US20100310454A1 - Combination of an anti-edb fibronectin antibody-il-2 fusion protein, and a molecule binding to b cells, b cell progenitors and /or their cancerous counterpart - Google Patents

Combination of an anti-edb fibronectin antibody-il-2 fusion protein, and a molecule binding to b cells, b cell progenitors and /or their cancerous counterpart Download PDF

Info

Publication number
US20100310454A1
US20100310454A1 US12/863,417 US86341708A US2010310454A1 US 20100310454 A1 US20100310454 A1 US 20100310454A1 US 86341708 A US86341708 A US 86341708A US 2010310454 A1 US2010310454 A1 US 2010310454A1
Authority
US
United States
Prior art keywords
antibody
combination
rituximab
combination according
lymphoma
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.)
Abandoned
Application number
US12/863,417
Other languages
English (en)
Inventor
Dario Neri
Hans Dietrich Menssen
Andreas Menrad
Christoph Schliemann
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of US20100310454A1 publication Critical patent/US20100310454A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/44Antibodies bound to carriers
    • 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
    • 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
    • A61K47/6813Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin the drug being a peptidic cytokine, e.g. an interleukin or interferon
    • 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
    • A61K47/6849Medicinal 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 receptor, a cell surface antigen or a cell surface determinant
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • 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
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3061Blood cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • 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

Definitions

  • the present invention relates to a combination of an anti-EDb fibronectin antibody-IL-2 fusion protein, and a molecule binding to B cells, B cell progenitors and/or their cancerous counterpart and uses thereof.
  • B-NHL B cell non-Hodgkin lymphoma
  • B-NHL B cell non-Hodgkin lymphoma
  • Rituximab (Rituxan®; R) is a chimeric monoclonal IgG1 antibody that directly binds to the CD20 cell surface epitope constitutively expressed on the cell surface of malignant and normal B cell populations.
  • rituximab (a) elicits antibody-dependent cellular cytotoxicity (ADCC), (b) induces lymphoma cell death through complement-dependent cytolysis (CDC) and/or complement-dependent cellular cytotoxicity, and (c) directly induces apoptosis following the engagement of CD20 by rituximab.
  • rituximab possibly has a vaccinal effect implemented via cross-presentation of lymphoma antigens from rituximab-killed malignant B cells by antigen-presenting cells and priming of lymphoma antigen-specific cytotoxic T cells (Selenko et al, 2001).
  • rituximab Treatment with rituximab as a single agent induces significant but moderate and short-lasting responses in patients with almost every subtype of B-cell lymphoma. However, its biggest benefit is seen when it is combined with induction chemotherapy regimens (Coiffier, 2006). Combined with standard chemotherapy, in particular with CHOP (cyclophosphamide, vincristine, adriamycin and prednisolone), rituximab at a dose of 375 mg/m 2 as a 90-min intravenous infusion on day 1 of each chemotherapy cycle even increases the cure rate of patients with diffuse large B cell lymphoma (DLBCL) to approximately 52% (Coiffier 2002, update of GELA OS data, ASH 2007) from 38% with chemotherapy alone.
  • DLBCL diffuse large B cell lymphoma
  • indolent lymphoma the addition of rituximab to every induction chemotherapy combination (FCM, CVP, CHOP, FND) has resulted in a significant increase in the overall response and complete remission rates as well as in a delay of the time to disease progression (Marcus, 2005; Hiddemann 2005).
  • adding rituximab to chemotherapy not always leads to improved clinical outcomes.
  • treatment with CHOP plus rituximab resulted in a similar progression-free survival and overall survival compared with patients on CHOP therapy alone (Lenz et al, 2005).
  • rituximab monotherapy In addition to its established role as a treatment to induce remissions (induction therapy) in B-NHL patients, rituximab monotherapy also has been evaluated as a maintenance therapy to consolidate responses or prolong remissions. Under the assumption that 25 mg rituximab/ml is the lowest acceptable serum concentration, a dose of 375 mg/m 2 rituximab infused every 3 months was found to be sufficient for rituximab maintenance therapy in a prospective pharmacokinetic study (Gordan, 2005).
  • rituximab maintenance therapy provides additional benefit to those patients in whom it was used as a part of the induction chemotherapy (e.g. R-CHOP).
  • rituximab in combination with chemotherapy (e.g. R-CHOP)
  • chemotherapy e.g. R-CHOP
  • approximately 50% of patients with relapsed/refractory CD20+ follicular lymphomas do not respond to initial treatment with rituximab (innate resistance; McLaughlin et al 1998), and about 60% of prior rituximab responding patients will not benefit from retreatment with rituximab (acquired resistance; Davis et al, 2000).
  • rituximab resistance represents a significant barrier to immuno- and immonochemotherapy of B-NHLs in terms of further improved clinical outcome.
  • Monoclonal antibodies targeting surface molecules other than CD20 in B-NHLs such as lumilixumab (anti-CD23), epratuzumab (anti-CD22), SGN-40 and HCD122 (both anti-CD40), galiximab (anti-CD80), apolizumab (Hu1D10), KRN848, 1D09C3 (all anti-HLA-DR) have shown promise in early clinical trials.
  • Novel anti-CD20 antibodies and antibodies directed against non-CD20 B-cell epitopes will have to demonstrate a significantly superior efficacy over rituximab to be considered successful, however, early clinical results with the most of these antibodies indicate incremental benefits, only.
  • anti B cell antibodies an rituximab in particular are being developed for the treatment of autoimmune diseases, including rheumatoid arthritis, Crohn's disease and autoimmune hemolytic anemia. (Assous et al, 2008).
  • EDB extra domain B of fibronectin
  • This 91-amino acid type III homology domain can be inserted into the fibronectin molecule during active tissue remodeling by a mechanism of alternative splicing (Zardi et al., supra).
  • EDB is essentially undetectable in healthy adult tissues but is highly abundant in the vasculature of many aggressive solid tumors.
  • the tumor-targeting ability of the high-affinity human antibody L19 (Pini et al., J Biol. Chem.
  • rIL-2 high dose P ⁇ 0.001.
  • L19-IL2 low dose vs. saline P ⁇ 0.00001
  • L19-IL2 low dose vs. rIL-2 1ow dose P ⁇ 0.00001
  • inducing CRs in 4 of 5 cases after 4 cycles of therapy whereas even a three-fold higher dose of the non-targeted cytokine combined with rituximab was only able to retard tumor growth (L19-IL2 low dose vs. rIL-2 high dose : P ⁇ 0.001).
  • the invention relates to a combination comprising at least
  • the molecule binding to B cells, B cell progenitors and/or their cancerous counterpart is specifically binding to CD20, CD23, CD22, CD40, CD80, HLA-DR or Hu1D10.
  • the molecule binding to B cells, B cell progenitors and/or their cancerous counterpart is selected from an antibody, antibody fragment or antibody mimetic.
  • Preferred is a molecule specifically binding to CD20, CD23, CD22, CD40 or CD80 which is a full-length antibody or antibody fragment, or a fusion protein thereof.
  • the antibody or antibody fragment, or fusion protein thereof is specifically binding to CD20.
  • the invention relates to a combination comprising at least
  • the invention relates to a combination comprising at least
  • the molecule specifically binding to cells expressing CD20 and/or specifically binding to CD20 is an antibody or antibody fragment specifically binding to CD20.
  • the antibody-part of (i) specifically binds to the EDb-domain of fibronectin (FN).
  • FN fibronectin
  • the antibody specifically recognizing EDb-fibronectin binds to a cryptic epitope.
  • An example for such antibody is the BC-1 antibody.
  • such antibody which binds to the EDb-domain of fibronectin exhibits a high affinity for the EDb-domain of FN, in particular, the antibody binds to the ED b fibronectin domain with nanomolar or subnanomolar affinity.
  • Such antibodies are known in the prior art and are e.g. described in WO99/58570.
  • the L19 antibody In particular preferred is the L19 antibody.
  • the antibody part specifically recognizing EDb fibronectin, in particular the L19 antibody can be employed in various antibody formats.
  • Preferred antibody formats are full IgG, Fab, (Fab′) 2 , scFv, diabody, minibody or small immunoprotein (SIP) format.
  • SIP small immunoprotein
  • the full IgG, scFv and SIP format for the L19 antibody Most preferred is the L19 antibody in the scFv format.
  • Several immunoprotein formats are known in the prior art, e.g. based on the CH3 domain or the ⁇ s2 -CH4 domain of IgE.
  • the preferred SIP format for L19 based on the ⁇ s2 -CH4 domain of IgE and L19 in full IgG format are for example described in WO03/076469.
  • the antibody-part contains at least one CDR sequence of the L19 antibody.
  • the antibody-part comprises the CDR sequences of the L19 antibody, in particular it comprises the sequences according to SEQ ID no. 6 to 11.
  • the antibody-part comprises the VL and VH chain of the L19 antibody. In a preferred embodiment, it comprises least one VH chain according to SEQ ID No. 1 or at least one VL chain according to SEQ ID No. 2. In an especially preferred embodiment, it comprises least one VH chain according to SEQ ID No. 1 and at least one VL chain according to SEQ ID No. 2.
  • the antibody-part comprises one VH chain according to SEQ ID No. 1 and one VL chain according to SEQ ID No. 2. In a further preferred embodiment, the antibody-part comprises two VH chains according to SEQ ID No. 1 and two VL chains according to SEQ ID No. 2.
  • VH and the VL chains are connected by an antibody linker.
  • the antibody linker comprises a sequence according to SEQ ID No. 3, or a sequence having at least 90% identity to the sequence according to SEQ. ID. No. 3.
  • the antibody-part specifically binding to EDb-fibronectin is fused to Interleukin-2. Both parts may be fused directly, or may be fused via a linker, in particular by a peptidic fusion protein linker.
  • the fusion protein linker has a length of 1 to 30 amino acids.
  • the fusion protein linker comprises a sequence according to SEQ ID No. 5.
  • the Interleukin-2 is human Interleukin-2 (human IL-2).
  • Interleukin-2 may be produced recombinantly or may be isolated from human tissue, preferably it is produced recombinantly (rIL-2).
  • the Interleukin-2 part comprises a sequence according to SEQ. ID. No. 4, or a functional variant thereof.
  • the fusion protein may be monomeric, or multimeric, e.g. dimeric. Dimeric or other multimeric forms may be formed covalently or non-covalently. E.g. L19(scFv)-IL2 may form non-covalent homodimers.
  • the fusion proteins are preferably produced recombinantly using methods known to the skilled person.
  • prokaryotic or eukaryotic expression systems e.g. yeast or mammalian expression systems, can be used.
  • the combination of the present invention further comprises a molecule binding to B cells, B cell progenitors and/or their cancerous counterpart.
  • the molecule binding to B cells, B cell progenitors and/or their cancerous counterpart is labelled, in particular radioactively labelled.
  • the labelling is a covalent labelling.
  • the labelled molecule binding to B cells, B cell progenitors and/or their cancerous counterpart is a radioactively labelled anti-CD20 antibody.
  • Various radioactive labels are used in medicine.
  • radioactive isotopes for labelling antibodies and proteins are 90 Y, 111 In and 131 I-labelled.
  • the anti-CD20 antibody is labelled with 90 Y, 111 In or 131 I.
  • the radioactively labelled anti-CD20 antibody is selected from Y-90-Ibritumomab-Tiuxetan (Y90-Zevalin® or -Zevalin®) and I-131 tositumomab (Bexxar®).
  • Y-90-Ibritumomab-Tiuxetan and its production is for example disclosed in EP 0 669 836 as Y2B8 (Yttrium-[90]-labeled 2B8-MX-DTPA).
  • the combination of the present invention further comprises a molecule specifically binding to CD20.
  • this molecule is an antibody or antibody fragment, or a fusion protein thereof.
  • anti-CD20 antibodies which exhibit ADCC activity.
  • the anti-CD20 antibody is selected from rituximab, Ocrelizumab, PRO131921, Veltuzumab, Ofatumumab, AME-133, and GA-101.
  • the antibodies specifically binding to CD20 are in full IgG, Fab, (Fab) 2 , scFv, diabody, minibody or small immunoprotein (SIP) format.
  • the anti-CD20 antibody may be monomeric or multimeric, e.g. dimeric. Multimeric antibodies may be homomeric or heteromeric. E.g. a bivalent antibody may be used, wherein one part specifically binds to CD20 and another part binds to another target. Also, the molecule specifically binding to CD20 may comprise further effectors, in particular it may be labelled radioactively. In this embodiment of the present invention, Zevalin® or Bexxar® may be used, as described above.
  • a particularly preferred anti-CD20 antibody is rituximab, in particular Rituxan® (also called MabThera® or IDEC-C2B8).
  • Rituxan® is a genetically engineered chimeric murine/human monoclonal antibody directed against the CD20 antigen found on the surface of normal and malignant B lymphocytes.
  • the antibody is an IgG1 kappa immunoglobulin containing murine light- and heavy-chain variable region sequences and human constant region sequences.
  • rituximab is disclosed e.g. in U.S. Pat. Nos. 5,843,439; 5,776,456 and 5,736,137.
  • the combination comprises rituximab and L19-IL2.
  • the L19 antibody is in scFv format.
  • L19-IL2 as described in Carnemolla et al., Blood. 2002; 99:1659-1665.
  • Another embodiment of the present invention relates to a combination as described above, for use as a medicament.
  • a further embodiment of the present invention relates to a combination as described above, for use as a medicament for treating cancer.
  • the cancer is a lymphoma, preferably a B-cell lymphoma. Most preferred is the use of the combination of the present invention for treating B-cell Non-Hodgkin lymphoma (B-NHL).
  • B-NHL B-cell Non-Hodgkin lymphoma
  • the B-cell lymphoma is refractory or relapsed B-cell lymphoma or a lymphoma resistant to rituximab-monotherapy.
  • the invention further relates to a method of treating cancer, wherein a combination of the present invention is administered to a cancer patient in therapeutically effective amount.
  • the cancer is a lymphoma, preferably a B-cell lymphoma, in particular a NHL.
  • a further embodiment of the present invention relates to a combination as described above, for use as a medicament for treating autoimmune diseases, in particular chronic autoimmune diseases.
  • the autoimmune disease is rheumatoid arthritis, Crohn's disease, colitis ulcerosa or autoimmune hemolytic anemia.
  • the patient can be any mammal, preferably the patient is a human.
  • intravenous e.g. intravenous, subcutaneous or intraperitoneal administration, wherein the intravenous administration is preferred.
  • the fusion protein specifically recognizing EDb fibronectin and the molecule binding to B cells, B cell progenitors and/or their cancerous counterpart may be administered at the same time or at different time points.
  • the combination may be administered once or several times to a patient. Also, it is possible, that one component of the combination is administered once, and the other component is administered several times.
  • rituximab and L19-IL2 are administered as combination therapy, they may be administered to a patient at the same time point, as this allows easier administration schedules.
  • rituximab and L19-IL2 both may be administered i.v. once or twice per day in time intervals ranging from few days up to 3 months. Also, one or more treatment rounds are possible.
  • rituximab may be administered in an amount of about 20 to 500 mg/m 2 , preferably in an amount of about 100 to 400 mg/m 2 , in particular of about 375 mg/m 2 rituximab per administration.
  • rituximab is administered on Day 1 of a 2-, 3-, or 4-week treatment schedule with up to 6-8 treatment cycles (remission induction), although other administration schedules are possible.
  • Therapeutic formulations of the active agents used in accordance with the present invention are prepared for storage by mixing an active agent having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Exemplary anti-CD20 antibody formulations are described in WO 98/56418.
  • This publication describes a liquid multidose formulation comprising 40 mg/mL rituximab, 25 mM acetate, 150 mM trehalose, 0.9% benzyl alcohol, 0.02% polysorbate 20 at pH 5.0 that has a minimum shelf life of two years storage at 2-8° C.
  • Another anti-CD20 formulation of interest comprises 10 mg/mL rituximab in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium citrate dihydrate, 0.7 mg/mL polysorbate 80, and Sterile Water for Injection, pH 6.5.
  • Lyophilized formulations adapted for subcutaneous administration are described in WO 97/04801. Such lyophilized formulations may be reconstituted with a suitable diluent to a high protein concentration and the reconstituted formulation may be administered subcutaneously to the patient to be treated herein.
  • the amount to be administered may vary.
  • the amount of L19-IL2 to be administered per administration is about 1 to 10 ⁇ 10 6 IU/m 2 , in particular about 5 to 50 ⁇ 10 6 IU/m 2 , especially about 10 to 30 ⁇ 10 6 IU/m 2 .
  • the administered amount varies over time; e.g. the amount of rituximab and/or L19-IL2 may be increased or decreased for one or more administration rounds.
  • a maintenance treatment in particular with rituximab or L19-IL2 alone, may follow the combination treatment phase.
  • chemoimmunotherapeutic regimens e.g. R-CHOP
  • Antibody linker is any linker, preferably a peptide linker, which is suitable for linking Vh and V1 domains. Suitable linkers are for example described in Bird et al, Science, 242, 423-426, 1988; Huston et al, PNAS USA, 85, 5879-5883, 1988, EP 0 573 551; EP 0 623679 and EP 0 318554, which documents are introduced by reference.
  • Fusion protein linkers are linkers suitable for linking an antibody or antibody-fragment and a second biologically active protein, preferably the linker is peptidic. Suitable linkers are described in EP 0 573 551; EP 0 623679 and EP 0 318554, which documents are introduced by reference. In particular, suitable linkers are described in EP 0 623679.
  • Specifically binding or “specifically recognizing” as used herein refers to binding to the corresponding target.
  • the binding molecule, antibody, antibody fragment or antibody mimetic binds with an affinity of at least about 1 ⁇ 10 ⁇ 7 M, preferably of at least about 1 ⁇ 10 ⁇ 9 M, and binds to the predetermined target with an affinity that is at least two-fold greater than its affinity for binding to a non-specific target (e.g. BSA, casein) other than the predetermined target or a closely-related target.
  • a non-specific target e.g. BSA, casein
  • Antibody as used herein encompasses full length antibodies, comprising native antibodies, monoclonal antibodies, polyclonal antibodies and multispecific antibodies (e.g., bispecific antibodies), human antibodies, humanized antibodies, chimeric antibodies, and full IgG antibodies, as well as antibody fragments.
  • antibody fragment refers to a portion of a full length antibody, in which a variable region or a functional capability is retained, namely the specific binding to the target.
  • antibody fragments include, but are not limited to, a Fab, Fab′, F(ab′)2, Fd, Fv, scFv and scFv-Fc fragment, a diabody, a linear antibody, small immunoprotein formats, a single-chain antibody, a minibody, a diabody formed from antibody fragments, and multispecific antibodies formed from antibody fragments.
  • Antibody fragments are usually smaller than full antibodies. Thereby, the pharmacokinetics are different and some antibody fragments only consist of one polypeptide chain, which can make production easier.
  • the antibody fragment is in scFv, (scFv) 2 , or small immunoprotein format.
  • the small immunoprotein format can be a format based on a CH3-domain (for example described in U.S. Pat. No. 5,837,821) or ⁇ S 2 CH4-domain of human IgE (for example described in WO 03/076469).
  • mAb refers to an antibody obtained from a population of substantially homogeneous antibodies; that is, the individual antibodies comprising the population that are identical except for naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic determinant, also referred to as an epitope. The modifier “monoclonal” is indicative of a substantially homogeneous population of antibodies directed to the identical epitope and is not to be construed as requiring production of the antibody by any particular method.
  • Monoclonal antibodies can be made by any technique or methodology known in the art; for example, the hybridoma method first described by Koehler et al., 1975, Nature 256:495, or recombinant DNA methods known in the art (see, e.g., U.S. Pat. No. 4,816,567).
  • monoclonal antibodies can also be isolated from phage antibody libraries, using techniques described in Clackson et al., 1991, Nature 352: 624-628, and Marks et al., 1991, J. Mol. Biol. 222: 581-597.
  • the antibodies in a preparation of polyclonal antibodies are typically a heterogeneous population of immunoglobulin isotypes and/or classes and also exhibit a variety of epitope specificity.
  • chimeric antibody as used herein is a type of monoclonal antibody in which a portion of or the complete amino acid sequence in one or more regions or domains of the heavy and/or light chain is identical with, homologous to, or a variant of the corresponding sequence in a monoclonal antibody from another species or belonging to another immunoglobulin class or isotype, or from a consensus sequence.
  • antibody fragments can be generated by enzymatic treatment of a full-length antibody. Papain digestion of antibodies produces two identical antigen-binding fragments called “Fab” fragments, each with a single antigen-binding site, and a residual “Fc” fragment, so called because of its ability to crystallize readily.
  • the Fab fragment also contains the constant domain of the light chain and the CH1 domain of the heavy chain. Pepsin treatment yields a F(ab′)2 fragment that has two antigen-binding sites and is still capable of cross-linking antigen.
  • Fab′ fragments differ from Fab fragments by the presence of a few additional residues at the C-terminus of the CH1 domain, including one or more cysteines from the antibody hinge region.
  • Fab-SH is the designation herein for a Fab′ in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab′)2 antibody fragments are pairs of Fab′ fragments linked by cysteine residues in the hinge region. Other chemical couplings of antibody fragments are also known.
  • “Fv” is a minimum antibody fragment that contains a complete antigen-recognition and binding site consisting of a dimer of one heavy and one light chain variable domain in tight, non-covalent association.
  • the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH VL dimer.
  • the six CDRs confer antigen-binding specificity to the antibody.
  • a “single-chain Fv” or “scFv” antibody fragment is a single chain Fv variant comprising the VH and VL domains of an antibody, in which the domains are present in a single polypeptide chain and which is capable of recognizing and binding antigen.
  • the scFv polypeptide optionally contains a polypeptide linker positioned between the VH and VL domains that enables the scFv to form a desired three-dimensional structure for antigen binding (see, e.g., Pluckthun, 1994, In The Pharmacology of Monoclonal Antibodies, Vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315).
  • diabodies refers to small antibody fragments having two antigen-binding sites. Each fragment contains a heavy chain variable domain (VH) concatenated to a light chain variable domain (VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the linked VH-VL domains are forced to pair with complementary domains of another chain, creating two antigen-binding sites.
  • VH heavy chain variable domain
  • VL light chain variable domain
  • Diabodies are described more fully, for example, in EP 404,097; WO 93/11161; and Hollinger et al., 1993, Proc. Nat. Acad. Sc. USA 90: 6444-6448.
  • a humanized antibody or a humanized antibody fragment includes an immunoglobulin amino acid sequence variant, or fragment thereof, which is capable of binding to a predetermined antigen and which, comprises one or more framework regions (FRs) having substantially the amino acid sequence of a human immunoglobulin and one or more CDRs having substantially the amino acid sequence of a non-human immunoglobulin.
  • This non-human amino acid sequence is referred to herein as an “import” sequence, which is typically taken from an “import” antibody domain, particularly a variable domain.
  • a humanized antibody includes at least the CDRs or HVLs of a non-human antibody, inserted between the FRs of a human heavy or light chain variable domain.
  • “Native antibodies” are defined herein as heterotetrameric glycoproteins, typically of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is covalently linked to a heavy chain by one disulfide bond to form a heterodimer. The heterotetramer is formed by covalent disulfide linkage between the two identical heavy chains of such heterodimers. Although the light and heavy chains are linked together by one disulfide bond, the number of disulfide linkages between the two heavy chains varies by immunoglobulin isotype. Each heavy and light chain also has regularly spaced intrachain disulfide bridges.
  • Each heavy chain has at the amino-terminus a variable domain (VH), followed by three or four constant domains (CH1, CH2, CH3, and CH4), as well as a hinge region between CH1 and CH2.
  • VH variable domain
  • CH1, CH2, CH3, and CH4 constant domains
  • Each light chain has two domains, an amino-terminal variable domain (VL) and a carboxy-terminal constant domain (CL).
  • VL domain associates non-covalently with the VH domain
  • the CL domain is commonly covalently linked to the CH1 domain via a disulfide bond.
  • Particular amino acid residues are believed to form an interface between the light and heavy chain variable domains (Chothia et al., 1985, J. Mol. Biol.
  • variable domains differ extensively in sequence among antibodies and contain residues that are directly involved in the binding and specificity of each particular antibody for its specific antigenic determinant. Hypervariability, both in the light chain and the heavy chain variable domains, is concentrated in three segments known as complementarity determining regions (CDRs) or hypervariable loops (HVLs). CDRs are defined by sequence comparison in Kabat et al., 1991, In: Sequences of Proteins of Immunological Interest, 5th Ed.
  • HVLs are structurally defined according to the three-dimensional structure of the variable domain, as described by Chothia and Lesk, 1987, J. Mol. Biol. 196: 901-917.
  • CDR-L1 is positioned at about residues 24-34, CDR-L2, at about residues 50-56, and CDR-L3, at about residues 89-97 in the light chain variable domain;
  • CDR-H1 is positioned at about residues 31-35, CDR-H2 at about residues 50-65, and CDR-H3 at about residues 95-102 in the heavy chain variable domain.
  • label refers to a detectable compound or composition that is conjugated directly or indirectly to the antibody.
  • the label may itself be detectable (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition that is detectable.
  • fibronectins are the product of the single FN gene, the resulting protein can exist in multiple forms which—apart from posttranslational modifications—arise from alternative splicing of its primary RNA transcript.
  • This polymorphism which leads to as many as 20 different isoforms in human FN, thereby generating FNs with different solubility, cell adhesive and ligand-binding properties, provides cells with the possibility to modify the composition of the extracellular matrix (ECM) in a tissue-specific manner.
  • ECM extracellular matrix
  • ED-B domain is to be understood as the extra-domain B of human fibronectin. It is often referred to as EDb, EIIIB or EDII.
  • Antibody mimetics are understood as binding molecules based on protein frameworks (“scaffolds”) which specifically bind to the target and which are distinct from antibodies and antibody fragments. Such scaffolds are described in Binz et al., 2005, Nat. Biotechnol. 23, 1257-1268. Antibody mimetics specifically binding to ED-B fibronectin are described in Grabulovski et al., J. Biol. Chem., 2007, 282:3196-3204.
  • Interleukin-2 refers to mammalian Interleukin-2, preferably human Interleukin-2 and functional variants thereof.
  • Functional variants of Interleukin-2 are variants of human Interleukin-2 which exhibit at least 10%, but more preferably more than 50%, and even more preferred more than 90% of the activity of native human Interleukin-2.
  • Interleukin-2 activities are activities of Inter-leukin-2 in biochemical assays or in vivo, in particular Interleukin-2 activity can be measured by the effect on proliferation and/or differentiation of activated T and B lymphocytes and of natural killer cells and/or induction of cytotoxic T cell activity and/or NK/lymphokine activated killer (LAK) anti-tumour activity (Meazza R, Marciano S, Sforzini S, et al. Analysis of IL-2 receptor expression and of the bio-logical effects of IL-2 gene transfection in small-cell lung cancer. Br. J. Cancer. 1996; 74: 788-795).
  • LAK NK/lymphokine activated killer
  • functional variants are cystein-125 muteins of Interleukin-2 as described in EP 0109748 and other muteins, including cystein muteins as described in EP136489, in particular serine 125-Interleukin-2.
  • the N-terminus of hIL.2 variants may be altered without significantly affecting the activity, in particular the N-terminal 1-5 amino acids, especially preferred the N-terminal Alanine may be deleted or altered, preferably deleted.
  • the Interleukin-2 may contain altered or deleted post-translational modifications, in particular the glycosylation pattern may be altered or missing. Different or absent glycosylation may be obtained e.g. either by mutating the sequence or by expression of the fusion protein in an appropriate host.
  • Aldesleukin which is approved for metastatic RCC, is unglycosylated des-alanyl-1, serine-125 human interleukine-2 produced in E. coli.
  • FIG. 1 Immunohistochemistry with L19 antibody reveals EDB expression in B-cell lymphoma xenografts. Immunohistochemical staining using the antibody L19, specific to EDB fibronectin, revealed a strong expression of this fibronectin isoform with a prominent vascular pattern of staining in Ramos lymphoma xenografts (left panel). The staining is similar to the staining pattern of L19 in solid tumors, as exemplified with the U87 glioblastoma xenograft (right panel). For negative control, the primary antibody was omitted. Scale bars, 100 ⁇ m.
  • FIG. 2 In vivo localization experiments: ex vivo immunofluorescence (A) and quantitative biodistribution studies (B). The in vivo targeting performance of the L19 antibody was tested in the subcutaneous SCID/Ramos lymphoma model.
  • A Lymphoma-bearing mice were injected with L19-SIP, chemically labeled with the fluorophore Cy3. The figure shows a 2-color fluorescence microscopic image of a lymphoma section 24 h after injection, confirming the antibody localization (red) on tumor vascular structures.
  • An anti-CD31 antibody has been applied ex vivo to outline endothelial cells and was detected with an Alexa Fluor 488 anti-rat IgG antibody (green).
  • FIG. 3 Effect of single-agent L19-IL2, unconjugated IL-2 and rituximab on lymphoma growth.
  • Ramos lymphoma xenografts were injected i.v. with either the vascular targeting L19-IL2 fusion protein ( ⁇ ; 20 ⁇ g), the corresponding dose of untargeted rIL-2 ( ⁇ ; 6.6 ⁇ g), rituximab ( ⁇ ; 200 ⁇ g), or control saline (x) on days 8, 11, 14, and 17 after tumor cell implantation.
  • FIG. 4 Therapeutic effect of L19-IL2 and unconjugated IL-2 in combination with rituximab.
  • SCID mice bearing established s.c. lymphoma xenografts were injected i.v. with either saline (x), 200 ⁇ g rituximab+low dose unconjugated IL-2 ( ⁇ ; 2.2 ⁇ g), 200 ⁇ g rituximab+high dose unconjugated IL-2 ( ⁇ ; 6.6 ⁇ g), 200 ⁇ g rituximab+low dose L19-IL2 ( ⁇ ; 6.6 ⁇ g, corresponding to 2.2 ⁇ g IL-2), or 200 ⁇ g rituximab+high dose L19-IL2 ( ⁇ ; 20 ⁇ g, corresponding to 6.6 ⁇ g IL-2) on days 8, 11, 14, and 17.
  • L19-IL2 in combination with rituximab was highly efficacious, inducing complete remissions in 4 of 5 mice in the low dose as well as in the high dose L19-IL2 group.
  • unconjugated rIL-2 in combination with rituximab did not induce tumor regressions and all tumors continued to grow. All mice with CRs remained tumor-free for a period of at least 42 days.
  • FIG. 5 Therapeutic effect of L19-IL2, IL-2 and rituximab in mono- and combination therapies in the disseminated lymphoma model.
  • SCID mice (n ⁇ 6) were injected i.v. with 2 ⁇ 10 6 Ramos lymphoma cells and treated 8 days later according to the following regimens: untargeted IL-2 (6.6 ⁇ g), L19-IL2 (20 ⁇ g), rituximab (200 ⁇ g), rituximab (200 ⁇ g)+IL-2 (6.6 ⁇ g), rituximab (200 ⁇ g)+L19-IL2 (20 ⁇ g), or control saline (In detail, to model systemic disease, SCID mice were injected i.v.
  • FIG. 6 Target validation in human lymphoma samples. EDB was found to be expressed in neovascular structures of human B-cell lymphoma entities, including the frequent subtypes diffuse large B-cell lymphoma and Burkitt lymphoma. Scale bars, 100 ⁇ m.
  • mice Six- to 8-week-old female CB17/lcr SCID mice were obtained from Charles River Laboratories (Sulzfeld, Germany). All mice were housed in microisolator units and provided with sterile food and water ad libitum throughout the studies.
  • the EBV-negative human B cell lymphoma cell line Ramos 44 was purchased from the American Type Culture Collection (ATCC, Manassas, Va.).
  • Cells were maintained in log-phase growth in RPMI 1640 medium adjusted to contain 2 mM L-glutamine, 10 mM HEPES, 1 mM sodium pyruvate, 4.5 g/L glucose, 1.5 g/L bicarbonate, 10% heat-inactivated fetal bovine serum, 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin.
  • the human follicular lymphoma cell line DoHH-2 was obtained from the German Resource Centre for Biological Material (DSMZ, Braunschweig, Germany).
  • L19 is a vascular targeting antibody directed against the EDB domain of fibronectin.
  • SIP format small immunoprotein
  • L19-IL2 fusion protein The expression, purification and characterization of L19 in SIP format (small immunoprotein) and the L19-IL2 fusion protein have been described previously in Borsi et al. (Int J Cancer. 2002; 102:75-85) and Carnemolla et al. (Blood. 2002; 99:1659-1665).
  • Recombinant human IL-2 Proleukin, 18 ⁇ 10 6 IU
  • was obtained from Prorero Pharma Liestal, Switzerland
  • the chimeric human IgG1 anti-CD20 monoclonal antibody rituximab (MabThera) from Roche (Reinach, Switzerland).
  • cryostat sections of frozen samples were fixed in ice-cold acetone, rehydrated in TBS (50 mmol/L Tris, 100 mmol/L NaCl pH 7.4), and blocked with 20% FCS (Invitrogen, Basel, Switzerland). L19-SIP was added onto the sections in a final concentration of 10 ⁇ g/mL. Bound primary antibody was detected with rabbit anti-human IgE antibody (Dako, Glostrup, Denmark) followed by biotinylated goat anti-rabbit IgG antibody (Biospa, Milan, Italy) and streptavidin-alkaline phosphatase complex (Biospa).
  • L19-SIP was labeled with Cy3-NHS ester, a fluorescent cyanine compound, following the manufacturer's recommendation (Amersham Pharmacia, D Weg, Switzerland).
  • 120 ⁇ g of L19-Cy3 conjugate were injected intravenously (i.v.) into the lateral tail vein of lymphoma-bearing mice.
  • Mice were sacrificed 24 h after injection, and tumors were excised, embedded in cryoembedding compound (Microm, Walldorf, Germany) and stored at ⁇ 80° C. 10 ⁇ m sections were cut, dried at 37° C. for 15 min and fixed with 4% paraformaldehyde for 15 min at room temperature.
  • Rat anti-CD31 antibody (BD Pharmingen) was applied to outline endothelial cells using Alexa Fluor 488 rabbit anti-rat IgG as secondary antibody (Invitrogen). Images were captured on an Axioskop 2 Mot plus microscope equipped with an AxioCam MRc camera (Zeiss).
  • L19-IL2 (6.6 and 20 ⁇ g, corresponding to 2.2 and 6.6 ⁇ g of “free” rIL-2, respectively), or unconjugated rIL-2 (2.2 and 6.6 ⁇ g) were administered in combination with rituximab (200 ⁇ g) by separate i.v. injections on days 8, 11, 14, and 17.
  • mice were treated with equimolar amounts of L19 in SIP (x.x ⁇ g) or IgG (x.x ⁇ g) format, alone or in combination with free rIL-2 (6.6 ⁇ g).
  • Treatment schedule for all agents was every third day for four (Ramos) or three (DoHH-2) injections in total (Q3Dx4 or Q3Dx3, respectively).
  • SCID mice were injected i.v. with 2 ⁇ 10 6 Ramos lymphoma cells resuspended in 200 ⁇ L PBS. Dissemination and growth of B-cell lymphoma was allowed to occur for 8 days before the initiation of therapy. Mice were randomly divided into 6 groups (n ⁇ 6) and injected i.v.
  • mice were monitored daily for the presence of hind-leg paralysis or signs of a deteriorating condition whereupon mice were sacrificed. Onset of paralysis or death were set as end points and survival of mice was recorded for analysis of therapeutic efficacy. Animal experiments using the disseminated lymphoma model were done in accordance with amendment 1 to the project license “Tumor Targeting”.
  • FIG. 2 a shows a 2-color fluorescence microscopic image of a lymphoma section, confirming the antibody localization on tumor vascular structures.
  • mice bearing s.c. implanted Ramos lymphoma xenografts were injected i.v. with radioiodinated preparations of L19-SIP.
  • L19 displayed an accumulation in the lymphoma tissue with absolute tumor uptake values of 4.7% ID/g 24 h after injection, but only moderate tumor-to-blood ratios of 2.1 at this time point (tumor-to-organ ratios ranging from 2.7 to 7.1).
  • tumor-to-blood ratios ranging from 2.7 to 7.1
  • the antibody was cleared from normal organs more rapidly, resulting in increased tumor-to-blood (4.8) and tumor-to-organ ratios (up to 17.3) and demonstrating a specific accumulation and retention of the antibody at the tumor site.
  • equimolar amounts of naked L19 in SIP or IgG format were therapeutically inactive when administered alone or in combination with free rIL-2, further reinforcing the concept that the therapeutic activity of L19-IL2 relied on the targeted delivery of the cytokine at the lymphoma site ( FIG. 7 ).
  • a combination therapy experiment was conducted according to the following scheme ( ⁇ 5 mice per group): 200 ⁇ g rituximab+2.2 ⁇ g unconjugated rIL-2 (“low dose”), 200 ⁇ g rituximab+6.6 ⁇ g unconjugated rIL-2 (“high dose”), 200 ⁇ g rituximab+6.6 ⁇ g L19-IL2 (“low dose”, corresponding to 2.2 ⁇ g rIL-2), 200 ⁇ g rituximab+20 ⁇ g L19-IL2 (“high dose”, corresponding to 6.6 ⁇ g rIL-2), or control saline.
  • injections were started on day 8 after tumor cell inoculation when palpable s.c. xenografts have developed and repeated every third day for 4 injections in total.
  • rituximab in combination with unconjugated rIL-2 caused significant tumor growth delay as compared to controls (rIL-2 low and high dose vs. saline: P ⁇ 0.001).
  • the combination of rituximab with the L19-IL2 fusion protein displayed a strikingly higher anti-lymphoma activity and induced complete eradications of established Ramos lymphomas in 4 of 5 mice in the high dose L19-IL2 group (L19-IL2 high dose vs.
  • rIL-2 low dose P ⁇ 0.00001
  • inducing CRs in 4 of 5 cases after 4 cycles of therapy whereas even a three-fold higher dose of the non-targeted cytokine combined with rituximab was only able to retard tumor growth (L19-IL2 low dose vs. rIL-2 high dose : P ⁇ 0.001).
  • animals having achieved a CR in the low dose L19-IL2 group eventually relapsed after remission duration of 21, 48, 50, and 81 days, respectively
  • all CRs in the higher dose L19-IL2 group were durable and all mice remained tumor-free for an observation period of one year. Two mice (one in the low dose and one in the high dose L19-IL2 group) did not achieve a CR but the tumor mass was reduced to less than 20 mm 3 .
  • L19-IL2 was effective as a single-agent in inhibiting lymphoma growth (P ⁇ 0.0001), yet without inducing tumor regressions, while the sum of its components (naked L19 and rIL-2) in equivalent doses showed no significant therapeutic activity.
  • SCID/Ramos lymphoma SCID mice inoculated i.v. with lymphoma cells regularly develop paralysis of the hind-legs, resulting from lymphoma manifestations in the spinal cord and indicating the terminal phase of the disease.
  • Ramos cells resulted in the development of hind-leg paralysis by day 26 in all cases in a pilot experiment, indicating an engraftment rate of 100% (data not shown).
  • the Kaplan-Meier survival curve is shown in FIG. 5 .
  • day 25 all saline-treated control mice succumbed to disseminated disease with a median survival time of 24 days.
  • the corresponding dose of single-agent L19-IL2 (20 ⁇ g) extended the median survival time to 29 days (P ⁇ 0.010, compared to non-targeted rIL-2) and was equally efficient as rituximab in delaying the appearance of the disease compared to saline-treated controls (median survival 29 and 30 days, respectively, vs. 24 days; P ⁇ 0.001 for both agents).
  • mice Two additional mice had to be sacrificed on day 73 and 79, respectively, due to lymphoma development in an axillary lymph node, yet without hind-leg paralysis. The three remaining mice were still disease-free 310 days after tumor cell inoculation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Physics & Mathematics (AREA)
  • Optics & Photonics (AREA)
  • Rheumatology (AREA)
  • Endocrinology (AREA)
  • Biomedical Technology (AREA)
  • Toxicology (AREA)
  • Transplantation (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Diabetes (AREA)
US12/863,417 2008-01-17 2008-11-08 Combination of an anti-edb fibronectin antibody-il-2 fusion protein, and a molecule binding to b cells, b cell progenitors and /or their cancerous counterpart Abandoned US20100310454A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP08075044.1 2008-01-17
EP08075044A EP2085095B1 (fr) 2008-01-17 2008-01-17 Combinaison de protéine de fusion anticorps de fibronectine anti-EDb-IL-2, et molécule se liant aux lymphocytes B, progéniteurs de lymphocytes B et/ou leur contrepartie cancéreuse
PCT/EP2008/009441 WO2009089858A1 (fr) 2008-01-17 2008-11-08 Association d'une protéine de fusion il-2-anticorps anti-edb de fibronectine, et d'une molécule se liant aux lymphocytes b, aux cellules progénitrices des lymphocytes b et/ou leur contrepartie cancéreuse

Publications (1)

Publication Number Publication Date
US20100310454A1 true US20100310454A1 (en) 2010-12-09

Family

ID=39427510

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/863,417 Abandoned US20100310454A1 (en) 2008-01-17 2008-11-08 Combination of an anti-edb fibronectin antibody-il-2 fusion protein, and a molecule binding to b cells, b cell progenitors and /or their cancerous counterpart
US12/270,174 Expired - Fee Related US8796426B2 (en) 2008-01-17 2008-11-13 Combination of an anti-EDb fibronectin antibody-IL-2 fusion protein, and a molecule binding to B cells, B cell progenitors and/or their cancerous counterpart
US14/306,992 Expired - Fee Related US9289470B2 (en) 2008-01-17 2014-06-17 Combination of an anti-EDb fibronectin antibody-IL-2 fusion protein, and a molecule binding to B cells, B cell progenitors and/or their cancerous counterpart

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/270,174 Expired - Fee Related US8796426B2 (en) 2008-01-17 2008-11-13 Combination of an anti-EDb fibronectin antibody-IL-2 fusion protein, and a molecule binding to B cells, B cell progenitors and/or their cancerous counterpart
US14/306,992 Expired - Fee Related US9289470B2 (en) 2008-01-17 2014-06-17 Combination of an anti-EDb fibronectin antibody-IL-2 fusion protein, and a molecule binding to B cells, B cell progenitors and/or their cancerous counterpart

Country Status (13)

Country Link
US (3) US20100310454A1 (fr)
EP (1) EP2085095B1 (fr)
JP (1) JP5140164B2 (fr)
KR (1) KR20100113572A (fr)
CN (1) CN101969980A (fr)
AT (1) ATE548052T1 (fr)
AU (1) AU2008347450B2 (fr)
BR (1) BRPI0822044A2 (fr)
CA (1) CA2711902C (fr)
ES (1) ES2382058T3 (fr)
MX (1) MX2010007716A (fr)
RU (1) RU2484845C2 (fr)
WO (1) WO2009089858A1 (fr)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE548052T1 (de) 2008-01-17 2012-03-15 Philogen Spa Kombination aus einem anti-edb-fibronectin- antikörper-il-2-fusionsprotein und einem b-zellen bindenden molekül, b-zellen-vorläufern und/oder deren krebserregendem gegenspieler
US20110318302A1 (en) * 2009-01-07 2011-12-29 Kathrin Schwager Cancer Treatment
AR078161A1 (es) 2009-09-11 2011-10-19 Hoffmann La Roche Formulaciones farmaceuticas muy concentradas de un anticuerpo anti cd20. uso de la formulacion. metodo de tratamiento.
DK3489255T3 (da) 2011-02-10 2021-08-23 Roche Glycart Ag Muterede interleukin-2-polypeptider
EP2672999A2 (fr) * 2011-02-10 2013-12-18 Roche Glycart AG Immunothérapie améliorée
EA201892619A1 (ru) 2011-04-29 2019-04-30 Роше Гликарт Аг Иммуноконъюгаты, содержащие мутантные полипептиды интерлейкина-2
CN108430511B (zh) * 2015-12-21 2021-06-04 合肥立方制药股份有限公司 一种药物设计方法和获得的药物及其应用
GB201621806D0 (en) 2016-12-21 2017-02-01 Philogen Spa Immunocytokines with progressive activation mechanism
WO2020070150A1 (fr) 2018-10-02 2020-04-09 Philogen S.P.A Immunoconjugués d'il2
EP3660039A1 (fr) 2018-11-30 2020-06-03 Philogen S.p.A. Immunoconjugués d'il2
WO2020249757A1 (fr) 2019-06-14 2020-12-17 Philogen S.P.A Immunoconjugués comprenant un dianticorps à chaîne unique et de l'interleukine-15 ou l'interleukine-15 et un domaine sushi du récepteur alpha de l'interleukine-15
KR102614063B1 (ko) 2020-12-24 2023-12-19 대화제약 주식회사 엑스트라도메인 b 피브로넥틴에 특이적으로 결합하는 신규한 항체
WO2023180409A1 (fr) * 2022-03-23 2023-09-28 Philogen S.P.A Preparation d'immunoconjugués d'il2
WO2024047237A1 (fr) 2022-09-01 2024-03-07 Philogen S.P.A. Polythérapie par tnf alpha et interleukine-2 pour le traitement du cancer de la peau hors mélanome

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5243029A (en) * 1986-04-09 1993-09-07 Fred Hutchinson Cancer Research Center Oncofetal structure of fibronectin
US20030045681A1 (en) * 1998-05-11 2003-03-06 Anthony J. Zelano Specific binding molecules for scintigraphy, conjugates containing them and therapeutic method for treatment of angiogenesis
US20030176663A1 (en) * 1998-05-11 2003-09-18 Eidgenossische Technische Hochscule Specific binding molecules for scintigraphy
US20040013640A1 (en) * 2000-02-24 2004-01-22 Luciano Zardi Compositions and methods for treatment of angiogenesis in pathological lesions
US20050074401A1 (en) * 2003-09-10 2005-04-07 Laura Borsi Selective targeting of tumor vasculature using radiolabelled antibody molecules
US20060251617A1 (en) * 2005-02-15 2006-11-09 Chiron Corporation Methods for treating lymphomas
US7273924B1 (en) * 1996-05-24 2007-09-25 Philogen S.P.A. Antibodies to the ED-B domain of fibronectin, their construction and uses
US20090208410A1 (en) * 2007-09-17 2009-08-20 Dietmar Berndorff Ed-b fibronectin as stratification marker for anti-tumor drugs

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IE56026B1 (en) 1982-10-19 1991-03-27 Cetus Corp Cysteine-depleted muteins of biologically active proteins
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
WO1985000817A1 (fr) 1983-08-10 1985-02-28 Amgen Expression microbienne de l'interleukine ii
JPH02500329A (ja) 1987-05-21 1990-02-08 クリエイテイブ・バイオマリキユールズ・インコーポレーテツド ターゲット化多機能蛋白質
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
DE69233068T2 (de) 1991-02-27 2004-01-22 Micromet Ag Serin-reiche peptidlinker
JPH07501451A (ja) 1991-11-25 1995-02-16 エンゾン・インコーポレイテッド 多価抗原結合タンパク質
CA2126967A1 (fr) 1992-11-04 1994-05-11 Anna M. Wu Nouvelle construction d'anticorps
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
ES2091684T3 (es) 1992-11-13 1996-11-01 Idec Pharma Corp Aplicacion terapeutica de anticuerpos quimericos y radiomarcados contra el antigeno de diferenciacion restringida de los linfocitos b humanos para el tratamiento del linfoma de las celulas b.
ES2435462T3 (es) 1995-07-27 2013-12-19 Genentech, Inc. Formulación de proteína liofilizada isotónica estable
AU740284B2 (en) 1997-06-13 2001-11-01 Genentech Inc. Stabilized antibody formulation
TWI259837B (en) 1998-05-11 2006-08-11 Eidgenossische Tech Hochscule Specific binding molecules for scintigraphy, conjugates containing them and therapeutic method for treatment of angiogenesis
JP2004529848A (ja) * 2000-09-07 2004-09-30 シエーリング アクチエンゲゼルシャフト EDb−フィブロネクチンドメイン(II)の受容体
CA2469045A1 (fr) * 2001-12-07 2003-06-19 Chiron Corporation Methodes therapeutiques de traitement d'un lymphome non hodgkinien
PT1483297E (pt) 2002-03-11 2010-03-29 Philogen Spa Anticorpos derivados de anti-ed-b l19 e direccionamento para a vasculatura tumoral
EP1842553A1 (fr) * 2006-04-07 2007-10-10 Bayer Schering Pharma Aktiengesellschaft Combinaison de la protéine de fusion entre un anticorps contre le domaine EDb de la fibronectine et interleukine 2 avec une autre petite molecule
ATE548052T1 (de) 2008-01-17 2012-03-15 Philogen Spa Kombination aus einem anti-edb-fibronectin- antikörper-il-2-fusionsprotein und einem b-zellen bindenden molekül, b-zellen-vorläufern und/oder deren krebserregendem gegenspieler

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5243029A (en) * 1986-04-09 1993-09-07 Fred Hutchinson Cancer Research Center Oncofetal structure of fibronectin
US7273924B1 (en) * 1996-05-24 2007-09-25 Philogen S.P.A. Antibodies to the ED-B domain of fibronectin, their construction and uses
US20080274099A1 (en) * 1996-05-24 2008-11-06 Dario Neri Antibodies of the ed-b domain of fibronectin, their construction and uses
US20030045681A1 (en) * 1998-05-11 2003-03-06 Anthony J. Zelano Specific binding molecules for scintigraphy, conjugates containing them and therapeutic method for treatment of angiogenesis
US20030176663A1 (en) * 1998-05-11 2003-09-18 Eidgenossische Technische Hochscule Specific binding molecules for scintigraphy
US20040013640A1 (en) * 2000-02-24 2004-01-22 Luciano Zardi Compositions and methods for treatment of angiogenesis in pathological lesions
US20050074401A1 (en) * 2003-09-10 2005-04-07 Laura Borsi Selective targeting of tumor vasculature using radiolabelled antibody molecules
US20090214423A1 (en) * 2003-09-10 2009-08-27 Laura Borsi Selective targeting of tumor vasculature using radiolabelled antibody molecules
US20060251617A1 (en) * 2005-02-15 2006-11-09 Chiron Corporation Methods for treating lymphomas
US20090208410A1 (en) * 2007-09-17 2009-08-20 Dietmar Berndorff Ed-b fibronectin as stratification marker for anti-tumor drugs

Also Published As

Publication number Publication date
JP5140164B2 (ja) 2013-02-06
MX2010007716A (es) 2010-08-04
RU2484845C2 (ru) 2013-06-20
US20140294723A1 (en) 2014-10-02
BRPI0822044A2 (pt) 2015-07-28
ATE548052T1 (de) 2012-03-15
CN101969980A (zh) 2011-02-09
AU2008347450B2 (en) 2013-07-25
ES2382058T3 (es) 2012-06-04
AU2008347450A1 (en) 2009-07-23
US20090202473A1 (en) 2009-08-13
EP2085095A1 (fr) 2009-08-05
JP2011509953A (ja) 2011-03-31
KR20100113572A (ko) 2010-10-21
WO2009089858A1 (fr) 2009-07-23
US9289470B2 (en) 2016-03-22
RU2010134360A (ru) 2012-02-27
US8796426B2 (en) 2014-08-05
CA2711902A1 (fr) 2009-07-23
CA2711902C (fr) 2017-07-18
EP2085095B1 (fr) 2012-03-07

Similar Documents

Publication Publication Date Title
US9289470B2 (en) Combination of an anti-EDb fibronectin antibody-IL-2 fusion protein, and a molecule binding to B cells, B cell progenitors and/or their cancerous counterpart
EP2289551A1 (fr) Immunothérapie de troubles auto-immunitaires utilisant des anticorps qui ciblent les lymphocytes B
US20200121588A1 (en) Method of Providing Subcutaneous Administration of Anti-CD38 Antibodies
KR20070100346A (ko) 크립토 결합 분자
JP2021525735A (ja) 抗cd37免疫コンジュゲート投薬レジメン
JP2019515904A (ja) 新血管新生に関連する障害を治療するための組成物および方法
ED Chamuleau et al. Monoclonal antibody therapy in haematological malignancies
CN111936170A (zh) 降低抗cd30抗体药物缀合物疗法的副作用的方法
Martin et al. Phase I to III Trials of Anti–B Cell Therapy in Non–Hodgkin's Lymphoma
Witkowska et al. Development of Anti-CD20 antigen-targeting therapies for B-cell lymphoproliferative malignancies-the state of the art
CN111526893A (zh) 减少抗cd30抗体药物偶联物疗法的副作用的方法
JP2016527200A (ja) Cd37抗体とクロラムブシルの併用
KR20220146488A (ko) 항-cd30 항체-약물 컨쥬게이트 및 비-호지킨 림프종의 치료를 위한 그의 용도
WO2010079161A1 (fr) Agent de déplétion des lymphocytes b pour le traitement de l'athérosclérose
Boyle et al. Ongoing development of monoclonal antibodies and antibody drug-conjugates in lymphoma
CN114728046A (zh) 联合抗cd30 adc、抗pd-1和化疗以治疗血液癌症
Satwani et al. Antibody-Targeted Therapy for Children, Adolescents and Young Adults with Hematological Malignancies
Candelaria Advances in the diagnosisand control of lymphomas
Davies et al. Epratuzumab
WO2017087843A1 (fr) Procédés de traitement de maladies prolifératives à l'aide de molécules de fusion d'anticorps et d'interféron obtenues par génie génétique

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION