US20110165154A1 - Compositions and methods using anti-cs1 antibodies to treat multiple myeloma - Google Patents

Compositions and methods using anti-cs1 antibodies to treat multiple myeloma Download PDF

Info

Publication number
US20110165154A1
US20110165154A1 US12/903,051 US90305110A US2011165154A1 US 20110165154 A1 US20110165154 A1 US 20110165154A1 US 90305110 A US90305110 A US 90305110A US 2011165154 A1 US2011165154 A1 US 2011165154A1
Authority
US
United States
Prior art keywords
agents
pharmaceutical composition
huluc63
administration
dexamethasone
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
US12/903,051
Inventor
Daniel Afar
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.)
AbbVie Biotherapeutics Inc
Original Assignee
Facet Biotech Corp
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 Facet Biotech Corp filed Critical Facet Biotech Corp
Priority to US12/903,051 priority Critical patent/US20110165154A1/en
Assigned to ABBOTT BIOTHERAPEUTICS CORP. reassignment ABBOTT BIOTHERAPEUTICS CORP. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: FACET BIOTECH CORPORATION
Publication of US20110165154A1 publication Critical patent/US20110165154A1/en
Priority to US15/091,977 priority patent/US20160206734A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/475Quinolines; Isoquinolines having an indole ring, e.g. yohimbine, reserpine, strychnine, vinblastine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule

Definitions

  • MM Multiple myeloma
  • the terms multiple myeloma and myeloma are used interchangeably to refer to the same condition.
  • the myeloma tumor, its products, and the host response to it result in a number of organ dysfunctions and symptoms of bone pain or fracture, renal failure, susceptibility to infection, anemia, hypocalcemia, and occasionally clotting abnormalities, neurologic symptoms and vascular manifestations of hyperviscosity. See D. Longo, in Harrison's Principles of Internal Medicine 14th Edition, p. 713 (McGraw-Hill, New York, 1998). No effective long-term treatment currently exists for MM.
  • MM malignant disease of plasma cells, manifested as hyperproteinemia, anemia, renal dysfunction, bone lesions, and immunodeficiency. MM is difficult to diagnose early because there may be no symptoms in the early stage. The disease has a progressive course with a median duration of survival of six months when no treatment is given. Systemic chemotherapy is the main treatment, and the current median of survival with chemotherapy is about three years, however fewer than 5% live longer than 10 years (See Anderson, K. et al., Annual Meeting Report 1999. Recent Advances in the Biology and Treatment of Multiple Myeloma (1999)).
  • Anti-CS1 antibodies that can be used in the methods and compositions are described in U.S. Patent Publication Nos. 2005/0025763 and 2006/0024296, the contents of which are incorporated herein by reference.
  • the anti-CS1 antibodies target CS1 (CD2-subsetl), which is also known as SLAMF7, CRACC, 19A, APEX-1, and FOAP12 (Genbank Accession Number NM — 021181.3).
  • CS1 is a glycoprotein that is highly expressed in bone marrow samples from patients diagnosed with MM.
  • anti-CS1 antibodies exhibit significant anti-myeloma activity (see, e.g., U.S. Patent Publication Nos. 2005/0025763 and 2006/0024296, the contents of which are incorporated herein by reference).
  • HuLuc63 effectively mediates lysis of myeloma cells via antibody dependent cellular cytotoxicity (ADCC) (see, e.g., U.S. Patent Publication No. 2005/0025763, the content of which is incorporated herein by reference).
  • ADCC antibody dependent cellular cytotoxicity
  • HuLuc63 significantly reduced tumor mass by more than 50% (see, e.g., U.S. Patent Publication No. 2005/0025763, the content of which is incorporated herein by reference).
  • the present disclosure relates to compositions and methods for treating patients diagnosed with Monoclonal Gammopathy of Undetermined Significance (MGUS), smoldering myeloma, asymptomatic MM, and symptomatic MM, ranging from newly diagnosed to late stage relapsed/refractory.
  • the methods relate to the administration of a pharmaceutical composition comprising an anti-CS1 antibody in combination with one or more therapeutic agents.
  • Anti-CS1 antibodies are typically administered intravenously at doses ranging from 0.5 to 20 mg/kg, from once a week to once a month.
  • One or more therapeutic agents can be administered concurrently, prior to, or following administration of an anti-CS1 antibody.
  • the agents can be administered separately or combined in a cocktail and administered together as a single composition.
  • the composition(s) can be administered by any means known in the art.
  • administration of the pharmaceutical compositions described herein increases the sensitivity of multiple myeloma cells to a therapeutic agent.
  • inclusion of an anti-CS1 antibody, such as HuLuc63 enhances the activity of therapeutic agents, such that lower doses can be used in the compositions and methods described herein.
  • administration of the pharmaceutical compositions described herein elicits at least one of the beneficial responses as defined by the European Group for Blood and Marrow transplantation (EBMT).
  • EBMT European Group for Blood and Marrow transplantation
  • administration of the pharmaceutical compositions described herein can result in a complete response, partial response, minimal response, no change, or plateau.
  • FIG. 1 depicts the anti-tumor activity of HuLuc63 alone and in combination with dexamethasone in an in vivo mouse multiple myeloma xenograft model
  • FIG. 2 depicts the anti-tumor activity of HuLuc63 alone, thalidomide alone, and HuLuc63 in combination with thalidomide in an in vivo mouse multiple myeloma xenograft model;
  • FIG. 3 depicts the anti-tumor activity of HuLuc63 alone, thalidomide/dexamethasone, and HuLuc63 in combination with thalidomide and dexamethasone in an in vivo mouse multiple myeloma xenograft model;
  • FIG. 4 depicts the anti-tumor activity of HuLuc63 alone and in combination with bevacizumab in an in vivo mouse multiple myeloma xenograft model.
  • compositions described herein combine anti-CS1 antibodies with one or more therapeutic agents at specific doses to potentiate or complement the anti-myeloma activities of the other.
  • suitable anti-CS1 antibodies include, but are not limited to, isolated antibodies that bind one or more of the three epitope clusters identified on CS1 and monoclonal antibodies produced by the hybridoma cell lines: Luc2, Luc3, Luc15, Luc22, Luc23, Luc29, Luc32, Luc34, Luc35, Luc37, Luc38, Luc39, Luc56, Luc60, Luc63, Luc69, LucX.1, LucX.2 or Luc90.
  • suitable anti-CS1 antibodies include isolated antibodies that bind one or more of the three epitope clusters identified on CS1 (SEQ ID NO: 1, Table 1 below; see, e.g., U.S. Patent Publication No. 2006/0024296, the content of which is incorporated herein by reference). As disclosed in U.S. Patent Publication No. 2006/0024296 and shown below in Table 1, the CS1 antibody binding sites have been grouped into 3 epitope clusters:
  • the pharmaceutical compositions include the anti-CS1 antibody HuLuc63.
  • HuLuc63 is a humanized recombinant monoclonal IgG1 antibody directed to human CS1.
  • the amino acid sequence for the heavy chain variable region (SEQ ID NO: 5) and the light chain variable region (SEQ ID NO: 6) for HuLuc63 is disclosed in U.S. Patent Publication No. 2005/0025763, the content of which is incorporated herein by reference, and in Table 1.
  • the synergistic effect permits one or more therapeutic agents to be administered in combination with one or more anti-CS1 antibodies at a reduced dosage, while retaining efficacy.
  • use of the compositions and methods described herein can reduce the deleterious side effects observed in conventional and novel treatment regimens used to treat MM when these agents are administered at their recommended dosages.
  • the synergistic effect permits one or more therapeutic agents to be administered in combination with one or more anti-CS1 antibodies at the approved dosage, but with greater than the expected efficacy.
  • compositions can be administered for the treatment of Monoclonal Gammopathy of Undetermined Significance (MGUS), smoldering myeloma, asymptomatic MM, and symptomatic MM, ranging from newly diagnosed to late stage relapsed/refractory.
  • MGUS Monoclonal Gammopathy of Undetermined Significance
  • smoldering myeloma asymptomatic MM
  • symptomatic MM ranging from newly diagnosed to late stage relapsed/refractory.
  • administration of the compositions results in a reduction in M-protein in serum or urine such that a plateau, no change, minimal, partial or complete response is observed as defined by the European Group for Blood and Marrow transplantation (EBMT).
  • EBMT European Group for Blood and Marrow transplantation
  • compositions that are beneficial in reducing tumor mass and/or regressing tumor growth, in patients diagnosed with multiple myeloma.
  • the pharmaceutical compositions can be used to treat other diseases characterized by the presence of monoclonal protein (M-protein, paraprotein) in the serum or urine.
  • M-protein monoclonal protein
  • an anti-CS1 antibody can be provided in a first pharmaceutical composition, and a therapeutic agent provided in a second composition.
  • an anti-CS1 antibody can be provided in a first pharmaceutical composition
  • one therapeutic agent can be provided in a second composition
  • the other therapeutic agent can be provided in a third composition.
  • an anti-CS1 antibody can be provided in one pharmaceutical composition and the therapeutic agents can be combined and provided in a second pharmaceutical composition.
  • one composition, comprising an anti-CS1 antibody and one or more therapeutic agents can be provided.
  • an anti-CS1 antibody is present in a pharmaceutical composition at a concentration sufficient to permit intravenous administration at 0.5 mg/kg to 20 mg/kg.
  • the concentration of an anti-CS1 antibody suitable for use in the compositions and methods described herein includes, but is not limited to, at least about 0.5 mg/kg, at least about 0.75 mg/kg, at least about 1 mg/kg, at least about 2 mg/kg, at least about 2.5 mg/kg, at least about 3 mg/kg, at least about 4 mg/kg, at least about 5 mg/kg, at least about 6 mg/kg, at least about 7 mg/kg, at least about 8 mg/kg, at least about 9 mg/kg, at least about 10 mg/kg, at least about 11 mg/kg, at least about 12 mg/kg, at least about 13 mg/kg, at least about 14 mg/kg, at least about 15 mg/kg, at least about 16 mg/kg, at least about 17 mg/kg, at least about 18 mg/kg, at least about 19 mg/kg, and at least about 15 mg/kg,
  • the anti-CS1 antibodies can be administered in single or multiple dose regimens. Generally, an anti-CS1 antibody is administered over a period of time from about 1 to about 24 hours, but is typically administered over a period of about 1 to 2 hours. Dosages can be repeated from about 1 to about 4 weeks or more, for a total of 4 or more doses. Typically, dosages are repeated once every week, once every other week, or once a month for a minimum of 4 doses to a maximum of 52 doses.
  • Determination of the effective dosage, total number of doses, and length of treatment with an anti-CS1 antibody is well within the capabilities of those skilled in the art, and can be determined using a standard dose escalation study to identify the maximum tolerated dose (MTh) (see, e.g., Richardson et al., 2002, Blood, 100(9):3063-3067, the content of which is incorporated herein by reference).
  • MTh maximum tolerated dose
  • one or more therapeutic agents are administered in combination with an anti-CS1 antibody.
  • the agents can be administered concurrently, prior to, or following administration of an anti-CS1 antibody.
  • an anti-CS1 antibody is administered prior to the administration of the therapeutic agents.
  • the anti-CS1 antibody can be administered approximately 0 to 60 days prior to the administration of the therapeutic agents.
  • an anti-CS1 antibody such as HuLuc63, is administered from about 30 minutes to about 1 hour prior to the administration of the therapeutic agents, or from about 1 hour to about 2 hours prior to the administration of the therapeutic agents, or from about 2 hours to about 4 hours prior to the administration of the therapeutic agents, or from about 4 hours to about 6 hours prior to the administration of the therapeutic agents, or from about 6 hours to about 8 hours prior to the administration of the therapeutic agents, or from about 8 hours to about 16 hours prior to the administration of the therapeutic agents, or from about 16 hours to 1 day prior to the administration of the therapeutic agents, or from about 1 to 5 days prior to the administration of the therapeutic agents, or from about 5 to 10 days prior to the administration of the therapeutic agents, or from about 10 to 15 days prior to the administration of the therapeutic agents, or from about 15 to 20 days prior to the administration of the therapeutic agents.
  • the anti-CS1 antibody is administered concurrently with the administration of the therapeutic agents.
  • an anti-CS1 antibody is administered following the administration of the therapeutic agents.
  • an anti-CS1 antibody such as HuLuc63
  • HuLuc63 can be administered approximately 0 to 60 days after the administration of the therapeutic agents.
  • HuLuc63 is administered from about 30 minutes to about 1 hour following the administration of the therapeutic agents, or from about 1 hour to about 2 hours following the administration of the therapeutic agents, or from about 2 hours to about 4 hours following the administration of the therapeutic agents, or from about 4 hours to about 6 hours following the administration of the therapeutic agents, or from about 6 hours to about 8 hours following the administration of the therapeutic agents, or from about 8 hours to about 16 hours following the administration of the therapeutic agents, or from about 16 hours to 1 day following the administration of the therapeutic agents, or from about 1 to 5 days following the administration of the therapeutic agents, or from about 5 to 10 days following the administration of the therapeutic agents, or from about 10 to 15 days following the administration of the therapeutic agents, or from about 15 to 20 days following the administration of the therapeutic agents, or from about 20 to 30 days following the administration of the therapeutic agents
  • Therapeutic agents that can be used in combination with the anti-CS1 antibodies described herein include, but are not limited to, targeted agents, conventional chemotherapy agents, hormonal therapy agents, and supportive care agents.
  • One or more therapeutic agents from the different classes e.g., targeted, conventional chemotherapeutic, hormonal, and supportive care, and/or subclasses can be combined in the compositions described herein.
  • the various classes described herein can be further divided into subclasses.
  • targeted agents can be separated into a number of different subclasses depending on their mechanism of action. As will be apparent to those of skill in the art, the agents can have more than one mechanism of action, and thus, could be classified into one or more subclasses.
  • the following subclasses have been identified: anti-angiogenic, inhibitors of growth factor signaling, immunomodulators, inhibitors of protein synthesis, folding and/or degradation, inhibitors of gene expression, pro-apoptotic agents, agents that inhibit signal transduction and agents with “other” mechanisms of action.
  • the mechanism of action for agents falling into the “other” subclass is unknown or poorly characterized.
  • targeted agents such as bevacizumab, sutinib, sorafenib, 2-methoxyestradiol or 2ME2, finasunate, PTK787, vandetanib, aflibercept, volociximab, etaracizumab (MEDI-522), cilengitide, erlotinib, cetuximab, panitumumab, gefitinib, trastuzumab, TKI258, CP-751,871, atacicept, rituximab, alemtuzumab, aldesleukine, atlizumab, tocilizumab, temsirolimus, everolimus, NPI-1387, MLNM3897, HCD122, SGN-40, HLL1, huN901-DM1, atiprimod, natalizumab, bortezomib, carfilzomib, NPI-00
  • conventional chemotherapy agents such as alklyating agents (e.g., oxaliplatin, carboplatin, cisplatin, cyclophosphamide, melphalan, ifosfamide, uramustine, chlorambucil, carmustine, mechloethamine, thiotepa, busulfan, temozolomide, dacarbazine), anti-metabolic agents (e.g., gemcitabine, cytosine arabinoside, Ara-C, capecitabine, 5FU (5-fluorouracil), azathioprine, mercaptopurine (6-MP), 6-thioguanine, aminopterin, pemetrexed, methotrexate), plant alkaloid and terpenoids (e.g., docetaxel, paclitaxel, vincristine, vinblastin, vinorelbine, vindesine, etoposide, VP-16, teniposide
  • hormonal agents such as anastrozole, letrozole, goserelin, tamoxifen, dexamethasone, prednisone, and prednisilone can be combined with an anti-CS1 antibody, such as HuLuc63 and used to treat MM.
  • an anti-CS1 antibody such as HuLuc63
  • supportive care agents such as pamidronate, zoledonic acid, ibandronate, gallium nitrate, denosumab, darbepotin alpha, epoetin alpha, eltrombopag, and pegfilgrastim can be combined with an anti-CS1 antibody, such as HuLuc63 and used to treat MM.
  • an anti-CS1 antibody such as HuLuc63
  • the therapeutic agents can be administered in any manner found appropriate by a clinician and are typically provided in generally accepted efficacious dose ranges, such as those described in the Physician Desk Reference, 56th Ed. (2002), Publisher Medical Economics, New Jersey.
  • a standard dose escalation study can be performed to identify the maximum tolerated dose (MTD) (see, e.g., Richardson, et al. 2002, Blood, 100(9):3063-3067, the content of which is incorporated herein by reference).
  • doses less than the generally accepted efficacious dose of a therapeutic agent can be used.
  • the composition comprises a dosage that is less than about 10% to 75% of the generally accepted efficacious dose range.
  • at least about 10% or less of the generally accepted efficacious dose range is used, at least about 15% or less, at least about 25%, at least about 30% or less, at least about 40% or less, at least about 50% or less, at least about 60% or less, at least about 75% or less, and at least about 90%.
  • the therapeutic agents can be administered singly or sequentially, or in a cocktail with other therapeutic agents, as described below.
  • the therapeutic agents can be administered orally, intravenously, systemically by injection intramuscularly, subcutaneously, intrathecally or intraperitoneally.
  • the therapeutic agents provided in the pharmaceutical composition(s) are selected from the group consisting of dexamethasone, thalidomide, vincristine, carmustine (BCNU), melphalan, cyclophosphamide, prednisone, doxorubicin, cisplatin, etoposide, bortezomib, lenalidomide, ara-C, and/or combinations thereof.
  • the pharmaceutical composition does not comprise bortezomib and/or lenalidomide.
  • two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of dexamethasone.
  • At least two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of dexamethasone and thalidomide.
  • dexamethasone and thalidomide are provided separately, such that a total of three pharmaceutical compositions are provided: a first comprising an anti-CS1 antibody such as HuLuc63, a second comprising dexamethasone, and a third comprising thalidomide.
  • compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of vincristine, doxorubicin and dexamethasone (e.g., VAD).
  • vincristine, doxorubicin and dexamethasone are provided separately.
  • compositions comprising other combinations of agents can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form.
  • compositions can be made: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63, a second comprising dexamethasone, and a third comprising vincristine and doxorubicin.
  • an anti-CS1 antibody such as HuLuc63
  • a second comprising dexamethasone
  • a third comprising vincristine and doxorubicin.
  • compositions comprising a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection, vincristine and dexamethasone (e.g., DVd).
  • an anti-CS1 antibody such as HuLuc63
  • a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection, vincristine and dexamethasone (e.g., DVd).
  • doxorubicin HCl liposome injection e.g., doxorubicin HCl liposome injection
  • vincristine and dexamethasone e.g., DVd
  • compositions can be made: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63, a second comprising doxorubicin HCl liposome injection, and a third comprising vincristine and dexamethasone.
  • an anti-CS1 antibody such as HuLuc63
  • doxorubicin HCl liposome injection a second comprising doxorubicin HCl liposome injection
  • third comprising vincristine and dexamethasone.
  • two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of cyclophosphamide.
  • compositions comprising a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide and etoposide (e.g., DT-PACE).
  • an anti-CS1 antibody such as HuLuc63
  • a second comprising a therapeutically effective amount of dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide and etoposide e.g., DT-PACE.
  • compositions comprising other combinations can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form.
  • dexamethasone and thalidomide could be provided in one composition and cisplatin, doxorubicin, cyclophosphamide and
  • compositions comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of vincristine, doxorubicin, dexamethasone, and cyclophosphamide.
  • an anti-CS1 antibody such as HuLuc63
  • a second comprising a therapeutically effective amount of vincristine, doxorubicin, dexamethasone, and cyclophosphamide.
  • compositions comprising other combinations can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form.
  • vincristine, doxorubicin, and cyclophosphamide could be provided as one composition and dexamethasone as a second.
  • compositions comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection, vincristine, dexamethasone, and thalidomide.
  • an anti-CS1 antibody such as HuLuc63
  • a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection, vincristine, dexamethasone, and thalidomide.
  • compositions comprising other combinations can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form.
  • vincristine, dexamethasone, and thalidomide could be provided as one composition and doxorubicin HCl liposome injection as a second.
  • compositions comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection and bortezomib.
  • an anti-CS1 antibody such as HuLuc63
  • a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection and bortezomib.
  • compositions comprising other combinations can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form.
  • doxorubicin HCl liposome injection can be provided as one composition and bortezomib as a second.
  • agents with the same mechanism of action as an anti-CS1 antibody such as HuMax-Cd38 (Genmab) can be provided in the pharmaceutical compositions described herein.
  • the pharmaceutical compositions comprise therapeutic agents with a mechanism of action that differs from an anti-CS1 antibody.
  • targeted agents that inhibit angiogenesis including, but not limited to, bevacizumab, sutinib, sorafenib, 2-methoxyestradiol or 2ME2, finasunate, PTK787, vandetanib, aflibercept, volociximab, etaracizumab (MEDI-522), cilengitide, can be used in the pharmaceutical compositions described herein.
  • agents that inhibit growth factor signaling including, but not limited to, erlotinib, cetuximab, panitumumab, gefitinib, trastuzumab, TKI258, CP-751,871, atacicept, can be used in the pharmaceutical compositions described herein.
  • immunomodulators including, but not limited to, rituximab, alemtuzumab, aldesleukine, atlizumab, tocilizumab, temsirolimus, everolimus, NPI-1387, MLNM3897, HCD122, SGN-40, HLL1, huN901-DM1, atiprimod, natalizumab, can be used in the pharmaceutical compositions described herein.
  • agents that inhibit protein synthesis, folding or degradation including but not limited to, bortezomib, carfilzomib, NPI-0052, tanespimycin, saquinavir mesylate, ritonavir, nelfinavir mesylate, indinavir sulfate, can be used in the pharmaceutical compositions described herein.
  • agents that inhibit gene expression including but not limited to, belinostat, LBH589, can be used in the pharmaceutical compositions described herein.
  • pro-apoptotic agents including but not limited to, mapatumumab, lexatumumab, AMG951, ABT-737, oblimersen, plitidepsin, can be used in the pharmaceutical compositions described herein.
  • agents that inhibit signal transduction including but not limited to, SCIO-469, P276-00, enzastaurin, tipifarnib, perifosine, imatinib, can be used in the pharmaceutical compositions described herein.
  • agents with other mechanisms of action including but not limited to, dasatinib, lenalidomide, thalidomide, simvastatin, and celecoxib can be used in the pharmaceutical compositions described herein.
  • compositions can exist as a solid, semi-solid, or liquid (e.g., suspensions or aerosols) dosage form.
  • the compositions are administered in unit dosage forms suitable for single administration of precise dosage amounts.
  • an anti-CS1 antibody can be packaged in dosages ranging from about 1 to 1000 mg.
  • an anti-CS1 antibody is packaged in a dosage at least about 1 mg, at least about 10 mg, at least about 20 mg, at least about 50 mg, at least about 100 mg, at least about 200 mg, at least about 300 mg, at least about 400 mg, at least about 500 mg, at least about 750 mg, at least about 1000 mg.
  • compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, nontoxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration.
  • diluents are selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological saline, Ringer's solution, dextrose solution, and Hank's solution.
  • composition or formulation can also include other carriers, adjuvants, or nontoxic, non-therapeutic, nonimmunogenic stabilizers and the like. Effective amounts of such diluent or carrier will be those amounts that are effective to obtain a pharmaceutically acceptable formulation in terms of solubility of components, or biological activity.
  • compositions described herein find use in treating MM.
  • compositions can be used to treat Monoclonal Gammopathy of Undetermined Significance (MGUS), smoldering myeloma, asymptomatic MM, and symptomatic MM, ranging from newly diagnosed to late stage relapsed/refractory.
  • MGUS Monoclonal Gammopathy of Undetermined Significance
  • smoldering myeloma asymptomatic MM
  • symptomatic MM ranging from newly diagnosed to late stage relapsed/refractory.
  • compositions can be combined with other treatment strategies, i.e., autologous stem cell transplantation and allogeneic effector cell transplantation, to develop an effective treatment strategy based on the stage of myeloma being treated (see, e.g., Multiple Myeloma Research Foundation, Multiple Myeloma: Stem Cell Transplantation 1-30 (2004); U.S. Pat. Nos. 6,143,292, and 5,928,639, Igarashi, et al. Blood 2004, 104(1): 170-177, Maloney, et al. 2003, Blood, 102(9): 3447-3454, Badros, et al. 2002, J Clin Oncol., 20:1295-1303, Tricot, et al. 1996, Blood, 87(3):1196-1198; the contents of which are incorporated herein by reference).
  • other treatment strategies i.e., autologous stem cell transplantation and allogeneic effector cell transplantation
  • A relatively normal renal function
  • B abnormal renal function, creatinine value ⁇ 2.0 mg/dL
  • ISS International Staging System
  • IPI international prognostic index
  • the ISS is based on the assessment of two blood test results, beta 2 -microglobulin ( ⁇ 2 -M) and albumin, which separates patients into three prognostic groups irrespective of type of therapy.
  • EBMT European Group for Blood and Marrow transplantation
  • bone marrow plasma cells should increase by >25% and at least 10% in absolute terms; MRI examination may be helpful in selected patients.
  • a “near complete response” is defined as the criteria for a “complete response” (CR), but with a positive immunofixation test.
  • a “very good partial response” is defined as a greater than 90% decrease in M protein (see, e.g., Multiple Myeloma Research Foundation, Multiple Myeloma: Treatment Overview 9 (2005)).
  • the degree to which administration of the compositions elicits a response in an individual clinically manifesting at least one symptom associated with MM depends in part, on the severity of disease, e.g., Stage I, II, or III, and in part, on whether the patient is newly diagnosed or has late stage refractory MM. Thus, in some embodiments, administration of the pharmaceutical composition elicits a complete response.
  • administration of the pharmaceutical composition elicits a very good partial response or a partial response.
  • administration of the pharmaceutical composition elicits a minimal response.
  • administration of the pharmaceutical composition prevents the disease from progressing, resulting in a response classified as “no change” or “plateau” by the EBMT.
  • compositions comprising an anti-CS1 antibody such as HuLuc63 and one or more therapeutic agents for treating individuals diagnosed with MM can be determined using art-standard techniques, such as a standard dose escalation study to identify the MTD (see, e.g., Richardson, et al. 2002, Blood, 100(9):3063-3067, the content of which is incorporated herein by reference).
  • anti-CS1 antibodies are administered intravenously.
  • Administration of the other therapeutic agents described herein can be by any means known in the art. Such means include oral, rectal, nasal, topical (including buccal and sublingual) or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration and will depend in part, on the available dosage form. For example, therapeutic agents that are available in a pill or capsule format typically are administered orally.
  • oral administration generally requires administration of a higher dose than does intravenous administration. Determination of the actual route of administration that is best in a particular case is well within the capabilities of those skilled in the art, and in part, will depend on the dose needed versus the number of times per month administration is required.
  • Factors affecting the selected dosage of an anti-CS1 antibody and the therapeutic agents used in the compositions and methods described herein include, but are not limited to, the type of agent, the age, weight, and clinical condition of the recipient patient, and the experience and judgment of the clinician or practitioner administering the therapy.
  • the selected dosage should be sufficient to result in no change, but preferably results in at least a minimal change.
  • An effective amount of a pharmaceutical agent is that which provides an objectively identifiable response, e.g., plateau, no change, minimal, partial, or complete, as noted by the clinician or other qualified observer, and as defined by the EBMT.
  • an anti-CS1 antibody is administered as a separate composition from the composition(s) comprising the therapeutic agents.
  • the therapeutic agents can each be administered as a separate composition, or combined in a cocktail and administered as a single combined composition.
  • the compositions comprising an anti-CS1 antibody and one or more therapeutic agents are administered concurrently.
  • an anti-CS1 antibody can be administered prior to the administration of composition(s) comprising the therapeutic agent(s).
  • an anti-CS1 antibody is administered following the administration of composition(s) comprising the therapeutic agent(s).
  • an anti-CS1 antibody is administered prior to or following the administration of the therapeutic agents
  • determination of the duration between the administration of an anti-CS1 antibody and administration of the agents is well within the capabilities of those skilled in the art, and in part, will depend on the dose needed versus the number of times per month administration is required.
  • Doses of anti-CS1 antibodies used in the methods described herein typically range between 0.5 mg/kg to 20 mg/kg.
  • Optimal doses for the therapeutic agents are the generally accepted efficacious doses, such as those described in the Physician Desk Reference, 56th Ed. (2002), Publisher Medical Economics, New Jersey.
  • Optimal doses for agents not described in the Physician Desk Reference can be determined using a standard dose escalation study to identify the MTD (see, e.g., Richardson, et al. 2002, Blood, 100(9):3063-3067, the contents of which are incorporated herein by reference).
  • an anti-CS1 antibody is present in a pharmaceutical composition at a concentration, or in a weight/volume percentage, or in a weight amount, suitable for intravenous administration at a dosage rate at least about 0.5 mg/kg, at least about 0.75 mg/kg, at least about 1 mg/kg, at least about 2 mg/kg, at least about 2.5 mg/kg, at least about 3 mg/kg, at least about 4 mg/kg, at least about 5 mg/kg, at least about 6 mg/kg, at least about 7 mg/kg, at least about 8 mg/kg, at least about 9 mg/kg, at least about 10 mg/kg, at least about 11 mg/kg, at least about 12 mg/kg, at least about 13 mg/kg, at least about 14 mg/kg, at least about 15 mg/kg, at least about 16 mg/kg, at least about 17 mg/kg, at least about 18 mg/kg, at least about 19 mg/kg, and at least about 20 mg/kg.
  • Dexamethasone is a corticosteroid that is used extensively for the treatment of MM in multiple lines of therapy. Dex has been used as monotherapy or in combination with a variety of agents, including lenalidomide, thalidomide, velcade or as part of the VAD (vincristine, doxorubicin, dexamethasone) regimen or the DVd liposomal doxorubicin, vincristine, short-schedule dexamethasone regimen.
  • VAD vincristine, doxorubicin, dexamethasone
  • Dex The mechanism of action of Dex involves direct induction of apoptosis of myeloma cells by activation of caspases (Chauhan et al., 1997, Oncogene 15:837-843; Chauhan et al., 1997, J. Biol. Chem. 272, 29995-29997; Chauhan et al., 2001, J. Biol. Chem., 276: 24453-24456).
  • mice When tumors reached an average size of about 100 mm 3 , animals were randomized into 3 groups of 8-10 mice each and were treated with 1 or 10 mg/kg of HuLuc63 or isotype control antibody administered intraperitoneally twice a week for a maximum of 6 doses.
  • Dex was administered intraperitoneally at a dose of 10 mg/kg twice a week for a maximum of 6 doses.
  • CS1 protein expression was examined on the OPM2 multiple myeloma cell line. No significant change in CS1 expression was observed pre- or post-treatment with HuLuc63, Dex, or with both agents.
  • the combination of HuLuc63 with Dex was tested for anti-myeloma activity in vivo.
  • OPM2 tumor-bearing mice were treated with sub-optimal doses of HuLuc63 (1 mg/kg), or isotype control antibody twice weekly for three weeks.
  • Dex was given twice a week at 10 mg/kg to mice receiving either isotype control antibody or HuLuc63.
  • the results showed significant anti-tumor activity of HuLuc63 alone and in combination with Dex (see, e.g., FIG. 1 ).
  • Mice in the combination treatment group exhibited significantly smaller tumors than in the HuLuc63 monotherapy group (see, e.g., FIG. 1 ).
  • Thalidomide is an immunomodulatory drug that is currently approved for the treatment of MM in combination with Dex.
  • the mechanism of action of Thal is not completely understood, but involves inhibition of angiogenesis, inhibition of growth and survival of stromal cells and tumor cells in the bone marrow, and altering the production of factors that influence the survival of myeloma cells (such as IL-6, IL-10, IL-4, IL-5, IL-12, IL-8, TNF-alpha).
  • CS1 protein expression was examined on the L363 multiple myeloma cell line. No significant change in CS1 expression was observed pre- or post-treatment with HuLuc63, Thal or with both agents.
  • the combination of HuLuc63 with Thal was tested for anti-myeloma activity in vivo.
  • L363 tumor-bearing mice were treated with HuLuc63, or isotype control antibody twice weekly for three weeks at 10 mg/kg.
  • Thal was given 5 days a week at 50 mg/kg for a maximum of 15 doses to mice receiving either isotype control antibody or HuLuc63.
  • Significant anti-tumor activity was observed with HuLuc63 alone, Thal alone and HuLuc63 in combination with Thal (see, e.g., FIG. 2 ).
  • Mice in the combination treatment group exhibited smaller tumors compared to the HuLuc63 and Thal monotherapy groups (see, e.g., FIG. 2 ).
  • Thal/Dex combination therapy is currently approved for front-line treatment of MM patients.
  • the combination of HuLuc63 with Thal/Dex was tested for anti-myeloma activity in vivo as described above.
  • the effect of Thal/Dex treatment on the expression of CS1 in MM cell lines and mouse xenograft tumors was examined by immunohistochemistry as described above.
  • L363 tumor-bearing mice were treated with HuLuc63, or isotype control antibody twice weekly for three weeks at 10 mg/kg.
  • Thal was given 5 days a week at 50 mg/kg for a maximum of 15 doses and Dex was given twice a week at 10 mg/kg for a maximum of 6 doses to mice receiving either isotype control antibody or HuLuc63.
  • the results showed significant anti-tumor activity of HuLuc63 alone, Thal/Dex alone and HuLuc63 in combination with Thal/Dex (see e.g., FIG. 3 ).
  • mice in the HuLuc63/Thal/Dex combination treatment group exhibited the smallest tumors, on average exhibiting a decrease of 50-70% in tumor size compared to HuLuc63 monotherapy or Thal/Dex therapy (see e.g., FIG. 3 ).
  • Bevacizumab is a monoclonal antibody that targets the endothelial growth factor VEGF. It works by inhibiting VEGF, resulting in the inhibition of new blood vessel formation in tumors. Bevacizumab is currently approved for the treatment of solid tumors, including metastatic colorectal cancer. Bevacizumab is currently not approved for the use in the treatment of MM.
  • HuLuc63 HuLuc63
  • Bevacizumab was tested for anti-myeloma activity in vivo, as described above.
  • L363 tumor-bearing mice were treated with HuLuc63, or isotype control antibody twice weekly for three weeks at 10 mg/kg.
  • Bevacizumab was given twice a week at 0.5 mg/kg for a maximum of 6 doses to mice receiving either isotype control antibody or HuLuc63.
  • Significant anti-tumor activity of HuLuc63 alone and in combination with Bevacizumab was observed (see e.g., FIG. 4 ). Bevacizumab alone did not exhibit significant anti-tumor activity.
  • mice receiving the combination of HuLuc63 with Bevacizumab exhibited significantly smaller tumors compared to the HuLuc63 monotherapy group, indicating that HuLuc63 and Bevacizumab may act synergistically to cause an anti-myeloma effect.
  • HuLuc63 will be given by intravenous injection (IV) at up to five dose levels ranging from 2.5 mg/kg to 20 mg/kg in combination with one or more therapeutic agents.
  • Patients will receive HuLuc63 once a week, with each dose infused over 1 hour, once every 10 days or once every two weeks, for a minimum of 4 doses and a maximum of 52 doses.
  • Drug combinations suitable for administration with HuLuc63 include, but are not limited to:
  • EBMT criteria will be assessed. If a patient has progressive disease, HuLuc63 will be discontinued and the other drug combinations may be withdrawn or continued at the discretion of the site investigator. If the patient has responded or has stable disease at Week 8-12, dosing with HuLuc63 and the drug combination will continue so that a maximum of 52 weeks of treatment are completed or disease progression occurs.
  • Approximately 15 to 30 patients in 5 cohorts will be enrolled for each drug combination. Each cohort will begin with 3 patients. If no dose-limiting toxicity (DLT) is noted within the first 4 weeks of treatment in any patient, enrollment will begin in the next higher cohort. If one patient has a DLT, 3 additional patients will be enrolled in the cohort. If no other patient in the cohort has a DLT, escalation to the next cohort may proceed. If a second patient in a cohort has a DLT, the maximum tolerated dose (MTD) has been reached.
  • DLT dose-limiting toxicity
  • a dose-limiting toxicity is defined using the National Cancer Center Institute Common Toxicity Criteria Version 3.0 (NCI CTCAE v3.0) as a grade 4 hematologic toxicity or hyperbilirubinemia, or a grade 3 toxicity in any other system considered related to HuLuc63 or the combination of HuLuc63 and any of the drugs listed in Table 1.
  • NCI CTCAE v3.0 National Cancer Center Institute Common Toxicity Criteria Version 3.0
  • 3 assessable patients must complete their first 4 weeks of dosing. If a DLT occurs, an additional three assessable patients will be accrued. Patients will be monitored for safety by assessing adverse events categorized by NCI CTCAE v3.0 and patients will be monitored for clinical activity using EBMT.
  • the maximally tolerated dose is defined as the highest dose studied for which the incidence of DLTs is ⁇ 33%.
  • the highest tolerated dose will be HuLuc63 at 20 mg/kg combined with the drugs at the doses listed above if no dose limiting toxicities are observed.

Abstract

Compositions and methods for treating MM are provided herein.

Description

    1. CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims benefit under 35 U.S.C. §119(e) to application Ser. Nos. 60/836,250 filed Aug. 7, 2006 and 60/856,144 filed Nov. 1, 2006, the contents of which are incorporated herein by reference.
  • 2. BACKGROUND
  • Multiple myeloma (“MM”) represents a malignant proliferation of plasma cells derived from a single clone. The terms multiple myeloma and myeloma are used interchangeably to refer to the same condition. The myeloma tumor, its products, and the host response to it result in a number of organ dysfunctions and symptoms of bone pain or fracture, renal failure, susceptibility to infection, anemia, hypocalcemia, and occasionally clotting abnormalities, neurologic symptoms and vascular manifestations of hyperviscosity. See D. Longo, in Harrison's Principles of Internal Medicine 14th Edition, p. 713 (McGraw-Hill, New York, 1998). No effective long-term treatment currently exists for MM. It is a malignant disease of plasma cells, manifested as hyperproteinemia, anemia, renal dysfunction, bone lesions, and immunodeficiency. MM is difficult to diagnose early because there may be no symptoms in the early stage. The disease has a progressive course with a median duration of survival of six months when no treatment is given. Systemic chemotherapy is the main treatment, and the current median of survival with chemotherapy is about three years, however fewer than 5% live longer than 10 years (See Anderson, K. et al., Annual Meeting Report 1999. Recent Advances in the Biology and Treatment of Multiple Myeloma (1999)).
  • While multiple myeloma is considered to be a drug-sensitive disease, almost all patients who initially respond to chemotherapy eventually relapse (See Anderson, K. et al., Annual Meeting Report 1999. Recent Advances in the Biology and Treatment of Multiple Myeloma (1999)). Since the introduction of melphalan and prednisone therapy for MM, numerous multi-drug chemotherapies including Vinca alkaloid, anthracycline, and nitrosourea-based treatments have been tested (See Case, D C et al., (1977) Am. J. Med 63:897 903); however, there has been little improvement in outcome over the past three decades (See Case, D C et al., (1977) Am, J. Med 63:897 903; Otsuki, T. et al, (2000) Cancer Res. 60:1). New methods of treatment, such as combination therapies utilizing monoclonal antibodies, therapeutic agents, and small molecule inhibitors of cellular receptors and/or proteins implicated in MM, are needed.
  • 3. SUMMARY
  • Described herein are compositions and methods useful for exploiting the anti-tumor properties of anti-CS1 antibodies. Anti-CS1 antibodies that can be used in the methods and compositions are described in U.S. Patent Publication Nos. 2005/0025763 and 2006/0024296, the contents of which are incorporated herein by reference. The anti-CS1 antibodies target CS1 (CD2-subsetl), which is also known as SLAMF7, CRACC, 19A, APEX-1, and FOAP12 (Genbank Accession Number NM021181.3). CS1, is a glycoprotein that is highly expressed in bone marrow samples from patients diagnosed with MM. In both in vitro and in vivo studies, anti-CS1 antibodies exhibit significant anti-myeloma activity (see, e.g., U.S. Patent Publication Nos. 2005/0025763 and 2006/0024296, the contents of which are incorporated herein by reference). By way of example, but not limitation, the anti-CS1 antibody, HuLuc63 effectively mediates lysis of myeloma cells via antibody dependent cellular cytotoxicity (ADCC) (see, e.g., U.S. Patent Publication No. 2005/0025763, the content of which is incorporated herein by reference). In a myeloma mouse tumor model, treatment with HuLuc63 significantly reduced tumor mass by more than 50% (see, e.g., U.S. Patent Publication No. 2005/0025763, the content of which is incorporated herein by reference).
  • The present disclosure relates to compositions and methods for treating patients diagnosed with Monoclonal Gammopathy of Undetermined Significance (MGUS), smoldering myeloma, asymptomatic MM, and symptomatic MM, ranging from newly diagnosed to late stage relapsed/refractory. In particular, the methods relate to the administration of a pharmaceutical composition comprising an anti-CS1 antibody in combination with one or more therapeutic agents. Anti-CS1 antibodies are typically administered intravenously at doses ranging from 0.5 to 20 mg/kg, from once a week to once a month.
  • One or more therapeutic agents, such as targeted agents, conventional chemotherapy agents, hormonal therapy agents, and supportive care agents and/or combinations thereof, can be administered concurrently, prior to, or following administration of an anti-CS1 antibody. The agents can be administered separately or combined in a cocktail and administered together as a single composition. The composition(s) can be administered by any means known in the art.
  • In some embodiments, administration of the pharmaceutical compositions described herein increases the sensitivity of multiple myeloma cells to a therapeutic agent. For example, inclusion of an anti-CS1 antibody, such as HuLuc63, enhances the activity of therapeutic agents, such that lower doses can be used in the compositions and methods described herein.
  • In some embodiments, administration of the pharmaceutical compositions described herein elicits at least one of the beneficial responses as defined by the European Group for Blood and Marrow transplantation (EBMT). For example, administration of the pharmaceutical compositions described herein can result in a complete response, partial response, minimal response, no change, or plateau.
  • 4. BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 depicts the anti-tumor activity of HuLuc63 alone and in combination with dexamethasone in an in vivo mouse multiple myeloma xenograft model;
  • FIG. 2 depicts the anti-tumor activity of HuLuc63 alone, thalidomide alone, and HuLuc63 in combination with thalidomide in an in vivo mouse multiple myeloma xenograft model;
  • FIG. 3 depicts the anti-tumor activity of HuLuc63 alone, thalidomide/dexamethasone, and HuLuc63 in combination with thalidomide and dexamethasone in an in vivo mouse multiple myeloma xenograft model; and
  • FIG. 4 depicts the anti-tumor activity of HuLuc63 alone and in combination with bevacizumab in an in vivo mouse multiple myeloma xenograft model.
  • 5. DETAILED DESCRIPTION
  • The compositions described herein combine anti-CS1 antibodies with one or more therapeutic agents at specific doses to potentiate or complement the anti-myeloma activities of the other. Examples of suitable anti-CS1 antibodies include, but are not limited to, isolated antibodies that bind one or more of the three epitope clusters identified on CS1 and monoclonal antibodies produced by the hybridoma cell lines: Luc2, Luc3, Luc15, Luc22, Luc23, Luc29, Luc32, Luc34, Luc35, Luc37, Luc38, Luc39, Luc56, Luc60, Luc63, Luc69, LucX.1, LucX.2 or Luc90. These monoclonal antibodies are named as the antibodies: Luc2, Luc3, Luc15, Luc22, Luc23, Luc29, Luc32, Luc34, Luc35, Luc37, Luc38, Luc39, Luc56, Luc60, Luc63, Luc69, LucX and Luc90, respectively, hereafter. Humanized versions are denoted by the prefix “hu” (see, e.g., U.S. Patent Publication Nos. 2005/0025763 and 2006/0024296, the contents of which are incorporated herein by reference).
  • In some embodiments, suitable anti-CS1 antibodies include isolated antibodies that bind one or more of the three epitope clusters identified on CS1 (SEQ ID NO: 1, Table 1 below; see, e.g., U.S. Patent Publication No. 2006/0024296, the content of which is incorporated herein by reference). As disclosed in U.S. Patent Publication No. 2006/0024296 and shown below in Table 1, the CS1 antibody binding sites have been grouped into 3 epitope clusters:
      • (1) the epitope defined by Luc90, which binds to hu50/mu50 (SEQ ID NO: 2). This epitope covers from about amino acid residue 23 to about amino acid residue 151 of human CS1. This epitope is resided within the domain 1 (V domain) of the extracellular domain. This epitope is also recognized by Luc34, LucX (including LucX.1 and LucX.2) and Luc69.
      • (2) the epitope defined by Luc38, which binds to mu25/hu75 (SEQ ID NO: 3) and hu50/mu50 (SEQ ID NO: 81). This epitope likely covers from about amino acid residue 68 to about amino acid residue 151 of human CS1. This epitope is also recognized by Luc5.
      • (3) the epitope defined by Luc 63, which binds to mu75/hu25 (SEQ ID NO: 4). This epitope covers from about amino acid residue 170 to about amino acid residue 227 of human CS1. This epitope is resided within domain 2 (C2 domain) of human CS1. This epitope is also recognized by Luc4, Luc12, Luc23, Luc29, Luc32 and Luc37.
  • The methods and pharmaceutical compositions are addressed in more detail below, but typically include at least one anti-CS1 antibody as described above. In some embodiments, the pharmaceutical compositions include the anti-CS1 antibody HuLuc63. HuLuc63 is a humanized recombinant monoclonal IgG1 antibody directed to human CS1. The amino acid sequence for the heavy chain variable region (SEQ ID NO: 5) and the light chain variable region (SEQ ID NO: 6) for HuLuc63 is disclosed in U.S. Patent Publication No. 2005/0025763, the content of which is incorporated herein by reference, and in Table 1.
  • TABLE 1
    SEQ ID
    NO: Amino Acid Sequence
    SEQ ID Met Ala Gly Ser Pro Thr Cys Leu Thr Leu Ile Tyr Ile Leu Trp Gln Leu
    NO: 1 Thr Gly Ser Ala Ala Ser Gly Pro Val Lys Glu Leu Val Gly Ser Val Gly
    Gly Ala Val Thr Phe Pro Leu Lys Ser Lys Val Lys Gln Val Asp Ser Ile
    Val Trp Thr Phe Asn Thr Thr Pro Leu Val Thr Ile Gln Pro Glu Gly Gly
    Thr Ile Ile Val Thr Gln Asn Arg Asn Arg Glu Arg Val Asp Phe Pro Asp
    Gly Gly Tyr Ser Leu Lys Leu Ser Lys Leu Lys Lys Asn Asp Ser Gly Ile
    Tyr Tyr Val Gly Ile Tyr Ser Ser Ser Leu Gln Gln Pro Ser Thr Gln Glu 
    Tyr Val Leu His Val Tyr Glu His Leu Ser Lys Pro Lys Val Thr Met Gly 
    Leu Gln Ser Asn Lys Asn Gly Thr Cys Val Thr Asn Leu Thr Cys Cys Met 
    Glu His Gly Glu Glu Asp Val Ile Tyr Thr Trp Lys Ala Leu Gly Gln Ala 
    Ala Asn Glu Ser His Asn Gly Ser Ile Leu Pro Ile Ser Trp Arg Trp Gly 
    Glu Ser Asp Met Thr Phe Ile Cys Val Ala Arg Asn Pro Val Ser Arg Asn 
    Phe Ser Ser Pro Ile Leu Ala Arg Lys Leu Cys Glu Gly Ala Ala Asp Asp 
    Pro Asp Ser Ser Met Val Leu Leu Cys Leu Leu Leu Val Pro Leu Leu Leu 
    Ser Leu Phe Val Leu Gly Leu Phe Leu Trp Phe Leu Lys Arg Glu Arg Gln 
    Glu Glu Tyr Ile Glu Glu Lys Lys Arg Val Asp Ile Cys Arg Glu Thr Pro 
    Asn Ile Cys Pro His Ser Gly Glu Asn Thr Glu Tyr Asp Thr Ile Pro His 
    Thr Asn Arg Thr Ile Leu Lys Glu Asp Pro Ala Asn Thr Val Tyr Ser Thr 
    Val Glu Ile Pro Lys Lys Met Glu Asn Pro His Ser Leu Leu Thr Met Pro 
    Asp Thr Pro Arg Leu Phe Ala Tyr Glu Asn Val Ile
    SEQ ID Met Ala Gly Ser Pro Thr Cys Leu Thr Leu Ile Tyr Ile Leu Trp Gln Leu
    NO: 2 Thr Gly Ser Ala Ala Ser Gly Pro Val Lys Glu Leu Val Gly Ser Val Gly
    Gly Ala Val Thr Phe Pro Leu Lys Ser Lys Val Lys Gln Val Asp Ser Ile
    Val Trp Thr Phe Asn Thr Thr Pro Leu Val Thr Ile Gln Pro Glu Gly Gly
    Thr Ile Ile Val Thr Gln Asn Arg Asn Arg Glu Arg Val Asp Phe Pro Asp
    Gly Gly Tyr Ser Leu Lys Leu Ser Lys Leu Lys Lys Asn Asp Ser Gly Ile
    Tyr Tyr Val Gly Ile Tyr Ser Ser Ser Leu Gln Gln Pro Ser Thr Gln Glu 
    Tyr Val Leu His Val Tyr Glu His Leu Ser Lys Pro Lys Val Thr Ile Asp 
    Arg Gln Ser Asn Lys Asn Gly Thr Cys Val Ile Asn Leu Thr Cys Ser Thr 
    Asp Gln Asp Gly Glu Asn Val Thr Tyr Ser Trp Lys Ala Val Gly Gln Gly 
    Asp Asn Gln Phe His Asp Gly Ala Thr Leu Ser Ile Ala Trp Arg Ser Gly 
    Glu Lys Asp Gln Ala Leu Thr Cys Met Ala Arg Asn Pro Val Ser Asn Ser 
    Phe Ser Thr Pro Val Phe Pro Gln Lys Leu Cys Glu Asp Ala Ala Thr Asp 
    Leu Thr Ser Leu Arg Gly
    SEQ ID Met Ala Arg Phe Ser Thr Tyr Ile Ile Phe Thr Ser Val Leu Cys Gln Leu
    NO: 3 Thr Val Thr Ala Ala Ser Gly Thr Leu Lys Lys Val Ala Gly Ala Leu Asp
    Gly Ser Val Thr Phe Thr Leu Asn Ile Thr Glu Ile Lys Val Asp Tyr Val
    Val Trp Thr Phe Asn Thr Phe Phe Leu Ala Met Val Lys Lys Asp Gly Gly
    Thr Ile Ile Val Thr Gln Asn Arg Asn Arg Glu Arg Val Asp Phe Pro Asp
    Gly Gly Tyr Ser Leu Lys Leu Ser Lys Leu Lys Lys Asn Asp Ser Gly Ile
    Tyr Tyr Val Gly Ile Tyr Ser Ser Ser Leu Gln Gln Pro Ser Thr Gln Glu 
    Tyr Val Leu His Val Tyr Glu His Leu Ser Lys Pro Lys Val Thr Met Gly 
    Leu Gln Ser Asn Lys Asn Gly Thr Cys Val Thr Asn Leu Thr Cys Cys Met 
    Glu His Gly Glu Glu Asp Val Ile Tyr Thr Trp Lys Ala Leu Gly Gln Ala 
    Ala Asn Glu Ser His Asn Gly Ser Ile Leu Pro Ile Ser Trp Arg Trp Gly 
    Glu Ser Asp Met Thr Phe Ile Cys Val Ala Arg Asn Pro Val Ser Arg Asn 
    Phe Ser Ser Pro Ile Leu Ala Arg Lys Leu Cys Glu Gly Ala Ala Asp Asp 
    Pro Asp Ser Ser Met Val
    SEQ ID Met Ala Arg Phe Ser Thr Tyr Ile Ile Phe Thr Ser Val Leu Cys Gln Leu
    NO: 4 Thr Val Thr Ala Ala Ser Gly Thr Leu Lys Lys Val Ala Gly Ala Leu Asp
    Gly Ser Val Thr Phe Thr Leu Asn Ile Thr Glu Ile Lys Val Asp Tyr Val
    Val Trp Thr Phe Asn Thr Phe Phe Leu Ala Met Val Lys Lys Asp Gly Val
    Thr Ser Gln Ser Ser Asn Lys Glu Arg Ile Val Phe Pro Asp Gly Leu Tyr
    Ser Met Lys Leu Ser Gln Leu Lys Lys Asn Asp Ser Gly Ala Tyr Arg Ala
    Glu Ile Tyr Ser Thr Ser Ser Gln Ala Ser Leu Ile Gln Glu Tyr Val Leu 
    His Val Tyr Lys His Leu Ser Arg Pro Lys Val Thr Ile Asp Arg Gln Ser 
    Asn Lys Asn Gly Thr Cys Val Ile Asn Leu Thr Cys Ser Thr Asp Gln Asp 
    Gly Glu Asn Val Thr Tyr Ser Trp Lys Ala Val Gly Gln Ala Ala Asn Glu 
    Ser His Asn Gly Ser Ile Leu Pro Ile Ser Trp Arg Trp Gly Glu Ser Asp 
    Met Thr Phe Ile Cys Val Ala Arg Asn Pro Val Ser Arg Asn Phe Ser Ser 
    Pro Ile Leu Ala Arg Lys Leu Cys Glu Gly Ala Ala Asp Asp Pro Asp Ser 
    Ser Met Val
    SEQ ID Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly Ser
    NO: 5 Leu Arg Leu Ser Cys Ala Ala Ser Gly Phe Asp Phe Ser Arg Tyr Trp Met
    Ser Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Ile Gly Glu Ile
    Asn Pro Asp Ser Ser Thr Ile Asn Tyr Ala Pro Ser Leu Lys Asp Lys Phe
    Ile Ile Ser Arg Asp Asn Ala Lys Asn Ser Leu Tyr Leu Gln Met Asn Ser
    Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys Ala Arg Pro Asp Gly Asn
    Tyr Trp Tyr Phe Asp Val Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ser
    SEQ ID Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly Asp
    NO: 6 Arg Val Thr Ile Thr Cys Lys Ala Ser Gln Asp Val Gly Ile Ala Val Ala
    Trp Tyr Gln Gln Lys Pro Gly Lys Val Pro Lys Leu Leu Ile Tyr Tip Ala
    Ser Thr Arg His Thr Gly Val Pro Asp Arg Phe Ser Gly Ser Gly Ser Gly
    Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro Glu Asp Val Ala Thr
    Tyr Tyr Cys Gln Gln Tyr Ser Ser Tyr Pro Tyr Thr Phe Gly Gln Gly Thr
    Lys Val Glu Ile Lys
  • At some doses, additive effects are seen; at other doses, synergistic effects are seen. In some embodiments, the synergistic effect permits one or more therapeutic agents to be administered in combination with one or more anti-CS1 antibodies at a reduced dosage, while retaining efficacy. Given that the side effects associated with the use of these agents are dose-dependent, use of the compositions and methods described herein can reduce the deleterious side effects observed in conventional and novel treatment regimens used to treat MM when these agents are administered at their recommended dosages.
  • In other embodiments, the synergistic effect permits one or more therapeutic agents to be administered in combination with one or more anti-CS1 antibodies at the approved dosage, but with greater than the expected efficacy.
  • The compositions can be administered for the treatment of Monoclonal Gammopathy of Undetermined Significance (MGUS), smoldering myeloma, asymptomatic MM, and symptomatic MM, ranging from newly diagnosed to late stage relapsed/refractory. Typically, administration of the compositions results in a reduction in M-protein in serum or urine such that a plateau, no change, minimal, partial or complete response is observed as defined by the European Group for Blood and Marrow transplantation (EBMT).
  • 5.2 Pharmaceutical Compositions
  • Provided herein are pharmaceutical compositions that are beneficial in reducing tumor mass and/or regressing tumor growth, in patients diagnosed with multiple myeloma. In addition, the pharmaceutical compositions can be used to treat other diseases characterized by the presence of monoclonal protein (M-protein, paraprotein) in the serum or urine.
  • In some embodiments, the various components of the compositions are provided separately. For example, an anti-CS1 antibody can be provided in a first pharmaceutical composition, and a therapeutic agent provided in a second composition. When the composition comprises two or more therapeutic agents, an anti-CS1 antibody can be provided in a first pharmaceutical composition, one therapeutic agent can be provided in a second composition and the other therapeutic agent can be provided in a third composition. In other embodiments, an anti-CS1 antibody can be provided in one pharmaceutical composition and the therapeutic agents can be combined and provided in a second pharmaceutical composition. In still other embodiments, one composition, comprising an anti-CS1 antibody and one or more therapeutic agents can be provided.
  • In typical embodiments, an anti-CS1 antibody is present in a pharmaceutical composition at a concentration sufficient to permit intravenous administration at 0.5 mg/kg to 20 mg/kg. In some embodiments, the concentration of an anti-CS1 antibody suitable for use in the compositions and methods described herein includes, but is not limited to, at least about 0.5 mg/kg, at least about 0.75 mg/kg, at least about 1 mg/kg, at least about 2 mg/kg, at least about 2.5 mg/kg, at least about 3 mg/kg, at least about 4 mg/kg, at least about 5 mg/kg, at least about 6 mg/kg, at least about 7 mg/kg, at least about 8 mg/kg, at least about 9 mg/kg, at least about 10 mg/kg, at least about 11 mg/kg, at least about 12 mg/kg, at least about 13 mg/kg, at least about 14 mg/kg, at least about 15 mg/kg, at least about 16 mg/kg, at least about 17 mg/kg, at least about 18 mg/kg, at least about 19 mg/kg, and at least about 20 mg/kg.
  • The anti-CS1 antibodies can be administered in single or multiple dose regimens. Generally, an anti-CS1 antibody is administered over a period of time from about 1 to about 24 hours, but is typically administered over a period of about 1 to 2 hours. Dosages can be repeated from about 1 to about 4 weeks or more, for a total of 4 or more doses. Typically, dosages are repeated once every week, once every other week, or once a month for a minimum of 4 doses to a maximum of 52 doses.
  • Determination of the effective dosage, total number of doses, and length of treatment with an anti-CS1 antibody is well within the capabilities of those skilled in the art, and can be determined using a standard dose escalation study to identify the maximum tolerated dose (MTh) (see, e.g., Richardson et al., 2002, Blood, 100(9):3063-3067, the content of which is incorporated herein by reference).
  • In some embodiments, one or more therapeutic agents are administered in combination with an anti-CS1 antibody. The agents can be administered concurrently, prior to, or following administration of an anti-CS1 antibody.
  • In some embodiments, an anti-CS1 antibody is administered prior to the administration of the therapeutic agents. For example, the anti-CS1 antibody can be administered approximately 0 to 60 days prior to the administration of the therapeutic agents. In some embodiments, an anti-CS1 antibody, such as HuLuc63, is administered from about 30 minutes to about 1 hour prior to the administration of the therapeutic agents, or from about 1 hour to about 2 hours prior to the administration of the therapeutic agents, or from about 2 hours to about 4 hours prior to the administration of the therapeutic agents, or from about 4 hours to about 6 hours prior to the administration of the therapeutic agents, or from about 6 hours to about 8 hours prior to the administration of the therapeutic agents, or from about 8 hours to about 16 hours prior to the administration of the therapeutic agents, or from about 16 hours to 1 day prior to the administration of the therapeutic agents, or from about 1 to 5 days prior to the administration of the therapeutic agents, or from about 5 to 10 days prior to the administration of the therapeutic agents, or from about 10 to 15 days prior to the administration of the therapeutic agents, or from about 15 to 20 days prior to the administration of the therapeutic agents, or from about 20 to 30 days prior to the administration of the therapeutic agents, or from about 30 to 40 days prior to the administration of the therapeutic agents, and from about 40 to 50 days prior to the administration of the therapeutic agents, or from about 50 to 60 days prior to the administration of the therapeutic agents.
  • In some embodiments, the anti-CS1 antibody is administered concurrently with the administration of the therapeutic agents.
  • In some embodiments, an anti-CS1 antibody is administered following the administration of the therapeutic agents. For example, an anti-CS1 antibody, such as HuLuc63, can be administered approximately 0 to 60 days after the administration of the therapeutic agents. In some embodiments, HuLuc63 is administered from about 30 minutes to about 1 hour following the administration of the therapeutic agents, or from about 1 hour to about 2 hours following the administration of the therapeutic agents, or from about 2 hours to about 4 hours following the administration of the therapeutic agents, or from about 4 hours to about 6 hours following the administration of the therapeutic agents, or from about 6 hours to about 8 hours following the administration of the therapeutic agents, or from about 8 hours to about 16 hours following the administration of the therapeutic agents, or from about 16 hours to 1 day following the administration of the therapeutic agents, or from about 1 to 5 days following the administration of the therapeutic agents, or from about 5 to 10 days following the administration of the therapeutic agents, or from about 10 to 15 days following the administration of the therapeutic agents, or from about 15 to 20 days following the administration of the therapeutic agents, or from about 20 to 30 days following the administration of the therapeutic agents, or from about 30 to 40 days following the administration of the therapeutic agents, and from about 40 to 50 days following the administration of the therapeutic agents, or from about 50 to 60 days following the administration of the therapeutic agents.
  • Therapeutic agents that can be used in combination with the anti-CS1 antibodies described herein include, but are not limited to, targeted agents, conventional chemotherapy agents, hormonal therapy agents, and supportive care agents. One or more therapeutic agents from the different classes, e.g., targeted, conventional chemotherapeutic, hormonal, and supportive care, and/or subclasses can be combined in the compositions described herein. The various classes described herein can be further divided into subclasses. By way of example, targeted agents can be separated into a number of different subclasses depending on their mechanism of action. As will be apparent to those of skill in the art, the agents can have more than one mechanism of action, and thus, could be classified into one or more subclasses. For purposes of the compositions and methods described herein, the following subclasses have been identified: anti-angiogenic, inhibitors of growth factor signaling, immunomodulators, inhibitors of protein synthesis, folding and/or degradation, inhibitors of gene expression, pro-apoptotic agents, agents that inhibit signal transduction and agents with “other” mechanisms of action. Typically, the mechanism of action for agents falling into the “other” subclass is unknown or poorly characterized.
  • For example, in some embodiments, targeted agents, such as bevacizumab, sutinib, sorafenib, 2-methoxyestradiol or 2ME2, finasunate, PTK787, vandetanib, aflibercept, volociximab, etaracizumab (MEDI-522), cilengitide, erlotinib, cetuximab, panitumumab, gefitinib, trastuzumab, TKI258, CP-751,871, atacicept, rituximab, alemtuzumab, aldesleukine, atlizumab, tocilizumab, temsirolimus, everolimus, NPI-1387, MLNM3897, HCD122, SGN-40, HLL1, huN901-DM1, atiprimod, natalizumab, bortezomib, carfilzomib, NPI-0052, tanespimycin, saquinavir mesylate, ritonavir, nelfinavir mesylate, indinavir sulfate, belinostat, LBH589, mapatumumab, lexatumumab, AMG951, ABT-737, oblimersen, plitidepsin, SCIO-469, P276-00, enzastaurin, tipifarnib, perifosine, imatinib, dasatinib, lenalidomide, thalidomide, simvastatin, and celecoxib can be combined with an anti-CS1 antibody, such as HuLuc63 and used to treat MM patients.
  • By way of another example, conventional chemotherapy agents, such as alklyating agents (e.g., oxaliplatin, carboplatin, cisplatin, cyclophosphamide, melphalan, ifosfamide, uramustine, chlorambucil, carmustine, mechloethamine, thiotepa, busulfan, temozolomide, dacarbazine), anti-metabolic agents (e.g., gemcitabine, cytosine arabinoside, Ara-C, capecitabine, 5FU (5-fluorouracil), azathioprine, mercaptopurine (6-MP), 6-thioguanine, aminopterin, pemetrexed, methotrexate), plant alkaloid and terpenoids (e.g., docetaxel, paclitaxel, vincristine, vinblastin, vinorelbine, vindesine, etoposide, VP-16, teniposide, irinotecan, topotecan), anti-tumor antibiotics (e.g., dactinomycin, doxorubicin, liposomal doxorubicin, daunorubicin, daunomycin, epirubicin, mitoxantrone, adriamycin, bleomycin, plicamycin, mitomycin C, caminomycin, esperamicins), and other agents (e.g., darinaparsin) can be combined with an anti-CS1 antibody, such as HuLuc63 and used to treat MM.
  • By way of another example, hormonal agents such as anastrozole, letrozole, goserelin, tamoxifen, dexamethasone, prednisone, and prednisilone can be combined with an anti-CS1 antibody, such as HuLuc63 and used to treat MM.
  • By way of another example, supportive care agents such as pamidronate, zoledonic acid, ibandronate, gallium nitrate, denosumab, darbepotin alpha, epoetin alpha, eltrombopag, and pegfilgrastim can be combined with an anti-CS1 antibody, such as HuLuc63 and used to treat MM.
  • The therapeutic agents can be administered in any manner found appropriate by a clinician and are typically provided in generally accepted efficacious dose ranges, such as those described in the Physician Desk Reference, 56th Ed. (2002), Publisher Medical Economics, New Jersey. In other embodiments, a standard dose escalation study can be performed to identify the maximum tolerated dose (MTD) (see, e.g., Richardson, et al. 2002, Blood, 100(9):3063-3067, the content of which is incorporated herein by reference).
  • In some embodiments, doses less than the generally accepted efficacious dose of a therapeutic agent can be used. For example, in various embodiments, the composition comprises a dosage that is less than about 10% to 75% of the generally accepted efficacious dose range. In some embodiments, at least about 10% or less of the generally accepted efficacious dose range is used, at least about 15% or less, at least about 25%, at least about 30% or less, at least about 40% or less, at least about 50% or less, at least about 60% or less, at least about 75% or less, and at least about 90%.
  • The therapeutic agents can be administered singly or sequentially, or in a cocktail with other therapeutic agents, as described below. The therapeutic agents can be administered orally, intravenously, systemically by injection intramuscularly, subcutaneously, intrathecally or intraperitoneally.
  • In some embodiments, the therapeutic agents provided in the pharmaceutical composition(s) are selected from the group consisting of dexamethasone, thalidomide, vincristine, carmustine (BCNU), melphalan, cyclophosphamide, prednisone, doxorubicin, cisplatin, etoposide, bortezomib, lenalidomide, ara-C, and/or combinations thereof.
  • In certain embodiments, however, the pharmaceutical composition does not comprise bortezomib and/or lenalidomide.
  • Accordingly, in some embodiments, two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of dexamethasone.
  • In some embodiments, at least two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of dexamethasone and thalidomide. In some embodiments, dexamethasone and thalidomide are provided separately, such that a total of three pharmaceutical compositions are provided: a first comprising an anti-CS1 antibody such as HuLuc63, a second comprising dexamethasone, and a third comprising thalidomide.
  • In some embodiments, at least two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of vincristine, doxorubicin and dexamethasone (e.g., VAD). In some embodiments, vincristine, doxorubicin and dexamethasone are provided separately. Provided that the agents retain their efficacy, compositions comprising other combinations of agents can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form. For example, a total of three compositions can be made: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63, a second comprising dexamethasone, and a third comprising vincristine and doxorubicin.
  • In some embodiments, at least two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection, vincristine and dexamethasone (e.g., DVd). Provided that the agents retain their efficacy, compositions comprising other combinations of agents can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form. For example, a total of three compositions can be made: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63, a second comprising doxorubicin HCl liposome injection, and a third comprising vincristine and dexamethasone.
  • In some embodiments, two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of cyclophosphamide.
  • In some embodiments, at least two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide and etoposide (e.g., DT-PACE). Provided that the agents retain their efficacy, compositions comprising other combinations can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form. By way of example, dexamethasone and thalidomide could be provided in one composition and cisplatin, doxorubicin, cyclophosphamide and etoposide provided in another composition.
  • In some embodiments, at least two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of vincristine, doxorubicin, dexamethasone, and cyclophosphamide. Provided that the agents retain their efficacy, compositions comprising other combinations can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form. By way of example, vincristine, doxorubicin, and cyclophosphamide could be provided as one composition and dexamethasone as a second.
  • In some embodiments, at least two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection, vincristine, dexamethasone, and thalidomide. Provided that the agents retain their efficacy, compositions comprising other combinations can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form. By way of example, vincristine, dexamethasone, and thalidomide could be provided as one composition and doxorubicin HCl liposome injection as a second.
  • In some embodiments, at least two pharmaceutical compositions are provided: a first comprising a therapeutically effective amount of an anti-CS1 antibody such as HuLuc63 and a second comprising a therapeutically effective amount of doxorubicin HCl liposome injection and bortezomib. Provided that the agents retain their efficacy, compositions comprising other combinations can be prepared depending in part, on dosage, route of administration, and whether the agents are provided in a solid, semi-solid or liquid form. By way of example, doxorubicin HCl liposome injection can be provided as one composition and bortezomib as a second.
  • In some embodiments, agents with the same mechanism of action as an anti-CS1 antibody, such as HuMax-Cd38 (Genmab) can be provided in the pharmaceutical compositions described herein.
  • In some embodiments, the pharmaceutical compositions comprise therapeutic agents with a mechanism of action that differs from an anti-CS1 antibody. For example, targeted agents that inhibit angiogenesis, including, but not limited to, bevacizumab, sutinib, sorafenib, 2-methoxyestradiol or 2ME2, finasunate, PTK787, vandetanib, aflibercept, volociximab, etaracizumab (MEDI-522), cilengitide, can be used in the pharmaceutical compositions described herein. In other embodiments, agents that inhibit growth factor signaling, including, but not limited to, erlotinib, cetuximab, panitumumab, gefitinib, trastuzumab, TKI258, CP-751,871, atacicept, can be used in the pharmaceutical compositions described herein. In other embodiments, immunomodulators including, but not limited to, rituximab, alemtuzumab, aldesleukine, atlizumab, tocilizumab, temsirolimus, everolimus, NPI-1387, MLNM3897, HCD122, SGN-40, HLL1, huN901-DM1, atiprimod, natalizumab, can be used in the pharmaceutical compositions described herein. In other embodiments, agents that inhibit protein synthesis, folding or degradation, including but not limited to, bortezomib, carfilzomib, NPI-0052, tanespimycin, saquinavir mesylate, ritonavir, nelfinavir mesylate, indinavir sulfate, can be used in the pharmaceutical compositions described herein. In other embodiments, agents that inhibit gene expression including but not limited to, belinostat, LBH589, can be used in the pharmaceutical compositions described herein. In other embodiments, pro-apoptotic agents including but not limited to, mapatumumab, lexatumumab, AMG951, ABT-737, oblimersen, plitidepsin, can be used in the pharmaceutical compositions described herein. In other embodiments, agents that inhibit signal transduction including but not limited to, SCIO-469, P276-00, enzastaurin, tipifarnib, perifosine, imatinib, can be used in the pharmaceutical compositions described herein. In other embodiments, agents with other mechanisms of action including but not limited to, dasatinib, lenalidomide, thalidomide, simvastatin, and celecoxib can be used in the pharmaceutical compositions described herein.
  • The pharmaceutical compositions can exist as a solid, semi-solid, or liquid (e.g., suspensions or aerosols) dosage form. Typically, the compositions are administered in unit dosage forms suitable for single administration of precise dosage amounts. For example, an anti-CS1 antibody can be packaged in dosages ranging from about 1 to 1000 mg. In some embodiments, an anti-CS1 antibody is packaged in a dosage at least about 1 mg, at least about 10 mg, at least about 20 mg, at least about 50 mg, at least about 100 mg, at least about 200 mg, at least about 300 mg, at least about 400 mg, at least about 500 mg, at least about 750 mg, at least about 1000 mg.
  • The compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, nontoxic carriers or diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration. The diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, physiological saline, Ringer's solution, dextrose solution, and Hank's solution.
  • In addition, the pharmaceutical composition or formulation can also include other carriers, adjuvants, or nontoxic, non-therapeutic, nonimmunogenic stabilizers and the like. Effective amounts of such diluent or carrier will be those amounts that are effective to obtain a pharmaceutically acceptable formulation in terms of solubility of components, or biological activity.
  • 5.3 Use of the Pharmaceutical Compositions to Treat Multiple Myeloma
  • The pharmaceutical compositions described herein find use in treating MM.
  • Typically, the compositions can be used to treat Monoclonal Gammopathy of Undetermined Significance (MGUS), smoldering myeloma, asymptomatic MM, and symptomatic MM, ranging from newly diagnosed to late stage relapsed/refractory.
  • The compositions can be combined with other treatment strategies, i.e., autologous stem cell transplantation and allogeneic effector cell transplantation, to develop an effective treatment strategy based on the stage of myeloma being treated (see, e.g., Multiple Myeloma Research Foundation, Multiple Myeloma: Stem Cell Transplantation 1-30 (2004); U.S. Pat. Nos. 6,143,292, and 5,928,639, Igarashi, et al. Blood 2004, 104(1): 170-177, Maloney, et al. 2003, Blood, 102(9): 3447-3454, Badros, et al. 2002, J Clin Oncol., 20:1295-1303, Tricot, et al. 1996, Blood, 87(3):1196-1198; the contents of which are incorporated herein by reference).
  • The staging system most widely used since 1975 has been the Durie-Salmon system, in which the clinical stage of disease (Stage I, II, or III) is based on four measurements (see, e.g., Durie and Salmon, 1975, Cancer, 36:842-854). These four measurements are: (1) levels of monoclonal (M) protein (also known as paraprotein) in the serum and/or the urine; (2) the number of lytic bone lesions; (3) hemoglobin values; and, (4) serum calcium levels. These three stages can be further divided according to renal function, classified as A (relatively normal renal function, serum creatinine value <2.0 mg/dL) and B (abnormal renal function, creatinine value ≧2.0 mg/dL). A new, simpler alternative is the International Staging System (ISS) (see, e.g., Greipp et al., 2003, “Development of an international prognostic index (IPI) for myeloma: report of the international myeloma working group”, The Hematology). The ISS is based on the assessment of two blood test results, beta2-microglobulin (β2-M) and albumin, which separates patients into three prognostic groups irrespective of type of therapy.
  • Administration of the pharmaceutical compositions at selected dosage ranges and routes typically elicits a beneficial response as defined by the European Group for Blood and Marrow transplantation (EBMT). Table 2 lists the EBMT criteria for response.
  • TABLE 2
    EBMT/IBMTR/ABMTR1 Criteria for Response
    Complete Response No M-protein detected in serum or urine by
    immunofixation for a minimum of 6 weeks and
    fewer than 5% plasma cells in bone marrow
    Partial Response >50% reduction in serum M-protein level
    and/or 90% reduction in urine free light chain
    excretion or reduction to <200 mg/24 hrs for 6
    weeks2
    Minimal Response 25-49% reduction in serum M-protein level
    and/or 50-89% reduction in urine free light
    chain excretion which still exceeds 200 mg/24 hrs
    for 6 weeks3
    No Change Not meeting the criteria or either minimal
    response or progressive disease
    Plateau No evidence of continuing myeloma-related
    organ or tissue damage, <25% change in M-
    protein levels and light chain excretion for 3
    months
    Progressive Disease Myeloma-related organ or tissue damage
    continuing despite therapy or its reappearance
    in plateau phase, >25% increase in serum M-
    protein level (>5 g/L) and/or >25% increase in
    urine M-protein level (>200 mg/24 hrs) and/or
    >25% increase in bone marrow plasma cells (at
    least 10% in absolute terms)2
    Relapse Reappearance of disease in patients previously
    in complete response, including detection of
    paraprotein by immunofixation
    1EBMT: European Group for Blood and Marrow transplantation; IBMTR: International Bone Marrow Transplant Registry; ABMTR: Autologous Blood and Marrow Transplant Registry.
    2For patients with non-secretory myeloma only, reduction of plasma cells in the bone marrow by >50% of initial number (partial response) or 25-49% of initial number (minimal response) is required.
    3In non-secretory myeloma, bone marrow plasma cells should increase by >25% and at least 10% in absolute terms; MRI examination may be helpful in selected patients.
  • Additional criteria that can be used to measure the outcome of a treatment include “near complete response” and “very good partial response”. A “near complete response” is defined as the criteria for a “complete response” (CR), but with a positive immunofixation test. A “very good partial response” is defined as a greater than 90% decrease in M protein (see, e.g., Multiple Myeloma Research Foundation, Multiple Myeloma: Treatment Overview 9 (2005)).
  • The degree to which administration of the compositions elicits a response in an individual clinically manifesting at least one symptom associated with MM, depends in part, on the severity of disease, e.g., Stage I, II, or III, and in part, on whether the patient is newly diagnosed or has late stage refractory MM. Thus, in some embodiments, administration of the pharmaceutical composition elicits a complete response.
  • In other embodiments, administration of the pharmaceutical composition elicits a very good partial response or a partial response.
  • In other embodiments, administration of the pharmaceutical composition elicits a minimal response.
  • In other embodiments, administration of the pharmaceutical composition prevents the disease from progressing, resulting in a response classified as “no change” or “plateau” by the EBMT.
  • Routes of administration and dosage ranges for compositions comprising an anti-CS1 antibody such as HuLuc63 and one or more therapeutic agents for treating individuals diagnosed with MM, can be determined using art-standard techniques, such as a standard dose escalation study to identify the MTD (see, e.g., Richardson, et al. 2002, Blood, 100(9):3063-3067, the content of which is incorporated herein by reference).
  • Typically, anti-CS1 antibodies are administered intravenously. Administration of the other therapeutic agents described herein can be by any means known in the art. Such means include oral, rectal, nasal, topical (including buccal and sublingual) or parenteral (including subcutaneous, intramuscular, intravenous and intradermal) administration and will depend in part, on the available dosage form. For example, therapeutic agents that are available in a pill or capsule format typically are administered orally. However, oral administration generally requires administration of a higher dose than does intravenous administration. Determination of the actual route of administration that is best in a particular case is well within the capabilities of those skilled in the art, and in part, will depend on the dose needed versus the number of times per month administration is required.
  • Factors affecting the selected dosage of an anti-CS1 antibody and the therapeutic agents used in the compositions and methods described herein, include, but are not limited to, the type of agent, the age, weight, and clinical condition of the recipient patient, and the experience and judgment of the clinician or practitioner administering the therapy. Generally, the selected dosage should be sufficient to result in no change, but preferably results in at least a minimal change. An effective amount of a pharmaceutical agent is that which provides an objectively identifiable response, e.g., plateau, no change, minimal, partial, or complete, as noted by the clinician or other qualified observer, and as defined by the EBMT.
  • Generally, an anti-CS1 antibody is administered as a separate composition from the composition(s) comprising the therapeutic agents. As discussed above, the therapeutic agents can each be administered as a separate composition, or combined in a cocktail and administered as a single combined composition. In some embodiments, the compositions comprising an anti-CS1 antibody and one or more therapeutic agents are administered concurrently. In other embodiments, an anti-CS1 antibody can be administered prior to the administration of composition(s) comprising the therapeutic agent(s). In yet other embodiments, an anti-CS1 antibody is administered following the administration of composition(s) comprising the therapeutic agent(s).
  • In those embodiments in which the an anti-CS1 antibody is administered prior to or following the administration of the therapeutic agents, determination of the duration between the administration of an anti-CS1 antibody and administration of the agents is well within the capabilities of those skilled in the art, and in part, will depend on the dose needed versus the number of times per month administration is required.
  • Doses of anti-CS1 antibodies used in the methods described herein typically range between 0.5 mg/kg to 20 mg/kg. Optimal doses for the therapeutic agents are the generally accepted efficacious doses, such as those described in the Physician Desk Reference, 56th Ed. (2002), Publisher Medical Economics, New Jersey. Optimal doses for agents not described in the Physician Desk Reference can be determined using a standard dose escalation study to identify the MTD (see, e.g., Richardson, et al. 2002, Blood, 100(9):3063-3067, the contents of which are incorporated herein by reference).
  • In some embodiments, an anti-CS1 antibody is present in a pharmaceutical composition at a concentration, or in a weight/volume percentage, or in a weight amount, suitable for intravenous administration at a dosage rate at least about 0.5 mg/kg, at least about 0.75 mg/kg, at least about 1 mg/kg, at least about 2 mg/kg, at least about 2.5 mg/kg, at least about 3 mg/kg, at least about 4 mg/kg, at least about 5 mg/kg, at least about 6 mg/kg, at least about 7 mg/kg, at least about 8 mg/kg, at least about 9 mg/kg, at least about 10 mg/kg, at least about 11 mg/kg, at least about 12 mg/kg, at least about 13 mg/kg, at least about 14 mg/kg, at least about 15 mg/kg, at least about 16 mg/kg, at least about 17 mg/kg, at least about 18 mg/kg, at least about 19 mg/kg, and at least about 20 mg/kg.
  • 6. EXAMPLES Example 1 HuLuc63 in Combination with Dexamethasone
  • Dexamethasone (Dex) is a corticosteroid that is used extensively for the treatment of MM in multiple lines of therapy. Dex has been used as monotherapy or in combination with a variety of agents, including lenalidomide, thalidomide, velcade or as part of the VAD (vincristine, doxorubicin, dexamethasone) regimen or the DVd liposomal doxorubicin, vincristine, short-schedule dexamethasone regimen. The mechanism of action of Dex involves direct induction of apoptosis of myeloma cells by activation of caspases (Chauhan et al., 1997, Oncogene 15:837-843; Chauhan et al., 1997, J. Biol. Chem. 272, 29995-29997; Chauhan et al., 2001, J. Biol. Chem., 276: 24453-24456).
  • The effect of HuLuc63 and Dex treatment on the expression of CS1 in MM cell lines and mouse xenograft tumors was examined by flow cytometry and immunohistochemistry respectively.
  • In vivo Xenograft Mouse Model: Methods and Results
  • Six- to eight-week old female IcrTac:ICR-Prkdcscid mice obtained from Taconic Farms (Germantown, N.Y.) were inoculated with 1×107 OPM2 or L363 cells (German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany) into the lower right flank. Caliper measurements were performed twice weekly to calculate tumor volume using the following formula: L×W×H/2, where L (length) is the longest side of the tumor in the plane of the animal's back, W (width) is the longest measurement perpendicular to the length and in the same plane and H (height) is taken at the highest point perpendicular to the back of the animal. When tumors reached an average size of about 100 mm3, animals were randomized into 3 groups of 8-10 mice each and were treated with 1 or 10 mg/kg of HuLuc63 or isotype control antibody administered intraperitoneally twice a week for a maximum of 6 doses.
  • Dex was administered intraperitoneally at a dose of 10 mg/kg twice a week for a maximum of 6 doses.
  • Tumor growth was monitored for a period of 1-2 months. Animal work was carried out under NIH guidelines (“Guide for the Care and Use of Laboratory Animals”) using protocols approved by IACUC at PDL BioPharma.
  • CS1 protein expression was examined on the OPM2 multiple myeloma cell line. No significant change in CS1 expression was observed pre- or post-treatment with HuLuc63, Dex, or with both agents. The combination of HuLuc63 with Dex was tested for anti-myeloma activity in vivo. OPM2 tumor-bearing mice were treated with sub-optimal doses of HuLuc63 (1 mg/kg), or isotype control antibody twice weekly for three weeks. Dex was given twice a week at 10 mg/kg to mice receiving either isotype control antibody or HuLuc63. The results showed significant anti-tumor activity of HuLuc63 alone and in combination with Dex (see, e.g., FIG. 1). Mice in the combination treatment group exhibited significantly smaller tumors than in the HuLuc63 monotherapy group (see, e.g., FIG. 1).
  • Example 2 HuLuc63 in Combination with Thalidomide
  • Thalidomide (Thal) is an immunomodulatory drug that is currently approved for the treatment of MM in combination with Dex. The mechanism of action of Thal is not completely understood, but involves inhibition of angiogenesis, inhibition of growth and survival of stromal cells and tumor cells in the bone marrow, and altering the production of factors that influence the survival of myeloma cells (such as IL-6, IL-10, IL-4, IL-5, IL-12, IL-8, TNF-alpha).
  • The effect of HuLuc63 and Thal treatment on the expression of CS1 in MM cell lines and mouse xenograft tumors was examined by flow cytometry and immunohistochemistry respectively, as described above.
  • CS1 protein expression was examined on the L363 multiple myeloma cell line. No significant change in CS1 expression was observed pre- or post-treatment with HuLuc63, Thal or with both agents. The combination of HuLuc63 with Thal was tested for anti-myeloma activity in vivo. L363 tumor-bearing mice were treated with HuLuc63, or isotype control antibody twice weekly for three weeks at 10 mg/kg. Thal was given 5 days a week at 50 mg/kg for a maximum of 15 doses to mice receiving either isotype control antibody or HuLuc63. Significant anti-tumor activity was observed with HuLuc63 alone, Thal alone and HuLuc63 in combination with Thal (see, e.g., FIG. 2). Mice in the combination treatment group exhibited smaller tumors compared to the HuLuc63 and Thal monotherapy groups (see, e.g., FIG. 2).
  • Example 3 HuLuc63 in Combination with Thalidomide/Dexamethasone
  • Thal/Dex combination therapy is currently approved for front-line treatment of MM patients. The combination of HuLuc63 with Thal/Dex was tested for anti-myeloma activity in vivo as described above. The effect of Thal/Dex treatment on the expression of CS1 in MM cell lines and mouse xenograft tumors was examined by immunohistochemistry as described above.
  • L363 tumor-bearing mice were treated with HuLuc63, or isotype control antibody twice weekly for three weeks at 10 mg/kg. Thal was given 5 days a week at 50 mg/kg for a maximum of 15 doses and Dex was given twice a week at 10 mg/kg for a maximum of 6 doses to mice receiving either isotype control antibody or HuLuc63. The results showed significant anti-tumor activity of HuLuc63 alone, Thal/Dex alone and HuLuc63 in combination with Thal/Dex (see e.g., FIG. 3). Mice in the HuLuc63/Thal/Dex combination treatment group exhibited the smallest tumors, on average exhibiting a decrease of 50-70% in tumor size compared to HuLuc63 monotherapy or Thal/Dex therapy (see e.g., FIG. 3).
  • Example 4 HuLuc63 in Combination with Bevacizumab
  • Bevacizumab is a monoclonal antibody that targets the endothelial growth factor VEGF. It works by inhibiting VEGF, resulting in the inhibition of new blood vessel formation in tumors. Bevacizumab is currently approved for the treatment of solid tumors, including metastatic colorectal cancer. Bevacizumab is currently not approved for the use in the treatment of MM.
  • To determine if an anti-angiogenic drug can enhance the antitumor effects of HuLuc63, a combination of HuLuc63 with Bevacizumab was tested for anti-myeloma activity in vivo, as described above. L363 tumor-bearing mice were treated with HuLuc63, or isotype control antibody twice weekly for three weeks at 10 mg/kg. Bevacizumab was given twice a week at 0.5 mg/kg for a maximum of 6 doses to mice receiving either isotype control antibody or HuLuc63. Significant anti-tumor activity of HuLuc63 alone and in combination with Bevacizumab was observed (see e.g., FIG. 4). Bevacizumab alone did not exhibit significant anti-tumor activity. However, mice receiving the combination of HuLuc63 with Bevacizumab exhibited significantly smaller tumors compared to the HuLuc63 monotherapy group, indicating that HuLuc63 and Bevacizumab may act synergistically to cause an anti-myeloma effect.
  • Example 5 Treatment of Patients Diagnosed with Multiple Myeloma
  • Proposed multi-center, open-label, multi-dose, dose escalation studies will be used to evaluate the combination of HuLuc63 and one or more therapeutic agents in patients with multiple myeloma after 1st, 2nd, or 3rd relapse. HuLuc63 will be given by intravenous injection (IV) at up to five dose levels ranging from 2.5 mg/kg to 20 mg/kg in combination with one or more therapeutic agents. Patients will receive HuLuc63 once a week, with each dose infused over 1 hour, once every 10 days or once every two weeks, for a minimum of 4 doses and a maximum of 52 doses.
  • Drug combinations suitable for administration with HuLuc63 include, but are not limited to:
      • 1) Melphalan+prednisone. Melphalan will be given in 28 day cycles, at 8 mg/m2/d on days 1 to 4, for a maximum number of 11 cycles. Prednisone will be given will be given in 28 day cycles, at 60 mg/m2/d on days 1 to 4, for a maximum number of 11 cycles;
      • 2) Melphalan+prednisone+thalidomide. Melphalan will be given in 28 day cycles, at 8 mg/m2/d on days 1 to 4, for a maximum number of 11 cycles. Prednisone will be given will be given in 28 day cycles, at 60 mg/m2/d on days 1 to 4, for a maximum number of 11 cycles. Thalidomide will be given in 28 day cycles, at 200 mg/d, for a maximum number of 11 cycles;
      • 3) Melphalan+prednisone+lenalidomide. Melphalan will be given in 28 to 42 day cycles, at 0.18 to 0.25 mg/kg for 4 days every 4 to 6 weeks, for a maximum number of 9 cycles. Prednisone will be given will be given in 28 to 42 day cycles, at 2 mg/kg for 4 days every 4 to 6 weeks, for a maximum number of 9 cycles, Lenalidomide will be given in 28 to 42 day cycles, at 5 to 10 mg/day, days 1 to 21 every 4 to 6 weeks, for a maximum number of 9 cycles;
      • 4) Melphalan+prednisone+bortezomib. Melphalan will be given in 42 day cycles, at 9 mg/m2/day on days 1 to 4, for a maximum number of 4 cycles. Prednisone will be given will be given in 42 day cycles, 60 mg/m2/d on days 1 to 4, for a maximum number of 4 cycles. Bortezomib will be given in 42 day cycles, at 1.3 mg/m2/day on days 1, 4, 8, 11, 22, 25, 29 and 32, for a maximum number of 4 cycles;
      • 5) Thalidomide+dexamethasone. Thalidomide will be given in 28 day cycles, at 200 mg/d, for a maximum number of 4 to 12 cycles. Dexamethasone will be given at 40 mg/day on days 1 to 4, 9 to 12, and 17 to 20, for a maximum number of 4 to 12 cycles;
      • 6) Cytoxan+prednisone. Cytoxan will be given in 28 day cycles, at 500 mg, once weekly, for a maximum number of 6 cycles. Prednisone will be given in 28 day cycles, at 100 mg/day, every other day, for a maximum number of 6 cycles;
      • 7) Cytoxan+prednisone+thalidomide. Cytoxan will be given in 28 day cycles, at 50 mg twice per day for 21 days, for a maximum number of 7 cycles. Prednisone will be given in 28 day cycles, at 100 mg/day, every other day, for a maximum number of 7 cycles. Thalidomide will be given in 28 day cycles, at 200 mg/d, for a maximum number of 7 cycles;
      • 8) Cytoxan+prednisone+lenalidomide. Cytoxan will be given in 28 clay cycles, at 300 to 700 mg on days 1 and 8, for a maximum number of 4 to 7 cycles. Prednisone will be given in 28 day cycles, at 50 mg/day every other day, for a maximum number of 4 to 7 cycles. Lenalidomide will be given in 28 day cycles, at 25 mg/day on days 1 to 21, for a maximum number of 4 to 7 cycles;
      • 9) Cytoxan+dexamethasone+lenalidomide. Cytoxan will be given in 28 day cycles, at 300 to 700 mg on days 1 and 8, for a maximum number of 4 to 7 cycles. Dexamethasone will be given in 28 day cycles, at 40 mg on days 1 to 4, 9 to 12 and 17 to 20, for a maximum number of 4 to 7 cycles. Lenalidomide will be given in 28 day cycles, at 25 mg/day on days 1 to 21, for a maximum number of 4 to 7 cycles;
      • 10) Cytoxan+prednisone+bortezomib. Cytoxan will be given in 28 day cycles, at 300 mg on days 1, 8, 15, and 22, for a maximum number of 4 to 7 cycles. Prednisone will be given in 28 day cycles, at 100 mg every other day, for a maximum number of 4 to 7 cycles. Bortezomib will be given in 28 day cycles, at 1.5 mg/m2/day, days 1, 8, and 15, for a maximum number of 4 to 7 cycles;
      • 11) Bevacizumab. Bevacizumab will be given in 14 day cycles, at 5 mg/kg, every 2 weeks, for a maximum number of 6 to 26 cycles;
      • 12) Tanespimycin+bortezomib. Tanespimycin will be given in 21 day cycles, at 100 to 340 mg/m2, twice weekly for 2 weeks, for a maximum of 4 to 8 cycles. Bortezomib will be given in 21 day cycles, at 0.7 to 1.3 mg/m2, twice weekly for 2 weeks, for a maximum number of 4 to 8 cycles;
      • 13) Doxil+bortezomib. Doxil will be given in 21 day cycles, at 30 mg/m2/day, at day 4, for a maximum number of 4 to 8 cycles. Bortezomib will be given in 21 day cycles, at 1.3 mg/m2, days 1, 4, 8 and 11, for a maximum number of 4 to 8 cycles; and,
      • 14) Dexamethasone. Dexamethasone can be given in 28 day cycles, at 40 mg/day, at days 1 to 4, 9 to 12, and 17 to 20, for a maximum number of 4 to 12 cycles, or alternately, in 28 day cycles, at 40 mg/day for 4 days, in 28 day cycles.
  • The above drug combinations can be given orally or by IV.
  • After 8-12 weeks of therapy, EBMT criteria will be assessed. If a patient has progressive disease, HuLuc63 will be discontinued and the other drug combinations may be withdrawn or continued at the discretion of the site investigator. If the patient has responded or has stable disease at Week 8-12, dosing with HuLuc63 and the drug combination will continue so that a maximum of 52 weeks of treatment are completed or disease progression occurs.
  • Approximately 15 to 30 patients in 5 cohorts will be enrolled for each drug combination. Each cohort will begin with 3 patients. If no dose-limiting toxicity (DLT) is noted within the first 4 weeks of treatment in any patient, enrollment will begin in the next higher cohort. If one patient has a DLT, 3 additional patients will be enrolled in the cohort. If no other patient in the cohort has a DLT, escalation to the next cohort may proceed. If a second patient in a cohort has a DLT, the maximum tolerated dose (MTD) has been reached.
  • A dose-limiting toxicity (DLT) is defined using the National Cancer Center Institute Common Toxicity Criteria Version 3.0 (NCI CTCAE v3.0) as a grade 4 hematologic toxicity or hyperbilirubinemia, or a grade 3 toxicity in any other system considered related to HuLuc63 or the combination of HuLuc63 and any of the drugs listed in Table 1. For dose escalation to the next cohort, 3 assessable patients must complete their first 4 weeks of dosing. If a DLT occurs, an additional three assessable patients will be accrued. Patients will be monitored for safety by assessing adverse events categorized by NCI CTCAE v3.0 and patients will be monitored for clinical activity using EBMT. The maximally tolerated dose (MTD) is defined as the highest dose studied for which the incidence of DLTs is <33%. The highest tolerated dose will be HuLuc63 at 20 mg/kg combined with the drugs at the doses listed above if no dose limiting toxicities are observed.
  • All publications, patents, patent applications and other documents cited in this application are hereby incorporated by reference in their entireties for all purposes to the same extent as if each individual publication, patent, patent application or other document were individually indicated to be incorporated by reference for all purposes.
  • While various specific embodiments have been illustrated and described, it will be appreciated that various changes can be made without departing from the spirit and scope of the invention(s).

Claims (33)

1. A pharmaceutical composition suitable for treating multiple myeloma in a subject, comprising a therapeutically effective amount of HuLuc63 in combination with one or more therapeutic agents, wherein said composition is capable of being administered in a single or multiple dose regimen.
2. A pharmaceutical composition suitable for treating multiple myeloma in a subject, comprising a therapeutically effective amount of HuLuc63 in combination with a one or more targeted agents, conventional chemotherapeutic agents, hormonal therapy agents, or supportive care agents, and/or combinations thereof, wherein said composition is capable of being administered in a single or multiple dose regimen.
3. The pharmaceutical composition according to claim 2, in which one or more conventional therapeutic agents are selected from the group consisting of thalidomide, vincristine, carmustine, melphalan, cyclophosphamide, doxorubicin, doxorubicin HCl liposome injection, cisplatin, etoposide, ara-C, and one or more hormonal therapy agents selected from the group consisting of dexamethasone and prednisone.
4. The pharmaceutical composition according to claim 2, in which one or more one or more targeted agents are selected from the group consisting of anti-angiogenic agents, inhibitors of growth factor signaling, immunomodulators, inhibitors of protein synthesis, folding and/or degradation, inhibitors of gene expression, pro-apoptotic agents, agents that inhibit signal transduction, other agents, and combinations thereof,
5. The pharmaceutical composition according to claim 3, comprising dexamethasone.
6. The pharmaceutical composition according to claim 3, comprising dexamethasone and thalidomide.
7. The pharmaceutical composition according to claim 3, comprising vincristine, doxorubicin and dexamethasone.
8. The pharmaceutical composition according to claim 3, comprising doxorubicin HCl liposome injection, vincristine and dexamethasone.
9. The pharmaceutical composition according to claim 3, comprising cyclophosphamide.
10. The pharmaceutical composition according to claim 3, comprising dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide and etoposide.
11. The pharmaceutical composition according to claim 3, comprising vincristine, doxorubicin, dexamethasone, and cyclophosphamide.
12. The pharmaceutical composition according to claim 3, comprising doxorubicin HCl liposome injection, vincristine, dexamethasone, and thalidomide.
13. The pharmaceutical composition according to claim 1 or 2, in which HuLuc63 is in an injectable form.
14. The pharmaceutical composition according to claim 12, containing from about 0.5 mg/kg to about 20 mg/kg of HuLuc63.
15. The pharmaceutical composition according to claim 1 or 2, in which administration of said pharmaceutical composition elicits a complete response.
16. The pharmaceutical composition according to claim 1 or 2, in which administration of said pharmaceutical composition elicits a very good partial response.
17. The pharmaceutical composition according to claim 1 or 2, in which administration of said pharmaceutical composition elicits a partial response.
18. The pharmaceutical composition according to claim 1 or 2, in which administration of said pharmaceutical composition elicits a minimal response.
19. A method for increasing sensitivity of a multiple myeloma cell to a therapeutic agent, comprising contacting the cell with a therapeutically effective amount of HuLuc63.
20. The method according to claim 19, further comprising contacting the cell with a therapeutically effective amount of one or more conventional therapeutic agents selected from the group consisting of thalidomide, vincristine, carmustine, melphalan, cyclophosphamide, doxorubicin, doxorubicin HCl liposome injection, cisplatin, etoposide, ara-C, and one or more hormonal therapy agents selected from the group consisting of dexamethasone and prednisone.
21. The method according to claim 19, wherein the multiple myeloma cell is resistant to the therapeutic agent.
22. The method according to claim 21, further comprising contacting said resistant cell with a therapeutic agent that the cell is sensitive to, said agent selected from the group consisting of one or more conventional therapeutic agents selected from the group consisting of thalidomide, vincristine, carmustine, melphalan, cyclophosphamide, doxorubicin, doxorubicin HCl liposome injection, cisplatin, etoposide, ara-C, and one or more hormonal therapy agents selected from the group consisting of dexamethasone and prednisone.
23. A method of treating multiple myeloma in a subject, the method comprising administering a first pharmaceutical composition comprising HuLuc63 and a second pharmaceutical composition comprising one or more targeted agents, conventional chemotherapeutic agents, hormonal therapy agents, or supportive care agents, or combinations thereof, wherein said compositions are capable of being administered in single or multiple dose regimens.
24. A method of treating multiple myeloma in a subject, the method comprising administering a therapeutically effective amount of HuLuc63 in combination with one or more targeted agents selected from the group consisting of anti-angiogenic agents, inhibitors of growth factor signaling, immunomodulators, inhibitors of protein synthesis, folding and/or degradation, inhibitors of gene expression, pro-apoptotic agents, agents that inhibit signal transduction, other agents, or combinations thereof, wherein said compositions are capable of being administered in single or multiple dose regimens.
25. The method according to claim 24, further comprising one or more conventional chemotherapeutic agents and one or more hormonal agents.
26. The method according to claim 25, in which one or more conventional therapeutic agents are selected from the group consisting of thalidomide, vincristine, carmustine, melphalan, cyclophosphamide, doxorubicin, doxorubicin HCl liposome injection, cisplatin, etoposide, ara-C, and one or more hormonal therapy agents selected from the group consisting of dexamethasone and prednisone.
27. The method according to claim 23, in which HuLuc63 is administered intravenously at a dosage from approximately 0.5 mg/kg to approximately 20 mg/kg.
28. The method according to claim 23, in which HuLuc63 is administered concurrently with the pharmaceutical composition comprising one or more of said targeted agents, conventional chemotherapeutic agents, hormonal therapy agents, supportive care agents, or combinations thereof.
29. The method according to claim 28, in which said administration elicits a complete response.
30. The method according to claim 28, in which said administration elicits a very good partial response.
31. The method according to claim 28, in which said administration elicits a partial response.
32. The method according to claim 28, in which said administration elicits a minimal response.
33. A method of treating multiple myeloma in a subject, comprising administering a therapeutically effective amount of HuLuc63, a therapeutically effective amount of one or more targeted agents, conventional chemotherapeutic agents, hormonal therapy agents, supportive care agents, or combinations thereof, and a pharmaceutically acceptable carrier, wherein said composition is capable of being administered in a single or multiple dose regimen.
US12/903,051 2006-08-07 2010-10-12 Compositions and methods using anti-cs1 antibodies to treat multiple myeloma Abandoned US20110165154A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/903,051 US20110165154A1 (en) 2006-08-07 2010-10-12 Compositions and methods using anti-cs1 antibodies to treat multiple myeloma
US15/091,977 US20160206734A1 (en) 2006-08-07 2016-04-06 Compositions and methods using anti-cs1 antibodies to treat multiple myeloma

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US83625006P 2006-08-07 2006-08-07
US85614406P 2006-11-01 2006-11-01
US11/835,245 US7842293B2 (en) 2006-08-07 2007-08-07 Compositions and methods using anti-CS1 antibodies to treat multiple myeloma
US12/903,051 US20110165154A1 (en) 2006-08-07 2010-10-12 Compositions and methods using anti-cs1 antibodies to treat multiple myeloma

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/835,245 Continuation US7842293B2 (en) 2006-08-07 2007-08-07 Compositions and methods using anti-CS1 antibodies to treat multiple myeloma

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/091,977 Continuation US20160206734A1 (en) 2006-08-07 2016-04-06 Compositions and methods using anti-cs1 antibodies to treat multiple myeloma

Publications (1)

Publication Number Publication Date
US20110165154A1 true US20110165154A1 (en) 2011-07-07

Family

ID=39033619

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/835,245 Active US7842293B2 (en) 2006-08-07 2007-08-07 Compositions and methods using anti-CS1 antibodies to treat multiple myeloma
US12/903,051 Abandoned US20110165154A1 (en) 2006-08-07 2010-10-12 Compositions and methods using anti-cs1 antibodies to treat multiple myeloma
US15/091,977 Abandoned US20160206734A1 (en) 2006-08-07 2016-04-06 Compositions and methods using anti-cs1 antibodies to treat multiple myeloma

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/835,245 Active US7842293B2 (en) 2006-08-07 2007-08-07 Compositions and methods using anti-CS1 antibodies to treat multiple myeloma

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/091,977 Abandoned US20160206734A1 (en) 2006-08-07 2016-04-06 Compositions and methods using anti-cs1 antibodies to treat multiple myeloma

Country Status (18)

Country Link
US (3) US7842293B2 (en)
EP (2) EP2068930B1 (en)
JP (4) JP2010500370A (en)
AU (1) AU2007281682B2 (en)
BR (1) BRPI0716475A2 (en)
CA (1) CA2660102C (en)
CY (1) CY1113309T1 (en)
DK (1) DK2068930T3 (en)
ES (1) ES2391790T3 (en)
HK (1) HK1129849A1 (en)
HR (1) HRP20120934T1 (en)
IL (1) IL196920A (en)
MX (1) MX2009001440A (en)
NO (1) NO345083B1 (en)
NZ (1) NZ574979A (en)
PL (1) PL2068930T3 (en)
PT (1) PT2068930E (en)
WO (1) WO2008019376A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014004376A2 (en) 2012-06-26 2014-01-03 Del Mar Pharmaceuticals Methods for treating tyrosine-kinase-inhibitor-resistant malignancies in patients with genetic polymorphisms or ahi1 dysregulations or mutations employing dianhydrogalactitol, diacetyldianhydrogalactitol, dibromodulcitol, or analogs or derivatives thereof
WO2021092044A1 (en) 2019-11-05 2021-05-14 Bristol-Myers Squibb Company M-protein assays and uses thereof
US11491154B2 (en) 2013-04-08 2022-11-08 Dennis M. Brown Therapeutic benefit of suboptimally administered chemical compounds

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8632772B2 (en) * 2006-08-07 2014-01-21 Dana-Farber Cancer Institute Methods of treating multiple myeloma using combination therapies based on anti-CS1 antibodies
NZ574979A (en) * 2006-08-07 2012-06-29 Abbott Biotherapeutics Corp Compositions and methods using anti-cs1 antibodies to treat multiple myeloma
EP3384919A1 (en) * 2008-06-02 2018-10-10 Dana Farber Cancer Institute, Inc. Cs1 peptides
JP2012507555A (en) 2008-10-31 2012-03-29 アボット バイオセラピューティクス コーポレイション Use of anti-CS1 antibodies in the treatment of rare diseases
EP2785731B1 (en) 2011-11-30 2019-02-20 AbbVie Biotechnology Ltd Vectors and host cells comprising a modified sv40 promoter for protein expression
EP2914278B1 (en) 2012-11-05 2021-06-02 Dana-Farber Cancer Institute, Inc. Xbp1, cd138, and cs1 peptides, pharmaceutical compositions that include the peptides, and methods of using such peptides and compositions
WO2015069703A1 (en) 2013-11-06 2015-05-14 Bristol-Myers Squibb Company Immunotherapeutic dosing regimens and combinations thereof
EP3066128B1 (en) 2013-11-06 2018-10-31 Bristol-Myers Squibb Company Combination of anti-kir and anti-cs1 antibodies to treat multiple myeloma
US10011657B2 (en) 2014-10-31 2018-07-03 Abbvie Biotherapeutics Inc. Anti-CS1 antibodies and antibody drug conjugates
JP2017537927A (en) 2014-12-04 2017-12-21 ブリストル−マイヤーズ スクイブ カンパニーBristol−Myers Squibb Company Combination of anti-CS1 and anti-PD1 antibodies to treat cancer (myeloma)
CA2990478A1 (en) 2015-06-29 2017-01-05 Bristol-Myers Squibb Company Immunotherapeutic dosing regimens comprising pomalidomide and an anti-cs1 antibody for treating cancer
US10537585B2 (en) 2017-12-18 2020-01-21 Dexcel Pharma Technologies Ltd. Compositions comprising dexamethasone

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050025763A1 (en) * 2003-05-08 2005-02-03 Protein Design Laboratories, Inc. Therapeutic use of anti-CS1 antibodies
US20060024296A1 (en) * 2003-05-08 2006-02-02 Protein Design Labs, Inc. Therapeutic use of anti-CS1 antibodies
US20080124332A1 (en) * 2006-08-07 2008-05-29 Pdl Biopharma, Inc. Compositions and methods using anti-cs1 antibodies to treat multiple myeloma

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL112969A (en) 1994-03-17 2001-05-20 Baxter Int Pharmaceutical compositions for the treatment of cancer comprising allogenic lymphocytes or their combination with a t-cell activator
DE69633668T2 (en) 1995-05-25 2006-03-09 Shimon Slavin ALLOGENIC CELL THERAPY FOR CANCER DUE TO ALLOGENIC STEM CELLS TRANSPLANTATION
US8632772B2 (en) * 2006-08-07 2014-01-21 Dana-Farber Cancer Institute Methods of treating multiple myeloma using combination therapies based on anti-CS1 antibodies
US20080095768A1 (en) 2006-08-07 2008-04-24 Pdl Biopharma, Inc. Use of allogeneic effector cells and anti-cs1 antibodies for selective killing of multiple myeloma cells

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050025763A1 (en) * 2003-05-08 2005-02-03 Protein Design Laboratories, Inc. Therapeutic use of anti-CS1 antibodies
US20060024296A1 (en) * 2003-05-08 2006-02-02 Protein Design Labs, Inc. Therapeutic use of anti-CS1 antibodies
US20080124332A1 (en) * 2006-08-07 2008-05-29 Pdl Biopharma, Inc. Compositions and methods using anti-cs1 antibodies to treat multiple myeloma
US7842293B2 (en) * 2006-08-07 2010-11-30 Facet Biotech Corporation Compositions and methods using anti-CS1 antibodies to treat multiple myeloma

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Beckman et al. (Can. 109:170-179 (2007)) *
Cespdes et al. (Clin. Transl. Oncol. 8(5):318-329 (2006)) *
Dennis (Nature 442:739-741 (2006)) *
Fujimori et al. (J. Nuc. Med. 31:1191-1198 (1990)) *
Rudnick et al. (Can. Biotherp. & Radiopharm. 24: 155-162 (2009)) *
Talmadge et al. (Am. J. Pathol 170(3):793-804 (2007)) *
Thurber et al. (Adv. Drug Deliv. Rev. 60:1421-1434 (2008)) *
Voskoglou-Nomikos (Clin. Can. Res. 9:4227-4239 (2003)) *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014004376A2 (en) 2012-06-26 2014-01-03 Del Mar Pharmaceuticals Methods for treating tyrosine-kinase-inhibitor-resistant malignancies in patients with genetic polymorphisms or ahi1 dysregulations or mutations employing dianhydrogalactitol, diacetyldianhydrogalactitol, dibromodulcitol, or analogs or derivatives thereof
US11491154B2 (en) 2013-04-08 2022-11-08 Dennis M. Brown Therapeutic benefit of suboptimally administered chemical compounds
WO2021092044A1 (en) 2019-11-05 2021-05-14 Bristol-Myers Squibb Company M-protein assays and uses thereof

Also Published As

Publication number Publication date
JP2010500370A (en) 2010-01-07
EP2068930B1 (en) 2012-09-19
PL2068930T3 (en) 2013-02-28
JP2017025077A (en) 2017-02-02
EP2425849A3 (en) 2012-05-09
IL196920A0 (en) 2011-08-01
US20160206734A1 (en) 2016-07-21
AU2007281682A1 (en) 2008-02-14
NZ574979A (en) 2012-06-29
BRPI0716475A2 (en) 2016-05-03
AU2007281682B2 (en) 2013-05-16
HK1129849A1 (en) 2009-12-11
HRP20120934T1 (en) 2012-12-31
JP6005205B2 (en) 2016-10-12
ES2391790T3 (en) 2012-11-29
CA2660102A1 (en) 2008-02-14
DK2068930T3 (en) 2012-10-29
IL196920A (en) 2013-11-28
US20080124332A1 (en) 2008-05-29
JP5775125B2 (en) 2015-09-09
NO345083B1 (en) 2020-09-21
WO2008019376A3 (en) 2008-07-10
CA2660102C (en) 2017-02-21
NO20090969L (en) 2009-04-28
US7842293B2 (en) 2010-11-30
WO2008019376A2 (en) 2008-02-14
EP2068930A2 (en) 2009-06-17
JP2015157823A (en) 2015-09-03
EP2425849A2 (en) 2012-03-07
CY1113309T1 (en) 2016-04-13
JP2014040421A (en) 2014-03-06
PT2068930E (en) 2012-10-23
MX2009001440A (en) 2009-04-15

Similar Documents

Publication Publication Date Title
US7842293B2 (en) Compositions and methods using anti-CS1 antibodies to treat multiple myeloma
US20170342150A1 (en) Method of treating multiple myeloma using combination therapies based on anti-cs1 antibodies
US20080095768A1 (en) Use of allogeneic effector cells and anti-cs1 antibodies for selective killing of multiple myeloma cells
CN101534860B (en) Compositions and methods using anti-cs1 antibodies to treat multiple myeloma

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBOTT BIOTHERAPEUTICS CORP., CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:FACET BIOTECH CORPORATION;REEL/FRAME:025445/0807

Effective date: 20100908

STCB Information on status: application discontinuation

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