US20150202287A1 - Combination therapies comprising anti-erbb3 agents - Google Patents

Combination therapies comprising anti-erbb3 agents Download PDF

Info

Publication number
US20150202287A1
US20150202287A1 US14/424,344 US201314424344A US2015202287A1 US 20150202287 A1 US20150202287 A1 US 20150202287A1 US 201314424344 A US201314424344 A US 201314424344A US 2015202287 A1 US2015202287 A1 US 2015202287A1
Authority
US
United States
Prior art keywords
inhibitor
trastuzumab
erbb2
lapatinib
cells
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
US14/424,344
Inventor
Bo Zhang
Charlotte McDonagh
Alexandra Huhalov
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.)
Merrimack Pharmaceuticals Inc
Original Assignee
Merrimack 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 Merrimack Pharmaceuticals Inc filed Critical Merrimack Pharmaceuticals Inc
Priority to US14/424,344 priority Critical patent/US20150202287A1/en
Publication of US20150202287A1 publication Critical patent/US20150202287A1/en
Assigned to MERRIMACK PHARMACEUTICALS, INC. reassignment MERRIMACK PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MCDONAGH, CHARLOTTE, ZHANG, BO, HUHALOV, ALEXANDRA
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/567Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in position 17 alpha, e.g. mestranol, norethandrolone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • A61K2039/507Comprising a combination of two or more separate antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • Estrogen receptors mediate intracellular signaling that can increase the frequency of cell division and drive tumor growth.
  • anti-endocrine therapies such as tamoxifen, fulvestrant, and letrozole have demonstrated significant efficacy in treating ER+ breast cancer patients, intrinsic or acquired resistance to such therapies has limited their success.
  • ErbB2 can function as a homodimer or as a heterodimer with related epidermal growth factor-type receptors such as ErbB3.
  • the ErbB2/ErbB3 heterodimer is the most potent ErbB receptor pairing with respect to strength of interaction, impact on receptor tyrosine phosphorylation, and effects on downstream intracellular signaling through mitogen activated protein kinase and phosphoinositide-3 kinase pathways.
  • Heregulin is the primary ligand for ErbB3, and activates signaling by ErbB2/ErbB3 heterodimers. Such signaling is believed to drive proliferation of cancer cells. Current ErbB2-targeted therapies do not effectively inhibit heregulin activated signaling.
  • MM-111 is a bispecific anti-ErbB2/anti-ErbB3 antibody that abrogates heregulin binding to ErbB2/ErbB3 and inhibits heregulin activation of ErbB2/ErbB3 without significantly affecting ErbB2 biological activity.
  • MM-111 inhibits ErbB3 phosphorylation, cell cycle progression, and tumor growth.
  • the co-administration of pluralities of anti-cancer drugs often provides better treatment outcomes than monotherapy.
  • Such outcomes can be subadditive, additive, or superadditive. That is to say that the combined effects of two anti-cancer drugs, each of which provides a quantifiable degree of benefit, can be less than, equal to, or greater than the sum of the benefits of each drug.
  • two drug, each of which when used alone to treat a lethal cancer provides an average one year extension of progression free survival could together provide a ⁇ 24 month extension (e.g., an 18 month extension), about a 24 month extension, or a >24 month extension (e.g., a 30 month extension) of progression free survival.
  • combination therapies for cancer treatment provide significantly subadditive outcomes. Outcomes that are near additive, additive, or superadditive are most desirable, but only occur rarely.
  • many drugs are known to alter the bioavailability, or otherwise affect the safety profile of other drugs when both drugs are co-administered. As new drugs are first used in combination therapies, unforeseen, hazardous drug-drug interactions may be observed that result in drug-drug interaction-mediated toxicity in the patient.
  • compositions effective for the inhibition of ErbB3 activation and also effective for the inhibition of estrogen receptor activation are also provided.
  • methods and compositions effective for the inhibition of ErbB3 activation and also effective for the inhibition of ErB1 and/or ErbB2 activation are useful for the treatment of tumors, e.g., malignant tumors, as well as for the treatment of other cancers.
  • a method of treating a subject with a malignant tumor where the tumor is an ErbB2 expressing or ErbB2 over-expressing tumor (e.g., HER2 +++ or HER2 ++ tumors) and the tumor may be a melanoma, clear cell sarcoma, head and neck, endometrial, prostate, breast, ovarian, esophageal, gastric, gastro-esophageal, colon, colorectal, lung, bladder, pancreatic, salivary gland, liver, skin, brain or renal tumor.
  • the tumor is an ErbB2 expressing or ErbB2 over-expressing tumor (e.g., HER2 +++ or HER2 ++ tumors) and the tumor may be a melanoma, clear cell sarcoma, head and neck, endometrial, prostate, breast, ovarian, esophageal, gastric, gastro-esophageal, colon, colorectal, lung, bladder, pancreatic, salivary gland,
  • the method comprises co-administering to the subject an effective amount an agent selected from i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) a MEK inhibitor, (e.g., selumetinib, trametinib, PD0325901, UO126), iv) a PI3 kinase inhibitor (e.g., BEZ235, BKM120, GDC0941), v) an AKT inhibitor (e.g., MK-2206, triciribine), vi) an mTOR inhibitor (e.g., BEZ235, AZD8055, everolimus, temsirolimus, sirolimus, ridaforolimus), vii) trastuzumab, viii) T-DM1, ix) capecitabine, x) cisplatin, and xi) MM-151; and combinations thereof, in combination with an effective amount of an anti-
  • the combination of the bispecific anti-ErbB2/anti-ErbB3 antibody and either the effective amount of an anti-estrogen agent or the effective amount of the receptor tyrosine kinase inhibitor, and optionally the effective amount of trastuzumab or T-DM1 is characterized as follows: when a first tissue culture medium is prepared comprising the bispecific anti-ErbB2/anti-ErbB3 antibody (e.g., the antibody comprising the amino acid sequence set forth in SEQ ID NO:1) at a first concentration and either the anti-estrogen agent at a second concentration or the receptor tyrosine kinase inhibitor (e.g., lapatinib) at a third concentration (wherein each concentration is the same or different as each other concentration), and the medium is contacted with cancer cells of a cell line in a cell culture, cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or the percentage of cells in the culture that are a
  • cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or the percentage of cells in the culture that are apoptotic is increased to a greater degree than cell growth, or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or percentage of cells in the culture that are apoptotic is increased, to a lesser degree when cancer cells of the cell line in a cell culture are contacted with each of a second medium that is essentially the same as the first medium except that it does not comprise a bispecific anti-ErbB2/anti-ErbB3 antibody, and a third medium that is essentially the same as the first medium except that it does not comprise any anti-estrogen agent and it does not comprise any receptor tyrosine kinase inhibitor.
  • all effective amounts are either mouse effective amounts or human effective amounts.
  • all effective amounts are mouse effective amounts and the combination of the bispecific anti-ErbB2/anti-ErbB3 antibody (optionally the antibody comprising the amino acid sequence set forth in SEQ ID NO:1) and either the effective amount of an anti-estrogen agent or the effective amount of the receptor tyrosine kinase inhibitor, is characterized as follows: when co-administered to BT474-M3 xenograft tumor bearing mice with a tumor of a measured volume, the combination is more effective at inhibiting tumor volume increase after 32 days of co-administration than is the mouse effective amount of the bispecific anti-ErbB2/anti-ErbB3 antibody administration without the co-administration of either the effective amount of an anti-estrogen agent or the effective amount of the receptor tyrosine kinase inhibitor.
  • a mouse effective amount of trastuzumab or T-DM1 is co-administered with the bispecific anti-
  • a bispecific anti-ErbB2/anti-ErbB3 antibody (optionally the antibody comprising SEQ ID NO:1) is provided for use in combination therapy of a cancer (optionally a melanoma, clear cell sarcoma, head and neck, endometrial, esophageal, gastro-esophageal junction, prostate, breast, ovarian, gastric, colon, colorectal, lung, bladder, pancreatic, salivary gland, liver, skin, brain or renal tumor), where the combination therapy comprises concomitant use of an effective amount an agent selected from i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor, vi) an mTOR inhibitor, vii) trastuzumab, viii) T-DM1, ix) capecitabine, x) cisplatin
  • an aqueous solution comprising a bispecific anti-ErbB2/anti-ErbB3 antibody (optionally an antibody comprising the amino acid sequence set forth in SEQ ID NO:1) at a first concentration and an agent selected from i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) an effective amount of a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor, vi) an mTOR inhibitor, vii) trastuzumab, viii) T-DM1, ix) capecitabine, x) cisplatin, and xi) MM-151; and combinations thereof, at a second concentration.
  • an anti-estrogen agent ii) a receptor tyrosine kinase inhibitor, iii) an effective amount of a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor,
  • a first tissue culture medium comprising the bispecific anti-ErbB2/anti-ErbB3 antibody at the first concentration and the agent at the second concentration, and the medium is contacted with cancer cells of a cell line in a cell culture, cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or percentage of cells in the culture that are apoptotic is increased.
  • cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or the percentage of cells in the culture that are apoptotic is increased to a lesser degree when cells of the cell line in a cell culture are contacted with a second tissue culture medium that is essentially the same as the first medium of except that it does not comprise the agent(s).
  • cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or the percentage of cells in the culture that are apoptotic is increased to a lesser degree when cells of the cell line in a cell culture are contacted with a third tissue culture medium that is essentially the same as the first medium of except that it does not comprise any bispecific anti-ErbB2/anti-ErbB3 antibody.
  • the aqueous solution is blood plasma in a subject, and the subject does not experience a toxicity that is sufficiently harmful to require a change in a therapy being administered to the subject, which toxicity is mediated by a drug-drug interaction in the subject between the bispecific anti-ErbB2/anti-ErbB3 antibody and the anti-estrogen agent or the receptor tyrosine kinase inhibitor.
  • the aqueous solution further comprises trastuzumab or T-DM1 at a third concentration.
  • the method, combination therapy, or aqueous solution does not comprise an aromatase inhibitor or an estrogen receptor antagonist.
  • the method, combination therapy, or aqueous solution comprises nab-paclitaxel.
  • the anti-estrogen agent may be an estrogen receptor antagonist (e.g., fulvestrant or tamoxifen) or an aromatase inhibitor (e.g., wherein the aromatase inhibitor is letrozole, exemestane, anastrozole, aminoglutethimide, testolactone, vorozole, formestane, or fadrozole.
  • the aromatase inhibitor is letrozole.
  • the receptor tyrosine kinase inhibitor is erlotinib, afatinib, dasatinib, gefitinib, imatinib, pazopinib, lapatinib, sunitinib, nilotinib or sorafenib.
  • the receptor tyrosine kinase inhibitor is lapatinib.
  • the bispecific anti ErbB2/anti-ErbB3 antibody is the A5-HSA-ML3.9, ML3.9-HSA-A5, A5-HSA-B1D2, B1D2-HSA-A5, B12-HSA-B1D2, B1D2-HSA-B12, A5-HSA-F5B6H2, F5B6H2-HSA-A5, H3-HSA-F5B6H2, F5B6H2-HSA-H3, F4-HSA-F5B6H2, F5B6H2-HSA-F4, B1D2-HSA-H3, H3-HSA-B1D2, or the antibody comprising the amino acid sequence set forth in SEQ ID NO:1.
  • Each embodiment and aspect thereof above may also further comprise use of capecitabine and/or cisplatin.
  • one or more of a)-i) that follow may optionally apply: a) the cell line is BT474-M3; b) the culture is a spheroid culture, c) paclitaxel or another taxane or another chemotherapeutic drug is co-administered, optionally in accordance with the manufacturer's directions, d) the agent i)-xi) is administered in accordance with the manufacturer's directions, e) the trastuzumab or T-DM1 is administered in accordance with the manufacturer's directions, f) the co-administration of the bispecific anti-ErbB2/anti-ErbB3 antibody with the agent g)-vi) produces an about additive or a superadditive effect, h) the bispecific anti-ErbB2/anti-ErbB3 antibody is the antibody comprising SEQ ID NO:1 and is administered in accordance with any of the regimens (e.g., modes, dosages, dosing intervals, loading and
  • FIG. 1 is a graph showing that the combination of MM-111 and tamoxifen inhibits tumor growth in vivo better than either MM-111 or tamoxifen does alone.
  • the x-axis shows time post tumor implant in days and the y-axis shows tumor volume in mm 3 . Mice were treated with inhibitors beginning on day 7 post BT474-M3 cell implant.
  • FIG. 2 is seven graphs showing that MM-111 combines positively with anti-estrogen drugs in inhibiting estrogen-stimulated spheroid growth in vitro.
  • FIG. 2 a shows the effect of MM-111, tamoxifen (4-hydroxytamoxifen or 4OHT), or MM-111 and tamoxifen on in vitro spheroid growth.
  • FIG. 2 b shows the effect of trastuzumab, tamoxifen, or trastuzumab and tamoxifen.
  • FIG. 2 c shows the effect of MM-111, fulvestrant (FVT), or MM-111 and fulvestrant.
  • FIG. 2 d shows the effect of trastuzumab, fulvestrant, or trastuzumab and fulvestrant.
  • FIG. 2 e shows the effect of MM-111, trastuzumab, or MM-111 and trastuzumab.
  • FIG. 2 f shows the effect of MM-111, trastuzumab, and tamoxifen combined compared to that of any of the double combinations.
  • FIG. 2 g shows the effect of MM-111, trastuzumab, and fulvestrant combined compared to that of any of the double combinations.
  • the x-axes are a log scale of each drug concentration for each experimental condition in nM and the y axis is spheroid size as % of control spheroid size.
  • FIG. 3 is seven graphs showing that MM-111 combines positively with anti-estrogen drugs in inhibiting heregulin (HRG)-stimulated spheroid growth in vitro.
  • FIG. 3 a shows the effect of MM-111, tamoxifen (4-hydroxytamoxifen or 4OHT), or MM-111 and tamoxifen.
  • FIG. 3 b shows the effect of trastuzumab, tamoxifen, or trastuzumab and tamoxifen.
  • FIG. 3 c shows the effect of MM-111, fulvestrant (FVT), or MM-111 and fulvestrant.
  • FIG. 3 d shows the effect of trastuzumab, fulvestrant, or trastuzumab and fulvestrant.
  • FIG. 3 e shows the effect of MM-111, trastuzumab, or MM-111 and trastuzumab.
  • FIG. 3 f shows the effect of MM-111, trastuzumab, and tamoxifen combined compared to that of any of the double combinations.
  • FIG. 3 g shows the effect of MM-111, trastuzumab, and fulvestrant combined compared to that of any of the double combinations.
  • the x-axes are a log scale of each drug concentration for each experimental condition in nM and the y axis is spheroid size as % of control spheroid size.
  • FIG. 4 is seven graphs showing that MM-111 combines positively with anti-estrogen drugs in inhibiting dual ligand (estrogen and heregulin)-stimulated spheroid growth in vitro.
  • FIG. 4 a shows the effect of MM-111, tamoxifen, or MM-111 and tamoxifen.
  • FIG. 4 b shows the effect of trastuzumab, tamoxifen, or trastuzumab and tamoxifen.
  • FIG. 4 c shows the effect of MM-111, fulvestrant (FVT), or MM-111 and fulvestrant.
  • FIG. 4 d shows the effect of trastuzumab, fulvestrant, or trastuzumab and fulvestrant.
  • FIG. 4 e shows the effect of MM-111, trastuzumab, or MM-111 and trastuzumab.
  • FIG. 4 f shows the effect of MM-111, trastuzumab, and tamoxifen combined compared to that of any of the double combinations.
  • FIG. 4 g shows the effect of MM-111, trastuzumab, and fulvestrant combined compared to that of any of the double combinations.
  • the x-axes are a log scale of each drug concentration for each experimental condition in nM and the y axis is spheroid size as % of control spheroid size.
  • FIG. 5 is a graph summarizing the effect of MM-111, trastuzumab, and tamoxifen combined compared to that of any of the double combinations or MM-111, trastuzumab, and fulvestrant combined compared to that of any of the double combinations at inhibiting single ligand (estrogen or heregulin) or dual-ligand (estrogen and heregulin)-stimulated spheroid growth in vitro.
  • the y-axis is % inhibition of spheroid size normalized to stimulated control.
  • FIG. 6 is a graph showing that the combination of MM-111 and lapatinib inhibits tumor growth in vivo.
  • the x-axis shows the time post tumor implant in days and the y-axis shows tumor volume in mm 3 . Mice were treated with inhibitors on day 7 post tumor implant.
  • FIG. 7 evaluates the ability of lapatinib to inhibit ErbB3 and AKT activation in heregulin-stimulated cells.
  • 7 a is a graph comparing computer-generated dose-response curves to experimental results in heregulin-stimulated BT474-M3 cells.
  • 7 b is a graph showing lapatinib inhibition (IC50) of ErbB3 and AKT activation in heregulin-stimulated and unstimulated cells following a 1-hour incubation with inhibitor.
  • FIG. 8 is a series of graphs showing MM-111 or lapatinib inhibition of ErbB3 ( 8 a ) or AKT ( 8 b ) activation in heregulin-stimulated cells incubated with inhibitor for 15 minutes, 1 hour, 4 hours, and 24 hours.
  • FIG. 8 c shows a comparison of IC50 for MM-111 and lapatinib at 1 hour and 24 hours for both BT474M3 cells and ZR75-30 cells.
  • FIG. 9 is a graph showing the effect of MM-111 and lapatinib combination treatment on AKT activation in heregulin-stimulated BT474-M3 cells.
  • FIG. 10 is a graph showing the effect of lapatinib on cell viability as a measure of proliferation of unstimulated and heregulin-stimulated BT474-M3 cells.
  • FIG. 11 is a graph showing the effect of MM-111, lapatinib, or the combination on BT474-M3 cell apoptosis. The number of dead cells, cells in late apoptosis, early apoptosis, and live cells was quantitated.
  • FIG. 12 is three graphs showing that MM-111 combines positively with anti-estrogen drugs and lapatinib in inhibiting dual ligand (estrogen (E2) and heregulin (HRG))-stimulated spheroid growth in vitro.
  • FIG. 12 a shows the effect of lapatinib alone or the combination of lapatinib and fulvestrant (FVT).
  • FIG. 12 b shows the effect of lapatinib alone or the combination of lapatinib and MM-111.
  • FIG. 12 c shows the effect of lapatinib alone, the combination of MM-111 and fulvestrant, or the triple combination of MM-111, FVT, and lapatinib.
  • Lapatinib is given in 3.3, 10, or 30 nM doses.
  • the x-axes are a log scale of each of MM-111 and/or FVT concentration in nM and the y axis is spheroid size as % of control (FBS alone) spheroid size.
  • FIG. 13 is four graphs showing that MM-111 combines positively with the aromatase inhibitor letrozole and the tyrosine kinase inhibitor lapatinib in heregulin (HRG) and androstenedione (A4)-stimulated BT474-M3-Aro cells that stably express human aromatase, which converts androstenedione to estrogen.
  • FIG. 13 a shows the effect of letrozole, MM-111, or the combination of letrozole and MM-111.
  • FIG. 13 b shows the effect of lapatinib, MM-111 or the combination of lapatinib and MM-111.
  • FIG. 13 c shows the effect of lapatinib, letrozole, or the combination of lapatinib and letrozole.
  • FIG. 13 d shows the effect of the dual combinations of MM-111 and letrozole, MM-111 and lapatinib, lapatinib and letrozole, and the triple combination of MM-111, lapatinib and letrozole.
  • the x-axes are a log scale of MM-111 concentration in nM.
  • the drug concentrations are a ratio of 10:20:1 MM-111 to letrozole to lapatinib.
  • the y axis is spheroid size as % of control spheroid size.
  • FIG. 14 is two graphs showing that MM-111 combines positively with the PI3K/mTOR inhibitor BEZ235 and trastuzumab in unstimulated ( FIG. 14 a ) and heregulin-stimulated ( FIG. 14 b ) NCI-N87 cells in vitro.
  • Cells were plated and either untreated (Control, open circle), treated with BEZ235 alone (open diamond), or treated with BEZ235 (5 nM FIG. 14 a, 20 nM FIG. 14 b ) and either trastuzumab (closed circle), MM-111 (closed square), or the combination of MM-111 and trastuzumab (closed diamond).
  • Closed circles may appear as squares or rectangles on trastuzumab curves when they comprise error bars.
  • the x-axes are a log scale of the concentrations of each of MM-111 and/or trastuzumab.
  • FIG. 15 is two graphs showing that MM-111 combines positively with the mTOR inhibitor everolimus and trastuzumab in unstimulated ( FIG. 15 a ) and heregulin-stimulated ( FIG. 15 b ) NCI-N87 cells in vitro.
  • Cells were plated and either untreated (Control, open circle), treated with everolimus alone (open diamond), or treated with everolimus (1 nM FIG. 15 a, 5 nM FIG. 15 b ) and either trastuzumab (closed circle), MM-111 (closed square), or the combination of MM-111 and trastuzumab (closed diamond). Closed circles may appear as squares or rectangles on trastuzumab curves when they comprise error bars.
  • the x-axes are a log scale of the concentrations of each of MM-111 and/or trastuzumab.
  • FIG. 16 is two graphs showing that MM-111 combines positively with the MEK inhibitor GSK1120212 (trametinib) in a mouse xenograft study.
  • Phospho-ERK was measured as a function of ERK activity via a Luminex® immunosandwich assay at 4 hours after the initial treatment ( FIG. 16A ) and 24 hours after the initial treatment ( FIG. 16B ) as a function of ERK activity via a Luminex immunosandwich assay.
  • the x-axes show type of treatment and the y-axes show mean fluorescent intensity.
  • FIG. 17 is two graphs showing that MM-111 combines positively with T-DM1 (ado-trastuzumab emtansine) in both the absence ( FIG. 17A ) and presence ( FIG. 17B ) of endogenous heregulin.
  • the x-axes show a log scale of T-DM1 concentration and the y-axes show viability as % of control.
  • FIG. 18 is a series of 3D graphs showing that MM-111 combines positively with TDM-1 (ado-trastuzumab emtansine) in the presence of exogenous heregulin.
  • NCI-N87 ( FIGS. 18A and 18B ) or BT-474-M3 ( FIGS. 18C and 18D ) cells were incubated with a dose range of both T-DM1 and heregulin, either without MM-111 ( FIGS. 18A and 18C ) or with MM-111 ( FIGS. 18B and 18D ).
  • the x-axes are the concentration of T-DM1
  • the y-axes are cell viability as % of untreated cells
  • the z-axes are heregulin concentration.
  • the data presented in these graphs is also set forth below in Tables 1-4.
  • bispecific anti-ErbB2/anti-ErbB3 antibodies are co-administered with one or more additional therapeutic agents (e.g. an aromatase inhibitor or tyrosine kinase inhibitor), to provide effective treatment to human patients having a cancer.
  • additional therapeutic agents e.g. an aromatase inhibitor or tyrosine kinase inhibitor
  • Such co-administrations beneficially have an additive or superadditive effect on suppressing tumor cell growth, which effect on suppressing tumor cell growth is measured, e.g., in a mouse xenograft model e.g., using BT-474 or NCI-N87 cells.
  • anti-ErbB3 agent refers to any therapeutic agent that binds to ErbB3 or binds to an ErbB3-specific ligand or blocks the expression of ErbB3, and thereby inhibits the activity of cellular signaling mediated by ErbB3.
  • types of anti-ErbB3 agents include antibodies, bispecific antibodies, ligand analogs, soluble forms of ErbB3 or the ErbB3 ectodomain, ErbB3 specific RNAi molecules, and similar biologic agents.
  • antibody describes a polypeptide comprising at least one antibody-derived antigen binding site (e.g., V H /V L region or Fv, or complementarity determining region—CDR) that specifically binds to a specific antigen, e.g., ErbB3.
  • V H /V L region or Fv, or complementarity determining region—CDR antibody-derived antigen binding site
  • Antibodies include whole antibodies and any antigen binding fragment, e.g., Fab or Fv, or a single chain fragment (e.g., scFv), as well as bispecific antibodies and similar engineered variants, human antibodies, humanized antibodies, chimeric antibodies Fabs, Fab′2s, ScFvs, SMIPs, Affibodies®, nanobodies, or a domain antibodies, and may be of any of the following isotypes: IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgAsec, IgD, and IgE.
  • IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgAsec, IgD, and IgE isotypes
  • the antibody may be a naturally occurring antibody or may be an antibody that has been altered (e.g., by mutation, deletion, substitution, conjugation to a non-antibody moiety).
  • an antibody may include one or more variant amino acids (compared to a naturally occurring antibody) which change a property (e.g., a functional property) of the antibody.
  • a property e.g., a functional property
  • numerous such alterations are known in the art which affect, e.g., half-life, effector function, and/or immune responses to the antibody in a patient.
  • antibody thus includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion,” e.g., Fabs) or single chains thereof (e.g., scFvs) as well as bispecific antibodies and similar engineered variants, provided that they retain the binding specificity of an antibody.
  • antigen binding fragment i.e., “antigen-binding portion,” e.g., Fabs
  • single chains thereof e.g., scFvs
  • an “anti-ErbB3 antibody” is an antibody that immunospecifically binds to the ectodomain of ErbB3 and an “anti-ErbB2 antibody” is an antibody that immunospecifically binds to the ectodomain of ErbB2.
  • the antibody may be an isolated antibody.
  • Such binding to ErbB3 or ErB2 exhibits a Kd with a value of no greater than 50 nM as measured by a surface plasmon resonance assay or a cell binding assay.
  • Exemplary anti-ErbB3 antibodies inhibit EGF-like ligand mediated phosphorylation of ErbB3, e.g., anti-ErbB2 antibodies that inhibit the binding of heregulin to ErbB2/ErbB3 heterodimers.
  • EGF-like ligands include EGF, TGF ⁇ , betacellulin, heparin-binding epidermal growth factor, biregulin, epigen, epiregulin, and amphiregulin, which typically bind to ErbB1 and induce heterodimerization of ErbB1 with ErbB3.
  • bispecific antibody refers to a protein comprising two antigen-binding sites, a first binding site exhibiting immunospecific binding to a first antigen or epitope and a second binding site exhibiting immunospecific binding to a second antigen or epitope distinct from the first.
  • An “anti-ErbB2/anti-ErbB3 bispecific antibody” is an antibody that comprises two binding sites, one that immunospecifically binds to the ectodomain of ErbB3 and another that immunospecifically binds to the ectodomain of ErbB2.
  • An exemplary bispecific ErbB3, ErbB2 antibody is an antibody comprising SEQ ID NO:1.
  • an “anti-estrogen agent” as used herein refers to an agent that prevents or reduces production of estrogen or prevents or reduces signaling mediated by estrogen receptors.
  • Anti-estrogen agents include but are not limited to estrogen receptor antagonists and aromatase inhibitors.
  • Estrogen receptor antagonists include but are not limited to raloxifene, fulvestrant, tamoxifen, afimoxifene (4-hydoroxytamoxifen), arzoxifene, toremifene, and lasofoxone.
  • the estrogen receptor antagonist is tamoxifen or fulvestrant.
  • Aromatase inhibitors work by blocking the synthesis of estrogen in an animal (e.g., a mouse or a human). This lowers estrogen levels in the animal and thereby inhibits the growth of estrogen-driven cancers.
  • aromatase inhibitors include but are not limited to exemestane, anastrozole, letrozole, aminoglutethimide, testolactone, vorozole, formestane, and fadrozole.
  • the aromatase inhibitor is exemestane or letrozole.
  • cancer any condition characterized by abnormal, unregulated, malignant cell growth.
  • malignant tumor is meant any cancer that takes the form of a tumor.
  • the term “effective amount” refers to an amount of a drug effective to achieve a desired effect, e.g., to ameliorate disease in a subject.
  • the effective amount of the drug may inhibit (e.g., slow to some extent, inhibit or stop) one or more of the following characteristics: cancer cell growth, cancer cell proliferation, cancer cell motility, cancer cell infiltration into peripheral organs, tumor metastasis, and tumor growth.
  • the effective amount of the drug may alternately do one or more of the following when administered to a subject: slow or stop tumor growth, reduce tumor size (e.g., volume or mass); relieve to some extent one or more of the symptoms associated with the cancer, extend progression free survival, result in an objective response (including a partial response or a complete response), and increase overall survival time.
  • the drug may prevent growth and/or kill existing cancer cells, it is cytostatic and/or cytotoxic.
  • a “mouse effective amount” refers to an amount of a drug effective to achieve a desired effect when the subject is a mouse.
  • a “human effective amount” refers to an amount of a drug effective to achieve a desired effect when the subject is a human patient.
  • combination therapy refers to the administration of at least two therapeutic agents to a subject either simultaneously or within a time period during which the effects of the earlier-administered therapeutic agent are still operative in the subject when a later-administered therapeutic agent is administered.
  • a “receptor tyrosine kinase inhibitor” as used herein refers to a member of a class of drugs that specifically inhibit receptor tyrosine kinases and thus reduce or eliminate the activation of various signal transduction pathways.
  • Receptor tyrosine kinase inhibitors useful for the treatment of cancer as disclosed herein include but are not limited to the small molecule inhibitors erlotinib, afatinib, dasatinib, gefitinib, imatinib, pazopinib, lapatinib, sunitinib, nilotinib and sorafenib.
  • Receptor tyrosine kinase inhibitors also include antibody-based therapeutics such as cetuximab, panitumumab, zalutumumab, nimotuzumab, and matuzumab).
  • the receptor tyrosine kinase inhibitor is lapatinib.
  • Dosage refers to parameters for administering a drug in defined quantities per unit time (e.g., per hour, per day, per week, per month, etc.) to a patient. Such parameters include, e.g., the size of each dose. Such parameters also include the configuration of each dose, which may be administered as one or more units, e.g., taken at a single administration, e.g., orally (e.g., as one, two, three or more pills, capsules, etc.) or injected (e.g., as a bolus). Dosage sizes may also relate to doses that are administered continuously (e.g., as an intravenous infusion over a period of minutes or hours).
  • Such parameters further include frequency of administration of separate doses, which frequency may change over time.
  • a “dosing cycle” or “dosing interval” is the period of time that comprises one cycle of treatment (e.g., 21 days or 28 days) for a dosing regimen.
  • Dose refers to an amount of a drug given in a single administration.
  • the components of the combination of the anti-ErbB2/ErbB3 antibody have an additive or superadditive effect on suppressing tumor cell growth, as compared to monotherapy with the anti-ErbB2/ErbB3 antibody or treatment with the other agents in the absence of antibody therapy.
  • additive is meant a result that is greater in extent (e.g., in the degree of reduction of tumor mitotic index or of tumor growth or in the degree of tumor shrinkage or the frequency and/or duration of symptom-free or symptom-reduced periods) than the best separate result achieved by monotherapy with each individual component, while “superadditive” is used to indicate a result that exceeds in extent the sum of such separate results.
  • the additive effect is measured as slowing or stopping of tumor growth or tumor cell proliferation.
  • the additive effect can also be measured as, e.g., reduction in size of a tumor, reduction of tumor mitotic index, reduction in number of metastatic lesions over time, increase in overall response rate, or increase in median or overall survival.
  • T C represents the delay in growth of the cells, which is the average time, in days, for the tumors of the treated group (T) and the tumors of the control group (C) to have reached a predetermined value (1 g, or 10 mL, for example), and Td represents the time, in days necessary for the volume of the tumor to double in the control animals.
  • a combination, used at its own maximum tolerated dose, in which each of the constituents is present at a dose generally less than or equal to its maximum tolerated dose exhibits therapeutic synergy when the log 10 cell kill is greater than the value of the log 10 cell kill of the best constituent when it is administered alone.
  • the log 10 cell kill of the combination exceeds the value of the log 10 cell kill of the best constituent of the combination by at least 0.1 log cell kill, at least 0.5 log cell kill, or at least 1.0 log cell kill.
  • Preferred cancer cells of cell lines are cells of ErbB2 expressing cell lines such as ErbB2 overexpressing cell lines, e.g., BT474-M3 (ATCC® # CRL-HTB-20TM, derived from breast ductal carcinoma cells), BT474-M3-Aro (BT474-M3 cells that stably express human aromatase), ZR75-30 (ATCC® # CRL1504TM, derived from breast ductal carcinoma cells), SKOV-3 (ATCC® # HTB-77TM, derived from metastatic ovarian adenocarcinoma cells), MCF7 (ATCC® # HTB-22TM) clone 18, MDA-MB-453 (ATCC® # HTB-131TM, derived from breast carcinoma cells), SK-BR-3 (ATCC® # HTB-30TM, derived from breast adenocarcinoma cells), and NCI-N87 (ATCC® # CRL-5822TM, derived from gastric carcinoma cells).
  • Cancers may include, for example, solid tumors such as: sarcomas (e.g., clear cell sarcoma), carcinomas (e.g., renal cell carcinoma), and lymphomas; tumors of the breast, colon, rectum, lung, oropharynx, hypopharynx, esophagus, gastric esophageal junction, stomach, pancreas, liver, bilecyst, bile duct, small intestine, urinary system (including the kidney, bladder, and epithelium of the urinary tract), female genital system (including the uterine neck, uterus, ovary, chorioma, and gestational trophoblast), male genital system (including the prostate, seminal vesicle, and testicles), endocrine glands (including the thyroid gland, adrenal gland, and pituitary body), skin (including angioma, melanoma, sarcoma originating from bone or soft tissue, and
  • a cancer may be an estrogen receptor positive (ER+) cancer.
  • ER+ estrogen receptor positive
  • Such cancers exemplify candidates for therapy regimens that include anti-estrogen agents.
  • Such cancers may include but are not limited to certain breast, ovarian, uterine, endometrial, lung, bone, brain, bladder, liver and urogenital cancers.
  • a cancer may be an ErbB2 gene-amplified cancer and/or an ErbB2-expressing or overexpressing cancer.
  • ErbB2 also known as HER2 or Neu, is a cell surface transmembrane receptor protein that generates intracellular signals (e.g., upon ligand activation) via its intracellular tyrosine kinase activity. In excess, such signals can promote oncogenesis e.g., by triggering cell division.
  • the ErbB2 gene is amplified and/or overexpressed in many types of human malignancies, including but not limited to breast, ovarian, endometrial, pancreatic, colorectal, prostate, salivary gland, kidney, and lung.
  • ErbB2 overexpressing cancers are designated a HER2 +++ or HER2 ++ depending on the level of ErbB2 overexpression, with HER2 +++ indicating the highest levels of HER2 expression.
  • HER2 +++ and HER2 ++ status are typically determined by an immunoassay such as immunohistochemistry, e.g., Herceptest®.
  • ErbB2 gene amplification may be determined by, e.g., FISH (fluorescence in situ hybridization), with HER2-amplified cancer cells being those that have more than two HER2 gene copies being HER2-amplified, and cells and/or tumors comprising HER2-amplified cancer cells being referred to as “FISH positive.”
  • bispecific anti-ErbB2, antiErbB3 antibodies that are scFv HSA conjugates are described in co-pending US patent publication No. 2011-0059076, and PCT publication Nos. WO2009/126920 and WO 2010/059315, MM-111 (also referred to as B2B3-1) and other bispecific anti-ErbB2/antiErbB3 antibodies that are scFv HSA conjugates and that are suitable for use in the methods and compositions provided herein, including the components of A5-HSA-ML3.9, ML3.9-HSA-A5, A5-HSA-B1D2, B1D2-HSA-A5, B12-HSA-B1D2, B1D2-HSA-B12, A5-HSA-F5B6H2, F5B6H2-HSA-A5, H3-HSA-F5B6H2, F5B6H2-HSA-H3, F4-HSA-F5B6H2, F5B6H2-HSA-F
  • MM-111 is currently undergoing clinical trials, including an open-label Phase 1/2 and pharmacologic study of MM-111 in patients with advanced, refractory HER2 positive cancers, an open-label Phase 1/2 trial of MM-111 in combination with trastuzumab (Herceptin®) in patients with advanced HER2 positive breast cancer, an open label, Phase 1/2 and pharmacologic study of MM-111 with five different combination treatments: MM-111 in combination with cisplatin, capecitabine, and trastuzumab, MM-111 in combination with lapatinib and trastuzumab, and MM-111 in combination with paclitaxel and trastuzumab, MM-111 in combination with lapatinib, paclitaxel and trastuzumab, and MM-111 in combination with docetaxel and
  • a bispecific anti-ErbB2/anti-ErbB3 antibody (e.g., MM-111) can be co-administered with other therapeutic agents, (e.g, an anti-estrogen receptor agent or a receptor tyrosine kinase inhibitor) prior to (e.g., neoadjuvant therapy), concurrent with, or following (e.g., adjuvant therapy) radiotherapy of, or surgical intervention to remove, a malignant tumor.
  • other therapeutic agents e.g, an anti-estrogen receptor agent or a receptor tyrosine kinase inhibitor
  • Additional therapeutic agents suitable for combination with anti-ErbB2/anti-ErbB3 antibodies may further include: 1) antibody EGFR inhibitors (e.g. MM-151, Sym004, cetuximab, panitumumab, zalutumumab, nimotuzumab, and matuzumab), additional small molecule tyrosine kinase inhibitors such as PKI-166, PD-158780, EKB-569 (pelitinib), tyrphostin AG-1478, and pan-HER kinase inhibitors (e.g.
  • antibody EGFR inhibitors e.g. MM-151, Sym004, cetuximab, panitumumab, zalutumumab, nimotuzumab, and matuzumab
  • additional small molecule tyrosine kinase inhibitors such as PKI-166, PD-158780, EKB-569 (pelitinib),
  • microtubule stabilizing agents e.g.
  • laulimalide epothilone A, epothilone B, discodermolide, eleutherobin, sarcodictyin A, sarcodictyin B, paclitaxel, nab-paclitaxel or docetaxel); antimetabolites such as 5-fluorouracil (5-FU) and capecitabine; 3) platinum-based therapeutics such as oxaliplatin, carboplatin and cisplatin; 4) mTOR inhibitors such as BEZ235 (a dual PI3K/mTOR inhibitor), AZD8055, everolimus, temsirolimus, siroplimus/rapamycin, and ridaforolimus; and 5) additional anti-ErbB2 therapeutic agents, such as trastuzumab, pertuzumab, and T-DM1 (ado-trastuzumab emtansine). Additional examples of therapeutic agents suitable for combination with anti-ErbB2/anti-ErbB3 antibodies may be
  • MM-111 is suitable for both large scale production and systemic therapy. MM-111 binds to ErbB2/ErbB3 heterodimers and forms a trimeric complex with ErbB2 and ErbB3, effectively inhibiting ErbB3 signaling.
  • the antitumor activity of MM-111 requires the presence of both ErbB2 and ErbB3, but is particularly dependent on ErbB2 expression.
  • the affinity of its ErbB2 antigen-binding site is about 30 times higher than the affinity of its ErbB3 antigen-binding site, but the ErbB2 antigen-binding site does not by itself inhibit ErbB2 activity when bound to ErbB2.
  • MM-111 is administered to human subjects (patients) at an interval measured in days, as a single loading dose of at least 20 mg/kg of MM-111 followed by at least seven day intervals (e.g., every 2 weeks) by at least one administration of a single maintenance dose of MM-111, where the maintenance dose is generally smaller than the loading dose, e.g., at least 5 mg/kg less than the loading dose.
  • BT474-M3 wild type cells 2000 cells/well are plated in Ultra Low Cluster 96-well plate (Costar). After overnight incubation, indicated treatments are introduced to the plate. Cells are continued to culture for six days. Spheroids are then examined by Nikon microscope and analyzed by MetaMorph Image Analysis Software (Molecular Devices). The spheroid size from cells cultured in medium containing 10% FBS is set as control.
  • BT474-M3 cells (2 ⁇ 10 7 cells per mice) are inoculated subcutaneously into Nu/Nu immunodeficient mice, which are implanted with an estrogen pellet (0.72 mg; 60-day release) one day before the experiment. Tumors are measured after seven days and mice are randomized into four groups: those treated with placebo, MM-111 (60 mg/kg, Q7D), 4-hydroxytamoxifen (5 mg; 60-day release pellet), and combination of MM-111 and 4-hydroxytamoxifen, respectively. Tumors are measured every three days and the experiment is ended at day 32.
  • mice were prepared in the xenograft model using the methods described above or minor variations thereof. Mice were inoculated with tumor forming BT474-M3 cells and on day 7 given a placebo (vehicle control), MM-111, tamoxifen, or a combination of MM-111 and tamoxifen and tumor growth was measured over time. As shown in FIG.
  • this in vivo BT474-M3 xenograft model showed resistance to tamoxifen treatment but when mice were given a combination of MM-111 and tamoxifen the combination treatment inhibited tumor growth to a significantly greater extent.
  • Statistical significance (p ⁇ 0.05) was observed for the combination group from day 28 onward when compared to vehicle control, from day 21 onward when compared to MM-111 and from day 25 onward when compared to tamoxifen.
  • Multicellular spheroids are used to simulate the growth and microenvironmental conditions of tumors in vitro.
  • spheroids of BT474-M3 cells were prepared using the methods described above or minor variations thereof and treated with an ErbB2-binding therapeutic and/or an anti-estrogen therapeutic.
  • Spheroids of estrogen-stimulated cells were treated with a dose range of MM-111, tamoxifen, or the combination of MM-111 and tamoxifen ( FIG. 2 a ); trastuzumab, tamoxifen or the combination of trastuzumab and tamoxifen ( FIG.
  • MM-111, fulvestrant, or the combination of MM-111 and fulvestrant FIG. 2 c
  • trastuzumab, fulvestrant, or the combination of trastuzumab and fulvestrant FIG. 2 d
  • MM-111, trastuzumab, or the combination of MM-111 and trastuzumab FIG. 2 e
  • MM-111, trastuzumab, fulvestrant and tamoxifen showed inhibitory effects on spheroid growth in the estrogen-stimulated BT474-M3 spheroid assay.
  • the combination of tamoxifen or fulvestrant with MM-111 ( FIGS. 2 a and 2 c , respectively) or trastuzumab ( FIGS. 2 b and 2 d , respectively) increased the degree of growth inhibition, as did the combination of MM-111 and trastuzumab ( FIG. 2 e ).
  • the inhibitory effects were increased still further when estrogen-stimulated spheroids were treated with the triple combination of MM-111, trastuzumab, and tamoxifen ( FIG. 2 f ) or MM-111, trastuzumab, and fulvestrant ( FIG. 2 g ) as compared to the double combinations of drugs.
  • spheroids of heregulin (HRG)-stimulated BT474-M3 cells were prepared using the methods described above or minor variations thereof and treated with a dose range of MM-111, tamoxifen, or the combination of MM-111 and tamoxifen ( FIG. 3 a ); trastuzumab, tamoxifen or the combination of trastuzumab and tamoxifen ( FIG. 3 b ); MM-111, fulvestrant, or the combination of MM-111 and fulvestrant ( FIG.
  • HRG heregulin
  • FIG. 3 c trastuzumab, fulvestrant, or the combination of trastuzumab and fulvestrant
  • FIG. 3 d a trastuzumab, a )
  • MM-111 MM-111 inhibited heregulin-induced spheroid growth but tamoxifen ( FIG. 3 a ), trastuzumab ( FIG. 3 b ), and fulvestrant ( FIG. 3 c ) did not inhibit heregulin stimulated spheroid growth. No significant combinational effect was observed when MM-111 was used with tamoxifen ( FIG. 3 a ) or fulvestrant ( FIG.
  • Dual ligand (estrogen and heregulin) stimulated spheroids were treated with a dose range of tamoxifen, MM-111 or the combination of MM-111 and tamoxifen ( FIG. 4 a ) or trastuzumab, tamoxifen or the combination of trastuzumab and tamoxifen ( FIG. 4 b ). While MM-111 and trastuzumab each inhibited spheroid growth ( FIG. 4 a ) the combination of MM-111 and tamoxifen showed greater inhibitory effects than either drug alone. In contrast, trastuzumab alone had no significant inhibitory effects and the combination of trastuzumab and tamoxifen showed similar effects to tamoxifen alone.
  • Dual ligand stimulated spheroids were then treated with a dose range of fulvestrant, MM-111 or the combination of MM-111 and fulvestrant ( FIG. 4 c ) or fulvestrant, trastuzumab, or a combination of fulvestrant or trastuzumab ( FIG. 4 d ).
  • MM-111 and fulvestrant each separately inhibited spheroid growth the combination of MM-111 and fulvestrant showed greater inhibitory effects than either drug alone ( FIG. 4 c ).
  • Trastuzumab alone had no significant inhibitory effects and the combination of trastuzumab and fulvestrant showed similar effects to tamoxifen alone ( FIG. 4 d ).
  • Dual ligand stimulated spheroids were then treated with MM-111, trastuzumab, or a combination of MM-111 and trastuzumab.
  • MM-111 showed greater inhibitory effects than trastuzumab in dual ligand-stimulated spheroid growth. Enhanced inhibitory effects were observed when both drugs were combined ( FIG. 4 e ).
  • the triple combination of MM-111, trastuzumab and either tamoxifen ( FIG. 4 f ) or fulvestrant ( FIG. 4 g ) showed similar inhibitory effects to those of MM-111 and trastuzumab in combination ( FIG. 4 e ) on estrogen- and heregulin-(dual ligand) stimulated spheroid growth.
  • Serum-starved cells are pre-incubated with serial dilutions of MM-111, lapatinib or combinations at doses and treatment times indicated, followed by stimulation with 5 nM heregulin 143 (R&D Systems, Minneapolis, Minn.) for 10 minutes.
  • Cell lysates are probed for phospho-ErbB3 (pErbB3), and phospho-AKT (pAKT) by ELISA as described previously (Schoeberl et al, 2009)
  • Inhibitor IC 50 values are calculated by fitting dose-response data to a 4-parameter sigmoidal curve (GraphPad Prism®, GraphPad Software, Inc., La Jolla, Calif.).
  • Cells (8,000/well) are seeded into 96-well plates and incubated overnight Inhibitor is added at doses indicated and cells are treated for 24 hours.
  • Inhibitor is added at doses indicated and cells are treated for 24 hours.
  • cells are serum-starved overnight prior to addition of inhibitor and 2 nM heregulin 1—(R&D Systems, Minneapolis, Minn.) is added 1 hour post-inhibitor treatment in media containing 5% FBS. Numbers of viable cells are measured as an indicator of cell proliferation using the CellTiter-Glo® Luminescent Cell Viability Assay (Promega, Madison, Wis.).
  • BT474-M3 cells (2000 cells/well) are plated in Ultra Low Cluster 96-well plate (Costar®, Corning, N.Y.). After overnight incubation, spheroids are treated with inhibitor at concentrations indicated for 72 hours. Spheroids are then trypsinized and combined with floating cells. Cells are washed twice with cold PBS and suspended in binding buffer (0.01 M HEPES, pH 7.4; 0.14 M NaCl; 2.5 mM CaCl 2 ). Cells are then stained with FITC-conjugated Annexin V and PI. Apoptotic cells are quantified on a FACSCaliburTM FACS machine.
  • Tumor xenografts are established by subcutaneous injection of BT474-M3 cells into the flank of 5-6 weeks old female athymic nude mice (nu/nu; Charles River Labs, Wilmington, Mass.). Mice receive a subcutaneous 60 day, slow-release estrogen implant in the opposite flank (0.72 mg pellet; Innovation Research of America, Sarasota, Fla.) 24 hours prior to the injection of cells. Once tumors reach a mean volume of 150-500 mm 3 , mice are randomized into groups of 8 or 10 and dosed by intraperitoneal injection once every three days with vehicle, MM-111 or lapatinib. For lapatinib combination studies, MM-111 is given once every seven days and lapatinib daily by gavage at doses indicated.
  • BT474-M3 cells were transfected with PS100010 vector containing human aromatase (gene accession No: NM — 000103.2). Cells with stable expression of aromatase (BT474-M3-Aro) were obtained after selection with 400 ⁇ g/ml geneticin.
  • BT474-M3-Aro cells (5000 cells/well) were plated in phenol red-free RPMI-1640 medium containing 5% charcoal-stripped FBS into 96-well plate. After overnight incubation, indicated treatments were introduced in the presence of androstenedione (A-4; 200 nM) and heregulin (HRG; 2 nM). After three days of treatment, cell viability was determined by WST-1 (Roche; Cat. #11 644 807 001) according to manufacturer's instruction. Cell viability in the presence of 5% charcoal-stripped FBS was set as control (100%).
  • MM-111 and lapatinib were each dosed at an optimal efficacious dose weekly and daily, respectively.
  • the percent change in tumor volume from day 40 to day 7 (inoculation) was calculated for each group ( FIG. 6 b ).
  • MM-111 The combination of MM-111 and lapatinib resulted in a percent change in tumor volume of ⁇ 69% (about 70%), reflecting tumor regressions, compared to ⁇ 11% (about 10%) for lapatinib and 14% (about 15%) for MM-111.
  • a dose range of lapatinib inhibition of pErbB3 activation was predicted using the computational modeling described above.
  • a dose range of lapatinib was applied to BT474-M3 cells followed by stimulation with 5 nM heregulin for 10 min.
  • the amount of pErbB3 was measured by ELISA using the methods described above or minor variations thereof.
  • Model-generated dose-response curves overlay the experimental data ( FIG. 7 a ).
  • a comparison of the inhibitory activity of lapatinib in heregulin-stimulated or unstimulated (basal) cells was performed to demonstrate that heregulin signaling perturbs the activity of lapatinib.
  • MM-111 is a More Potent Inhibitor of HRG-Driven ErbB3 and AKT Phosphorylation than Lapatinib
  • BT474-M3 In order to compare the ability of MM-111 and lapatinib to inhibit heregulin-induced ErbB3 activation, BT474-M3, or an additional ErbB2 overexpressing breast tumor cell line, ZR75-30 (ATCC® #
  • Good reduction by MM-111 of pAKT levels (inhibition of AKT phosphorylation) in BT474-M3 (IC 50 10) ( FIG.
  • the ability of MM-111 to inhibit heregulin-induced ErbB3 activation (phosphorylation) was superior to lapatinib by greater than an order of magnitude and the relative IC 50 for each inhibitor ( FIG. 8 c ) was consistent following up to 24 hours incubation with inhibitors, indicating treatment times had little effect on the potency of the inhibitors.
  • MM-111 combined with lapatinib on pAKT inhibition (reduction of pAKT levels) was assessed in heregulin-stimulated BT474-M3 cells.
  • Cells were incubated for 2 hours with a dose range of MM-111, lapatinib or their combination and pAKT was measured by ELISA.
  • MM-111 and lapatinib was extremely effective, inhibiting pAKT well below basal levels at therapeutically relevant concentrations ( FIG. 9 ).
  • Treatment with either MM-111 (1 ⁇ M) or lapatinib (1 ⁇ M) alone resulted in similar levels of pAKT inhibition (see FIG. 8 b ) while the combination resulted in about 20% more inhibition of pAKT.
  • lapatinib The effect of lapatinib on cell proliferation was measured in unstimulated and heregulin-stimulated BT474-M3 cells.
  • Cells grown in serum or in serum plus 2 nM heregulin were treated with lapatinib across a dose range for 24 hours.
  • Lapatinib treatment resulted in about a 50% inhibition of unstimulated cells but its effect was reduced to about 23% inhibition in heregulin-stimulated BT474-M3 cells ( FIG. 10 ).
  • MM-111 The effect of the MM-111 combination with lapatinib on apoptosis was assessed in a BT474-M3 spheroid model.
  • Spheroids were prepared using the methods described above or minor variations thereof and treated with MM-111 (100 nM), lapatinib (33 nM), or a combination of 100 nM MM-111 and 33 nM lapatinib.
  • Cells were then stained with Annexin V and propidium iodide (PI) and quantitated using FACS ( FIG. 11 , Table 2).
  • MM-111 Combines Positively with Anti-Estrogen Drugs and Lapatinib in Inhibiting Dual Ligand (Estrogen and Heregulin)-Stimulated Spheroid Growth
  • spheroids of estrogen and heregulin-stimulated BT474-M3 cells were prepared using the methods described above or minor variations thereof and treated with 3.3 nM, 10 nM, or 30 nM lapatinib, either alone or in combination with a dose range of fulvestrant (FVT) ( FIG. 12 a ); 3.3 nM, 10 nM, or 30 nM lapatinib, either alone or in combination with a dose range of MM-111 ( FIG.
  • MM-111 Combines Positively with Anti-Estrogen Drugs in Inhibiting Spheroid Growth in BT474-M3 Cells Overexpressing Human Androstenedione
  • Androstenedione is a steroid hormone that is converted to estrogen by aromatase.
  • aromatase-expressing cells were treated in the presence of androstenedione (A4) and heregulin (HRG) with MM-111, letrozole, or the combination of MM-111 or letrozole ( FIG. 13 a ); MM-111, lapatinib, or the combination of MM-111 and lapatinib ( FIG. 13 b ); lapatinib, letrozole, or the combination of lapatinib and letrozole ( FIG.
  • MM-111 is prepared as a formulation containing 25 mg/ml MM-111 in a sterile aqueous solution comprising 20 mM L-histidine hydrochloride, 150 mM sodium chloride, pH 6.5, which is stored at 2-8° C.
  • MM-111 must be brought to room temperature prior to administration.
  • Containers e.g., vials
  • MM-111 must not be shaken.
  • the appropriate quantity of MM-111 is removed from the container, diluted in 250 mL of 0.9% normal saline and administered as an infusion using a low protein binding in-line filter (e.g., a 0.22 micrometer filter).
  • a low protein binding in-line filter e.g., a 0.22 micrometer filter
  • MM-111 is initially administered over about 90 minutes (first administration). In the absence of an infusion reaction, subsequent doses are administered over about 60 minutes.
  • a patient's body weight at the start of a dosing cycle is used to calculate the dose used throughout the cycle. Should a patient's body weight change by more than 10%, a new total dose is calculated to reflect this change.
  • Preferred plasma concentrations of MM-111 achieved during treatment are at least 106 mg/L. It has now been discovered that certain combinations of dose frequency and dosage will achieve and maintain this plasma concentration during the course of treatment in at least half, and preferably in more than 60%, 70% or 80% of treated patients.
  • a higher initial dose (loading dose—LD) is given, followed as defined intervals by at least one maintenance dose (MD).
  • Intervals of dosing in days are typically indicated as QxD, wherein x represents an integer, so that a QxD of 7 indicates dosing every 7 days.
  • Table 3A, Table 3B, and Table 3C below show doses and dosing intervals of the invention.
  • the indicated loading doses are optional—initial doses are preferably made at the indicated loading dose (LD), but may (e.g., as directed or at the physician's discretion) be made at the maintenance dose (MD).
  • Table 3A provides a set of exemplary dosing intervals, loading doses and maintenance doses.
  • Table 3B provides a variation of Table 3A allowing for dosage variability (indicated as “about”) of up to +/ ⁇ 3 mg/mL.
  • Table 3C appears below and provides a more extensive set of exemplary dosing intervals, loading doses and maintenance doses.
  • the top figure is the integer x in the interval QxD (e.g., 18 as the top figure in a cell indicates a dosing interval of Q18D or every 18 days)
  • the middle figure represents the (optional) loading dose (LD) in mg/kg
  • the bottom figure represents the maintenance dose (MD) in mg/kg.
  • the top cell in Table 3A indicates a dosing interval (QxD) of once every seven days, a loading dose (optional) of 25 mg per kg of patient body weight, and a maintenance dose of 20 mg per kg of patient body weight; while the cell furthest to the right on the top row of Table 3C indicates a dosing interval (QxD) of once every seven days, a loading dose (optional) of 30 mg per kg of patient body weight, and a maintenance dose of 15 mg per kg of patient body weight.
  • QxD dosing interval
  • TKIs selective tyrosine kinase inhibitors
  • lapatinib is administered at a dosage of 1000 to 1500 mg in 250 mg tablets taken once daily.
  • Lapatinib is often used in combination with another cancer medication, capecitabine, which is taken for 14 day periods with one week in between.
  • a modified dosing schedule is used wherein an increased dose of lapatinib is administered on days 1-5 of a 14 day cycle, said increased dose being a higher dose than the standard dose of 1000 to 1500 mg/day.
  • the higher lapatinib dose is between 2000 and 9000 mg/d.
  • higher lapatinib dose might be 2000, 2250, 3375, 3000, 3250, 3500, 3750, 4000, 4250, 4500, 4750, 5000, 5250, 5500, 5750, 6000, 6250, 6500, 6750, 7000, 7250, 7500, 7750, 8000, 8250, 8500, 8750, or 9000 mg/day, and so on.
  • a loading dose is given on day 1 of the 14-day cycle that is a higher dose than that given on subsequent days, the maintenance dose.
  • a loading dose given on day 1 of the 14 day cycle might be 7000 mg/day, followed by a maintenance dose of 3000 mg/day.
  • Non-limiting examples of loading dose and maintenance dose combinations are listed in Table 4 below.
  • MM-111 is administered as described in Example 15.
  • the treatment further comprises trastuzumab.
  • Trastuzumab is typically given with an initial loading dose followed by a maintenance dose.
  • trastuzumab may be dosed at a loading dose of 8 mg/kg followed by a maintenance dose of 6 mg/kg every three weeks.
  • MM-111 with cisplatin, capecitabine, and trastuzumab is done, for example, by the following method or minor variations thereof.
  • Cisplatin is administered on day 1 of each 21-day cycle by intravenous (i.v.) infusion over two hours, at a dose of 80 mg/m 2 .
  • Capecitabine is administered orally, twice daily, at a dose of 1000 mg/m 2 .
  • Up to 21-day cycles of cisplatin and capecitabine are administered.
  • Trastuzumab is administered i.v. at week 1 at an 8 mg/kg loading dose over 90 minutes, followed by a maintenance dose of 6 mg/kg every 21 days over 30-90 minutes.
  • MM-111 is administered as described in the above Examples. For example, MM-111 is administered i.v. over 90 minutes for the first dose and then weekly over 60 minutes thereafter.
  • MM-111 is administered i.v. at a 4 mg/kg loading dose on week 1 over 90 minutes, followed by a 2 mg/kg weekly maintenance dose thereafter.
  • Lapatinib is given by mouth either at 1000 mg daily doses or at the one of the dose regimens described in Example 13.
  • MM-111 is administered as described in the above Examples. For example, MM-111 is administered i.v. over 90 minutes for the first dose and then weekly over 60 minutes thereafter.
  • MM-111 is administered, for example, by the following method or minor variations thereof.
  • Patients are administered therapy on a 28-day treatment cycle.
  • Paclitaxel dosing begins on day 1 of cycle 1.
  • Paclitaxel is administered at 80 mg/m 2 weekly, as an i.v. infusion over 60 minutes.
  • Trastuzumab is administered at a 4 mg/kg loading dose on week 1, i.v. over 90 minutes, followed by a 2 mg/kg weekly maintenance dose thereafter.
  • MM-111 is administered as described in the above Examples. For example, MM-111 is administered i.v. over 90 minutes for the first dose and then weekly over 60 minutes thereafter.
  • MM-111 (at dosages described herein; see, e.g., Example 15) can be administered in combination with one or more additional agents to a patient in need thereof for the treatment of a cancer.
  • MM-111 can be administered in combination with MM-151 (oligoclonal anti-EGFR mixture), TDM-1 (ado-trastuzumab emtansine, an antibody-drug conjugate of the antibody trastuzumab linked to maytansine derivative (DM1)), an mTOR inhibitor (e.g., AZD8055, sirolimus, everolimus, temsirolimus, ridaforolimus, or the dual PI3K/mTOR inhibitor BEZ235), and combinations thereof.
  • MM-151 oligoclonal anti-EGFR mixture
  • TDM-1 anti-trastuzumab emtansine
  • DM1 maytansine derivative
  • an mTOR inhibitor e.g., AZD8055, sirolimus,
  • MM-151 is an oligoclonal therapeutic that is a mixture of three fully human monoclonal antibodies designed to bind to non-overlapping epitopes of the epidermal growth factor receptor, or EGFR (also known as ErbB1).
  • An oligoclonal therapeutic is a mixture of two or more distinct monoclonal antibodies.
  • MM-151 is disclosed, e.g., in copending PCT Application No. PCT/US12/45235.
  • MM-111 can be administered in the same dosage form as MM-151, TDM-1, and/or the mTOR inhibitor(s), or the agents can be administered in separate dosage forms.
  • MM-111 and one or more of MM-151, TDM-1, and/or the mTOR inhibitor(s) is administered to a patient for the treatment of a malignant tumor, e.g., an ErbB2-expressing or ErbB2 over-expressing tumor (e.g., HER or HER tumors).
  • a malignant tumor e.g., an ErbB2-expressing or ErbB2 over-expressing tumor (e.g., HER or HER tumors).
  • the tumor may be a melanoma, clear cell sarcoma, head and neck, endometrial, prostate, breast, ovarian, gastric, colon, colorectal, lung, bladder, pancreatic, salivary gland, liver, skin, brain or renal tumor.
  • MM-111 and MM-151 are co-administered to treat a solid tumor (e.g., an advanced refractory solid tumor) in a patient in need thereof.
  • a solid tumor e.g., an advanced refractory solid tumor
  • MM-111 combined with trastuzumab and the dual PI3K/mTOR inhibitor BEZ235 was assessed by the methods described above or with minor variations thereof in unstimulated and heregulin-stimulated NCI-N87 cells in vitro.
  • Cells were incubated for 2 hours with a dose range of MM-111, trastuzumab, or their combination, in the presence of BEZ235 as shown in FIG. 14 .
  • Cells were plated and either untreated (Control, open circle), treated with BEZ235 alone (open diamond), or treated with BEZ235 (5 nM FIG. 14 a, 20 nM FIG.
  • MM-111 combined with the mTOR inhibitor everolimus and trastuzumab was assessed in unstimulated ( FIG. 15 a ) and heregulin-stimulated ( FIG. 15 b ) BT474M3 cells in vitro.
  • Cells were plated and either untreated (Control, open circle), treated with everolimus alone (open diamond), or treated with everolimus (1 nM FIG. 15 a, 5 nM FIG. 15 b ) and either trastuzumab (closed circle), MM-111 (closed square), or the combination of MM-111 and trastuzumab (closed diamond).
  • Treatment with the triple combination of MM-111, trastuzumab, and everolimus resulted in a significant reduction in cell number compared to control cells in both the presence and absence of heregulin stimulation.
  • MM-111 at dosages described herein (see, e.g., Example 15), can be administered in combination with one or more MAP/ERK kinase (MEK)/phosphatidylinositol 3-kinase (PI3k)/AKT pathway inhibitors to a patient in need thereof for the treatment of a cancer.
  • MEK MAP/ERK kinase
  • PI3k phosphatidylinositol 3-kinase
  • MM-111 can be administered in the same dosage form as the MEK/PI3k/AKT pathway inhibitor(s) or these agents can be administered in separate dosage forms.
  • MM-111 and a MEK/PI3k/AKT pathway inhibitor is administered to a patient for the treatment of a malignant tumor, e.g., an ErbB2-expressing or ErbB2 over-expressing tumor (e.g., HER +++ or HER ++ tumors).
  • a malignant tumor e.g., an ErbB2-expressing or ErbB2 over-expressing tumor (e.g., HER +++ or HER ++ tumors).
  • the tumor may be a melanoma, clear cell sarcoma, head and neck, endometrial, prostate, breast, ovarian, gastric, colon, colorectal, lung, bladder, pancreatic, salivary gland, liver, skin, brain or renal tumor.
  • MM-111 and the MEK inhibitor trametinib were tested in a mouse xenograft model as described below:
  • BT474-M3 cells are transduced with full length HRG1- ⁇ 1 (Origene)-constructs (encoding PAC-PA-turboGFP for selection) by lentiviral infection.
  • a HRG overexpressing polyclonal cell line (BT474-M3-GFP-HRG) is established after puromycin selection and sorting for high GFP expressing cell population.
  • a control cell line (BT474-M3-GFP) is engineered to express EGFP in the same manner. Cells are maintained in culture in RPMI supplemented with 10% FBS, penicillin, streptomycin and puromycin.
  • MM-111 is produced in-house at Merrimack Pharmaceuticals and MK2206, trametinib, and UO126 (UO126-EtOH) are purchased from Selleckchem. 7-9 week old female NCRNU-mice (Taconic) are implanted subcutaneously with 0.72 mg 60-day release 17 ⁇ -estradiol pellets (Innovative Research of America).
  • mice 48 hours after implantation of the ⁇ -estradiol pellets, 15 ⁇ 106 BT474-M3-GFP-HRG cells were implanted in the second axillary mammary fat pad.
  • tumors reached an average volume of 450-500 mm3 (day 0)
  • mice were randomized into groups to receive control (phosphate buffered saline, intraperitoneally), MM-111 (48 mg/kg, intraperitoneally using phosphate buffered saline as vehicle), trametinib (3 mg/kg, by oral gavage in 0.5% methylcellulose/0.1% Tween®-80) or a combination of MM-111 and trametinib.
  • mice A set (3 per group) of mice are euthanized 4 h after the initial treatment. Tumors were excised and snap-frozen in liquid nitrogen for subsequent protein analysis. Another set of animals continued on their assigned treatment regimens for another 7 days receiving phosphate buffered saline control (intraperitoneally) on days 3 and 6, MM-111 (48 mg/kg, intraperitoneally) on days 3 and 6, trametinib (3 mg/kg, by oral gavage) on days 1, 2, 5 and 6. 24 hours after the last treatment, tumors were excised and snap-frozen in liquid nitrogen for subsequent protein analysis.
  • phosphate buffered saline control intraperitoneally
  • MM-111 48 mg/kg, intraperitoneally
  • trametinib 3 mg/kg, by oral gavage
  • Frozen tumor samples were pulverized and protein lysates were prepared in Tissue Extraction Reagent (Invitrogen) supplemented with Protease Inhibitor Cocktail Set III (Calbiochem, EMD) and HaltTM Phosphatase Inhibitor Cocktail (Thermo Scientific). Lysates were diluted in assay buffer (1% Bovine Serum Albumin in Tris buffered saline, pH 7.4, containing 0.1% Tween-20) to 1 mg/ml concentration and analyzed for phopsho- and total Erk1/2 using Luminex® immunosandwich assay on a FLEXMAP 3D instrument (Luminex).
  • assay buffer 1% Bovine Serum Albumin in Tris buffered saline, pH 7.4, containing 0.1% Tween-20
  • MM-111 at dosages described herein (see, e.g., Example 15), can be administered in combination with one or more additional ErbB2-targeted therapeutic agents, such as trastuzumab, T-DM1, pertuzumab, etc.
  • additional ErbB2-targeted therapeutic agents such as trastuzumab, T-DM1, pertuzumab, etc.
  • cells with ( FIG. 17B ) and without ( FIG. 17A ) heregulin (HRG) were treated with a dose escalation of T-DM1 and then treated with MM-111 as follows.
  • NCI-N87 cells are transduced with full length HRG1- ⁇ 1 (Origene)-constructs (encoding PAC-PA-turboGFP for selection) by lentiviral infection.
  • a HRG overexpressing polyclonal cell line (NCI-N87-GFP-HRG) is established after puromycin selection and sorting for high GFP expressing cell population.
  • a control cell line (NCI-N87-GFP) is engineered to express EGFP in the same manner. Cells are maintained in culture in RPMI supplemented with 10% fetal bovine serum, penicillin, streptomycin and puromycin (1 ⁇ g/ml).
  • MM-111 is manufactured at Merrimack Pharmaceuticals. Recombinant human NRG-1- ⁇ 1 EGF domain is from R&D Systems. T-DM1 (KADCYLA, Genentech) is obtained from pharmacy. CellTiter-Glo® Luminescent Cell Viability Assay is from Promega.
  • NCI-N87 or BT-474-M3 cells were seeded on 384-well flat clear bottom polystyrene microplates in 20 ⁇ l of culture media (RPMI supplemented with 10% fetal bovine serum and penicillin and streptomycin). Cells were allowed to adhere 16-20 hours at +37° C., 5% CO 2 . Cells were stimulated for 4 hours by adding 5 ⁇ l of culture media containing recombinant human NRG-1- ⁇ 1 EGF domain (HRG1) bringing the final HRG1 stimulation concentration to 0, 0.313, 0.625, 1.25, 2.5, 5 or 10 nM.
  • HRG1 human NRG-1- ⁇ 1 EGF domain
  • T-DM1 was added in 5 ⁇ l of culture media with or without MM-111 bringing the T-DM1 concentration to 0, 0.001524, 0.004572, 0.013717, 0.041152, 0.123457, 0.37037, 1.11111, 3.333333 or 10 ⁇ g/ml and the MM-111 concentration to 0 or 1 ⁇ M. All conditions were assayed in biological quadruplicates. Cells were incubated for 72 h at +37° C., 5% CO 2 .
  • Cell viability was determined by CellTiter-Glo® Luminescent Cell Viability Assay by adding 20 ⁇ l of reconstituted CellTiter-Glo reagent per well, incubating plated for 10 minutes at RT in an orbital shaker and reading the luminescence signal using BiotekTM Synergy H1 Hybrid Reader.
  • NCI-N87-GFP or NCI-N87-GFP-HRG1 cells were seeded on 384-well flat clear bottom polystyrene microplates in 20 ⁇ l of culture media (RPMI supplemented with 10% fetal bovine serum and penicillin and streptomycin). Cells were allowed to adhere 16-20 hours at +37° C., 5% CO2.
  • T-DM1 was added in 5 ⁇ l of culture media with or without MM-111 bringing the T-DM1 concentration to 0, 0.001524, 0.004572, 0.013717, 0.041152, 0.123457, 0.37037, 1.11111, 3.333333 or 10 ⁇ g/ml and the MM-111 concentration to 0 or 1 ⁇ M.
  • Cells were incubated for 72 h at +37° C., 5% CO 2 .
  • Cell viability was determined by CellTiter-Glo® Luminescent Cell Viability Assay by adding 20 ul of reconstituted CellTiter-Glo reagent per well, incubating plated for 10 minutes at RT in an orbital shaker and reading the luminescence signal using Biotek Synergy H1 Hybrid Reader.
  • NCI-N87 cells and BT-474-M3 cells were prepared as described above and treated with a dose range of both T-DM1 and heregulin in the presence and absence of MM-111 (3D graphs, FIG. 18 ).
  • Cell viability was tested and numbers are given as % of control and summarized in Tables 1-4 below.
  • FIG. 18 while exogenous HRG decreases the activity of T-DM1, co-treatment of the cells with a combination of MM-111 and T-DM1 greatly reduced cell viability in the presence of endogenous heregulin in both NCI-N87 ( FIG. 18B ) and BT-474-M3 cells ( FIG. 18D ).
  • the Table and Appendix below describe effective an anti-estrogen agents, receptor tyrosine kinase inhibitors; MEK/PI3 kinase/AKT inhibitors, and mTOR inhibitors that can be used in the methods and compositions of the invention.
  • the bispecific anti-ErbB2/anti-ErbB3 antibody co-administered in combination with an agent selected from i) an effective amount of an anti-estrogen agent; ii) an effective amount of a receptor tyrosine kinase inhibitor; iii) a MEK/PI3 kinase/AKT inhibitor; iv) MM-151; v) an mTOR inhibitor; and/or vi) trastuzumab or T-DM1, and combinations thereof, can be further co-administered with at least a third antineoplastic agent selected from any of those disclosed in the Table and Appendix below.
  • Exemplary antineoplastic agents for treatment of breast cancer in combination with a bispecific anti-ErbB2/anti-ErbB3 antibody Exemplary Agent Therapeutic Class (Generic/Tradename) Exemplary Dose Mitotic Inhibitors paclitaxel (TAXOL ®; ABRAXANE ®) 175 mg/m 2 docetaxel (TAXOTERE ®) 60-100 mg/m 2 Topoisomerase Inhibitors camptothecin topotecan hydrochloride (HYCAMTIN etoposide (EPOSIN ®) Alkylating Agents cyclophosphamide (CYTOXAN ®) 600 mg/m 2 Platinum-Based Agents Cisplatin 20-100 mg/m 2 carboplatin (PARAPLATIN ®) 300 mg/m 2 nedaplatin (AQUPLA ®) oxaliplatin (ELOXATIN ®) 65-85 mg/m 2 satraplatin (SPERA ®) triplatin t
  • RNR Ribonucleotide Reductase Inhibitor
  • Ranimustine Satraplatin Semustine Streptozocin Temozolomide Treosulfan Triaziquone Triethylene Melamine ThioTEPA Bedford, Abraxis, Teva Triplatin tetranitrate Trofosfamide Uramustine Antimetabolites 5-azacytidine Flourouracil (5-FU)/Capecitabine 6-mercaptopurine (Mercaptopurine, 6-MP) 6-Thioguanine (6-TG) Purinethol ® Teva Cytosine Arabinoside (Cytarabine, Thioguanine ® GlaxoSmithKline Ara-C) Azathioprine Azasan ® AAIPHARMA LLC Capecitabine XELODA ® HLR (Roche) Cladribine (2-CdA, 2- Leustatin ® Ortho Biotech chlorodeoxyadenosine) 5-Trifluoromethyl-2′-deoxyuridine Fludar
  • Vandetanib (AZD647) Zactima AstraZeneca Vatalanib; PTK-787 Novartis; Bayer Schering Pharma XL184, NSC718781 Exelixis, GSK Microtubule-Targeting Agents Colchicine Docetaxel Taxotere ® Sanofi-Aventis US Ixabepilone IXEMPRA TM Bristol-Myers Squibb Larotaxel Sanofi-aventis Ortataxel Spectrum Pharmaceuticals Nanoparticle paclitaxel (ABI-007) Abraxane ® Abraxis BioScience, Inc.
  • Paclitaxel Taxol ® Bristol-Myers Squibb Tesetaxel Genta Vinblastine sulfate Velban ® Lilly Vincristine Oncovin ® Lilly Vindesine sulphate Eldisine ® Lilly Vinflunine Pierre Fabre Vinorelbine tartrate Navelbine ® Pierre Fabre mTOR Inhibitors
  • Deforolimus AP23573, MK 8669, ARIAD Pharmaceuticals, Inc Ridaforolimus
  • Everolimus RAD001, RAD001C
  • Certican Afinitor Novartis Sirolimus (Rapamycin) Rapamune ® Wyeth Pharma Temsirolimus (CCI-779)
  • Irinotecan HCL Camptosar ® Pharmacia & Upjohn Mitoxantrone HCL Novantrone EMD Serono Rubitecan Teniposide (VM-26) Vumon ® Bristol-Myers Squibb Topotecan HCL Hycamtin ® GlaxoSmithKline Chemotherapeutic Agents Adriamycin, 5-Fluorouracil, Cytoxin, Bleomycin, Mitomycin C, Daunomycin, Carminomycin, Aminopterin, Dactinomycin, Mitomycins, Esperamicins Clofarabine, Mercaptopurine, Pentostatin, Thioguanine, Cytarabine, Decitabine, Floxuridine, Gemcitabine (Gemzar), Enocitabine, Sapacitabine Hormonal Therapies Abarelix Plenaxis TM Amgen Abiraterone acetate CB7630 BTG plc Afimoxifene TamoGel Ascend Therapeutics,
  • PKT Protein Kinase B
  • AKT Inhibitor Triciribine AKT DECIPHERA Deciphera Pharmaceuticals, LLC Perifosine (KRX0401, D-21266) Keryx Biopharmaceuticals, Inc., AEtenta Zentaris, Inc. Perifosine with Docetaxel Keryx Biopharmaceuticals, Inc., AEtenta Zentaris, Inc. Perifosine with Gemcitabine AEtenta Zentaris, Inc. Perifosine with Paclitaxel Keryx Biopharmaceuticals, Inc, AEtenta Zentaris, Inc. Protein Kinase-B inhibitor DEVELOGEN DeveloGen AG PX316 Oncothyreon, Inc. RX0183 Rexahn Pharmaceuticals, Inc.
  • Enzastaurin HCL (LY317615) Enzastaurin Eli Lilly LY294002/Wortmannin Eli Lilly PI3K Inhibitors SEMAFORE Semafore Pharmaceuticals PX866 Oncothyreon, Inc. SF1126 Semafore Pharmaceuticals VMD-8000 VM Discovery, Inc. XL147 Exelixis, Inc. XL147 with XL647 Exelixis, Inc. XL765 Exelixis, Inc.

Abstract

Disclosed are methods and compositions for inhibiting the growth of a tumor (e.g., a malignant 5 tumor) in a subject. In particular, combination therapies for treating a tumor in a subject by coadministering an agent selected from i) an effective amount of an anti-estrogen agent; ii) an effective amount of a receptor tyrosine kinase inhibitor; iii) an effective amount of a MEK/PI3 kinase/AKT inhibitor; iv) an effective amount of MM-151; v) an effective amount of an mTOR inhibitor; and/or vi) an effective amount of trastuzumab or T-DM 1, and/or combinations thereof; and an effective amount of a 10 bispecific anti-ErbB2/anti-ErbB3 antibody.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims benefit of U.S. Provisional Application No. 61/695,242, filed Aug. 30, 2012, which is hereby incorporated by reference in its entirety.
  • FIELD OF THE INVENTION
  • The various aspects of the invention disclosed herein relate to methods and compositions for the treatment of cancers.
  • BACKGROUND OF THE INVENTION
  • Approximately 75% of breast cancers are estrogen receptor (ER) positive. Other cancers are also ER positive (ER+). Estrogen receptors mediate intracellular signaling that can increase the frequency of cell division and drive tumor growth. Although anti-endocrine therapies such as tamoxifen, fulvestrant, and letrozole have demonstrated significant efficacy in treating ER+ breast cancer patients, intrinsic or acquired resistance to such therapies has limited their success.
  • The prevalence of amplification of the human epidermal growth factor receptor 2 (HER2, or ErbB2) in breast cancer and other cancers has resulted in the research and development of drugs that have ErbB2 as a therapeutic target. Although both the anti-ErbB2 monoclonal antibody trastuzumab, the anti-ErbB2 monoclonal antibody drug conjugate T-DM1 (ado-trastuzumab emtansine) and the ErbB1/ErbB2 dual receptor tyrosine kinase inhibitor lapatinib have met with success in the clinic, many patients fail to benefit from these drugs. Additionally, the majority of patients with tumors that initially respond will eventually recrudesce after extended treatment using these therapies.
  • ErbB2 can function as a homodimer or as a heterodimer with related epidermal growth factor-type receptors such as ErbB3. The ErbB2/ErbB3 heterodimer is the most potent ErbB receptor pairing with respect to strength of interaction, impact on receptor tyrosine phosphorylation, and effects on downstream intracellular signaling through mitogen activated protein kinase and phosphoinositide-3 kinase pathways. Heregulin is the primary ligand for ErbB3, and activates signaling by ErbB2/ErbB3 heterodimers. Such signaling is believed to drive proliferation of cancer cells. Current ErbB2-targeted therapies do not effectively inhibit heregulin activated signaling. MM-111 is a bispecific anti-ErbB2/anti-ErbB3 antibody that abrogates heregulin binding to ErbB2/ErbB3 and inhibits heregulin activation of ErbB2/ErbB3 without significantly affecting ErbB2 biological activity. In preclinical models of HER-2+ gastric, breast, ovarian and lung cancers, MM-111 inhibits ErbB3 phosphorylation, cell cycle progression, and tumor growth.
  • Thus, a need exists for therapies and therapeutic strategies providing improved inhibition of ErbB3 activation (e.g., ligand-induced activation) as well as for therapies and therapeutic strategies providing improved inhibition of estrogen receptor signaling activity or of ErB1 and ErbB2 receptor signaling activity, as well as inhibition of the downstream intracellular signaling pathways such as the mitogen activated protein kinase and phosphoinositide-3 kinase pathways.
  • In the treatment of cancers, the co-administration of pluralities of anti-cancer drugs (combination therapy) often provides better treatment outcomes than monotherapy. Such outcomes can be subadditive, additive, or superadditive. That is to say that the combined effects of two anti-cancer drugs, each of which provides a quantifiable degree of benefit, can be less than, equal to, or greater than the sum of the benefits of each drug. For example, two drug, each of which when used alone to treat a lethal cancer provides an average one year extension of progression free survival, could together provide a <24 month extension (e.g., an 18 month extension), about a 24 month extension, or a >24 month extension (e.g., a 30 month extension) of progression free survival. Typically, combination therapies for cancer treatment provide significantly subadditive outcomes. Outcomes that are near additive, additive, or superadditive are most desirable, but only occur rarely. In addition, many drugs are known to alter the bioavailability, or otherwise affect the safety profile of other drugs when both drugs are co-administered. As new drugs are first used in combination therapies, unforeseen, hazardous drug-drug interactions may be observed that result in drug-drug interaction-mediated toxicity in the patient.
  • Thus approaches for safely administering combination therapies comprising administration of ErbB2/ErbB3 heterodimer-targeted agents for cancer treatment, and especially combinations that yield near-additive, additive, or superadditive outcomes are needed.
  • SUMMARY OF THE INVENTION
  • Provided herein are methods and compositions effective for the inhibition of ErbB3 activation and also effective for the inhibition of estrogen receptor activation. Also provided are methods and compositions effective for the inhibition of ErbB3 activation and also effective for the inhibition of ErB1 and/or ErbB2 activation. These methods and compositions are useful for the treatment of tumors, e.g., malignant tumors, as well as for the treatment of other cancers.
  • In a first embodiment, a method of treating a subject with a malignant tumor is provided, where the tumor is an ErbB2 expressing or ErbB2 over-expressing tumor (e.g., HER2+++ or HER2++ tumors) and the tumor may be a melanoma, clear cell sarcoma, head and neck, endometrial, prostate, breast, ovarian, esophageal, gastric, gastro-esophageal, colon, colorectal, lung, bladder, pancreatic, salivary gland, liver, skin, brain or renal tumor. The method comprises co-administering to the subject an effective amount an agent selected from i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) a MEK inhibitor, (e.g., selumetinib, trametinib, PD0325901, UO126), iv) a PI3 kinase inhibitor (e.g., BEZ235, BKM120, GDC0941), v) an AKT inhibitor (e.g., MK-2206, triciribine), vi) an mTOR inhibitor (e.g., BEZ235, AZD8055, everolimus, temsirolimus, sirolimus, ridaforolimus), vii) trastuzumab, viii) T-DM1, ix) capecitabine, x) cisplatin, and xi) MM-151; and combinations thereof, in combination with an effective amount of an anti-ErbB3 agent, e.g., a bispecific anti-ErbB2/anti-ErbB3 antibody (e.g., an antibody comprising the amino acid sequence set forth in SEQ ID NO:1). Additional agents for use in combination with the anti-ErbB3 agent, e.g., a bispecific anti-ErbB2/anti-ErbB3 antibody, are described in the Appendix.
  • In one aspect, the combination of the bispecific anti-ErbB2/anti-ErbB3 antibody and either the effective amount of an anti-estrogen agent or the effective amount of the receptor tyrosine kinase inhibitor, and optionally the effective amount of trastuzumab or T-DM1, is characterized as follows: when a first tissue culture medium is prepared comprising the bispecific anti-ErbB2/anti-ErbB3 antibody (e.g., the antibody comprising the amino acid sequence set forth in SEQ ID NO:1) at a first concentration and either the anti-estrogen agent at a second concentration or the receptor tyrosine kinase inhibitor (e.g., lapatinib) at a third concentration (wherein each concentration is the same or different as each other concentration), and the medium is contacted with cancer cells of a cell line in a cell culture, cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or the percentage of cells in the culture that are apoptotic is increased. In certain aspects, cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or the percentage of cells in the culture that are apoptotic is increased to a greater degree than cell growth, or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or percentage of cells in the culture that are apoptotic is increased, to a lesser degree when cancer cells of the cell line in a cell culture are contacted with each of a second medium that is essentially the same as the first medium except that it does not comprise a bispecific anti-ErbB2/anti-ErbB3 antibody, and a third medium that is essentially the same as the first medium except that it does not comprise any anti-estrogen agent and it does not comprise any receptor tyrosine kinase inhibitor.
  • In another aspect, all effective amounts are either mouse effective amounts or human effective amounts. In another aspect, all effective amounts are mouse effective amounts and the combination of the bispecific anti-ErbB2/anti-ErbB3 antibody (optionally the antibody comprising the amino acid sequence set forth in SEQ ID NO:1) and either the effective amount of an anti-estrogen agent or the effective amount of the receptor tyrosine kinase inhibitor, is characterized as follows: when co-administered to BT474-M3 xenograft tumor bearing mice with a tumor of a measured volume, the combination is more effective at inhibiting tumor volume increase after 32 days of co-administration than is the mouse effective amount of the bispecific anti-ErbB2/anti-ErbB3 antibody administration without the co-administration of either the effective amount of an anti-estrogen agent or the effective amount of the receptor tyrosine kinase inhibitor. In another aspect, a mouse effective amount of trastuzumab or T-DM1 is co-administered with the bispecific anti-ErbB2/anti-ErbB3 antibody.
  • In a second embodiment, a bispecific anti-ErbB2/anti-ErbB3 antibody (optionally the antibody comprising SEQ ID NO:1) is provided for use in combination therapy of a cancer (optionally a melanoma, clear cell sarcoma, head and neck, endometrial, esophageal, gastro-esophageal junction, prostate, breast, ovarian, gastric, colon, colorectal, lung, bladder, pancreatic, salivary gland, liver, skin, brain or renal tumor), where the combination therapy comprises concomitant use of an effective amount an agent selected from i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor, vi) an mTOR inhibitor, vii) trastuzumab, viii) T-DM1, ix) capecitabine, x) cisplatin, and xi) MM-151; and combinations thereof.
  • In a third embodiment, an aqueous solution is provided comprising a bispecific anti-ErbB2/anti-ErbB3 antibody (optionally an antibody comprising the amino acid sequence set forth in SEQ ID NO:1) at a first concentration and an agent selected from i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) an effective amount of a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor, vi) an mTOR inhibitor, vii) trastuzumab, viii) T-DM1, ix) capecitabine, x) cisplatin, and xi) MM-151; and combinations thereof, at a second concentration. In certain aspects, when a first tissue culture medium is prepared comprising the bispecific anti-ErbB2/anti-ErbB3 antibody at the first concentration and the agent at the second concentration, and the medium is contacted with cancer cells of a cell line in a cell culture, cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or percentage of cells in the culture that are apoptotic is increased. In certain aspects, cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or the percentage of cells in the culture that are apoptotic is increased to a lesser degree when cells of the cell line in a cell culture are contacted with a second tissue culture medium that is essentially the same as the first medium of except that it does not comprise the agent(s). In another aspect, cell growth or cell proliferation or production of pErbB3 or production of pAKT in the cells is inhibited, or the percentage of cells in the culture that are apoptotic is increased to a lesser degree when cells of the cell line in a cell culture are contacted with a third tissue culture medium that is essentially the same as the first medium of except that it does not comprise any bispecific anti-ErbB2/anti-ErbB3 antibody.
  • In another aspect, the aqueous solution is blood plasma in a subject, and the subject does not experience a toxicity that is sufficiently harmful to require a change in a therapy being administered to the subject, which toxicity is mediated by a drug-drug interaction in the subject between the bispecific anti-ErbB2/anti-ErbB3 antibody and the anti-estrogen agent or the receptor tyrosine kinase inhibitor.
  • In another aspect, the aqueous solution further comprises trastuzumab or T-DM1 at a third concentration.
  • In another aspect, the method, combination therapy, or aqueous solution does not comprise an aromatase inhibitor or an estrogen receptor antagonist. In one embodiment the method, combination therapy, or aqueous solution comprises nab-paclitaxel.
  • In each embodiment and aspect thereof above, the anti-estrogen agent may be an estrogen receptor antagonist (e.g., fulvestrant or tamoxifen) or an aromatase inhibitor (e.g., wherein the aromatase inhibitor is letrozole, exemestane, anastrozole, aminoglutethimide, testolactone, vorozole, formestane, or fadrozole. Preferably the aromatase inhibitor is letrozole. Also in each embodiment and aspect thereof above, the receptor tyrosine kinase inhibitor is erlotinib, afatinib, dasatinib, gefitinib, imatinib, pazopinib, lapatinib, sunitinib, nilotinib or sorafenib. Preferably the receptor tyrosine kinase inhibitor is lapatinib. Also in each embodiment and aspect thereof above, the bispecific anti ErbB2/anti-ErbB3 antibody is the A5-HSA-ML3.9, ML3.9-HSA-A5, A5-HSA-B1D2, B1D2-HSA-A5, B12-HSA-B1D2, B1D2-HSA-B12, A5-HSA-F5B6H2, F5B6H2-HSA-A5, H3-HSA-F5B6H2, F5B6H2-HSA-H3, F4-HSA-F5B6H2, F5B6H2-HSA-F4, B1D2-HSA-H3, H3-HSA-B1D2, or the antibody comprising the amino acid sequence set forth in SEQ ID NO:1. Each embodiment and aspect thereof above may also further comprise use of capecitabine and/or cisplatin.
  • In each embodiment and aspect thereof above, one or more of a)-i) that follow may optionally apply: a) the cell line is BT474-M3; b) the culture is a spheroid culture, c) paclitaxel or another taxane or another chemotherapeutic drug is co-administered, optionally in accordance with the manufacturer's directions, d) the agent i)-xi) is administered in accordance with the manufacturer's directions, e) the trastuzumab or T-DM1 is administered in accordance with the manufacturer's directions, f) the co-administration of the bispecific anti-ErbB2/anti-ErbB3 antibody with the agent g)-vi) produces an about additive or a superadditive effect, h) the bispecific anti-ErbB2/anti-ErbB3 antibody is the antibody comprising SEQ ID NO:1 and is administered in accordance with any of the regimens (e.g., modes, dosages, dosing intervals, loading and maintenance doses and dosing schemes) described in Examples 12 and 13, below, i) the lapatinib is administered in accordance with any of the regimens (e.g., modes, dosages, dosing intervals, loading and maintenance doses and dosing schemes) described in Example 16, below.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph showing that the combination of MM-111 and tamoxifen inhibits tumor growth in vivo better than either MM-111 or tamoxifen does alone. The x-axis shows time post tumor implant in days and the y-axis shows tumor volume in mm3. Mice were treated with inhibitors beginning on day 7 post BT474-M3 cell implant.
  • FIG. 2 is seven graphs showing that MM-111 combines positively with anti-estrogen drugs in inhibiting estrogen-stimulated spheroid growth in vitro. FIG. 2 a shows the effect of MM-111, tamoxifen (4-hydroxytamoxifen or 4OHT), or MM-111 and tamoxifen on in vitro spheroid growth. FIG. 2 b shows the effect of trastuzumab, tamoxifen, or trastuzumab and tamoxifen. FIG. 2 c shows the effect of MM-111, fulvestrant (FVT), or MM-111 and fulvestrant. FIG. 2 d shows the effect of trastuzumab, fulvestrant, or trastuzumab and fulvestrant. FIG. 2 e shows the effect of MM-111, trastuzumab, or MM-111 and trastuzumab. FIG. 2 f shows the effect of MM-111, trastuzumab, and tamoxifen combined compared to that of any of the double combinations. FIG. 2 g shows the effect of MM-111, trastuzumab, and fulvestrant combined compared to that of any of the double combinations. The x-axes are a log scale of each drug concentration for each experimental condition in nM and the y axis is spheroid size as % of control spheroid size.
  • FIG. 3 is seven graphs showing that MM-111 combines positively with anti-estrogen drugs in inhibiting heregulin (HRG)-stimulated spheroid growth in vitro. FIG. 3 a shows the effect of MM-111, tamoxifen (4-hydroxytamoxifen or 4OHT), or MM-111 and tamoxifen. FIG. 3 b shows the effect of trastuzumab, tamoxifen, or trastuzumab and tamoxifen. FIG. 3 c shows the effect of MM-111, fulvestrant (FVT), or MM-111 and fulvestrant. FIG. 3 d shows the effect of trastuzumab, fulvestrant, or trastuzumab and fulvestrant. FIG. 3 e shows the effect of MM-111, trastuzumab, or MM-111 and trastuzumab. FIG. 3 f shows the effect of MM-111, trastuzumab, and tamoxifen combined compared to that of any of the double combinations. FIG. 3 g shows the effect of MM-111, trastuzumab, and fulvestrant combined compared to that of any of the double combinations. The x-axes are a log scale of each drug concentration for each experimental condition in nM and the y axis is spheroid size as % of control spheroid size.
  • FIG. 4 is seven graphs showing that MM-111 combines positively with anti-estrogen drugs in inhibiting dual ligand (estrogen and heregulin)-stimulated spheroid growth in vitro. FIG. 4 a shows the effect of MM-111, tamoxifen, or MM-111 and tamoxifen. FIG. 4 b shows the effect of trastuzumab, tamoxifen, or trastuzumab and tamoxifen. FIG. 4 c shows the effect of MM-111, fulvestrant (FVT), or MM-111 and fulvestrant. FIG. 4 d shows the effect of trastuzumab, fulvestrant, or trastuzumab and fulvestrant. FIG. 4 e shows the effect of MM-111, trastuzumab, or MM-111 and trastuzumab. FIG. 4 f shows the effect of MM-111, trastuzumab, and tamoxifen combined compared to that of any of the double combinations. FIG. 4 g shows the effect of MM-111, trastuzumab, and fulvestrant combined compared to that of any of the double combinations. The x-axes are a log scale of each drug concentration for each experimental condition in nM and the y axis is spheroid size as % of control spheroid size.
  • FIG. 5 is a graph summarizing the effect of MM-111, trastuzumab, and tamoxifen combined compared to that of any of the double combinations or MM-111, trastuzumab, and fulvestrant combined compared to that of any of the double combinations at inhibiting single ligand (estrogen or heregulin) or dual-ligand (estrogen and heregulin)-stimulated spheroid growth in vitro. The y-axis is % inhibition of spheroid size normalized to stimulated control.
  • FIG. 6 is a graph showing that the combination of MM-111 and lapatinib inhibits tumor growth in vivo. The x-axis shows the time post tumor implant in days and the y-axis shows tumor volume in mm3. Mice were treated with inhibitors on day 7 post tumor implant.
  • FIG. 7 evaluates the ability of lapatinib to inhibit ErbB3 and AKT activation in heregulin-stimulated cells. 7 a is a graph comparing computer-generated dose-response curves to experimental results in heregulin-stimulated BT474-M3 cells. 7 b is a graph showing lapatinib inhibition (IC50) of ErbB3 and AKT activation in heregulin-stimulated and unstimulated cells following a 1-hour incubation with inhibitor.
  • FIG. 8 is a series of graphs showing MM-111 or lapatinib inhibition of ErbB3 (8 a) or AKT (8 b) activation in heregulin-stimulated cells incubated with inhibitor for 15 minutes, 1 hour, 4 hours, and 24 hours. FIG. 8 c shows a comparison of IC50 for MM-111 and lapatinib at 1 hour and 24 hours for both BT474M3 cells and ZR75-30 cells.
  • FIG. 9 is a graph showing the effect of MM-111 and lapatinib combination treatment on AKT activation in heregulin-stimulated BT474-M3 cells.
  • FIG. 10 is a graph showing the effect of lapatinib on cell viability as a measure of proliferation of unstimulated and heregulin-stimulated BT474-M3 cells.
  • FIG. 11 is a graph showing the effect of MM-111, lapatinib, or the combination on BT474-M3 cell apoptosis. The number of dead cells, cells in late apoptosis, early apoptosis, and live cells was quantitated.
  • FIG. 12 is three graphs showing that MM-111 combines positively with anti-estrogen drugs and lapatinib in inhibiting dual ligand (estrogen (E2) and heregulin (HRG))-stimulated spheroid growth in vitro. FIG. 12 a shows the effect of lapatinib alone or the combination of lapatinib and fulvestrant (FVT). FIG. 12 b shows the effect of lapatinib alone or the combination of lapatinib and MM-111. FIG. 12 c shows the effect of lapatinib alone, the combination of MM-111 and fulvestrant, or the triple combination of MM-111, FVT, and lapatinib. Lapatinib is given in 3.3, 10, or 30 nM doses. The x-axes are a log scale of each of MM-111 and/or FVT concentration in nM and the y axis is spheroid size as % of control (FBS alone) spheroid size.
  • FIG. 13 is four graphs showing that MM-111 combines positively with the aromatase inhibitor letrozole and the tyrosine kinase inhibitor lapatinib in heregulin (HRG) and androstenedione (A4)-stimulated BT474-M3-Aro cells that stably express human aromatase, which converts androstenedione to estrogen. FIG. 13 a shows the effect of letrozole, MM-111, or the combination of letrozole and MM-111. FIG. 13 b shows the effect of lapatinib, MM-111 or the combination of lapatinib and MM-111. FIG. 13 c shows the effect of lapatinib, letrozole, or the combination of lapatinib and letrozole. FIG. 13 d shows the effect of the dual combinations of MM-111 and letrozole, MM-111 and lapatinib, lapatinib and letrozole, and the triple combination of MM-111, lapatinib and letrozole. The x-axes are a log scale of MM-111 concentration in nM. The drug concentrations are a ratio of 10:20:1 MM-111 to letrozole to lapatinib. The y axis is spheroid size as % of control spheroid size.
  • FIG. 14 is two graphs showing that MM-111 combines positively with the PI3K/mTOR inhibitor BEZ235 and trastuzumab in unstimulated (FIG. 14 a) and heregulin-stimulated (FIG. 14 b) NCI-N87 cells in vitro. Cells were plated and either untreated (Control, open circle), treated with BEZ235 alone (open diamond), or treated with BEZ235 (5 nM FIG. 14 a, 20 nM FIG. 14 b) and either trastuzumab (closed circle), MM-111 (closed square), or the combination of MM-111 and trastuzumab (closed diamond). Closed circles (trastuzumab) may appear as squares or rectangles on trastuzumab curves when they comprise error bars. The x-axes are a log scale of the concentrations of each of MM-111 and/or trastuzumab.
  • FIG. 15 is two graphs showing that MM-111 combines positively with the mTOR inhibitor everolimus and trastuzumab in unstimulated (FIG. 15 a) and heregulin-stimulated (FIG. 15 b) NCI-N87 cells in vitro. Cells were plated and either untreated (Control, open circle), treated with everolimus alone (open diamond), or treated with everolimus (1 nM FIG. 15 a, 5 nM FIG. 15 b) and either trastuzumab (closed circle), MM-111 (closed square), or the combination of MM-111 and trastuzumab (closed diamond). Closed circles may appear as squares or rectangles on trastuzumab curves when they comprise error bars. The x-axes are a log scale of the concentrations of each of MM-111 and/or trastuzumab.
  • FIG. 16 is two graphs showing that MM-111 combines positively with the MEK inhibitor GSK1120212 (trametinib) in a mouse xenograft study. Phospho-ERK was measured as a function of ERK activity via a Luminex® immunosandwich assay at 4 hours after the initial treatment (FIG. 16A) and 24 hours after the initial treatment (FIG. 16B) as a function of ERK activity via a Luminex immunosandwich assay. The x-axes show type of treatment and the y-axes show mean fluorescent intensity.
  • FIG. 17 is two graphs showing that MM-111 combines positively with T-DM1 (ado-trastuzumab emtansine) in both the absence (FIG. 17A) and presence (FIG. 17B) of endogenous heregulin. The x-axes show a log scale of T-DM1 concentration and the y-axes show viability as % of control.
  • FIG. 18 is a series of 3D graphs showing that MM-111 combines positively with TDM-1 (ado-trastuzumab emtansine) in the presence of exogenous heregulin. NCI-N87 (FIGS. 18A and 18B) or BT-474-M3 (FIGS. 18C and 18D) cells were incubated with a dose range of both T-DM1 and heregulin, either without MM-111 (FIGS. 18A and 18C) or with MM-111 (FIGS. 18B and 18D). The x-axes are the concentration of T-DM1, the y-axes are cell viability as % of untreated cells, and the z-axes are heregulin concentration. The data presented in these graphs is also set forth below in Tables 1-4.
  • DETAILED DESCRIPTION
  • As herein provided, bispecific anti-ErbB2/anti-ErbB3 antibodies (e.g., MM-111) are co-administered with one or more additional therapeutic agents (e.g. an aromatase inhibitor or tyrosine kinase inhibitor), to provide effective treatment to human patients having a cancer. Such co-administrations beneficially have an additive or superadditive effect on suppressing tumor cell growth, which effect on suppressing tumor cell growth is measured, e.g., in a mouse xenograft model e.g., using BT-474 or NCI-N87 cells.
  • The term “anti-ErbB3 agent” refers to any therapeutic agent that binds to ErbB3 or binds to an ErbB3-specific ligand or blocks the expression of ErbB3, and thereby inhibits the activity of cellular signaling mediated by ErbB3. Non-limiting examples of types of anti-ErbB3 agents include antibodies, bispecific antibodies, ligand analogs, soluble forms of ErbB3 or the ErbB3 ectodomain, ErbB3 specific RNAi molecules, and similar biologic agents.
  • The term “antibody” describes a polypeptide comprising at least one antibody-derived antigen binding site (e.g., VH/VL region or Fv, or complementarity determining region—CDR) that specifically binds to a specific antigen, e.g., ErbB3. “Antibodies” include whole antibodies and any antigen binding fragment, e.g., Fab or Fv, or a single chain fragment (e.g., scFv), as well as bispecific antibodies and similar engineered variants, human antibodies, humanized antibodies, chimeric antibodies Fabs, Fab′2s, ScFvs, SMIPs, Affibodies®, nanobodies, or a domain antibodies, and may be of any of the following isotypes: IgG1, IgG2, IgG3, IgG4, IgM, IgA1, IgA2, IgAsec, IgD, and IgE. The antibody may be a naturally occurring antibody or may be an antibody that has been altered (e.g., by mutation, deletion, substitution, conjugation to a non-antibody moiety). For example, an antibody may include one or more variant amino acids (compared to a naturally occurring antibody) which change a property (e.g., a functional property) of the antibody. For example, numerous such alterations are known in the art which affect, e.g., half-life, effector function, and/or immune responses to the antibody in a patient. The term “antibody” thus includes whole antibodies and any antigen binding fragment (i.e., “antigen-binding portion,” e.g., Fabs) or single chains thereof (e.g., scFvs) as well as bispecific antibodies and similar engineered variants, provided that they retain the binding specificity of an antibody.
  • An “anti-ErbB3 antibody” is an antibody that immunospecifically binds to the ectodomain of ErbB3 and an “anti-ErbB2 antibody” is an antibody that immunospecifically binds to the ectodomain of ErbB2. The antibody may be an isolated antibody. Such binding to ErbB3 or ErB2 exhibits a Kd with a value of no greater than 50 nM as measured by a surface plasmon resonance assay or a cell binding assay. Exemplary anti-ErbB3 antibodies inhibit EGF-like ligand mediated phosphorylation of ErbB3, e.g., anti-ErbB2 antibodies that inhibit the binding of heregulin to ErbB2/ErbB3 heterodimers. EGF-like ligands include EGF, TGFα, betacellulin, heparin-binding epidermal growth factor, biregulin, epigen, epiregulin, and amphiregulin, which typically bind to ErbB1 and induce heterodimerization of ErbB1 with ErbB3.
  • The term “bispecific antibody” as used herein refers to a protein comprising two antigen-binding sites, a first binding site exhibiting immunospecific binding to a first antigen or epitope and a second binding site exhibiting immunospecific binding to a second antigen or epitope distinct from the first. An “anti-ErbB2/anti-ErbB3 bispecific antibody” is an antibody that comprises two binding sites, one that immunospecifically binds to the ectodomain of ErbB3 and another that immunospecifically binds to the ectodomain of ErbB2. An exemplary bispecific ErbB3, ErbB2 antibody is an antibody comprising SEQ ID NO:1.
  • An “anti-estrogen agent” as used herein refers to an agent that prevents or reduces production of estrogen or prevents or reduces signaling mediated by estrogen receptors. Anti-estrogen agents include but are not limited to estrogen receptor antagonists and aromatase inhibitors. Estrogen receptor antagonists include but are not limited to raloxifene, fulvestrant, tamoxifen, afimoxifene (4-hydoroxytamoxifen), arzoxifene, toremifene, and lasofoxone. Preferably, the estrogen receptor antagonist is tamoxifen or fulvestrant. Aromatase inhibitors work by blocking the synthesis of estrogen in an animal (e.g., a mouse or a human). This lowers estrogen levels in the animal and thereby inhibits the growth of estrogen-driven cancers. Examples of aromatase inhibitors include but are not limited to exemestane, anastrozole, letrozole, aminoglutethimide, testolactone, vorozole, formestane, and fadrozole. Preferably, the aromatase inhibitor is exemestane or letrozole.
  • By “cancer” is meant any condition characterized by abnormal, unregulated, malignant cell growth.
  • By “malignant tumor” is meant any cancer that takes the form of a tumor.
  • The term “effective amount” refers to an amount of a drug effective to achieve a desired effect, e.g., to ameliorate disease in a subject. Where the disease is a cancer, the effective amount of the drug may inhibit (e.g., slow to some extent, inhibit or stop) one or more of the following characteristics: cancer cell growth, cancer cell proliferation, cancer cell motility, cancer cell infiltration into peripheral organs, tumor metastasis, and tumor growth. Where the disease is a cancer, the effective amount of the drug may alternately do one or more of the following when administered to a subject: slow or stop tumor growth, reduce tumor size (e.g., volume or mass); relieve to some extent one or more of the symptoms associated with the cancer, extend progression free survival, result in an objective response (including a partial response or a complete response), and increase overall survival time. To the extent the drug may prevent growth and/or kill existing cancer cells, it is cytostatic and/or cytotoxic.
  • A “mouse effective amount” refers to an amount of a drug effective to achieve a desired effect when the subject is a mouse.
  • A “human effective amount” refers to an amount of a drug effective to achieve a desired effect when the subject is a human patient.
  • The terms “combination therapy,” “concomitant use,” “co-administration,” co-administering,” “co-administered,” and the like, refer to the administration of at least two therapeutic agents to a subject either simultaneously or within a time period during which the effects of the earlier-administered therapeutic agent are still operative in the subject when a later-administered therapeutic agent is administered.
  • A “receptor tyrosine kinase inhibitor” as used herein refers to a member of a class of drugs that specifically inhibit receptor tyrosine kinases and thus reduce or eliminate the activation of various signal transduction pathways. Receptor tyrosine kinase inhibitors useful for the treatment of cancer as disclosed herein include but are not limited to the small molecule inhibitors erlotinib, afatinib, dasatinib, gefitinib, imatinib, pazopinib, lapatinib, sunitinib, nilotinib and sorafenib. Receptor tyrosine kinase inhibitors also include antibody-based therapeutics such as cetuximab, panitumumab, zalutumumab, nimotuzumab, and matuzumab). Preferably, the receptor tyrosine kinase inhibitor is lapatinib.
  • “Dosage” or “dosing regimen” refers to parameters for administering a drug in defined quantities per unit time (e.g., per hour, per day, per week, per month, etc.) to a patient. Such parameters include, e.g., the size of each dose. Such parameters also include the configuration of each dose, which may be administered as one or more units, e.g., taken at a single administration, e.g., orally (e.g., as one, two, three or more pills, capsules, etc.) or injected (e.g., as a bolus). Dosage sizes may also relate to doses that are administered continuously (e.g., as an intravenous infusion over a period of minutes or hours). Such parameters further include frequency of administration of separate doses, which frequency may change over time. A “dosing cycle” or “dosing interval” is the period of time that comprises one cycle of treatment (e.g., 21 days or 28 days) for a dosing regimen.
  • “Dose” refers to an amount of a drug given in a single administration.
  • In combination, the components of the combination of the anti-ErbB2/ErbB3 antibody have an additive or superadditive effect on suppressing tumor cell growth, as compared to monotherapy with the anti-ErbB2/ErbB3 antibody or treatment with the other agents in the absence of antibody therapy. By “additive” is meant a result that is greater in extent (e.g., in the degree of reduction of tumor mitotic index or of tumor growth or in the degree of tumor shrinkage or the frequency and/or duration of symptom-free or symptom-reduced periods) than the best separate result achieved by monotherapy with each individual component, while “superadditive” is used to indicate a result that exceeds in extent the sum of such separate results. In one embodiment, the additive effect is measured as slowing or stopping of tumor growth or tumor cell proliferation. The additive effect can also be measured as, e.g., reduction in size of a tumor, reduction of tumor mitotic index, reduction in number of metastatic lesions over time, increase in overall response rate, or increase in median or overall survival.
  • One non-limiting example of a measure by which effectiveness of a therapeutic treatment can be quantified is by calculating the log 10 cell kill, which is determined according to the following equation:

  • log 10 cell kill=TC(days)/3.32×Td
  • in which T C represents the delay in growth of the cells, which is the average time, in days, for the tumors of the treated group (T) and the tumors of the control group (C) to have reached a predetermined value (1 g, or 10 mL, for example), and Td represents the time, in days necessary for the volume of the tumor to double in the control animals. When applying this measure, a product is considered to be active if log 10 cell kill is greater than or equal to 0.7 and a product is considered to be very active if log 10 cell kill is greater than 2.8. Using this measure, a combination, used at its own maximum tolerated dose, in which each of the constituents is present at a dose generally less than or equal to its maximum tolerated dose, exhibits therapeutic synergy when the log 10 cell kill is greater than the value of the log 10 cell kill of the best constituent when it is administered alone. In an exemplary case, the log 10 cell kill of the combination exceeds the value of the log 10 cell kill of the best constituent of the combination by at least 0.1 log cell kill, at least 0.5 log cell kill, or at least 1.0 log cell kill.
  • Preferred cancer cells of cell lines are cells of ErbB2 expressing cell lines such as ErbB2 overexpressing cell lines, e.g., BT474-M3 (ATCC® # CRL-HTB-20™, derived from breast ductal carcinoma cells), BT474-M3-Aro (BT474-M3 cells that stably express human aromatase), ZR75-30 (ATCC® # CRL1504™, derived from breast ductal carcinoma cells), SKOV-3 (ATCC® # HTB-77™, derived from metastatic ovarian adenocarcinoma cells), MCF7 (ATCC® # HTB-22™) clone 18, MDA-MB-453 (ATCC® # HTB-131™, derived from breast carcinoma cells), SK-BR-3 (ATCC® # HTB-30™, derived from breast adenocarcinoma cells), and NCI-N87 (ATCC® # CRL-5822™, derived from gastric carcinoma cells).
  • Cancers may include, for example, solid tumors such as: sarcomas (e.g., clear cell sarcoma), carcinomas (e.g., renal cell carcinoma), and lymphomas; tumors of the breast, colon, rectum, lung, oropharynx, hypopharynx, esophagus, gastric esophageal junction, stomach, pancreas, liver, bilecyst, bile duct, small intestine, urinary system (including the kidney, bladder, and epithelium of the urinary tract), female genital system (including the uterine neck, uterus, ovary, chorioma, and gestational trophoblast), male genital system (including the prostate, seminal vesicle, and testicles), endocrine glands (including the thyroid gland, adrenal gland, and pituitary body), skin (including angioma, melanoma, sarcoma originating from bone or soft tissue, and Kaposi's sarcoma), brain and meninges (including astrocytoma, neuroastrocytoma, spongioblastoma, retinoblastoma, neuroma, neuroblastoma, neurinoma and neuroblastoma), nerves, and eyes.
  • A cancer may be an estrogen receptor positive (ER+) cancer. Such cancers exemplify candidates for therapy regimens that include anti-estrogen agents. Such cancers may include but are not limited to certain breast, ovarian, uterine, endometrial, lung, bone, brain, bladder, liver and urogenital cancers.
  • A cancer may be an ErbB2 gene-amplified cancer and/or an ErbB2-expressing or overexpressing cancer. ErbB2, also known as HER2 or Neu, is a cell surface transmembrane receptor protein that generates intracellular signals (e.g., upon ligand activation) via its intracellular tyrosine kinase activity. In excess, such signals can promote oncogenesis e.g., by triggering cell division. The ErbB2 gene is amplified and/or overexpressed in many types of human malignancies, including but not limited to breast, ovarian, endometrial, pancreatic, colorectal, prostate, salivary gland, kidney, and lung. ErbB2 overexpressing cancers are designated a HER2+++ or HER2++ depending on the level of ErbB2 overexpression, with HER2+++ indicating the highest levels of HER2 expression. HER2+++ and HER2++ status are typically determined by an immunoassay such as immunohistochemistry, e.g., Herceptest®. ErbB2 gene amplification may be determined by, e.g., FISH (fluorescence in situ hybridization), with HER2-amplified cancer cells being those that have more than two HER2 gene copies being HER2-amplified, and cells and/or tumors comprising HER2-amplified cancer cells being referred to as “FISH positive.”
  • A number of bispecific anti-ErbB2, antiErbB3 antibodies that are scFv HSA conjugates are described in co-pending US patent publication No. 2011-0059076, and PCT publication Nos. WO2009/126920 and WO 2010/059315, MM-111 (also referred to as B2B3-1) and other bispecific anti-ErbB2/antiErbB3 antibodies that are scFv HSA conjugates and that are suitable for use in the methods and compositions provided herein, including the components of A5-HSA-ML3.9, ML3.9-HSA-A5, A5-HSA-B1D2, B1D2-HSA-A5, B12-HSA-B1D2, B1D2-HSA-B12, A5-HSA-F5B6H2, F5B6H2-HSA-A5, H3-HSA-F5B6H2, F5B6H2-HSA-H3, F4-HSA-F5B6H2, F5B6H2-HSA-F4, B1D2-HSA-H3, and H3-HSA-B1D2. Other suitable bispecific anti-ErbB2/antiErbB3 antibodies are disclosed and claimed in U.S. Pat. Nos. 7,332,580 and 7,332,585. MM-111 is currently undergoing clinical trials, including an open-label Phase 1/2 and pharmacologic study of MM-111 in patients with advanced, refractory HER2 positive cancers, an open-label Phase 1/2 trial of MM-111 in combination with trastuzumab (Herceptin®) in patients with advanced HER2 positive breast cancer, an open label, Phase 1/2 and pharmacologic study of MM-111 with five different combination treatments: MM-111 in combination with cisplatin, capecitabine, and trastuzumab, MM-111 in combination with lapatinib and trastuzumab, and MM-111 in combination with paclitaxel and trastuzumab, MM-111 in combination with lapatinib, paclitaxel and trastuzumab, and MM-111 in combination with docetaxel and trastuzumab, and an open label, Phase 2 study of MM-111 and paclitaxel with or without trastuzumab in patients with HER-2 expressing carcinomas of the distal esophagus, gastroesophageal junction and stomach.
  • A bispecific anti-ErbB2/anti-ErbB3 antibody (e.g., MM-111) can be co-administered with other therapeutic agents, (e.g, an anti-estrogen receptor agent or a receptor tyrosine kinase inhibitor) prior to (e.g., neoadjuvant therapy), concurrent with, or following (e.g., adjuvant therapy) radiotherapy of, or surgical intervention to remove, a malignant tumor.
  • Additional therapeutic agents suitable for combination with anti-ErbB2/anti-ErbB3 antibodies may further include: 1) antibody EGFR inhibitors (e.g. MM-151, Sym004, cetuximab, panitumumab, zalutumumab, nimotuzumab, and matuzumab), additional small molecule tyrosine kinase inhibitors such as PKI-166, PD-158780, EKB-569 (pelitinib), tyrphostin AG-1478, and pan-HER kinase inhibitors (e.g. CI-1033 (canertinib, PD 183805), AC480/BMS-599626, HM781-36B, AZD8931 (sapitinib) and PF299804 (dacomitinib)); 2) microtubule stabilizing agents (e.g. laulimalide, epothilone A, epothilone B, discodermolide, eleutherobin, sarcodictyin A, sarcodictyin B, paclitaxel, nab-paclitaxel or docetaxel); antimetabolites such as 5-fluorouracil (5-FU) and capecitabine; 3) platinum-based therapeutics such as oxaliplatin, carboplatin and cisplatin; 4) mTOR inhibitors such as BEZ235 (a dual PI3K/mTOR inhibitor), AZD8055, everolimus, temsirolimus, siroplimus/rapamycin, and ridaforolimus; and 5) additional anti-ErbB2 therapeutic agents, such as trastuzumab, pertuzumab, and T-DM1 (ado-trastuzumab emtansine). Additional examples of therapeutic agents suitable for combination with anti-ErbB2/anti-ErbB3 antibodies may be found in Table 5 and the Appendix below.
  • MM-111 is suitable for both large scale production and systemic therapy. MM-111 binds to ErbB2/ErbB3 heterodimers and forms a trimeric complex with ErbB2 and ErbB3, effectively inhibiting ErbB3 signaling. The antitumor activity of MM-111 requires the presence of both ErbB2 and ErbB3, but is particularly dependent on ErbB2 expression. The affinity of its ErbB2 antigen-binding site is about 30 times higher than the affinity of its ErbB3 antigen-binding site, but the ErbB2 antigen-binding site does not by itself inhibit ErbB2 activity when bound to ErbB2. The strong binding of MM-111 to ErbB2 places the ErbB3 antigen-binding site in close proximity to bound ErbB2/ErbB3 heterodimer, resulting in an avidity effect that potentiates the binding of the ErbB3 antigen-binding site to the heterodimer ErbB3, whereby a biological effect is produced. MM-111 is administered to human subjects (patients) at an interval measured in days, as a single loading dose of at least 20 mg/kg of MM-111 followed by at least seven day intervals (e.g., every 2 weeks) by at least one administration of a single maintenance dose of MM-111, where the maintenance dose is generally smaller than the loading dose, e.g., at least 5 mg/kg less than the loading dose.
  • EXAMPLES
  • The following examples are provided by way of illustration only and not by way of limitation. Those of skill in the art will readily recognize a variety of non-critical parameters that could be changed or modified to yield essentially the same or similar results.
  • MM-111 in Combination with Anti-Estrogen Therapeutics
  • Methods: Spheroid In Vitro Tumor Model Assay
  • BT474-M3 wild type cells (2000 cells/well) are plated in Ultra Low Cluster 96-well plate (Costar). After overnight incubation, indicated treatments are introduced to the plate. Cells are continued to culture for six days. Spheroids are then examined by Nikon microscope and analyzed by MetaMorph Image Analysis Software (Molecular Devices). The spheroid size from cells cultured in medium containing 10% FBS is set as control.
  • Xenograft Model
  • BT474-M3 cells (2×107 cells per mice) are inoculated subcutaneously into Nu/Nu immunodeficient mice, which are implanted with an estrogen pellet (0.72 mg; 60-day release) one day before the experiment. Tumors are measured after seven days and mice are randomized into four groups: those treated with placebo, MM-111 (60 mg/kg, Q7D), 4-hydroxytamoxifen (5 mg; 60-day release pellet), and combination of MM-111 and 4-hydroxytamoxifen, respectively. Tumors are measured every three days and the experiment is ended at day 32.
  • Example 1 MM-111 and Tamoxifen Combination Therapy Inhibits Tumor Growth In Vivo
  • In order to compare the effect of MM-111 and tamoxifen combination therapy on tumor growth in vivo, estrogen stimulated mice were prepared in the xenograft model using the methods described above or minor variations thereof. Mice were inoculated with tumor forming BT474-M3 cells and on day 7 given a placebo (vehicle control), MM-111, tamoxifen, or a combination of MM-111 and tamoxifen and tumor growth was measured over time. As shown in FIG. 1, this in vivo BT474-M3 xenograft model showed resistance to tamoxifen treatment but when mice were given a combination of MM-111 and tamoxifen the combination treatment inhibited tumor growth to a significantly greater extent. Statistical significance (p<0.05) was observed for the combination group from day 28 onward when compared to vehicle control, from day 21 onward when compared to MM-111 and from day 25 onward when compared to tamoxifen.
  • Example 2 MM-111 Combines Positively with Anti-Estrogen Drugs in Inhibiting Estrogen-Stimulated Spheroid Growth
  • Multicellular spheroids are used to simulate the growth and microenvironmental conditions of tumors in vitro. To further investigate the ability of MM-111 to inhibit cell growth when in combination with anti-estrogen drugs, spheroids of BT474-M3 cells were prepared using the methods described above or minor variations thereof and treated with an ErbB2-binding therapeutic and/or an anti-estrogen therapeutic. Spheroids of estrogen-stimulated cells were treated with a dose range of MM-111, tamoxifen, or the combination of MM-111 and tamoxifen (FIG. 2 a); trastuzumab, tamoxifen or the combination of trastuzumab and tamoxifen (FIG. 2 b); MM-111, fulvestrant, or the combination of MM-111 and fulvestrant (FIG. 2 c); trastuzumab, fulvestrant, or the combination of trastuzumab and fulvestrant (FIG. 2 d); or MM-111, trastuzumab, or the combination of MM-111 and trastuzumab (FIG. 2 e). When used as single agent alone, MM-111, trastuzumab, fulvestrant and tamoxifen showed inhibitory effects on spheroid growth in the estrogen-stimulated BT474-M3 spheroid assay. The combination of tamoxifen or fulvestrant with MM-111 (FIGS. 2 a and 2 c, respectively) or trastuzumab (FIGS. 2 b and 2 d, respectively) increased the degree of growth inhibition, as did the combination of MM-111 and trastuzumab (FIG. 2 e). The inhibitory effects were increased still further when estrogen-stimulated spheroids were treated with the triple combination of MM-111, trastuzumab, and tamoxifen (FIG. 2 f) or MM-111, trastuzumab, and fulvestrant (FIG. 2 g) as compared to the double combinations of drugs.
  • Example 3 MM-111 Combines Positively with Anti-Estrogen Drugs in Inhibiting Heregulin-Stimulated Spheroid Growth
  • To further investigate the ability of MM-111 to inhibit cell growth when in combination with anti-estrogen drugs, spheroids of heregulin (HRG)-stimulated BT474-M3 cells were prepared using the methods described above or minor variations thereof and treated with a dose range of MM-111, tamoxifen, or the combination of MM-111 and tamoxifen (FIG. 3 a); trastuzumab, tamoxifen or the combination of trastuzumab and tamoxifen (FIG. 3 b); MM-111, fulvestrant, or the combination of MM-111 and fulvestrant (FIG. 3 c); trastuzumab, fulvestrant, or the combination of trastuzumab and fulvestrant (FIG. 3 d); or MM-111, trastuzumab, or the combination of MM-111 and trastuzumab (FIG. 3 e). MM-111 inhibited heregulin-induced spheroid growth but tamoxifen (FIG. 3 a), trastuzumab (FIG. 3 b), and fulvestrant (FIG. 3 c) did not inhibit heregulin stimulated spheroid growth. No significant combinational effect was observed when MM-111 was used with tamoxifen (FIG. 3 a) or fulvestrant (FIG. 3 c). The combination of trastuzumab and either tamoxifen (FIG. 3 b) or fulvestrant (FIG. 3 d) failed to show inhibitory activity significantly greater than either drug alone. As shown in FIG. 3 e, MM-111 but not trastuzumab showed inhibitory activity in heregulin-stimulated spheroid growth. Improved inhibitory effects were observed when both drugs were combined. In comparison to the double combination of either MM-111 or trastuzumab with tamoxifen or fulvestrant, the triple combination of MM-111, trastuzumab and either tamoxifen (FIG. 3 f) or fulvestrant (FIG. 3 g) showed similar inhibitory effects as those of MM-111 and trastuzumab in combination (FIG. 3 e) on heregulin-stimulated spheroid growth.
  • Example 4 MM-111 Combines Positively with Anti-Estrogen Drugs in Inhibiting Dual Ligand (Estrogen and Heregulin)-Stimulated Spheroid Growth
  • Dual ligand (estrogen and heregulin) stimulated spheroids were treated with a dose range of tamoxifen, MM-111 or the combination of MM-111 and tamoxifen (FIG. 4 a) or trastuzumab, tamoxifen or the combination of trastuzumab and tamoxifen (FIG. 4 b). While MM-111 and trastuzumab each inhibited spheroid growth (FIG. 4 a) the combination of MM-111 and tamoxifen showed greater inhibitory effects than either drug alone. In contrast, trastuzumab alone had no significant inhibitory effects and the combination of trastuzumab and tamoxifen showed similar effects to tamoxifen alone.
  • Dual ligand stimulated spheroids were then treated with a dose range of fulvestrant, MM-111 or the combination of MM-111 and fulvestrant (FIG. 4 c) or fulvestrant, trastuzumab, or a combination of fulvestrant or trastuzumab (FIG. 4 d). Again, while MM-111 and fulvestrant each separately inhibited spheroid growth the combination of MM-111 and fulvestrant showed greater inhibitory effects than either drug alone (FIG. 4 c). Trastuzumab alone had no significant inhibitory effects and the combination of trastuzumab and fulvestrant showed similar effects to tamoxifen alone (FIG. 4 d).
  • Dual ligand stimulated spheroids were then treated with MM-111, trastuzumab, or a combination of MM-111 and trastuzumab. MM-111 showed greater inhibitory effects than trastuzumab in dual ligand-stimulated spheroid growth. Enhanced inhibitory effects were observed when both drugs were combined (FIG. 4 e).
  • In comparison to the double combination of MM-111 or trastuzumab with tamoxifen or fulvestrant, the triple combination of MM-111, trastuzumab and either tamoxifen (FIG. 4 f) or fulvestrant (FIG. 4 g) showed similar inhibitory effects to those of MM-111 and trastuzumab in combination (FIG. 4 e) on estrogen- and heregulin-(dual ligand) stimulated spheroid growth.
  • The data in the preceding Examples demonstrate that combination therapies comprising MM-111 and an anti-estrogen therapeutic are more effective than each of these therapies alone. The percent of spheroid growth inhibition induced by each treatment under estrogen or heregulin stimulation is summarized in FIG. 5 and Table 1. MM-111 was required for inhibition of spheroids stimulated with heregulin. For each stimulated condition tested, the triple combination resulted in the greatest inhibition of spheroid growth, providing a percent inhibition ranging from about 70% to about 90%.
  • TABLE 1
    Percent inhibitor induced maximal spheroid growth inhibition
    MM-111 + MM-111 + Trastuzumab + Triple
    Trastuzumab anti-estrogen anti-estrogen combination
    Tamoxifen combination
    E2 54% 49% 55% 73%
    HRG 65% 43%  0% 71%
    E2 + HRG 46% 43% 36% 79%
    Fulvestrant combination
    E2 54% 49% 55% 77%
    HRG
    64% 34%  4% 71%
    E2 + HRG 46% 57% 47% 88%

    The percent of spheroid growth inhibition (normalized to untreated, stimulated control) was determined for 104 doses of inhibitor treatment.
  • The combination of MM-111 and tamoxifen resulted in potent inhibition of tumor growth in vivo. Taken together, these data demonstrate that the combination of MM-111 and anti-estrogen therapies results in potent anti-tumor effects in vitro and in vivo.
  • MM-111 in Combination with Lapatinib
  • Methods Computational Modeling
  • A computational model of HRG-induced phospho-ErbB3 signaling, as well as a model of lapatinib, was used as previously described (Schoeberl, et al 2009).
  • Cell Signaling Assay
  • Serum-starved cells are pre-incubated with serial dilutions of MM-111, lapatinib or combinations at doses and treatment times indicated, followed by stimulation with 5 nM heregulin 143 (R&D Systems, Minneapolis, Minn.) for 10 minutes. Cell lysates are probed for phospho-ErbB3 (pErbB3), and phospho-AKT (pAKT) by ELISA as described previously (Schoeberl et al, 2009) Inhibitor IC50 values are calculated by fitting dose-response data to a 4-parameter sigmoidal curve (GraphPad Prism®, GraphPad Software, Inc., La Jolla, Calif.).
  • Cell Proliferation Assay
  • Cells (8,000/well) are seeded into 96-well plates and incubated overnight Inhibitor is added at doses indicated and cells are treated for 24 hours. For experiments with ligand stimulation, cells are serum-starved overnight prior to addition of inhibitor and 2 nM heregulin 1—(R&D Systems, Minneapolis, Minn.) is added 1 hour post-inhibitor treatment in media containing 5% FBS. Numbers of viable cells are measured as an indicator of cell proliferation using the CellTiter-Glo® Luminescent Cell Viability Assay (Promega, Madison, Wis.).
  • Apoptosis Assay
  • BT474-M3 cells (2000 cells/well) are plated in Ultra Low Cluster 96-well plate (Costar®, Corning, N.Y.). After overnight incubation, spheroids are treated with inhibitor at concentrations indicated for 72 hours. Spheroids are then trypsinized and combined with floating cells. Cells are washed twice with cold PBS and suspended in binding buffer (0.01 M HEPES, pH 7.4; 0.14 M NaCl; 2.5 mM CaCl2). Cells are then stained with FITC-conjugated Annexin V and PI. Apoptotic cells are quantified on a FACSCalibur™ FACS machine.
  • Xenograft Efficacy Studies
  • Tumor xenografts are established by subcutaneous injection of BT474-M3 cells into the flank of 5-6 weeks old female athymic nude mice (nu/nu; Charles River Labs, Wilmington, Mass.). Mice receive a subcutaneous 60 day, slow-release estrogen implant in the opposite flank (0.72 mg pellet; Innovation Research of America, Sarasota, Fla.) 24 hours prior to the injection of cells. Once tumors reach a mean volume of 150-500 mm3, mice are randomized into groups of 8 or 10 and dosed by intraperitoneal injection once every three days with vehicle, MM-111 or lapatinib. For lapatinib combination studies, MM-111 is given once every seven days and lapatinib daily by gavage at doses indicated.
  • Aromatase-Overexpressing BT474-M3 Cells and Proliferation Assay
  • BT474-M3 cells were transfected with PS100010 vector containing human aromatase (gene accession No: NM000103.2). Cells with stable expression of aromatase (BT474-M3-Aro) were obtained after selection with 400 μg/ml geneticin. For cell proliferation assay, BT474-M3-Aro cells (5000 cells/well) were plated in phenol red-free RPMI-1640 medium containing 5% charcoal-stripped FBS into 96-well plate. After overnight incubation, indicated treatments were introduced in the presence of androstenedione (A-4; 200 nM) and heregulin (HRG; 2 nM). After three days of treatment, cell viability was determined by WST-1 (Roche; Cat. #11 644 807 001) according to manufacturer's instruction. Cell viability in the presence of 5% charcoal-stripped FBS was set as control (100%).
  • Example 5 The Combination of MM-111 and Lapatinib Inhibits Tumor Growth In Vivo
  • The combination of MM-111 with lapatinib was investigated in vivo in the BT474-M3 breast cancer xenograft model using the methods described above or minor variations thereof. MM-111 and lapatinib were each dosed at an optimal efficacious dose weekly and daily, respectively. The combination of MM-111 and lapatinib provided more potency compared to either drug alone, reaching statistical significance for MM-111 (p=3.9×10-4) and lapatinib (p=5.1×10-3) on day 13 (FIG. 6). The percent change in tumor volume from day 40 to day 7 (inoculation) was calculated for each group (FIG. 6 b). The combination of MM-111 and lapatinib resulted in a percent change in tumor volume of −69% (about 70%), reflecting tumor regressions, compared to −11% (about 10%) for lapatinib and 14% (about 15%) for MM-111.
  • Example 6 Simulations Predict Lapatinib has Suboptimal Activity in Inhibiting Heregulin-Driven pErbB3 and pAKT
  • A dose range of lapatinib inhibition of pErbB3 activation was predicted using the computational modeling described above. A dose range of lapatinib was applied to BT474-M3 cells followed by stimulation with 5 nM heregulin for 10 min. The amount of pErbB3 was measured by ELISA using the methods described above or minor variations thereof. Model-generated dose-response curves overlay the experimental data (FIG. 7 a). A comparison of the inhibitory activity of lapatinib in heregulin-stimulated or unstimulated (basal) cells was performed to demonstrate that heregulin signaling perturbs the activity of lapatinib. Untreated and heregulin-stimulated cells were probed for pErbB3 and pAKT and the IC50 was calculated (FIG. 7 b). These data show that lapatinib alone is not an effective inhibitor of heregulin-activated signaling.
  • Example 7 MM-111 is a More Potent Inhibitor of HRG-Driven ErbB3 and AKT Phosphorylation than Lapatinib
  • In order to compare the ability of MM-111 and lapatinib to inhibit heregulin-induced ErbB3 activation, BT474-M3, or an additional ErbB2 overexpressing breast tumor cell line, ZR75-30 (ATCC® #
  • CRL-1504®), cells were incubated with serial dilutions of either inhibitor for 15 minutes, 1 hour, 4 hours, and 24 hours followed by stimulation with 5 nM heregulin for 10 min Amounts of pAKT and pErbB3 were measured by ELISA essentially as described. MM-111 potently reduced pErbB3 levels (inhibited ErbB3 phosphorylation) in BT474-M3 (IC50=3 nM) cells (FIG. 8 a) and ZR75-30 cells (IC50=5 nM) (FIG. 8 c). Good reduction by MM-111 of pAKT levels (inhibition of AKT phosphorylation) in BT474-M3 (IC50=10) (FIG. 8 b) and in ZR75-30 cells (IC50=4 nM) (FIG. 8 d) was also observed. The ability of MM-111 to inhibit heregulin-induced ErbB3 activation (phosphorylation) was superior to lapatinib by greater than an order of magnitude and the relative IC50 for each inhibitor (FIG. 8 c) was consistent following up to 24 hours incubation with inhibitors, indicating treatment times had little effect on the potency of the inhibitors.
  • Example 8 The Combination of MM-111 and Lapatinib Potently Inhibits pAKT
  • The effect of MM-111 combined with lapatinib on pAKT inhibition (reduction of pAKT levels) was assessed in heregulin-stimulated BT474-M3 cells. Cells were incubated for 2 hours with a dose range of MM-111, lapatinib or their combination and pAKT was measured by ELISA. In the presence of heregulin, the combination of MM-111 and lapatinib was extremely effective, inhibiting pAKT well below basal levels at therapeutically relevant concentrations (FIG. 9). Treatment with either MM-111 (1 μM) or lapatinib (1 μM) alone resulted in similar levels of pAKT inhibition (see FIG. 8 b) while the combination resulted in about 20% more inhibition of pAKT.
  • Example 9 The Ability of Lapatinib to Inhibit Cell Proliferation is Perturbed Under Heregulin-Stimulated Conditions
  • The effect of lapatinib on cell proliferation was measured in unstimulated and heregulin-stimulated BT474-M3 cells. Cells grown in serum or in serum plus 2 nM heregulin were treated with lapatinib across a dose range for 24 hours. Lapatinib treatment resulted in about a 50% inhibition of unstimulated cells but its effect was reduced to about 23% inhibition in heregulin-stimulated BT474-M3 cells (FIG. 10).
  • Example 10 Treatment with the Combination of MM-111 and Lapatinib Results in Increased Apoptosis
  • The effect of the MM-111 combination with lapatinib on apoptosis was assessed in a BT474-M3 spheroid model. Spheroids were prepared using the methods described above or minor variations thereof and treated with MM-111 (100 nM), lapatinib (33 nM), or a combination of 100 nM MM-111 and 33 nM lapatinib. Cells were then stained with Annexin V and propidium iodide (PI) and quantitated using FACS (FIG. 11, Table 2). Cell populations staining positive with Annexin V and PI were quantified as late apoptotic, cell populations staining positive with Annexin V but not PI were quantified as early apoptotic, cell populations staining positive for PI but not Annexin V were quantified as dead cells and populations of cells not stained with either Annexin V or PI were considered alive and not apoptotic (Table 2). Spheroids that were treated with both MM-111 and lapatinib had a higher number of total apoptotic cells (about 46%) compared to those treated with only lapatinib (about 31%) or only MM-111 (about 20%; FIG. 10).
  • TABLE 2
    Percent cell population after treatment with MM-111,
    lapatinib or the combination
    Live cells Early apoptosis Late apoptosis Dead cells
    Control 75.2 17.3 7.2 0.42
    MM-111 78.9 12.9 7.5 0.74
    Lapatinib 67.9 16.8 14.5 0.73
    Combination 52.1 30.0 16.2 1.74
  • Example 11 MM-111 Combines Positively with Anti-Estrogen Drugs and Lapatinib in Inhibiting Dual Ligand (Estrogen and Heregulin)-Stimulated Spheroid Growth
  • To further investigate the ability of MM-111 to inhibit cell growth when in combination with both anti-estrogen drugs and tyrosine kinase inhibitors, spheroids of estrogen and heregulin-stimulated BT474-M3 cells were prepared using the methods described above or minor variations thereof and treated with 3.3 nM, 10 nM, or 30 nM lapatinib, either alone or in combination with a dose range of fulvestrant (FVT) (FIG. 12 a); 3.3 nM, 10 nM, or 30 nM lapatinib, either alone or in combination with a dose range of MM-111 (FIG. 12 b); or 3.3 nM, 10 nM, or 30 nM lapatinib, either alone or in combination with a dose range of both MM-111 and fulvestrant (FIG. 12 c). In the presence of dual ligand stimulation the combination of lapatinib and FVT did not greatly increase inhibition of spheroid growth over lapatinib alone (FIG. 12 a). In contrast, the addition of MM-111 greatly increased the sensitivity of the spheroids to lapatinib treatment (FIG. 12 b), and the triple combination of lapatinib, FVT and MM-111 showed an even greater increase of spheroid growth inhibition over lapatinib alone.
  • Example 12 MM-111 Combines Positively with Anti-Estrogen Drugs in Inhibiting Spheroid Growth in BT474-M3 Cells Overexpressing Human Androstenedione
  • Androstenedione is a steroid hormone that is converted to estrogen by aromatase. To further investigate the ability of MM-111 to inhibit spheroid growth, aromatase-expressing cells were treated in the presence of androstenedione (A4) and heregulin (HRG) with MM-111, letrozole, or the combination of MM-111 or letrozole (FIG. 13 a); MM-111, lapatinib, or the combination of MM-111 and lapatinib (FIG. 13 b); lapatinib, letrozole, or the combination of lapatinib and letrozole (FIG. 13 c); and each of the dual combination plus the triple combination of MM-111, lapatinib, and letrozole (FIG. 13 d). In cells treated with A4 and HRG, the letrozole treatment did not result in significant inhibition of spheroid cell growth as compared to control (untreated) cells, whereas cells treated with MM-111 alone or the combination of MM-111 and letrozole inhibited cell proliferation to a similar extent (FIG. 13 a). Lapatinib treatment of the cells did not result in growth inhibition except at high concentrations, whereas treatment with MM-111 alone or in combination resulted in similar levels of cell growth inhibition except in higher concentrations where the combination showed increased inhibition of cell growth over either of the single treatments (FIG. 13 b). Treatment with lapatinib alone, letrozole alone, or the combination of lapatinib and letrozole did not result in significant cell growth inhibition except at high concentration (FIG. 13 c) Similarly, as shown in FIG. 13 d, the double combination of lapatinib and letrozole resulted in cell growth inhibition only at high drug concentration. In contrast the dual combinations of MM-111 and letrozole or MM-111 and lapatinib both showed an increase in cell growth inhibition as compared to control, and the triple combination of MM-111, lapatinib, and letrozole inhibited cell growth to an even greater degree.
  • Example 13 Amino Acid Sequence of MM-111 (SEQ ID NO:1)
  • QVQLQESGGGLVKPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWVANINRDGSASYYVDS
    VKGRFTISRDDAKNSLYLQMNSLRAEDTAVYYCARDRGVGYFDLWGRGTLVTVSSASTGGGGS
    GGGGSGGGGSQSALTQPASVSGSPGQSITISCTGTSSDVGGYNFVSWYQQHPGKAPKLMIYDVS
    DRPSGVSDRFSGSKSGNTASLIISGLQADDEADYYCSSYGSSSTHVIFGGGTKVTVLGAASDAHK
    SEVAHRFKDLGEENFKALVLIAFAQYLQQSPFEDHVKLVNEVTEFAKTCVADESAENCDKSLHT
    LFGDKLCTVATLRETYGEMADCCAKQEPERNECFLQHKDDNPNLPRLVRPEVDVMCTAFHDNE
    ETFLKKYLYEIARRHPYFYAPELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAK
    QRLKCASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDLLECADDRA
    DLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPADLPSLAADFVESKDVCKNYAEAK
    DVFLGMFLYEYARRHPDYSVVLLLRLAKTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNL
    IKQNCELFEQLGEYKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAED
    YLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPKEFQAETFTFHADICTL
    SEKERQIKKQTALVELVKHKPKATKEQLKAVMDDFAAFVEKCCKADDKETCFAEEGKKLVAAS
    QAALGLAAALQVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIAWVRQMPGKGLEYMGLIYP
    GDSDTKYSPSFQGQVTISVDKSVSTAYLQWSSLKPSDSAVYFCARHDVGYCTDRTCAKWPEWL
    GVWGQGTLVTVSSGGGGSSGGGSGGGGSQSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSW
    YQQLPGTAPKLLIYDHTNRPAGVPDRFSGSKSGTSASLAISGFRSEDEADYYCASWDYTLSGWVF
    GGGTKLTVLG
  • Dosing and Administration of MM-111 in Combination with One or More Additional Therapeutics Example 14 Mode of Administration of MM-111
  • MM-111 is prepared as a formulation containing 25 mg/ml MM-111 in a sterile aqueous solution comprising 20 mM L-histidine hydrochloride, 150 mM sodium chloride, pH 6.5, which is stored at 2-8° C.
  • MM-111 must be brought to room temperature prior to administration. Containers (e.g., vials) of MM-111 must not be shaken. The appropriate quantity of MM-111 is removed from the container, diluted in 250 mL of 0.9% normal saline and administered as an infusion using a low protein binding in-line filter (e.g., a 0.22 micrometer filter).
  • MM-111 is initially administered over about 90 minutes (first administration). In the absence of an infusion reaction, subsequent doses are administered over about 60 minutes.
  • A patient's body weight at the start of a dosing cycle is used to calculate the dose used throughout the cycle. Should a patient's body weight change by more than 10%, a new total dose is calculated to reflect this change.
  • Example 15 Dosage and Administration of MM-111
  • Preferred plasma concentrations of MM-111 achieved during treatment are at least 106 mg/L. It has now been discovered that certain combinations of dose frequency and dosage will achieve and maintain this plasma concentration during the course of treatment in at least half, and preferably in more than 60%, 70% or 80% of treated patients.
  • In certain embodiments a higher initial dose (loading dose—LD) is given, followed as defined intervals by at least one maintenance dose (MD). Intervals of dosing in days are typically indicated as QxD, wherein x represents an integer, so that a QxD of 7 indicates dosing every 7 days. Table 3A, Table 3B, and Table 3C below show doses and dosing intervals of the invention. In Table 3A, Table 3B, and Table 3C the indicated loading doses are optional—initial doses are preferably made at the indicated loading dose (LD), but may (e.g., as directed or at the physician's discretion) be made at the maintenance dose (MD). Table 3A provides a set of exemplary dosing intervals, loading doses and maintenance doses. Table 3B provides a variation of Table 3A allowing for dosage variability (indicated as “about”) of up to +/−3 mg/mL. Table 3C appears below and provides a more extensive set of exemplary dosing intervals, loading doses and maintenance doses. In each cell of Table 3A, Table 3B, and Table 3C, the top figure is the integer x in the interval QxD (e.g., 18 as the top figure in a cell indicates a dosing interval of Q18D or every 18 days), the middle figure represents the (optional) loading dose (LD) in mg/kg, and the bottom figure represents the maintenance dose (MD) in mg/kg. Thus the top cell in Table 3A indicates a dosing interval (QxD) of once every seven days, a loading dose (optional) of 25 mg per kg of patient body weight, and a maintenance dose of 20 mg per kg of patient body weight; while the cell furthest to the right on the top row of Table 3C indicates a dosing interval (QxD) of once every seven days, a loading dose (optional) of 30 mg per kg of patient body weight, and a maintenance dose of 15 mg per kg of patient body weight.
  • TABLE 3B
     7
    about 25 
    about 20 
     7
    about 40 
    about 30 
    14
    about 60 
    about 44 
    14
    about 90 
    about 75 
    21
    about 120
    about 105
  • TABLE 3A
    7
    25
    20
    7
    40
    30
    14
    60
    45
    14
    90
    75
    21
    120
    105
  • TABLE 3C
    7 7 7 7 7 7 7 7 7 7 7 7 7
    10 15 20 25 30 15 20 25 30 35 20 25 30
    5 5 5 5 5 10 10 10 10 10 15 15 15
    7 7 7 7 7 7 7 7 7 7 7 7 7
    35 40 25 30 35 40 45 30 35 40 45 50 55
    15 15 20 20 20 20 20 25 25 25 25 25 25
    7 7 14 14 14 14 14 14 14 14 14 14 14
    60 65 35 40 45 50 55 60 65 70 75 40 45
    25 25 30 30 30 30 30 30 30 30 30 35 35
    14 14 14 14 14 14 14 14 14 14 14 14 14
    50 55 60 65 70 75 45 50 55 60 65 70 75
    35 35 35 35 35 35 40 40 40 40 40 40 40
    14 14 14 14 14 14 14 14 14 14 14 14 14
    50 55 60 65 70 75 55 60 65 70 75 60 65
    45 45 45 45 45 45 50 50 50 50 50 55 55
    14 14 14 14 14 14 14 14 21 21 21 21 21
    70 75 65 70 75 70 75 75 60 65 70 65 70
    55 55 60 60 60 65 65 70 55 55 55 60 60
    21 21 21 21 21 21
    75 70 75 80 85 90
    60 65 70 75 80 85
  • Example 16 Dosage and Administration of MM-111 with Lapatinib and Trastuzumab
  • Treatment for patients with trastuzumab-refractory HER2-overexpressing breast cancer is a critical unmet need in the field of breast oncology, and novel approaches to address this need are required. Although selective tyrosine kinase inhibitors (TKIs) have been highly effective for the treatment of certain tyrosine kinase oncogene-driven cancers, their clinical anti-tumor efficacy in the treatment of HER2-driven breast cancer has been disappointing despite adequate biodistribution and apparent target inhibition. Two completed phase II trials using the most potent HER2 TKI, lapatinib, have reported response rates of only 4%-8% in patients with trastuzumab-refractory HER2-overexpressing breast cancer. It is now known that the effective treatment of HER2+ breast cancer is more complex and resilient than previously thought. Recent evidence has highlighted the role of HER3 and a robust signal buffering capacity inherent in the HER2-HER3 tumor driver that protects it against a two log inhibition of HER2 catalytic activity, placing it beyond the therapeutic index of even the most potent tyrosine kinase inhibitors (TKIs).
  • Typically, lapatinib is administered at a dosage of 1000 to 1500 mg in 250 mg tablets taken once daily. Lapatinib is often used in combination with another cancer medication, capecitabine, which is taken for 14 day periods with one week in between.
  • In order to test whether the full inactivation of the HER2-HER3 driver can be achieved with much higher TKI dosing at an intermittent dosing schedule is more efficacious than continuous dosing, a modified dosing schedule is used wherein an increased dose of lapatinib is administered on days 1-5 of a 14 day cycle, said increased dose being a higher dose than the standard dose of 1000 to 1500 mg/day. In some embodiments, the higher lapatinib dose is between 2000 and 9000 mg/d. For example, higher lapatinib dose might be 2000, 2250, 3375, 3000, 3250, 3500, 3750, 4000, 4250, 4500, 4750, 5000, 5250, 5500, 5750, 6000, 6250, 6500, 6750, 7000, 7250, 7500, 7750, 8000, 8250, 8500, 8750, or 9000 mg/day, and so on.
  • In certain embodiments a loading dose is given on day 1 of the 14-day cycle that is a higher dose than that given on subsequent days, the maintenance dose. For example, a loading dose given on day 1 of the 14 day cycle might be 7000 mg/day, followed by a maintenance dose of 3000 mg/day. Non-limiting examples of loading dose and maintenance dose combinations are listed in Table 4 below.
  • MM-111 is administered as described in Example 15. In some embodiments the treatment further comprises trastuzumab. Trastuzumab is typically given with an initial loading dose followed by a maintenance dose. For example, trastuzumab may be dosed at a loading dose of 8 mg/kg followed by a maintenance dose of 6 mg/kg every three weeks.
  • TABLE 4
    Exemplary lapatinib dosing schedule: loading dose (top number)
    and maintenance dose (bottom number) in mg/d
    2000 2000 2000 2500 2500 2500 3000 3000 3000 3000 3000 3500 3500
    1000 1500 2000 1000 1500 2000 1000 1500 2000 2500 3000 1000 1500
    3500 3500 3500 4000 4000 4000 4000 4000 4000 4500 4500 4500 4500
    2000 2500 3000 1000 1500 2000 2500 3000 3500 1000 1500 2000 2500
    4500 4500 4500 5000 5000 5000 5000 5000 5000 5000 5000 5500 5500
    3000 3500 4000 1000 1500 2000 2500 3000 3500 4000 4500 1000 1500
    5500 5500 5500 5500 5500 5500 5500 6000 6000 6000 6000 6000 6000
    2000 2500 3000 3500 4000 4500 5000 1000 1500 2000 2500 3000 3500
    6000 6000 6000 6000 7500 7500 7500 7500 7500 7500 7500 7500 7500
    4000 4500 5000 5500 1000 1500 2000 2500 3000 3500 4000 4500 5000
    7500 7500 7500 7500 8000 8000 8000 8000 8000 8000 8000 8000 8000
    5500 6000 6500 7000 1000 1500 2000 2500 3000 3500 4000 4500 5000
    8000 8000 8000 8000 8000 9000 9000 9000 9000 9000 9000 9000 9000
    5500 6000 6500 7000 7500 1000 1500 2000 2500 3000 3500 4000 4500
    9000 9000 9000 9000 9000 9000 9000 9000
    5000 5500 6000 6500 7000 7500 8000 8500
  • Example 17 Dosage and Administration of MM-111 with Cisplatin, Capecitabine, and Trastuzumab
  • Administration of MM-111 with cisplatin, capecitabine, and trastuzumab is done, for example, by the following method or minor variations thereof.
  • Patients are administered therapy on a 21-day treatment cycle. Cisplatin is administered on day 1 of each 21-day cycle by intravenous (i.v.) infusion over two hours, at a dose of 80 mg/m2. Capecitabine is administered orally, twice daily, at a dose of 1000 mg/m2. Up to 21-day cycles of cisplatin and capecitabine are administered. Trastuzumab is administered i.v. at week 1 at an 8 mg/kg loading dose over 90 minutes, followed by a maintenance dose of 6 mg/kg every 21 days over 30-90 minutes. MM-111 is administered as described in the above Examples. For example, MM-111 is administered i.v. over 90 minutes for the first dose and then weekly over 60 minutes thereafter.
  • Example 18 Dosage and Administration of MM-111 with Lapatinib and Trastuzumb
  • Administration of MM-111 with lapatinib and trastuzumab is done, for example, by the following method or minor variations thereof. Trastuzumab is administered i.v. at a 4 mg/kg loading dose on week 1 over 90 minutes, followed by a 2 mg/kg weekly maintenance dose thereafter. Lapatinib is given by mouth either at 1000 mg daily doses or at the one of the dose regimens described in Example 13. MM-111 is administered as described in the above Examples. For example, MM-111 is administered i.v. over 90 minutes for the first dose and then weekly over 60 minutes thereafter.
  • Example 19 Dosage and Administration of MM-111 with Paclitaxel and Trastuzumab
  • Administration of MM-111 with paclitaxel and trastuzumab is done, for example, by the following method or minor variations thereof. Patients are administered therapy on a 28-day treatment cycle. Paclitaxel dosing begins on day 1 of cycle 1. Paclitaxel is administered at 80 mg/m2 weekly, as an i.v. infusion over 60 minutes. Trastuzumab is administered at a 4 mg/kg loading dose on week 1, i.v. over 90 minutes, followed by a 2 mg/kg weekly maintenance dose thereafter. MM-111 is administered as described in the above Examples. For example, MM-111 is administered i.v. over 90 minutes for the first dose and then weekly over 60 minutes thereafter.
  • Example 20 Coadministration of MM-111 and Other Therapeutic Agents
  • MM-111 (at dosages described herein; see, e.g., Example 15) can be administered in combination with one or more additional agents to a patient in need thereof for the treatment of a cancer. In particular, MM-111 can be administered in combination with MM-151 (oligoclonal anti-EGFR mixture), TDM-1 (ado-trastuzumab emtansine, an antibody-drug conjugate of the antibody trastuzumab linked to maytansine derivative (DM1)), an mTOR inhibitor (e.g., AZD8055, sirolimus, everolimus, temsirolimus, ridaforolimus, or the dual PI3K/mTOR inhibitor BEZ235), and combinations thereof.
  • MM-151 is an oligoclonal therapeutic that is a mixture of three fully human monoclonal antibodies designed to bind to non-overlapping epitopes of the epidermal growth factor receptor, or EGFR (also known as ErbB1). An oligoclonal therapeutic is a mixture of two or more distinct monoclonal antibodies. MM-151 is disclosed, e.g., in copending PCT Application No. PCT/US12/45235.
  • MM-111 can be administered in the same dosage form as MM-151, TDM-1, and/or the mTOR inhibitor(s), or the agents can be administered in separate dosage forms.
  • In an embodiment, MM-111 and one or more of MM-151, TDM-1, and/or the mTOR inhibitor(s) is administered to a patient for the treatment of a malignant tumor, e.g., an ErbB2-expressing or ErbB2 over-expressing tumor (e.g., HER or HER tumors). The tumor may be a melanoma, clear cell sarcoma, head and neck, endometrial, prostate, breast, ovarian, gastric, colon, colorectal, lung, bladder, pancreatic, salivary gland, liver, skin, brain or renal tumor.
  • In another embodiment, MM-111 and MM-151 are co-administered to treat a solid tumor (e.g., an advanced refractory solid tumor) in a patient in need thereof.
  • The effect of MM-111 combined with trastuzumab and the dual PI3K/mTOR inhibitor BEZ235 on cell proliferation was assessed by the methods described above or with minor variations thereof in unstimulated and heregulin-stimulated NCI-N87 cells in vitro. Cells were incubated for 2 hours with a dose range of MM-111, trastuzumab, or their combination, in the presence of BEZ235 as shown in FIG. 14. Cells were plated and either untreated (Control, open circle), treated with BEZ235 alone (open diamond), or treated with BEZ235 (5 nM FIG. 14 a, 20 nM FIG. 14 b) and either trastuzumab (closed circle), MM-111 (closed square), or the combination of MM-111 and trastuzumab (closed diamond). Treatment with the triple combination of MM-111, trastuzumab, and BEZ235 resulted in a significant reduction in cell number compared to control cells in both the presence and absence of heregulin stimulation.
  • The effect of MM-111 combined with the mTOR inhibitor everolimus and trastuzumab was assessed in unstimulated (FIG. 15 a) and heregulin-stimulated (FIG. 15 b) BT474M3 cells in vitro. Cells were plated and either untreated (Control, open circle), treated with everolimus alone (open diamond), or treated with everolimus (1 nM FIG. 15 a, 5 nM FIG. 15 b) and either trastuzumab (closed circle), MM-111 (closed square), or the combination of MM-111 and trastuzumab (closed diamond). Treatment with the triple combination of MM-111, trastuzumab, and everolimus resulted in a significant reduction in cell number compared to control cells in both the presence and absence of heregulin stimulation.
  • Example 21 Combination Treatment with MM-111 and MEK/PI3k/AKT Pathway Inhibitors
  • MM-111, at dosages described herein (see, e.g., Example 15), can be administered in combination with one or more MAP/ERK kinase (MEK)/phosphatidylinositol 3-kinase (PI3k)/AKT pathway inhibitors to a patient in need thereof for the treatment of a cancer. MM-111 can be administered in the same dosage form as the MEK/PI3k/AKT pathway inhibitor(s) or these agents can be administered in separate dosage forms.
  • In one embodiment, MM-111 and a MEK/PI3k/AKT pathway inhibitor is administered to a patient for the treatment of a malignant tumor, e.g., an ErbB2-expressing or ErbB2 over-expressing tumor (e.g., HER+++ or HER++ tumors). The tumor may be a melanoma, clear cell sarcoma, head and neck, endometrial, prostate, breast, ovarian, gastric, colon, colorectal, lung, bladder, pancreatic, salivary gland, liver, skin, brain or renal tumor.
  • To determine whether the combination of MM-111 with a MEK inhibitor results in decreased Erk1/2 activity in a tumor model, MM-111 and the MEK inhibitor trametinib, and were tested in a mouse xenograft model as described below:
  • Cell Culture and Xenograft Study
  • BT474-M3 cells are transduced with full length HRG1-β1 (Origene)-constructs (encoding PAC-PA-turboGFP for selection) by lentiviral infection. A HRG overexpressing polyclonal cell line (BT474-M3-GFP-HRG) is established after puromycin selection and sorting for high GFP expressing cell population. A control cell line (BT474-M3-GFP) is engineered to express EGFP in the same manner. Cells are maintained in culture in RPMI supplemented with 10% FBS, penicillin, streptomycin and puromycin. MM-111 is produced in-house at Merrimack Pharmaceuticals and MK2206, trametinib, and UO126 (UO126-EtOH) are purchased from Selleckchem. 7-9 week old female NCRNU-mice (Taconic) are implanted subcutaneously with 0.72 mg 60-day release 17 β-estradiol pellets (Innovative Research of America).
  • MM-111 Combination Treatment with Trametinib
  • 48 hours after implantation of the β-estradiol pellets, 15×106 BT474-M3-GFP-HRG cells were implanted in the second axillary mammary fat pad. When tumors reached an average volume of 450-500 mm3 (day 0), mice were randomized into groups to receive control (phosphate buffered saline, intraperitoneally), MM-111 (48 mg/kg, intraperitoneally using phosphate buffered saline as vehicle), trametinib (3 mg/kg, by oral gavage in 0.5% methylcellulose/0.1% Tween®-80) or a combination of MM-111 and trametinib. A set (3 per group) of mice are euthanized 4 h after the initial treatment. Tumors were excised and snap-frozen in liquid nitrogen for subsequent protein analysis. Another set of animals continued on their assigned treatment regimens for another 7 days receiving phosphate buffered saline control (intraperitoneally) on days 3 and 6, MM-111 (48 mg/kg, intraperitoneally) on days 3 and 6, trametinib (3 mg/kg, by oral gavage) on days 1, 2, 5 and 6. 24 hours after the last treatment, tumors were excised and snap-frozen in liquid nitrogen for subsequent protein analysis. Frozen tumor samples were pulverized and protein lysates were prepared in Tissue Extraction Reagent (Invitrogen) supplemented with Protease Inhibitor Cocktail Set III (Calbiochem, EMD) and Halt™ Phosphatase Inhibitor Cocktail (Thermo Scientific). Lysates were diluted in assay buffer (1% Bovine Serum Albumin in Tris buffered saline, pH 7.4, containing 0.1% Tween-20) to 1 mg/ml concentration and analyzed for phopsho- and total Erk1/2 using Luminex® immunosandwich assay on a FLEXMAP 3D instrument (Luminex). Antibodies from Cell Signaling Technologies were used to detect total Erk1/2 (3374) and pT202/Y204-sites on Erk1/2 (4370BF). 4 h after the start of treatment, addition of MM-111 to trametinib showed a trend to decreasing Erk1/2 activity (decreasing pT202/Y204-Erk1/2, p=0.086, Student's t-test) compared to treatment withtrametinib alone (FIG. 16A). 24 h after the last treatment, addition of MM-111 to trametinib significantly decreased Erk1/2 activity (decreasing pT202/Y204-Erk1/2, p<0.001, Student's t-test) compared to treatment with trametinib alone (FIG. 16B).
  • Example 22 Combination Treatment with MM-111 and ErbB2-Targeted Therapeutic Agents
  • MM-111, at dosages described herein (see, e.g., Example 15), can be administered in combination with one or more additional ErbB2-targeted therapeutic agents, such as trastuzumab, T-DM1, pertuzumab, etc. In order to assess the effect of MM-111 in combination with an additional ErbB2-targeted therapeutic, cells with (FIG. 17B) and without (FIG. 17A) heregulin (HRG) were treated with a dose escalation of T-DM1 and then treated with MM-111 as follows.
  • Materials
  • NCI-N87 cells are transduced with full length HRG1-β1 (Origene)-constructs (encoding PAC-PA-turboGFP for selection) by lentiviral infection. A HRG overexpressing polyclonal cell line (NCI-N87-GFP-HRG) is established after puromycin selection and sorting for high GFP expressing cell population. A control cell line (NCI-N87-GFP) is engineered to express EGFP in the same manner. Cells are maintained in culture in RPMI supplemented with 10% fetal bovine serum, penicillin, streptomycin and puromycin (1 μg/ml).
  • MM-111 is manufactured at Merrimack Pharmaceuticals. Recombinant human NRG-1-β1 EGF domain is from R&D Systems. T-DM1 (KADCYLA, Genentech) is obtained from pharmacy. CellTiter-Glo® Luminescent Cell Viability Assay is from Promega.
  • 700 NCI-N87 or BT-474-M3 cells were seeded on 384-well flat clear bottom polystyrene microplates in 20 μl of culture media (RPMI supplemented with 10% fetal bovine serum and penicillin and streptomycin). Cells were allowed to adhere 16-20 hours at +37° C., 5% CO2. Cells were stimulated for 4 hours by adding 5 μl of culture media containing recombinant human NRG-1-β1 EGF domain (HRG1) bringing the final HRG1 stimulation concentration to 0, 0.313, 0.625, 1.25, 2.5, 5 or 10 nM. After 4 hours, T-DM1 was added in 5 μl of culture media with or without MM-111 bringing the T-DM1 concentration to 0, 0.001524, 0.004572, 0.013717, 0.041152, 0.123457, 0.37037, 1.11111, 3.333333 or 10 μg/ml and the MM-111 concentration to 0 or 1 μM. All conditions were assayed in biological quadruplicates. Cells were incubated for 72 h at +37° C., 5% CO2. Cell viability was determined by CellTiter-Glo® Luminescent Cell Viability Assay by adding 20 μl of reconstituted CellTiter-Glo reagent per well, incubating plated for 10 minutes at RT in an orbital shaker and reading the luminescence signal using Biotek™ Synergy H1 Hybrid Reader.
  • 700 NCI-N87-GFP or NCI-N87-GFP-HRG1 cells were seeded on 384-well flat clear bottom polystyrene microplates in 20 μl of culture media (RPMI supplemented with 10% fetal bovine serum and penicillin and streptomycin). Cells were allowed to adhere 16-20 hours at +37° C., 5% CO2. T-DM1 was added in 5 μl of culture media with or without MM-111 bringing the T-DM1 concentration to 0, 0.001524, 0.004572, 0.013717, 0.041152, 0.123457, 0.37037, 1.11111, 3.333333 or 10 μg/ml and the MM-111 concentration to 0 or 1 μM. All conditions were assayed in biological quadruplicates. Cells were incubated for 72 h at +37° C., 5% CO2. Cell viability was determined by CellTiter-Glo® Luminescent Cell Viability Assay by adding 20 ul of reconstituted CellTiter-Glo reagent per well, incubating plated for 10 minutes at RT in an orbital shaker and reading the luminescence signal using Biotek Synergy H1 Hybrid Reader.
  • As shown in FIG. 17, In the absence of endogenous heregulin, treatment of cells with T-DM1 reduces cell viability up to approximately 54.6% with 0.1 μg/mL T-DM1, at which point the remaining cells become resistant. The addition of MM-111 restores sensitivity of the cells to T-DM1. This effect is significantly increased in the presence of endogenous heregulin where treatment of cells with T-DM1 reduces cell viability up to approximately 47.3% with 0.04111152 μg/mL T-DM1, at which point the remaining cells become resistant. The addition of MM-111 restores sensitivity of the cells to T-DM1 further reducing cell viability by approximately 30% to approximately 80% (FIG. 17B).
  • To test the combination therapy with endogenous heregulin (HRG), NCI-N87 cells and BT-474-M3 cells were prepared as described above and treated with a dose range of both T-DM1 and heregulin in the presence and absence of MM-111 (3D graphs, FIG. 18). Cell viability was tested and numbers are given as % of control and summarized in Tables 1-4 below. As can be seen in FIG. 18, while exogenous HRG decreases the activity of T-DM1, co-treatment of the cells with a combination of MM-111 and T-DM1 greatly reduced cell viability in the presence of endogenous heregulin in both NCI-N87 (FIG. 18B) and BT-474-M3 cells (FIG. 18D).
  • TABLE 1
    NCI-N87 cells without MM-111
    T-
    HRG DM1
    (nM) 0 0.001524 0.004572 0.013717 0.041152 0.123457 0.37037 1.111111 3.333333 10 (μg/ml)
    0 100.0001 93.36976 75.62197 51.2155 50.04061 42.64956 46.5761 42.8517 40.80667 43.84619
    0.313 105.2683 100.3208 92.59442 79.62461 74.71907 64.7733 50.76463 55.8238 57.24998 57.69454
    0.625 110.9354 106.4739 106.0159 84.03995 61.45342 71.29415 58.90545 65.075 62.77755 67.86139
    1.25 105.9291 102.2426 94.15791 81.15374 64.90047 66.01914 69.29675 69.40256 58.90233 68.76759
    2.5 95.5206 90.80036 84.09815 83.76757 69.34041 74.67369 67.23904 72.16217 66.05516 65.68811
    5 99.9019 94.98233 89.09868 80.41984 71.01775 73.54437 68.99217 75.72861 64.94796 63.21516
    10 114.0075 88.14249 101.4929 80.41022 69.82144 64.32714 61.13489 62.15488 59.45985 68.79469
  • TABLE 2
    NCI-N87 cells with MM-111
    T-
    HRG DM1
    (nM) 0 0.001524 0.004572 0.013717 0.041152 0.123457 0.37037 1.111111 3.333333 10 (μg/ml)
    0 99.99994 95.67672 78.87085 64.23961 46.34934 41.8017 32.86071 35.79531 35.56626 32.20002
    0.313 101.4158 98.3686 87.83576 74.46012 52.5808 44.82781 41.52759 37.6864 34.50943 36.02112
    0.625 105.024 108.5211 86.78032 66.85997 55.32827 45.12097 40.0349 38.27817 38.6461 36.36282
    1.25 110.2781 107.8277 99.94741 73.38117 51.00783 43.11336 43.031 40.2014 37.52794 35.5527
    2.5 118.0867 117.2117 99.28302 90.13422 63.70836 52.75964 43.73032 41.26843 36.41708 38.52877
    5 126.5145 128.6171 118.6616 95.04545 70.74315 51.52235 43.87818 43.0434 42.54104 40.95163
    10 146.1417 140.3695 122.2993 95.17032 70.81566 52.81674 47.2081 43.46258 42.60197 42.12973
  • TABLE 3
    BT-474-M3 cells without MM-111
    T-
    HRG DM1
    (nM) 0 0.001524 0.004572 0.013717 0.041152 0.123457 0.37037 1.111111 3.333333 10 (μg/ml)
    0 100 113.2862 98.62592 94.0068 59.46435 46.28945 46.03028 40.94169 38.39982 37.44111
    0.313 119.8347 116.4401 115.5001 120.231 97.75329 98.67097 94.19504 95.98138 95.80208 84.02226
    0.625 123.4093 117.8323 116.7982 112.1619 108.0231 104.9562 107.4638 97.33409 94.15624 94.01721
    1.25 116.9189 108.1483 107.0419 108.9035 104.0392 109.1902 91.67416 94.82696 100.5096 88.67155
    2.5 114.6797 113.1709 104.357 114.8767 108.3647 96.44198 100.0466 99.7191 99.2052 89.80503
    5 117.0592 123.3681 119.6607 111.0487 111.2081 98.30082 102.3885 99.45587 90.43802 99.39723
    10 116.0498 130.2768 115.6098 111.159 104.5468 101.7859 99.51807 101.389 98.00674 87.40396
  • TABLE 4
    BT-474-M3 cells with MM-111
    T-
    HRG DM1
    (nM) 0 0.001524 0.004572 0.013717 0.041152 0.123457 0.37037 1.111111 3.333333 10 (μg/ml)
    0 100.0001 105.1766 103.9555 103.6039 77.39243 66.65368 57.99249 52.81845 61.04489 49.52486
    0.313 116.4349 118.4495 115.8389 106.7296 91.78999 68.56924 61.57601 59.03206 58.89703 52.39394
    0.625 123.4726 120.7381 123.5142 115.1022 91.45484 68.63193 61.65257 53.11713 54.6567 47.24296
    1.25 132.3855 131.1941 131.9924 125.6464 100.4235 78.41731 64.38723 54.49712 56.83025 54.51656
    2.5 131.572 136.6084 146.7699 134.055 109.0098 80.11692 73.45718 64.84097 55.26672 53.33503
    5 146.0121 152.0284 151.8639 138.9854 116.4752 86.2649 70.99611 74.74774 61.73628 55.30379
    10 157.2453 154.9672 149.674 145.9666 121.6431 91.79307 81.23282 69.08484 71.97691 61.17057
  • Endnotes
  • While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth.
  • All patents patent applications and publications mentioned herein are incorporated by reference to the same extent as if each independent patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety. In particular, WO 2012/116317 is incorporated herein by reference in its entirety.
  • APPENDIX Anticancer Agents
  • The Table and Appendix below describe effective an anti-estrogen agents, receptor tyrosine kinase inhibitors; MEK/PI3 kinase/AKT inhibitors, and mTOR inhibitors that can be used in the methods and compositions of the invention.
  • The bispecific anti-ErbB2/anti-ErbB3 antibody co-administered in combination with an agent selected from i) an effective amount of an anti-estrogen agent; ii) an effective amount of a receptor tyrosine kinase inhibitor; iii) a MEK/PI3 kinase/AKT inhibitor; iv) MM-151; v) an mTOR inhibitor; and/or vi) trastuzumab or T-DM1, and combinations thereof, can be further co-administered with at least a third antineoplastic agent selected from any of those disclosed in the Table and Appendix below.
  • TABLE 5
    Exemplary antineoplastic agents for treatment of breast cancer in combination with a
    bispecific anti-ErbB2/anti-ErbB3 antibody.
    Exemplary Agent
    Therapeutic Class (Generic/Tradename) Exemplary Dose
    Mitotic Inhibitors paclitaxel (TAXOL ®; ABRAXANE ®) 175 mg/m2
    docetaxel (TAXOTERE ®) 60-100 mg/m2
    Topoisomerase Inhibitors camptothecin
    topotecan hydrochloride (HYCAMTIN
    etoposide (EPOSIN ®)
    Alkylating Agents cyclophosphamide (CYTOXAN ®) 600 mg/m2
    Platinum-Based Agents Cisplatin 20-100 mg/m2
    carboplatin (PARAPLATIN ®) 300 mg/m2
    nedaplatin (AQUPLA ®)
    oxaliplatin (ELOXATIN ®) 65-85 mg/m2
    satraplatin (SPERA ®)
    triplatin tetranitrate
    Selective Estrogen Modulators (SERM) tamoxifen (NOLVADEX ®) 20-40 mg/day
    raloxifene (EVISTA ®) 60 mg/day
    toremifene (FARESTON ®)
    Antimetabolites methotrexate 40 mg/m2
    Fluorouracil (5-FU) 500 mg/m2
    Raltitrexed
    Antitumor Antibiotics Doxorubicin (ADRIAMYCIN ®) 40-75 mg/m2
    epirubicin (ELLENCE ®) 60-120 mg/m2
    Aromatase Inhibitors aminoglutethimide (CYTADREN ®) 250-2000 mg/day
    anastrozole (ARIMIDEX ®) 1 mg/day
    letrozole (FEMARA ®) 2.5 mg/day
    Vorozole
    exemestane (AROMASIN ®) 25-50 mg/day
    Testolactone
    fadrozole (AFEMA ®)
    Anti-VEGF Agents bevacizumab (AVASTIN ®) 10 mg/kg
    Anti-ErbB2 (HER2/neu) Agents trastuzumab (HERCEPTIN ®) 2-8 mg/kg
    Pertuzumab (OMNITARG ®)
    Anti-ErbB3 (HER3) Agents U3-1287 (AMG 888)
  • APPENDIX Anticancer Agents
  • Other anticancer agents for combination
    with a bispecific anti-ErbB2/anti-ErbB3
    antibody Brand Name(s) Manufacturer/Proprietor
    Anti-IGF1R Antibodies
    AMG 479 (fully humanized mAb) Amgen
    IMCA12 (fully humanized mAb) ImClone
    NSC-742460 Dyax
    19D12 (fully humanized mAb)
    CP751-871 (fully humanized mAb) Pfizer
    H7C10 (humanized mAb)
    alphaIR3 (mouse)
    scFV/FC (mouse/human chimera)
    EM/164 (mouse)
    MK-0646, F50035 Pierre Fabre Medicament, Merck
    Small Molecules Targeting IGF1R
    NVP-AEW541 Novartis
    BMS-536,924 (1H-benzoimidazol-2-yl)- Bristol-Myers Squibb
    1H-pyridin-2-one)
    BMS-554,417 Bristol-Myers Squibb
    Cycloligan
    TAE226
    PQ401
    Anti-EGFR Antibodies
    INCB7839 Incyte
    Bevacizumab Avastin ® Genentech
    Cetuximab Erbitux ® EMCLONE
    mAb 806
    Matuzumab (EMD72000)
    Nimotuzumab (TheraCIM)
    Panitumumab Vectibix ® Amgen
    MM-151 Merrimack
    Sym004 Symphogen
    Zalutumumab Humax
    Anti-ErbB3 Therapeutics
    U3-1287/AMG888 U3 Pharma/Amgen
    MM-121 Merrimack Pharmaceuticals
    Anti-ErbB2 Therapeutics
    trastuzumab Herceptin ® Genentech
    HKI-272—neratinib Wyeth
    KOS-953—tanespimycin Kosan Biosciences
    T-DM1—ado-trastuzumab emtansine Kadcyla ® Genentech
    Her/ErbB Dimerization Inhibitors
    2C4, R1273—Pertuzumab Omnitarg ® Genentech, Roche
    Small Molecules Targeting EGFR
    CI-1033 (PD 183805) Pfizer, Inc.
    EKB-569
    Gefitinib IRESSA ™ AstraZeneca
    Lapatinib (GW572016) GlaxoSmithKline
    Lapatinib Ditosylate Tykerb ® SmithKline Beecham
    Erlotinib HCl (OSI-774) Tarceva ® OSI Pharms
    PD158780
    PKI-166 Novartis
    Tyrphostin AG 1478 (4-(3-Chloroanillino)-
    6,7-dimethoxyquinazoline)
    Afatinib (BIBW 2992) Boehringer Ingelheim
    Small Molecules Targeting MEK
    CI-1040 (PD184352)
    AZD6244 (selumetinib)
    RDEA119 (BAY 869766)
    GSK1120212 (trametinib) Glaxo Smith Kline
    PD-0325901
    GDC-0973 Genentech
    UO126-EtOH Cell Signaling Technology
    Anti-cMet Antibody Therapies
    AVEO (AV299) AVEO
    AMG102 Amgen
    5D5 (OA-5D5) Genentech
    Small Molecules Targeting cMet
    PHA665752
    ARQ-650RP ArQule
    ARQ 197 ArQule
    Alkyaling Agents
    BCNU→ 1,3-bis t2-chloroethyl)-
    nitrosourea
    Bendamustine
    Busulfan Myleran GlaxoSmithKline
    Carboplatin Paraplatin Bristol-Myers Squibb
    Carboquone
    Carmustine
    CCNU→ 1,-(2-chloroethyl)-3-cyclohexyl-
    1-nitrosourea (methyl CCNU)
    Chlorambucil Leukeran ® Smithkline Beecham
    Chlormethine
    Cisplatin (Cisplatinum, CDDP) Platinol Bristol-Myers
    Cytoxan Bristol-Myers Squibb
    Cyclophosphamide Neosar Teva Parenteral
    Dacarbazine (DTIC)
    Fotemustine
    Hexamethylmelamine (Altretamine, HMM) Hexalen ® MGI Pharma, Inc.
    Ifosfamide Mitoxana ® ASTA Medica
    Lomustine
    Mannosulfan
    Melphalan Alkeran ® GlaxoSmithKline
    Nedaplatin
    Nimustine
    Oxaliplatin Eloxatin ® Sanofi-Aventis US
    Prednimustine,
    Procarbazine HCL Matulane Sigma-Tau Pharmaceuticals, Inc.
    Ribonucleotide Reductase Inhibitor (RNR)
    Ranimustine
    Satraplatin
    Semustine
    Streptozocin
    Temozolomide
    Treosulfan
    Triaziquone
    Triethylene Melamine
    ThioTEPA Bedford, Abraxis, Teva
    Triplatin tetranitrate
    Trofosfamide
    Uramustine
    Antimetabolites
    5-azacytidine
    Flourouracil (5-FU)/Capecitabine
    6-mercaptopurine
    (Mercaptopurine, 6-MP)
    6-Thioguanine (6-TG) Purinethol ® Teva
    Cytosine Arabinoside (Cytarabine, Thioguanine ® GlaxoSmithKline
    Ara-C)
    Azathioprine Azasan ® AAIPHARMA LLC
    Capecitabine XELODA ® HLR (Roche)
    Cladribine (2-CdA, 2- Leustatin ® Ortho Biotech
    chlorodeoxyadenosine)
    5-Trifluoromethyl-2′-deoxyuridine
    Fludarabine phosphate Fludara ® Bayer Health Care
    Floxuridine (5-fluoro-2) FUDR ® Hospira, Inc.
    Methotrexate sodium Trexall Barr
    Pemetrexed Alimta ® Lilly
    Pentostatin Nipent ® Hospira, Inc.
    Raltitrexed Tomudex ® AstraZeneca
    Tegafur
    Aromatose Inhibitor
    Ketoconazole
    Glucocorticoids
    Dexamethasone Decadron ® Dexasone, Wyeth, Inc.
    Diodex, Hexadrol, Maxidex
    Prednisolone
    Prednisone Deltasone, Orasone, Liquid
    Pred, Sterapred ®
    Immunotherapeutics
    Alpha interferon
    Angiogenesis Inhibitor Avastin ® Genentech
    IL-12→ Interleukin 12
    IL-2→ Interleukin 2 (Aldesleukin) Proleukin ® Chiron
    Receptor Tyrosine Kinase Inhibitors
    AMG 386 Amgen
    Axitinib ((AG-013736) Pfizer, Inc
    Bosutinib (SKI-606) Wyeth
    Brivanib alalinate (BMS-582664) BMS
    Cediranib ( AZD2171) Recentin AstraVeneca
    Dasatinib (BMS-354825) Sprycel ® Bristol-Myers Squibb
    Imatinib mesylate Gleevec Novartis
    Lestaurtinib (CEP-701) Cephalon
    Motesanib diphosphate (AMG-706) Amgen/Takeda
    Nilotinib hydrochloride monohydrate Tasigna ® Novartis
    Pazopanib HCL (GW786034) Armala GSK
    Semaxanib (SU5416) Pharmacia,
    Sorafenib tosylate Nexavar ® Bayer
    Sunitinib malate Sutent ® Pfizer, Inc.
    Vandetanib (AZD647) Zactima AstraZeneca
    Vatalanib; PTK-787 Novartis; Bayer Schering Pharma
    XL184, NSC718781 Exelixis, GSK
    Microtubule-Targeting Agents
    Colchicine
    Docetaxel Taxotere ® Sanofi-Aventis US
    Ixabepilone IXEMPRA ™ Bristol-Myers Squibb
    Larotaxel Sanofi-aventis
    Ortataxel Spectrum Pharmaceuticals
    Nanoparticle paclitaxel (ABI-007) Abraxane ® Abraxis BioScience, Inc.
    Paclitaxel Taxol ® Bristol-Myers Squibb
    Tesetaxel Genta
    Vinblastine sulfate Velban ® Lilly
    Vincristine Oncovin ® Lilly
    Vindesine sulphate Eldisine ® Lilly
    Vinflunine Pierre Fabre
    Vinorelbine tartrate Navelbine ® Pierre Fabre
    mTOR Inhibitors
    Deforolimus (AP23573, MK 8669, ARIAD Pharmaceuticals, Inc
    Ridaforolimus)
    Everolimus (RAD001, RAD001C) Certican ®, Afinitor Novartis
    Sirolimus (Rapamycin) Rapamune ® Wyeth Pharma
    Temsirolimus (CCI-779)
    BEZ235 Torisel ® Wyeth Pharma
    AZD8055
    Protein Synthesis Inhibitor
    L-asparaginase Elspar ® Merck & Co.
    Somatostatin Analogue
    Octreotide acetate Sandostatin ® Novartis
    Topoisomerase Inhibitors
    Actinomycin D
    Camptothecin (CPT)
    Belotecan
    Daunorubicin citrate Daunoxome ® Gilead
    Doxorubicin hydrochloride Doxil ® Alza
    Vepesid ® Bristol-Myers Squibb
    Etoposide Etopophos Hospira, Bedford, Teva Parenteral,
    Etc.
    Irinotecan HCL (CPT-11) Camptosar ® Pharmacia & Upjohn
    Mitoxantrone HCL Novantrone EMD Serono
    Rubitecan
    Teniposide (VM-26) Vumon ® Bristol-Myers Squibb
    Topotecan HCL Hycamtin ® GlaxoSmithKline
    Chemotherapeutic Agents
    Adriamycin, 5-Fluorouracil, Cytoxin,
    Bleomycin, Mitomycin C, Daunomycin,
    Carminomycin, Aminopterin,
    Dactinomycin, Mitomycins, Esperamicins
    Clofarabine, Mercaptopurine, Pentostatin,
    Thioguanine, Cytarabine, Decitabine,
    Floxuridine, Gemcitabine (Gemzar),
    Enocitabine, Sapacitabine
    Hormonal Therapies
    Abarelix Plenaxis ™ Amgen
    Abiraterone acetate CB7630 BTG plc
    Afimoxifene TamoGel Ascend Therapeutics, Inc.
    Anastrazole Arimidex ® AstraZeneca
    Aromatase inhibitor Atamestane plus toremifene Intarcia Therapeutics, Inc.
    Arzoxifene Eli Lilly & Co.
    Asentar; DN 101 Novartis; Oregon Health & Science
    Univ.
    Bicalutamide Casodex ® AstraZeneca
    Buserelin Suprefact ® Sanofi Aventis
    Cetrorelix Cetrotide ® EMD Serono
    Exemestane Aromasin ® Pfizer
    Exemestane Xtane Natco Pharma, Ltd.
    Fadrozole (CGS 16949A )
    Flutamide Eulexin ® Schering
    Flutamide Prostacur Laboratorios Almirall, S.A.
    Fulvestrant Faslodex ® AstraZeneca
    Goserelin acetate Zoladex ® AstraZeneca
    Letrozole Femara ® Novartis
    Letrozole (CGS20267) Femara Chugai Pharmaceutical Co., Ltd.
    Letrozole Estrochek Jagsonpal Pharmaceuticals, Ltd.
    Letrozole Letrozole Indchemie Health Specialities
    Leuprolide acetate Eligard ® Sanofi Aventis
    Leuprolide acetate Leopril VHB Life Sciences, Inc.
    Leuprolide acetate Lupron ®/Lupron Depot TAP Pharma
    Leuprolide acetate Viador Bayer AG
    Megestrol acetate Megace ® Bristol-Myers Squibb
    Magestrol acetate Estradiol Valerate Jagsonpal Pharmaceuticals, Ltd.
    (Delestrogen)
    Medroxyprogesterone acetate Veraplex Combiphar
    MT206 Medisyn Technologies, Inc.
    Nafarelin
    Nandrolone decanoate Zestabolin Mankind Pharma, Ltd.
    Nilutamide Nilandron ® Aventis Pharmaceuticals
    Raloxifene HCL Evista ® Lilly
    Tamoxifen Taxifen Yung Shin Pharmaceutical
    Tamoxifen Tomifen Alkem Laboratories, Ltd.
    Tamoxifen citrate Nolvadex AstraZeneca
    Tamoxifen citrate Soltamox EUSA Pharma, Inc.
    Tamoxifen citrate Tamoxifen citrate Sopharma JSCo.
    SOPHARMA
    Toremifene citrate Fareston ® GTX, Inc.
    Triptorelin pamoate Trelstar ® Watson Labs
    Triptorelin pamoate Trelstar Depot Paladin Labs, Inc.
    Protein Kinase B (PKB) Inhibitors
    Akt Inhibitor ASTEX Astex Therapeutics
    Akt Inhibitors NERVIANO Nerviano Medical Sciences
    AKT Kinase Inhibitor TELIK Telik, Inc.
    AKT Inhibitor Triciribine
    AKT DECIPHERA Deciphera Pharmaceuticals, LLC
    Perifosine (KRX0401, D-21266) Keryx Biopharmaceuticals, Inc.,
    AEtenta Zentaris, Inc.
    Perifosine with Docetaxel Keryx Biopharmaceuticals, Inc.,
    AEtenta Zentaris, Inc.
    Perifosine with Gemcitabine AEtenta Zentaris, Inc.
    Perifosine with Paclitaxel Keryx Biopharmaceuticals, Inc,
    AEtenta Zentaris, Inc.
    Protein Kinase-B inhibitor DEVELOGEN DeveloGen AG
    PX316 Oncothyreon, Inc.
    RX0183 Rexahn Pharmaceuticals, Inc.
    RX0201 Rexahn Pharmaceuticals, Inc.
    VQD002 VioQuest Pharmaceuticals, Inc.
    XL418 Exelixis, Inc.
    ZEN027 AEtenta Zentaris, Inc.
    Phosphatidylinositol 3-Kinase (PI3K)
    Inhibitors
    BEZ235 Novartis AG
    BGT226 Novartis AG
    CAL101 Calistoga Pharmaceuticals, Inc.
    CHR4432 Chroma Therapeutics, Ltd.
    Erk/PI3K Inhibitors ETERNA AEterna Zentaris, Inc.
    GDC0941 Genentech Inc./Piramed
    Limited/Roche Holdings, Ltd.
    Enzastaurin HCL (LY317615) Enzastaurin Eli Lilly
    LY294002/Wortmannin Eli Lilly
    PI3K Inhibitors SEMAFORE Semafore Pharmaceuticals
    PX866 Oncothyreon, Inc.
    SF1126 Semafore Pharmaceuticals
    VMD-8000 VM Discovery, Inc.
    XL147 Exelixis, Inc.
    XL147 with XL647 Exelixis, Inc.
    XL765 Exelixis, Inc.
    PI-103 Roche/Piramed
    BKM120
    Cvelin-dependent kinase inhibitors
    CYC200, r-roscovitine Seliciclib Cyclacel Pharma
    NSC-649890, L86-8275, HMR-1275 Alvocidib NCI
    TLR9, CD289
    IMOxine Merck KGaA
    HYB2055 Idera
    IMO-2055 Isis Pharma
    1018 ISS Dynavax Technologies/UCSF
    PF-3512676 Pfizer
    Enzyme Inhibitor
    Lonafarnib (SCH66336) Sarasar SuperGen, U Arizona
    Anti-TRAIL
    AMG-655 Aeterna Zentaris, Keryx Biopharma
    Apo2L/TRAIL, AMG951 Genentech, Amgen
    Apomab (fully humanized mAb Genentech
    Other
    Imprime PGG Biothera
    CHR-2797 AminopeptidaseM1 Chroma Therapeutics
    E7820, NSC 719239 Integrin-alpha2 Eisai
    INCB007839 ADAM 17, TACE Incyte
    CNF2024, BIIB021 Hsp90 Biogen Idec
    MP470, HPK-56 Kit/Met/Ret Shering-Plough
    SNDX-275/MS-275 HDAC Syndax
    Zarnestra, Tipifarnib, R115777 Ras Janssen Pharma
    Volociximab; Eos 200-4, M200 alpha581 integrin Biogen Idec; Eli Lilly/UCSF/PDL
    BioPharma
    Apricoxib (TP2001) ICOX-2 Inhibitor Daiichi Sankyo; Tragara Pharma

Claims (29)

What is claimed is:
1. A method of treating a subject having a malignant tumor, the method comprising administering to the subject an effective amount a combination therapy comprising and effective amount of each of one or more agents selected from i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor, vi) an mTOR inhibitor, vii) trastuzumab, viii) ado-trastuzumab emtansine, ix) capecitabine, x) cisplatin, and xi) nab-paclitaxel; and an effective amount of an agent that is a bispecific anti-ErbB2/anti-ErbB3 antibody that comprises the amino acid sequence set forth in SEQ ID NO:1.
2. The method of claim 1, wherein the anti-estrogen agent is an estrogen receptor antagonist or an aromatase inhibitor.
3. The method of claim 2, wherein the estrogen receptor antagonist is fulvestrant or tamoxifen.
4. The method of claim 2, wherein the aromatase inhibitor is letrozole, exemestane, anastrozole, aminoglutethimide, testolactone, vorozole, formestane, or fadrozole.
5. The method of claim 4, wherein the aromatase inhibitor is letrozole.
6. The method of claim 1, wherein the administration to the subject of the combination therapy does not create a drug-drug interaction-mediated toxicity in the subject.
7. The method of claim 1, wherein the administration to the subject of the combination therapy creates a substantially additive or superadditive effect as compared to the separate administration of each of the combination therapy agents alone.
8. The method of any one of claims 1 to 7, wherein the receptor tyrosine kinase inhibitor is erlotinib, afatinib, dasatinib, gefitinib, imatinib, pazopinib, lapatinib, sunitinib, nilotinib, or sorafenib.
9. The method of claim 8, wherein the receptor tyrosine kinase inhibitor is lapatinib.
10. The method of any one of claims 1 to 9, further comprising co-administration to the patient of an effective amount of either or both of capecitabine and cisplatin.
11. Use of a bispecific anti-ErbB2/anti-ErbB3 antibody for combination therapy together with one or more agents selected from i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor, vi) an mTOR inhibitor, vii) trastuzumab, viii) ado-trastuzumab emtansine, ix) capecitabine, and x) cisplatini) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor, vi) an mTOR inhibitor, vii) trastuzumab, viii) ado-trastuzumab emtansine, ix) capecitabine, and x) cisplatin; for treatment of a malignant tumor.
12. The use of claim 11, wherein the anti-estrogen agent is an estrogen receptor antagonist or an aromatase inhibitor.
13. The use of claim 12, wherein the estrogen receptor antagonist is fulvestrant or tamoxifen.
14. The use of claim 12, wherein the aromatase inhibitor chosen from the group consisting of letrozole, exemestane, anastrozole, aminoglutethimide, testolactone, vorozole, formestane, and fadrozole.
15. The use of claim 14, wherein the aromatase inhibitor is letrozole.
16. The use of any one of claims 11 to 15, wherein the receptor tyrosine kinase inhibitor is chosen from the group consisting of erlotinib, afatinib, dasatinib, gefitinib, imatinib, pazopinib, lapatinib, sunitinib, nilotinib and sorafenib.
17. The use of claim 16, wherein the receptor tyrosine kinase inhibitor is lapatinib.
18. An aqueous solution comprising a bispecific anti-ErbB2/anti-ErbB3 antibody and one or more of i) an anti-estrogen agent, ii) a receptor tyrosine kinase inhibitor, iii) a MEK inhibitor, iv) a PI3 kinase inhibitor, v) an AKT inhibitor, vi) an mTOR inhibitor, vii) trastuzumab, viii) ado-trastuzumab emtansine, ix) capecitabine, x) cisplatin, and xi) nab-paclitaxel.
19. A method of inhibiting the growth of a malignant tumor comprising tumor cells, said method comprising contacting the tumor cells with the aqueous solution of claim 18.
20. The method of claim 1, wherein said MEK inhibitor is selumetinib, trametinib, UO126, or PD0325901, said PI3K inhibitor is BKM-120 or GDC-0941, and said AKT inhibitor is triciribine or MK-2206.
21. The use of claim 11, wherein said MEK inhibitor is selumetinib, trametinib, UO126, or PD0325901, said PI3K inhibitor is BKM-120 or GDC-0941, and said AKT inhibitor is triciribine or MK-2206.
22. The aqueous solution of claim 18, wherein said MEK inhibitor is selumetinib, trametinib, UO126, or PD0325901, said PI3K inhibitor is BKM-120 or GDC-0941, and said AKT inhibitor is triciribine or MK-2206.
23. The method of claim 1, comprising co-administering the MEK inhibitor and the bispecific anti-ErbB2/anti-ErbB3 antibody.
24. The method of claim 1, comprising co-administering the PI3K inhibitor and the bispecific anti-ErbB2/anti-ErbB3 antibody.
25. The method of claim 1, comprising co-administering the mTOR inhibitor and the bispecific anti-ErbB2/anti-ErbB3 antibody.
26. The method of claim 1, comprising co-administering trastuzumab or ado-trastuzumab emtansine and the bispecific anti-ErbB2/anti-ErbB3 antibody.
27. The method of claim 1, wherein said mTOR inhibitor is BEZ235, AZD8055, everolimus, temsirolimus, sirolimus, or ridaforolimus.
28. The use of claim 11, wherein said mTOR inhibitor is BEZ235, AZD8055, everolimus, temsirolimus, sirolimus, or ridaforolimus.
29. The aqueous solution of claim 18, wherein said mTOR inhibitor is BEZ235, AZD8055, everolimus, temsirolimus, sirolimus, or ridaforolimus.
US14/424,344 2012-08-30 2013-08-30 Combination therapies comprising anti-erbb3 agents Abandoned US20150202287A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/424,344 US20150202287A1 (en) 2012-08-30 2013-08-30 Combination therapies comprising anti-erbb3 agents

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261695242P 2012-08-30 2012-08-30
PCT/US2013/057714 WO2014036520A1 (en) 2012-08-30 2013-08-30 Combination therapies comprising anti-erbb3 agents
US14/424,344 US20150202287A1 (en) 2012-08-30 2013-08-30 Combination therapies comprising anti-erbb3 agents

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/057714 A-371-Of-International WO2014036520A1 (en) 2012-08-30 2013-08-30 Combination therapies comprising anti-erbb3 agents

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/630,452 Continuation US20180066065A1 (en) 2012-08-30 2017-06-22 Combination therapies comprising anti-erbb3 agents

Publications (1)

Publication Number Publication Date
US20150202287A1 true US20150202287A1 (en) 2015-07-23

Family

ID=50184467

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/424,344 Abandoned US20150202287A1 (en) 2012-08-30 2013-08-30 Combination therapies comprising anti-erbb3 agents
US14/015,776 Active US9345766B2 (en) 2012-08-30 2013-08-30 Combination therapies comprising anti-ERBB3 agents
US15/160,523 Abandoned US20160326262A1 (en) 2012-08-30 2016-05-20 Combination therapies comprising anti-erbb3 agents
US15/630,452 Abandoned US20180066065A1 (en) 2012-08-30 2017-06-22 Combination therapies comprising anti-erbb3 agents

Family Applications After (3)

Application Number Title Priority Date Filing Date
US14/015,776 Active US9345766B2 (en) 2012-08-30 2013-08-30 Combination therapies comprising anti-ERBB3 agents
US15/160,523 Abandoned US20160326262A1 (en) 2012-08-30 2016-05-20 Combination therapies comprising anti-erbb3 agents
US15/630,452 Abandoned US20180066065A1 (en) 2012-08-30 2017-06-22 Combination therapies comprising anti-erbb3 agents

Country Status (2)

Country Link
US (4) US20150202287A1 (en)
WO (1) WO2014036520A1 (en)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2009008656A (en) 2007-02-16 2009-11-10 Merrimack Pharmaceuticals Inc Antibodies against erbb3 and uses thereof.
KR20110112301A (en) * 2008-11-18 2011-10-12 메리맥 파마슈티컬즈, 인크. Human serum albumin linkers and conjugates thereof
DK2544680T3 (en) 2010-03-11 2015-04-27 Merrimack Pharmaceuticals Inc USE OF ErbB3 INHIBITORS IN TREATMENT OF TRIPLE-NEGATIVE BREAST CANCER
WO2015100459A2 (en) 2013-12-27 2015-07-02 Merrimack Pharmaceuticals, Inc. Biomarker profiles for predicting outcomes of cancer therapy with erbb3 inhibitors and/or chemotherapies
US10184006B2 (en) 2015-06-04 2019-01-22 Merrimack Pharmaceuticals, Inc. Biomarkers for predicting outcomes of cancer therapy with ErbB3 inhibitors
CA3007644A1 (en) 2015-12-11 2017-06-15 Regeneron Pharmaceuticals, Inc. Methods for reducing or preventing growth of tumors resistant to egfr and/or erbb3 blockade
WO2017102789A1 (en) * 2015-12-17 2017-06-22 F. Hoffmann-La Roche Ag Combination therapy of anti-her3 antibodies and anti-her2 antibodies

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003070234A1 (en) * 2002-02-19 2003-08-28 Case Western Reserve University Alkylating agent combinations in the treatment of cancer
WO2012079093A2 (en) * 2010-12-10 2012-06-14 Merrimack Pharmaceuticals, Inc. Dosage and administration of bispecific scfv conjugates

Family Cites Families (145)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5800815A (en) 1903-05-05 1998-09-01 Cytel Corporation Antibodies to P-selectin and their uses
US4450103A (en) 1982-03-01 1984-05-22 Cetus Corporation Process for recovering human IFN-β from a transformed microorganism
US4588585A (en) 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US4737462A (en) 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4959314A (en) 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US5869620A (en) 1986-09-02 1999-02-09 Enzon, Inc. Multivalent antigen-binding proteins
DE3785186T2 (en) 1986-09-02 1993-07-15 Enzon Lab Inc BINDING MOLECULE WITH SINGLE POLYPEPTIDE CHAIN.
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US4704692A (en) 1986-09-02 1987-11-03 Ladner Robert C Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
DE3856559T2 (en) 1987-05-21 2004-04-29 Micromet Ag Multifunctional proteins with predetermined objectives
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5753627A (en) 1988-12-05 1998-05-19 Novartis Ag Use of certain complexed somatostatin peptides for the invivo imaging of somatostatin receptor-positive tumors and metastasis
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5705363A (en) 1989-03-02 1998-01-06 The Women's Research Institute Recombinant production of human interferon τ polypeptides and nucleic acids
ATE92107T1 (en) 1989-04-29 1993-08-15 Delta Biotechnology Ltd N-TERMINAL FRAGMENTS OF HUMAN SERUM ALBUMIN-CONTAINING FUSION PROTEINS.
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
FR2650598B1 (en) 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
US5730978A (en) 1989-09-01 1998-03-24 Fred Hutchinson Cancer Research Center Inhibition of lymphocyte adherence with α4β1-specific antibodies
US6033665A (en) 1989-09-27 2000-03-07 Elan Pharmaceuticals, Inc. Compositions and methods for modulating leukocyte adhesion to brain endothelial cells
US5292658A (en) 1989-12-29 1994-03-08 University Of Georgia Research Foundation, Inc. Boyd Graduate Studies Research Center Cloning and expressions of Renilla luciferase
US5525491A (en) 1991-02-27 1996-06-11 Creative Biomolecules, Inc. Serine-rich peptide linkers
WO1992015682A1 (en) 1991-02-27 1992-09-17 Creative Biomolecules, Inc. Serine-rich peptide linkers
US6025165A (en) 1991-11-25 2000-02-15 Enzon, Inc. Methods for producing multivalent antigen-binding proteins
JPH07501451A (en) 1991-11-25 1995-02-16 エンゾン・インコーポレイテッド Multivalent antigen binding protein
FR2686620B1 (en) 1992-01-27 1995-06-23 Rhone Poulenc Rorer Sa HUMAN SERUM-ALBUMIN, PREPARATION AND USE.
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
EP1997894B1 (en) 1992-02-06 2011-03-30 Novartis Vaccines and Diagnostics, Inc. Biosynthetic binding protein for cancer marker
WO1994012520A1 (en) 1992-11-20 1994-06-09 Enzon, Inc. Linker for linked fusion polypeptides
SG44845A1 (en) 1993-01-12 1997-12-19 Biogen Inc Recombitant anti-vla4 antibody molecules
CN1074243A (en) 1993-02-06 1993-07-14 北京中化生物技术研究所 Interleukin 6-interleukin-22 fusion rotein and method for making and purposes
US5780594A (en) 1993-03-01 1998-07-14 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Biologically active protein fragments containing specific binding regions of serum albumin or related proteins
US5928904A (en) 1993-09-07 1999-07-27 Smithkline Beecham Corporation DNA encoding recombinant IL4 antibodies useful in treatment of IL4 mediated disorders
US5914110A (en) 1993-09-07 1999-06-22 Smithkline Beecham Corporation Recombinant IL4 antibodies useful in treatment of IL4 mediated disorders
US5733782A (en) 1993-10-25 1998-03-31 Creative Biomolecules, Inc. Methods and compositions for high protein production from non-native DNA
US5821231A (en) 1993-12-06 1998-10-13 Cytel Corporation CS-1 peptidomimetics, compositions and methods of using same
US5936065A (en) 1993-12-06 1999-08-10 Cytel Corporation CS-1 peptidomimetics, compositions and methods of using the same
US5840299A (en) 1994-01-25 1998-11-24 Athena Neurosciences, Inc. Humanized antibodies against leukocyte adhesion molecule VLA-4
US8715645B2 (en) 1994-05-27 2014-05-06 The Regents Of The University Of Colorado Viral vectors encoding apoptosis-inducing proteins and methods for making and using the same
US5622701A (en) 1994-06-14 1997-04-22 Protein Design Labs, Inc. Cross-reacting monoclonal antibodies specific for E- and P-selectin
US5811391A (en) 1994-08-25 1998-09-22 Cytel Corporation Cyclic CS-1 peptidomimetics, compositions and methods of using same
US5874540A (en) 1994-10-05 1999-02-23 Immunomedics, Inc. CDR-grafted type III anti-CEA humanized mouse monoclonal antibodies
US5763733A (en) 1994-10-13 1998-06-09 Enzon, Inc. Antigen-binding fusion proteins
US5670356A (en) 1994-12-12 1997-09-23 Promega Corporation Modified luciferase
US6306840B1 (en) 1995-01-23 2001-10-23 Biogen, Inc. Cell adhesion inhibitors
US5656730A (en) 1995-04-07 1997-08-12 Enzon, Inc. Stabilized monomeric protein compositions
EP2258726A1 (en) 1995-06-14 2010-12-08 The Regents of the University of California High affinity human antibodies to c-erbB-2
GB9526733D0 (en) 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US5968511A (en) 1996-03-27 1999-10-19 Genentech, Inc. ErbB3 antibodies
US6686350B1 (en) 1996-07-25 2004-02-03 Biogen, Inc. Cell adhesion inhibitors
US6423512B1 (en) 1996-07-26 2002-07-23 Novartis Ag Fusion polypeptides
MY120425A (en) 1996-07-26 2005-10-31 Novartis Ag Fusion polypeptides
WO1998048837A1 (en) 1997-04-30 1998-11-05 Enzon, Inc. Polyalkylene oxide-modified single chain polypeptides
US6191269B1 (en) 1997-05-30 2001-02-20 The Regents Of The University Of California Selective induction of cell death by delivery of amino-terminal interleukin-1-α pro-piece polypeptide
EP0991619B1 (en) 1997-06-23 2003-09-10 Tanabe Seiyaku Co., Ltd. Inhibitors of alpha 4-beta 1 mediated cell adhesion
US6455550B1 (en) 1997-08-22 2002-09-24 Hoffmann-La Roche Inc. N-alkanoylphenylalanine derivatives
US6229011B1 (en) 1997-08-22 2001-05-08 Hoffman-La Roche Inc. N-aroylphenylalanine derivative VCAM-1 inhibitors
US6197794B1 (en) 1998-01-08 2001-03-06 Celltech Therapeutics Limited Phenylalanine derivatives
US6329372B1 (en) 1998-01-27 2001-12-11 Celltech Therapeutics Limited Phenylalanine derivatives
US6171809B1 (en) 1998-01-29 2001-01-09 Packard Instrument Company Method and compositions for detecting luciferase biological samples
WO1999051743A1 (en) 1998-04-03 1999-10-14 Chugai Seiyaku Kabushiki Kaisha Humanized antibody against human tissue factor (tf) and process for constructing humanized antibody
GB9811159D0 (en) 1998-05-22 1998-07-22 Celltech Therapeutics Ltd Chemical compounds
EE04639B1 (en) 1998-05-28 2006-06-15 Biogen, Inc. A cell adhesion inhibiting compound and a pharmaceutical composition comprising it
GB9812088D0 (en) 1998-06-05 1998-08-05 Celltech Therapeutics Ltd Chemical compounds
US6685617B1 (en) 1998-06-23 2004-02-03 Pharmacia & Upjohn Company Inhibitors of α4β1 mediated cell adhesion
US6355678B1 (en) 1998-06-29 2002-03-12 Parker Hughes Institute Inhibitors of the EGF-receptor tyrosine kinase and methods for their use
GB9821061D0 (en) 1998-09-28 1998-11-18 Celltech Therapeutics Ltd Chemical compounds
ES2257884T3 (en) 1998-10-16 2006-08-01 Biogen Idec Ma Inc. CONJUGADOS DE POLIMERO AND INTERFERON BETA-1A AND ITS USES.
US6333396B1 (en) 1998-10-20 2001-12-25 Enzon, Inc. Method for targeted delivery of nucleic acids
JP3180906B2 (en) 1998-11-12 2001-07-03 日本電気株式会社 Manufacturing method of magnetoresistive composite head
PT1154993E (en) 1999-02-18 2005-01-31 Hoffmann La Roche THIOAMIDE DERIVATIVES
US6265572B1 (en) 1999-04-20 2001-07-24 Hoffmann-La Roche Inc. Pyrrolidincarbonylamino cyclic disulfide anti-inflammatory agents
US7534866B2 (en) 2005-10-19 2009-05-19 Ibc Pharmaceuticals, Inc. Methods and compositions for generating bioactive assemblies of increased complexity and uses
US6756378B2 (en) 1999-06-30 2004-06-29 Pharmacopeia Drug Discovery, Inc. VLA-4 inhibitor compounds
US7067313B1 (en) 1999-07-14 2006-06-27 D. Collen Research Foundation Ligands for use in therapeutic compositions for the treatment of hemostasis disorders
US6365619B1 (en) 1999-07-22 2002-04-02 Novartis Ag Treatment of arteriosclerosis
PL354063A1 (en) 1999-08-13 2003-12-15 Biogen, Inc.Biogen, Inc. Cell adhesion inhibitors
US6531122B1 (en) 1999-08-27 2003-03-11 Maxygen Aps Interferon-β variants and conjugates
EP1094110A1 (en) 1999-10-21 2001-04-25 Leadd B.V. Apoptosis inducing proteinaceous substance
WO2001036376A1 (en) 1999-11-18 2001-05-25 Ajinomoto Co., Inc. Novel phenylalanine derivatives
US6380387B1 (en) 1999-12-06 2002-04-30 Hoffmann-La Roche Inc. 4-Pyrimidinyl-n-acyl-l phenylalanines
US6388084B1 (en) 1999-12-06 2002-05-14 Hoffmann-La Roche Inc. 4-pyridinyl-n-acyl-l-phenylalanines
DE19962936A1 (en) 1999-12-24 2001-06-28 Bayer Ag New beta-aminoacid amide derivatives, are integrin antagonists useful for treating inflammatory, autoimmune or immunological disorders e.g. asthma, diabetes or rheumatoid arthritis
WO2001051487A1 (en) 1999-12-28 2001-07-19 Pfizer Products Inc. Non-peptidyl inhibitors of vla-4 dependent cell binding useful in treating inflammatory, autoimmune, and respiratory diseases
US7446174B2 (en) 2001-03-02 2008-11-04 Xencor, Inc. Protein based TNF-α variants for the treatment of TNF-α related disorders
WO2001070670A1 (en) 2000-03-23 2001-09-27 Ajinomoto Co., Inc. Novel phenylalanine derivative
US6946134B1 (en) 2000-04-12 2005-09-20 Human Genome Sciences, Inc. Albumin fusion proteins
WO2001079442A2 (en) 2000-04-12 2001-10-25 Human Genome Sciences, Inc. Albumin fusion proteins
US20050100991A1 (en) 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
EP1289552B1 (en) 2000-05-19 2006-03-29 The Center for Blood Research, INC. Methods for treating hemostatic disorders by soluble p-selectin
GB0027328D0 (en) 2000-06-23 2000-12-27 Aventis Pharma Inc Bioengineered vehicles for targeted nucleic acid delivery
US6787636B1 (en) 2000-07-14 2004-09-07 New Century Pharmaceuticals, Inc. Modified serum albumin with reduced affinity for nickel and copper
US20040247565A1 (en) 2000-07-19 2004-12-09 Chih-Ping Liu Method of treatment using interferon-tau
US7015216B2 (en) 2000-07-21 2006-03-21 Elan Pharmaceuticals, Inc. Heteroaryl-β-alanine derivatives as alpha 4 integrin inhibitors
US6689781B2 (en) 2000-07-21 2004-02-10 Elan Pharmaceuticals, Inc. Phenylalanine derivatives as alpha 4 integrin inhibitors
BRPI0113331B8 (en) 2000-08-18 2021-05-25 Ajinomoto Kk phenylalanine derivatives or their pharmaceutically acceptable salts, alpha 4 integrin antagonist, therapeutic agent or preventive agent for inflammatory diseases, and pharmaceutical composition
WO2002022563A1 (en) 2000-09-14 2002-03-21 Toray Industries, Inc. Urea derivative and adhesive-molecule inhibitor containing the same as active ingredient
DE60122555T2 (en) 2000-09-25 2007-07-12 Toray Industries, Inc. SPIRO COMPOUNDS AND THESE ADHESION MOLECULAR HIBITORS CONTAINING ACTIVE INGREDIENTS
US6818216B2 (en) 2000-11-28 2004-11-16 Medimmune, Inc. Anti-RSV antibodies
SK286991B6 (en) 2001-02-22 2009-09-07 Celltech R&D Limited Phenylalanine enamide derivatives, pharmaceutical composition containing the compounds and the use of them
ES2180416B1 (en) 2001-03-12 2004-06-01 BIOTOOLS BIOTECHNOLOGICAL &amp; MEDICAL LABORATORIES, S.A. PROCEDURE FOR THE PREPARATION OF STABILIZED REACTION MIXTURES, TOTAL OR PARTIALLY DESIRED, THAT INCLUDE, AT LEAST, ONE ENZYME, REACTION MIXES AND KITS CONTAINING THEM.
US20090226429A1 (en) 2001-05-25 2009-09-10 Human Genome Sciences, Inc. Antibodies That Immunospecifically Bind to TRAIL Receptors
WO2003007889A2 (en) 2001-07-17 2003-01-30 Research Development Foundation Therapeutic agents comprising pro-apoptotic proteins
JP4164871B2 (en) 2001-07-26 2008-10-15 味の素株式会社 Novel phenylpropionic acid derivatives
AU2002326531A1 (en) * 2001-08-03 2003-02-17 The Trustees Of The University Of Pennsylvania Monoclonal antibodies to activated erbb family members and methods of use thereof
US6900292B2 (en) 2001-08-17 2005-05-31 Lee-Hwei K. Sun Fc fusion proteins of human erythropoietin with increased biological activities
EP1454898A4 (en) 2001-12-13 2006-12-13 Ajinomoto Kk Novel phenylalanine derivative
KR101271635B1 (en) 2001-12-21 2013-06-12 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
CA2484556A1 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
JP5424521B2 (en) 2001-12-21 2014-02-26 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド Albumin fusion protein
AU2003207464A1 (en) 2002-01-04 2003-07-24 Xencor Dominant negative proteins and methods thereof
CN1656095A (en) 2002-03-22 2005-08-17 东丽株式会社 Spiro derivatives and adhesion molecule inhibitors comprising the same as the active ingredient
US7332585B2 (en) 2002-04-05 2008-02-19 The Regents Of The California University Bispecific single chain Fv antibody molecules and methods of use thereof
US7332580B2 (en) 2002-04-05 2008-02-19 The Regents Of The University Of California Bispecific single chain Fv antibody molecules and methods of use thereof
GB0216571D0 (en) 2002-07-17 2002-08-28 Celltech R&D Ltd Chemical compounds
GB0216568D0 (en) 2002-07-17 2002-08-28 Celltech R&D Ltd Chemical compounds
US7314613B2 (en) 2002-11-18 2008-01-01 Maxygen, Inc. Interferon-alpha polypeptides and conjugates
US20040115204A1 (en) 2002-12-11 2004-06-17 Fanger Gary R. Antibodies to treat cancer
WO2004082640A2 (en) 2003-03-19 2004-09-30 New Century Pharmaceuticals, Inc. Human serum albumin conjugates with therapeutic compounds
US7446125B2 (en) 2003-04-09 2008-11-04 Universidad Del Pais Vasco Derivatives of nitroproline compounds, processes of manufacture and uses in the treatment of integrin mediated disorders
WO2005035719A2 (en) 2003-10-03 2005-04-21 Board Of Control Of Michigan Technological University Modified luciferase
US7638331B2 (en) 2004-01-02 2009-12-29 The Administration of the Tulane Rducation Fund Directed apoptosis in COX-2 overexpressing cancer cells through expression targeted gene delivery
EP2360186B1 (en) 2004-04-13 2017-08-30 F. Hoffmann-La Roche AG Anti-P-selectin antibodies
WO2005117973A2 (en) 2004-05-05 2005-12-15 Merrimack Pharmaceuticals, Inc. Bispecific binding agents for modulating biological activity
US8921528B2 (en) 2004-06-01 2014-12-30 Domantis Limited Bispecific fusion antibodies with enhanced serum half-life
TW200610754A (en) 2004-06-14 2006-04-01 Daiichi Seiyaku Co Vla-4 inhibitor
US7196112B2 (en) 2004-07-16 2007-03-27 Biogen Idec Ma Inc. Cell adhesion inhibitors
US20060018859A1 (en) 2004-07-16 2006-01-26 Carter Daniel C Modified human serum albumin with reduced or eliminated affinity to chemical or biological contaminants at Cys 34
CN101072581A (en) 2004-10-05 2007-11-14 健泰科生物技术公司 Therapeutic agents with decreased toxicity
AP2007004034A0 (en) 2004-12-31 2007-06-30 Genentech Inc Polypeptides sthat bind br3 and uses thereof
CN101163501A (en) 2005-02-23 2008-04-16 梅里麦克制药股份有限公司 Bispecific binding agents for modulating biological activity
US20070243163A1 (en) 2006-02-17 2007-10-18 Chih-Ping Liu Respiratory tract delivery of interferon-tau
WO2008014493A2 (en) 2006-07-28 2008-01-31 Drc Computer Corporation Configurable processor module accelerator using a progrmmable logic device
MX2009002523A (en) 2006-09-08 2009-03-20 Ambrx Inc Modified human plasma polypeptide or fc scaffolds and their uses.
AU2007294805A1 (en) 2006-09-14 2008-03-20 Human Genome Sciences, Inc. Albumin fusion proteins
US8580263B2 (en) 2006-11-21 2013-11-12 The Regents Of The University Of California Anti-EGFR family antibodies, bispecific anti-EGFR family antibodies and methods of use thereof
WO2008076949A2 (en) 2006-12-15 2008-06-26 Concert Pharmaceuticals Inc. Quinazoline derivatives and methods of treatment
CN102057054B (en) * 2008-04-11 2015-06-10 梅里麦克制药股份有限公司 Human serum albumin linkers and conjugates thereof
KR20110112301A (en) 2008-11-18 2011-10-12 메리맥 파마슈티컬즈, 인크. Human serum albumin linkers and conjugates thereof
WO2011133668A2 (en) * 2010-04-20 2011-10-27 President And Fellows Of Harvard College Methods and compositions for the treatment of cancer
US9238080B2 (en) 2010-05-21 2016-01-19 Merrimack Pharmaceuticals, Inc. Bi-specific fusion proteins

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003070234A1 (en) * 2002-02-19 2003-08-28 Case Western Reserve University Alkylating agent combinations in the treatment of cancer
WO2012079093A2 (en) * 2010-12-10 2012-06-14 Merrimack Pharmaceuticals, Inc. Dosage and administration of bispecific scfv conjugates

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ClinicalTrials.gov Archive (NCT01304784, 2/24/2011) *
Lu et al (Clinical Cancer Research, 2007, Vol. 13, pp. 5883-5888) *

Also Published As

Publication number Publication date
US20180066065A1 (en) 2018-03-08
US20140079703A1 (en) 2014-03-20
US20160326262A1 (en) 2016-11-10
US9345766B2 (en) 2016-05-24
WO2014036520A1 (en) 2014-03-06

Similar Documents

Publication Publication Date Title
US20140056898A1 (en) Combination therapies comprising anti-erbb3 agents
US20180066065A1 (en) Combination therapies comprising anti-erbb3 agents
EP2729172B1 (en) Antibodies against epidermal growth factor receptor (egfr) and uses thereof
AU2012257727B2 (en) Method for EGFR directed combination treatment of cancer
EP2815765A1 (en) Overcoming resistance to ERBB pathway inhibitors
WO2011140254A1 (en) Antibodies against epidermal growth factor receptor (egfr) and uses thereof
US20170314079A1 (en) Antibodies against epidermal growth factor receptor (egfr) and uses thereof
US20190336599A1 (en) Combination of ramucirumab and merestinib for use in treatment of colorectal cancer
US20230321087A1 (en) Fgfr inhibitor combination therapies
TW202224682A (en) Methods and compositions comprising a krasg12c inhibitor and a vegf inhibitor for treating solid tumors
WO2024003837A1 (en) Use of anti-egfr/anti-met antibody to treat gastric or esophageal cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: MERRIMACK PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZHANG, BO;MCDONAGH, CHARLOTTE;HUHALOV, ALEXANDRA;SIGNING DATES FROM 20150429 TO 20150904;REEL/FRAME:036685/0402

STCB Information on status: application discontinuation

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