US20060045877A1 - Immunoconjugates targeting syndecan-1 expressing cells and use thereof - Google Patents

Immunoconjugates targeting syndecan-1 expressing cells and use thereof Download PDF

Info

Publication number
US20060045877A1
US20060045877A1 US10/975,434 US97543404A US2006045877A1 US 20060045877 A1 US20060045877 A1 US 20060045877A1 US 97543404 A US97543404 A US 97543404A US 2006045877 A1 US2006045877 A1 US 2006045877A1
Authority
US
United States
Prior art keywords
immunoconjugate
cells
antibody
targeting
syndecan
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
US10/975,434
Inventor
Viktor Goldmakher
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.)
Biotest AG
Immunogen Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US10/975,434 priority Critical patent/US20060045877A1/en
Assigned to IMMUNOGEN, INC. reassignment IMMUNOGEN, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GOLDMAKHER, VIKTOR S.
Assigned to BIOTEST AG reassignment BIOTEST AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: IMMUNOGEN, INC.
Publication of US20060045877A1 publication Critical patent/US20060045877A1/en
Priority to US11/735,644 priority patent/US8840898B2/en
Assigned to BIOTEST AG (ONE-HALF INTEREST) reassignment BIOTEST AG (ONE-HALF INTEREST) CORRECTIVE ASSIGNMENT TO CORRECT THE CONVEYANCE OF INTEREST FROM IMMUNOGEN, INC. TO BIOTEST AG PREVIOUSLY RECORDED ON REEL 015929 FRAME 0785. ASSIGNOR(S) HEREBY CONFIRMS THE IMMUNOGEN, INC., CONVEYS AN UNDIVIDED ONE-HALF OF THE RIGHT, TITLE AND INTEREST IN AND TO THIS APPLICATION TO BIOTEST AG.. Assignors: IMMUNOGEN, INC. (ONE-HALF INTEREST)
Priority to US14/327,888 priority patent/US20150010585A1/en
Assigned to IMMUNOGEN, INC. reassignment IMMUNOGEN, INC. CHANGE OF ADDRESS FOR IMMUNOGEN, INC Assignors: IMMUNOGEN, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention pertains to immunoconjugates and their use in different indications.
  • the present invention relates to immunoconjugates, the delivery of their effector molecule(s) to target sites and the site specific release of the effector molecule(s) in, at or near target cells, tissues and organs.
  • the present invention relates to immunoconjugates comprising one or more syndecan-1 targeting agent and highly potent effector molecules, which are attached to the targeting agent.
  • the effector molecule is activated by cleavage/dissociation from the targeting agent portion of the immunoconjugate in, at or near the target cells, tissues or organs.
  • Immunoconjugates comprise at least one targeting agent attached to at least one effector molecule.
  • Such immunoconjugates can be categorized according to their effector molecules into, for example, drug immunoconjugates, immunotoxin conjugate and radioimmunoconjugates (Payne, 2003).
  • Efficiency in killing cells is one key factor in the usefulness of an immunoconjugate.
  • Efficiency can be influenced by the potency of the effector molecule (Blättler and Chari, 2001), by the ability of the effector to retain its potency (Chari et al., 1995; Liu et al., 1996; Ojima et al., 2002; Senter et al., 2002 and Sievers and Linenberger, 2001), By the tumor accessibility (Charter, 2001), by the level of expression of the target antigen on the target cell, targeting agent affinity, and by the ability of the target cell to internalize the immunoconjugate (Wargalla, 1989). In the initial development period of immunoconjugates, the efficiencies of conjugates having a drug as an effector molecule often were disappointing compared to the free drug.
  • cytotoxic drug or the toxin is highly cytotoxic, the immunoconjugate has to reach its target site without adversely affecting the host on its way. Accordingly, if the immunoconjugate circulates, for example, in the bloodstream to reach its target site, then this should occur without a substantial release of active drug. Thus, ideally, a highly cytotoxic drug or toxin of an immunoconjugate is only activated upon reaching its target.
  • the immunoconjugate has to be able to selectively interact with the target cells. Particularly for in vivo applications, it is critical that the immunoconjugate does not have substantial adverse effects on essential non-target cells. Thus, both the cellular target of the immunoconjugate and the targeting agent of the immunoconjugate have to be carefully selected to ensure specificity (Blättler and Chari, 2001).
  • immunoconjugates comprising targeting antibodies demonstrate pharmacokinetic and tissue distribution characteristics similar to those of corresponding antibodies (Xie, 2003).
  • MYLOTARG a conjugate of an anti-CD33 humanized monoclonal antibody and the highly cytotoxic DNA-damaging agent calicheamicin, has been recently approved by the FDA as the first drug immunoconjugate for clinical treatment of certain indications of myeloid leukemia (Bross, 2001; Hamann, 2002; Dowell, 2001).
  • At least one targeting agent selectively targeting cell-surface expressed syndecan-1
  • the cytotoxicity of the effector molecule, in its native form, on cells targeted by said targeting antibody may be higher or about the same as the cytotoxicity of the immunoconjugate on said targeted cells.
  • the effector molecule may have, in its native form, a potency of about 10 ⁇ 14 -10 ⁇ 7 , preferably a potency of about 10 ⁇ 13 -10 ⁇ 7 M, of about 10 ⁇ 12 -10 ⁇ 7 M, of about 10 ⁇ 12 -10 ⁇ 8 M, most preferably of about 10 ⁇ 11 -10 ⁇ 10 M, which includes any narrower potency ranges encompassed by the ranges specified above, such as, but not limited to, a potency of about 10 ⁇ 11 -10 ⁇ 10 M.
  • the effector molecule may be a maytansinoid, in particular DM1, DM3 or DM4, a CC1065 analogue, a calicheamicin or a taxane.
  • the effector molecule may have a molecular weight of less than 5 kDa, in particular less than 2 kDa, more in particular less than 1 kDa and in between about 600 and about 800 Da.
  • the targeting agent may be a targeting antibody, which includes fragments of antibodies, or non-immunoglobulin targeting molecule.
  • the targeting antibody may be derived from an antibody that internalizes poorly.
  • the targeting antibody may be derived from the antibody B-B4.
  • the present invention is also directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of the immunoconjugate described above and one or more pharmaceutically acceptable excipients.
  • the present invention is also directed to a kit comprising, in separate containers, pharmaceutical compositions for use in combination to inhibit, delay and/or prevent the growth of tumors and/or spread of tumor cells, wherein one container comprises an effective amount of above described pharmaceutical composition, and wherein, a separate container comprises a second pharmaceutical composition comprising an effective amount of an agent for the inhibition, delay and/or prevention of the growth of tumors and/or spread of tumor cells, and one or more pharmaceutically acceptable excipients.
  • the agent in said second pharmaceutical composition may be a chemotherapeutic agent or another immunoconjugate.
  • the invention is directed to a method for treating, inhibiting, delaying and/or preventing the growth of tumor cells in a cell culture containing syndecan-1 expressing tumor cells and non-tumor cells, comprising administering an effective amount of the above described immunoconjugate.
  • the effective amount induces, in certain embodiments, cell death or continuous cell cycle arrest of said syndecan-1 expressing tumor cells.
  • the cells in said cell culture may be obtained from a cancer patient and may, after administration of said effective amount of said immunoconjugate, be reimplanted into said cancer patient.
  • the cells in said cell culture may be isolated from a patient suffering from an hematologic malignancy and/or a solid tumor comprising syndecan-1 expressing cells, in particular from a patient suffering from one of the following: multiple myeloma, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma, Hodgkin's and non-Hodgkin's lymphoma, chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML), a solid tissue sarcoma or a colon carcinoma.
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphoblastic leukemia
  • AML acute myeloblastic leukemia
  • the present invention is also directed to a method of inhibiting, delaying and/or preventing the growth of a tumor comprising syndecan-1 expressing tumor cells and/or spread of syndecan-1 expressing tumor cells in a patient in need thereof, comprising
  • the invention is also directed to a method for inhibiting, delaying and/or preventing the growth of a tumor and/or spread of malignant tumor cells in a patient in need thereof, comprising
  • the present invention is also directed to a method for treating a subject having a condition that would benefit from the selective suppression of myeloma cell survival, the method comprising:
  • FIGS. 1A-1F show the expression of CD138 in multiple myeloma (MM) cells.
  • FIGS. 2A-2C show the effect of B-B4-DM1 in comparison with that produced by the naked antibody or by non-conjugated drug on survival of CD138 + and CD138 ⁇ MM cells.
  • FIGS. 3A to 3 C show the inhibitory effect of B-B4-DM1 on proliferation of CD138 + and CD138 ⁇ cells adherent to bone marrow stromal cells (BMSCs).
  • BMSCs bone marrow stromal cells
  • FIGS. 4A and 4B show the survival and cell cycle effects of B-B4-DM1 on CD138 + MM cells.
  • FIGS. 5A to 5 D show the activity of B-B4-DM1 in a tumor xenograft model of human CD138 + multiple myeloma.
  • FIGS. 6A to F show the activity of B-B4-DM1 on large tumor xenografts of human CD138 + OPM multiple myeloma.
  • FIG. 7A shows the expression of GFP in the cells (GFP stands for Green Fluorescent Protein).
  • FIGS. 7B to 7 F show the activity of B-B4-DM1 on GFP + human MM xenografts.
  • FIGS. 8A to 8 C show that B-B4-DM1 reduces MM tumor burden in SCID-hu hosts implanted with patient MM cells.
  • FIGS. 9A to 9 C show that B-B4-DM1 increases survival in SCID-hu hosts implanted with the Ocy-My5 MM cell line.
  • the present invention relates to immunoconjugates and the delivery of their effector molecule(s) to target sites and the site specific release of effector(s) molecule in, at or near target cells, tissues and organs. More particularly, the present invention relates to immunoconjugates comprising syndecan-1 targeting agents and potent effector molecules.
  • the effector molecules are covalently linked, chelated or otherwise associated with the targeting agent.
  • the effector molecules may be activated by cleavage/dissociation from the targeting agent portion of the immunoconjugate at the target site.
  • the immunoconjugates according to the present invention are administered to a subject in need of therapeutic treatment or to cells isolated from such a subject in need of therapeutic treatment.
  • the effector molecule or molecules may be released from the immunoconjugate by cleavage/dissociation in, at or close to the target cell, tissue or organ.
  • the immunoconjugate comprises the antibody B-B4 and at least one highly cytotoxic drug or toxin as an effector molecule and is administered to a patient with cancer.
  • a therapeutically effective amount of the immunoconjugate is administered intravenously to a patient so that it concentrates in the cancer cells.
  • the effector molecule or molecules are released from the antibody target by natural means.
  • the immunoconjugate comprises the antibody B-B4 and at least one highly cytotoxic drug and is administered to a cell population isolated from a patient with cancer.
  • a cell death or continuous cell cycle arrest inducing amount of the immunoconjugate is administered to the cell population so that it concentrates in the cancerous cells.
  • the effector molecule or molecules are released from the targeting antibody by natural means or external means to induce cell death or continuous cell cycle arrest in the cancer cells.
  • the immunoconjugate comprises the antibody B-B4 and at least one highly cytotoxic drug or an immunotoxin as an effector molecule and is administered to a patient with cancer.
  • a therapeutically effective amount of the immunoconjugate is administered intravenously to a patient so that it concentrates in the cancerous cells.
  • the effector molecule or molecules are released from the antibody target by an external means to induce cell death or continuous cell cycle arrest in the cancer cells.
  • a targeting agent according to the present invention is able to associate with a molecule expressed by a target cell and includes peptides and non-peptides.
  • targeting agents according to the present invention include targeting antibodies and non-immunoglobulin targeting molecules, which may be based on non-immunoglobulin proteins, including, but not limited to, AFFILIN® molecules, ANTICALINS® and AFFIBODIES®.
  • Non-immunoglobulin targeting molecules also include non-peptidic targeting molecules including targeting DNA and RNA oligonucleotides (aptamers).
  • a targeting antibody according to the present invention is or is based on a natural antibody or is produced synthetically or by genetic engineering and binds to an antigen on a cell or cells (target cell(s)) of interest.
  • a targeting antibody according to the present invention includes a monoclonal antibody, a polyclonal antibody, a multispecific antibody (for example, a bispecific antibody), or an antibody fragment.
  • the targeting antibody may be engineered to, for example, improve its affinity to the target cells (Ross, 2003) or diminish its immunogenicity.
  • the targeting antibody may be attached to a liposomal formulation including effector molecules (Carter, 2003).
  • An antibody fragment comprises a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody.
  • antibody fragments according to the present invention include Fab, Fab′, F(ab′) 2 , and Fv fragments, but also diabodies; domain antibodies (dAb) (Ward, 1989; U.S. Pat. No. 6,005,079); linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • a single chain variable fragment antibody scFv
  • the heavy and light chains VH and VL
  • VH and VL can be linked by a short amino acid linker having, for example, the sequence (glycine 4 serine) n , which has sufficient flexibility to allow the two domains to assemble a functional antigen binding pocket. Addition of various signal sequences may allow for more precise targeting of the targeting antibody.
  • variable regions for constructing the scFv can, if a mAb against a target of interest is available, be obtained by RT-PCR which clones out the variable regions from mRNA extracted from the parent hybridoma. Alternatively, the scFv can be generated de novo by phage display technology (Smith, 2001).
  • a bispecific antibody according to the present invention may, for example, have at least one arm that is reactive against a target tissue and one arm that is reactive against a linker moiety (U.S. Patent Publication 20020006379).
  • a bispecific antibody according to the present invention may also bind to more than one antigen on a target cell (Carter, 2003).
  • An antibody according to the present invention may be modified by, for example, introducing cystein residues to introduce thiol groups (Olafsen, 2004).
  • the targeting antibody may be derived from any source and may be, but is not limited to, a camel antibody, a murine antibody, a chimeric human/mouse antibody or a chimeric human/monkey antibody, in particular, a chimeric human/monkey antibody with the monkey portion stemming from a cynomolgus monkey.
  • Humanized antibodies are antibodies that contain sequences derived from a human-antibody and from a non-human antibody and are also within the scope of the present invention. Suitable methods for humanizing antibodies include CDR-grafting (complementarity determining region grafting) (EP 0 239 400; WO 91/09967; U.S. Pat. Nos.
  • CDR-grafting the mouse complementarity-determining regions (CDRs) from, for example, mAb B-B4 are grafted into human variable frameworks, which are then joined to human constant regions, to create a human B-B4 antibody.
  • CDRs mouse complementarity-determining regions
  • the resurfacing technology uses a combination of molecular modeling, statistical analysis and mutagenesis to alter the non-CDR surfaces of antibody variable regions to resemble the surfaces of known antibodies of the target host.
  • Strategies and methods for the resurfacing of antibodies, and other methods for reducing immunogenicity of antibodies within a different host, are disclosed, for example, in U.S. Pat. No. 5,639,641.
  • Human antibodies can be made by a variety of methods known in the art including phage display methods. See also U.S. Pat. Nos.
  • Fully human antibodies may also been used. Those antibodies can be selected by the phage display approach, where CD138 or an antigenic determinant thereof is used to selectively bind phage expressing, for example, B-B4 variable regions (see, Krebs, 2001). This approach is advantageously coupled with an affinity maturation technique to improve the affinity of the antibody.
  • the targeting antibody is, in its unconjugated form, moderately or poorly internalizable. Moderate internalization constitutes about 30% to about 75% internalization of antibody, poor internalization constitutes about 0.01% to up to about 30% internalization after 3 hours incubation at 37° C.
  • the targeting antibody binds to CD138, for example, antibodies B-B4, BC/B-B4, B-B2, DL-101, 1 D4, MI15, 1.BB.210, 2Q1484, 5F7, 104-9, 281-2 in particular B-B4.
  • the targeting antibody binds primarily to cell-surface expressed CD138.
  • the targeting antibody does not substantially bind non-cell-surface expressed CD138.
  • a targeting antibody such as “B-B4 targeting antibody”
  • this targeting antibody has the binding specificity of the antibody B-B4.
  • a targeting antibody is said to be “derived from” a specified antibody, this means that this targeting antibody has the binding specificity of this antibody, but might take any form consistent with the above description of a targeting antibody.
  • a targeting antibody is said to do something “selectively” such as “selectively targeting cell-surface expressed syndcan-1” or, to be “selective” for something, this means that there is a significant selectivity (i.e. a higher affinity towards CD138-positive cells compared with CD138-negative cells) for, in case of the example provided, cell-surface expressed syndecan-1, compared to any other antigens and adverse side effects in a given environment are substantially avoided due to this selectivity.
  • Non-immunoglobulin targeting molecules include targeting molecues derived from non-immunoglobulin proteins as well as non-peptidic targeting molecules.
  • Small non-immunoglobulin proteins which are included in this definition are designed to have specific affinities towards, in particular surface expressed CD138.
  • These small non-immunoglobulin proteins include scaffold based engineered molecules such as Affilin® molecules that have a relatively low molecular weight such as between 10 kDa and 20 kDa.
  • Appropriate scaffolds include, for example, gamma crystalline. Those molecules have, in their natural state, no specific binding activity towards the target molecules.
  • Non-immunoglobulin targeting molecules are derived from lipocalins, and include, for example ANTICALINS®, which resemble in structure somewhat immunoglobulins. However, lipocalins are composed of a single polypeptide chain with 160 to 180 amino acid residues.
  • the binding pocket of lipocalins can be reshaped to recognize a molecule of interest with high affinity and specificity (see, for example, Beste et al., 1999).
  • Artificial bacterial receptors such as those marketed under the trademark Affibody® (Affibody AB) are also within the scope of the present invention. These artificial bacterial receptor molecules are small, simple proteins and may be composed of a three-helix bundle based on the scaffold of one of the IgG-binding domains of Protein A ( Staylococcus aureus ). These molecules have binding properties similar to many immunoglobulins, but are substantially smaller, having a molecular weight often not exceeding 10 kDa and are also comparatively stable.
  • Non-peptidic targeting molecules include, but are limited to, to DNA and RNA oligonucleotides that bind to CD138 (aptamers).
  • Effector molecule is a molecule or a derivative, or an analogue thereof that is attached to a targeting agent and exerts a desired effect, for example apoptosis, or another type of cell death, or a continuous cell cycle arrest on the target cell or cells.
  • Effector molecules according to the present invention include molecules that can exert desired effects in a target cell and include, but are not limited to, toxins, drugs, in particular low molecular weight cytotoxic drugs, radionuclides, biological response modifiers, pore-forming agents, cytotoxic enzymes, prodrug activating enzymes, antisense oligonucleotides, antibodies or cytokines as well as functional derivatives or analogues/fragments thereof.
  • the effector increases internal effector delivery of the immunoconjugate, in particular when the natural form of the antibody on which the targeting antibody of the immunoconjugate is based is poorly internalizable.
  • the effector is, in its native form, non-selective.
  • the effector has high non-selective toxicity, including systemic toxicity, when in its native form.
  • the “native form” of an effector molecule of the present invention is an effector molecule before being attached to the targeting agent to form an immunoconjugate.
  • the non-selective toxicity of the effector molecule is substantially eliminated upon conjugation to the targeting agent.
  • the effector molecule causes, upon reaching the target cell, death or continuous cell cycle arrest in the target cell.
  • a drug-effector molecule according to the present invention includes, but is not limited to, a drug including, for example, small highly cytotoxic drugs that act as inhibitors of tubulin polymerization such as maytansinoids, dolastatins, auristatin and crytophycin; DNA alkylating agents like CC-1065 analogues or derivatives (U.S. Pat. Nos.
  • An effector maytansinoid includes maytansinoids of any origin, including, but not limited to synthetic maytansinol and maytansinol analogue and derivative.
  • Doxorubicin, daunomycin, methotrexate, vinblastine, neocarzinostatin, macromycin, trenimon and ⁇ -amanitin are some other effector molecules within the scope of the present invention.
  • antisense DNA molecules as effector molecules.
  • effector or effector molecule
  • Maytansine is a natural product originally derived from the Ethiopian shrub Maytenus serrata (Remillard, 1975; U.S. Pat. No. 3,896,111). This drug inhibits tubulin polymerization, resulting in mitotic block and cell death (Remillard, 1975; Bhattacharyya, 1977; Kupchan, 1978).
  • the cytotoxicity of maytansine is 200-1000-fold higher than that of anti-cancer drugs in clinical use that affect tubulin polymerization, such as Vinca alkaloids or taxol.
  • clinical trials of maytansine indicated that it lacked a therapeutic window due to its high systemic toxicity.
  • Maytansine and maytansinoids are highly cytotoxic but their clinical use in cancer therapy has been greatly limited by their severe systemic side-effects primarily attributed to their poor selectivity for tumors. Clinical trials with maytansine showed serious adverse effects on the central nervous system and gastrointestinal system.
  • Maytansinoids have also been isolated from other plants including seed tissue of Trewia nudiflora (U.S. Pat. No. 4,418,064)
  • Certain microbes also produce maytansinoids, such as maytansinol and C-3 maytansinol esters (U.S. Pat. No. 4,151,042).
  • the present invention is directed to maytansinoids of any origin, including synthetic maytansinol and maytansinol analogues which are disclosed, for example, in U.S. Pat. Nos. 4,137,230; 4,248,870; 4,256,746; 4,260,608; 4,265,814; 4,294,757; 4,307,016; 4,308,268; 4,308,269; 4,309,428; 4,313,946; 4,315,929; 4,317,821; 4,322,348; 4,331,598; 4,361,650; 4,362,663; 4,364,866; 4,371,533; 4,424,219 and 4,151,042.
  • the maytansinoid is a thiol-containing maytansinoid and is more preferably produced according to the processes disclosed in U.S. Pat. No. 6,333,410 to Chari et al or in Chari et al. (Chari, 1992).
  • DM-1 (N 2 -deacetyl-N 2 -(3-mercapto-1-oxopropyl)-maytansine) is a preferred effector molecule in the context of the present invention.
  • DM1 is 3- to 10-fold more cytotoxic than maytansine, and has been converted into a pro-drug by linking it via disulfide bond(s) to a monoclonal antibody directed towards a tumor-associated antigen.
  • Certain of these conjugates (sometimes called “tumor activated prodrugs” (TAPs)) are not cytotoxic in the blood compartment, since they are activated upon associating with a target cells and internalized, thereby releasing the drug (Blättler, 2001).
  • maytansinoids comprise a side chain that contains a sterically hindered thiol bond such as, but not limited to, maytansinoids N 2′ -deacetyl-N 2′ -(4-mercapto-1-oxopentyl)-maytansine, also referred to as “DM3,” and N 2′ -deacetyl-N 2′ -(4-methyl-4-mercapto-1-oxopentyl)-maytansine, also referred to as “DM4.”
  • maytansinoids N 2′ -deacetyl-N 2′ -(4-mercapto-1-oxopentyl)-maytansine also referred to as “DM3”
  • N 2′ -deacetyl-N 2′ -(4-methyl-4-mercapto-1-oxopentyl)-maytansine also referred to as “DM4.”
  • DNA alkylating agents are also particularly preferred as effector molecules and include, but are not limited to, CC-1065 analogues or derivatives.
  • CC-1065 is a potent antitumor-antibiotic isolated from cultures of Streptomyces zelensis and has been shown to be exceptionally cytotoxic in vitro (U.S. Pat. No. 4,169,888).
  • CC-1065 analogues or derivatives described in U.S. Pat. Nos. 5,475,092, 5,585,499 and 5,739,350.
  • modified CC-1065 analogues or derivatives as described in U.S. Pat. No.
  • CC-1065 analogues or derivatives as described, for example, in U.S. Pat. No. 6,756,397 are also within the scope of the present invention.
  • CC-1065 analogues or derivatives may, for example, be synthesized as described in U.S. Pat. No. 6,534,660.
  • Taxanes are mitotic spindle poisons that inhibit the depolymerization of tubulin, resulting in an increase in the rate of microtubule assembly and cell death.
  • Taxanes that are within the scope of the present invention are, for example, disclosed in U.S. Pat. Nos. 6,436,931; 6,340,701; 6,706,708 and U.S. Patent Publications 20040087649; 20040024049 and 20030004210.
  • Other taxanes are disclosed, for example, in U.S. Pat. No. 6,002,023, U.S. Pat. No. 5,998,656, U.S. Pat. No.
  • Calicheamicin effector molecules include gamma 1I, N-acetyl calicheamicin and other derivatives of calicheamicin.
  • Calicheamicin binds in a sequence-specific manner to the minor groove of DNA, undergoes rearrangement and exposes free radicals, leading to breakage of double-stranded DNA, resulting in cell apoptosis and death.
  • a calicheamicin effector molecule that can be used in the context of the present invention is described in U.S. Pat. No. 5,053,394.
  • An immunoconjugate according to the present invention comprises at least one targeting agent, in particular targeting antibody, and one effector molecule.
  • the immunoconjugate might comprise further molecules for example for stabilization.
  • the term “conjugate” is generally used to define the operative association of the targeting agent with one or more effector molecules and is not intended to refer solely to any type of operative association, and is particularly not limited to chemical “conjugation”. So long as the targeting agent is able to bind to the target site and the attached effector functions sufficiently as intended, particularly when delivered to the target site, any mode of attachment will be suitable.
  • the conjugation methods according to the present invention include, but are not limited to, direct attachment of the effector molecule to the targeting antibody, with or without prior modification of the effector molecule and/or the targeting antibody or attachment via linkers.
  • Linkers can be categorized functionally into, for example, acid labile, photosensitive, enzyme cleavable linkers etc.
  • Other suitable linkers may include disulfide bonds and non-cleavable bonds, such as, but not limited to Sulfosuccinimidyl maleimidomethyl cyclohexane carboxylate (SMCC), which is a heterobifunctional linker capable of linking compounds with SH-containing compounds.
  • SMCC Sulfosuccinimidyl maleimidomethyl cyclohexane carboxylate
  • Bifunctional and heterobifunctional linker molecules such as carbohydrate-directed heterobifunctional linker molecules, such as S-(2-thiopyridyl)-L-cysteine hydrazide (TPCH), are also within the scope of the present invention (Vogel, 2004).
  • the effector molecule such as a maytansinoid, may be conjugated to the targeting antibody via a two reaction step process, including as a first step modification of the targeting antibody with a cross-linking reagent such as N-succinimidyl pyridyidithiopropionate (SPDP) to introduce dithiopyridyl groups into the targeting antibody.
  • SPDP N-succinimidyl pyridyidithiopropionate
  • a reactive maytansinoid having a thiol group such as DM1
  • a reactive maytansinoid having a thiol group such as DM1
  • DM1 a reactive maytansinoid having a thiol group
  • one-step conjugation processes such as the one disclosed in U.S. Patent Publication 20030055226 to Chari et al are also within the scope of the present invention.
  • multiple effector molecules of the same or different kind are attached to a targeting antibody.
  • CC-1065 analogues or derivatives may be conjugated to the targeting agent via for example PEG linking groups as described in U.S. Pat. No. 6,716,821.
  • Calicheamicins may be conjugated to the targeting antibodies via linkers (U.S. Pat. No. 5,877,296 and U.S. Pat. No. 5,773,001) or according to the conjugation methods disclosed in U.S. Pat. No. 5,712,374 and U.S. Pat. No. 5,714,586. Another preferred method for preparing calicheamicin conjugates is disclosed in U.S. Patent Publication 20040082764.
  • the immunoconjugates of the present invention also include recombinant fusion proteins.
  • the present invention takes advantage of the property of antibodies, in particular monoclonal antibodies, to bind to specific antigen targets, in particular, the property of certain antibodies to bind to CD138.
  • CD138 or sydecan-1 (also described as SYND1; SYNDECAN; SDC; SCD1; CD138 ANTIGEN, SwissProt accession number: P18827 human) is a membrane glycoprotein that was originally described to be present on cells of epithelial origin, and subsequently found on hematopoietic cells (Sanderson, 1989).
  • CD138 is highly expressed on the majority of MM cells, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma cells and cells of Hodgkin's and non-Hodgkin's lymphomas, chronic lymphocytic leukemia (CLL) (Horvathova, 1995), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML) (Seftalioglu, 2003 (a); Seftalioglu, 2003 (b)), solid tissue sarcomas, colon carcinomas as well as other hematologic malignancies and solid tumors that express syndecan-1 (Carbone et al., 1999; Sebestyen et al., 1999; Han et al., 2004; Charnaux et al., 2004; O'Connell et al., 2004; Orosz and Kopper, 2001).
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphoblastic leukemia
  • cancers that have been shown to be positive for CD138 expression are many ovarian adenocarcinomas, transitional cell bladder carcinomas, kidney clear cell carcinomas, squamous cell lung carcinomas; breast carcinomas and uterine cancers (see, for example, Davies et al., 2004; Barbareschi et al., 2003; Mennerich et al., 2004; Anttonen et al., 2001; Wijdenes, 2002).
  • CD138 expression is restricted to plasma cells (Wijdenes, 1996; Chilosi, 1999) and is not expressed on peripheral blood lymphocytes, monocytes, granulocytes, and red blood cells.
  • CD34 + stem and progenitor cells do not express CD138 and anti-CD138 mAbs do not affect the number of colony forming units in hematopoietic stem cell cultures (Wijdenes, 1996).
  • CD138 is mainly expressed on simple and stratified epithelia within the lung, liver, skin, kidney and gut. Only a weak staining was seen on endothelial cells (Bernfield, 1992; Vooijs, 1996). It has been reported that CD138 exists in polymorphic forms in human lymphoma cells (Gattei, 1999).
  • B-B4, BC/B-B4, B-B2, DL-101, 1 D4, MI15, 1.BB.210, 2Q1484, 5F7, 104-9, 281-2 in particular B-B4 have been reported to be specific to CD138.
  • B-B4, 1D4 and MI15 recognized both the intact molecule and the core protein of CD138 and were shown to recognize either the same or closely related epitopes (Gattei, 1999).
  • B-B4 has the advantage of not recognizing soluble CD138, but only CD138 in membrane bound form (Wijdenes, 2002).
  • B-B4 a murine IgG1 mAb, binds to a linear epitope between residues 90-95 of the core protein on human syndecan-1 (CD138) (Wijdenes, 1996; Dore, 1998). Consistent with the expression pattern of CD138, B-B4 was shown to strongly react with plasma cell line RPM18226, but not to react with endothelial cells. Also consistent with the expression pattern of CD138, B-B4 also reacted with epithelial cells lines A431 (keratinocyte derived) and HepG2 (hepatocyte derived). An immunotoxin B-B4-saporin was also highly toxic towards the plasma cell line RPM18226, in fact considerably more toxic than free saporin.
  • B-B4-saporin showed only toxicity towards cell line A431, although in a clonogenic assay B-B4 saporin showed no inhibitory effect on the outgrowth of A431 cells (Vooijs, 1996).
  • Other researchers reported lack of specificity of MM-associated antigens against tumors (Couturier, 1999).
  • the reactivity of B-B4 with tissue of various organs is shown in Table 1, the reactivity of B-B4 with cell lines of different origins is shown in Table 2.
  • the reactivity was determined by immunohistochemistry (Table 1) and cytofluorography (Table 2). The number of (+) signs indicate the intensity of the reaction.
  • immunoconjugates on a cellular level has been described, for example, for huC242-DM1 (Immunogen, Inc.), an immunoconjugate comprising the antibody huC242 and the maytansinoid DM1, an inhibitor of tubulin polymerization described above.
  • the activity of this immunoconjugate at the cellular level was described to include the following steps: (1) binding of the immunoconjugate to the antigen expressed on a cancer cell, (2) the internalization of the conjugate-antigen complex by the cancer cell, and (3) release of DM1, thereby allowing DM1 to reach its intracellular target tubulin and to inhibit tubulin polymerization (Xie, 2003).
  • TEPs tumor activated prodrugs
  • the present invention is useful in the treatment of, but is not limited to, cancers, in particular, multiple myeloma, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma, Hodgkin's and non-Hodgkin's lymphomas, chronic lymphocytic leukemia (CLL) (Horvathova, 1995), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML) (Seftalioglu, 2003 (a); Seftalioglu, 2003 (b)), solid tissue sarcomas, colon carcinomas as well as other hematologic malignancies and solid tumors that express syndecan-1 (Carbone et al., 1999; Sebestyen et al., 1999; Han et al., 2004; Charnaux et al., 2004; O'Connell et al., 2004; Orosz and Kopper, 2001).
  • cancers in particular, multiple myelom
  • the immunoconjugates according to the present invention can be administered by any route, including intravenously, parenterally, orally, intramuscularly, intrathecally or as an aerosol.
  • the mode of delivery will depend on the desired effect. A skilled artisan will readily know the best route of administration for a particular treatment in accordance with the present invention. The appropriate dosage will depend on the route of administration and the treatment indicated, and can readily be determined by a skilled artisan in view of current treatment protocols.
  • compositions containing an immunoconjugate of the present invention as the active ingredient can be prepared according to conventional pharmaceutical compounding techniques. See, for example, Remington's Pharmaceutical Sciences, 17th Ed. (1985, Mack Publishing Co., Easton, Pa.). Typically, an antagonistic amount of active ingredient will be admixed with a pharmaceutically acceptable carrier.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, for example, intravenous, oral, parenteral, intrathecal, transdermal, or by aerosol.
  • the immunoconjugate can be formulated into solid or liquid preparations such as capsules, pills, tablets, lozenges, melts, powders, suspensions or emulsions.
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, suspending agents, and the like in the case of oral liquid preparations (such as, for example, suspensions, elixirs and solutions); or carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations (such as, for example, powders, capsules and tablets).
  • tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be sugar-coated or enteric-coated by standard techniques.
  • the active agent must be stable to passage through the gastrointestinal tract. If necessary, suitable agents for stable passage can be used, and may include phospholipids or lecithin derivatives described in the literature, as well as liposomes, microparticles (including microspheres and macrospheres).
  • the immunoconjugate may be dissolved in a pharmaceutical carrier and administered as either a solution or a suspension.
  • suitable carriers are water, saline, phosphate buffer solution (PBS), dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative or synthetic origin.
  • PBS phosphate buffer solution
  • the carrier may also contain other ingredients, for example, preservatives, suspending agents, solubilizing agents, buffers and the like.
  • the immunoconjugate When the immunoconjugate are being administered intracerebroventricularly or intrathecally, they may also be dissolved in cerebrospinal fluid.
  • MM is treated as follows, with use of the B-B4-DM1 conjugate as an example.
  • This example is not intended to limit the present invention in any manner, and a skilled artisan could readily determine other immunoconjugates of the present invention and other treatment regimes which could be utilized for the treatment of diseases such as MM.
  • the immunoconjugate Due to the selective expression of CD138 on patient MM cells on via the blood stream accessible cells, the specificity of B-B4 and the stability of the B-B4-DM1 conjugate in the bloodstream, the immunoconjugate removes the systemic toxicity of DM1 and provides an opportunity to target the delivery of the DM1-effector molecule(s).
  • the immunoconjugates of this invention provide a means for the effective administration of the effector molecules to cell sites where the effector molecules can be released from the immunoconjugates. This targeted delivery and release provides a significant advance in the treatment of multiple myeloma, for which current chemotherapy methods sometimes provide incomplete remission.
  • solid tumors may also be treated as follows with use of B-B4-DM1, as an example.
  • This example is not intended to limit the present invention in any manner, and a skilled artisan could readily determine other immunoconjugates of the present invention and other treatment regimes which could be utilized for the treatment of solid tumors.
  • the tumor is first treated to reduce the size of the tumor, for example chemotherapeutically or radioactively.
  • Subsequent administration of the immunoconjugates of this invention provides a means for eliminating residual cancer cells.
  • the administration of the immunoconjugate allows specific targeting of these residual cells and release of the effector molecules at the target site. This targeted delivery and release provides a significant advance in the treatment of residual cancer cells of solid tumors, for which current chemotherapy methods sometimes provide incomplete remission.
  • the thiol-containing maytansinoid DM1 was synthesized from the microbial fermentation product ansamitocin P-3, as previously described by Chari (Chari et al, 1992). Characterization of murine B-B4 (Wijdenes, 1996) and preparation of humanized C242 (huC242) (Roguska, 1994) have been previously described. Antibody-drug conjugates were prepared as described by Liu et al (Liu, 1996). An average of 3.5 DM1 molecules was linked per antibody molecule.
  • CD138 + dexamethasone (Dex)-sensitive MM.1S and Dex-resistant MM.1R, Ocy-My5, OPM1 and OPM2 human MM cell lines and CD138 ⁇ Waldenstrom Macroglobulinemia (WM) WSU-WM and the lymphoma (LB) SUDHL4 cell lines were used.
  • Cell lines were cultured in RPMI-1640 medium (GIBCO) supplemented with 10% fetal bovine serum (FBS; Hyclone, Logan, Utah), L-glutamine, penicillin, and streptomycin (GIBCO) (denoted below as RPMI complete medium).
  • Plasma cells (PC) and bone marrow (BM) cells were isolated using Ficoll-Hypaque density gradient sedimentation from BM aspirates, obtained from MM patients following informed consent. BM cells were separated. BMSCs were obtained by long-term cultures of BM cells (4-8 weeks) in RPMI 1640 medium supplemented with 20% FBS.
  • CD138 on normal plasma cells and patient MM cells was evalutated.
  • BM aspirate samples from normal donors and patients with MM were treated with 0.86% ammonium chloride to lyse red blood cells.
  • PC were then isolated by positive immunomagnetic bead selection using anti-CD138 antibodies and Magnet Assisted Cell Sorting (“MACS,” Miltenyi Biotech). Purity of plasma cells (>95%) was assessed by flow cytometric (Becton-Dickinson “FACSort”) monitoring for CD38 + /CD45 lo phenotype as well as forward and side scatter and morphological characteristics.
  • flow cytometric Becton-Dickinson “FACSort”
  • cRNA cRNA was hybridized to Human Genome U133 (HG-U133) GeneChip® arrays (Affymetrix, Inc.) representing approximately 33,000 human genes, and GeneChip® arrays were scanned on a GeneArray® Scanner (Affymetrix, Inc., Santa Clara, Calif.). Normalization of arrays and calculation of expression values was performed using the DNA-Chip Analyzer (“dChip”) program. Arrays were normalized based on relative signal produced for an invariant subset of genes. This model-based method was used for probe selection and computing expression values. By pooling hybridization information across multiple arrays, it was possible to assess standard errors for the expression level indexes. This approach also allowed automatic probe selection in the analysis stage to reduce errors due to cross-hybridizing probes and image contamination.
  • HG-U133 Human Genome U133
  • GeneChip® arrays GeneChip® arrays representing approximately 33,000 human genes
  • GeneChip® arrays were scanned on a GeneArray® Scanner (Affymetri
  • B-B4 antibody Internalization of B-B4 antibody was examined with a cultured CD138 + cell line by flow cytometry and under a fluorescent microscope.
  • the antibody was modified by Alexa 488 dye (Molecular Probes), and the fluorescence of the non-internalized antibody bound to cells was quenched by exposure to an “anti-Alexa antibody” (Molecular Probes).
  • Alexa 488 dye Molecular Probes
  • B-B4 was poorly internalized.
  • B-B4-DM1 The effect of B-B4-DM1 on cell proliferation was measured by the extent of [3H]-thymidine (NEN Life Science Products, Boston, Mass.) incorporation.
  • Cells (2 ⁇ 10 4 cells/well) were incubated in 96-well culture plates in the presence of 70%-80% confluent BMSCs at 37° C. with or without a test-agent (in triplicate wells).
  • [ 3 H]-thymidine (0.5 ⁇ Ci) was then added to each well for the last 8 h.
  • Cells were harvested onto glass filters with an automatic cell harvester (Cambridge Technology, Cambridge, Mass.) and counted using a Micro-Beta® Trilux counter (Wallac, Gaithersburgh, Md.).
  • FITC-labeled Annexin V and propidium iodide PI was carried out to detect induction of apoptotic cell death by B-B4-DM1.
  • cells were washed with PBS and re-suspended in 100 ⁇ l of HEPES buffer containing Annexin V-FITC and propidium iodide (PI) (Annexin V-FLUOS staining kit; Roche Diagnostic, Indianapolis, Ind.).
  • PI Annexin V-FLUOS staining kit
  • mice were treated intavenously (i.v.) with vehicle alone (PBS), unconjugated B-B4 (13.3 ⁇ g/ml), B-B4-DM1 (conjugate containing 75 or 150 ⁇ g DM1/Kg per day), or control huC242-DM1 (150 ⁇ g DM1/Kg per day), for a total of 3 days.
  • PBS vehicle alone
  • B-B4-DM1 conjuggate containing 75 or 150 ⁇ g DM1/Kg per day
  • control huC242-DM1 150 ⁇ g DM1/Kg per day
  • Human fetal bones were obtained from products of conceptions of second trimester abortions in compliance with state and federal regulations (Advanced Bioscience Resourses, ABR; Alameda, Calif.).
  • the implantation of human fetal long bone grafts into SCID mice to produce SCID-hu mice has been previously described (McCune et al, 1988; Namikawa et al, 1988; Kyoizumi et al, 1993; Akkina et al, 1994; Chen et al, 1994; Sandhu et al, 1996; Urashima, 1997).
  • the femurs or tibias of 19 to 23 gestational week fetuses were cut into fragments and implanted s.c.
  • Blood 50-100 ⁇ l was withdrawn from the tail vein for measurement of human paraprotein in murine serum using ELISA (Bethyl, Montgomery, Tex.). Goat anti-human ⁇ and ⁇ antisera were used for capture and goat anti-human ⁇ or ⁇ HRP conjugates were used for detection.
  • ELISA Bethyl, Montgomery, Tex.
  • Excised bone grafts were fixed in 10% buffered formalin; skeletal tissues were decalcified with 14% EDTA and embedded in paraffin by previously described standard techniques (Sasaki, 1995). Sections were then stained with H & E (Hematoxylin and eosin) for histopathological examination. Immunoperoxidase studies were performed on paraffin sections using an indirect technique as described (Urashima, 1997). Rabbit anti-human ⁇ and ⁇ antisera were used for detection of MM cells in fetal bone.
  • CD138 is expressed on patient MM cells and is the most important target Ag for identification and selection of these cells.
  • previous reports show heterogenous CD138 expression on MM cells (Wijdenes, 1996; Dhodapkar, 1998; Witzig, 1996; Schneider, 1997; Rawstron, 1997).
  • the expression of CD138 on patient MM cells was measured by gene profiling and flow cytometry.
  • FIG. 1A shows the individual fold increase in intensity of CD138 gene expression compared to normal PC; FIG.
  • CD138 was expressed in all 15 MM specimens (100%) examined at a 95 ⁇ 8-fold mean increase in intensity relative to normal plasma cells.
  • FIG. 1D shows the percentage of patients expressing CD138 + MM cells, as determined by flow cytometry on fresh BM aspirate samples.
  • FIG. 1E shows the percentage of CD138 + MM cells in CD138 + patients on fresh BM aspirates and
  • FIG. 1F shows MFI of CD138 + or CD138 ⁇ MM cells within CD38 bright CD45 lo population.
  • FIG. 1D 18 of 25 patients (72%) expressed CD138, with a mean of 68 ⁇ 31% CD138 + cells ( FIG. 1E ) and MFI of 1234 ⁇ 539 (range: 166-2208) ( FIG. 1F ). Taken together, these results indicate that CD138 is highly expressed in patient MM cells.
  • B-B4-DM1 The effects of B-B4-DM1 on survival of CD138 + (MM-1S, MM-1R, Ocy-My5) and CD138 ⁇ cells (SUDHL-4 and WSU-WM) were determined using an MTT assay. MM cell lines were exposed to unconjugated B-B4 mAb ( FIG. 2A ), immunoconjugate B-B4-DM1 ( FIG. 2B ), or free DM1 drug at equimolar concentrations ( FIG. 2C ). Cell survival was measured using an MTT assay. Data (mean ⁇ SD of triplicate experiments) are shown in FIGS. 2A to 2 C as percentage of untreated controls.
  • CD138 + MM cell lines MM-1S, MM-1R and Ocy-My5 were evaluated as well as CD138 ⁇ cell lines including the lymphoma cell line SUDHL4 and the Waldestrom's Macroglobulinemia cell line WSU-WM.
  • MM cells Since adhesion of MM cells to BMSC (bone marrow stromal cells) protects MM cells against drug-induced apoptosis, the effect of B-B4-DM1 on proliferation of CD138 + (Ocy-My5) MM and CD138 ⁇ (SUDHL-4) LB cells adherent to BMSC was evaluated.
  • BMSC bone marrow stromal cells
  • Cell proliferation was measured by [ 3 H]thymidine incorporation following 72 h treatment with B-B4-DM1 (10 nM). Values represent the mean [ 3 H]-TdR incorporation (cpm) of triplicate cultures.
  • B-B4-DM1 (10 nM) significantly inhibited the proliferation of CD138 + Ocy-My5 cells, but had no significant effect on CD138 ⁇ SUDHL-4 cells. Unconjugated B-B4 did not exert any significant effect, whereas free DM1 (10 nM) was cytotoxic to both cell lines.
  • FIG. 3C shows the cytotoxic activity of B-B4-DM1 (10 nM) on CD138 + /CD56 + patient MM cells cultured with BMSCs using flow cytometry. Following 72 h of treatment with the immunoconjugate, >90% reduction in the MM cells was observed. Taken together, these results indicate that B-B4-DM1 overcomes cell adhesion mediated drug resistance (CAM-DR).
  • CAM-DR cell adhesion mediated drug resistance
  • CD138 + Ocy-My5 MM cells were incubated with B-B4-DM1 (10 nM) for 72 h. Apoptotic cell death was then measured by staining with annexin V and PI and flow cytometric analysis.
  • FIG. 4A shows the induction of apoptotic cell death in CD138 + Ocy-My5 MM cells after 48 h exposure to B-B4-DM1 (10 nM). Percentages of stained cells are reported in each quadrant.
  • FIG. 4A shows a significant increase in both annexin V + /PI ⁇ and annexin V + /PI + fractions in CD138 + cells exposed to B-B4-DM1 and free DM1, whereas no significant differences were detected in cells treated with unconjugated mAb alone.
  • FIG. 4B shows the effects of B-B4-DM1 treatment on the cell cycle.
  • Ocy-My5 MM cells were exposed to B-B4 mAb (13.3 ⁇ g/ml) or B-B4-DM1 (10 nM) for 48 h, labeled with PI, and analyzed using flow cytometry. Percentages of cells in the S-phase (S) and G2/M phase (G2) are indicated. As shown in FIG. 4B , B-B4 mAb alone had no significant effect on the proportion of cells in G2/M phase compared to untreated cells (15% vs 16%), whereas exposure of MM cells to B-B4-DM1 induced a majority (88%) of cells into the G2/M phase.
  • a human MM s.c. xenograft model in SCID mice was used to study the in vivo activity of B-B4-DM1 against CD138 + OPM1 cells.
  • the therapeutic efficacy of B-B4-DM1 was measured in mice bearing large palpable tumors (average size 453 ⁇ 74 mm 3 ). Animals were treated daily i.v.
  • FIGS. 6A to 6 F also shows the results obtained when CB-17 SCID mice were inoculated s.c. in the interscapular area with 5 ⁇ 10 6 OPM1 MM cells.
  • FIG. 7A shows a flow cytometry analysis of GFP + OPM1 cells, indicating a ⁇ 2-log difference in MFI of transfected cells.
  • FIGS. 7B and 7B show results from five animals being injected with 5 ⁇ 10 6 GFP + cells, monitored with whole-body fluorescence imaging for tumor development, and then treated with B-B4-DM1 (150 ⁇ g DM1/kg). Tumor sizes were determined directly by imaging the GFP-expressing tumor.
  • FIGS. 7 C and D are representative whole-body fluorescence imaging from a mouse treated with B-B4-DM1.
  • FIGS. 7E and 7F are negative images of the representative mouse. As seen in FIGS. 7B and 7C to 7 F, B-B4-DM1 induced significant regressions of GFP + tumors, confirming high activity of the immunoconjugate against CD138 + MM cells.
  • FIGS. 8A and 8B the results of monitoring mice for changes in levels of human ⁇ chain as an indicator of disease burden are shown.
  • the Figure shows a significant reduction of ⁇ levels after treatment with B-B4-DM1.
  • treatment with B-B4-DM1 induced a significant reduction of human paraprotein, whereas human paraprotein continued to rise in mice treated with control antibody.
  • FIGS. 9A and 9B show representative human bone sections after implantation of Ocy-My5 cells and before treatment. Sections are respectively stained by H & E and with anti- ⁇ mAb. Finally, the activity of B-B4-DM1 on survival of tumor bearing mice was studied.
  • FIG. 9A and 9B show representative human bone sections after implantation of Ocy-My5 cells and before treatment. Sections are respectively stained by H & E and with anti- ⁇ mAb.
  • FIG. 9C shows the survival of mice measured from the first day of treatment to the day of death or sacrifice.
  • Figure shows a significant prolongation of survival after treatment with B-B4-DM1
  • B-B4-DM1 150 ⁇ g DM1/kg
  • control huC242-DM1 150 ⁇ g DM1/kg

Abstract

Immunoconjugates comprising a targeting agent selectively targeting cell-surface expressed syndecan-1 and at least one effector molecule as well as in vitro and in vivo methods of using those immunocomjugates are disclosed. The effector molecule may have, in its native form, high non-selective cytotoxicity, but substantially no non-selective cytotoxicity when part of said immunoconjugate. Targeting agents include the antibody B-B4 as well as other agents that bind cell-surface expressed syndecan-1.

Description

    BACKGROUND
  • This invention pertains to immunoconjugates and their use in different indications. In particular, the present invention relates to immunoconjugates, the delivery of their effector molecule(s) to target sites and the site specific release of the effector molecule(s) in, at or near target cells, tissues and organs. More particularly, the present invention relates to immunoconjugates comprising one or more syndecan-1 targeting agent and highly potent effector molecules, which are attached to the targeting agent. The effector molecule is activated by cleavage/dissociation from the targeting agent portion of the immunoconjugate in, at or near the target cells, tissues or organs.
  • The publications and other materials, including patents, used herein to illustrate the invention and, in particular, to provide additional details respecting the practice are incorporated by reference. For convenience, the publications are referenced in the following text by author and date and are listed alphabetically by author in the appended bibliography.
  • A substantial body of research has concentrated on the development of systems in which an effector agent can be selectively delivered to a desired location or cell population, i.e., a system for a more targeted treatment of ailments with fewer toxic side effects. In spite of considerable progress that has been achieved, many of those delivery systems for the treatment of various diseases, for example, the treatment of cancer, are still often ineffective or subject the patient to considerable risk.
  • Immunoconjugates comprise at least one targeting agent attached to at least one effector molecule. Such immunoconjugates can be categorized according to their effector molecules into, for example, drug immunoconjugates, immunotoxin conjugate and radioimmunoconjugates (Payne, 2003).
  • Efficiency in killing cells is one key factor in the usefulness of an immunoconjugate. Efficiency can be influenced by the potency of the effector molecule (Blättler and Chari, 2001), by the ability of the effector to retain its potency (Chari et al., 1995; Liu et al., 1996; Ojima et al., 2002; Senter et al., 2002 and Sievers and Linenberger, 2001), By the tumor accessibility (Charter, 2001), by the level of expression of the target antigen on the target cell, targeting agent affinity, and by the ability of the target cell to internalize the immunoconjugate (Wargalla, 1989). In the initial development period of immunoconjugates, the efficiencies of conjugates having a drug as an effector molecule often were disappointing compared to the free drug.
  • In response, immunoconjugates with highly cytotoxic effector toxin molecules were constructed. However, while the efficiencies of this new generation of immunoconjugates were much improved, they were often immunogenic in humans, inducing neutralizing antibodies both to the toxin protein and to the mouse monoclonal antibody. In response, “humanized” antibodies conjugated to nonimmunogenic effector molecules were developed (Payne, 2003).
  • In the context of both highly cytotoxic drugs and toxins conjugated to a targeting agent, systemic toxicity has to be considered. If the cytotoxic drug or the toxin is highly cytotoxic, the immunoconjugate has to reach its target site without adversely affecting the host on its way. Accordingly, if the immunoconjugate circulates, for example, in the bloodstream to reach its target site, then this should occur without a substantial release of active drug. Thus, ideally, a highly cytotoxic drug or toxin of an immunoconjugate is only activated upon reaching its target.
  • Specificity is another factor critical for the usability of an immunoconjugate. The immunoconjugate has to be able to selectively interact with the target cells. Particularly for in vivo applications, it is critical that the immunoconjugate does not have substantial adverse effects on essential non-target cells. Thus, both the cellular target of the immunoconjugate and the targeting agent of the immunoconjugate have to be carefully selected to ensure specificity (Blättler and Chari, 2001).
  • It has also been considered important that immunoconjugates comprising targeting antibodies demonstrate pharmacokinetic and tissue distribution characteristics similar to those of corresponding antibodies (Xie, 2003).
  • First successes have been achieved with immunoconjugates. For example, MYLOTARG, a conjugate of an anti-CD33 humanized monoclonal antibody and the highly cytotoxic DNA-damaging agent calicheamicin, has been recently approved by the FDA as the first drug immunoconjugate for clinical treatment of certain indications of myeloid leukemia (Bross, 2001; Hamann, 2002; Dowell, 2001).
  • However, there remains a need to develop effective immunoconjugates for a wide array of indications.
  • SUMMARY OF THE INVENTION
  • The present invention pertains in one embodiment to an immunoconjugate comprising
  • at least one targeting agent selectively targeting cell-surface expressed syndecan-1,
  • at least one effector molecule,
        • wherein the effector molecule has, in its native form, high non-selective cytotoxicity,
        • wherein the targeting agent is functionally attached to said effector molecule to form the immunoconjugate, and
        • wherein the effector molecule has substantially no non-selective cytotoxicity when part of said immunoconjugate.
  • The cytotoxicity of the effector molecule, in its native form, on cells targeted by said targeting antibody may be higher or about the same as the cytotoxicity of the immunoconjugate on said targeted cells. The effector molecule may have, in its native form, a potency of about 10−14-10−7, preferably a potency of about 10−13-10−7M, of about 10−12-10−7M, of about 10−12-10−8M, most preferably of about 10−11-10−10 M, which includes any narrower potency ranges encompassed by the ranges specified above, such as, but not limited to, a potency of about 10−11-10−10 M. The effector molecule may be a maytansinoid, in particular DM1, DM3 or DM4, a CC1065 analogue, a calicheamicin or a taxane. In certain embodiments, the effector molecule may have a molecular weight of less than 5 kDa, in particular less than 2 kDa, more in particular less than 1 kDa and in between about 600 and about 800 Da.
  • The targeting agent may be a targeting antibody, which includes fragments of antibodies, or non-immunoglobulin targeting molecule.
  • The targeting antibody may be derived from an antibody that internalizes poorly. In certain embodiments, the targeting antibody may be derived from the antibody B-B4.
  • The present invention is also directed to a pharmaceutical composition comprising an effective amount of the immunoconjugate described above and one or more pharmaceutically acceptable excipients.
  • The present invention is also directed to a kit comprising, in separate containers, pharmaceutical compositions for use in combination to inhibit, delay and/or prevent the growth of tumors and/or spread of tumor cells, wherein one container comprises an effective amount of above described pharmaceutical composition, and wherein, a separate container comprises a second pharmaceutical composition comprising an effective amount of an agent for the inhibition, delay and/or prevention of the growth of tumors and/or spread of tumor cells, and one or more pharmaceutically acceptable excipients. The agent in said second pharmaceutical composition may be a chemotherapeutic agent or another immunoconjugate.
  • In one embodiment, the invention is directed to a method for treating, inhibiting, delaying and/or preventing the growth of tumor cells in a cell culture containing syndecan-1 expressing tumor cells and non-tumor cells, comprising administering an effective amount of the above described immunoconjugate. The effective amount induces, in certain embodiments, cell death or continuous cell cycle arrest of said syndecan-1 expressing tumor cells. The cells in said cell culture may be obtained from a cancer patient and may, after administration of said effective amount of said immunoconjugate, be reimplanted into said cancer patient. The cells in said cell culture may be isolated from a patient suffering from an hematologic malignancy and/or a solid tumor comprising syndecan-1 expressing cells, in particular from a patient suffering from one of the following: multiple myeloma, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma, Hodgkin's and non-Hodgkin's lymphoma, chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML), a solid tissue sarcoma or a colon carcinoma.
  • The present invention is also directed to a method of inhibiting, delaying and/or preventing the growth of a tumor comprising syndecan-1 expressing tumor cells and/or spread of syndecan-1 expressing tumor cells in a patient in need thereof, comprising
    • administering to the patient at least one immunoconjugate in a growth of the tumor and/or spreading of the tumor cells inhibiting or reducing amount,
      wherein the immunoconjugate selectively inhibits, delays or prevents the growth and/or spread of syndecan-1 expressing cells. The patient may, in this embodiment of the invention, suffer from an hematologic malignancy and/or a solid tumor comprising syndecan-1 expressing cells, in particular from one of the following: multiple myeloma, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma, Hodgkin's and non-Hodgkin's lymphoma, chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML), a solid tissue sarcoma or a colon carcinoma. An effector molecule of the immunoconjugate may, in this embodiment, exhibit, in its native form, high non-selective cytotoxicity.
  • The invention is also directed to a method for inhibiting, delaying and/or preventing the growth of a tumor and/or spread of malignant tumor cells in a patient in need thereof, comprising
    • (a) administering to the patient one or more cancer drugs and/or radiation in an amount effective to reduce tumor load; and
    • (b) administering to the patient at least one immunoconjugate in a growth of a tumor and/or spreading of tumor cells inhibiting, delaying or preventing amount,
      wherein the immunoconjugate selectively inhibits, delays or prevents the growth and/or spread of syndecan-1 expressing cells; (a) and (b) may hereby be performed consecutively in two consecutive treatment regimes. The drug of (a) and the immunoconjugate of (b) may also be administered in a single administration step.
  • The present invention is also directed to a method for treating a subject having a condition that would benefit from the selective suppression of myeloma cell survival, the method comprising:
    • (a) providing at least one immunoconjugate that selectively binds to syndecan-1 expressed on myeloma cells; and
    • (b) administering the immunoconjugate to the subject to selectively decrease survival or growth of said myeloma cells of the subject. The immunoconjugate may comprise a B-B4 targeting antibody. The immunoconjugate may, in this embodiment, comprise a maytansinoid effector molecule. The selective suppression of myeloma cell survival may also induce growth arrest or apoptosis in myeloma cells.
    BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A-1F show the expression of CD138 in multiple myeloma (MM) cells.
  • FIGS. 2A-2C show the effect of B-B4-DM1 in comparison with that produced by the naked antibody or by non-conjugated drug on survival of CD138+ and CD138 MM cells.
  • FIGS. 3A to 3C show the inhibitory effect of B-B4-DM1 on proliferation of CD138+ and CD138 cells adherent to bone marrow stromal cells (BMSCs).
  • FIGS. 4A and 4B show the survival and cell cycle effects of B-B4-DM1 on CD138+ MM cells.
  • FIGS. 5A to 5D show the activity of B-B4-DM1 in a tumor xenograft model of human CD138+ multiple myeloma.
  • FIGS. 6A to F show the activity of B-B4-DM1 on large tumor xenografts of human CD138+ OPM multiple myeloma.
  • FIG. 7A shows the expression of GFP in the cells (GFP stands for Green Fluorescent Protein). FIGS. 7B to 7F show the activity of B-B4-DM1 on GFP+ human MM xenografts.
  • FIGS. 8A to 8C show that B-B4-DM1 reduces MM tumor burden in SCID-hu hosts implanted with patient MM cells.
  • FIGS. 9A to 9C show that B-B4-DM1 increases survival in SCID-hu hosts implanted with the Ocy-My5 MM cell line.
  • DETAILED DESCRIPTION OF VARIOUS AND PREFERRED EMBODIMENTS
  • The present invention relates to immunoconjugates and the delivery of their effector molecule(s) to target sites and the site specific release of effector(s) molecule in, at or near target cells, tissues and organs. More particularly, the present invention relates to immunoconjugates comprising syndecan-1 targeting agents and potent effector molecules. The effector molecules are covalently linked, chelated or otherwise associated with the targeting agent. The effector molecules may be activated by cleavage/dissociation from the targeting agent portion of the immunoconjugate at the target site.
  • The immunoconjugates according to the present invention are administered to a subject in need of therapeutic treatment or to cells isolated from such a subject in need of therapeutic treatment. The effector molecule or molecules may be released from the immunoconjugate by cleavage/dissociation in, at or close to the target cell, tissue or organ.
  • As one example, the immunoconjugate comprises the antibody B-B4 and at least one highly cytotoxic drug or toxin as an effector molecule and is administered to a patient with cancer. In this example, a therapeutically effective amount of the immunoconjugate is administered intravenously to a patient so that it concentrates in the cancer cells. The effector molecule or molecules are released from the antibody target by natural means.
  • As a second example, the immunoconjugate comprises the antibody B-B4 and at least one highly cytotoxic drug and is administered to a cell population isolated from a patient with cancer. In this example, a cell death or continuous cell cycle arrest inducing amount of the immunoconjugate is administered to the cell population so that it concentrates in the cancerous cells. The effector molecule or molecules are released from the targeting antibody by natural means or external means to induce cell death or continuous cell cycle arrest in the cancer cells.
  • As a third example, the immunoconjugate comprises the antibody B-B4 and at least one highly cytotoxic drug or an immunotoxin as an effector molecule and is administered to a patient with cancer. In this example, a therapeutically effective amount of the immunoconjugate is administered intravenously to a patient so that it concentrates in the cancerous cells. The effector molecule or molecules are released from the antibody target by an external means to induce cell death or continuous cell cycle arrest in the cancer cells.
  • Targeting agent: A targeting agent according to the present invention is able to associate with a molecule expressed by a target cell and includes peptides and non-peptides. In particular, targeting agents according to the present invention include targeting antibodies and non-immunoglobulin targeting molecules, which may be based on non-immunoglobulin proteins, including, but not limited to, AFFILIN® molecules, ANTICALINS® and AFFIBODIES®. Non-immunoglobulin targeting molecules also include non-peptidic targeting molecules including targeting DNA and RNA oligonucleotides (aptamers).
  • Targeting antibody: A targeting antibody according to the present invention is or is based on a natural antibody or is produced synthetically or by genetic engineering and binds to an antigen on a cell or cells (target cell(s)) of interest. A targeting antibody according to the present invention includes a monoclonal antibody, a polyclonal antibody, a multispecific antibody (for example, a bispecific antibody), or an antibody fragment. The targeting antibody may be engineered to, for example, improve its affinity to the target cells (Ross, 2003) or diminish its immunogenicity. The targeting antibody may be attached to a liposomal formulation including effector molecules (Carter, 2003). An antibody fragment comprises a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody. Examples of antibody fragments according to the present invention include Fab, Fab′, F(ab′)2, and Fv fragments, but also diabodies; domain antibodies (dAb) (Ward, 1989; U.S. Pat. No. 6,005,079); linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. In a single chain variable fragment antibody (scFv) the heavy and light chains (VH and VL) can be linked by a short amino acid linker having, for example, the sequence (glycine4serine)n, which has sufficient flexibility to allow the two domains to assemble a functional antigen binding pocket. Addition of various signal sequences may allow for more precise targeting of the targeting antibody. Addition of the light chain constant region (CL) may allow dimerization via disulphide bonds, giving increased stability and avidity. Variable regions for constructing the scFv can, if a mAb against a target of interest is available, be obtained by RT-PCR which clones out the variable regions from mRNA extracted from the parent hybridoma. Alternatively, the scFv can be generated de novo by phage display technology (Smith, 2001). A bispecific antibody according to the present invention may, for example, have at least one arm that is reactive against a target tissue and one arm that is reactive against a linker moiety (U.S. Patent Publication 20020006379). A bispecific antibody according to the present invention may also bind to more than one antigen on a target cell (Carter, 2003). An antibody according to the present invention may be modified by, for example, introducing cystein residues to introduce thiol groups (Olafsen, 2004).
  • In accordance with the present invention, the targeting antibody may be derived from any source and may be, but is not limited to, a camel antibody, a murine antibody, a chimeric human/mouse antibody or a chimeric human/monkey antibody, in particular, a chimeric human/monkey antibody with the monkey portion stemming from a cynomolgus monkey. Humanized antibodies are antibodies that contain sequences derived from a human-antibody and from a non-human antibody and are also within the scope of the present invention. Suitable methods for humanizing antibodies include CDR-grafting (complementarity determining region grafting) (EP 0 239 400; WO 91/09967; U.S. Pat. Nos. 5,530,101; and 5,585,089), veneering or resurfacing (EP 0 592 106; EP 0 519 596; Padian, 199; Studnicka et al., 1994; Roguska et al., 1994), chain shuffling (U.S. Pat. No. 5,565,332) and Delmmunosation™ (Biovation, LTD). In CDR-grafting, the mouse complementarity-determining regions (CDRs) from, for example, mAb B-B4 are grafted into human variable frameworks, which are then joined to human constant regions, to create a human B-B4 antibody. Several antibodies humanized by CDR-grafting are now in clinical use, including MYLOTARG (Sievers et al., 2001) and HECEPTIN (Pegram et al, 1998).
  • The resurfacing technology uses a combination of molecular modeling, statistical analysis and mutagenesis to alter the non-CDR surfaces of antibody variable regions to resemble the surfaces of known antibodies of the target host. Strategies and methods for the resurfacing of antibodies, and other methods for reducing immunogenicity of antibodies within a different host, are disclosed, for example, in U.S. Pat. No. 5,639,641. Human antibodies can be made by a variety of methods known in the art including phage display methods. See also U.S. Pat. Nos. 4,444,887, 4,716,111, 5,545,806, and 5,814,318; and international patent application publications WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741.
  • Fully human antibodies may also been used. Those antibodies can be selected by the phage display approach, where CD138 or an antigenic determinant thereof is used to selectively bind phage expressing, for example, B-B4 variable regions (see, Krebs, 2001). This approach is advantageously coupled with an affinity maturation technique to improve the affinity of the antibody.
  • In one embodiment, the targeting antibody is, in its unconjugated form, moderately or poorly internalizable. Moderate internalization constitutes about 30% to about 75% internalization of antibody, poor internalization constitutes about 0.01% to up to about 30% internalization after 3 hours incubation at 37° C. In another preferred embodiment the targeting antibody binds to CD138, for example, antibodies B-B4, BC/B-B4, B-B2, DL-101, 1 D4, MI15, 1.BB.210, 2Q1484, 5F7, 104-9, 281-2 in particular B-B4. Preferably the targeting antibody binds primarily to cell-surface expressed CD138. In another embodiment, the targeting antibody does not substantially bind non-cell-surface expressed CD138. When, in the context of the present invention, the name of a specific antibody is combined with the term “targeting antibody” such as “B-B4 targeting antibody,” this means that this targeting antibody has the binding specificity of the antibody B-B4. If a targeting antibody is said to be “derived from” a specified antibody, this means that this targeting antibody has the binding specificity of this antibody, but might take any form consistent with the above description of a targeting antibody. If, in the context of the present invention, for example, a targeting antibody is said to do something “selectively” such as “selectively targeting cell-surface expressed syndcan-1” or, to be “selective” for something, this means that there is a significant selectivity (i.e. a higher affinity towards CD138-positive cells compared with CD138-negative cells) for, in case of the example provided, cell-surface expressed syndecan-1, compared to any other antigens and adverse side effects in a given environment are substantially avoided due to this selectivity.
  • Non-immunoglobulin targeting molecules: Non-immunoglobulin targeting molecules according to the present invention include targeting molecues derived from non-immunoglobulin proteins as well as non-peptidic targeting molecules. Small non-immunoglobulin proteins which are included in this definition are designed to have specific affinities towards, in particular surface expressed CD138. These small non-immunoglobulin proteins include scaffold based engineered molecules such as Affilin® molecules that have a relatively low molecular weight such as between 10 kDa and 20 kDa. Appropriate scaffolds include, for example, gamma crystalline. Those molecules have, in their natural state, no specific binding activity towards the target molecules. By engineering the protein surfaces through locally defined randomization of solvent exposed amino acids, completely new binding sites are created. Former non-binding proteins are thereby transformed into specific binding proteins. Such molecules can be specifically designed to bind a target, such as CD138, and allow for specific delivery of one or more effector molecules (see, scil Proteins GmbH at www.scilproteins.com, 2004). Another kind of non-immunoglobulin targeting molecules are derived from lipocalins, and include, for example ANTICALINS®, which resemble in structure somewhat immunoglobulins. However, lipocalins are composed of a single polypeptide chain with 160 to 180 amino acid residues. The binding pocket of lipocalins can be reshaped to recognize a molecule of interest with high affinity and specificity (see, for example, Beste et al., 1999). Artificial bacterial receptors such as those marketed under the trademark Affibody® (Affibody AB) are also within the scope of the present invention. These artificial bacterial receptor molecules are small, simple proteins and may be composed of a three-helix bundle based on the scaffold of one of the IgG-binding domains of Protein A (Staylococcus aureus). These molecules have binding properties similar to many immunoglobulins, but are substantially smaller, having a molecular weight often not exceeding 10 kDa and are also comparatively stable. Suitable artificial bacterial receptor molecules are, for example, described in U.S. Pat. Nos. 5,831,012; 6,534,628 and 6,740,734. Non-peptidic targeting molecules include, but are limited to, to DNA and RNA oligonucleotides that bind to CD138 (aptamers).
  • Effector molecule: An effector molecule according to the present invention is a molecule or a derivative, or an analogue thereof that is attached to a targeting agent and exerts a desired effect, for example apoptosis, or another type of cell death, or a continuous cell cycle arrest on the target cell or cells. Effector molecules according to the present invention include molecules that can exert desired effects in a target cell and include, but are not limited to, toxins, drugs, in particular low molecular weight cytotoxic drugs, radionuclides, biological response modifiers, pore-forming agents, cytotoxic enzymes, prodrug activating enzymes, antisense oligonucleotides, antibodies or cytokines as well as functional derivatives or analogues/fragments thereof.
  • In a preferred embodiment, the effector increases internal effector delivery of the immunoconjugate, in particular when the natural form of the antibody on which the targeting antibody of the immunoconjugate is based is poorly internalizable. In another preferred embodiment the effector is, in its native form, non-selective. In certain embodiments the effector has high non-selective toxicity, including systemic toxicity, when in its native form. The “native form” of an effector molecule of the present invention is an effector molecule before being attached to the targeting agent to form an immunoconjugate. In another preferred embodiment, the non-selective toxicity of the effector molecule is substantially eliminated upon conjugation to the targeting agent. In another preferred embodiment, the effector molecule causes, upon reaching the target cell, death or continuous cell cycle arrest in the target cell. A drug-effector molecule according to the present invention includes, but is not limited to, a drug including, for example, small highly cytotoxic drugs that act as inhibitors of tubulin polymerization such as maytansinoids, dolastatins, auristatin and crytophycin; DNA alkylating agents like CC-1065 analogues or derivatives (U.S. Pat. Nos. 5,475,092; 5,585,499; 6,716,821) and duocarmycin; enediyne antibiotics such as calicheamicin and esperamicin; and potent taxoid (taxane) drugs (Payne, 2003). Maytansinoids and calicheamicins are particularly preferred. An effector maytansinoid includes maytansinoids of any origin, including, but not limited to synthetic maytansinol and maytansinol analogue and derivative. Doxorubicin, daunomycin, methotrexate, vinblastine, neocarzinostatin, macromycin, trenimon and α-amanitin are some other effector molecules within the scope of the present invention. Also within the scope of the present invention are antisense DNA molecules as effector molecules. When the name of, for example, a specific drug or class of drugs is combined herein with the term “effector” or “effector molecule,” reference is made to an effector of an immunoconjugate according to the present invention that is based on the specified drug or class of drugs.
  • Maytansine is a natural product originally derived from the Ethiopian shrub Maytenus serrata (Remillard, 1975; U.S. Pat. No. 3,896,111). This drug inhibits tubulin polymerization, resulting in mitotic block and cell death (Remillard, 1975; Bhattacharyya, 1977; Kupchan, 1978). The cytotoxicity of maytansine is 200-1000-fold higher than that of anti-cancer drugs in clinical use that affect tubulin polymerization, such as Vinca alkaloids or taxol. However, clinical trials of maytansine indicated that it lacked a therapeutic window due to its high systemic toxicity. Maytansine and maytansinoids are highly cytotoxic but their clinical use in cancer therapy has been greatly limited by their severe systemic side-effects primarily attributed to their poor selectivity for tumors. Clinical trials with maytansine showed serious adverse effects on the central nervous system and gastrointestinal system.
  • Maytansinoids have also been isolated from other plants including seed tissue of Trewia nudiflora (U.S. Pat. No. 4,418,064)
  • Certain microbes also produce maytansinoids, such as maytansinol and C-3 maytansinol esters (U.S. Pat. No. 4,151,042).
  • The present invention is directed to maytansinoids of any origin, including synthetic maytansinol and maytansinol analogues which are disclosed, for example, in U.S. Pat. Nos. 4,137,230; 4,248,870; 4,256,746; 4,260,608; 4,265,814; 4,294,757; 4,307,016; 4,308,268; 4,308,269; 4,309,428; 4,313,946; 4,315,929; 4,317,821; 4,322,348; 4,331,598; 4,361,650; 4,362,663; 4,364,866; 4,371,533; 4,424,219 and 4,151,042.
  • In a preferred embodiment, the maytansinoid is a thiol-containing maytansinoid and is more preferably produced according to the processes disclosed in U.S. Pat. No. 6,333,410 to Chari et al or in Chari et al. (Chari, 1992).
  • DM-1 (N2-deacetyl-N2-(3-mercapto-1-oxopropyl)-maytansine) is a preferred effector molecule in the context of the present invention. DM1 is 3- to 10-fold more cytotoxic than maytansine, and has been converted into a pro-drug by linking it via disulfide bond(s) to a monoclonal antibody directed towards a tumor-associated antigen. Certain of these conjugates (sometimes called “tumor activated prodrugs” (TAPs)) are not cytotoxic in the blood compartment, since they are activated upon associating with a target cells and internalized, thereby releasing the drug (Blättler, 2001). Several antibody-DM1 conjugates have been developed (Payne, 2003), and been evaluated in clinical trials. For example, huC242-DM1 treatment in colorectal cancer patients was well tolerated, did not induce any detectable immune response, and had a long circulation time (Tolcher, 2003).
  • Other particularly preferred maytansinoids comprise a side chain that contains a sterically hindered thiol bond such as, but not limited to, maytansinoids N2′-deacetyl-N2′-(4-mercapto-1-oxopentyl)-maytansine, also referred to as “DM3,” and N2′-deacetyl-N2′-(4-methyl-4-mercapto-1-oxopentyl)-maytansine, also referred to as “DM4.”
  • DNA alkylating agents are also particularly preferred as effector molecules and include, but are not limited to, CC-1065 analogues or derivatives. CC-1065 is a potent antitumor-antibiotic isolated from cultures of Streptomyces zelensis and has been shown to be exceptionally cytotoxic in vitro (U.S. Pat. No. 4,169,888). Within the scope of the present invention are, for examples the CC-1065 analogues or derivatives described in U.S. Pat. Nos. 5,475,092, 5,585,499 and 5,739,350. As the person skilled in the art will readily appreciate, modified CC-1065 analogues or derivatives as described in U.S. Pat. No. 5,846,545 and prodrugs of CC-1065 analogues or derivatives as described, for example, in U.S. Pat. No. 6,756,397 are also within the scope of the present invention. In certain embodiments of the invention, CC-1065 analogues or derivatives may, for example, be synthesized as described in U.S. Pat. No. 6,534,660.
  • Another group of compounds that make preferred effector molecules are taxanes, especially highly potent ones and those that contain thiol or disulfide groups. Taxanes are mitotic spindle poisons that inhibit the depolymerization of tubulin, resulting in an increase in the rate of microtubule assembly and cell death. Taxanes that are within the scope of the present invention are, for example, disclosed in U.S. Pat. Nos. 6,436,931; 6,340,701; 6,706,708 and U.S. Patent Publications 20040087649; 20040024049 and 20030004210. Other taxanes are disclosed, for example, in U.S. Pat. No. 6,002,023, U.S. Pat. No. 5,998,656, U.S. Pat. No. 5,892,063, U.S. Pat. No. 5,763,477, U.S. Pat. No. 5,705,508, U.S. Pat. No. 5,703,247 and U.S. Pat. No. 5,367,086. As the person skilled in the art will appreciate, PEGylated taxanes such as the ones described in U.S. Pat. No. 6,596,757 are also within the scope of the present invention.
  • Calicheamicin effector molecules according to the present invention include gamma 1I, N-acetyl calicheamicin and other derivatives of calicheamicin. Calicheamicin binds in a sequence-specific manner to the minor groove of DNA, undergoes rearrangement and exposes free radicals, leading to breakage of double-stranded DNA, resulting in cell apoptosis and death. One example of a calicheamicin effector molecule that can be used in the context of the present invention is described in U.S. Pat. No. 5,053,394.
  • Immunoconjugate: An immunoconjugate according to the present invention comprises at least one targeting agent, in particular targeting antibody, and one effector molecule. The immunoconjugate might comprise further molecules for example for stabilization. For immunoconjugates, the term “conjugate” is generally used to define the operative association of the targeting agent with one or more effector molecules and is not intended to refer solely to any type of operative association, and is particularly not limited to chemical “conjugation”. So long as the targeting agent is able to bind to the target site and the attached effector functions sufficiently as intended, particularly when delivered to the target site, any mode of attachment will be suitable. The conjugation methods according to the present invention include, but are not limited to, direct attachment of the effector molecule to the targeting antibody, with or without prior modification of the effector molecule and/or the targeting antibody or attachment via linkers. Linkers can be categorized functionally into, for example, acid labile, photosensitive, enzyme cleavable linkers etc. Other suitable linkers may include disulfide bonds and non-cleavable bonds, such as, but not limited to Sulfosuccinimidyl maleimidomethyl cyclohexane carboxylate (SMCC), which is a heterobifunctional linker capable of linking compounds with SH-containing compounds. Bifunctional and heterobifunctional linker molecules, such as carbohydrate-directed heterobifunctional linker molecules, such as S-(2-thiopyridyl)-L-cysteine hydrazide (TPCH), are also within the scope of the present invention (Vogel, 2004). The effector molecule, such as a maytansinoid, may be conjugated to the targeting antibody via a two reaction step process, including as a first step modification of the targeting antibody with a cross-linking reagent such as N-succinimidyl pyridyidithiopropionate (SPDP) to introduce dithiopyridyl groups into the targeting antibody. In a second step, a reactive maytansinoid having a thiol group, such as DM1, may be added to the modified antibody, resulting in the displacement of the thiopyridyl groups in the modified antibody, and the production of disulfide-linked cytotoxic maytansinoid/antibody conjugate (U.S. Pat. No. 5,208,020). However, one-step conjugation processes such as the one disclosed in U.S. Patent Publication 20030055226 to Chari et al are also within the scope of the present invention. In one embodiment of the present invention multiple effector molecules of the same or different kind are attached to a targeting antibody.
  • CC-1065 analogues or derivatives may be conjugated to the targeting agent via for example PEG linking groups as described in U.S. Pat. No. 6,716,821.
  • Calicheamicins may be conjugated to the targeting antibodies via linkers (U.S. Pat. No. 5,877,296 and U.S. Pat. No. 5,773,001) or according to the conjugation methods disclosed in U.S. Pat. No. 5,712,374 and U.S. Pat. No. 5,714,586. Another preferred method for preparing calicheamicin conjugates is disclosed in U.S. Patent Publication 20040082764.
  • The immunoconjugates of the present invention also include recombinant fusion proteins.
  • The present invention takes advantage of the property of antibodies, in particular monoclonal antibodies, to bind to specific antigen targets, in particular, the property of certain antibodies to bind to CD138.
  • CD138 or sydecan-1 (also described as SYND1; SYNDECAN; SDC; SCD1; CD138 ANTIGEN, SwissProt accession number: P18827 human) is a membrane glycoprotein that was originally described to be present on cells of epithelial origin, and subsequently found on hematopoietic cells (Sanderson, 1989). In malignant hematopoiesis, CD138 is highly expressed on the majority of MM cells, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma cells and cells of Hodgkin's and non-Hodgkin's lymphomas, chronic lymphocytic leukemia (CLL) (Horvathova, 1995), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML) (Seftalioglu, 2003 (a); Seftalioglu, 2003 (b)), solid tissue sarcomas, colon carcinomas as well as other hematologic malignancies and solid tumors that express syndecan-1 (Carbone et al., 1999; Sebestyen et al., 1999; Han et al., 2004; Charnaux et al., 2004; O'Connell et al., 2004; Orosz and Kopper, 2001).
  • Other cancers that have been shown to be positive for CD138 expression are many ovarian adenocarcinomas, transitional cell bladder carcinomas, kidney clear cell carcinomas, squamous cell lung carcinomas; breast carcinomas and uterine cancers (see, for example, Davies et al., 2004; Barbareschi et al., 2003; Mennerich et al., 2004; Anttonen et al., 2001; Wijdenes, 2002).
  • In the normal human hematopoietic compartment, CD138 expression is restricted to plasma cells (Wijdenes, 1996; Chilosi, 1999) and is not expressed on peripheral blood lymphocytes, monocytes, granulocytes, and red blood cells. In particular, CD34+ stem and progenitor cells do not express CD138 and anti-CD138 mAbs do not affect the number of colony forming units in hematopoietic stem cell cultures (Wijdenes, 1996). In non-hematopoietic compartments, CD138 is mainly expressed on simple and stratified epithelia within the lung, liver, skin, kidney and gut. Only a weak staining was seen on endothelial cells (Bernfield, 1992; Vooijs, 1996). It has been reported that CD138 exists in polymorphic forms in human lymphoma cells (Gattei, 1999).
  • Monoclonal antibodies antibodies B-B4, BC/B-B4, B-B2, DL-101, 1 D4, MI15, 1.BB.210, 2Q1484, 5F7, 104-9, 281-2 in particular B-B4 have been reported to be specific to CD138. Of those B-B4, 1D4 and MI15 recognized both the intact molecule and the core protein of CD138 and were shown to recognize either the same or closely related epitopes (Gattei, 1999). B-B4 has the advantage of not recognizing soluble CD138, but only CD138 in membrane bound form (Wijdenes, 2002).
  • B-B4, a murine IgG1 mAb, binds to a linear epitope between residues 90-95 of the core protein on human syndecan-1 (CD138) (Wijdenes, 1996; Dore, 1998). Consistent with the expression pattern of CD138, B-B4 was shown to strongly react with plasma cell line RPM18226, but not to react with endothelial cells. Also consistent with the expression pattern of CD138, B-B4 also reacted with epithelial cells lines A431 (keratinocyte derived) and HepG2 (hepatocyte derived). An immunotoxin B-B4-saporin was also highly toxic towards the plasma cell line RPM18226, in fact considerably more toxic than free saporin. However, from the two epithelial cell lines tested, B-B4-saporin showed only toxicity towards cell line A431, although in a clonogenic assay B-B4 saporin showed no inhibitory effect on the outgrowth of A431 cells (Vooijs, 1996). Other researchers reported lack of specificity of MM-associated antigens against tumors (Couturier, 1999).
  • The reactivity of B-B4 with tissue of various organs is shown in Table 1, the reactivity of B-B4 with cell lines of different origins is shown in Table 2. The reactivity was determined by immunohistochemistry (Table 1) and cytofluorography (Table 2). The number of (+) signs indicate the intensity of the reaction.
    TABLE 1
    Reactivity of B-B4 with tissues of various
    organs (immunohistochemistry)
    Organ Tissue B-B4
    Blood Normal plasma cells +++
    Blood MM patient cells +++
    Kidney Tubular epithelium
    Kidney Glomerular
    Kidney Urothelium ++
    Kidney Smooth muscle of hilus
    Liver Sinusoid endothelium
    Liver Biliary epithelium
    Liver Hepatocytes ++
    Lung Alveolar epithelium ++
    Lung Bronchial epithelium +
    Lung Blood vessel
    Lung Bronchial gland ++
    Duodenum Crypts epithelium ++
    Duodenum Glands ++
    Duodenum Chorion lymphocytes ++
    Duodenum Smooth muscle
    Duodenum Blood vessels
    Heart Myocytes ++ (cytoplasmic)
    Spleen Red pulp +
    Various organs Muscle
    Various organs Connective tissue
    Various organs Nervous tissue
    Various organs Epithelium +++
    Various organs Endothelium +
    Cell lines MM cell lines +++
  • TABLE 2
    Reactivity of B-B4 with cell lines of
    different origins (Cytofluorography)
    Cell line Cell type B-B4
    RPMI 8226 Multiple myeloma +++
    U266 Multiple myeloma +++
    UM-1 Multiple myeloma +++
    XG-1 Multiple myeloma +++
    Daudi EBV-infected LCL
    Ramos EBV-infected LCL
    Jijoye EBV-infected LCL
    BJAB Burkitt lymphoma
    Raji Burkitt lymphoma
    BTL-1 LCL
    BTL-6 LCL
    KM-3 Pre-B +
    REH Pre-B
    NALM-6 Pre-B +
    ROS Pre-B
    697 Pre-B
    CEM T-cell
    Jurkat T-cell
    HL-60 Myeloid
    U937 Myeloid +
    HEL Myeloid
    KG1A Myeloid
    K562 Erythroid ++
    A341 Epithelial +++
    HepG Hepatocytic ++
    HUVEC Endothelial +
    Peripheral blood Monocyte
    Peripheral blood B-cell (CD19+)
    Peripheral blood T-cell (CD3+)
    Peripheral blood Granulocytes
    Bone marrow (CD34+, CD33+, CD19+, CD20+, CD10+,
    CD3+, CD19+, CD14+, CD38+) cells
    Bone marrow Plasma cells ++
    Bone marrow Myeloma cells/CD38 high +++
    Tonsil (CD19+, CD38+) cells
    Patient sample ALL B-cell
    Patient sample CLL B-cell
    Patient sample Reed-Sternberg cell ++
    Hodgkin
  • The activity of immunoconjugates on a cellular level has been described, for example, for huC242-DM1 (Immunogen, Inc.), an immunoconjugate comprising the antibody huC242 and the maytansinoid DM1, an inhibitor of tubulin polymerization described above. The activity of this immunoconjugate at the cellular level was described to include the following steps: (1) binding of the immunoconjugate to the antigen expressed on a cancer cell, (2) the internalization of the conjugate-antigen complex by the cancer cell, and (3) release of DM1, thereby allowing DM1 to reach its intracellular target tubulin and to inhibit tubulin polymerization (Xie, 2003). This multi-step attachment, internalization and release model forms the rationale behind the development of tumor activated prodrugs (TAPs) (Immunogen, 2003). Similar uptake mechanisms have been described for immunoconjugates based on anti-PSCA antibodies, which were reported to be internalized via caveolae (Ross, 2002).
  • The present invention is useful in the treatment of, but is not limited to, cancers, in particular, multiple myeloma, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma, Hodgkin's and non-Hodgkin's lymphomas, chronic lymphocytic leukemia (CLL) (Horvathova, 1995), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML) (Seftalioglu, 2003 (a); Seftalioglu, 2003 (b)), solid tissue sarcomas, colon carcinomas as well as other hematologic malignancies and solid tumors that express syndecan-1 (Carbone et al., 1999; Sebestyen et al., 1999; Han et al., 2004; Charnaux et al., 2004; O'Connell et al., 2004; Orosz and Kopper, 2001).
  • The immunoconjugates according to the present invention can be administered by any route, including intravenously, parenterally, orally, intramuscularly, intrathecally or as an aerosol. The mode of delivery will depend on the desired effect. A skilled artisan will readily know the best route of administration for a particular treatment in accordance with the present invention. The appropriate dosage will depend on the route of administration and the treatment indicated, and can readily be determined by a skilled artisan in view of current treatment protocols.
  • Pharmaceutical compositions containing an immunoconjugate of the present invention as the active ingredient can be prepared according to conventional pharmaceutical compounding techniques. See, for example, Remington's Pharmaceutical Sciences, 17th Ed. (1985, Mack Publishing Co., Easton, Pa.). Typically, an antagonistic amount of active ingredient will be admixed with a pharmaceutically acceptable carrier. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, for example, intravenous, oral, parenteral, intrathecal, transdermal, or by aerosol.
  • For oral administration, the immunoconjugate can be formulated into solid or liquid preparations such as capsules, pills, tablets, lozenges, melts, powders, suspensions or emulsions. In preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, suspending agents, and the like in the case of oral liquid preparations (such as, for example, suspensions, elixirs and solutions); or carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations (such as, for example, powders, capsules and tablets). Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be sugar-coated or enteric-coated by standard techniques. The active agent must be stable to passage through the gastrointestinal tract. If necessary, suitable agents for stable passage can be used, and may include phospholipids or lecithin derivatives described in the literature, as well as liposomes, microparticles (including microspheres and macrospheres).
  • For parenteral administration, the immunoconjugate may be dissolved in a pharmaceutical carrier and administered as either a solution or a suspension. Illustrative of suitable carriers are water, saline, phosphate buffer solution (PBS), dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative or synthetic origin. The carrier may also contain other ingredients, for example, preservatives, suspending agents, solubilizing agents, buffers and the like. When the immunoconjugate are being administered intracerebroventricularly or intrathecally, they may also be dissolved in cerebrospinal fluid.
  • In accordance with the present invention, MM is treated as follows, with use of the B-B4-DM1 conjugate as an example. This example is not intended to limit the present invention in any manner, and a skilled artisan could readily determine other immunoconjugates of the present invention and other treatment regimes which could be utilized for the treatment of diseases such as MM. Due to the selective expression of CD138 on patient MM cells on via the blood stream accessible cells, the specificity of B-B4 and the stability of the B-B4-DM1 conjugate in the bloodstream, the immunoconjugate removes the systemic toxicity of DM1 and provides an opportunity to target the delivery of the DM1-effector molecule(s). The immunoconjugates of this invention provide a means for the effective administration of the effector molecules to cell sites where the effector molecules can be released from the immunoconjugates. This targeted delivery and release provides a significant advance in the treatment of multiple myeloma, for which current chemotherapy methods sometimes provide incomplete remission.
  • In accordance with the present invention, in particular solid tumors may also be treated as follows with use of B-B4-DM1, as an example. This example is not intended to limit the present invention in any manner, and a skilled artisan could readily determine other immunoconjugates of the present invention and other treatment regimes which could be utilized for the treatment of solid tumors. The tumor is first treated to reduce the size of the tumor, for example chemotherapeutically or radioactively. Subsequent administration of the immunoconjugates of this invention provides a means for eliminating residual cancer cells. The administration of the immunoconjugate allows specific targeting of these residual cells and release of the effector molecules at the target site. This targeted delivery and release provides a significant advance in the treatment of residual cancer cells of solid tumors, for which current chemotherapy methods sometimes provide incomplete remission.
  • The present invention is further described by reference to the following Examples, which are offered by way of illustration and are not intended to limit the invention in any manner. Standard techniques well known in the art or the techniques specifically described below were utilized.
  • Materials and Methods
  • Preparation of mAb-DM1 Conjugate
  • The thiol-containing maytansinoid DM1 was synthesized from the microbial fermentation product ansamitocin P-3, as previously described by Chari (Chari et al, 1992). Characterization of murine B-B4 (Wijdenes, 1996) and preparation of humanized C242 (huC242) (Roguska, 1994) have been previously described. Antibody-drug conjugates were prepared as described by Liu et al (Liu, 1996). An average of 3.5 DM1 molecules was linked per antibody molecule.
  • Cell Lines and Patient Cells
  • CD138+ dexamethasone (Dex)-sensitive MM.1S and Dex-resistant MM.1R, Ocy-My5, OPM1 and OPM2 human MM cell lines and CD138 Waldenstrom Macroglobulinemia (WM) WSU-WM and the lymphoma (LB) SUDHL4 cell lines were used. Cell lines were cultured in RPMI-1640 medium (GIBCO) supplemented with 10% fetal bovine serum (FBS; Hyclone, Logan, Utah), L-glutamine, penicillin, and streptomycin (GIBCO) (denoted below as RPMI complete medium). Plasma cells (PC) and bone marrow (BM) cells were isolated using Ficoll-Hypaque density gradient sedimentation from BM aspirates, obtained from MM patients following informed consent. BM cells were separated. BMSCs were obtained by long-term cultures of BM cells (4-8 weeks) in RPMI 1640 medium supplemented with 20% FBS.
  • Gene Expression Analysis and Data Analysis: Expression of CD138 in MM Patients
  • Expression of CD138 on normal plasma cells and patient MM cells was evalutated. BM aspirate samples from normal donors and patients with MM were treated with 0.86% ammonium chloride to lyse red blood cells. PC were then isolated by positive immunomagnetic bead selection using anti-CD138 antibodies and Magnet Assisted Cell Sorting (“MACS,” Miltenyi Biotech). Purity of plasma cells (>95%) was assessed by flow cytometric (Becton-Dickinson “FACSort”) monitoring for CD38+/CD45lo phenotype as well as forward and side scatter and morphological characteristics.
  • Total RNA was isolated from 5×106 cells utilizing an “RNeasy® kit” (Qiagen Inc., Valencia, Calif.). Total RNA (10-15 μg) was reverse-transcribed to get cDNA using the “Superscript® II RT kit” (Invitrogen Life Technologies, Carlsbad, Calif.). cDNA was used in an in vitro transcription reaction to synthesize biotin-labeled cRNA utilizing “ENZO® RNA labeling kit” (Enzo Diagnostics, Inc., Farmingdale, N.Y.). Labeled cRNA was purified with the “RNeasy® Mini-kit” (Qiagen Inc., Valencia, Calif.) and quantitated. Purified cRNA (15 μg) was hybridized to Human Genome U133 (HG-U133) GeneChip® arrays (Affymetrix, Inc.) representing approximately 33,000 human genes, and GeneChip® arrays were scanned on a GeneArray® Scanner (Affymetrix, Inc., Santa Clara, Calif.). Normalization of arrays and calculation of expression values was performed using the DNA-Chip Analyzer (“dChip”) program. Arrays were normalized based on relative signal produced for an invariant subset of genes. This model-based method was used for probe selection and computing expression values. By pooling hybridization information across multiple arrays, it was possible to assess standard errors for the expression level indexes. This approach also allowed automatic probe selection in the analysis stage to reduce errors due to cross-hybridizing probes and image contamination.
  • Antibody Internalization
  • Internalization of B-B4 antibody was examined with a cultured CD138+ cell line by flow cytometry and under a fluorescent microscope. The antibody was modified by Alexa 488 dye (Molecular Probes), and the fluorescence of the non-internalized antibody bound to cells was quenched by exposure to an “anti-Alexa antibody” (Molecular Probes). Thus, semi-quantitatively discrimination between surface-bound and internalized antibody was possible. B-B4 was poorly internalized.
  • Colorimetric Survival Assay
  • Survival of CD138+ and CD138 cells upon administration of B-B4-DM1, B-B4 and DM1 was examined using a tetrazolium colorimetric assay (CellTiter 96® Non-Radioactive Cell Proliferation Assay; Promega, WI), as previously described (Mossmann, 1983). Cells (1×104) were plated in 24-well plates in 1 ml RPMI complete medium and then treated as indicated. At the end of each treatment, cells were incubated with 150 μl of Dye Solution and then incubated for 4 h at 37° C. A solubilization/stop solution was then added to each well under vigorous pipetting to dissolve the formazan crystals. Absorbance was measured at 570 nm, and cell viability was estimated as percentage of untreated controls. All experiments were repeated 3 times, and each experimental condition was repeated in triplicate wells in each experiment. Data reported are average values±SD of 3 representative experiments.
  • Cell Proliferation Assay
  • The effect of B-B4-DM1 on cell proliferation was measured by the extent of [3H]-thymidine (NEN Life Science Products, Boston, Mass.) incorporation. Cells (2×104 cells/well) were incubated in 96-well culture plates in the presence of 70%-80% confluent BMSCs at 37° C. with or without a test-agent (in triplicate wells). [3H]-thymidine (0.5 μCi) was then added to each well for the last 8 h. Cells were harvested onto glass filters with an automatic cell harvester (Cambridge Technology, Cambridge, Mass.) and counted using a Micro-Beta® Trilux counter (Wallac, Gaithersburgh, Md.).
  • Detection of Apoptosis
  • Dual staining with FITC-labeled Annexin V and propidium iodide (PI) was carried out to detect induction of apoptotic cell death by B-B4-DM1. After treatment of 1×106 tumor cells for 48 h, cells were washed with PBS and re-suspended in 100 □l of HEPES buffer containing Annexin V-FITC and propidium iodide (PI) (Annexin V-FLUOS staining kit; Roche Diagnostic, Indianapolis, Ind.). Following 15 min incubation at room temperature, cells were analyzed using a Coulter Epics XL flow cytometer for the presence of an Annexin V-FITC-positive/PI-negative apoptotic cell population.
  • Cell Cycle Analysis
  • 1×106 MM cells were incubated with or without a test-agent for 48 h, washed with PBS, permeabilized by a 30 min exposure to 70% ethanol at 4° C., incubated with PI (50-μg/mL) in 0.5 ml PBS containing 20 U/mL Rnase A (Roche) for 30 min at room temperature, and analyzed for DNA content by cell-associated fluorescence using a flow cytometer and CellQuest™ software.
  • In Vivo Activity
  • Human MM Xenograft Murine Model
  • In this model, CB-17 SCID mice were subcutaneously (s.c.) inoculated in the interscapular area with 5×106 OPM1 or OPM2 cells in 100 μl of RPMI-1640 medium. Treatment was initiated after the detection of palpable tumors. Tumor growth was measured weekly in two dimensions using a caliper, and volume was expressed in mm3 using the formula: V=0.5a×b2, where a and b are the long and short diameter of the tumor, respectively. Tumor size was evaluated from the first day of treatment until day of first sacrifice. The survival time is defined as the time interval between start of the experiment and either death or day of sacrifice. Mice were treated intavenously (i.v.) with vehicle alone (PBS), unconjugated B-B4 (13.3 μg/ml), B-B4-DM1 (conjugate containing 75 or 150 μg DM1/Kg per day), or control huC242-DM1 (150 μg DM1/Kg per day), for a total of 3 days. In addition, two mice bearing very large tumors (average size of 1309±60 mm3) were treated with B-B4-DM1 (150 μg DM1/kg per day) for a total of 3 days and observed for changes in the tumor size.
  • Autofluorescencent GFP+ Human MM Xenograft Model
  • Procedures for stably transfection of green fluorescent protein (GFP) in tumor cells and use have been previously described (Yang, 1999; Yang 2000). Five mice were injected s.c. with GFP+ OPM1 cells as described above. Mice were monitored by whole-body fluorescence imaging using “Illumatool Bright Light System LT-9900” (Lightools Research, Encinitas, Calif.). After accurate cutaneous shave of tumor area, fluorescence imaging results were digitally captured by a Sony® DSC-P5™ digital camera (Sony, New York, N.Y.) and analyzed with Adobe PhotoShop® 4.0.
  • SCID-hu Mouse Model
  • Human fetal bones were obtained from products of conceptions of second trimester abortions in compliance with state and federal regulations (Advanced Bioscience Resourses, ABR; Alameda, Calif.). The implantation of human fetal long bone grafts into SCID mice to produce SCID-hu mice has been previously described (McCune et al, 1988; Namikawa et al, 1988; Kyoizumi et al, 1993; Akkina et al, 1994; Chen et al, 1994; Sandhu et al, 1996; Urashima, 1997). In brief, the femurs or tibias of 19 to 23 gestational week fetuses were cut into fragments and implanted s.c. into SCID mice. After approximately 8 weeks, 2 to 5×106 BM cells from a MM patient or Ocy-My5 MM cells were injected in 50 μl PBS directly into human bone of SCID-hu hosts. Production and level of human paraprotein in mouse serum was an indicator of myeloma engrafinent and growth. At least 2 consecutive measurements, of increasing levels of circulating human immunoglobulin (huIg), signified human MM cell growth.
  • Measurement of Serum Paraprotein Concentration
  • Blood (50-100 μl) was withdrawn from the tail vein for measurement of human paraprotein in murine serum using ELISA (Bethyl, Montgomery, Tex.). Goat anti-human λ and κ antisera were used for capture and goat anti-human λ or κ HRP conjugates were used for detection.
  • Histopatological Analysis
  • Excised bone grafts were fixed in 10% buffered formalin; skeletal tissues were decalcified with 14% EDTA and embedded in paraffin by previously described standard techniques (Sasaki, 1995). Sections were then stained with H & E (Hematoxylin and eosin) for histopathological examination. Immunoperoxidase studies were performed on paraffin sections using an indirect technique as described (Urashima, 1997). Rabbit anti-human λ and κ antisera were used for detection of MM cells in fetal bone.
  • Statistical Analysis
  • Statistical significance of differences was determined using Student's t-test. Differences were considered significant when p<0.05.
  • Results and Discussion
  • CD138 is expressed on patient MM cells and is the most important target Ag for identification and selection of these cells. However previous reports show heterogenous CD138 expression on MM cells (Wijdenes, 1996; Dhodapkar, 1998; Witzig, 1996; Schneider, 1997; Rawstron, 1997). The expression of CD138 on patient MM cells was measured by gene profiling and flow cytometry. FIGS. 1A to 1C show the CD138 gene expression profiles of normal plasma cells (n=3) and patient MM cells (n=15) measured utilizing HG-U133 GeneChip® array (Affymetrix) data. FIG. 1A shows the individual fold increase in intensity of CD138 gene expression compared to normal PC; FIG. 1B shows the mean of intensity of CD138 gene expression in normal PC (n=3) and patient MM cells (n=15) and FIG. 1C the mean fold increase intensity of CD138 gene expression in MM cells (n=15) compared to normal PC (n=3). As can be seen, CD138 was expressed in all 15 MM specimens (100%) examined at a 95±8-fold mean increase in intensity relative to normal plasma cells.
  • Furthermore, flow cytometry was used to assess cell surface expression of CD138 on MM cells from 25 patients. Expression of CD138 on the CD38brightCD45lo cell population was assessed both by percentage of positive cells, and by mean fluorescence intensity (MFI). FIG. 1D shows the percentage of patients expressing CD138+ MM cells, as determined by flow cytometry on fresh BM aspirate samples. FIG. 1E shows the percentage of CD138+ MM cells in CD138+ patients on fresh BM aspirates and FIG. 1F shows MFI of CD138+ or CD138 MM cells within CD38brightCD45lo population. As can be seen from FIG. 1D 18 of 25 patients (72%) expressed CD138, with a mean of 68±31% CD138+ cells (FIG. 1E) and MFI of 1234±539 (range: 166-2208) (FIG. 1F). Taken together, these results indicate that CD138 is highly expressed in patient MM cells.
  • These results were consistent with previous reports showing CD138 expression on 60% and 100% of cases (Horvathova, 1995; Wijdenes, 1996). Possible explanations for the variability in CD138 detection by flow cytometry are the rapid shedding of protein during flow cytometric manipulation of specimens, the high turnover rate of the molecule on the cell membrane, lack of cell surface antigen (Ag) in pre-apoptotic plasma cells or Ag expression dependent on stage of the cell cycle (Clement, 1995). By immunohistochemistry, CD138 has been reported to be highly sensitive and specific marker of MM cells in 100% of BM biopsies (Chilosi, 1999). These data support the potential value of CD138 as a target for immunotherapeutic approaches in MM.
  • The effects of B-B4-DM1 on survival of CD138+ (MM-1S, MM-1R, Ocy-My5) and CD138 cells (SUDHL-4 and WSU-WM) were determined using an MTT assay. MM cell lines were exposed to unconjugated B-B4 mAb (FIG. 2A), immunoconjugate B-B4-DM1 (FIG. 2B), or free DM1 drug at equimolar concentrations (FIG. 2C). Cell survival was measured using an MTT assay. Data (mean±SD of triplicate experiments) are shown in FIGS. 2A to 2C as percentage of untreated controls. CD138+ MM cell lines MM-1S, MM-1R and Ocy-My5 were evaluated as well as CD138 cell lines including the lymphoma cell line SUDHL4 and the Waldestrom's Macroglobulinemia cell line WSU-WM.
  • As can be seen from FIG. 2B, treatment with B-B4-DM1 (1-50 nM) induced growth inhibition in CD138+ tumor cells in a time- and dose-dependent manner. This effect was clearly detected after 72 h in all CD138+ cells. B-B4-DM1 treatment of CD138+ OPM1 and OPM2 MM cells further confirmed these observations (data not shown). In contrast, B-B4-DM1 (1-50 nM) was not toxic to CD138 cells, even after treatment for 96 h. To confirm that inhibitory activity of the immunoconjugate is specifically related to mAb-delivered cytotoxicity, the effect of equimolar concentrations of B-B4 antibody or unconjugated drug DM1 were tested. Even the highest concentrations of B-B4 did not affect the growth of cells at 96 h, (FIG. 2A), whereas free DM1 was equally and highly cytotoxic in both CD138+ and CD138 cell lines (FIG. 2C). These data indicate that activity of the immunoconjugate is not related to the differential sensitivity of cells to the drug nor the intrinsic properties of the antibody.
  • Since adhesion of MM cells to BMSC (bone marrow stromal cells) protects MM cells against drug-induced apoptosis, the effect of B-B4-DM1 on proliferation of CD138+ (Ocy-My5) MM and CD138 (SUDHL-4) LB cells adherent to BMSC was evaluated.
  • Ocy-My5 (FIG. 3A) or SUDHL-4 (FIG. 3B) cells (2×104) were seeded on 70%-80% confluent BMSC for 24 h. Cell proliferation was measured by [3H]thymidine incorporation following 72 h treatment with B-B4-DM1 (10 nM). Values represent the mean [3H]-TdR incorporation (cpm) of triplicate cultures. As seen in FIGS. 3A and 3B, B-B4-DM1 (10 nM) significantly inhibited the proliferation of CD138+ Ocy-My5 cells, but had no significant effect on CD138 SUDHL-4 cells. Unconjugated B-B4 did not exert any significant effect, whereas free DM1 (10 nM) was cytotoxic to both cell lines.
  • CD56, another CD associated with MM, and CD138 expression was evaluated by flow cytometry. Previous experiments established that B-B4-DM1, even at a concentrations as high as 240 μM, did not affect binding of FITC-labeled anti-CD138 antibody to CD138-expressing cells. FIG. 3C shows the cytotoxic activity of B-B4-DM1 (10 nM) on CD138+/CD56+ patient MM cells cultured with BMSCs using flow cytometry. Following 72 h of treatment with the immunoconjugate, >90% reduction in the MM cells was observed. Taken together, these results indicate that B-B4-DM1 overcomes cell adhesion mediated drug resistance (CAM-DR).
  • To determine whether apoptotic cell death occurs in cells exposed to the immunoconjugate, CD138+ Ocy-My5 MM cells were incubated with B-B4-DM1 (10 nM) for 72 h. Apoptotic cell death was then measured by staining with annexin V and PI and flow cytometric analysis.
  • FIG. 4A shows the induction of apoptotic cell death in CD138+ Ocy-My5 MM cells after 48 h exposure to B-B4-DM1 (10 nM). Percentages of stained cells are reported in each quadrant. FIG. 4A shows a significant increase in both annexin V+/PI and annexin V+/PI+ fractions in CD138+ cells exposed to B-B4-DM1 and free DM1, whereas no significant differences were detected in cells treated with unconjugated mAb alone. FIG. 4B shows the effects of B-B4-DM1 treatment on the cell cycle. Ocy-My5 MM cells were exposed to B-B4 mAb (13.3 μg/ml) or B-B4-DM1 (10 nM) for 48 h, labeled with PI, and analyzed using flow cytometry. Percentages of cells in the S-phase (S) and G2/M phase (G2) are indicated. As shown in FIG. 4B, B-B4 mAb alone had no significant effect on the proportion of cells in G2/M phase compared to untreated cells (15% vs 16%), whereas exposure of MM cells to B-B4-DM1 induced a majority (88%) of cells into the G2/M phase.
  • A human MM s.c. xenograft model in SCID mice was used to study the in vivo activity of B-B4-DM1 against CD138+ OPM1 cells. In this model, the therapeutic efficacy of B-B4-DM1 was measured in mice bearing large palpable tumors (average size 453±74 mm3). Animals were treated daily i.v. for 3 consecutive days with vehicle alone (PBS 9phosphate buffered saline); n=5), unconjugated B-B4 (13.3 μg/ml; n=5), B-B4-DM1 (150 μg DM1/kg; n=5), or control huC242-DM1 (150 μg DM1/kg; n=5) which does not bind OPM1 cells. Tumor size and overall survival were monitored serially in this cohorts.
  • FIG. 5 shows the results obtained after CB-17 SCID mice were inoculated s.c. in the interscapular area with 5×106 OPM1 (A and B) or OPM2 (C and D) MM cells. Mice were treated i.v. with B-B4-DM1 or control mAbs for 3 consecutive days. Tumor volume was assessed in two dimensions using an caliper eletronic, and the volume was expressed in mm3 using the formula: V=0.5a×b2, where a and b are the long and short diameter of the tumor, respectively. Tumor volume and survival were calculated as described previously.
  • As shown in FIGS. 5A and 5B, vehicle alone, unconjugated B-B4 and huC242-DM1, had no significant effect on tumor growth (panel A) or survival (panel B). Importantly, treatment with 150 μg/kg of B-B4-DM1 induced tumor regression and a significant increase in survival (p<0.001). We also studied the effect induced by B-B4-DM1 (75 or 150 μg DM1/kg; n=10) against OPM2 MM cells. As shown in FIGS. 5C and D, treatment with 75 μg/kg of B-B4-DM1 induced a significant delay in tumor growth, and 150 μg/kg of B-B4-DM1 completely inhibited tumor growth. A significant increase in survival was also observed in mice treated at both dose levels (p<0.05) relative to animals treated with vehicle or huC242-DM1 alone. To confirm the activity of B-B4-DM1 (150 μg DM1/kg), animals bearing a significant burden of disease (average tumor size was 1309±60 mm3) were treated. FIGS. 6A to 6F also shows the results obtained when CB-17 SCID mice were inoculated s.c. in the interscapular area with 5×106 OPM1 MM cells. Again, mice were treated i.v. with B-B4-DM1 (150 μg DM1/kg) for a total of 3 consecutive days. Tumor volume was measured in two dimensions using a caliper, and the volume was expressed in mm3 using the formula: V=0.5a×b2, where a and b are the long and short diameter of the tumor, respectively.
  • As shown in FIGS. 6A to 6F, significant tumor regression was induced by B-B4-DM1 treatment. Taken together, these results indicate that B-B4-DM1 is highly active in controlling tumor growth in a murine xenograft model of human MM.
  • Since expression of reporter genes encoding fluorescent proteins are sensitive method for in vivo detection of localized tumor growth as well as distant metastasis, the OPM1 MM cells were transfected with green fluorescent protein (GFP) and B-B4-DM1 activity was further characterized. In particular, mice were injected s.c. with GFP+ OPM1 cells, followed by serial whole-body fluorescence imaging to assess development of GFP+ tumors. Mice were then treated with B-B4-DM1 (150 μg DM1/kg; n=5). FIG. 7A shows a flow cytometry analysis of GFP+ OPM1 cells, indicating a ˜2-log difference in MFI of transfected cells. FIG. 7B shows results from five animals being injected with 5×106 GFP+ cells, monitored with whole-body fluorescence imaging for tumor development, and then treated with B-B4-DM1 (150 μg DM1/kg). Tumor sizes were determined directly by imaging the GFP-expressing tumor. FIGS. 7 C and D are representative whole-body fluorescence imaging from a mouse treated with B-B4-DM1. FIGS. 7E and 7F are negative images of the representative mouse. As seen in FIGS. 7B and 7C to 7F, B-B4-DM1 induced significant regressions of GFP+ tumors, confirming high activity of the immunoconjugate against CD138+ MM cells.
  • Since the SCID-hu model of MM accurately reproduces the pathological behaviours of the disease, the efficacy of B-B4-DM1 treatment was tested in (i) SCID-hu mice injected with patient MM cells and (ii) SCID-hu mice injected with Ocy-My5 MM cell line (Urashima, 1997). The activity of the immunoconjugate on disease confined to the human fetal bone chip implanted s.c. in mice was studied. Four mice with patient MM cells growing in human bone environment increasing serum huIg levels, were treated with either B-B4-DM1 (150 μg DM1/kg) or the control huC242-DM1 (150 μg DM1/kg).
  • In FIGS. 8A and 8B the results of monitoring mice for changes in levels of human κ chain as an indicator of disease burden are shown. The Figure shows a significant reduction of κ levels after treatment with B-B4-DM1. FIG. 8C shows final human κ chain levels (mean±SD) (n=4) after treatment. As seen in FIGS. 8A to 8C, treatment with B-B4-DM1 induced a significant reduction of human paraprotein, whereas human paraprotein continued to rise in mice treated with control antibody.
  • The activity of B-B4-DM1 after injection of Ocy-My5 cells in human fetal bone, tumor cell growth in bone (FIGS. 9A and 9B) and subsequent spread to surrounding tissues was also studied (Urashima, 1997) were treated with either B-B4-DM1 (150 μg DM1/kg) or the control huC242-DM1 (150 μg DM1/kg). FIGS. 9A and 9B show representative human bone sections after implantation of Ocy-My5 cells and before treatment. Sections are respectively stained by H & E and with anti-λ mAb. Finally, the activity of B-B4-DM1 on survival of tumor bearing mice was studied. FIG. 9C shows the survival of mice measured from the first day of treatment to the day of death or sacrifice. Figure shows a significant prolongation of survival after treatment with B-B4-DM1 As seen in FIG. 9C, treatment with B-B4-DM1 (150 μg DM1/kg) induced a significant prolongation in survival compared with control huC242-DM1 (150 μg DM1/kg) therapy. Taken together, these results confirm in vivo B-B4-DM1 activity in preclinical models which mimic many features of human MM.
  • It will be appreciated that the methods and compositions of the instant invention can be incorporated in the form of a variety of embodiments, only a few of which are disclosed herein. It will be apparent to the artisan that other embodiments exist and do not depart from the spirit of the invention. Thus, the described embodiments are illustrative and should not be construed as restrictive.
  • REFERENCES
    • Akkina R K, Rosenblatt J D, Campbell A G, Chen I S, Zack J A. Modeling human lymphoid precursor cell gene therapy in the SCID-hu mouse. Blood. 1994; 84:1393-1398.
    • Anttonen A, Heikkila P, Kajanti M, Jalkanen M, Joensuu H. High syndecan-1 expression is associated with favourable outcome in squamous cell lung carcinoma treated with radical surgery. Lung Cancer. 2001 June; 32(3):297-305.
    • Barbareschi M, Maisonneuve P, Aldovini D, Cangi M G, Pecciarini L, Angelo Mauri F, Veronese S, Caffo O, Lucenti A, Palma P D, Galligioni E, Doglioni C. High syndecan-1 expression in breast carcinoma is related to an aggressive phenotype and to poorer prognosis. Cancer. 2003 Aug. 1; 98(3):474-83.
    • Bernfield M, Kokenyesi R, Kato M, Hinkes M T, Spring J, Gallo R L, Lose E J. Biology of the syndecans: a family of transmembrane heparan sulfate proteoglycans. Annu Rev Cell Biol. 1992; 8:365-393.
    • Beste G, Schmidt F S, Stibora T, Skerra A. Small antibody-like proteins with prescribed ligand specificities derived from the lipocalin fold. Proc. Natl. Acad. Sci. USA. 1999: 96, 1898-1903.
    • Bhattacharyya B, Wolff J. Maytansine binding to the vinblastine sites of tubulin. FEBS Lett. 1977; 75:159-162.
    • Blättler W A and Chari R V J. Drugs to Enhance the Therapeutic Potency of Anticancer Antibodies: Antibody-Drug Conjugates as Tumor-Activated Prodrugs. In: Ojima, I., Vite, G. D. and Altmann, K.-H., Editors, 2001. Anticancer Agents-Frontiers in Cancer Chemotherapy, American Chemical Society, Washington, D.C., pp. 317-338.
    • Bross P F, Beitz J, Chen G, Chen X H, Duffy E, Kieffer L, Roy S, Sridhara R, Rahman A, Williams G, Pazdur R. Approval summary: gemtuzumab ozogamicin in relapsed acute myeloid leukemia. Clin Cancer Res. 2001; 7:1490-1496.
    • Carbone A, Gaidano G, Gloghini A, Ferlito A, Rinaldo A, Stein H. AIDS-related plasma-blastic lymphomas of the oral cavity and jaws: a diagnostic dilemma. Ann. Otol. Rhinol. Laryngol. 1999; 108: 95-99.
    • Carter P. Improving the efficacy of antibody-based cancer therapies. Nat Rev Cancer. 2001; 1:118-129.
    • Chari R V, Martell B A, Gross J L, Cook S B, Shah S A, Blattler W A, McKenzie S J, Goldmacher V S. Immunoconjugates containing novel maytansinoids: promising anticancer drugs. Cancer Res. 1992; 52:127-131.
    • Chari R V, Jackel K A, Bourret L A, Derr S M, Tadayoni B M, Mattocks K M, Shah S A, Liu C, Blättler W A and Goldmacher V S. Enhancement of the selectivity and antitumor efficacy of a CC-1065 analogue through immunoconjugate formation. Cancer Res. 1995; 55: 4079-4084.
    • Charnaux N, Brule S, Chaigneau T, Saffar L, Sutton A, Hamon M, Prost C, Lievre N, Vita C, Gattegno L. RANTES (CCL5) induces a CCR5-dependent accelerated shedding of syndecan-1 (CD138) and syndecan-4 from HeLa cells and forms complexes with the shed ectodomains of these proteoglycans as well as with those of CD44. Glycobiology. 2004 Sep. 8 [Epub ahead of print]
    • Chen B P, Galy A, Kyoizumi S, Namikawa R, Scarborough J, Webb S, Ford B, Cen D Z, Chen S C. Engraftment of human hematopoietic precursor cells with secondary transfer potential in SCID-hu mice. Blood. 1994; 84:2497-2505.
    • Chilosi M, Adami F, Lestani M, Montagna L, Cimarosto L, Semenzato G, Pizzolo G, Menestrina F. CD138/syndecan-1: a useful immunohistochemical marker of normal and neoplastic plasma cells on routine trephine bone marrow biopsies. Mod Pathol. 1999; 12:1101-1106.
    • Clement C, Vooijs, W. C., Klein, B., and Wijdenes, J. In: al. SFSe, ed. J. Leukocyte Typing V. Oxford: Oxford University Press; 1995:714-715.
    • Couturier O, Faivre-Chauvet A; Filippovich I V; Thedréz P, Saï-Maurel C; Bardiés M; Mishra A K; Gauvrit M; Blain G; Apostolidis C; Molinet R; Abbe J C; Bateille R; Wijdenes J; Chatal J F; Cherel M; Validation of 213Bi-alpha radioimmunotherapy for multiple myeloma. Clinical Cancer Research 5(10 Suppl.) (October 1999) 3165s-3170s.
    • Davies E J et al., Blackhall F H, Shanks J H, David G, McGown A T, Swindell R, Slade R J, Martin-Hirsch P, Gallagher J T, Jayson G C. Distribution and Clinical Significance of Heparan Sulfate Proteoglycans in Ovarian Cancer Clin Cancer Res. 2004; 10(15):5178-86.
    • Dhodapkar M V, Abe E, Theus A, Lacy M, Langford J K, Barlogie B, Sanderson R D. Syndecan-1 is a multifunctional regulator of myeloma pathobiology: control of tumor cell survival, growth, and bone cell differentiation. Blood. 1998; 91:2679-2688.
    • Dore J M, Morard F, Vita N, Wijdenes J. Identification and location on syndecan-1 core protein of the epitopes of B-B2 and B-B4 monoclonal antibodies. FEBS Lett. 1998; 426:67-70.
    • Dowell J A, Korth-Bradley J, Liu H, King S P, Berger M S. Pharmacokinetics of gemtuzumab ozogamicin, an antibody-targeted chemotherapy agent for the treatment of patients with acute myeloid leukemia in first relapse. J Clin Pharmacol. 2001; 41:1206-1214.
    • Edinger M, Sweeney T J, Tucker A A, Olomu A B, Negrin R S, Contag C H. Noninvasive assessment of tumor cell proliferation in animal models. Neoplasia. 1999; 1:303-310.
    • Gattei V, Godeas C, Degan M, Rossi F M, Aldinucci D, Pinto A. Characterization of Anti-CD138 monoclonal antibodies as tools for investigating the molecular polymorphism of syndecan-1 in human lymphoma cells. Br J Haematol. 1999; 104:152-162.
    • Hamann P R, Hinman L M, Beyer C F, Lindh D, Upeslacis J, Flowers D A, Bernstein I. An anti-CD33 antibody-calicheamicin conjugate for treatment of acute myeloid leukemia. Choice of linker. Bioconjug Chem. 2002; 13:40-46.
    • Han I, Park H, Oh E S. New insights into syndecan-2 expression and tumourigenic activity in colon carcinoma cells. J Mol Histol. 2004: 35(3):319-26.
    • Horvathova M, Gaillard, J.-P., Liutard, J., Duperray, C., Lavabre-Bertrand, T., Bourquard, P et al. In: al. SFSe, ed. Leucocyte Typing V. Oxford: Oxford University Press; 1995: 713-714.
    • Krebs B, Rauchenberger R, Reiffert S, Rothe C, Tesar M, Thomassen E, Cao M, Dreier T, Fischer D, Hoss A et al. High-throughput generation and engineering of recombinant human antibodies. 2001. J. Immunol. Methods 254, pp. 67-84.
    • Kupchan S M, Sneden A T, Branfman A R, Howie G A, Rebhun L I, Mclvor W E, Wang R W, Schnaitman T C. Structural requirements for antileukemic activity among the naturally occurring and semisynthetic maytansinoids. J Med. Chem. 1978; 21:31-37.
    • Kyoizumi S, Baum C M, Kaneshima H, McCune J M, Yee E J, Namikawa R. Implantation and maintenance of functional human bone marrow in SCID-hu mice. Blood. 1992; 79:1704-1711.
    • Kyoizumi S, Murray L J, Namikawa R. Preclinical analysis of cytokine therapy in the SCID-hu mouse. Blood. 1993; 81:1479-1488.
    • Liu C, Tadayoni B M, Bourret L A, Mattocks K M, Derr S M, Widdison W C, Kedersha N L, Ariniello P D, Goldmacher V S, Lambert J M, Blattler W A, Chari R V. Eradication of large colon tumor xenografts by targeted delivery of maytansinoids. Proc Natl Acad Sci USA. 1996; 93:8618-8623.
    • McCune J M, Namikawa R, Kaneshima H, Shultz L D, Lieberman M, Weissman I L. The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function. Science. 1988; 241:1632-1639.
    • Mennerich D, Vogel A, Klaman I, Dahl E, Lichtner R B, Rosenthal A, Pohlenz H D, Thierauch K H, Sommer A. Shift of syndecan-1 expression from epithelial to stromal cells during progression of solid tumours. Eur J Cancer. 2004 June; 40(9):1373-82.
    • Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983; 65:55-63.
    • Namikawa R, Ueda R, Kyoizumi S. Growth of human myeloid leukemias in the human marrow environment of SCID-hu mice. Blood. 1993; 82:2526-2536.
    • O'Connell F P, Pinkus J L, Pinkus G S. CD138 (Syndecan-1), a Plasma Cell Marker Immunohistochemical Profile in Hematopoietic and Nonhematopoietic Neoplasms. Am J Clin Pathol 2004; 121:254-263.
    • Ojima I, Geng X, Wu X, Qu C, Borella C P, Xie H, Wilhelm S D, Leece B A, Bartle L M, Goldmacher V S and Chari R V. Tumor-specific novel taxoid-monoclonal antibody conjugates. 2002. J. Med. Chem. 45, pp. 5620-5623.
    • Olafsen, T, Cheung, C C, Yazaki, P J, Li L, Sundaresan G, Gambhir S S, Sherman, M A, Williams, L E, Shively, J E, Raubitschek, A A, and Wu, A M. Covalent disulfide-linked anti-CEA diabody allows site-specific conjugation and radiolabeling for tumor targeting applications. 2004; Prot. Eng. Design & Selection 17:1: 21-27.
    • Orosz Z, Kopper L. Syndecan-1 expression in different soft tissue tumours. Anticancer Res. 2001: 21 (1B):733-7.
    • Padlan, EA. A possible procedure for reducing the immunogenicity of antibody variable domains while preserving their ligand-binding properties. Mol. Immunol. 1991; 28: 489-498.
    • Payne G. Progress in immunoconjugate cancer therapeutics. Cancer Cell. 2003; 3:207-212.
    • Pegram M D, Lipton A, Hayes D F, Weber B L, Baselga J M, Tripathy D, Baly D, Baughman S A, Twaddell T, Glaspy J A and Slamon D J. Phase II study of receptor-enhanced chemosensitivity using recombinant humanized anti-p185HER2/neu monoclonal antibody plus cisplatin in patients with HER2/neu-overexpressing metastatic breast cancer refractory to chemotherapy treatment. 1998. J. Clin. Oncol. 16, pp. 2659-2671.
    • Rawstron A C, Owen R G, Davies F E, Johnson R J, Jones R A, Richards S J, Evans P A, Child J A, Smith G M, Jack A S, Morgan G J. Circulating plasma cells in multiple myeloma: characterization and correlation with disease stage. Br J Haematol. 1997; 97:46-55.
    • Remillard S, Rebhun L I, Howie G A, Kupchan S M. Antimitotic activity of the potent tumor inhibitor maytansine. Science. 1975; 189:1002-1005.
    • Roguska M A, Pedersen J T, Keddy C A, Henry A H, Searle S J, Lambert J M, Goldmacher V S, Blattler W A, Rees A R, Guild B C. Humanization of murine monoclonal antibodies through variable domain resurfacing. Proc Natl Acad Sci USA. 1994; 91:969-973.
    • Ross S, Spencer S D, Holcomb I, Tan C, Hongo J, Devaux B, Rangell L, Keller G A, Schow P, Steeves R M, Lutz R J, Frantz G, Hillan K, Peale F, Tobin P, Eberhard D, Rubin M A, Lasky L A, Koeppen H. Prostate stem cell antigen as therapy target: tissue expression and in vivo efficacy of an immunoconjugate. Cancer Res. 2002 May 1; 62(9):2546-53.
    • Ross J S, Gray K, Gray G, Worland P J, Rolfe M. Anticancer Antibodies, Am J Clin Path. (Apr. 17, 2003).
    • Sanderson R D, Lalor P, Bernfield M. B lymphocytes express and lose syndecan at specific stages of differentiation. Cell Regul. 1989; 1:27-35.
    • Sandhu J S, Clark B R, Boynton E L, Atkins H, Messner H, Keating A, Hozumi N. Human hematopoiesis in SCID mice implanted with human adult cancellous bone. Blood. 1996; 88:1973-1982.
    • Sasaki A, Boyce B F, Story B, Wright K R, Chapman M, Boyce R, Mundy G R, Yoneda T. Bisphosphonate risedronate reduces metastatic human breast cancer burden in bone in nude mice. Cancer Res. 1995; 55:3551-3557.
    • Schneider U, van Lessen A, Huhn D, Serke S. Two subsets of peripheral blood plasma cells defined by differential expression of CD45 antigen. Br J Haematol. 1997; 97:56-64.
    • Sebestyen A, Berczi L, Mihalik R, Paku S, Matolcsy A, Kopper L. Syndecan-1 (CD138) expression in human non-Hodgkin lymphomas. Br J Haematol. 1999; 104(2):412-9.
    • Seftalioglu A, Karakus S. Syndecan-1/CD138 expression in normal myeloid, acute lymphoblastic and myeloblastic leukemia cells. Acta Histochem. 2003; 105:213-221.
    • Seftalioglu A, Karakus S, Dundar S, Can B, Erdemli E, Irmak M K, Oztas E, Korkmaz C, Yazar F, Cavusoglu I. Syndecan-1 (CD138) expression in acute myeloblastic leukemia cells—an immuno electron microscopic study. Acta Oncol. 2003; 42:71-74.
    • Senter P D, Doronina S, Cerveny C, Chace D, Francisco J, Klussman K, Mendelsohn B, Meyer D, Siegall C B, Thompson J et al. (2002). Cures and regressions of established tumors with monoclonal antibody auristatin conjugates. Abstract #2062, American Assoication for Cancer Res. (San Francisco, Calif.: American Association for Cancer Res.), 414.
    • Sievers E L, Larson R. A., Stadtmauer, E. A., Estey, E., Lowenberg, B., Dombret, H., Karanes, C., Theobald, M., Bennett, J. M., Sherman, M. L. et al. Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive acute myeloid leukemia in first relapse. 2001. J. Clin. Oncol. 19, pp. 3244-3254.
    • Sievers E L and Linenberger M. Mylotarg: antibody-targeted chemotherapy comes of age. 2001. Curr. Opin. Oncol. 13, pp. 522-527.
    • Smith R., Single chain antibody variable region fragments; www.stanford.edu/˜smithr/science/scfv.html (last updated on May, 2001). Studnicka G M, Soares S, Better M, Williams R E, Nadell R, Horwitz A H. Human-engineered monoclonal antibodies retain full specific binding activity by preserving non-CDR complementarity-modulating residues. Protein Eng. 1994: 7(6): 805-814.
    • Tolcher A W, Ochoa L, Hammond L A, Patnaik A, Edwards T, Takimoto C, Smith L, de Bono J, Schwartz G, Mays T, Jonak Z L, Johnson R, DeWitte M, Martino H, Audette C, Maes K, Chari R V, Lambert J M, Rowinsky E K. Cantuzumab mertansine, a maytansinoid immunoconjugate directed to the CanAg antigen: a phase 1, pharmacokinetic, and biologic correlative study. J Clin Oncol. 2003; 21:211-222.
    • Urashima M, Chen B P, Chen S, Pinkus G S, Bronson R T, Dedera D A, Hoshi Y, Teoh G, Ogata A, Treon S P, Chauhan D, Anderson K C. The development of a model for the homing of multiple myeloma cells to human bone marrow. Blood. 1997; 90:754-765.
    • Vogel C W. Preparation of immunoconjugates using antibody oligosaccharide moieties. Methods in Molecular Biology: Bioconjugation protocols strategies and methods. 2004; 283:087-108.
    • Vooijs W C, Post J, Wijdenes J, Schuurman H J, Bolognesi A, Polito L, Stirpe F, Bast E J, de Gast G C. Efficacy and toxicity of plasma-cell-reactive monoclonal antibodies B-B2 and B-B4 and their immunotoxins. Cancer Immunol Immunother. 1996; 42:319-328.
    • Ward, E. S., D. Gussow, A. D. Griffiths, P. T. Jones, and G. Winter. Binding activities of a repertoire of single immunoglobin variable domains secreted from Escherichia coli. Nature. 1989. 341:544-546.
    • Wargalla U C, Reisfeld R A. Rate of internalization of an immunotoxin correlates with cytotoxic activity against human tumor cells. Proc. Natl. Acad. Sci. USA. 1989; 86:5146-5150.
    • Wijdenes J, Vooijs W C, Clement C, Post J, Morard F, Vita N, Laurent P, Sun R X, Klein B, Dore J M. A plasmocyte selective monoclonal antibody (B-B4) recognizes syndecan-1. Br J Haematol. 1996; 94:318-323.
    • Wijdenes J, Dore J M, Clement C, Vermot-Desroches C. CD138, J Biol Regul Homeost Agents. 2002 April-June; 16(2):152-5.
    • Witzig T E, Kimlinger T K, Ahmann G J, Katzmann J A, Greipp P R. Detection of myeloma cells in the peripheral blood by flow cytometry. Cytometry. 1996; 26:113-120.
    • Xie H, Audette C, Hoffee M, Lambert J M, Blättler W. Pharmacokinetics and biodistribution of the antitumor immunoconjugate, cantuzumab mertansine (huC242-DM1), and its two components in mice. J Pharmacol Exp Ther. 2004 March; 308(3): 1073-82.
    • Yang M, Jiang P, An Z, Baranov E, Li L, Hasegawa S, Al-Tuwaijri M, Chishima T, Shimada H, Moossa A R, Hoffman R M. Genetically fluorescent melanoma bone and organ metastasis models. Clin Cancer Res. 1999; 5:3549-3559.
    • Yang M, Baranov E, Jiang P, Sun F X, Li X M, Li L, Hasegawa S, Bouvet M, Al-Tuwaijri M, Chishima T, Shimada H, Moossa A R, Penman S, Hoffman R M. Whole-body optical imaging of green fluorescent protein-expressing tumors and metastases. Proc Natl Acad Sci USA. 2000; 97:1206-1211.

Claims (37)

1. An immunoconjugate comprising:
at least one targeting agent selectively targeting cell-surface expressed syndecan-1,
at least one effector molecule,
wherein said effector molecule has, in its native form, high non-selective cytotoxicity,
wherein said targeting agent is functionally attached to said effector molecule to form said immunoconjugate, and
wherein said effector molecule has substantially no non-selective cytotoxicity when part of said immunoconjugate.
2. The immunoconjugate of claim 1, wherein the cytotoxicity of said effector molecule, in its native form, on cells targeted by said targeting agent is higher or about the same as the cytotoxicity of said immunoconjugate on said targeted cells.
3. The immunoconjugate of claim 1, wherein said effector molecule has, in its native form, a potency of about 10−11-10−8 M.
4. The immunoconjugate of claim 1, wherein said effector molecule is a maytansinoid, a CC1065 analogue, a calicheamicin or a taxane.
5. The immunoconjugate of claim 4, wherein said effector molecule is a maytansinoid.
6. The immunoconjugate of claim 5, wherein said maytansinoid is DM1.
7. The immunoconjugate of claim 5, wherein said maytansinoid is DM3.
8. The immunoconjugate of claim 5, wherein said maytansinoid is DM4.
9. The immunoconjugate of claim 1, wherein said effector molecule has a molecular weight of less than 5 kDa.
10. The immunoconjugate of claim 9, wherein said effector molecule has a molecular weight of between about 600 and about 800 Da.
11. The immunoconjugate of claim 1, wherein said targeting agent is a targeting antibody or non-immunoglobulin targeting molecule.
12. The immunoconjugate of claim 11, wherein said targeting agent is a targeting antibody.
13. The immunoconjugate of claim 12, wherein said targeting antibody is an antibody fragment.
14. The immunoconjugate of claim 12, wherein said targeting antibody is derived from an antibody that internalizes poorly.
15. The immunoconjugate of claim 12, wherein said targeting antibody is derived from the antibody B-B4.
16. A pharmaceutical composition comprising an effective amount of the immunoconjugate according to claim 1 and one or more pharmaceutically acceptable excipients.
17. A kit comprising, in separate containers, pharmaceutical compositions for use in combination to inhibit, delay and/or prevent the growth of tumors and/or spread of tumor cells, wherein one container comprises an effective amount of the pharmaceutical composition of claim 16, and wherein, a separate container comprises a second pharmaceutical composition comprising an effective amount of an agent for the inhibition, delay and/or prevention of the growth of tumors and/or spread of tumor cells, and one or more pharmaceutically acceptable excipients.
18. The kit of claim 17, wherein said agent in said second pharmaceutical composition is a chemotherapeutic agent or another immunoconjugate.
19. A method for treating inhibiting, delaying and/or preventing the growth of tumor cells in a cell culture containing syndecan-1 expressing tumor cells and non-tumor cells comprising
administering a growth of syndecan-1 expressing tumor cells inhibiting, delaying and/or preventing effective amount of the immunoconjugate of claim 1.
20. The method of claim 19, wherein said effective amount induces cell death or continuous cell cycle arrest in said syndecan-1 expressing tumor cells.
21. The method of claim 19, wherein said cells in said cell culture are obtained from a cancer patient and wherein, after administration of said effective amount of said immunoconjugate, the cells of said cell culture are reimplanted into said cancer patient.
22. The method of claim 19, wherein the cells in said cell culture were isolated from a patient suffering from an hematologic malignancy and/or a solid tumor comprising syndecan-1 expressing cells.
23. The method of claim 22, wherein said patient is suffering from one of the following: multiple myeloma, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma, Hodgkin's and non-Hodgkin's lymphoma, chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML), a solid tissue sarcoma or a colon carcinoma.
24. The method of claim 23, wherein said patient is suffering from multiple myeloma.
25. A method for inhibiting, delaying and/or preventing the growth of a tumor comprising syndecan-1 expressing tumor cells and/or spread of syndecan-1 expressing tumor cells in a patient in need thereof, comprising
administering to said patient at least one immunoconjugate in a growth of said tumor and/or spreading of said tumor cells inhibiting or reducing amount,
wherein said immunoconjugate comprises at least one effector molecule and at least one targeting agent, and
wherein said immunoconjugate selectively inhibits, delays or prevents the growth and/or spread of syndecan-1 expressing cells.
26. The method of claim 25, wherein said patient suffers from an hematologic malignancy and/or a solid tumor comprising syndecan-1 expressing cells.
27. The method of claim 26, wherein said patient suffers from one of the following: multiple myeloma, ovarian carcinoma, kidney carcinoma, gall bladder carcinoma, breast carcinoma, prostate cancer, lung cancer, colon carcinoma, Hodgkin's and non-Hodgkin's lymphoma, chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), acute myeloblastic leukemia (AML), solid tissue sarcoma or colon carcinoma.
28. The method of claim 27, wherein the disease is multiple myeloma.
29. The method of claim 25, wherein said effector molecule exhibits, in its native form, high non-selective cytotoxicity.
30. The method of claim 25, wherein said effector molecule of said immunoconjugate is a toxin, cytotoxic enzyme, low molecular weight cytotoxic drug, a pore-forming agent, biological response modifier, prodrug activating enzyme, an antibody, cytokine or a radionuclide.
31. A method for inhibiting, delaying and/or preventing the growth of a tumor and/or spread of malignant tumor cells in a patient in need thereof, comprising
(a) administering to said patient one or more cancer drugs and/or radiation in an amount effective to reduce tumor load; and
(b) administering to said patient at least one immunoconjugate in a growth of a tumor and/or spreading of tumor cells inhibiting, delaying or preventing amount,
wherein said immunoconjugate selectively inhibits, delays or prevents the growth and/or spread of syndecan-1 expressing cells.
32. The method of claim 31, wherein (a) and (b) are performed consecutively in two consecutive treatment regimes.
33. The method of claim 31, wherein the at least one drug of (a) and the immunoconjugate of (b) are administered in a single administration step.
34. A method for treating a subject having a condition that would benefit from the selective suppression of myeloma cell survival, the method comprising:
(a) providing at least one immunoconjugate that selectively binds to syndecan-1 expressed on myeloma cells; and
(b) administering the immunoconjugate to the subject to selectively decrease survival or growth of said myeloma cells of said subject.
35. The method of claim 34, wherein the immunoconjugate comprises a B-B4 targeting antibody.
36. The method of claim 34, wherein the immunoconjugate comprises a maytansinoid effector molecule.
37. The method of claim 34, wherein said selective suppression of myeloma cell survival induces growth arrest or cell death in myeloma cells.
US10/975,434 2004-08-30 2004-10-29 Immunoconjugates targeting syndecan-1 expressing cells and use thereof Abandoned US20060045877A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/975,434 US20060045877A1 (en) 2004-08-30 2004-10-29 Immunoconjugates targeting syndecan-1 expressing cells and use thereof
US11/735,644 US8840898B2 (en) 2004-08-30 2007-04-16 Immunoconjugates targeting syndecan-1 expressing cells and use thereof
US14/327,888 US20150010585A1 (en) 2004-08-30 2014-07-10 Immunoconjugates targeting syndecan-1 expressing cells and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US60539404P 2004-08-30 2004-08-30
US10/975,434 US20060045877A1 (en) 2004-08-30 2004-10-29 Immunoconjugates targeting syndecan-1 expressing cells and use thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/735,644 Division US8840898B2 (en) 2004-08-30 2007-04-16 Immunoconjugates targeting syndecan-1 expressing cells and use thereof

Publications (1)

Publication Number Publication Date
US20060045877A1 true US20060045877A1 (en) 2006-03-02

Family

ID=35997729

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/975,434 Abandoned US20060045877A1 (en) 2004-08-30 2004-10-29 Immunoconjugates targeting syndecan-1 expressing cells and use thereof
US11/735,644 Expired - Fee Related US8840898B2 (en) 2004-08-30 2007-04-16 Immunoconjugates targeting syndecan-1 expressing cells and use thereof
US14/327,888 Abandoned US20150010585A1 (en) 2004-08-30 2014-07-10 Immunoconjugates targeting syndecan-1 expressing cells and use thereof

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/735,644 Expired - Fee Related US8840898B2 (en) 2004-08-30 2007-04-16 Immunoconjugates targeting syndecan-1 expressing cells and use thereof
US14/327,888 Abandoned US20150010585A1 (en) 2004-08-30 2014-07-10 Immunoconjugates targeting syndecan-1 expressing cells and use thereof

Country Status (3)

Country Link
US (3) US20060045877A1 (en)
AU (1) AU2004224925C1 (en)
CA (1) CA2486285C (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080280779A1 (en) * 2006-09-26 2008-11-13 Shaughnessy Jr John D Gene expression profiling based identification of genomic signatures of multiple myeloma and uses thereof
US20090169570A1 (en) * 2007-12-26 2009-07-02 Benjamin Daelken Methods and agents for improving targeting of cd138 expressing tumor cells
US20090175863A1 (en) * 2007-12-26 2009-07-09 Elmar Kraus Agents targeting cd138 and uses thereof
US20090181038A1 (en) * 2007-12-26 2009-07-16 Gregor Schulz Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
US20090232810A1 (en) * 2007-12-26 2009-09-17 Elmar Kraus Immunoconjugates targeting cd138 and uses thereof
WO2010019227A1 (en) * 2008-08-13 2010-02-18 George Mason Intellectual Properties, Inc. Ex vivo therapeutic screening of living bone marrow cells for multiple myeloma
WO2010126551A1 (en) 2009-04-30 2010-11-04 Immunogen, Inc. Potent conjugates and hydrophilic linkers
US20110123554A1 (en) * 2009-05-06 2011-05-26 Frank Osterroth Uses of immunoconjugates targeting cd138
EP3266469A2 (en) 2008-04-30 2018-01-10 ImmunoGen, Inc. Cross-linkers and their uses
US10117932B2 (en) 2011-12-08 2018-11-06 Biotest Ag Uses of immunoconjugates targeting CD138
EP3480202A1 (en) 2009-06-03 2019-05-08 ImmunoGen, Inc. Conjugation methods
WO2024000262A1 (en) * 2022-06-29 2024-01-04 京东方科技集团股份有限公司 Probe and kit for early diagnosis of diffuse large b-cell lymphoma
US11945868B2 (en) 2017-10-02 2024-04-02 Visterra, Inc. Antibody molecules to CD138 and uses thereof

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070054332A1 (en) * 2005-08-10 2007-03-08 Alan Rapraeger Syndecan 1 ectodomain inhibits cancer
AU2013201618B2 (en) * 2009-05-06 2016-06-02 Biotest Ag Uses of immunoconjugates targeting CD138
EA201991268A3 (en) 2011-03-29 2020-01-31 Иммуноджен, Инк. OBTAINING MAYTANSINOID-ANTIBODIES CONJUGATES IN ONE-STEP METHOD
WO2012135517A2 (en) 2011-03-29 2012-10-04 Immunogen, Inc. Preparation of maytansinoid antibody conjugates by a one-step process
SG10201702737TA (en) 2012-10-04 2017-05-30 Immunogen Inc Use of a pvdf membrane to purify cell-binding agent cytotoxic agent conjugates
CA2966932A1 (en) 2014-11-19 2016-05-26 Immunogen, Inc. Process for preparing cell-binding agent-cytotoxic agent conjugates
RU2611685C2 (en) * 2015-07-20 2017-02-28 Илья Владимирович Духовлинов Humanized monoclonal antibody specific to syndecan-1
RU2018130108A (en) 2016-02-05 2020-03-06 Иммуноджен, Инк. EFFECTIVE METHOD FOR PRODUCING CONJUGATES BINDING A CELL AGENT CYTOTOXIC AGENT
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. Multispecific molecules that bind to bcma and uses thereof
CA3108754A1 (en) 2018-08-06 2020-02-13 Daiichi Sankyo Company, Limited Combination of antibody-drug conjugate and tubulin inhibitor
KR20230049463A (en) * 2021-10-06 2023-04-13 주식회사 이뮤노로지컬디자이닝랩 Recombinant protein which recognizes CD138 and the composition comprising the same for treating cancer

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444887A (en) * 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4761111A (en) * 1981-09-18 1988-08-02 Brown Andrew M Automobile lifting and towing equipment
US5208020A (en) * 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5565332A (en) * 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US5639641A (en) * 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US5814318A (en) * 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US6005079A (en) * 1992-08-21 1999-12-21 Vrije Universiteit Brussels Immunoglobulins devoid of light chains
US6080777A (en) * 1992-01-31 2000-06-27 The Trustees Of Columbia University In The City Of New York Taxol as a radiation sensitizer
US20020006379A1 (en) * 1998-06-22 2002-01-17 Hansen Hans J. Production and use of novel peptide-based agents for use with bi-specific antibodies
US20030109682A1 (en) * 2001-09-07 2003-06-12 Daniel Santi Maytansines and maytansine conjugates
US20040126379A1 (en) * 2002-08-21 2004-07-01 Boehringer Ingelheim International Gmbh Compositions and methods for treating cancer using cytotoxic CD44 antibody immunoconjugates and chemotherapeutic agents
US20040241817A1 (en) * 2003-01-22 2004-12-02 Glycart Biotechnology Ag Fusion constructs and use of same to produce antibodies with increased Fc receptor binding affinity and effector function
US20050123549A1 (en) * 2003-07-21 2005-06-09 Immunogen Inc. CA6 antigen-specific cytotoxic conjugate and methods of using the same
US20050169933A1 (en) * 2003-10-10 2005-08-04 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
US20090169570A1 (en) * 2007-12-26 2009-07-02 Benjamin Daelken Methods and agents for improving targeting of cd138 expressing tumor cells
US20090175863A1 (en) * 2007-12-26 2009-07-09 Elmar Kraus Agents targeting cd138 and uses thereof
US20090181038A1 (en) * 2007-12-26 2009-07-16 Gregor Schulz Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
US20090232810A1 (en) * 2007-12-26 2009-09-17 Elmar Kraus Immunoconjugates targeting cd138 and uses thereof
US20100028346A1 (en) * 2008-06-16 2010-02-04 Immunogen Inc. Novel synergistic effects

Family Cites Families (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3896111A (en) 1973-02-20 1975-07-22 Research Corp Ansa macrolides
US4151042A (en) 1977-03-31 1979-04-24 Takeda Chemical Industries, Ltd. Method for producing maytansinol and its derivatives
US4169888A (en) 1977-10-17 1979-10-02 The Upjohn Company Composition of matter and process
US4137230A (en) 1977-11-14 1979-01-30 Takeda Chemical Industries, Ltd. Method for the production of maytansinoids
US4307016A (en) 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4265814A (en) 1978-03-24 1981-05-05 Takeda Chemical Industries Matansinol 3-n-hexadecanoate
JPS5562090A (en) 1978-10-27 1980-05-10 Takeda Chem Ind Ltd Novel maytansinoid compound and its preparation
JPS55164687A (en) 1979-06-11 1980-12-22 Takeda Chem Ind Ltd Novel maytansinoid compound and its preparation
US4256746A (en) 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
JPS5566585A (en) 1978-11-14 1980-05-20 Takeda Chem Ind Ltd Novel maytansinoid compound and its preparation
JPS55102583A (en) 1979-01-31 1980-08-05 Takeda Chem Ind Ltd 20-acyloxy-20-demethylmaytansinoid compound
JPS55162791A (en) 1979-06-05 1980-12-18 Takeda Chem Ind Ltd Antibiotic c-15003pnd and its preparation
JPS55164685A (en) 1979-06-08 1980-12-22 Takeda Chem Ind Ltd Novel maytansinoid compound and its preparation
JPS55164686A (en) 1979-06-11 1980-12-22 Takeda Chem Ind Ltd Novel maytansinoid compound and its preparation
US4309428A (en) 1979-07-30 1982-01-05 Takeda Chemical Industries, Ltd. Maytansinoids
JPS5645483A (en) 1979-09-19 1981-04-25 Takeda Chem Ind Ltd C-15003phm and its preparation
EP0028683A1 (en) 1979-09-21 1981-05-20 Takeda Chemical Industries, Ltd. Antibiotic C-15003 PHO and production thereof
JPS5645485A (en) 1979-09-21 1981-04-25 Takeda Chem Ind Ltd Production of c-15003pnd
WO1982001188A1 (en) 1980-10-08 1982-04-15 Takeda Chemical Industries Ltd 4,5-deoxymaytansinoide compounds and process for preparing same
US4313946A (en) 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4315929A (en) 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
JPS57192389A (en) 1981-05-20 1982-11-26 Takeda Chem Ind Ltd Novel maytansinoid
US4418064A (en) 1982-09-29 1983-11-29 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids: treflorine, trenudine, and N-methyltrenudone
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US5053394A (en) 1988-09-21 1991-10-01 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
ATE139258T1 (en) 1990-01-12 1996-06-15 Cell Genesys Inc GENERATION OF XENOGENE ANTIBODIES
AU648313B2 (en) 1990-04-25 1994-04-21 Pharmacia & Upjohn Company Novel CC-1065 analogs
DE69233482T2 (en) 1991-05-17 2006-01-12 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
US5998656A (en) 1991-09-23 1999-12-07 Florida State University C10 tricyclic taxanes
US5200534A (en) 1992-03-13 1993-04-06 University Of Florida Process for the preparation of taxol and 10-deacetyltaxol
ES2149768T3 (en) 1992-03-25 2000-11-16 Immunogen Inc CONJUGATES OF BINDING AGENTS OF CELLS DERIVED FROM CC-1065.
US5703247A (en) 1993-03-11 1997-12-30 Virginia Tech Intellectual Properties, Inc. 2-Debenzoyl-2-acyl taxol derivatives and methods for making same
US5475011A (en) 1993-03-26 1995-12-12 The Research Foundation Of State University Of New York Anti-tumor compounds, pharmaceutical compositions, methods for preparation thereof and for treatment
US6740734B1 (en) 1994-01-14 2004-05-25 Biovitrum Ab Bacterial receptor structures
SE9400088D0 (en) 1994-01-14 1994-01-14 Kabi Pharmacia Ab Bacterial receptor structures
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5763477A (en) 1994-07-22 1998-06-09 Dr. Reddy's Research Foundation Taxane derivatives from 14-β-hydroxy-10 deacetylbaccatin III
CA2214993A1 (en) 1995-03-10 1996-09-19 Qun Y. Zheng Cephalomannine epoxide, its analogues and a method for preparing the same
EP0822830B1 (en) 1995-04-27 2008-04-02 Amgen Fremont Inc. Human anti-IL-8 antibodies, derived from immunized xenomice
EP0823941A4 (en) 1995-04-28 2001-09-19 Abgenix Inc Human antibodies derived from immunized xenomice
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
CA2273194C (en) 1996-12-03 2011-02-01 Abgenix, Inc. Transgenic mammals having human ig loci including plural vh and vk regions and antibodies produced therefrom
TR199902553T2 (en) 1997-04-14 2000-03-21 Micromet Gesellschaft F�R Biomedizinische Forschung Mbh New methods and uses for the production of antigen receptors against the human body.
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
NZ517772A (en) 1999-11-24 2004-03-26 Immunogen Inc Cytotoxic agents comprising taxanes and their therapeutic use
US6333410B1 (en) 2000-08-18 2001-12-25 Immunogen, Inc. Process for the preparation and purification of thiol-containing maytansinoids
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
US20020193569A1 (en) * 2001-06-04 2002-12-19 Idec Pharmaceuticals Corporation Bispecific fusion protein and method of use for enhancing effector cell killing of target cells
US6716821B2 (en) 2001-12-21 2004-04-06 Immunogen Inc. Cytotoxic agents bearing a reactive polyethylene glycol moiety, cytotoxic conjugates comprising polyethylene glycol linking groups, and methods of making and using the same
US6534660B1 (en) 2002-04-05 2003-03-18 Immunogen, Inc. CC-1065 analog synthesis
US6756397B2 (en) 2002-04-05 2004-06-29 Immunogen, Inc. Prodrugs of CC-1065 analogs
CN100482277C (en) 2002-05-02 2009-04-29 惠氏控股公司 Calicheamicin derivative-carrier conjugates
US6596757B1 (en) 2002-05-14 2003-07-22 Immunogen Inc. Cytotoxic agents comprising polyethylene glycol-containing taxanes and their therapeutic use
US7390898B2 (en) 2002-08-02 2008-06-24 Immunogen Inc. Cytotoxic agents containing novel potent taxanes and their therapeutic use
CN102573916B (en) 2009-05-06 2017-05-17 生物测试股份公司 Uses of immunoconjugates targeting cd138

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444887A (en) * 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4761111A (en) * 1981-09-18 1988-08-02 Brown Andrew M Automobile lifting and towing equipment
US5530101A (en) * 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5585089A (en) * 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5208020A (en) * 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US5814318A (en) * 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5565332A (en) * 1991-09-23 1996-10-15 Medical Research Council Production of chimeric antibodies - a combinatorial approach
US6080777A (en) * 1992-01-31 2000-06-27 The Trustees Of Columbia University In The City Of New York Taxol as a radiation sensitizer
US6005079A (en) * 1992-08-21 1999-12-21 Vrije Universiteit Brussels Immunoglobulins devoid of light chains
US5639641A (en) * 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US20020006379A1 (en) * 1998-06-22 2002-01-17 Hansen Hans J. Production and use of novel peptide-based agents for use with bi-specific antibodies
US20030109682A1 (en) * 2001-09-07 2003-06-12 Daniel Santi Maytansines and maytansine conjugates
US20040126379A1 (en) * 2002-08-21 2004-07-01 Boehringer Ingelheim International Gmbh Compositions and methods for treating cancer using cytotoxic CD44 antibody immunoconjugates and chemotherapeutic agents
US20040241817A1 (en) * 2003-01-22 2004-12-02 Glycart Biotechnology Ag Fusion constructs and use of same to produce antibodies with increased Fc receptor binding affinity and effector function
US20050123549A1 (en) * 2003-07-21 2005-06-09 Immunogen Inc. CA6 antigen-specific cytotoxic conjugate and methods of using the same
US20050169933A1 (en) * 2003-10-10 2005-08-04 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
US20090169570A1 (en) * 2007-12-26 2009-07-02 Benjamin Daelken Methods and agents for improving targeting of cd138 expressing tumor cells
US20090175863A1 (en) * 2007-12-26 2009-07-09 Elmar Kraus Agents targeting cd138 and uses thereof
US20090181038A1 (en) * 2007-12-26 2009-07-16 Gregor Schulz Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
US20090232810A1 (en) * 2007-12-26 2009-09-17 Elmar Kraus Immunoconjugates targeting cd138 and uses thereof
US20100028346A1 (en) * 2008-06-16 2010-02-04 Immunogen Inc. Novel synergistic effects

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Chen et al, BioTechniques 49:513-515, 2010. *
Supiot et al, Cancer 94:1202-9, 2002 *
Vooijs et al, Cancer Immuno Immunother 42:319-328, 1996, abstract only *

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080280779A1 (en) * 2006-09-26 2008-11-13 Shaughnessy Jr John D Gene expression profiling based identification of genomic signatures of multiple myeloma and uses thereof
US9387261B2 (en) * 2007-12-26 2016-07-12 Biotest Ag Immunoconjugates targeting CD138 and uses thereof
KR101626416B1 (en) * 2007-12-26 2016-06-01 바이오테스트 아게 Agents targeting cd138 and uses thereof
US20090181038A1 (en) * 2007-12-26 2009-07-16 Gregor Schulz Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
US20090232810A1 (en) * 2007-12-26 2009-09-17 Elmar Kraus Immunoconjugates targeting cd138 and uses thereof
AU2008339913B2 (en) * 2007-12-26 2014-03-20 Biotest Ag Methods and agents for improving targeting of CD138 expressing tumor cells
KR20100100989A (en) * 2007-12-26 2010-09-15 바이오테스트 아게 Agents targeting cd138 and uses thereof
RU2547939C2 (en) * 2007-12-26 2015-04-10 Биотест Аг Anti-cd138 immunoconjugates and using them
JP2015038158A (en) * 2007-12-26 2015-02-26 バイオテスト・アクチエンゲゼルシヤフト Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
JP2011507933A (en) * 2007-12-26 2011-03-10 バイオテスト・アクチエンゲゼルシヤフト Methods for reducing cytotoxic side effects and improving efficacy of immune complexes
US20170081418A1 (en) * 2007-12-26 2017-03-23 Biotest Ag Immunoconjugates targeting cd138 and uses thereof
US9446146B2 (en) * 2007-12-26 2016-09-20 Biotest Ag Methods and agents for improving targeting of CD138 expressing tumor cells
RU2486203C2 (en) * 2007-12-26 2013-06-27 Биотест Аг Method for improving targeted action of cd138 expressing tumour cells and agents to this effect
AU2008339910B2 (en) * 2007-12-26 2014-01-09 Biotest Ag Agents targeting CD138 and uses thereof
AU2008339911B2 (en) * 2007-12-26 2014-02-27 Biotest Ag Immunoconjugates targeting CD138 and uses thereof
KR101654822B1 (en) * 2007-12-26 2016-09-06 바이오테스트 아게 Immunoconjugates targeting cd138 and uses thereof
US20090169570A1 (en) * 2007-12-26 2009-07-02 Benjamin Daelken Methods and agents for improving targeting of cd138 expressing tumor cells
EP2238169A1 (en) * 2007-12-26 2010-10-13 Biotest AG Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
US9011864B2 (en) * 2007-12-26 2015-04-21 Biotest Ag Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
TWI501778B (en) * 2007-12-26 2015-10-01 Biotest Ag Immunoconjugates targeting cd138 and uses thereof
US9221914B2 (en) * 2007-12-26 2015-12-29 Biotest Ag Agents targeting CD138 and uses thereof
KR20160028515A (en) * 2007-12-26 2016-03-11 바이오테스트 아게 Immunoconjugates targeting cd138 and uses thereof
US20090175863A1 (en) * 2007-12-26 2009-07-09 Elmar Kraus Agents targeting cd138 and uses thereof
EP3266469A2 (en) 2008-04-30 2018-01-10 ImmunoGen, Inc. Cross-linkers and their uses
US20110207627A1 (en) * 2008-08-13 2011-08-25 George Mason University Ex vivo therapeutics screening of living bone marrow cells for multiple myeloma
WO2010019227A1 (en) * 2008-08-13 2010-02-18 George Mason Intellectual Properties, Inc. Ex vivo therapeutic screening of living bone marrow cells for multiple myeloma
WO2010126551A1 (en) 2009-04-30 2010-11-04 Immunogen, Inc. Potent conjugates and hydrophilic linkers
US9289509B2 (en) 2009-05-06 2016-03-22 Biotest Ag Uses of immunoconjugates targeting CD138
US20110123554A1 (en) * 2009-05-06 2011-05-26 Frank Osterroth Uses of immunoconjugates targeting cd138
EP3480202A1 (en) 2009-06-03 2019-05-08 ImmunoGen, Inc. Conjugation methods
US10117932B2 (en) 2011-12-08 2018-11-06 Biotest Ag Uses of immunoconjugates targeting CD138
US11945868B2 (en) 2017-10-02 2024-04-02 Visterra, Inc. Antibody molecules to CD138 and uses thereof
WO2024000262A1 (en) * 2022-06-29 2024-01-04 京东方科技集团股份有限公司 Probe and kit for early diagnosis of diffuse large b-cell lymphoma

Also Published As

Publication number Publication date
US20150010585A1 (en) 2015-01-08
AU2004224925B2 (en) 2011-03-17
CA2486285A1 (en) 2006-02-28
AU2004224925C1 (en) 2011-07-21
CA2486285C (en) 2017-03-07
US20070183971A1 (en) 2007-08-09
US8840898B2 (en) 2014-09-23
AU2004224925A1 (en) 2006-03-16

Similar Documents

Publication Publication Date Title
US8840898B2 (en) Immunoconjugates targeting syndecan-1 expressing cells and use thereof
TWI524897B (en) Method of decreasing cytotoxic side-effects and improving efficacy of immunoconjugates
US9221914B2 (en) Agents targeting CD138 and uses thereof
US9387261B2 (en) Immunoconjugates targeting CD138 and uses thereof
US9446146B2 (en) Methods and agents for improving targeting of CD138 expressing tumor cells
AU2013203245A1 (en) Agents targeting CD138 and uses thereof
AU2013204492A1 (en) Immunoconjugates targeting CD138 and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIOTEST AG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:IMMUNOGEN, INC.;REEL/FRAME:015929/0785

Effective date: 20050404

Owner name: IMMUNOGEN, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GOLDMAKHER, VIKTOR S.;REEL/FRAME:015929/0730

Effective date: 20050404

AS Assignment

Owner name: BIOTEST AG (ONE-HALF INTEREST), GERMANY

Free format text: CORRECTIVE ASSIGNMENT TO CORRECT THE CONVEYANCE OF INTEREST FROM IMMUNOGEN, INC. TO BIOTEST AG PREVIOUSLY RECORDED ON REEL 015929 FRAME 0785. ASSIGNOR(S) HEREBY CONFIRMS THE IMMUNOGEN, INC., CONVEYS AN UNDIVIDED ONE-HALF OF THE RIGHT, TITLE AND INTEREST IN AND TO THIS APPLICATION TO BIOTEST AG.;ASSIGNOR:IMMUNOGEN, INC. (ONE-HALF INTEREST);REEL/FRAME:025694/0829

Effective date: 20050404

AS Assignment

Owner name: IMMUNOGEN, INC., MASSACHUSETTS

Free format text: CHANGE OF ADDRESS FOR IMMUNOGEN, INC;ASSIGNOR:IMMUNOGEN, INC.;REEL/FRAME:033705/0069

Effective date: 20080304

STCV Information on status: appeal procedure

Free format text: ON APPEAL -- AWAITING DECISION BY THE BOARD OF APPEALS

STCV Information on status: appeal procedure

Free format text: BOARD OF APPEALS DECISION RENDERED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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