[go: up one dir, main page]

WO2025059162A1 - Activateur car contenant des variants d'il-2 pour améliorer la fonctionnalité de cellules car-t - Google Patents

Activateur car contenant des variants d'il-2 pour améliorer la fonctionnalité de cellules car-t Download PDF

Info

Publication number
WO2025059162A1
WO2025059162A1 PCT/US2024/046173 US2024046173W WO2025059162A1 WO 2025059162 A1 WO2025059162 A1 WO 2025059162A1 US 2024046173 W US2024046173 W US 2024046173W WO 2025059162 A1 WO2025059162 A1 WO 2025059162A1
Authority
WO
WIPO (PCT)
Prior art keywords
car
engager
cells
amino acid
bcma
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/US2024/046173
Other languages
English (en)
Inventor
Heydar MORAVEJ
Mohammad RASHIDIAN
Taha RAKHSHANDEHROO
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.)
Dana Farber Cancer Institute Inc
Original Assignee
Dana Farber Cancer Institute Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Dana Farber Cancer Institute Inc filed Critical Dana Farber Cancer Institute Inc
Publication of WO2025059162A1 publication Critical patent/WO2025059162A1/fr
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • A61K40/31Chimeric antigen receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/40Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
    • A61K40/41Vertebrate antigens
    • A61K40/42Cancer antigens
    • A61K40/4202Receptors, cell surface antigens or cell surface determinants
    • A61K40/421Immunoglobulin superfamily
    • A61K40/4211CD19 or B4
    • 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
    • A61K40/4202Receptors, cell surface antigens or cell surface determinants
    • A61K40/4214Receptors for cytokines
    • A61K40/4215Receptors for tumor necrosis factors [TNF], e.g. lymphotoxin receptor [LTR], CD30
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • 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
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K40/00
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction

Definitions

  • IL-2R The cognate IL-2 receptor (IL-2R) includes three subunits, namely IL-2 subunit alpha (IL-2R ⁇ ; CD25), IL-2 subunit beta (IL-2R ⁇ ; CD122), and the IL-2 subunit gamma (IL-2R ⁇ ; CD132).
  • IL-2 may also induce an alternative differentiation pathway of T cells, resulting in the generation of distinct “better” effector CD8 + T cells (Hashimoto et al., Nature 610(7930):173-181 (2022)). This process may rely, at least in part, on IL-2 binding to IL-2R ⁇ . Additionally, IL-2R ⁇ - biased agonists may drive T cells towards a terminally differentiated state (Codarri et al., Nature 610(7930):161-172 (2022)). [0005] Known IL-2 variants may have different affinities for different IL-2R subunits and may offer advantages in connection with therapies.
  • a weak affinity IL-2 (muIL2) that contains the amino acid substitutions of H16A and F42A, relative to wild-type IL-2, has increased selectivity for T cells and less toxicity through reduced global IL-2 binding and activation of all IL- 2R expressing cells.
  • the binding affinity of muIL2 for human IL2R ⁇ and IL2R ⁇ is decreased 110- and 3-fold, respectively, compared with wild-type IL-2 (Quayle et al., Clin. Cancer Res.26(8):1953- 1964 (2020)).
  • Challenges persist with IL-2-based therapies in the clinic, with many clinical trials failing to meet their primary endpoints (Raeber et al., Ebiomedicine 90:104539 pp. 1-25 (2023)).
  • CD19 is a B-cell co-receptor expressed on B cells and a wide variety of blood-borne malignancies.
  • CD19 CAR T cells were initially approved for the treatment of acute lymphoblastic leukemia (ALL) and have subsequently been approved for Burkitt’s Lymphoma and Mantle Cell Lymphoma.
  • BCMA is a receptor expressed on the surface of B-cell lineage cells and a major marker of multiple myeloma (MM).
  • MM is associated with an uncontrollable expansion of plasma cells in the bone marrow, which can progress to extra-medullary lesions forming elsewhere in the body.
  • BCMA CAR T cell therapy has shown great promise against MM with studies showing an overall response rate of 80% even in patients with extra-medullary lesions (Gagelmann et al., Eur. J. Haematol.104(4):318-327 (2020)).
  • challenges persist with CAR T cell therapy.
  • a recent meta-analysis of 22 CAR T cell clinical studies highlighted its ineffectiveness in solid tumors with a poor average overall response rate of 9% (Hou et al., Dis. Markers 2019:3425291 pp. 1-11 (2019)).
  • IL-2 variants chimeric antigen receptor (CAR)-engagers containing an IL-2 variant, and methods of enhancing activity of chimeric CAR immune cells are 2 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 expected to address the above needs.
  • the IL-2 variants per se or connected with another active moiety, such as an antibody or binding fragment thereof, augment anti-cancer therapy.
  • the CAR- engagers containing an IL-2 variant enhance CAR immune cells, driving them toward generation of memory cells, and preventing exhaustion.
  • a first aspect of the present disclosure is directed to an IL-2 variant, which differs from wild type IL-2 (SEQ ID NO: 102) in terms of from three to eight amino acid substitutions selected from amino acid residues H16, D20, R38, F42, Y45, E62, L72, and V91 of SEQ ID NO: 1.
  • the three to eight amino acid substitutions are selected from H16A, H16R, H16S, D20A, D20Q, R38D, F42A, Y45A, E62N, L72G, and V91H.
  • Related aspects include nucleic acids encoding the IL-2 variants, vectors containing the nucleic acid, cells transformed with the vector, methods of making the IL-2 variants, pharmaceutical compositions containing the IL-2 variants, and uses thereof to treat cancer.
  • One such use involves enhancement of adoptive cell therapy such as CAR T therapy.
  • another aspect of the present disclosure is directed to a chimeric antigen receptor (CAR)- engager designed for use with CAR-immune cell therapy such as CAR-T therapy, wherein the CAR- engager contains a first moiety comprising that binds an epitope on an extracellular domain (ED) of the CAR connected to a second moiety comprising an immune effector domain comprising the IL- 2 variant.
  • the ED includes one or more extracellular binding domains (EBDs) of the CAR and any other extracellular portions of the CAR, e.g., a linker that connects antibody fragments, or EBDs, etc.
  • EBDs extracellular binding domains
  • connection between the first moiety and the second moiety may be peptidic or non-peptidic (covalent), such that the CAR-engager may be a continuous protein or polypeptide, or a moiety that contains two proteinaceous entities connected by a covalent bond.
  • Another aspect of the present disclosure is directed to a heterodimeric CAR-engager, containing a first moiety that binds an epitope on an EB of the CAR connected to first dimerization domain, and a second moiety containing an immune effector domain which comprises the IL-2 variant connected to a second dimerization domain, wherein the first and the second dimerization domains bind to form a heterodimeric CAR-engager.
  • nucleic acids that encode the CAR-engager in the embodiments wherein the CAR-engager is a continuous protein
  • nucleic acids that encode the second moiety of the heterodimeric CAR-engager protein vectors containing the nucleic acids encoding the CAR- engager protein, vectors containing the first and/or second proteins of the heterodimeric CAR- engager, cells transformed with the vector(s), pharmaceutical compositions that contain the CAR- engager and a pharmaceutically acceptable carrier, and methods of making the CAR-engager protein.
  • a further aspect of the present disclosure is directed to a method of treating cancer.
  • the method entails administering to a subject a first course of an effective amount of CAR-engager therapy.
  • the subject will have received a prior administration of immune cells that express a CAR that contains an extracellular domain that binds an antigen present on a cancer cell that contains the ectodomain, and the ectodomain of the CAR-engager, a transmembrane domain, and an intracellular domain comprising a stimulatory domain.
  • Working examples e.g., Example 11, disclosed herein demonstrate that CAR-engagers with different IL-2 variants enhance CAR immune cells that target BCMA and CD19, drives them toward generation of memory cells, and prevents exhaustion of the CAR-immune cells.
  • the working examples also demonstrate that the timing and dosage amounts of the CAR-engager therapy may optimize its effects on the prior CAR immune cell therapy in terms enhancing CAR immune cells that target a cancer antigen by driving them toward generation of memory immune cells, and exhaustion-preventative proliferation of the CAR immune cells. And thus, the working examples present a hypothesis of a fundamental mechanism of action as between the CAR-engager and the CAR immune cells.
  • the working examples demonstrate that the two binding events, namely the binding between the immune effector domain of the CAR-engager and the cognate receptor on the immune cells, and the binding between the moiety of the CAR-engager that binds the EB of the CAR, produce a synergistic, molecular “cross-talk” between the intracellular domain (endodomain) of the cognate receptor and the endodomain stimulatory regions of the CAR, respectively, that results in production of IFN- ⁇ and TNF- ⁇ , and ultimately the generation of memory immune cells, and exhaustion-preventative proliferation of the CAR immune cells.
  • FIG. 1 schematically illustrates the domains of a CAR-engager according to some embodiments that contains a first moiety containing an antigen that is used as a target for a CAR, 4 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 such as the ectodomain of CD19 or BCMA on the surface of cancer cells, a dimerization domain, such as CH3, and a second moiety containing an immune cell effector domain (ICE).
  • the CAR- engager may be a monomer, a dimer, or a multimer.
  • FIGs.2A – 2I are a set of illustrations and line plots that show three CAR-engagers.
  • FIG.2A – 2I are a set of illustrations and line plots that show three CAR-engagers.
  • FIG.2A schematically illustrates a CAR-engager that contains a BCMA ectodomain and a Neo2/15 synthetic cytokine immune cell effector domain.
  • FIG.2B schematically illustrates a CAR-engager that contains a BCMA ectodomain and two weak affinity mutated IL-2 (mIL2) synthetic cytokine immune cell effector domains.
  • FIG. 2C schematically illustrates a CAR-engager that contains a BCMA ectodomain and a 4-1BBL immune cell effector domain.
  • FIG.2D is a line plot that shows dose-dependent staining of CAR T cells that bind CD19 or non-transduced T cells (NT T cells) with CAR-engager or control proteins.
  • FIG.2E is a line plot that shows dose-dependent staining of CAR T cells that bind BCMA or non-transduced T cells (NT T cells) with CAR-engager or control proteins.
  • FIGs.2F and 2G are a line and bar plot, respectively, that together show dose-dependent activation of CAR T cells after CAR-engager treatment.
  • FIG. 2H is a line plot showing that the BCMA-muIL2 CAR-engager does not block the killing efficacy of the CAR T cells.
  • FIG. 2I is a line plot that shows phosphorylation of signal transducer and activator of transcription (STAT5) in the BCMA CAR T cells.
  • FIGs.3A – 3B are a set of illustrations and line plots showing the effects of CAR-engagers on non-transduced T cells.
  • FIG.3A schematically illustrates the experimental design.
  • FIG.3B is a set of line plots that show T cell count and carboxyfluorescein succinimidyl ester (CFSE) staining of non-transduced, activated T cells treated with teceleukin, a CAR-engager containing an BCMA ectodomain and two mutated weak affinity IL-2 (muIL2), or CAR-engager containing an BCMA ectodomain and a Neoleukin domain.
  • CFSE carboxyfluorescein succinimidyl ester
  • FIG. 4A – 4C are a set of illustrations and line plots showing that CAR-engagers specifically activate CAR T cells.
  • FIG.4A schematically illustrates the experimental design.
  • FIG. 4B is a bar plot that shows the percentage of CD69 + anti-BCMA CAR-transduced, activated T cells after treatment with BCMA CAR-engager, BCMA CAR-engager without an immune cell effector domain control, or a non-antigen-specific CAR-engager control.
  • FIG. 4C is a bar plot that shows the percentage of CD69 + anti-CD19 CAR-transduced, activated T cells after treatment with CD19 CAR-engager or a non-antigen-specific CAR-engager control.
  • FIG.5 is a line plot showing that CAR-engagers do not inhibit BCMA CAR T cell killing and that the percentage of OPM2 target cell survival after incubation with CAR T cells and CAR- engagers (red) or non-transduced T cells (blue).
  • FIGs. 6A – 6C are a set of illustrations and photographs showing that CAR-engagers reduce tumor burden in vivo.
  • FIG.6A schematically illustrates the experimental design.
  • FIGs.6B – 6C are a set of photographs that show tumor burden in mice before and after CAR T cell infusion and CAR-engager treatment.
  • FIGs.7A – 7C are a set of flow cytometry plots showing the tumor burden in mice after CAR T cell infusion and CAR-engager treatment.
  • FIG. 7A is a set of flow cytometry plots that shows OPM2 tumor burden in blood, spleen, and lymph nodes.
  • FIG.7B is a set of flow cytometry plots that shows OPM2 tumor burden in bone marrow and lung.
  • FIG.7C is a set of flow cytometry plots that shows OPM2 tumor burden in liver, kidney, and the eye tumor site.
  • eGFP OPM2 cells
  • PerCP signal control is shown on the x-axis.
  • FIGs.8A – 8C are a set of flow cytometry plots showing human CD45 + and CAR- T cells in mice after CAR T cell infusion and CAR-engager treatment.
  • FIG.8A is a set of flow cytometry plots that shows CAR T cells in blood, spleen, and lymph nodes.
  • FIG.8B is a set of flow cytometry plots that shows CAR T cells in bone marrow and lung.
  • FIG.8C is a set of flow cytometry plots that shows CAR T cells in the liver, kidney, and the eye tumor site.
  • CD45 staining is shown on the y- axis and CAR-engager labeled with AF647 staining is shown on the x-axis.
  • FIGs.9A –9E are a set of schematics, line plots, and box plots showing that CAR-engager treatment results in enhanced activity and persistence of CAR T cells in vivo.
  • FIG.9A is a line plot that shows circulating half-life of the BCMA CAR-engagers.
  • FIG.9B schematically illustrates the experimental design.
  • FIGs. 9C and 9D are a set of flow cytometry plots and box plots that show selective expansion and persistence of BCMA CAR T cells.
  • FIG. 9E is a box plot that shows the percentage of CD8 + CAR T cells after CAR-engager treatment.
  • FIGSNE stochastic neighbor embedding
  • FIG.10E is a set 6 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 of bar plots that show generation of memory CAR T cells.
  • FIGs. 10F and 10G are a set of flow cytometric plots and bar plots showing that a substantial number of CAR T cells two months post- CAR T cell injection.
  • FIG.10H is a line plot showing that mice maintained consistent body weight throughout the experiment.
  • FIG.10I is a set of flow cytometric plots that show CAR T cells from CAR-engager treated mice have a stem-cell memory phenotype.
  • FIG. 10E is a set 6 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 of bar plots that show generation of memory CAR T cells.
  • FIGs. 10F and 10G are a set of flow cytometric plots and bar plots showing that a substantial number of C
  • FIG. 10J is a set of tSNE plots displaying FlowSOM defined clusters among persisting BCMA CAR T cells.
  • FIGs. 11A – 11E are a set of schematics, photographs, and line, bar, and tSNE plots showing that CAR-engager treatment results in CAR T cell persistence in vivo.
  • FIG. 11A schematically illustrates the experimental design.
  • FIG.11B is a set of photographs that show tumor burden in mice before and after CAR T cell infusion and CAR-engager treatment.
  • FIG.11C is a set of flow cytometric plots showing persistence of CAR T cells.
  • FIG.11D is a set of bar plots showing in vitro killing assays of persistent T cells.
  • FIG.11E is a set of t-SNE plots showing immune cell markers from CD8 + T cells.
  • FIGs. 12A – 12B is a schematic and a set of bar plots showing that CAR-E treatment expands CAR T cells in vivo in the absence of tumor cells.
  • FIG.12A schematically illustrates the experimental design.
  • FIG. 12B is a set of bar plots that show counts of CAR T cells 30 days-post injection.
  • FIGs.13A – 13B are a set of flow cytometry plots showing that neither the BCMA-muIL2 nor the VHH-muIL2 treatment exhibited binding to any specific population within human PBMCs.
  • FIG. 13A is a set of flow cytometry plots showing that BCMA-muIL2 does not bind to human PBMCs.
  • FIG.13B is a set of flow cytometry plots showing that VHH-muIL2 does not bind to human PBMCs.
  • FIGs. 14A – 14C are a set of photomicrographs and dot plots showing specific binding and gradual internalization of the BCMA-muIL2 in CAR T cells.
  • FIG. 14A – 14C are a set of photomicrographs and dot plots showing specific binding and gradual internalization of the BCMA-muIL2 in CAR T cells.
  • FIG. 14A is a set of photomicrographs that show cells stained with CellTracker Blue CMAC, incubated with the indicated treatment, each treatment labeled with Alexa647 (BCMA-muIL2) or dsRed (VHH- muIL2) for 1 to 5 hours and imaged. Photomicrographs are representative of >100 cell images.
  • FIG. 14B is a dot plot that shows quantitative analysis of the imaged cells.
  • FIG. 14C is a dot plot that 7 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 shows the correlation between Alexa647 mean intensity (BCMA-muIL2) and dsRed mean intensity (VHH-muIL2). [00031] FIGs.
  • FIGS. 15A – 15C are a set of flow cytometry plots showing individual flow cytometric data corresponding to the pooled data presented in FIG. 9D.
  • FIG. 15A is a set of flow cytometry results from mice treated solely with CAR T cells.
  • FIG.15B is a set of flow cytometry results from mice treated with CAR T cells and VHH-muIL2.
  • FIG.15C is a set of flow cytometry results from mice treated with CAR T cells and BCMA-muIL2.
  • FIGs. 16A – 16C are a set of flow cytometry plots showing individual flow cytometric data of the mice shown in FIGs.10A –10J.
  • FIG.16A is a set of flow cytometric results from mice treated solely with CAR T cells.
  • FIG.16B is a set of flow cytometry results from mice treated with CAR T cells and VHH-muIL2.
  • FIG.16C is a set of flow cytometry results from mice treated with CAR T cells and BCMA-muIL2.
  • FIGs. 17A – 17C are a set of bar, line, and tSNE plots showing human T cell-derived cytokines in the serum of mice that received OPM2 cancer cells followed by a low dose of CAR T cells.
  • FIG.17A is a bar plot that shows levels of IFN ⁇ , GM-CSF, and TNF ⁇ . Serum samples were diluted at a ratio of 1:40.
  • FIG.17B is a set of line plots that shows IFN ⁇ levels between the BCMA-muIL2 group and the VHH-muIL2 group (error bars represent mean with standard deviation).
  • FIG. 17C is a set of Flt-SNE mapping of CAR T cells derived from the PBS, BCMA-muIL2 and VHH-muIL2 treated mice showing the expression of ten immune cell markers.
  • FIG.18 is a set of t-SNE mapping of CD4 + CAR + T cells derived from the five BCMA- muIL2 CAR-E treated mice showing the expression of nine immune cell markers.
  • FIGs. 19A – 19G are a set of flow cytometry, tSNE, bar, violin, and pie plots and heatmaps showing single-cell RNA sequencing analyses elucidate BCMA-muIL2 effect on CAR T cells.
  • FIG.19A is a set of flow cytometry plots showing CAR + cells analyzed 89 days after CAR-T administration.
  • FIG. 19B is a tSNE plot that shows data after Harmony algorithm, showing proportion of CD4, CD8, and proliferating (CD4 and CD8) cells.
  • FIG.19C is a tSNE plot that shows split between the groups treated with BCMA-muIL2 or VHH-muIL2 treatments.
  • FIG.19D is a set of heatmaps of significantly differentially expressed genes in CD4 + CAR T cells and CD8 + CAT T cells after the indicated treatment.
  • FIG.19E is a set of violin plots of the gene scores between CD8 8 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 and CD4 cells, the scores being constructed using the normalized expression of the different genes for each phenotype in FIG. 19D.
  • FIG. 19F is a set of pie plots that shows the diversity of T-cell receptor (TCR) clonotypes.
  • FIG.19G is a bar plot that shows clonotype diversity within a sample’s total cell count. [00036] FIGs.
  • FIGS. 20A – 20B are a set of schematics and flow cytometry plots showing that CAR- engager treatment results in enhanced organ trafficking of CAR T cells in vivo.
  • FIG. 20A schematically illustrates the experimental design.
  • FIG. 20B is a set of flow cytometry plots that show selective trafficking, expansion, and persistence of BCMA CAR T cells.
  • FIGs.21A – 21C are a set of line and bar plots showing the effects of CAR-engagers on non-transduced T cells and CAR T cells.
  • FIGs.21A and 21B are a line and bar plot, respectively, that together show dose-dependent activation of CAR T cells after CAR-engager treatment (FIG.21A) and that the CAR-engager does not activate non-transduced T cells (FIG. 21B).
  • FIG.21C is a line plot showing that the CD19-muIL2 CAR-engager does not block the killing efficacy of the CD19 CAR T cells or non-transduced T cells (NT T cells).
  • FIGs. 22A – 22B are a set a of line plots showing activation of CAR T cells by CAR- engager variants.
  • FIG. 22A is a line plot showing that CAR-engager variant treatment results in STAT5 phosphorylation.
  • FIG.22B is a line plot showing that CAR-engager variant treatment results in CD69 expression on CAR T cells.
  • FIGs.23A – 23B are a set of line plots showing activation of rested CAR T cells by CAR- engager variants.
  • FIG. 23A is a line plot showing that CAR-engager variant treatment results in STAT5 phosphorylation in rested CAR T cells.
  • FIG.23B is a line plot showing that CAR-engager variant treatment results in CD69 expression on rested CAR T cells.
  • FIGs.24A – 24B are a set of line plots showing the percentage of phosphorylated STAT5 (pSTAT5) positive BCMA CAR T Cells containing FDA-approved CAR constructs induced by BCMA CAR-engagers.
  • FIG. 24A is a line plot that shows pSTAT5 positive BCMA CAR T cells containing the FDA-approved idecabtagene vicleucel (Ide-cel) construct.
  • FIG. 24B is a line plot showing pSTAT5 positive BCMA CAR T cells containing the FDA-approved Ciltacabtagene autoleucel (Cilta-cel) construct.
  • FIGs.25A – 25F are a set of line plots showing the percentage of phosphorylated STAT5 (pSTAT5) positive BCMA CAR T Cells containing FDA-approved CAR constructs induced by BCMA-muIL2 CAR-engagers.
  • FIGs.25A – 25C are a set of line plots that show pSTAT5 positive BCMA CAR T cells containing the FDA-approved Ide-cel construct.
  • FIGs.25D – 25F are a set of line plots that show pSTAT5 positive BCMA CAR T cells containing the FDA-approved Cilta-cel construct. For incubation of 24 hours, CAR T cells were washed at 2 hours to remove unbound any CAR-engager.
  • FIGs. 26A – 26B are a set of line plots showing BCMA CAR-engager dose-dependent activation of rested BCMA CAR T cells through CD69 expression.
  • FIG. 26A is a line plot that shows pSTAT5 positive BCMA CAR T cells containing the FDA-approved Ide-cel construct.
  • FIG. 26B is a line plot showing pSTAT5 positive BCMA CAR T cells containing the FDA-approved cilta-cel construct.
  • CD69 serves as an activation marker for human T cells.
  • FIGs. 27A – 27B are a set of line plots showing BCMA CAR-engager dose-dependent activation of rested BCMA CAR T cells through CD69 expression.
  • FIG. 27A is a line plot that shows pSTAT5 positive BCMA CAR T cells containing the FDA-approved idecabtagene vicleucel construct.
  • FIG. 27B is a line plot showing pSTAT5 positive BCMA CAR T cells containing the FDA-approved ciltacabtagene autoleucel (Cilta-cel) construct.
  • FIGs. 28A – 28B are a set of line plots showing BCMA CAR-engager dose-dependent activation of rested BCMA CAR T cells through CD69 expression.
  • FIG. 28A is a line plot that shows pSTAT5 positive BCMA CAR T cells containing the FDA-approved idecabtagene vicleucel construct.
  • FIG. 28B is a line plot showing pSTAT5 positive BCMA CAR T cells containing the FDA-approved ciltacabtagene autoleucel construct.
  • FIGs.29A – 29C are a set of line plots showing induction of CAR T cell proliferation in vitro from CAR-engager treatment with reduced impact on non-transduced T cells as compared to wild-type IL-2.
  • FIGs. 29A – 29B are a set of line plots that show incubation of activated BCMA CAR T cells with the ciltacabtagene autoleucel CAR construct, with varying concentrations of BCMA CAR-engager treatment, followed by assessment of CAR T cell proliferation using flow cytometry after 3 days.
  • the dashed line represents the number of CAR T cells without any treatment.
  • FIGs.30A – 30C are a set of line plots showing bio-layer interferometry (BLI) associate and dissociation between CAR-engagers containing the triple mutant IL-2 V7 or CAR-engagers containing the double-mutant IL-2 U4 exhibits lower affinity for IL-2Ra as compared to wild-type IL-2.
  • BBI bio-layer interferometry
  • FIG.30A is a line plot that shows BLI between wild-type IL-2 (Teceleukin) and IL-2Ra.
  • FIG. 30B is a line plot that shows BLI between the V7 CAR-engager and IL-2Ra.
  • FIG.30C is a line plot that shows BLI between the CAR-engager containing the U4 CAR-engager and IL-2Ra.
  • FIGs.31A – 31B are a set of schematics and line plots showing that the CAR-engagers containing IL-2 variants expand CAR-T cells in vivo, and that the expansion levels correlate with in vitro pSTAT5 signaling.
  • FIG.31A schematically illustrates the experimental design.
  • FIG.31B is a set of line plots that show CD4 and CD8 CAR T cells circulating in the blood. Mice were bled for 5 weeks on days 11, 18, 28, 35, 42 post injection, and CD4 and CD8 CAR T-cells were counted. All the three tested CAR-engagers expanded CAR T cells in vivo as compared to the control, PBS treatment group. V7 and Y2 CAR-engagers expanded CAR T cells more than X12 CAR-engagers.
  • FIGs.32A – 32C are a set of schematics, photographs, and line plots showing BCMA-V7 CAR-engagers enhances CAR-T cell activity when administered at day 3 or even day 14 post- CAR T-cell injection.
  • FIG. 32A schematically illustrates the experimental design.
  • Another cohort of mice received 6 doses of V7 CAR-engager treatment starting from day 3 post-injection of CAR-T cells, administered on days 3, 6, 10, 14, 21, 28.
  • the third cohort of mice received 6 doses of treatment starting from day 14, which is the post- CRS window in patients, and on days 14, 18, 21, 28, 35, 42.
  • FIG.32B is a set of photographs that shows BLI imaging on the indicated days to assess tumor growth in the different cohorts.
  • FIG.32C is a line plot that shows CAR T cells circulating in the blood. Mice were bled for 6 weeks, once per week starting from day 14, and CAR T cells were counted. V7 CAR-engager treatment expanded CAR T cells in both cohorts that received treatment, compared to the PBS treatment group.
  • FIGs. 33A – 33M are a set of schematics, line, bar, and dot plots showing substantial activation and transcriptomic changes in CAR T cells after CAR-E treatment.
  • FIG. 33A is a line plot that shows CAR-E induction of pSTAT5 activity in CAR T cells with either the full CAR 11 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 construct or the CAR-ICD- ⁇ construct.
  • FIGs.33B – 33D are a set of line plots that show CAR-E CD69 (FIG. 33B), IFN- ⁇ (FIG.33C), and TNF- ⁇ (FIG. 33D) staining in BCMA CAR T cells and BCMA CAR-ICD- ⁇ T cells.
  • FIGs.33E – 33G are a set of line plots that show CD69 (FIG.33E), IFN- ⁇ (FIG.
  • FIG. 33F schematically illustrates the experimental design for in vivo assessment of the efficacy of CAR-E on BCMA CAR T cells and BCMA CAR-ICD- ⁇ T cells.
  • FIG. 33I is a set of bar plots that show expansion and persistence of BCMA CAR T cells and BCMA CAR-ICD- ⁇ T cells in mouse organs 1 month after CAR T cell injection; **** P ⁇ 0.0001.
  • FIG.33J is a volcano Plot that shows the highest upregulated genes in CD8 + CAR T cells 4 hours after CAR- E treatment.
  • FIG. 33K is a volcano plot that shows the highest upregulated genes in CD8 + CAR- ICD- ⁇ T cells 4 hours after CAR-E treatment.
  • FIG. 33L is a heatmap that shows gene expression changes in CD8 + and CD4 + T cells after 4 hours of CAR-E treatment.
  • FIG.33M is a heatmap that shows gene expression changes in CD8 + and CD4 + T cells after 2 and 24 hours of CAR-E treatment.
  • FIG.34 is a set of bar plots showing transcriptome changes in CAR T cells after CAR-E treatment.
  • FIGs.35A – 35G are a set of schematics, photographs, line, and pie graphs showing that lower doses of CAR-engagers enhance CAR T cell activity and promote functional memory.
  • FIG.35A – 35G are a set of schematics, photographs, line, and pie graphs showing that lower doses of CAR-engagers enhance CAR T cell activity and promote functional memory.
  • FIG.35B is a set of photographs that show bioluminescence imaging (BLI) of monitored tumor burdens.
  • FIG. 35C is a survival analyses showing that all CAR-engager treated mice survived for the duration of the experiment.
  • FIG.35D is a line graph that shows the quantification of BLI analyses from FIG.35B.
  • FIG.35E is a line graph that shows flow cytometric analyses of CAR T-cell presence in blood.
  • FIG.35F is a line graph that shows IFN- ⁇ levels.
  • FIG.35G is a set of pie graphs that show the CAR T cells in the bone marrow and spleen of mice treated with CAR-engager.
  • FIGs.36A – 36G are a set of schematics, bar, and line graphs showing treatment of CAR- engager expands CAR T cells in in vivo in the absence of tumor cells in a dose-dependent manner.
  • FIG. 36A schematically illustrates the experimental design.
  • FIGs. 36B – 36C are a set of bar and line graphs showing flow cytometric analysis of spleen and bone marrow tissues harvested 30 days post-injection of CAR T cells.
  • FIG. 36D – 36E are a set of bar graphs that show analyses of persisting CAR T cells and different subsets of memory CAR T cells in the spleen and bone marrow 12 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 of the CAR-E-treated mice.
  • FIG. 36F is a bar graph showing that both the antigen and the low- affinity IL-2 components of CAR-engager are essential for its impact.
  • FIG. 37 a set of flow cytometry plots showing anti-human-CD45 and BCMA-CAR staining in blood samples of mice that received human CAR T cells with and without CAR-E treatment.
  • FIGS. 39A – 39D are a set of heatmaps and tSNE plots showing CAR-E promotion of phyotypic diversity of bone marrow and splenocyte-derived CAR T cells.
  • FIG. 39A – 39D are a set of heatmaps and tSNE plots showing CAR-E promotion of phyotypic diversity of bone marrow and splenocyte-derived CAR T cells.
  • FIG. 39A is a heatmap that shows eight FLOWSOM-derived metaclusters in bone marrow samples.
  • FIG.39B is a heatmap that shows eight FLOWSOM-derived metaclusters in spleen samples.
  • FIG.39C is a tSNE plot that shows CAR T populations in bone marrow samples.
  • FIG. 39D is a tSNE plot that shows CAR T populations in spleen samples.
  • FIGs. 40A – 40D are a set of flow cytometry plots showing anti-human-CD45 and BCMA-CAR staining in individual mice that received human CAR T cells and CAR-E treatment.
  • FIG.40A is a set of flow cytometry plots of mice that received CAR T cells and PBS control.
  • FIG. 40B is a set of flow cytometry plots of mice that received CAR T cells and 2 mg/kg BCMA-muIL2.
  • FIG.40C is a set of flow cytometry plots of mice that received CAR T cells and 4 mg/kg BCMA- muIL2.
  • FIG.40D is a set of flow cytometry plots of mice that received CAR T cells and 8 mg/kg BCMA-muIL2.
  • FIGs. 41A – 41E are a set of flow cytometry plots showing anti-human-CD45 and BCMA-CAR staining in individual mice that received human CAR T cells and CAR-E treatment.
  • FIG. 41A is a set of flow cytometry plots of mice that received CAR T cells and BCMA-CH3 control.
  • FIG.41B is a set of flow cytometry plots of mice that received CAR T cells and low dose IL-2.
  • FIG.41C is a set of flow cytometry plots of mice that received CAR T cells and VHH-muIL2.
  • FIG. 41D is a set of flow cytometry plots of mice that received CAR T cells and BCMA-muIL2.
  • FIG.41E is a set of flow cytometry plots of mice that received CAR T cells only.
  • FIG.42 is a set of line plots showing CAR-engagers each containing an immune cell effector domain containing an U4, V6, V7, or Y2 IL-2 variant and their selective and potent induction of proliferation in CAR T cells but not non-transduced T cells.
  • FIG.43 is a set of line plots showing phosphorylation of STAT5 in CAR T cells and non- transduced T cells following CAR-E treatment.
  • FIG. 44 is a set of line plots showing CD69 expression on CAR T cells and non- transduced T cells following CAR-E treatment.
  • FIG.45 is a set of line plots showing TNF- ⁇ and IFN- ⁇ secretion from CAR T cells and non-transduced T cells following CAR-E treatment.
  • FIG.46A – 46B are a set of line plots showing CAR-E binding to IL-2R ⁇ and IL-2R ⁇ by Biolayer Interferometry. DETAILED DESCRIPTION OF THE DISCLOSURE [00063] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which the subject matter herein belongs. As used in the specification and the appended claims, unless specified to the contrary, the following terms have the meaning indicated to facilitate the understanding of the present disclosure.
  • transitional term “comprising,” which is synonymous with “including,” “containing,” or “characterized by,” is inclusive or open-ended and does not exclude additional, unrecited elements or method steps.
  • the transitional phrase “consisting of” excludes any element or method step not specified in the claim (or the specific element or method step with which the phrase “consisting of” is associated).
  • the transitional phrase “consisting essentially of” limits the scope of a claim to the specified elements and method or steps and “unrecited elements and method steps that do not materially affect the basic and novel characteristic(s)” of the claimed disclosure.
  • the disclosure provides an IL-2 variant, which differs from wild type IL-2 (SEQ ID NO: 102) in terms of from three to eight amino acid substitutions selected from amino acid residues H16, D20, R38, F42, Y45, E61, E62, L72, and V91 of SEQ ID NO: 102.
  • the three to eight amino acid substitutions are selected from H16A, H16R, H16S, D20A, D20Q, R38D, F42A, Y45A, E61A, E62N, L72G, and V91H.
  • IL-2 variant refers to non-naturally occurring variant of IL-2 capable of binding to the cognate IL-2 receptor(s) on an immune cell and initiating signal transduction through that receptor to achieve substantially the same effect as the naturally occurring cytokine.
  • the amino acid sequence of wild-type IL-2 is set forth below (SEQ ID NO: 102): 1 aptssstkkt qlqlehllld lqmilnginn yknpkltrml tfkfympkka telkhlqcle 61 eelkpleevl nlaqsknfhl rprdlisnin vivlelkgse ttfmceyade tativeflnr 121 witfcqsiis tlt [00070] All descriptions of the IL-2 variants of the present disclosure are made in reference to SEQ ID NO:102.
  • the IL-2 variant includes a first amino acid substitution (of the three to eight amino acid substitutions), from N-terminus to C-terminus, is selected from H16A, H16R, H16S, D20A, or D20Q, a second amino acid substitution is selected from R38D, F42A, and V91H, and a third amino acid substitution is selected from Y45A and E62N. [00071] In some embodiments, the IL-2 variant includes a first amino acid substitution of H16A, H16R, or H16S. In other embodiments, the IL-2 variant includes a first amino acid substitution of D20A or D20Q.
  • the IL-2 variant includes a second amino acid substitution of F42A.
  • the IL-2 variant includes a first amino acid substitution of H16A, H16R, or H16S and a second amino acid substitution of F42A.
  • the IL-2 variant includes a first amino acid substitution of D20A or D20Q and a second amino acid substitution of F42A.
  • the IL-2 variant includes a third amino acid substitution of Y45A. In other embodiments, the third amino acid substitution is E62N.
  • the IL-2 variant includes a first amino acid substitution of H16A, H16R, or H16S, a second amino acid substitution of F42A, and a third amino acid substitution of Y45A.
  • the IL-2 variant includes a first amino acid substitution of D20A or D20Q, a second amino acid substitution of F42A, and a third amino acid substitution of Y45A.
  • the IL-2 variant includes a first amino acid substitution of H16A, H16R, or H16S, a second amino acid substitution of F42A, and a third amino acid substitution of Y45A.
  • the IL-2 variant includes a first amino acid substitution of D20A or D20Q, a second amino acid substitution of F42A, and a third amino acid substitution of Y45A. [00076] In some embodiments, the IL-2 variant includes a fourth amino acid substitution selected from R38D, E61A, L72G, and V91H. [00077] In some embodiments, the IL-2 variant includes a first amino acid substitution of H16A, H16R, or H16S, a second amino acid substitution of F42A, a third amino acid substitution of Y45A or E62N, and a fourth amino acid substitution selected from D20A, D20Q, R38D, Y45A, E61A, L72G, and V91H.
  • the IL-2 variant includes a first amino acid substitution of H16A, H16R, or H16S, a second amino acid substitution of F42A, a third amino acid substitution of Y45A or E62N, a fourth amino acid substitution selected from D20A, D20Q, E61A, L72G, and V91H, and a fifth amino acid substitution of R38D.
  • the IL-2 variant includes a first amino acid substitution of H16A, H16R, or H16S, a second amino acid substitution of F42A, a third amino acid substitution of Y45A or E62N, a fourth amino acid substitution selected from D20A, D20Q, E61A, and V91H, a fifth amino acid substitution of R38D, and a sixth amino acid substitution of L72G.
  • the IL-2 variant includes a first amino acid substitution of H16A, H16R, or H16S, a second amino acid substitution of F42A, a third amino acid substitution of Y45A, 16 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 a fourth amino acid substitution selected from D20A, D20Q, E61A, and V91H, a fifth amino acid substitution of R38D, a sixth amino acid substitution of L72G, and a seventh amino acid substitution of E62N.
  • the IL-2 variant has amino acid substitutions D20Q, F42A, and Y45A, and has the amino acid sequence set forth below (SEQ ID NO: 112). Amino acid substitutions relative to wild-type IL-2 for SEQ ID NOs: 112-123 are boxed in the sequences below.
  • the IL-2 variant has amino acid substitutions H16A, F42A, and E62N, and has the amino acid sequence set forth below (SEQ ID NO: 113): 1 aptssstkkt qlqleallld lqmilnginn yknpkltrml takfympkka telkhlqcle 61 enlkpleevl nlaqsknfhl rprdlisnin vivlelkgse
  • CAR-engagers may also reduce the cellular dose needed for CAR immune cell therapy, which may result in reduced adverse side effects (e.g., cytokine release syndrome) caused by the larger doses typically used in the clinic and therefore, the CAR-engagers are also referred herein as CAR-enhancers or CAR-E.
  • CAR-enhancers CAR-E.
  • CAR-engager binding to CAR is reversible, the CAR engager does not induce immune synapse formation of CAR on the CAR immune cell surface. Therefore, CAR- engagers do not block CAR-mediated killing of cancer cells.
  • CAR immune cells often do not persist in the body during minimal residual disease (MRD), which, as known in the art, is associated with limited cancer antigens.
  • MRD minimal residual disease
  • the disclosure provides a CAR-E which contains a first proteinaceous moiety and a second proteinaceous moiety.
  • the first proteinaceous moiety binds an epitope on the extracellular domain (ED) of a CAR.
  • the first proteinaceous moiety binds an epitope present on the extracellular binding domain (EBD) of the CAR that binds a cancer antigen on the surface of a cancer cell.
  • the second proteinaceous moiety which is an immune effector domain, comprises an IL-2 variant of the present disclosure.
  • CAR-engager is a contiguous protein, where the first proteinaceous entity and the proteinaceous entity are connected by a peptide bond. In some embodiments, the first proteinaceous entity and the second proteinaceous entity are connected by click chemistry. [00096] In some embodiments, the CAR-engager is formulated and administered as a monomeric protein or proteinaceous entity. In other embodiments, the CAR-engager is formulated and administered in the form of a dimer, either as a homodimer or a heterodimer protein or proteinaceous entity.
  • the first proteinaceous moiety of the CAR-engager is designed to bind an epitope present on the extracellular binding domain of the CAR that targets an antigen on the surface of a cancer cell.
  • the first moiety of the CAR-engager is an ectodomain that binds an EBD of a CAR presented on an immune cell.
  • the ectodomain of a cancer antigen is the portion of the antigen on the surface of a cancer cell that binds a T cell receptor or a CAR on 19 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 an immune cell.
  • the CAR-engager may include the entire extracellular domain of a cancer antigen.
  • Ectodomains may be derived from (e.g., identified in) a cancer antigen in accordance with standard techniques. See, e.g., and Gershoni et al., Biodrugs 21(3):145-156 (2007) and Francino-Urdaniz and Whitehead, RSC Chem. Biol. 2(6):1580-1589 (2021).
  • the term “derived from” as used herein when referring to a protein and nucleic acid refers to a sequence that originates and is identified from the sequence of a parent (e.g., wild-type or endogenous) protein and nucleic acid, respectively.
  • a sequence derived from a parent sequence may be a portion (fragment) of the parent sequence and/or may vary by at least one position from the parent amino acid or nucleotide sequence.
  • Protein variants may include amino acid substitutions, insertions, and/or deletions.
  • an amino acid sequence derived from a parent sequence may constitute a fragment of the parent sequence and be identical for a specific range of amino acids of the parent but does not include amino acids outside that specific region.
  • Nucleic acid variants may include substitutions or in-frame insertions or deletions (i.e., insertions or deletions that do not result in downstream frame shift of the nucleic acid codons).
  • Unique cancer antigens may be determined by known methods. For example, cancer genomes can be compared with normal cell genomes to identify neoantigens. In some embodiments, caner transcriptomes are compared to normal cell transcriptomes. Computational methods may then be utilized to identify suitable binding sites for a CAR. Most often the CAR binds a portion of the extracellular domain of an antigen. In some embodiments, the CAR and the corresponding cancer antigen are known in the art. [000100] In some embodiments, the ectodomain of the CAR-engager contains the entire extracellular domain of a cancer antigen.
  • the CAR-engager contains a portion of the extracellular domain of a cancer antigen which is targeted by a CAR.
  • the ectodomain of the CAR- engager contains the extracellular domain of BCMA.
  • the amino acid sequence of a representative CAR-engager that contains a BCMA extracellular domain is MLQMAGQCSQNEYFDSLLHACIPCQLRCSSNTPPLTCQRYCNASVTNSVKGTNA (SEQ ID NO: 1).
  • the ectodomain of the CAR-engager contains two repetitions of the extracellular domain of BCMA.
  • the amino acid sequence of a representative CAR-engager that contains two repetitions of the BCMA extracellular domain is set forth below (SEQ ID NO: 2): 1 mlqmagqcsq neyfdsllha cipcqlrcss ntppltcqry cnasvtnsvk gtnagggsgg 61 gsprgsgggs mlqmagqcsq neyfdsllha cipcqlrcss ntppltcqry cnasvtnsvk 121 gtn [000103]
  • the ectodomain of the CAR- engager contains a variant of the extracellular domain of CD19.
  • amino acid sequence of a representative CAR-engager that contains a variant of CD19 extracellular domain is set forth below (SEQ ID NO: 3): 1 peeplvvkve egdeawlpcl kgtsdgptqq ltwsresplk pflkvsfgvp glgvhvrpna 61 vslvisnvsq qmggfylcqp gppsekawqp gwtvnvegsg elfrwnvsdl gglgcglknr 121 ssegpsspsg klmspklyvw akdrpeiweg eppclpprds lnqslsrdmt vapgstlwls 181 cgvppdsvsr gplswthvhp kgpksllsle lkddrpardm wvt
  • the ectodomain of the CAR-engager is KDRPEIWEGEPP (SEQ ID NO: 106), which corresponds to amino acid residues 142-153 of SEQ ID NO: 5.
  • the ectodomain of the CAR- engager contains at least a portion of the extracellular domain of CD20.
  • the amino acid sequence of a representative CAR-engager that contains a CD20 extracellular domain is KISHFLKMESLNFIRAHTPYINIYNCEPANPSEKNSPSTQYCYSIQS (SEQ ID NO: 6).
  • the ectodomain of the CAR- engager contains at least a portion of the extracellular domain of CD22.
  • the amino acid sequence of a representative CAR-engager that contains a CD22 extracellular domain is set forth below (SEQ ID NO: 7): 1 dsskwvfehp etlyawegac vwipctyral dgdlesfilf hnpeynknts kfdgtrlyes 61 tkdgkvpseq krvqflgdkn knctlsihpv hlndsgqlgl rmesktekwm erihlnvser 121 pfpphiqlpp eiqesqevtl tcllnfscyg ypiqlqwlle gvpmrqaavt stsltik
  • the ectodomain of the CAR- engager contains the Ig domains 2 and 3 (2-3) of CD22.
  • the amino acid sequence of a representative CAR-engager that contains Ig domains 2-3 of CD22 is set forth below (SEQ ID NO: 103): 1 phiqlppeiq esqevtltcl lnfscygypi qlqwllegvp mrqaavtsts ltiksvftrs 61 elkfspqwsh hgkivtcqlq dadgkflsnd tvqpkleikv tpsdaivreg dsvtmtcevs 121 ssnpeyttvs wlkdgtslkk qntftlnlre vtkdqsgkyc cqvsndvgpg rseevflq [000111] In some embodiments,
  • the amino acid sequence of a representative CAR-engager that contains Ig domain 3 of CD22 is set forth below (SEQ ID NO: 104): 1 pkleikvtps daivregdsv tmtcevsssn peyttvswlk dgtslkkqnt ftlnlrevtk 61 dqsgkyccqv sndvgpgrse evflq [000112]
  • the ectodomain of the CAR-engager contains the Ig domains 5 through 7 (5-7) of CD22.
  • amino acid sequence of a representative CAR-engager that contains Ig domains 5-7 of CD22 is set forth below (SEQ ID NO: 105): 1 pkkvttviqn pmpiregdtv tlscnynssn psvtryewkp hgaweepslg vlkiqnvgwd 61 nttiacaacn swcswaspva lnprdvrvrk ikplseihsg nsvslqcdfs sshpkevqff 121 wekngrllgk esqlnfdsis pedagsyscw vnnsigqtas prrlrvsmsp gdqvmegksa 181 tltcesdanp pvshytwfdw nnqslpyhsq klrlepvkvq
  • the amino acid sequence of a representative CAR-engager that contains a Claudin 18.2 first extracellular domain is set forth below (SEQ ID NO: 8): 1 dqwstqdlyn npvtavfnyq glwrscvres sgftecrgyf tllglpamlq avr [000114]
  • the amino acid sequence of a representative CAR-engager that contains a Claudin 18.2 second extracellular domain is set forth below (SEQ ID NO: 9): 1 vtnfwmstan mytgmggmvq tvqtrytfga a [000115]
  • the ectodomain of the CAR- engager contains at least a portion of the extracellular domain of SLAMF7.
  • the amino acid sequence of a representative CAR-engager that contains a SLAMF7 extracellular domain is set forth below (SEQ ID NO: 10): 1 sgpvkelvgs vggavtfplk skvkqvdsiv wtfnttplvt iqpeggtiiv tqnrnrervd 61 fpdggyslkl sklkkndsgi yyvgiysssl qqpstqeyvl hvyehlskpk vtmglqsnkn 121 gtcvtnltcc mehgeedviy twkalgqaan eshngsilpi swrwgesdmt ficvarnpvs 181 rnfsspilar klcegaaddp dssm [000116]
  • the CAR targets PD-1 the CAR targets
  • a representative CAR-engager that contains a PD-1 extracellular domain is set forth below (SEQ ID NO: 11): 1 fldspdrpwn pptfspallv vtegdnatft csfsntsesf vlnwyrmsps nqtdklaafp 61 edrsqpgqdc rfrvtqlpng rdfhmsvvra rrndsgtylc gaislapkaq ikeslraelr 121 vterraevpt ahpspsprpa gqfqtlv [000117]
  • the ectodomain of the CAR-engager contains a variant of the extracellular domain of PD-1.
  • the ectodomain of the CAR-engager contains the N-loop of PD-1.
  • the amino acid sequence of a representative CAR-engager that contains the N- loop of the PD-1 extracellular domain is LDSPDRPWNP (SEQ ID NO: 107), which corresponds to amino acid residues 2-11 of SEQ ID NO: 11.
  • the ectodomain of the CAR-engager contains the CD-loop of PD- 1.
  • the amino acid sequence of a representative CAR-engager that contains the CD-loop of the PD- 1 extracellular domain is NQTDKLAAFPEDRSQPGQDCRFRVTQ (SEQ ID NO: 108), which corresponds to amino acid residues 51-76 of SEQ ID NO: 11.
  • the ectodomain of the CAR-engager contains at least a portion of the extracellular domain of KIT.
  • amino acid sequence of a representative CAR-engager that contains a KIT extracellular domain is set forth below (SEQ ID NO: 12): 1 qpsvspgeps ppsihpgksdassivgdeir llctdpgfvk wtfeildetn enkqnewite 61 kaeatntgky tctnkhglsn siyvfvrdpa klflvdrsly gkedndtlvr cpltdpevtn 121 yslkgcqgkp lpkdlrfipd pkagimiksv krayhrlclh csvdqegksv lsekfilkvr 181 pafkavpvvs vskasyllre geeftvtcti kdvssvyst wkrensqtkl qekyn
  • amino acid sequence of a representative CAR-engager that contains a TROP2 extracellular domain is set forth below (SEQ ID NO: 13): 1 htaaqdnctc ptnkmtvcsp dgpggrcqcr algsgmavdc stltskclll karmsapkna 61 rtlvrpseha lvdndglydp dcdpegrfka rqcnqtsvcw cvnsvgvrrt dkgdlslrcd 121 elvrthhili dlrhrptaga fnhsdldael rrlfreryrl hpkfvaavhy eqptiqielr 181 qntsqkaagd vdigdaayyf erdikgeslf qgrggldlr
  • the amino acid sequence 31 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 of a representative CAR-engager that contains a CD38 extracellular domain is set forth below (SEQ ID NO: 14): 1 vprwrqqwsg pgttkrfpet vlarcvkyte ihpemrhvdc qsvwdafkga fiskhpcnit 61 eedyqplmkl gtqtvpcnki llwsrikdla hqftqvqrdm ftledtllgy laddltwcge 121 fntskinyqs cpdwrkdcsn npvsvfwktv srrfaeaacd vvhvmlngsr skifdknstf 181 gsvevhnlqp
  • the amino acid sequence of a representative CAR-engager that contains MSLN is set forth below (SEQ ID NO: 15): 1 malptarpll gscgtpalgs llfllfslgw vqpsrtlage tgqeaapldg vlanppniss 61 lsprqllgfp caevsglste rvrelavala qknvklsteq lrclahrlse ppedldalpl 121 dlllflnpda fsgpqactrf fsritkanvd llprgaperq rllpaalacw gvrgsllsea 181 dvralgglac dlpgrfvaes aevllprlvs cpgpldqdqq eaaraalqgg gppygppstw 241 svstmdalrg ll
  • the ectodomain of the CAR-engager contains a portion of the MSLN protein.
  • the ectodomain of the CAR-engager is IPXGYLVLDLSMQEALS (SEQ ID NO: 17), where X is any amino acid.
  • the ectodomain of the CAR-engager is YXVXDLSMQEL (SEQ ID NO: 18), where X is any amino acid.
  • CAR-E first moiety CAR-binding antibodies and derivatives thereof [000124]
  • the first moiety of the CAR-engager may be an antibody that binds an epitope on the ED of the CAR, or an ED-binding derivative thereof.
  • Antibody derivatives include antibody fragments (e.g., a scFv and nanobody fragments).
  • the first moiety of the CAR- engager is an anti-anti-CD19 antibody binding moiety that binds an epitope on a CAR the EBD of which binds CD19.
  • a representative anti-anti-CD19 binding moiety is set forth below (SEQ ID NO: 124): 32 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 1 qvqlqqpgae lvrpgasvkl scktsgysft rywmnwvkqr pgqglewigm ihpsdsetrl 61 nqkfkdkatl tvdnssstay mqlssptsed savyycasiy yeeawgqgtl vtvsaggggs 121 ggggsggggs diqmtqspas lsasvgetvt itcrasgnih nylawyqqkq gkspqllvyn aktladsvps rfsgsgsgtq yslkinslqp edfgsyy
  • the first moiety of the CAR-engager may be an anti-(G4S) binding moiety that binds an epitope on a CAR the linker of which has at least two repeats of GGGGS (SEQ ID NO: 71).
  • a representative heavy chain variable region (VH) of an anti-(G4S) binding moiety is set forth below (SEQ ID NO: 125): 1 qsvkesggrl vtpgtpltlt ctvsgfslss naidwvrqap gkglewigil grsgstyyas 61 wakgrftisr tssttvdlki tspttedtat yfcarhfylw gpgtlvtvss [000127]
  • a representative light chain variable region (VL) of an anti-(G4S) binding moiety is set forth below (SEQ ID NO: 126): 1 aqvltqtasp vsaavggtvt incqasqsvy snylswyqqk pgqppkllma ttstlepgvp 61 srfkgsgsgt
  • a representative anti- ⁇ light chain binding moiety is set forth below (SEQ ID NO: 110).
  • Anti-anti-mouse binding moieties are known in the art, 33 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 see, Kochenderfer et al., J. Immunother. 32(7):689-702 (2009) and Cheng et al., Cytometry A. 103(1):16-26 (2023).
  • Additional antibodies and derivatives thereof that bind CAR EDs that may be useful are known in the art, see, e.g., U.S. Patent 9,701,758 and U.S. Patent Application Publication 2005/0287148, both of which are incorporated herein by reference in their entireties.
  • the second moiety is an immune cell effector domain that comprises an IL-2 variant as described herein.
  • the IL-2 variant binds the one or more of the cognate receptor subunits of IL-2R ⁇ , IL-2R ⁇ , and IL-2R ⁇ on the immune cell (containing the CAR). This binding event modulates the activity of the CAR-immune cell.
  • modulate(s),” and “modulation” as used herein embrace both activation and inhibition of the CAR immune cell.
  • the CAR-engager contains a plurality (i.e., two, three, or more) of immune cell effector domains, wherein at least one of which is an IL-2 variant described herein, and wherein any two or more of immune effector domains may be the same as or different from each other.
  • the CAR-engager contains two immune effector domains containing two IL-2 variants as disclosed herein. In some embodiments, the CAR-engager contains three immune effector domains containing three IL-2 variants as disclosed herein.
  • the second immune cell effector domain is a cytokine, an immune cell-activating moiety, or an immune cell-inhibiting moiety, and variants and fragments thereof, that bind to their cognate targets on the immune cells.
  • cytokine includes low molecular weight extracellular polypeptides/glycoproteins that promote, modulate, and regulate the immune response (i.e., increase or decrease activity, differentiation, or proliferation).
  • Representative examples of cytokines include chemokines, interferons (IFNs), interleukins (ILs), lymphokines and tumor necrosis factors (TNFs).
  • immune cell activating variant of a cytokine as used herein refers to non-naturally occurring variant of a cytokine capable of binding to a cytokine receptor on an immune cell and initiating signal transduction through that receptor to achieve substantially the same effect as the naturally occurring cytokine.
  • the second immune cell effector domain is an immune cell activating moiety, e.g., immune cell activating cytokines and immune cell-activating variants and 34 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 fragments thereof.
  • Immune cell activating moieties activate, promote, or maintain the activity of immune cells.
  • the second immune cell effector domain is derived from CD40, CD48, CD58, CD70, CD80, CD86, CD112, glucocorticoid-induced TNFR-related protein ligand (GITRL; TNFSF18), herpesvirus entry mediator (HVEM; TNFSF14), Semaphorin 3B (SEMAA; SEMA3B), Signaling lymphocytic activation molecule family member 1 (SLAM; SLAMF1; CD150), T cell immunoglobulin and mucin domain containing 4 (TIM4), TNF superfamily member 4 (TNFSF4; OX40L), TNF superfamily member 8 (TNFSF8; CD30L), interleukin-2 (IL-2), IL-7, IL-9, IL-10, IL-12, IL-15, IL-18, IL-21, IL-27, CCL21, 4-1BBL (also known as TNF superfamily member 9; TNFSF9), or an immune cell-activating variant
  • GITRL TNFSF18
  • the amino acid sequences of representative immune cell activating moieties are from which the second immune cell effector domain may be derived are provided at the NCBI Accession numbers set forth in Table 2, and are incorporated herein by reference.
  • Table 2 Gene Name, Symbols, and NCBI Accession Numbers of Representative Immune Cell- Activating Proteins Gene Name Gene Symbols Protein Accession No(s).
  • the immune cell effector domain contains the weak affinity variant of IL-2 (muIL2), having the amino acid sequence set forth below (SEQ ID NO: 19): 1 aptssstkkt qlqleallld lqmilnginn yknpkltrml takfympkka telkhlqcle 61 eelkpleevl nlaqsknfhl rprdlisnin vivlelkgse ttfmceyade tativeflnr 121 witfcqsiis tlt [000138]
  • the second immune cell effector domain is an IL-2 variant that has an H16A substitution (i.e., an alanine (A) at amino acid residue 16 in place of the histidine (H)) and
  • the second immune cell effector domain contains at least a portion of the weak affinity IL-2 variant, having together, the amino acid sequence set forth below (SEQ ID NO: 20): 1 aptssstkkt qlqleallld lqmilnginn yknpkltrml takfympkka telkhlqcle 61 eelkpleevl nlaqsknfhl rprdlisnin vivlelkgse ttfmceyade tativeflnr 121 witfcqsiis tltggggsgg ggsggggggsgg ggsaptsst kktqlqleal lldlqmilng 181 innyknpklt rmltakfymp kkatelkhlq cleeelkple evlnlaqskn fhlrpr
  • the second immune cell effector domain contains at least a portion of the wild-type IL-2, having the amino acid sequence set forth below (SEQ ID NO: 102): 1 aptssstkkt qlqlehllld lqmilnginn yknpkltrml tfkfympkka telkhlqcle 61 eelkpleevl nlaqsknfhl rprdlisnin vivlelkgse ttfmceyade tativeflnr 121 witfcqsiis tlt [000141]
  • the muIL2 (SEQ ID NO: 19) second immune cell effector domain has a dissociation constant (KD) of about 1200 nM for IL-2R ⁇ (CD25), representing a 110-fold decrease as compared to wild type
  • the second immune cell effector contains at least a portion of IL- 7.
  • the amino acid sequence of a representative IL-7 is set forth below (SEQ ID NO: 21): 1 mfhvsfryif glpplilvll pvassdcdie gkdgkqyesv lmvsidqlld smkeigsncl 61 nnefnffkrh icdankegmf lfraarklrq flkmnstgdf dlhllkvseg ttillnctgq 121 vkgrkpaalg eaqptkslee nkslkeqkkl ndlcflkrll qeiktcwnki lmgtkeh [000143]
  • the second immune cell effector domain contains at least a portion of IL-18.
  • the second immune cell effector domain contains at least a portion of IL-21.
  • the second immune cell effector domain contains at least a portion of IL-27.
  • amino acid sequence of a representative IL-27 is set forth below (SEQ ID NO: 25): 1 mgqtagdlgw rlsllllpll lvqagvwgfp rppgrpqlsl qelrreftvs lhlarkllse 61 vrgqahrfae shlpgvnlyl lplgeqlpdv sltfqawrrl sdperlcfis ttlqpfhall 121 gglgtqgrwt nmermqlwam rldlrdlqrh lrfqvlaagf nlpeeeeeee eeeeerkgl 181 lpgalgsalq gpaqvswpql lstyrllhsl elvlsravre llllskaghs vwplgfptls
  • the second immune cell effector domain contains two repeats of Neo-2/15, having the amino acid sequence of SEQ ID NO: 26. [000149] In some embodiments, the second immune cell effector domain contains at least a portion of 4-1BBL. 4-1BBL is also known as TNF ligand superfamily member 9 (TNFSF9).
  • the amino acid sequence of a representative 4-1BBL is provided at NCBI Accession No. NP_003802, incorporated herein by reference.
  • the second immune cell effector domain 38 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 contains at least a portion of the extracellular domain of 4-1BBL.
  • the second immune cell effector domain contains a portion of the extracellular domain of 4-1BBL, having the amino acid sequence set forth below (SEQ ID NO: 27): 1 dpaglldlrq gmfaqlvaqn vllidgplsw ysdpglagvs ltgglsyked tkelvvakag 61 vyyvffqlel rrvvagegsg svslalhlqp lrsaagaaal altvdlppas searnsafgf 121 qgrllhlsag qrlgvhlhte ararhawqlt qgatvlglfr vtpeipa [000150]
  • the CAR-engager contains three immune cell effector domains, e.g., wherein the first immune cell effector domain is an IL-2 variant and the second and the third immune cell effector domains are the extra
  • the second immune cell effector domain may be a fragment, e.g., a single-chain variable antibody fragment (scFv), that binds and activates the CAR immune cell.
  • the second immune cell effector domain is an scFv that binds an epitope on 4- 1BB, CD2, CD27, CD28, CD30 (TNFRSF8), CD40L, CD226, CTLA4, GITR, IL-2R, LIGHT, OX40, PD-1, TIM2, SLAM, or TIM1.
  • the second immune cell effector domain is derived from a commercially available anti-CTLA4 antibody, antibody fragment, or derivative thereof, e.g., bavunalimab (formerly pavunalimab/XmAb 22841), botensilimab, cadonilimab, ipilimumab (Yervoy®), quavonlimab, tremelimumab (Imjudo®), volrustomig, vudalimab, or zalifrelimab.
  • the second immune cell effector domain is a scFv that binds CTLA4.
  • the amino acid sequences of representative heavy and light chains of antibodies that bind CTLA4 are set forth in Table 3.
  • Table 3 Amino acid Sequences of Representative anti-CTLA Antibody Heavy and Light Chains botensilimab heavy chain (SEQ ID NO: 28) 1 evqlvesggg lvkpggslrl scaasgftfs sysmnwvrqa pgkglewvss isssssyiyy s l d t t q s p t Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 1 qvqlvesggg vvqpgrslrl scaasgftfs sytmhwvrqa pgkglewvtf isydgnnkyy 61 adsvkgrfti srdnskntly lqmnslraed taiyycartg wlgpfdywgq gt
  • the second immune cell effector domain is derived from a commercially available anti-OX40 antibody, antibody fragment (e.g., scFv), or derivative thereof, e.g., tavolimab, or vonlerolizumab (Pogalizumab; MOXR 0916).
  • scFv antibody fragment
  • tavolimab tavolimab
  • vonlerolizumab Pogalizumab; MOXR 0916.
  • Table 4 Amino acid Sequences of Representative anti-OX40 Antibody Heavy and Light Chains tavolimab heavy chain (SEQ ID NO: 38) 1 qvqlqesgpg lvkpsqtlsl tcavyggsfs sgywnwirkh pgkgleyigy isyngityhn s s g y e r s p t y t y v y k g s p t [ 000 56] n some embod ments, t e mmune ce e ector doma n conta ns t e V av ng t e amino acid sequence set forth below (SEQ ID NO 42): 1 diqmtqspss lsasvgdrvt itcrasqdis nylnwyqqkp gkapklliyy tsrlrsgvps 61 r
  • the second immune cell effector domain is derived from a commercially available anti-PD-1 antibody, antibody fragment (e.g., scFv), or derivative thereof, e.g., atezolizumab, avelumab, bintrafusp alfa, cosibelimab, danburstotug, durvalumab (Imfinzi®), inbakicept, lodapolimab, pimivalimab, or socazolimab.
  • Table 5 Amino Acid Sequences of Representative anti-PD-1 Antibody Heavy and Light Chains atezolizumab heavy chain (SEQ ID NO: 44) 1 evqllesggg lvqpggslrl scaasgftfs syimmwvrqa pgkglewvss iypsggitfy v t s 42 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 121 psdeqlksgt asvvcllnnf ypreakvqwk vdnalqsgns qesvteqdsk dstyslsstl 181 tlskadyekh kvyacevthq glsspvtksf nrgec pimivalimab heavy chain (SEQ ID NO: 50) [ ving the amino acid sequence set forth below
  • Immune cell inhibiting moieties repress or block immune cell activity and function.
  • the immune cell-inhibiting moiety may be derived from CD80, CD86, CD112, CD155, CD276 (B7-H3), Ceacam-1, FGL1, galectin- 3, HLA-E, HVEM, PD-L1, PD-L2, VISTA, or VTCN1 (B7-H4).
  • the amino acid sequences of representative immune cell-inhibiting proteins from which the second immune cell effector domain may be derived are provided at the NCBI Accession numbers set forth in Table 6, and are incorporated herein by reference.
  • the second immune cell effector domain contains at least a portion of the extracellular domain of CD86.
  • amino acid sequence of a representative CD155 extracellular domain is set forth below (SEQ ID NO: 56): 1 wpppgtgdvv vqaptqvpgf lgdsvtlpcy lqvpnmevth vsqltwarhg esgsmavfhq 61 tqgpsysesk rlefvaarlg aelrnaslrm fglrvedegn ytclfvtfpq gsrsvdiwlr 121 vlakpqntae vqkvqltgep vpmarcvstg grppaqitwh sdlggmpnts qvpgflsgtv 181 tvtslwilvp ssqvdgknvt ckvehesfek pqlltvnltv yyppevsisg ydnnw
  • the amino acid sequence of a representative galecin-3 extracellular domain is set forth below (SEQ ID NO: 60): 1 madnfslhda lsgsgnpnpq gwpgawgnqp agaggypgas ypgaypgqap pgaypgqapp 61 gaypgapgay pgapapgvyp gppsgpgayp ssgqpsatga ypatgpygap agplivpynl 121 plpggvvprm litilgtvkp nanrialdfq rgndvafhfn prfnennrrv ivcntkldnn 181 wgreerqsvf pfesgkpfki qvlvepdhfk vavndahllq ynhrvkklne isklgisgdi 241 dl
  • the second immune cell effector domain contains at least a portion of the extracellular domain of nectin-2 (CD112, HVEB).
  • amino acid sequence of a representative nectin-2 extracellular domain is set forth below (SEQ ID NO: 63): 1 qdvrvqvlpe vrgqlggtve lpchllppvp glyislvtwq rpdapanhqn vaafhpkmgp 61 sfpspkpgse rlsfvsakqs tgqdteaelq datlalhglt fürgnytce fatfpkgsvr 121 gmtwlrviak pknqaeaqkv tfsqdpttva lciskegrpp ariswlssld weaketqvsg 181 tlagtvtvts rftlvpsgra dgvtvtckve hesfeepali pvtlsvrypp evsisgyddn 241 wyl
  • the amino acid sequence of a representative PD- L1extracellular domain is set forth below (SEQ ID NO: 64): 1 ftvtvpkdly vveygsnmti eckfpvekql dlaalivywe medkniiqfv hgeedlkvqh 61 ssyrqrarll kdqlslgnaa lqitdvklqd agvyrcmisy ggadykritv kvnapynkin 121 qrilvvdpvt seheltcqae gypkaeviwt ssdhqvlsgk tttnskree klfnvtstlr 181 intttneify ctfrrldpee nhtaelvipe lplahppner [000173]
  • the second immune cell effector domain contains at least
  • the second immune cell effector domain contains at least a portion of the extracellular domain of VTCN1 (B7-H4).
  • VTCN1 The amino acid sequence of a representative VTCN1 is set forth below (SEQ ID NO: 66): 1 liigfgisgr hsitvttvas agnigedgil sctfepdikl sdiviqwlke gvlglvhefk 61 egkdelseqd emfrgrtavf adqvivgnas lrlknvqltd agtykcyiit skgkgnanle 121 yktgafsmpe vnvdynasse tlrceaprwf pqptvvwasq vdqganfsev sntsfelnse 181 nvtmkvvsvl ynvtinntys cmiendiaka tgdikvtese ikrrshlqll nskas Dimerization domain 47 Include Draft Include
  • the CAR-engager forms and is administered in the form of a homodimer or a homo-multimer.
  • the homodimer thus contains two CAR-engager entities.
  • the order of the first moiety, the second moiety and the dimerization domain are not critical.
  • the dimerization domain is disposed between the first moiety and the second moiety.
  • the CAR-engager is in the form of a heterodimer, which contains a first moiety connected to a first dimerization domain and a second moiety connected to a second dimerization domain.
  • the first and second dimerization domains dimerize the first and second moieties to form a heterodimer.
  • the first and second dimerization domains contain a knob-in-hole configuration.
  • One of the dimerization domains contains a protuberance (knob) and the other dimerization domain contains a cavity (hole) that is sterically compensatory to the protuberance, where the tertiary structure of the protuberance is positionable within the tertiary structure of the cavity.
  • Dimerization domains with knob-in-hole configurations may have directed amino acid mutations where the protuberance is an amino acid that has a larger side chain volume than present on a dimerization domain derived from a natural source (e.g., IgA, IgD, IgG, IgM, or IgE) and the cavity is an amino acid that has a smaller side chain volume than present on a dimerization domain derived from a natural source.
  • the protuberance is an amino acid change from a threonine (T) to a lysine (K) and the corresponding cavity is an amino acid change from a leucine (L) to an aspartic acid (D) or a lysine (K).
  • the first dimerization domain contains two amino acid substitutions, for example, a threonine (T) to a lysine (K) and a leucine (L) to a lysine (K), while the second dimerization domain contains a leucine (L) to an aspartic acid (D) or a glutamic acid (E) and a tyrosine (Y) to a glutamic acid (E) or aspartic acid (D).
  • the knob-in-hole dimerization domains are based on opposed charges.
  • the first dimerization domain contains a positively charged amino acid and the second dimerization domain contains a negatively charged amino acid sterically opposable to the positively charged amino acid on the first dimerization domain.
  • Additional protuberance and cavity arrangements are known in the art. See, e.g., U.S. Patents 5,821,333, 7,183,076, 8,642,745, 9,248,182, 9,309,311, 9,527,927, 9,562,109, 9,890,204, 48 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 10,138,303, and 11,168,344 and U.S.
  • the dimerization domains may be derived from IgA, IgD, IgG, IgM, or IgE.
  • the first and the second dimerization domains may contain the same or different amino acid sequences, provided that they bind each other.
  • the first and the second dimerization domains are the IgG1 constant heavy (CH) 3 domain.
  • the amino acid sequence of a representative IgG1 CH3 domain is set forth below (SEQ ID NO: 67): 1 epkspksadk thtapqprep qvytlppsrd eltknqvslt clvkgfypsd iavewesngq 61 pennykttpp vldsdgsffl yskltvdksr wqqgnvfscs vmhealhnhy tqkslslspg 121 k [000182]
  • the first and the second dimerization domains are the IgG1 constant heavy CH2 domain.
  • the amino acid sequence of a representative IgG1 CH2 domain is set forth below (SEQ ID NO: 68): 1 pcpapellgg psvflfppkp kdtlmisrtp evtcvvvdvs hedpevkfnw yvdgvevhna 61 ktkpreeqyn styrvvsvlt vlhqdwlngk eykckvsnka lpapiektis kak [000183]
  • the first and the second dimerization domains are the IgG1 CH2 and CH3 domains.
  • the CH2 and CH3 domains may be interconnected by a linker.
  • the first, the second, or both the first and the second dimerization domains contain a fragment crystallizable region (Fc).
  • the Fc contains L234A and L235A substitutions relative to wild-type Fc that abolishes binding of the Fc to (1) the Fc- ⁇ receptor and (2) the complement component 1q (C1q), referred herein as a “silent Fc”.
  • the silent Fc maintains binding to the neonatal Fc receptor (FcRn) (and therefore extending circulatory half-life of CAR-E which contains the silent Fc to several days).
  • Silent Fc also provides a stabilizing effect to the CAR- E (comparable to the stabilizing effect of wild-type Fc).
  • the silent Fc also has a P329G substation; these three L234A, L235A, and P329G substitutions are also commonly referred to as “PG-LALA”.
  • Linkers [000184] In some embodiments, the CAR-engager contains one or more linkers.
  • a linker is “flexible” when the peptide bonds allow rotation of the amino acid residues within the linker and allow the first and the second moieties to move and bind to their respective cognate receptors on the 49 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 CAR-expressing immune cell or steric spacing (i.e., a spacer) between the first and the second moieties.
  • a linker may be disposed between any two CAR-engager components (also referred to herein as domains, entities, moieties, portions).
  • a linker may be disposed between the dimerization domain and the adjacent domain.
  • a linker may be disposed between the dimerization domain and the second moiety.
  • the CAR-engager contains two linkers, where a first linker is disposed between the first moiety and the dimerization domain, and a second linker is disposed between the dimerization domain and the second moiety.
  • the linker comprises an amino acid having the sequence GGGX, GGGGX (SEQ ID NO: 69), or GSSGSX (SEQ ID NO: 70), where X is any nucleotide, typically either cysteine (C) or serine (S), or repeating sequence thereof.
  • the linker has the amino acid sequence GGGGS (SEQ ID NO: 71), GSPRG (SEQ ID NO: 72), GGGGSGGGGS (SEQ ID NO: 73), GGGGSGGGGSGGGGS (SEQ ID NO: 74), GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 75), GSPRGGGGSGGGGSGGGGS (SEQ ID NO: 76), GSTSGSGKPGSGEGSTKG (SEQ ID NO: 77), KESGSVSSEQLAQFRSLD (SEQ ID NO: 78), EGKSSGSGSESKST (SEQ ID NO: 79), or GSAGSAAGSGEF (SEQ ID NO: 80).
  • the linker may be derived from IgA, IgD, IgE, IgG, or IgM. In some embodiments, the linker may be derived from the hinge region of CD3 ⁇ , CD4, CD8 ⁇ , CD28, IgG1, IgG2, or IgG4. Amino acid sequences of representative linkers are listed in Table 7. Table 7: Amino acid Sequences of Representative Linkers Linker Sequence A 50 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 [000189] In some embodiments, the CAR-engager is in the form of a fusion protein, where the components are linked by peptide bonds.
  • the CAR-engager contains proteinaceous entities that may be covalently connected interconnected by click chemistry, which is a type of chemical connection formed by a method of controlled chemical ligation.
  • the connection may be an azide-alkyne connection, an oxime or hydrazine connection, a tetrazine-transcyclooctene connection, an azide-nitrone connection, a thiol-alkene connection, an alkene-tetrazole connection, an alkene-tetrazine connection, an alkene-azide connection, a conjugated diene-alkene connection, or an isonitrile-tetrazine connection.
  • CAR-E or the IL-2 variant may be encoded in a nucleic acid which is expressed in a cell to produce the CAR-E.
  • nucleic acid refers to a polymer of nucleotides, each of which are organic molecules consisting of a nucleoside (a nucleobase and a five-carbon sugar) and a phosphate.
  • nucleotide unless specifically stated or obvious from context, includes nucleosides that have a ribose sugar (i.e., a ribonucleotide that forms ribonucleic acid, RNA) or a 2’-deoxyribose sugar (i.e., a deoxyribonucleotide that forms deoxyribonucleic acid, DNA).
  • Nucleotides serve as the monomeric units of nucleic acid polymers or polynucleotides.
  • the four nucleobases in DNA are guanine (G), adenine (A), cytosine (C) and thymine (T).
  • the four nucleobases in RNA are guanine (G), adenine (A), cytosine (C) and uracil (U).
  • Nucleic acids are linear chains of nucleotides (e.g., at least 3 nucleotides) chemically bonded by a series of ester linkages between the phosphoryl group of one nucleotide and the hydroxyl group of the sugar (i.e., ribose or 2’-deoxyribose) in the adjacent nucleotide.
  • nucleotides e.g., at least 3 nucleotides
  • nucleic acid sequence of a representative wild-type IL-2 is set forth below (NCBI Accession No. NM_000586.4; SEQ ID NO: 111;): 51 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 1 ctatcaccta agtgtgggct aatgtaacaa agagggattt cacctacatc cattcagtca 61 gtctttgggg gtttaaagaa attccaaaga gtcatcagaa gaggaaaaat gaaggtaatg 121 ttttttcaga caggtaaagt ctttgaaaat atgtgtaata tgtaaacat tttgacaccc 181 ccataatatt ttccagaat tacagtata aattgcatc
  • nucleic acid encoding the CAR-engager includes a signal peptide- encoding nucleic acid disposed 5’ to the nucleic acid encoding the first moiety.
  • signal peptide refers to a short (e.g., 5-30 or 10-100 amino acids long) stretch of amino acids that directs the transport of the protein during translation.
  • CAR-engagers containing a signal peptide will be secreted from the cell. Typically, the signal peptide is cleaved from the CAR-engager before secretion.
  • the signal peptide may be connected to the nucleic acid encoding the first moiety or the nucleic acid encoding the second moiety.
  • the signal peptide may be derived from Ig- ⁇ -3 heavy chain (IGHG3), albumin, CD8 ⁇ , CD33, erythropoietin (EPO), IL-2, human or mouse Ig-kappa chain V- III (IgK VIII), tissue plasminogen activator (tPA), or secreted alkaline phosphatase (SEAP).
  • IGHG3 Ig- ⁇ -3 heavy chain
  • EPO erythropoietin
  • IL-2 human or mouse Ig-kappa chain V- III
  • IgK VIII tissue plasminogen activator
  • SEAP secreted alkaline phosphatase
  • Signal peptides may also be synthetic (i.e., non-naturally occurring). Amino acid sequences of representative signal peptides are listed in Table 8.
  • Table 8 Amino acid Sequences of Representative Signal Peptides Signal peptide Sequence 52 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 CD33 (SEQ ID NO: 92) MPLLLLLPLLWAGALA EPO (SEQ ID NO: 93) MGVHECPAWLWLLLSLLSLPLGLPVLG [000196] IL-2 variant-encoding nucleic acids and CAR-engager-encoding nucleic acids may be introduced into a cell by a suitable vector.
  • the CAR-encoding nucleic acids may be introduced into one or more cells by separate vectors.
  • a vector is configured so as to contain the elements necessary to effect transport into the immune cell and effect expression of the nucleic acid(s) after transformation.
  • Such elements include an origin of replication, a poly-A tail sequence, a selectable marker, and one or more suitable sites for the insertion of the nucleic acid sequences, such as a multiple cloning site (MCS), one or more suitable promoters, each promoter operatively linked to the insertion sites of the nucleic acid sequences and the selectable marker, and additional optional regulatory elements.
  • MCS multiple cloning site
  • promoter refers to a nucleic acid sequence that regulates, directly or indirectly, the transcription of a corresponding nucleic acid coding sequence to which it is operably linked, which in the context of the present disclosure, is an IL-2 variant, and in other aspects, a CAR-engager protein that contains the variant.
  • a promoter may function alone to regulate transcription, or it may act in concert with one or more other regulatory sequences (e.g., enhancers or silencers, or regulatory elements that may be present in the nucleic acid sequences or the vector). Promoters are located near the transcription start sites of genes, on the same strand and upstream on the DNA (toward the 5’ region of the sense strand).
  • Promoters typically range from about 100-1000 base pairs in length. 53 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 [000198]
  • the term “operatively linked” as used herein is to be understood that a nucleic acid sequence is spatially situated or disposed in the vector relative to another nucleic acid sequence, e.g., a promoter is operatively linked to drive the expression of a nucleic acid coding sequence (e.g., the CAR-engager-encoding nucleic acid sequence).
  • a single vector contains a single promoter operatively linked to the CAR-engager-encoding nucleic acid of the enhancer.
  • a single vector contains a single promoter operatively linked to the first moiety-encoding nucleic acid and the second moiety-encoding nucleic acid.
  • the nucleic acids are separated by a nucleic acid encoding a self-cleaving peptide or an internal ribosome entry site (IRES).
  • the single vector contains a first promoter operatively liked to the first moiety- encoding nucleic acid and a second promoter operatively liked to the first moiety -encoding nucleic acid.
  • two vectors are constructed.
  • a first vector contains a promoter operatively linked to the first moiety-encoding nucleic acid and a second vector contains a promoter operatively linked to the second moiety-encoding nucleic acid.
  • the vector contains a strong mammalian promoter, for example a cytomegalovirus (CMV) promoter, a simian virus 40 (SV40) early promoter, synthetic promoters (e.g., RPBSA (synthetic, from Sleeping Beauty), or CAG (synthetic, CMV early enhancer element, chicken ⁇ -Actin, and splice acceptor of rabbit ⁇ -Globin)) or promoters derived from the ⁇ -actin, phosphoglycerate kinase (PGK), or factor EF1 ⁇ genes.
  • CMV cytomegalovirus
  • SV40 simian virus 40
  • synthetic promoters e.g., RPBSA (synthetic, from Sleeping Beauty), or CAG (sy
  • the promoter may contain a core region located close to the nucleic acid coding sequence.
  • the promoter is modified to remove methylation sensitive motifs (e.g., a cytosine nucleotide is followed by a guanine nucleotide, or “CpG”), or by the addition of a regulatory sequence that binds transcriptional factors that repress DNA methylation.
  • the vector includes A/T-rich, nuclear matrix interacting sequences, known as scaffold matrix attachment regions (S/MAR), which enhance transformation efficiency and improve the stability of transgene expression.
  • the vector is a viral vector, for example, a retroviral vector, a lentiviral vector, an adenoviral vector, a herpesvirus vector, an adenovirus, or an adeno-associated virus (AAV) vector.
  • lentiviral vectors have been described, for example, in U.S. Patents 5,665,577, 5,981,276, 6,013,516, 7,090,837, 8,119,119 and 10,954,530.
  • the vector is a non-viral vector, representative examples of which include plasmids, mRNA, linear single stranded (ss) DNA or linear double stranded (ds) DNA, minicircles, and transposon-based vectors, such as Sleeping Beauty (SB)-based vectors and piggyBac(PB)-based vectors.
  • the vector may include both viral and non- viral elements.
  • the vector is a plasmid.
  • the plasmid may also contain other elements e.g., that facilitate transport and expression of the nucleic acid in an immune cell.
  • the plasmid may be linearized with restriction enzymes, in vitro transcribed to produce mRNA, and then modified with a 5’ cap and a 3’ poly-A tail.
  • the vector multiple plasmids, a first plasmid encoding a first proteinaceous entity (e.g., the ectodomain of the CAR-engager) and a second plasmid encoding a second proteinaceous entity (e.g., the immune effector domain of the CAR-engager).
  • the IL-2 variants and CAR-Es may be expressed in a genetically modified (or transformed) cell containing a vector that contains a nucleic acid encoding an IL-2 variant, the CAR- E, or components of the CAR-E for the purpose of making and purifying protein.
  • Cells useful for the cloning and other manipulations of these vectors are conventional. Cells from various strains of E. coli may be used for replication of the vectors and other steps in the construction of the CAR-engagers of this disclosure.
  • Suitable host cells or cell lines for the expression of the nucleic acids encoding the IL-2 variants and the nucleic acid-encoding CAR-engagers include eukaryotic cells.
  • the cells are a mammalian cell line.
  • the cells are mammalian cells such as CHO (e.g., DG44, CHO-S), fibroblast cells (e.g., 3T3, COS), embryonic cells (e.g., PER.C6, HEK (e.g., HEK.293)), somatic cell hybrids (e.g., Sp2/0), and cancer cells, for example, myeloma cells (e.g., NS0 (NS zero)).
  • the nucleic acids encoding the CAR- engager is expressed in a CHO or a myeloma cell.
  • Human cells may be used, thus enabling the expressed CAR-engager to be modified with human glycosylation patterns.
  • suitable mammalian cells and methods for transformation, culture, amplification, screening and product production and purification are known in the art. See, e.g., Green et al., eds., Molecular Cloning: A Laboratory Manual, 5 th ed., Cold Spring Harbor Laboratory Press, New York, 2012. 55 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 [000208]
  • the cells are prokaryotic.
  • Prokaryotic (i.e., bacterial) cells may prove useful as host cells suitable for the expression of the nucleic acids encoding CAR-engagers (see, e.g., Pluckthun, Immunol. Rev.130:151-188 (1992)).
  • any CAR-engagers produced in a bacterial cell would be screened for retention of function (e.g., CAR binding ability of an expressed CAR-E).
  • the CAR-engager expressed by the bacterial cell was produced in a properly folded form, that bacterial cell would be a desirable host, or in alternative embodiments the CAR-engager may express in the bacterial host and then be subsequently re-folded.
  • various strains of E. Coli used for expression are well-known as host cells in the field of biotechnology.
  • Various strains of B. Subtilis, Streptomyces, other bacilli and the like may also be employed.
  • the IL-2 variant or the CAR-engager protein is isolated from the cell (e.g., cell lysates) or from the medium in which the cell is cultured. Protein isolation techniques are known in the art.
  • Representative isolation techniques include chromatography, affinity chromatography, nickel- nitrilotriacetic acid (Ni-NTA) affinity chromatography, high performance liquid chromatography (HPLC), hydroxylapatite chromatography, protein A- Sepharose, gel electrophoresis, and dialysis.
  • the affinity chromatography resin is a Protein A affinity chromatography resin or a Protein G affinity chromatography resin. Additional protein isolation systems and methods are known in the art. See, e.g., U.S. Patents 516,9936, 6,267,958, 8,357,778, 9,630,165, 9,708,399, 10,023,608, 10,207,229, 11,369,703, and 11,390,668, U.S.
  • compositions containing an IL-2 variant or CAR-E may be formulated in a pharmaceutically acceptable carrier.
  • an effective amount refers to a sufficient amount of an IL-2 variant or a CAR-engager to provide the desired effect, e.g., which may include any one or more of killing at least cancer cell, reversing, alleviating, ameliorating, inhibiting, diminishing, slowing down, arresting, stabilizing, achieving remission, or preventing the onset, progression, development, severity or recurrence of a symptom, complication or condition, or biochemical indicia associated with a cancer.
  • IL-2 variant and the CAR-engager administered to a subject will vary between wide limits, depending 56 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 upon the location, type, and severity of the cancer, the age, body weight, and condition of the individual to be treated, etc. A physician will ultimately determine appropriate doses to be used.
  • the IL-2 variant and/or the CAR-engager in the pharmaceutical composition may be in the form of a monomer (in embodiments lacking a dimerization domain), homodimer, or heterodimer, as described herein. [000211] IL-2 variants of the present disclosure may be used alone or in combination with another active agents to treat cancer.
  • High-dose IL-2 treatment can overcome regulatory T cell (Treg)-associated IL-2 trapping and allow extra IL-2 to activate tumor infiltrating lymphocytes (TILs) for treating cancer, for example, metastatic renal cell carcinoma and melanoma.
  • TILs tumor infiltrating lymphocytes
  • patients who respond to high-dose IL-2 treatment frequently suffer from intolerable toxicities (See, e.g., Li et al., Nat. Commun.8(1):1762 (2017)), which has limited its clinical use.
  • IL-2 variants of the present disclosure may constitute a more attractive alternative in terms of binding to IL-2R ⁇ on Tregs (See, e.g., Mott et al., J. Mol. Biol.247(5):979-94 (1995)) or increasing the binding to IL-2R ⁇ on effector cells (See, e.g., Levin et al., Nature 484(7395):529-33 (2012) and Sun et al., Nat. Commun. 10(1):3874 pp. 1-12 (2019)).
  • IL-2 variants of the present disclosure may be conjugated to other active moieties, such as antibodies. See, Ren, et al., supra.
  • the IL-2 variants of the present disclosure may be fused to an anti–PD-1 antibody. These fusion proteins may show better intratumoral T cell binding, more potent antitumor effects, and may also overcome PD-L1 therapy resistance.
  • the IL-2 variants may also be conjugated to anti-tumor antibodies. See, e.g., Sun et al., Nat. Commun.10(1):3874 pp.1-12 (2019).
  • Compositions may be provided as sterile solid or liquid preparations.
  • Solid preparations may be reconstituted and diluted into a liquid preparation before use, e.g., with carriers containing isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous solutions, which may be buffered to a selected pH.
  • Liquid carriers include aqueous and non-aqueous carriers alike. Representative examples of liquid carriers include sterile water for injection, saline, Lactated Ringer Injection solution, phosphate buffered saline, soluble sugars (e.g., dextrose), dimethyl sulfoxide (DMSO), ethanol, and suitable mixtures thereof.
  • the liquid carrier includes a protein dissolved or dispersed therein, representative examples include serum albumin (e.g., human serum albumin, recombinant human albumin), gelatin, and casein.
  • the liquid carrier includes a water-miscible polyol (e.g., glycerol, propylene glycol, liquid 57 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 polyethylene glycol, and the like).
  • the compositions are typically isotonic, i.e., they have the same osmotic pressure as blood.
  • Citric acid, sodium chloride, sugars, polyalcohols, and isotonic electrolyte solutions may be used to achieve the desired isotonicity.
  • other excipients may be added, e.g., wetting, dispersing, or emulsifying agents, gelling and viscosity enhancing agents, preservatives and the like as known in the art.
  • the compositions include citric acid, ethylenediaminetetraacetic acid (EDTA), and polysorbate 20 with a pH range between about 6.8 to about 7.2.
  • the methods may entail administration of the IL-2 variant, per se, or in the form of a fusion with another active moiety, such as an antibody or binding fragment thereof, that binds a receptor on an immune cell, e.g., a TIL, or antibody that binds an epitope on an antigen present on a tumor cell.
  • the administration of the IL-2 variant is used to treat kidney cancer (e.g., metastatic renal cell carcinoma), melanoma, colon cancer, lung cancer, or ovarian cancer.
  • the method entails administering to a subject in need thereof a pharmaceutical composition containing a CAR-engager described herein.
  • Cancers treatable in accordance with the disclosed methods broadly include hematopoietic cancers and cancers characterized by the presence of a solid tumor.
  • subject or “patient” as used herein includes all members of the animal kingdom prone (or disposed) to or suffering from the indicated cancer. In some embodiments, the subject is a human.
  • a subject “having cancer” or “in need of” treatment broadly embraces subjects who have been positively diagnosed, including subjects having active disease who may have been previously treated with one or more rounds of therapy, and subjects who are not currently being treated (e.g., in remission) but who might still be at risk of relapse, and subjects who have not been positively diagnosed but who are predisposed to cancer (e.g., on account of the basis of prior medical history and/or family medical history, or who otherwise present with a one or more risk factors such that a medical professional might reasonably suspect that the subject was predisposed to cancer).
  • treat refers to the aft-recognized indicia of therapeutic efficacy, intervention, process performed on, or the administration of an active agent to the subject in need thereof with the therapeutic objective (“therapeutic effect”) of reversing, alleviating, ameliorating, inhibiting, diminishing, slowing down, arresting, stabilizing, or preventing the onset, progression, development, metastases, severity or recurrence of a symptom, improvement in survival time, total/complete or partial remission, complication or condition, or biochemical indicia associated with cancer.
  • therapeutic objective therapeutic effect
  • the cancer is a hematopoietic cancer.
  • Representative hematological cancers include plasma cell neoplasm (e.g., myeloma, multiple myeloma, relapsed or refractory multiple myeloma, plasma cell myeloma, extramedullary multiple myeloma, monoclonal gammopathy of unknown significance (MUGS), asymptomatic smoldering multiple myeloma, or solitary plasmacytoma), lymphoma (e.g., Hodgkin’s lymphoma (HL), non-Hodgkin’s lymphoma, Burkitt lymphoma, Waldenstrom macroglobulinemia, plasmablastic lymphoma, plasmacytoid lymphoma, B-cell lymphoma, high-grade B-cell lymphoma
  • plasma cell neoplasm e.g., myeloma, multiple myeloma, relapsed
  • the cancer is characterized by the presence of a solid tumor.
  • the cancer is a bladder cancer (e.g.,transitional cell carcinoma, also called urothelial carcinoma), kidney cancer (e.g., renal cell carcinoma (RCC), kidney renal clear cell carcinoma (KIRC), transitional cell cancer, or Wilms tumor), skin cancer (e.g., melanoma, skin cutaneous melanoma (SKCM), basal cell carcinoma, and squamous cell carcinoma of the skin), lung cancer (e.g., small cell lung cancer, non-small cell lung cancer, including lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC)), head and neck cancer (e.g., squamous cell carcinoma of the head and neck (SCCHN) also called head and neck squamous cell carcinoma (HNSC), laryngeal and hypopharyngeal cancer, nasal cavity and paranasal sinus cancer, na
  • a bladder cancer e.g.,
  • the disclosed methods treat malignant mesothelioma, ovarian cancer, breast cancer (e.g., TNBC), pancreatic cancer, lung cancer, liver cancer, glioblastoma, gastric cancer, endometrial cancer, cervical cancer, biliary cancer, uterine serous carcinoma, cholangiocarcinoma, neuroblastoma, sarcoma, or melanoma.
  • the disclosed methods further include an anti-CD19 immune cell therapy, a CAR-E that binds CD19, and are used to treat HL, non-Hodgkin’s lymphoma, ALL, CLL, chronic lymphocytic leukemia, Burkitt lymphoma, DLBCL, PMBL, high-grade B-cell lymphoma, FL, MCL, or multiple myeloma (MM).
  • HL non-Hodgkin’s lymphoma
  • ALL CLL
  • chronic lymphocytic leukemia Burkitt lymphoma
  • DLBCL DLBCL
  • PMBL high-grade B-cell lymphoma
  • FL FL
  • MCL multiple myeloma
  • the disclosed methods further include anti-BCMA immune cell therapy and a CAR-E that binds BCMA, and are used to treat MM, HL, non-Hodgkin’s lymphoma, acute myeloid leukemia (AML), chronic myeloid leukemia (CML), plasma cell leukemia, SLE, acute AMR, chronic AMR, or AL-amyloidosis.
  • the cancer is characterized as being in a state of minimal residual disease (MRD).
  • MRD is a state at which a cancer patient has a small number of cancer cells that remain in the body after treatment.
  • the number of remaining cells may be so small that they do not cause any physical signs or symptoms of the cancer, and often may not be detectable through traditional methods, such as viewing cells under a microscope and/or by tracking abnormal serum proteins in the blood.
  • the amount of cancer antigens present in a subject in a state of MRD are limited. And this limited presence of the cancer antigen may not adequately support the proliferation and efficacy of CAR immune cells.
  • the additional presence of a CAR-engager presents the CAR immune cells with not only additional cancer antigen, but also a supportive IL-2 variant, and in some embodiments, a second immune cell effector domain, that may modulate the activity of the CAR immune cell to promote proliferation, efficacy, and/or persistence.
  • the subject receiving an administration of CAR-engager is in a state of MRD.
  • the method of treating cancer involves treatment of a state of minimal residual disease (MRD) in the subject.
  • the method of treating cancer involves elimination of MRD in the subject.
  • MRD minimal residual disease
  • samples from either a blood draw or a bone marrow aspiration may be used. The most widely used tests to measure MRD are flow cytometry, polymerase chain reaction (PCR) and next-generation sequencing.
  • the CAR extracellular domain is derived from a commercially available anti-BCMA antibody, BCMA-binding fragment, or derivative thereof, e.g., belantamab (Blenrep®), linvoseltamab (REGN5458), pacanalotamab (AMG 420), pavurutamab (AMG 701), and teclistamab (Tecvayli®).
  • the extracellular domain of the CAR binds the BCMA ectodomain of the CAR-engager that has the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2.
  • the CAR extracellular domain contains a single variable heavy (VHH) or variant thereof.
  • the CAR extracellular domain is derived from a commercially available anti-Claudin 18.2 antibody, anti-Claudin 18.2-binding fragment, or derivatives thereof, e.g., osemitamab and zolbetuximab (Vyloy®).
  • the extracellular domain of the CAR binds a Claudin 18.2 ectodomain of the CAR-engager.
  • the CAR-engager contains a Claudin 18.2 ectodomain that has any one of the amino acid sequences SEQ ID NOs: 8-9. [000238]
  • the CAR binds SLAMF7.
  • the extracellular domain of the CAR binds the SLAMF7 63 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 ectodomain of the CAR-engager. In some embodiments, the extracellular domain of the CAR binds the SLAMF7 ectodomain of the CAR-engager having the amino acid sequence SEQ ID NO: 10. [000239] In some embodiments, the CAR binds PD-1. CAR extracellular domains that bind to PD- 1 are known in the art. See, e.g., U.S. Patents 10,124,023 and 11,136,392, and U.S.
  • the CAR extracellular domain is derived from a commercially available anti-PD-1 antibody, anti- PD-1-binding fragment, or derivative thereof, e.g., balstilimab, budigalimab, cadonilimab, cemiplimab (Libtayo®), cetrelimab, dostarlimab (Jemperli®), izuralimab, nivolumab (Opdivo®), pacmilimab, pembrolizumab (Keytruda®), penpulimab, peresolimab, pidilizumab, retifanlimab, rosnilimab, sintilimab, spartalizumab, tislelizumab, toripalimab, volrustomig, vudalimab, zeluvalimab, and zi
  • balstilimab e.g., balstilimab, budigalimab, cadoni
  • the extracellular domain of the CAR binds a PD-1 ectodomain of the CAR-engager. In some embodiments, the extracellular domain of the CAR binds the PD-1 ectodomain of the CAR-engager that has any one of the amino acid sequences SEQ ID NO: 11 and 107-108. [000240] In some embodiments, the CAR binds Mast/stem cell growth factor receptor Kit (KIT; also known as Receptor tyrosine kinase KIT proto-oncogene). CAR extracellular domains that bind KIT are known in the art. See, e.g., U.S.
  • KIT Mast/stem cell growth factor receptor Kit
  • the CAR extracellular domain is derived from a commercially available anti-KIT antibody, anti-KIT-binding fragment, or derivative thereof, e.g., barzolvolimab.
  • the extracellular domain of the CAR binds the KIT ectodomain of the CAR-engager.
  • the extracellular domain of the CAR binds the KIT ectodomain of the CAR-engager having the amino acid sequence SEQ ID NO: 12. [000241]
  • the CAR binds TROP2.
  • CAR extracellular domains that bind TROP2 are known in the art. See, e.g., U.S. Patents 11,602,525, and 11,768,203, and U.S. Patent Application Publications 2018/0296689, 2021/0169852, and 2022/0204582.
  • the CAR extracellular domain is derived from a commercially available anti-TROP2 antibody, anti- TROP2-binding fragment, or derivatives thereof, e.g., datopotamab and sacituzumab.
  • the extracellular domain of the CAR binds a TROP2 ectodomain of the CAR-engager.
  • the CAR-engager contains a TROP2 ectodomain that has the amino acid sequence SEQ ID NO: 13. 64 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 [000242]
  • the CAR binds CD38.
  • CAR extracellular domains that bind CD38 are known in the art. See, e.g., U.S. Patents 10,709,775, 10,799,536, 10,836,998, and 11,365,394 and U.S.
  • the CAR extracellular domain is derived from a commercially available anti-CD38 antibody, anti-CD38-binding fragment, or derivative thereof, e.g., daratumumab (Darzalex®), isatuximab (Sarclisa®), and mezagitamab.
  • the extracellular domain of the CAR binds the CD38 ectodomain of the CAR- engager.
  • the extracellular domain of the CAR binds the CD38 ectodomain of the CAR-engager having the amino acid sequence SEQ ID NO: 14. [000243] In some embodiments, the CAR binds MSLN. CAR extracellular domains that bind MSLN are known in the art. See, e.g., U.S. Patents 10,550,179, 10,640,569, 10,730,954, 11,648,268, and 11,702,472, and U.S.
  • the CAR extracellular domain is derived from a commercially available anti-MSLN antibody, anti-MSLN-binding fragment, or derivative thereof, e.g., amatuximab.
  • the extracellular domain of the CAR binds a MSLN ectodomain of the CAR-engager.
  • the extracellular domain of the CAR binds the MSLN ectodomain of the CAR-engager that has any one of the amino acid sequences SEQ ID NO: 15 and 17-18.
  • the intracellular domain of the CAR contains a signaling domain that enables intracellular signaling and immune cell function.
  • the signaling domain may include a primary signaling domain and/or a co-stimulatory signaling domain.
  • the intracellular domain is capable of delivering a signal approximating that of natural ligation of an ITAM- containing molecule or receptor complex such as a TCR receptor complex.
  • the signaling domain includes a plurality, e.g., 2 or 3, costimulatory signaling domains, e.g., selected from 4-1BB, CD3 ⁇ , CD28, CD27, ICOS, and OX40.
  • the signaling domain may include a CD3 ⁇ domain as a primary signaling domain, and any of the following pairs of co-stimulatory signaling domains from the extracellular to the intracellular direction: 4-1BB-CD27; CD27-4-1BB; 4-1BB-CD28; CD28-4-1BB; OX40- CD28; CD28-OX40; 4-1BB-CD3 ⁇ ; CD3 ⁇ -4-1BB; CD28-CD3 ⁇ ; CD3 ⁇ -CD28; CD28-4-1BB and 4- 1BB-CD28.
  • the primary signaling domain is derived from CD3 ⁇ , CD27, CD28, CD40, KIR2DS2, MyD88, or OX40.
  • the co-stimulatory signaling 65 include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 domain is derived from one or more of CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD4, CD5, CD8 ⁇ , CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD40, CD45, CD68, CD72, CD80, CD86, CD137 (4-1BB; TNFRSF9), CD154, CLEC-1, 4-1BB, DAP10 (hematopoietic cell signal transducer ((HCST)), DAP12 (TYROBP), Dectin-1, Fc ⁇ RI, Fc ⁇ RI, Fc ⁇ RII, Fc ⁇ RIII, IL-2RB, ICOS, KIR2DS2, MyD88, OX40, and ZAP70.
  • HCST hematopoietic cell signal transducer
  • DAP12 TYROBP
  • a representative CAR with a CD3 ⁇ stimulatory signaling domain is the FDA-approved CAR-expressing immune cells tisagenlecleucel (Kymriah®).
  • Representative CARs with CD3 ⁇ and 4-1BB co-stimulatory signaling domains are the FDA-approved CAR-expressing immune cells idecabtagene vicleucel (Abecma®), lisocabtagene maraleucel (Breyanzi®), and ciltacabtagene autoleucel (Carvykti®).
  • Representative CARs with CD28 and CD3 ⁇ co-stimulatory signaling domains are the FDA-approved CAR-expressing immune cells brexucabtagene autoleucel (Tecartus®) and axicabtagene ciloleucel (Yescarta®).
  • the CAR immune cell is a T cell.
  • the CAR immune cell is a NK cell. Additional CAR immune cells are known in the art, e.g., U.S.
  • the CAR immune cells may be autologous or allogeneic.
  • immune cells or progenitors thereof can be isolated from a tissue of body fluid from one subject prior to administration to the same subject (autologous) or a different, compatible subject (allogeneic). Most typically, the CAR immune cells are administered once.
  • the number of CAR immune cells administered to a subject will vary between wide limits, depending upon the location, type, and severity of the cancer, the age, body weight, and condition of the individual to be treated, etc. A physician will ultimately determine appropriate number of cells and doses to be used. Typically, the CAR immune cells will be given in a single, one-time dose.
  • Dosage amounts (e.g., numbers) of CAR immune cells effective to treat cancer are known in the ar. In some embodiments, the effective number of the CAR immune cells is between about 1 ⁇ 10 4 to about 1 ⁇ 10 10 cells per subject. In some embodiments, the effective number of the CAR immune cells is between about 1 ⁇ 10 5 to about 1 ⁇ 10 10 cells per subject.
  • the effective number of the CAR immune cells is about the number of cells given in FDA-approved 66 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 CAR T cell therapies, which is between about 1 ⁇ 10 6 to about 1 ⁇ 10 10 cells per kg of subject body weight. In some embodiments, the effective number of the CAR immune cells is between about 1 ⁇ 10 5 to about 6 ⁇ 10 8 cells per kg of subject body weight. [000251] Since the CAR-engager promotes functionality and persistence of CAR immune cells, CAR therapy that contemplates coordinate administration of the CAR-engager may entail use of fewer CAR immune cells compared to FDA-approved CAR T cell therapies.
  • the CAR immune cells may be administered to a subject for the treatment of a cancer by any medically acceptable route.
  • the CAR immune cells are typically delivered intravenously, although they may also be introduced into other convenient sites (e.g., to an affected organ or tissue) or modes, as determined by an attending physician.
  • Administration of the CAR-E [000253] Broadly, the order in which the CAR-engager and CAR immune cells are administered during the same course of treatment may be varied, provided that they are able to interact in vivo and cause the desired effect.
  • the CAR-engager is co-administered substantially simultaneously to the subject with the CAR immune cells. In some embodiments, the CAR-engager is contacted with the CAR immune cells in vitro before co-administration to the subject. In some embodiments, the CAR-engager is administered to the subject subsequent to the administration of the CAR immune cells. In some embodiments, the CAR-engager is administered to the subject prior to the administration of the CAR immune cells.
  • the methods entail administration of an effective amount of the CAR-engager to a cancer patient who had received, is receiving, or will receive an administration of immune cells containing a CAR that contains an extracellular domain to which the CAR-engager binds, a transmembrane domain, and an intracellular domain comprising a stimulatory domain.
  • the order in which the CAR-engager and CAR immune cells are administered during the same course of treatment may not be critical, provided that they are able to interact in vivo and cause the desired effect.
  • the CAR-engager is co-administered substantially simultaneously to the subject with the CAR immune cells.
  • the CAR-engager is contacted with 67 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 the CAR immune cells in vitro before co-administration to the subject.
  • the CAR-engager is administered to the subject prior to the administration of the CAR immune cells.
  • the CAR-engager is administered to the subject subsequent to the administration of the CAR immune cells, e.g., upon a determination that the CAR-immune cells have lost vitality or persistence in the subject. This determination may be made in accordance with known techniques.
  • a sample is obtained from the subject after the administration of the immune cells.
  • the concentration of immune cells present within the sample may be used to calculate the difference between the concentration of the immune cells administered to the subject and the concentration of the immune cell measured in the sample.
  • the CAR-engager may be administered once the measured immune cell concentration is less than the administered immune cell concentration. In some embodiments, the CAR-engager is administered once the measured immune cell concentration is less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 25%, less than 10%, or less than 5% of the administered concentration of immune cells.
  • administration of the CAR-engager is conducted at least about 1 week, at least about 2 weeks, at least about 1 month, at least about 2 months, at least, at least about 3 months, at least about 6 months, at least about 9 months, or at least about a year after administering the CAR-immune cells.
  • the CAR-engager is administered for at least one cycle, for example, administered once a week, once every two weeks, once every three weeks. The cycle may be repeated, for example, for 2 cycles, 3 cycles, 5 cycles, or8 cycles.
  • the CAR-engager is administered for a period of consecutive days before cyclic administration, for example, administered once a day for five days and once every three weeks thereafter. In some embodiments, the CAR-engager is administered once every 3 weeks (a 21-day cycle) as an infusion over about 30 to about 90 minutes. In some embodiments, the CAR-engager is administered for 5 consecutive days every 21 days and repeated for 8 cycles. [000258] In some embodiments, the CAR-engager is administered as an intravenous infusion over a period of time. Representative infusion times are 30 minutes, 60 minutes, and 90 minutes. In some embodiments, the infusion time is between 30 and 60 minutes.
  • the first administration is infused into a patient for 90 minutes and subsequent administrations are infused into a patient for 30 minutes.
  • the treatment method may entail administration of a first course of CAR-engager therapy that is initiated up to about 6 months after administration of the CAR immune cell therapy.
  • the term “effective amount” as used herein refers to a sufficient amount of CAR-engager to provide the desired effect, e.g., the amount of a CAR-engager to bind to a CAR-expressing immune cell.
  • the first course of CAR-engager therapy is initiated at any time up to about 4 years after the CAR immune cell therapy. In some embodiments, the first course of CAR- engager therapy is initiated at any time up to about 3 years after the CAR immune cell therapy. In some embodiments, the first course of CAR-engager therapy is initiated at any time up to about 2 years after the CAR immune cell therapy. In some embodiments, the first course of CAR-engager therapy is initiated at any time up to about 1 year after the CAR immune cell therapy. In some embodiments, the first course of CAR-engager therapy is initiated at any time up to about 9 months after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated at any time up to about 6 months after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated at any time up to about 5 months after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated at any time up to about 4 months after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated at any time up to about 3 months after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated at any time up to about 2 months after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated at any time up to about 1 month after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated at any time up to about 4 weeks after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated at any time up to about 3 weeks after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated about 2 weeks after the CAR immune cell therapy.
  • the first course of CAR-engager therapy is initiated 2 weeks after the CAR immune cell therapy.
  • the first course of CAR-engager therapy entails administering a total of about 1 to about 6 doses (e.g., 2 doses, 3 doses, 4 doses, 5 doses, or 6 doses) of the CAR- engager.
  • the first course of the CAR-engager therapy entails administration of about 1 dose per week, about 2 doses per week, about 3 doses per week, or about 4 doses per week.
  • the first course of CAR-engager therapy is conducted over a period of time of about 1 to about 3 weeks with administration of about 1 to about 3 doses of CAR-engager per week.
  • the dosage amounts of the CAR-engager administered during the first course of CAR- engager therapy may range from about 1 to about 8 mg/kg of patient body weight.
  • the dosage (effective amount) of the CAR-engager is about 1 mg/kg, 2 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, or about 8 mg/kg.
  • the present methods further include administration of a second, subsequent course of CAR-engager therapy to the subject.
  • the subject may have relapsed, or is at risk of relapse.
  • the CAR-engager administered in the second course of CAR- engager therapy may be the same as or different from the CAR-engager administered in the first course of CAR-engager therapy.
  • the CAR-engager administered in the second course of CAR- engager therapy may be administered within the same period of time after the CAR immune cell therapy as described above or in the same amounts of time described above, but after the first course of CAR-engager therapy.
  • Combination Therapy [000274]
  • the present methods may include co-administration of another anti-cancer therapy.
  • co-administered includes substantially contemporaneous administration, by the same or separate dosage forms, or sequentially, e.g., as part of the same treatment regimen or by way of successive treatment regimens.
  • the first of the two therapies may still be detectable at effective concentrations at the site of treatment.
  • the sequence and time interval may be determined such that they can act together (e.g., synergistically to provide an increased benefit than if they were administered otherwise).
  • the therapeutics may be administered at the same 70 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 time or sequentially in any order at different points in time; however, if not administered at the same time, they may be administered sufficiently close in time so as to provide the desired therapeutic effect, which may be in a synergistic fashion.
  • the terms are not limited to the administration of the active agents at exactly the same time.
  • the subject may also have had an additional anti-cancer therapy.
  • the additional therapy may be administered (1) prior to CAR immune cell therapy, (2) after the CAR immune cell therapy but before the first course of CAR-E therapy, (3) after the first course of CAR-E therapy but before the second course of CAR-E therapy, or (4) after the second course of CAR-E therapy.
  • the additional anti-cancer therapy is chemotherapy, radiotherapy, immunotherapy, targeted therapy, pro-apoptotic therapy, or cell cycle regulation therapy, therapy with thalidomide, lenalidomide, bortezomib, and/or melphalan.
  • Expansion and differentiation agents may also be provided prior to, during, or after administration of the CAR immune cells to increase differentiation, expansion, and/or persistence of the CAR immune cells (e.g., T cells and NK cells).
  • CAR immune cells e.g., T cells and NK cells.
  • Anti-cancer agents that may be used in combination with the IL-2 variants and/or chimeric antigen receptor (CAR)-engagers are known in the art. See, e.g., U.S. Patent No.9,101,622 (Section 5.2 thereof).
  • an "anti-cancer” agent is capable of negatively affecting cancer in a subject, for example, by killing cancer cells, inducing apoptosis in cancer cells, reducing the growth rate of cancer cells, reducing the incidence or number of metastases, reducing tumor size, inhibiting tumor growth, reducing the blood supply to a tumor or cancer cells, promoting an immune response against cancer cells or a tumor, preventing or inhibiting the progression of cancer, or increasing the lifespan of a subject with cancer. More generally, these other compositions would be provided in a combined amount effective to kill or inhibit proliferation of cancerous cells. This process may involve contacting the cancer cells with recipient cells and the agent(s) or multiple factor(s) at the same time.
  • the IL-2 variants and/or CAR-engagers of the present disclosure are used in conjunction with or following prior therapies such as chemotherapeutic, 71 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 radiotherapeutic, immunotherapeutic intervention, targeted therapy, pro-apoptotic therapy, or cell cycle regulation therapy.
  • the IL-2 variants and/or CAR-engagers of the present disclosure are used in conjunction with high-dose chemotherapy prior to the administering of the genetically modified immune cells.
  • bone marrow cells or peripheral blood stem cells are administered subsequent to the high-dose chemotherapy.
  • the IL-2 variants and/or CAR-engagers of the present disclosure are used in conjunction with an effective amount of thalidomide, lenalidomide, bortezomib, or a combination thereof.
  • Additional potentiating treatments that may be used in conjunction with the IL-2 variants and/or CAR-engagers of the present disclosure include melphalan.
  • Melphalan (Alkeran®, Evomela®), an alkylating antineoplastic agent, is used for high-dose conditioning prior to hematopoietic stem cell transplant in patients with multiple myeloma, as well as for palliative treatment of multiple myeloma and for the palliation of unresectable epithelial carcinoma of the ovary.
  • Melphalan is also used to treat AL amyloidosis, neuroblastoma, rhabdomyosarcoma, breast cancer, ocular retinoblastoma, some conditioning regiments before bone marrow transplant, and in some cases, malignant melanoma.
  • Melphalan may be administered in pill form by mouth.
  • Immunotherapy including immune checkpoint inhibitors may be employed to treat a diagnosed cancer.
  • Immune checkpoint molecules include, for example, PD-1, PDL1, CTLA4, KIR, TIGIT, TIM-3, LAG-3, BTLA, VISTA, CD47, and NKG2A.
  • Clinically available examples of immune checkpoint inhibitors include durvalumab (Imfinzi®), atezolizumab (Tecentriq®), and avelumab (Bavencio®).
  • PD-1 inhibitors include nivolumab (Opdivo®), pembrolizumab (Keytruda®), and cemiplimab (Libtayo®). Additional inhibitors that may be useful in the practice of the present disclosure are known in the art. See, e.g., U.S. Patent Application Publications 2012/0321637, 2014/0194442, and 2020/0155520. 72 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 Chemotherapy [000283] Anti-cancer therapies also include a variety of combination therapies with both chemical and radiation-based treatments.
  • Combination chemotherapies include, for example, Abraxane®, altretamine, docetaxel, Herceptin®, methotrexate, Novantrone®, Zoladex®, cisplatin (CDDP), carboplatin, procarbazine, mechlorethamine, cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, raloxifene, estrogen receptor binding agents, Taxol®, gemcitabien, Navelbine®, farnesyl-protein tansferase inhibitors, transplatinum, 5-fluorouracil, vincristine, vinblastine and methotrexate, or any analog or derivative variant of the foregoing and also combinations
  • Radiotherapy also include radiation-based, DNA-damaging treatments.
  • Combination radiotherapies include what are commonly known as gamma-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells which cause a broad range of damage on DNA, on the replication and repair of DNA, and on the assembly and maintenance of chromosomes. Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells and will be determined by the attending physician.
  • Radiotherapy may include external or internal radiation therapy.
  • External radiation therapy involves a radiation source outside the subject’s body and sending the radiation toward the area of the cancer within the body.
  • Internal radiation therapy uses a radioactive substance sealed in needles, seeds, wires, or catheters that are placed directly into or near the cancer.
  • the lentivirus was harvested at 48, 72, and 96 hours (h) post transfection, sedimented at 20,000 x g for 2h, and resuspended in optiMEM media. A new batch of HEK293 cells was then subjected to three rounds of transduction with the virus. Cells were allowed to recover in DMEM complete media and were subjected to puromycin selection to retain only cells that integrated the lentivirus plasmid. Cells were then expanded in four 15 cm culture dishes until they reached confluency, washed carefully with PBS, and incubated in serum-free DMEM for 24 to 48h. The supernatant was harvested, and protein expression was confirmed via SDS-PAGE and immunoblotting.
  • Proteins were purified by adsorption onto a nickel nitriloacetic acid (Ni-NTA) metal affinity column. Non- specifically bound proteins were removed by washing with 40 mM imidazole. The imidazole concentration was increased to 250 mM, allowing recovery of the protein of interest. The protein was further purified via size-exclusion chromatography and were stored in 50 mM HEPES buffer, pH 7.5 at -80 °C until use. [000289] Some CAR-engagers were isolated by passage through an affinity chromatography resin, typically in the presence of a neutral phosphate buffer.
  • the affinity chromatography resin was then subjected to an acidic buffer with a pH of about 3 to about 4, thereby washing CAR-engager off of the affinity chromatography resin.
  • a basic buffer may be used to neutralize the acidic buffer, then a tangential flow filtration of the neutralized buffer can be performed with a formulation buffer, to isolate a concentrated and purified solution containing the CAR-engager.
  • the CAR construct that binds human BCMA contains an scFv derived from the anti-human BCMA antibody clone MSK54, followed by human 39BB and CD3 ⁇ intracellular signaling domains.
  • the human signaling CAR constructs were transduced into HeLa cells that stably produce gamma-retrovirus pseudotyped with the envelope of the feline endogenous virus (RD114), which has been shown to transduce human hematopoietic cells (HSC) with high efficiency (Ward et al., Mol. Ther. 8(5):804-12 (2003)).
  • High viral titer clones were isolated by 74 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 limiting dilution.
  • the high expression clone was seeded and grown in DMEM complete media containing 10% FBS until 80% confluency. Media was exchanged with RPMI complete media containing 10% FBS. After 24 hours, the virus-containing media was harvested, sterile-filtered using a 0.45 ⁇ m PES filter, and utilized for producing CAR T cells. [000291]
  • the production of CAR T cells was adapted from previous studies. See, for example, Li et al., Methods Mol. Biol.1514:111-118 (2017).
  • whole blood was obtained from apheresis leukoreduction collars of platelet healthy donors, due to a high number of viable white blood cells.
  • the whole blood was subjected to centrifugation through a Ficoll gradient to isolate PBMCs.
  • Whole PBMCs were utilized without selecting for CD8 + T cells.
  • PBMCs were resuspended in RPMI media containing 10% Fetal Bovine Serum (FBS), 200 IU/mL IL-2, 60 ng/mL IL-7, 10 ng/mL IL-15, 2 ⁇ g/mL anti-human CD3 (OKT3 clone), and 0.5 ⁇ g/mL anti-human CD28 (CD28.1 clone) at a cell concentration of 4 ⁇ 10 6 cells/mL in 3 mL of media per well in a 6-well plate.
  • FBS Fetal Bovine Serum
  • mice were used to establish a multiple myeloma mouse model in NSG mice.
  • In vivo experiments were initiated by tail vein intravenous injection of 1 ⁇ 10 6 OPM2 cells expressing GFP and Firefly Luciferase followed by biweekly Bioluminescent Imaging (BLI).
  • BLI Bioluminescent Imaging
  • mice were intravenously injected through the tail vein with CAR T cells.
  • BLI was performed biweekly afterwards to assess tumor burden. Quantification was measured using photons/sec using Aura software.
  • 75 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 [000293] Organ analysis.
  • mice were sacrificed and spleen, bone marrow, blood, liver, kidney, and lungs were harvested and weighed.
  • the liver, kidney, and lungs were minced, digested with collagenase (final concentration of 1 ⁇ g/mL collagenase), and incubated at 37 °C for 1 h.
  • Spleens were crushed, and bone marrow was aspirated using a 30-gauge insulin needle. All processed cells were pushed through a 70 ⁇ m strainer to produce a single cell suspension of cells. Cells were resuspended in 1 mL of ammonium-chloride- potassium (ACK) lysis buffer to deplete the sample of red blood cells for 2 m at room temperature.
  • ACK ammonium-chloride- potassium
  • the resulting single cell suspensions were washed with fluorescence-activated single cell sorting (FACS) buffer of PBS and 0.5% BSA, stained, and analyzed using flow cytometry.
  • FACS fluorescence-activated single cell sorting
  • the OPM2 cell line which endogenously express BCMA, were engineered to express green fluorescent protein (GFP) and Firefly Luciferase.
  • GFP green fluorescent protein
  • Firefly Luciferase PBMC
  • Peripheral Blood Mononuclear Cells were obtained by Ficoll gradient of apheresis leukoreduction collars of platelet healthy donors.
  • Tumor burden was assessed using the IVIS® Lumina Series III (Perkin Elmer) after intraperitoneal injection of D-Luciferin (150 mg/kg, from a 15 mg/ml solution) at the indicated time. Each mouse was imaged in groups of up to five mice in the supine position at the same time points (5 min). BLI intensity was analyzed by Aura imaging analysis software (Spectral Instruments Imaging). Peripheral blood from mice was obtained 76 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 by submandibular bleeding in an EDTA-coated tube and analyzed for CAR T cell detection and expansion. In brief, volume of the blood was determined in order to calculate absolute values.
  • the samples were incubated with the indicated treatment, labeled with Alexa647, at the specified time and temperature.
  • the cells were imaged using the Leica THUNDER Imager.
  • the intensity cut-off for the AlexaFluor647 channel was set at 2000 for all images, except for the VHH-muIL2 samples shown on the right (condition 4), for which, the image sensitivity was enhanced by 25-fold (intensity cut- off 80) to visualize the Alexa647 signal.
  • ELISA ELISA analyses were performed to measure the levels of human T cell-derived cytokines in the serum of mice that received OPM2 cancer cells followed by a low dose of CAR T cells, as shown in FIG. 19A.
  • pSTAT5 assay Initially, CAR T were incubated in complete RPMI media without the presence of cytokines (rested) for 24 h. Cells were then stained with CellTraceTM Blue (Invitrogen) or CellTrackerTM Red (Invitrogen) for 30 minutes at 37 °C for the indicated conditions. Cells were washed once with complete RPMI + 10% FBS media. CAR T cells stained with CellTrackerTM Red were blocked using recombinant BCMA-CH3 (100 nM) for 20 minutes on ice while cells stained with CellTraceTM Blue were not blocked.
  • CellTraceTM Blue Invitrogen
  • CellTrackerTM Red Invitrogen
  • Cells were then washed once with complete RPMI + 10% FBS media followed by seeding of approximately 2 ⁇ 10 5 cells (either the two separately stained cells co-cultured or separately cultured) per well of a 96-well plate in the presence of serial dilutions of treatments or cytokine controls at 37 °C. After 5 minutes of incubation, cells were immediately fixed with 1.5% formaldehyde in PBS for 10 minutes at room temperature. Cells were then permeabilized with ice cold 100% methanol for 20 minutes on ice 4 °C.
  • Phenotype of the cells was then determined using projection of our sample to the Seurat pbmc_multimodal dataset using the FindTransferAnchors() and FindQuery() workflow.
  • the FindMarkers() function was used to find differentially expressed genes between chosen groups.
  • the heatmaps were generated with the DoHeatmap() function with a downsampling of 500 cells.
  • Gene Scores were obtained by creating a list of the genes of interest which was given as the feature for the function AddModuleScore().
  • a 96 well plate was seeded with approximately 2 ⁇ 10 5 cells/well in the presence of 10 nM treatment or controls at 37 °C for 2 hours. Cells were washed with FACS buffer and left at 37 °C for 2 or 22 hours (4 hour and 24-hour timepoints). Cells were stained with anti-CD8-FITC (1:50, Biolegend), anti-CD4-PE/Dazzle594TM (1:50, Biolegend) and Alexa647 labeled BCMA and sorted on the Sony Sorter MA900.10,000 CD4 and 10,000 CD8 cells were sorted per condition. SMART- Seq mRNA library preparation kit (Takara Bio) was utilized to generate mRNA libraries, with each replicate tagged with a unique index.
  • RNA Seq-Analysis Gene counts for the samples were obtained by trimming the fastQ files and transcript quantification using the RNAlysis software. Gene names were obtained from the homo sapiens ensembl database with biomaRt, and differential expression between different conditions was determined using the DESeq2 pipeline. Volcano plots were drawn using the EnhancedVolcano library, with a cutoffs at logFC >
  • GSEA was performed using the pipeline of the fgsea package with the ranking metric being -log 10 (p-value)*sign (fold change) and basing the computations on the hallmark pathways of the MSigDB collection.
  • Example 2 BCMA-containing CAR-engager in vitro characterization.
  • a Fusion protein consisting of the human BCMA ectodomain was fused with a two low- affinity mutated human IL-2 (muIL2) domains, and to improve pharmacokinetics and enhance stability, the CH3 domain (Feige et al., Trends Biochem.
  • IL-2 induces an alternative differentiation pathway of T cells, resulting in the generation of distinct “better effector” CD8 + T cells (Hashimoto et al., Nature 610(7930):173-181 (2022)). This process may rely, at least in part, on IL-2 binding to IL- 2R ⁇ . Additionally, IL-2R ⁇ -biased agonists may drive T cells towards a terminally differentiated state (Codarri et al., Nature 610(7930):161-172 (2022)).
  • CAR-engagers may be able to overcome the need for IL-2R ⁇ in the alternative differentiation pathway by anchoring the low-affinity IL-2 on the surface of the CAR T cells via the antigen-to-CAR binding, thereby promoting the generation of memory CAR T cells.
  • a potential synergistic effect between CAR signaling and IL-2 signaling may also exist.
  • BCMA CAR-engager Minimal binding of BCMA CAR-engager to non-transduced T cells was observed as well as minimal binding of VHH-muIL2, a control construct which replaces the BCMA ectodomain with an irrelevant nanobody (VHH) in the CAR-engager CH3-muIL2 construct.
  • VHH irrelevant nanobody
  • This observation further suggests that the binding of the CAR-engager to CAR T cells is primarily driven by the BCMA antigen, and that the muIL2 exhibits weak binding to both CAR T and non-transduced T cells.
  • the BCMA-muIL2 CAR-engager did not exhibit binding to any immune cell populations in human peripheral blood mononuclear cells (PBMCs) (FIGs.15A – 15B).
  • PBMCs peripheral blood mononuclear cells
  • BCMA CAR-engager the functional effects of BCMA CAR-engager on BCMA CAR T cells were evaluated.
  • BCMA CAR T cells were incubated with varying concentrations of the BCMA-muIL2 CAR-engager for 24 h, followed by assessment 80 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 of the expression of the CD69 activation marker (Cibrián and Sánchez-Madrid, Eur. J. Immunol. 47(6):946-953 (2017)) using flow cytometric analysis.
  • OPM2 cells were co-incubated with BCMA CAR T cells (shown in red) or non- transduced T cells (shown in gray) (E:T ratio 1:1; 30,000 cells of each) in the presence of varying concentrations of the BCMA-muIL2 CAR-E treatment.
  • Live (PI-) OPM2 cells were counted 48 hours later, with an N of 3 for each of the experiments. Error bars in FIG.2H represent mean with standard deviation. Without being bound by theory, this finding might be attributed to the reversibility of CAR-engager binding to CAR, while the killing process, which involves the clustering effect and synapse formation between CAR and cancer antigen, is an irreversible event.
  • the BCMA CAR-engager selectively induces STAT5 activity in CAR T cells through the cis-delivery of the low-affinity IL-2 to the same targeted CAR T cells.
  • IL-2 is known to exhibit strong activity on T cells, and the phosphorylation of STAT5 both serves as a reliable indicator of IL-2/IL-2R engagement and correlates with downstream effects such as phenotypic marker expression and cell proliferation (Jones et al., J. Immunol.205(7):1721-1730 (2020)).
  • BCMA CAR T cells were exposed to varying concentrations of the BCMA CAR-engager.
  • Wild-type IL-2 exhibited a lower EC50 (approximately 0.001 nM) suggesting a difference in signaling kinetics.
  • the two-step process involved in STAT5 activity mediated by the BCMA-muIL2 CAR-engager involves: (i) binding of the antigen-to-CAR on T cell surfaces and (ii) subsequent interaction of the low-affinity IL-2 with nearby IL-2R, which, without being bound by theory, may be the reason for the measured difference.
  • wild-type IL-2 requires only binding to IL-2R, enabling it to more rapidly induce STAT5 activity.
  • the VHH-muIL2 can activate STAT5 solely through the low- affinity IL-2, which may explain its requirement for higher concentrations to induce STAT5 activity in T cells.
  • BCMA CAR T cells pre-blocked with BCMA-CH3 showed a significant decrease in pSTAT5 levels to the same degree as control VHH-muIL2, validating that the potency of the CAR- engager is mediated by antigen-to-CAR binding (FIG.2I).
  • FOG.2I antigen-to-CAR binding
  • non- blocked and pre-blocked BCMA CAR T cells were co-cultured in the presence of varying concentrations of CAR-engager.
  • Pre-blocked CAR T cells had lower pSTAT5 levels compared to their co-cultured non-blocked CAR T cells, indicating that CAR-engager affects the targeted CAR T cells (cis-activation) but not adjacent cells.
  • BCMA CAR T cells were treated with the indicated treatments for 5 min at 37 °C followed by STAT5 phosphorylation assessment.
  • peripheral blood mononuclear cells PBMCs
  • PBMCs peripheral blood mononuclear cells
  • the activated T cells were treated with teceleukin (recombinant human IL-2 without glycosyl units), a CAR-engager that contains a N-terminal ectodomain that binds BCMA ( ⁇ 7 kDa), a CH3 domain ( ⁇ 14 kDa), and two repeats of the weak affinity variant of IL-2 immune cell effector domain, with the overall structure of BCMA-CH3-muIL2-muIL2 and referred herein as BCMA-muIL2, or a CAR-engager that contains an ectodomain that binds BCMA, a CH3 domain, and the Neo-2/15 immune cell effector domain, with the overall structure of BCMA-CH3-Neo-2/15, referred to herein as BCMA-Neo-2/15, for 4 days.
  • teceleukin recombinant human IL-2 without glycosyl units
  • a CAR-engager that contains a N-terminal ectodomain that binds BCMA (
  • T cells were counted and stained with carboxyfluorescein succinimidyl ester (CFSE) and analyzed for mean fluorescence intensity (MFI) of CFSE to determine T cell division.
  • CAR-engagers BCMA-muIL2 or BCMA-Neo-2/15 effected T cell counts and division (CFSE staining), similar to teceleukin, which is known to activate T cells.
  • the CAR-engager treatment resulted in less sensitivity as compared to the teceleukin treatment.
  • Systemic administration of IL-2 is associated with severe side effects (Rosenberg, J. Immunol. 192(12):5451-5458 (2014); Dutcher et al., J.
  • Example 4 CAR-engagers activate CAR T cells specifically through the ectodomain [000311] To show that CAR-engager immune cell effector domains stimulate immune cells, the following experiment was performed. As illustrated in FIG.4A, PBMCs were stimulated with anti- CD3, anti-CD28, IL-2, IL-7, and IL-15 to produce activated T cells, which were then transduced with a vector containing a CAR. The activated CAR-expressing T cells (CAR T cells) were rested for 24 hours and then treated with CAR-engagers containing an immune cell effector domain or CAR-engagers lacking an immune cell effector domain as an ectodomain control.
  • CAR T cells activated CAR-expressing T cells
  • CAR-engagers containing BCMA ectodomain, a CH3 domain, and containing either 4- 1BBL (BCMA-41BBL), weak affinity IL-2 (BCMA-muIL2), or Neo-2/15 (BCMA-Neo-2/15) immune cell effector domains were tested for T cell activation.
  • BCMA-41BBL 4- 1BBL
  • BCMA-muIL2 weak affinity IL-2
  • Neo-2/15 BCMA-Neo-2/15
  • ectodomain specificity control containing a nanobody that binds FN1 (clone NJB2, abbreviated NJB2-VHH) fused to a CH3 domain, and the Neo-2/15 stimulatory (NJB2-VHH-Neo-2/15) was tested for T cell activation.
  • the ectodomain specificity controls have similar overall structure as the CAR-engagers used in this experiment (protein domain-CH3-muIL2-muIL2 or protein domain-CH3-Neo-2/15).
  • Ectodomain specificity controls and CAR-engagers were incubated with CAR T cells for 10 hours, and the cells were stained for CD69 as an activation marker and measured by flow cytometry.
  • CAR-engagers containing a cancer antigen ectodomain specifically activate CAR T cells that expresses a CAR that recognizes CAR-engager’s cancer antigen ectodomain.
  • Example 5 CAR-engagers stimulate CAR T cell killing target cells 84 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 [000314] To show that CAR-engagers do not inhibit CAR T l cell killing, the following experiment was performed. CAR T cells were produced as described above and co-incubated with CAR- engagers and BCMA + multiple myeloma cancer cells.
  • CAR T cells were incubated with OPM2 BCMA + cells at an E:T ratio of 1:1 for 1 day and analyzed for target cell survival as compared to target cells without T cell coincubation (FIG.5A).
  • BCMA ectodomain CAR-engagers with either a muIL2 (BCMA-CH3-muIL2) or a 4- 1BBL (BCMA-CH3-41BBL) immune cell effector domain did not inhibit killing of OPM2 cells (FIG.5B).
  • Example 6 CAR-engagers reduce tumor burden, extend survival, and extend CAR T cell persistence in vivo [000316] To show that CAR-engagers reduce tumor burden, extend CAR T cell in vivo persistence, and extend survival, the following experiment was performed. 1 ⁇ 10 6 OPM2 BCMA + multiple myeloma cancer cells were intravenously (i.v.) injected into NOD-scid IL2R ⁇ null (NSG) mice 10 days before infusing a suboptimal dose of 5 ⁇ 10 5 anti-BCMA CAR T cells by i.v. injection.
  • mice were treated twice weekly for two weeks, followed by once weekly with 200 ⁇ g/mouse of CAR-engager containing a BCMA ectodomain, a CH3 domain, and two weak affinity IL-2 immune cell effector domains (BCMA-CH3-muIL2-muIL2) by intraperitoneal (i.p.) injection (FIG.6A).
  • CAR-engager containing a BCMA ectodomain, a CH3 domain, and two weak affinity IL-2 immune cell effector domains (BCMA-CH3-muIL2-muIL2) by intraperitoneal (i.p.) injection.
  • FIG.6A two weak affinity IL-2 immune cell effector domains
  • CAR-engager treated group all mice completely cleared OPM2 tumor cells from the bone marrow, as no signal was detected by imaging.
  • FIGs. 7A-7C show flow cytometry with GFP on the y-axis.
  • GFP + OPM2 cells were detected at similar levels in the bone marrow, lung (FIG.7B), and liver (FIG.7C) of the no-CAR control mouse and the CAR-only mice.
  • FIG.7A One CAR-only mouse had significant levels of OPM2 cells in the blood and spleen (FIG.7A).
  • FIGs.7A The one mouse that received CAR T cell and CAR-engager treatment that developed an eye tumor had no to little OPM2 cells in the blood or spleen (FIG.7A), bone marrow or lung (FIG 7B), and liver (FIG. 7C).
  • FIGS. 8A – 8C show flow cytometry with anti-CD45 on the y-axis and BCMA + -CH3 tagged with Alexa Flour TM 647 (AF647) on the x-axis.
  • CD45 + CAR + T cells only persisted in CAR T cell and CAR-engager treated mice.
  • CAR-only treated mice had little to no CD45 + cells in all organs tested (FIGs.8A-8C).
  • CAR T cell + CAR-engager treated mice had CD45 + cells that also stained positive for the BCMA cancer antigen (which is also the CAR binding target) tagged with AF647, as shown on the x-axis.
  • CD45 + AF647 + double positive CAR T cells were detected in the blood, spleen, and lymph node (FIG. 8A), bone marrow and lung (FIG. 8B), and liver and kidney (FIG.8C). CD45 + single positive cells were only detected in large numbers in liver and kidney (FIG.8C). Little to no CD45 + AF647 + double positive CAR T cells were detected in the eye tumor site (FIG.8C). These results indicated that CAR-engagers reduce tumor burden, extend CAR T cell in vivo persistence, and extend survival.
  • Example 7 CAR-engager fate [000322] The CAR-engagers bind CAR T cells at the cell surface at 4 °C, and slowly internalize at 37 °C.
  • BCMA CAR T cells were exposed to AlexaFluor647-labeled BCMA-muIL2, BCMA-CH3, or VHH-muIL2 at a concentration of 2 nM. The cells were incubated at either 4 °C or 37 °C for various time intervals, followed by fixation and subsequent microscopy imaging.
  • the cytoplasm mean intensity was reported, as the dsRed is expressed inside the cell, whereas for the AlexaFluor 647 channel, the mean intensity for the entire cell was measured. [000323]
  • the CAR-engager rapidly clears from the circulation. Pulsing CAR T-cells with the CAR- engager treatment, where pulsing involves periods of stimulation followed by periods of resting, is superior to prolonged exposure to CAR-engagers as extended exposure can lead to exhaustion or the generation of terminally differentiated CAR T cells.
  • a CAR-engager with a short circulation half-life can be more effective at expanding CAR T cells, driving generation of memory CAR T cells, decrease potential competition with tumor antigen for CAR binding, and enhanced safety profile in patients. Therefore, the CH3 domain of IgG1 was used in the CAR-engager platform. Pharmacokinetic studies illustrated that the circulatory half-life of the CAR-engager was short (1- 1.5 hours) (FIG. 9A).
  • the sera were then obtained by centrifugation and used for the subsequent analysis.
  • An ELISA was performed to determine the concentration of the treatments in the sera.
  • the ELISA plates were coated with 5 ⁇ g/ml anti-His6 antibody overnight, followed by incubation with the sera for 2 h at room temperature.
  • An anti-FLAG HRP antibody was next used for the detection; the CAR-engager was engineered to have FLAG and His6 tags at the C-terminus. Based on the collected five time points, the initial concentration of the treatment in the sera was estimated to be 20% higher than the first (30 min) collected time point.
  • the BCMA CAR-E was >90% and >99% cleared from the circulation in 8 h and 24 h, respectively.
  • the BCMA CAR-engager enhances activity and persistence of CAR T-cells in a multiple myeloma (MM) model.
  • MM multiple myeloma
  • BCMA CAR T cells Two weeks after OPM2 cell injection, freshly prepared BCMA CAR T cells (0.5 million CAR + cells containing an 39BB-CD3 ⁇ CAR construct) were intravenously administered.
  • a cohort of mice received the BCMA-muIL2 CAR-engager treatment (FIG.9B).
  • BCMA-muIL2 CAR-E treatment 200 ⁇ g was administered twice per week for two weeks, followed by once per week until the endpoint. After one month or longer, mice were euthanized, and flow cytometric analyses were performed on the harvested organs.
  • BCMA CAR T cells were detected by co-staining with an anti-human CD45 antibody and Alexa647-labeled BCMA antigen. Similar results were obtained in repeated experiments. Additional control cohorts received VHH-muIL2 treatment with the same dose and schedule as BCMA-muIL2.
  • FIG.9D shows pooled data from these experiments.
  • Data were analyzed by group mean comparisons using one-way ANOVA and subsequent Tukey post-hoc analysis. Individual flow graphs for the pooled data are shown in FIGs.17A – 17C. Error bars represent mean with standard deviation.
  • Analysis of blood samples collected at various time points revealed a substantial expansion of CAR T cells in the circulation following CAR-engager treatment, with the peak expansion observed at week 4 (FIG. 10D).
  • Flow cytometric analyses of blood samples revealed robust expansion of CAR T cells in the treatment group compared to PBS or VHH-muIL2 cohorts.
  • mice treated with CAR-engager exhibited no signs of toxicity based on clinical observations and weight measurements (FIG. 10H).
  • Subsequent analysis conducted two months after CAR T cell injection demonstrated a substantial presence of CAR T cells, including memory CAR T cells, in the CAR-engager treated mice (FIGs.10F-10J, FIGs.18A – 18C, and FIGs.19A – 19C).
  • CAR T cells were detected in the spleen; however, these mice succumbed to tumor growth at around 20 days post-CAR T cell injection.
  • the BCMA-muIL2 89 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 treatment had also increased the presence of CAR T cells in bone marrow compared to PBS or VHH-muIL2 cohorts, but the difference was less significant than spleen. Data were analyzed by two-way ANOVA with Tukey’s multiple comparisons test. *P ⁇ 0.05, ***P ⁇ 0.001, ****P ⁇ 0.0001. Individual flow data are shown in FIGs.18A – 18C. Error bars represent mean with S.E.M.
  • FIG. 19A – 19C The Flt-SNE mapping shown in FIG. 19C is of CAR T cells derived from the PBS, BCMA-muIL2 and VHH-muIL2 treated mice as shown in FIG. 10A.
  • CD45 + , ⁇ -BCMA-CAR + immune cells from the mice were concatenated to form a total of ⁇ 9800 (PBS spleen), ⁇ 8100 (BCMA-muIL2 spleen), ⁇ 7600 (PBS bone marrow), ⁇ 14200 (BCMA-muIL2 bone marrow), ⁇ 1420 (VHH-muIL2 Bone Marrow).
  • the entire high dimensional dataset was merged to create a single Flt-SNE map for each condition with the signal strength of various phenotypic markers defining specific immune phenotypes expressed with a blue-green-yellow-red continuous color scale.
  • FltSNE was conducted with the following parameters: max iterations: 1000, theta: 0.5, learning rate: 200, perplexity: 20. There were inadequate numbers of CAR T cells in VHH-muIL2 cohort spleen to conduct Flt-SNE. To enhance visibility, the dots representing the VHH-muIL2 bone marrow samples were enlarged, as fewer cells were detectable in these mice. In the BCMA-muIL2 treated mice, the majority of CAR + cells were CD8 + cells, while in the VHH-muIL2 samples, CD4 + cells constituted the majority of CAR + cells.
  • CAR + cells in the CAR+PBS cohort exhibited low or no expression of CD45RA, CD45RO, 90 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 or CD62L, whereas the BCMA-muIL2 treated mice showed a CAR + population with elevated expression levels of these memory markers.
  • the treatment not only facilitates robust proliferation and eradication of tumor cells using low doses of CAR T cells but also promotes the development of long-lasting memory cells, demonstrating the efficacy of BCMA-muIL2 CAR-engager treatment in enhancing the clearance of tumor cells by CAR T cells, and generation of long-lasting memory cells.
  • Bone marrow and splenocytes were analyzed via flow cytometry to detect and quantify CAR-expressing T cells and the bone marrow cells or splenocytes were co-incubated with OPM2 target cells at various E:T ratios (1:1 and 2:1) based on CAR-expressing cells. Survival was determined 24, 48, and 72 hours later using flow cytometric analysis. The three-month-old CAR T cells demonstrated efficient killing of tumor cells and long-term functionality (FIG.11D).
  • CD45 + , CD8 + , ⁇ -BCMA- CAR + cells from the five mice were concatenated to form a total of ⁇ 17600 (spleen) and ⁇ 10800 (bone marrow) cells.
  • the entire high dimensional dataset (excluding the CD45, CD8, and CD4 parameters) was merged to create a single tSNE map with the signal strength of six phenotypic markers defining specific immune phenotypes expressed with a blue-green-yellow-red continuous color scale.
  • tSNE analysis was performed using 1000 iterations, a perplexity of 30 and a learning rate of 1237 and 756 for spleen and bone marrow respectively.
  • CD45 + , CD4 + , ⁇ -BCMA-CAR + immune cells from the five mice were concatenated to form a total of ⁇ 9000 (spleen) and ⁇ 6600 (bone marrow) cells.
  • the entire high dimensional dataset (excluding the CD45, CD8, and CD4 parameters) was merged to create a single t-SNE map with the signal strength of six phenotypic markers defining specific immune phenotypes expressed with a blue-green-yellow-red continuous color scale.
  • tSNE analysis was performed using 1000 iterations, a perplexity of 30 and a learning rate of 630 and 466 for spleen and bone marrow, respectively.
  • CAR + cells were sorted after staining with BCMA-AlexaFluor647 and TotalSeq-C hashing antibodies from BCMA-muIL2 or VHH-muIL2 treated mice as shown in the red and green boxes, respectively in FIG.21A.5000 CAR + cells from BCMA-muIL2 mice bone marrow and spleen and 2500 CAR + cells from VHH- muIL2 mice bone marrow and spleen were loaded onto the 10X channel.
  • the scRNAseq analysis revealed that the predominant population of persistent CAR T cells in the BCMA-muIL2-treated mouse consisted of CD8 + T cells (FIGs.
  • FIG. 21D shows significantly differentially expressed genes between CAR-engager treatment and VHH conditions in CD8 and CD4 CAR T cells, split among different relevant conditions.
  • Genes marked with an * are the significantly differentially expressed genes between BCMA-muIL2 and VHH-muIL2 treated mice in the subset of interest. This was evidenced by elevated expression levels of granzyme family genes, other cytotoxicity-associated genes, and MHC class II genes.
  • CAR-engager expands CAR T cells in the absence of tumor antigens
  • CAR T cell expansion typically occurs following infusion in patients, with peak expansion observed around 10-14 days post-infusion (Rodriguez-Otero et al., N. Engl. J. Med. 388(11):1002-1014 (2023)).
  • Eradication of minimal residual disease (MRD) facilitates long-lasting and complete responses.
  • Example 11 CAR-engager containing immune cell effector domain IL-2 variants
  • the binding strength of the IL-2 variant to the CAR immune cell may have significant impacts on the efficacy of CAR-engager therapy.
  • CAR-engagers IL-2 variant containing different IL-2 variants were generated and integrated into CAR-engagers and are summarized in Table 9. Amino acid substitutions are relative to the natural (wild-type) sequence of human IL-2 (NCBI Accession No. NP_000577; SEQ ID NO: 102).
  • some of the CAR-engagers contain different dimerization domains, including CH3, and silent fragment crystallizable region (Fc).
  • Silent Fc contains at least two mutations relative to wild-type Fc that abolish Fc- ⁇ receptor and complement component 1q (C1q) binding, while maintaining the stabilizing effect and neonatal Fc receptor (FcRn) binding of wild-type Fc.
  • the at least two mutations include L234A and L235A, and are commonly referred to as (“LALA”).
  • CAR-engager variant E4 has a dimerization domain containing IgG1 with the silent Fc mutations of L234A and L235A (“LALA”) and further lacks an IL-2 variant.
  • the CAR-engager variant E4 was developed and used as a control molecule.
  • CAR-engagers containing a single IL-2 variant are listed in Table 9 as “One-IL-2” while CAR-engagers containing two repeats of IL-2 variants are listed in the below table as “IL-2.”
  • variants Q3, A8, A10, and A16 have two repeats of the indicated IL-2 variant on each molecule of CAR-engager.
  • the CAR-engager may be present as a dimer due to the presence of a dimerization domain.
  • CAR-engagers containing a wild-type CD19 SEQ ID NO: 5
  • CD19 wt wild-type CD19
  • CAR-engagers containing the variant of CD19 that has the amino acid sequence of SEQ ID NO: 3, are listed in Table 9 as “CD19.”
  • Table 1 CAR-engager ICE Variants
  • Variant CAR-engager structure IL-2 amino acid substitutions relative Activity 95 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024
  • FIG. 22A shows the percentage of BCMA CAR T cells stained positive for phosphorylated STAT5 (pSTAT5) after treatment by select CAR-engager ICE variants described in Table 9, as determined by flow cytometry.
  • FIG. 22B shows dose-dependent activation of rested BCMA CAR T cells after CAR- engager treatment after 24 hours.
  • CD69 serves as an activation marker for human T cells.
  • the dashed line at 0 nM represents the background expression of CD69 in the absence of any treatment or cytokines.
  • the dashed line “+Cytokine” indicates the CD69 expression 96 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 level of non-rested CAR T cells incubated in the presence of IL-2, IL-7, and IL-15 cytokines. The dashed lines are included as additional negative and positive controls.
  • FIG.23A shows STAT5 phosphorylation in BCMA CAR T cells after CAR-engager ICE variant treatment.
  • Several of the IL-2 variants exhibited the ability to induce STAT5 activity in BCMA CAR T cells.
  • FIGs. 24A –24B show STAT5 phosphorylation in BCMA CAR T cells induced by treatment with the BCMA CAR-engagers detailed in Table 9.
  • the CAR T cells contain the FDA- approved BCMA CAR constructs idecabtagene vicleucel (Ide-cel; ABECMA®) or ciltacabtagene autoleucel (Cilta-cel; CARVYKTI®).
  • the percentage of BCMA CAR T cells stained positive for phosphorylated STAT5 (pSTAT5) as determined by flow cytometry are shown.
  • FIGs. 25A –25F show STAT5 phosphorylation in BCMA CAR T cells induced by treatment with the BCMA-muIL2 CAR-engagers detailed in Table 9.
  • the CAR T cells contain the FDA-approved BCMA CAR constructs Ide-cel or Cilta-cel.
  • the percentage of BCMA CAR T cells stained positive for pSTAT5 as determined by flow cytometry are shown.
  • FIGs. 26A – 26B show dose-dependent activation of rested BCMA CAR T cells by treatment with the BCMA CAR-engagers detailed in Table 9.
  • the CAR T cells contain the FDA- approved BCMA CAR constructs Ide-cel or Cilta-cel.
  • FIGs. 27A – 28B show dose-dependent activation of rested BCMA CAR T cells after treatment with additional BCMA CAR-engagers detailed in Table 9.
  • the CAR T cells contain the FDA-approved BCMA CAR constructs Ide-cel or Cilta-cel.
  • FIGs.29A – 29C show that select CAR-engagers induce CAR T cell proliferation in vitro with reduced impact on non-transduced T cells as compared to wild-type IL-2.
  • FIG. 29A shows incubation of activated BCMA CAR T cells containing the Cilta-cel CAR construct, treated with increasing concentrations of BCMA CAR-engager, followed by assessment of CAR T cell proliferation using flow cytometry after 3 days.
  • the dashed line in FIG.29A represents the number of CAR T cells that received no treatment.
  • FIG.29B shows a repetition of the experiment show in FIG. 29A with additional CAR-engagers.
  • FIG. 29A shows incubation of activated BCMA CAR T cells containing the Cilta-cel CAR construct, treated with increasing concentrations of BCMA CAR-engager, followed by assessment of CAR T cell proliferation using flow cytometry after 3 days.
  • the dashed line in FIG.29A represents the number of CAR T cells that received no treatment
  • 29C shows a similar experiment evaluating the impact of CAR-engager on the cellular proliferation of normal, non-transduced T cells.
  • CAR- 98 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 engagers induce proliferation in non-transduced T cells significantly less than wild-type IL-2.
  • CAR- engagers containing the triple mutant IL-2 (V7, V9, Y2, and Y9) induced significantly less proliferation in normal T cells as compared to CAR-engagers containing the double-mutant IL-2 (U4).
  • FIG.42 shows that CAR-engagers induce CAR T cell proliferation in vitro with no little to no impact on non-transduced T cells.
  • BCMA CAR-engagers containing immune cell effector domains containing a U4, V7, Y2, V6, X12, X15, or U8 IL-2 variant were examined for their ability to induce proliferation of anti-BCMA CAR T cells or non-transduced T cell controls.
  • Wild-type IL- 2 a non-targeting CAR-engager with an immune cell effector domain containing E3 IL-2 variant (VHH-cH3-IL2), and a BCMA CAR-engager without an immune cell effector domain (BCMA- CH3) were used as controls.
  • FIGs. 30A – 30C show that the V7 CAR-engager exhibits lower affinity for IL-2R ⁇ as compared to wild-type IL-2 or the U4 CAR-engager.
  • BLI analyses of association and dissociation between wild-type IL-2 (Teceleukin®), BCMA-muIL2 CAR-engager (U4; double mutant IL-2), or BCMA-muIL2 CAR-engager (V7; triple mutant IL-2) molecules and IL-2R ⁇ are shown.
  • FIGs. 31A – 31B show the newly developed CAR-engager containing IL-2 variants induce proliferation of CAR-T cells in vivo, and that the proliferation levels correlate with in vitro pSTAT5 signaling.
  • FIG. 31A schematically illustrations the experimental setup; mice received OPM2 cancer cells, followed by 0.5 million CAR-T cells (Cilta-cel) one week later. Mice were bled for 5 weeks on days 11, 18, 28, 35, and 42 and CD4 and CD8 CAR T-cells were counted (FIG. 31B).
  • FIGs. 32A – 32C show that the V7 CAR-engager enhances CAR-T cell activity when administered 3 or even 14 days post-injection of CAR T cells.
  • Another mouse cohort received 6 doses of V7 CAR-engager starting from 3 days post-injection of CAR T cells, 99 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 administered on days 3, 6, 10, 14, 21, 28.
  • the third mouse cohort received 6 doses of CAR-engagers starting from day 14, which is the post-CRS window in patients, and on days 14, 18, 21, 28, 35, 42.
  • BLI imaging was performed on the indicated days to assess tumor growth in the different cohorts (FIG.32B). Mice were bled for 6 weeks, once per week starting from day 14, and CAR T cells were counted (FIG. 32C).
  • FIGs.43 – 45 show that CAR-engagers containing IL-2 variants induce in vitro pSTAT5 signaling (FIG. 43) and gene expression (FIGs. 44 and 45).
  • CAR T cells (about 60% of CAR + T cells) and non-transduced T cells were incubated with increasing concentrations of CAR-engagers containing an immune effector domain containing an IL-2 variant (including U4, V7, Y2, V6, X12, X15, U8) and controls (wild-type IL-2 and a CAR-engager without an immune effector domain (BCMA-CH3)).
  • pSTAT5 activity was assessed by flow cytometry after 30 minutes of CAR-engager treatment (FIG. 43).
  • CD69 expression was assessed by flow cytometric analysis after a 2-hour incubation of T cells with CAR-engagers, followed by a wash, and a further 22-hour incubation (FIG.44).
  • TNF- ⁇ and IFN- ⁇ production was assessed by multiplex ELISA via flow cytometry after a 2-hour incubation of T cells with CAR-engagers, followed by a wash, and a further 22-hour incubation and media harvesting (FIG.45). Each condition included 3 technical replicates. T cells were obtained from PBMCs from a single healthy donor.
  • FIGs.46A – 46B show that CAR-engagers containing IL-2 variants bind to IL-2R ⁇ and IL-2R ⁇ by biolayer interferometry.
  • a biosensor tip was coated with dimeric IL-2R ⁇ (FIG.46A) or IL-2R ⁇ (FIG.46B), and incubated with CAR-engagers containing an immune effector domain containing an IL-2 variant (including U4, V7, Y2, V6, X12, X15, U8) or controls (monomeric wild-type IL-2 (positive control), CAR-E with wild-type IL-2 (positive control), and a CAR-engager with monomeric V7 IL-2 variant)) for the indicated times.
  • the release rate was assessed by transferring the biosensor tip into the washing solution. Each condition included 3 technical replicates.
  • Example 12 CD19 CAR-E does not inhibit killing efficacy of CD19 CAR T cells
  • CAR-engagers that bind BCMA were observed to bind but not inhibit killing efficacy of BCMA CAR T cells (FIGs.2D – 2H).
  • a killing assay was conducted using CD19 CAR T cells 100 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 and patient-derived CD19 + leukemia cells in the presence of varying concentrations of the CD19 CAR-engager.
  • Live (PI-) Nalm6 cells were counted 48 hours later, with an N of 3 for each of the experiments.
  • the experimental findings disclosed herein with CD19 are consistent with the findings in BCMA cancer models and BCMA CAR-E, above.
  • Example 13 CAR-engagers effectively enhance CAR immune cell efficacy and persistence at low doses on initial treatment and tumor rechallenge [000361]
  • Mice were intravenously injected with OPM2 cells (1 ⁇ 10 6 , i.v.). A week later, CAR T cells (0.5 ⁇ 10 6 , i.v.) were administered. Two weeks post-injection of CAR T cells, the mice were divided into two cohorts, with mice receiving the CAR-E treatment or not. The treatment group received four doses of CAR-E treatment (4 mg/kg per dose) on days 14, 18, 21, and 28.
  • Cytokine assessments also revealed elevated levels of IFN- ⁇ in the CAR-E treatment cohort by sandwich ELISA on 1:40 diluted serum samples collected the same days when CAR-T counts were assessed as shown FIG.22E (FIG.35F).
  • One of the CAR-E treated mice (M5) showed liver relapse on day 60 (FIG. 35B). Regardless, all the five CAR-E treated mice underwent re-challenge, using 1 million of the liver metastasis-derived OPM2 cells, on day 60 to assess the generation of memory CAR-T cells. All mice showed considerably less signal compared to the na ⁇ ve control mice (FIG.35B; see day 66).
  • mice While the mice exhibited liver signals, none showed bone marrow signals, suggesting the presence 101 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 of functional memory CAR T cells in the bone marrow inhibiting tumor growth.
  • CAR-E treatment could contribute to re-expanding CAR T cells and controlling tumor growth in liver metastasis, the mice were re-treated with CAR-E (4 mg/kg) on days 68, 70, 74, 77, and 80. Impressively, all mice successfully cleared the liver metastasis, indicating that CAR-E could facilitate the re-expansion and trafficking of CAR T cells to eliminate tumor cells (FIG. 35B; see day 83).
  • mice that showed relapse on day 60 also cleared the tumor from liver after re-challenge, although it exhibited some signal on the last day of the experiment.
  • Example 14 CAR T cell expansion in the absence of tumor antigens is dose-dependent.
  • CAR T cell expansion occurs post-infusion in patients, with peak expansion observed around 10-14 days post-infusion (Rodriguez-Otero et al., N. Engl. J. Med.388(11):1002- 1014 (2023)). This expansion is driven by antigen availability and the tumor-killing process, which promotes CAR T cell proliferation (Turtle et al., J. Clin.
  • the CAR-E mechanism of action may be independent of tumor cells and antigens presented thereon and may thus expand CAR T cells in the absence of tumor cells (and therefore tumor antigen), addressing the critical clinical challenge of limited in vivo CAR T cell expansion post-infusion.
  • mice were injected solely with 0.25 million BCMA CAR T cells in the absence of tumor cells.
  • Organs were collected one-month post-injection of CAR T cells, and flow cytometric analyses were conducted to assess the presence of CAR T cells. [000369] Further, the results demonstrated that CAR-E impact on CAR T cells is dose-dependent (FIGs.36B – 36C).
  • TEM cells 103 Include Draft Include Date Include Time VIA EFS Attorney Docket No.046094-788001WO Date of Deposit: September 11, 2024 are CD45RA- CD45RO + CCR7-; TEMRA cells are CD45RA + CD45RO + CCR7-; TSCM cells are CD45RA + CD45RO + CCR7 + ; TCM cells are CD45RA- CD45RO + CCR7 + ; TNaive cells are CD45RA + CD45RO- CCR7 + .
  • the experiment in panels FIGs. 36A – 36E utilized PBMCs from one donor. Overall, these findings demonstrate that CAR-E treatment induces the expansion of CAR T cells and enables the CAR T cells develop diverse memory phenotypes, regardless of the presence of tumor cells.
  • the antigen-only, BCMA-CH3 treatment cohort did not result in the expansion or persistence of CAR T cells compared to the CAR-E treatment cohort, further validating that both the ectodomain (e.g., BCMA antigen) component and the immune cell effector domain (e.g., low-affinity IL-2) component of the CAR-E molecule are necessary for its impact on CAR T cells (FIG.36F – 36G and 41A – 41E).
  • Mice received CAR T cells and different treatments (4 mg/kg) following a schedule similar to that shown in FIG. 36A.
  • the BCMA-CH3 antigen, VHH-muIL2, or the low-dose wild-type IL-2 treatments did not result in expansion or persistence of CAR T cells compared to the CAR-engager-treated cohort. Error bars represent mean with standard deviation.
  • the experiment in panels FIGs.36F – 36G utilized PBMCs from two donors. The Kruskal-Wallis test was used for each subset of CAR T cells and total T cells. Subsequently, post-hoc Dunn’s analysis was conducted to compare each group with the treatment group. The table in FIG.23G displays the adjusted p-values. This aligns with the in vitro and in vivo analyses disclosed herein.
  • Example 15 The efficacy of the CAR-E requires signaling through both the CAR and the IL-2R intracellular signaling domains [000373] To better understand the mechanism of action of CAR-E, it was investigated whether the effects on CAR T cells are solely mediated by anchoring the low-affinity IL-2 onto CAR T cells through BCMA-to-CAR binding or if it involves simultaneous engagement of both IL-2R and CAR intracellular signaling domains. For this purpose, BCMA CAR T cells were made using a BCMA CAR construct lacking the 41BB-CD3 ⁇ intracellular signaling domain but retaining an identical extracellular ectodomain (referred to as CAR-Intracellular domain deletion, or CAR-ICD- ⁇ ).
  • Dasatinib a lymphocyte cell-specific protein-tyrosine kinase (LCK) inhibitor, and Ruxolitinib, a Janus kinase (JAK) inhibitor, both individually and in combination, significantly inhibited the impact of the CAR-E molecule on CAR T cells (FIGs.33E – 33G), further suggesting that the CAR-E molecules engages both CAR and IL-2R receptors and activating their intracellular signaling pathways.
  • LCK lymphocyte cell-specific protein-tyrosine kinase
  • Ruxolitinib a Janus kinase (JAK) inhibitor
  • CAR-ICD- ⁇ T cells were treated with the BCMA-muIL2 CAR-E molecule. Treatments were removed after 2 hours to mimic in vivo conditions, and cells were subjected to bulk RNA-sequencing either 2 or 22 hours later. [000376] Remarkably, the CAR-E induced substantial transcriptomic changes, demonstrating quantitatively larger fold-changes compared to all other conditions, including wild-type IL-2 (FIGs.33J – 33N).
  • GSEA gene set enrichment analysis

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Toxicology (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Plant Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne de nouveaux variants d'IL-2 à faible affinité et leurs utilisations, y compris une thérapie cellulaire adoptive par amplification de la puissance et de la durée de cellules immunitaires CAR pour traiter le cancer.
PCT/US2024/046173 2023-09-11 2024-09-11 Activateur car contenant des variants d'il-2 pour améliorer la fonctionnalité de cellules car-t Pending WO2025059162A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202363537660P 2023-09-11 2023-09-11
US63/537,660 2023-09-11
US202463664020P 2024-06-25 2024-06-25
US63/664,020 2024-06-25

Publications (1)

Publication Number Publication Date
WO2025059162A1 true WO2025059162A1 (fr) 2025-03-20

Family

ID=92931887

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/046173 Pending WO2025059162A1 (fr) 2023-09-11 2024-09-11 Activateur car contenant des variants d'il-2 pour améliorer la fonctionnalité de cellules car-t

Country Status (1)

Country Link
WO (1) WO2025059162A1 (fr)

Citations (148)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US183076A (en) 1876-10-10 Improvement in bias-measures
US5169936A (en) 1989-04-14 1992-12-08 Biogen, Inc. Protein purification on immobilized metal affinity resins effected by elution using a weak ligand
US5665577A (en) 1989-02-06 1997-09-09 Dana-Farber Cancer Institute Vectors containing HIV packaging sequences, packaging defective HIV vectors, and uses thereof
US5821333A (en) 1995-03-01 1998-10-13 Genetech, Inc. Method for making heteromultimeric polypeptides
US5906936A (en) 1988-05-04 1999-05-25 Yeda Research And Development Co. Ltd. Endowing lymphocytes with antibody specificity
US5981276A (en) 1990-06-20 1999-11-09 Dana-Farber Cancer Institute Vectors containing HIV packaging sequences, packaging defective HIV vectors, and uses thereof
US6013516A (en) 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US20050079170A1 (en) 2001-09-14 2005-04-14 Fabrice Le Gall Dimeric and multimeric antigen binding structure
US20050287148A1 (en) 1995-01-17 2005-12-29 Malaya Chatterjee Monoclonal antibody 1A7 and use for the treatment of melanoma and small cell carcinoma
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
US7090837B2 (en) 2003-01-21 2006-08-15 The Salk Institute For Biological Studies Compositions and methods for tissue specific targeting of lentivirus vectors
US20080090995A1 (en) 2005-02-14 2008-04-17 Ge Healthcare Bio-Sciences Ab Liquid Chromatography Method
US7375234B2 (en) 2002-05-30 2008-05-20 The Scripps Research Institute Copper-catalysed ligation of azides and acetylenes
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
US20090069561A1 (en) 2004-06-30 2009-03-12 The Scripps Research Institute Click chemistry route to triazole dendrimers
US7741465B1 (en) 1992-03-18 2010-06-22 Zelig Eshhar Chimeric receptor genes and cells transformed therewith
US20110294148A1 (en) 1999-04-09 2011-12-01 Hvidovre Hospital Tissue inhibitor of matrix metalloproteinases type-1 (timp-1) as a cancer marker and postoperative marker for minimal residual disease or recurrent disease in patients with a prior history of cancer
US8101238B2 (en) 2004-07-22 2012-01-24 The Scripps Research Institute Polymeric materials via click chemistry
US8119119B2 (en) 2004-06-25 2012-02-21 Centre National De La Recherche Scientifique Non-integrative and non-replicative lentivirus, preparation and uses thereof
US8124353B2 (en) 2006-07-14 2012-02-28 Ac Immune S.A. Methods of treating and monitoring disease with antibodies
US20120244075A1 (en) 2011-03-24 2012-09-27 Nanoprobes, Inc. 5 NM Nickel-NTA-Gold Nanoparticles
US20120321637A1 (en) 2011-06-20 2012-12-20 The Board Of Regents Of The University Of Texas System Combination cancer therapy with herv inhibition
US8357778B2 (en) 2007-05-21 2013-01-22 Nomadic Bioscience Co., Ltd. Polypeptide, an affinity chromatography material, and a method for separating and/or purifying immunoglobulin
US8372986B2 (en) 2005-09-30 2013-02-12 The Scripps Research Institute Ruthenium-catalyzed cycloaddition of alkynes and organic azides
US8389282B2 (en) 2007-03-30 2013-03-05 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred T lymphocytes
US8394914B2 (en) 2007-08-24 2013-03-12 Board Of Trustees Of Michigan State University Functional polyglycolide nanoparticles derived from unimolecular micelles
US8399645B2 (en) 2003-11-05 2013-03-19 St. Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
US20130089554A1 (en) 2009-12-29 2013-04-11 Emergent Product Development Seattle, Llc RON Binding Constructs and Methods of Use Thereof
US20140024111A1 (en) 2008-01-07 2014-01-23 Amgen Inc. Host cell for making antibody fc-heterodimeric molecules using electrostatic steering effects
US8642745B2 (en) 1997-05-02 2014-02-04 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US20140194442A1 (en) 2011-01-12 2014-07-10 Boehringer Ingelheim International Gmbh Anticancer therapy
US8877170B2 (en) 2009-02-21 2014-11-04 Sofradim Production Medical device with inflammatory response-reducing coating
US8927736B2 (en) 2009-08-11 2015-01-06 The Scripps Research Institute Copper catalyzed cycloaddition of organic azides and 1-haloalkynes
US8927682B2 (en) 2007-08-24 2015-01-06 Board Of Trustees Of Michigan State University Functionalization of polyglycolides by “click” chemistry
US20150175711A1 (en) 2012-06-15 2015-06-25 Sinomab Bioscience Limited Anti-cd22 anti-idiotypic antibodies and uses thereof
US9101622B2 (en) 2002-05-17 2015-08-11 Celgene Corporation Methods for treating newly diagnosed multiple myeloma 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione in combination with dexamethasone
US9139649B2 (en) 2014-02-25 2015-09-22 Immunomedics, Inc. Humanized anti-CD22 antibody
US9181343B2 (en) 2012-07-19 2015-11-10 Redwood Bioscience Inc. Antibody specific for CD22 and methods of use thereof
US9248182B2 (en) 2012-04-20 2016-02-02 Merus B.V. Methods and means for the production of Ig-like molecules
US9309311B2 (en) 2009-04-27 2016-04-12 Oncomed Pharmaceuticals, Inc. Method for making Heteromultimeric molecules
US20160131655A1 (en) 2011-04-21 2016-05-12 Boehringer Ingelheim International Gmbh Bcma-based stratification and therapy for multiple myeloma patients
US9422351B2 (en) 2011-11-03 2016-08-23 The Trustees Of The University Of Pennsylvania Isolated B7-H4 specific compositions and methods of use thereof
US9528160B2 (en) 2008-11-07 2016-12-27 Adaptive Biotechnolgies Corp. Rare clonotypes and uses thereof
US9527927B2 (en) 2011-12-20 2016-12-27 Medimmune, Llc Modified polypeptides for bispecific antibody scaffolds
US9562109B2 (en) 2010-11-05 2017-02-07 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US9629877B2 (en) 2013-05-14 2017-04-25 Board Of Regents, The University Of Texas System Human application of engineered chimeric antigen receptor (CAR) T-cells
US9630165B2 (en) 2014-01-17 2017-04-25 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
US20170158760A1 (en) 2008-10-20 2017-06-08 Abbvie Inc. Isolation and Purification of Antibodies Using Protein A Affinity Chromatography
US9701758B2 (en) 2013-05-24 2017-07-11 Board Of Regents, The University Of Texas System Anti-CD19 scFv (FMC63) polypeptide
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US20170226216A1 (en) 2014-07-24 2017-08-10 Bluebird Bio, Inc. Bcma chimeric antigen receptors
US9790267B2 (en) 2011-11-08 2017-10-17 The Trustees Of The University Of Pennsylvania Glypican-3-specific antibody and uses thereof
US20170296623A1 (en) 2014-12-17 2017-10-19 Cellectis INHIBITORY CHIMERIC ANTIGEN RECEPTOR (iCAR OR N-CAR) EXPRESSING NON-T CELL TRANSDUCTION DOMAIN
US20170335281A1 (en) 2014-03-15 2017-11-23 Novartis Ag Treatment of cancer using chimeric antigen receptor
US9885298B2 (en) 2015-10-15 2018-02-06 Toyota Jidosha Kabushiki Kaisha Fuel injection control device for engine
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US9896547B2 (en) 2012-12-03 2018-02-20 The Scripps Research Institute Polymerization method and polymers formed therewith
US20180086843A1 (en) 2012-04-26 2018-03-29 Bioatla, Llc Anti-cd22 antibodies
US20180133296A1 (en) 2015-04-17 2018-05-17 David Maxwell Barrett Methods for improving the efficacy and expansion of chimeric antigen receptor?expressing cells
WO2018119114A1 (fr) * 2016-12-22 2018-06-28 Cue Biopharma, Inc. Polypeptides multimères modulateurs des lymphocytes t et leurs méthodes d'utilisation
US20180187149A1 (en) 2015-06-25 2018-07-05 Icell Gene Therapeutics Llc CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS OF USE THEREOF
US10023608B1 (en) 2013-03-13 2018-07-17 Amgen Inc. Protein purification methods to remove impurities
US10072088B2 (en) 2015-08-17 2018-09-11 Janssen Pharmaceutica, Nv Anti-BCMA antibodies and uses thereof
WO2018170168A1 (fr) * 2017-03-15 2018-09-20 Cue Biopharma, Inc. Procédés pour moduler une réponse immunitaire
US20180296689A1 (en) 2009-02-13 2018-10-18 Immunomedics, Inc. Neoadjuvant use of antibody-drug conjugates
US10124023B2 (en) 2013-02-26 2018-11-13 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
US10138303B2 (en) 2010-05-14 2018-11-27 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
US10189903B2 (en) 2012-02-13 2019-01-29 Seattle Children's Hospital Bispecific chimeric antigen receptors and methods of use thereof to treat cancer
US10207229B2 (en) 2012-03-16 2019-02-19 Board Of Trustees Of Michigan State University Functionalization of a porous membrane with an adsorbed polyacid
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells
US10280462B2 (en) 2002-10-11 2019-05-07 Jacobus Johannes Maria van Dongen Nucleic acid amplification primers for PCR-based clonality studies
US20190135894A1 (en) 2015-06-25 2019-05-09 iCell Gene Therapeuticics LLC COMPOUND CHIMERIC ANTIGEN RECEPTOR (cCAR) TARGETING MULTIPLE ANTIGENS, COMPOSITIONS AND METHODS OF USE THEREOF
US20190135937A1 (en) 2017-11-03 2019-05-09 Sorrento Therapeutics, Inc. CD-38 Directed Chimeric Antigen Receptor Constructs
US20190151365A1 (en) 2016-07-28 2019-05-23 Novartis Ag Combination therapies of chimeric antigen receptors and pd-1 inhibitors
US20190276492A1 (en) 2012-02-15 2019-09-12 Hoffmann-La Roche Inc. Fc-receptor based affinity chromatography
US10421817B1 (en) 2019-01-17 2019-09-24 Beijing Mabworks Biotech Co., Ltd. Antibodies binding human Claudin 18.2 and uses thereof
US10442867B2 (en) 2017-07-31 2019-10-15 Lentigen Technology, Inc. Compositions and methods for treating cancer with anti-CD19/CD20 immunotherapy
US20190359727A1 (en) 2017-02-17 2019-11-28 Fred Hutchinson Cancer Research Center Combination therapies for treatment of bcma-related cancers and autoimmune disorders
US10494435B2 (en) 2008-04-04 2019-12-03 The Government Of The United States America As Represented By The Secretary Of The Department Of Health And Human Services Human monoclonal antibodies specific for CD22
US20190375815A1 (en) 2017-01-31 2019-12-12 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
US20190381171A1 (en) 2017-02-17 2019-12-19 Unum Therapeutics Inc. Co-use of anti-bcma antibody and antibody-coupled t cell receptor (actr) in cancer therapy and b cell disorders
US10533055B2 (en) 2014-08-28 2020-01-14 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for CD19
US20200024342A9 (en) 2016-06-24 2020-01-23 Icell Gene Therapeutics Llc CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS THEREOF
US10550179B2 (en) 2017-01-09 2020-02-04 Lentigen Technology Inc. Compositions and methods for treating cancer with anti-mesothelin immunotherapy
US20200048359A1 (en) 2017-02-28 2020-02-13 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
US20200055948A1 (en) 2017-04-28 2020-02-20 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
US20200071397A1 (en) 2018-05-31 2020-03-05 Washington University Chimeric antigen receptor t cells (car-t) for the treatment of cancer
US10640569B2 (en) 2013-12-19 2020-05-05 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
US20200155520A1 (en) 2015-11-19 2020-05-21 Genentech, Inc. Methods of treating cancer using b-raf inhibitors and immune checkpoint inhibitors
US10683369B2 (en) 2015-08-03 2020-06-16 Engmab Sàrl Monoclonal antibodies against BCMA
US10709775B2 (en) 2015-08-11 2020-07-14 Cellectis Cells for immunotherapy engineered for targeting CD38 antigen and for CD38 gene inactivation
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
US20200255803A1 (en) 2017-09-27 2020-08-13 Gracell Biotechnologies (Shanghai) Co., Ltd. Engineered immune cell capable of inducing secretion of anti-cd47 antibody
US20200281974A1 (en) 2017-09-26 2020-09-10 Longwood University Pd1-specific chimeric antigen receptor as an immunotherapy
US20200281973A1 (en) 2016-03-04 2020-09-10 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
US20200308541A1 (en) 2017-10-12 2020-10-01 Icell Gene Therapeutics, Llc Compound Chimeric Antigen Receptor (CCAR) Targeting Multiple Antigents, Compositions and Methods of Use Thereof
US10799536B2 (en) 2016-12-09 2020-10-13 Onk Therapeutics Limited Method of treating multiple myeloma using natural killer cells expressing a chimeric antigen receptor for CD38
US10815301B2 (en) 2015-10-15 2020-10-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Service Anti-CD30 chimeric antigen receptors
US20200339699A1 (en) 2018-02-01 2020-10-29 Innovent Biologics (Suzhou) Co., Ltd. Fully humanized anti-b cell maturation antigen (bcma) single-chain antibody and use thereof
US10836998B2 (en) 2014-02-14 2020-11-17 Cellectis Cells for immunotherapy engineered for targeting antigen present both on immune cells and pathological cells
US20200360431A1 (en) 2017-11-15 2020-11-19 Novartis Ag Bcma-targeting chimeric antigen receptor, cd19-targeting chimeric antigen receptor, and combination therapies
WO2020252418A2 (fr) * 2019-06-14 2020-12-17 Cugene, Inc. Nouveaux variants d'interleukines-2 pour le traitement du cancer
US20200392248A1 (en) 2018-02-23 2020-12-17 Beijing Meikang Geno-Immune Biotechnology Co., Ltd. A cd19-based chimeric antigen receptor and application thereof
US20210047402A1 (en) 2019-06-14 2021-02-18 Teneobio, Inc. Multispecific heavy chain antibodies binding to cd22 and cd3
US10934363B2 (en) 2015-08-11 2021-03-02 Legend Biotech Usa Inc. Chimeric antigen receptors based on single domain antibodies and methods of use thereof
US20210061877A1 (en) 2014-12-19 2021-03-04 Dana-Farber Cancer Institute, Inc. Chimeric antigen receptors and methods of use thereof
US20210079057A1 (en) 2017-06-13 2021-03-18 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US10954530B2 (en) 2015-03-02 2021-03-23 Autolus Limited Retroviral and lentiviral vectors
US20210095022A1 (en) 2017-12-22 2021-04-01 TaneoBio, Inc. Heavy chain antibodies binding to cd22
US11028185B2 (en) 2011-06-28 2021-06-08 Whitehead Institute For Biomedical Research Using sortases to install click chemistry handles for protein ligation
US20210169852A1 (en) 2018-07-27 2021-06-10 Daiichi Sankyo Company, Limited Protein recognizing drug moiety of antibody-drug conjugate
US20210206815A1 (en) 2016-08-11 2021-07-08 Repligen Corporation Alkaline stable fc-binding proteins for affinity chromatography
US11066457B2 (en) 2017-02-08 2021-07-20 Cellular Biomedicine Group Hk Limited Construction of chimeric antigen receptor targeting CD20 antigen and activity identification of engineered T cells thereof
US20210230548A1 (en) 2018-05-03 2021-07-29 Board Of Regents, The University Of Texas System Natural killer cells engineered to express chimeric antigen receptors with immune checkpoint blockade
US11091588B2 (en) 2015-04-24 2021-08-17 The Penn State Research Foundation Clickable waterborne polymers and click-crosslinked waterborne polymers, clickable functional compounds, click functionalized waterborne polymers, and uses thereof
US20210253729A1 (en) 2017-07-25 2021-08-19 Board Of Regents, The University Of Texas System Enhanced chimeric antigen receptors and use thereof
US11098118B2 (en) 2018-05-18 2021-08-24 Lanova Medicines Limited Company Anti-claudin 18.2 antibodies and uses thereof
US20210300986A1 (en) 2017-12-05 2021-09-30 The Medical Research Infrastrutrure And Health Services Fund Of The Tel Aviv Medecal Center T-cells comprising two different chimeric antigen receptors and uses thereof
US11136392B2 (en) 2015-06-23 2021-10-05 Memorial Sloan-Kettering Cancer Center PD-1 immune modulating agents
US11160833B2 (en) 2014-12-15 2021-11-02 The Regents Of The University Of California Bispecific OR-gate chimeric antigen receptor responsive to CD19 and CD20
US11168344B2 (en) 2005-03-31 2021-11-09 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US20220056101A1 (en) 2018-11-27 2022-02-24 Duke University Anti-lmp2 tcr-t cell therapy for the treatment of ebv-associated cancers
US20220073643A1 (en) 2018-12-28 2022-03-10 Nanjing GenScript Biotech Co., Ltd. Claudin18.2 binding moieties and uses thereof
WO2022087149A2 (fr) * 2020-10-22 2022-04-28 Gilead Sciences, Inc. Protéines de fusion d'interleukine-2-fc et méthodes d'utilisation
US11352460B2 (en) 2019-11-25 2022-06-07 Covestro Llc Waterborne azido-alkyne click compositions
WO2022125711A1 (fr) * 2020-12-09 2022-06-16 Asher Biotherapeutics, Inc. Construction de cytokine ciblée pour la thérapie cellulaire génétiquement modifiée
US11365394B2 (en) 2017-12-22 2022-06-21 Fate Therapeutics, Inc. Enhanced immune effector cells and use thereof
US20220193138A1 (en) 2019-04-25 2022-06-23 Purdue Research Foundation Engineered natural killer cells redirected toward purinergic signaling, constructs thereof, and methods for using the same
US11369703B2 (en) 2018-08-31 2022-06-28 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
US11390668B2 (en) 2009-10-20 2022-07-19 Abbvie Inc. Isolation and purification of anti-IL-13 antibodies using protein a affinity chromatography
US20220273710A1 (en) 2018-05-15 2022-09-01 Autolus Limited A cd79-specific chimeric antigen receptor
US11433100B2 (en) 2020-08-20 2022-09-06 A2 Biotherapeutics, Inc. Compositions and methods for treating ceacam positive cancers
US11439665B2 (en) 2020-03-17 2022-09-13 Cellular Biomedicine Group Hk Limited Combined chimeric antigen receptor targeting CD19 and CD20 and application thereof
US11485782B2 (en) 2018-03-14 2022-11-01 Beijing Xuanyi Pharmasciences Co., Ltd. Anti-claudin 18.2 antibodies
US20220380852A1 (en) 2019-08-27 2022-12-01 Fundación Para La Investigación Biomédica Del Hospital Universitario 12 De Octubre Method for determining the presence or absence of minimal residual disease (mrd) in a subject who has been treated for a disease
US11541127B2 (en) 2015-04-15 2023-01-03 Astellas Pharma, Inc. Drug conjugates comprising antibodies against claudin 18.2
US11602525B2 (en) 2014-04-25 2023-03-14 Rinat Neuroscience Corp. Antibody-drug conjugates with high drug loading
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
US11633426B2 (en) 2014-10-20 2023-04-25 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
US11648268B2 (en) 2015-12-09 2023-05-16 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of using same
US20230192840A1 (en) 2018-12-28 2023-06-22 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Antibody and use thereof
US11702472B2 (en) 2014-06-06 2023-07-18 Memorial Sloan-Kettering Cancer Center Method of reducing mesothelin-expressing tumor burden by administration of T cells comprising mesothelin-targeted chimeric antigen receptors
US11713346B2 (en) 2015-05-11 2023-08-01 Biontech Cell & Gene Therapies Gmbh Claudin-18.2-specific immunoreceptors and T cell epitopes
US20230242877A1 (en) 2020-01-21 2023-08-03 Nanjing Bioheng Biotech Co., Ltd Immune cell comprising chimeric antigen receptor and use thereof
US11768203B2 (en) 2016-03-31 2023-09-26 University Of Southern California Highly sensitive and specific luciferase based reporter assay for antigen detection
WO2024159087A1 (fr) * 2023-01-26 2024-08-02 Dana-Farber Cancer Institute, Inc. Développement d'une plateforme d'activateurs-récepteurs chimériques de l'antigène (car) pour améliorer la fonctionnalité et/ou la persistance de lymphocytes t car

Patent Citations (155)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US183076A (en) 1876-10-10 Improvement in bias-measures
US5906936A (en) 1988-05-04 1999-05-25 Yeda Research And Development Co. Ltd. Endowing lymphocytes with antibody specificity
US5665577A (en) 1989-02-06 1997-09-09 Dana-Farber Cancer Institute Vectors containing HIV packaging sequences, packaging defective HIV vectors, and uses thereof
US5169936A (en) 1989-04-14 1992-12-08 Biogen, Inc. Protein purification on immobilized metal affinity resins effected by elution using a weak ligand
US5981276A (en) 1990-06-20 1999-11-09 Dana-Farber Cancer Institute Vectors containing HIV packaging sequences, packaging defective HIV vectors, and uses thereof
US7741465B1 (en) 1992-03-18 2010-06-22 Zelig Eshhar Chimeric receptor genes and cells transformed therewith
US20050287148A1 (en) 1995-01-17 2005-12-29 Malaya Chatterjee Monoclonal antibody 1A7 and use for the treatment of melanoma and small cell carcinoma
US5821333A (en) 1995-03-01 1998-10-13 Genetech, Inc. Method for making heteromultimeric polypeptides
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6013516A (en) 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
US8642745B2 (en) 1997-05-02 2014-02-04 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US20110294148A1 (en) 1999-04-09 2011-12-01 Hvidovre Hospital Tissue inhibitor of matrix metalloproteinases type-1 (timp-1) as a cancer marker and postoperative marker for minimal residual disease or recurrent disease in patients with a prior history of cancer
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
US20050079170A1 (en) 2001-09-14 2005-04-14 Fabrice Le Gall Dimeric and multimeric antigen binding structure
US9101622B2 (en) 2002-05-17 2015-08-11 Celgene Corporation Methods for treating newly diagnosed multiple myeloma 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione in combination with dexamethasone
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
US7375234B2 (en) 2002-05-30 2008-05-20 The Scripps Research Institute Copper-catalysed ligation of azides and acetylenes
US7763736B2 (en) 2002-05-30 2010-07-27 The Scripps Research Institute Copper-catalysed ligation of azides and acetylenes
US9302997B2 (en) 2002-05-30 2016-04-05 The Scripps Research Institute Copper-catalysed ligation of azides and acetylenes
US10280462B2 (en) 2002-10-11 2019-05-07 Jacobus Johannes Maria van Dongen Nucleic acid amplification primers for PCR-based clonality studies
US7090837B2 (en) 2003-01-21 2006-08-15 The Salk Institute For Biological Studies Compositions and methods for tissue specific targeting of lentivirus vectors
US8399645B2 (en) 2003-11-05 2013-03-19 St. Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
US8119119B2 (en) 2004-06-25 2012-02-21 Centre National De La Recherche Scientifique Non-integrative and non-replicative lentivirus, preparation and uses thereof
US20090069561A1 (en) 2004-06-30 2009-03-12 The Scripps Research Institute Click chemistry route to triazole dendrimers
US8101238B2 (en) 2004-07-22 2012-01-24 The Scripps Research Institute Polymeric materials via click chemistry
US20080090995A1 (en) 2005-02-14 2008-04-17 Ge Healthcare Bio-Sciences Ab Liquid Chromatography Method
US11168344B2 (en) 2005-03-31 2021-11-09 Chugai Seiyaku Kabushiki Kaisha Methods for producing polypeptides by regulating polypeptide association
US8372986B2 (en) 2005-09-30 2013-02-12 The Scripps Research Institute Ruthenium-catalyzed cycloaddition of alkynes and organic azides
US8124353B2 (en) 2006-07-14 2012-02-28 Ac Immune S.A. Methods of treating and monitoring disease with antibodies
US8389282B2 (en) 2007-03-30 2013-03-05 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred T lymphocytes
US8357778B2 (en) 2007-05-21 2013-01-22 Nomadic Bioscience Co., Ltd. Polypeptide, an affinity chromatography material, and a method for separating and/or purifying immunoglobulin
US8394914B2 (en) 2007-08-24 2013-03-12 Board Of Trustees Of Michigan State University Functional polyglycolide nanoparticles derived from unimolecular micelles
US8927682B2 (en) 2007-08-24 2015-01-06 Board Of Trustees Of Michigan State University Functionalization of polyglycolides by “click” chemistry
US20140024111A1 (en) 2008-01-07 2014-01-23 Amgen Inc. Host cell for making antibody fc-heterodimeric molecules using electrostatic steering effects
US10494435B2 (en) 2008-04-04 2019-12-03 The Government Of The United States America As Represented By The Secretary Of The Department Of Health And Human Services Human monoclonal antibodies specific for CD22
US20170158760A1 (en) 2008-10-20 2017-06-08 Abbvie Inc. Isolation and Purification of Antibodies Using Protein A Affinity Chromatography
US9528160B2 (en) 2008-11-07 2016-12-27 Adaptive Biotechnolgies Corp. Rare clonotypes and uses thereof
US20180296689A1 (en) 2009-02-13 2018-10-18 Immunomedics, Inc. Neoadjuvant use of antibody-drug conjugates
US8877170B2 (en) 2009-02-21 2014-11-04 Sofradim Production Medical device with inflammatory response-reducing coating
US9890204B2 (en) 2009-04-07 2018-02-13 Hoffmann-La Roche Inc. Trivalent, bispecific antibodies
US9309311B2 (en) 2009-04-27 2016-04-12 Oncomed Pharmaceuticals, Inc. Method for making Heteromultimeric molecules
US8927736B2 (en) 2009-08-11 2015-01-06 The Scripps Research Institute Copper catalyzed cycloaddition of organic azides and 1-haloalkynes
US11390668B2 (en) 2009-10-20 2022-07-19 Abbvie Inc. Isolation and purification of anti-IL-13 antibodies using protein a affinity chromatography
US20130089554A1 (en) 2009-12-29 2013-04-11 Emergent Product Development Seattle, Llc RON Binding Constructs and Methods of Use Thereof
US10138303B2 (en) 2010-05-14 2018-11-27 Rinat Neuroscience Corp. Heterodimeric proteins and methods for producing and purifying them
US9562109B2 (en) 2010-11-05 2017-02-07 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the Fc domain
US20140194442A1 (en) 2011-01-12 2014-07-10 Boehringer Ingelheim International Gmbh Anticancer therapy
US20120244075A1 (en) 2011-03-24 2012-09-27 Nanoprobes, Inc. 5 NM Nickel-NTA-Gold Nanoparticles
US20160131655A1 (en) 2011-04-21 2016-05-12 Boehringer Ingelheim International Gmbh Bcma-based stratification and therapy for multiple myeloma patients
US20120321637A1 (en) 2011-06-20 2012-12-20 The Board Of Regents Of The University Of Texas System Combination cancer therapy with herv inhibition
US11028185B2 (en) 2011-06-28 2021-06-08 Whitehead Institute For Biomedical Research Using sortases to install click chemistry handles for protein ligation
US9422351B2 (en) 2011-11-03 2016-08-23 The Trustees Of The University Of Pennsylvania Isolated B7-H4 specific compositions and methods of use thereof
US9790267B2 (en) 2011-11-08 2017-10-17 The Trustees Of The University Of Pennsylvania Glypican-3-specific antibody and uses thereof
US9527927B2 (en) 2011-12-20 2016-12-27 Medimmune, Llc Modified polypeptides for bispecific antibody scaffolds
US10189903B2 (en) 2012-02-13 2019-01-29 Seattle Children's Hospital Bispecific chimeric antigen receptors and methods of use thereof to treat cancer
US20190276492A1 (en) 2012-02-15 2019-09-12 Hoffmann-La Roche Inc. Fc-receptor based affinity chromatography
US10207229B2 (en) 2012-03-16 2019-02-19 Board Of Trustees Of Michigan State University Functionalization of a porous membrane with an adsorbed polyacid
US9248182B2 (en) 2012-04-20 2016-02-02 Merus B.V. Methods and means for the production of Ig-like molecules
US20180086843A1 (en) 2012-04-26 2018-03-29 Bioatla, Llc Anti-cd22 antibodies
US20150175711A1 (en) 2012-06-15 2015-06-25 Sinomab Bioscience Limited Anti-cd22 anti-idiotypic antibodies and uses thereof
US20220220198A1 (en) 2012-07-19 2022-07-14 Redwood Bioscience, Inc. Antibody specific for cd22 and methods of use thereof
US9181343B2 (en) 2012-07-19 2015-11-10 Redwood Bioscience Inc. Antibody specific for CD22 and methods of use thereof
US9896547B2 (en) 2012-12-03 2018-02-20 The Scripps Research Institute Polymerization method and polymers formed therewith
US10124023B2 (en) 2013-02-26 2018-11-13 Memorial Sloan-Kettering Cancer Center Compositions and methods for immunotherapy
US10023608B1 (en) 2013-03-13 2018-07-17 Amgen Inc. Protein purification methods to remove impurities
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US9629877B2 (en) 2013-05-14 2017-04-25 Board Of Regents, The University Of Texas System Human application of engineered chimeric antigen receptor (CAR) T-cells
US9701758B2 (en) 2013-05-24 2017-07-11 Board Of Regents, The University Of Texas System Anti-CD19 scFv (FMC63) polypeptide
US10640569B2 (en) 2013-12-19 2020-05-05 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
US9630165B2 (en) 2014-01-17 2017-04-25 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
US10836998B2 (en) 2014-02-14 2020-11-17 Cellectis Cells for immunotherapy engineered for targeting antigen present both on immune cells and pathological cells
US9139649B2 (en) 2014-02-25 2015-09-22 Immunomedics, Inc. Humanized anti-CD22 antibody
US20170335281A1 (en) 2014-03-15 2017-11-23 Novartis Ag Treatment of cancer using chimeric antigen receptor
US11602525B2 (en) 2014-04-25 2023-03-14 Rinat Neuroscience Corp. Antibody-drug conjugates with high drug loading
US11702472B2 (en) 2014-06-06 2023-07-18 Memorial Sloan-Kettering Cancer Center Method of reducing mesothelin-expressing tumor burden by administration of T cells comprising mesothelin-targeted chimeric antigen receptors
US11084880B2 (en) 2014-07-21 2021-08-10 Novartis Ag Anti-BCMA chimeric antigen receptor
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
US20220064316A1 (en) 2014-07-21 2022-03-03 The Trustees Of The University Of Pennsylvania Treatment of cancer using humanized anti-bcma chimeric antigen receptor
US20170226216A1 (en) 2014-07-24 2017-08-10 Bluebird Bio, Inc. Bcma chimeric antigen receptors
US10533055B2 (en) 2014-08-28 2020-01-14 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for CD19
US11633426B2 (en) 2014-10-20 2023-04-25 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
US11160833B2 (en) 2014-12-15 2021-11-02 The Regents Of The University Of California Bispecific OR-gate chimeric antigen receptor responsive to CD19 and CD20
US20170296623A1 (en) 2014-12-17 2017-10-19 Cellectis INHIBITORY CHIMERIC ANTIGEN RECEPTOR (iCAR OR N-CAR) EXPRESSING NON-T CELL TRANSDUCTION DOMAIN
US20210061877A1 (en) 2014-12-19 2021-03-04 Dana-Farber Cancer Institute, Inc. Chimeric antigen receptors and methods of use thereof
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells
US10954530B2 (en) 2015-03-02 2021-03-23 Autolus Limited Retroviral and lentiviral vectors
US11541127B2 (en) 2015-04-15 2023-01-03 Astellas Pharma, Inc. Drug conjugates comprising antibodies against claudin 18.2
US20180133296A1 (en) 2015-04-17 2018-05-17 David Maxwell Barrett Methods for improving the efficacy and expansion of chimeric antigen receptor?expressing cells
US11091588B2 (en) 2015-04-24 2021-08-17 The Penn State Research Foundation Clickable waterborne polymers and click-crosslinked waterborne polymers, clickable functional compounds, click functionalized waterborne polymers, and uses thereof
US11713346B2 (en) 2015-05-11 2023-08-01 Biontech Cell & Gene Therapies Gmbh Claudin-18.2-specific immunoreceptors and T cell epitopes
US11136392B2 (en) 2015-06-23 2021-10-05 Memorial Sloan-Kettering Cancer Center PD-1 immune modulating agents
US20180187149A1 (en) 2015-06-25 2018-07-05 Icell Gene Therapeutics Llc CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS OF USE THEREOF
US20190135894A1 (en) 2015-06-25 2019-05-09 iCell Gene Therapeuticics LLC COMPOUND CHIMERIC ANTIGEN RECEPTOR (cCAR) TARGETING MULTIPLE ANTIGENS, COMPOSITIONS AND METHODS OF USE THEREOF
US10683369B2 (en) 2015-08-03 2020-06-16 Engmab Sàrl Monoclonal antibodies against BCMA
US10934363B2 (en) 2015-08-11 2021-03-02 Legend Biotech Usa Inc. Chimeric antigen receptors based on single domain antibodies and methods of use thereof
US10709775B2 (en) 2015-08-11 2020-07-14 Cellectis Cells for immunotherapy engineered for targeting CD38 antigen and for CD38 gene inactivation
US10072088B2 (en) 2015-08-17 2018-09-11 Janssen Pharmaceutica, Nv Anti-BCMA antibodies and uses thereof
US9885298B2 (en) 2015-10-15 2018-02-06 Toyota Jidosha Kabushiki Kaisha Fuel injection control device for engine
US10815301B2 (en) 2015-10-15 2020-10-27 The United States Of America, As Represented By The Secretary, Department Of Health And Human Service Anti-CD30 chimeric antigen receptors
US20200155520A1 (en) 2015-11-19 2020-05-21 Genentech, Inc. Methods of treating cancer using b-raf inhibitors and immune checkpoint inhibitors
US11648268B2 (en) 2015-12-09 2023-05-16 Memorial Sloan Kettering Cancer Center Immune cell compositions and methods of using same
US20200281973A1 (en) 2016-03-04 2020-09-10 Novartis Ag Cells expressing multiple chimeric antigen receptor (car) molecules and uses therefore
US11768203B2 (en) 2016-03-31 2023-09-26 University Of Southern California Highly sensitive and specific luciferase based reporter assay for antigen detection
US20200283534A1 (en) 2016-06-24 2020-09-10 iCell Gene Therapeuticics LLC CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS THEREOF
US20200024342A9 (en) 2016-06-24 2020-01-23 Icell Gene Therapeutics Llc CHIMERIC ANTIGEN RECEPTORS (CARs), COMPOSITIONS AND METHODS THEREOF
US20190151365A1 (en) 2016-07-28 2019-05-23 Novartis Ag Combination therapies of chimeric antigen receptors and pd-1 inhibitors
US20210206815A1 (en) 2016-08-11 2021-07-08 Repligen Corporation Alkaline stable fc-binding proteins for affinity chromatography
US20210046118A1 (en) 2016-12-09 2021-02-18 Onk Therapeutics Limited Engineered natural killer cells and uses thereof
US10799536B2 (en) 2016-12-09 2020-10-13 Onk Therapeutics Limited Method of treating multiple myeloma using natural killer cells expressing a chimeric antigen receptor for CD38
WO2018119114A1 (fr) * 2016-12-22 2018-06-28 Cue Biopharma, Inc. Polypeptides multimères modulateurs des lymphocytes t et leurs méthodes d'utilisation
US10550179B2 (en) 2017-01-09 2020-02-04 Lentigen Technology Inc. Compositions and methods for treating cancer with anti-mesothelin immunotherapy
US20190375815A1 (en) 2017-01-31 2019-12-12 Novartis Ag Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
US11066457B2 (en) 2017-02-08 2021-07-20 Cellular Biomedicine Group Hk Limited Construction of chimeric antigen receptor targeting CD20 antigen and activity identification of engineered T cells thereof
US20190359727A1 (en) 2017-02-17 2019-11-28 Fred Hutchinson Cancer Research Center Combination therapies for treatment of bcma-related cancers and autoimmune disorders
US20190381171A1 (en) 2017-02-17 2019-12-19 Unum Therapeutics Inc. Co-use of anti-bcma antibody and antibody-coupled t cell receptor (actr) in cancer therapy and b cell disorders
US20200048359A1 (en) 2017-02-28 2020-02-13 Novartis Ag Shp inhibitor compositions and uses for chimeric antigen receptor therapy
WO2018170168A1 (fr) * 2017-03-15 2018-09-20 Cue Biopharma, Inc. Procédés pour moduler une réponse immunitaire
US20200055948A1 (en) 2017-04-28 2020-02-20 Novartis Ag Cells expressing a bcma-targeting chimeric antigen receptor, and combination therapy with a gamma secretase inhibitor
US10730954B2 (en) 2017-05-12 2020-08-04 Harpoon Therapeutics, Inc. MSLN targeting trispecific proteins and methods of use
US20210079057A1 (en) 2017-06-13 2021-03-18 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using fusion proteins
US20210253729A1 (en) 2017-07-25 2021-08-19 Board Of Regents, The University Of Texas System Enhanced chimeric antigen receptors and use thereof
US10442867B2 (en) 2017-07-31 2019-10-15 Lentigen Technology, Inc. Compositions and methods for treating cancer with anti-CD19/CD20 immunotherapy
US20200281974A1 (en) 2017-09-26 2020-09-10 Longwood University Pd1-specific chimeric antigen receptor as an immunotherapy
US20200255803A1 (en) 2017-09-27 2020-08-13 Gracell Biotechnologies (Shanghai) Co., Ltd. Engineered immune cell capable of inducing secretion of anti-cd47 antibody
US20200308541A1 (en) 2017-10-12 2020-10-01 Icell Gene Therapeutics, Llc Compound Chimeric Antigen Receptor (CCAR) Targeting Multiple Antigents, Compositions and Methods of Use Thereof
US11618787B2 (en) 2017-10-31 2023-04-04 Janssen Biotech, Inc. Methods of treating high risk multiple myeloma
US20190135937A1 (en) 2017-11-03 2019-05-09 Sorrento Therapeutics, Inc. CD-38 Directed Chimeric Antigen Receptor Constructs
US20200360431A1 (en) 2017-11-15 2020-11-19 Novartis Ag Bcma-targeting chimeric antigen receptor, cd19-targeting chimeric antigen receptor, and combination therapies
US20210300986A1 (en) 2017-12-05 2021-09-30 The Medical Research Infrastrutrure And Health Services Fund Of The Tel Aviv Medecal Center T-cells comprising two different chimeric antigen receptors and uses thereof
US20210095022A1 (en) 2017-12-22 2021-04-01 TaneoBio, Inc. Heavy chain antibodies binding to cd22
US11365394B2 (en) 2017-12-22 2022-06-21 Fate Therapeutics, Inc. Enhanced immune effector cells and use thereof
US20200339699A1 (en) 2018-02-01 2020-10-29 Innovent Biologics (Suzhou) Co., Ltd. Fully humanized anti-b cell maturation antigen (bcma) single-chain antibody and use thereof
US20200392248A1 (en) 2018-02-23 2020-12-17 Beijing Meikang Geno-Immune Biotechnology Co., Ltd. A cd19-based chimeric antigen receptor and application thereof
US11485782B2 (en) 2018-03-14 2022-11-01 Beijing Xuanyi Pharmasciences Co., Ltd. Anti-claudin 18.2 antibodies
US20210230548A1 (en) 2018-05-03 2021-07-29 Board Of Regents, The University Of Texas System Natural killer cells engineered to express chimeric antigen receptors with immune checkpoint blockade
US20220273710A1 (en) 2018-05-15 2022-09-01 Autolus Limited A cd79-specific chimeric antigen receptor
US11098118B2 (en) 2018-05-18 2021-08-24 Lanova Medicines Limited Company Anti-claudin 18.2 antibodies and uses thereof
US20200071397A1 (en) 2018-05-31 2020-03-05 Washington University Chimeric antigen receptor t cells (car-t) for the treatment of cancer
US20210169852A1 (en) 2018-07-27 2021-06-10 Daiichi Sankyo Company, Limited Protein recognizing drug moiety of antibody-drug conjugate
US11369703B2 (en) 2018-08-31 2022-06-28 Genzyme Corporation Sterile chromatography resin and use thereof in manufacturing processes
US20220056101A1 (en) 2018-11-27 2022-02-24 Duke University Anti-lmp2 tcr-t cell therapy for the treatment of ebv-associated cancers
US20220073643A1 (en) 2018-12-28 2022-03-10 Nanjing GenScript Biotech Co., Ltd. Claudin18.2 binding moieties and uses thereof
US20230192840A1 (en) 2018-12-28 2023-06-22 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Antibody and use thereof
US10421817B1 (en) 2019-01-17 2019-09-24 Beijing Mabworks Biotech Co., Ltd. Antibodies binding human Claudin 18.2 and uses thereof
US20220193138A1 (en) 2019-04-25 2022-06-23 Purdue Research Foundation Engineered natural killer cells redirected toward purinergic signaling, constructs thereof, and methods for using the same
WO2020252418A2 (fr) * 2019-06-14 2020-12-17 Cugene, Inc. Nouveaux variants d'interleukines-2 pour le traitement du cancer
US20210047402A1 (en) 2019-06-14 2021-02-18 Teneobio, Inc. Multispecific heavy chain antibodies binding to cd22 and cd3
US20220380852A1 (en) 2019-08-27 2022-12-01 Fundación Para La Investigación Biomédica Del Hospital Universitario 12 De Octubre Method for determining the presence or absence of minimal residual disease (mrd) in a subject who has been treated for a disease
US11352460B2 (en) 2019-11-25 2022-06-07 Covestro Llc Waterborne azido-alkyne click compositions
US20230242877A1 (en) 2020-01-21 2023-08-03 Nanjing Bioheng Biotech Co., Ltd Immune cell comprising chimeric antigen receptor and use thereof
US11439665B2 (en) 2020-03-17 2022-09-13 Cellular Biomedicine Group Hk Limited Combined chimeric antigen receptor targeting CD19 and CD20 and application thereof
US11433100B2 (en) 2020-08-20 2022-09-06 A2 Biotherapeutics, Inc. Compositions and methods for treating ceacam positive cancers
WO2022087149A2 (fr) * 2020-10-22 2022-04-28 Gilead Sciences, Inc. Protéines de fusion d'interleukine-2-fc et méthodes d'utilisation
WO2022125711A1 (fr) * 2020-12-09 2022-06-16 Asher Biotherapeutics, Inc. Construction de cytokine ciblée pour la thérapie cellulaire génétiquement modifiée
WO2024159087A1 (fr) * 2023-01-26 2024-08-02 Dana-Farber Cancer Institute, Inc. Développement d'une plateforme d'activateurs-récepteurs chimériques de l'antigène (car) pour améliorer la fonctionnalité et/ou la persistance de lymphocytes t car

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
"NCBI", Database accession no. NP_000577
CHENG ET AL., CYTOMETRY A, vol. 103, no. 1, 2023, pages 16 - 26
CIBRIANSANCHEZ-MADRID, EUR. J. IMMUNOL., vol. 47, no. 6, 2017, pages 946 - 953
DIAB ET AL., J. CLIN. ONCOL, vol. 41, no. 30, 2023, pages 4756 - 4767
DUTCHER ET AL., J. IMMUNOTHER. CANCER, vol. 2, no. 1, 2014, pages 1 - 23
FEIGE ET AL., TRENDS BIOCHEM. SCI., vol. 35, no. 4, 2010, pages 189 - 198
FRAIETTA ET AL., NAT. MED, vol. 24, no. 5, 2018, pages 563 - 571
FRANCINO-URDANIZWHITEHEAD, RSC CHEM. BIOL., vol. 2, no. 6, 2021, pages 1580 - 1589
GAGELMANN ET AL., EUR. J. HAEMATOL, vol. 104, no. 4, 2020, pages 318 - 327
GARDNER ET AL., BLOOD, vol. 129, no. 25, 2017, pages 3322 - 3331
GERSHONI ET AL., BIODRUGS, vol. 21, no. 3, 2007, pages 145 - 156
HASHIMOTO ET AL., NATURE, vol. 610, no. 7930, 2022, pages 161 - 172
HOSSAIN ET AL., BLOOD, vol. 132, 2018, pages 490 - 490
HOU ET AL., DIS. MARKERS 2019, vol. 3425291, 2019, pages 1 - 11
JONES ET AL., J. IMMUNOL., vol. 205, no. 7, 2020, pages 1721 - 1730
KATZ ET AL., CLIN. CANCER RES., vol. 21, no. 14, 2015, pages 3149 - 59
KOCHENDERFER ET AL., J. IMMUNOTHER, vol. 32, no. 7, 2009, pages 689 - 702
LAURENT ET AL., NAT. COMMUN, vol. 6, no. 7333, 2015, pages 1 - 12
LEE ET AL., LEUKEMIA, vol. 35, no. 1, 2021, pages 255 - 258
LEVIN ET AL., NATURE, vol. 484, no. 7395, 2012, pages 529 - 33
LI ET AL., METHODS MOL. BIOL, vol. 1514, 2017, pages 111 - 118
MATT ET AL., J. MOL. BIOL., vol. 247, no. 5, 1995, pages 979 - 94
PACHELLA ET AL., J. ADV. PRACT. ONCOL, vol. 6, no. 3, 2015, pages 212 - 221
PLUCKTHUN, IMMUNOL. REV, vol. 130, 1992, pages 151 - 188
QUAYLE ET AL., CLIN. CANCER RES, vol. 26, no. 8, 2020, pages 1953 - 1964
QUAYLE ET AL., CLIN. CANCER RES., vol. 26, no. 8, 2020, pages 1953 - 1964
RAEBER ET AL., EBIOMEDICINE, vol. 90, no. 104539, 2023, pages 1 - 25
RAJE ET AL., N. ENGL. J. MED, vol. 380, no. 18, 2019, pages 1726 - 1737
REN ET AL., J. CLIN. INVEST, vol. 132, no. 3, 2022, pages 1 - 13
RODRIGUEZ-OTERO ET AL., N. ENGL. J. MED, vol. 388, no. 11, 2023, pages 1002 - 1014
ROEX ET AL., J. HEMATOL. ONCOL, vol. 13, no. 1, 2020, pages 164
ROSENBERG, J. IMMUNOL., vol. 192, no. 12, 2014, pages 5451 - 5458
SMITH ET AL., MOL. THER, vol. 26, no. 6, 2018, pages 1447 - 1456
SUN ET AL., NAT. COMMUN, vol. 10, no. 1, 2019, pages 1762 - 12
TRAUNECKER ET AL., EMBO J., vol. 10, no. 12, 1991, pages 3655 - 9
TURTLE ET AL., J. CLIN. INVEST, vol. 126, no. 6, 2016, pages 2123 - 38
WARD ET AL., MOL. THER, vol. 8, no. 5, 2003, pages 804 - 12

Similar Documents

Publication Publication Date Title
US20250230217A1 (en) Methods for improving the efficacy and expansion of immune cells
ES3003809T3 (en) Chimeric antigen receptors specific for b-cell maturation antigen and encoding polynucleotides
JP7217970B2 (ja) 融合タンパク質を用いてt細胞受容体をリプログラミングするための組成物及び方法
JP2024008968A (ja) 複数の抗原を標的とするcompoundキメラ抗原受容体(cCAR)の組成物およびその使用方法
CN110177803A (zh) 用于使用融合蛋白进行tcr重新编程的组合物和方法
CN110267677A (zh) 使用与原m2巨噬细胞分子抑制剂组合的嵌合抗原受体治疗癌症
EP3619234A1 (fr) Compositions et méthodes pour thérapies par cellules adoptives
TW202023580A (zh) 使用靶特異性融合蛋白進行tcr再程式化之組合物及方法
CN116829194A (zh) 用于工程化细胞疗法的靶向细胞因子构建体
CN105392888A (zh) 使用人源化抗cd19嵌合抗原受体治疗癌症
CN115135674A (zh) 树突细胞激活性嵌合抗原受体和其用途
JP7524465B2 (ja) 免疫細胞機能の改善
WO2022006451A2 (fr) Compositions et procédés de reprogrammation de tcr faisant intervenir des protéines de fusion et des anticorps anti-pd1
CN113412276A (zh) 二聚化剂调节的免疫受体复合物
JP7690597B2 (ja) 免疫細胞機能の改善
US20230242638A1 (en) Chimeric antigen receptor targeting cldn18.2 and use thereof
IL297147A (en) Methods and Uses Related to Transgenic Cell Therapy with a Chimeric Antigen Receptor Directed to a B-Cell Maturation Antigen
WO2022056321A1 (fr) Compositions et procédés pour la reprogrammation de tcr au moyen de protéines de fusion spécifiques gpc3
JP2023524811A (ja) Cd70特異的融合タンパク質を使用したtcrリプログラミングのための組成物及び方法
WO2022056304A1 (fr) Compositions et méthodes pour la reprogrammation de tcr au moyen de protéines de fusion spécifiques de la nectine-4
WO2024030970A2 (fr) Édition génique des gènes cibles pour améliorer les fonctions des cellules tueuses naturelles
WO2024159087A1 (fr) Développement d'une plateforme d'activateurs-récepteurs chimériques de l'antigène (car) pour améliorer la fonctionnalité et/ou la persistance de lymphocytes t car
WO2025059162A1 (fr) Activateur car contenant des variants d'il-2 pour améliorer la fonctionnalité de cellules car-t
Yu et al. CAR NK cell therapy for solid tumors: Potential and challenges
WO2023034220A2 (fr) Compositions et procédés de reprogrammation de tcr à l'aide de protéines de fusion et de cxcr6

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: 24782701

Country of ref document: EP

Kind code of ref document: A1