WO2024173376A2 - Combination therapy comprising multispecific gamma delta tcr antibodies - Google Patents
Combination therapy comprising multispecific gamma delta tcr antibodies Download PDFInfo
- Publication number
- WO2024173376A2 WO2024173376A2 PCT/US2024/015571 US2024015571W WO2024173376A2 WO 2024173376 A2 WO2024173376 A2 WO 2024173376A2 US 2024015571 W US2024015571 W US 2024015571W WO 2024173376 A2 WO2024173376 A2 WO 2024173376A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cancer
- seq
- antigen
- dose
- sequence
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/30—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
- C07K16/3069—Reproductive system, e.g. ovaria, uterus, testes, prostate
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/3955—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2809—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against the T-cell receptor (TcR)-CD3 complex
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/28—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
- C07K16/2803—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
- C07K16/2827—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
- A61K2039/507—Comprising a combination of two or more separate antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/54—Medicinal preparations containing antigens or antibodies characterised by the route of administration
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/31—Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
Definitions
- the present disclosure relates to methods and medical uses for the treatment of cancer comprising administration of a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V52 T cell receptor in combination with a common gamma chain cytokine and/or an immune checkpoint inhibitor.
- Gamma-delta (yb) T cells are T cells that express a T cell receptor (TOR) that is made up of a gamma chain and a delta chain.
- TOR T cell receptor
- the majority of y6 T cells express TCRs comprising Vy9 and V ⁇ 52 chains.
- VY9V ⁇ 52 T cells can react against a wide array of pathogens and tumor cells. This broad reactivity is understood to be conferred by phosphoantigens that are able to specifically activate this T-cell subset in a TOR dependent fashion.
- the broad antimicrobial and anti-tumor reactivity of VY9V ⁇ 52 T-cells suggest a direct involvement in immune control of cancers and infections.
- VY9V ⁇ 52 T cells can be useful in the treatment of infections or cancer as these may promote VY9V ⁇ 52 T cell reactivity towards the pathogen or infected cells or cancer cell.
- WO 2015/156673 describes antibodies that bind VY9V ⁇ 52 TCRS and are capable of activating VY9V ⁇ 52 T cells.
- WO 2020/060405 and WO 2022/008646 describe multispecific antibodies that bind both VY9V ⁇ 52 T cells and a human cancer antigen and thus have the potential to recruit VY9V ⁇ 52 T cells to cancer cells and thus stimulate a therapeutic effect.
- There is still a need for improved methods for the treatment of cancer using such multispecific antibodies wherein the methods achieve an efficacious and long-lasting response in a broad population of cancer patients while minimizing adverse effects.
- the present disclosure provides a method of treating cancer in a subject in need thereof comprising administering (i) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigenbinding region capable of binding a human Vy9V52 T cell receptor; and (ii) a dose of a common gamma chain cytokine.
- the common gamma chain cytokine is IL-2.
- the IL-2 is administered at a dose of less than 3 MIU per day.
- the common gamma chain cytokine is IL-15.
- the first antigen-binding region binds to a human cancer antigen selected from CD1d, PSMA, CD40, CD123, 5T4, Nectin-4, EGFR, and CD33. In some embodiments, the first antigen binding region binds to human PSMA.
- the present disclosure provides a method of treating cancer in a subject in need thereof comprising administering (i) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigenbinding region capable of binding a human Vy9V ⁇ 52 T cell receptor; and (ii) an anti-PD1 or an anti-PD-L1 antibody.
- the method comprises administration of pembrolizumab.
- the first antigen-binding region binds to a human cancer antigen selected from CD1d, PSMA, CD40, CD123, 5T4, Nectin-4, EGFR, and CD33.
- the first antigen binding region binds to human PSMA.
- the present disclosure provides a method of treating cancer in a subject in need thereof comprising administering (i) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigenbinding region capable of binding a human Vy9V ⁇ 52 T cell receptor; (ii) an anti-PD1 or an anti- PD-L1 antibody; and (iii) a common gamma chain cytokine.
- the common gamma chain cytokine is IL-2.
- the IL-2 is administered at a dose of less than 3 MIU per day.
- the common gamma chain cytokine is IL-15.
- the method comprises administration of pembrolizumab.
- the first antigen-binding region binds to a human cancer antigen selected from CD1d, PSMA, CD40, CD123, 5T4, Nectin-4, EGFR, and CD33.
- the first antigen binding region binds to human PSMA.
- the present disclosure provides a kit for the treatment of cancer comprising: (a) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor; and (b) a common gamma chain cytokine, wherein the kit optionally comprises instructions for use.
- the common gamma chain cytokine is IL-2.
- the dose of IL-2 is less than 3 MIU per day.
- the common gamma chain cytokine is IL-15.
- the first antigen-binding region binds to a human cancer antigen selected from CD1d, PSMA, CD40, CD123, 5T4, Nectin-4, EGFR, and CD33. In some embodiments, the first antigen binding region binds to human PSMA.
- the present disclosure provides a kit for the treatment of cancer comprising: (a) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor; and (b) an anti-PD1 or anti-PD-L1 antibody, wherein the kit optionally comprises instructions for use.
- the anti-PD1 antibody is pembrolizumab.
- the first antigen-binding region binds to a human cancer antigen selected from CD1d, PSMA, CD40, CD123, 5T4, Nectin-4, EGFR, and CD33.
- the first antigen binding region binds to human PSMA.
- the present disclosure provides a kit for the treatment of cancer comprising: (a) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor; (b) an anti-PD1 or anti-PD-L1 antibody; and (c) a common gamma chain cytokine, wherein the kit optionally comprises instructions for use.
- the common gamma chain cytokine is IL-2.
- the dose of IL-2 is less than 3 MIU per day.
- the common gamma chain cytokine is IL-15.
- the first antigen-binding region binds to a human cancer antigen selected from CD1d, PSMA, CD40, CD123, 5T4, Nectin-4, EGFR, and CD33. In some embodiments, the first antigen binding region binds to human PSMA. In some embodiments, the anti-PD1 antibody is pembrolizumab.
- Fig. 1 shows Prostate Specific Antigen (PSA) responses as % change from baseline. Shown is best PSA response and, for those patients with no PSA decline, PSA value at Cycle 5 Day 1 . Dose Levels (DL) for each patient are indicated.
- PSA Prostate Specific Antigen
- Fig. 2 shows PSA levels over time in Pt N304.
- FIG. 3A - Fig. 3D show changes in Vy9V ⁇ 52 T cell-related pharmacodynamic parameters over time after administration of LAVA-1207.
- FIG. 4A - Fig. 4B show various pharmacokinetic parameters of LAVA- 1207.
- Fig. 5A - Fig. 5B show a summary of adverse events during dose escalation trial of
- Fig. 6A - Fig. 6E show changes in PD1 expression and frequency of Vy9V ⁇ 52 T cells over time and maximum observed receptor occupancy of LAVA-1207.
- Fig. 6A shows relative change in PD1+ Vy9V ⁇ 52 T cells after administration of LAVA-1207 compared to baseline for cohort 4.
- Fig. 6B shows relative change in MFI of PD1 on Vy9V ⁇ 52 T cells compared to baseline for cohort 4.
- Fig. 6C shows relative change of Vy9V52 T cells (compared to baseline, set at 100) from the completed cohort 6A2; dashed lines indicate administration of IL-2.
- Fig. 6A shows relative change in PD1+ Vy9V ⁇ 52 T cells after administration of LAVA-1207 compared to baseline for cohort 4.
- Fig. 6B shows relative change in MFI of PD1 on Vy9V ⁇ 52 T cells compared to baseline for cohort 4.
- Fig. 6C shows relative change of Vy9V52 T cells (compared to baseline, set at 100
- FIG. 6D shows Vy9V ⁇ 52 T cells per ml of blood over time from completed cohort 6A2; dashed lines indicate administration of IL-2.
- Fig. 6E shows the increase in the percentage of LAVA-1207 receptor occupancy on Vy9V ⁇ 52 T cells in the completed cohorts.
- Fig. 7A - Fig. 7C shows percentages of Vy9V ⁇ 52 T cells (Fig. 7A), of PD1+ Vy9V ⁇ 52 T cells (Fig. 7B), and of CD3+ T cells (Fig. 7C) in prostate cancer patient PBMCs, malignant (m), and non-malignant (n.m.) areas of the patient prostate.
- Fig. 8 shows upregulation of PD1 expression on Vy9V ⁇ 52 T cells activated with a V62- bsTCE.
- Fig. 9 shows an exemplary dosing schedule of the disclosed embodiments.
- Fig. 10 shows exemplary dose cohorts of LAVA-1207 and LAVA-1207 + IL-2.
- FIG. 11A-FIG. 11 B show the effect of a multispecific binding agent on Vy9V ⁇ 52 T cell expansion.
- FIG. 11 A shows the increase in Vy9V ⁇ 52 T cell frequency as a percentage of CD3 + T cells after incubation with IL-2 and either medium alone, pamidronate or the multispecific binding agent.
- FIG. 11 B shows the fold expansion of Vy9V ⁇ 52 T cells over baseline after incubation with IL-2 and either medium alone, pamidronate or the multispecific binding agent.
- Fig. 12A-Fig. 12E show the Vy9V52-T cell frequency and phenotype of patient-derived tumor and non-tumor tissue and the cytolytic activity of Vy9V52-T cells against patient-derived tumor and non-tumor tissue in the presence of LAVA-1207.
- Fig. 12A shows the frequency of Vy9V52-T cells as a percentage of T cells in patient PBMC, patient-derived malignant, and non-malignant tissue.
- Fig. 12B shows the phenotype of PBMC-, malignant-, and non- malignant tissue-derived Vy9V52-T cells and CD3+ cells, Fig.
- FIG. 12C shows LAVA-1207- mediated increase in Vy9V52-T cell degranulation (as assessed by CD107a expression) when malignant tissue is incubated with LAVA-1207 and PBMC.
- Fig. 12D shows the lack of LAVA- 1207-mediated increase in Vy9V52-T cell degranulation when non-malignant tissue is incubated with LAVA-1207 and PBMC.
- Fig. 12E shows the increase in tumor cell lysis when malignant tissue is incubated with LAVA-1207 and PBMCs as opposed to non-malignant cells.
- Fig. 13A-Fig. 13C show the phenotypical analysis of ligands and matched coreceptors on patient-derived tumor and non-tumor tissue and Vy9V52-T cells.
- Fig. 13A shows differences in PSMA, BTN3A, and BTN2A1 expression.
- Fig. 13B shows differences in Nectin- 2, PVR, MIC-A/MIC-B, ULPB1 , ULBP3, ULBP2/5/6, and HLA-E expression.
- Fig. 13C shows the frequency of Vy9V ⁇ 52 T cells that express the DNAM-1 , NKG2D, or NKG2A receptors in PBMC, malignant, and non-malignant tissue.
- Fig. 14A-Fig.14H show the effect of blocking the DNAM-1 or NKG2D receptors on LAVA-1207-mediated Vy9V52-T cell cytokine production and degranulation as well as lysis of prostate cancer cell lines. Effects on IFNy (Fig. 14A), TNF (Fig. 14B), IL-2 (Fig. 14C), IL-4 (Fig. 14D) are shown, as well as effects on Vy9V ⁇ 52 T cell degranulation (Fig. 14E), tumor cell lysis (Fig. 14F) in the presence or absence of DNAM-1 blocking antibodies.
- Fig. 14G shows the effect of blocking the NKG2D receptor on Vy9V ⁇ 52 T cell degranulation.
- Fig. 14H shows the effect of blocking the NKG2D receptor on tumor cell lysis.
- Fig. 15A-Fig. 15B show the effects of blocking NKG2A (Fig. 15A) or BTN3A (Fig. 15B) on LAVA-1207-mediated T cell activation.
- Fig. 16 shows the effect of either anti-DNAM-1 or anti-NKG2D blocking antibodies on LAVA-1207-mediated Vy9V ⁇ 52 T cell degranulation using patient prostate tumor tissue.
- Fig. 17A-Fig. 17B show that NCG mice implanted with prostate cancer cells alone or admixed with PBMCs and treated with LAVA- 1207 had a reduction in tumor growth volume (Fig. 17A) and an increased survival (Fig. 17B) as compared to mice that were only implanted with prostate cancer cells and treated with either PBMCs or PBS or PSMA-V52-Fc bsTCE.
- FIG. 18A-Fig. 18B show that labeling LAVA-1207 with FITC does not impair its binding to either Vy9V52-T cells (Fig. 18A) or prostate cancer cells (Fig. 18B).
- FIG. 19A-Fig. 19B show exemplary immunohistochemistry staining showing tissues positive for PSMA and binding by LAVA- 1207 (Fig. 19A) and tissues negative for PSMA and an absence of binding by LAVA-1207 (Fig. 19B). Arrows indicate potential V52+ T cells (positive binding of LAVA-1207).
- Fig. 20 illustrates the different CDR numbering systems (Kabat, Chothia, IMGT, and Combined) for the 6H4 VHH.
- Vy9V52-T cells represent a conserved T cell subset (approximately 1-5% of all cluster of differentiation [CD]3+ T cells) that can induce cell death in a wide range of malignant cells in an H LA-peptide-independent fashion (Lo Presti et al. 2017; de Weerdt et al. 2018; Kunzmann, Bauer, and Wilhelm 1999; Gertner-Dardenne et al. 2012).
- Vy9V52-T cells have properties of both the innate and adaptive immune systems, they serve as a functional bridge between these two critical systems to impact tumor killing. As such, not only do they have the capability to be activated for immediate and potent killing of tumor cells, they also have the potential to contribute to a cascade response in which downstream activation of innate immune cells such as natural killer (NK) cells and adaptive immune cells such as classical (alpha-beta) T cells can result in additional tumor killing. Finally, activated Vy9V52-T cells can take up, process and present antigen, which may lead to priming of the adaptive immune system resulting in immunological memory with the potential to provide deep and durable antitumor responses.
- NK natural killer
- adaptive immune cells such as classical (alpha-beta) T cells
- BTN2A1 butyrophilin
- BTN3A1 butyrophilin2A1
- Phosphoantigens accumulate upon dysregulation of the mevalonate pathway and are upregulated during cellular stress, such as malignant transformation, or through pharmacological manipulation with e.g. aminobisphosphonates.
- Vy9V52-T cells In addition, the presence of other cell surface receptors such as NKG2D allow Vy9V52-T cells to interact with cells expressing NKG2D ligands MICA/B and ULBPs (Vantourout and Hayday 2013; Kong et al. 2009; Rincon-Orozco et al. 2005). Following activation, Vy9V52-T cell functions include cytotoxicity, secretion of chemokines and pro-inflammatory cytokines and antigen presentation (Vantourout and Hayday 2013; Brandes, Willimann, and Moser 2005; Devilder et al. 2006).
- Vy9V52-T cells Since the presence of Vy9V52-T cells in solid tumors strongly correlates with patient survival (Gentles et al. 2015; Tosolini et al. 2017), approaches that improve targeting and activation of Vy9V52-T cells to tumors are thought to have the potential for the development of novel, efficacious and safe cancer treatments (de Bruin et al. 2016).
- Interleukin (I L)-2 is a cytokine known to promote T cell proliferation and function (Nada et al. 2017; Chen et al. 2022; Li and David Pauza 2011).
- the rationale for exploring IL-2 as an immune modulator for LAVA-1207 is based on its known T-cell stimulatory effect.
- IL-2 has been shown to induce rapid expansion of Vy9V52-T cells in vitro and to also support Vy9V52- T cell expansion in both non-human primates and humans exposed to phosphoantigen stimulation (Nada et al. 2017; Sicard et al. 2005; Wilhelm et al. 2003; Dieli et al. 2007; Meraviglia et al.
- IL-2 and other common gamma chain cytokines such as IL-15, could therefore expand and enhance the availability of Vy9V52-T cells in patients treated with multispecific antibodies described herein (including LAVA-1207) to promote more potent responses towards tumor cells.
- human V52 when used herein, refers to the rearranged 52 chain of the Vy9V52-T cell receptor (TCR).
- UniProtKB - A0JD36 (A0JD36_HUMAN) gives an example of a variable TRDV2 sequence.
- human VY9 when used herein, refers to the rearranged y9 chain of the VY9V52-T cell receptor (TCR).
- UniProtKB - Q99603_HUMAN gives an example of a variable TRGV9 sequence.
- PSMA Prostate-Specific Membrane Antigen protein
- CD1d refers to the human CD1d protein (UniProtKB - P15813 (CD1 D_HUMAN)).
- CD40 refers to the CD40 protein, also known as tumor necrosis factor receptor superfamily member 5 (UniProtKB - P25942 (TNR5_HUMAN)), Isoform I.
- CD123 when used herein, refers to the human CD123 protein, also termed interleukin-3 receptor alpha chain (NCBI Reference Sequence: NP_002174.1).
- IL-2 refers to an interleukin-2 protein.
- UniProtKB - P60568 provides the WT human IL-2 amino acid sequence.
- the term IL-2 is inclusive of variants of the WT sequence.
- IL-15 refers to an interleukin-15 protein.
- UniProtKB-P40933 provides the WT human IL-15 amino acid sequence.
- the term IL-15 is inclusive of variants of the WT sequence.
- 5T4 refers to the human Trophoblast glycoprotein (UniProtKB - Q13641).
- human Nectin-4 when used herein, refers to the human Nectin-4 protein (UniProt Q96NY8 ⁇ NECT4_HUMAN).
- EGFR refers to the human EGFR protein (UniProtKB - P00533 (EGFR_HUMAN)).
- CD33 refers to the human CD33 protein, also known as Siglec3 (UniProt P20138 (CD33_HUMAN)).
- immunoglobulin as used herein is intended to refer to a class of structurally related glycoproteins consisting of two pairs of polypeptide chains, one pair of light (L) chains and one pair of heavy (H) chains, all four potentially inter-connected by disulfide bonds.
- immunoglobulin heavy chain “heavy chain of an immunoglobulin” or “heavy chain” as used herein is intended to refer to one of the chains of an immunoglobulin.
- a heavy chain is typically comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region (abbreviated herein as CH) which defines the isotype of the immunoglobulin.
- the heavy chain constant region typically is comprised of three domains, CH1 , CH2, and CH3.
- the heavy chain constant region further comprises a hinge region.
- the two heavy chains are interconnected via disulfide bonds in the hinge region.
- each light chain is typically comprised of several regions; a light chain variable region (VL) and a light chain constant region (CL).
- VL light chain variable region
- CL light chain constant region
- the VH and VL regions may be subdivided into regions of hypervariability (or hypervariable regions which may be hypervariable in sequence and/or form structurally defined loops), also termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FRs).
- CDRs complementarity determining regions
- Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
- CDR sequences may be determined by use of various methods, e.g. the methods provided by Chothia and Lesk (1987) J. Mol. Biol. 196:901 or Kabat et al. (1991) Sequence of protein of immunological interest, fifth edition. NIH publication. Various methods for CDR determination and amino acid numbering can be compared on www.abysis.org (UCL).
- isotype refers to the immunoglobulin (sub)class (for instance lgG1 , lgG2, lgG3, lgG4, IgD, IgA, IgE, or IgM) or any allotype thereof, such as lgG1m(za) and lgG1 m(f) that is encoded by heavy chain constant region genes.
- immunoglobulin subclass
- Each heavy chain isotype can be combined with either a kappa (K) or lambda (A) light chain.
- K kappa
- A lambda
- An antibody of the present disclosure can possess any isotype.
- antibody is intended to refer to an immunoglobulin molecule, a fragment of an immunoglobulin molecule, or a derivative of either thereof, which has the ability to specifically bind to an antigen under typical physiological conditions with a half-life of significant periods of time, such as at least about 30 minutes, at least about 45 minutes, at least about one hour, at least about two hours, at least about four hours, at least about 8 hours, at least about 12 hours, about 24 hours or more, about 48 hours or more, about 3, 4, 5, 6, 7 or more days, etc., or any other relevant functionally-defined period (such as a time sufficient to induce, promote, enhance, and/or modulate a physiological response associated with antibody binding to the antigen and/or time sufficient for the antibody to recruit an effector activity).
- significant periods of time such as at least about 30 minutes, at least about 45 minutes, at least about one hour, at least about two hours, at least about four hours, at least about 8 hours, at least about 12 hours, about 24 hours or more, about 48 hours or more, about 3, 4, 5, 6, 7
- antigen-binding region and “antigen-binding domain” are used interchangeably herein and refer to a portion of an antibody which interacts with an antigen.
- Antigen-binding domains may comprise variable regions of both the heavy and light chains of the immunoglobulin molecule or may be a single-domain antigen-binding region, e.g. a heavy chain variable region only.
- the constant regions of an antibody if present, may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system and components of the complement system such as 01 q, the first component in the classical pathway of complement activation.
- the Fc region of an immunoglobulin is defined as the fragment of an antibody which would be typically generated after digestion of an antibody with papain which includes the two CH2-CH3 regions of an immunoglobulin and a connecting region, e.g. a hinge region.
- the constant domain of an antibody heavy chain defines the antibody isotype, e.g. lgG1 , lgG2, lgG3, lgG4, lgA1 , lgA2, IgM, IgD, or IgE.
- the Fc-region mediates the effector functions of antibodies with cell surface receptors called Fc receptors and proteins of the complement system.
- hinge region as used herein is intended to refer to the hinge region of an immunoglobulin heavy chain.
- the hinge region of a human IgG 1 antibody corresponds to amino acids 216-230 according to the Ell numbering.
- CH2 region or “CH2 domain” as used herein is intended to refer to the CH2 region of an immunoglobulin heavy chain.
- CH2 region of a human IgG 1 antibody corresponds to amino acids 231-340 according to the Ell numbering.
- the CH2 region may also be any of the other subtypes as described herein.
- CH3 region or “CH3 domain” as used herein is intended to refer to the CH3 region of an immunoglobulin heavy chain.
- CH3 region of a human IgG 1 antibody corresponds to amino acids 341-447 according to the Ell numbering.
- the CH3 region may also be any of the other subtypes as described herein.
- antibody as used herein, unless otherwise stated or clearly contradicted by context, includes fragments of an antibody that retain the ability to specifically bind to the antigen. It has been shown that the antigen-binding function of an antibody may be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antibody” include (i) a Fab’ or Fab fragment, i.e. a monovalent fragment consisting of the VL, VH, CL and CH1 domains, or a monovalent antibody as described in WO 2007/059782; (ii) F(ab')2 fragments, i.e.
- bivalent fragments comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting essentially of the VH and CH1 domains; and (iv) a Fv fragment consisting essentially of the VL and VH domains of a single arm of an antibody.
- the two domains of the Fv fragment, VL and VH are coded for by separate genes, they may be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain antibodies or single chain Fv (scFv), see for instance Bird et al., Science 242, 423-426 (1988) and Huston et al., PNAS USA 85, 5879-5883 (1988)).
- single chain antibodies are encompassed within the term antibody unless otherwise indicated by context.
- fragments are generally included within the meaning of antibody, they collectively and each independently are unique features of the present disclosure, exhibiting different biological properties and utility.
- the term antibody also includes polyclonal antibodies, monoclonal antibodies (mAbs), chimeric antibodies and humanized antibodies, and antibody fragments provided by any known technique, such as enzymatic cleavage, peptide synthesis, and recombinant techniques.
- the first antigenbinding region or the second antigen-binding region, or both is a single domain antibody.
- Single domain antibodies are well known to the skilled person, see e.g. Hamers-Casterman et al. (1993) Nature 363:446, Roovers et al. (2007) Curr Opin Mol Ther 9:327 and Krah et al.
- Single domain antibodies comprise a single CDR1 , a single CDR2, and a single CDR3.
- single domain antibodies are variable fragments of heavy-chain-only antibodies, antibodies that naturally do not comprise light chains, single domain antibodies derived from conventional antibodies, and engineered antibodies.
- Single domain antibodies may be derived from any species.
- single domain antibodies can be derived from antibodies raised in Camelidae species, for example in camel, dromedary, llama, alpaca and guanaco. Like a whole antibody, a single domain antibody is able to bind selectively to a specific antigen.
- Single domain antibodies may contain only the variable domain of an immunoglobulin chain, i.e. CDR1 , CDR2 and CDR3 and framework regions. Such antibodies are also called Nanobody®, or VHH.
- parent antibody is to be understood as an antibody which is identical to an antibody according to the present disclosure, but wherein the parent antibody does not have one or more of the specified mutations.
- An “antibody variant” or a “variant of a parent antibody” of the present disclosure is an antibody molecule which comprises one or more mutations as compared to a “parent antibody”.
- Amino acid substitutions may exchange a native amino acid for another naturally- occurring amino acid, or for a non-naturally- occurring amino acid derivative.
- the amino acid substitution may be conservative or non-conservative. In the context of the present disclosure, conservative substitutions may be defined by substitutions within the classes of amino acids reflected in one or more of TABLE 1 , TABLE 2, and TABLE 3.
- parent protein is to be understood as a protein which is identical to a protein according to the present disclosure, but wherein the parent protein does not have one or more of the specified mutations.
- a “variant” or “protein variant” of the present disclosure is a protein which comprises one or more amino acid mutations as compared to a “parent protein”.
- a substitution in a variant is indicated as: Original amino acid - position - substituted amino acid;
- the three-letter code, or one letter code, are used, including the codes Xaa and X to indicate amino acid residue.
- the notation “T366W” means that the variant comprises a substitution of threonine with tryptophan in the variant amino acid position corresponding to the amino acid in position 366 in the parent antibody.
- a substitution embraces a substitution into any one of the other nineteen natural amino acids, or into other amino acids, such as non-natural amino acids.
- a substitution of amino acid T in position 366 includes each of the following substitutions: 366A, 366C, 366D, 366G, 366H, 366F, 366I, 366K, 366L, 366M, 366N, 366P, 366Q, 366R, 366S, 366E, 366V, 366W, and 366Y.
- full-length antibody when used herein, refers to an antibody which contains all heavy and light chain constant and variable domains corresponding to those that are normally found in a wild-type antibody of that isotype.
- chimeric antibody refers to an antibody wherein the variable region is derived from a non-human species (e.g. derived from rodents) and the constant region is derived from a different species, such as human. Chimeric antibodies may be generated by genetic engineering. Chimeric monoclonal antibodies for therapeutic applications are developed to reduce antibody immunogenicity.
- humanized antibody refers to a genetically engineered non-human antibody, which contains human antibody constant domains and non-human variable domains modified to contain a high level of sequence homology to human variable domains. This can be achieved by grafting of the six non-human antibody complementarity-determining regions (CDRs), which together form the antigen binding site, onto a homologous human acceptor framework region (FR). In order to fully reconstitute the binding affinity and specificity of the parental antibody, the substitution of framework residues from the parental antibody (i.e. the non-human antibody) into the human framework regions (back-mutations) may be required. Structural homology modeling may help to identify the amino acid residues in the framework regions that are important for the binding properties of the antibody.
- CDRs complementarity-determining regions
- FR homologous human acceptor framework region
- a humanized antibody may comprise non-human CDR sequences, primarily human framework regions optionally comprising one or more amino acid back-mutations to the non-human amino acid sequence, and, optionally, fully human constant regions.
- additional amino acid modifications which are not necessarily back-mutations, may be introduced to obtain a humanized antibody with preferred characteristics, such as affinity and biochemical properties. Humanization of non-human therapeutic antibodies is performed to minimize its immunogenicity in man while such humanized antibodies at the same time maintain the specificity and binding affinity of the antibody of non-human origin.
- multispecific antibody refers to an antibody having specificities for at least two different, such as at least three, typically non-overlapping, epitopes. Such epitopes may be on the same or on different target antigens. If the epitopes are on different targets, such targets may be on the same cell or different cells or cell types.
- a multispecific antibody may comprise one or more single-domain antibodies.
- bispecific antibody refers to an antibody having specificities for two different, typically non-overlapping, epitopes. Such epitopes may be on the same or different targets. If the epitopes are on different targets, such targets may be on the same cell or different cells or cell types. A bispecific antibody may comprise one or two single-domain antibodies.
- bispecific T cell engager or “bsTCE” refers to a bispecific antibody having specificities for two different epitopes, in which one of the epitopes is expressed by a T cell.
- Examples of different classes of multispecific, such as bispecific, antibodies include but are not limited to (i) IgG-like molecules with complementary CH3 domains to force heterodimerization; (ii) recombinant IgG-like dual targeting molecules, wherein the two sides of the molecule each contain the Fab fragment or part of the Fab fragment of at least two different antibodies; (iii) IgG fusion molecules, wherein full length IgG antibodies are fused to extra Fab fragment or parts of Fab fragment; (iv) Fc fusion molecules, wherein single chain Fv molecules or stabilized diabodies are fused to heavy-chain constant- domains, Fc-regions or parts thereof; (v) Fab fusion molecules, wherein different Fab- fragments are fused together, fused to heavy-chain constant-domains, Fc-regions or parts thereof; and (vi) ScFv-and diabody-based and heavy chain antibodies (e.g., domain antibodies, Nanobodies®) wherein different single chain Fv molecules or
- IgG-like molecules with complementary CH3 domains molecules include but are not limited to the Triomab® (Trion Pharma/Fresenius Biotech), the Knobs-into-Holes (Genentech), CrossMAbs (Roche) and the electrostatically-matched (Amgen, Chugai, Oncomed), the LLIZ-Y (Genentech, Wranik et al. J. Biol. Chem. 2012, 287(52): 43331-9, doi: 10.1074/jbc.M 112.397869.
- IgG-like dual targeting molecules include but are not limited to Dual Targeting (DT)-lg (GSK/Domantis, WO 2009/058383), Two-in-one Antibody (Genentech, Bostrom, et al 2009. Science 323, 1610-1614), Cross-linked Mabs (Karmanos Cancer Center), mAb2 (F-Star), ZybodiesTM (Zyngenia, LaFleur et al. MAbs. 2013 Mar- Apr;5(2):208-18), approaches with common light chain, K Bodies (Novlmmune, WO 2012/023053) and CovX-body® (CovX/Pfizer, Doppalapudi, V.R., et al 2007. Bioorg. Med. Chem. Lett. 17,501-506).
- DT Dual Targeting
- GSK/Domantis WO 2009/058383
- Two-in-one Antibody Genentech, Bostrom, et al 2009. Science 323, 1610-1614
- Cross-linked Mabs
- IgG fusion molecules include but are not limited to Dual Variable Domain (DVD)-lg (Abbott), Dual domain double head antibodies (Unilever; Sanofi Aventis), IgG-like Bispecific (ImClone/Eli Lilly, Lewis et al. Nat Biotechnol. 2014 Feb;32(2):191-8), Ts2Ab (Medlmmune/AZ, Dimasi et al. J Mol Biol.
- DVD Dual Variable Domain
- UPD Dual domain double head antibodies
- IgG-like Bispecific ImClone/Eli Lilly, Lewis et al. Nat Biotechnol. 2014 Feb;32(2):191-8
- Ts2Ab Medlmmune/AZ, Dimasi et al. J Mol Biol.
- Fc fusion molecules include but are not limited to ScFv/Fc Fusions (Academic Institution, Pearce et al Biochem Mol Biol Int. 1997 Sep;42(6):1179), SCORPION (Emergent BioSolutions/T rubion, Blankenship JW, et al. AACR 100th Annual meeting 2009 (Abstract #5465); Zymogenetics/BMS, WO2010111625), Dual Affinity Retargeting Technology (Fc-DARTTM) (MacroGenics) and Dual(ScFv)2-Fab (National Research Center for Antibody Medicine - China).
- Fab fusion bispecific antibodies include but are not limited to F(ab)2 (Medarex/AMGEN), Dual-Action or Bis-Fab (Genentech), Dock-and-Lock® (DNL) (ImmunoMedics), Bivalent Bispecific (Biotecnol) and Fab-Fv (UCB-Celltech).
- ScFv-, diabody-based and domain antibodies include but are not limited to Bispecific T Cell Engager (BiTE®) (Micromet, Tandem Diabody (Tandab) (Affimed), Dual Affinity Retargeting Technology (DARTTM) (MacroGenics), Single-chain Diabody (Academic, Lawrence FEBS Lett. 1998 Apr 3;425(3):479-84), TCR-like Antibodies (AIT, ReceptorLogics), Human Serum Albumin ScFv Fusion (Merrimack, W02010059315) and COMBODY molecules (Epigen Biotech, Zhu et al. Immunol Cell Biol. 2010 Aug;88(6):667-75), dual targeting nanobodies® (Ablynx, Hmila et al., FASEB J. 2010), dual targeting heavy chain only domain antibodies.
- BiTE® Bispecific T Cell Engager
- DARTTM Dual Affinity Retargeting Technology
- Single-chain Diabody Academic, Lawrence
- the multispecific antibody used in the present disclosure is in a VHH-Fc format, i.e. the antibody comprises two or more single-domain antigen-binding regions that are linked to each other via a human Fc region dimer.
- each singledomain antigen-binding region is fused to an Fc region polypeptide and the two fusion polypeptides form a dimeric bispecific antibody via disulfide bridges in the hinge region.
- Such constructs typically do not contain full, or any, CH1 or light chain sequences.
- Figure 12B of W006064136 provides an illustration of an example of this format.
- binds In the context of antibody binding to an antigen, the terms “binds”, “capable of binding” or “specifically binds” refer to the binding of an antibody to a predetermined antigen or target (e.g. human V ⁇ 52 or human PSMA) to which binding typically is with an apparent affinity corresponding to a KD of about 10' 6 M or less, e.g. 10' 7 M or less, such as about 10' 8 M or less, such as about 10' 9 M or less, about 10' 10 M or less, or about 10- 11 M or even less, e.g. when determined using flow cytometry.
- a predetermined antigen or target e.g. human V ⁇ 52 or human PSMA
- KD values can be determined using for instance surface plasmon resonance (SPR) technology in a BIAcore T200 or bio-layer interferometry (BLI) in an Octet RED96 instrument using the antigen as the ligand and the binding moiety or binding molecule as the analyte.
- SPR surface plasmon resonance
- BSA bio-layer interferometry
- Octet RED96 instrument using the antigen as the ligand and the binding moiety or binding molecule as the analyte.
- Specific binding means that the antibody binds to the predetermined antigen with an affinity corresponding to a KD that is at least tenfold lower, such as at least 100-fold lower, for instance at least 1 ,000 fold lower, such as at least 10,000 fold lower, for instance at least 100,000 fold lower than its affinity for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely- related antigen.
- a non-specific antigen e.
- the degree with which the affinity is lower is dependent on the KD of the binding moiety or binding molecule, so that when the KD of the binding moiety or binding molecule is very low (that is, the binding moiety or binding molecule is highly specific), then the degree with which the affinity for the antigen is lower than the affinity for a non-specific antigen may be at least 10,000-fold.
- KD KD
- M refers to the dissociation equilibrium constant of a particular interaction between the antigen and the binding moiety or binding molecule.
- Capable of binding the V ⁇ 52 chain of a VY9V52-TCR” or “binding the V ⁇ 52 chain of a VY9V52-TCR” or the like means that the antibody can bind the V ⁇ 52 chain as a separate molecule and/or as part of a VY9V52-TCR. However, the antibody will not bind to the VY9 chain as a separate molecule.
- Capable of binding the VY9 chain of a VY9V52-TCR” or “binding the VY9 chain of a VY9V52-TCR” or the like means that the antibody can bind the VY9 chain as a separate molecule and/or as part of a VY9V52-TCR. However, the antibody will not bind to the V ⁇ 52 chain as a separate molecule.
- “competition” or “able to compete” or “competes” refers to any detectably significant reduction in the propensity for a particular binding molecule (e.g. an PSMA binding antibody) to bind a particular binding partner (e.g. PSMA) in the presence of another molecule (e.g. a different PSMA antibody) that binds the binding partner.
- a particular binding molecule e.g. an PSMA binding antibody
- a particular binding partner e.g. PSMA
- competition means an at least about 25 percent reduction, such as an at least about 50 percent, e.g.
- Additional methods for determining binding specificity by competitive inhibition may be found in for instance Harlow et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1988), Colligan et al., eds., Current Protocols in Immunology, Greene Publishing Assoc, and Wiley InterScience N. Y., (1992, 1993), and Muller, Meth. Enzymol. 92, 589-601 (1983)).
- the method or use of the present disclosure may involve the use of an antibody that binds to the same epitope on a target (e.g. V ⁇ 52 or PSMA) as antibody described herein.
- a target e.g. V ⁇ 52 or PSMA
- mapping antibody epitopes on target antigens known in the art, including but not limited to: crosslinking coupled mass spectrometry, allowing identification of peptides that are part of the epitope, and X-ray crystallography identifying individual residues on the antigen that form the epitope.
- Epitope residues can be determined as being all amino acid residues with at least one atom less than or equal to 5 A from the antibody.
- epitope cutoff distance was chosen as the epitope cutoff distance to allow for atoms within a van der Waals radius plus a possible water- mediated hydrogen bond.
- epitope residues can be determined as being all amino acid residues with at least one atom less than or equal to 8 A. Less than or equal to 8 A is chosen as the epitope cutoff distance to allow for the length of an extended arginine amino acid.
- Crosslinking coupled mass spectrometry begins by binding the antibody and the antigen with a mass labeled chemical crosslinker. Next the presence of the complex is confirmed using high mass MALDI detection. Because after crosslinking chemistry the Ab/Ag complex is extremely stable, many various enzymes and digestion conditions can be applied to the complex to provide many different overlapping peptides.
- Identification of these peptides is performed using high resolution mass spectrometry and MS/MS techniques. Identification of the crosslinked peptides is determined using mass tag linked to the crosslinking reagents. After MS/MS fragmentation and data analysis, peptides that are crosslinked and are derived from the antigen are part of the epitope, while peptides derived from the antibody are part of the paratope. All residues between the most N- and C-terminal crosslinked residue from the individual crosslinked peptides found are considered to be part of the epitope or paratope.
- first and second antigen-binding regions when used herein do not refer to their orientation I position in the antibody, i.e. they have no meaning with regard to the N- or C-terminus.
- first and second only serve to correctly and consistently refer to the two different antigen-binding regions in the claims and the description.
- the percent identity between two nucleotide or amino acid sequences may e.g. be determined using the algorithm of E. Meyers and W. Miller, Comput. Appl. Biosci 4, 11-17 (1988) which has been incorporated into the ALIGN program (version 2.0), using a PAM 120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
- Treatment refers to the administration of an effective amount of an antibody, common gamma chain cytokine, and/or immune checkpoint inhibitor according to the present disclosure with the purpose of easing, ameliorating, arresting, eradicating (curing) or preventing symptoms or disease states.
- an "effective amount” or “an amount effective to treat” refer to an amount of an agent described herein (e.g., a multispecific antibody, common gamma chain cytokine, and/or immune checkpoint inhibitor, either alone or in combination) effective, at dosages and for periods of time necessary, to achieve a desired therapeutic result (e.g. , prevent or treat cancer in an individual).
- An effective amount of a polypeptide, such as an antibody may vary according to factors such as the disease stage, age, sex, and weight of the individual, the timing of administration, route of administration; the duration of the treatment; drugs used in combination; the judgment of the prescribing physician; and like factors known in the medical arts.
- an effective amount is also one in which any toxic or detrimental effects of the antibody are outweighed by the therapeutically beneficial effects.
- Administration may be carried out by any suitable route, but will typically be parenteral, such as intravenous, intramuscular, or subcutaneous.
- the size of the dose will also be determined by the existence, nature, and extent of any adverse side- effects that might accompany the administration of a particular active, and the desired physiological effect. It will be appreciated by one of skill in the art that various diseases or disorders could require prolonged treatment involving multiple administrations, perhaps using various rounds of administration.
- a “dosing interval” refers to the frequency of administration of a particular agent (e.g., the frequency of administration of a multispecific antibody, common gamma chain cytokine, or immune checkpoint inhibitor) during the course of treatment.
- the present disclosure provides methods of treating a cancer in a subject in need thereof comprising administering to the subject (i) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor and (ii) a common gamma chain cytokine.
- common gamma chain cytokine refers to a cytokine that binds the common gamma chain receptor (common yc), also known as CD132.
- Common gamma chain cytokines include IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21.
- the methods and kits described herein utilize IL-2 or IL-15.
- IL-2 and IL-15 bind a common heterodimeric receptor composed of the I L-2/15Rp (CD122) and the common y chains. The specificity of action of both cytokines is conferred by their corresponding a receptor chains, IL- 2Ra (CD25) and IL-15Ra (CD215).
- the present disclosure provides methods of treating a cancer in a subject in need thereof comprising administering to the subject (i) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor and (ii) IL- 2.
- the IL-2 used in the method of the present disclosure may be recombinant IL-2.
- IL-2 may be Aldesleukin (Proleukin®) for example presented in a 5 mL vial with 1.3 mg Aldesleukin/Proleukin (22 x 10 6 IU (international units) per vial), wherein the product can be reconstituted using sterile water for injection.
- Other suitable forms of IL-2 include NKTR-214 (Bempegaldesleukin/Nektar), ALK 4230 (nemvaleukin/Aalkermes-Reliant), SAR444245 - THOR 707/Synthorx/Sanofi) and XTX-202 (Xilio).
- the IL-2 is a variant IL-2.
- the variant IL-2 binds to the IL-2Rbeta and gamma chains, but does not bind or demonstrates reduced/abrogated binding to the IL-2Ralpha chain (e.g. NL201 from Neoleukin, Silva et al. De novo design of potent and selective mimics of IL-2 and IL-15. Nature 565, 186-191 (2019), and MDNA11 from Medicenna).
- the IL- 2 and/or variants thereof is pegylated.
- IL-2 may be administered via any suitable administration route, for example subcutaneously.
- the multispecific antibody is administered prior to the administration of IL-2.
- IL-2 is administered at least once, at least twice, at least three time, at least four time, at least five times, at least six times, or at least seven times after the administration of the multispecific antibody.
- the method comprises administration of a multispecific antibody followed by 1 , 2, 3, 4, 5, 6, 7, or more daily doses of IL-2.
- the multispecific antibody is administered concurrently with the administration of IL-2.
- the IL-2 is administered prior to the administration of the multispecific antibody.
- IL-2 or a variant thereof may be administered at any suitable dose.
- IL-2 may for example be given at a dose of between 0.1 and 5 MIU (0.1 and 5x10 6 IU), between 0.1 and 2.5 MIU/day, between 0.1 and 3.5 MIU/day, between 0.1 and 4.5 MIU, between 0.5 and 3.5 MIU/day, between 0.5 and 2.5 MIU/day, or between 0.5 and 1.5 MIU/day.
- IL-2 is given at a dose of 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, or 1.0 MIU.
- IL-2 is given at a daily dose of 1.0 MIU.
- IL-2 is given at a dose of 1.1 , 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1 , 2.2, 2.3, 2.4, 2.5, 2.6, 2.7,
- IL-2 is given at a dose of 2.0 MIU. In some embodiments, IL-2 is given at a dose of 3.0 MIU.
- LDSC IL-2 Low dose subcutaneous interleukin-2
- LDSC IL-2 Low dose subcutaneous interleukin-2
- IL-2 is given as a “low-dose”.
- a low dose of IL-2 refers to a dose that is less than 3MIU. In some embodiments, IL-2 is given at a dose of less than 3.0 MIU.
- IL-2 is given at a dose of less than 2.0 MIU. In some embodiments, IL-2 is given at a dose of less than 1.0 MIU. IL-2 may also be given multiple times daily, for example twice daily at a dose of between 0.1 and 5 MIU (0.1 and 5x10 6 IU), such as a dose of between 0.2 and 2 MIU, e.g. 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 1.1 , 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8,
- 0.1 and 5 MIU 0.1 and 5x10 6 IU
- IL-2 in IL-2 is dosed at different amounts depending on the treatment cycle. In some embodiments, IL-2 is dosed at a lower amount during the first treatment cycle and the dose is increased during subsequent treatment cycles.
- IL-2 may be administered with any suitable dosing interval, for example a dosing interval of 1 day to 1 month, for example a dosing interval of 1 to 21 days, such as 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days or a dose interval of between 7 and 21 days, such as 7 or 14 days.
- the dosing of the IL-2 is multiple times daily, for example the dose of IL-2 can be dosed 1 , 2, 3, 4, 5, or 6 times daily. In some embodiments, the IL-2 is dosed once daily. In some embodiments, the IL-2 is dosed twice daily.
- the present disclosure provides methods of treating a cancer in a subject in need thereof comprising administering to the subject (i) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor and (ii) IL- 15.
- IL-15 Suitable forms of IL-15 are known in the art. See e.g., Int J Mol Sci. 2022 Jul; 23(13): 7311 , which is herein incorporated by reference in its entirety.
- the IL- 15 is a heterodimer of IL-15 and IL-15 receptor alpha (hetlL-15), which is also referred to as NIZ985.
- the IL-15 is N-803, formerly known as ALT-803, which is an IL-15 variant complexed with a human IL-15Ra sushi domain-Fc fusion protein.
- the IL-15 is SOT101 , also referred to as Nanrilkefusp alfa, which is a human fusion protein comprising the cytokine IL-15 and the high-affinity binding sushi+ domain of IL- 15 receptor alpha (IL-15Ra).
- the IL-15 is NKTR-255, which is a polyethylene glycol-conjugate of rhlL-15.
- NKTR-255 is dosed at 1.5 pg/kg, 3.0 pg/kg, or 3.0/6.0 pg/kg.
- the IL-15 and/or variants thereof is pegylated.
- the multispecific antibody is administered prior to the administration of IL-15.
- IL-15 is administered at least once, at least twice, at least three times, at least four times, at least five times, at least six times, or at least seven times after the administration of the multispecific antibody.
- the method comprises administration of a multispecific antibody followed by 1 , 2, 3, 4, 5, 6, 7, or more daily doses of IL-15.
- the multispecific antibody is administered concurrently with the administration of IL-15.
- the IL-15 is administered prior to the administration of the multispecific antibody.
- IL-15 may be administered at any suitable dose.
- the IL-15 is given at a dose of between 0.001-1000 MIU (0.001 and 1000x10 6 IU)).
- the dose of IL-15 is between 0.01-100 MIU.
- the dose of IL-15 is between 0.1-10 Mill.
- the dose of IL-15 is between 0.5-5 Mill.
- the dose of IL-15 is between 1-5 MIU.
- the dose of IL-15 is between 2-5 MIU.
- the IL-15 is given at a dose of between 0.5 and 1.5 MIU/day, e.g.
- IL-15 may also be given multiple times daily, for example twice daily at a dose of between 0.1 and 5 MIU (0.1 and 5 10 6 IU), such as a dose of between 0.2 and 2MIU/day, such as between 0.5 and 1.5 MIU, e.g. 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 1.1 , 1.2, 1.3, 1 ,4 or 1.5 MIU.
- the IL-15 is given at a dose of between 0.01-40 pg/kg. In some embodiments, the dose of IL-15 is between 0.05-20 pg/kg. In some embodiments, the dose of IL-15 is between 0.1-10 pg/kg. In some embodiments, the dose of IL-15 is between 1-15 pg/kg.
- the dose is any dose between 1 and 15 pg/kg, such as 1 pg/kg, 1.5 pg/kg, 2 pg/kg, 2.5 pg/kg, 3 pg/kg, 3.5 pg/kg, 4 pg/kg, 4.5 pg/kg, 5 pg/kg, 5.5 pg/kg, 6 pg/kg, 6.5 pg/kg, 7 pg/kg, 7.5 pg/kg, 8 pg/kg, 8.5 pg/kg, 9 pg/kg, 9.5 pg/kg, 10 pg/kg, 10.5 pg/kg, 11 pg/kg, 11.5 pg/kg, 12 pg/kg, 12.5 pg/kg, 13 pg/kg, 13.5 pg/kg, 14 pg/kg, 14.5 pg/kg, or 15 pg/kg.
- in IL-15 is dosed at different amounts depending on
- IL-15 may be administered with any suitable dosing interval, for example a dosing interval of 1 day to 1 month, for example a dosing interval of 1 to 21 days, such as 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days or a dose interval of between 7 and 21 days, such as 7 or 14 days.
- the dosing of the IL-15 is multiple times daily, for example the dose of IL-15 can be dosed 1 , 2, 3, 4, 5, or 6 times daily.
- the IL-15 is dosed once daily.
- the IL-15 is dosed twice daily.
- the present disclosure provides methods of treating a cancer in a subject in need thereof comprising administering to the subject (i) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor and (ii) an immune checkpoint inhibitor.
- Immune checkpoint inhibitors are known in the art, including agents that target PD-1 , PD-L1 , CTLA4, LAG-3, TIM-3, TIGIT, and/or NKG2A. In some embodiments, the immune checkpoint inhibitors target PD-1 or PD-L1
- the immune checkpoint inhibitor is an anti-PD1 antibody or an anti-PD-L1 antibody.
- the anti-PD1 antibody is selected from pembrolizumab (Keytruda®), nivolumab (Opdivo®), ZynzTM (retifanlimab), JemperliTM (dostarlimuab), cemiplimab (Libtayo®), Tislelizumab (BGB-A317), and Sintilimab (IBI308).
- the anti-PD-L1 antibody is selected from atezolizumab (Tecentriq®), durvalumab (Imfinzi®), and avelumab (Bavencio®). In some embodiments, the anti-PD1 antibody is pembrolizumab.
- the immune checkpoint inhibitor is an anti-CTLA4 antibody.
- the anti-CTLA4 antibody is ipilimumab (Yervoy®).
- the immune checkpoint inhibitor is an inhibitor of LAG3.
- the inhibitor is an anti-LAG3 antibody selected from relatlimab, Tebotelimab, or Favezelimab. Additional LAG-3 antibody and inhibitors are summarized in Huo et al., The promising immune checkpoint LAG-3 in cancer immunotherapy: from basic research to clinical application. Front Immunol. 2022 Jul 26;13:956090, incorporated herein by reference.
- the immune checkpoint inhibitor is an inhibitor of TIM3 or TIGIT.
- the immune checkpoint inhibitor is an inhibitor of NKG2A.
- the inhibitor of NKG2A is Monalizumab.
- the multispecific antibody is administered prior to the administration of the immune checkpoint inhibitor (e.g., an anti-PD1 antibody). In some embodiments, the multispecific antibody is administered concurrently with the administration of immune checkpoint inhibitor (e.g., an anti-PD1 antibody). In some embodiments, the immune checkpoint inhibitor (e.g., an anti-PD1 antibody) is administered prior to the administration of the multispecific antibody.
- the immune checkpoint inhibitor e.g., an anti-PD1 antibody
- the immune checkpoint inhibitor is administered at any suitable dose and dosing interval according to the agent in use.
- the immune checkpoint inhibitor is an antibody and is given at a dose of between 100 and 1000, such as a dose of between 200-800 mg, such as dose between 300-500 mg, such as a dose of 400 mg.
- the dosing is every 6 weeks.
- the immune checkpoint inhibitor is an anti-PD1 antibody.
- the immune checkpoint inhibitor antibody is administered according to the instructions on the approved label.
- the anti-PD1 antibody is pembrolizumab.
- pembrolizumab is administered at a dose specified on the drug label for the appropriate indication.
- the pembrolizumab label is available at accessdata.fda.gov/drugsatfda_docs/label/2021/125514s096lbl.pdf.
- pembrolizumab is dosed at 400 mg IV over 30 minutes every 6 weeks.
- the nivolumab label is available at accessdata.fda.gov/drugsatfda_docs/label/2018/125554s058lbl.pdf. Additional drug labels for approved products are available at the FDA website.
- the anti-PD1 or anti-PD-L1 antibody may be administered with any suitable dosing interval, for example a dosing interval of 1 day to 2 month, for example a dosing interval of 1 to 50 days, such as 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 28, 35, 42, or 49 days or a dose interval of between 7 and 49 days, such as 7, 14, 21 , 28, 35, or 42 days.
- the anti-PD1 antibody is administered every 42 days ⁇ e.g., every 6 weeks).
- the multispecific antibody is administered prior to the administration of the immune checkpoint inhibitor (e.g., an anti-PD1 antibody). In some embodiments, the multispecific antibody is administered concurrently with the administration of immune checkpoint inhibitor (e.g., an anti-PD1 antibody). In some embodiments, the immune checkpoint inhibitor (e.g., an anti-PD1 antibody) is administered prior to the administration of the multispecific antibody.
- the immune checkpoint inhibitor e.g., an anti-PD1 antibody
- the immune checkpoint inhibitor (e.g., an anti-PD1 antibody) is used in combination with a common gamma chain cytokine (e.g., IL-2 or IL-15) described above.
- the multispecific antibody is administered prior to the administration of the immune checkpoint inhibitor (e.g., an anti-PD1 antibody) and IL-2 or IL- 15.
- the multispecific antibody is administered concurrently with the administration of immune checkpoint inhibitor (e.g., an anti-PD1 antibody) and IL-2 or IL-15.
- the immune checkpoint inhibitor e.g., an anti-PD1 antibody
- IL-2 or IL-15 are administered prior to the administration of the multispecific antibody.
- IL-2 or IL-15 and/or anti-PD1 or anti-PD-L1 antibody may be administered daily for at least 1 day, such as 2, 3, 4, 5, 6, 7 or more days after administration of the multispecific antibody, for example starting the day after administration of the multispecific antibody.
- the multispecific antibody may be administered on day 1 and IL-2 or IL-15 and/or anti-PD1 or anti- PD-L1 antibody may be administered on day 2 or on day 2, 3 and 4.
- Such a cycle may be repeated at least 1 time, such as 2, 3, 4, 5, 6, 7, 8 or more times, e.g. with intervals between the multispecific antibody administration of 14 days.
- the multispecific antibody may be administered with any suitable dosing interval, for example a dosing interval of 1 day to 1 month, for example a dosing interval of 1 to 21 days, such as 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days or a dose interval of between 7 and 21 days, such as 7 or 14 days. In some embodiments, the multispecific antibody is administered every 14 days. [113] The multispecific antibody may be administered at any suitable dose.
- the multispecific antibody is administered at a dose of at least 1 pg, at least 2 pg, at least 3 pg, at least 4 pg, at least 5 pg, at least 6 pg, at least 7 pg, at least 8 pg, at least 9 pg, at least 10 pg, at least 11 pg, at least 12 pg, at least 13 pg, at least 14 pg, at least 15 pg, at least 16 pg, at least 17 pg, at least 18 pg, at least 19 pg, at least 20 pg, at least 25 pg, at least 30 pg, at least 35 pg, at least 40 pg, or at least 45 pg.
- the multispecific antibody is administered at a dose of at least 100 pg, at least 110 pg, at least 120 pg, or at least 130 pg. In some embodiments, the multispecific antibody is administered at a dose of at least 300 pg, at least 310 pg, at least 320 pg, at least 330 pg, at least 340 pg, at least 350 pg, at least 360 pg, at least 370 pg, at least 380 pg, at least 390 pg, or at least 400 pg.
- the multispecific antibody is administered at a dose of at least 500 pg, at least 510 pg, at least 520 pg, at least 530 pg, at least 540 pg, at least 550 pg, at least 560 pg, at least 570 pg, at least 580 pg, at least 590 pg, or at least 600 pg.
- the multispecific antibody is administered at a dose of at least 750 pg, at least 760 pg, at least 770 pg, at least 780 pg, at least 790 pg, at least 800 pg, at least 810 pg, at least 820 pg, at least 830 pg, at least 840 pg, or at least 850 pg.
- the multispecific antibody is administered at a dose of at least 1000 pg, at least 1.1 mg, at least 1.2 mg, at least 1.3 mg, at least 1.4 mg, at least 1.5 mg, at least 1.6 mg, at least 1.7 mg, at least 1.8 mg, at least 1.9 mg, or at least 2 mg.
- the multispecific antibody is administered at a dose of at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 4.5 mg, at least 5 mg, at least 5.5 mg, at least 6 mg, at least 6.5 mg, at least 7 mg, at least 7.5 mg, at least 8 mg, at least 8.5 mg, at least 9 mg, at least 9.5 mg, or at least 10 mg.
- the multispecific antibody is administered at a dose of at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 60 mg, at least 70 mg, at least 80 mg, at least 90 mg, or at least 100 mg.
- he multispecific antibody is administered at a dose of at least 110 mg, at least 120 mg, at least 130 mg, at least 140 mg, at least 150 mg, at least 160 mg, at least 170 mg, at least 180 mg, at least 190 mg, or at least 200 mg. In some embodiments, the multispecific antibody is administered at a dose between 360 micrograms and 10 mg per administration. In some embodiments, the multispecific antibody is administered at a dose between 10 mg and 100 mg per administration.
- the multispecific antibody is administered once every 14 days at a dose of at least 1 pg, at least 2 pg, at least 3 pg, at least 4 pg, at least 5 pg, at least 6 pg, at least 7 pg, at least 8 pg, at least 9 pg, at least 10 pg, at least 11 pg, at least 12 pg, at least 13 pg, at least 14 pg, at least 15 pg, at least 16 pg, at least 17 pg, at least 18 pg, at least 19 pg, at least 20 pg, at least 25 pg, at least 30 pg, at least 35 pg, at least 40 pg, or at least 45 pg.
- the multispecific antibody is administered once every 14 days at a dose of at least 100 pg, at least 110 pg, at least 120 pg, or at least 130 pg. In some embodiments, the multispecific antibody is administered once every 14 days at a dose of at least 300 pg, at least 310 pg, at least 320 pg, at least 330 pg, at least 340 pg, at least 350 pg, at least 360 pg, at least 370 pg, at least 380 pg, at least 390 pg, or at least 400 pg.
- the multispecific antibody is administered once every 14 days at a dose of at least 500 pg, at least 510 pg, at least 520 pg, at least 530 pg, at least 540 pg, at least 550 pg, at least 560 pg, at least 570 pg, at least 580 pg, at least 590 pg, or at least 600 pg.
- the multispecific antibody is once every 14 days administered at a dose of at least 750 pg, at least 760 pg, at least 770 pg, at least 780 pg, at least 790 pg, at least 800 pg, at least 810 pg, at least 820 pg, at least 830 pg, at least 840 pg, or at least 850 pg.
- the multispecific antibody is administered once every 14 days at a dose of at least 1000 pg, at least 1.1 mg, at least 1.2 mg, at least 1.3 mg, at least 1.4 mg, at least 1.5 mg, at least 1 .6 mg, at least 1.7 mg, at least 1.8 mg, at least 1.9 mg, or at least 2 mg.
- the multispecific antibody is administered once every 14 days at a dose of at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 4.5 mg, at least 5 mg, at least 5.5 mg, at least 6 mg, at least 6.5 mg, at least 7 mg, at least 7.5 mg, at least 8 mg, at least 8.5 mg, at least 9 mg, at least 9.5 mg, or at least 10 mg. In some embodiments, the multispecific antibody is administered once every 14 days at a dose between 360 micrograms and 10 mg per administration.
- the multispecific antibody is administered twice every 14 days at a dose of at least 1 pg, at least 2 pg, at least 3 pg, at least 4 pg, at least 5 pg, at least 6 pg, at least 7 pg, at least 8 pg, at least 9 pg, at least 10 pg, at least 11 pg, at least 12 pg, at least 13 pg, at least 14 pg, at least 15 pg, at least 16 pg, at least 17 pg, at least 18 pg, at least 19 pg, at least 20 pg, at least 25 pg, at least 30 pg, at least 35 pg, at least 40 pg, or at least 45 pg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 100 pg, at least 110 pg, at least 120 pg, or at least 130 pg. In some embodiments, the multispecific antibody is administered twice every 14 days at a dose of at least 300 pg, at least 310 pg, at least 320 pg, at least 330 pg, at least 340 pg, at least 350 pg, at least 360 pg, at least 370 pg, at least 380 pg, at least 390 pg, or at least 400 pg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 500 pg, at least 510 pg, at least 520 pg, at least 530 pg, at least 540 pg, at least 550 pg, at least 560 pg, at least 570 pg, at least 580 pg, at least 590 pg, or at least 600 pg.
- the multispecific antibody is twice every 14 days administered at a dose of at least 750 pg, at least 760 pg, at least 770 pg, at least 780 pg, at least 790 pg, at least 800 pg, at least 810 pg, at least 820 pg, at least 830 pg, at least 840 pg, or at least 850 pg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 1000 pg, at least 1.1 mg, at least 1.2 mg, at least 1.3 mg, at least 1.4 mg, at least 1.5 mg, at least 1 .6 mg, at least 1.7 mg, at least 1.8 mg, at least 1.9 mg, or at least 2 mg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 4.5 mg, at least 5 mg, at least 5.5 mg, at least 6 mg, at least 6.5 mg, at least 7 mg, at least 7.5 mg, at least 8 mg, at least 8.5 mg, at least 9 mg, at least 9.5 mg, or at least 10 mg. In some embodiments, the multispecific antibody is administered twice every 14 days at a dose between 360 micrograms and 10 mg per administration.
- the multispecific antibody is administered at a dose of at least
- I pg/kg at least 2 pg/kg, at least 3 pg/kg, at least 4 pg/kg, at least 5 pg/kg, at least 6 pg/kg, at least 7 pg/kg, at least 8 pg/kg, at least 9 pg/kg, at least 10 pg/kg, at least 11 pg/kg, at least 12 pg/kg, at least 13 pg/kg, at least 14 pg/kg, at least 15 pg/kg, at least 16 pg/kg, at least 17 pg/kg, at least 18 pg/kg, at least 19 pg/kg, at least 20 pg/kg, at least 25 pg/kg, at least 30 pg/kg, at least 35 pg/kg, at least 40 pg/kg, or at least 45 pg/kg.
- the multispecific antibody is administered at a dose of at least 100 pg/kg, at least 110 pg/kg, at least 120 pg/kg, or at least 130 pg/kg. In some embodiments, the multispecific antibody is administered at a dose of at least 300 pg/kg, at least 310 pg/kg, at least 320 pg/kg, at least 330 pg/kg, at least 340 pg/kg, at least 350 pg/kg, at least 360 pg/kg, at least 370 pg/kg, at least 380 pg/kg, at least 390 pg/kg, or at least 400 pg/kg.
- the multispecific antibody is administered at a dose of at least 500 pg/kg, at least 510 pg/kg, at least 520 pg/kg, at least 530 pg/kg, at least 540 pg/kg, at least 550 pg/kg, at least 560 pg/kg, at least 570 pg/kg, at least 580 pg/kg, at least 590 pg/kg, or at least 600 pg/kg.
- the multispecific antibody is administered at a dose of at least 750 pg/kg, at least 760 pg/kg, at least 770 pg/kg, at least 780 pg/kg, at least 790 pg/kg, at least 800 pg/kg, at least 810 pg/kg, at least 820 pg/kg, at least 830 pg/kg, at least 840 pg/kg, or at least 850 pg/kg.
- the multispecific antibody is administered at a dose of at least 1000 pg/kg, at least 1.1 mg/kg, at least 1.2 mg/kg, at least 1.3 mg/kg, at least 1.4 mg/kg, at least 1.5 mg/kg, at least 1.6 mg/kg, at least 1.7 mg/kg, at least 1.8 mg/kg, at least 1.9 mg/kg, or at least 2 mg/kg.
- the multispecific antibody is administered at a dose of at least 2.5 mg/kg, at least 3 mg/kg, at least 3.5 mg/kg, at least 4 mg/kg, at least 4.5 mg/kg, at least 5 mg/kg, at least 5.5 mg/kg, at least 6 mg/kg, at least 6.5 mg/kg, at least 7 mg/kg, at least 7.5 mg/kg, at least 8 mg/kg, at least 8.5 mg/kg, at least 9 mg/kg, at least 9.5 mg/kg, or at least 10 mg/kg.
- the multispecific antibody is administered once every 14 days at a dose of at least 1 pg/kg, at least 2 pg/kg, at least 3 pg/kg, at least 4 pg/kg, at least 5 pg/kg, at least 6 pg/kg, at least 7 pg/kg, at least 8 pg/kg, at least 9 pg/kg, at least 10 pg/kg, at least
- the multispecific antibody is administered once every 14 days at a dose of at least 100 pg/kg, at least 110 pg/kg, at least 120 pg/kg, or at least 130 pg/kg.
- the multispecific antibody is administered once every 14 days at a dose of at least 300 pg/kg, at least 310 pg/kg, at least 320 pg/kg, at least 330 pg/kg, at least 340 pg/kg, at least 350 pg/kg, at least 360 pg/kg, at least 370 pg/kg, at least 380 pg/kg, at least 390 pg/kg, or at least 400 pg/kg.
- the multispecific antibody is administered once every 14 days at a dose of at least 500 pg/kg, at least 510 pg/kg, at least 520 pg/kg, at least 530 pg/kg, at least 540 pg/kg, at least 550 pg/kg, at least 560 pg/kg, at least 570 pg/kg, at least 580 pg/kg, at least 590 pg/kg, or at least 600 pg/kg.
- the multispecific antibody is administered once every 14 days at a dose of at least 750 pg/kg, at least 760 pg/kg, at least 770 pg/kg, at least 780 pg/kg, at least 790 pg/kg, at least 800 pg/kg, at least 810 pg/kg, at least 820 pg/kg, at least 830 pg/kg, at least 840 pg/kg, or at least 850 pg/kg.
- the multispecific antibody is administered once every 14 days at a dose of at least 1000 pg/kg, at least 1.1 mg/kg, at least 1.2 mg/kg, at least 1.3 mg/kg, at least 1.4 mg/kg, at least 1.5 mg/kg, at least 1.6 mg/kg, at least 1.7 mg/kg, at least 1.8 mg/kg, at least 1.9 mg/kg, or at least 2 mg/kg.
- the multispecific antibody is administered once every 14 days at a dose of at least 2.5 mg/kg, at least 3 mg/kg, at least 3.5 mg/kg, at least 4 mg/kg, at least 4.5 mg/kg, at least 5 mg/kg, at least 5.5 mg/kg, at least 6 mg/kg, at least 6.5 mg/kg, at least 7 mg/kg, at least 7.5 mg/kg, at least 8 mg/kg, at least 8.5 mg/kg, at least 9 mg/kg, at least 9.5 mg/kg, or at least 10 mg/kg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 1 pg/kg, at least 2 pg/kg, at least 3 pg/kg, at least 4 pg/kg, at least 5 pg/kg, at least 6 pg/kg, at least 7 pg/kg, at least 8 pg/kg, at least 9 pg/kg, at least 10 pg/kg, at least 11 pg/kg, at least 12 pg/kg, at least 13 pg/kg, at least 14 pg/kg, at least 15 pg/kg, at least 16 pg/kg, at least 17 pg/kg, at least 18 pg/kg, at least 19 pg/kg, at least 20 pg/kg, at least 25 pg/kg, at least 30 pg/kg, at least 35 pg/kg, at least 40 pg/kg, or at least 45 pg/kg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 100 pg/kg, at least 110 pg/kg, at least 120 pg/kg, or at least 130 pg/kg. In some embodiments, the multispecific antibody is administered twice every 14 days at a dose of at least 300 pg/kg, at least 310 pg/kg, at least 320 pg/kg, at least 330 pg/kg, at least 340 pg/kg, at least 350 pg/kg, at least 360 pg/kg, at least 370 pg/kg, at least 380 pg/kg, at least 390 pg/kg, or at least 400 pg/kg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 500 pg/kg, at least 510 pg/kg, at least 520 pg/kg, at least 530 pg/kg, at least 540 pg/kg, at least 550 pg/kg, at least 560 pg/kg, at least 570 pg/kg, at least 580 pg/kg, at least 590 pg/kg, or at least 600 pg/kg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 750 pg/kg, at least 760 pg/kg, at least 770 pg/kg, at least 780 pg/kg, at least 790 pg/kg, at least 800 pg/kg, at least 810 pg/kg, at least 820 pg/kg, at least 830 pg/kg, at least 840 pg/kg, or at least 850 pg/kg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 1000 pg/kg, at least 1.1 mg/kg, at least 1.2 mg/kg, at least 1.3 mg/kg, at least 1.4 mg/kg, at least 1.5 mg/kg, at least 1.6 mg/kg, at least 1.7 mg/kg, at least 1.8 mg/kg, at least 1.9 mg/kg, or at least 2 mg/kg.
- the multispecific antibody is administered twice every 14 days at a dose of at least 2.5 mg/kg, at least 3 mg/kg, at least 3.5 mg/kg, at least 4 mg/kg, at least 4.5 mg/kg, at least 5 mg/kg, at least 5.5 mg/kg, at least 6 mg/kg, at least 6.5 mg/kg, at least 7 mg/kg, at least 7.5 mg/kg, at least 8 mg/kg, at least 8.5 mg/kg, at least 9 mg/kg, at least 9.5 mg/kg, or at least 10 mg/kg.
- CRS cytokine response syndrome
- AMG 160 and HPN424 aim to direct cytotoxic T cells to prostate tumors by targeting CD3 and PSMA simultaneously.
- Step dosing involves administering one or more priming doses of a multispecific antibody prior to the target dose.
- the priming dose is a lower dose of the multispecific antibody than the target dose. This allows the immune system to be primed in a stepwise manner in an effort to prevent an uncontrolled inflammatory response at the target dose (Ball et al, 2023).
- the term “priming dose” or as used herein refers to a dose of a multispecific antibody described herein that primes a subject for administration of a target dose of the multispecific antibody such that the target dose does not result in one or more therapy- related toxicities such as CRS.
- a “target dose” as used herein refers to the therapeutically effective dose of a multispecific antibody described herein or the dose of a multispecific antibody that is being evaluated for use as a therapeutically effective dose (e.g., in the dose escalation studies described herein).
- the priming dose is 25% or less of the target dose.
- the priming dose is 25% or less, 20% or less, 15% or less, 10% or less, or 5% or less of the target dose.
- the priming dose is 5%, 6%, 7%, 8%, 9%, 10%, 11 %, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% of the target dose.
- the priming dose is 15% of the target dose.
- the priming dose is 50% of the target dose.
- the priming dose is 1 pg, 2 pg, 3 pg, 4 pg, 5 pg, 10 pg, 15 pg, 20 pg, 25 pg, 30 pg, 35 pg, 40 pg, 45 pg, 50 pg, 60 pg, 70 pg, 80 pg, 90 pg, 100 pg, 110 pg, 120 pg, 130 pg, 140 pg, 150 pg, 160 pg, 170 pg, 180 pg, 190 pg, 200 pg, 210 pg, 220 pg, 230 pg, 240 pg, 250 pg, 260 pg, 270 pg, 280 pg, 290 pg, 300 pg, 310 pg, 320 pg, 330 pg, 340 pg, 350 pg, 360 pg, 370
- the first priming dose is 120 pg
- the second priming dose is 360 pg.
- the target dose of the multispecific antibody is 120 pg and the priming dose is 18 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 540 pg and the priming dose is 81 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 800 pg and the priming dose is 120 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 800 pg and the priming doses are 120 pg and 360 pg.
- the target dose of the multispecific antibody is 1200 pg and the priming dose is 180 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 1200 pg and the priming doses are 120 pg and 360 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 1800 pg and the priming dose is 180 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 1800 pg and the priming doses are 120 pg and 360 pg.
- the target dose of the multispecific antibody is 1800 pg and the priming dose is 180 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 1800 pg and the priming doses are 120 pg and 360 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 3600 pg and the priming dose is 180 pg. In some embodiments, the target dose of the multispecific antibody (e.g., LAVA-1207) is 3600 pg and the priming doses are 120 pg and 360 pg.
- the two or more priming doses are administered 1 day apart. In some embodiments, the two or more priming doses are administered 2 days apart. In some embodiments, the two or more priming doses are administered 3 days apart. In some embodiments, the two or more priming doses are administered 4 days apart. In some embodiments, the two or more priming doses are administered 5 days apart. In some embodiments, the two or more priming doses are administered 6 days apart. some embodiments, the two or more priming doses are administered 7 days apart. In some embodiments, the two or more priming doses are administered 8 days apart. In some embodiments, the two or more priming doses are administered 9 days apart.
- the two or more priming doses are administered 10 days apart. In some embodiments, the two or more priming doses are administered 11 days apart. In some embodiments, the two or more priming doses are administered 12 days apart. In some embodiments, the two or more priming doses are administered 13 days apart. In some embodiments, the two or more priming doses are administered 14 days apart. In some embodiments, the first priming dose is administered on day 1 , and the second priming dose is administered on day 4. In some embodiments, the first priming does is administered on day 1 , and the second priming dose is administered on day 8.
- the last priming dose is administered 1 day before the target dose. In some embodiments, the last priming dose is administered 2 days before the target dose. In some embodiments, the last priming dose is administered 3 days before the target dose. In some embodiments, the last priming dose is administered 4 days before the target dose. In some embodiments, the last priming dose is administered 5 days before the target dose. In some embodiments, the last priming dose is administered 6 days before the target dose. In some embodiments, the last priming dose is administered 7 days before the target dose. In some embodiments, the last priming dose is administered 8 days before the target dose. In some embodiments, the last priming dose is administered 9 days before the target dose.
- the last priming dose is administered 10 days before the target dose. In some embodiments, the last priming dose is administered 11 days before the target dose. In some embodiments, the last priming dose is administered 12 days before the target dose. In some embodiments, the last priming dose is administered 13 days before the target dose. In some embodiments, the last priming dose is administered 14 days before the target dose.
- the first priming dose is administered on day 1
- the second priming dose is administered on day 4
- the target dose is administered on day 8.
- the first priming does is administered on day 1
- the second priming dose is administered on day 8
- the target dose is administered on day 15.
- the multispecific antibody is LAVA- 1207 and the first treatment cycle comprises administration of one or more priming doses and one or more target doses.
- the first treatment cycle comprises administration of one priming dose and one or more target doses. In some embodiments, the first treatment cycle comprises administration of two priming doses and one or more target doses. In some embodiments, the first treatment cycle comprises administration of three priming doses and one or more target doses. In some embodiments, the first treatment cycle comprises administration of one priming dose and one target dose. In some embodiments, the first treatment cycle comprises administration of two priming doses and one target dose. In some embodiments, the first treatment cycle comprises administration of three priming doses and one target dose.
- the multispecific antibody is LAVA-1207 and the second treatment cycle comprises administration of one or more priming doses and one or more target doses.
- the second treatment cycle comprises administration of one priming dose and one or more target doses.
- the second treatment cycle comprises administration of two priming doses and one or more target doses.
- the second treatment cycle comprises administration of three priming doses and one or more target doses.
- the second treatment cycle comprises administration of one priming dose and one target dose.
- the second treatment cycle comprises administration of two priming doses and one target dose.
- the second treatment cycle comprises administration of three priming doses and one target dose.
- the multispecific antibody is LAVA- 1207 and the first and second treatment cycles comprise administration of one or more priming doses and one or more target doses. In some embodiments, the first and second treatment cycles comprise administration of one priming dose and one or more target doses. In some embodiments, the first and second treatment cycles comprise administration of two priming doses and one or more target doses. In some embodiments, the first and second treatment cycles comprise administration of three priming doses and one or more target doses. In some embodiments, the first and second treatment cycles comprise administration of one priming dose and one target dose. In some embodiments, the first and second treatment cycles comprise administration of two priming doses and one target dose. In some embodiments, the first and second treatment cycles comprise administration of three priming doses and one target dose.
- each agent may be administered one or more times.
- the administration of the multispecific antibody (MSAb) as the 1 st agent may be administration of a single dose of a MSAb or multiple (e.g., 2, 3, 4, 5, 6, or more) doses of the MSAb over the course of one or more days
- the administration of the cytokine as the 2 nd agent may be administration of a single dose of a cytokine or multiple (e.g., 2, 3, 4, 5, 6, or more) doses of the cytokine over the course of one or more days.
- cytokine in TABLE 4 is intended to refer to a common gamma chain cytokine (e.g., IL-2 or IL-15).
- Reference to a MSAb includes bispecific T cell engagers (bsTCE). Inclusion of two agents as either the first or second agent is intended to refer to the concurrent administration of the listed agents
- MSAb multispecific antibody
- ICI immune checkpoint inhibitor
- a “treatment cycle” refers to a period of treatment (e.g., administration of a multispecific antibody, common gamma chain cytokine, or immune checkpoint inhibitor) optionally followed by a period of rest (e.g., wherein no therapeutic agents are administered).
- a full course of treatment may comprise one or more treatment cycles, e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, or more treatment cycle.
- a treatment cycle may be 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 or more days long.
- an exemplary course of treatment may comprise the following:
- the dosing interval of the multispecific antibody over the full course of treatment is every 14 days.
- an immune checkpoint inhibitor e.g., an anti-PD1 antibody
- an anti-PD1 antibody can be administered at a variety of points throughout the course of treatment.
- an anti-PD1 antibody can be administered before, concurrent with, or after a dose of LAVA-1207.
- an anti-PD1 antibody can be administered before, concurrent with, or after a dose of IL-2 and/or IL-15.
- the treatment regimen for LAVA- 1207 comprises administration of one or more premedications.
- the premedications are typically administered prior to administration of a target dose of LAVA- 1207.
- the premedications are administered with a priming dose of LAVA-1207.
- Predmedications can include one or more of an antipyretic (e.g., paracetamol or acetaminophen), an antihistamine (e.g. s diphenhydramine), and a corticosteroid (e.g., dexamethasone or methylprednisolone).
- the premedications include each of paracetamol or acetaminophen, diphenhydramine, and dexamethasone or methylprednisolone. In some embodiments, the premedications are given in combination with hydration (e.g. 1000 mL or more of fluids).
- methylprednisolone is administered at a dose of 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, or 50 mg. In some embodiments, methylprednisolone is administered at a dose of 20 mg. In some embodiments, dexamethasone is administered at a dose of 0.5 mg, 1 mg, 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 7 mg, 8 mg, 9 mg, or 10 mg. In some embodiments, dexamethasone is administered at a dose of 4 mg.
- acetaminophen is administered at a dose of 100 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg, 1000 mg, 1100 mg, 1200 mg, 1300 mg, 1400 mg, 1500 mg, 1600 mg, 1700 mg, 1800 mg, 1900 mg, or 2000 mg.
- diphenhydramine is administered at a dose of 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 110 mg, 120 mg, 130 mg, 140 mg, or 150 mg.
- premedications are administered 4, 3, 2, or 1 hours before the first dose of LAVA-1207.
- One or more of the premedications may be administered after LAVA- 1207 administrations have begun based on physician opinion.
- premedications are administered 4, 3, 2, or 1 hours before any of the LAVA-1207 doses (priming doses or target doses) shown in TABLE 5.
- a subject’s response to treatment with LAVA- 1207 can be monitored by subject sample collection prior to and during treatment with LAVA-1207.
- blood samples are taken from a subject prior to the start of treatment with LAVA-1207 and additional blood samples are taken at various timepoints after treatment has been initiated.
- These blood samples can be analyzed for pharmacodynamic characteristics including LAVA-1207 binding to Vy9V62 T cells, Vy9V ⁇ 52 T-cell activation status, general immune cell analysis (e.g., activation status and frequency of B cells, T cells, NK cells, and monocytes), cytokine levels, and anti-drug antibodies.
- additional blood samples are taken for use in cell-free DNA analysis of tumor mutation burden, tumor load, and/or circulating tumor cells.
- the methods of the present disclosure comprise administration of LAVA-1207 at a dose of 1.5 pg, 4.5 pg, 13.5 pg, 40 pg, 120 pg, 360 pg, 540 pg, 800 pg, 1.2 mg, 1.8 mg, 3.6 mg, 5.4 mg, 7.2 mg, 8.1 mg, 12 mg, 21 mg, 65 mg, or 200 mg once per 2 week treatment cycle for a total of 12 cycles.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first and second treatment cycle comprise administration of LAVA- 1207 at a dose of 1.5 pg, 4.5 pg, 13.5 pg, 40 pg, 120 pg, 360 pg, 540 pg, 800 pg, 1.2 mg, 1.8 mg, 3.6 mg, 5.4 mg, 7.2 mg, 8.1 mg, 12 mg, 21 mg, 65 mg, or 200 mg on the first day of the treatment cycle followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU for a total of 12 cycles.
- Cycles 5-12 comprise administration of LAVA-1207 at a dose of 1.5 pg, 4.5 pg, 13.5 pg, 40 pg, 120 pg, 360 pg, 540 pg, 800 pg, 1.2 mg, 1.8 mg, 3.6 mg, 5.4 mg, 7.2 mg, 8.1 mg, 12 mg, 21 mg, 65 mg, or 200mg, on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose of 120 pg on the first and second day of the treatment cycle, followed by administration of a target dose of 800 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA-1207 at a target dose of 800 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 800 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose of 120 pg on the first day of the treatment cycle, and a priming dose of 360 pg on the second day of the treatment cycle, followed by administration of a target dose of 800 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 Mill.
- the second, third, and fourth treatment cycles comprise administration of LAVA-1207 at a target dose of 800 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of 800 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 1200 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA- 1207 at a target dose of 1200 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of 1200 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose of 120 pg on the first day of the treatment cycle, and a priming dose of 360 pg on the second day of the treatment cycle, followed by administration of a target dose of 1200 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA-1207 at a target dose of 1200 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of 1200 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 1800 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA- 1207 at a target dose of 1800 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 1800 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 3600 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA- 1207 at a target dose of 3600 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 3600 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 5400 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA- 1207 at a target dose of 5400 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 5400 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 7200 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA- 1207 at a target dose of 7200 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 7200 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 8100 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA- 1207 at a target dose of 8100 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 8100 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 12000 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA-1207 at a target dose of 12000 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 Mill.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 12000 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 65000 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA-1207 at a target dose of 65000 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 65000 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise 12 2-week treatment cycles, wherein the first treatment cycle comprises administration of LAVA- 1207 at a priming dose on the first and second day of the treatment cycle, followed by administration of a target dose of 200000 pg on the third day of the treatment cycle, followed by 1 or 3 days of once daily doses of IL-2 at 1 MIU.
- the second, third, and fourth treatment cycles comprise administration of LAVA-1207 at a target dose of 200000 pg on the first day of the treatment cycle followed by 3 days of once daily doses of IL-2 at 1 MIU.
- Cycles 5-12 comprise administration of LAVA-1207 at a target dose of a target dose of 200000 pg on the first day of the treatment cycle.
- the methods of the present disclosure comprise at least 4 treatment cycles, wherein the first cycle is 4 weeks and the second - fourth cycles are two weeks each.
- the first cycle comprises administration of a first priming dose of 120 pg LAVA-1207 on day 1 and administration of a target dose of 800 pg LAVA-1207 on day 15, wherein the target dose is administered on day 1 of each of cycles 2-4.
- the first cycle comprises administration of a first priming dose of 120 pg LAVA- 1207 on day 1 , a second priming dose of 360 pg LAVA-1207 on day 8, and a target dose of 800 pg LAVA- 1207 on day 15, wherein the target dose is administered on day 1 of each of cycles 2- 4.
- the first cycle comprises administration of a first priming dose of 120 pg LAVA-1207 on day 1 , a second priming dose of 360 pg LAVA-1207 on day 8, and a target dose of 1200 pg LAVA- 1207 on day 15, wherein the target dose is administered on day 1 of each of cycles 2-4.
- the first cycle comprises administration of a first priming dose of 120 pg LAVA-1207 on day 1 , a second priming dose of 360 pg LAVA-1207 on day 8, and a target dose of 800 pg LAVA-1207 on day 15, followed by 1 MIU of IL-2 on day 16, wherein the target dose is administered on day 1 of each of cycles 2-4, followed by 1 MIU of IL-2 on day 2 of each of cycles 2-4.
- the first cycle comprises administration of a first priming dose of 120 pg LAVA-1207 on day 1, a second priming dose of 360 pg LAVA-1207 on day 8, and a target dose of 800 pg LAVA-1207 on day 15, followed by 1 MIU of IL-2 on day 16, 17, and 18, wherein the target dose is administered on day 1 of each of cycles 2-4, followed by 1 MIU of IL-2 on day 2, 3, and 4 of each of cycles 2-4.
- the first cycle comprises administration of a first priming dose of 120 pg LAVA-1207 on day 1 , a second priming dose of 360 pg LAVA-1207 on day 8, and a target dose of 1200 pg LAVA-1207 on day 15, followed by 1 MIU of IL-2 on day 16, wherein the target dose is administered on day 1 of each of cycles 2-4, followed by 1 MIU of IL-2 on day 2 of each of cycles 2-4.
- the first cycle comprises administration of a first priming dose of 120 pg LAVA-1207 on day 1 , a second priming dose of 360 pg LAVA-1207 on day 8, and a target dose of 1200 pg LAVA-1207 on day 15, followed by 1 MIU of IL-2 on day 16, 17, and 18, wherein the target dose is administered on day 1 of each of cycles 2-4, followed by 1 MIU of IL-2 on day 2, 3, and 4 of each of cycles 2-4.
- the present disclosure provides a method of treating cancer in a subject in need thereof comprising administering a multispecific binding agent descried herein or a composition comprising the same and (i) a common gamma chain cytokine (e.g., IL-2 or IL-15) and/or (ii) an immune checkpoint inhibitor (e.g., an anti-PD1 or anti-PDL1 antibody).
- a multispecific binding agent descried herein or a composition comprising the same and (i) a common gamma chain cytokine (e.g., IL-2 or IL-15) and/or (ii) an immune checkpoint inhibitor (e.g., an anti-PD1 or anti-PDL1 antibody).
- a multispecific binding agent descried herein or a composition comprising the same and (i) a common gamma chain cytokine (e.g., IL-2 or IL-15) and/or (ii) an immune checkpoint inhibitor (e.g., an anti-PD1
- treating refers to the treatment of a cancer in a mammal, e.g., in a human, including (a) inhibiting the cancer, i.e., arresting cancer development or preventing cancer progression; (b) relieving the cancer, i.e., causing regression of the cancer state or relieving one or more symptoms of the cancer; and (c) curing the cancer, i.e., remission of one or more cancer symptoms.
- treatment may refer to a short-term (e.g., temporary and/or acute) and/or a long-term (e.g., sustained) reduction in one or more cancer symptoms.
- treatment results in an improvement or remediation of the symptoms of the cancer.
- the improvement is an observable or measurable improvement or may be an improvement in the general feeling of well-being of the subject.
- the subject may be a neonate, a juvenile, or an adult.
- Mammalian species that may be treated with the present methods include canines and felines; equines; bovines; ovines; etc. and primates, particularly humans.
- Animal models, particularly small mammals e.g. mice, rats, guinea pigs, hamsters, rabbits, etc. may be used for experimental investigations.
- the subject is a human.
- the cancer is one in which the target antigen is expressed on the tumor neo-vasculature or tumor-associated endothelial cells of primary or metastatic tumors, including colorectal cancer, lung cancer, non-small cell lung cancer, endometrial and ovarian cancer, uterine cancer, uterine corpus endometrial carcinoma, gastric cancer, urothelial carcinomas, hepatocellular carcinoma, oral squamous cancer, thyroid tumors and glioblastomas, or adenoid cystic carcinoma of the head and neck, head and neck squamous cell carcinoma, prostate cancer, non-metastatic prostate cancer, metastatic prostate cancer, therapy refractory metastatic castration resistant prostate cancer, glioblastoma multiform, or blastic plasmacytoid dendritic neoplasm.
- the target antigen is expressed on the tumor neo-vasculature or tumor-associated endothelial cells of primary or metastatic tumors, including colorectal cancer, lung cancer, non
- the cancer is a hematological malignancy such as T cell lymphoma, multiple myeloma, acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, mantle cell lymphoma, myelodysplastic syndrome pre-B acute lymphoblastic leukemia, B cell lymphoma, smoldering myeloma, myelomonocytic leukemias, lymphoplasmacytic lymphoma, hairy cell leukemia, splenic marginal zone lymphoma, non-Hodgkin's lymphoma, Hodgkin's lymphoma, follicular lymphoma, pancreatic cancer, colon cancer, B-cell acute lymphoblastic leukemia, B-cell lymphoma/leukemia, B-cell chronic lymphoproliferative disorders, Burkitt lymphoma or B acute lymphoblastic leuk
- T cell lymphoma
- the cancer is a solid tumor cancer, such as renal cell carcinoma, melanoma, breast cancer (including triple-negative breast cancer), gastro-esophageal cancer, small bowel carcinoma, central nervous system tumors, medulloblastomas, hepatocellular carcinoma, glioma, neuroblastoma, bladder cancer, sarcoma, penile cancer, basal cell carcinoma, merkel cell carcinoma, neuroendocrine carcinoma, neuroendocrine tumors, carcinoma of unknown primary (CUP), thymoma, vulvar cancer, cervical carcinoma, testicular cancer, cholangiocarcinoma, appendicular carcinoma, mesothelioma, ampullary carcinoma, anal cancer, or choriocarcinoma.
- a solid tumor cancer such as renal cell carcinoma, melanoma, breast cancer (including triple-negative breast cancer), gastro-esophageal cancer, small bowel carcinoma, central nervous system tumors, medulloblastomas, hepatocellular carcinoma, gli
- Administration of the agents described herein can occur by injection, irrigation, inhalation, consumption, electro-osmosis, hemodialysis, iontophoresis, and other methods known in the art.
- administration route is local or systemic.
- administration route is intraarterial, intracranial, intradermal, intraduodenal, intrammamary, intrameningeal, intraperitoneal, intrathecal, intramuscular, intratumoral, intravenous, intravitreal, ophthalmic, parenteral, spinal, subcutaneous, ureteral, urethral, vaginal, or intrauterine.
- the administration route is by infusion (e.g., continuous or bolus).
- infusion e.g., continuous or bolus
- methods for local administration that is, delivery to the site of injury or disease, include through an Ommaya reservoir, e.g. for intrathecal delivery (See e.g., US Patent Nos. 5,222,982 and 5,385,582, incorporated herein by reference); by bolus injection, e.g. by a syringe, e.g. into a joint; by continuous infusion, e.g. by cannulation, such as with convection (See e.g., US Patent Application Publication No.
- the administration route is by topical administration or direct injection.
- subjects are selected for treatment according to the methods described herein by one or more exclusion and/or inclusion criteria.
- Inclusion/exclusion criteria may include: age, sex, infection status (e.g., hepatis B or C), prior treatment regimens (e.g., immunosuppressants or aminobisphosphonates), presence of malignancies other than the cancer intended for treatment, and general health status.
- subjects are selected for treatment with LAVA-1207 by one or more of the following inclusion criteria:
- CT computed tomography
- MRI magnetic resonance imaging
- HBsAg hepatitis B surface antigen
- Subjects who are positive for anti-HBc or hepatitis C antibody may be included if they have a negative polymerase chain reaction (PCR) within 6 weeks prior to initial multispecific antibody administration. A subject who is PCR positive will be excluded.
- PCR polymerase chain reaction
- Unstable cardiovascular function is defined as: (a) symptomatic ischemia, or (b) uncontrolled clinically significant conduction abnormalities (i.e. ventricular tachycardia on antiarrhythmic agents are excluded; 1st degree atrioventricular block or asymptomatic left anterior fascicular block/right bundle branch block is not excluded), or (c) congestive heart failure New York Heart Association Class > 3, or (d) myocardial infarction within 3 months;
- the multispecific antibody comprises a first antigenbinding domain capable of binding a human cancer antigen and a second antigen-binding domain capable of binding a human Vy9V ⁇ 52 T cell receptor by binding either the Vy9 or V02 chain (e.g., a bispecific antibody).
- the multispecific antibody comprises a first antigenbinding domain that capable of binding a human cancer antigen, a second antigen-binding domain capable of binding a human Vy9V52 T cell receptor, and a third antigen-binding domain capable of binding to a third target antigen or epitope.
- the multispecific antibody further comprises a fourth, fifth, or sixth antigen-binding domain.
- any of the first, second, third, fourth, fifth, or sixth antigen-binding domains of the multispecific, antibody may be a single-domain antibody, such as a VHH.
- the first and second antigen binding domains of the multispecific antibody are VHHs.
- Antibodies may be formulated with pharmaceutically-acceptable excipients in accordance with conventional techniques such as those disclosed in Rowe et al. 2012 Handbook of Pharmaceutical Excipients, ISBN 9780857110275).
- the pharmaceutically-acceptable excipient as well as any other carriers, diluents or adjuvants should be suitable for the antibodies and the chosen mode of administration.
- Suitability for excipients and other components of pharmaceutical compositions is determined based on the lack of significant negative impact on the desired biological properties of the chosen antibody or pharmaceutical composition of the present disclosure (e.g., less than a substantial impact (10% or less relative inhibition, 5% or less relative inhibition, etc.) upon antigen binding).
- a pharmaceutical composition may include diluents, fillers, salts, buffers, detergents (e.g., a nonionic detergent, such as Tween-20 or Tween-80), stabilizers (e.g., sugars or protein-free amino acids), preservatives, tissue fixatives, solubilizers, and/or other materials suitable for inclusion in a pharmaceutical composition.
- detergents e.g., a nonionic detergent, such as Tween-20 or Tween-80
- stabilizers e.g., sugars or protein-free amino acids
- preservatives e.g., tissue fixatives, solubilizers, and/or other materials suitable for inclusion in a pharmaceutical composition.
- Further pharmaceutically- acceptable excipients include any and all suitable solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonicity agents, antioxidants and absorption-delaying agents and the like that are physiologically compatible with an antibody of the present disclosure.
- the multispecific antibody may be formulated at a strength of about 1 mg/mL.
- the formulation includes 1-20 mM histidine.
- the formulation includes 0.1-10 mM methionine.
- the formulation includes 50-500 mM sucrose.
- the formation includes 0.01-0.05% polysorbate 80.
- the formulation is at a pH 5.0-7.5.
- the multispecific antibody is formulated at a strength of 1 mg/mL.
- the formulation includes 10 mM histidine.
- the formulation includes 1 mM methionine. In some embodiments, the formulation includes 280 mM sucrose. In some embodiments, the formulation includes 0.02 % polysorbate 80. In some embodiments, the formulation is at pH 6.0.
- the multispecific antibody may be formulated at a strength of 1 mg/mL in 10 mM histidine, 1 mM methionine, 280 mM sucrose, 0.02 % polysorbate 80, pH 6.0. The multispecific antibody may be administered via any suitable administration route, for example intravenously or subcutaneously.
- Multispecific antibodies for use in the present disclosure are typically produced recombinantly, i.e. by expression of nucleic acid constructs encoding the antibodies in suitable host cells, followed by purification of the produced recombinant antibody from the cell culture.
- Nucleic acid constructs can be produced by standard molecular biological techniques well-known in the art. The constructs are typically introduced into the host cell using an expression vector. Suitable nucleic acid constructs and expression vectors are known in the art.
- Host cells suitable for the recombinant expression of antibodies are well-known in the art, and include CHO, H EK- 293, Expi293F, PER-C6, NS/0 and Sp2/0 cells.
- any of the multispecific antibodies of this disclosure are modified to reduce or eliminate the formation of pyroglutamic acid at the N-terminus of the modified multispecific antibodies as compared to the unmodified multispecific antibodies of this disclosure.
- the glutamate residue (E) or glutamine (Q) residue is modified to an aspartic acid residue (D).
- any of the multispecific antibodies of the present disclosure may comprise an aspartic acid (D) residue in the first position.
- the first antigen-binding region and second antigen-binding region of the multispecific antibody used in the present disclosure may be covalently linked via a peptide linker, e.g. a linker having a length of from 1 to 20 amino acids, e.g. from 1 to 10 amino acids, such as 2, 3, 4, 5, 6, 7, 8 or 10 amino acids.
- the peptide linker may comprise or consist of four glycine residues followed by a serine residue.
- the first antigen-binding region may be located N- terminally or C-terminally of the second antigen-binding region.
- the first and second antigen-binding regions may be linked or directly fused to Fc domain monomers.
- the multispecific or bispecific antibody used in the present disclosure is capable of binding a human Vy9V52 T cell receptor.
- the multispecific antibody is capable of binding to human V52.
- the term “human V02”, when used herein, refers to the rearranged 52 chain of the Vy9V62-T cell receptor (TOR).
- UniProtKB - A0JD36 (A0JD36_HUMAN) gives an example of a variable TRDV2 sequence.
- Antigen-binding domains that specifically bind to V52+ T cell receptors are known in the art.
- V52 antigen binding domain sequences are shown in TABLE 6A, TABLE 6B, TABLE 6C, and TABLE 6D.
- Table 6B Exemplary V52 TCR-specific CDR Sequences (IMGT)
- Table 6C Exemplary V52 TCR-specific CDR Sequences (Chothia)
- CDR sequences of a given binding agent as determined by multiple numbering systems are compiled into a single sequence that encompasses the entirety of each of the CDR amino acid ranges in the variable region.
- CDRs for the 6H4 VHH described in Tables 6A, 6B, and 6C are shown in FIG. 20.
- the N- and C-terminal ranges for each CDR region are indicated by dashed lines.
- the combined CDR sequences covering the entirety of these ranges are shown in the last row.
- Illustrative combined CDRs for V52 antigen-binding domains are shown in Table 6D.
- a 6H4 VHH comprising a CDR1 amino acid sequence comprising SEQ ID NO: 26, a CDR2 amino acid sequence comprising SEQ ID NO: 27, and a CDR3 amino acid sequence comprising SEQ ID NO: 28 as defined by the Kabat numbering system
- a 6H4 VHH comprising a CDR1 amino acid sequence comprising SEQ ID NO: 45, a CDR2 amino acid sequence comprising SEQ ID NO: 46, and a CDR3 amino acid sequence comprising SEQ ID NO: 47 as defined by the IMGT numbering system
- (2) a 6H4 VHH comprising a CDR1 amino acid sequence comprising SEQ ID NO: 63, a CDR2 amino acid sequence comprising SEQ ID NO: 64, and a CDR3 amino acid sequence comprising SEQ ID NO: 65 as defined by the Chothia numbering system
- a 6H4 VHH comprising a CDR1 amino acid sequence comprising SEQ ID NO: 26
- the multispecific antibody is capable of binding to human Vy9.
- human Vy9 when used herein, refers to the rearranged Vy9 chain of the Vy9V62-T cell receptor (TOR).
- UniProtKB - Q99603 (TRGV9_HUMAN) gives an example of a variable Vy9 sequence.
- Exemplary Vy9V ⁇ 52 TCR antigen-binding domain sequences are known in the art, see e.g. WO 2015/156673.
- the (second) antigen-binding domain capable of binding the human Vy9V ⁇ 52 T cell receptor may comprise the VH CDR1 sequence of SEQ ID NO:5, the VH CDR2 sequence of SEQ ID NO: 6 and the VH CDR3 sequence of SEQ ID NO: 7.
- the second antigen-binding region may comprise an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 8.
- the second antigen-binding region may comprise an amino acid sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 9.
- the second antigen-binding domain comprises or consists of SEQ ID NO: 8.
- the second antigen-binding domain comprises or consists of SEQ ID NO: 9.
- the (second) antigen-binding domain capable of binding the human Vy9V52 T cell receptor may comprise the VH CDR1 sequence of SEQ ID NO: 10, the VH CDR2 sequence of SEQ ID NO: 11 and the VH CDR3 sequence of SEQ ID NO: 12.
- the second antigen-binding region may comprise an amino acid sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 13.
- the second antigen-binding domain comprises or consists of SEQ ID NO: 13.
- the (second) antigen-binding domain capable of binding the human Vy9V52 T cell receptor may comprise the VH CDR1 sequence of SEQ ID NO: 14, the VH CDR2 sequence of SEQ ID NO: 15 and the VH CDR3 sequence of SEQ ID NO: 16.
- the second antigen-binding region may comprise an amino acid sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 17.
- the second antigen-binding domain comprises or consists of SEQ ID NO: 17.
- the (second) antigen-binding domain capable of binding the human Vy9V ⁇ 52 T cell receptor may comprise the VH CDR1 sequence of SEQ ID NO: 18, the VH CDR2 sequence of SEQ ID NO: 19 and the VH CDR3 sequence of SEQ ID NO: 20.
- the second antigen-binding region may comprise an amino acid sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 21.
- the second antigen-binding domain comprises or consists of SEQ ID NO: 21.
- the (second) antigen-binding domain capable of binding the human Vy9V ⁇ 52 T cell receptor may comprise the VH CDR1 sequence of SEQ ID NO: 22, the VH CDR2 sequence of SEQ ID NO: 23 and the VH CDR3 sequence of SEQ ID NO: 24.
- the second antigen-binding region may comprise an amino acid sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 25.
- the second antigen-binding domain comprises or consists of SEQ ID NO: 25.
- the multispecific antibody used in the method of the present disclosure competes (i.e. is able to compete) for binding to V ⁇ 52 with an antibody having a sequence selected from SEQ ID NOs: 4, 8, 9, 13, 17, 21 , 25, and 29.
- the multispecific antibody used in the method of the present disclosure may bind to the same epitope on human V52 as an antibody having a sequence selected from SEQ ID NOs: 4, 8, 9, 13, 17, 21 , 25, and 29.
- the multispecific or bispecific antibody used in the present disclosure is capable of binding a human cancer antigen.
- the human cancer antigen may for example be an antigen associated with solid tumors or an antigen associated with hematological cancer disease. Human cancer antigens may be exclusively expressed on malignant cells or overexpressed on malignant cells relative to healthy cells.
- the human cancer antigen is selected from HER2/neu, EGFR, CA- 125, PSA, CD44, EpCAM, CEACAM5, GD2, CD1d, CD40, CD123, 5T4, Nectin-4, PSMA, B7-H3, RAGE, CA72-4, HE4, Glypican-3, PSMA, MUC1 , CD133, EGFRvlll, CAIX, CD24, CD166, CD47, CD70, CD133, CD276 (B7-H3) , CD271 , CD146, CD164, CD123, CD38, CD166, CD24, CD29, CD49f, CD56, CD20, CD71 , CD98, CD99, CD147, CD166, CD200, CD184 (CXCR4), CD44v6, CD271 , CD276, CD304 (BDCA-4), CD326 (EpCAM), and CD338 (ABCB5).
- the human cancer antigen is selected from the group consisting of PSMA, CD1d, CD40, CD123, 5T4, and Nectin-4. In some embodiments, the human cancer antigen is selected from the group consisting of PSMA, CD1d, CD40, CD123, 5T4, EGFR, and Nectin-4. In some embodiments, the human cancer antigen is selected from PSMA, CD1d, CD40, CD123, 5T4, EGFR, CD33, and Nectin-4. Exemplary cancer antigen-specific CDR and VHH sequences are provided in TABLE 7A, TABLE 7B, TABLE 7C, and TABLE 7D below.
- Table 7 A Exemplary cancer antigen-specific CDR (Kabat) and VHH Sequences
- Table 7B Exemplary cancer antigen-specific CDR Sequences (Chothia)
- Table 7C Exemplary cancer antigen-specific CDR Sequences (IMGT)
- Table 7D Exemplary cancer antigen-specific CDR Sequences (Combined)
- the human cancer antigen may be PSMA.
- Multispecific antibodies having antigen-binding regions capable of binding a human PSMA suitable for use in the present disclosure have for example been described in WO 2022/008646 (herein incorporated by reference).
- the (first) antigen-binding region of the multispecific antibody used in the present disclosure may comprise the VH CDR1 sequence of SEQ ID NO: 87, the VH CDR2 sequence of SEQ ID NO: 88 and the VH CDR3 sequence of SEQ ID NO: 89.
- the first antigen-binding region may comprise an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 90.
- the first antigen-binding region comprises or consists of SEQ ID NO: 90.
- the multispecific antibody used in the method of the present disclosure may compete (i.e. be able to compete) for binding to PSMA with an antibody having the sequence set forth in SEQ ID NO:90.
- the multispecific antibody may bind the same epitope on human PSMA as an antibody having the sequence set forth in SEQ ID NO:90.
- the multispecific antibody used in the present disclosure may comprise a first antigen-binding region and a second antigen-binding region, wherein the first antigen-binding region comprises the CDR1 sequence set forth in SEQ ID NO: 87 , the CDR2 sequence set forth in SEQ ID NO:88 and the CDR3 sequence set forth in SEQ ID NO:89 and wherein the second antigen-binding region comprises the CDR1 sequence set forth in SEQ ID NO:5, the CDR2 sequence set forth in SEQ ID NO:6 and the CDR3 sequence set forth in SEQ ID NO:7.
- the multispecific antibody used in the present disclosure may comprise a first antigen-binding region and a second antigen-binding region, wherein the first antigen-binding region comprises the sequence set forth in SEQ ID NO:90 and the second antigen-binding region comprises the sequence set forth in SEQ ID NO:8.
- the second antigen-binding domain comprises any of the CDR combinations recited in Tables 6A-6D.
- the multispecific antibody may comprise or consist of the sequences set forth in SEQ ID NO:334 and SEQ ID NO:335.
- Such a multispecific antibody is referred to herein as LAVA-1207 or PSMA-V62-Fc. Sequences for LAVA-1207 are provided below in TABLE 8. Antigen binding domains are shown in underlined text; hinge regions are in bold an italicized text; DEL sequence is shown in boxed text; Fc domains are in regular text with mutations relative to WT I gG 1 in bold and underlined text.
- the multispecific antibody of the present disclosure may be capable of mediating killing of PSMA-expressing cells, e.g. LNCaP cells, 22Rv1 cells or VCaP cells through activation of Vy9V32 T cells.
- the multispecific antibody may be capable of inducing killing of LNCaP cells through activation of Vy9V62 T cells with an EC50 value of 50 pM or less, such as 25 pM or less, e.g. 20 pM or less, such as 15 pM or less, e.g. 10 pM or less, or even 5 pM or less, such as 2 pM or less or 1 pM or less when tested as described in Example 6 of WO 2022/008646.
- the multispecific antibody may be capable of inducing killing of LNCaP, 22Rv1 or VCaP cells through activation of Vy9V52 T cells with an EC50 value of 50 pM or less, such as 25 pM or less, e.g. 20 pM or less, such as 15 pM or less when tested after 24 hours as described in Example 13 of WO 2022/008646, preferably both at a 1 :1 and a 1 :10 effector to target cell ratio.
- the multispecific antibody of the present disclosure may be capable of binding to the PSMA positive prostate cancer cell line LNCaP with an EC50 of 50 nM or less, such as 20 nM or less, e.g. 10 nM of less, when tested as described in Example 7 of WO 2022/008646.
- the multispecific antibody of the present disclosure may further or alternatively be capable of binding to Vy9V52 T cells with an EC50 of 10 nM or less, such as 5 nM or less, e.g. 2 nM of less, when tested as described in Example 7 of WO 2022/008646.
- the multispecific antibody may be capable of binding to recombinant human PSMA protein with a KD value of 100 nM or less, such as 50 nM or less, when tested as described in Example 11 of WO 2022/008646.
- the multispecific antibody of the present disclosure may further or alternatively be capable of binding to human Vy9V52-Fcwith a KD value of 10 nM or less, such as 5 nM or less, e.g. 2 nM or less, such as 1 nM or less when tested as described in Example 11 of WO 2022/008646.
- the multispecific antibody may be capable of mediating killing of human PSMA- expressing cells from a prostate cancer patient. Killing of human PSMA-expressing cells from a prostate cancer patient may e.g. be determined as described in Example 10 of WO 2022/008646.
- the multispecific antibody of the present disclosure may further or alternatively be capable of mediating specific cell death of more than 25%, such as more than 50%, at a concentration of 50 nM, as determined in the assays described in Example 10 or Example 14 of WO 2022/008646.
- the multispecific antibody may not be capable of mediating killing of PSMA-negative cells, such as PSMA negative human cells.
- the multispecific antibody may not induce IL-2, IL-4, IL-6, IL- 10 or TN Fa in whole blood from healthy donors at concentrations up to 280 nM, when tested as described in Example 16 of WO 2022/008646.
- the multispecific antibody may alternatively or in addition induce more than 10-fold less IL-8 and/or more than 50-fold less IFNy than Campath® in whole blood from healthy donors when tested as described in Example 16 of WO 2022/008646.
- the multispecific antibody used in the present disclosure may comprise a first antigenbinding region capable of binding PSMA and be for use in the treatment of prostate cancer, such as non-metastatic or metastatic prostate cancer, for example metastatic castration resistant prostate cancer, such as therapy refractory metastatic castration resistant prostate cancer.
- the multispecific antibody may comprise a first antigen-binding region capable of binding PSMA and be used for the treatment of prostate cancer in patients that have previously received at least one or more treatments for prostate cancer.
- the one or more treatments are selected from taxane-based chemotherapy, androgen receptor targeted therapy treatment, with an androgen biosynthesis inhibitor (e.g. abiraterone, enzalutamide, and/or apalutamide), treatment with a PARP inhibitor, and treatment with lutetium/radiolabeled PSMA.
- an androgen biosynthesis inhibitor e.g. abiraterone, enzalutamide, and/or apalutamide
- the multispecific antibody capable of binding PSMA may be for use in the treatment of cancers in which PSMA is expressed on the tumor neo-vasculature or tumor- associated endothelial cells of primary or metastatic tumors including from colorectal cancer, lung cancer, breast cancer, endometrial and ovarian cancer, gastric cancer, renal cell cancer, urothelial cancer, hepatocellular cancer, oral squamous cancer, thyroid tumors and glioblastomas.
- the multispecific antibody capable of binding PSMA may be for use in the treatment of adenoid cystic carcinoma of the head and neck.
- the human cancer antigen is CD1d.
- Antibodies having antigenbinding regions capable of binding a human CD1d suitable for incorporation into a multispecific antibody to be used in the present disclosure have for example been described in WO 2016/122320 and WO 2020/060405 (both herein incorporated by reference).
- the (first) antigen-binding region of the multispecific antibody used in the present disclosure may comprise the VH CDR1 sequence of SEQ ID NO:95, the VH CDR2 sequence of SEQ ID NO: 96 and the VH CDR3 sequence of SEQ ID NO: 97.
- the first antigen-binding region comprises an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 98.
- the first antigen-binding region comprises an amino acid sequence comprising or consisting of SEQ ID NO: 98.
- the multispecific antibody used in the method of the present disclosure may compete (i.e. be able to compete) for binding to CD1d with an antibody having the sequence set forth in SEQ ID NO: 98.
- the multispecific antibody may bind the same epitope on human CD1d as an antibody having the sequence set forth in SEQ ID NO: 98.
- the multispecific antibody used in the present disclosure may comprise a first antigenbinding region and a second antigen-binding region, wherein the first antigen-binding region comprises the CDR1 sequence set forth in SEQ ID NO: 95, the CDR2 sequence set forth in SEQ ID NO: 96 and the CDR3 sequence set forth in SEQ ID NO: 97 and wherein the second antigenbinding region comprises the CDR1 sequence set forth in SEQ ID NO: 5, the CDR2 sequence set forth in SEQ ID NO: 6 and the CDR3 sequence set forth in SEQ ID NO: 7.
- the second antigen-binding domain comprises any of the CDR combinations recited in Tables 6A-6D.
- the multispecific antibody used in the disclosure may comprise a first antigen-binding region capable of binding CD1d and be for use in the treatment of hematological malignancies such as T cell lymphoma, multiple myeloma, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, mantle cell lymphoma, B cell lymphoma, smoldering myeloma, Hodgkin lymphoma, myelomonocytic leukemias, lymphoplasmacytic lymphoma, hairy cell leukemia, or splenic marginal zone lymphoma, or solid tumors, such as renal cell carcinoma, melanoma, colorectal carcinoma, head and neck cancer, breast cancer, prostate cancer, lung cancer, pancreatic cancer, gastro-esophageal cancer, small bowel carcinoma, central nervous system tumors, medulloblastomas, hepatocellular carcinoma, ovarian cancer, glioma,
- the human cancer antigen is CD40.
- Antibodies having antigenbinding regions capable of binding a human CD40 suitable for incorporation into a multispecific antibody to be used in the present disclosure have for example been described in WO 2020/159368 (herein incorporated by reference).
- the (first) antigen-binding region of the multispecific antibody used in the present disclosure may comprise the VH CDR1 sequence of SEQ ID NO: 99, the VH CDR2 sequence of SEQ ID NO: 100, and the VH CDR3 sequence of SEQ ID NO: 101.
- the first antigen-binding region comprises an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 102.
- the first antigen-binding region comprises an amino acid sequence comprising or consisting of SEQ ID NO: 102.
- the multispecific antibody used in the method of the present disclosure may compete (i.e. be able to compete) for binding to CD40 with an antibody having the sequence set forth in SEQ ID NO: 102.
- the multispecific antibody may bind the same epitope on human CD40 as an antibody having the sequence set forth in SEQ ID NO: 102.
- the multispecific antibody used in the present disclosure may comprise a first antigenbinding region and a second antigen-binding region, wherein the first antigen-binding region comprises the CDR1 sequence set forth in SEQ ID NO: 99, the CDR2 sequence set forth in SEQ ID NO: 100 and the CDR3 sequence set forth in SEQ ID NO: 101 and wherein the second antigenbinding region comprises the CDR1 sequence set forth in SEQ ID NO: 5, the CDR2 sequence set forth in SEQ ID NO: 6 and the CDR3 sequence set forth in SEQ ID NO: 7.
- the second antigen-binding domain comprises any of the CDR combinations recited in Tables 6A-6D.
- the multispecific antibody used in the present disclosure may comprise a first antigenbinding region capable of binding CD40 and be for use in the treatment of chronic lymphocytic leukemia, multiple myeloma, non-Hodgkin's lymphoma, Hodgkin's lymphoma, follicular lymphoma, head and neck cancer, pancreatic cancer, ovarian cancer, lung cancer, breast cancer, colon cancer, prostate cancer, B-cell lymphoma/leukemia, Burkitt lymphoma or B acute lymphoblastic leukemia.
- the human cancer antigen is CD123.
- Antibodies having antigenbinding regions capable of binding a human CD123 suitable for incorporation into a multispecific antibody to be used in the present disclosure have for example been described in WO 2022/180271 (herein incorporated by reference).
- the (first) antigen-binding region of the multispecific antibody used in the present disclosure may comprise the VH CDR1 sequence of SEQ ID NO: 103, the VH CDR2 sequence of SEQ ID NO:104 and the VH CDR3 sequence of SEQ ID NO:105.
- the first antigen-binding region comprises an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to SEQ ID NO: 106.
- the first antigen-binding region comprises an amino acid sequence comprising or consisting of SEQ ID NO: 106.
- the multispecific antibody used in the method of the present disclosure may compete (i.e. be able to compete) for binding to CD123 with an antibody having the sequence set forth in SEQ ID NO: 106.
- the multispecific antibody may bind the same epitope on human CD123 as an antibody having the sequence set forth in SEQ ID NO: 106.
- the multispecific antibody used in the present disclosure may comprise a first antigenbinding region and a second antigen-binding region, wherein the first antigen-binding region comprises the CDR1 sequence set forth in SEQ ID NO: 103, the CDR2 sequence set forth in SEQ ID NO: 104 and the CDR3 sequence set forth in SEQ ID NO: 105 and wherein the second antigenbinding region comprises the CDR1 sequence set forth in SEQ ID NO: 5, the CDR2 sequence set forth in SEQ ID NO: 6 and the CDR3 sequence set forth in SEQ ID NO: 7.
- the second antigen-binding domain comprises any of the CDR combinations recited in Tables 6A-6D.
- the multispecific antibody used in the present disclosure may comprise a first antigenbinding region capable of binding CD123 and be for use in the treatment of acute myeloid leukemia, B-cell acute lymphoblastic leukemia, hairy cell leukemia, Hodgkin lymphoma, blastic plasmacytoid dendritic cell neoplasm, chronic myeloid leukemia, chronic lymphocytic leukemia, B-cell chronic lymphoproliferative disorders or myelodysplastic syndrome.
- the human cancer antigen is Nectin-4.
- the (first) antigen-binding region of the multispecific antibody used in the present disclosure may comprise the VH CDR1 sequence selected from SEQ ID NOs: 107, 11 , 115, 119, 123, 127, and 131 , the VH CDR2 sequence selected from SEQ ID NOs: 108, 112, 116, 120, 124, 128, and 132 and the VH CDR3 sequence selected from SEQ ID NOs: 109, 113, 117, 121 , 125, 129, 133.
- the first antigen-binding region comprises an amino acid sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to a sequence selected from SEQ ID NO: 110, 114, 118, 122, 126, 130, and 134. In some embodiments, the first antigenbinding region comprises an amino acid sequence comprising or consisting of a sequence selected from SEQ ID NO: 110, 114, 118, 122, 126, 130, and 134.
- the multispecific antibody used in the method of the present disclosure may competes (i.e. be able to compete) for binding to Nectin-4 with an antibody having a sequence selected from SEQ ID NO: 110, 114, 118, 122, 126, 130, and 134.
- the multispecific antibody may bind the same epitope on human Nectin-4 as an antibody having a sequence selected from SEQ ID NO: 110, 114, 118, 122, 126, 130, and 134.
- the multispecific antibody used in the present disclosure may comprise a first antigenbinding region capable of binding Nectin-4 and be for use in the treatment of bladder cancer, breast cancer, renal cancer, prostate cancer, ovarian cancer, esophageal cancer, head and neck cancer, lung cancer, pancreatic cancer, gastric cancer, thyroid cancer, colorectal cancer, cholangiocarcinoma, or uterine corpus endometrial carcinoma.
- 5T4 Multispecific Antibodies capable of binding Nectin-4 and be for use in the treatment of bladder cancer, breast cancer, renal cancer, prostate cancer, ovarian cancer, esophageal cancer, head and neck cancer, lung cancer, pancreatic cancer, gastric cancer, thyroid cancer, colorectal cancer, cholangiocarcinoma, or uterine corpus endometrial carcinoma.
- the human cancer antigen is 5T4.
- the (first) antigen-binding region of the multispecific antibody used in the present disclosure may comprise the VH CDR1 sequence selected from SEQ ID NOs: 135, 139, 143, 147, 151, and 159, the VH CDR2 sequence selected from SEQ ID NOs: 136, 140, 144, 148, 152, 156, and 160, and the VH CDR3 sequence selected from SEQ ID NOs: 137, 141 , 145, 149, 153, 157, and 161.
- the first antigen-binding region comprises an amino acid sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to a sequence selected from SEQ ID NO: 138, 142, 146, 150, 154, 158, and 162. In some embodiments, the first antigen-binding region comprises an amino acid sequence comprising or consisting a sequence selected from SEQ ID NO: 138, 142, 146, 150, 154, 158, and 162.
- the multispecific antibody used in the method of the present disclosure may compete (i.e. be able to compete) for binding to 5T4 with an antibody having a sequence selected from SEQ ID NO: 138, 142, 146, 150, 154, 158, and 162.
- the multispecific antibody may bind the same epitope on human 5T4 as an antibody having a sequence selected from SEQ ID NO: 138, 142, 146, 150, 154, 158, and 162.
- the multispecific antibody used in the methods of present disclosure may comprise a first antigen-binding region capable of binding 5T4 and be for use in the treatment bladder cancer, cervical cancer, non-small cell lung cancer, mesothelioma, head and neck squamous cell carcinoma, glioblastoma multiform, esophageal cancer, pancreatic cancer, breast cancer, including triple-negative breast cancer, colorectal cancer, gastric cancer, ovarian cancer, uterine cancer, prostate cancer, renal cancer, esophageal cancer or pre-B acute lymphoblastic leukemia.
- the human cancer antigen is EGFR.
- the (first) antigen-binding region of the multispecific antibody used in the present disclosure may comprise the VH CDR1 sequence of SEQ ID NO: 91 , the VH CDR2 sequence of SEQ ID NO: 92, and the VH CDR3 sequence of SEQ ID NO: 93.
- the first antigen-binding region comprises an amino acid sequence having at least 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% sequence identity to a sequence selected from SEQ ID NO: 94.
- the first antigen-binding region comprises an amino acid sequence comprising or consisting of SEQ ID NO: 94.
- the multispecific antibody used in the method of the present disclosure my i.e. be able to compete) for binding to EGFR with an antibody having the sequence set forth in SEQ ID NO: 94.
- the multispecific antibody may bind the same epitope on human EGFR as an antibody having the sequence set forth in SEQ ID NO: 94.
- the multispecific antibody used in the present disclosure may comprise a first antigenbinding region and a second antigen-binding region, wherein the first antigen-binding region comprises the CDR1 sequence set forth in SEQ ID NO: 91, the CDR2 sequence set forth in SEQ ID NO: 92 and the CDR3 sequence set forth in SEQ ID NO: 93 and wherein the second antigenbinding region comprises the CDR1 sequence set forth in SEQ ID NO: 5, the CDR2 sequence set forth in SEQ ID NO: 6 and the CDR3 sequence set forth in SEQ ID NO: 7..
- the second antigen-binding region comprises a CDR combination selected from Tabled 6A-6D.
- the multispecific antibody used in the methods of present disclosure may comprise a first antigen-binding region capable of binding EGFR and be for use in the treatment of primary or metastatic colon or colorectal cancer, peritoneal cancer, liver cancer, head and neck squamous cell carcinoma (HNSCC), non-small cell lung carcinoma (NSCLC), squamous cell carcinoma of the skin.
- HNSCC head and neck squamous cell carcinoma
- NSCLC non-small cell lung carcinoma
- the EGFR multispecific antibody may be able to activate human Vy9 ⁇ /52 T cells. Activation of Vy9V52 T cells may be measured through measuring alterations in gene-expression and/or (surface) marker expression (e.g., activation markers, such as CD25, CD69, or CD107a) and/or secretory protein (e.g., cytokines or chemokines) profiles.
- the EGFR multispecific antibody may be able to increase the number of cells positive for CD107a at least 2-fold, such as at least 5-fold, for example when tested as described in Example 9 in WO 2022/122973 (herein incorporated by reference), e.g.
- the EGFR multispecific antibody of the present disclosure may have an EC50 value for increasing the percentage of CD107a positive cells of 100 pM or less, such as 50 pM or less, e.g. 25 pM or less, such as 20 pM or less, e.g. 15 pM or less when tested for example using Vy9V52 T cells and A431 target cells as described in Example 9 of WO 2022/122973.
- the multispecific antibody used in the present disclosure may further comprise a half-life extension domain that extends the serum half-life of the multispecific antibody.
- a half-life extension domain that extends the serum half-life of the multispecific antibody.
- examples for means to extend serum half-life of the binding agents of the disclosure include peptides, proteins or domains of proteins, which are fused or otherwise attached to the multispecific antibodies.
- the group of peptides, proteins or protein domains includes peptides binding to other proteins with preferred pharmacokinetic profile in the human body such as serum albumin (see WO 2009/127691).
- serum albumin refers to the albumin protein present in human blood plasma. Human serum albumin is the most abundant protein in the blood. It constitutes about half of the blood serum protein.
- a human serum albumin has the sequence of UniProt ID NO: P02768.
- the multispecific antibody may have a terminal half-life that is longer than about 168 hours when administered to a human subject.
- the terminal half-life may be 336 hours or longer.
- the “terminal half-life” of an antibody when used herein refers to the time taken for the serum concentration of the polypeptide to be reduced by 50%, in vivo in the final phase of elimination.
- Half-life extension domains include larger domains of proteins or complete proteins includes e.g. the fusion of human serum albumin, variants or mutants of human serum albumin (see WO 2011/051489, WO 2012/059486, WO 2012/150319, WO 2013/135896, WO 2014/072481 , WO 2013/075066) or domains thereof as well as the fusion of constant region of immunoglobulins (Fc domains) and variants thereof, as described herein.
- Such variants of Fc domains may be optimized/modified in order to allow the desired pairing of dimers or multimers, to abolish Fc receptor binding (e.g., the Fcg receptor), to enhance binding to FcRn, or for other reasons.
- a further concept known in the art to extend the half-life of small protein compounds in the human body is the pegylation of those compounds such as the polypeptide or binding agent of the present disclosure.
- the half-life extension domain may be an Fc domain (also referred to as an “Fc region”).
- Fc domain describes the minimum region (in the context of a larger polypeptide) or smallest protein folded structure (in the context of an isolated protein) that can bind to or be bound by an Fc receptor (FcR).
- Fc domain monomer describes the single chain protein that, when associated with another Fc domain monomer, forms a functional Fc domain. The association of two Fc domain monomers creates one Fc domain. When two Fc domain monomers associate, the resulting Fc domain has Fc receptor binding activity. Thus, an Fc domain is a dimeric structure that can bind an Fc receptor.
- all references herein to a “variant Fc domain” are to be understood as referring to a dimeric Fc domain, in which each Fc domain monomer comprises the referenced mutation.
- Fc domain as used herein includes the polypeptides comprising the constant region of an antibody excluding the first constant region immunoglobulin domain.
- Fc domain refers to the last two constant region immunoglobulin domains (CH2, CH3) of IgG and optionally the flexible hinge N-terminal to these domains.
- all references to amino acid positions in Fc domains and Fc domain monomers are according to the EU index as set forth in Kabat (1991 , NIH Publication 91-3242, National Technical Information Service, Springfield, Va.).
- polymorphisms have been observed at a number of Fc domain positions, including but not limited to Kabat 270, 272, 312, 315, 356, and 358, and thus slight differences between the sequences provided herein and sequences in the art may exist.
- the Fc domain may be derived from any of a variety of different antibody isotypes, including but not limited to, a wild-type or modified lgG1 , lgG2, lgG3, lgG4. IgA, IgE, or IgM. In some embodiments, the Fc domain is derived from a human lgG1.
- the DEL polymorph comprises the amino acids D-E-L at positions 356, 357, and 358, respectively (e.g., SEQ ID NO: 337).
- the EEM polymorph comprises the amino acids E-E-M at positions 356, 357, and 358, respectively (e.g., SEQ ID NO: 336).
- Two binding agents that are otherwise identical except for the presence of a DEL Fc domain or an EEM Fc domain are expected to demonstrate similar properties in terms of ligand binding and therapeutic efficacy.
- the Fc domain is a DEL Fc domain.
- the Fc domain is an EEM Fc domain.
- the Fc domain is a variant Fc domain that forms a variant Fc domain with a desirable property, such as increased half-life, compared to naturally occurring (wild-type) Fc sequences.
- a “variant Fc domain” refers to a non-naturally occurring Fc domain, for example an Fc domain comprising one or more non-naturally occurring amino acid residues, one or more amino acid substitutions relative to a wild-type human constant domain, or one or more amino acid deletion, addition and/or modification.
- the serum half-life of binding agents comprising Fc domains may be increased by increasing the binding affinity of the Fc domain for FcRn.
- the Fc domain variant has enhanced serum half-life relative to a comparable molecule.
- the Fc domain variant comprises at least one amino acid substitution at one or more positions selected from the group consisting of M252Y, S254T and T256E (referred to herein as “YTE”; e.g., SEQ ID NO: 338 and 339).
- YTE amino acid substitution at one or more positions selected from the group consisting of M252Y, S254T and T256E
- the Fc domain variant comprises a Y at position 252 (referred to herein as “Y”, e.g., SEQ ID NO: 340 and 341).
- the Fc domain variant may comprise a non-naturally occurring amino acid residue at additional and/or alternative positions known to one skilled in the art (see, e.g., U.S. Pat. Nos. 5,624,821 ; 6,277,375; 6,737,056; PCT Patent Publications WO 01/58957; WO 02/06919; WO 04/016750; WO 04/029207; WO 04/035752; WO 04/074455; WO 04/099249; WO 04/063351 ; WO 05/070963; WO 05/040217, WO 05/092925, and WO 06/020114).
- the Fc domain monomer allows assembly of two or more polypeptide chains in a covalent manner, for example by disulfide linking between cysteine residues. In this way the Fc domain monomer acts as a dimerization domain, allowing assembly of two polypeptide chains to form a dimer.
- such dimers comprise two polypeptides, each polypeptide including an antigen-binding domain described herein linked to an Fc domain monomer, thereby forming a bivalent binding agent.
- such dimers comprise two polypeptides, one polypeptide comprising an antigen-binding domain described herein linked to an Fc domain monomer, and one polypeptide comprising an Fc domain monomer, thereby forming a monovalent binding agent.
- the Fc domain may be a heterodimer comprising two Fc monomers, wherein the first antigen-binding domain is fused to the first Fc monomer and the second antigen-binding region is fused to the second Fc monomer and wherein the first and second Fc monomers comprise asymmetric amino acid mutations that favor the formation of heterodimers over the formation of homodimers (see e.g., Ridgway et al. (1996) 'Knobs-into-holes' engineering of antibody CH3 domains for heavy chain heterodimerization. Protein Eng 9:617).
- the CH3 regions of the Fc monomers may comprise said asymmetric amino acid mutations, for example the first Fc polypeptide may comprise a T366W substitution (e.g., “knob” mutations), and the second Fc polypeptide may comprise T366S, L368A and Y407V substitutions (e.g., “hole” mutations), or vice versa, wherein the amino acid positions correspond to human lgG1 according to the EU numbering system. Furthermore, the cysteine residues at position 220 in the first and second Fc polypeptides may have been deleted or substituted, wherein the amino acid position corresponds to human lgG1 according to the EU numbering system. See e.g., SEQ ID NOs: 343 and 344.
- first and/or second Fc monomers may contain mutations that render the antibody inert, i.e. unable to, or having reduced ability to, mediate Fc effector functions.
- the inert Fc domain may in addition not be able to bind C1q.
- the first and second Fc monomers may comprise a mutation at position 234 and/or 235, for example the first and second Fc monomer may comprise an L234F and an L235E substitution, wherein the amino acid positions correspond to human lgG1 according to the EU numbering system. See e.g., SEQ ID NO: 342.
- Fc domain monomer sequences are shown in TABLE 9. Hinge sequences are shown in bold and italicized text; DEL and EEM polymorphisms are shown in boxed text; mutations relative to WT IgG 1 are shown in bold and underlined text. Table 9: Fc domain sequences*
- the Fc domains described in TABLE 9 comprise a wild type lgG1 hinge (e.g., SEQ ID NO: 347 - EPKSCDKTHTCPPCP). In some embodiments, the Fc domains described in TABLE 9 comprise a modified lgG1 hinge (e.g., SEQ ID NO: 348 - AAASDKTHTCPPCP).
- the present disclosure provides a kit for the treatment of cancer comprising: (a) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V62 T cell receptor, as described herein; and (b) a dose of a common gamma cytokine (e.g., IL-2, IL-15, or a variant thereof), optionally comprising instructions for use.
- a common gamma cytokine e.g., IL-2, IL-15, or a variant thereof
- the present disclosure provides a kit for the treatment of cancer comprising: (a) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor, as described herein; and (b) an immune checkpoint inhibitor, optionally comprising instructions for use.
- the immune checkpoint inhibitor is an anti- PD1 antibody (e.g., pembrolizumab).
- kits for the treatment of cancer comprising: (a) a multispecific antibody comprising a first antigen-binding region capable of binding a human cancer antigen and a second antigen-binding region capable of binding a human Vy9V ⁇ 52 T cell receptor, as described herein; (b) an immune checkpoint inhibitor (e.g., an anti- PD1 antibody such as pembrolizumab); and (c) a common gamma cytokine (e.g., IL-2 or IL-15), optionally comprising instructions for use.
- an immune checkpoint inhibitor e.g., an anti- PD1 antibody such as pembrolizumab
- a common gamma cytokine e.g., IL-2 or IL-15
- the kit may further comprise a solvent for the dilution of the multispecific antibody, the common gamma chain cytokine, and/or the immune checkpoint inhibitor.
- Example 1 Dose escalation of LAVA-1207 in metastatic castration resistant prostate cancer (mCRPC) patients
- LAVA-1207 is a bispecific humanized antibody of 78 kDa that engages Vy9V52-T cells and PSMA+ tumor cells.
- LAVA-1207 is a disulfide-linked heterodimer of two fusion proteins set forth in SEQ ID NO: 334 and SEQ ID NO:335, further described and characterized in WO 2022/008646 (incorporated herein by reference).
- This study was an open-label, multi-center, Phase 1/2a dose escalation study in patients with therapy-refractory, metastatic, castration-resistant prostate cancer (mCRPC).
- LAVA-1207 is administered via intravenous infusion every 2 weeks. Dose levels are as follows: 1.5 pg (starting dose) (Dose Level (DL) 1), 4.5 pg (DL2), 13.5 pg (DL3), 40 pg (DL4), 120 pg (DL5), 360 pg (DL6), 540 pg (DL7), and 800 pg (DL8)
- Anti-tumour activity was determined by evaluating prostate-specific antigen (PSA) changes in blood samples of patients upon treatment with LAVA-1207. Several patients showed stable or reduced PSA levels after LAVA-1207 treatment (Fig. 1). Furthermore, the baseline Vy9V62-T cell frequencies as percentage of total T cells were determined for each patient (not shown). At least in some instances, PSA decline was observed patients with high Vy9V52-T cells as a percentage of total T cells. See N304 PSA levels in Fig. 2 and N304 Vy9V52- T cell relative changes in Fig. 3A. Fig.
- FIG. 3A shows a pronounced drop in Vy9V52-T cell frequency in the peripheral blood 2 hr after dosing, likely reflective of Vy9V62-T cell migration/re-distribution with subsequent recovery.
- Fig. 3C and Fig. 3D also show Vy9V62-T cell activation markers (CD25 - Fig. 3C and CD69 - Fig. 3D) upregulated following dosing and that Receptor occupancy (RO - Fig. 3B) was detectable up to day 14 after Eol, with peak levels ranging from 6.1% to 12.6% in dose level 4 (40pg).
- Example 2 Evaluation of the safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity, and antitumor activity of a PSMA targeting bispecific y5-T cell engager, in patients with therapy refractory metastatic castration resistant prostate cancer
- This trial is an open-label, multi-center, Phase 1 and 2a dose escalation trial with an expansion cohort to investigate the safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity and preliminary antitumor activity of LAVA-1207 in patients with therapy refractory metastatic castration resistant prostate cancer (mCRPC).
- mCRPC therapy refractory metastatic castration resistant prostate cancer
- the trial starts with an open-label, dose-escalation part (Part 1) to determine the recommended Phase 2 dose (RP2D).
- Part 2 The second part of the trial (Part 2) is an open-label expansion cohort at the RP2D and schedule, in which the number of patients will be expanded to confirm safety in a patient population with therapy refractory mCRPC with measurable disease.
- LAVA-1207 will be administered with or without low dose subcutaneous IL-2 (referred to as LDSC IL-2), and optionally pembrolizumab may be administered.
- escalating dose levels are envisaged, including cohorts that include priming doses. Eligible patients will receive sequentially higher doses of LAVA-1207 in subsequent dose cohorts and will continue receiving LAVA-1207 until 24 weeks or up to disease progression, unacceptable toxicity or withdrawal of consent or otherwise as specified in the investigational medicinal product (IMP) discontinuation criteria.
- IMP investigational medicinal product
- Patients in a dose cohort will receive LAVA- 1207 as intravenous (IV) infusions at the same dose in a dosing interval of 2 weeks (equal to one cycle) consecutively without interruption, except when necessary to manage adverse events (AEs).
- IV intravenous
- the infusion duration for this biologic product will initially be 2 hours in the first cycle, 1 hour in the 2 nd cycle and 30 minutes in subsequent cycles.
- DLT period a dose-limiting-toxicity
- Dose escalation will continue until the RP2D can be defined by an optimal biological active dose of LAVA-1207 or until a maximum tolerated dose (MTD) has been determined, whichever is reached first.
- An optimal biological active dose is defined as a safe dose that demonstrates the greatest pharmacological activity of LAVA-1207. Considerations for pharmacological activity include the evaluation of effects on Vy9V52-T cells such as binding of LAVA-1207 to Vy9V52-T cells and changes in the frequency or activation status of Vy9V52-T cells as assessed in peripheral blood.
- IL-2 will be administered subcutaneously (SC) as a single dose (cohort A1) or as 3 doses (cohort A2) starting at 24 hours after the start of LAVA-1207 infusion (see Table 12).
- Table 12 Exploratory dose cohorts with LAVA-1207 and LDSC IL-2 a) LDSC IL-2 administration of 1 x 10 6 IU (1 MIU/day) - one day - at 24 hours after LAVA-1207 start of infusion (Sol). Note: the allowed time window is -1 hour before - and +3 hours at 24 hours after LAVA-1207 Sol. b) LDSC IL-2 administration of 1 x 10 e IU (1 MIU/day) - every day for 3 days - at 24 -, 48-, and 72 hours after LAVA-1207 start of infusion. Note: the allowed time window is -1 hour before and +3 hours after each specified timepoint.
- LDSC IL-2 will occur during the first 4 cycles of LAVA-1207 treatment only (e.g. up to 8 weeks).
- IL-2 is expected to increase the number of Vy9V52-T cells, thereby increasing the total number of Vy9V52 -T cells available for LAVA-1207 mediated lysis of PSMA expressing prostate cancer cells.
- cohorts will be initiated in with an anti-PD-L1 antibody at 400 mg intravenously over 30 minutes every 6 weeks [Q6W] in addition to the target dose of the multispecific antibody starting at Cycle 2 (second target dose). Treatment will be administered for a period not to exceed 2 years (18 cycles of the anti-PD-L1 antibody or until one or more discontinuation criteria have been met.
- the dose of pembrolizumab will remain constant at 400 mg Q6W for each dose level of the multispecific antibody.
- This trial will comprise additional cohorts to further investigate the safety, tolerability, immunogenicity, pharmacokinetics and pharmacodynamics of LAVA-1207 in combination with IL- 2 and to investigate the preliminary antitumor activity in patients with therapy refractory mCRPC in measurable disease.
- Primary objectives of the dose escalation cohort will include: (a) Investigate the safety and tolerability of LAVA-1207 in patients with therapy refractory mCRPC.
- Primary objectives of the expansion cohort will include:
- CTCs circulating tumor cells
- ctDNA circulating tumor DNA
- Primary endpoints for the dose escalation for LAVA-1207 alone, LAVA-1207 plus LDSC IL-2, and LAVA-1207 plus pembrolizumab will include:
- Primary endpoints for the expansion cohort for LAVA-1207 alone, LAVA-1207 plus LDSC IL-2, and LAVA-1207 plus pembrolizumab will include:
- Patient should have failed at least 1 line of taxane-based chemotherapy or is deemed medically unsuitable to be treated with a taxane regimen.
- Patient should have received a 2nd generation or later androgen receptor targeted therapy/ androgen biosynthesis inhibitor (e.g. abiraterone, enzalutamide, and/or apalutamide). Progression on novel antiandrogen therapy may have occurred in the non- metastatic CRPC setting.
- abiraterone, enzalutamide, and/or apalutamide e.g. abiraterone, enzalutamide, and/or apalutamide
- Bone scintigraphy appearance of 2 or more new metastatic lesions.
- Patient should have undergone bilateral orchiectomy or should be on continuous androgen-deprivation therapy (ADT) with a gonadotropin-releasing hormone agonist or antagonist (surgical or medical castration).
- ADT continuous androgen-deprivation therapy
- gonadotropin-releasing hormone agonist or antagonist surgical or medical castration
- hepatitis B surface antigen [HBsAg] and hepatitis B core antibody (anti-HBc) negative, and hepatitis C virus antibody Patients who are positive for anti-HBc or hepatitis C antibody may be included if they have a negative polymerase chain reaction (PCR) within 6 weeks prior to initial IMP administration. Those who are PCR positive will be excluded.
- PCR polymerase chain reaction
- Unstable cardiovascular function defined as: (a) symptomatic ischemia, or (b) uncontrolled clinically significant conduction abnormalities (i.e. ventricular tachycardia on antiarrhythmic agents are excluded; 1 st degree atrioventricular block or asymptomatic left anterior fascicular block/right bundle branch block is not excluded), or (c) congestive heart failure New York Heart Association Class > 3, or (d) myocardial infarction within 3 months.
- Previous treatment with antitumor therapies 2 weeks prior to initial IMP for radiotherapy and androgen receptor targeted therapy/ androgen biosynthesis inhibitor, and 4 weeks for systemic chemotherapy or targeted-/ immunotherapy.
- Treatment phase planned treatment duration of 24 weeks. At the request of the treating physician and in consultation with the sponsor, continued access to IMP, alone or in combination with pembrolizumab, might be offered beyond the planned treatment phase for individual patients with ongoing disease control.
- treatment duration will be for up to 18 cycles of pembrolizumab (approximately 24 months) or until any discontinuation criteria are met.
- Post-treatment phase A follow-up phase of 120 days after the last dose of IMP for each patient with an end of treatment (EoT) visit between 14 and 42 days after the last dose of IMP for each patient.
- EoT end of treatment
- LAVA-1207 is a concentrate for solution for infusion and will be administered as IV infusion with a 14-day dosing interval.
- the infusion duration will be 2 hours (+15 minutes) for any priming dose(s) and for the first target dose, followed by 1 hour for the second target dose and 30 minutes in subsequent cycles.
- a step-dosing regimen during Cycle 1 will start at Expanded Cohort 8 onwards.
- LDSC IL-2 will be administered as a single dose or as 3 daily doses starting at 24 hours (-1 hr- and +2 hrs) after the start of LAVA-1207 infusion for a total duration of 4 cycles.
- patients may continue treatment upon resolution of a (serious) (S)AE to a Grade 1 AE or be withdrawn from the trial.
- LAVA-1207 has safely reached a dose of 1200 pg in therapy refractory mCRPC patients.
- LAVA-1207 alone, LAVA-1207 + LDSC IL-2, and LAVA-1207 + pembrolizumab
- Blood samples will be collected to determine LAVA-1207 concentrations and evaluate the following pharmacodynamic markers: binding of LAVA- 1207 to Vy9V52-T cells, Vy9V52-T cells (activation status and frequency) and general immune cell analysis (activation status and frequency of B-cells, T-cell subsets (CD4+ , CD8+ and Treg), natural killer (NK) cells and monocytes) and induction of cytokines (e.g. interleukin [IL]-1P, IL-2, IL 6, IL-8, tumor necrosis factor [TNF]-a, interferon [IFN]-y and Granulocyte-macrophage colony-stimulating factor [GM- CSF]).
- cytokines e.g. interleukin [IL]-1P, IL-2, IL 6, IL-8, tumor necrosis factor [TNF]-a, interferon [IFN]-y and Granulocyte-macrophage colony-stimulating factor [GM-
- Imaging will be performed for:
- Pharmacokinetics Individual patient serum LAVA-1207 concentrations will be used to derive pharmacokinetic parameters using standard non-compartmental methods. [300] Pharmacodynamics: Pharmacodynamic markers and possible induction of cytokines will be analyzed descriptively.
- DCR complete response (CR)+partial response (PR)+ stable disease (SD)) for patients with measurable disease (in Part 1 and 2) at 8, 16 and 24 weeks.
- PSA reduction defined as any decline of PSA level compared to baseline
- PSA response defined as > 50% decline of PSA level compared to baseline
- Progression free survival will be descriptively analyzed with using a Kaplan-Meier approach. Progression free survival is defined as the time from start of treatment until progression (soft-tissue disease progression [by iRECIST], bone lesion progression [by PCWG3 criteria]), or death.
- the DEC consists of minimally the medical monitor, minimally one of the principal investigators and a representative of the sponsor. [310] The trial design outlined in Example 2 can be repeated, wherein IL-15 is substituted for IL-2.
- Fig. 6A-Fig. 6B shows increased PD-1 expression on Vy9V52-T cells after treatment with LAVA-1207.
- Fig. 7A shows percentages of Vy9V32 T cells in untreated non-malignant (n.m), malignant (m) areas of the prostate of patients with prostate cancer, and prostate cancer patient PBMC samples.
- Fig. 7B and Fig. 7C shows PD-1 expression on Vy9V62 T cells and CD3+ T cells from the same prostate cancer patient samples.
- Fig. 8 shows that Vy9V ⁇ 52 T cells activated in vitro with a V62-bsTCE (bispecific T cell engager) upregulate PD-1.
- V62-bsTCE bispecific T cell engager
- Example 4 Exemplary LAVA-1207 and IL-2 dosing regimen
- Fig. 9 and Fig. 10 show exemplary dosing regimen of LAVA-1207 and IL-2.
- pembrolizumab may also be included.
- the patient in cycle one, is administered a priming dose of LAVA-1207 at day -14 and at day -7. Then at day 0 the patient is administered a target dose of LAVA-1207. Then at days 1 , 2, and 3 the patient is administered a low dose of IL- 2. This cycle is repeated without the priming dose 3 more time every 2 weeks, and then the patient is administered only a target dose of LAVA-1207 for each subsequent cycle.
- a similar exemplary dosing regimen may be performed, using IL-15 instead of IL-2.
- Example 5 LAVA-1207 triggers activation of Vy9V52-T cells in patient-derived tumor tissue and PBMCs and induces selective prostate tumor cell lysis
- PBMC Peripheral blood mononuclear cells
- LymphoprepTM AXI-1114547, Fresenius density gradient centrifugation. Blood samples were obtained from Sanquin (Amsterdam, The Netherlands) in case of healthy donors or from prostate cancer patients under written informed consent from the Amsterdam UMC (location VUmc, Amsterdam, The Netherlands).
- PBMC were processed for phenotypic analysis using flow cytometry or resuspended in RPMI medium complete for functional experiments.
- Healthy donor-derived Vy9V62-T cells were isolated from PBMC and expanded.
- Vy9V62-T cells were isolated from PBMC using the EasySepTM human gamma/delta T cell isolation kit (19255, STEMCELL Technologies) and directly used in functional experiments (referred to as non-expanded Vy9V62-T cells).
- Non-malignant and malignant tissue samples were collected from radical prostatectomies of patients with non-metastatic prostate cancer. In combination with clinical and radiological findings provided by treating physicians, the pathologist assessed whether the tissue pieces were non-malignant or malignant by macroscopic analysis.
- Tissues were cut into small pieces, resuspended in dissociation medium composed of Iscove’s Modified Dulbecco’s Medium (IMDM, 12440053, Gibco) supplemented with 0.1% DNAse I (10104159001 , Roche), 0.14% Collagenase A (10103586001, Roche), 100 lU/mL PSG and 5% FCS, transferred to a sterile flask, and then incubated with a magnetic stirrer for 45 minutes at 37° C. Cell suspensions were subsequently strained with a 100 pM cell strainer (352360, BD Falcon).
- IMDM Modified Dulbecco’s Medium
- LNCaP PSMA+ clone FGC, CRL-1740
- PC3 PSMA-, CRL-1435
- ATCC American Type Culture Collection
- VCaP PSMA+, 06020201-1VL
- 22Rv1 PSMA+, 5092802
- RPMI Roswell Park Memorial Institute
- PD-1 expression did not differ across Vy9V52-T cells in PBMC, non-malignant and malignant prostate tissue. Comparatively, the percentage of all CD3+ cells that expressed CD69, 4-1 BB and PD-1 was significantly higher in non-malignant and malignant prostate tissue as compared to PBMC. See FIG. 12B.
- LAVA- 1207 (PSMA-V62-Fc) was incubated with malignant (PSMA expression MFI mean ⁇ SEM; 5.7 ⁇ 2.4) and non-malignant prostate tissue (PSMA expression MFI mean ⁇ SEM; 0.7 ⁇ 0.3), alone or co-cultured with autologous patient PBMCs for 24 hours.
- PSMA expression MFI mean ⁇ SEM; 5.7 ⁇ 2.4 malignant
- PSMA expression MFI mean ⁇ SEM 0.7 ⁇ 0.3
- 50 nM LAVA- 1207 increased the degranulation in both the malignant prostate tissue (Tl tissue-infiltrated) and PBMCs (FIG. 12C).
- Example 6 NKG2D and DNAM-1 receptor ligand interactions contribute to LAVA-1207- induced Vy9V52-T cell degranulation and tumor cell lysis
- ULBP-1 (NKG2D ligand) was expressed at similar levels between the malignant and non-malignant prostate tissue.
- the frequency of the NKG2D and NKG2A receptor expression on Vy9V52-T cells did vary significantly between malignant and non-malignant prostate tissue, and the percentage of Vy9V62-T cells positive for DNAM-1 receptor was lower in malignant prostate tissue than in non- malignant prostate tissue. See FIG. 13A-FIG. 13C.
- Expanded healthy donor-derived Vy9V52-T cells were cultured with prostate cell lines (LNCaP, VCaP, or 22Rv1) for 4 hours after a 30 minute pre-incubation with 10 pg/ml Fc receptor block (130-059-901 , Miltenyi Biotec) and a-DNAM-1 (clone DX11 , 559787, BD Bioscience), a- NKG2D (clone 149810, MAB139-100, R&D Systems) or a-NKG2A (Monalizumab, PX-TA1392- 100UG, Proteogenics) blocking antibodies were added 30 minutes before incubation with 0.001- 5 nM LAVA-1207.
- Vy9V52-T cell degranulation (as measured by CD107a expression), cytokine production (IFN-y, TNF, IL-2, and IL-4), and tumor cell lysis all increased with increasing concentration of LAVA-1207.
- cytokine production IFN-y, TNF, IL-2, and IL-4
- tumor cell lysis all increased with increasing concentration of LAVA-1207.
- neutralizing antibodies directed towards the DNAM- 1 or NKG2D receptors this increase could be partially mitigated except at the highest (saturating) concentrations of LAVA-1207.
- FIG. 14A- FIG. 14H Neutralizing antibodies to NKG2A and BTN3A did not have a significant impact on the LAVA-1207-mediated Vy9V52-T cell degranulation. See FIG. 15A-FIG. 15B.
- DNAM-1 and NKG2D receptor interactions can contribute to Vy9V52-T cell reactivity and prostate cancer lysis at non-saturating LAVA- 1207 concentrations.
- Example 7 LAVA-1207 triggers in vivo antitumor activity in a xenograft prostate cancer model using human PBMC
- TGI tumor growth inhibition
- LAVA-1207 was FITC-labeled by Squarix GmbH (Marl, Germany) using a controlled reaction with NHS-coupled FITC.
- the average number of fluorochrome molecules conjugated per protein (F/P) was 4.3.
- Tissue-reactivity was assessed by Charles River Laboratories (Evreux, France) by immunohistochemistry (IHC) using the FITC-labeled-LAVA-1207 in a panel of 41 different frozen normal human tissues (adrenal, bone marrow, breast/ mammary gland, cecum, cerebellum, cerebral cortex, colon, duodenum, endothelium [vessels], eye, esophagus, fallopian tube [oviduct], gall bladder, heart [ventricle], ileum, jejunum, kidney [cortex], liver, lung, lymph node, muscle [striated, skeletal], peripheral nerve, ovary, pancreas, parotid, parathyroid pituitary, placenta, prostate, rectum, skin, spinal cord, spleen, stomach, testis, thymus, thyroid, tonsil, ureter, urinary bladder, uterus [cervix] and uterus [endometrium]) and peripheral blood smears (three donors
- Frozen sections were air-dried, fixed in zinc formalin (4087236, Microm Microtech), and rinsed using Milipore water. Test samples were incubated with 0, 3 or 20 pg/ml FITC-labeled-LAVA-1207 followed by 2 pg/ml goat a-FITC antibody (01-40-01 , KPL) after which antibody block (760-4204, Discovery) was applied.
- the ChromoMab DAB kit (760-159, Discovery) was used for detection according to manufacturer recommendations. Test samples were then stained with hematoxylin II (790-2208, Ventana) and bluing reagent (760- 2037, Ventana), washed, dehydrated and mounted. Slides were evaluated by a pathologist using a light microscope (Olympus BX51).
Landscapes
- Health & Medical Sciences (AREA)
- Immunology (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Endocrinology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Mycology (AREA)
- Microbiology (AREA)
- Engineering & Computer Science (AREA)
- Animal Behavior & Ethology (AREA)
- Gynecology & Obstetrics (AREA)
- Pregnancy & Childbirth (AREA)
- Reproductive Health (AREA)
- Cell Biology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Description
Claims
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP24757537.6A EP4665766A2 (en) | 2023-02-13 | 2024-02-13 | Combination therapy comprising multispecific gamma delta tcr antibodies |
Applications Claiming Priority (12)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363445171P | 2023-02-13 | 2023-02-13 | |
| US63/445,171 | 2023-02-13 | ||
| US202363471647P | 2023-06-07 | 2023-06-07 | |
| US63/471,647 | 2023-06-07 | ||
| US202363543364P | 2023-10-10 | 2023-10-10 | |
| US63/543,364 | 2023-10-10 | ||
| US202363547960P | 2023-11-09 | 2023-11-09 | |
| US63/547,960 | 2023-11-09 | ||
| US202463618073P | 2024-01-05 | 2024-01-05 | |
| US63/618,073 | 2024-01-05 | ||
| US202463624652P | 2024-01-24 | 2024-01-24 | |
| US63/624,652 | 2024-01-24 |
Publications (2)
| Publication Number | Publication Date |
|---|---|
| WO2024173376A2 true WO2024173376A2 (en) | 2024-08-22 |
| WO2024173376A3 WO2024173376A3 (en) | 2024-10-17 |
Family
ID=92420644
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/015571 Ceased WO2024173376A2 (en) | 2023-02-13 | 2024-02-13 | Combination therapy comprising multispecific gamma delta tcr antibodies |
Country Status (2)
| Country | Link |
|---|---|
| EP (1) | EP4665766A2 (en) |
| WO (1) | WO2024173376A2 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2025012118A3 (en) * | 2023-07-07 | 2025-04-10 | LAVA Therapeutics N.V. | 5t4 binding agents and methods of use |
Family Cites Families (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EA039086B1 (en) * | 2014-04-10 | 2021-12-01 | Лава Терапьютикс Н.В. | Immunoglobulins binding human v9v2 t cell receptors |
| CN108699605A (en) * | 2015-06-03 | 2018-10-23 | 爱兰细胞技术公司 | The adjoint method and kit of therapy based on IL-2 and the therapy based on mescenchymal stem cell |
| CN116437954A (en) * | 2020-07-31 | 2023-07-14 | 埃克塞里艾克西斯公司 | Combinations Used to Treat Cancer |
-
2024
- 2024-02-13 WO PCT/US2024/015571 patent/WO2024173376A2/en not_active Ceased
- 2024-02-13 EP EP24757537.6A patent/EP4665766A2/en active Pending
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2025012118A3 (en) * | 2023-07-07 | 2025-04-10 | LAVA Therapeutics N.V. | 5t4 binding agents and methods of use |
Also Published As
| Publication number | Publication date |
|---|---|
| EP4665766A2 (en) | 2025-12-24 |
| WO2024173376A3 (en) | 2024-10-17 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP7379561B2 (en) | Chimeric antigens and T cell receptors and methods of use | |
| AU2018233976B2 (en) | Fc-optimized anti-CD25 for tumour specific cell depletion | |
| CN112513080B (en) | VISTA antigen binding molecules | |
| JP2023126946A (en) | Compositions comprising a combination of an anti-LAG-3 antibody, a PD-1 pathway inhibitor, and an immunotherapeutic agent | |
| CN105636986B (en) | Methods and compositions comprising combinations of VEGF antagonists and anti-CTLA-4 antibodies | |
| KR20200037388A (en) | Proteins that bind NKG2D, CD16, and EGFR, HLA-E, CCR4, or PD-L1 | |
| KR20190123299A (en) | Multispecific Binding Proteins Targeting CAIX, ANO1, Mesothelin, TROP2, CEA, or Cladin-18.2 | |
| JP2019515661A (en) | BCMA binding molecules and methods of use thereof | |
| JP2019509055A (en) | Multispecific immunomodulatory antigen-binding construct | |
| KR20200038530A (en) | Proteins that bind NKG2D, CD16 and tumor-associated antigens | |
| JP2018503399A (en) | Multispecific immunomodulatory antigen-binding construct | |
| KR20190120770A (en) | Proteins That Bind PSMA, NKG2D, and CD16 | |
| JP7458973B2 (en) | Combination of novel antibodies and Treg depleting antibodies with immunostimulatory antibodies | |
| KR20190120781A (en) | Proteins That Bind to CD123, NKG2D, and CD16 | |
| CN118791614A (en) | T cell activating antibodies | |
| JP2022553129A (en) | Antibodies against poliovirus receptor (PVR) and uses thereof | |
| EP4665766A2 (en) | Combination therapy comprising multispecific gamma delta tcr antibodies | |
| WO2022117569A1 (en) | A ccr8 antagonist antibody in combination with a lymphotoxin beta receptor agonist antibody in therapy against cancer | |
| US20240124607A1 (en) | Proteins binding nkg2d, cd16, and ceacam5 | |
| US20250161355A1 (en) | Proteins for modulating cell therapy products and methods of use thereof | |
| KR20250089513A (en) | Use of anti-CLDN4-anti-CD137 bispecific antibodies in combination with PD-1 signaling inhibitors in cancer therapy | |
| US20240409641A1 (en) | Dual acting cd1d immunoglobulin for treatment of myelodysplastic syndromes | |
| WO2025231372A2 (en) | Methods for treating multiple myeloma with car-t cells and bispecific antibodies | |
| WO2025064716A1 (en) | T-cell modulatory polypeptides and methods of use thereof | |
| CN116670169A (en) | Multispecific binding compounds that bind to PD-L1 |
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: 24757537 Country of ref document: EP Kind code of ref document: A2 |
|
| ENP | Entry into the national phase |
Ref document number: 2025546468 Country of ref document: JP Kind code of ref document: A |
|
| WWE | Wipo information: entry into national phase |
Ref document number: 2024757537 Country of ref document: EP |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24757537 Country of ref document: EP Kind code of ref document: A2 |
|
| ENP | Entry into the national phase |
Ref document number: 2024757537 Country of ref document: EP Effective date: 20250915 |