EP1877083A2 - Antikörper zur behandlung von krebserkrankungen - Google Patents

Antikörper zur behandlung von krebserkrankungen

Info

Publication number
EP1877083A2
EP1877083A2 EP06750858A EP06750858A EP1877083A2 EP 1877083 A2 EP1877083 A2 EP 1877083A2 EP 06750858 A EP06750858 A EP 06750858A EP 06750858 A EP06750858 A EP 06750858A EP 1877083 A2 EP1877083 A2 EP 1877083A2
Authority
EP
European Patent Office
Prior art keywords
egfr
antibody
mmc
cells
binding molecule
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.)
Withdrawn
Application number
EP06750858A
Other languages
English (en)
French (fr)
Inventor
Le Sun
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.)
Welson Pharmaceuticals Inc
Original Assignee
Welson Pharmaceuticals Inc
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 Welson Pharmaceuticals Inc filed Critical Welson Pharmaceuticals Inc
Publication of EP1877083A2 publication Critical patent/EP1877083A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • 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
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies

Definitions

  • EGFR Epidermal growth factor receptor
  • Activation of the EGFR autocrine growth pathway in cancer cells can be attributable to several mechanisms, namely, EGFR overexpression, increased concentration of ligand(s), decreased phosphatase activity, decreased receptor turnover, and the presence of aberrant receptors.
  • Administration of blocking anti-EGFR antibodies only is not so therapeutic to those cancers independent to ligand binding.
  • Currently-used anticancer drugs are unable to recognize tumor cells from normal cells. The amount of an anticancer drug required to achieve a clinically effective level of cell kill often causes severe damage to actively propagating nonmalignant cells such as cells of the gastrointestinal tract and bone marrow, resulting in a variety of undesirable side effects.
  • the present invention features additional anti-EGFR immunoconjugates and methods of using the same for suppressing the growth of tumor cells and for treating cancers of epithelial origin.
  • the present invention features EGFR-binding molecules.
  • Each molecule comprises an antigen-binding fragment of an anti-EGFR monoclonal antibody and a cytotoxic agent.
  • an EGFR-binding molecule of the present invention is a monoclonal antibody LAl or LA22 conjugated with a cytotoxic agent.
  • the present invention also features humanized or engineered antibodies comprising an antigen-binding fragment of LAl or LA22 conjugated with mitomycin C, pingyangmycin, or other anti-cellular agents.
  • the present invention features other anti-EGFR antibodies conjugated with mitomycin C or pingyangmycin. These antibodies can be monoclonal antibodies, human antibodies, humanized or engineered antibodies, synthetic antibodies, or single chain antibodies.
  • the EGFR-binding molecules or antibodies of the present invention can be used to kill or inhibit the growth of cancer cells. These methods comprise contacting cancer cells with an EGFR-binding molecule or antibody of the present invention.
  • the EGFR-binding molecules or antibodies of the present invention can be used to treat cancers. These methods comprise administrating an effective amount of an EGFR-binding molecule or antibody of the present invention to a subject in need thereof. Cancers or cancer cells amenable to the present invention include those of epithelial origin.
  • the present invention also features pharmaceutical compositions comprising an EGFR-binding molecule or antibody of the present invention.
  • FIGURE 1 demonstrates the cytotoxic effects of anti-EGFR monoclonal antibodies conjugated with pingyangmycin or mitomycin C on human epidermoid carcinoma cells.
  • FIGURE 2 illustrates the dose-dependent cytotoxic effect of monoclonal antibody LA22 conjugated with pingyangmycin or mitomycin C on human epidermoid carcinoma cells.
  • FIGURE 4 shows the dose-dependent cytotoxic effect of monoclonal antibody
  • FIGURE 5 demonstrates the dose-dependent cytotoxic effect of monoclonal antibody LAl conjugated with pingyangmycin or mitomycin C on human lung cells.
  • FIGURE 6 illustrates the in vivo tumor toxicity effect of LA22-MMC on female Balb/c mice.
  • FIGURE 6A shows the treatment scheme of LA22-MMC immunoconjugate on A431 xenografts and the inhibitory effect of tumor growth in vivo.
  • FIGURE 6B summarizes the tumor size and percent of tumor inhibition of each of the treatment groups (control, MMC, and LA22-MMC at 0.0032 mg/kg and 0.08 mg/kg).
  • FIGURE 7 shows the biodistribution of LA22-MMC immunoconjugate on tumor-bearing mice after 125 I-radiolabeling and immunoscintigraphy.
  • FIGURE 8 compares the in vitro cytotoxic effect of LA22-MMC immunoconjugate on human epidermoid carcinoma cells with Erbitux -MMC and
  • Herceptin ® -MMC Herceptin ® -MMC.
  • the present invention features anti-EGFR monoclonal antibodies LAl or
  • LA22 conjugated with cytotoxic or anticellular agents Both LAl and LA22 are commercially available from Upstate USA, Inc., Charlottesville, VA. Monoclonal antibody LA22 is also described in U.S. Patent No. 5,459,061, which is incorporated herein by reference in its entirety.
  • Non-limiting examples of suitable cytotoxic or anti-cellular agents include chemotherapeutic agents, radioisotopes, and cytotoxins.
  • Specific examples of cytotoxic or anti-cellular agents include, but are not limited to, cytotoxic antibiotics (such as mitomycin C or pingyangmycin), hormones (such as steroids), anti-metabolites (such as cytosine arabinoside, fluorouracil, methotrexate or aminopterin), anthracycline, vinca alkaloids, demecolcine, etoposide, mithramycin, alkylating agents (such as chlorambucil or melphalan), doxorubicin, daunomycin, methotrexate, vinblastine, neocarzinostatin, macromycin, trenimon, ⁇ -amanitin, ricin, ricin A-chain, daunorubicin, taxol, ethiduim bromide, mitomycin, etoposide, ten
  • LAl or LA22 monoclonal antibodies can also be conjugated to an anti-cancer pro-drug activating enzyme capable of converting a pro-drug to its active form. See, for example, U.S. Patent No. 4,975,287, which is incorporated herein by reference in its entirety.
  • Linking or coupling one or more cytotoxic moieties to an antibody can be achieved by a variety of mechanisms, for example, covalent binding, affinity binding, intercalation, coordinate binding and complexation.
  • Preferred binding methods are those involving covalent binding, such as using chemical cross-linkers, natural peptides or disulfide bonds.
  • the covalent binding can be achieved either by direct condensation of existing side chains or by the incorporation of external bridging molecules.
  • Many bivalent or polyvalent agents are useful in coupling protein molecules to other proteins, peptides or amine functions.
  • Examples of coupling agents are carbodiimides, diisocyanates, glutaraldehyde, diazobenzenes, and hexamethylene diamines. This list is not intended to be exhaustive of the various coupling agents known in the art but, rather, is exemplary of the more common coupling agents that may be used.
  • an antibody is first derivatized, following by attachment of the cytotoxic component to the derivatized product.
  • the term "derivatize” refers to the chemical modification of the antibody substrate with a suitable cross-linking agent.
  • cross-linking agents include the disulfide-bond containing linkers SPDP (N-succinimidyl-3-(2-pyr ⁇ dyldithio)propionate) and SMPT (4-succinimidyl-oxycarbonyl- ⁇ -methyl- ⁇ (2-pyridyldithio)toluene).
  • Biologically-releasable bonds can also be employed in constructing a clinically-active immunoconjugate, such that the cytotoxic moiety is capable of being released from the antibody once it has entered the target cell.
  • Numerous types of linking constructs are known, including simply direct disulfide bond formation between sulfhydryl groups contained on amino acids such as cysteine, or otherwise introduced into respective protein structures, and disulfide linkages using available or designed linker moieties.
  • Numerous types of disulfide-bond containing linkers are known, which can successfully be employed to conjugate cytotoxic moieties to antibodies.
  • the present invention also features anti-EGFR molecules comprising the antigen-binding fragments of LA22 or LAl monoclonal antibodies conjugated with cytotoxic or anticellular agents. These antigen-binding fragments can include or consist of the scFv, diabody, minibody, Fab, F(ab') 2 , or Fv fragments of LA22 or LAl 1 monoclonal antibodies.
  • the anti-EGFR molecules of the present invention are humanized or engineered monoclonal antibodies prepared from monoclonal antibodies LA22 or LAl. Humanized or engineered antibodies are particularly desirable for therapeutic treatment of human subjects.
  • Humanized or engineered forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, or antigen-binding fragments thereof (such as scFv, diabody, minibody, Fab, or F(ab') 2 ), which contain minimal sequence derived from non-human immunoglobulin.
  • Humanized or engineered antibodies are derived from human immunoglobulins in which the residues forming the complementary determining regions (CDRs) are replaced by the residues from CDRs of a non-human antibody, such as LA22 or LAl.
  • CDRs complementary determining regions
  • Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized or engineered antibodies which recognize a selected epitope can be generated using a technique referred to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a murine antibody
  • the methods, as described in U.S. Patent Nos. 6,639,055 and 6,794,132, both of which are incorporated herein by reference, can also be used to prepare humanized or engineered antibodies.
  • the present invention features other anti-EGFR antibodies conjugated with mitomycin C, pingyangmycin, or other cytotoxic antibiotics.
  • Antibodies suitable for this purpose include, but are not limited to, polyclonal, monoclonal, monospecific, poly-specific, non-specific, humanized or engineered, human, single-chain, chimeric, synthetic, recombinant, hybrid, mutated, grafted, or in vitro generated antibodies.
  • an antibody of the present invention binds to an epitope in the extracellular domain of EGFR and has an EGFR-binding affinity of at least 10 "5 M “1 , 10 "6 M “1 , 10 "7 M “1 , 10 “ 8 M “1 , 10 “9 M “1 , 10 '10 M '1 , or stronger.
  • the scFv, diabody, minibody, Fab, F(ab') 2 , or Fv fragments of these antibodies can also be used for the conjugation of mitomycin C, pingyangmycin, or other cytotoxic antibiotics.
  • a pharmaceutical composition of the present invention typically includes an EGFR-binding molecule of the present invention (e.g., mitomycin C- or pingyangmycin-conjugated LA22 or LAl) and a pharmacologically-acceptable carrier.
  • EGFR-binding molecule of the present invention e.g., mitomycin C- or pingyangmycin-conjugated LA22 or LAl
  • pharmacologically-acceptable carrier includes any and all solvents, dispersion media, coatings, anti-bacterial and anti-fungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically-active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the vectors or cells of the present invention, its use in therapeutic or prophylactic compositions is contemplated. Supplementary active ingredients also can be incorporated into the compositions.
  • Administration of a pharmaceutical composition of the present invention can be by way of any common route so long as the target tissue is available via that route. This includes oral, nasal, buccal, rectal, vaginal or topical. Alternatively, administration may be by orthotopic, intradermal, subcutaneous, intramuscular, intraperitoneal, intratumoral, circumferentially, catheterization, or intravenous injection.
  • a pharmaceutical composition can also be administered parenterally or intraperitoneally.
  • Solutions of the active compounds as free base or pharmacologically- acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form is sterile and fluid to the extent that easy syringability exists. It is preferably also stable under the conditions of manufacture and storage and preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, or vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various anti-bacterial or anti-fungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal or the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating an EGFR-binding molecule of the present invention in the required amount in an appropriate solvent with various other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle, which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying or freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • an EGFR-binding molecule of the present invention can be incorporated with excipients and used in the form of non-ingestible mouthwashes and dentifrices.
  • a mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (Dobell's Solution).
  • an EGFR-binding molecule may be incorporated into an anti-septic wash containing sodium borate, glycerin and potassium bicarbonate.
  • An EGFR-binding molecule may also be dispersed in dentifrices, including: gels, pastes, powders, or slurries.
  • An EGFR- binding molecule may be added in a therapeutically or prophylactically effective amount to a paste dentifrice that may include water, binders, abrasives, flavoring agents, foaming agents, or humectants.
  • compositions of the present invention may be formulated in a neutral or salt form.
  • Pharmaceutically-acceptable salts include the acid addition salts formed with the free amino groups of the protein or formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • compositions or solutions Upon formulation, compositions or solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically or prophylactically effective.
  • the formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like.
  • parenteral administration in an aqueous solution for example, the solution can be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous, and intraperitoneal administration.
  • sterile aqueous media which can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage could be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see, for example, REMINGTON'S PHARMACEUTICAL SCIENCES (15th Edition), pages 1035- 1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated.
  • the EGFR-binding molecules of the present invention can be used to kill or inhibit the growth of cancer cells.
  • the EGFR-binding molecules of the present invention can also be used to treat cancers in patients or animals.
  • cancers that are amenable to the present invention include, but are not limited to, cancers of epithelial origin, such as glioblastoma, cancer of the lung, breast, head and neck, and bladder, or other epidermoid cancers.
  • the EGFR-binding molecule of the present invention.
  • the EGFR-binding molecule would be provided in an amount effective to kill or inhibit proliferation of the tumor cells.
  • the EGFR-binding molecules can also be combined with other traditional therapies to treat cancers.
  • Agents or factors suitable for use in a combined therapy are any chemical compound or treatment method that induces DNA damage when applied to a cell. Such agents and factors include radiation and waves that induce DNA damage such as, ⁇ - irradiation, X-rays, UV-irradiation, microwaves, electronic emissions, and the like.
  • Chemotherapeutic agents contemplated to be of use include, e.g., adriamycin, 5- fluorouracil (5FU), etoposide (VP- 16), camptothecin, actinomycin-D, mitomycin C, cisplatin (CDDP), and in some cases, hydrogen peroxide.
  • LA22 hybridoma (ATCC No. HB 10342) cells at 2x10 4 cell density were injected to Balb/c nude mice. Monoclonal antibodies from ascites was purified using Protein G affinity agarose beads (Upstate, Lake Placid, NY). Briefly, ascites fluid was diluted 1 : 1 with PBS (pH 7.4) and centrifuged at 14,00Og for 4 min. Supernatant was carefully collected and loaded onto a Protein G column (containing 2 mL Protein G agarose) and a 10 mL PBS was then applied to the column to wash unbound components.
  • Bound mAb LA22 was eluted with elution buffer (50 mM glycine-HCl, pH 2.7) and transferred into PBS (pH 7.4) by Desalting Column (Pierce, Rockford, IL). Aliquots were analyzed for purity with SDS- PAGE. Concentration of mAb LA22 was determined to be at 1 mg/ml of mouse IgG, which was equivalent to an O.D. (optical density) reading of 1.40. [0050] The A431 human epidermoid carcinoma cells, which express large quantities of EGFR, and A549 human lung adenocarcinoma cells were used for in vitro cytotoxicity assays.
  • DMEM Dulbecco's Modified Eagle medium
  • FBS fetal bovine serum
  • MMC (10 mg in 5 mL PBS) was reacted with 0.9 mL of SPDP (Pierce, Rockford, IL) stock (10 mg in 1 mL DMF) at 4°C for 6 h. The resulting solution was stored in small aliquots at -80°C until further use.
  • mAb LA22 5.5 mg in 3 mL of PBS
  • mAb LAl was reduced by adding 46 ⁇ L of 2-iminothiolane (4.4 mg dissolved in 1 mL DMF) for 1 h under nitrogen. The reduced antibody was purified by size exclusion chromatography using a desalting column (Pierce, Rockford, IL), that was pre-equilibrated with PBS.
  • LA22-MMC or LA22-PMC
  • FIGURE 2 illustrates that anti-EGFR mAb LA22-PYM or LA22- MMC induced the death of A431 human cancer cells in a dose-dependent manner.
  • Another LA22-MMC cytotoxicity assay was performed with additional controls.
  • the A431 cells, as well as human lung adenocarcinoma A549 cells were plated (2x10 3 cells/well, 100 ⁇ L/well) in 96-well cell culture plates (Corning, Corning, NY) in the presence of DMEM medium and 10% FBS for 4 h at 37 °C, 5% CO 2 .
  • MMC, naked mAb LA22 or LA22-MMC immunoconjugate in serum-free DMEM medium (100 ⁇ L) were added onto the individual wells and incubated for 4 days at 37°C and 5% CO 2 . The supernatants were discarded first. For dead cells control, 100 ⁇ L of cold 95% ethanol was added into the well for 1 min then discarded.
  • MTT solutions (USB, Cleveland, OH; 10% in DMEM medium) were added (100 ⁇ L/well) and incubated for 4 h at 37°C. The supernatants were discarded and DMSO was added (100 ⁇ L/well) to dissolve the precipitates. Optical density was read at 570 nm using a microplate reader (Bio-Rad, Hercules, CA). [0056] , The above-mentioned inhibition of A431/A549 in vitro cell growth was used as a measure for drug activity in vitro and was determined by exposing cultured cells (2x10 3 cells/well) for 4 days at 37°C with MMC, LA22-MMC or LA22.
  • the tested MMC concentrations ranged from 0.03 ⁇ g/mL to 30 ⁇ g/mL.
  • the dosage of the LA22-MMC conjugate for cytotoxicity was calculated according to the total amount of MMC added in chemical coupling reaction. Because the efficiency of chemical coupling reactions was not 100%, theoretically the real MMC concentration linked to LA22 was lower than indicated concentration. The dosage of LA22 was the same as LA22-MMC.
  • Non-specific mouse IgG (as a negative control) was labeled with MMC and added into A431 cells to evaluate its cytotoxicity on cell growth in vitro. The result showed that non-specific mouse IgG-MMC immunoconjugates did not kill cultured cells as efficiently as LA22-MMC, which proved that the cytotoxicity of LA22-MMC can be mediated via LA22-EGFR binding (data not included).
  • A549 cell line (ATCC No. CCL-185).
  • A549 cells were seeded at 2,000 cells/well of 96-well plate in DMEM-10% FBS.
  • various amounts of LAl, LAl-MMC, or MMC-SPDP were added to the cell cultures.
  • the cell numbers in each well were determined by MTT assay. The numbers were expressed as the percentage of the cell death (FIGURE 5).
  • LAl-MMC induced the death of A549 human lung cancer cells in a dose-dependent manner. No significant cell death was observed with mAb LAl alone. Conjugation with mitomycin C significantly increased the cell killing potency of LAl.
  • mice treated with LA22-MMC (0.08 mg/kg) inhibited tumor growth in vivo when compared with control untreated mice.
  • MMC (0.08 mg/kg) injection was potent to inhibit tumor growth, the efficiency was much lower than LA22-MMC (0.08 mg/kg), which suggested the great advantage of LA22-mediated specific tumor targeting.
  • Low dosage administration (0.0032 mg/kg) of LA22-MMC was not potent as the high-dosage of LA22-MMC administration (0.08 mg/kg), which suggested the dose-dependent effect of LA22-MMC immunoconjugate.
  • mice were sacrificed and the tumors were weighed (FIGURE 6B).
  • the nude mice- bearing A431/A549 xenografts were injected intravenously with 17.6 MBq ( ⁇ 20 ⁇ g, 0.3 mL) of 125 I-LA22-MMC, and placed prone on a two-head ⁇ -camera (E.CAM, Siemens, Kunststoff, Germany) equipped with a parallel-hole, low-energy, high-resolution collimator.
  • the posterior images were acquired at different time points post-injection for the visual observation of in vivo distribution of LA22-MMC, and stored digitally in a 128 x 128 matrix.
  • the acquisition count limits were set at 1000 K.
  • LA22-MMC first distributed in the circulation system and blood-rich tissues such as liver or heart (4h imaging); gradually the labeled immunoconjugates enriched on the tumor sites (1 day to 4 days). After 4 days, LA22-MMC mainly enriched on the tumor sites.
  • Erbitux ® and Herceptin ® two commercially-available mAb drugs for cancer therapy, target EGFR and HER2, respectively. Both EGFR and HER2 belong to the ErbB receptor family (Mendelsohn, J. et ah, Oncogene, 19:6550-7565, 2000).
  • Erbitux ® and Herceptin ® an in vitro cytotoxicity assay using the MTT method was conducted. The conjugation of MMC with Erbitux ® (ImClone, New York, NY) or Herceptin ® (Genentech, San Francisco, CA) or mouse IgG was done as described above.
  • Erbitux ® and Herceptin ® were labeled with MMC and evaluated for their cytotoxicities.
  • A431 (FIGURE 8A) or A549 (FIGURE 8B) cells were seeded into 96-well cell culture plates. Naked antibodies or immunoconjugates were added to wells and incubated for 4 days at 37°C and 5% CO 2 . Viable cells were measured using the MTT assay. Using similar antibody concentrations, LA22-MMC prodrug killed A431 and A549 cells more efficiently than Erbitux ® -MMC or Herceptin ® -MMC (see FIGURES 8A & 8B) .

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Cell Biology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
EP06750858A 2005-04-27 2006-04-20 Antikörper zur behandlung von krebserkrankungen Withdrawn EP1877083A2 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US67509405P 2005-04-27 2005-04-27
PCT/US2006/014941 WO2006116001A2 (en) 2005-04-27 2006-04-20 Antibodies for the treatment of cancers

Publications (1)

Publication Number Publication Date
EP1877083A2 true EP1877083A2 (de) 2008-01-16

Family

ID=37215266

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06750858A Withdrawn EP1877083A2 (de) 2005-04-27 2006-04-20 Antikörper zur behandlung von krebserkrankungen

Country Status (3)

Country Link
EP (1) EP1877083A2 (de)
CN (1) CN101277716A (de)
WO (1) WO2006116001A2 (de)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2009008909A (es) 2007-03-01 2009-08-28 Symphogen As Composiciones de anticuerpo recombinante anti-receptor de factor de crecimiento epidermico.
WO2010022736A2 (en) 2008-08-29 2010-03-04 Symphogen A/S Recombinant anti-epidermal growth factor receptor antibody compositions
CN104059148B (zh) * 2013-03-18 2016-09-07 回而生医药科技(北京)有限公司 人源化抗人表皮生长因子受体抗体及其应用
US10611833B2 (en) 2014-03-07 2020-04-07 Welson Pharmaceuticals, Inc. Humanized anti-human epidermal growth factor receptor antibody and application thereof
CN112176013B (zh) * 2019-07-02 2022-08-12 华东理工大学 一种提高平阳霉素生物合成的效率的方法

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2006116001A2 *

Also Published As

Publication number Publication date
WO2006116001A3 (en) 2006-12-21
CN101277716A (zh) 2008-10-01
WO2006116001A2 (en) 2006-11-02

Similar Documents

Publication Publication Date Title
US8586049B2 (en) Antibody-drug conjugates
CN111712520A (zh) 磷脂酰肌醇聚糖3抗体及其偶联物
US20210017295A1 (en) Bispecific binding agents and uses thereof
WO2017196764A1 (en) Antibody-drug conjugate of an anti-glypican-3 antibody and a tubulysin analog, preparation and uses
JP2023036874A (ja) イムノコンジュゲートにおけるメチオニン酸化を防止する方法
WO2006116001A2 (en) Antibodies for the treatment of cancers
US20100047164A1 (en) Anti-Tumor Antibodies
EP4129335A1 (de) Anti-c-met-antikörper-wirkstoff-konjugat und anwendungen davon
KR20230065935A (ko) 치료적 접합체
JP2020532523A (ja) 抗egfr抗体薬物コンジュゲート(adc)及びその使用
Shao et al. Inhibition of human tumor xenograft growth in nude mice by a conjugate of monoclonal antibody LA22 to epidermal growth factor receptor with anti-tumor antibiotics mitomycin C
US20180186891A1 (en) Anti-fibroblast activation protein (fap) antibodies for treatment and diagnosis
JP7130243B2 (ja) 二重特異性抗体を用いた抗体薬物複合体プラットホーム
JP2020532543A (ja) 抗egfr抗体薬物コンジュゲート(adc)及びその使用
WO2023048231A1 (ja) 腫瘍細胞を死滅させるための医薬
WO2023048233A1 (ja) 腫瘍細胞を死滅させるための医薬
JP2024519585A (ja) 抗p-カドヘリン抗体を含む抗体コンジュゲートおよびその使用
CA3070610A1 (en) Methods and compositions for targeting disease related cell surface receptors using radiolabeled and/or cytotoxin labelled antibodies
KR20130138608A (ko) 위치 특이적 항체-싸이토톡신 결합체 및 이의 용도
Chauhan et al. Journal Homepage:-www. journalijar. com
NZ615308B2 (en) Antibody-drug conjugates

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20071122

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

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

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20081101