EP4240424A1 - Composition comprenant une combinaison d'un inhibiteur de point de contrôle immunitaire et d'un conjugué anticorps-amatoxine pour une utilisation dans le traitement du cancer - Google Patents

Composition comprenant une combinaison d'un inhibiteur de point de contrôle immunitaire et d'un conjugué anticorps-amatoxine pour une utilisation dans le traitement du cancer

Info

Publication number
EP4240424A1
EP4240424A1 EP21802741.5A EP21802741A EP4240424A1 EP 4240424 A1 EP4240424 A1 EP 4240424A1 EP 21802741 A EP21802741 A EP 21802741A EP 4240424 A1 EP4240424 A1 EP 4240424A1
Authority
EP
European Patent Office
Prior art keywords
antibody
composition
amatoxin
conjugate
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21802741.5A
Other languages
German (de)
English (en)
Inventor
Michael Kulke
Torsten HECHLER
Andreas Pahl
Christoph Müller
Susanne Werner-Simon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Heidelberg Pharma Research GmbH
Original Assignee
Heidelberg Pharma Research GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Heidelberg Pharma Research GmbH filed Critical Heidelberg Pharma Research GmbH
Publication of EP4240424A1 publication Critical patent/EP4240424A1/fr
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6817Toxins
    • A61K47/6831Fungal toxins, e.g. alpha sarcine, mitogillin, zinniol or restrictocin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6867Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from a cell of a blood cancer
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • Composition Comprising a Combination of Immune Checkpoint Inhibitor and Antibody- Amatoxin Conjugate for Use in Cancer Therapy
  • the present application relates to the field of cancer immunotherapy.
  • the present application relates to a composition comprising (a) at least one immune checkpoint inhibitor and (b) at least one conjugate, wherein said conjugate is comprising (i) a target binding moiety, (ii) at least one amatoxin, and (iii) optionally at least one linker connecting said target binding moiety with said at least one amatoxin.
  • the present application further relates to said composition for use in treating a patient having a cancer, and to a pharmaceutical formulation comprising said composition and additional excipients, as well as to methods of producing and using said composition.
  • Regulation of the immune system is mediated by a variety of mechanisms, including regulatory cells of the innate and adaptive immune system, like regulatory T cells (Treg), myeloid-derived suppressor cells (MDSCs) and M2-type macrophages; regulatory cytokines, such as IL- 10 and TGF-p; and immune checkpoints that control T cell activation.
  • regulatory cells of the innate and adaptive immune system like regulatory T cells (Treg), myeloid-derived suppressor cells (MDSCs) and M2-type macrophages; regulatory cytokines, such as IL- 10 and TGF-p; and immune checkpoints that control T cell activation.
  • Treg regulatory T cells
  • MDSCs myeloid-derived suppressor cells
  • M2-type macrophages regulatory cytokines, such as IL- 10 and TGF-p
  • immune checkpoints that control T cell activation.
  • Such cells and molecules can be exploited as a mechanism of immune subversion during development of cancer and chronic infectious diseases.
  • Immune checkpoints are receptors on the cell membrane of T lymphocytes which modulate the immune reactivity of said cells.
  • Tumor cells often activate anti-inflammatory immune checkpoint pathways via respective ligands that suppress anti-tumor immune responses, thus evading immuno- surveillance and progressing tumor growth.
  • ICIs Immune checkpoint inhibitors
  • monoclonal antibodies which bind to anti-inflammatory immune checkpoints or their ligands and can interrupt this tumor suppression strategy by reactivating the immune system and, hence, reestablishing its capacity to combat tumors.
  • ICIs have been shown to be clinically effective in a variety of tumor types (Dyck and Mills, 2017; Darwin et al, 2018).
  • CTLA-4 is predominantly expressed in T cells
  • PD-1 is expressed in activated T cells, B cells, and certain myeloid cells.
  • CTLA-4 works in the priming phase of T-cell activation and limits early T-cell activation
  • PD-1 works later, during the effector phase, mostly in peripheral tissues where T cells encounter PD-1 ligands (Dyck and Mills, 2017).
  • CTLA-4 one of the first inhibitory receptors identified as playing a role in suppression of T-cell responses, is structurally similar to CD28 and binds to CD80 and CD86 at a higher affinity than CD28. It has been suggested that CTLA-4 expression interferes with T-cell activation by reducing the CD28-mediated costimulatory (second) signal, leading to T cell anergy; anergic T cells have limited effector function (Dyck and Mills, 2017).
  • CTLA-4 expression and function is intrinsically linked with T-cell activation; CTLA-4 is immediately upregulated following T cell receptor (TCR) engagement (signal 1).
  • TCR T cell receptor
  • Blocking CTLA-4 in vivo has been shown to inhibit CTLA-4 binding to CD80/86 and to promote anti-tumoral immunity by inhibiting Treg cells and enhancing effector T-cell function (Wei et al, 2018).
  • the Programmed Cell Death Protein 1 (PD-1) molecule consists of an intracellular domain which has potential phosphorylation sites within an immune tyrosine-based inhibitory motif (ITIM) and an immune receptor inhibitory tyrosine-based switch motif (ITSM), a hydrophobic transmembrane region, and an extracellular IgV domain (Li et al, 2016).
  • ITIM immune tyrosine-based inhibitory motif
  • ITAM immune receptor inhibitory tyrosine-based switch motif
  • An activated switch motif (ITSM) is required for the inhibitory effect of PD-1 on active T cells; PD-1 ligand binding leads to recruitment of the tyrosine phosphatase SHP-2 to the ITSM motif and thus interferes with TCR downstream signalling.
  • PD-1 ligand binding leads to interference with signalling molecules such as PIP-3 kinase and Ras which are important for T-cell proliferation, cytokine secretion and metabolism, and furthermore induces metabolic alterations in T effector cells and promotes induction of Treg cells.
  • signalling molecules such as PIP-3 kinase and Ras which are important for T-cell proliferation, cytokine secretion and metabolism, and furthermore induces metabolic alterations in T effector cells and promotes induction of Treg cells.
  • PD-1 ligand binding can also lead to T-cell exhaustion.
  • PD-1 has been detected on T cells, Tregs, exhausted T cells, B cells, activated monocytes, dendritic cells (DCs), natural killer (NK) cells, natural killer T (NKT) cells, epithelial cells, and tumor cells (Li et al, 2016).
  • PD-1 expression on T cells is induced by antigen stimulation.
  • PD-1 mainly exerts its inhibitory effect on T cells in the periphery.
  • Two ligands of PD-1 have been identified, PD-L1 (CD274) and PD-L2 (CD273).
  • tumor cells and myeloid cells are thought to be the main cell types mediating T-cell suppression through PD-1 binding (Li et al, 2016; Dyck and Mills, 2017).
  • lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin and mucindomain containing-3 (TIM-3), T cell immunoglobulin and ITIM domain (TIGIT), V-domain Ig suppressor of T cell activation (VISTA), CD96, and BTLA (CD272) (Qin et al, 2019).
  • LAG-3 lymphocyte activation gene-3
  • TIM-3 T cell immunoglobulin and mucindomain containing-3
  • T cell immunoglobulin and ITIM domain T cell immunoglobulin and ITIM domain
  • VISTA V-domain Ig suppressor of T cell activation
  • CD96 CD96
  • BTLA CD272
  • LAG-3 is usually expressed on activated CD4-positive and CD8-positive T cells, Tregs, a subpopulation of natural killer (NK) cells, B cells, and plasmacytoid dendritic cells (pDCs).
  • NK natural killer
  • pDCs plasmacytoid dendritic cells
  • Thl T helper 1
  • MHC-II, galectin-3, LSECtin, a-synuclein, and fibrinogen-like protein 1 (FGL1) have been described to interact with LAG-3.
  • TIM-3 also called hepatitis A virus cellular receptor 2 (HAVCR2) belongs to the Ig super- family with an N-terminal Ig variable region (IgV)-like domain, a membrane-proximal mucin-like domain containing sites for O-linked glycosylation (glycosylated mucin domain), a single transmembrane region and a C -terminal cytoplasmic tail.
  • IgV Ig variable region
  • a single transmembrane region a single transmembrane region
  • C cytoplasmic tail a C cytoplasmic tail.
  • Expression of TIM-3 is not limited to T cells, it is known to be expressed on different types of immune cells, including B cells, Tregs, NK cells, DCs, monocytes, and macrophages.
  • TIM-3 Four distinct ligands have been reported to bind to the IgV domain of TIM-3, including galectin-9, high-mobility group protein Bl (HMGB1), carcinoembryonic antigen cell adhesion molecule 1 (Ceacam-1), and phosphatidyl serine (PtdSer), wherein galectin-9 and HMGBlare soluble ligands, while Ceacam-1 and PtdSer belong to surface ligands.
  • HMGB1 high-mobility group protein Bl
  • Ceacam-1 carcinoembryonic antigen cell adhesion molecule 1
  • PtdSer phosphatidyl serine
  • the TIGIT protein includes an extracellular IgV region, a transmembrane domain, and a cytoplasmic tail that harbors an ITIM and an immunoglobulin tail tyrosine (ITT)-like phosphorylation motif.
  • ITIM immunoglobulin tail tyrosine
  • TIGIT immunoglobulin tail tyrosine
  • the expression of TIGIT has been demonstrated to be tightly restricted to lymphocytes, mainly on T cell subsets (including Tregs and memory T cells) and NK cells.
  • TIGIT binds two ligands, namely CD155 (PVR or Necl-5) and CD112 (nectin-2, also known as PRR2 or PVRL2) with different affinity.
  • TIGIT exerts its immunosuppressive effects by competing for ligands with other counterparts like CD266 (DNAM-1).
  • CD226 delivers a positive co-stimulatory signal
  • TIGIT delivers inhibitory signals into the T cell (Qin et al, 2019).
  • Therapeutic monoclonal antibodies which have been approved by regulatory authorities as immune checkpoint inhibitors are including Ipilimumab directed against CTLA-4; Nivolumab and Pembrolizumab directed against PD-1; and Atezolizumab, Avelumab, Durvalumab and Cemiplimab directed against PD-L1 (Singh et al, 2020).
  • the tumor types for which immune checkpoint blockade therapies have been approved by regulatory authorities are including melanoma, squamous and non-squamous non-small cell lung cancer, metastatic small cell lung cancer, renal cell carcinoma, Hodgkin lymphoma, urothelial carcinoma, head and neck squamous cell carcinoma, Merkel cell carcinoma, hepatocellular carcinoma, gastric and gastroesophageal carcinoma, metastatic colorectal cancer, primary mediastinal B cell lymphoma, recurrent or metastatic cervical cancer, and metastatic cutaneous squamous cell carcinoma (Wei et al, 2018; Singh et al, 2020).
  • combination therapies comprising, e.g., Ipilimumab and Nivolumab have been used.
  • ICIs have been employed in combination with cancer vaccines typically consisting of a source of cancer antigens and adjuvants that activate innate immune cells like dendritic cells; cancer vaccines aim to generate tumor-specific T cells that kill tumor cells via secretion of IFN- ⁇ or lytic granules.
  • ICIs have also been used in combination with radiotherapy and in combination with histone deacetylase inhibitors, which can induce direct tumor cytotoxicity and improve tumor immunogenicity in some cancer types.
  • ICIs have been regarded as one of the most important developments in cancer therapy over the past decade.
  • a significant part of patients did not respond (Dyck and Mills, 2017).
  • irAEs immune-related adverse events
  • ICD immunogenic cell death
  • a cell death modality that does stimulate an immune response against dead-cell antigens, in particular deriving from cancer cells, as compared to non-immunogenic cancer cell death, for example by apoptosis.
  • ICD involves changes in the composition of the cell surface as well as the release of soluble mediators; these signals operate on a series of receptors expressed by dendritic cells, thus stimulating their presentation of tumor antigens to T cells (Kroemer et al, 2013). This cell death modality originally has been observed and studied in the context of anti-cancer chemotherapy.
  • ICD has been found to be characterized by a combination of alterations in the composition of the plasma membrane of dying cells and in the composition of the microenvironment (Fig. 3). These alterations result from premortem stress and subsequent cellular disintegration. ICD is obligatorily preceeded by two types of stress, which are endoplasmic reticulum (ER) stress and autophagy as an adaptive response to stress.
  • ER stress due to, e.g., chemotherapy, calreticulin (CRT), whose largest fraction is normally secluded in the ER lumen, heat shock proteins and other ER proteins are exposed at the outer surface of the plasma membrane.
  • ATP adenosine triphosphate
  • HMGB1 non-histone chromatin protein high-mobility group box 1
  • CRT, ATP and HMGB1 interact with CD91 (low density lipoprotein receptor-related protein 1, LRP1), P2RX7 (purinergic receptor) and TLR4 (Toll-like receptor 4) receptors, respectively, which are expressed by dendritic cells and promote engulfment of dying cells, production of cytokines like IL-1 ⁇ , and presentation of tumor antigens.
  • CD91 low density lipoprotein receptor-related protein 1, LRP1
  • P2RX7 purinergic receptor
  • TLR4 Toll-like receptor 4
  • the patient's dying cancer cells hence, operate as a vaccine that stimulates a tumor-specific immune response, characterized by DC recruitment, increased number and activity of T lymphocytes as well as increased ratio of cytotoxic CD8-positive T lymphocytes (CTLs) over regulatory T cells (Tregs) within the tumor, which in turn can control residual cancer cells (Kroemer et al, 2013).
  • Distinct chemotherapeutic agents are not equivalent in their capacity to induce ICD.
  • ADCs Antibody drug conjugates
  • amatoxins antibody-targeted amatoxin conjugates
  • tumor antigen-specific antibodies antibody fragments or derivatives or antibody-like proteins
  • amatoxin-based ADCs comprising tumor antigen-specific antibody as target binding moiety induces immuno- genic cell death.
  • the inventors have further observed a synergistic effect of an amatoxin-based ADCs and an immune checkpoint inhibitor with regard to their tumor-cell killing activities in vivo.
  • composition for use in the treatment of cancer or chronic infectious disease, said composition comprising
  • composition for use
  • composition for use
  • pharmaceutically acceptable buffers for use
  • surfactants for use
  • diluents carriers
  • excipients fillers
  • binders for use
  • lubricants for use
  • glidants for use
  • disintegrants for use
  • preservatives for use
  • Fig- 1 Structural formulae of various amatoxins.
  • the numbers in bold type (1 to 8) designate the standard numbering of the eight amino acids forming the amatoxin.
  • the standard designations of the atoms in amino acids 1, 3, and 4 are also shown (Greek letters a to y, Greek letters a to 8, and numbers from 1 ’ to 7’, respectively).
  • Fig. 2 Principal concept of immunogenic cell death.
  • cancer cells responding to ICD inducers expose CRT on the outer leaflet of their plasma membrane at a preapoptotic stage, and secrete ATP during apoptosis.
  • cells undergoing ICD release the nuclear protein HMGB1 as their membranes become permeabilized during secondary necrosis.
  • CRT, ATP, and HMGB1 bind to the receptors CD91, P2RX7, and TLR4, respectively.
  • ATP adenosine triphosphate
  • CRT calreticulin
  • CTL cytotoxic CD8+ T lymphocyte
  • DC dendritic cell
  • HMGB1 high-mobility group box 1
  • IFN interferon
  • IL interleukin
  • TLR Toll-like receptor
  • Fig. 3 Principal concept of immune checkpoint inhibition.
  • MHC presentation of peptide antigen to the T cell receptor provides a first signal for T cell activation (1).
  • CD80 on the antigen-presenting cell and CD28 on the T cell, the T cell receives a second, costimulatory activation signal (2).
  • CTLA-4 competes with CD28 for binding to CD80, and delivers an inhibitory signal to the T cell (3).
  • PD-1 receptor on the T cell binds to PD-L1, delivering an inhibitory signal to the T cell (4).
  • Tumor cells employ the use of these mechanisms in order to prevent T cells from clearing malignant cells.
  • T cells remain active after identifying tumor cells and can clear them from the host.
  • CD cluster of differentiation
  • CTLA-4 cytotoxic T lymphocyte associated antigen-4
  • MHC major histocompatibility complex
  • PD-1 programmed cell death protein- 1
  • PD-L1 programmed cell death protein- 1
  • TCR T-cell receptor
  • Fig. 4 Immunogenic cell death induced by antibody-targeted amatoxin conjugates (ATACs).
  • Immunogenic cell death (ICD) markers calreticulin (CRT), adenosin tri-phosphate (ATP), and high-mobility group box 1 protein (HMGB1) were determined.
  • Amanitin-conjugated ADCs induce the secretion of ICD markers in a target- dependent manner.
  • (A, B, C) Her2 -positive cell line BT474, and (D, E, F) CD79b-positive cell line BJAB were exposed to no compound (first bar), 100 nM Maytansine (second bar), 100 nM amanitin (third bar), 50 nM Anti-Her2-amanitin conjugate (fourth bar) and 50 nM Anti-CD79b-amanitin conjugate (fifth bar), respectively; and (A), (D) CRT-exposing (positive) cells; (B), (E) ATP secretion; and (C), (F) HMGB1 release were assessed.
  • Fig. 5 Synergistic action of Avelumab and Anti-CD19-Amatoxin conjugate in the presence of peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • Fig. 6 Synergistic action of Avelumab and Anti-CD 19- Amatoxin conjugate in the absence or presence of peripheral blood mononuclear cells (PBMCs).
  • PBMCs peripheral blood mononuclear cells
  • Fig- 7. Overview of cytotoxic potency of anti-HER2 ATACs in different cell lines.
  • Anti- HER2-LALA-D265C antibodies conjugated to linker-comprising amatoxins XIXa, XVIIIa, Xllla, Xlla, XXIIa, XXIa (the corresponding conjugated amatoxins coupled to the respective anti-HER2-LALA-D265C antibody are designated XlXb, XVIIIb, Xlllb, Xllb, XXIIb, XXIb wherein the anti-HER2 antibody corresponds to the antibody in said formulae.).
  • Fig 8. Efficacy of anti-Her2 ATACs in a s.c. JIMT-1 Xenograft model.
  • Jimt-1 xenograft model female NMRI nude mice were inoculated with 5x10 6 Jimt-1 breast cancer cells (Mol Cancer Ther. 2004 Dec;3(12): 1585-92) per mouse subcutaneously in the right flank.
  • At a mean tumor vol. of ⁇ 120 mm 3 animals were allocated to respective experimental groups on day 0.
  • the animals received a single intravenous dose of amanitin based anti-Her2 antibody drug conjugates (ADCs) as indicated. Tumor volume and body weight were determined twice per week.
  • ADCs amanitin based anti-Her2 antibody drug conjugates
  • Fig. 9 Synergistic action of pemprolizumab and anti-CD19-Amatoxin conjugate treatment in vivo in a Raji xenograft mouse model system.
  • Tumor volume was assessed at various time points after tumor cell inoculation. Error bars represent the SEM.
  • Fig. 10 Survival plot of mice depicting the synergistic effect of a combination treatment of anti-CTLA4 treatment and anti-CD19-amatoxin conjugate in a Raji xenograft mouse model.
  • Anti-CD19 ATAC was administered at a dose of 0.1 mg/kg or 0.3 mg/kg single dose on day 0, ipilimumab was administered at a dose of 4mg/kg on days 0, 3, 6, 8, and 10.
  • Combination treatment was done using anti-CD19 ATAC at a dose of 0.1 mg/kg, or 0.3 mg/kg (both single dose) on day 0, and ipilimumab at a dose of 4mg/kg on days 0, 3, 6, 8, 10.
  • Experimental details are provided in Example 6.
  • embodiments disclosed herein are not meant to be understood as individual embodiments which would not relate to one another.
  • Features discussed with one embodiment are meant to be disclosed also in connection with other embodiments shown herein. If, in one case, a specific feature is not disclosed with one embodiment, but with another, the skilled person would understand that does not necessarily mean that said feature is not meant to be disclosed with said other embodiment. The skilled person would understand that it is the gist of this application to disclose said feature also for the other embodiment, but that just for purposes of clarity and to keep the specification in a manageable volume this has not been done.
  • the content of the prior art documents referred to herein is incorporated by reference. This refers, particularly, for prior art documents that disclose standard or routine methods. In that case, the incorporation by reference has mainly the purpose to provide sufficient enabling disclosure and avoid lengthy repetitions.
  • a pharmaceutical composition comprising
  • composition for use in the treatment of cancer comprising
  • compositions for use in the treatment of a chronic infectious disease comprising
  • Immune checkpoints also called immune checkpoint receptors, control T-cell activation and thus prevent overshooting inflammation and autoimmune diseases, but also suppress antitumor immune responses.
  • immune checkpoint inhibitor or simply “checkpoint inhibitor” or “ICI” refers to any agent or compound that, either directly or indirectly, decreases the level of or inhibits the function of an immune checkpoint receptor protein or molecule found on the surface of an immune cell (for example, a T cell), or to any agent or compound that, either directly or indirectly, decreases the level of or inhibits the function of a ligand that binds to said immune checkpoint receptor protein or molecule, either as a soluble compound or on the surface of an immune cell-inhibitory cell.
  • Such an inhibitory cell can be, for example, a cancer cell, a regulatory T cell, a tolerogenic antigen presenting cell, a myeloid-derived suppressor cells, a tumor-associated macrophage, or a cancer- associated fibroblast.
  • Said ligand is typically capable of binding the immune checkpoint receptor protein or molecule on the immune cell.
  • a non-limiting example of an immune checkpoint receptor protein-ligand pair is PD-1, PD-L1.
  • PD-1 is an immune checkpoint receptor protein found on T-cells.
  • PD-L1 which can be over-expressed by cancer cells, binds to PD-1 and helps the cancer cells to evade the host immune system attack.
  • an immune checkpoint inhibitor prevents the PD- 1/PD-L1 interaction by either blocking the PD- 1 on the T cell (i.e.. acts as a PD-I inhibitor) or the PD-L1 on the cancer cell (i.e., acts as a PD-L1 inhibitor), thereby maintaining or restoring anti-tumor T-cell activity or blocking inhibitory cancer cell activity.
  • immune checkpoint inhibitors are antagonists of an immune inhibitory receptor, such PD-1, which inhibit, in this case, the PD-1 or PD-L1 in the PD-1/PD-L1 pathway.
  • PD-1 or PD-L1 inhibitors include, without limitation, humanized or human antibodies antagonizing or blocking human PD-1 function such as pembrolizumab, pidilizumab, cemiplimab, JTX-4014, spartalizumab, sintilimab (IBI308), dostarlimab (TSR-042, WBP- 285), INCMGA00012 (MGA012), AMP-224, PD1-1, PD1-2, PD1-3, PD1-4, PD1-5, BCD- 100, AGEN-2034, Toripalimab (TAB001, JS001), or AMP-514 (MEDI0680), as well as fully human antibodies such as the PD-1 blocking nivolumab or blocking PD-L1 such as
  • Pembrolizumab (formerly also known as lambrolizumab; trade name Keytruda; also known as MK-3475) disclosed e.g. in Hamid, O. et al. (2013) New England Journal of Medicine 369(2): 134-44, is a humanized IgG4 monoclonal antibody that binds to PD-1; it contains a mutation at C228P designed to prevent Fc-mediated cytotoxicity.
  • Pembrolizumab is e.g. disclosed in US 8,354,509 and W02009/114335. It is approved by the FDA for the treatment of patients suffering from unresectable or metastatic melanoma and patients with metastatic NSCLC.
  • Nivolumab (CAS Registry Number: 946414-94-4; BMS-936558 or MDX1106b) is a fully human IgG4 monoclonal antibody which specifically blocks PD-1, lacking detectable antibody-dependent cellular toxicity (ADCC).
  • ADCC antibody-dependent cellular toxicity
  • Nivolumab is e.g. disclosed in US 8,008,449 and W02006/121168. It has been approved by the FDA for the treatment of patients suffering from unresectable or metastatic melanoma, metastatic NSCLC and advanced renal cell carcinoma.
  • Pidilizumab (CT-011; Cure Tech) is a humanized IgGlk monoclonal antibody that binds to PD-1. Pidilizumab is e.g. disclosed in W02009/101611.
  • PD1-1 to PD1-5 refer to anti-PD-1 antibodies as disclosed in WO2018/220169.
  • Ipilumumab (CAS Registry Number: 477202-00-9, which may also be referred to as 10D1, or MDX010, MDX-101) is a human IgGl antibody that binds Cytotoxic T-lymphocyte antigen- 4 (CTLA4).
  • CTLA-4 is an inhibitory molecule that competes with the stimulatory CD28 for binding to B7 on antigen presenting cells.
  • CTLA-4 and CD28 are both presented on the surface of T-cells.
  • Ipilimumab is a human IgGl that binds CTLA-4, preventing the inhibition of T-cell mediated immune responses to tumors. Ipilimumab is e.g. disclosed in WO 01/14424 as antibody “10D1”.
  • Immune checkpoint receptors or molecules include, without limitation, e.g., PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, VISTA, 0X40, GITR, ICOS, CD276 (B7-H3), B7-H4 (VTCN1), IDO, KIR, CD122, CD137 , CD94/NKG2A, CD80, CD86, Galectin-3, LSECtin, CD112, Ceacam-1, Gal-9, PtdSer, HMGB1, HVEM, CD 155 and BTLA (CD272).
  • An immune checkpoint inhibitor according to the present invention may e.g. be a small molecule (organic) compound or a large molecule such as a peptide or a nucleic acid.
  • small molecule immune checkpoint inhibitors according to the invention include CA-170, including its precursor AUNP-12, as disclosed in W015033301 Al; or e.g. BMS-8 (CAS number 1675201-90-7).
  • an immune checkpoint inhibitor is an antibody, or an antigen binding fragement thereof, or an antigen binding derivative thereof.
  • the immune checkpoint inhibitor is a monoclonal antibody, or an antigen binding fragment thereof, or an antigen binding derivative thereof.
  • amatoxin includes all cyclic peptides composed of 8 amino acids as isolated from the genus Amanita and described in Wieland, T. and Faulstich H. (Wieland T, Faulstich H., CRC Crit Rev Biochem.
  • a “derivative” of a compound refers to a species having a chemical structure that is similar to the compound, yet containing at least one chemical group not present in the compound and/or deficient of at least one chemical group that is present in the compound.
  • the compound to which the derivative is compared is known as the “parent” compound.
  • a “derivative” may be produced from the parent compound in one or more chemical reaction steps.
  • an “analogue” of a compound is structurally related but not identical to the compound and exhibits at least one activity of the compound.
  • the compound to which the analogue is compared is known as the “parent” compound.
  • the afore-mentioned activities include, without limitation: binding activity to another compound; inhibitory activity, e.g. enzyme inhibitory activity; toxic effects; activating activity, e.g. enzyme-activating activity. It is not required that the analogue exhibits such an activity to the same extent as the parent compound.
  • a compound is regarded as an analogue within the context of the present application, if it exhibits the relevant activity to a degree of at least 1% (more preferably at least 5%, more preferably at least 10%, more preferably at least 20%, more preferably at least 30%, more preferably at least 40%, and more preferably at least 50%) of the activity of the parent compound.
  • an “analogue of an amatoxin”, as it is used herein, refers to a compound that is structurally related to any one of a-amanitin, ⁇ -amanitin, ⁇ - amanitin, ⁇ - amanitin, amanin, amaninamide, amanullin, and amanullinic acid and that exhibits at least 1% (more preferably at least 5%, more preferably at least 10%, more preferably at least 20%, more preferably at least 30%, more preferably at least 40%, and more preferably at least 50%) of the inhibitory activity against mammalian RNA polymerase II as compared to at least one of a-amanitin, ⁇ -amanitin, ⁇ -amanitin, £-amanitin, amanin, amaninamide, amanullin, and amanullinic acid.
  • an “analogue of an amatoxin” suitable for use in the present invention may even exhibit a greater inhibitory activity against mammalian RNA polymerase II than any one of a-amanitin, ⁇ -amanitin, ⁇ -amanitin, 8-amanitin, amanin, amaninamide, amanullin, or amanullinic acid.
  • the inhibitory activity might be measured by determining the concentration at which 50% inhibition occurs (IC50 value).
  • the inhibitory activity against mammalian RNA polymerase II can be determined indirectly by measuring the inhibitory activity on cell proliferation.
  • a “semisynthetic analogue” refers to an analogue that has been obtained by chemical synthesis using compounds from natural sources (e.g. plant materials, bacterial cultures, fungal cultures or cell cultures) as starting material.
  • natural sources e.g. plant materials, bacterial cultures, fungal cultures or cell cultures
  • a “semisynthetic analogue” of the present invention has been synthesized starting from a compound isolated from a mushroom of the Amanitaceae family.
  • a “synthetic analogue” refers to an analogue synthesized by so-called total synthesis from small (typically petrochemical) building blocks. Usually, this total synthesis is carried out without the aid of biological processes.
  • the amatoxin of said conjugate can be selected from the group consisting of ⁇ -amanitin, ⁇ -amanitin, ⁇ -amanitin, ⁇ -amanitin, amanin, amaninamide, amanullin, amanullinic acid, and analogues, derivatives and salts thereof.
  • amatoxins are defined as peptides or depsipeptides that inhibit mammalian RNA polymerase II.
  • Preferred amatoxins are those with a functional group (e.g. a carboxylic group, an amino group, a hydroxy group, a thiol or a thiol-capturing group) that can be reacted with linker molecules or target-binding moieties as defined below.
  • the term “amanitins” particularly refers to bicyclic structure that are based on an aspartic acid or asparagine residue in position 1, a proline residue, particularly a hydroxyproline residue in position 2, an isoleucine, hydroxyisoleucine or dihydroxyisoleucine in position 3, a tryptophan or hydroxytryptophan residue in position 4, glycine residues in positions 5 and 7, an isoleucine residue in position 6, and a cysteine residue in position 8, particularly a derivative of cysteine that is oxidized to a sulfoxide or sulfone derivative (for the numbering and representative examples of amanitins, see Figure 1), and furthermore includes all chemical derivatives thereof; further all semisynthetic analogues thereof; further all synthetic analogues thereof built from building blocks according to the master structure of the natural compounds (cyclic, 8 amino acids), further all synthetic or semisynthetic analogues containing non-hydroxylated amino acids instead
  • target-binding moiety refers to any molecule or part of a molecule that can specifically bind to a target molecule or target epitope.
  • Preferred target- binding moieties in the context of the present application are (i) antibodies or antigen-binding fragments thereof; (ii) antibody-like proteins; and (iii) nucleic acid aptamers.
  • “Target-binding moieties” suitable for use in the present invention typically have a molecular mass of 40 000 Da (40 kDa) or more.
  • a “linker” in the context of the present application refers to a molecule that increases the distance between two components, e.g.
  • the linker may serve another purpose as it may facilitate the release of the amatoxin specifically in the cell being targeted by the target binding moiety. It is preferred that the linker and preferably the bond between the linker and the amatoxin on one side and the bond between the linker and the target binding moiety or antibody on the other side is stable under the physiological conditions outside the cell, e.g. the blood, while it can be cleaved inside the cell, in particular inside the target cell, e.g. cancer cell. To provide this selective stability, the linker may comprise functionalities that are preferably pH-sensitive or protease sensitive.
  • the bond linking the linker to the target binding moiety may provide the selective stability.
  • a linker has a length of at least 1, preferably of 1-30 atoms length (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 atoms), wherein one side of the linker has been reacted with the amatoxin and, the other side with a target-binding moiety.
  • a linker preferably is a C 1-30 -alkyl, C 1-30 -heteroalkyl, C 2-30 - alkenyl, C 2-30 -heteroalkenyl, C 2-30 -alkynyl, C2-30-heteroalkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, aralkyl, or a heteroaralkyl group, optionally substituted.
  • the linker may contain one or more structural elements such as amide, ester, ether, thioether, disulfide, hydrocarbon moieties and the like. The linker may also contain combinations of two or more of these structural elements.
  • each one of these structural elements may be present in the linker more than once, e.g. twice, three times, four times, five times, or six times.
  • the linker may comprise a disulfide bond. It is understood that the linker has to be attached either in a single step or in two or more subsequent steps to the amatoxin and the target binding moiety. To that end the linker to be will carry two groups, preferably at a proximal and distal end, which can (i) form a covalent bond to a group, preferably an activated group on an amatoxin or the target binding-peptide or (ii) which is or can be activated to form a covalent bond with a group on an amatoxin.
  • linker if the linker is present, it is preferred that chemical groups are at the distal and proximal end of the linker, which are the result of such a coupling reaction, e.g. an ester, an ether, a urethane, a peptide bond etc.
  • the presence of a “linker” is optional, i.e. the toxin may be directly linked to a residue of the target-binding moiety in some embodiments of the target-binding moiety toxin conjugate.
  • said immune checkpoint inhibitor and/or the target binding moiety of said conjugate is selected from the group consisting of
  • an antibody preferably a monoclonal antibody
  • an antigen-binding fragment thereof preferably a variable domain (Fv), a Fab fragment or an F(ab)2 fragment,
  • Said antibody, or antigen-binding fragment thereof, or antigen-binding derivative thereof can be a murine, a chimeric, a humanized or a human antibody, or antigen-binding fragment, or antigen-binding derivative thereof, respectively.
  • antibody shall refer to a protein consisting of one or more polypeptide chains encoded by immunoglobulin genes or fragments of immunoglobulin genes or cDNAs derived from the same.
  • Said immunoglobulin genes include the light chain kappa, lambda and heavy chain alpha, delta, epsilon, gamma and mu constant region genes as well as any of the many different variable region genes.
  • the basic immunoglobulin (antibody) structural unit is usually a tetramer composed of two identical pairs of polypeptide chains, the light chains (L, having a molecular weight of about 25 kDa) and the heavy chains (H, having a molecular weight of about 50-70 kDa).
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated as VH or V H ) and a heavy chain constant region (abbreviated as CH or C H ).
  • the heavy chain constant region is comprised of three domains, namely CHI, CH2 and CH3.
  • Each light chain contains a light chain variable region (abbreviated as VL or V L ) and a light chain constant region (abbreviated as CL or C L ).
  • the VH and VL regions can be further subdivided into regions of hypervariability, which are also called complementarity determining regions (CDR) interspersed with regions that are more conserved called framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL region is composed of three CDRs and four FRs arranged from the amino terminus to the carboxy terminus in the order of FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains form a binding domain that interacts with an antigen.
  • the CDRs are most important for binding of the antibody or the antigen binding portion thereof.
  • the FRs can be replaced by other sequences, provided the three-dimensional structure which is required for binding of the antigen is retained. Structural changes of the construct most often lead to a loss of sufficient binding to the antigen.
  • antigen binding portion“ of the (monoclonal) antibody refers to one or more fragments of an antibody which retain the ability to specifically bind to the CD20 antigen in its native form.
  • antigen binding portions of the antibody include a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains, an F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfid bridge at the hinge region, an Fd fragment consisting of the VH and CHI domain, an Fv fragment consisting of the VL and VH domains of a single arm of an antibody, and a dAb fragment which consists of a VH domain and an isolated complementarity determining region (CDR).
  • CDR complementarity determining region
  • the antibody, or antibody fragment or antibody derivative thereof, according to the present invention can be a monoclonal antibody.
  • the antibody can be of the IgA, IgD, IgE, IgG or IgM isotype.
  • mAb monoclonal antibody
  • a monoclonal antibody displays a single binding specificity and affinity for a particular epitope.
  • human monoclonal antibody refers to an antibody displaying a single binding specificity which has variable and constant regions derived from or based on human germline immunoglobulin sequences or derived from completely synthetic sequences.
  • the method of preparing the monoclonal antibody is not relevant for the binding specificity.
  • such antibody is selected from the group consisting of IgG, IgD, IgE, IgA and/or IgM, or a fragment or derivative thereof, more preferably such antibody is an IgG type antibody or fragment or derivative thereof.
  • fragment shall refer to fragments of such antibody retaining target binding capacities, e.g., a CDR (complementarity determining region), a hypervariable region, a variable domain (Fv), an IgG heavy chain (consisting of VH, CHI, hinge, CH2 and CH3 regions), an IgG light chain (consisting of VL and CL regions), and/or a Fab and/or F(ab) 2 .
  • CDR complementarity determining region
  • Fv variable domain
  • IgG heavy chain consististing of VH, CHI, hinge, CH2 and CH3 regions
  • IgG light chain consististing of VL and CL regions
  • Fab and/or F(ab) 2 Fab and/or F(ab) 2 .
  • antigen-binding derivative or “derivative” shall refer to protein constructs being structurally different from, but still having some structural relationship to, the common antibody concept, e.g.
  • a protein composed of a peptide scaffold and at least one of the CDRs of the original antibody it is derived from.
  • Examples include e.g. scFv, Fab and/or F(ab)2, as well as bi-, tri- or higher specific antibody constructs. All these items are explained below.
  • antibody derivatives known to the skilled person are Diabodies, Camelid Antibodies, Domain Antibodies, bivalent homodimers with two chains consisting of scFvs, IgAs (two IgG structures joined by a J chain and a secretory component), shark antibodies, antibodies consisting of new world primate framework plus non-new world primate CDR, dimerised constructs comprising CH3+VL+VH, other scaffold protein formats comprising CDRs, and antibody conjugates (e.g., antibody, or fragments or derivatives thereof, linked to a drug, a toxin, a cytokine, an aptamer, a nucleic acid such as a desoxyribonucleic acid (DNA) or ribonucleic acid (RNA), a therapeutic polypeptide, a radioisotope or a label).
  • Said scaffold protein formats may comprise, for example, antibody-like proteins such as ankyrin and affilin proteins and others.
  • antibody-like protein refers to a protein that has been engineered (e.g. by mutagenesis of Ig loops) to specifically bind to a target molecule.
  • an antibody-like protein comprises at least one variable peptide loop attached at both ends to a protein scaffold. This double structural constraint greatly increases the binding affinity of the antibody-like protein to levels comparable to that of an antibody.
  • the length of the variable peptide loop typically consists of 10 to 20 amino acids.
  • the scaffold protein may be any protein having good solubility properties.
  • the scaffold protein is a small globular protein.
  • Antibody-like proteins include without limitation affibodies, anticalins, and designed ankyrin repeat proteins (Binz et al., 2005). Antibody-like proteins can be derived from large libraries of mutants, e.g. by panning from large phage display libraries, and can be isolated in analogy to regular antibodies. Also, antibody-like binding proteins can be obtained by combinatorial mutagenesis of surface-exposed residues in globular proteins.
  • Fab relates to an IgG fragment comprising the antigen binding region, said fragment being composed of one constant and one variable domain from each heavy and light chain of the antibody.
  • F(ab) 2 relates to an IgG fragment consisting of two Fab fragments connected to one another by disulfide bonds.
  • scFv relates to a single-chain variable fragment being a fusion of the variable regions of the heavy and light chains of immunoglobulins, linked together with a short linker, usually comprising serine (S) and/or glycine (G) residues.
  • S serine
  • G glycine
  • Modified antibody formats are for example bi- or trispecific antibody constructs, antibody- based fusion proteins, immunoconjugates and the like.
  • IgG, scFv, Fab and/or F(ab)2 are antibody formats which are well known to the skilled person. Related enabling techniques are available from respective textbooks.
  • said antibody, or antigen- binding fragment thereof or antigen-binding derivative thereof is a murine, a chimeric, a humanized or a human antibody, or antigen-binding fragment or antigen-binding derivative thereof, respectively.
  • Monoclonal antibodies (mAb) derived from mouse may cause unwanted immunological side- effects due to the fact that they contain a protein from another species which may elicit antibodies.
  • antibody humanization and maturation methods have been designed to generate antibody molecules with minimal immunogenicity when applied to humans, while ideally still retaining specificity and affinity of the non- human parental antibody (for review see Almagro and Fransson 2008).
  • CDR grafting the framework regions of a mouse mAb are replaced by corresponding human framework regions.
  • W0200907861 discloses the generation of humanized forms of mouse antibodies by linking the CDR regions of non-human antibodies to human constant regions by recombinant DNA technology.
  • humanized antibody relates to an antibody, a fragement or a derivative thereof, in which at least a portion of the constant regions and/or the framework regions, and optionally a portion of CDR regions, of the antibody is derived from or adjusted to human immunoglobulin sequences.
  • the antibodies, the antibody fragments or antibody derivatives thereof, disclosed herein can comprise humanized sequences, in particular of the preferred VH- and VL-based antigen- binding region which maintain appropriate ligand affinity.
  • the amino acid sequence modifications to obtain said humanized sequences may occur in the CDR regions and/or in the framework regions of the original antibody and/or in antibody constant region sequences.
  • Said antibody, or antibody fragment or antibody derivative thereof can be glycosylated.
  • the glycan can be an N-linked oligosaccharide chain at asparagin 297 of the heavy chain.
  • the antibodies or fragments or derivatives of the present invention may be produced by transfection of a host cell with an expression vector comprising the coding sequence for the antibody according to the invention.
  • the expression vector or recombinant plasmid is produced by placing the coding antibody sequences under control of suitable regulatory genetic elements, including promoter and enhancer sequences like, e.g., a CMV promoter.
  • Heavy and light chain sequences might be expressed from individual expression vectors which are co-transfected, or from dual expression vectors.
  • Said transfection may be a transient transfection or a stabile transfection.
  • the transfected cells are subsequently cultivated to produce the transfected antibody construct.
  • stabile transfection is performed, then stable clones secreting antibodies with properly associated heavy and light chains are selected by screening with an appropriate assay, such as, e.g., ELISA, subcloned, and propagated for future production.
  • said immune checkpoint inhibitor binds to an immune checkpoint receptor selected from the group consisting of PD-1, CTLA-4, LAG-3, TIGIT, TIM-3, VISTA, BTLA (CD272), 0X40 (CD 134), B7-H4 (VTCN1), CD96, CD278 (ICOS), CD94/NKG2A and CD 160, or to a ligand of an immune checkpoint receptor selected from the group consisting of PD-L1, PD-L2, CD80, CD86, Galectin-3, LSECtin, CD112, Ceacam-1, Gal-9, PtdSer, HMGB1, HVEM, CD 155, OX40L, CD275 (ICOSLG),.
  • an immune checkpoint receptor selected from the group consisting of PD-1, CTLA-4, LAG-3, TIGIT, TIM-3, VISTA, BTLA (CD272), 0X40 (CD 134), B7-H4 (VTCN1), CD96, CD278 (
  • the composition according to the present invention comprises an immune checkpoint inhibitor, wherein said immune checkpoint inhibitor is an antibody selected from the group consisting of nivolumab, pembrolizumab, pidilizumab, cemiplimab, JTX-4014, spartalizumab, sintilimab (IBI308), dostarlimab (TSR-042, WBP-285), INCMGA00012 (MGA012), AMP-224, PD1-1, PD1-2, PD1-3, PD1-4, PD1-5, BCD- 100, AGEN-2034, Toripalimab (TAB001, JS001), or AMP-514 MEDI0680 nivolumab, avelumab, durvalumab, cosibelimab (CK-301), WBP-3155 (CS1001), atezolizumab or CX-072, or an antigen-binding fragment thereof, or an antigen-binding derivative thereof
  • the antibody is one of avelumab, nivolumab, ipilimumab, pembrolizumab, or an antigen-binding fragment thereof, or an antigen-binding derivative thereof.
  • the composition according to the present invention as disclosed herein comprises a combination of two or more immune checkpoint inhibitors, e.g. two, three, four, five, six immune checkpoint inhibitors as disclosed herein, preferably the composition comprises a combination of two immune checkpoint inhibitors.
  • the composition according to the invention as disclosed herein comprises two or more immune checkpoint inhibitors that target different immune checkpoints, e.g. CTLA-4 and PD-1/PD-L1, PD-1/PD-L1 and TIGIT, PD-1/ PD-L1 and 0X40, PD-1/PD-L1 and VISTA, CTLA4 and TIGIT, CTLA4 and 0X40.
  • compositions according to the invention may e.g. comprise one of the following combinations of immune checkpoint inhibtors:
  • CTLA4 - PD-1/PD-L1
  • BMS986178 in combination with one of nivolumab, avelumab, pembrolizumab, pidilizumab, PD1-1, PD1-2, PD1-3, PD1-4, PD1-5, durvalumab, atezolizumab
  • CI-8993 in combination with one of nivolumab, avelumab, pembrolizumab, pidilizumab, PD1-1, PD1-2, PD1-3, PD1-4, PD1-5, durvalumab, atezolizumab
  • MEDI0562 in combination with one of nivolumab, avelumab, pembrolizumab, pidilizumab, PD1-1, PD1-2, PD1-3, PD1-4, PD1-5, durvalumab, atezolizumab, or PF04518600 in combination with one of nivolumab, avelumab, pembrolizumab, pidilizumab, PD1-1, PD1-2, PD1-3, PD1-4, PD1-5, durvalumab, atezolizumab
  • MBG453 in combination with one of of of nivolumab, avelumab, pembrolizumab, pidilizumab, PD1-1, PD1-2, PD1-3, PD1-4, PD1-5, durvalumab, atezolizumab
  • Ipilimumab in combination with one of MED 10562, or PF04518600.
  • the composition according to the present invention comprises a target binding moiety, wherein said target binding moiety of said conjugate binds to a target molecule on the cell surface of cancer cells wherein the target molecule is one of PSMA, CD 19, CD37, CD269, sialyl Lewis a , HER-2/neu, epithelial cell adhesion molecule (EpCAM).
  • target molecule is one of PSMA, CD 19, CD37, CD269, sialyl Lewis a , HER-2/neu, epithelial cell adhesion molecule (EpCAM).
  • PSMA prostate specific membrane antigen
  • GCPII glutamate carboxypeptidase II
  • NAALADase I N-acetyl-L-aspartyl-L-glutamate peptidase I
  • NAAG N-acetyl-aspartylglutamate
  • PSMA is a membrane-bound cell surface peptidase that plays different physiological roles and is expressed in various tissues such as the prostate, kidney, small intestine, central and peripheral nervous system. It is highly expressed by malignant prostate epithelial cells and vascular endothelial cells of numerous solid tumor malignancies, including glioblastomas, breast and bladder cancers.
  • CD 19“ as disclosed above refers to B-lymphocyte antigen CD 19, also known as CD 19 molecule (Cluster of Differentiation 19), B-Lymphocyte Surface Antigen B4, T-Cell Surface Antigen Leu- 12 and CVID3 which is a transmembrane protein that in humans is encoded by the gene CD 19.
  • CD 19 is a biomarker for B lymphocyte development, lymphoma diagnosis and can be utilized as a target for leukemia immunotherapies.
  • CD37 refers to a protein that in humans is encoded by the CD37 gene and is a member of the “tetraspanin” superfamily or transmembrane 4 superfamily. Tetraspanins are characterized by the presence of four conserved transmembrane domains that are considered as “molecular facilitators” of signaling transduction, involved in a wide range of biological processes including cell growth, survival, adhesion, cell-cell communication, and trafficking, intercellular communication via exosomes, tumorigenesis, metastasis, and regulation of immune responses.
  • Tetraspanin members have also been described to have functional roles in a wide array of cellular processes, including cell motility, development and differentiation, activation, proliferation, migration and tumor invasion (Hemler 2001; Xu-Monette et al. 2016). Increased CD37 expression was found in B cell malignancies (Zou et al. 2018). Most B-cell malignancies express CD37, including B-cell non-Hodgkin lymphoma (NHL) and B-cell chronic lymphocytic leukemia (B-CLL).
  • NHL B-cell non-Hodgkin lymphoma
  • B-CLL B-cell chronic lymphocytic leukemia
  • Target molecule CD269 refers to B-cell maturation antigen (BCMA or BCM), also known as tumor necrosis factor receptor superfamily member 17 (TNFRSF17), which is a protein that in humans is encoded by the TNFRSF17 gene. CD269 is implicated in leukemia, lymphomas, and multiple myeloma.
  • BCMA or BCM B-cell maturation antigen
  • TNFRSF17 tumor necrosis factor receptor superfamily member 17
  • Sialyl Lewis a (also referred to as CD 15) as disclosed above refers to a tetrasaccharide composed of a sialic acid, fucose and an N-acetyllactosamine. Sialyl Lewis a mediates phagocytosis and chemotaxis, found on neutrophils; expressed in patients with Hodgkin disease, as well as in some B-cell chronic lymphocytic leukemias, acute lymphoblastic leukemias, and most acute nonlymphocytic leukemias. Sialyl Lewis a is present on almost all Reed-Sternberg cells, and can be used in immunohistochemistry to identify the presence of such cells in biopsies which is diagnostic of Hodgkin's lymphoma.
  • HER-2/neu refers to the receptor tyrosine-protein kinase erbB-2, also known as CD340 (cluster of differentiation 340), proto-oncogene Neu, Erbb2 (rodent), or ERBB2 (human), is a protein that in humans is encoded by the ERBB2 gene.
  • ERBB is abbreviated from erythroblastic oncogene B, a gene isolated from avian genome. It is also frequently called HER2 (from human epidermal growth factor receptor 2) or HER2/neu.
  • HER2 is a member of the human epidermal growth factor receptor (HER/EGFR/ERBB) family.
  • HER2 HER2 dimerization of the receptor results in the autophosphorylation of tyrosine residues within the cytoplasmic domain of the receptors and initiates a variety of signaling pathways leading to cell proliferation and tumorigenesis.
  • Amplification or overexpression of HER2 occurs in approximately 15-30% of breast cancers and 10-30% of gastric/gastroesophageal cancers and serves as a prognostic and predictive biomarker.
  • EpCAM as disclosed above refers to “epithelial cell adhesion molecule“ which is a transmembrane glycoprotein mediating Ca2+-independent homotypic cell-cell adhesion in epithelia.
  • EpCAM is a glycosylated type I membrane protein having a molecular weight of 30-40kD and in humans is endcoded by the EPCAM gene. The sequence of the EpCAM molecule predicts the presence of three potential N-linked glycosylation sites. It is composed of 314 amino acids.
  • EpCAM consists of an extracellular domain (242 amino acids) with epidermal growth factor (EGF)- and thyroglobulin repeat-like domains, a single transmembrane domain (23 amino acids), and a short intracellular domain (26 amino acids).
  • EpCAM is involved in cell signaling, migration, proliferation, and differentiation. In additon, EpCAM has oncogenic potential via its capacity to upregulate c-myc, e-fabp, and cyclins A & E. EpCAM may also be referred to as Ep-CAM, 17-1A, HEA125, MK-1, GA733-2, EGP- 2, EGP34, KSA, TROP-1, ESA, or KS1/4. EpCAM is expressed exclusively in epithelia and epithelial-derived neoplasms and can be used as diagnostic marker for various cancers.
  • target molecule and “target epitope”, respectively, refers to an antigen and an epitope of an antigen, respectively, that is specifically bound by a target-binding moiety.
  • target molecule is a tumour- associated antigen, in particular an antigen or an epitope which is present on the surface of one or more tumour cell types in an increased concentration and/or in a different steric configuration as compared to the surface of non-tumour cells.
  • said antigen or epitope is present on the surface of one or more tumour cell types, but not on the surface of non-tumour cells.
  • the target-binding moiety specifically binds to an epitope of an antigen selected from: PSMA, CD19, CD37, CD269, sialyl Lewis a , HER- 2/neu , epithelial cell adhesion molecule (EpCAM).
  • said antigen or epitope is preferentially expressed on cells involved in autoimmune diseases.
  • the target-binding moiety specifically binds to an epitope of the IL-6 receptor (IL-6R).
  • IL-6R IL-6 receptor
  • the composition according to the present invention as disclosed herein comprises a conjugate comprising a target binding moiety, wherein said target binding moiety of said conjugate is an antibody having an Fc region comprising at least one mutation selected from the group consisting of D265C, D265A, A118C, L234A, or L235A (according to the EU numbering system).
  • EU index as in Kabat or “EU numbering system” refers to the numbering of the human IgGl EU antibody (see e.g. Edelman et al., 1969, Proc Natl Acad Sci USA 63:78-85, hereby entirely incorporated by reference).
  • the antibody representing the target-binding moiety of a conjugate according to the present invention as disclosed herein comprises an Fc region carrying a D265C mutation and said linker, if present, or said amatoxin is connected to said antibody via one or two of the D265C residues of said antibody, preferably via a disulfide linkage.
  • said antibody representing the target-binding moiety of said conjugate has been genetically engineered to comprise a heavy chain 118Cys, a heavy chain 239Cys, or heavy chain 265Cys according to the EU numbering system, preferably a heavy chain 265Cys according to the EU numbering system, and wherein said linker, if present, or said amatoxin is connected to said antibody via said heavy chain 118Cys, or said heavy chain 239Cys, or heavy chain 265Cys residue, respectively.
  • the antibody representing the target-binding moiety of said conjugate according to the invention as disclosd herein comprises an Fc region which comprises a L234 mutation, a L235 mutation and a D265 mutation.
  • the antibody representing the target-binding moiety of said conjugate according to the invention as disclosd herein comprises an Fc region comprising a L234A, L235A and a D265C mutation (according to EU numbering system).
  • the term “genetically engineered” or “genetic engineering” relates to the modification of the amino acid sequence or part thereof of a given or natural polypeptide or protein in the sense of nucleotide and/or amino acid substitution, insertion, deletion or reversion, or any combinations thereof, by gene technological methods such as e.g. site- directed mutagenesis as described in Biochem. J. (1986) 237, 1-7, or J Biol Chem. 2015 Jan 30; 290(5): 2577-2592.
  • amino acid substitution relates to modifications of the amino acid sequence of the protein, wherein one or more amino acids are replaced with the same number of different amino acids, producing a protein which contains a different amino acid sequence than the original protein.
  • a conservative amino acid substitution is understood to relate to a substitution which due to similar size, charge, polarity and/or conformation does not significantly affect the structure and function of the protein.
  • Groups of conservative amino acids in that sense represent, e.g., the non-polar amino acids Gly, Ala, Vai, I1e and Leu; the aromatic amino acids Phe, Trp and Tyr; the positively charged amino acids Lys, Arg and His; and the negatively charged amino acids Asp and Glu.
  • Exemplary amino acid substitutions are presented in Table 1 below:
  • said linker, if present, or said amatoxin is connected to said antibody via any of the natural Cys residues of said antibody, preferably via a disulfide linkage.
  • the conjugate according to the present invention can have a cytotoxic activity of an IC50 better than 10x10 -9 M, 9x10 -9 M, 8x10 -9 M, 7x10 -9 M, 6x10 -9 M, 5x10 -9 M, 4x10 -9 M, 3x10 -9 M, 2x10 -9 M, preferably better than 10x10 -10 M, 9x10 -10 M, 8x10 -10 M, 7x10 -10 M, 6x10 -10 M, 5x10 -10 M, 4x10 -10 M, 3x10 -10 M, 2x10 -10 M, and more preferably better than 10x10 -11 M, 9x10 -11 M, 8x10 -11 M, 7x10 -11 M, 6x10 -11 M, 5x10 -11 M,
  • the linker of said conjugate, if present, or said target binding moiety is connected to said amatoxin via (i) the ⁇ C-atom of amatoxin amino acid 1, or (ii) the 5 C-atom of amatoxin amino acid 3, or (iii) the 6’-C-atom of amatoxin amino acid 4.
  • said conjugate in the composition as described comprises a linker.
  • Said linker can be a stable (non-cleavable) or a cleavable linker.
  • the cleavable linker can be selected from the group consisting of an enzymatically cleavable linker, preferably a protease-cleavable linker, and a chemically cleavable linker, preferably a linker comprising a disulfide bridge.
  • a “cleavable linker” is understood as comprising at least one cleavage site.
  • cleavage site shall refer to a moiety that is susceptible to specific cleavage at a defined position under particular conditions. Said conditions are, e.g., specific enzymes or a reductive environment in specific body or cell compartments.
  • non-cleavable linker is understood not to be subject to enzymatical cleavage by e.g. cathepsin B and is released from the conjugates of the invention during degradation (e.g., lysosomal degradation) of the antibody moiety of the conjugate of the invention inside the target cell.
  • the cleavage site is an enzymatically cleavable moiety comprising two or more amino acids.
  • said enzymatically cleavable moiety comprises a valine-alanine (Vai- Ala), valine-citrulline (Val- Cit), valine-lysine (Val-Lys), valine-arginine (Val-Arg) dipeptide, a phenylalanine-lysine- glycine-proline-leucin-glycine (Phe Lys Gly Pro Leu Gly) or alanine-alanine-proline-valine (Ala Ala Pro Vai) peptide, or a ⁇ -glucuronide or ⁇ -galactoside.
  • the enzymatically cleavable moiety may also be referred to as linker.
  • the enzymatically cleavable moiety according to the invention comprises a dipeptide selected from Phe-Lys, Val-Lys, Phe-Ala, Vai-Ala, Phe-Cit and Val-Cit, particularly wherein the cleavable linker further comprises a p-aminobenzyl (PAB) spacer between the dipeptides and the amatoxin:
  • the conjugates of the invention as disclosed herein can comprise an enzymatically cleavable moiety which comprises any one of the dipeptides-PAB moieties Phe-Lys-PAB, Val-LysPAB, Phe-Ala-PAB, Val-Ala-PAB, Phe-Cit-PAB, or Val-Cit-PAB as disclosed above.
  • the cleavable moiety of the conjugates of the invention comprises the dipeptide-PAB moiety Val-Ala-PAB.
  • the cleavable moieties or linkers of the invention as disclosed above comprise a thiol reactive group, selected from bromo acetamide, iodo acetamide, methylsulfonylbenzothiazole, 4,6-dichloro-l,3,5-triazin-2-ylamino group methyl- sulfonyl phenyltetrazole or methylsulfonyl phenyloxadiazole, pyridine-2-thiol, 5- nitropyridine-2-thiol, methanethiosulfonate, or a maleimide.
  • a thiol reactive group selected from bromo acetamide, iodo acetamide, methylsulfonylbenzothiazole, 4,6-dichloro-l,3,5-triazin-2-ylamino group methyl- sulfonyl phenyltetrazole or methylsulfonyl
  • the thiol reactive group is a maleimide (maleimidyl moiety) as depicted below:
  • the linker of the invention comprises the structure (i) prior to coupling, or (ii) following the coupling to an antibody as disclosed herein.
  • said cleavage site can be cleavable by at least one protease selected from the group consisting of cysteine protease, metalloprotease, serine protease, threonine protease, and aspartic protease.
  • Cysteine proteases also known as thiol proteases, are proteases that share a common catalytic mechanism that involves a nucleophilic cysteine thiol in a catalytic triad or dyad.
  • Metalloproteases are proteases whose catalytic mechanism involves a metal. Most metalloproteases require zinc, but some use cobalt.
  • the metal ion is coordinated to the protein via three ligands.
  • the ligands co-ordinating the metal ion can vary with histidine, glutamate, aspartate, lysine, and arginine.
  • the fourth coordination position is taken up by a labile water molecule.
  • Serine proteases are enzymes that cleave peptide bonds in proteins; serine serves as the nucleophilic amino acid at the enzyme's active site. Serine proteases fall into two broad categories based on their structure: chymotrypsin-like (trypsin-like) or subtilisin-like.
  • Threonine proteases are a family of proteolytic enzymes harbouring a threonine (Thr) residue within the active site.
  • the prototype members of this class of enzymes are the catalytic subunits of the proteasome, however, the acyltransferases convergently evolved the same active site geometry and mechanism.
  • Aspartic proteases are a catalytic type of protease enzymes that use an activated water molecule bound to one or more aspartate residues for catalysis of their peptide substrates. In general, they have two highly conserved aspartates in the active site and are optimally active at acidic pH. Nearly all known aspartyl proteases are inhibited by pepstatin.
  • the cleavable site is cleavable by at least one agent selected from the group consisting of Cathepsin A or B, matrix metalloproteinases (MMPs), elastases, ⁇ -glucuronidase and ⁇ -galactosidase, preferably Cathepsin B.
  • MMPs matrix metalloproteinases
  • elastases elastases
  • ⁇ -glucuronidase ⁇ -galactosidase
  • ⁇ -galactosidase preferably Cathepsin B.
  • the cleavage site is a disulfide bond and specific cleavage is conducted by a reductive environment, e.g., an intracellular reductive environment, such as, e.g., acidic pH conditions.
  • a reductive environment e.g., an intracellular reductive environment, such as, e.g., acidic pH conditions.
  • the conjugate of the invention as disclosed herein comprises a non-cleavable linker.
  • each C 1 -C 6 alkylene, C 1 -C 6 heteroalkylene, C 2 -C 6 alkenylene, C 2 -C 6 heteroalkenylene, C 2 -C 6 alkynylene, C 2 -C 6 heteroalkynylene, C 3 -C 6 cycloalkylene, heterocycloalkylene, arylene, or heteroarylene of the non-cleavable linker as disclosed herein may optionally be interrupted by one or more heteroatoms selected from O, S and N and may e.g.
  • substituents independently selected for each occasion from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, alkaryl, alkyl heteroaryl, amino, ammonium, acyl, acyloxy, acylamino, aminocarbonyl, alkoxycarbonyl, ureido, carbamate, aryl, heteroaryl, sulfinyl, sulfonyl, hydroxyl, alkoxy, sulfanyl, halogen, carboxy, trihalomethyl, cyano, hydroxy, mercapto, and nitro.
  • the non-cleavable linker of the conjugate of the invention comprises a -(CH 2 ) n - unit, where n is an integer from, 2-12, e.g., 2-6, e.g. n is 1, 2, 3, 4, 5, or 6.
  • the non-cleavable linker of the conjugate of the invention comprises -(CH 2 ) n - wherein n is 6 and the linker is represented by the formula:
  • the non-cleavable linkers of the invention as disclosed herein further comprise a thiol-reactive group.
  • the thio-reactive group of said non-cleavable linkers as disclosed above may e.g. be selected from bromo acetamide, iodo acetamide, methylsulfonylbenzothiazole, 4,6-dichloro-l,3,5-triazin-2-ylamino group methyl-sulfonyl phenyltetrazole or methylsulfonyl phenyloxadiazole, pyridine-2 -thiol, 5- nitropyridine-2- thiol, methanethiosulfonate, or a maleimide.
  • the thiol reactive group is a maleimide (maleimidyl moiety) as disclosed above.
  • the non-cleavable linker comprising said maleimide may e.g. have the following structure, whereby the wavy line at the linker terminus indicates the point of attachment to the amatoxin:
  • the meleimidyl moiety of e.g. cleavable or non-cleavable linker as disclosed herein comprise the structure: whereby the wavy line represents the attachment site of a cleavable or non-cleavable linker as disclosed herein.
  • conjugates of the invention comprising a cleavable or non-cleavable linker which further comprise a thiol-reactive group may be coupled to a naturally occurring sulfhydryl moiety in the antibody of the conjugate, or said cleavable or non-cleavable linker of the conjugates of the invention comprising a thiol-reactive group may be coupled to a sulfhydryl moiety which has been introduced into the antibody by genetic engineering as described in e.g. Nat Biotechnol. 2008 Aug;26(8):925-32.
  • the cleavable or non-cleavable linker as disclosed herein which comprise a thio-reactive group are coupled to sulfhydryl moieties that have been introduced into the Fc region of the antibody of the conjugate of the invention by genetic engineering.
  • Preferred positions within the Fc region of said antibody at which sulfhydryl moieties may be introduced comprise D265, or Al 18 (according to EU numbering), more preferably D265.
  • the conjugate in said composition comprises any of the following compounds of formulas (I) to (XI), respectively, as linker-amatoxin moieties:
  • the conjugates of the invention are preferably synthesized by reacting thiol groups of antibodies with a compound according to any one of formula Xlla to XXIIa containing a maleimide moiety as reactive cap.
  • reactive cap refers to a chemical moiety which reactacts with e.g. thiol groups of the antibody to covalenty link the the compounds of formula (Xlla) to (XXIIa) to the antibody.
  • the compounds according to formula (Xlla) - (XXIIa) of the invention are used for generating or manufacturing antibody-drug conjugates (ADCs), more specifically antibody-targeted amatoxin conjugates.
  • ADCs of the invention as disclosed herein may also be referred to as “ATACs”.
  • compounds (Xlla) to (XXIIa) are reacted with target binding moieties, such as e.g. antibodies, e.g. human IgGl antibodies, under suitable conditions to form ATACs.
  • target binding moieties such as e.g. antibodies, e.g. human IgGl antibodies
  • the compounds of the invention according to formula (Xlla) - (XXIIa) can e.g. be reacted with target binding moieties, such as e.g.
  • said conjugates of the invention are compound according to any one of formula (Xllb) to (XXIIb) as disclosed hereinbelow:
  • n is preferably from 1 to 10, preferably from 1, 2, 3 to 4, preferably from 1, 2 to 5, preferably from 4 to 7, preferably from 8 to 10.
  • said conjugate is a compound according to any one of formula XIII to XXII:
  • amatoxin linker moieties are coupled to s-amino groups of naturally occurring lysine residues of said antibody, and wherein n is preferably from 1 to 8, preferably from 1, 2, 3 to 4, preferably from 2 to 5, preferably from 5 to 7.
  • said conjugate is a compound according to any one of formula XXIII, XXIV, Xlllb, XXIIb, XXb, and XVIb:
  • amatoxin linker moieties are coupled to the thiol groups of cysteine residues of the antibody, and wherein n is preferably from 1 to 10, or e.g. wherein n is from 2, 4 to 6, more preferably wherein n is 1, 2, 4, or 8.
  • said composition comprises at least one one of the immune checkpoint inhibitors avelumab, nivolumab, pembrolizumab, durvalumab, or ipilimumab and a conjugate comprising an antibody directed against HER2 (aHER2-LALA-D265C) and an amatoxin according to formula XXIb, XXIIb, Xllb, Xlllb, XVIIIb, or XlXb, wherein the antibody as denoted in the respective amatoxin structures as disclosed above is the ⁇ HER2-LALA-D265C as disclosed herein.
  • said composition comprises one of
  • the composition of the invention comprises the antibody as disclosed above coupled to one amatoxin as disclosed above and at least one immune checkpoint inhibitor as disclosed above.
  • said composition comprises at least one one of the immune checkpoint inhibitors avelumab, nivolumab, pembrolizumab, durvalumab, or ipilimumab and a conjugate comprising an antibody directed against CD19 (chiBCE19-D265C) and an amatoxin according to formula XXIII, XXII, XXb, XXIIb, XXIV, Xlllb, or XVIb , wherein the antibody as denoted in the respective amatoxin structures as disclosed above is CD 19 (chiBCE19-D265C).
  • said composition comprises one of
  • the composition of the invention comprises the antibody as disclosed above coupled to one amatoxin as disclosed above and at least one immune checkpoint inhibitor as disclosed above.
  • said composition according to the invention comprises at least one one of the immune checkpoint inhibitors avelumab, nivolumab, pembrolizumab, durvalumab, or ipilimumab and a conjugate comprising an antibody directed against PSMA and an amatoxin according to formula XXIII, XXII, XXb, XXIIb, XXIV, Xlllb, or XVIb, wherein the antibody as denoted in the respective amatoxin structures as disclosed above is an anti-PSMA antibody.
  • said composition comprises one of
  • composition of the invention comprises the antibody as disclosed above coupled to one amatoxin as disclosed above and at least one immune checkpoint inhibitor as disclosed above.
  • the anti-PSMA antibody of the conjugate of the invention as disclosed above is an anti-PSMA antibody as disclosed in WO 2020/025564.
  • the antibody of the conjugate of the invention as disclosed herein is one of 3-F11-varl, 3-Fl l-var2, 3-Fl l-var3, 3-Fl l-var4, 3-Fl l-var5, 3-Fl l-var6, 3-F11- var7, 3-Fl l-var8, 3-Fl l-var9, 3-Fl l-varl0, 3-F11-varl 1, 3-Fl l-varl2, 3-Fl l-varl3, 3-F11- varl4, 3-F11-varl 15, or 3-Fl l-varl6, preferably, the antibody of the conjugate of the invention is one of 3-F11-varl, 3-Fl l-varl3, or 3-Fl l-varl6 as disclosed in WO 2020/025564.
  • the anti-PSMA antibody as disclosed above for use in the composition of the invention comprises at least one mutation in its Fc region selected from L234A, L235A and D265C (numbering according to EU nomenclature), preferably L234A, L235A and D265C.
  • said composition according to the invention comprises at least one of the immune checkpoint inhibitors avelumab, nivolumab, pembrolizumab, durvalumab, or ipilimumab and a conjugate comprising an antibody directed against CD37 and an amatoxin according to formula XXIII, XXII, XXb, XXIIb, XXIV, Xlllb, or XVIb, wherein the antibody as denoted in the respective amatoxin structures as disclosed above is an anti-CD37 antibody.
  • said composition comprises one of
  • composition of the invention comprises the antibody as disclosed above coupled to one amatoxin as disclosed above and at least one immune checkpoint inhibitor as disclosed above.
  • the anti-CD37 antibody as disclosed above for use in the composition of the invention comprises at least one mutation in its Fc region selected from L234A, L235A and D265C (numbering according to EU nomenclature), preferably L234A, L235A and D265C.
  • said composition according to the invention comprises at least one of the immune checkpoint inhibitors avelumab, nivolumab, pembrolizumab, durvalumab, or ipilimumab and a conjugate comprising an antibody directed against CD269 and an amatoxin according to formula XXIII, XXII, XXb, XXIIb, XXIV, Xlllb, or XVIb, wherein the antibody as denoted in the respective amatoxin structures as disclosed above is an anti-CD269 antibody
  • said composition comprises one of
  • composition of the invention comprises the antibody as disclosed above coupled to one amatoxin as disclosed above and at least one immune checkpoint inhibitor as disclosed above.
  • the anti-CD269 antibody of the conjugate of the invention as disclosed above is the humanized antibody J22.9-ISY as disclosed in WO20 18/115466 and wherein the conjugate comprises an amatoxin linker-moiety according to formula (I), or wherein the conjugate is represented by formula (Xllb) as disclosed herein.
  • the anti-CD269 antibody as disclosed above for use in the composition of the invention may e.g. comprise at least one mutation in its Fc region selected from L234A, L235A and D265C (numbering according to EU nomenclature), preferably the Fc region of said antibody comprises the mutations L234A, L235A and D265C.
  • said composition according to the invention comprises at least one of the immune checkpoint inhibitors avelumab, nivolumab, pembrolizumab, durvalumab, or ipilimumab and a conjugate comprising an antibody directed against sialyl Lewis a and an amatoxin according to formula XXIII, XXII, XXb, XXIIb, XXIV, Xlllb, or XVIb wherein the antibody as denoted in the respective amatoxin structures as disclosed above is an anti- a sialyl Lewis a antibody.
  • said composition comprises one of
  • the composition of the invention comprises the antibody as disclosed above coupled to one amatoxin as disclosed above and at least one immune checkpoint inhibitor as disclosed above.
  • the anti- sialyl Lewis a antibody as disclosed above for use in the composition of the invention comprises at least one mutation in its Fc region selected from L234A, L235A and D265C (numbering according to EU nomenclature), preferably L234A, L235A and D265C.
  • said composition according to the invention comprises at least one one of the immune checkpoint inhibitors avelumab, nivolumab, pembrolizumab, durvalumab or ipilimumab and a conjugate comprising an antibody directed against EpCAM and an amatoxin according to formula XXIII, XXII, XXb, XXIIb, XXIV, Xlllb, or XVIb, wherein the antibody as denoted in the respective amatoxin structures as disclosed above is an anti- EpCAM antibody.
  • said composition comprises one of Accordingly, the composition of the invention comprises the antibody as disclosed above coupled to one amatoxin as disclosed above and at least one immune checkpoint inhibitor as disclosed above.
  • the anti-EpCAM antibody as disclosed above for use in the composition of the invention comprises at least one mutation in its Fc region selected from L234A, L235A and D265C (numbering according to EU nomenclature), preferably L234A, L235A and D265C.
  • the conjugate for use in the composition of the invention as disclosed above comprises or is according to formula Xllb, XlVb, XVIIIb, XXb, or XVIIb.
  • said composition according to the invention comprises at least one one of the immune checkpoint inhibitors avelumab, nivolumab, pembrolizumab, durvalumab, or ipilimumab and a conjugate according to formula XXIII, XXII, XXb, XXIIb, XXIV, Xlllb, or XVIb.
  • said composition comprises at least one of
  • composition of the invention comprises at least a conjugate as disclosed above and at least one immune checkpoint inhibitor as disclosed above.
  • the present invention pertains a conjugate according to formula (Xllb), (Xlllb), (XlVb), (XVb), (XVII)), (XVIIb), (XVIIIb), (XlXb), (XXb), (XXIb), (XXIIb) for use in the manufacture of a composition of the invention.
  • the present invention pertains to an immune checkpoint inhibitor selected from the group of avelumab, nivolumab, pembrolizumab, ipilimumab, or durvalumab for use in the manufacture of a composition according to the invention as disclosed herein.
  • a pharmaceutical formulation comprising the composition (for use) as described, further comprising one or more pharmaceutically acceptable buffers, surfactants, diluents, carriers, excipients, fillers, binders, lubricants, glidants, disintegrants, adsorbents, and/or preservatives.
  • said pharmaceutical formulation may be ready for administration, while in lyophilised form said formulation can be transferred into liquid form prior to administration, e.g., by addition of water for injection which may or may not comprise a preservative such as for example, but not limited to, benzyl alcohol, antioxidants like vitamin A, vitamin E, vitamin C, retinyl palmitate, and selenium, the amino acids cysteine and methionine, citric acid and sodium citrate, synthetic preservatives like the parabens methyl paraben and propyl paraben.
  • a preservative such as for example, but not limited to, benzyl alcohol, antioxidants like vitamin A, vitamin E, vitamin C, retinyl palmitate, and selenium, the amino acids cysteine and methionine, citric acid and sodium citrate, synthetic preservatives like the parabens methyl paraben and propyl paraben.
  • Said pharmaceutical formulation may further comprise one or more stabilizer, which may be, e.g., an amino acid, a sugar polyol, a disaccharide and/or a polysaccharide.
  • Said pharmaceutical formulation may further comprise one or more surfactant, one or more isotonizing agents, and/or one or more metal ion chelator, and/or one or more preservative.
  • the pharmaceutical formulation as described herein can be suitable for at least intravenous, intramuscular or subcutaneous administration.
  • said conjugate according to the present invention may be provided in a depot formulation which allows the sustained release of the biologically active agent over a certain period of time.
  • a primary packaging such as a prefilled syringe or pen, a vial, or an infusion bag is provided, which comprises said formulation according to the previous aspect of the invention.
  • the prefilled syringe or pen may contain the formulation either in lyophilised form (which has then to be solubilised, e.g., with water for injection, prior to administration), or in aqueous form.
  • Said syringe or pen is often a disposable article for single use only, and may have a volume between 0.1 and 20 ml.
  • the syringe or pen may also be a multi-use or multi-dose syringe or pen.
  • Said vial may also contain the formulation in lyophilised form or in aqueous form and may serve as a single or multiple use device. As a multiple use device, said vial can have a bigger volume.
  • Said infusion bag usually contains the formulation in aqueous form and may have a volume between 20 and 5000 ml.
  • the composition for use in the treatment of cancer, or the pharmaceutical formulation as described relates to a cancer which is selected from the group consisting of melanoma, squamous and non-squamous non-small cell lung cancer, metastatic small cell lung cancer, renal cell carcinoma, Hodgkin lymphoma, B lymphocyte-associated malignancies, urothelial carcinoma, head and neck squamous cell carcinoma, Merkel cell carcinoma, hepatocellular carcinoma, gastric and gastroesophageal carcinoma, metastatic colorectal cancer, multiple myeloma, primary mediastinal B cell lymphoma, recurrent or metastatic cervical cancer, and metastatic cutaneous squamous cell carcinoma, prostate cancer, breast cancer, including triple-negative breast cancer (TNBC).
  • TNBC triple-negative breast cancer
  • the present invention relates to said composition or pharmaceutical formulation as disclosed herein for use in the treatment of B lymphocyte- associated malignancies, in particular for use in the treatment of non-Hodgkin’s lymphoma, follicular lymphoma, diffuse large B cell non-Hodgkin’s lymphoma, and chronic lymphocytic leukaemia.
  • the present invention pertains to an immune checkpoint inhibitor as disclosed herein for use in a composition of the invention, or in a pharmaceutical composition of the invention. Accordingly, the present invention pertains to avelumab, nivolumab, pembrolizumab, ipilimumab, PD1-1, PD1-2, PD1-3, PD1-4, PD1-5, pidilizumab, cemiplimab, JTX-4014, spartalizumab, sintilimab (IBI308), dostarlimab, Toripalimab, durvalumab, or atezolizumab for use in a composition or pharmaceutical formulation according to the invention as disclosed herein.
  • the present invention pertains to the use of avelumab, nivolumab, pembrolizumab, ipilimumab, or durvalumab in a composition or pharmaceutical formulation according to the invention as disclosed herein.
  • the invention further relates to a method for the treatment of cancer in a human subject in need thereof, wherein the method comprises administering to the subject a composition comprising (a) at least one immune checkpoint inhibitor and (b) at least one conjugate, wherein said conjugate is comprising (i) a target binding moiety, (ii) at least one amatoxin, and (iii) optionally at least one linker connecting said target binding moiety with said at least one amatoxin.
  • the method of treatment of cancer in a human subject in need thereof as disclosed above comprises administering the composition or the pharmaceutical formulation as disclosed above for the treatment of melanoma, squamous and non-squamous non-small cell lung cancer, metastatic small cell lung cancer, renal cell carcinoma, Hodgkin lymphoma, urothelial carcinoma, head and neck squamous cell carcinoma, Merkel cell carcinoma, hepatocellular carcinoma, gastric and gastroesophageal carcinoma, metastatic colorectal cancer, primary mediastinal B cell lymphoma, recurrent or metastatic cervical cancer, and metastatic cutaneous squamous cell carcinoma.
  • the method of treating cancer as disclosed above comprises adiministering the immune checkpoint inhibitor and the conjugate of the composition as disclosed herein sequentially or simultaneously.
  • the checkpoint inhibitor may be administered first, followed by the administration of the conjugate, alternatively, the conjugate of the invention may e.g. be administered first followed by the administration of the immune checkpoint inhibitor.
  • the immune checkpoint inhibitor and the conjugate of the invention are administered intravenously (i.v.) sequentially or simultaneously as disclosed above.
  • the term “simultaneous administration” as used herein refers to the administration of the immune checkpoint inhibitor and of the conjugate of the invention on the same day, e.g. within 1 hour, 2 hours, 4 hours, 6 hours, 8 hours, 12 hours, 18 hours, 20 hours, or 23 hours apart from each other.
  • the invention further relates to the use of a composition comprising (a) at least one immune checkpoint inhibitor and (b) at least one conjugate, wherein said conjugate is comprising (i) a target binding moiety, (ii) at least one amatoxin, and (iii) optionally at least one linker connecting said target binding moiety with said at least one amatoxin, for the treatment of cancer.
  • step 1 product 7.99mg (8.54 ⁇ mol) of step 1 product were dissolved in 665pl dry DMF. At ambient temperature 127.98 ⁇ l of a solution of TBTU in dry DMF were added. Afterwards a solution of 128.04pl of DIPEA in dry DMF was added (stock solution: 52.25pl DIPEA in 1500pl dry DMF). The resulted solution was stirred for 1 minute at ambient temperature under argon. Then a solution of 128.05 ⁇ l of BMP-Val-Ala-PAB-NH 2 (WO2018115466) was added. The resulted reaction mixture was stirred for 2h at room temperature. The solvent was removed under reduced pressure. The crude product was purified by HPLC to yield 7.34mg (63%) of the pure product.
  • Antibodies were conjugated to the amatoxin linker conjugates by means of the so-called Thiomab technology.
  • the conjugation takes place by conjugation of the maleimide residue of the toxin linker construct to the free SH group of a cysteine residue in the antibody, as shown in the following reaction scheme:
  • the antibodies used in the present experiments comprise a D265C substitution in both Fc domains, in order to provide a cystein residue that has such free SH group.
  • the respective technology is disclosed in WO2016/142049 Al, the content of which is incorporated herein by reference, and which results in a homogenous product with a fixed drug to antibody ration (“DAR”) of 2 and a site specific conjugation.
  • DAR drug to antibody ration
  • the Her2-positive cell line BT474 is a human breast cancer cell line.
  • the CD79b-positive cell line BJAB is a human Burkitt lymphoma-derived B cell line.
  • Maytansine 100 nM
  • Amanitin 100 nM
  • Anti-HER2-Amanitin conjugate 50 nM antibody
  • Anti- CD79b-Amanitin conjugate 50 nM antibody
  • BT474 cells were assayed at 24, 48, and 72 h post-treatment.
  • the adherent BT474 cells were lifted with a solution of highly purified, recombinant cell-dissociation enzymes (TrypLE, Thermo- Fisher) for 2 min. at 37°C.
  • Both BT474 and BJAB cells were washed with PBS/2% FBS and then fixed in 0.5% paraformaldehyde for 5 min. After washing twice with cold PBS/2% FBS, cells were incubated for 5 min. with an Fc receptor binding inhibitor (FC block, eBioscience), blocking Fc receptor-mediated non-specific binding, followed by 30-min.
  • FC block Fc receptor binding inhibitor
  • Culture media was collected at 24, 48, and 72 h post-treatment. Analytes were captured on a Nunc Maxisorp 96-well plate coated at pH 9 with 1 pg/mL Anti-HMGBl antibody (clone 1D5, Sigma), and plates were blocked with casein buffer (ThermoFisher). A standard curve was generated using recombinant human HMGB1 protein (R&D Systems) added to culture media that had been incubated at 37°C/5% CO 2 for the same number of days as the experimental samples. This step had the effect of normalizing background signals observed in fresh culture media. Media from the experimental samples were transferred to fresh tubes, centrifuged gently to pellet debris, and then added to the prepared plate along with the standard curve.
  • R&D Systems recombinant human HMGB1 protein
  • Amanitin-conjugated antibody-drug conjugates induced exposure and secretion of said ICD markers, respectively, in a target-dependent manner.
  • Example 4 Synergistic Cytotoxic Action of AT AC and Immune Checkpoint Inhibitor in vivo
  • the cytotoxic activity of the combination comprising an ICI and an ATAC has been assessed by use of a tumor mouse model in vivo.
  • the study consisted of 6 experimental groups with 12 animals each.
  • CD 19-positive Raji cells human Burkitt lymphoma, DSMZ
  • PBMCs human peripheral blood mononuclear cells
  • Table 1 Mean Tumorvolumes at day 31 post tumor cell inoculation. TV: tumor volumes;
  • PBMCs peripheral blood mononuclear cells
  • Tumor volumes were measured twice per week by calliper measurements, and body weights were determined in parallel. Animals were sacrificed and necropsies were performed when either tumor volumes were >1600 mm 3 or when the mice needed to be sacrificed due to ethical reasons (according to German animal welfare legislation). Table 2: Experimental Details of the Study in Example 4
  • the combination of the CD19-specific ATAC chiBCE19-D265C-XIIb at a dose of 0.3 mg/kg body weight with the PD-L1 -specific antibody Avelumab at a dose of 20 mg/kg body weight yielded a higher reduction in tumor volumes in vivo as compared to the reduction achieved by the CD19-specific ATAC chiBCE19-D265C-XIIb alone.
  • CD 19- positive Raji cells human Burkitt lymphoma, DSMZ
  • PBMCs German Red Cross
  • Fig. 9 illustrate that the synergistic cytotoxic effect of ATAC and anti-PD-1 Immune Checkpoint Inhibitor in vivo depends on the presence of human PBMCs in the mouse model used and that an effective dose for the anti- CD 19 ATAC is needed in order to induce immunogenic cell death which synergizes with the activity of the immune checkpoint inhibitor.
  • Example 6 Efficacy of anti-CD19 ATAC chiBCE19-D265C-XIIb and Ipilimumab in a Raji Tumor Xenograft Model in NOD/SCID Mice Reconstituted with Human PBMCs
  • CD 19- positive Raji cells human Burkitt lymphoma, DSMZ
  • PBMCs German Red Cross
  • Fig. 10 illustrate a synergistic effect on survival in animals that received a combination treatment of CTLA4 immune checkpoint inhibitor and anti-CD19-ATAC (group 6).
  • the results indicate that an effective amount of the anti-CD19 ATAC is needed to induce immunogenic cell death which in combination with a CTLA4 checkpoint inhibitor (e.g. Ipilimumab) acts synergistically as evidenced by prolonged survival of the respective treatment group.
  • a CTLA4 checkpoint inhibitor e.g. Ipilimumab

Landscapes

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

Abstract

La présente invention concerne une composition comprenant (a) au moins un inhibiteur de point de contrôle immunitaire et (b) au moins un conjugué, ledit conjugué comprenant (i) une fraction de liaison à une cible, (ii) au moins une amatoxine, et (iii) éventuellement au moins un lieur reliant ladite fraction de liaison à une cible à ladite ou auxdites amatoxines. La présente invention concerne en outre ladite composition destinée à être utilisée dans le traitement d'un patient atteint d'un cancer, et une formulation pharmaceutique comprenant ladite composition et des excipients supplémentaires, ainsi que des méthodes de production et d'utilisation de ladite composition.
EP21802741.5A 2020-11-04 2021-11-04 Composition comprenant une combinaison d'un inhibiteur de point de contrôle immunitaire et d'un conjugué anticorps-amatoxine pour une utilisation dans le traitement du cancer Pending EP4240424A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20205794 2020-11-04
PCT/EP2021/080688 WO2022096604A1 (fr) 2020-11-04 2021-11-04 Composition comprenant une combinaison d'un inhibiteur de point de contrôle immunitaire et d'un conjugué anticorps-amatoxine pour une utilisation dans le traitement du cancer

Publications (1)

Publication Number Publication Date
EP4240424A1 true EP4240424A1 (fr) 2023-09-13

Family

ID=73288362

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21802741.5A Pending EP4240424A1 (fr) 2020-11-04 2021-11-04 Composition comprenant une combinaison d'un inhibiteur de point de contrôle immunitaire et d'un conjugué anticorps-amatoxine pour une utilisation dans le traitement du cancer

Country Status (10)

Country Link
EP (1) EP4240424A1 (fr)
JP (1) JP2023548522A (fr)
KR (1) KR20230104653A (fr)
CN (1) CN116744969A (fr)
AU (1) AU2021373298A1 (fr)
CA (1) CA3195572A1 (fr)
CL (1) CL2023001266A1 (fr)
IL (1) IL302034A (fr)
MX (1) MX2023005192A (fr)
WO (1) WO2022096604A1 (fr)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3208117A1 (fr) * 2021-03-19 2022-09-22 Torsten HECHLER Conjugues d'anticorps d'amatoxine specifiques de lymphocytes b
US20240165257A1 (en) 2022-11-01 2024-05-23 Heidelberg Pharma Research Gmbh Anti-gucy2c antibody and uses thereof

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
AU784012B2 (en) 1999-08-24 2006-01-12 E. R. Squibb & Sons, L.L.C. Human CTLA-4 antibodies and their uses
PT2439273T (pt) 2005-05-09 2019-05-13 Ono Pharmaceutical Co Anticorpos monoclonais humanos para morte programada 1 (pd- 1) e métodos para o tratamento de cancro utilizando anticorpos anti-pd-1 isoladamente ou em combinação com outros imunoterapêuticos
NZ600758A (en) 2007-06-18 2013-09-27 Merck Sharp & Dohme Antibodies to human programmed death receptor pd-1
DE202007018529U1 (de) 2007-07-07 2008-12-04 Chamalow S.A. Implantierbarer Funkfrequenzdefibrillator R.F.
US8747847B2 (en) 2008-02-11 2014-06-10 Curetech Ltd. Monoclonal antibodies for tumor treatment
EP2262837A4 (fr) 2008-03-12 2011-04-06 Merck Sharp & Dohme Protéines de liaison avec pd-1
EP3485912A1 (fr) 2009-04-08 2019-05-22 Faulstich, Heinz, Dr. Constituants thérapeutiques contenant de l'amatoxine de liaison à la surface cellulaire destinés à la thérapie des tumeurs
WO2015033301A1 (fr) 2013-09-06 2015-03-12 Aurigene Discovery Technologies Limited Dérivés 1,3,4-oxadiazole et 1,3,4-thiadiazole servant d'immunomodulateurs
EP4115910A1 (fr) 2015-03-09 2023-01-11 Heidelberg Pharma Research GmbH Conjugués amatoxine-anticorps
EP3222292A1 (fr) 2016-03-03 2017-09-27 Heidelberg Pharma GmbH Conjugués d'amanitine
US11446388B2 (en) 2016-12-23 2022-09-20 Heidelberg Pharma Research Gmbh Amanitin antibody conjugates
EP3630822A1 (fr) 2017-06-02 2020-04-08 Boehringer Ingelheim International GmbH Polythérapie anticancéreuse
JP2021521202A (ja) * 2018-04-13 2021-08-26 ハイデルベルク ファルマ リサーチ ゲゼルシャフト ミット ベシュレンクテル ハフツング 固形腫瘍の治療のための標的化されたアマトキシン複合体
CN112533956A (zh) 2018-07-31 2021-03-19 海德堡医药研究有限责任公司 针对psma的人源化抗体

Also Published As

Publication number Publication date
AU2021373298A1 (en) 2023-06-08
IL302034A (en) 2023-06-01
JP2023548522A (ja) 2023-11-17
WO2022096604A1 (fr) 2022-05-12
CL2023001266A1 (es) 2023-12-15
CA3195572A1 (fr) 2022-05-12
KR20230104653A (ko) 2023-07-10
MX2023005192A (es) 2023-05-15
CN116744969A (zh) 2023-09-12

Similar Documents

Publication Publication Date Title
CN111164100A (zh) 白介素-21突变蛋白和治疗方法
EA035987B1 (ru) ПОЛИПЕПТИДЫ, СОДЕРЖАЩИЕ Fc С ИЗМЕНЕННЫМ ГЛИКОЗИЛИРОВАНИЕМ И СНИЖЕННОЙ АФФИННОСТЬЮ К Fc-ГАММА РЕЦЕПТОРАМ
TWI718427B (zh) 對cd123具專一性之抗體和抗體-藥物共軛體及彼等之用途
CN110869394A (zh) 工程改造的抗体化合物及其缀合物
EP4240424A1 (fr) Composition comprenant une combinaison d'un inhibiteur de point de contrôle immunitaire et d'un conjugué anticorps-amatoxine pour une utilisation dans le traitement du cancer
CN117946277A (zh) 多特异性抗体及其制备和使用方法
US20230330248A1 (en) Antibody-drug conjugate including novel cyclic dinucleotide derivative
KR102281405B1 (ko) 항-vista 항체 및 이의 용도
CN112566937A (zh) 对cd3特异性的抗体及其用途
CN112739340A (zh) 抗cd5抗体药物缀合物(adc)在同种异体细胞疗法中的用途
TW202233248A (zh) 組合療法
JP2023533793A (ja) 治療用抗体およびそれらの使用
US20220370632A1 (en) B-lymphocyte specific amatoxin antibody conjugates
CN113811329A (zh) 抗pd-1抗体和其用途
US20220133902A1 (en) Composition Comprising a Combination of Immune Checkpoint Inhibitor and Antibody-Amatoxin Conjugate for Use in Cancer Therapy
KR20230009397A (ko) B7-h3 항체 약물 콘쥬게이트를 단독으로 또는 조합하여 사용하는 방법
CA3078812A1 (fr) Anticorps anti-glyco-muc1 et leurs utilisations
EP4375300A1 (fr) Composition pharmaceutique et son utilisation
CN116801871A (zh) 组合疗法
JP2022552349A (ja) Bリンパ球特異的アマトキシン抗体コンジュゲート
JP2023100255A (ja) 治療用抗体およびそれらの使用
WO2024023771A1 (fr) Polythérapies pour le traitement du cancer
KR20230107478A (ko) 치료 항체 및 그의 용도
CN112996816A (zh) 抗糖-muc1抗体及其用途

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230525

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40097583

Country of ref document: HK

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20240226