the complete wildtype human CD30 cytoplasmic domain.
  • the polypeptide comprises a truncated cytoplasmic domain of CD30, optionally wherein the truncated cytoplasmic domain of CD30 comprises or consists of at least one CD30 TRAF binding motif.
  • wildtype human CD30 cytoplasmic domain may relate to a polypeptide comprising amino acid sequence 407-595 of UniProtKB database entry P28908 ⁇ TNR8_HUMAN, as set forth e.g. in SEQ ID No. 6. It is herewith envisaged that the cytoplasmic polypeptide domain, region or motif of wildtype CD30 as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the amino acid sequence as set forth in SEQ ID No. 13. In particular, all amino acid substitutions that maintain the functional activity of the cytoplasmic polypeptide domain, region or motif of wildtype CD30 are envisaged.
  • the polypeptide of the chimeric PD1 receptor may have an amino acid sequence with at least 85% identity to the amino acids as set forth in SEQ ID No.: 14.
  • a chimeric PD1 receptor as herein provided may e.g. comprise a polypeptide having an amino acid sequence with at least 85% or at least 86%, or at least 87%, or at least 88%, or at least 89% or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96% or at least 97%, or at least 98%, or at least 99%, or 100% identity to the amino acids as set forth in any one of the SEQ ID No’s selected from the group consisting of SEQ ID No. 12, 14, 28, and 35.
  • a chimeric transmembrane receptor as herein provided may have one or more conservative amino acid substitutions relative to an amino acid sequence as set forth in any one of the SEQ ID No’s selected from the group consisting of SEQ ID No. 12, 14, 28 and 35.
  • the invention provides a (isolated) nucleic acid encoding for the chimeric transmembrane receptor as herein provided.
  • the invention provides a vector comprising a nucleic acid comprising a nuclear acid sequence encoding for a chimeric PD1 switch receptor as herein provided.
  • polynucleotide or “nucleic acid” as used herein comprises a sequence of polyribonucleotides and polydeoxribonucleotides, e.g. modified or unmodified RNA or DNA, each in single-stranded and/or double-stranded form linear or circular, or mixtures thereof, including hybrid molecules.
  • the nucleic acids according to this invention thus comprise DNA (such as dsDNA, ssDNA, cDNA), RNA (such as dsRNA, ssRNA, mRNA ivtRNA), combinations thereof or derivatives (such as RNA) thereof.
  • a polynucleotide may comprise a conventional phosphodiester bond or a non- conventional bond (e.g., an amide bond, such as found in peptide nucleic acids (RNA)).
  • the polynucleotides of the invention may also contain one or more modified bases, such as, for example, tritylated bases and unusual bases such as inosine. Other modifications, including chemical, enzymatic, or metabolic modifications, are also conceivable, as long as a binding molecule of the invention can be expressed from the polynucleotide.
  • the polynucleotide may be provided in isolated form as defined elsewhere herein.
  • a polynucleotide may include regulatory sequences such as transcription control elements (including promoters, enhancers, operators, repressors, and transcription termination signals), ribosome binding site, introns, or the like.
  • the present invention provides a polynucleotide comprising or consisting of a nucleic acid that is at least about 80 %, about 85 %, about 90 %, about 91 %, about 92 %, about 93 %, about 94 %, about 95 %, about 96 %, about 97 %, about 98 %, about 99 %, or 100 % identical to a reference polynucleotide sequence selected from the group consisting of sequences as depicted in SEQ ID Nos.15, 16, 34, and 36.
  • polynucleotides described above may or may not comprise additional or altered nucleotide sequences encoding e.g., altered amino acid residues.
  • the polynucleotides may further encode fusion polypeptides, fragments, variants and other derivatives of the chimeric transmembrane receptors described herein.
  • the nucleic acid sequences of the vectors of the present invention may be codon-optimized for optimal expression in the desired host T-cell, e.g. a human lymphocyte; or for expression in bacterial, yeast or insect T-cells that are particularly envisaged for the expression of a soluble TCR of the invention.
  • Codon-optimization refers to the exchange in a sequence of interest of codons that are generally rare in highly expressed genes of a given species by codons that are generally frequent in highly expressed genes of such species, such codons encoding the same amino acids as the codons that are being exchanged. Selection of optimum codons thus depends on codon usage of the host genome and the presence of several desirable and undesirable sequence motifs.
  • a “vector” as understood herein relates to a nucleic acid molecule used as a vehicle to transfer (foreign) genetic material into a host T-cell where it can for instance be replicated and/or expressed.
  • the vector may be a viral vector or a non-viral vector.
  • Viral vectors may be selected from adenoviruses, poxviruses, alphaviruses, arenaviruses, flaviviruses, rhabdoviruses, retroviruses, lentiviruses, herpesviruses, paramyxoviruses, picornaviruses, and combinations thereof.
  • Viruses used for transfection of T-cells may include naturally occurring viruses as well as artificial viruses. Viruses may be either an enveloped or non-enveloped virus. Parvoviruses (such as AAVs) are examples of non-enveloped viruses.
  • the viruses may be enveloped viruses.
  • the viruses used for transfection of T-cells may be retroviruses and in particular lentiviruses.
  • Viral envelope proteins that can promote viral infection of eukaryotic cells may comprise HIV-1 derived lentiviral vectors (LVs) pseudotyped with envelope glycoproteins (GPs) from the vesicular stomatitis virus (VSV-G), the modified feline endogenous retrovirus (RD114TR), and the modified gibbon ape leukemia virus (GALVTR).
  • LVs HIV-1 derived lentiviral vectors pseudotyped with envelope glycoproteins (GPs) from the vesicular stomatitis virus (VSV-G), the modified feline endogenous retrovirus (RD114TR), and the modified gibbon ape leukemia virus (GALVTR).
  • GPs envelope glycoproteins
  • VSV-G vesicular stomatitis virus
  • RD114TR modified feline endogenous retrovirus
  • GALVTR gibbon ape leukemia virus
  • viruses such as parvoviruses, including adeno-associated viruses (AAV), thereby
  • RD114 env chimeric envelope protein RD114pro or RDpro (which is an RD114-HIV chimera that was constructed by replacing the R peptide cleavage sequence of RD114 with the HIV-1 matrix/capsid (MA/CA) cleavage sequence, such as described in Bell et al. Experimental Biology and Medicine 2010; 235: 1269-1276; the content of which is incorporated herein by reference), baculovirus GP64 env (such as described in Wang et al. J. Virol.
  • Non-viral vectors may comprise a naked nucleic acid such as naked plasmid DNA, cationic lipids, synthetic polycationic polymers, dendrimers, synthetic peptides such as cell-penetrating peptides (CPP’s), P-1,3- glucans, or combinations thereof.
  • a naked nucleic acid such as naked plasmid DNA, cationic lipids, synthetic polycationic polymers, dendrimers, synthetic peptides such as cell-penetrating peptides (CPP’s), P-1,3- glucans, or combinations thereof.
  • vector encompasses, without limitation, plasmids, viral vectors (including retroviral vectors, lentiviral vectors, adenoviral vectors, vaccinia virus vectors, polyoma virus vectors, and adenovirus- associated vectors (AAV)), phages, phagemids, cosmids and artificial chromosomes (including BACs and YACs).
  • viral vectors including retroviral vectors, lentiviral vectors, adenoviral vectors, vaccinia virus vectors, polyoma virus vectors, and adenovirus- associated vectors (AAV)
  • phages phagemids
  • cosmids and artificial chromosomes including BACs and YACs.
  • the vector itself is generally a nucleotide sequence, commonly a DNA sequence that comprises an insert (transgene) and a larger sequence that serves as the “backbone” of the vector.
  • Engineered vectors typically comprise an origin for autonomous replication in the host-cells (if stable expression of the polynucleotide is desired), selection markers, and restriction enzyme cleavage sites (e.g. a multiple cloning site, MCS).
  • the vector may additionally comprise promoters, genetic markers, reporter genes, targeting sequences, other regulatory elements, and/or protein purification tags. As known to those skilled in the art, large numbers of suitable vectors are known to those of skill in the art and many are commercially available.
  • the vector may further comprise a nucleic acid encoding a chimeric antigen receptor (CAR).
  • CAR chimeric antigen receptor
  • the vector may further comprise a nucleic acid encoding a T-cell receptor comprising a TCRa chain and a TCRp chain.
  • the engineered T-cell receptor may be a recombinant/engineered T-cell receptor.
  • the vector may further comprise a nucleic acid encoding for a CD8 Co-receptor.
  • the CD 8 Co-receptor may be a wildtype CD 8 Co-receptor. It is contemplated that the nucleic acid may encode e.g. a CD8a and a CD8p Co-receptor.
  • An advantage of incorporation of CD8 co-receptor into the vector is the resulting option of achieving a coordinated CD4+ and CD8+ TCR-T cell response in adoptive cell therapy which broadens and deepens clinical responses.
  • the CD8 Co-receptor may be a chimeric CD8 Co-receptor with CD8 Co-receptor functionality.
  • the expression human wildtype CD8a Co-receptor relates to a protein having an amino acid sequence according to UniProtKB database entry No. P01732 • CD8A_HUMAN, as set forth e.g. in SEQ ID No. 17. It is further understood that the expression human wildtype CD8p Co-receptor relates to a protein having an amino acid sequence according to UniProtKB database entry No. P10966 • CD8B_HUMAN, as set forth e.g. in SEQ ID No. 18.
  • a chimeric human CD8 Co-receptor polypeptide as referred to herein may comprise a single chain polypeptide comprising both an CD8a IG-like domain region together with an CD8p IG-like domain region, and may be able to maintain the function of an CD8a IG-like domain region and an CD8p IG-like domain region being present on individual, separate polypeptides in a wildtype CD8ap co-receptor.
  • T-cells as herein provided comprising the chimeric PD1 receptor, the engineered T-cell receptor and, in addition, a chimeric CD8 Co- receptor therefore exhibit enhanced T-cell activation, proliferation, cytokine production, and cytotoxicity, ultimately improving the therapeutic efficacy of TCR-T -cell therapy.
  • the vector may further include one or more multicistronic element(s) and the multicistronic element(s) may be positioned, for example, between any two nucleic acid sequences encoding for the chimeric PD1 receptor, and the optional TCRa, TCRp, and CD8 Coreceptor.
  • the multicistronic element(s) may include a sequence encoding a ribosome skip element selected from among a T2A, a P2A, a E2A or a F2A or an internal ribosome entry site (IRES).
  • self-cleaving 2A peptide refers to relatively short peptides (of the order of 20 amino acids long, depending on the virus of origin) acting co- translationally, by preventing the formation of a normal peptide bond between the glycine and last proline, resulting in the ribosome skipping to the next codon, and the nascent peptide cleaving between the Gly and Pro. After cleavage, the short 2A peptide remains fused to the C-terminus of the 'upstream’ protein, while the proline is added to the N- terminus of the 'downstream’ protein.
  • Self-cleaving 2A peptide may be selected from porcine teschovirus-1 (P2A), equine rhinitis A virus (E2A), Thosea asigna virus (T2A), foot-and-mouth disease virus (F2A), or any combination thereof.
  • P2A porcine teschovirus-1
  • E2A equine rhinitis A virus
  • T2A Thosea asigna virus
  • F2A foot-and-mouth disease virus
  • an (isolated) T-cell comprising a nucleic acid encoding for the chimeric human PD1 receptor of the present invention.
  • the T-cell may further comprise a nucleic acid encoding for an engineered T-cell receptor or a CAR.
  • the expressions “engineered T-cell receptor” and “recombinant T-cell receptor”, respectively, are to be distinguished from a “CAR” T-cell receptor.
  • engineered/recombinant TCRs recognize HLA-presented peptides derived from proteins of all cellular compartments.
  • the expressions “engineered T-cell receptor” and “recombinant T-cell receptor”, respectively, are understood to embrace TCRs that are not naturally expressed by the recited T cell (e.g., TCRs that are exogenous to the T cell and that are introduced into the T cell genome by way of a genetic engineering technique described herein).
  • the T-cell may further comprise a nucleic acid encoding for a recombinant CD8 Co-receptor.
  • the one or more nucleic acid may have been stably integrated into the genome of the T-cell, e.g. by targeted knock-in utilizing e.g. CRISPR/Cas9.
  • a T-cell may express the chimeric PD1 receptor as herein described.
  • the T-cell may further express an engineered T-cell receptor or a CAR.
  • the T-cell may, in some embodiments, further express a CD8 co-receptor such as a wildtype CD8 co-receptor or a chimeric CD8 co-receptor.
  • the chimeric CD8 co-receptor may be a chimeric receptor having functionality of a wildtype CD8 Co- receptor.
  • the chimeric CD8 co-receptor may have the same MHC-complex binding functionality as a wildtype CD8 Co-receptor.
  • the T-cell may be a CD4 T-cell, and further the CD4 T-cell may additionally expresses a recombinant human CD8 co-receptor, such as e.g. a CD8a and CD8£ receptor, or a chimeric CD8 co-receptor.
  • a recombinant human CD8 co-receptor such as e.g. a CD8a and CD8£ receptor, or a chimeric CD8 co-receptor.
  • the vector and/or the nucleic acid as herein described may have been introduced into the T-cell.
  • the (isolated) T-cells may be generated using various methods, including those recognized in the literature.
  • a polynucleotide encoding an expression cassette that comprises a tumor recognition, or another type of recognition moiety, and that also encodes for a chimeric PD1 receptor as herein described and, optionally, the engineered T-cell receptor or a CAR and, optionally, a CD8 Co-receptor may be stably introduced into the T-cell by a transposon/transposase system or a viral-based gene transfer system, such as a lentiviral or a retroviral system, or another suitable method, such as transfection, electroporation, transduction, lipofection, calcium phosphate (CaPCU), Nano engineered substances, such as Ormosil, mRNA-based therapy, viral delivery methods, including adenoviruses, retroviruses, lentiviruses, adeno-associated viruses, or another suitable method. It is envisaged that T
  • the T-cells may be transfected by means known in the art including lipofection (liposome-based transfection), electroporation, calcium phosphate transfection, biolistic particle delivery (e.g., gene guns), microinjection, or combinations thereof.
  • lipofection liposome-based transfection
  • electroporation calcium phosphate transfection
  • biolistic particle delivery e.g., gene guns
  • microinjection microinjection
  • Various methods of transfecting cells are known in the art. See, e.g., Sambrook & Russell (Eds.) Molecular Cloning: A Laboratory Manual (3rd Ed.) Volumes 1-3 (2001) Cold Spring Harbor Laboratory Press; Ramamoorth & Narvekar “Non Viral Vectors in Gene Therapy- An Overview.” JCIinDiagn Res. (2015) 9(1): GE01-GE06.
  • the cell may be a a T-cell, y5 T-cell, and/or a natural killer T-cell.
  • the a T-cell may be a CD4 T-cell, or the ap T-cell may be a CD8 T-cell, or the y5 T-cell may comprise e.g. a Vy1 chain or a Vy2 chain, or may be e.g. a Vy9V52+ T-cell.
  • the T-cell may express a chimeric PD1 receptor as herein described.
  • the T-cell may further express an engineered T-cell receptor or a CAR.
  • the T-cell may be a CD4 T-cell that further expresses a CD8 Co-receptor, e.g. both CD8a and CD8p Co-receptor, or any engineered protein exhibiting CD8 Co-receptor functionality.
  • the T-cells may further express an engineered T-cell receptor.
  • Engineered T-cells of the present disclosure can be used to treat a subject in need of treatment for a condition, for example, a cancer described herein.
  • the T-cells may be ap T-cells or y3 T-cells that express the chimeric PD1 receptor polypeptide as described herein, and furthermore an engineered TCR.
  • the T- cells may further express a CD8 Co-receptor such as a wildtype or chimeric CD8 co- receptor.
  • T-cells described herein may be used to treat a cancer, including solid tumors and hematologic malignancies. For example, “hot” tumors or “cold” tumors may be treated by the T-cells herewith provided.
  • the engineered T-cell receptor as herein described may specifically bind a MAGE antigen family member, such as MAGE-A1 or Mage-A4, or wherein the engineered T-cell receptor may specifically bind an antigen selected from the group consisting of a PRAME antigen, a NY-ESO-1 antigen, a GP100 antigen, an AFP antigen, a Col6A3 antigen, an HPV-16 antigen, a WT1 antigen, an HA1 antigen, an HA2 antigen, a mutated KRAS antigen, a mutated NRAS antigen, a mutated HRAS antigen, a mutated TP53 antigen, and an EGFR antigen.
  • a PRAME antigen such as MAGE-A1 or Mage-A4
  • a GP100 antigen such as MAGE-A1 or Mage-A4
  • an antigen selected from the group consisting of a PRAME antigen, a NY-ESO-1 antigen,
  • the expression “mutated” with respect to specific tumor antigens as herein used relates to well-known mutations within the epitope region of the respective protein, polypeptide or peptide that has been correlated with expression in a human cancer.
  • the T-cells described herein may also be used to treat an infectious disease.
  • the T-cells described herein may be used to treat an infectious disease, an infectious disease may be caused by a virus.
  • the T-cells described herein may be used to treat an immune disease, such as an autoimmune disease.
  • the T-cells may be ap T-cells or y ⁇ T-cells that express a chimeric PD1 receptor as described herein, and optionally an engineered TCR, and optionally a CD8 Co-receptor such as a wildtype or chimeric CD8 co-receptor.
  • the T-cell may be derived from a healthy subject, or the T-cell may be derived from a patient suffering from a disease.
  • the T-cell may be derived from an induced pluripotent stem cell (iPSCs).
  • iPSCs induced pluripotent stem cell
  • an isolated T-cell wherein the T-cell expresses a chimeric PD1 receptor comprising a polypeptide, wherein said polypeptide comprises at least one PD1 polypeptide region having at least 60% sequence identity with a polypeptide domain, a polypeptide region or a polypeptide motif of a human PD1 receptor as set forth in SEQ ID No. 1 , wherein said human PD1 polypeptide region comprises a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif of CD2, wherein the T-cell further expresses a recombinant T- cell receptor.
  • an isolated T-cell wherein the T-cell expresses a chimeric PD1 receptor comprising a polypeptide, wherein said polypeptide comprises at least one PD1 polypeptide comprising a PD1 extracellular ligand binding domain; further wherein said polypeptide comprises at least one non-PD1 co-stimulatory cytoplasmic polypeptide domain, region or motif of CD2, wherein the T- cell further expresses a recombinant T-cell receptor.
  • T-cells comprising a chimeric PD1-receptor including the costimulatory domain of CD2, as well as a PD1 extracellular ligand binding domain, thus being engineered for use as a switch receptor, is able to turn negative signals e.g. present in a tumor microenvironment into positive signals for T-cell activation.
  • the inventors could show for the first time that, if expressed in T-cells which further express an engineered T-cell receptor, the chimeric PD1 receptor comprising the at least one co-stimulatory domain of CD2 which is linked to the PD1 polypeptide region that is still capable to bind to its natural ligand, the chimeric transmembrane receptor is conveying resistance to the T-cell to the immunosuppressive tumor microenvironment.
  • the T-cells according to the fifteenth or sixteenth aspect of the invention exhibit less TCR-T exhaustion and depletion through apoptosis, and show stimulated TCR-T proliferation and functional activity.
  • the chimeric PD1 -receptor of the T-cells according to the fifteenth or sixteenth aspect may comprise a PD1 transmembrane domain as described with respect to the first aspect of the present invention.
  • the transmembrane region of the chimeric PD1 receptor of the T-cell in accordance with the fifteenth or sixteenth aspect may be from a co-stimulatory polypeptide.
  • the chimeric PD1 receptor of the T-cell in accordance with the fifteenth or sixteenth aspect may comprise a transmembrane polypeptide region of CD2.
  • the transmembrane domain of wildtype CD2 as referred to herein may relate to a polypeptide comprising amino acid sequence 210-235 of UniProtKB database entry P06729 ⁇ CD2_HUMAN, as set forth e.g. in SEQ ID No. 19.
  • the transmembrane domain of wildtype CD2 as referred to herein may relate to a polypeptide having an amino acid sequence as set forth in SEQ ID NO: 20.
  • the transmembrane domain as included in the chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to the transmembrane domain of the wildtype CD2 receptor.
  • all amino acid substitutions that maintain the functional activity of the wildtype transmembrane domain are envisaged.
  • the transmembrane polypeptide region of the chimeric PD1 receptor may have at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least
  • the chimeric PD1 receptor of the T-cell in accordance with the fifteenth or sixteenth aspect may further comprise at least one linker region.
  • This may be e.g. a polypeptide linker region.
  • Such linker(s) may be included e.g. between functional domains/regions/motifs of the chimeric transmembrane receptor. It may be a linker region naturally occurring e.g. in wildtype PD1 receptor, or e.g. in co-stimulatory proteins, e.g. in costimulatory proteins from which the costimulatory domain of the receptor is derived.
  • polypeptide linker regions may be included between the transmembrane domain and the ligand binding domain, and/or between the transmembrane domain and the IgV domain of the chimeric PD1 receptor, and/or between the transmembrane domain and the at least one intracellular co-stimulatory domain, and/or between individual co-stimulatory domains (in embodiments comprising more than one co-stimulatory domains).
  • Such linker region may comprise 1-100 amino acids, or e.g. 1-80 amino acids, or e.g. 1-50 amino acids, or e.g. 5-100 amino acids.
  • a linker region of the chimeric transmembrane receptor as herein provided may comprise the amino acid sequence as set forth in SEQ ID No.9 (GGGS)n or as set forth in Seq ID No. 10 (GGGGS) n , wherein n is between 0 and 20, or wherein n is between 0 and 10, or where n is between 0 and 5, or where n is between 3 and 5.
  • each (polypeptide) linker known in the art is herewith envisaged as being potentially included in the chimeric PD1 switch receptor of the T-cells according to the fifteenth or sixteenth aspect.
  • the chimeric transmembrane receptor polypeptide of the T-cells may comprise at least one costimulatory cytoplasmic polypeptide domain or cytoplasmic polypeptide motif of CD2.
  • cytoplasmic co-stimulatory domains of CD2 may be fused to the at least one PD1 polypeptide region comprising a functional PD1 receptor extracellular ligand binding domain, in order to generate a functional chimeric PD1 switch receptor capable of redirecting the signaling pathways triggered by PD1 receptor engagement with at least one PD1 ligand such that - instead of inducing inhibitory pathways in the cell - binding of the at least one PD1 ligand promotes T-cell activation, persistence and enhanced anti-tumor responses of a T-cell expressing the chimeric transmembrane receptors as herein provided, if the T-cell - at the same time - comprises/expresses an engineered T-cell receptor.
  • the T-cells expressing the chimeric PD1 receptor according to the fifteenth or sixteenth aspect may comprise e.g. at least one complete cytoplasmic domain of CD2.
  • the at least one cytoplasmic polypeptide domain or cytoplasmic polypeptide motif of CD2 may have an amino acid sequence having at least 70%, or at least 71%, or at least 72%, or at least 73%, or at least 74%, or at least 75%, or at least 76%, or at least 77%, or at least 78%, or at least 79%, or at least 80%, or at least 81%, or at least 82%, or at least 83%, or at least 84%, or at least 85%, or at least 86%, or at least 87%, or at least 88%, or at least 89% or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96% or at least 97%
  • T-cell comprising the chimeric PD1 receptor together with the engineered TCR according to the fifteenth or sixteenth aspect may exhibit sustained or enhanced cytotoxicity and/or cytokine secretion upon binding a PD1 ligand.
  • T-cells comprising the chimeric transmembrane receptor in accordance with the fifteenth or sixteenth aspect may be capable of increasing resistance to PD1 ligand expressing cancer cells.
  • the chimeric PD1 receptors of the T-cells may comprise any functional combination of costimulatory cytoplasmic polypeptide domain(s) motif(s) and/or region(s) of the cytoplasmic polypeptide domain, region or motif selected from the group consisting of CD2 and other co-stimulatory proteins.
  • the chimeric PD1 receptor of the T-cells may comprise at least one cytoplasmic polypeptide domain, region or motif of CD2.
  • a cytoplasmic polypeptide region of CD2 may comprise the complete cytoplasmic domain of wildtype human CD2.
  • the cytoplasmic polypeptide region of CD2 included in the chimeric PD1 receptor may comprise at least one functional, co-stimulatory motif/domain/region of the complete wildtype human CD2 cytoplasmic domain.
  • wildtype human CD2 cytoplasmic domain as referred to herein may relate to a polypeptide comprising amino acid sequence 236-351 of UniProtKB database entry P06729 ⁇ CD2_HUMAN, as set forth e.g. in SEQ ID No. 19. It is herewith envisaged that the cytoplasmic polypeptide domain, region or motif of wildtype CD2 as included in the chimeric PD1 receptor of the T-cell as herein provided may have one or more conservative amino acid substitutions relative to the amino acid sequence as set forth in SEQ ID No. 21. In particular, all amino acid substitutions that maintain the functional activity of the cytoplasmic polypeptide domain, region or motif of wildtype CD2 are envisaged.
  • the polypeptide of the chimeric PD1 receptor may have an amino acid sequence with at least 85% identity to the amino acids as set forth in SEQ ID NO.: 22.
  • a chimeric PD1 receptor as herein provided may e.g. comprise a polypeptide having an amino acid sequence with at least 85% or at least 86%, or at least 87%, or at least 88%, or at least 89% or at least 90%, or at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96% or at least 97%, or at least 98%, or at least 99%, or 100% identity to the amino acids as set forth in SEQ ID No. 22.
  • a chimeric PD1 receptor as herein provided may have one or more conservative amino acid substitutions relative to an amino acid sequence as set forth in SEQ ID No. 22.
  • the cell may be a ap T-cell, yo T-cell, and/or a natural killer T-cell.
  • the ap T-cell may be a CD4 T-cell, or the ap T-cell may be a CD8 T-cell, or the y5 T-cell may comprise e.g. a Vy1 chain or a Vy2 chain, or may be e.g. a Vy9V82+ T-cell.
  • the T-cell may express a chimeric PD1 receptor as herein described.
  • the T-cell may further express an engineered T-cell receptor or a CAR.
  • the T-cell may be a CD 4 T-cell that further expresses a CD8 Co-receptor, e.g. both CD8a and CD8p Co-receptor, or any engineered protein exhibiting CD8 Co-receptor functionality.
  • the T-cells further express an engineered T-cell receptor.
  • Engineered T-cells of the present disclosure can be used to treat a subject in need of treatment for a condition, for example, a cancer described herein.
  • the T-cells may be o.p T-cells or y5 T-cells that express the chimeric PD1 receptor polypeptide as described herein, and furthermore an engineered TCR.
  • the T-cells may further express a CD8 Co-receptor such as a wildtype or chimeric CD8 co-receptor.
  • T- cells described herein may be used to treat a cancer, including solid tumors and hematologic malignancies. For example, “hot” tumors or “cold” tumors may be treated by the T-cells herewith provided.
  • the engineered T-cell receptor as herein described may specifically bind a MAGE antigen family member, such as MAGE-A1 or Mage-A4, or wherein the engineered T-cell receptor may specifically bind an antigen selected from the group consisting of a PRAME antigen, a NY-ESO-1 antigen, a GP100 antigen, an AFP antigen, a Col6A3 antigen, an HPV-16 antigen, a WT1 antigen, an HA1 antigen, an HA2 antigen, a mutated KRAS antigen, a mutated NRAS antigen, a mutated HRAS antigen, a mutated TP53 antigen, and an EGFR antigen.
  • a PRAME antigen such as MAGE-A1 or Mage-A4
  • a GP100 antigen such as MAGE-A1 or Mage-A4
  • an antigen selected from the group consisting of a PRAME antigen, a NY-ESO-1 antigen,
  • the expression “mutated” with respect to specific tumor antigens as herein used relates to well-known mutations within the epitope region of the respective protein, polypeptide or peptide that has been correlated with expression in a human cancer.
  • the T-cells described herein may also be used to treat an infectious disease.
  • the T-cells described herein may be used to treat an infectious disease, an infectious disease may be caused a virus.
  • the T-cells described herein may be used to treat an immune disease, such as an autoimmune disease.
  • the T-cells may be o.p T-cells or T-cells that express a chimeric transmembrane receptor as described herein, and an engineered TCR, and optionally a CD8 Co-receptor such as a wildtype or chimeric CD8 co-receptor.
  • the T-cell of the fifteenth or sixteenth aspect may be derived from a healthy subject, or the T-cell may be derived from a patient suffering from a disease.
  • the T-cell may be derived from an induced pluripotent stem cell (iPSCs).
  • iPSCs induced pluripotent stem cell
  • kits comprising means to prepare the T-cells as herein provided.
  • this invention relates to a pharmaceutical composition comprising the T-cell provided by the present invention.
  • the pharmaceutical composition may further comprise an adjuvant, excipient, buffer, diluent, carrier, stabilizer or combination thereof.
  • a pharmaceutical composition comprising T-cells which express the chimeric PD1 receptor as herein described.
  • the T-cells may further express an engineered T-cell receptor or a CAR.
  • the pharmaceutical composition may further comprise CD4 T-cells expressing said chimeric transmembrane receptor, expressing an engineered T-cell receptor, and further expressing a recombinant CD8 Co-receptor, such as e.g. a CD8a receptor and a CD8 receptor, or a chimeric CD8 receptor.
  • the pharmaceutical composition may further comprise one or more pharmaceutically acceptable carriers.
  • Any pharmaceutically acceptable carrier can be used, as long as the carrier does not impact the viability of the T-cells to be administered is suitable for the chosen route of administration of the pharmaceutical composition.
  • the pharmaceutical acceptable carrier may be a physiological saline solution, optionally with components such as human serum albumin that can improve the viability of the T-cells that express the chimeric PD1 receptor. It is also possible that the chimeric transmembrane receptor expressing T-cells are stored, after their manufacture, in frozen form, for example at a temperature of between -20°C and -80 °C. In this case, the pharmaceutical composition may contain cryo-protectants that have been added to protect the cells from being damaged by the freezing process.
  • cryoprotectants examples include glycerol, DMSO. These cryoprotectants can be used together with crystalloid solutions such as commercially available HypoThermosol® or PlasmaLyte-A solution which are both approved for infusion and are available in pharmaceutical grade.
  • media of the “CryoStor family” include media of the “CryoStor family”, commercially available animal protein-free defined cryopreservation media from Biolife Solutions such as CyroStor2 (CS2, an optimized freeze media pre-formulated with 2% DMSO), CyroStor5 (CS5, an optimized freeze media pre-formulated with 5% DMSO), or CyroStorlO (CS10, an optimized freeze media pre-formulated with 10% DMSO).
  • a method for preparing a T-cell for immunotherapy comprising
  • the method may comprise transforming, transfecting (e.g. electroporating) or transducing the isolated T-cells with the vector.
  • transforming, transfecting e.g. electroporating
  • transducing the isolated T-cells with the vector.
  • a method for treating a patient having a disease comprising introducing in vivo the vector as herein disclosed into a T-cell of the patient.
  • the nucleic acid may be a DNA or an mRNA.
  • the vector may be - for example - a nonreplicating viral vector.
  • the nucleic acid may be mRNA, and the mRNA may be in vivo introduced into the T-cell of the patient using nanoparticles, such as lipid nanoparticles.
  • the disease may be e.g. an autoimmune disease or a cancer.
  • a cancer treated by the method may be selected from the group consisting of non-small cell lung cancer, small cell lung cancer, pancreatic cancer, ovarian cancer, melanoma, breast cancer, liver cancer, kidney cancer, esophageal cancer, brain cancer, gastric cancer, Merkel cell carcinoma, leukemia, urinary bladder cancer, uterine cancer, colorectal cancer, gallbladder cancer, bile duct cancer, and prostate cancer.
  • the cancer treated may be a solid tumor.
  • the lung cancer may be, but is not limited to, non-small cell lung cancer (NSCLC), including squamous cell carcinoma of the lung, adenocarcinoma of the lung, large cell carcinoma of the lung and other histologic types of NSCLC or small cell lung cancer.
  • NSCLC non-small cell lung cancer
  • the breast cancer may be, but is not limited to, ductal breast cancer, ductal-invasive breast cancer, invasive breast cancer, tubular breast cancer, medullary breast cancer or combinations thereof.
  • the gastric cancer may be gastric adenocarcinoma or squamous cell cancer.
  • the sarcoma cancer may be, but is not limited to, chondrosarcoma cancer, osteosarcoma cancer or combinations thereof.
  • the adenoma cancer may include, but is also not limited to, gastric adenocarcinoma, pancreatic adenocarcinoma or combinations thereof.
  • the cancer cells may express at least one ligand of PD1.
  • a method for increasing cytotoxicity of a T-cell in adoptive cell therapy comprising introducing a vector or nucleic acid as herein provided into the T-cell.
  • the T-cell receptor may be a recombinant T- cell receptor that specifically binds a tumor specific antigen.
  • this may be a PRAME antigen, or a MAGE antigen such as e.g. a Mage-A1 antigen.
  • Example 1 In-vitro T-cell killing analysis of T-cells according to the present invention transduced with chimeric PD1 receptor polypeptides according to the present invention and an engineered T-cell receptor
  • chimeric PD1 receptor constructs have been used to transduce CD8 T-cells together with a HLA-I restricted TCR raised against either MAGE-A1 or PRAME.
  • Purified transduced T-cells were used in an in-vitro T-cell killing assay with PD-L1 -overexpressing Corl_23-A2-NLR cells for evaluating cytotoxicity of the transduced T-cells.
  • Table 2 as presented below summarize the chimeric PD1 receptor constructs that have been generated by the inventors in a schematic representation: [00197] Table 2
  • CYP relates to the origin of the cytoplasmic domain of the chimeric receptor encoded by the plasmid created by the Inventors.
  • TM relates to the origin of the transmembrane domain of the chimeric receptor encoded by the plasmid, and
  • EC relates to the origin of the extracellular domain of the chimeric receptor encoded by the plasmid created by the Inventors.
  • PBMCs from a healthy donor buffy coat were isolated by density gradient centrifugation with Lymphoprep.
  • Purified polyclonal CD8 T-cells were obtained by positive selection with anti-CD8+ microbeads.
  • CD3 T-cells were activated using TransAct in presence of IL-7/IL-15. Two days post activation, CD8 T-cells were separately transduced with either HLA-I restricted TCR raised against MAGE-A1 (MAGE-A1_TCR) alone, or together with different versions of the PD1 Switch receptors (for the experiments according to Fig.2 and 3).
  • MAGE-A1_TCR HLA-I restricted TCR raised against MAGE-A1
  • CD8 T-cells were separately transduced with either a TCR raised against PRAME (PRAME_TCR) alone, or together with different versions of the PD1 Switch receptors.
  • PRAME_TCR PRAME
  • the HLA-I restricted TCR raised against MAGE-A1 (MAGE-A1_TCR) as used herein has been described e.g. in WO 2014/118236, which is herewith incorporated by reference in its entirety.
  • the HLA-I restricted TCR raised against MAGE-A1 as used herein relates to “TCR1367” as described in WO 2014/118236.
  • the CDR sequences of the respective a and p chain of “TCR1367” as used herein are further described -for example - in WO 2023/083864, which is herewith incorporated by reference in its entirety.
  • Transduced CD8 T-cells were further expanded, and at Day 9 the transduced fraction was positively selected using CD34 microbeads.
  • Purified transduced T-cells were cultured for further expansion and were harvested and cryopreserved at Day 10.
  • T-cell characterization was based on transgene expression levels using FACS and killing assay.
  • the in-vitro T-cell killing assay was performed according to the method described e.g. by Kalbasi, A., Siurala, M., Su, L.L. et al. “Potentiating adoptive cell therapy using synthetic IL-9 receptors”. Nature 607, 360-365 (2022).
  • the human TCR T-cell repetitive killing assay was conducted using IncuCyte Live Cell Analysis. Using PDL1 overexpressing CorL23-A2-NLR cells, 1x10 4 tumor cells were plated per well in 96-well plates.
  • Untransduced (mock), or transduced human T-cells were added in triplicates at 1 to 2 E:T ratio (for the experiments shown by Fig 2) or 1:4 E:T ratio (for the experiments shown by Fig 3).
  • the in-vitro T-cell killing assay was performed according to the method described e.g. by Kalbasi, A., Siurala, M., Su, L.L. et al. “Potentiating adoptive cell therapy using synthetic IL-9 receptors”. Nature 607, 360-365 (2022).
  • the human TCR T-cell repetitive killing assay was conducted using IncuCyte Live Cell Analysis. Using PDL1 expressing NCI-H1703 cells, 1x10 4 tumor cells were plated per well in 96-well plates.
  • Extracellular surface staining was performed for 30 minutes at 4°C in flow cytometry FACS buffer (BD Bioscience).
  • the following antibodies were used: from BioLegend: CD8a (clone HIT8a); from Invitrogen: CD34 (clone QBEND10), CD34 (clone 4H11); from Miltenyi Biotec: CD8a (clone REA734), from Beckman Coulter: TCRBV3S1 V£3.
  • anti-Human PD-1(CD279) (Clone EH12.1)
  • PE-conjugated HLA-A*02 01 specific MAGE-A1 MHC tetramer (KVLEYVIKV; SEQ ID No.
  • Fig. 2 shows the results of the killing assay with the chimeric PD1 receptors in PD-L1 -overexpressing CorL23-A2-NLR cells, wherein an E:T ratio of 1 :2 was used
  • Fig. 3 shows the results of the killing assay with the chimeric PD1 receptors in PD-L1 -overexpressing CorL23-A2-NLR cells, wherein an E:T ratio of 1 :4 was used.
  • “Mock” relates to mock-transduced T-cell fraction
  • “Cancer cells” relate to the sample merely comprising PD-L1 -overexpressing CorL23-A2-NLR cells
  • “TCR tag” relates to CD8 T-cell fraction transduced with HLA-I restricted TCR raised against MAGE-A1 (MAGE-A1_TCR)
  • “TCR+PD1 DN” relates to CD8 T-cell fraction transduced with HLA-I restricted TCR raised against MAGE-A1 (MAGE-A1_TCR) together with a PD1 receptor that lacks its cytoplasmic domain ( pl_1258)
  • “TCR_PD1-ICOS_SwR” relates to CD8 T- cell fraction transduced with HLA-I restricted TCR raised against MAGE-A1 (MAGE- A1_TCR) together with a chimeric PD1 receptor comprising a PD1 extracellular polypeptide region and an ICOS transmembrane and cytoplasm
  • Fig. 5 shows the results of the killing assay with chimeric PD1 receptors in PD-L1 -expressing NCI-H1703 cells, wherein an E:T ratio of 2:1 was used.
  • PRAME TCR only relates to CD8 T-cell fraction transduced with HLA-I restricted TCR raised against PRAME (PRAME_TCR);
  • PRAME TCR + PD1- ICOS relates to CD8 T-cell fraction transduced with HLA-I restricted TCR raised against PRAME (PRAME_TCR) together with a chimeric PD1 receptor comprising a PD1 extracellular polypeptide region, a PD1 transmembrane domain and an ICOS cytoplasmic polypeptide region (pl_2352 in Fig 4);
  • PRAME TCR + PD1-41BB relates to CD8 T-cell fraction transduced with HLA-I restricted TCR raised against PRAME (PRAME_TCR) together with a chimeric PD1 receptor comprising a PD1
  • CD8 cells transduced with vectors comprising nucleic acids encoding for different chimeric PD1 receptors as herein provided have been checked for expression of the chimeric PD1 receptor.
  • the respective chimeric PD1 receptors used in Flow Cytometry Analysis correspond to the respective constructs according to Table 2 as shown above.
  • PD1dnV1 and Mock transduced cells have been used as controls.
  • the chimeric PD1 receptors comprising both a transmembrane domain and a cytoplasmic domain of TLR2, TLR4, CD40L, GITR, IL-6R- beta and 4-1 BB, respectively, were not expressed. All other chimeric PD1 receptors as listed in Table 2 show high expression in the transduced CD8 T-cells.
  • the increase killing activity is also achieved with an engineered HLA-I restricted TCR raised against PRAME together with a chimeric PD1 receptor comprising a non - PD1 costimulatory cytoplasmic polypeptide domain, motif or region of ICOS or 41 BB.
  • a non - PD1 co-stimulatory cytoplasmic polypeptide domain, motif or region of CD40 wherein said costimulatory region comprises full length wildtype CD40 cytoplasmic fused to at least one further (additional) CD40 costimulatory motif (i.e. the CD40 TRAF6 domain and- additionally - the CD40 TRAF123 domain as present in construct pl_2355).
  • the results described above demonstrate - in principle - suitability of the chimeric PD1 switch receptor polypeptides as herein provided for improving adoptive cell therapy (ACT).
  • the chimeric PD1 receptor polypeptides of the present invention e.g. in combination with an engineered T-cell receptor, may be functional in providing improved resistance to the T-cell in immunosuppressive tumor microenvironment, in preventing T-cell exhaustion and/or depletion through apoptosis; and in stimulating T-cell proliferation and functional activity, such as increased cytotoxicity.
  • fusion of at least one non-PD1 polypeptide region, wherein the at least one non-PD1 polypeptide region comprises at least one costimulatory cytoplasmic polypeptide domain, region or motif of CD2, HVEM, CD30 or CD40, to a PD1 polypeptide region that comprises the PD1 ligand binding domain is able to act like a “switch” receptor, by turning negative signals into positive signals, thereby enhancing cytotoxicity of a T-cell in presence of tumor cells that express at least one PD1 ligand.
  • Engineered T-cells expressing the chimeric PD1 receptor polypeptides together with an engineered T-cell receptor as provided herein exhibit an improved killing activity compared to control samples in presence of a PD1 ligand.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Toxicology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne entre autres un récepteur de protéine 1 de mort cellulaire programmée (PD1) chimérique ; comprenant un polypeptide, ledit polypeptide comprenant au moins une région polypeptidique PD1 comprenant un domaine de liaison au ligand extracellulaire PD1 ; en outre ledit polypeptide comprenant au moins un domaine, région ou motif polypeptidique cytoplasmique costimulateur non-PD1, ledit au moins un domaine, région ou motif polypeptidique cytoplasmique étant un domaine, région ou un motif polypeptidique cytoplasmique de CD30, CD40, médiateur d'entrée de l'herpèsvirus (HVEM), ou CD2. L'invention concerne également des acides nucléiques, des vecteurs et des lymphocytes T correspondants comprenant ou exprimant les récepteurs chimériques, une composition pharmaceutique comprenant les lymphocytes T, et des méthodes de préparation d'un lymphocyte T pour une immunothérapie et pour le traitement d'une maladie, respectivement, dans lesquels le récepteur PD1 chimérique est utilisé.
PCT/EP2025/052975 2024-03-13 2025-02-05 Récepteur de protéine 1 de mort cellulaire programmée chimérique Pending WO2025190565A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP24163116 2024-03-13
EP24163116.7 2024-03-13

Publications (1)

Publication Number Publication Date
WO2025190565A1 true WO2025190565A1 (fr) 2025-09-18

Family

ID=90365095

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2025/052975 Pending WO2025190565A1 (fr) 2024-03-13 2025-02-05 Récepteur de protéine 1 de mort cellulaire programmée chimérique

Country Status (1)

Country Link
WO (1) WO2025190565A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013019615A2 (fr) 2011-07-29 2013-02-07 The Trustees Of The University Of Pennsylvania Récepteurs de commutation par costimulation
WO2014118236A2 (fr) 2013-01-29 2014-08-07 Max-Delbrück-Centrum Für Moledulare Medizin (Mdc) Berlin-Buch Constructions à avidité élevée de reconnaissance d'antigènes
WO2023083864A1 (fr) 2021-11-09 2023-05-19 T-Knife Gmbh Méthodes de sélection d'un patient pour le traitement d'une tumeur solide positive pour mage-a1, de prédiction de la sensibilité d'un patient diagnostiqué comme atteint d'une tumeur solide positive pour mage-a1 au traitement de cette tumeur et de traitement d'un patient diagnostiqué comme atteint d'une telle tumeur solide positive pour mage-a1, ainsi que des compositions pharmaceutiques et des kits de diagnostic correspondants
WO2024041761A1 (fr) * 2022-08-23 2024-02-29 Medigene Immunotherapies Gmbh Combinaison de récepteurs de lymphocytes t spécifiques de ny-eso-1 et de récepteurs chimériques de costimulation

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013019615A2 (fr) 2011-07-29 2013-02-07 The Trustees Of The University Of Pennsylvania Récepteurs de commutation par costimulation
WO2014118236A2 (fr) 2013-01-29 2014-08-07 Max-Delbrück-Centrum Für Moledulare Medizin (Mdc) Berlin-Buch Constructions à avidité élevée de reconnaissance d'antigènes
WO2023083864A1 (fr) 2021-11-09 2023-05-19 T-Knife Gmbh Méthodes de sélection d'un patient pour le traitement d'une tumeur solide positive pour mage-a1, de prédiction de la sensibilité d'un patient diagnostiqué comme atteint d'une tumeur solide positive pour mage-a1 au traitement de cette tumeur et de traitement d'un patient diagnostiqué comme atteint d'une telle tumeur solide positive pour mage-a1, ainsi que des compositions pharmaceutiques et des kits de diagnostic correspondants
WO2024041761A1 (fr) * 2022-08-23 2024-02-29 Medigene Immunotherapies Gmbh Combinaison de récepteurs de lymphocytes t spécifiques de ny-eso-1 et de récepteurs chimériques de costimulation

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR LABORATORY PRESS, pages: 1 - 3
ALTSCHUL, S. F. ET AL., NUCL. ACIDS RES., vol. 25, 1997, pages 3389 - 3402
BELL ET AL., EXPERIMENTAL BIOLOGY AND MEDICINE, vol. 235, 2010, pages 1269 - 1276
CHO HYUN-IL ET AL: "Abstract 5506: CD30 co-stimulatory domain enhances the efficacy of chimeric antigen receptor-engineered [gamma][delta]T cells", CANCER RESEARCH, vol. 82, no. 12_Supplement, 15 June 2022 (2022-06-15), pages 5506 - 5506, XP055952025, ISSN: 0008-5472, DOI: 10.1158/1538-7445.AM2022-5506 *
HYUN-IL ET AL: "Abstract 1788: A novel costimulatory signaling domain for chimeric antigen receptor-engineering | Cancer Research | American Association for Cancer Research", CANCER RESEARCH, vol. 83, no. 7_Supplement, 4 April 2023 (2023-04-04), pages 1788 - 1788, XP093275717, ISSN: 0008-5472, DOI: 10.1158/1538-7445.AM2023-1788 *
KALBASI, A.SIURALA, M.SU, L.L. ET AL.: "Potentiating adoptive cell therapy using synthetic IL-9 receptors", NATURE, vol. 607, 2022, pages 360 - 365, XP037900195, DOI: 10.1038/s41586-022-04801-2
KOBOLD S ET AL: "Impact of a New Fusion Receptor on PD-1-Mediated Immunosuppression in Adoptive T Cell Therapy", JOURNAL OF THE NATIONAL CANCER INSTITUTE, OXFORD UNIVERSITY PRESS, GB, vol. 107, no. 8, DJV146, 23 June 2015 (2015-06-23), pages 1 - 10, XP002743492, ISSN: 0027-8874, [retrieved on 20150801], DOI: 10.1093/JNCI/DJV146 *
LORENZINI THEO ET AL: "Rational design of PD-1-CD28 immunostimulatory fusion proteins for CAR T cell therapy", BRITISH JOURNAL OF CANCER, vol. 129, no. 4, 4 July 2023 (2023-07-04), London, pages 696 - 705, XP093179121, ISSN: 0007-0920, DOI: 10.1038/s41416-023-02332-9 *
MERTEN ET AL., J. VIROL., vol. 79, 2005, pages 834 - 840
RAMAMOORTHNARVEKAR: "Non Viral Vectors in Gene Therapy- An Overview", JCLINDIAGN RES., vol. 9, no. 1, 2015, pages GE01 - GE06
SAILER NADJA ET AL: "T-Cells Expressing a Highly Potent PRAME-Specific T-Cell Receptor in Combination with a Chimeric PD1-41BB Co-Stimulatory Receptor Show a Favorable Preclinical Safety Profile and Strong Anti-Tumor Reactivity", CANCERS, vol. 14, no. 8, 1998, 14 April 2022 (2022-04-14), pages 1 - 18, XP093001527, DOI: 10.3390/cancers14081998 *
WANG ET AL., J. VIROL., vol. 81, 2007, pages 10869 - 10878

Similar Documents

Publication Publication Date Title
US12351617B2 (en) Immune cells comprising truncated NKG2D chimeric receptors
US11034763B2 (en) Flag tagged CD19-CAR-T cells
EP2771355B1 (fr) CELLULE EFFECTRICE MODIFIÉE (OU RÉCEPTEUR CHIMÉRIQUE) POUR TRAITER LA NÉOPLASIE EXPRIMANT LE DISALOGANGLIOSIDE Gd2
US12258381B2 (en) Activating chimeric receptors and uses thereof in natural killer cell immunotherapy
CN109306016B (zh) 共表达细胞因子il-7的nkg2d-car-t细胞及其用途
CN108004259B (zh) 靶向b细胞成熟抗原的嵌合抗原受体及其用途
KR20140127829A (ko) 2세대 키메라 항원 수용체에서 cd2 시그널링 도메인의 용도
KR20180021137A (ko) 키메라 항원 수용체 (car), 조성물 및 이의 사용 방법
JP6762485B2 (ja) 抗グリピカン−1−免疫抗原受容体
WO2018068766A1 (fr) Récepteur antigénique chimérique ciblant le cd19, son procédé de préparation et son application
US20190192573A1 (en) Anti-osteosarcoma car-t derived from the antibody oi-3
CN115925989A (zh) 长效的双靶点嵌合抗原受体、核酸分子、重组载体、细胞及其应用
Ren-Heidenreich et al. Comparison of the TCR ζ-chain with the FcR γ-chain in chimeric TCR constructs for T cell activation and apoptosis
JP2022542051A (ja) 養子免疫療法のための組成物および方法
JP6842688B2 (ja) キメラ抗原受容体
WO2025190565A1 (fr) Récepteur de protéine 1 de mort cellulaire programmée chimérique
WO2025191067A1 (fr) Récepteur transmembranaire chimérique comprenant au moins immunorécepteur de lymphocytes t avec une region polypeptidique des domaines ig et itim (tigit), des lymphocytes t exprimant le récepteur de commutateur tigit humain chimérique, des vecteurs avec des acides nucléiques codant pour le récepteur tigit, des kits pour préparer les lymphocytes t, ainsi que des compositions pharmaceutiques correspondantes et des méthodes pour traiter un patient ayant une maladie et pour augmenter la cytotoxicité d'un lymphocyte t dans une thérapie cellulaire adoptive
WO2025045752A1 (fr) Récepteur de commutateur cd 95 humain chimérique, lymphocytes t exprimant un tel récepteur conjointement avec un récepteur de lymphocytes t modifié, vecteurs respectifs, kits, compositions pharmaceutiques et méthodes pour traiter un patient présentant une maladie
CN108165568B (zh) 一种培养CD19CAR-iNKT细胞方法及用途
CN115960256A (zh) 一种长效的嵌合抗原受体、载体及其构建方法和应用
WO2025027088A1 (fr) Co-récepteur cd8 humain chimérique amélioré, acide nucléique codant pour le co-récepteur cd8 humain chimérique amélioré, vecteurs correspondants, lymphocytes t isolés transduits avec l'acide nucléique ou vecteurs correspondants et kits pour les préparer, compositions pharmaceutiques correspondantes et méthodes de traitement d'un patient atteint d'une maladie
WO2025027087A1 (fr) Co-récepteur cd8 humain chimérique, acide nucléique codant pour le co-récepteur cd8 humain chimérique, vecteurs correspondants, lymphocytes t isolés transduits avec l'acide nucléique ou vecteurs correspondants et kits pour les préparer, ainsi que compositions pharmaceutiques correspondantes et méthodes de traitement d'un patient ayant une maladie
TW202237825A (zh) 針對同種異體細胞療法的通用嵌合抗原受體表現之免疫細胞
KR102568745B1 (ko) 세포외소포체 분비능이 증진된 면역세포 및 이를 활용한 면역 항암요법
JP7054181B2 (ja) キメラ抗原受容体

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: 25708343

Country of ref document: EP

Kind code of ref document: A1