WO2013063001A1 - Therapeutic combinations and methods of treating melanoma - Google Patents

Therapeutic combinations and methods of treating melanoma Download PDF

Info

Publication number
WO2013063001A1
WO2013063001A1 PCT/US2012/061533 US2012061533W WO2013063001A1 WO 2013063001 A1 WO2013063001 A1 WO 2013063001A1 US 2012061533 W US2012061533 W US 2012061533W WO 2013063001 A1 WO2013063001 A1 WO 2013063001A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
etbr
mpk
seq
inhibitor
Prior art date
Application number
PCT/US2012/061533
Other languages
French (fr)
Inventor
Paul Polakis
Jyoti Asundi
Suzanna CLARK
Original Assignee
Genentech, Inc.
F. Hoffmann-La Roche Ag
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
Priority to CN201280065175.XA priority Critical patent/CN104039340B/en
Priority to EA201490879A priority patent/EA201490879A1/en
Priority to SG11201401815XA priority patent/SG11201401815XA/en
Priority to KR1020147013878A priority patent/KR20140097205A/en
Application filed by Genentech, Inc., F. Hoffmann-La Roche Ag filed Critical Genentech, Inc.
Priority to CA2850034A priority patent/CA2850034A1/en
Priority to US14/354,362 priority patent/US20140341916A1/en
Priority to JP2014538892A priority patent/JP6251682B2/en
Priority to AU2012328980A priority patent/AU2012328980A1/en
Priority to BR112014009953A priority patent/BR112014009953A2/en
Priority to EP12842985.9A priority patent/EP2776051A4/en
Priority to IN3062CHN2014 priority patent/IN2014CN03062A/en
Priority to MX2014004991A priority patent/MX2014004991A/en
Publication of WO2013063001A1 publication Critical patent/WO2013063001A1/en
Priority to IL232135A priority patent/IL232135A0/en
Priority to MA37028A priority patent/MA35645B1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/437Heterocyclic 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 five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • C07K16/2869Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against hormone receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3053Skin, nerves, brain
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule

Definitions

  • This invention concerns in general, a treatment of melanoma by using certain antibody and small molecule drug combinations.
  • melanomas can demonstrate molecular variations, for example in certain signal transduction pathways necessary for cell responsiveness to growth factors. Therefore, rather than treating melanoma as a single disease, at have been made to stratify it into molecular subtypes in order to treat each subtype with the most appropriate therapies.
  • the MAPK pathway is a phosphorylation-driven signal transduction cascade that couples intracellular responses to the binding of growth factors to cell surface receptors. This pathway regulates several processes including cell proliferation and differentiation, and is often dysregulated in a variety of cancers. (Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer. 2004;4:937-947).
  • the classical MAPK pathway consists of RAS, RAF, MEK and ERK, where RAS triggers the formation of a RAF/MEK/ERK kinase complex which then drives transcription of key regulators through protein phosphorylation.
  • RAS triggers the formation of a RAF/MEK/ERK kinase complex which then drives transcription of key regulators through protein phosphorylation.
  • the inhibition of MAPK signaling with agents targeted toward critical proteins in the pathway has the potential to inhibit growth in a variety of tumor types (Wong K-K et al., Recent developments in anti-cancer agents targeting the Ras/Raf/MEK/ERK pathway. Recent Pat Anticancer Drug Discov. 2009;4:28-35.
  • GDC-0973 (a.k.a. XL518) is a potent and highly selective small molecule inhibitor of MEK1/2, a MAPK kinase that activates ERK1/2 (Johnston S. XL518, a potent, selective, orally bioavailable MEK1 inhibitor, downregulates the Ras/Raf/MEK/ERK pathway in vivo, resulting in tumor growth inhibition and regression in preclinical models.
  • AACR-NCI-EORTC Symposium on Molecular Targets and Cancer Therapeutics October 22, 2007; San Francisco, CA. Abstract C209.
  • vemurafenib also known as Zelboraf®, which is a B-Raf enzyme inhibitor
  • Zelboraf® which is a B-Raf enzyme inhibitor
  • Vemurafenib has been shown to cause programmed cell death in melanoma cell lines (Sala E, et al., BRAF silencing by short hairpin RNA or chemical blockade by PLX4032 leads to different responses in melanoma and thyroid carcinoma cells. Mol. Cancer Res. 6 (5): 751-9 (May 2008).
  • Vemurafenib interrupts the B-Raf/MEK step on the B-Raf/MEK/ERK pathway if the B-Raf has the common V600E mutation.
  • Vemurafenib is effective in melanoma patients whose cancer has a V600E BRAF mutation (that is, at amino acid position number 600 on the B-RAF protein, the normal valine is replaced by glutamic acid). About 60% of melanomas have the V600E BRAF mutation.
  • the present invention contemplates a method of tumor growth inhibition (TGI) in a subject suffering from melanoma comprising administering to the subject an effective amount of an anti- endothelin B receptor (ETBR) antibody drug conjugate in combination with an effective amount of a MAP kinase inhibitor.
  • TGI tumor growth inhibition
  • the combination of an anti-ETBR antibody drug conjugate and a MAP kinase inhibitor is synergistic.
  • the TGI is greater than the TGI seen using an anti-ETBR antibody drug conjugate alone or greater than the TGI seen using a MAP kinase inhibitor alone.
  • the TGI is about 10% greater, or about 15%> greater, or about 20% greater, or about 25% greater, or about 30% greater, or about 35% greater, or about 40% greater, or about 45% greater, or about 50% greater, or about 55%o greater, or about 60% greater, or about 65% greater, or about 70% greater than use of an anti-ETBR antibody drug conjugate alone or the TGI is about 10% greater, or about 15% greater, or about 20% greater, or about 25% greater, or about 30% greater, or about 35% greater, or about 40% greater, or about 45%o greater, or about 50% greater, or about 55%> greater, or about 60% greater, or about 65%> greater, or about 70%) greater than use of a MAP kinase inhibitor alone.
  • the anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
  • the anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDR1 is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDR1 is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
  • the anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9.
  • a further aspect of this method also includes an anti-ETBR antibody also having a VL which is SEQ ID NO: 8.
  • the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is a cytotoxic agent that is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes, and wherein said cytotoxin is a toxin.
  • the toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin.
  • the toxin is a maytansinoid.
  • the MAP kinase inhibitor is a BRAF inhibitor.
  • the BRAF inhibitor is propane- 1- sulfonic acid ⁇ 3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -amide.
  • the BRAF inhibitor has the following chemical structure:
  • the MAP kinase inhibitor is a MEK inhibitor.
  • the MEK inhibitor is (S)-(3,4- difuoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone.
  • the MEK inhibitor has the following chemical structure:
  • a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody is described.
  • said melanoma is ETBR positive.
  • said melanoma is metastatic.
  • said subject has not had prior therapy with a MAP kinase inhibitor.
  • said subject has a V600E BRAF gene mutation or said subject is BRAF wildtype, having no V600E BRAF mutation.
  • the subject has not had prior therapy with a MAP kinase inhibitor.
  • the combination of an anti-ETBR antibody and a MAP kinase inhibitor is synergistic.
  • the TGI is greater than the TGI seen using an anti-ETBR antibody alone or greater than the TGI seen using a MAP kinase inhibitor alone.
  • the TGI is about 10% greater, or about 15%> greater, or about 20% greater, or about 25% greater, or about 30% greater, or about 35% greater, or about 40% greater, or about 45% greater, or about 50%) greater, or about 55% greater, or about 60% greater, or about 65% greater, or about 70% greater than use of an anti-ETBR antibody alone or the TGI is about 10% greater, or about 15% greater, or about 20% greater, or about 25% greater, or about 30% greater, or about 35% greater, or about 40% greater, or about 45%) greater, or about 50% greater, or about 55%) greater, or about 60%) greater, or about 65% greater, or about 70%) greater than use of a MAP kinase inhibitor alone.
  • the anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
  • the anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDR1 is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDR1 is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
  • the anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9.
  • a further aspect of this method also includes an anti-ETBR antibody also having a VL which is SEQ ID NO: 8.
  • the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes, and wherein said cytotoxin is a toxin.
  • the toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin.
  • the toxin is a maytansinoid.
  • the MAP kinase inhibitor is a BRAF inhibitor.
  • the BRAF inhibitor is propane- 1 -sulfonic acid ⁇ 3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -amide.
  • the BRAF inhibitor has the following chemical structure:
  • the MAP kinase inhibitor is a MEK inhibitor.
  • the MEK inhibitor is (S)-(3,4- difuoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone.
  • the MEK inhibitor has the following chemical structure:
  • a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein the MAP kinase inhibitor is administered first to said subject in need thereof.
  • said anti-ETBR antibody drug conjugate is administered after administration of said MAP kinase inhibitor.
  • contemplated methods of the invention include where the anti-ETBR antibody and the MAP kinase inhibitor are administered simultaneously.
  • a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein the anti-ETBR antibody drug conjugate and the MAP kinase inhibitor are administered sequentially, wherein the anti-ETBR antibody drug conjugate is administered to the subject first and the MAP kinase inhibitor is administered to the subject after administration of the anti-ETBR antibody drug conjugate.
  • a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein the MAP kinase inhibitor is administered to the subject first and the anti-ETBR antibody drug conjugate is administered to the subject after administration of the MAP kinase inhibitor.
  • a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein said anti-ETBR antibody drug conjugate is administered intraveneously.
  • the anti-ETBR antibody drug conjugate is dosed at about 0.1 mpk, or about 0.2 mpk, or about 0.3 mpk, or about 0.5 mpk, or about 1 mpk, or about 5 mpk, or about 10 mpk, or about 15 mpk, or about 20 mpk, or about 25 mpk, or about 30 mpk in the claimed methods of the invention.
  • a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein the MAP kinase inhibitor is administered orally.
  • the BRAF inhibitor is dosed at about 1 mpk, or about 2 mpk, or about 3 mpk, or about 4 mpk, or about 5 mpk, or about 6 mpk, or about 7 mpk, or about 8 mpk, or about 9 mpk, or about 10 mpk, or about 11 mpk, or about 12 mpk, or about 15 mpk, or about 20 mpk or about 30 mpk in the claimed methods of the invention.
  • an article of manufacture is used for TGI in a subject suffering from melanoma comprising a package comprising an anti-ETBR antibody drug conjugate composition and a MAP kinase inhibitor composition.
  • said anti- ETBR antibody drug conjugate specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
  • said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDRl is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDRl is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
  • an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9.
  • an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO: 7 or 9 and the VL is SEQ ID NO: 8.
  • the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes.
  • the cytotoxin is a toxin wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin. In one aspect, the toxin is a maytansinoid.
  • the MAP kinase inhibitor is a BRAF inhibitor.
  • the BRAF inhibitor is propane- 1 -sulfonic acid ⁇ 3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3- carbonyl]-2,4-difluoro-phenyl ⁇ -amide.
  • the BRAF inhibitor has the following chemical structure:
  • the MAP kinase inhibitor is a MEK inhibitor.
  • the MEK inhibitor is (S)-(3,4- difuoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone.
  • the MEK inhibitor has the following chemical structure:
  • an article of manufacture for treating melanoma in a subject comprising a package comprising an anti-ETBR antibody drug conjugate composition and a MAP kinase inhibitor composition. It is further contemplated that said anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
  • said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDRl is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDRl is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
  • VH CDRl is SEQ ID NO:1
  • VH CDR2 is SEQ ID NO:3
  • VL CDR2 is SEQ ID NO:5
  • VL CDR3 is SEQ ID NO:6.
  • an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9.
  • an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO: 7 or 9 and the VL is SEQ ID NO: 8.
  • the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes.
  • the cytotoxin is a toxin wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin.
  • the toxin is a maytansinoid.
  • the MAP kinase inhibitor is a BRAF inhibitor.
  • the BRAF inhibitor is propane- 1 -sulfonic acid ⁇ 3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3- carbonyl]-2,4-difluoro-phenyl ⁇ -amide.
  • the BRAF inhibitor has the following chemical structure:
  • the MAP kinase inhibitor is a MEK inhibitor.
  • the MEK inhibitor is (S)-(3,4-dimoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l - yl)methanone.
  • the MEK inhibitor has the following chemical structure:
  • one aspect of the invention is use of an anti-ETBR antibody drug conjugate and a MAP kinase inhibitor in the preparation of a medicament for TGI of a melanoma. It is further contemplated that said anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
  • said anti- ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDR1 is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDR1 is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
  • VH CDR1 is SEQ ID NO: l
  • VH CDR2 is SEQ ID NO:2
  • VH CDR3 is SEQ ID NO:3
  • VL CDR1 is SEQ ID NO:4
  • VL CDR2 is SEQ ID NO:5
  • VL CDR3 is SEQ ID NO:6.
  • an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9.
  • an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO: 7 or 9 and the VL is SEQ ID NO: 8.
  • the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes.
  • the cytotoxin is a toxin wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin.
  • the toxin is a maytansinoid.
  • the MAP kinase inhibitor is a BRAF inhibitor.
  • the BRAF inhibitor is propane- 1 -sulfonic acid ⁇ 3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3- carbonyl]-2,4-difluoro-phenyl ⁇ -amide.
  • the BRAF inhibitor has the following chemical structure:
  • the MAP kinase inhibitor is a MEK inhibitor.
  • the MEK inhibitor is (S)-(3,4-dimoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l - yl)methanone.
  • the MEK inhibitor has the following chemical structure:
  • one aspect of the invention is use of an article of manufacture comprising an anti-ETBR antibody drug conjugate composition and a MAP kinase inhibitor composition in the preparation of a medicament for TGI of a melanoma. It is further contemplated that said anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
  • said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDRl is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDRl is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
  • VH CDRl is SEQ ID NO:1
  • VH CDR2 is SEQ ID NO:3
  • VL CDR2 is SEQ ID NO:5
  • VL CDR3 is SEQ ID NO:6.
  • an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9.
  • an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO: 7 or 9 and the VL is SEQ ID NO: 8.
  • the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes.
  • the cytotoxin is a toxin wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin.
  • the toxin is a maytansinoid.
  • the MAP kinase inhibitor is a BRAF inhibitor.
  • the BRAF inhibitor is propane- 1 -sulfonic acid ⁇ 3-[5-(4-chlorophenyl)-lH- pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl ⁇ -amide. Further, it is contemplated that the BRAF inhibitor has the following chemical structure:
  • the MAP kinase inhibitor is a MEK inhibitor.
  • the MEK inhibitor is (S)-(3,4-difuoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3- (piperidin-2yl)azetidin-l -yl)methanone.
  • the MEK inhibitor has the following chemical structure:
  • Figure 1 is a schematic of the MAP kinase pathway.
  • Figure 2 demonstrates the relationship of receptor level to ADC cell killing in vitro.
  • the indicated number of receptor copies/cell was estimated by Scatchard analysis.
  • Panel A shows cell killing by anti-ET B R ADC titration for the melanoma cell line UACC-257X2.2 and panel B for melanoma cell line A2058.
  • the indicated concentrations of anti-ET B R ADC Hu5E9vl-vc-MMAE
  • control IgG-vc- MMAE or equivalent amount of PBS vehicle control were incubated with cells for 5 days and relative cell viability (y-axis) assessed using CellTiter-Glo.
  • Figure 3 shows the in vivo efficacy of anti-ETBR ADC in xenografts mouse models.
  • Subcutaneous tumors were established in mice inoculated with UACC-257X2.2 (Panel A) or A2058 (Panel B) cells. When tumor volumes reached approximately 200 mm 3 (day 0), animals were given a single IV injection of either control ADC (Control-vc-MMAE) or anti-ETBR ADC (Hu5E9vl -vc- MMAE) at the indicated doses. Average tumor volumes with standard deviations were determined from 10 animals per groups (indicated on graph).
  • FIG. 4 shows ET B R expression in UACC-257X2.2 melanoma cells treated for 24h with varying concentrations of BRAFi-945.
  • Panel A shows ETBR transcript normalized to RPL19 transcript.
  • Panel B shows the expression of total ETBR and GAPDH (Control) protein in 50 ⁇ g whole cell lysates.
  • Panel C shows surface ETBR protein expression in live cells as seen by flow cytometry, where the first peak indicates cells treated to secondary detection reagent alone, the middle peak indicates cells untreated with BRAF inhibitor, and the last peak indicates BRAF inhibitor treated cells.
  • Figure 5 shows in vivo combination efficacy of anti-ET B R ADC (Hu5E9vl-vc-MMAE) and BRAFi-945 against UACC-257X2.2 melanoma xenograft mouse models at varying doses.
  • Subcutaneous tumors were established in mice inoculated with UACC -257X2.2 cell lines. When tumor volumes reached approximately 200 mm 3 (day 0), animals were dosed orally once a day for 21 days with BRAFi- 945 or vehicle control. On day 1 (after two doses of BRAFi-945), animals were given a single IV injection of either vehicle or anti-ET B R ADC at the indicated doses. Average tumor volumes with standard deviations were determined from 10 animals per group.
  • Panel A shows a 1 mpk BRAFi-945 and 1 mpk anti-ET B R-ADC (Hu5E9vl -vc-MMAE) combination
  • panel B shows a 1 mpk BRAFi-945 and 3 mpk anti-ET B R-ADC (Hu5E9vl -vc-MMAE) combination
  • panel C shows a 6 mpk BRAFi-945 and 1 mpk anti-ET B R-ADC (Hu5E9vl -vc-MMAE) combination
  • panel D shows a 6 mpk BRAFi-945 and 3 mpk anti-ET B R-ADC (Hu5E9vl -vc-MMAE) combination
  • panel E shows a 20 mpk BRAFi-945 and 3 mpk anti-ET B R-ADC (Hu5E9vl -vc- MMAE) combination.
  • Figure 6 shows ETBR expression in COLO 829 melanoma cells treated for 24h with varying concentrations of BRAF inhibitor RG7204.
  • Panel A shows ETBR transcript normalized to RPL19 transcript.
  • Panel B shows expression of total ETBR and GAPDH (Control) protein in 50 ⁇ g whole cell lysates.
  • Panel C shows surface ETBR protein expression in live cells as seen by flow cytometry, where the first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with BRAF inhibitor and the third peak indicates BRAF inhibitor treated cells.
  • Figure 7 demonstrates the in vivo combination efficacy of anti-ET B R ADC and BRAF inhibitor RG7204 against COLO 829 melanoma xenografts mouse model.
  • Subcutaneous tumors were established in mice inoculated with COLO 829 melanoma cell lines. When tumor volumes reached approximately 200 mm 3 (day 0), animals were dosed orally twice a day for 21 days with RG7204. On day 1 (after three doses of RG7204), animals were given a single IV injection of either vehicle or anti-ET B R ADC (Hu5E9vl -vc-MMAE) at the indicated doses. Average tumor volumes with standard deviations were determined from 9 animals per group.
  • panel A shows 3 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 30 mpk of RG7204;
  • panel B shows 1 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 30 mpk of RG7204;
  • panel C shows 1 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 10 mpk of RG7204;
  • panel D shows 3 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 10 mpk of RG7204.
  • Figure 8 shows ETBR expression in A2058 melanoma cells treated for 24h with varying concentrations of BRAF inhibitor RG7204.
  • Panel A shows ETBR transcript normalized to RPL19 transcript;
  • panel B shows expression of total ETBR and GAPDH (Control) protein in 100 ⁇ g whole cell lysates;
  • panel C shows surface ETBR protein expression in live cells as seen by flow cytometry.
  • the first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with BRAF inhibitor, and the third peak indicates BRAF inhibitor treated cells.
  • Figure 9 demonstrates in vivo combination efficacy of anti-ET B R ADC (Hu5E9vl -vc-MMAE) and BRAF inhibitor RG7204 against A2058 melanoma xenograft mouse models.
  • Subcutaneous tumors were established in mice inoculated with A2058 melanoma cell lines. When tumor volumes reached approximately 200 mm 3 (day 0), animals were dosed orally twice a day for 21 days with RG7204. On day 1 (after three doses of RG7204), animals were given a single IV injection of either vehicle or anti- ET B R ADC (Hu5E9vl -vc-MMAE) at the indicated doses. Average tumor volumes with standard deviations were determined from 10 animals per group.
  • Panel A shows 6 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 10 mpk of RG7204;
  • panel B shows 6 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 30 mpk of RG7204;
  • panel C shows 3 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 10 mpk of RG7204;
  • panel D shows 3 mpk of anti-ET B R-ADC (Hu5E9vl -vc- MMAE) in combination with 30 mpk of RG7204.
  • Figure 10 shows Western blot experiments performed with BRAFi RG7204 showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and ⁇ -tubulin in 25 to 100 ⁇ g whole cell lysates from IPC-298 melanoma cells.
  • Figure 11 shows surface ETBR protein expression in IPC-298 live cells as seen by flow cytometry after incubation with 0.1 ⁇ , 1 ⁇ and 10 ⁇ of BRAFi RG7204 (panels A, B and C respectively).
  • the first peak indicates cells treated to secondary detection reagent alone, the second peak indicates BRAF inhibitor treated cells and the third peak indicates cells untreated with BRAF inhibitor.
  • Figure 12 shows Western blot experiments performed with MEKi-623 (panel A) and MEKi-973 (panel B) at concentrations of 0 ⁇ , 0.01 ⁇ , 0.1 ⁇ and 1 ⁇ showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and ⁇ -tubulin in 50 ⁇ g whole cell lysates from COL0829 melanoma cells.
  • Figure 13 shows surface ETBR protein expression in COLO 829 live cells as seen by flow cytometry after incubation with 0.01 ⁇ (panels A and D), 0.1 ⁇ (panels B and E) and 1 ⁇ (panel C and F) of MEKi-623 (panels A, B and C respectively) or MEKi-973 (panels D, E and F respectively).
  • the first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with MEK inhibitor and the third peak indicates MEK inhibitor treated cells.
  • Figure 14 shows ETBR mRNA expression in A2058 melanoma cells treated for 24h with varying concentrations of MEKi-623 (panel A) or MEKi-973 (panel B), normalized to RPL19 transcript.
  • Figure 15 shows Western blot experiments performed with MEKi-623 (panel A) and MEKi-973 (panel B) at concentrations of 0 ⁇ , 0.01 ⁇ , 0.1 ⁇ and 1 ⁇ showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and ⁇ -tubulin in 50-100 ⁇ g whole cell lysates from A2058 melanoma cells.
  • Figure 16 shows surface ETBR protein expression in A2058 live cells as seen by flow cytometry after incubation with 0.01 ⁇ (panels A and D), 0.1 ⁇ (panels B and E) and 1 ⁇ (panel C and F) of MEKi-623 (panels A, B and C respectively) or MEKi-973 (panels D, E and F respectively).
  • the first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with MEK inhibitor and the third peak indicates MEK inhibitor treated cells.
  • Figure 17 demonstrates in vivo combination efficacy of anti-ET B R ADC (Hu5E9vl -vc-MMAE) and MEKi-973 against A2058 melanoma xenograft mouse models.
  • Subcutaneous tumors were established in mice inoculated with A2058 melanoma cell lines. When tumor volumes reached approximately 200 mm 3 (day 0), animals were dosed orally once a day for 21 days with MEKi-973. On day 1 (after two doses of MEKi-973), animals were given a single IV injection of either vehicle or anti- ET B R ADC (Hu5E9vl -vc-MMAE) at the indicated doses. Average tumor volumes with standard deviations were determined from 9 animals per group.
  • Panel A shows 7.5 mpk of anti-gD ADC (control) in combination with 7.5 mpk of MEKi-973 as compared to a vehicle control and anti-gD ADC alone
  • panel B shows 6 mpk of anti-ET B R- ADC (Hu5E9vl -vc-MMAE) in combination with 7.5 mpk of MEKi-973 as compared to a vehicle control and 7.5 mpk MEKi-973 alone (GDC-0973) or 6 mpk of anti-ET B R-ADC alone.
  • Figure 18 shows ET B R transcript expression in SK23-MEL melanoma cells treated for 24h with varying concentrations of MEKi-623 (panel A) or MEKi-973 (panel B), which were normalized to RPL19 transcript.
  • Figure 19 shows Western blot experiments performed with MEKi-623 (panel A) and MEKi-973 (panel B) at concentrations of 0 ⁇ , 0.01 ⁇ , 0.1 ⁇ and 1 ⁇ showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and ⁇ -tubulin in 50 ⁇ g whole cell lysates from SK23- MEL melanoma cells.
  • Figure 20 shows surface ETBR protein expression in live SK23-MEL cells as seen by flow cytometry after incubation with 0.01 ⁇ (panels A and D), 0.1 ⁇ (panels B and E) and 1 ⁇ (panel C and F) of MEKi-623 (panels A, B and C respectively) or MEKi-973 (panels D, E and F respectively).
  • the first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with MEK inhibitor and the third peak indicates MEK inhibitor treated cells.
  • Figure 21 demonstrates in vivo combination efficacy of anti-ET B R ADC (Hu5E9vl-vc-MMAE) and MEKi-973 against SK23-MEL melanoma xenograft mouse models.
  • Panel A shows 6 mpk of anti-gD ADC (control) in combination with 7.5 mpk of MEKi-973 as compared to a vehicle control, 7.5 mpk MEKi-973 and 6 mpk anti-gD ADC alone
  • panel B shows 6 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 7.5 mpk of MEKi-973 ("Combination") as compared to a vehicle control and 7.5 mpk MEKi-973 alone or 6 mpk of anti-ET B R-ADC alone.
  • Panel C shows 3 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 3 mpk of MEKi-973 ("Combination"), as compared to a vehicle control, 3 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) or 3 mpk of MEKi-973.
  • Panel D shows 3 mpk of anti-ET B R-ADC (Hu5E9vl -vc- MMAE) in combination with 7.5 mpk of MEKi-973 ("Combination") as compared to a vehicle control and 7.5 mpk MEKi-973 alone or 3 mpk of anti-ET B R-ADC alone.
  • Panel E shows 6 mpk of anti-ET B R- ADC (Hu5E9vl -vc-MMAE) in combination with 3 mpk of MEKi-973 ("Combination") as compared to a vehicle control and 3 mpk MEKi-973 alone or 6 mpk of anti-ET B R-ADC alone.
  • Figure 22 shows Western blot experiments performed with MEKi-623 (panel A) and MEKi-973 (panel B) at concentrations of 0 ⁇ , 0.01 ⁇ , 0.1 ⁇ and 1 ⁇ showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and ⁇ -tubulin in 25-100 ⁇ g whole cell lysates from IPC-298 melanoma cells.
  • Figure 23 shows surface ETBR protein expression in live IPC-298 cells as seen by flow cytometry after incubation with 0.01 ⁇ (panels A and D), 0.1 ⁇ (panels B and E) and 1 ⁇ (panel C and F) of MEKi-623 (panels A, B and C respectively) or MEKi-973 (panels D, E and F respectively).
  • the first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with MEK inhibitor and the third peak indicates MEK inhibitor treated cells.
  • Figure 24 demonstrates in vivo combination efficacy of anti-ET B R ADC (Hu5E9vl -vc-MMAE) and MEKi-623 against IPC-298 melanoma xenograft mouse models.
  • Panel A shows 6 mpk of anti-gD ADC (control) in combination with 1 mpk of MEKi-623 as compared to a vehicle control, 1 mpk MEKi-623 and 6 mpk anti-gD ADC alone
  • Panel B shows 6 mpk of anti-ET B R-ADC (Hu5E9vl -vc-MMAE) in combination with 1 mpk of MEKi- 623 ("Combination") as compared to a vehicle control and 1 mpk MEKi-623 alone or 6 mpk of anti- ET B R-ADC alone.
  • FIG. 25 in vivo combination efficacy of anti-ET B R ADC (Hu5E9vl -vc-MMAE) and MEKi- 973 against IPC-298 melanoma xenograft mouse models.
  • Panel A shows 6 mpk of anti-gD ADC (control) in combination with 7.5 mpk of MEKi-973
  • Figure 26 depicts expression of phosphorylated erk and total erk protein in COLO 829 tumors treated with either vehicle or 30mpk BRAFi RG7204.
  • Figure 27 depicts ETBR transcript expression in COLO 829 tumors treated with BRAFi RG7204 (panel A) and in A2058 tumors treated with MEKi-973 for 3 days (panel B).
  • Panel A shows ETBR transcript normalized to control GAPDH in COLO 829 cell line, in COLO 892 tumors treated with either vehicle control or 10 mpk or 30 mpk RG7204.
  • Panel B shows ETBR transcript normalized to control Hprtl in A2058 tumors treated with either vehicle control or 5 or 10 mpk of MEKi-973.
  • an "acceptor human framework” for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below.
  • An acceptor human framework "derived from” a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less.
  • the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
  • Binding affinity refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
  • binding affinity refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
  • An "affinity matured” antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.
  • HVRs hypervariable regions
  • anti-ETBR antibody and "an antibody that binds to ETBR” refer to an antibody that is capable of binding the endothelin B receptor (ETBR) with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting ETBR.
  • the extent of binding of an anti-ETBR antibody to an unrelated, non-ETBR protein is less than about 10% of the binding of the antibody to ETBR as measured, e.g., by a radioimmunoassay (RIA).
  • RIA radioimmunoassay
  • an antibody that binds to ETBR has a dissociation constant (Kd) of ⁇ 1 ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 "8 M or less, e.g. from 10 "8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • Kd dissociation constant
  • an anti-ETBR antibody binds to an epitope of ETBR that is conserved among ETBR from different species.
  • antibody herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
  • an "antibody fragment” refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds.
  • antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab') 2 ; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
  • an "antibody that binds to the same epitope" as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more.
  • An exemplary competition assay is provided herein.
  • BRAF refers to a serine/threonine-protein kinase B-Raf, also known as proto-oncogene B-Raf or v-Raf murine sarcoma viral oncogene homolog Bl, which is a protein that in humans is encoded by the BRAF gene.
  • B-Raf protein is involved in sending signals in cells and in cell growth.
  • BRAF inhibitor or "BRAFi” as used herein refers to any number of known small molecule drug compounds which can inhibit or interrupt the B-Raf/MEK step on the B-Raf/MEK/ERK pathway.
  • suitable BRAFi may include, but are not limited to, those described in
  • Another example may be, but is not limited to, GSK 2118436, having a CAS registry number 405554-55- 4, which is also known as 5-[2-[4-[2-(Dimethylamino)ethoxy]phenyl]-5-(4-pyridinyl)-lH-imidazol-4-yl]- 2,3-dihydro-lH-inden-l-one oxime.
  • chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
  • the "class" of an antibody refers to the type of constant domain or constant region possessed by its heavy chain.
  • the heavy chain constant domains that correspond to the different classes of immunoglobulins are called ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , respectively.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction.
  • Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 , Zr 89 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatic
  • Antibody effector functions refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell- mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
  • an "effective amount" of an agent refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • ETBR refers to any native endothelin B receptor (ETBR) from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g., mice and rats), unless otherwise indicated.
  • the term encompasses "full-length,” unprocessed ETBR as well as any form of ETBR that results from processing in the cell.
  • the term also encompasses naturally occurring variants of ETBR, e.g., splice variants or allelic variants.
  • amino acid sequence of an exemplary human ETBR is shown in SEQ ID NO: 10 (see Nakamuta M et al., Cloning and Sequence Analysis of a cDNA encoding Human non-selective type of endothelin receptor, Biochem Biophys Res Commun. 1991 May 31 : 177(l):34-9).
  • anti-ETBR antibody - ADC refers to any anti-ETBR antibody described herein that is conjugated to a toxin.
  • toxins include, but are not limited to, maytansinoids or specifically monomethylauristatin (MMAE).
  • MMAE monomethylauristatin
  • An anti-ETBR antibody- ADC is contemplated as a species of "anti-ETBR antibodies of the invention”.
  • Fc region herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region.
  • the term includes native sequence Fc regions and variant Fc regions.
  • a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain.
  • the C-terminal lysine (Lys447) of the Fc region may or may not be present.
  • numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.
  • FR Framework or "FR” refers to variable domain residues other than hypervariable region (HVR) residues.
  • the FR of a variable domain generally consists of four FR domains: FRl, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FRl -H 1 (L 1 )-FR2-H2(L2)-FR3 -H3 (L3)-FR4.
  • VH or VL
  • BRAFi-945" refers to a B-Raf enzyme inhibitor that is 4- amino-N-(6-chloro-2-fluoro-3-(3-fluoro propyl sulfonamido) phenyl)thieno[3,2-d]pyrimidine-7- carboxamide and has a structure having the following formula as disclosed in Example 15 of
  • host cell refers to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells.
  • Host cells include “transformants” and “transformed cells,” which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
  • a "human antibody” is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
  • a "human consensus framework” is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
  • the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences.
  • the subgroup of sequences is a subgroup as in Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, NIH Publication 91-3242, Bethesda MD (1991), vols. 1-3.
  • the subgroup is subgroup kappa I as in Kabat et al., supra.
  • the subgroup III is subgroup III as in Kabat et al., supra.
  • a “humanized” antibody refers to a chimeric antibody comprising amino acid residues from non- human HVRs and amino acid residues from human FRs.
  • a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody.
  • a humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody.
  • a "humanized form" of an antibody, e.g., a non-human antibody refers to an antibody that has undergone humanization.
  • hypervariable region refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops ("hypervariable loops").
  • native four-chain antibodies comprise six HVRs; three in the VH (HI, H2, H3), and three in the VL (LI, L2, L3).
  • HVRs generally comprise amino acid residues from the hypervariable loops and/or from the "complementarity determining regions" (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition.
  • CDRs complementarity determining regions
  • Exemplary hypervariable loops occur at amino acid residues 26-32 (LI), 50-52 (L2), 91-96 (L3), 26-32 (HI), 53-55 (H2), and 96-101 (H3).
  • Exemplary CDRs CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3 occur at amino acid residues 24-34 of LI, 50-56 of L2, 89- 97 of L3, 31-35B of HI, 50-65 of H2, and 95-102 of H3.
  • CDRs generally comprise the amino acid residues that form the hypervariable loops.
  • CDRs also comprise "specificity determining residues,” or "SDRs,” which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated- CDRs, or a-CDRs.
  • Exemplary a-CDRs (a-CDR-Ll, a-CDR-L2, a-CDR-L3, a-CDR-Hl, a-CDR-H2, and a-CDR-H3) occur at amino acid residues 31-34 of LI, 50-55 of L2, 89-96 of L3, 31-35B of HI, 50-58 of H2, and 95-102 of H3.
  • HVR residues and other residues in the variable domain are numbered herein according to Kabat et al., supra.
  • an “immunoconjugate” is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
  • mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats).
  • domesticated animals e.g., cows, sheep, cats, dogs, and horses
  • primates e.g., humans and non-human primates such as monkeys
  • rabbits e.g., mice and rats
  • rodents e.g., mice and rats.
  • the individual or subject is a human.
  • an "isolated" antibody is one which has been separated from a component of its natural environment.
  • an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC).
  • electrophoretic e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis
  • chromatographic e.g., ion exchange or reverse phase HPLC
  • An "isolated" nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment.
  • An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location.
  • isolated nucleic acid encoding an anti-ETBR antibody refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
  • MAP kinase mitogen-activated protein kinase
  • CMGC CDK/MAPK/GSK3/CLK
  • ERK1/2 pathway of mammals is probably the best characterized MAPK system.
  • the most important upstream activators of this pathway are the Raf proteins (A-Raf, B-Raf or c-Raf), the key mediators of response to growth factors (EGF, FGF, PDGF, etc.).
  • MEK MAPK/ERK kinase
  • MEK inhibitor refers to any number of known small molecule drug compounds which can inhibit or interrupt the MEK step on the MAP kinase pathway.
  • suitable MEKi may include, but are not limited to, those described as MEKi-623, MEKi-973, or GSK1120212.
  • MEKi-973 refers to a MEK inhibitor (S)-(3,4-difuoro-2-((2-fluoro-4- iodo henyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone, having the structure:
  • the term "monoclonal antibody” as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts.
  • polyclonal antibody preparations typically include different antibodies directed against different determinants (epitopes)
  • each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen.
  • the modifier "monoclonal” indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method.
  • the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein.
  • a “naked antibody” refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel. The naked antibody may be present in a pharmaceutical formulation.
  • Native antibodies refer to naturally occurring immunoglobulin molecules with varying structures.
  • native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and CH3).
  • VH variable region
  • VL variable region
  • the light chain of an antibody may be assigned to one of two types, called kappa ( ⁇ ) and lambda ( ⁇ ), based on the amino acid sequence of its constant domain.
  • package insert is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
  • Percent (%) amino acid sequence identity with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared.
  • % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2.
  • the ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087.
  • the ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code.
  • the ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
  • the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B is calculated as follows: 100 times the fraction X/Y
  • composition refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • RG7204 refers to a B-Raf enzyme inhibitor that has a molecular formula of C23H 18 CIF 2 N 3 C>3S and the following structure:
  • treatment refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis.
  • antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
  • variable region refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen.
  • the variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs).
  • FRs conserved framework regions
  • HVRs hypervariable regions
  • antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano et al., J. Immunol. 150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).
  • vector refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked.
  • the term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced.
  • Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors.”
  • the invention is based, in part, on antibodies that bind to ETBR.
  • Antibodies of the invention are useful, e.g., for the treatment of melanoma.
  • an anti-ETBR antibody comprises at least one, two, three, four, five, or six CDRs selected from (a) CDR-L1 (KS S Q SLLD SDGKTYLN, SEQ ID NO:7), (b) CDR-L2 (LVSKLDS, SEQ ID NO:8), (c) CDR-L3 (WQGTHFPYT; SEQ ID NO:9), (d) CDR-H1 (GYTFTSYWMQ; SEQ ID NO: l), (e) CDR- H2 (TIYPGDGDTSYAQKFKG; SEQ ID NO:2), and (f) CDR-H3 (WGYAYDIDN; SEQ ID NO:3).
  • an anti-ETBR antibody is humanized.
  • an anti-ETBR antibody comprises CDRs as in any of the above embodiments, and further comprises an acceptor human framework, e.g. a human immunoglobulin framework or a human consensus framework.
  • the invention provides an isolated anti-ETBR antibody having the VL amino acid sequence of SEQ ID NO:8, and the VH amino acid sequence of SEQ ID NO:7.
  • the invention provides an anti-ETBR antibody having a VL sequence of SEQ ID NO: 8 and a VH amino acid sequence of SEQ ID NO:9.
  • an anti-ETBR antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:7 or 9.
  • VH heavy chain variable domain
  • a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti- ETBR antibody comprising that sequence retains the ability to bind to ETBR.
  • an anti-ETBR antibody comprising a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 8.
  • VL light chain variable domain
  • a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-ETBR antibody comprising that sequence retains the ability to bind to ETBR.
  • substitutions e.g., conservative substitutions
  • insertions, or deletions relative to the reference sequence
  • an anti-ETBR antibody comprising that sequence retains the ability to bind to ETBR.
  • a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 8.
  • the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
  • an anti-ETBR antibody comprising a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above.
  • the antibody comprises the VH and VL sequences in SEQ ID NO: 7 or 9 and SEQ ID NO:8, respectively, including post-translational modifications of those sequences.
  • the invention provides an antibody that binds to the same epitope as an anti- ETBR antibody provided herein.
  • an antibody is provided that binds to the same epitope as an anti-ETBR antibody comprising a VH sequence of SEQ ID NO: 7 or 9 and a VL sequence of SEQ ID NO: 8.
  • an anti-ETBR antibody is provided that binds to an epitope within an N-terminal extracellular domain #1 fragment of ETBR consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
  • an anti-ETBR antibody is a monoclonal antibody, including a chimeric, humanized or human antibody.
  • an anti-ETBR antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab')2 fragment.
  • the antibody is a full length antibody, e.g., an intact IgGl antibody or other antibody class or isotype as defined herein.
  • an anti-ETBR antibody according to any of the above embodiments may incorporate any of the features, singly or in combination, as described in Sections 1-7 below:
  • an antibody provided herein has a dissociation constant (Kd) of ⁇ 1 ⁇ , ⁇ 100 nM, ⁇ 10 nM, ⁇ 1 nM, ⁇ 0.1 nM, ⁇ 0.01 nM, or ⁇ 0.001 nM (e.g. 10 ⁇ 8 M or less, e.g. from 10 ⁇ 8 M to 10 "13 M, e.g., from 10 "9 M to 10 "13 M).
  • Kd dissociation constant
  • Kd is measured by a radiolabeled antigen binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay.
  • Solution binding affinity of Fabs for antigen is measured by equilibrating Fab with a minimal concentration of ( 125 I) -labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (see, e.g., Chen et al., J. Mol. Biol. 293:865-881(1999)).
  • MICROTITER ® multi-well plates (Thermo Scientific) are coated overnight with 5 ⁇ g/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (pH 9.6), and subsequently blocked with 2%> (w/v) bovine serum albumin in PBS for two to five hours at room temperature (approximately 23°C).
  • a non-adsorbent plate (Nunc #269620)
  • 100 pM or 26 pM [ 125 I]-anti gen are mixed with serial dilutions of a Fab of interest (e.g., consistent with assessment of the anti-VEGF antibody, Fab-12, in Presta et al., Cancer Res.
  • the Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation at room temperature (e.g., for one hour). The solution is then removed and the plate washed eight times with 0.1% polysorbate 20 (TWEEN-20 ® ) in PBS. When the plates have dried, 150 ⁇ /well of scintillant (MICROSCINT-20TM; Packard) is added, and the plates are counted on a TOPCOUNTTM gamma counter (Packard) for ten minutes. Concentrations of each Fab that give less than or equal to 20% of maximal binding are chosen for use in competitive binding assays.
  • Kd is measured using surface plasmon resonance assays using a BIACORE ® -2000 or a BIACORE ® -3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at -10 response units (RU).
  • CM5 carboxymethylated dextran biosensor chips
  • EDC N-ethyl-N'- (3-dimethylaminopropyl)- carbodiimide hydrochloride
  • NHS N-hydroxysuccinimide
  • Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 ⁇ g/ml (-0.2 ⁇ ) before injection at a flow rate of 5 ⁇ /minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 tiM) are injected in PBS with 0.05% polysorbate 20 (TWEEN-20TM) surfactant (PBST) at 25°C at a flow rate of approximately 25 ⁇ /min. Association rates (k on ) and dissociation rates (k 0 ff) are calculated using a simple one-to-one
  • the on-rate can be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity
  • a spectrometer such as a stop-flow equipped spectrophometer (Aviv Instruments) or a 8000-series SLM- AMINCOTM spectrophotometer (ThermoSpectronic) with a stirred cuvette.
  • an antibody provided herein is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab') 2 , Fv, and scFv fragments, and other fragments described below. For a review of certain antibody fragments, see Hudson et al. Nat. Med. 9: 129-134 (2003). For a review of scFv fragments, see, e.g., Pluckthun, in The Pharmacology of
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161 ; Hudson et al., Nat. Med. 9: 129-134 (2003); and Hollinger et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al., Nat. Med. 9: 129-134 (2003).
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516 Bl).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
  • recombinant host cells e.g. E. coli or phage
  • an antibody provided herein is a chimeric antibody.
  • Certain chimeric antibodies are described, e.g., in U.S. Patent No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, 81 :6851-6855 (1984)).
  • a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • a chimeric antibody is a "class switched" antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
  • a chimeric antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • HVRs e.g., CDRs, (or portions thereof) are derived from a non-human antibody
  • FRs or portions thereof
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the HVR residues are derived
  • Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the "best-fit” method (see, e.g., Sims et al. J. Immunol.
  • framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol., 151 :2623 (1993)); human mature (somatically mutated) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, Front. Biosci. 13: 1619-1633 (2008)); and framework regions derived from screening FR libraries (see, e.g., Baca et al., J Biol. Chem. 272: 10678-10684 (1997) and Rosok et al., J. Biol. Chem. 271 :22611-22618 (1996)).
  • an antibody provided herein is a human antibody.
  • Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin. Pharmacol. 5: 368-74 (2001) and Lonberg, Curr. Opin.
  • Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human
  • Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse- human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol, 133: 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications , pp. 51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al., J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-cell hybridoma technology are also described in Li et al.. Proc. Natl Acad. Sci.
  • Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
  • Antibodies of the invention may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in Hoogenboom et al. m Methods in Molecular Biology 178: 1-37 (O'Brien et al., ed., Human Press, Totowa, NJ, 2001) and further described, e.g., in the McCafferty et al., Nature 348:552-554; Clackson et al., Nature 352: 624-628 (1991); Marks et al., J. Mol. Biol.
  • phage display methods repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al., Ann. Rev. Immunol, 12: 433-455 (1994). Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments. Libraries from immunized sources provide high-affinity antibodies to the immunogen without the requirement of constructing hybridomas.
  • PCR polymerase chain reaction
  • the naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al., EMBO J, 12: 725-734 (1993).
  • naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol, 227: 381-388 (1992).
  • Patent publications describing human antibody phage libraries include, for example: US Patent No. 5,750,373, and US Patent Publication Nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360.
  • Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
  • an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody.
  • Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites.
  • one of the binding specificities is for ETBR and the other is for any other antigen.
  • bispecific antibodies may bind to two different epitopes of ETBR.
  • Bispecific antibodies may also be used to localize cytotoxic agents to cells which express ETBR.
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments.
  • Multispecific antibodies include, but are not limited to, recombinant co- expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983)), WO 93/08829, and Traunecker et al., EMBO J. 10: 3655 (1991)), and "knob-in-hole” engineering (see, e.g., U.S. Patent No. 5,731,168). Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US Patent No.
  • Engineered antibodies with three or more functional antigen binding sites are also included herein (see, e.g. US 2006/0025576A1).
  • the antibody or fragment herein also includes a "Dual Acting FAb” or “DAF” comprising an antigen binding site that binds to ETBR as well as another, different antigen (see, US 2008/0069820, for example).
  • amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
  • antibody variants having one or more amino acid substitutions are provided.
  • Sites of interest for substitutional mutagenesis include the HVRs and FRs.
  • Conservative substitutions are shown in Table 1 under the heading of "conservative substitutions.” More substantial changes are provided in Table 1 under the heading of "exemplary substitutions,” and as further described below in reference to amino acid side chain classes.
  • Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, or improved ADCC or CDC. TABLE 1
  • Amino acids may be grouped according to common side -chain properties:hydrophobic:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody).
  • a parent antibody e.g. a humanized or human antibody
  • the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody.
  • An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display- based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
  • Alterations may be made in HVRs, e.g., to improve antibody affinity. Such alterations may be made in HVR "hotspots," i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol.
  • Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in Hoogenboom et al. m Methods in Molecular Biology 178: 1-37 (O'Brien et al., ed., Human Press, Totowa, NJ, (2001).)
  • affinity maturation diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis).
  • a secondary library is then created.
  • the library is then screened to identify any antibody variants with the desired affinity.
  • Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.
  • substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen.
  • conservative alterations e.g., conservative substitutions as provided herein
  • Such alterations may be outside of HVR "hotspots" or SDRs.
  • each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
  • a useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham and Wells (1989) Science, 244: 1081-1085.
  • a residue or group of target residues e.g., charged residues such as arg, asp, his, lys, and glu
  • a neutral or negatively charged amino acid e.g., alanine or polyalanine
  • Further substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions.
  • a crystal structure of an antigen- antibody complex to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution.
  • Variants may be screened to determine whether they contain the desired properties.
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • the carbohydrate attached thereto may be altered.
  • Native antibodies produced by mammalian cells typically comprise a branched, biantennary
  • oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15:26-32 (1997).
  • the oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure.
  • modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
  • antibody variants having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region.
  • the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5% to 65% or from 20% to 40%.
  • the amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example.
  • Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ⁇ 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd).
  • WO2002/031140 Okazaki et al. J. Mol. Biol. 336: 1239-1249 (2004); Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004).
  • Examples of cell lines capable of producing defucosylated antibodies include Led 3 CHO cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys.
  • knockout cell lines such as alpha-l,6-fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004); Kanda, Y. et al., Biotechnol. Bioeng., 94(4):680-688 (2006); and WO2003/085107).
  • Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean-Mairet et al.); US Patent No. 6,602,684 (Umana et al.); and US 2005/0123546 (Umana et al.). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided.
  • Such antibody variants may have improved CDC function.
  • Such antibody variants are described, e.g., in WO 1997/30087 (Patel et al.); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.).
  • one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant.
  • the Fc region variant may comprise a human Fc region sequence ⁇ e.g., a human IgGl, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification ⁇ e.g. a substitution) at one or more amino acid positions.
  • the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious.
  • In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities.
  • Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability.
  • NK cells express FcyRIII only, whereas monocytes express FcyRI, FcyRII and FcyRIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-492 (1991).
  • Non- limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No. 5,500,362 (see, e.g. Hellstrom, I. et al. Proc. Natl Acad. Sci. USA 83:7059-7063 (1986)) and Hellstrom, I et al., Proc.
  • non-radioactive assays methods may be employed (see, for example, ACTITM non-radioactive cytotoxicity assay for flow cytometry
  • PBMC peripheral blood mononuclear cells
  • NK Natural Killer
  • ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. Proc. Natl Acad. Sci. USA 95:652-656 (1998).
  • Clq binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity.
  • a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J. Immunol. Methods 202: 163 (1996); Cragg, M.S. et al., Blood 101 : 1045-1052 (2003); and Cragg, M.S. and M.J. Glennie, Blood 103:2738- 2743 (2004)).
  • FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et al., Int'l. Immunol. 18(12): 1759-1769 (2006)).
  • Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056).
  • Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US Patent No. 7,332,581).
  • an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).
  • alterations are made in the Fc region that result in altered ⁇ i.e., either improved or diminished) Clq binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No. 6,194,551, WO 99/51642, and Idusogie et al. J. Immunol. 164: 4178-4184 (2000).
  • CDC Complement Dependent Cytotoxicity
  • Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No. 7,371,826).
  • cysteine engineered antibodies e.g., "thioMAbs”
  • one or more residues of an antibody are substituted with cysteine residues.
  • the substituted residues occur at accessible sites of the antibody.
  • reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker- drug moieties, to create an immunoconjugate, as described further herein.
  • any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region.
  • Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No.
  • an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n- vinyl
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
  • Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567.
  • isolated nucleic acid encoding an anti-ETBR antibody described herein is provided.
  • Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody).
  • one or more vectors e.g., expression vectors
  • a host cell comprising such nucleic acid is provided.
  • a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NS0, Sp20 cell).
  • a method of making an anti-ETBR antibody comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • nucleic acid encoding an antibody is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein.
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. See also Charlton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ, 2003), pp. 245-254, describing expression of antibody fragments in E. coli.
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22: 1409-1414 (2004), and Li et al., Nat. Biotech. 24:210-215 (2006).
  • Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIESTM technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, Biol. Reprod.
  • monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al., Annals N. Y. Acad. Sci. 383:44-68 (1982); MRC 5 cells; and FS4 cells.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR " CHO cells (Urlaub et al., Proc. Natl. Acad. Sci.
  • Anti-ETBR antibodies provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.
  • an antibody of the invention is tested for its antigen binding activity, e.g., by known methods such as ELISA, Western blot, etc.
  • competition assays may be used to identify an antibody that competes with, for example, Hu5E9v. l or Hu5E9v.2 for binding to ETBR.
  • a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by Hu5E9v.l or Hu5E9v.2.
  • epitope e.g., a linear or a conformational epitope
  • Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) "Epitope Mapping Protocols," in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ).
  • anti-ETBR antibodies described herein specifically bind an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
  • immobilized ETBR is incubated in a solution comprising a first labeled antibody that binds to ETBR (e.g., Hu5E9v.l or Hu5E9v.2) and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to ETBR.
  • the second antibody may be present in a hybridoma supernatant.
  • immobilized ETBR is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to ETBR, excess unbound antibody is removed, and the amount of label associated with immobilized ETBR is measured.
  • assays are provided for identifying whether anti-ETBR antibodies and/or BRAFi compounds have biological activity.
  • Biological activity may include those described in the Examples, e.g., in vitro melanoma cell survival assays or in vivo xenograft models in which melanoma cell lines are transplanted into nude mice and tumor growth inhibition (TGI) is assessed.
  • TGI tumor growth inhibition
  • the invention also provides immunoconjugates comprising an anti-ETBR antibody herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • cytotoxic agents such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
  • the invention also provides immunoconjugates (interchangeably referred to as "antibody-drug conjugates,” or "ADCs”) comprising an antibody conjugated to one or more cytotoxic agents, such as a chemotherapeutic agent, a drug, a growth inhibitory agent, a toxin (e.g., a protein toxin, an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • cytotoxic agents such as a chemotherapeutic agent, a drug, a growth inhibitory agent, a toxin (e.g., a protein toxin, an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • Immunoconjugates have been used for the local delivery of cytotoxic agents, i.e., drugs that kill or inhibit the growth or proliferation of cells, in the treatment of cancer (Xie et al (2006) Expert. Opin. Biol. Ther. 6(3):281-291 ; Kovtun et al (2006) Cancer Res. 66(6):3214-3121 ; Law et al (2006) Cancer Res. 66(4):2328-2337; Lambert, J. (2005) Curr. Opinion in Pharmacology 5:543-549; Wu et al (2005) Nature Biotechnology 23(9): 1137-1146; Payne, G.
  • cytotoxic agents i.e., drugs that kill or inhibit the growth or proliferation of cells
  • Immunoconjugates allow for the targeted delivery of a drug moiety to a tumor, and intracellular accumulation therein, where systemic administration of unconjugated drugs may result in unacceptable levels of toxicity to normal cells as well as the tumor cells sought to be eliminated (Baldwin et al., Lancet (Mar. 15, 1986) pp. 603-05; Thorpe (1985) "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review," in Monoclonal Antibodies '84: Biological And Clinical
  • Efforts to design and refine ADC have focused on the selectivity of monoclonal antibodies (mAbs) as well as drug mechanism of action, drug-linking, drug/antibody ratio (loading), and drug-releasing properties (McDonagh (2006) Protein Eng. Design & Sel.; Doronina et al (2006) Bioconj. Chem. 17: 114-124; Erickson et al (2006) Cancer Res. 66(8): l-8; Sanderson et al (2005) Clin. Cancer Res. 11 :843-852; Jeffrey et al (2005) J. Med. Chem. 48: 1344-1358; Hamblett et al (2004) Clin. Cancer Res. 10:7063-7070).
  • the toxins may exert their cytotoxic effects by mechanisms including tubulin binding, DNA binding, or topoisomerase inhibition. Some cytotoxic drugs tend to be inactive or less active when conjugated to large antibodies or protein receptor ligands.
  • ZEVALIN® is an antibody-radioisotope conjugate composed of a murine IgGl kappa monoclonal antibody directed against the CD20 antigen found on the surface of normal and malignant B lymphocytes and 11 lln or 90Y radioisotope bound by a thiourea linker-chelator (Wiseman et al (2000) Eur. Jour. Nucl. Med. 27(7):766-77; Wiseman et al (2002) Blood 99(12):4336-42; Witzig et al (2002) J. Clin. Oncol.
  • ZEVALIN has activity against B-cell non-Hodgkin's Lymphoma (NHL), administration results in severe and prolonged cytopenias in most patients.
  • MYLOTARGTM (gemtuzumab ozogamicin, Wyeth Pharmaceuticals), an antibody-drug conjugate composed of a huCD33 antibody linked to calicheamicin, was approved in 2000 for the treatment of acute myeloid leukemia by injection (Drugs of the Future (2000) 25(7):686; US Patent Nos.
  • ADCs Antibody-drug conjugates (ADCs) composed of the maytansinoid, DM1, linked to trastuzumab show potent anti-tumor activity in HER2-overexpressing trastuzumab-sensitive and -resistant tumor cell lines and xenograft models of human cancer.
  • T-DM1 Trastuzumab-MCC-DMl
  • ADC HER2 antibody-drug conjugate
  • BC metastatic breast cancer
  • auristatin peptides auristatin E (AE) and monomethylauristatin (MMAE), synthetic analogs of dolastatin, were conjugated to chimeric monoclonal antibodies cBR96 (specific to Lewis Y on carcinomas) and cACIO (specific to CD30 on hematological malignancies) (Doronina et al (2003) Nature Biotechnol. 21(7):778-784) and are under therapeutic development.
  • an immunoconjugate comprises an antibody and a chemotherapeutic agent or other toxin.
  • Chemotherapeutic agents useful in the generation of immunoconjugates are described herein (e.g., above). Enzymatically active toxins and fragments thereof can also be used and are described herein.
  • an immunoconjugate comprises an antibody and one or more small molecule drug moieties (toxins), including, but not limited to, small molecule drugs such as a calicheamicin, maytansinoid, dolastatin, auristatin, anthracycline, taxane, trichothecene, and CC1065, and the derivatives of these drugs that have cytotoxic activity. Examples of such immunoconjugates are discussed in further detail below.
  • An immunoconjugate (or "antibody-drug conjugate” (“ADC”)) of the invention may be of Formula I, below, wherein an antibody is conjugated (i.e., covalently attached) to one or more drug moieties (D) through an optional linker (L).
  • the antibody may be conjugated to the drug either directly or via a linker.
  • p is the average number of drug moieties per antibody, which can range, e.g., from about 1 to about 20 drug moieties per antibody, and in certain embodiments, from 1 to about 8 drug moieties per antibody.
  • a linker may comprise one or more linker components.
  • exemplary linker components include 6- maleimidocaproyl (“MC”), maleimidopropanoyl (“MP”), valine-citrulline (“val-cit” or “vc”), alanine- phenylalanine (“ala-phe”), p-aminobenzyloxycarbonyl (a "PAB”), N-Succinimidyl 4-(2-pyridylthio) pentanoate (“SPP”), N-succinimidyl 4-(N-maleimidomethyl) cyclohexane-1 carboxylate (“SMCC”), and N-Succinimidyl (4-iodo-acetyl) aminobenzoate (“SIAB”).
  • MC 6- maleimidocaproyl
  • MP maleimidopropanoyl
  • val-cit valine-citrulline
  • alanine- phenylalanine ala-p
  • a linker may be a "cleavable linker," facilitating release of a drug in the cell.
  • an acid-labile linker e.g., hydrazone
  • protease-sensitive linker e.g., peptidase-sensitive
  • photolabile linker e.g., dimethyl linker or disulfide -containing linker
  • dimethyl linker or disulfide -containing linker e.g., Cancer Research 52: 127-131 (1992); U.S. Patent No. 5,208,020
  • a linker is as shown in the following Formula II:
  • A is a stretcher unit, and a is an integer from 0 to 1 ; W is an amino acid unit, and w is an integer from 0 to 12; Y is a spacer unit, and y is 0, 1, or 2; and Ab, D, and p are defined as above for Formula I.
  • linkers are described in US 2005-0238649 Al, which is expressly incorporated herein by reference.
  • a linker component may comprise a "stretcher unit” that links an antibody to another linker component or to a drug moiety.
  • stretcher units are shown below (wherein the wavy line indicates sites of covalent attachment to an antibody):
  • a linker component may comprise an amino acid unit.
  • the amino acid unit allows for cleavage of the linker by a protease, thereby facilitating release of the drug from the immunoconjugate upon exposure to intracellular proteases, such as lysosomal enzymes. See, e.g., Doronina et al. (2003) Nat. Biotechnol. 21 :778-784.
  • Exemplary amino acid units include, but are not limited to, a dipeptide, a tripeptide, a tetrapeptide, and a pentapeptide.
  • Exemplary dipeptides include: valine-citrulline (vc or val-cit), alanine-phenylalanine (af or ala-phe); phenylalanine-lysine (fk or phe-lys); or N-methyl-valine-citrulline (Me-val-cit).
  • Exemplary tripeptides include: glycine -valine-citrulline (gly-val-cit) and glycine-glycine-glycine (gly-gly-gly).
  • An amino acid unit may comprise amino acid residues that occur naturally, as well as minor amino acids and non- naturally occurring amino acid analogs, such as citrulline.
  • Amino acid units can be designed and optimized in their selectivity for enzymatic cleavage by a particular enzyme, for example, a tumor- associated protease, cathepsin B, C and D, or a plasmin protease.
  • a linker component may comprise a "spacer" unit that links the antibody to a drug moiety, either directly or by way of a stretcher unit and/or an amino acid unit.
  • a spacer unit may be "self-immolative” or a "non-self-immolative.”
  • a "non-self-immolative" spacer unit is one in which part or all of the spacer unit remains bound to the drug moiety upon enzymatic (e.g., proteolytic) cleavage of the ADC.
  • non-self-immolative spacer units include, but are not limited to, a glycine spacer unit and a glycine-glycine spacer unit.
  • peptidic spacers susceptible to sequence-specific enzymatic cleavage are also contemplated.
  • enzymatic cleavage of an ADC containing a glycine -glycine spacer unit by a tumor-cell associated protease would result in release of a glycine-glycine-drug moiety from the remainder of the ADC.
  • the glycine- glycine-drug moiety is then subjected to a separate hydrolysis step in the tumor cell, thus cleaving the glycine-glycine spacer unit from the drug moiety.
  • a "self-immolative" spacer unit allows for release of the drug moiety without a separate hydrolysis step.
  • a spacer unit of a linker comprises a p-aminobenzyl unit.
  • a p-aminobenzyl alcohol is attached to an amino acid unit via an amide bond, and a carbamate, methylcarbamate, or carbonate is made between the benzyl alcohol and a cytotoxic agent. See, e.g., Hamann et al. (2005) Expert Opin. Ther. Patents (2005) 15: 1087-1 103.
  • the spacer unit is p-aminobenzyloxycarbonyl (PAB).
  • the phenylene portion of a p-amino benzyl unit is substituted with Qm, wherein Q is -Ci-Cg alkyl, -0-(Ci-Cg alkyl), -halogen,- nitro or -cyano; and m is an integer ranging from 0-4.
  • Q is -Ci-Cg alkyl, -0-(Ci-Cg alkyl), -halogen,- nitro or -cyano
  • m is an integer ranging from 0-4.
  • self-immolative spacer units further include, but are not limited to, aromatic compounds that are electronically similar to p-aminobenzyl alcohol ⁇ see, e.g., US 2005/0256030 Al), such as 2-aminoimidazol-5 -methanol derivatives (Hay et al. (1999) Bioorg. Med. Chem. Lett.
  • Spacers can be used that undergo cyclization upon amide bond hydrolysis, such as substituted and unsubstituted 4- aminobutyric acid amides (Rodrigues et al., Chemistry Biology, 1995, 2, 223); appropriately substituted bicyclo[2.2.1] and bicyclo[2.2.2] ring systems (Storm, et al., J. Amer. Chem. Soc , 1972, 94, 5815); and 2-aminophenylpropionic acid amides (Amsberry, et al., J. Org. Chem., 1990, 55, 5867).
  • Elimination of amine -containing drugs that are substituted at the a-position of glycine are also examples of self-immolative spacers useful in ADCs.
  • a spacer unit is a branched bis(hydroxymethyl)styrene (BHMS) unit as depicted below, which can be used to incorporate and release multiple drugs.
  • BHMS branched bis(hydroxymethyl)styrene
  • Q is -Ci-Cg alkyl, -0-(Ci-Cg alkyl), -halogen, -nitro or -cyano;
  • m is an integer ranging from 0-4;
  • n is 0 or 1 ; and
  • p ranges raging from 1 to about 20.
  • linker L may be a dendritic type linker for covalent attachment of more than one drug moiety through a branching, multifunctional linker moiety to an antibody (Sun et al (2002) Bioorganic & Medicinal Chemistry Letters 12:2213-2215; Sun et al (2003) Bioorganic & Medicinal Chemistry 1 1 : 1761 -1768).
  • Dendritic linkers can increase the molar ratio of drug to antibody, i.e. loading, which is related to the potency of the ADC.
  • a cysteine engineered antibody bears only one reactive cysteine thiol group, a multitude of drug moieties may be attached through a dendritic linker.
  • Linkers components including stretcher, spacer, and amino acid units, may be synthesized by methods known in the art, such as those described in US 2005-0238649 Al .
  • an immunoconjugate comprises an antibody conjugated to one or more maytansinoid molecules.
  • Maytansinoids are mitototic inhibitors which act by inhibiting tubulin polymerization. Maytansine was first isolated from the east African shrub Maytenus serrata (U.S. Patent No. 3896111). Subsequently, it was discovered that certain microbes also produce maytansinoids, such as maytansinol and C-3 maytansinol esters (U.S. Patent No. 4,151,042). Synthetic maytansinol and derivatives and analogues thereof are disclosed, for example, in U.S. Patent Nos.
  • Maytansinoid drug moieties are attractive drug moieties in antibody-drug conjugates because they are: (i) relatively accessible to prepare by fermentation or chemical modification or derivatization of fermentation products, (ii) amenable to derivatization with functional groups suitable for conjugation through non-disulfide linkers to antibodies, (iii) stable in plasma, and (iv) effective against a variety of tumor cell lines.
  • Maytansine compounds suitable for use as maytansinoid drug moieties are well known in the art and can be isolated from natural sources according to known methods or produced using genetic engineering techniques (see Yu et al (2002) PNAS 99:7968-7973). Maytansinol and maytansinol analogues may also be prepared synthetically according to known methods.
  • Exemplary maytansinoid drug moieties include those having a modified aromatic ring, such as: C-19-dechloro (US Pat. No. 4256746) (prepared by lithium aluminum hydride reduction of ansamytocin P2); C-20-hydroxy (or C-20-demethyl) +/-C-19-dechloro (US Pat. Nos. 4361650 and 4307016) (prepared by demethylation using Streptomyces or Actinomyces or dechlorination using LAH); and C-20- demethoxy, C-20-acyloxy (-OCOR), +/-dechloro (U.S. Pat. No. 4,294,757) (prepared by acylation using acyl chlorides), and those having modifications at other positions.
  • C-19-dechloro (US Pat. No. 4256746) (prepared by lithium aluminum hydride reduction of ansamytocin P2)
  • C-20-hydroxy (or C-20-demethyl) +/-C-19-dechloro US Pat
  • Exemplary maytansinoid drug moieties also include those having modifications such as: C-9-SH (US Pat. No. 4424219) (prepared by the reaction of maytansinol with H 2 S or P 2 S 5 ); C-14- alkoxymethyl(demethoxy/CH 2 OR)(US 4331598); C-14-hydroxymethyl or acyloxymethyl (CH 2 OH or CH 2 OAc) (US Pat. No. 4450254) (prepared from Nocardia); C-15-hydroxy/acyloxy (US 4364866) (prepared by the conversion of maytansinol by Streptomyces); C-15-methoxy (US Pat. Nos.
  • R may independently be H or a Ci-C 6 alkyl.
  • the alkylene chain attaching the amide group to the sulfur atom may be methanyl, ethanyl, or propyl, i.e., m is 1, 2, or 3 (US 633410; US 5208020; Chari et al (1992) Cancer Res. 52: 127-131 ; Liu et al (1996) Proc. Natl. Acad. Sci USA 93:8618-8623).
  • the maytansinoid drug moiety will have the following stereochemistry:
  • Exemplary embodiments of maytansinoid drug moieities include: DM1 ; DM3; and DM4, having the structures:
  • Exemplary antibody-drug conjugates where DM1 is linked through a BMPEO linker to group of the antibody have the structure and abbreviation:
  • Immunoconjugates containing maytansinoids, methods of making the same, and their therapeutic use are disclosed, for example, in U.S. Patent Nos. 5,208,020, 5,416,064, US 2005/0276812 Al, and European Patent EP 0 425 235 Bl, the disclosures of which are hereby expressly incorporated by reference. Liu et al. Proc. Natl. Acad. Sci. USA 93:8618-8623 (1996) describe immunoconjugates comprising a maytansinoid designated DM1 linked to the monoclonal antibody C242 directed against human colorectal cancer.
  • the conjugate was found to be highly cytotoxic towards cultured colon cancer cells, and showed antitumor activity in an in vivo tumor growth assay.
  • Chari et al. Cancer Research 52: 127-131 (1992) describe immunoconjugates in which a maytansinoid was conjugated via a disulfide linker to the murine antibody A7 binding to an antigen on human colon cancer cell lines, or to another murine monoclonal antibody TA. l that binds the HER-2/neu oncogene.
  • the cytotoxicity of the TA.l- maytansonoid conjugate was tested in vitro on the human breast cancer cell line SK-BR-3, which expresses 3 x 10 5 HER-2 surface antigens per cell.
  • the drug conjugate achieved a degree of cytotoxicity similar to the free maytansinoid drug, which could be increased by increasing the number of maytansinoid molecules per antibody molecule.
  • the A7 -maytansinoid conjugate showed low systemic cytotoxicity in mice.
  • Antibody-maytansinoid conjugates are prepared by chemically linking an antibody to a maytansinoid molecule without significantly diminishing the biological activity of either the antibody or the maytansinoid molecule. See, e.g., U.S. Patent No. 5,208,020 (the disclosure of which is hereby expressly incorporated by reference). An average of 3-4 maytansinoid molecules conjugated per antibody molecule has shown efficacy in enhancing cytotoxicity of target cells without negatively affecting the function or solubility of the antibody, although even one molecule of toxin/antibody would be expected to enhance cytotoxicity over the use of naked antibody. Maytansinoids are well known in the art and can be synthesized by known techniques or isolated from natural sources.
  • Suitable maytansinoids are disclosed, for example, in U.S. Patent No. 5,208,020 and in the other patents and nonpatent publications referred to hereinabove.
  • Preferred maytansinoids are maytansinol and maytansinol analogues modified in the aromatic ring or at other positions of the maytansinol molecule, such as various maytansinol esters.
  • Antibody-maytansinoid conjugates comprising the linker component SMCC may be prepared as disclosed in US 2005/0276812 Al, "Antibody-drug conjugates and Methods.”
  • the linkers comprise disulfide groups, thioether groups, acid labile groups, photolabile groups, peptidase labile groups, or esterase labile groups, as disclosed in the above-identified patents. Additional linkers are described and exemplified herein.
  • Conjugates of the antibody and maytansinoid may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4- (N-maleimidomethyl) cyclohexane-l-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HQ), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluor
  • the coupling agent is N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP) (Carlsson et al., Biochem. J. 173:723-737 (1978)) or N-succinimidyl-4-(2-pyridylthio)pentanoate (SPP) to provide for a disulfide linkage.
  • SPDP N-succinimidyl-3-(2-pyridyldithio) propionate
  • SPP N-succinimidyl-4-(2-pyridylthio)pentanoate
  • the linker may be attached to the maytansinoid molecule at various positions, depending on the type of the link.
  • an ester linkage may be formed by reaction with a hydroxyl group using conventional coupling techniques. The reaction may occur at the C-3 position having a hydroxyl group, the C-14 position modified with hydroxymethyl, the C-15 position modified with a hydroxyl group, and the C-20 position having a hydroxyl group.
  • the linkage is formed at the C-3 position of maytansinol or a maytansinol analogue.
  • an immunoconjugate comprises an antibody conjugated to dolastatin or a dolastatin peptidic analog or derivative, e.g., an auristatin (US Pat. Nos. 5635483; 5780588).
  • Dolastatins and auristatins have been shown to interfere with microtubule dynamics, GTP hydrolysis, and nuclear and cellular division (Woyke et al (2001) Antimicrob. Agents and Chemother. 45(12):3580-3584) and have anticancer (US Pat. No.5663149) and antifungal activity (Pettit et al (1998) Antimicrob. Agents Chemother. 42:2961-2965).
  • the dolastatin or auristatin drug moiety may be attached to the antibody through the N (amino) terminus or the C (carboxyl) terminus of the peptidic drug moiety (WO
  • Exemplary auristatin embodiments include the N-terminus linked monomethylauristatin drug moieties DE and DF (US 7498298).
  • R 2 is selected from H and C r C 8 alkyl
  • R 3 is selected from H, C C 8 alkyl, C 3 -C 8 carbocycle, aryl, C C 8 alkyl-aryl, C C 8 alkyl-(C 3 -C 8 carbocycle), C3-C 8 heterocycle and Ci-C 8 alkyl-(C3-C 8 heterocycle);
  • R 4 is selected from H, C C 8 alkyl, C 3 -C 8 carbocycle, aryl, C C 8 alkyl-aryl, C C 8 alkyl-(C 3 -C 8 carbocycle), C 3 -C 8 heterocycle and Ci-C 8 alkyl-(C3-C 8 heterocycle);
  • R 5 is selected from H and methyl; or R 4 and R 5 jointly form a carbocyclic ring and have the formula -(CR a R b ) n - wherein R a and R b are independently selected from H, Q-C 8 alkyl and C 3 -C 8 carbocycle and n is selected from 2, 3, 4, 5 and 6;
  • R 6 is selected from H and C r C 8 alkyl
  • R 7 is selected from H, C r C 8 alkyl, C 3 -C 8 carbocycle, aryl, C r C 8 alkyl-aryl, C r C 8 alkyl-(C 3 -C 8 carbocycle), C 3 -C 8 heterocycle and C r C 8 alkyl-(C 3 -C 8 heterocycle);
  • each R 8 is independently selected from H, OH, C r C 8 alkyl, C 3 -C 8 carbocycle and 0-(C r C 8 alkyl);
  • R 9 is selected from H and C r C 8 alkyl
  • R 10 is selected from aryl or C 3 -C 8 heterocycle
  • Z is O, S, NH, or NR 12 , wherein R 12 is C C 8 alkyl;
  • R 11 is selected from H, C C 2 o alkyl, aryl, C 3 -C 8 heterocycle, -(R 13 0) m -R 14 , or -(R 13 0) m - CH(R 15 ) 2 ;
  • m is an integer ranging from 1 -1000;
  • R 13 is C 2 -C 8 alkyl
  • R 14 is H or C r C 8 alkyl
  • each occurrence of R 15 is independently H, COOH, -(CH 2 ) n -N(R 16 ) 2 , -(CH 2 ) n -S0 3 H, or -(CH 2 ) n - S0 3 -C r C 8 alkyl;
  • each occurrence of R 16 is independently H, C C 8 alkyl, or -(CH 2 ) n -COOH;
  • R 18 is selected from -C(R 8 ) 2 -C(R 8 ) 2 -aryl, -C(R 8 ) 2 -C(R 8 ) 2 -(C 3 -C 8 heterocycle), and
  • n is an integer ranging from 0 to 6.
  • R 3 , R 4 and R 7 are independently isopropyl or sec-butyl and R 5 is -H or methyl.
  • R 3 and R 4 are each isopropyl, R 5 is -H, and R 7 is sec-butyl.
  • R 2 and R 6 are each methyl, and R 9 is -H.
  • each occurrence of R 8 is -OCH 3 .
  • R 3 and R 4 are each isopropyl, R 2 and R 6 are each methyl, R 5 is -H, R 7 is sec-butyl, each occurrence of R 8 is -OCH 3 , and R 9 is -H.
  • Z is -O- or -NH-.
  • R 10 is aryl.
  • R 10 is -phenyl.
  • when Z is -0-, R 11 is -H, methyl or t-butyl.
  • R 11 is -CH(R 15 ) 2 , wherein R 15 is -(CH 2 ) n -N(R 16 ) 2 , and R 16 is -C r C 8 alkyl or -(CH 2 ) n -COOH.
  • Z is -NH
  • R 11 is -CH(R 15 ) 2 , wherein R 15 is -(CH 2 ) n -S0 3 H.
  • An exemplary auristatin embodiment of formula D E is MMAE, wherein the wavy line indicates the covalent attachment to a linker (L) of an antibody-drug conjugate:
  • An exemplary auristatin embodiment of formula D F is MMAF, wherein the wavy line indicates the covalent attachment to a linker (L) of an antibody-drug conjugate (see US 7498298 and Doronina et al. (2006
  • Other exemplary embodiments include monomethylvaline compounds having phenylalanine carboxy modifications at the C-terminus of the pentapeptide auristatin drug moiety (WO 2007/008848) and monomethylvaline compounds having phenylalanine sidechain modifications at the C-terminus of the pentapeptide auristatin drug moiety (WO 2007/008603).
  • hydrophilic groups including but not limited to, triethylene glycol esters (TEG), as shown above, can be attached to the drug moiety at R 11 .
  • TEG triethylene glycol esters
  • ADCs of Formula I comprising an auristatin/dolastatin or derivative thereof are described in US 7498298 and Doronina et al. (2006) Bioconjugate Chem. 17: 114-124, which is expressly incorporated herein by reference.
  • Exemplary embodiments of ADCs of Formula I comprising MMAE or MMAF and various linker components have the following structures and abbreviations (wherein "Ab” is an antibody; p is 1 to about 8, "Val-Cit” is a valine -citrulline dipeptide; and "S” is a sulfur atom:
  • Exemplary embodiments of ADCs of Formula I comprising MMAF and various linker components further include Ab-MC-PAB-MMAF and Ab-PAB-MMAF.
  • immunoconjugates comprising MMAF attached to an antibody by a linker that is not proteolytically cleavable have been shown to possess activity comparable to immunoconjugates comprising MMAF attached to an antibody by a proteolytically cleavable linker. See, Doronina et al. (2006) Bioconjugate Chem. 17: 114-124. In such instances, drug release is believed to be effected by antibody degradation in the cell. Id.
  • peptide-based drug moieties can be prepared by forming a peptide bond between two or more amino acids and/or peptide fragments.
  • Such peptide bonds can be prepared, for example, according to the liquid phase synthesis method (see E. Schroder and K. Liibke, "The Peptides", volume 1, pp 76-136, 1965, Academic Press) that is well known in the field of peptide chemistry.
  • Auristatin/dolastatin drug moieties may be prepared according to the methods of: US 2005-0238649 Al ; US Pat. No.5635483; US Pat. No.5780588; Pettit et al (1989) J. Am. Chem. Soc. 111 :5463-5465; Pettit et al (1998) Anti-Cancer Drug Design 13:243-277; Pettit, G.R., et al. Synthesis, 1996, 719-725; Pettit et al (1996) J. Chem. Soc. Perkin Trans. 1 5:859-863; and Doronina (2003) Nat. Biotechnol. 21(7):778-784.
  • auristatin/dolastatin drug moieties of formula D F may be prepared using methods described in US 7498298 and Doronina et al. (2006)
  • Auristatin/dolastatin drug moieties of formula D E such as MMAE and derivatives thereof, may be prepared using methods described in Doronina et al. (2003) Nat. Biotech. 21 :778-784.
  • Drug-linker moieties MC-MMAF, MC-MMAE, MC-vc-PAB-MMAF, and MC-vc-PAB- MMAE may be conveniently synthesized by routine methods, e.g., as described in Doronina et al. (2003) Nat. Biotech. 21 :778-784, and US 7498298, and then conjugated to an antibody of interest.
  • the immunoconjugate comprises an antibody conjugated to one or more calicheamicin molecules.
  • the calicheamicin family of antibiotics are capable of producing double- stranded DNA breaks at sub-picomolar concentrations.
  • For the preparation of conjugates of the calicheamicin family see U.S. Pat. Nos. 5,712,374, 5,714,586, 5,739,116, 5,767,285, 5,770,701, 5,770,710, 5,773,001, 5,877,296 (all to American Cyanamid Company).
  • Structural analogues of calicheamicin which may be used include, but are not limited to, ⁇ , ⁇ 2 ', ⁇ 3 ', N-acetyl-y , PSAG and ⁇ ' ⁇ (Hinman et al., Cancer Research 53:3336-3342 (1993), Lode et al., Cancer Research 58:2925-2928 (1998), and the aforementioned U.S. patents to American Cyanamid).
  • Another anti-tumor drug to which the antibody can be conjugated is QFA, which is an antifolate.
  • QFA Another anti-tumor drug to which the antibody can be conjugated.
  • Both calicheamicin and QFA have intracellular sites of action and do not readily cross the plasma membrane. Therefore, cellular uptake of these agents through antibody-mediated internalization greatly enhances their cytotoxic effects.
  • Enzymatically active toxins and fragments thereof which can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin and the tricothecenes. See, for example, WO 93/21232 published October 28, 1993.
  • the present invention further contemplates an immunoconjugate formed between an antibody and a compound with nucleolytic activity (e.g., a ribonuclease or a DNA endonuclease such as a deoxyribonuclease; DNase).
  • a compound with nucleolytic activity e.g., a ribonuclease or a DNA endonuclease such as a deoxyribonuclease; DNase.
  • an immunoconjugate may comprise a highly radioactive atom.
  • a variety of radioactive isotopes are available for the production of radioconjugated antibodies. Examples include At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 Zr 89 and radioactive isotopes of Lu.
  • the immunoconjugate When used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc 99m or I 123 , or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123, iodine-131, indium-I l l, fluorine-19, carbon-13, nitrogen- 15, oxygen- 17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • the radio- or other labels may be incorporated in the immunoconjugate in known ways.
  • the peptide may be biosynthesized or may be synthesized by chemical amino acid synthesis using suitable amino acid precursors involving, for example, fluorine- 19 in place of hydrogen.
  • Labels such as tc 99m or I 123 , Re 186 , Re 188 Zr 89 and In 111 can be attached via a cysteine residue in the peptide.
  • Yttrium-90 can be attached via a lysine residue.
  • the IODOGEN method (Fraker et al (1978) Biochem. Biophys. Res. Commun. 80: 49-57 can be used to incorporate iodine-123. "Monoclonal Antibodies in Immunoscintigraphy" (Chatal, CRC Press 1989) describes other methods in detail.
  • an immunoconjugate may comprise an antibody conjugated to a prodrug-activating enzyme that converts a prodrug (e.g., a peptidyl chemotherapeutic agent, see WO 81/01145) to an active drug, such as an anti-cancer drug.
  • a prodrug e.g., a peptidyl chemotherapeutic agent, see WO 81/01145
  • an active drug such as an anti-cancer drug.
  • ADPT antibody- dependent enzyme -mediated prodrug therapy
  • Enzymes that may be conjugated to an antibody include, but are not limited to, alkaline phosphatases, which are useful for converting phosphate -containing prodrugs into free drugs; arylsulfatases, which are useful for converting sulfate - containing prodrugs into free drugs; cytosine deaminase, which is useful for converting non-toxic 5- fluorocytosine into the anti-cancer drug, 5-fluorouracil; proteases, such as serratia protease, thermolysin, subtilisin, carboxypeptidases and cathepsins (such as cathepsins B and L), which are useful for converting peptide-containing prodrugs into free drugs; D-alanylcarboxypeptidases, which are useful for converting prodrugs that contain D-amino acid substituents; carbohydrate-cleaving enzymes such as ⁇ - galactosidase and neuraminidase, which are useful for converting
  • Drug loading is represented by p, the average number of drug moieties per antibody in a molecule of Formula I. Drug loading may range from 1 to 20 drug moieties (D) per antibody.
  • ADCs of Formula I include collections of antibodies conjugated with a range of drug moieties, from 1 to 20. The average number of drug moieties per antibody in preparations of ADC from conjugation reactions may be characterized by conventional means such as mass spectroscopy, ELISA assay, and HPLC. The quantitative distribution of ADC in terms of p may also be determined. In some instances, separation, purification, and characterization of homogeneous ADC where p is a certain value from ADC with other drug loadings may be achieved by means such as reverse phase HPLC or electrophoresis.
  • p may be limited by the number of attachment sites on the antibody.
  • an antibody may have only one or several cysteine thiol groups, or may have only one or several sufficiently reactive thiol groups through which a linker may be attached.
  • higher drug loading e.g. p >5
  • the drug loading for an ADC of the invention ranges from 1 to about 8; from about 2 to about 6; or from about 3 to about 5. Indeed, it has been shown that for certain ADCs, the optimal ratio of drug moieties per antibody may be less than 8, and may be about 2 to about 5. See US 2005-0238649 Al .
  • an antibody may contain, for example, lysine residues that do not react with the drug-linker intermediate or linker reagent, as discussed below. Generally, antibodies do not contain many free and reactive cysteine thiol groups which may be linked to a drug moiety; indeed most cysteine thiol residues in antibodies exist as disulfide bridges.
  • an antibody may be reduced with a reducing agent such as dithiothreitol (DTT) or tricarbonylethylphosphme (TCEP), under partial or total reducing conditions, to generate reactive cysteine thiol groups.
  • DTT dithiothreitol
  • TCEP tricarbonylethylphosphme
  • an antibody is subjected to denaturing conditions to reveal reactive nucleophilic groups such as lysine or cysteine.
  • the loading (drug/antibody ratio) of an ADC may be controlled in different ways, e.g., by: (i) limiting the molar excess of drug-linker intermediate or linker reagent relative to antibody, (ii) limiting the conjugation reaction time or temperature, and (iii) partial or limiting reductive conditions for cysteine thiol modification.
  • the resulting product is a mixture of ADC compounds with a distribution of one or more drug moieties attached to an antibody.
  • the average number of drugs per antibody may be calculated from the mixture by a dual ELISA antibody assay, which is specific for antibody and specific for the drug.
  • Individual ADC molecules may be identified in the mixture by mass spectroscopy and separated by HPLC, e.g. hydrophobic interaction chromatography (see, e.g., McDonagh et al (2006) Prot. Engr. Design & Selection 19(7):299-307; Hamblett et al (2004) Clin. Cancer Res.
  • a homogeneous ADC with a single loading value may be isolated from the conjugation mixture by electrophoresis or chromatography.
  • An ADC of Formula I may be prepared by several routes employing organic chemistry reactions, conditions, and reagents known to those skilled in the art, including: (1) reaction of a nucleophilic group of an antibody with a bivalent linker reagent to form Ab-L via a covalent bond, followed by reaction with a drug moiety D; and (2) reaction of a nucleophilic group of a drug moiety with a bivalent linker reagent, to form D-L, via a covalent bond, followed by reaction with a nucleophilic group of an antibody.
  • Nucleophilic groups on antibodies include, but are not limited to: (i) N-terminal amine groups, (ii) side chain amine groups, e.g. lysine, (iii) side chain thiol groups, e.g. cysteine, and (iv) sugar hydroxyl or amino groups where the antibody is glycosylated.
  • Amine, thiol, and hydroxyl groups are nucleophilic and capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleimide groups. Certain antibodies have reducible interchain disulfides, i.e. cysteine bridges.
  • Antibodies may be made reactive for conjugation with linker reagents by treatment with a reducing agent such as DTT (dithiothreitol) or tricarbonylethylphosphine (TCEP), such that the antibody is fully or partially reduced.
  • a reducing agent such as DTT (dithiothreitol) or tricarbonylethylphosphine (TCEP)
  • TCEP tricarbonylethylphosphine
  • Each cysteine bridge will thus form, theoretically, two reactive thiol nucleophiles.
  • Additional nucleophilic groups can be introduced into antibodies through modification of lysine residues, e.g., by reacting lysine residues with 2-iminothiolane (Traut's reagent), resulting in conversion of an amine into a thiol.
  • Reactive thiol groups may be introduced into an antibody by introducing one, two, three, four, or more cysteine residues (e.
  • Antibody-drug conjugates of the invention may also be produced by reaction between an electrophilic group on an antibody, such as an aldehyde or ketone carbonyl group, with a nucleophilic group on a linker reagent or drug.
  • an electrophilic group on an antibody such as an aldehyde or ketone carbonyl group
  • nucleophilic groups on a linker reagent or drug include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide.
  • an antibody is modified to introduce electrophilic moieties that are capable of reacting with nucleophilic subsituents on the linker reagent or drug.
  • the sugars of glycosylated antibodies may be oxidized, e.g. with periodate oxidizing reagents, to form aldehyde or ketone groups which may react with the amine group of linker reagents or drug moieties.
  • the resulting imine Schiff base groups may form a stable linkage, or may be reduced, e.g. by borohydride reagents to form stable amine linkages.
  • reaction of the carbohydrate portion of a glycosylated antibody with either galactose oxidase or sodium meta-periodate may yield carbonyl (aldehyde and ketone) groups in the antibody that can react with appropriate groups on the drug (Hermanson, Bioconjugate Techniques).
  • antibodies containing N-terminal serine or threonine residues can react with sodium meta-periodate, resulting in production of an aldehyde in place of the first amino acid (Geoghegan & Stroh, (1992) Bioconjugate Chem. 3: 138-146; US
  • Such an aldehyde can be reacted with a drug moiety or linker nucleophile.
  • Nucleophilic groups on a drug moiety include, but are not limited to: amine, thiol, hydroxyl, hydrazide, oxime, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide groups capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleimide groups.
  • the compounds of the invention expressly contemplate, but are not limited to, ADC prepared with the following cross-linker reagents: BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo- SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc., Rockford, IL., U.S.A.
  • Immunoconjugates comprising an antibody and a cytotoxic agent may also be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane-l -carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p- azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)- ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate),
  • a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987).
  • Carbon- 14-labeled l-isothiocyanatobenzyl-3- methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See W094/11026.
  • a fusion protein comprising an antibody and a cytotoxic agent may be made, e.g., by recombinant techniques or peptide synthesis.
  • a recombinant DNA molecule may comprise regions encoding the antibody and cytotoxic portions of the conjugate either adjacent to one another or separated by a region encoding a linker peptide which does not destroy the desired properties of the conjugate.
  • an antibody may be conjugated to a "receptor” (such as streptavidin) for utilization in tumor pre -targeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand” (e.g., avidin) which is conjugated to a cytotoxic agent (e.g., a
  • the invention further provides pharmaceutical formulations comprising at least one antibody of the invention and/or at least one immunoconjugate thereof.
  • a pharmaceutical formulation comprises 1) an antibody of the invention and/or an immunoconjugate thereof, and 2) a pharmaceutically acceptable carrier.
  • a pharmaceutical formulation comprises 1) an antibody of the invention and/or an immunoconjugate thereof, and optionally, 2) at least one additional therapeutic agent. Additional therapeutic agents include, but are not limited to, those described below.
  • compositions comprising an antibody or immunoconjugate of the invention are prepared for storage by mixing the antibody or immunoconjugate having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers ⁇ Remington 's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)) in the form of aqueous solutions or lyophilized or other dried formulations.
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, histidine and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as
  • octadecyldimethylbenzyl ammonium chloride hexamethonium chloride; benzalkonium chloride, benzethonium chloride); phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins;
  • hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes ⁇ e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEENTM, PLURONICSTM or polyethylene glycol (PEG).
  • Pharmaceutical formulations to be used for in vivo administration are generally sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • Active ingredients may also be entrapped in microcapsule prepared, for example, by
  • coacervation techniques or by interfacial polymerization for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody or immunoconjugate of the invention, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3- hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated antibodies or immunoconjugates remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37°C, resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved.
  • stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • An antibody may be formulated in any suitable form for delivery to a target cell/tissue.
  • antibodies may be formulated as immunoliposomes.
  • a "liposome” is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes.
  • Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA 82:3688 (1985); Hwang et al., Proc. Natl Acad. Sci. USA 77:4030 (1980); U.S. Pat. Nos. 4,485,045 and 4,544,545; and
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al., J. Biol. Chem. 257:286-288 (1982) via a disulfide interchange reaction. A chemotherapeutic agent is optionally contained within the liposome. See Gabizon et al., J. National Cancer Inst. 81(19): 1484 (1989).
  • an immunoconjugate comprises an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • an enzymatically active toxin or fragment thereof including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (
  • an immunoconjugate comprises an antibody as described herein conjugated to a radioactive atom to form a radioconjugate.
  • a variety of radioactive isotopes are available for the production of radioconjugates. Examples include Zr 89 , At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 ,
  • the radioconjugate when used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc99m or 1123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123 again, iodine-131, indium-I l l, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
  • NMR nuclear magnetic resonance
  • Conjugates of an antibody and cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4- (N-maleimidomethyl) cyclohexane-l-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HQ), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds
  • a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987).
  • Carbon- 14-labeled l-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX- DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See
  • the linker may be a "cleavable linker" facilitating release of a cytotoxic drug in the cell.
  • a "cleavable linker” facilitating release of a cytotoxic drug in the cell.
  • an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Res. 52: 127-131 (1992); U.S. Patent No. 5,208,020) may be used.
  • the immunuoconjugates or ADCs herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo- GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl- (4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc.,
  • compositions of an anti-ETBR antibody as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions.
  • Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arg
  • sHASEGP soluble neutral-active hyaluronidase glycoproteins
  • rHuPH20 HYLENEX ® , Baxter International, Inc.
  • Certain exemplary sHASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968.
  • a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
  • Aqueous antibody formulations include those described in US Patent No. 6,171,586 and
  • the formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other.
  • active ingredients for example, it may be desirable to further provide a BRAF inhibitor, a MEK inhibitor or an anti-CTLA-4 antibody, ipilimumab.
  • Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
  • Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin- microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
  • the formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
  • any of the anti-ETBR antibodies provided herein may be used in therapeutic methods.
  • an anti-ETBR antibody for use as a medicament is provided.
  • the methods provide for an anti-ETBR antibody in combination with a BRAF inhibitor as useful as a medicament.
  • such a combination is useful in treating melanoma and/or metastatic melanoma.
  • an anti-ETBR antibody in combination with a BRAF inhibitor for use in a method of treatment is provided.
  • the invention provides an anti-ETBR antibody for use in a method of treating an individual having melanoma and/or metastatic melanoma comprising administering to the individual an effective amount of the anti-ETBR antibody and an effective amount of a BRAF inhibitor.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below, to the combination described.
  • the invention provides an anti- ETBR antibody in combination with a BRAF inhibitor for use in tumor growth inhibition (TGI).
  • TGI tumor growth inhibition
  • the invention provides an anti-ETBR antibody in combination with a BRAF inhibitor for use in a method of inhibiting tumor growth in a subject comprising administering to the subject an effective of the anti-ETBR antibody in combination with a BRAF inhibitor to inhibit tumor growth.
  • a "subject" according to any of the above embodiments is preferably a human.
  • the invention provides for the use of an anti-ETBR antibody in combination with a BRAF inhibitor in the manufacture or preparation of a medicament.
  • the medicament is for treatment of melanoma and/or metastatic melanoma.
  • the medicament is for use in a method of treating melanoma and/or metastatic melanoma comprising administering to an individual having melanoma and/or metastatic melanoma an effective amount of the medicament.
  • the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below.
  • the medicament is for tumor growth inhibition.
  • the medicament is for use in a method of tumor growth inhibition in an individual comprising administering to the individual an amount effective of the medicament to inhibit tumor growth.
  • An "individual" according to any of the above embodiments may be a human.
  • the invention provides pharmaceutical formulations comprising any of the anti-ETBR antibodies provided herein, e.g., in combination with a BRAF inhibitor for use in any of the above therapeutic methods.
  • a pharmaceutical formulation comprises any of the anti- ETBR antibodies provided herein in combination with a BRAF inhibitor and a pharmaceutically acceptable carrier.
  • a pharmaceutical formulation comprises any of the anti- ETBR antibodies provided herein in combination with a BRAF inhibitor and at least one additional therapeutic agent, e.g., as described below.
  • Antibodies of the invention can be used either alone or in combination with other agents in a therapy.
  • an antibody of the invention may be co-administered with at least one additional therapeutic agent.
  • an additional therapeutic agent is a BRAF inhibitor, a MEK inhibitor, or an anti-CTLA-4 antibody, such as, for example, ipilimumab.
  • Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant.
  • Antibodies of the invention can also be used in combination with radiation therapy.
  • An antibody of the invention (and any additional therapeutic agent, such as, for example, a BRAF inhibitor) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration.
  • Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic.
  • Various dosing schedules including but not limited to single or multiple administrations over various time -points, bolus administration, and pulse infusion are contemplated herein.
  • the BRAF inhibitor may be administered orally, in either tablet or capsule or liquid form.
  • Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners.
  • the antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
  • an antibody of the invention when used alone or in combination with one or more other additional therapeutic agents, will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician.
  • the antibody is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 ⁇ g/kg to 15 mg/kg (e.g. O.
  • lmg/kg-lOmg/kg can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion.
  • One typical daily dosage might range from about 1 ⁇ g/kg to 100 mg/kg or more, depending on the factors mentioned above.
  • the treatment would generally be sustained until a desired suppression of disease symptoms occurs.
  • One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg.
  • one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient.
  • Such doses may be administered intermittently, e.g.
  • Every week or every three weeks e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody.
  • An initial higher loading dose, followed by one or more lower doses may be administered.
  • other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
  • an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above comprises a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition is an antibody of the invention.
  • the label or package insert indicates that the composition is used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent.
  • the article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition.
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • Example 1 In vitro evaluations of specific cell killing by an anti-ETBR ADC
  • the anti-ETBR antibody- ADC candidate Hu5E9vl -ADC was evaluated in vitro on melanoma cell lines expressing either relatively low ETBR copy number, in the case of cell line A2058 (Obtained from American Type Culture Collection) or high ETBR copy number, in the case of cell line UACC- 257X2.2.
  • the UACC-257X2.2 cell line is a derivative of the parental UACC-257 cell line (NCI- Frederick Cancer DCT Tumor Repository) optimized for growth in vivo. Parental UACC-257 cells were injected subcutaneously in the right flank of female NCr nude mice, one tumor was harvested and dissociated grown in vitro resulting in the UACC-257X1.2 cell line.
  • the UACC-257X1.2 line was injected again subcutaneously in the right flank of female NCr nude mice in an effort to improve the growth of the cell line.
  • a tumor from this study was collected and again adapted for in vitro growth to generate the UACC-257X2.2 cell line.
  • This cell line expresses high levels of ET B R as determined by flow cytometry.
  • the relationship of receptor levels to Hu5E9vl-vc-MMAE cell killing in these cell lines was evaluated as follows.
  • the melanoma cell lines A2058 and UACC-257X2.2 were grown in appropriate media at 37°C and 5% C0 2 .
  • Hu5E9vl-ADC Hu5E9vl-ADC concentrations was added to triplicate wells. Five days later, cell survival was determined using CellTiter-Glo Luminescent Cell Viability Reagent (G7572; Promega Corporation) and with an EnVision 2101 Mutilabel Reader (Perkin- Elmer).
  • Example 2 In vivo evaluations of specific tumor killing by an anti-ETBR ADC
  • melanoma cell lines A2058 and UACC- 257X2.2 were selected as suitable models for in vivo anti-tumor activity studies that represent a wide range of ETBR expression.
  • the UACC-257X2.2 melanoma cell line is a derivative of the parental UACC-257 melanoma cell line (National Cancer Institute (NCI)) optimized for growth in vivo.
  • NCI National Cancer Institute
  • UACC-257 cells were injected subcutaneously in the right flank of female NCr nude mice, one tumor was harvested and grown in vitro resulting in the UACC-257X1.2 cell line.
  • the UACC-257X1.2 line was injected again subcutaneously in the right flank of female NCr nude mice in an effort to improve the growth of the cell line.
  • a tumor from this study was collected and again adapted for in vitro growth to generate the UACC-257X2.2 cell line.
  • This cell line and tumors derived from this line express ETBR comparable to the parental cell line UACC-257 (data not shown).
  • the anti-ETBR ADC candidate Hu5E9vl -ADC was administered as a single intravenous (IV) injection on day 0 at 1 mpk, 3 mpk, or 6 mpk (mg/kg).
  • a control ADC antibody and vehicle control were also administered. Average tumor volumes with standard deviations were determined from 10 animals per group. Tumor volumes were measured twice per week until study end.
  • the primer-probe set for RPL19 is as follows:
  • the primer-probe set for ETBR is as follows:
  • FACS fluorescence-activated cell sorting
  • Example 4 Effect of BRAF inhibitor drugs on in vivo efficacy of anti-ETBR ADC
  • Tumor Volume (mm3) (length x width 2 ) x 0.5
  • a TGI value of 100% indicates tumor stasis, of > 1% but ⁇ 100% indicates tumor growth delay, and of > 100% indicates tumor regression.
  • UIs uncertainty intervals
  • the random sample is composed of 1000 simulated realizations of the fitted-mixed model, where the %TGI has been recalculated for each realization.
  • UI uncertainty intervals
  • the %TGI has been recalculated for each realization.
  • the reported UI is the value for which 95% of the time, the recalculated values of %TGI will fall in this region given the fitted model.
  • the 2.5 and 97.5 percentiles of the simulated distribution were used as the upper and lower UIs.
  • Example 5 Dose Testing Anti-ETBR ADC and BRAFi combinations in vivo in COLO 829 Xenografts
  • Tumors were grown to an average size of approximately 200 mm 3 , whereupon animals were randomized into groups of 10 each.
  • Table 3 summarizes the three melanoma xenograft models tested at varying doses, as described above, to demonstrate the combination effects, expressed as a percent delta (last column) of the combination use of anti-ETBR ADC with a BRAF inhibitor as compared to either the percent TGI of the anti-ETBR ADC as a single agent or the percent TGI of a BRAF inhibitor as a single agent.
  • the percent TGI was calculated using a Linear Mixed Effects (LME) modeling approach, as described above.
  • LME Linear Mixed Effects
  • ETBR transcript and protein total protein and cell surface protein
  • COL0829 BRAF V600E
  • A2058 BRAF V600E
  • SK23-MEL BRAF WT /RAS WT
  • IPC-298 BRAF WT /RAS C61L
  • Primer-probe sets were designed with primers flanking a fluorogenic probe dual labeled with Reporter dye FAM and quencher dye TAMRA.
  • the primer-probe set for RPL19 is as follows:
  • the primer-probe set for ETBR is as follows:
  • FACS fluorescence-activated cell sorting
  • results for A2058 are shown in Figure 16A-F
  • results for COL0829 are shown in Figure 13A-F
  • results for SK23-MEL are shown in Figure 20A-F
  • results for IPC- 298 are shown in Figure 23A-F.
  • Example 8 Effect of MEK inhibitor drugs on in vivo efficacy of anti-ETBR ADC
  • Example 7 Given the results demonstrated in Example 7 above, the impact of the MEK inhibitors described herein on the in vivo efficacy of an anti-ET B R ADC was tested. To do this, the in vivo efficacy for various combinations of Hu5E9vl -ADC and MEKi-623 and/or MEKi-973 were evaluated against A2058 and SK-MEL-23 and IPC-298 melanoma in vivo models, performed as described above in Example 4. An appropriate methylcellulose twee vehicle control (0.5% methylcellulose, 0.2% Tween-80 (MCT) or MEK inhibitor at doses of 1 mpk, 3 mpk or 7.5 mpk were administered orally once a day x 21 days beginning on study Day 0. A single 3 mpk or 6 mpk dose of Hu5E9vl-ADC or control, a histidine buffer #8, was administered intraveneously (after two doses of a MEK inhibitor) via tail vein at study Day 1.
  • MCT methylcellulose
  • Example 9 PD studies of A2058 and COLO 829 melanoma xenografts
  • A2058 or COLO 829 tumors were grown to an average size of approximately 200 mm 3 , whereupon animals were randomized into groups of 5-6 each.
  • an appropriate vehicle control Kerel LF
  • RG7204 at doses of 10 mpk or 30 mpk were administered twice a day for 3 days ( Figure 27A).
  • an appropriate vehicle control or MEKi-973 at doses of 5mpk and lOmpk were administered orally once a day for 3 days (Figure 27B). Flash frozen tumors harvested at end of study were homogenized and processed for RNA and/or protein.
  • Taqman assays were set up using reagents from Applied Biosystems (ABI, Foster City, CA) and assayed using 7500 Real Time PCR machine and software from ABI. Primer-probe sets were designed with primers flanking a fluorogenic probe dual labeled with Reporter dye FAM and quencher dye TAMRA. ETBR transcript levels in the tumors were normalized against transcript levels of reference genes such as Hprtl
  • the primer-probe set for reference gene Hprtl (hypoxanthine phosphoribosyltransferase 1) is as follows: [0380] Forward primer -5' CAC ATC AAA GAC AGC ATC TAA GAA (SEQ ID NO: 17); Reverse primer-5' CAA GTT GGA AAA TAC AGT CAA CAT T (SEQ ID NO: 18) and probe-5' TTT TGT TCT GTC CTG GAA TTA TTT TAG TAG TGT TTC A (SEQ ID NO: 19).
  • the primer-probe set for ETBR is as follows:
  • the primer-probe set for reference gene GAPDH (Glyceraldehyde 3 phosphate dehydrogenase) is as follows:
  • Figure 27A shows that BRAFi induces ETBR niRNA in vivo as compared to control vehicle.
  • Figure 27B shows that MEKi-973 induces ETBR mRNA in vivo as compared to control vehicle as well.
  • Phosphorylated erk and total erk protein levels were evaluated in the tumors by western blotting using the following reagents: for detection of proteins: anti-Phospho-p44/42 MAPK (Erkl/2)
  • BRAFi appears to inhibit Perk in vivo as compared to control.

Abstract

The invention provides therapeutic combinations of anti-ETBR antibodies and MAP kinase inhibitors and methods of using the same to treat melanoma.

Description

THERAPEUTIC COMBINATIONS AND METHODS OF TREATING MELANOMA
RELATED APPLICATIONS
[0001] This application claims the benefit under 35 USC 119(e) of U.S. Provisional Application Number 61/552,893 filed 28 October 2011 and U.S. Provisional Application Number 61/678,978 filed 2 August 2012, the contents of which are incorporated herein by reference.
FIELD OF THE INVENTION
[0002] This invention concerns in general, a treatment of melanoma by using certain antibody and small molecule drug combinations.
BACKGROUND
[0003] Melanoma is an aggressive form of skin cancer that has recently undergone an alarming increase in incidence (Thompson JF et al., Cutaneous melanoma in the era of molecular profiling. Lancet 2009;374:362-5). Although cures can be achieved with surgical resection of localized lesions, the advanced stages of melanoma are only poorly responsive to currently approved therapies. The 5-year survival rate for stage IV metastatic melanoma is approximately 10% (Thompson, Lancet 2009). New therapeutic approaches, including antisense to Bcl2, antibodies to CTLA4, small molecule RAF kinase inhibitors, and adoptive immunotherapy, are currently in clinical testing for metastatic melanoma (Ascierto PA et al., Melanoma: a model for testing new agents in combination therapies. J Transl Med 2010;8:38^15). The results from some of these recent studies seem to be encouraging, but a durable impact on overall survival may require therapeutic combinations including additional new agents.
[0004] However, it is recognized that melanomas can demonstrate molecular variations, for example in certain signal transduction pathways necessary for cell responsiveness to growth factors. Therefore, rather than treating melanoma as a single disease, at have been made to stratify it into molecular subtypes in order to treat each subtype with the most appropriate therapies.
[0005] One subtype of melanoma harbors aberrations in the MAP kinase pathway. The MAPK pathway is a phosphorylation-driven signal transduction cascade that couples intracellular responses to the binding of growth factors to cell surface receptors. This pathway regulates several processes including cell proliferation and differentiation, and is often dysregulated in a variety of cancers. (Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer. 2004;4:937-947). The classical MAPK pathway consists of RAS, RAF, MEK and ERK, where RAS triggers the formation of a RAF/MEK/ERK kinase complex which then drives transcription of key regulators through protein phosphorylation. The inhibition of MAPK signaling with agents targeted toward critical proteins in the pathway has the potential to inhibit growth in a variety of tumor types (Wong K-K et al., Recent developments in anti-cancer agents targeting the Ras/Raf/MEK/ERK pathway. Recent Pat Anticancer Drug Discov. 2009;4:28-35.
[0006] Inappropriate activation of the MEK/ERK pathway promotes cell growth in the absence of exogenous growth factors. GDC-0973 (a.k.a. XL518) is a potent and highly selective small molecule inhibitor of MEK1/2, a MAPK kinase that activates ERK1/2 (Johnston S. XL518, a potent, selective, orally bioavailable MEK1 inhibitor, downregulates the Ras/Raf/MEK/ERK pathway in vivo, resulting in tumor growth inhibition and regression in preclinical models. Presented at: AACR-NCI-EORTC Symposium on Molecular Targets and Cancer Therapeutics; October 22, 2007; San Francisco, CA. Abstract C209). As a consequence, the oncogenic signal from cell surface, Ras and Raf, to ERK is interrupted. Sustained inhibition of ERK activation translates into decreased proliferation and induction of apoptosis. In multiple preclinical studies, GDC-0973 has been shown to inhibit cell growth and induce tumor regression.
[0007] Recently, vemurafenib, also known as Zelboraf®, which is a B-Raf enzyme inhibitor, was approved by the U.S. Food and Drug Administration for the treatment of late-stage melanoma. Vemurafenib has been shown to cause programmed cell death in melanoma cell lines (Sala E, et al., BRAF silencing by short hairpin RNA or chemical blockade by PLX4032 leads to different responses in melanoma and thyroid carcinoma cells. Mol. Cancer Res. 6 (5): 751-9 (May 2008). Vemurafenib interrupts the B-Raf/MEK step on the B-Raf/MEK/ERK pathway if the B-Raf has the common V600E mutation. Vemurafenib is effective in melanoma patients whose cancer has a V600E BRAF mutation (that is, at amino acid position number 600 on the B-RAF protein, the normal valine is replaced by glutamic acid). About 60% of melanomas have the V600E BRAF mutation. Melanoma cells without this mutation do not appear to be inhibited by vemurafenib; it paradoxically stimulates normal BRAF and may promote tumor growth (Hatzivassiliou G et al., RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature 464 (7287): 431-5; Halaban R et al., PLX4032, a Selective BRAF (V600E) Kinase Inhibitor, Activates the ERK Pathway and Enhances Cell Migration and Proliferation of BRAF (WT) Melanoma Cells. Pigment Cell Melanoma Res 23 (2): 190-200 (February 2010). While clinical trials revealed an improved survival, improved objective response rate, and improved progression-free survival for those patients treated with vemurafenib as compared to DTIC, disease recurrence is likely (Nazarian R. et al., Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature Vol:468, Pages: 973-977 (16 December 2010)).
[0008] Despite advances in melanoma cancer therapy, there is a great need for additional therapeutic treatments capable of effectively inhibiting neoplastic cell growth. Accordingly, it is an objective of the present invention to identify combinations of therapeutic agents to produce compositions of matter useful in the therapeutic treatment of melanoma cancers. SUMMARY
[0009] The present invention contemplates a method of tumor growth inhibition (TGI) in a subject suffering from melanoma comprising administering to the subject an effective amount of an anti- endothelin B receptor (ETBR) antibody drug conjugate in combination with an effective amount of a MAP kinase inhibitor.
In one aspect, the combination of an anti-ETBR antibody drug conjugate and a MAP kinase inhibitor is synergistic. In another aspect, with respect to the synergistic combination, the TGI is greater than the TGI seen using an anti-ETBR antibody drug conjugate alone or greater than the TGI seen using a MAP kinase inhibitor alone. In yet a further aspect, with respect to the synergistic combination, the TGI is about 10% greater, or about 15%> greater, or about 20% greater, or about 25% greater, or about 30% greater, or about 35% greater, or about 40% greater, or about 45% greater, or about 50% greater, or about 55%o greater, or about 60% greater, or about 65% greater, or about 70% greater than use of an anti-ETBR antibody drug conjugate alone or the TGI is about 10% greater, or about 15% greater, or about 20% greater, or about 25% greater, or about 30% greater, or about 35% greater, or about 40% greater, or about 45%o greater, or about 50% greater, or about 55%> greater, or about 60% greater, or about 65%> greater, or about 70%) greater than use of a MAP kinase inhibitor alone.
[0010] In another aspect of the invention described above, the anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10. In yet another aspect of the claimed method, the anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDR1 is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDR1 is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6. In still another aspect of the claimed method, the anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9. A further aspect of this method also includes an anti-ETBR antibody also having a VL which is SEQ ID NO: 8.
[0011] In one aspect of the claimed method described above, the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is a cytotoxic agent that is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes, and wherein said cytotoxin is a toxin. In another aspect of the claimed invention, the toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin. In yet another aspect of the invention, the toxin is a maytansinoid.
[0012] In one aspect of the claimed method described above, the MAP kinase inhibitor is a BRAF inhibitor. In yet another aspect of the claimed method described above, the BRAF inhibitor is propane- 1- sulfonic acid {3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl} -amide. In another aspect, the BRAF inhibitor has the following chemical structure:
Figure imgf000006_0001
[0013] In another aspect of the claimed method described above, the MAP kinase inhibitor is a MEK inhibitor. In yet another aspect of the claimed method described above, the MEK inhibitor is (S)-(3,4- difuoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone. In still another aspect of the claimed method described above, the MEK inhibitor has the following chemical structure:
Figure imgf000006_0002
[0014] In one aspect of the invention, it is contemplated that a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody is described. In another aspect of the claimed method described above, said melanoma is ETBR positive. In yet another aspect of the claimed method, said melanoma is metastatic. In still a further aspect of the claimed method, said subject has not had prior therapy with a MAP kinase inhibitor. In yet a further aspect of the claimed method, said subject has a V600E BRAF gene mutation or said subject is BRAF wildtype, having no V600E BRAF mutation. In still a further aspect of the claimed method, the subject has not had prior therapy with a MAP kinase inhibitor.
[0015] In another aspect of the claimed method described above, the combination of an anti-ETBR antibody and a MAP kinase inhibitor is synergistic. In another aspect, with respect to the synergistic combination, the TGI is greater than the TGI seen using an anti-ETBR antibody alone or greater than the TGI seen using a MAP kinase inhibitor alone. In yet a further aspect, with respect to the synergistic combination, the TGI is about 10% greater, or about 15%> greater, or about 20% greater, or about 25% greater, or about 30% greater, or about 35% greater, or about 40% greater, or about 45% greater, or about 50%) greater, or about 55% greater, or about 60% greater, or about 65% greater, or about 70% greater than use of an anti-ETBR antibody alone or the TGI is about 10% greater, or about 15% greater, or about 20% greater, or about 25% greater, or about 30% greater, or about 35% greater, or about 40% greater, or about 45%) greater, or about 50% greater, or about 55%) greater, or about 60%) greater, or about 65% greater, or about 70%) greater than use of a MAP kinase inhibitor alone.
[0016] In another aspect of the invention described above, the anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10. In yet another aspect of the claimed method, the anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDR1 is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDR1 is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6. In still another aspect of the claimed method, the anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9. A further aspect of this method also includes an anti-ETBR antibody also having a VL which is SEQ ID NO: 8.
[0017] In one aspect of the claimed method described above, the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes, and wherein said cytotoxin is a toxin. In another aspect of the claimed invention, the toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin. In yet another aspect of the invention, the toxin is a maytansinoid.
[0018] In one aspect of the claimed method described above, the MAP kinase inhibitor is a BRAF inhibitor. In yet another aspect of the method described above, the BRAF inhibitor is propane- 1 -sulfonic acid {3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl} -amide. In another aspect, the BRAF inhibitor has the following chemical structure:
Figure imgf000007_0001
[0019] In another aspect of the claimed method described above, the MAP kinase inhibitor is a MEK inhibitor. In yet another aspect of the claimed method described above, the MEK inhibitor is (S)-(3,4- difuoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone. In still another aspect of the claimed method described above, the MEK inhibitor has the following chemical structure:
Figure imgf000008_0001
[0020] In one aspect of the invention, it is contemplated that a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein the MAP kinase inhibitor is administered first to said subject in need thereof. In another aspect of the invention, said anti-ETBR antibody drug conjugate is administered after administration of said MAP kinase inhibitor. Alternatively, contemplated methods of the invention include where the anti-ETBR antibody and the MAP kinase inhibitor are administered simultaneously.
[0021] In one aspect of the invention, it is contemplated that a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein the anti-ETBR antibody drug conjugate and the MAP kinase inhibitor are administered sequentially, wherein the anti-ETBR antibody drug conjugate is administered to the subject first and the MAP kinase inhibitor is administered to the subject after administration of the anti-ETBR antibody drug conjugate.
[0022] In one aspect of the invention, it is contemplated that a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein the MAP kinase inhibitor is administered to the subject first and the anti-ETBR antibody drug conjugate is administered to the subject after administration of the MAP kinase inhibitor.
[0023] In furtherance of the above contemplated aspects of the invention, it is further contemplated that a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein said anti-ETBR antibody drug conjugate is administered intraveneously. It is also contemplated that the anti-ETBR antibody drug conjugate is dosed at about 0.1 mpk, or about 0.2 mpk, or about 0.3 mpk, or about 0.5 mpk, or about 1 mpk, or about 5 mpk, or about 10 mpk, or about 15 mpk, or about 20 mpk, or about 25 mpk, or about 30 mpk in the claimed methods of the invention.
[0024] In furtherance of the above contemplated aspects of the invention, it is further contemplated that a method of treating melanoma comprising administering to a subject in need thereof a therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody drug conjugate is described, wherein the MAP kinase inhibitor is administered orally. It is also contemplated that the BRAF inhibitor is dosed at about 1 mpk, or about 2 mpk, or about 3 mpk, or about 4 mpk, or about 5 mpk, or about 6 mpk, or about 7 mpk, or about 8 mpk, or about 9 mpk, or about 10 mpk, or about 11 mpk, or about 12 mpk, or about 15 mpk, or about 20 mpk or about 30 mpk in the claimed methods of the invention. [0025] In one aspect of the invention, it is contemplated that an article of manufacture is used for TGI in a subject suffering from melanoma comprising a package comprising an anti-ETBR antibody drug conjugate composition and a MAP kinase inhibitor composition. It is further contemplated that said anti- ETBR antibody drug conjugate specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10. Alternatively, it is also contemplated that said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDRl is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDRl is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6. A further alternative that is contemplated is an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9. In yet another alternative, an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO: 7 or 9 and the VL is SEQ ID NO: 8. In another aspect of the article of manufacture, the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. In one further aspect, the cytotoxin is a toxin wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin. In one aspect, the toxin is a maytansinoid.
[0026] In one aspect of the method of treating melanoma described above, it is also contemplated that the MAP kinase inhibitor is a BRAF inhibitor. In yet another aspect of the method described above, the BRAF inhibitor is propane- 1 -sulfonic acid {3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3- carbonyl]-2,4-difluoro-phenyl} -amide. Further, it is contemplated that the BRAF inhibitor has the following chemical structure:
Figure imgf000009_0001
[0027] In another aspect of the claimed method described above, the MAP kinase inhibitor is a MEK inhibitor. In yet another aspect of the claimed method described above, the MEK inhibitor is (S)-(3,4- difuoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone. In still another aspect of the claimed method described above, the MEK inhibitor has the following chemical structure:
Figure imgf000010_0001
[0028] In one aspect of the invention, it is contemplated that an article of manufacture for treating melanoma in a subject comprising a package comprising an anti-ETBR antibody drug conjugate composition and a MAP kinase inhibitor composition. It is further contemplated that said anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10. Alternatively, it is also contemplated that said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDRl is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDRl is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6. A further alternative that is contemplated is an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9. In yet another alternative, an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO: 7 or 9 and the VL is SEQ ID NO: 8. In another aspect of the article of manufacture, the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. In one further aspect, the cytotoxin is a toxin wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin. In one aspect, the toxin is a maytansinoid.
[0029] In one aspect of the article of manufacture described above, it is also contemplated that the MAP kinase inhibitor is a BRAF inhibitor. In yet another aspect of the article of manufacture described above, the BRAF inhibitor is propane- 1 -sulfonic acid {3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3- carbonyl]-2,4-difluoro-phenyl} -amide. Further, it is contemplated that the BRAF inhibitor has the following chemical structure:
Figure imgf000010_0002
[0030] In another aspect of the article of manufacture described above, the MAP kinase inhibitor is a MEK inhibitor. In yet another aspect of the article of manufacture described above, the MEK inhibitor is (S)-(3,4-dimoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l - yl)methanone. In still another aspect of the article of manufacture described above, the MEK inhibitor has the following chemical structure:
Figure imgf000011_0001
[0031] It is contemplated that one aspect of the invention is use of an anti-ETBR antibody drug conjugate and a MAP kinase inhibitor in the preparation of a medicament for TGI of a melanoma. It is further contemplated that said anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10. Alternatively, it is also contemplated that said anti- ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDR1 is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDR1 is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6. A further alternative that is contemplated is an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9. In yet another alternative, an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO: 7 or 9 and the VL is SEQ ID NO: 8. In another aspect of the article of manufacture, the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. In one further aspect, the cytotoxin is a toxin wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin. In one aspect, the toxin is a maytansinoid.
[0032] In one aspect of the use of the medicament described above, it is also contemplated that the MAP kinase inhibitor is a BRAF inhibitor. In yet another aspect of the use of the medicament described above, the BRAF inhibitor is propane- 1 -sulfonic acid {3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3- carbonyl]-2,4-difluoro-phenyl} -amide. Further, it is contemplated that the BRAF inhibitor has the following chemical structure:
Figure imgf000012_0001
[0033] In another aspect of the use of the medicament described above, the MAP kinase inhibitor is a MEK inhibitor. In yet another aspect of the use of the medicament described above, the MEK inhibitor is (S)-(3,4-dimoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l - yl)methanone. In still another aspect of the use of the medicament described above, the MEK inhibitor has the following chemical structure:
Figure imgf000012_0002
[0034] It is contemplated that one aspect of the invention is use of an article of manufacture comprising an anti-ETBR antibody drug conjugate composition and a MAP kinase inhibitor composition in the preparation of a medicament for TGI of a melanoma. It is further contemplated that said anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10. Alternatively, it is also contemplated that said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDRl is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDRl is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6. A further alternative that is contemplated is an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9. In yet another alternative, an anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO: 7 or 9 and the VL is SEQ ID NO: 8. In another aspect of the use of the article of manufacture, the anti-ETBR antibody is conjugated to a cytotoxin, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes. In one further aspect, the cytotoxin is a toxin wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin. In one aspect, the toxin is a maytansinoid.
[0035] In one aspect of the use of the article of manufacture described above, it is also contemplated that the MAP kinase inhibitor is a BRAF inhibitor. In yet another aspect of the use of the article of manufacture described above, the BRAF inhibitor is propane- 1 -sulfonic acid {3-[5-(4-chlorophenyl)-lH- pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl}-amide. Further, it is contemplated that the BRAF inhibitor has the following chemical structure:
Figure imgf000013_0001
[0036] In another aspect of the use of the article of manufacture described above, the MAP kinase inhibitor is a MEK inhibitor. In yet another aspect of the use of the article of manufacture described above, the MEK inhibitor is (S)-(3,4-difuoro-2-((2-fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3- (piperidin-2yl)azetidin-l -yl)methanone. In still another aspect of the use of the article of manufacture described above, the MEK inhibitor has the following chemical structure:
Figure imgf000013_0002
BRIEF DESCRIPTION OF THE FIGURES [0037] Figure 1 is a schematic of the MAP kinase pathway.
[0038] Figure 2 demonstrates the relationship of receptor level to ADC cell killing in vitro. The indicated number of receptor copies/cell was estimated by Scatchard analysis. Panel A shows cell killing by anti-ETBR ADC titration for the melanoma cell line UACC-257X2.2 and panel B for melanoma cell line A2058. The indicated concentrations of anti-ETBR ADC (Hu5E9vl-vc-MMAE), control IgG-vc- MMAE, or equivalent amount of PBS vehicle control were incubated with cells for 5 days and relative cell viability (y-axis) assessed using CellTiter-Glo.
[0039] Figure 3 shows the in vivo efficacy of anti-ETBR ADC in xenografts mouse models.
Subcutaneous tumors were established in mice inoculated with UACC-257X2.2 (Panel A) or A2058 (Panel B) cells. When tumor volumes reached approximately 200 mm3 (day 0), animals were given a single IV injection of either control ADC (Control-vc-MMAE) or anti-ETBR ADC (Hu5E9vl -vc- MMAE) at the indicated doses. Average tumor volumes with standard deviations were determined from 10 animals per groups (indicated on graph).
[0040] Figure 4 shows ETBR expression in UACC-257X2.2 melanoma cells treated for 24h with varying concentrations of BRAFi-945. Panel A shows ETBR transcript normalized to RPL19 transcript. Panel B shows the expression of total ETBR and GAPDH (Control) protein in 50μg whole cell lysates. Panel C shows surface ETBR protein expression in live cells as seen by flow cytometry, where the first peak indicates cells treated to secondary detection reagent alone, the middle peak indicates cells untreated with BRAF inhibitor, and the last peak indicates BRAF inhibitor treated cells.
[0041] Figure 5 shows in vivo combination efficacy of anti-ETBR ADC (Hu5E9vl-vc-MMAE) and BRAFi-945 against UACC-257X2.2 melanoma xenograft mouse models at varying doses. Subcutaneous tumors were established in mice inoculated with UACC -257X2.2 cell lines. When tumor volumes reached approximately 200 mm3 (day 0), animals were dosed orally once a day for 21 days with BRAFi- 945 or vehicle control. On day 1 (after two doses of BRAFi-945), animals were given a single IV injection of either vehicle or anti-ETBR ADC at the indicated doses. Average tumor volumes with standard deviations were determined from 10 animals per group. Drug and dosage information are as indicated: Panel A shows a 1 mpk BRAFi-945 and 1 mpk anti-ETBR-ADC (Hu5E9vl -vc-MMAE) combination; panel B shows a 1 mpk BRAFi-945 and 3 mpk anti-ETBR-ADC (Hu5E9vl -vc-MMAE) combination; panel C shows a 6 mpk BRAFi-945 and 1 mpk anti-ETBR-ADC (Hu5E9vl -vc-MMAE) combination; panel D shows a 6 mpk BRAFi-945 and 3 mpk anti-ETBR-ADC (Hu5E9vl -vc-MMAE) combination; and panel E shows a 20 mpk BRAFi-945 and 3 mpk anti-ETBR-ADC (Hu5E9vl -vc- MMAE) combination.
[0042] Figure 6 shows ETBR expression in COLO 829 melanoma cells treated for 24h with varying concentrations of BRAF inhibitor RG7204. Panel A shows ETBR transcript normalized to RPL19 transcript. Panel B shows expression of total ETBR and GAPDH (Control) protein in 50μg whole cell lysates. Panel C shows surface ETBR protein expression in live cells as seen by flow cytometry, where the first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with BRAF inhibitor and the third peak indicates BRAF inhibitor treated cells.
[0043] Figure 7 demonstrates the in vivo combination efficacy of anti-ETBR ADC and BRAF inhibitor RG7204 against COLO 829 melanoma xenografts mouse model. Subcutaneous tumors were established in mice inoculated with COLO 829 melanoma cell lines. When tumor volumes reached approximately 200 mm3 (day 0), animals were dosed orally twice a day for 21 days with RG7204. On day 1 (after three doses of RG7204), animals were given a single IV injection of either vehicle or anti-ETBR ADC (Hu5E9vl -vc-MMAE) at the indicated doses. Average tumor volumes with standard deviations were determined from 9 animals per group. Drug and dosage information are as indicated: panel A shows 3 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 30 mpk of RG7204; panel B shows 1 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 30 mpk of RG7204; panel C shows 1 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 10 mpk of RG7204; and panel D shows 3 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 10 mpk of RG7204.
[0044] Figure 8 shows ETBR expression in A2058 melanoma cells treated for 24h with varying concentrations of BRAF inhibitor RG7204. Panel A shows ETBR transcript normalized to RPL19 transcript; panel B shows expression of total ETBR and GAPDH (Control) protein in 100μg whole cell lysates; and panel C shows surface ETBR protein expression in live cells as seen by flow cytometry. The first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with BRAF inhibitor, and the third peak indicates BRAF inhibitor treated cells.
[0045] Figure 9 demonstrates in vivo combination efficacy of anti-ETBR ADC (Hu5E9vl -vc-MMAE) and BRAF inhibitor RG7204 against A2058 melanoma xenograft mouse models. Subcutaneous tumors were established in mice inoculated with A2058 melanoma cell lines. When tumor volumes reached approximately 200 mm3 (day 0), animals were dosed orally twice a day for 21 days with RG7204. On day 1 (after three doses of RG7204), animals were given a single IV injection of either vehicle or anti- ETBR ADC (Hu5E9vl -vc-MMAE) at the indicated doses. Average tumor volumes with standard deviations were determined from 10 animals per group. Drug and dosage information indicated on each graph as follows: Panel A shows 6 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 10 mpk of RG7204; panel B shows 6 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 30 mpk of RG7204; panel C shows 3 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 10 mpk of RG7204; and panel D shows 3 mpk of anti-ETBR-ADC (Hu5E9vl -vc- MMAE) in combination with 30 mpk of RG7204.
[0046] Figure 10 shows Western blot experiments performed with BRAFi RG7204 showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and β-tubulin in 25 to 100μg whole cell lysates from IPC-298 melanoma cells.
[0047] Figure 11 shows surface ETBR protein expression in IPC-298 live cells as seen by flow cytometry after incubation with 0.1 μΜ, 1 μΜ and 10 μΜ of BRAFi RG7204 (panels A, B and C respectively). The first peak indicates cells treated to secondary detection reagent alone, the second peak indicates BRAF inhibitor treated cells and the third peak indicates cells untreated with BRAF inhibitor.
[0048] Figure 12 shows Western blot experiments performed with MEKi-623 (panel A) and MEKi-973 (panel B) at concentrations of 0 μΜ, 0.01 μΜ, 0.1 μΜ and 1 μΜ showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and β-tubulin in 50μg whole cell lysates from COL0829 melanoma cells.
[0049] Figure 13 shows surface ETBR protein expression in COLO 829 live cells as seen by flow cytometry after incubation with 0.01 μΜ (panels A and D), 0.1 μΜ (panels B and E) and 1 μΜ (panel C and F) of MEKi-623 (panels A, B and C respectively) or MEKi-973 (panels D, E and F respectively). The first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with MEK inhibitor and the third peak indicates MEK inhibitor treated cells.
[0050] Figure 14 shows ETBR mRNA expression in A2058 melanoma cells treated for 24h with varying concentrations of MEKi-623 (panel A) or MEKi-973 (panel B), normalized to RPL19 transcript.
[0051] Figure 15 shows Western blot experiments performed with MEKi-623 (panel A) and MEKi-973 (panel B) at concentrations of 0 μΜ, 0.01 μΜ, 0.1 μΜ and 1 μΜ showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and β-tubulin in 50-100μg whole cell lysates from A2058 melanoma cells.
[0052] Figure 16 shows surface ETBR protein expression in A2058 live cells as seen by flow cytometry after incubation with 0.01 μΜ (panels A and D), 0.1 μΜ (panels B and E) and 1 μΜ (panel C and F) of MEKi-623 (panels A, B and C respectively) or MEKi-973 (panels D, E and F respectively). The first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with MEK inhibitor and the third peak indicates MEK inhibitor treated cells.
[0053] Figure 17 demonstrates in vivo combination efficacy of anti-ETBR ADC (Hu5E9vl -vc-MMAE) and MEKi-973 against A2058 melanoma xenograft mouse models. Subcutaneous tumors were established in mice inoculated with A2058 melanoma cell lines. When tumor volumes reached approximately 200 mm3 (day 0), animals were dosed orally once a day for 21 days with MEKi-973. On day 1 (after two doses of MEKi-973), animals were given a single IV injection of either vehicle or anti- ETBR ADC (Hu5E9vl -vc-MMAE) at the indicated doses. Average tumor volumes with standard deviations were determined from 9 animals per group. Drug and dosage information indicated on each graph as follows: Panel A shows 7.5 mpk of anti-gD ADC (control) in combination with 7.5 mpk of MEKi-973 as compared to a vehicle control and anti-gD ADC alone; panel B shows 6 mpk of anti-ETBR- ADC (Hu5E9vl -vc-MMAE) in combination with 7.5 mpk of MEKi-973 as compared to a vehicle control and 7.5 mpk MEKi-973 alone (GDC-0973) or 6 mpk of anti-ETBR-ADC alone.
[0054] Figure 18 shows ETBR transcript expression in SK23-MEL melanoma cells treated for 24h with varying concentrations of MEKi-623 (panel A) or MEKi-973 (panel B), which were normalized to RPL19 transcript.
[0055] Figure 19 shows Western blot experiments performed with MEKi-623 (panel A) and MEKi-973 (panel B) at concentrations of 0 μΜ, 0.01 μΜ, 0.1 μΜ and 1 μΜ showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and β-tubulin in 50μg whole cell lysates from SK23- MEL melanoma cells.
[0056] Figure 20 shows surface ETBR protein expression in live SK23-MEL cells as seen by flow cytometry after incubation with 0.01 μΜ (panels A and D), 0.1 μΜ (panels B and E) and 1 μΜ (panel C and F) of MEKi-623 (panels A, B and C respectively) or MEKi-973 (panels D, E and F respectively). The first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with MEK inhibitor and the third peak indicates MEK inhibitor treated cells. [0057] Figure 21 demonstrates in vivo combination efficacy of anti-ETBR ADC (Hu5E9vl-vc-MMAE) and MEKi-973 against SK23-MEL melanoma xenograft mouse models. Drug and dosage information indicated on each graph as follows: Panel A shows 6 mpk of anti-gD ADC (control) in combination with 7.5 mpk of MEKi-973 as compared to a vehicle control, 7.5 mpk MEKi-973 and 6 mpk anti-gD ADC alone; panel B shows 6 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 7.5 mpk of MEKi-973 ("Combination") as compared to a vehicle control and 7.5 mpk MEKi-973 alone or 6 mpk of anti-ETBR-ADC alone. Panel C shows 3 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 3 mpk of MEKi-973 ("Combination"), as compared to a vehicle control, 3 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) or 3 mpk of MEKi-973. Panel D shows 3 mpk of anti-ETBR-ADC (Hu5E9vl -vc- MMAE) in combination with 7.5 mpk of MEKi-973 ("Combination") as compared to a vehicle control and 7.5 mpk MEKi-973 alone or 3 mpk of anti-ETBR-ADC alone. Panel E shows 6 mpk of anti-ETBR- ADC (Hu5E9vl -vc-MMAE) in combination with 3 mpk of MEKi-973 ("Combination") as compared to a vehicle control and 3 mpk MEKi-973 alone or 6 mpk of anti-ETBR-ADC alone.
[0058] Figure 22 shows Western blot experiments performed with MEKi-623 (panel A) and MEKi-973 (panel B) at concentrations of 0 μΜ, 0.01 μΜ, 0.1 μΜ and 1 μΜ showing expression of total ETBR, Perk and erk proteins and control proteins GAPDH and β-tubulin in 25-100μg whole cell lysates from IPC-298 melanoma cells.
[0059] Figure 23 shows surface ETBR protein expression in live IPC-298 cells as seen by flow cytometry after incubation with 0.01 μΜ (panels A and D), 0.1 μΜ (panels B and E) and 1 μΜ (panel C and F) of MEKi-623 (panels A, B and C respectively) or MEKi-973 (panels D, E and F respectively). The first peak indicates cells treated to secondary detection reagent alone, the second peak indicates cells untreated with MEK inhibitor and the third peak indicates MEK inhibitor treated cells.
[0060] Figure 24 demonstrates in vivo combination efficacy of anti-ETBR ADC (Hu5E9vl -vc-MMAE) and MEKi-623 against IPC-298 melanoma xenograft mouse models. Drug and dosage information indicated on each graph as follows: Panel A shows 6 mpk of anti-gD ADC (control) in combination with 1 mpk of MEKi-623 as compared to a vehicle control, 1 mpk MEKi-623 and 6 mpk anti-gD ADC alone; panel B shows 6 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 1 mpk of MEKi- 623 ("Combination") as compared to a vehicle control and 1 mpk MEKi-623 alone or 6 mpk of anti- ETBR-ADC alone.
[0061] Figure 25 in vivo combination efficacy of anti-ETBR ADC (Hu5E9vl -vc-MMAE) and MEKi- 973 against IPC-298 melanoma xenograft mouse models. Drug and dosage information indicated on each graph as follows: Panel A shows 6 mpk of anti-gD ADC (control) in combination with 7.5 mpk of MEKi-973 as compared to a vehicle control, 7.5 mpk MEKi-973 and 6 mpk anti-gD ADC alone; panel B shows 6 mpk of anti-ETBR-ADC (Hu5E9vl -vc-MMAE) in combination with 7.5 mpk of MEKi-973 ("Combination") as compared to a vehicle control and 7.5 mpk MEKi-973 alone or 6 mpk of anti-ETBR- ADC alone.
[0062] Figure 26 depicts expression of phosphorylated erk and total erk protein in COLO 829 tumors treated with either vehicle or 30mpk BRAFi RG7204. [0063] Figure 27 depicts ETBR transcript expression in COLO 829 tumors treated with BRAFi RG7204 (panel A) and in A2058 tumors treated with MEKi-973 for 3 days (panel B). Panel A shows ETBR transcript normalized to control GAPDH in COLO 829 cell line, in COLO 892 tumors treated with either vehicle control or 10 mpk or 30 mpk RG7204. Panel B shows ETBR transcript normalized to control Hprtl in A2058 tumors treated with either vehicle control or 5 or 10 mpk of MEKi-973.
DETAILED DESCRIPTION OF EMBODIMENTS OF THE INVENTION
I. DEFINITIONS
[0064] An "acceptor human framework" for the purposes herein is a framework comprising the amino acid sequence of a light chain variable domain (VL) framework or a heavy chain variable domain (VH) framework derived from a human immunoglobulin framework or a human consensus framework, as defined below. An acceptor human framework "derived from" a human immunoglobulin framework or a human consensus framework may comprise the same amino acid sequence thereof, or it may contain amino acid sequence changes. In some embodiments, the number of amino acid changes are 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less. In some embodiments, the VL acceptor human framework is identical in sequence to the VL human immunoglobulin framework sequence or human consensus framework sequence.
[0065] "Affinity" refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). Unless indicated otherwise, as used herein, "binding affinity" refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen). The affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (Kd). Affinity can be measured by common methods known in the art, including those described herein. Specific illustrative and exemplary embodiments for measuring binding affinity are described in the following.
[0066] An "affinity matured" antibody refers to an antibody with one or more alterations in one or more hypervariable regions (HVRs), compared to a parent antibody which does not possess such alterations, such alterations resulting in an improvement in the affinity of the antibody for antigen.
[0067] The terms "anti-ETBR antibody" and "an antibody that binds to ETBR" refer to an antibody that is capable of binding the endothelin B receptor (ETBR) with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting ETBR. In one embodiment, the extent of binding of an anti-ETBR antibody to an unrelated, non-ETBR protein is less than about 10% of the binding of the antibody to ETBR as measured, e.g., by a radioimmunoassay (RIA). In certain embodiments, an antibody that binds to ETBR has a dissociation constant (Kd) of < 1 μΜ, < 100 nM, < 10 nM, < 1 nM, < 0.1 nM, < 0.01 nM, or < 0.001 nM (e.g. 10"8 M or less, e.g. from 10"8 M to 10"13 M, e.g., from 10"9 M to 10"13 M). In certain embodiments, an anti-ETBR antibody binds to an epitope of ETBR that is conserved among ETBR from different species. [0068] The term "antibody" herein is used in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity.
[0069] An "antibody fragment" refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. scFv); and multispecific antibodies formed from antibody fragments.
[0070] An "antibody that binds to the same epitope" as a reference antibody refers to an antibody that blocks binding of the reference antibody to its antigen in a competition assay by 50% or more, and conversely, the reference antibody blocks binding of the antibody to its antigen in a competition assay by 50% or more. An exemplary competition assay is provided herein.
[0071] The term "BRAF" as used herein refers to a serine/threonine-protein kinase B-Raf, also known as proto-oncogene B-Raf or v-Raf murine sarcoma viral oncogene homolog Bl, which is a protein that in humans is encoded by the BRAF gene. The B-Raf protein is involved in sending signals in cells and in cell growth.
[0072] The term "BRAF inhibitor" or "BRAFi" as used herein refers to any number of known small molecule drug compounds which can inhibit or interrupt the B-Raf/MEK step on the B-Raf/MEK/ERK pathway. Examples of suitable BRAFi may include, but are not limited to, those described in
International Patent Application PCT/US2010/047007 filed August 27, 2010, in International Patent Application PCT/US2010/046975 filed August 27, 2010; in International Patent Application
PCT/US2010/046952 filed August 27, 2010; in International Patent Application PCT/US2010/046955 filed August 27, 2010; and in International Patent Application PCT/US2006/024361 filed June 21, 2006. Another example may be, but is not limited to, GSK 2118436, having a CAS registry number 405554-55- 4, which is also known as 5-[2-[4-[2-(Dimethylamino)ethoxy]phenyl]-5-(4-pyridinyl)-lH-imidazol-4-yl]- 2,3-dihydro-lH-inden-l-one oxime.
[0073] The term "chimeric" antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species.
[0074] The "class" of an antibody refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGi, IgG2, IgG3, IgG i, IgAi, and IgA2. The heavy chain constant domains that correspond to the different classes of immunoglobulins are called α, δ, ε, γ, and μ, respectively.
[0075] The term "cytotoxic agent" as used herein refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction. Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At211, 1131, 1125, Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 , Zr89 and radioactive isotopes of Lu); chemotherapeutic agents or drugs (e.g., methotrexate, adriamicin, vinca alkaloids (vincristine, vinblastine, etoposide), doxorubicin, melphalan, mitomycin C, chlorambucil, daunorubicin or other intercalating agents); growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; antibiotics; toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof; and the various antitumor or anticancer agents disclosed below.
[0076] "Effector functions" refer to those biological activities attributable to the Fc region of an antibody, which vary with the antibody isotype. Examples of antibody effector functions include: Clq binding and complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell- mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g. B cell receptor); and B cell activation.
[0077] An "effective amount" of an agent, e.g., a pharmaceutical formulation, refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
[0078] The term "ETBR," as used herein, refers to any native endothelin B receptor (ETBR) from any vertebrate source, including mammals such as primates (e.g. humans) and rodents (e.g., mice and rats), unless otherwise indicated. The term encompasses "full-length," unprocessed ETBR as well as any form of ETBR that results from processing in the cell. The term also encompasses naturally occurring variants of ETBR, e.g., splice variants or allelic variants. The amino acid sequence of an exemplary human ETBR is shown in SEQ ID NO: 10 (see Nakamuta M et al., Cloning and Sequence Analysis of a cDNA encoding Human non-selective type of endothelin receptor, Biochem Biophys Res Commun. 1991 May 31 : 177(l):34-9).
[0079] The term "anti-ETBR antibody - ADC" as used herein, refers to any anti-ETBR antibody described herein that is conjugated to a toxin. Such toxins include, but are not limited to, maytansinoids or specifically monomethylauristatin (MMAE). An anti-ETBR antibody- ADC is contemplated as a species of "anti-ETBR antibodies of the invention".
[0080] The term "Fc region" herein is used to define a C-terminal region of an immunoglobulin heavy chain that contains at least a portion of the constant region. The term includes native sequence Fc regions and variant Fc regions. In one embodiment, a human IgG heavy chain Fc region extends from Cys226, or from Pro230, to the carboxyl-terminus of the heavy chain. However, the C-terminal lysine (Lys447) of the Fc region may or may not be present. Unless otherwise specified herein, numbering of amino acid residues in the Fc region or constant region is according to the EU numbering system, also called the EU index, as described in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD, 1991.
[0081] "Framework" or "FR" refers to variable domain residues other than hypervariable region (HVR) residues. The FR of a variable domain generally consists of four FR domains: FRl, FR2, FR3, and FR4. Accordingly, the HVR and FR sequences generally appear in the following sequence in VH (or VL): FRl -H 1 (L 1 )-FR2-H2(L2)-FR3 -H3 (L3)-FR4. [0082] The terms "full length antibody," "intact antibody," and "whole antibody" are used herein interchangeably to refer to an antibody having a structure substantially similar to a native antibody structure or having heavy chains that contain an Fc region as defined herein.
[0083] The term "945" or BRAFi-945" as used herein refers to a B-Raf enzyme inhibitor that is 4- amino-N-(6-chloro-2-fluoro-3-(3-fluoro propyl sulfonamido) phenyl)thieno[3,2-d]pyrimidine-7- carboxamide and has a structure having the following formula as disclosed in Example 15 of
International Patent Application PCT/US2010/046955 filed August 27, 2010 which is incorporated herein by reference in its entirety:
Figure imgf000021_0001
[0084] The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include "transformants" and "transformed cells," which include the primary transformed cell and progeny derived therefrom without regard to the number of passages. Progeny may not be completely identical in nucleic acid content to a parent cell, but may contain mutations. Mutant progeny that have the same function or biological activity as screened or selected for in the originally transformed cell are included herein.
[0085] A "human antibody" is one which possesses an amino acid sequence which corresponds to that of an antibody produced by a human or a human cell or derived from a non-human source that utilizes human antibody repertoires or other human antibody-encoding sequences. This definition of a human antibody specifically excludes a humanized antibody comprising non-human antigen-binding residues.
[0086] A "human consensus framework" is a framework which represents the most commonly occurring amino acid residues in a selection of human immunoglobulin VL or VH framework sequences.
Generally, the selection of human immunoglobulin VL or VH sequences is from a subgroup of variable domain sequences. Generally, the subgroup of sequences is a subgroup as in Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, NIH Publication 91-3242, Bethesda MD (1991), vols. 1-3. In one embodiment, for the VL, the subgroup is subgroup kappa I as in Kabat et al., supra. In one embodiment, for the VH, the subgroup is subgroup III as in Kabat et al., supra.
[0087] A "humanized" antibody refers to a chimeric antibody comprising amino acid residues from non- human HVRs and amino acid residues from human FRs. In certain embodiments, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody. A humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. A "humanized form" of an antibody, e.g., a non-human antibody, refers to an antibody that has undergone humanization.
[0088] The term "hypervariable region" or "HVR," as used herein, refers to each of the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops ("hypervariable loops"). Generally, native four-chain antibodies comprise six HVRs; three in the VH (HI, H2, H3), and three in the VL (LI, L2, L3). HVRs generally comprise amino acid residues from the hypervariable loops and/or from the "complementarity determining regions" (CDRs), the latter being of highest sequence variability and/or involved in antigen recognition. Exemplary hypervariable loops occur at amino acid residues 26-32 (LI), 50-52 (L2), 91-96 (L3), 26-32 (HI), 53-55 (H2), and 96-101 (H3). (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987).) Exemplary CDRs (CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3) occur at amino acid residues 24-34 of LI, 50-56 of L2, 89- 97 of L3, 31-35B of HI, 50-65 of H2, and 95-102 of H3. (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991).) With the exception of CDR1 in VH, CDRs generally comprise the amino acid residues that form the hypervariable loops. CDRs also comprise "specificity determining residues," or "SDRs," which are residues that contact antigen. SDRs are contained within regions of the CDRs called abbreviated- CDRs, or a-CDRs. Exemplary a-CDRs (a-CDR-Ll, a-CDR-L2, a-CDR-L3, a-CDR-Hl, a-CDR-H2, and a-CDR-H3) occur at amino acid residues 31-34 of LI, 50-55 of L2, 89-96 of L3, 31-35B of HI, 50-58 of H2, and 95-102 of H3. (See Almagro and Fransson, Front. Biosci. 13: 1619-1633 (2008).) Unless otherwise indicated, HVR residues and other residues in the variable domain (e.g., FR residues) are numbered herein according to Kabat et al., supra.
[0089] An "immunoconjugate" is an antibody conjugated to one or more heterologous molecule(s), including but not limited to a cytotoxic agent.
[0090] An "individual" or "subject" is a mammal. Mammals include, but are not limited to, domesticated animals (e.g., cows, sheep, cats, dogs, and horses), primates (e.g., humans and non-human primates such as monkeys), rabbits, and rodents (e.g., mice and rats). In certain embodiments, the individual or subject is a human.
[0091] An "isolated" antibody is one which has been separated from a component of its natural environment. In some embodiments, an antibody is purified to greater than 95% or 99% purity as determined by, for example, electrophoretic (e.g., SDS-PAGE, isoelectric focusing (IEF), capillary electrophoresis) or chromatographic (e.g., ion exchange or reverse phase HPLC). For review of methods for assessment of antibody purity, see, e.g., Flatman et al., J. Chromatogr. B 848:79-87 (2007).
[0092] An "isolated" nucleic acid refers to a nucleic acid molecule that has been separated from a component of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in cells that ordinarily contain the nucleic acid molecule, but the nucleic acid molecule is present extrachromosomally or at a chromosomal location that is different from its natural chromosomal location. [0093] "Isolated nucleic acid encoding an anti-ETBR antibody" refers to one or more nucleic acid molecules encoding antibody heavy and light chains (or fragments thereof), including such nucleic acid molecule(s) in a single vector or separate vectors, and such nucleic acid molecule(s) present at one or more locations in a host cell.
[0094] The term "mitogen-activated protein kinase" (MAP kinase) as used herein refers to the serine/threonine-specific protein kinases belonging to the CMGC (CDK/MAPK/GSK3/CLK) kinase group. The ERK1/2 pathway of mammals is probably the best characterized MAPK system. The most important upstream activators of this pathway are the Raf proteins (A-Raf, B-Raf or c-Raf), the key mediators of response to growth factors (EGF, FGF, PDGF, etc.).
[0095] The term "MAPK/ERK kinase" (MEK) as used herein refers to a tyrosine kinase which occupies a central role in the MAPK pathway. Expression of constitutive ly active forms of MEK leads to transformation of cell lines.
[0096] The term "MEK inhibitor" (MEKi) as used herein refers to any number of known small molecule drug compounds which can inhibit or interrupt the MEK step on the MAP kinase pathway. Examples of suitable MEKi may include, but are not limited to, those described as MEKi-623, MEKi-973, or GSK1120212.
[0097] The term "MEKi-973" as used herein refers to a MEK inhibitor (S)-(3,4-difuoro-2-((2-fluoro-4- iodo henyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone, having the structure:
Figure imgf000023_0001
[0098] The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen. Thus, the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary methods for making monoclonal antibodies being described herein. [0099] A "naked antibody" refers to an antibody that is not conjugated to a heterologous moiety (e.g., a cytotoxic moiety) or radiolabel. The naked antibody may be present in a pharmaceutical formulation.
[0100] "Native antibodies" refer to naturally occurring immunoglobulin molecules with varying structures. For example, native IgG antibodies are heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light chains and two identical heavy chains that are disulfide-bonded. From N- to C-terminus, each heavy chain has a variable region (VH), also called a variable heavy domain or a heavy chain variable domain, followed by three constant domains (CHI, CH2, and CH3). Similarly, from N- to C-terminus, each light chain has a variable region (VL), also called a variable light domain or a light chain variable domain, followed by a constant light (CL) domain. The light chain of an antibody may be assigned to one of two types, called kappa (κ) and lambda (λ), based on the amino acid sequence of its constant domain.
[0101] The term "package insert" is used to refer to instructions customarily included in commercial packages of therapeutic products, that contain information about the indications, usage, dosage, administration, combination therapy, contraindications and/or warnings concerning the use of such therapeutic products.
[0102] "Percent (%) amino acid sequence identity" with respect to a reference polypeptide sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the reference polypeptide sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid sequence identity values are generated using the sequence comparison computer program ALIGN-2. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office, Washington D.C., 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, California, or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
[0103] In situations where ALIGN-2 is employed for amino acid sequence comparisons, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows: 100 times the fraction X/Y
[0104] where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A. Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as described in the immediately preceding paragraph using the ALIGN-2 computer program.
[0105] The term "pharmaceutical formulation" refers to a preparation which is in such form as to permit the biological activity of an active ingredient contained therein to be effective, and which contains no additional components which are unacceptably toxic to a subject to which the formulation would be administered.
[0106] A "pharmaceutically acceptable carrier" refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject., A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
[0107] The term "RG7204" refers to a B-Raf enzyme inhibitor that has a molecular formula of C23H18CIF2N3C>3S and the following structure:
Figure imgf000025_0001
[0108] As used herein, "treatment" (and grammatical variations thereof such as "treat" or "treating") refers to clinical intervention in an attempt to alter the natural course of the individual being treated, and can be performed either for prophylaxis or during the course of clinical pathology. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. In some embodiments, antibodies of the invention are used to delay development of a disease or to slow the progression of a disease.
[0109] The term "variable region" or "variable domain" refers to the domain of an antibody heavy or light chain that is involved in binding the antibody to antigen. The variable domains of the heavy chain and light chain (VH and VL, respectively) of a native antibody generally have similar structures, with each domain comprising four conserved framework regions (FRs) and three hypervariable regions (HVRs). (See, e.g., Kindt et al. Kuby Immunology, 6th ed., W.H. Freeman and Co., page 91 (2007).) A single VH or VL domain may be sufficient to confer antigen-binding specificity. Furthermore, antibodies that bind a particular antigen may be isolated using a VH or VL domain from an antibody that binds the antigen to screen a library of complementary VL or VH domains, respectively. See, e.g., Portolano et al., J. Immunol. 150:880-887 (1993); Clarkson et al., Nature 352:624-628 (1991).
[0110] The term "vector," as used herein, refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes the vector as a self-replicating nucleic acid structure as well as the vector incorporated into the genome of a host cell into which it has been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors."
II. COMPOSITIONS AND METHODS
[0111] In one aspect, the invention is based, in part, on antibodies that bind to ETBR. Antibodies of the invention are useful, e.g., for the treatment of melanoma.
[0112] Exemplary Anti-ETBR Antibodies
[0113] In one aspect, the invention provides isolated antibodies that bind to ETBR. In certain embodiments, an anti-ETBR antibody comprises at least one, two, three, four, five, or six CDRs selected from (a) CDR-L1 (KS S Q SLLD SDGKTYLN, SEQ ID NO:7), (b) CDR-L2 (LVSKLDS, SEQ ID NO:8), (c) CDR-L3 (WQGTHFPYT; SEQ ID NO:9), (d) CDR-H1 (GYTFTSYWMQ; SEQ ID NO: l), (e) CDR- H2 (TIYPGDGDTSYAQKFKG; SEQ ID NO:2), and (f) CDR-H3 (WGYAYDIDN; SEQ ID NO:3).
[0114] In any of the above embodiments, an anti-ETBR antibody is humanized. In one embodiment, an anti-ETBR antibody comprises CDRs as in any of the above embodiments, and further comprises an acceptor human framework, e.g. a human immunoglobulin framework or a human consensus framework. In another aspect, the invention provides an isolated anti-ETBR antibody having the VL amino acid sequence of SEQ ID NO:8, and the VH amino acid sequence of SEQ ID NO:7. In yet another aspect, the invention provides an anti-ETBR antibody having a VL sequence of SEQ ID NO: 8 and a VH amino acid sequence of SEQ ID NO:9.
[0115] In another aspect, an anti-ETBR antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:7 or 9. In certain embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti- ETBR antibody comprising that sequence retains the ability to bind to ETBR. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO:7 or 9. In certain embodiments, substitutions, insertions, or deletions occur in regions outside the CDRs (i.e., in the FRs). [0116] In another aspect, an anti-ETBR antibody is provided, wherein the antibody comprises a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100%) sequence identity to the amino acid sequence of SEQ ID NO: 8. In certain embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-ETBR antibody comprising that sequence retains the ability to bind to ETBR. In certain embodiments, a total of 1 to 10 amino acids have been substituted, inserted and/or deleted in SEQ ID NO: 8. In certain embodiments, the substitutions, insertions, or deletions occur in regions outside the HVRs (i.e., in the FRs).
[0117] In another aspect, an anti-ETBR antibody is provided, wherein the antibody comprises a VH as in any of the embodiments provided above, and a VL as in any of the embodiments provided above. In one embodiment, the antibody comprises the VH and VL sequences in SEQ ID NO: 7 or 9 and SEQ ID NO:8, respectively, including post-translational modifications of those sequences.
[0118] In a further aspect, the invention provides an antibody that binds to the same epitope as an anti- ETBR antibody provided herein. For example, in certain embodiments, an antibody is provided that binds to the same epitope as an anti-ETBR antibody comprising a VH sequence of SEQ ID NO: 7 or 9 and a VL sequence of SEQ ID NO: 8. In certain embodiments, an anti-ETBR antibody is provided that binds to an epitope within an N-terminal extracellular domain #1 fragment of ETBR consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
[0119] In a further aspect of the invention, an anti-ETBR antibody according to any of the above embodiments is a monoclonal antibody, including a chimeric, humanized or human antibody. In one embodiment, an anti-ETBR antibody is an antibody fragment, e.g., a Fv, Fab, Fab', scFv, diabody, or F(ab')2 fragment. In another embodiment, the antibody is a full length antibody, e.g., an intact IgGl antibody or other antibody class or isotype as defined herein.
[0120] In a further aspect, an anti-ETBR antibody according to any of the above embodiments may incorporate any of the features, singly or in combination, as described in Sections 1-7 below:
[0121] Antibody Affinity
[0122] In certain embodiments, an antibody provided herein has a dissociation constant (Kd) of < 1 μΜ, < 100 nM, < 10 nM, < 1 nM, < 0.1 nM, < 0.01 nM, or < 0.001 nM (e.g. 10~8 M or less, e.g. from 10~8 M to 10"13 M, e.g., from 10"9 M to 10"13 M).
[0123] In one embodiment, Kd is measured by a radiolabeled antigen binding assay (RIA) performed with the Fab version of an antibody of interest and its antigen as described by the following assay. Solution binding affinity of Fabs for antigen is measured by equilibrating Fab with a minimal concentration of (125I) -labeled antigen in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate (see, e.g., Chen et al., J. Mol. Biol. 293:865-881(1999)). To establish conditions for the assay, MICROTITER® multi-well plates (Thermo Scientific) are coated overnight with 5 μg/ml of a capturing anti-Fab antibody (Cappel Labs) in 50 mM sodium carbonate (pH 9.6), and subsequently blocked with 2%> (w/v) bovine serum albumin in PBS for two to five hours at room temperature (approximately 23°C). In a non-adsorbent plate (Nunc #269620), 100 pM or 26 pM [125I]-anti gen are mixed with serial dilutions of a Fab of interest (e.g., consistent with assessment of the anti-VEGF antibody, Fab-12, in Presta et al., Cancer Res. 57:4593-4599 (1997)). The Fab of interest is then incubated overnight; however, the incubation may continue for a longer period (e.g., about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation at room temperature (e.g., for one hour). The solution is then removed and the plate washed eight times with 0.1% polysorbate 20 (TWEEN-20®) in PBS. When the plates have dried, 150 μΐ/well of scintillant (MICROSCINT-20™; Packard) is added, and the plates are counted on a TOPCOUNT™ gamma counter (Packard) for ten minutes. Concentrations of each Fab that give less than or equal to 20% of maximal binding are chosen for use in competitive binding assays.
[0124] According to another embodiment, Kd is measured using surface plasmon resonance assays using a BIACORE®-2000 or a BIACORE ®-3000 (BIAcore, Inc., Piscataway, NJ) at 25°C with immobilized antigen CM5 chips at -10 response units (RU). Briefly, carboxymethylated dextran biosensor chips (CM5, BIACORE, Inc.) are activated with N-ethyl-N'- (3-dimethylaminopropyl)- carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Antigen is diluted with 10 mM sodium acetate, pH 4.8, to 5 μg/ml (-0.2 μΜ) before injection at a flow rate of 5 μΐ/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 tiM) are injected in PBS with 0.05% polysorbate 20 (TWEEN-20™) surfactant (PBST) at 25°C at a flow rate of approximately 25 μΐ/min. Association rates (kon) and dissociation rates (k0ff) are calculated using a simple one-to-one
Langmuir binding model (BIACORE ® Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (Kd) is calculated as the ratio koff kon See, e.g., Chen et al., J. Mol. Biol. 293:865-881 (1999). If the on-rate exceeds 106 M" 1 s"
1 by the surface plasmon resonance assay above, then the on-rate can be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity
(excitation = 295 nm; emission = 340 nm, 16 nm band-pass) at 25°C of a 20 nM anti-antigen antibody (Fab form) in PBS, pH 7.2, in the presence of increasing concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped spectrophometer (Aviv Instruments) or a 8000-series SLM- AMINCO™ spectrophotometer (ThermoSpectronic) with a stirred cuvette.
[0125] Antibody Fragments
[0126] In certain embodiments, an antibody provided herein is an antibody fragment. Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab')2, Fv, and scFv fragments, and other fragments described below. For a review of certain antibody fragments, see Hudson et al. Nat. Med. 9: 129-134 (2003). For a review of scFv fragments, see, e.g., Pluckthun, in The Pharmacology of
Monoclonal Antibodies , vol. 113, Rosenburg and Moore eds., (Springer-Verlag, New York), pp. 269-315 (1994); see also WO 93/16185; and U.S. Patent Nos. 5,571,894 and 5,587,458. For discussion of Fab and F(ab')2 fragments comprising salvage receptor binding epitope residues and having increased in vivo half-life, see U.S. Patent No. 5,869,046.
[0127] Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161 ; Hudson et al., Nat. Med. 9: 129-134 (2003); and Hollinger et al., Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al., Nat. Med. 9: 129-134 (2003).
[0128] Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, MA; see, e.g., U.S. Patent No. 6,248,516 Bl).
[0129] Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g. E. coli or phage), as described herein.
[0130] Chimeric and Humanized Antibodies
[0131] In certain embodiments, an antibody provided herein is a chimeric antibody. Certain chimeric antibodies are described, e.g., in U.S. Patent No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, 81 :6851-6855 (1984)). In one example, a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region. In a further example, a chimeric antibody is a "class switched" antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
[0132] In certain embodiments, a chimeric antibody is a humanized antibody. Typically, a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody. Generally, a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences. A humanized antibody optionally will also comprise at least a portion of a human constant region. In some embodiments, some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
[0133] Humanized antibodies and methods of making them are reviewed, e.g., in Almagro and
Fransson, Front. Biosci. 13: 1619-1633 (2008), and are further described, e.g., in Riechmann et al., Nature 332:323-329 (1988); Queen et al., Proc. Natl Acad. Sci. USA 86: 10029-10033 (1989); US Patent Nos. 5, 821,337, 7,527,791, 6,982,321, and 7,087,409; Kashmiri et al, Methods 36:25-34 (2005) (describing SDR (a-CDR) grafting); Padlan, Mol. Immunol. 28:489-498 (1991) (describing
"resurfacing"); Dall'Acqua et al., Methods 36:43-60 (2005) (describing "FR shuffling"); and Osbourn et al., Methods 36:61-68 (2005) and Klimka et al., Br. J. Cancer, 83:252-260 (2000) (describing the "guided selection" approach to FR shuffling). [0134] Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the "best-fit" method (see, e.g., Sims et al. J. Immunol. 151 :2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol., 151 :2623 (1993)); human mature (somatically mutated) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, Front. Biosci. 13: 1619-1633 (2008)); and framework regions derived from screening FR libraries (see, e.g., Baca et al., J Biol. Chem. 272: 10678-10684 (1997) and Rosok et al., J. Biol. Chem. 271 :22611-22618 (1996)).
[0135] Human Antibodies
[0136] In certain embodiments, an antibody provided herein is a human antibody. Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin. Pharmacol. 5: 368-74 (2001) and Lonberg, Curr. Opin.
Immunol. 20:450-459 (2008).
[0137] Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human
immunoglobulin loci, which replace the endogenous immunoglobulin loci, or which are present extrachromosomally or integrated randomly into the animal's chromosomes. In such transgenic mice, the endogenous immunoglobulin loci have generally been inactivated. For review of methods for obtaining human antibodies from transgenic animals, see Lonberg, Nat. Biotech. 23: 1117-1125 (2005). See also, e.g., U.S. Patent Nos. 6,075,181 and 6,150,584 describing XENOMOUSE™ technology; U.S. Patent No. 5,770,429 describing HuMAB® technology; U.S. Patent No. 7,041,870 describing K-M MOUSE® technology, and U.S. Patent Application Publication No. US 2007/0061900, describing VELOCIMOUSE® technology). Human variable regions from intact antibodies generated by such animals may be further modified, e.g., by combining with a different human constant region.
[0138] Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse- human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol, 133: 3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications , pp. 51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al., J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-cell hybridoma technology are also described in Li et al.. Proc. Natl Acad. Sci. USA, 103:3557-3562 (2.006). Additional methods include those described, for example, in U.S. Patent No. 7,189,826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas). Human hybridoma technology (Trioma technology) is also described in Vollmers and Brandlein, Histology and Histopathology, 20(3):927-937 (2005) and Vollmers and Brandlein, Methods and Findings in Experimental and Clinical Pharmacology, 27(3): 185- 91 (2005). [0139] Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
[0140] Library-Derived Antibodies
[0141] Antibodies of the invention may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in Hoogenboom et al. m Methods in Molecular Biology 178: 1-37 (O'Brien et al., ed., Human Press, Totowa, NJ, 2001) and further described, e.g., in the McCafferty et al., Nature 348:552-554; Clackson et al., Nature 352: 624-628 (1991); Marks et al., J. Mol. Biol. 222: 581-597 (1992); Marks and Bradbury, m Methods in Molecular Biology 248: 161-175 (Lo, ed., Human Press, Totowa, NJ, 2003); Sidhu et al., J. Mol. Biol. 338(2): 299-310 (2004); Lee et al., J. Mol. Biol. 340(5): 1073-1093 (2004); Fellouse, Proc. Natl. Acad. Sci. USA 101(34): 12467-12472 (2004); and Lee et al., J. Immunol. Methods 284(1-2): 119-132(2004).
[0142] In certain phage display methods, repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al., Ann. Rev. Immunol, 12: 433-455 (1994). Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments. Libraries from immunized sources provide high-affinity antibodies to the immunogen without the requirement of constructing hybridomas. Alternatively, the naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al., EMBO J, 12: 725-734 (1993).
Finally, naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol, 227: 381-388 (1992). Patent publications describing human antibody phage libraries include, for example: US Patent No. 5,750,373, and US Patent Publication Nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360.
[0143] Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
[0144] Multispecific Antibodies
[0145] In certain embodiments, an antibody provided herein is a multispecific antibody, e.g. a bispecific antibody. Multispecific antibodies are monoclonal antibodies that have binding specificities for at least two different sites. In certain embodiments, one of the binding specificities is for ETBR and the other is for any other antigen. In certain embodiments, bispecific antibodies may bind to two different epitopes of ETBR. Bispecific antibodies may also be used to localize cytotoxic agents to cells which express ETBR. Bispecific antibodies can be prepared as full length antibodies or antibody fragments. [0146] Techniques for making multispecific antibodies include, but are not limited to, recombinant co- expression of two immunoglobulin heavy chain-light chain pairs having different specificities (see Milstein and Cuello, Nature 305: 537 (1983)), WO 93/08829, and Traunecker et al., EMBO J. 10: 3655 (1991)), and "knob-in-hole" engineering (see, e.g., U.S. Patent No. 5,731,168). Multi-specific antibodies may also be made by engineering electrostatic steering effects for making antibody Fc-heterodimeric molecules (WO 2009/089004A1); cross-linking two or more antibodies or fragments (see, e.g., US Patent No. 4,676,980, and Brennan et al., Science, 229: 81 (1985)); using leucine zippers to produce bi-specific antibodies (see, e.g., Kostelny et al., J. Immunol, 148(5): 1547-1553 (1992)); using "diabody" technology for making bispecific antibody fragments (see, e.g., Hollinger et al., Proc. Natl Acad. Sci. USA, 90:6444- 6448 (1993)); and using single-chain Fv (sFv) dimers (see,e.g. Gruber et al., J. Immunol, 152:5368 (1994)); and preparing trispecific antibodies as described, e.g., in Tutt et al. J. Immunol 147: 60 (1991).
[0147] Engineered antibodies with three or more functional antigen binding sites, including "Octopus antibodies," are also included herein (see, e.g. US 2006/0025576A1).
[0148] The antibody or fragment herein also includes a "Dual Acting FAb" or "DAF" comprising an antigen binding site that binds to ETBR as well as another, different antigen (see, US 2008/0069820, for example).
[0149] Antibody Variants
[0150] In certain embodiments, amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody. Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
[0151] Substitution, Insertion, and Deletion Variants
[0152] In certain embodiments, antibody variants having one or more amino acid substitutions are provided. Sites of interest for substitutional mutagenesis include the HVRs and FRs. Conservative substitutions are shown in Table 1 under the heading of "conservative substitutions." More substantial changes are provided in Table 1 under the heading of "exemplary substitutions," and as further described below in reference to amino acid side chain classes. Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, or improved ADCC or CDC. TABLE 1
Figure imgf000033_0001
[0153] Amino acids may be grouped according to common side -chain properties:hydrophobic:
Norleucine, Met, Ala, Val, Leu, He;
neutral hydrophilic: Cys, Ser, Thr, Asn, Gin;
acidic: Asp, Glu;
basic: His, Lys, Arg;
residues that influence chain orientation: Gly, Pro;
aromatic: Trp, Tyr, Phe.
[0154] Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
[0155] One type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody). Generally, the resulting variant(s) selected for further study will have modifications (e.g., improvements) in certain biological properties (e.g., increased affinity, reduced immunogenicity) relative to the parent antibody and/or will have substantially retained certain biological properties of the parent antibody. An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display- based affinity maturation techniques such as those described herein. Briefly, one or more HVR residues are mutated and the variant antibodies displayed on phage and screened for a particular biological activity (e.g. binding affinity).
[0156] Alterations (e.g., substitutions) may be made in HVRs, e.g., to improve antibody affinity. Such alterations may be made in HVR "hotspots," i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury, Methods Mol. Biol.
207: 179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity. Affinity maturation by constructing and reselecting from secondary libraries has been described, e.g., in Hoogenboom et al. m Methods in Molecular Biology 178: 1-37 (O'Brien et al., ed., Human Press, Totowa, NJ, (2001).) In some embodiments of affinity maturation, diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis). A secondary library is then created. The library is then screened to identify any antibody variants with the desired affinity. Another method to introduce diversity involves HVR-directed approaches, in which several HVR residues (e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted.
[0157] In certain embodiments, substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind antigen. For example, conservative alterations (e.g., conservative substitutions as provided herein) that do not substantially reduce binding affinity may be made in HVRs. Such alterations may be outside of HVR "hotspots" or SDRs. In certain embodiments of the variant VH and VL sequences provided above, each HVR either is unaltered, or contains no more than one, two or three amino acid substitutions.
[0158] A useful method for identification of residues or regions of an antibody that may be targeted for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham and Wells (1989) Science, 244: 1081-1085. In this method, a residue or group of target residues (e.g., charged residues such as arg, asp, his, lys, and glu) are identified and replaced by a neutral or negatively charged amino acid (e.g., alanine or polyalanine) to determine whether the interaction of the antibody with antigen is affected. Further substitutions may be introduced at the amino acid locations demonstrating functional sensitivity to the initial substitutions. Alternatively, or additionally, a crystal structure of an antigen- antibody complex to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution. Variants may be screened to determine whether they contain the desired properties.
[0159] Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an antibody with an N-terminal methionyl residue. Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g. for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
[0160] Glycosylation variants
[0161] In certain embodiments, an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
[0162] Where the antibody comprises an Fc region, the carbohydrate attached thereto may be altered. Native antibodies produced by mammalian cells typically comprise a branched, biantennary
oligosaccharide that is generally attached by an N-linkage to Asn297 of the CH2 domain of the Fc region. See, e.g., Wright et al. TIBTECH 15:26-32 (1997). The oligosaccharide may include various carbohydrates, e.g., mannose, N-acetyl glucosamine (GlcNAc), galactose, and sialic acid, as well as a fucose attached to a GlcNAc in the "stem" of the biantennary oligosaccharide structure. In some embodiments, modifications of the oligosaccharide in an antibody of the invention may be made in order to create antibody variants with certain improved properties.
[0163] In one embodiment, antibody variants are provided having a carbohydrate structure that lacks fucose attached (directly or indirectly) to an Fc region. For example, the amount of fucose in such antibody may be from 1% to 80%, from 1% to 65%, from 5% to 65% or from 20% to 40%. The amount of fucose is determined by calculating the average amount of fucose within the sugar chain at Asn297, relative to the sum of all glycostructures attached to Asn 297 (e. g. complex, hybrid and high mannose structures) as measured by MALDI-TOF mass spectrometry, as described in WO 2008/077546, for example. Asn297 refers to the asparagine residue located at about position 297 in the Fc region (Eu numbering of Fc region residues); however, Asn297 may also be located about ± 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300, due to minor sequence variations in antibodies. Such fucosylation variants may have improved ADCC function. See, e.g., US Patent Publication Nos. US 2003/0157108 (Presta, L.); US 2004/0093621 (Kyowa Hakko Kogyo Co., Ltd). Examples of publications related to "defucosylated" or "fucose-deficient" antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002/0164328; US
2004/0093621; US 2004/0132140; US 2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119; WO 2003/084570; WO 2005/035586; WO 2005/035778; WO2005/053742;
WO2002/031140; Okazaki et al. J. Mol. Biol. 336: 1239-1249 (2004); Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004). Examples of cell lines capable of producing defucosylated antibodies include Led 3 CHO cells deficient in protein fucosylation (Ripka et al. Arch. Biochem. Biophys. 249:533-545 (1986); US Pat Appl No US 2003/0157108 Al, Presta, L; and WO 2004/056312 Al, Adams et al, especially at Example 11), and knockout cell lines, such as alpha-l,6-fucosyltransferase gene, FUT8, knockout CHO cells (see, e.g., Yamane-Ohnuki et al. Biotech. Bioeng. 87: 614 (2004); Kanda, Y. et al., Biotechnol. Bioeng., 94(4):680-688 (2006); and WO2003/085107).
[0164] Antibodies variants are further provided with bisected oligosaccharides, e.g., in which a biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody variants may have reduced fucosylation and/or improved ADCC function. Examples of such antibody variants are described, e.g., in WO 2003/011878 (Jean-Mairet et al.); US Patent No. 6,602,684 (Umana et al.); and US 2005/0123546 (Umana et al.). Antibody variants with at least one galactose residue in the oligosaccharide attached to the Fc region are also provided. Such antibody variants may have improved CDC function. Such antibody variants are described, e.g., in WO 1997/30087 (Patel et al.); WO 1998/58964 (Raju, S.); and WO 1999/22764 (Raju, S.).
[0165] Fc region variants
[0166] In certain embodiments, one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein, thereby generating an Fc region variant. The Fc region variant may comprise a human Fc region sequence {e.g., a human IgGl, IgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification {e.g. a substitution) at one or more amino acid positions.
[0167] In certain embodiments, the invention contemplates an antibody variant that possesses some but not all effector functions, which make it a desirable candidate for applications in which the half life of the antibody in vivo is important yet certain effector functions (such as complement and ADCC) are unnecessary or deleterious. In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities. For example, Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability. The primary cells for mediating ADCC, NK cells, express FcyRIII only, whereas monocytes express FcyRI, FcyRII and FcyRIII. FcR expression on hematopoietic cells is summarized in Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol. 9:457-492 (1991). Non- limiting examples of in vitro assays to assess ADCC activity of a molecule of interest is described in U.S. Patent No. 5,500,362 (see, e.g. Hellstrom, I. et al. Proc. Natl Acad. Sci. USA 83:7059-7063 (1986)) and Hellstrom, I et al., Proc. Natl Acad. Sci. USA 82: 1499-1502 (1985); 5,821,337 (see Bruggemann, M. et al., J. Exp. Med. 166:1351-1361 (1987)). Alternatively, non-radioactive assays methods may be employed (see, for example, ACTI™ non-radioactive cytotoxicity assay for flow cytometry
(CellTechnology, Inc. Mountain View, CA; and CytoTox 96® non-radioactive cytotoxicity assay (Promega, Madison, WI). Useful effector cells for such assays include peripheral blood mononuclear cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, e.g., in a animal model such as that disclosed in Clynes et al. Proc. Natl Acad. Sci. USA 95:652-656 (1998). Clq binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity. See, e.g., Clq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402. To assess complement activation, a CDC assay may be performed (see, for example, Gazzano-Santoro et al., J. Immunol. Methods 202: 163 (1996); Cragg, M.S. et al., Blood 101 : 1045-1052 (2003); and Cragg, M.S. and M.J. Glennie, Blood 103:2738- 2743 (2004)). FcRn binding and in vivo clearance/half life determinations can also be performed using methods known in the art (see, e.g., Petkova, S.B. et al., Int'l. Immunol. 18(12): 1759-1769 (2006)).
[0168] Antibodies with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Patent No. 6,737,056). Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (US Patent No. 7,332,581).
[0169] Certain antibody variants with improved or diminished binding to FcRs are described. (See, e.g., U.S. Patent No. 6,737,056; WO 2004/056312, and Shields et al., J. Biol. Chem. 9(2): 6591 -6604 (2001).)
[0170] In certain embodiments, an antibody variant comprises an Fc region with one or more amino acid substitutions which improve ADCC, e.g., substitutions at positions 298, 333, and/or 334 of the Fc region (EU numbering of residues).
[0171] In some embodiments, alterations are made in the Fc region that result in altered {i.e., either improved or diminished) Clq binding and/or Complement Dependent Cytotoxicity (CDC), e.g., as described in US Patent No. 6,194,551, WO 99/51642, and Idusogie et al. J. Immunol. 164: 4178-4184 (2000).
[0172] Antibodies with increased half lives and improved binding to the neonatal Fc receptor (FcRn), which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al., J. Immunol. 117:587 (1976) and Kim et al., J. Immunol. 24:249 (1994)), are described in US2005/0014934A1 (Hinton et al.). Those antibodies comprise an Fc region with one or more substitutions therein which improve binding of the Fc region to FcRn. Such Fc variants include those with substitutions at one or more of Fc region residues: 238, 256, 265, 272, 286, 303, 305, 307, 311, 312, 317, 340, 356, 360, 362, 376, 378, 380, 382, 413, 424 or 434, e.g., substitution of Fc region residue 434 (US Patent No. 7,371,826).
[0173] See also Duncan & Winter, Nature 322:738-40 (1988); U.S. Patent No. 5,648,260; U.S. Patent No. 5,624,821 ; and WO 94/29351 concerning other examples of Fc region variants.
[0174] Cysteine engineered antibody variants
[0175] In certain embodiments, it may be desirable to create cysteine engineered antibodies, e.g., "thioMAbs," in which one or more residues of an antibody are substituted with cysteine residues. In particular embodiments, the substituted residues occur at accessible sites of the antibody. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker- drug moieties, to create an immunoconjugate, as described further herein. In certain embodiments, any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy chain Fc region. Cysteine engineered antibodies may be generated as described, e.g., in U.S. Patent No.
7,521,541. [0176] Antibody Derivatives
[0177] In certain embodiments, an antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available. The moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers. Non- limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n- vinyl
pyrrolidone)poly ethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, polyoxyethylated polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof. Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water. The polymer may be of any molecular weight, and may be branched or unbranched. The number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
[0178] In another embodiment, conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided. In one embodiment, the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)). The radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
[0179] Recombinant Methods and Compositions
[0180] Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Patent No. 4,816,567. In one embodiment, isolated nucleic acid encoding an anti-ETBR antibody described herein is provided. Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody (e.g., the light and/or heavy chains of the antibody). In a further embodiment, one or more vectors (e.g., expression vectors) comprising such nucleic acid are provided. In a further embodiment, a host cell comprising such nucleic acid is provided. In one such embodiment, a host cell comprises (e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody. In one embodiment, the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell (e.g., Y0, NS0, Sp20 cell). In one embodiment, a method of making an anti-ETBR antibody is provided, wherein the method comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
[0181] For recombinant production of an anti-ETBR antibody, nucleic acid encoding an antibody, e.g., as described above, is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell. Such nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
[0182] Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein. For example, antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed. For expression of antibody fragments and polypeptides in bacteria, see, e.g., U.S. Patent Nos. 5,648,237, 5,789,199, and 5,840,523. (See also Charlton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ, 2003), pp. 245-254, describing expression of antibody fragments in E. coli.) After expression, the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
[0183] In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized," resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22: 1409-1414 (2004), and Li et al., Nat. Biotech. 24:210-215 (2006).
[0184] Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
[0185] Plant cell cultures can also be utilized as hosts. See, e.g., US Patent Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIES™ technology for producing antibodies in transgenic plants).
[0186] Vertebrate cells may also be used as hosts. For example, mammalian cell lines that are adapted to grow in suspension may be useful. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, Biol. Reprod. 23:243-251 (1980)); monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al., Annals N. Y. Acad. Sci. 383:44-68 (1982); MRC 5 cells; and FS4 cells. Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR" CHO cells (Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); and myeloma cell lines such as Y0, NS0 and Sp2/0. For a review of certain mammalian host cell lines suitable for antibody production, see, e.g., Yazaki and Wu, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, NJ), pp. 255-268 (2003).
[0187] Assays
[0188] Anti-ETBR antibodies provided herein may be identified, screened for, or characterized for their physical/chemical properties and/or biological activities by various assays known in the art.
[0189] Binding assays and other assays
[0190] In one aspect, an antibody of the invention is tested for its antigen binding activity, e.g., by known methods such as ELISA, Western blot, etc.
[0191] In another aspect, competition assays may be used to identify an antibody that competes with, for example, Hu5E9v. l or Hu5E9v.2 for binding to ETBR. In certain embodiments, such a competing antibody binds to the same epitope (e.g., a linear or a conformational epitope) that is bound by Hu5E9v.l or Hu5E9v.2. Detailed exemplary methods for mapping an epitope to which an antibody binds are provided in Morris (1996) "Epitope Mapping Protocols," in Methods in Molecular Biology vol. 66 (Humana Press, Totowa, NJ). In one aspect of the invention, anti-ETBR antibodies described herein specifically bind an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
[0192] In an exemplary competition assay, immobilized ETBR is incubated in a solution comprising a first labeled antibody that binds to ETBR (e.g., Hu5E9v.l or Hu5E9v.2) and a second unlabeled antibody that is being tested for its ability to compete with the first antibody for binding to ETBR. The second antibody may be present in a hybridoma supernatant. As a control, immobilized ETBR is incubated in a solution comprising the first labeled antibody but not the second unlabeled antibody. After incubation under conditions permissive for binding of the first antibody to ETBR, excess unbound antibody is removed, and the amount of label associated with immobilized ETBR is measured. If the amount of label associated with immobilized ETBR is substantially reduced in the test sample relative to the control sample, then that indicates that the second antibody is competing with the first antibody for binding to ETBR. See Harlow and Lane (1988) Antibodies: A Laboratory Manual ch.14 (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY).
[0193] Activity assays
[0194] In one aspect, assays are provided for identifying whether anti-ETBR antibodies and/or BRAFi compounds have biological activity. Biological activity may include those described in the Examples, e.g., in vitro melanoma cell survival assays or in vivo xenograft models in which melanoma cell lines are transplanted into nude mice and tumor growth inhibition (TGI) is assessed.
[0195] Immunoconjugates
[0196] The invention also provides immunoconjugates comprising an anti-ETBR antibody herein conjugated to one or more cytotoxic agents, such as chemotherapeutic agents or drugs, growth inhibitory agents, toxins (e.g., protein toxins, enzymatically active toxins of bacterial, fungal, plant, or animal origin, or fragments thereof), or radioactive isotopes.
[0197] The invention also provides immunoconjugates (interchangeably referred to as "antibody-drug conjugates," or "ADCs") comprising an antibody conjugated to one or more cytotoxic agents, such as a chemotherapeutic agent, a drug, a growth inhibitory agent, a toxin (e.g., a protein toxin, an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
[0198] Immunoconjugates have been used for the local delivery of cytotoxic agents, i.e., drugs that kill or inhibit the growth or proliferation of cells, in the treatment of cancer (Xie et al (2006) Expert. Opin. Biol. Ther. 6(3):281-291 ; Kovtun et al (2006) Cancer Res. 66(6):3214-3121 ; Law et al (2006) Cancer Res. 66(4):2328-2337; Lambert, J. (2005) Curr. Opinion in Pharmacology 5:543-549; Wu et al (2005) Nature Biotechnology 23(9): 1137-1146; Payne, G. (2003) i 3:207-212; Syrigos and Epenetos (1999) Anticancer Research 19:605-614; Niculescu-Duvaz and Springer (1997) Adv. Drug Deliv. Rev. 26: 151- 172; U.S. Pat. No. 4,975,278). Immunoconjugates allow for the targeted delivery of a drug moiety to a tumor, and intracellular accumulation therein, where systemic administration of unconjugated drugs may result in unacceptable levels of toxicity to normal cells as well as the tumor cells sought to be eliminated (Baldwin et al., Lancet (Mar. 15, 1986) pp. 603-05; Thorpe (1985) "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review," in Monoclonal Antibodies '84: Biological And Clinical
Applications (A. Pinchera et al., eds) pp. 475-506. Both polyclonal antibodies and monoclonal antibodies have been reported as useful in these strategies (Rowland et al., (1986) Cancer Immunol. Immunother. 21 : 183-87). Drugs used in these methods include daunomycin, doxorubicin, methotrexate, and vindesine (Rowland et al., (1986) supra). Toxins used in antibody-toxin conjugates include bacterial toxins such as diphtheria toxin, plant toxins such as ricin, small molecule toxins such as geldanamycin (Mandler et al (2000) J. Nat. Cancer Inst. 92(19): 1573-1581 ; Mandler et al (2000) Bioorganic & Med. Chem. Letters 10: 1025-1028; Mandler et al (2002) Bioconjugate Chem. 13:786-791), maytansinoids (EP 1391213; Liu et al., (1996) Proc. Natl. Acad. Sci. USA 93:8618-8623), and calicheamicin (Lode et al (1998) Cancer Res. 58:2928; Hinman et al (1993) Cancer Res. 53:3336-3342). Efforts to design and refine ADC have focused on the selectivity of monoclonal antibodies (mAbs) as well as drug mechanism of action, drug-linking, drug/antibody ratio (loading), and drug-releasing properties (McDonagh (2006) Protein Eng. Design & Sel.; Doronina et al (2006) Bioconj. Chem. 17: 114-124; Erickson et al (2006) Cancer Res. 66(8): l-8; Sanderson et al (2005) Clin. Cancer Res. 11 :843-852; Jeffrey et al (2005) J. Med. Chem. 48: 1344-1358; Hamblett et al (2004) Clin. Cancer Res. 10:7063-7070). The toxins may exert their cytotoxic effects by mechanisms including tubulin binding, DNA binding, or topoisomerase inhibition. Some cytotoxic drugs tend to be inactive or less active when conjugated to large antibodies or protein receptor ligands.
[0199] ZEVALIN® (ibritumomab tiuxetan, Biogen/Idec) is an antibody-radioisotope conjugate composed of a murine IgGl kappa monoclonal antibody directed against the CD20 antigen found on the surface of normal and malignant B lymphocytes and 11 lln or 90Y radioisotope bound by a thiourea linker-chelator (Wiseman et al (2000) Eur. Jour. Nucl. Med. 27(7):766-77; Wiseman et al (2002) Blood 99(12):4336-42; Witzig et al (2002) J. Clin. Oncol. 20(10):2453-63; Witzig et al (2002) J. Clin. Oncol. 20(15):3262-69). Although ZEVALIN has activity against B-cell non-Hodgkin's Lymphoma (NHL), administration results in severe and prolonged cytopenias in most patients. MYLOTARG™ (gemtuzumab ozogamicin, Wyeth Pharmaceuticals), an antibody-drug conjugate composed of a huCD33 antibody linked to calicheamicin, was approved in 2000 for the treatment of acute myeloid leukemia by injection (Drugs of the Future (2000) 25(7):686; US Patent Nos. 4970198; 5079233; 5585089; 5606040; 5693762; 5739116; 5767285; 5773001). Antibody-drug conjugates (ADCs) composed of the maytansinoid, DM1, linked to trastuzumab show potent anti-tumor activity in HER2-overexpressing trastuzumab-sensitive and -resistant tumor cell lines and xenograft models of human cancer.
Trastuzumab-MCC-DMl (T-DM1) is currently undergoing evaluation in phase II clinical trials in patients whose disease is refractory to HER2-directed therapies (Beeram et al (2007) "A phase I study of trastuzumab-MCC-DMl (T-DM1), a first-in-class HER2 antibody-drug conjugate (ADC), in patients (pts) with HER2+ metastatic breast cancer (BC)", American Society of Clinical Oncology 43rd:June 02 (Abs 1042; Krop et al, European Cancer Conference ECCO, Poster 2118, September 23-27, 2007, Barcelona;US 7097840; US 2005/0276812; US 2005/0166993). The auristatin peptides, auristatin E (AE) and monomethylauristatin (MMAE), synthetic analogs of dolastatin, were conjugated to chimeric monoclonal antibodies cBR96 (specific to Lewis Y on carcinomas) and cACIO (specific to CD30 on hematological malignancies) (Doronina et al (2003) Nature Biotechnol. 21(7):778-784) and are under therapeutic development.
[0200] In certain embodiments, an immunoconjugate comprises an antibody and a chemotherapeutic agent or other toxin. Chemotherapeutic agents useful in the generation of immunoconjugates are described herein (e.g., above). Enzymatically active toxins and fragments thereof can also be used and are described herein.
[0201] In certain embodiments, an immunoconjugate comprises an antibody and one or more small molecule drug moieties (toxins), including, but not limited to, small molecule drugs such as a calicheamicin, maytansinoid, dolastatin, auristatin, anthracycline, taxane, trichothecene, and CC1065, and the derivatives of these drugs that have cytotoxic activity. Examples of such immunoconjugates are discussed in further detail below.
[0202] Exemplary Immunoconjugates
[0203] An immunoconjugate (or "antibody-drug conjugate" ("ADC")) of the invention may be of Formula I, below, wherein an antibody is conjugated (i.e., covalently attached) to one or more drug moieties (D) through an optional linker (L).
Ab-(L-D)p I
[0204] Accordingly, the antibody may be conjugated to the drug either directly or via a linker. In Formula I, p is the average number of drug moieties per antibody, which can range, e.g., from about 1 to about 20 drug moieties per antibody, and in certain embodiments, from 1 to about 8 drug moieties per antibody.
[0205] Exemplary Linkers
[0206] A linker may comprise one or more linker components. Exemplary linker components include 6- maleimidocaproyl ("MC"), maleimidopropanoyl ("MP"), valine-citrulline ("val-cit" or "vc"), alanine- phenylalanine ("ala-phe"), p-aminobenzyloxycarbonyl (a "PAB"), N-Succinimidyl 4-(2-pyridylthio) pentanoate ("SPP"), N-succinimidyl 4-(N-maleimidomethyl) cyclohexane-1 carboxylate ("SMCC"), and N-Succinimidyl (4-iodo-acetyl) aminobenzoate ("SIAB"). Various linker components are known in the art, some of which are described below.
[0207] A linker may be a "cleavable linker," facilitating release of a drug in the cell. For example, an acid-labile linker (e.g., hydrazone), protease-sensitive (e.g., peptidase-sensitive) linker, photolabile linker, dimethyl linker or disulfide -containing linker (Chari et al., Cancer Research 52: 127-131 (1992); U.S. Patent No. 5,208,020) may be used.
[0208] In certain embodiments, a linker is as shown in the following Formula II:
-Aa— Ww— Yy- II
[0209] wherein A is a stretcher unit, and a is an integer from 0 to 1 ; W is an amino acid unit, and w is an integer from 0 to 12; Y is a spacer unit, and y is 0, 1, or 2; and Ab, D, and p are defined as above for Formula I. Exemplary embodiments of such linkers are described in US 2005-0238649 Al, which is expressly incorporated herein by reference.
[0210] In some embodiments, a linker component may comprise a "stretcher unit" that links an antibody to another linker component or to a drug moiety. Exemplary stretcher units are shown below (wherein the wavy line indicates sites of covalent attachment to an antibody):
Figure imgf000043_0001
Figure imgf000043_0002
Figure imgf000043_0003
MPEG O
[0211] In some embodiments, a linker component may comprise an amino acid unit. In one such embodiment, the amino acid unit allows for cleavage of the linker by a protease, thereby facilitating release of the drug from the immunoconjugate upon exposure to intracellular proteases, such as lysosomal enzymes. See, e.g., Doronina et al. (2003) Nat. Biotechnol. 21 :778-784. Exemplary amino acid units include, but are not limited to, a dipeptide, a tripeptide, a tetrapeptide, and a pentapeptide. Exemplary dipeptides include: valine-citrulline (vc or val-cit), alanine-phenylalanine (af or ala-phe); phenylalanine-lysine (fk or phe-lys); or N-methyl-valine-citrulline (Me-val-cit). Exemplary tripeptides include: glycine -valine-citrulline (gly-val-cit) and glycine-glycine-glycine (gly-gly-gly). An amino acid unit may comprise amino acid residues that occur naturally, as well as minor amino acids and non- naturally occurring amino acid analogs, such as citrulline. Amino acid units can be designed and optimized in their selectivity for enzymatic cleavage by a particular enzyme, for example, a tumor- associated protease, cathepsin B, C and D, or a plasmin protease.
[0212] In some embodiments, a linker component may comprise a "spacer" unit that links the antibody to a drug moiety, either directly or by way of a stretcher unit and/or an amino acid unit. A spacer unit may be "self-immolative" or a "non-self-immolative." A "non-self-immolative" spacer unit is one in which part or all of the spacer unit remains bound to the drug moiety upon enzymatic (e.g., proteolytic) cleavage of the ADC. Examples of non-self-immolative spacer units include, but are not limited to, a glycine spacer unit and a glycine-glycine spacer unit. Other combinations of peptidic spacers susceptible to sequence-specific enzymatic cleavage are also contemplated. For example, enzymatic cleavage of an ADC containing a glycine -glycine spacer unit by a tumor-cell associated protease would result in release of a glycine-glycine-drug moiety from the remainder of the ADC. In one such embodiment, the glycine- glycine-drug moiety is then subjected to a separate hydrolysis step in the tumor cell, thus cleaving the glycine-glycine spacer unit from the drug moiety.
[0213] A "self-immolative" spacer unit allows for release of the drug moiety without a separate hydrolysis step. In certain embodiments, a spacer unit of a linker comprises a p-aminobenzyl unit. In one such embodiment, a p-aminobenzyl alcohol is attached to an amino acid unit via an amide bond, and a carbamate, methylcarbamate, or carbonate is made between the benzyl alcohol and a cytotoxic agent. See, e.g., Hamann et al. (2005) Expert Opin. Ther. Patents (2005) 15: 1087-1 103. In one embodiment, the spacer unit is p-aminobenzyloxycarbonyl (PAB). In certain embodiments, the phenylene portion of a p-amino benzyl unit is substituted with Qm, wherein Q is -Ci-Cg alkyl, -0-(Ci-Cg alkyl), -halogen,- nitro or -cyano; and m is an integer ranging from 0-4. Examples of self-immolative spacer units further include, but are not limited to, aromatic compounds that are electronically similar to p-aminobenzyl alcohol {see, e.g., US 2005/0256030 Al), such as 2-aminoimidazol-5 -methanol derivatives (Hay et al. (1999) Bioorg. Med. Chem. Lett. 9:2237) and ortho- or para-aminobenzylacetals. Spacers can be used that undergo cyclization upon amide bond hydrolysis, such as substituted and unsubstituted 4- aminobutyric acid amides (Rodrigues et al., Chemistry Biology, 1995, 2, 223); appropriately substituted bicyclo[2.2.1] and bicyclo[2.2.2] ring systems (Storm, et al., J. Amer. Chem. Soc , 1972, 94, 5815); and 2-aminophenylpropionic acid amides (Amsberry, et al., J. Org. Chem., 1990, 55, 5867). Elimination of amine -containing drugs that are substituted at the a-position of glycine (Kingsbury, et al., J. Med. Chem. , 1984, 27, 1447) are also examples of self-immolative spacers useful in ADCs.
[0214] In one embodiment, a spacer unit is a branched bis(hydroxymethyl)styrene (BHMS) unit as depicted below, which can be used to incorporate and release multiple drugs.
Figure imgf000045_0001
enzymatic
cleavage
2 drugs
[0215] wherein Q is -Ci-Cg alkyl, -0-(Ci-Cg alkyl), -halogen, -nitro or -cyano; m is an integer ranging from 0-4; n is 0 or 1 ; and p ranges raging from 1 to about 20.
[0216] In another embodiment, linker L may be a dendritic type linker for covalent attachment of more than one drug moiety through a branching, multifunctional linker moiety to an antibody (Sun et al (2002) Bioorganic & Medicinal Chemistry Letters 12:2213-2215; Sun et al (2003) Bioorganic & Medicinal Chemistry 1 1 : 1761 -1768). Dendritic linkers can increase the molar ratio of drug to antibody, i.e. loading, which is related to the potency of the ADC. Thus, where a cysteine engineered antibody bears only one reactive cysteine thiol group, a multitude of drug moieties may be attached through a dendritic linker.
[0217] Exemplary linker components and combinations thereof are shown below in the context of ADCs of Formula II:
Figure imgf000045_0002
Figure imgf000046_0001
Figure imgf000046_0002
[0218] Linkers components, including stretcher, spacer, and amino acid units, may be synthesized by methods known in the art, such as those described in US 2005-0238649 Al .
[0219] Exemplary Drug Moieties
[0220] Maytansine and maytansinoids
[0221] In some embodiments, an immunoconjugate comprises an antibody conjugated to one or more maytansinoid molecules. Maytansinoids are mitototic inhibitors which act by inhibiting tubulin polymerization. Maytansine was first isolated from the east African shrub Maytenus serrata (U.S. Patent No. 3896111). Subsequently, it was discovered that certain microbes also produce maytansinoids, such as maytansinol and C-3 maytansinol esters (U.S. Patent No. 4,151,042). Synthetic maytansinol and derivatives and analogues thereof are disclosed, for example, in U.S. Patent Nos. 4,137,230; 4,248,870; 4,256,746; 4,260,608; 4,265,814; 4,294,757; 4,307,016; 4,308,268; 4,308,269; 4,309,428; 4,313,946; 4,315,929; 4,317,821; 4,322,348; 4,331,598; 4,361,650; 4,364,866; 4,424,219; 4,450,254; 4,362,663; and 4,371,533.
[0222] Maytansinoid drug moieties are attractive drug moieties in antibody-drug conjugates because they are: (i) relatively accessible to prepare by fermentation or chemical modification or derivatization of fermentation products, (ii) amenable to derivatization with functional groups suitable for conjugation through non-disulfide linkers to antibodies, (iii) stable in plasma, and (iv) effective against a variety of tumor cell lines.
[0223] Maytansine compounds suitable for use as maytansinoid drug moieties are well known in the art and can be isolated from natural sources according to known methods or produced using genetic engineering techniques (see Yu et al (2002) PNAS 99:7968-7973). Maytansinol and maytansinol analogues may also be prepared synthetically according to known methods.
[0224] Exemplary maytansinoid drug moieties include those having a modified aromatic ring, such as: C-19-dechloro (US Pat. No. 4256746) (prepared by lithium aluminum hydride reduction of ansamytocin P2); C-20-hydroxy (or C-20-demethyl) +/-C-19-dechloro (US Pat. Nos. 4361650 and 4307016) (prepared by demethylation using Streptomyces or Actinomyces or dechlorination using LAH); and C-20- demethoxy, C-20-acyloxy (-OCOR), +/-dechloro (U.S. Pat. No. 4,294,757) (prepared by acylation using acyl chlorides), and those having modifications at other positions.
[0225] Exemplary maytansinoid drug moieties also include those having modifications such as: C-9-SH (US Pat. No. 4424219) (prepared by the reaction of maytansinol with H2S or P2S5); C-14- alkoxymethyl(demethoxy/CH2 OR)(US 4331598); C-14-hydroxymethyl or acyloxymethyl (CH2OH or CH2OAc) (US Pat. No. 4450254) (prepared from Nocardia); C-15-hydroxy/acyloxy (US 4364866) (prepared by the conversion of maytansinol by Streptomyces); C-15-methoxy (US Pat. Nos. 4313946 and 4315929) (isolated from Trewia nudlflora); C-18-N-demethyl (US Pat. Nos. 4362663 and 4322348) (prepared by the demethylation of maytansinol by Streptomyces); and 4,5-deoxy (US 4371533) (prepared by the titanium trichloride/LAH reduction of maytansinol).
[0226] Many positions on maytansine compounds are known to be useful as the linkage position, depending upon the type of link. For example, for forming an ester linkage, the C-3 position having a hydroxyl group, the C-14 position modified with hydroxymethyl, the C-15 position modified with a hydroxyl group and the C-20 position having a hydroxyl group are all suitable.
[0227] M
Figure imgf000047_0001
[0228] where the wavy line indicates the covalent attachment of the sulfur atom of the maytansinoid drug moiety to a linker of an ADC. R may independently be H or a Ci-C6 alkyl. The alkylene chain attaching the amide group to the sulfur atom may be methanyl, ethanyl, or propyl, i.e., m is 1, 2, or 3 (US 633410; US 5208020; Chari et al (1992) Cancer Res. 52: 127-131 ; Liu et al (1996) Proc. Natl. Acad. Sci USA 93:8618-8623).
[0229] All stereoisomers of the maytansinoid drug moiety are contemplated for the compounds of the invention, i.e. any combination of R and S configurations at the chiral carbons of D (US 7276497; US 6913748; US 6441 163; US 633410 (RE39151); US 5208020; Widdison et al (2006) J. Med. Chem. 49:4392-4408, which are incorporated by reference in their entirety). In one embodiment, the maytansinoid drug moiety will have the following stereochemistry:
Figure imgf000048_0001
[0230] Exemplary embodiments of maytansinoid drug moieities include: DM1 ; DM3; and DM4, having the structures:
Figure imgf000048_0002
Figure imgf000049_0001
Figure imgf000049_0002
[0231] wherein the wavy line indicates the covalent attachment of the sulfur atom of the drug to a linker (L) of an antibody-drug conjugate. HERCEPTIN® (trastuzumab) linked by SMCC to DM1 has been reported (WO 2005/037992; US 2005/0276812; US 2005/016993).
[0232] Other exemplary maytansinoid antibody-drug conjugates have the following structures and abbreviations, (wherein Ab is antibody and p is 1 to about 8):
Figure imgf000050_0001
Ab-SMCC-DMl
[0233] Exemplary antibody-drug conjugates where DM1 is linked through a BMPEO linker to group of the antibody have the structure and abbreviation:
Figure imgf000050_0002
where Ab is antibody; n is 0, 1 , or 2; and p is 1 , 2, 3, or 4. [0234] Immunoconjugates containing maytansinoids, methods of making the same, and their therapeutic use are disclosed, for example, in U.S. Patent Nos. 5,208,020, 5,416,064, US 2005/0276812 Al, and European Patent EP 0 425 235 Bl, the disclosures of which are hereby expressly incorporated by reference. Liu et al. Proc. Natl. Acad. Sci. USA 93:8618-8623 (1996) describe immunoconjugates comprising a maytansinoid designated DM1 linked to the monoclonal antibody C242 directed against human colorectal cancer. The conjugate was found to be highly cytotoxic towards cultured colon cancer cells, and showed antitumor activity in an in vivo tumor growth assay. Chari et al. Cancer Research 52: 127-131 (1992) describe immunoconjugates in which a maytansinoid was conjugated via a disulfide linker to the murine antibody A7 binding to an antigen on human colon cancer cell lines, or to another murine monoclonal antibody TA. l that binds the HER-2/neu oncogene. The cytotoxicity of the TA.l- maytansonoid conjugate was tested in vitro on the human breast cancer cell line SK-BR-3, which expresses 3 x 105 HER-2 surface antigens per cell. The drug conjugate achieved a degree of cytotoxicity similar to the free maytansinoid drug, which could be increased by increasing the number of maytansinoid molecules per antibody molecule. The A7 -maytansinoid conjugate showed low systemic cytotoxicity in mice.
[0235] Antibody-maytansinoid conjugates are prepared by chemically linking an antibody to a maytansinoid molecule without significantly diminishing the biological activity of either the antibody or the maytansinoid molecule. See, e.g., U.S. Patent No. 5,208,020 (the disclosure of which is hereby expressly incorporated by reference). An average of 3-4 maytansinoid molecules conjugated per antibody molecule has shown efficacy in enhancing cytotoxicity of target cells without negatively affecting the function or solubility of the antibody, although even one molecule of toxin/antibody would be expected to enhance cytotoxicity over the use of naked antibody. Maytansinoids are well known in the art and can be synthesized by known techniques or isolated from natural sources. Suitable maytansinoids are disclosed, for example, in U.S. Patent No. 5,208,020 and in the other patents and nonpatent publications referred to hereinabove. Preferred maytansinoids are maytansinol and maytansinol analogues modified in the aromatic ring or at other positions of the maytansinol molecule, such as various maytansinol esters.
[0236] There are many linking groups known in the art for making antibody-maytansinoid conjugates, including, for example, those disclosed in U.S. Patent No. 5208020 or EP Patent 0 425 235 Bl; Chari et al. Cancer Research 52: 127-131 (1992); and US 2005/016993 Al, the disclosures of which are hereby expressly incorporated by reference. Antibody-maytansinoid conjugates comprising the linker component SMCC may be prepared as disclosed in US 2005/0276812 Al, "Antibody-drug conjugates and Methods." The linkers comprise disulfide groups, thioether groups, acid labile groups, photolabile groups, peptidase labile groups, or esterase labile groups, as disclosed in the above-identified patents. Additional linkers are described and exemplified herein.
[0237] Conjugates of the antibody and maytansinoid may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4- (N-maleimidomethyl) cyclohexane-l-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HQ), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as l,5-difluoro-2,4-dinitrobenzene). In certain embodiments, the coupling agent is N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP) (Carlsson et al., Biochem. J. 173:723-737 (1978)) or N-succinimidyl-4-(2-pyridylthio)pentanoate (SPP) to provide for a disulfide linkage.
[0238] The linker may be attached to the maytansinoid molecule at various positions, depending on the type of the link. For example, an ester linkage may be formed by reaction with a hydroxyl group using conventional coupling techniques. The reaction may occur at the C-3 position having a hydroxyl group, the C-14 position modified with hydroxymethyl, the C-15 position modified with a hydroxyl group, and the C-20 position having a hydroxyl group. In one embodiment, the linkage is formed at the C-3 position of maytansinol or a maytansinol analogue.
[0239] Auristatins and dolastatins
[0240] In some embodiments, an immunoconjugate comprises an antibody conjugated to dolastatin or a dolastatin peptidic analog or derivative, e.g., an auristatin (US Pat. Nos. 5635483; 5780588). Dolastatins and auristatins have been shown to interfere with microtubule dynamics, GTP hydrolysis, and nuclear and cellular division (Woyke et al (2001) Antimicrob. Agents and Chemother. 45(12):3580-3584) and have anticancer (US Pat. No.5663149) and antifungal activity (Pettit et al (1998) Antimicrob. Agents Chemother. 42:2961-2965). The dolastatin or auristatin drug moiety may be attached to the antibody through the N (amino) terminus or the C (carboxyl) terminus of the peptidic drug moiety (WO
02/088172).
[0241] Exemplary auristatin embodiments include the N-terminus linked monomethylauristatin drug moieties DE and DF (US 7498298).
[0242]
Figure imgf000052_0001
[0243] wherein the wavy line of DE and DF indicates the covalent attachment site to an antibody or antibody-linker component, and independently at each location:
[0244] R2 is selected from H and CrC8 alkyl; [0245] R3 is selected from H, C C8 alkyl, C3-C8 carbocycle, aryl, C C8 alkyl-aryl, C C8 alkyl-(C3-C8 carbocycle), C3-C8 heterocycle and Ci-C8 alkyl-(C3-C8 heterocycle);
[0246] R4 is selected from H, C C8 alkyl, C3-C8 carbocycle, aryl, C C8 alkyl-aryl, C C8 alkyl-(C3-C8 carbocycle), C3-C8 heterocycle and Ci-C8 alkyl-(C3-C8 heterocycle);
[0247] R5 is selected from H and methyl; or R4 and R5 jointly form a carbocyclic ring and have the formula -(CRaRb)n- wherein Ra and Rb are independently selected from H, Q-C8 alkyl and C3-C8 carbocycle and n is selected from 2, 3, 4, 5 and 6;
[0248] R6 is selected from H and CrC8 alkyl;
[0249] R7 is selected from H, CrC8 alkyl, C3-C8 carbocycle, aryl, CrC8 alkyl-aryl, CrC8 alkyl-(C3-C8 carbocycle), C3-C8 heterocycle and CrC8 alkyl-(C3-C8 heterocycle);
[0250] each R8 is independently selected from H, OH, CrC8 alkyl, C3-C8 carbocycle and 0-(CrC8 alkyl);
[0251] R9 is selected from H and CrC8 alkyl;
[0252] R10 is selected from aryl or C3-C8 heterocycle;
[0253] Z is O, S, NH, or NR12, wherein R12 is C C8 alkyl;
[0254] R11 is selected from H, C C2o alkyl, aryl, C3-C8 heterocycle, -(R130)m-R14, or -(R130)m- CH(R15)2;
[0255] m is an integer ranging from 1 -1000;
[0256] R13 is C2-C8 alkyl;
[0257] R14 is H or CrC8 alkyl;
[0258] each occurrence of R15 is independently H, COOH, -(CH2)n-N(R16)2, -(CH2)n-S03H, or -(CH2)n- S03-CrC8 alkyl;
[0259] each occurrence of R16 is independently H, C C8 alkyl, or -(CH2)n-COOH;
[0260] R18 is selected from -C(R8)2-C(R8)2-aryl, -C(R8)2-C(R8)2-(C3-C8 heterocycle), and
-C(R8)2-C(R8)2-(C3-C8 carbocycle); and n is an integer ranging from 0 to 6.
[0261] In one embodiment, R3, R4 and R7 are independently isopropyl or sec-butyl and R5 is -H or methyl. In an exemplary embodiment, R3 and R4 are each isopropyl, R5 is -H, and R7 is sec-butyl. In yet another embodiment, R2 and R6 are each methyl, and R9 is -H. In still another embodiment, each occurrence of R8 is -OCH3. In an exemplary embodiment, R3 and R4 are each isopropyl, R2 and R6 are each methyl, R5 is -H, R7 is sec-butyl, each occurrence of R8 is -OCH3, and R9 is -H. In one embodiment, Z is -O- or -NH-. In one embodiment, R10 is aryl. In an exemplary embodiment, R10 is -phenyl. In an exemplary embodiment, when Z is -0-, R11 is -H, methyl or t-butyl. In one embodiment, when Z is -NH, R11 is -CH(R15)2, wherein R15 is -(CH2)n-N(R16)2, and R16 is -CrC8 alkyl or -(CH2)n-COOH. In another embodiment, when Z is -NH, R11 is -CH(R15)2, wherein R15 is -(CH2)n-S03H.
[0262] An exemplary auristatin embodiment of formula DE is MMAE, wherein the wavy line indicates the covalent attachment to a linker (L) of an antibody-drug conjugate:
Figure imgf000054_0001
[0263] An exemplary auristatin embodiment of formula DF is MMAF, wherein the wavy line indicates the covalent attachment to a linker (L) of an antibody-drug conjugate (see US 7498298 and Doronina et al. (2006
Figure imgf000054_0002
MMAF
[0264] Other exemplary embodiments include monomethylvaline compounds having phenylalanine carboxy modifications at the C-terminus of the pentapeptide auristatin drug moiety (WO 2007/008848) and monomethylvaline compounds having phenylalanine sidechain modifications at the C-terminus of the pentapeptide auristatin drug moiety (WO 2007/008603).
[0265] Other drug moieties include the following MMAF derivatives, wherein the wavy line indicates the cov
Figure imgf000054_0003
Figure imgf000055_0001
Figure imgf000056_0001
[0266] In one aspect, hydrophilic groups including but not limited to, triethylene glycol esters (TEG), as shown above, can be attached to the drug moiety at R11. Without being bound by any particular theory, the hydrophilic groups assist in the internalization and non-agglomeration of the drug moiety.
[0267] Exemplary embodiments of ADCs of Formula I comprising an auristatin/dolastatin or derivative thereof are described in US 7498298 and Doronina et al. (2006) Bioconjugate Chem. 17: 114-124, which is expressly incorporated herein by reference. Exemplary embodiments of ADCs of Formula I comprising MMAE or MMAF and various linker components have the following structures and abbreviations (wherein "Ab" is an antibody; p is 1 to about 8, "Val-Cit" is a valine -citrulline dipeptide; and "S" is a sulfur atom:
Figure imgf000056_0002
Ab-MC-vc-PAB-MMAF
Figure imgf000056_0003
Ab-MC-vc-PAB-MMAE
Figure imgf000056_0004
Ab-MC-MMAE
Figure imgf000057_0001
Ab-MC-MMAF
[0268] Exemplary embodiments of ADCs of Formula I comprising MMAF and various linker components further include Ab-MC-PAB-MMAF and Ab-PAB-MMAF. Interestingly,
immunoconjugates comprising MMAF attached to an antibody by a linker that is not proteolytically cleavable have been shown to possess activity comparable to immunoconjugates comprising MMAF attached to an antibody by a proteolytically cleavable linker. See, Doronina et al. (2006) Bioconjugate Chem. 17: 114-124. In such instances, drug release is believed to be effected by antibody degradation in the cell. Id.
[0269] Typically, peptide-based drug moieties can be prepared by forming a peptide bond between two or more amino acids and/or peptide fragments. Such peptide bonds can be prepared, for example, according to the liquid phase synthesis method (see E. Schroder and K. Liibke, "The Peptides", volume 1, pp 76-136, 1965, Academic Press) that is well known in the field of peptide chemistry.
Auristatin/dolastatin drug moieties may be prepared according to the methods of: US 2005-0238649 Al ; US Pat. No.5635483; US Pat. No.5780588; Pettit et al (1989) J. Am. Chem. Soc. 111 :5463-5465; Pettit et al (1998) Anti-Cancer Drug Design 13:243-277; Pettit, G.R., et al. Synthesis, 1996, 719-725; Pettit et al (1996) J. Chem. Soc. Perkin Trans. 1 5:859-863; and Doronina (2003) Nat. Biotechnol. 21(7):778-784.
[0270] In particular, auristatin/dolastatin drug moieties of formula DF, such as MMAF and derivatives thereof, may be prepared using methods described in US 7498298 and Doronina et al. (2006)
Bioconjugate Chem. 17: 114-124. Auristatin/dolastatin drug moieties of formula DE, such as MMAE and derivatives thereof, may be prepared using methods described in Doronina et al. (2003) Nat. Biotech. 21 :778-784. Drug-linker moieties MC-MMAF, MC-MMAE, MC-vc-PAB-MMAF, and MC-vc-PAB- MMAE may be conveniently synthesized by routine methods, e.g., as described in Doronina et al. (2003) Nat. Biotech. 21 :778-784, and US 7498298, and then conjugated to an antibody of interest.
[0271] Calicheamicin
[0272] In other embodiments, the immunoconjugate comprises an antibody conjugated to one or more calicheamicin molecules. The calicheamicin family of antibiotics are capable of producing double- stranded DNA breaks at sub-picomolar concentrations. For the preparation of conjugates of the calicheamicin family, see U.S. Pat. Nos. 5,712,374, 5,714,586, 5,739,116, 5,767,285, 5,770,701, 5,770,710, 5,773,001, 5,877,296 (all to American Cyanamid Company). Structural analogues of calicheamicin which may be used include, but are not limited to, γ , α2', α3', N-acetyl-y , PSAG and θ'ι (Hinman et al., Cancer Research 53:3336-3342 (1993), Lode et al., Cancer Research 58:2925-2928 (1998), and the aforementioned U.S. patents to American Cyanamid). Another anti-tumor drug to which the antibody can be conjugated is QFA, which is an antifolate. Both calicheamicin and QFA have intracellular sites of action and do not readily cross the plasma membrane. Therefore, cellular uptake of these agents through antibody-mediated internalization greatly enhances their cytotoxic effects.
[0273] Other cytotoxic agents
[0274] Other antitumor agents that can be conjugated to an antibody include anthracyclines (Kratz et al (2006) Current Med. Chem. 13:477-523; Jeffrey et al (2006) Bioorganic & Med. Chem. Letters 16:358- 362; Torgov et al (2005) Bioconj. Chem. 16:717-721 ; Nagy et al (2000) Proc. Natl. Acad. Sci. 97:829- 834; Dubowchik et al (2002) Bioorg. & Med. Chem. Letters 12: 1529-1532; King et al (2002) J. Med. Chem. 45:4336-4343; US 6630579), BCNU, streptozocin, vincristine and 5-fluorouracil, the family of agents known collectively as the LL-E33288 complex, described in US Pat. Nos. 5,053,394, 5,770,710, as well as esperamicins (US Pat. No. 5,877,296).
[0275] Enzymatically active toxins and fragments thereof which can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin and the tricothecenes. See, for example, WO 93/21232 published October 28, 1993.
[0276] The present invention further contemplates an immunoconjugate formed between an antibody and a compound with nucleolytic activity (e.g., a ribonuclease or a DNA endonuclease such as a deoxyribonuclease; DNase).
[0277] In certain embodiments, an immunoconjugate may comprise a highly radioactive atom. A variety of radioactive isotopes are available for the production of radioconjugated antibodies. Examples include At211, 1131, 1125, Y90, Re186, Re188, Sm153, Bi212, P32, Pb212 Zr89 and radioactive isotopes of Lu. When the immunoconjugate is used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc99m or I123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123, iodine-131, indium-I l l, fluorine-19, carbon-13, nitrogen- 15, oxygen- 17, gadolinium, manganese or iron.
[0278] The radio- or other labels may be incorporated in the immunoconjugate in known ways. For example, the peptide may be biosynthesized or may be synthesized by chemical amino acid synthesis using suitable amino acid precursors involving, for example, fluorine- 19 in place of hydrogen. Labels such as tc99m or I123, Re186, Re188 Zr89 and In111 can be attached via a cysteine residue in the peptide.
Yttrium-90 can be attached via a lysine residue. The IODOGEN method (Fraker et al (1978) Biochem. Biophys. Res. Commun. 80: 49-57 can be used to incorporate iodine-123. "Monoclonal Antibodies in Immunoscintigraphy" (Chatal, CRC Press 1989) describes other methods in detail.
[0279] In certain embodiments, an immunoconjugate may comprise an antibody conjugated to a prodrug-activating enzyme that converts a prodrug (e.g., a peptidyl chemotherapeutic agent, see WO 81/01145) to an active drug, such as an anti-cancer drug. Such immunoconjugates are useful in antibody- dependent enzyme -mediated prodrug therapy ("ADEPT"). Enzymes that may be conjugated to an antibody include, but are not limited to, alkaline phosphatases, which are useful for converting phosphate -containing prodrugs into free drugs; arylsulfatases, which are useful for converting sulfate - containing prodrugs into free drugs; cytosine deaminase, which is useful for converting non-toxic 5- fluorocytosine into the anti-cancer drug, 5-fluorouracil; proteases, such as serratia protease, thermolysin, subtilisin, carboxypeptidases and cathepsins (such as cathepsins B and L), which are useful for converting peptide-containing prodrugs into free drugs; D-alanylcarboxypeptidases, which are useful for converting prodrugs that contain D-amino acid substituents; carbohydrate-cleaving enzymes such as β- galactosidase and neuraminidase, which are useful for converting glycosylated prodrugs into free drugs; β-lactamase, which is useful for converting drugs derivatized with β-lactams into free drugs; and penicillin amidases, such as penicillin V amidase and penicillin G amidase, which are useful for converting drugs derivatized at their amine nitrogens with phenoxyacetyl or phenylacetyl groups, respectively, into free drugs. Enzymes may be covalently bound to antibodies by recombinant DNA techniques well known in the art. See, e.g., Neuberger et al., Nature 312:604-608 (1984).
[0280] Drug Loading
[0281] Drug loading is represented by p, the average number of drug moieties per antibody in a molecule of Formula I. Drug loading may range from 1 to 20 drug moieties (D) per antibody. ADCs of Formula I include collections of antibodies conjugated with a range of drug moieties, from 1 to 20. The average number of drug moieties per antibody in preparations of ADC from conjugation reactions may be characterized by conventional means such as mass spectroscopy, ELISA assay, and HPLC. The quantitative distribution of ADC in terms of p may also be determined. In some instances, separation, purification, and characterization of homogeneous ADC where p is a certain value from ADC with other drug loadings may be achieved by means such as reverse phase HPLC or electrophoresis.
[0282] For some antibody-drug conjugates, p may be limited by the number of attachment sites on the antibody. For example, where the attachment is a cysteine thiol, as in the exemplary embodiments above, an antibody may have only one or several cysteine thiol groups, or may have only one or several sufficiently reactive thiol groups through which a linker may be attached. In certain embodiments, higher drug loading, e.g. p >5, may cause aggregation, insolubility, toxicity, or loss of cellular permeability of certain antibody-drug conjugates. In certain embodiments, the drug loading for an ADC of the invention ranges from 1 to about 8; from about 2 to about 6; or from about 3 to about 5. Indeed, it has been shown that for certain ADCs, the optimal ratio of drug moieties per antibody may be less than 8, and may be about 2 to about 5. See US 2005-0238649 Al .
[0283] In certain embodiments, fewer than the theoretical maximum of drug moieties are conjugated to an antibody during a conjugation reaction. An antibody may contain, for example, lysine residues that do not react with the drug-linker intermediate or linker reagent, as discussed below. Generally, antibodies do not contain many free and reactive cysteine thiol groups which may be linked to a drug moiety; indeed most cysteine thiol residues in antibodies exist as disulfide bridges. In certain embodiments, an antibody may be reduced with a reducing agent such as dithiothreitol (DTT) or tricarbonylethylphosphme (TCEP), under partial or total reducing conditions, to generate reactive cysteine thiol groups. In certain embodiments, an antibody is subjected to denaturing conditions to reveal reactive nucleophilic groups such as lysine or cysteine.
[0284] The loading (drug/antibody ratio) of an ADC may be controlled in different ways, e.g., by: (i) limiting the molar excess of drug-linker intermediate or linker reagent relative to antibody, (ii) limiting the conjugation reaction time or temperature, and (iii) partial or limiting reductive conditions for cysteine thiol modification.
[0285] It is to be understood that where more than one nucleophilic group reacts with a drug-linker intermediate or linker reagent followed by drug moiety reagent, then the resulting product is a mixture of ADC compounds with a distribution of one or more drug moieties attached to an antibody. The average number of drugs per antibody may be calculated from the mixture by a dual ELISA antibody assay, which is specific for antibody and specific for the drug. Individual ADC molecules may be identified in the mixture by mass spectroscopy and separated by HPLC, e.g. hydrophobic interaction chromatography (see, e.g., McDonagh et al (2006) Prot. Engr. Design & Selection 19(7):299-307; Hamblett et al (2004) Clin. Cancer Res. 10:7063-7070; Hamblett, K.J., et al. "Effect of drug loading on the pharmacology, pharmacokinetics, and toxicity of an anti-CD30 antibody-drug conjugate," Abstract No. 624, American Association for Cancer Research, 2004 Annual Meeting, March 27-31 , 2004, Proceedings of the AACR, Volume 45, March 2004; Alley, S.C., et al. "Controlling the location of drug attachment in antibody-drug conjugates," Abstract No. 627, American Association for Cancer Research, 2004 Annual Meeting, March 27-31, 2004, Proceedings of the AACR, Volume 45, March 2004). In certain embodiments, a homogeneous ADC with a single loading value may be isolated from the conjugation mixture by electrophoresis or chromatography.
[0286] Certain Methods of Preparing Immunconjugates
[0287] An ADC of Formula I may be prepared by several routes employing organic chemistry reactions, conditions, and reagents known to those skilled in the art, including: (1) reaction of a nucleophilic group of an antibody with a bivalent linker reagent to form Ab-L via a covalent bond, followed by reaction with a drug moiety D; and (2) reaction of a nucleophilic group of a drug moiety with a bivalent linker reagent, to form D-L, via a covalent bond, followed by reaction with a nucleophilic group of an antibody.
Exemplary methods for preparing an ADC of Formula I via the latter route are described in US 2005- 0238649 Al, which is expressly incorporated herein by reference.
[0288] Nucleophilic groups on antibodies include, but are not limited to: (i) N-terminal amine groups, (ii) side chain amine groups, e.g. lysine, (iii) side chain thiol groups, e.g. cysteine, and (iv) sugar hydroxyl or amino groups where the antibody is glycosylated. Amine, thiol, and hydroxyl groups are nucleophilic and capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleimide groups. Certain antibodies have reducible interchain disulfides, i.e. cysteine bridges.
Antibodies may be made reactive for conjugation with linker reagents by treatment with a reducing agent such as DTT (dithiothreitol) or tricarbonylethylphosphine (TCEP), such that the antibody is fully or partially reduced. Each cysteine bridge will thus form, theoretically, two reactive thiol nucleophiles. Additional nucleophilic groups can be introduced into antibodies through modification of lysine residues, e.g., by reacting lysine residues with 2-iminothiolane (Traut's reagent), resulting in conversion of an amine into a thiol. Reactive thiol groups may be introduced into an antibody by introducing one, two, three, four, or more cysteine residues (e.g., by preparing variant antibodies comprising one or more non- native cysteine amino acid residues).
[0289] Antibody-drug conjugates of the invention may also be produced by reaction between an electrophilic group on an antibody, such as an aldehyde or ketone carbonyl group, with a nucleophilic group on a linker reagent or drug. Useful nucleophilic groups on a linker reagent include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide. In one embodiment, an antibody is modified to introduce electrophilic moieties that are capable of reacting with nucleophilic subsituents on the linker reagent or drug. In another embodiment, the sugars of glycosylated antibodies may be oxidized, e.g. with periodate oxidizing reagents, to form aldehyde or ketone groups which may react with the amine group of linker reagents or drug moieties. The resulting imine Schiff base groups may form a stable linkage, or may be reduced, e.g. by borohydride reagents to form stable amine linkages. In one embodiment, reaction of the carbohydrate portion of a glycosylated antibody with either galactose oxidase or sodium meta-periodate may yield carbonyl (aldehyde and ketone) groups in the antibody that can react with appropriate groups on the drug (Hermanson, Bioconjugate Techniques). In another embodiment, antibodies containing N-terminal serine or threonine residues can react with sodium meta-periodate, resulting in production of an aldehyde in place of the first amino acid (Geoghegan & Stroh, (1992) Bioconjugate Chem. 3: 138-146; US
5362852). Such an aldehyde can be reacted with a drug moiety or linker nucleophile.
[0290] Nucleophilic groups on a drug moiety include, but are not limited to: amine, thiol, hydroxyl, hydrazide, oxime, hydrazine, thiosemicarbazone, hydrazine carboxylate, and arylhydrazide groups capable of reacting to form covalent bonds with electrophilic groups on linker moieties and linker reagents including: (i) active esters such as NHS esters, HOBt esters, haloformates, and acid halides; (ii) alkyl and benzyl halides such as haloacetamides; (iii) aldehydes, ketones, carboxyl, and maleimide groups.
[0291] The compounds of the invention expressly contemplate, but are not limited to, ADC prepared with the following cross-linker reagents: BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo-GMBS, sulfo-KMUS, sulfo-MBS, sulfo- SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl-(4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc., Rockford, IL., U.S.A.
[0292] Immunoconjugates comprising an antibody and a cytotoxic agent may also be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl) cyclohexane-l -carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HC1), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p- azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)- ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as l,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987). Carbon- 14-labeled l-isothiocyanatobenzyl-3- methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See W094/11026.
[0293] Alternatively, a fusion protein comprising an antibody and a cytotoxic agent may be made, e.g., by recombinant techniques or peptide synthesis. A recombinant DNA molecule may comprise regions encoding the antibody and cytotoxic portions of the conjugate either adjacent to one another or separated by a region encoding a linker peptide which does not destroy the desired properties of the conjugate.
[0294] In yet another embodiment, an antibody may be conjugated to a "receptor" (such as streptavidin) for utilization in tumor pre -targeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g., avidin) which is conjugated to a cytotoxic agent (e.g., a
radionucleotide).
[0295] Pharmaceutical Formulations
[0296] In one aspect, the invention further provides pharmaceutical formulations comprising at least one antibody of the invention and/or at least one immunoconjugate thereof. In some embodiments, a pharmaceutical formulation comprises 1) an antibody of the invention and/or an immunoconjugate thereof, and 2) a pharmaceutically acceptable carrier. In some embodiments, a pharmaceutical formulation comprises 1) an antibody of the invention and/or an immunoconjugate thereof, and optionally, 2) at least one additional therapeutic agent. Additional therapeutic agents include, but are not limited to, those described below.
[0297] Pharmaceutical formulations comprising an antibody or immunoconjugate of the invention are prepared for storage by mixing the antibody or immunoconjugate having the desired degree of purity with optional physiologically acceptable carriers, excipients or stabilizers {Remington 's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)) in the form of aqueous solutions or lyophilized or other dried formulations. Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, histidine and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as
octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride); phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins;
hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes {e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). Pharmaceutical formulations to be used for in vivo administration are generally sterile. This is readily accomplished by filtration through sterile filtration membranes.
[0298] Active ingredients may also be entrapped in microcapsule prepared, for example, by
coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980).
[0299] Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody or immunoconjugate of the invention, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and y ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable
microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)-3- hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated antibodies or immunoconjugates remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37°C, resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S-S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
[0300] An antibody may be formulated in any suitable form for delivery to a target cell/tissue. For example, antibodies may be formulated as immunoliposomes. A "liposome" is a small vesicle composed of various types of lipids, phospholipids and/or surfactant which is useful for delivery of a drug to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of biological membranes. Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA 82:3688 (1985); Hwang et al., Proc. Natl Acad. Sci. USA 77:4030 (1980); U.S. Pat. Nos. 4,485,045 and 4,544,545; and
W097/38731 published October 23, 1997. Liposomes with enhanced circulation time are disclosed in U.S. Patent No. 5,013,556.
[0301] Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter. Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al., J. Biol. Chem. 257:286-288 (1982) via a disulfide interchange reaction. A chemotherapeutic agent is optionally contained within the liposome. See Gabizon et al., J. National Cancer Inst. 81(19): 1484 (1989).
[0302] In another embodiment, an immunoconjugate comprises an antibody as described herein conjugated to an enzymatically active toxin or fragment thereof, including but not limited to diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
[0303] In another embodiment, an immunoconjugate comprises an antibody as described herein conjugated to a radioactive atom to form a radioconjugate. A variety of radioactive isotopes are available for the production of radioconjugates. Examples include Zr89, At211, 1131, 1125, Y90, Re186, Re188, Sm153,
212 32 212
Bi , P , Pb and radioactive isotopes of Lu. When the radioconjugate is used for detection, it may comprise a radioactive atom for scintigraphic studies, for example tc99m or 1123, or a spin label for nuclear magnetic resonance (NMR) imaging (also known as magnetic resonance imaging, mri), such as iodine-123 again, iodine-131, indium-I l l, fluorine-19, carbon-13, nitrogen-15, oxygen-17, gadolinium, manganese or iron.
[0304] Conjugates of an antibody and cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyridyldithio) propionate (SPDP), succinimidyl-4- (N-maleimidomethyl) cyclohexane-l-carboxylate (SMCC), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HQ), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as toluene 2,6-diisocyanate), and bis-active fluorine compounds (such as l,5-difluoro-2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987). Carbon- 14-labeled l-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX- DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See
W094/11026. The linker may be a "cleavable linker" facilitating release of a cytotoxic drug in the cell. For example, an acid-labile linker, peptidase-sensitive linker, photolabile linker, dimethyl linker or disulfide-containing linker (Chari et al., Cancer Res. 52: 127-131 (1992); U.S. Patent No. 5,208,020) may be used.
[0305] The immunuoconjugates or ADCs herein expressly contemplate, but are not limited to such conjugates prepared with cross-linker reagents including, but not limited to, BMPS, EMCS, GMBS, HBVS, LC-SMCC, MBS, MPBH, SBAP, SIA, SIAB, SMCC, SMPB, SMPH, sulfo-EMCS, sulfo- GMBS, sulfo-KMUS, sulfo-MBS, sulfo-SIAB, sulfo-SMCC, and sulfo-SMPB, and SVSB (succinimidyl- (4-vinylsulfone)benzoate) which are commercially available (e.g., from Pierce Biotechnology, Inc.,
Rockford, IL., U.S. A).
[0306] Pharmaceutical Formulations
[0307] Pharmaceutical formulations of an anti-ETBR antibody as described herein are prepared by mixing such antibody having the desired degree of purity with one or more optional pharmaceutically acceptable carriers (Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized formulations or aqueous solutions. Pharmaceutically acceptable carriers are generally nontoxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride; benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes); and/or non-ionic surfactants such as polyethylene glycol (PEG). Exemplary pharmaceutically acceptable carriers herein further include insterstitial drug dispersion agents such as soluble neutral-active hyaluronidase glycoproteins (sHASEGP), for example, human soluble PH-20 hyaluronidase glycoproteins, such as rHuPH20 (HYLENEX®, Baxter International, Inc.). Certain exemplary sHASEGPs and methods of use, including rHuPH20, are described in US Patent Publication Nos. 2005/0260186 and 2006/0104968. In one aspect, a sHASEGP is combined with one or more additional glycosaminoglycanases such as chondroitinases.
[0308] Exemplary lyophilized antibody formulations are described in US Patent No. 6,267,958.
Aqueous antibody formulations include those described in US Patent No. 6,171,586 and
WO2006/044908, the latter formulations including a histidine-acetate buffer.
[0309] The formulation herein may also contain more than one active ingredients as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. For example, it may be desirable to further provide a BRAF inhibitor, a MEK inhibitor or an anti-CTLA-4 antibody, ipilimumab. Such active ingredients are suitably present in combination in amounts that are effective for the purpose intended.
[0310] Active ingredients may be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin- microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington's Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980). [0311] Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g. films, or microcapsules.
[0312] The formulations to be used for in vivo administration are generally sterile. Sterility may be readily accomplished, e.g., by filtration through sterile filtration membranes.
[0313] Therapeutic Methods and Compositions
[0314] Any of the anti-ETBR antibodies provided herein may be used in therapeutic methods.
[0315] In one aspect, an anti-ETBR antibody for use as a medicament is provided. In another aspect, the methods provide for an anti-ETBR antibody in combination with a BRAF inhibitor as useful as a medicament. In further aspects, such a combination is useful in treating melanoma and/or metastatic melanoma. In certain embodiments, an anti-ETBR antibody in combination with a BRAF inhibitor for use in a method of treatment is provided. In certain embodiments, the invention provides an anti-ETBR antibody for use in a method of treating an individual having melanoma and/or metastatic melanoma comprising administering to the individual an effective amount of the anti-ETBR antibody and an effective amount of a BRAF inhibitor. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below, to the combination described. In further embodiments, the invention provides an anti- ETBR antibody in combination with a BRAF inhibitor for use in tumor growth inhibition (TGI). In certain embodiments, the invention provides an anti-ETBR antibody in combination with a BRAF inhibitor for use in a method of inhibiting tumor growth in a subject comprising administering to the subject an effective of the anti-ETBR antibody in combination with a BRAF inhibitor to inhibit tumor growth. A "subject" according to any of the above embodiments is preferably a human.
[0316] In a further aspect, the invention provides for the use of an anti-ETBR antibody in combination with a BRAF inhibitor in the manufacture or preparation of a medicament. In one embodiment, the medicament is for treatment of melanoma and/or metastatic melanoma. In a further embodiment, the medicament is for use in a method of treating melanoma and/or metastatic melanoma comprising administering to an individual having melanoma and/or metastatic melanoma an effective amount of the medicament. In one such embodiment, the method further comprises administering to the individual an effective amount of at least one additional therapeutic agent, e.g., as described below. In a further embodiment, the medicament is for tumor growth inhibition. In a further embodiment, the medicament is for use in a method of tumor growth inhibition in an individual comprising administering to the individual an amount effective of the medicament to inhibit tumor growth. An "individual" according to any of the above embodiments may be a human.
[0317] In a further aspect, the invention provides pharmaceutical formulations comprising any of the anti-ETBR antibodies provided herein, e.g., in combination with a BRAF inhibitor for use in any of the above therapeutic methods. In one embodiment, a pharmaceutical formulation comprises any of the anti- ETBR antibodies provided herein in combination with a BRAF inhibitor and a pharmaceutically acceptable carrier. In another embodiment, a pharmaceutical formulation comprises any of the anti- ETBR antibodies provided herein in combination with a BRAF inhibitor and at least one additional therapeutic agent, e.g., as described below.
[0318] Antibodies of the invention can be used either alone or in combination with other agents in a therapy. For instance, an antibody of the invention may be co-administered with at least one additional therapeutic agent. In certain non-limiting embodiments, an additional therapeutic agent is a BRAF inhibitor, a MEK inhibitor, or an anti-CTLA-4 antibody, such as, for example, ipilimumab.
[0319] Such combination therapies noted above encompass combined administration (where two or more therapeutic agents are included in the same or separate formulations), and separate administration, in which case, administration of the antibody of the invention can occur prior to, simultaneously, and/or following, administration of the additional therapeutic agent and/or adjuvant. Antibodies of the invention can also be used in combination with radiation therapy.
[0320] An antibody of the invention (and any additional therapeutic agent, such as, for example, a BRAF inhibitor) can be administered by any suitable means, including parenteral, intrapulmonary, and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous administration. Dosing can be by any suitable route, e.g. by injections, such as intravenous or subcutaneous injections, depending in part on whether the administration is brief or chronic. Various dosing schedules including but not limited to single or multiple administrations over various time -points, bolus administration, and pulse infusion are contemplated herein. Alternatively, the BRAF inhibitor may be administered orally, in either tablet or capsule or liquid form.
[0321] Antibodies of the invention would be formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The antibody need not be, but is optionally formulated with one or more agents currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of antibody present in the formulation, the type of disorder or treatment, and other factors discussed above. These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.
[0322] For the prevention or treatment of disease, the appropriate dosage of an antibody of the invention (when used alone or in combination with one or more other additional therapeutic agents) will depend on the type of disease to be treated, the type of antibody, the severity and course of the disease, whether the antibody is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the antibody, and the discretion of the attending physician. The antibody is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g. O. lmg/kg-lOmg/kg) of antibody can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 μg/kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of the antibody would be in the range from about 0.05 mg/kg to about 10 mg/kg. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g. every week or every three weeks (e.g. such that the patient receives from about two to about twenty, or e.g. about six doses of the antibody). An initial higher loading dose, followed by one or more lower doses may be administered. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.
[0323] Articles of Manufacture
[0324] In another aspect of the invention, an article of manufacture containing materials useful for the treatment, prevention and/or diagnosis of the disorders described above is provided. The article of manufacture comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV solution bags, etc. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition which is by itself or combined with another composition effective for treating, preventing and/or diagnosing the condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). At least one active agent in the composition is an antibody of the invention. The label or package insert indicates that the composition is used for treating the condition of choice. Moreover, the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises an antibody of the invention; and (b) a second container with a composition contained therein, wherein the composition comprises a further cytotoxic or otherwise therapeutic agent. The article of manufacture in this embodiment of the invention may further comprise a package insert indicating that the compositions can be used to treat a particular condition. Alternatively, or additionally, the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically- acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes. TABLE 2: SEQUENCES
Figure imgf000069_0001
DIITMDYKGSYLRICLLHPVQKTAFMQFYKTAKDWWLFSF
YFCLPLAITAFFYTLMTCEM
LRKKSGMQIALNDHLKQRREVAKTVFCLVLVFALCWLPL HL SRILKLTL YNQNDPNRCEL
LSFLLVLDYIGINMASLNSCINPIALYLVSKRFK CFKSC LCC WCQ SFEEKQ SLEEKQ S CLKFKANDHGYDNFRS SNKYS S S
EXAMPLES
[0325] The following are examples of methods and compositions of the invention. It is understood that various other embodiments may be practiced, given the general description provided above.
[0326] Example 1 : In vitro evaluations of specific cell killing by an anti-ETBR ADC
[0327] The anti-ETBR antibody- ADC candidate Hu5E9vl -ADC was evaluated in vitro on melanoma cell lines expressing either relatively low ETBR copy number, in the case of cell line A2058 (Obtained from American Type Culture Collection) or high ETBR copy number, in the case of cell line UACC- 257X2.2. The UACC-257X2.2 cell line is a derivative of the parental UACC-257 cell line (NCI- Frederick Cancer DCT Tumor Repository) optimized for growth in vivo. Parental UACC-257 cells were injected subcutaneously in the right flank of female NCr nude mice, one tumor was harvested and dissociated grown in vitro resulting in the UACC-257X1.2 cell line. The UACC-257X1.2 line was injected again subcutaneously in the right flank of female NCr nude mice in an effort to improve the growth of the cell line. A tumor from this study was collected and again adapted for in vitro growth to generate the UACC-257X2.2 cell line. This cell line expresses high levels of ETBR as determined by flow cytometry. The relationship of receptor levels to Hu5E9vl-vc-MMAE cell killing in these cell lines was evaluated as follows.
[0328] The melanoma cell lines A2058 and UACC-257X2.2 were grown in appropriate media at 37°C and 5% C02. To assess the effects of Hu5E9vl-ADC on cell viability, cells were plated at 1,500 per well in 50 μΕ of normal growth medium in 96-well clear-bottom black plates. Twenty-four hours later, an additional 50 μΕ of culture medium with serial dilutions of Hu5E9vl-ADC concentrations was added to triplicate wells. Five days later, cell survival was determined using CellTiter-Glo Luminescent Cell Viability Reagent (G7572; Promega Corporation) and with an EnVision 2101 Mutilabel Reader (Perkin- Elmer).
[0329] A determination of antibody binding sites per cell was performed (Scatchard analysis): The affinity constant and the number of cell surface binding sites for each antibody were estimated by incubating the melanoma cells for 4 h on ice with a fixed concentration of 125I-labeled Hu5E9vl-ADC combined with increasing concentrations of unlabeled Hu5E9vl -ADC. The data were analyzed by nonlinear curve fitting using an analysis program method.
[0330] As shown in Figure 2A and 2B, by Scatchard analysis, the number of Hu5E9vl-ADC binding sites on A2058 and UACC-257X2.2 was estimated at 1,582 sites and 33,939 sites per cell, respectively. Titration of these cell lines with the anti-ETBR ADC candidate showed specific cell killing relative to control ADC that was generally proportional to the level of ETBR expression.
[0331] Example 2: In vivo evaluations of specific tumor killing by an anti-ETBR ADC
[0332] Based on the studies described in Example 1 above, melanoma cell lines A2058 and UACC- 257X2.2 were selected as suitable models for in vivo anti-tumor activity studies that represent a wide range of ETBR expression. The UACC-257X2.2 melanoma cell line is a derivative of the parental UACC-257 melanoma cell line (National Cancer Institute (NCI)) optimized for growth in vivo.
Specifically, parental UACC-257 cells were injected subcutaneously in the right flank of female NCr nude mice, one tumor was harvested and grown in vitro resulting in the UACC-257X1.2 cell line. The UACC-257X1.2 line was injected again subcutaneously in the right flank of female NCr nude mice in an effort to improve the growth of the cell line. A tumor from this study was collected and again adapted for in vitro growth to generate the UACC-257X2.2 cell line. This cell line and tumors derived from this line express ETBR comparable to the parental cell line UACC-257 (data not shown).
[0333] Next, efficacy studies were performed using the melanoma cells lines in the xenografts mouse models described above. All studies were conducted in accordance with the Guide for the Care and Use of Laboratory Animals (Ref: Institute of Laboratory Animal Resources (NIH publication no. 85-23), Washington, DC: National Academies Press; 1996). 10- to 14-week-old female CRL Nu/Nu or NCr nude mice from Charles River Laboratories were inoculated subcutaneously in the dorsal right flank with either 5 X 106 UACC-257X2.2 cells in HBSS with Matrigel or 5 X 106 A2058 cells in HBSS with Matrigel. When tumor volumes reached approximately 200 mm3 (day 0), animals were randomized into groups of 10 each.
[0334] For single agent efficacy studies, the anti-ETBR ADC candidate Hu5E9vl -ADC was administered as a single intravenous (IV) injection on day 0 at 1 mpk, 3 mpk, or 6 mpk (mg/kg). A control ADC antibody and vehicle control were also administered. Average tumor volumes with standard deviations were determined from 10 animals per group. Tumor volumes were measured twice per week until study end.
[0335] The results are shown in Figures 3A for the high ETBR copy number UACC-257X2.2 cell line and 3B for the low ETBR copy number cell line A2058. Consistent with the in vitro cell killing experiments described in Example 1, the UACC-257X2.2 xenograft tumors were more responsive to the Hu5E9vl-ADC. While efficacy was not apparent for the group dosed at 1 mg/kg, sustained tumor regression was observed in response to single dose of 3 and 6 mg/kg Hu5E9vl-ADC (Figure 3 A).
[0336] Doses of 3 and 6 mg/kg of Hu5E9vl-ADC, 6mg/kg control ADC or vehicle control were administered to animals bearing the low ETBR copy number A2058 tumors. A partial reduction in tumor burden was observed at the high dose of 6 mpk of the Hu5E9vl-ADC relative to the matching dose of control ADC or vehicle. Efficacy was not apparent for the group dosed at 3 mg/kg of Hu5E9vl-ADC. Thus, a reduction of tumor burden in the A2058 xenograft model that represents the low end of the spectrum of ETBR expression in human melanomas (Asundi et al, 2011) suggests that efficacy can be achieved with the candidate Hu5E9vl-ADC as a single agent in tumors that correspond to the full expression range of ETBR encountered in human melanomas.
[0337] Example 3 : Effect of BRAF inhibitor drugs on the expression levels of ETBR
[0338] The effect of BRAF inhibitor drugs on the expression level of ETBR transcript and protein (total protein and cell surface protein) was evaluated in a variety of melanoma cells representing various genetic backgrounds of melanoma, such as mutant for RAF(Y600E), wild-type for BRAF and mutant for RAS (Q61L).
[0339] Melanoma cell lines UACC-257X2.2, A2058, COLO 829, IPC-298 (ATCC) were treated with a BRAF inhibitor drug ("BRAFi"), specifically RG7204 at varying concentrations by adding the appropriate drug volume to cells in culture for 24h on four-well dishes.
[0340] To determine the effect of BRAFi on ETBR and control ribosomal protein LI 9 (RPL19) transcript levels, the following experiments were performed. Cells treated with RG7204 for 24h were harvested from plates by scraping and processed for total RNA using Qiashredder and RNeasy mini kits (79654, 74104 from Qiagen, Valencia, CA). Taqman assays were set up using reagents from Applied Biosystems (ABI, Foster City, CA) and assayed using 7500 Real Time PCR machine and software from ABL Primer-probe sets were designed with primers flanking a fluorogenic probe dual labeled with Reporter dye FAM and quencher dye TAMRA.
[0341] The primer-probe set for RPL19 is as follows:
[0342] Forward primer -5' AGC GGA TTC TCA TGG AAC A (SEQ ID NO: 11); Reverse primer-5' CTG GTC AGC CAG GAG CTT (SEQ ID NO: 12) and probe-5' TCC ACA AGC TGA AGG CAG ACA AGG (SEQ ID NO: 13).
[0343] The primer-probe set for ETBR is as follows:
[0344] Forward primer- 5'TCA CTG AAT TCC TGC ATT AAC C (SEQ ID NO: 14), reverse primer- 5'GCA TAA GCA TGA CTT AAA GCA GTT (SEQ ID NO: 15) and probe-5' AAT TGC TCT GTA TTT GGT GAG CAA AAG ATT CAA (SEQ ID NO: 16).
[0345] The results for UACC-257X2.2 are shown in Figure 4A, the results for A2058 are shown in Figure 8A, and the results for COLO 829 are shown in Figure 6A. These results demonstrate that treatment with BRAFi RG7204 for 24 hours appears to increase the ETBR transcripts in all cell lines tested, as compared to control cells to which no BRAFi RG7204 was added.
[0346] To test whether the increase in ETBR transcripts due to BRAFi treatment also results in any changes in ETBR total protein levels, Western blot experiments were performed on the same cell lines treated with BRAFi RG7204 as described above. For Western blotting, the following reagents were used: for detection of proteins: an anti-ETBR in-house generated monoclonal antibody 1H1.8.5, anti- Phospho-p44/42 MAPK (Erkl/2) (Thr202/Tyr204) antibody (9101, Cell Signaling Technology), anti- p44/42 MAPK (Erkl/2) antibody (9102, Cell Signaling Technology) and as controls, a rabbit polyclonal anti-GAPDH (glyceraldehyde-3 -phosphate dehydrogenase) antibody (PA1 -987; Affinity Bioreagents) and mouse monoclonal anti- β-Tubulin antibody (556321, BD Pharmingen).The results for UACC- 257X2.2 are shown in Figure 4B, the results for A2058 are shown in Figure 8B, the results for COLO 829 are shown in Figure 6B and the results for IPC-298 are shown in Figure 10. These results demonstrate that treatment with BRAFi RG7204 for 24 hours appears to increase the ETBR total protein levels in the UACC-257X2.2, the A2058, and COLO 829 cell lines tested, as compared to control cells to which no BRAFi was added. However, with respect to the cell line IPC-298 which is wild-type for BRAF and mutant for RAS (Q61L), BRAFi does not appear to increase ETBR levels as compared over the various BRAFi dose levels, rather it appears to activate the levels of phosphor-ERK, as shown in Figure 10.
[0347] To determine whether the observed increases in total ETBR protein level due to BRAFi treatment also results in an increase of ETBR surface protein levels, a fluorescence-activated cell sorting (FACS) analysis was performed. Cells were harvested in PBS with 2.5 mmol/L EDTA and washed in PBS buffer containing 1% FBS. All subsequent steps were carried out at 4°C. Cells were incubated for 1 hour with 3μg/mL anti ETBR antibody Hu5E9vl, followed by anti-human IgG fluorescent detection reagent (Al 1013; Invitrogen). Cells were then analyzed with a FACS Calibur flow cytometer (BD Biosciences). The results for UACC-257X2.2 are shown in Figure 4C, the results for A2058 are shown in Figure 8C, and the results for COLO 829 are shown in Figure 6C. These results demonstrate that treatment with BRAFi RG7204 for 24h appears to increase the surface levels of ETBR protein expressed in all cell lines tested, as compared to control cells to which no BRAFi was added. However, with respect to cell line IPC-298, BRAFi RG7204 appears to reduce ETBR levels at all dose levels tested, as shown in Figure 11A-C.
[0348] Example 4: Effect of BRAF inhibitor drugs on in vivo efficacy of anti-ETBR ADC
[0349] Given the results demonstrated in Example 3 above, the impact of the BRAF inhibitor drug on the in vivo efficacy of an anti-ETBR ADC was tested. To do this, the in vivo efficacy for various combinations of Hu5E9vl-ADC and BRAFi-945 were evaluated against the UACC-257X2.2 melanoma model described above. Tumors were grown to an average size of approximately 200 mm3, whereupon animals were randomized into groups of 10 each. An appropriate vehicle control (Klucel LF) or BRAFi- 945 at doses of lmpk, 6 mpk or 20 mpk were administered orally once a day x 21 days beginning on study Day 0. A single 1 mpk or 3 mpk dose of Hu5E9vl -ADC or control, a histidine buffer #8, was administered intraveneously (after two doses of 945) via tail vein at study Day 1.
[0350] The average tumor volumes were determined from 10 animals per group. Tumor volumes were measured twice per week until study end. Tumor volumes were measured in two dimensions (length and width) using UltraCal IV calipers (Model 54 10 1 11, Fred V. Fowler Company; Newton, MA). The following formula was used with Excel, version 12.2.8 (Microsoft; Redmond, WA) to calculate tumor volume: Tumor Volume (mm3) = (length x width2) x 0.5
[0351] To analyze the repeated measurement of tumor volumes from the same animals over time, a mixed-modeling Linear Mixed Effects (LME) approach was used (Pinheiro et al. 2009). This approach can address both repeated measurements and a modest drop-out rate due to non-treatment-related termination of animals prior to study end. Cubic regression splines were used to fit a non-linear profile to the time courses of log2 tumor volume at each dose level. These non- linear profiles were then related to dose within the mixed model. Tumor growth inhibition (TGI) as a percentage of vehicle was calculated as percent area under the fitted curve (AUC) per day in relation to the vehicle, using the following formula:
Figure imgf000074_0001
[0352] Using this formula, a TGI value of 100% indicates tumor stasis, of > 1% but < 100% indicates tumor growth delay, and of > 100% indicates tumor regression. To get uncertainty intervals (UIs) for %TGI, the fitted curve and the fitted covariance matrix were used to generate a random sample as an approximation to the distribution of %TGI. The random sample is composed of 1000 simulated realizations of the fitted-mixed model, where the %TGI has been recalculated for each realization. Here, in the reported UI is the value for which 95% of the time, the recalculated values of %TGI will fall in this region given the fitted model. The 2.5 and 97.5 percentiles of the simulated distribution were used as the upper and lower UIs.
[0353] The results are shown in Figures 5A, 5B, 5C, 5D and 5E. All combinations of the Hu5E9vl- ADC and BRAFi-945 demonstrated better efficacy than either drug as a single agent alone. The two drugs combined at the lowest levels tested to give combination efficacy that was almost indistinguishable from the combination efficacy achieved at the highest dose levels tested.
[0354] Example 5: Dose Testing Anti-ETBR ADC and BRAFi combinations in vivo in COLO 829 Xenografts
[0355] The study described in the example above allowed a refinement of the evaluation of in vivo combination efficacy of Hu5E9vl -ADC with the BRAF inhibitor drug RG7204. A lack of antagonism between the drugs was anticipated, thus the combination efficacy of the drugs were tested at lower doses. The COLO 829 xenograft model was chosen as representative of medium levels of ETBR expression, further increasing the stringency of the combination studies. Tumors were grown to an average size of approximately 200 mm3, whereupon animals were randomized into groups of 9 each. An appropriate vehicle control (Klucel LF) or G00044364.1-12 (RG7204) at doses of 10 mpk or 30 mpk were administered orally twice a day for 21 days starting on day 0. A single dose of Hu5E9vl -ADC at either 1 mpk or 3 mpk or control, a histidine buffer #8, was administered intravenously on day 1 (after three doses of RG7204). The results are shown in Figures 7A, 7B, 7C and 7D.
[0356] Mid range doses of both drugs (30mg/kg RG7204 and 3 mg/kg of Hu5E9vl-ADC) combined well together to give combination efficacy greater than either drug alone. Other combinations of the two drugs at lower doses trended similarly, with the sole exception of the lowest doses tested in combination. [0357] Example 6: Dose Testing Anti-ETBR ADC and BRAFi combinations in vivo in A2058
Xenografts
[0358] The efficacy of Hu5E9vl -ADC and RG7204 in the A2058 xenograft model was tested. This model is of particular interest due to the high level of stringency it represents. The A2058 xenograft model represents the lower end of the ETBR expression spectrum found in melanoma patients, thereby making it a challenging model for achieving anti-ETBR ADC efficacy. Further, in spite of its BRAF V600E mutational status, this model has been demonstrated to be non-responsive to RG7204 with an in vitro killing efficacy of >20μΜ (data not shown).
[0359] Tumors were grown to an average size of approximately 200 mm3, whereupon animals were randomized into groups of 10 each. An appropriate vehicle control (Klucel LF) or RG7204 at doses of 10 mpk or 30 mpk were administered orally twice a day for 21 days starting on day 0. A single dose of Hu5E9vl-ADC at either 3 mpk or 6 mpk or control, a histidine buffer #8, was administered
intraveneously into the tail vein on day 1 (after three doses of RG7204). The results are shown in Figures 9A, 9B, 9C, and 9D.
[0360] The results show that in all cases tested, the combination of Hu5E9vl-ADC and RG7204 demonstrated greater efficacy than any single agent alone. A 10 mg/kg dose of RG7204 alone did not show single agent efficacy against the A2058 model (see Figures 9A and 9C). However, when combined with a 6 mg/kg dose of Hu5E9vl-ADC, a better efficacy was achieved than with either agent alone (see Figures 9A and 9C). The combination of 10 mg/kg RG7204 with 6 mg/kg of Hu5E9vl-ADC (Figure 9A) demonstrated combination efficacy almost indistinguishable from the combination efficacy achieved at the highest dose levels tested, i.e., 30 mpk RG7204 and 6 mpk Hu5E9vl-ADC as shown in Figure 9B.
[0361] Table 3 summarizes the three melanoma xenograft models tested at varying doses, as described above, to demonstrate the combination effects, expressed as a percent delta (last column) of the combination use of anti-ETBR ADC with a BRAF inhibitor as compared to either the percent TGI of the anti-ETBR ADC as a single agent or the percent TGI of a BRAF inhibitor as a single agent. The percent TGI was calculated using a Linear Mixed Effects (LME) modeling approach, as described above.
[0362] Example 7: Effect of MEK inhibitor drugs on the expression levels of ETBR
[0363] The effect of MEK inhibitor drugs on the expression level of ETBR transcript and protein (total protein and cell surface protein) was evaluated in a variety of melanoma cells that are either BRAF wild- type or mutational and/or RAS wild-type or mutational: COL0829 (BRAFV600E), A2058 (BRAFV600E), SK23-MEL (BRAFWT/RASWT), or IPC-298 (BRAFWT/RASC61L).
[0364] Melanoma cell lines A2058, COLO 829, SK23-MEL and IPC-298 (ATCC) were treated with a MEK inhibitor drug ("MEKi-973" or "MEKi-623"), at varying concentrations (0 μΜ, 0.01 μΜ, 0.1 μΜ or 1 μΜ) by adding the appropriate drug volume to cells in culture for 24h on four-well dishes.
[0365] To determine the effect of MEKi on ETBR transcript levels, the following experiments were performed as described above in Example 3. Cells treated with either MEKi-973 for 24h were harvested from plates by scraping and processed for total RNA using Qiashredder and RNeasy mini kits (79654, 74104 from Qiagen, Valencia, CA). Taqman assays were set up using reagents from Applied Biosystems (ABI, Foster City, CA) and assayed using 7500 Real Time PCR machine and software from ABI.
Primer-probe sets were designed with primers flanking a fluorogenic probe dual labeled with Reporter dye FAM and quencher dye TAMRA.
[0366] The primer-probe set for RPL19 is as follows:
[0367] Forward primer -5' AGC GGA TTC TCA TGG AAC A (SEQ ID NO: 11); Reverse primer-5' CTG GTC AGC CAG GAG CTT (SEQ ID NO: 12) and probe-5' TCC ACA AGC TGA AGG CAG ACA AGG (SEQ ID NO : 13) .
[0368] The primer-probe set for ETBR is as follows:
[0369] Forward primer- 5'TCA CTG AAT TCC TGC ATT AAC C (SEQ ID NO: 14), reverse primer- 5'GCA TAA GCA TGA CTT AAA GCA GTT (SEQ ID NO: 15) and probe-5' AAT TGC TCT GTA TTT GGT GAG CAA AAG ATT CAA (SEQ ID NO: 16).
[0370] The results for A2058 are shown in Figure 14A treated with MEKi-623 and 14B treated with MEKi-973 at the indicated doses. These results demonstrate that treatment with a MEK inhibitor for 24 hours appears to increase the ETBR transcripts, as compared to control cells to which no MEK inhibitor was added.
[0371] To test whether the increase in ETBR transcripts due to MEKi treatment also results in any changes in ETBR total protein levels, Western blot experiments were performed on cell lines COL0829 (BRAFV600E), A2058 (BRAFV600E), SK23-MEL (BRAFWT/RASWT), or IPC-298 (BRAFWT/RASC61L) which were treated with MEKi-973 as described above. For Western blotting, the following reagents were used: for detection of proteins: an anti-ETBR in-house generated monoclonal antibody 1H1.8.5., anti-Phospho-p44/42 MAPK (Erkl/2) (Thr202/Tyr204) antibody (9101, Cell Signaling Technology), anti-p44/42 MAPK (Erkl/2) antibody (9102, Cell Signaling Technology) and as controls, a rabbit polyclonal anti-GAPDH (glyceraldehyde-3 -phosphate dehydrogenase) antibody (PA1 -987; Affinity Bioreagents) and mouse monoclonal anti- β-Tubulin antibody (556321, BD Pharmingen). The results for A2058 are shown in Figure 15A treated with MEKi-623 and 15B treated with MEKi-973, the results for COLO 829 are shown in Figure 12A treated with MEKi-623 and 12B treated with MEKi-973, the results for SK23-MEL are shown in Figure 19A treated with MEKi-623 and 19B treated with MEKi-973, and the results for IPC-298 are shown in Figure 22A treated with MEKi-623 and 22B treated with MEKi- 973. These results demonstrate that treatment with MEKi-623 or MEKi-973 for 24 hours appears to increase the ETBR total protein levels in all the cell lines tested, as compared to control cells to which no MEKi was added.
[0372] To determine whether the observed increases in total ETBR protein level due to MEKi treatment also results in an increase of ETBR surface protein levels, a fluorescence-activated cell sorting (FACS) analysis was performed as described above. Cells were harvested in PBS with 2.5 mmol/L EDTA and washed in PBS buffer containing 1% FBS. All subsequent steps were carried out at 4°C. Cells were incubated for 1 hour with 3μg/mL anti ETBR antibody Hu5E9vl, followed by anti-human IgG fluorescent detection reagent (Al 1013; Invitrogen). Cells were then analyzed with a FACS Calibur flow cytometer (BD Biosciences). The results for A2058 are shown in Figure 16A-F, results for COL0829 are shown in Figure 13A-F, the results for SK23-MEL are shown in Figure 20A-F and the results for IPC- 298 are shown in Figure 23A-F. These results demonstrate that treatment with MEKi-623 or MEKi-973 for 24h appears to increase the surface levels of ETBR protein expressed in all cell lines tested, as compared to control cells to which no MEK inhibitor was added.
[0373] Example 8: Effect of MEK inhibitor drugs on in vivo efficacy of anti-ETBR ADC
[0374] Given the results demonstrated in Example 7 above, the impact of the MEK inhibitors described herein on the in vivo efficacy of an anti-ETBR ADC was tested. To do this, the in vivo efficacy for various combinations of Hu5E9vl -ADC and MEKi-623 and/or MEKi-973 were evaluated against A2058 and SK-MEL-23 and IPC-298 melanoma in vivo models, performed as described above in Example 4. An appropriate methylcellulose twee vehicle control (0.5% methylcellulose, 0.2% Tween-80 (MCT) or MEK inhibitor at doses of 1 mpk, 3 mpk or 7.5 mpk were administered orally once a day x 21 days beginning on study Day 0. A single 3 mpk or 6 mpk dose of Hu5E9vl-ADC or control, a histidine buffer #8, was administered intraveneously (after two doses of a MEK inhibitor) via tail vein at study Day 1.
[0375] The results are shown in Figures 17A-B, Figure 21, Figure 24 and Figure 25. Surprisingly, all combinations of the Hu5E9vl -ADC and MEK inhibitors tested demonstrated efficacy greater than the additive efficacy of either drug as a single agent alone.
[0376] Example 9: PD studies of A2058 and COLO 829 melanoma xenografts
[0377] Tumors collected at the end of studies represented in Figure 7 (COLO 829 vs combination anti- ETBR-ADC and BRAFi RG7204) did not show an increase of ETBR. This could be due to the fact that the timing of the tumor collection (day 34) was well past the wash out period of the BRAFi drug administered. In order to evaluate whether the in vitro effects of BRAFi/MEKi on cell lines, (i.e. increase of ETBR and decrease of Perk), also occurs in vivo, and therefore allows for a greater efficacy of anti- ETBR ADC and BRAFi/MEKi in combination, the following experiments were performed.
[0378] A2058 or COLO 829 tumors were grown to an average size of approximately 200 mm3, whereupon animals were randomized into groups of 5-6 each. For the BRAFi PD study, an appropriate vehicle control (Klucel LF) or RG7204 at doses of 10 mpk or 30 mpk were administered twice a day for 3 days (Figure 27A). For the MEKi PD study, an appropriate vehicle control or MEKi-973 at doses of 5mpk and lOmpk were administered orally once a day for 3 days (Figure 27B). Flash frozen tumors harvested at end of study were homogenized and processed for RNA and/or protein. Taqman assays were set up using reagents from Applied Biosystems (ABI, Foster City, CA) and assayed using 7500 Real Time PCR machine and software from ABI. Primer-probe sets were designed with primers flanking a fluorogenic probe dual labeled with Reporter dye FAM and quencher dye TAMRA. ETBR transcript levels in the tumors were normalized against transcript levels of reference genes such as Hprtl
(hypoxanthine phosphoribosyltransferase 1) or GAPDH (glyceraldehyde 3 phosphate dehydrogenase) using primer and probe sets that were specific to the human homologs of these genes.
[0379] The primer-probe set for reference gene Hprtl (hypoxanthine phosphoribosyltransferase 1) is as follows: [0380] Forward primer -5' CAC ATC AAA GAC AGC ATC TAA GAA (SEQ ID NO: 17); Reverse primer-5' CAA GTT GGA AAA TAC AGT CAA CAT T (SEQ ID NO: 18) and probe-5' TTT TGT TCT GTC CTG GAA TTA TTT TAG TAG TGT TTC A (SEQ ID NO: 19).
[0381] The primer-probe set for ETBR is as follows:
[0382] Forward primer- 5'TCA CTG AAT TCC TGC ATT AAC C (SEQ ID NO: 14), reverse primer- 5'GCA TAA GCA TGA CTT AAA GCA GTT (SEQ ID NO: 15) and probe-5' AAT TGC TCT GTA TTT GGT GAG CAA AAG ATT CAA (SEQ ID NO: 16).
[0383] The primer-probe set for reference gene GAPDH (Glyceraldehyde 3 phosphate dehydrogenase) is as follows:
[0384] Forward primer -5' GAA GAT GGT GAT GGG ATT TC (SEQ ID NO:20), Reverse primer-5' GAA GGT GAA GGT CGG AGT C (SEQ ID NO:21), and probe-5' CAA GCT TCC CGT TCT CAG CC (SEQ ID NO:22).
[0385] Figure 27A shows that BRAFi induces ETBR niRNA in vivo as compared to control vehicle. Figure 27B shows that MEKi-973 induces ETBR mRNA in vivo as compared to control vehicle as well.
[0386] Phosphorylated erk and total erk protein levels were evaluated in the tumors by western blotting using the following reagents: for detection of proteins: anti-Phospho-p44/42 MAPK (Erkl/2)
(Thr202/Tyr204) antibody (9101, Cell Signaling Technology), anti-p44/42 MAPK (Erkl/2) antibody (9102, Cell Signaling Technology) and mouse monoclonal anti- β-Tubulin antibody (556321, BD Pharmingen) as control (Figure 26). Here, BRAFi appears to inhibit Perk in vivo as compared to control.
TABLE 3: PERCENT TUMOR GROWTH INHIBITION (TGI) SUMMARY
Figure imgf000079_0001
[0387] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, the descriptions and examples should not be construed as limiting the scope of the invention. The disclosures of all patent and scientific literature cited herein are expressly incorporated in their entirety by reference.

Claims

WHAT IS CLAIMED IS:
1. A method of tumor growth inhibition (TGI) in a subject suffering from melanoma comprising
administering to the subject an effective amount of an anti-endothelin B receptor (ETBR) antibody in combination with an effective amount of a MAP kinase inhibitor.
2. The method of claim 1, wherein said combination is synergistic.
3. The method of claim 1, wherein said TGI is greater than the TGI seen using an anti-ETBR antibody alone.
4. The method of claim 1, wherein said TGI is greater than the TGI seen using a MAP kinase inhibitor alone.
5. The method of claim 3, wherein the TGI is about 10% greater, or about 15%> greater, or about 20%> greater, or about 25%> greater, or about 30%> greater, or about 35%> greater, or about 40%> greater, or about 45%o greater, or about 50%> greater, or about 55%> greater, or about 60%> greater, or about 65%> greater, or about 70%> greater than use of an anti-ETBR antibody alone.
6. The method of claim 4, wherein the TGI is about 10%> greater, or about 15%> greater, or about 20%> greater, or about 25%> greater, or about 30%> greater, or about 35%> greater, or about 40%> greater, or about 45%o greater, or about 50%> greater, or about 55%> greater, or about 60%> greater, or about 65%> greater, or about 70%> greater than use of a MAP kinase inhibitor alone.
7. The method of claim 1, wherein said anti-ETBR antibody specifically binds an ETBR epitope
consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
8. The method of claim 1, wherein said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDR1 is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDR1 is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
9. The method of claim 1, wherein said anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9.
10. The method of claim 9, wherein said VL is SEQ ID NO:8.
11. The method of claim 1 , wherein said anti-ETBR antibody is conjugated to a cytotoxin.
12. The method of claim 11, wherein said cytotoxin is cytotoxic agent is selected from the group
consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes.
13. The method of claim 12, wherein said cytotoxin is a toxin.
14. The method of claim 13, wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin.
15. The method of claim 14, wherein said toxin is a maytansinoid.
16. The method of claim 1, wherein said MAP kinase inhibitor is a BRAF inhibitor.
17. The method of claim 16, wherein said BRAF inhibitor is propane-1 -sulfonic acid {3-[5-(4- chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl} -amide.
18. The method of claim 16, wherein said BRAF inhibitor has the following chemical structure:
Figure imgf000082_0001
19. The method of claim 1, wherein said MAP kinase inhibitor is a MEK inhibitor.
20. The method of claim 19, wherein said MEK inhibitor is (S)-(3,4-difuoro-2-((2-fluoro-4- iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l -yl)methanone.
21. The method of claim 19, wherein said MEK inhibitor has the following chemical structure:
Figure imgf000082_0002
22. A method of treating melanoma comprising administering to a subject in need thereof a
therapeutically effective amount of a MAP kinase inhibitor and an anti-ETBR antibody.
23. The method of claim 22, wherein said combination is synergistic.
24. The method of claim 22, wherein said TGI is greater than the TGI seen using an anti-ETBR
antibody alone.
25. The method of claim 22, wherein said TGI is greater than the TGI seen using a MAP kinase
inhibitor alone.
26. The method of claim 24, wherein the TGI is about 10% greater, or about 15%> greater, or about 20%> greater, or about 25%> greater, or about 30%> greater, or about 35%> greater, or about 40%> greater, or about 45%o greater, or about 50%> greater, or about 55%> greater, or about 60%> greater, or about 65%> greater, or about 70%> greater than use of an anti-ETBR antibody alone.
27. The method of claim 25, wherein the TGI is about 10%> greater, or about 15%> greater, or about 20%> greater, or about 25%> greater, or about 30%> greater, or about 35%> greater, or about 40%> greater, or about 45%o greater, or about 50%> greater, or about 55%> greater, or about 60%> greater, or about 65%> greater, or about 70%> greater than use of a MAP kinase inhibitor alone.
28. The method of claim 22, wherein said anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
29. The method of claim 22, wherein said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDRl is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDRl is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
30. The method of claim 22, wherein said anti-ETBR antibody has a variable heavy chain and a
variable light chain, wherein said VH is SEQ ID NO:7 or 9.
31. The method of claim 30, wherein said VL is SEQ ID NO: 8.
32. The method of claim 22, wherein said anti-ETBR antibody is conjugated to a cytotoxin.
33. The method of claim 32, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes.
34. The method of claim 33, wherein said cytotoxin is a toxin.
35. The method of claim 34, wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin.
36. The method of claim 35, wherein said toxin is a maytansinoid.
37. The method of claim 22, wherein said MAP kinase inhibitor is a BRAF inhibitor.
38. The method of claim 37, wherein said BRAF inhibitor is propane-1 -sulfonic acid {3-[5-(4- chlorophenyl)- 1 H-pyrrolo [2,3 -b]pyridine-3 -carbonyl] -2,4-difluoro-phenyl} -amide.
39. The method of claim 37, wherein said BRAF inhibitor has the following chemical structure:
Figure imgf000083_0001
40. The method of claim 22, wherein said MAP kinase inhibitor is a MEK inhibitor.
41. The method of claim 40, wherein said MEK inhibitor is (S)-(3,4-difuoro-2-((2-fluoro-4- iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l -yl)methanone.
42. Th method of claim 40, wherein said MEK inhibitor has the following chemical structure:
Figure imgf000083_0002
43. The method of claim 22, wherein said melanoma is ETBR positive.
44. The method of claim 22, wherein said melanoma is metastatic.
45. The method of claim 22, wherein said subject has not had prior therapy with a MAP kinase
inhibitor.
46. The method of claim 22, wherein said subject has a V600E BRAF gene mutation.
47. The method of claim 22, wherein said subject is V600E wildtype.
48. The method of claim 22, wherein the MAP kinase inhibitor is administered first to said subject in need thereof.
49. The method of claim 22, wherein said anti-ETBR antibody is administered after administration of said MAP kinase inhibitor.
50. The method of claim 22, wherein the anti-ETBR antibody and the MAP kinase inhibitor are administered simultaneously.
51. The method of claim 22, wherein the anti-ETBR antibody and the MAP kinase inhibitor are administered sequentially.
52. The method of claim 51, wherein the anti-ETBR antibody is administered to the subject first.
53. The method of claim 52, wherein the MAP kinase inhibitor is administered to the subject after administration of the anti-ETBR antibody.
54. The method of claim 51, wherein the MAP kinase inhibitor is administered to the subject first.
55. The method of claim 54, wherein the anti-ETBR antibody is administered to the subject after administration of the MAP kinase inhibitor.
56. The method of claim 22, wherein said anti-ETBR antibody is administered intraveneously.
57. The method of claim 22, wherein said anti-ETBR antibody is dosed at about 0.1 mpk, or about 0.2 mpk, or about 0.3 mpk, or about 0.5 mpk, or about 1 mpk, or about 5 mpk, or about 10 mpk, or about 15 mpk, or about 20 mpk, or about 25 mpk, or about 30 mpk.
58. The method of claim 22, wherein the MAP kinase inhibitor is administered orally.
59. The method of claim 22, wherein the MAP kinase inhibitor is dosed at about 1 mpk, or about 2 mpk, or about 3 mpk, or about 4 mpk, or about 5 mpk, or about 6 mpk, or about 7 mpk, or about 8 mpk, or about 9 mpk, or about 10 mpk, or about 11 mpk, or about 12 mpk, or about 15 mpk, or about 20 mpk or about 30 mpk.
60. An article of manufacture for TGI in a subject suffering from melanoma comprising a package comprising an anti-ETBR antibody composition and a MAP kinase inhibitor composition.
61. An article of manufacture for treating melanoma in a subject comprising a package comprising an anti-ETBR antibody composition and a MAP kinase inhibitor composition.
62. The article of manufacture of claim 60 or 61, wherein said anti-ETBR antibody specifically binds an ETBR epitope consisting of amino acids number 64 to 101 of SEQ ID NO: 10.
63. The article of manufacture of claim 60 or 61, wherein said anti-ETBR antibody has three variable heavy chain CDRs and three variable light chain CDRs wherein VH CDR1 is SEQ ID NO: l, VH CDR2 is SEQ ID NO:2, VH CDR3 is SEQ ID NO:3 and wherein VL CDR1 is SEQ ID NO:4, VL CDR2 is SEQ ID NO:5, VL CDR3 is SEQ ID NO:6.
64. The article of manufacture of claim 60 or 61, wherein said anti-ETBR antibody has a variable heavy chain and a variable light chain, wherein said VH is SEQ ID NO:7 or 9.
65. The article of manufacture of claim 64, wherein said VL is SEQ ID NO: 8.
66. The article of manufacture of claim 60 or 61, wherein said anti-ETBR antibody is conjugated to a cytotoxin.
67. The article of manufacture of claim 66, wherein said cytotoxin is cytotoxic agent is selected from the group consisting of toxins, antibiotics, radioactive isotopes and nucleolytic enzymes.
68. The article of manufacture of claim 67, wherein said cytotoxin is a toxin.
69. The article of manufacture of claim 68, wherein said toxin is selected from the group consisting of maytansinoid, calicheamicin and auristatin.
70. The article of manufacture of claim 69, wherein said toxin is a maytansinoid.
71. The article of manufacture of claim 60 or 61, wherein said MAP kinase inhibitor is a BRAF
inhibitor.
72. The article of manufacture of claim 71, wherein said BRAF inhibitor is propane- 1 -sulfonic acid {3-[5-(4-chlorophenyl)-lH-pyrrolo[2,3-b]pyridine-3-carbonyl]-2,4-difluoro-phenyl} -amide.
73. The article of manufacture of claim 71, wherein said BRAF inhibitor has the following chemical structure:
Figure imgf000085_0001
74. The article of manufacture of claim 60 or 61, wherein said MAP kinase inhibitor is a MEK
inhibitor.
75. The article of manufacture of claim 74, wherein said MEK inhibitor is (S)-(3,4-difuoro-2-((2- fluoro-4-iodophenyl)amino)phenyl)(3-hydoxy-3-(piperidin-2yl)azetidin-l-yl)methanone.
76. The article of manufacture of claim 74, wherein said MEK inhibitor has the following chemical structure:
Figure imgf000085_0002
Use of an article of manufacture of claim 60 or 61 in the preparation of a medicament for TGI of a melanoma.
PCT/US2012/061533 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma WO2013063001A1 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
US14/354,362 US20140341916A1 (en) 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma
SG11201401815XA SG11201401815XA (en) 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma
KR1020147013878A KR20140097205A (en) 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma
AU2012328980A AU2012328980A1 (en) 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma
CA2850034A CA2850034A1 (en) 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma
EA201490879A EA201490879A1 (en) 2011-10-28 2012-10-24 THERAPEUTIC COMBINATIONS AND METHODS OF TREATING MELANOMA
JP2014538892A JP6251682B2 (en) 2011-10-28 2012-10-24 Combinations and methods of treatment for melanoma treatment
CN201280065175.XA CN104039340B (en) 2011-10-28 2012-10-24 Treat melanomatous method and therapeutic combination
BR112014009953A BR112014009953A2 (en) 2011-10-28 2012-10-24 tumor growth inhibition method, melanoma treatment, industrialized article and use
EP12842985.9A EP2776051A4 (en) 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma
IN3062CHN2014 IN2014CN03062A (en) 2011-10-28 2012-10-24
MX2014004991A MX2014004991A (en) 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma.
IL232135A IL232135A0 (en) 2011-10-28 2014-04-22 Therapeutic combinations and methods of treating melanoma
MA37028A MA35645B1 (en) 2011-10-28 2014-05-13 Therapeutic associations and methods of treating melanoma

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161552893P 2011-10-28 2011-10-28
US61/552,893 2011-10-28
US201261678978P 2012-08-02 2012-08-02
US61/678,978 2012-08-02

Publications (1)

Publication Number Publication Date
WO2013063001A1 true WO2013063001A1 (en) 2013-05-02

Family

ID=48168400

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/061533 WO2013063001A1 (en) 2011-10-28 2012-10-24 Therapeutic combinations and methods of treating melanoma

Country Status (18)

Country Link
EP (1) EP2776051A4 (en)
JP (2) JP6251682B2 (en)
KR (1) KR20140097205A (en)
CN (1) CN104039340B (en)
AR (1) AR088509A1 (en)
AU (1) AU2012328980A1 (en)
BR (1) BR112014009953A2 (en)
CA (1) CA2850034A1 (en)
CL (1) CL2014001092A1 (en)
CO (1) CO7151543A2 (en)
EA (1) EA201490879A1 (en)
IL (1) IL232135A0 (en)
IN (1) IN2014CN03062A (en)
MX (1) MX2014004991A (en)
PE (1) PE20142312A1 (en)
SG (1) SG11201401815XA (en)
TW (1) TW201325613A (en)
WO (1) WO2013063001A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9216170B2 (en) 2012-03-19 2015-12-22 Hoffmann-La Roche Inc. Combination therapy for proliferative disorders
WO2016055907A1 (en) * 2014-10-10 2016-04-14 Pfizer Inc. Synergistic auristatin combinations
US9724413B2 (en) 2011-08-01 2017-08-08 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
WO2017220739A1 (en) 2016-06-24 2017-12-28 Commissariat A L'energie Atomique Et Aux Energies Alternatives Antibody directed against the endothelin receptor beta sub-type
US10946093B2 (en) 2014-07-15 2021-03-16 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2994544C (en) * 2015-08-03 2021-03-30 Enb Therapeutics, Llc Compositions and methods for treating cancers associated with etbr activation

Citations (148)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US633410A (en) 1898-09-22 1899-09-19 George A Ames Ice-cutter.
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US3896111A (en) 1973-02-20 1975-07-22 Research Corp Ansa macrolides
US4137230A (en) 1977-11-14 1979-01-30 Takeda Chemical Industries, Ltd. Method for the production of maytansinoids
US4151042A (en) 1977-03-31 1979-04-24 Takeda Chemical Industries, Ltd. Method for producing maytansinol and its derivatives
US4248870A (en) 1978-10-27 1981-02-03 Takeda Chemical Industries, Ltd. Maytansinoids and use
US4256746A (en) 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
US4260608A (en) 1978-11-14 1981-04-07 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and methods of use thereof
WO1981001145A1 (en) 1979-10-18 1981-04-30 Univ Illinois Hydrolytic enzyme-activatible pro-drugs
US4265814A (en) 1978-03-24 1981-05-05 Takeda Chemical Industries Matansinol 3-n-hexadecanoate
US4294757A (en) 1979-01-31 1981-10-13 Takeda Chemical Industries, Ltd 20-O-Acylmaytansinoids
US4307016A (en) 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4308268A (en) 1979-06-11 1981-12-29 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and method of use thereof
US4308269A (en) 1979-06-11 1981-12-29 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and method of use thereof
US4309428A (en) 1979-07-30 1982-01-05 Takeda Chemical Industries, Ltd. Maytansinoids
US4313946A (en) 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4315929A (en) 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
US4317821A (en) 1979-06-08 1982-03-02 Takeda Chemical Industries, Ltd. Maytansinoids, their use and pharmaceutical compositions thereof
US4322348A (en) 1979-06-05 1982-03-30 Takeda Chemical Industries, Ltd. Maytansinoids
US4331598A (en) 1979-09-19 1982-05-25 Takeda Chemical Industries, Ltd. Maytansinoids
US4362663A (en) 1979-09-21 1982-12-07 Takeda Chemical Industries, Ltd. Maytansinoid compound
US4364866A (en) 1979-09-21 1982-12-21 Takeda Chemical Industries, Ltd. Maytansinoids
US4371533A (en) 1980-10-08 1983-02-01 Takeda Chemical Industries, Ltd. 4,5-Deoxymaytansinoids, their use and pharmaceutical compositions thereof
US4424219A (en) 1981-05-20 1984-01-03 Takeda Chemical Industries, Ltd. 9-Thiomaytansinoids and their pharmaceutical compositions and use
US4450254A (en) 1980-11-03 1984-05-22 Standard Oil Company Impact improvement of high nitrile resins
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4970198A (en) 1985-10-17 1990-11-13 American Cyanamid Company Antitumor antibiotics (LL-E33288 complex)
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5053394A (en) 1988-09-21 1991-10-01 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
US5079233A (en) 1987-01-30 1992-01-07 American Cyanamid Company N-acyl derivatives of the LL-E33288 antitumor antibiotics, composition and methods for using the same
WO1993001161A1 (en) 1991-07-11 1993-01-21 Pfizer Limited Process for preparing sertraline intermediates
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1993021232A1 (en) 1992-04-10 1993-10-28 Research Development Foundation IMMUNOTOXINS DIRECTED AGAINST c-erbB-2 (HER-2/neu) RELATED SURFACE ANTIGENS
WO1994011026A2 (en) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
US5362852A (en) 1991-09-27 1994-11-08 Pfizer Inc. Modified peptide derivatives conjugated at 2-hydroxyethylamine moieties
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
EP0425235B1 (en) 1989-10-25 1996-09-25 Immunogen Inc Cytotoxic agents comprising maytansinoids and their therapeutic use
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
WO1997030087A1 (en) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation of glycosylated antibodies
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
WO1997038731A1 (en) 1996-04-18 1997-10-23 The Regents Of The University Of California Immunoliposomes that optimize internalization into target cells
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5739116A (en) 1994-06-03 1998-04-14 American Cyanamid Company Enediyne derivatives useful for the synthesis of conjugates of methyltrithio antitumor agents
US5750373A (en) 1990-12-03 1998-05-12 Genentech, Inc. Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
WO1998058964A1 (en) 1997-06-24 1998-12-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
WO1999022764A1 (en) 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
WO1999051642A1 (en) 1998-04-02 1999-10-14 Genentech, Inc. Antibody variants and fragments thereof
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
WO2001029246A1 (en) 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
US6313276B1 (en) * 1991-07-12 2001-11-06 Shionogi Sieyaku Kabushiki Kaisha Human endothelin receptor
WO2002031140A1 (en) 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
WO2002088172A2 (en) 2001-04-30 2002-11-07 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US6630579B2 (en) 1999-12-29 2003-10-07 Immunogen Inc. Cytotoxic agents comprising modified doxorubicins and daunorubicins and their therapeutic use
WO2003085107A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
WO2003084570A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. DRUG CONTAINING ANTIBODY COMPOSITION APPROPRIATE FOR PATIENT SUFFERING FROM FcϜRIIIa POLYMORPHISM
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
EP1391213A1 (en) 2002-08-21 2004-02-25 Boehringer Ingelheim International GmbH Compositions and methods for treating cancer using maytansinoid CD44 antibody immunoconjugates and chemotherapeutic agents
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
WO2004056312A2 (en) 2002-12-16 2004-07-08 Genentech, Inc. Immunoglobulin variants and uses thereof
US20050014934A1 (en) 2002-10-15 2005-01-20 Hinton Paul R. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
US20050016993A1 (en) 2003-04-17 2005-01-27 Koskey James Donald Heated pet mat
US20050079574A1 (en) 2003-01-16 2005-04-14 Genentech, Inc. Synthetic antibody phage libraries
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
WO2005035778A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY USING RNA INHIBITING THE FUNCTION OF α1,6-FUCOSYLTRANSFERASE
WO2005037992A2 (en) 2003-10-10 2005-04-28 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
US20050119455A1 (en) 2002-06-03 2005-06-02 Genentech, Inc. Synthetic antibody phage libraries
US20050123546A1 (en) 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
US6913748B2 (en) 2002-08-16 2005-07-05 Immunogen, Inc. Cross-linkers with high reactivity and solubility and their use in the preparation of conjugates for targeted delivery of small molecule drugs
US20050166993A1 (en) 2004-01-29 2005-08-04 Viken James P. Automatic fluid exchanger
US20050238649A1 (en) 2003-11-06 2005-10-27 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
US20050256030A1 (en) 2004-02-23 2005-11-17 Bainian Feng Heterocyclic self-immolative linkers and conjugates
US20050260186A1 (en) 2003-03-05 2005-11-24 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20050266000A1 (en) 2004-04-09 2005-12-01 Genentech, Inc. Variable domain library and uses
US20050276812A1 (en) 2004-06-01 2005-12-15 Genentech, Inc. Antibody-drug conjugates and methods
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
WO2006044908A2 (en) 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulation in histidine-acetate buffer
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
US7097840B2 (en) 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2007008848A2 (en) 2005-07-07 2007-01-18 Seattle Genetics, Inc. Monomethylvaline compounds having phenylalanine carboxy modifications at the c-terminus
WO2007008603A1 (en) 2005-07-07 2007-01-18 Seattle Genetics, Inc. Monomethylvaline compounds having phenylalanine side-chain modifications at the c-terminus
US7189826B2 (en) 1997-11-24 2007-03-13 Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
US20070061900A1 (en) 2000-10-31 2007-03-15 Murphy Andrew J Methods of modifying eukaryotic cells
US20070117126A1 (en) 1999-12-15 2007-05-24 Genentech, Inc. Shotgun scanning
US20070160598A1 (en) 2005-11-07 2007-07-12 Dennis Mark S Binding polypeptides with diversified and consensus vh/vl hypervariable sequences
US7276497B2 (en) 2003-05-20 2007-10-02 Immunogen Inc. Cytotoxic agents comprising new maytansinoids
US20070237764A1 (en) 2005-12-02 2007-10-11 Genentech, Inc. Binding polypeptides with restricted diversity sequences
US20070292936A1 (en) 2006-05-09 2007-12-20 Genentech, Inc. Binding polypeptides with optimized scaffolds
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
WO2008077546A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
US20090002360A1 (en) 2007-05-25 2009-01-01 Innolux Display Corp. Liquid crystal display device and method for driving same
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
US7527791B2 (en) 2004-03-31 2009-05-05 Genentech, Inc. Humanized anti-TGF-beta antibodies
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
US20090226925A1 (en) * 2005-05-20 2009-09-10 Grebe Stefan K G Methods for Detecting Circulating Tumor Cells
US20100003240A1 (en) * 1999-05-04 2010-01-07 New York University Cancer treatment with endothelin receptor antagonists
US20100249096A1 (en) * 2005-10-07 2010-09-30 Exelixis, Inc. Azetidines as MEK Inhibitors for the Treatment of Proliferative Diseases
US20110105521A1 (en) * 2008-07-11 2011-05-05 Novartis Ag Combination of (a) a phosphoinositide 3-kinase inhibitor and (b) a modulator of ras/raf/mek pathway
US20110206702A1 (en) * 2010-02-23 2011-08-25 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MA33974B1 (en) * 2009-10-12 2013-02-01 Hoffmann La Roche COMBINATIONS OF A PI3K INHIBITOR AND A MEK INHIBITOR

Patent Citations (160)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US633410A (en) 1898-09-22 1899-09-19 George A Ames Ice-cutter.
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US3896111A (en) 1973-02-20 1975-07-22 Research Corp Ansa macrolides
US4151042A (en) 1977-03-31 1979-04-24 Takeda Chemical Industries, Ltd. Method for producing maytansinol and its derivatives
US4137230A (en) 1977-11-14 1979-01-30 Takeda Chemical Industries, Ltd. Method for the production of maytansinoids
US4361650A (en) 1978-03-24 1982-11-30 Takeda Chemical Industries, Ltd. Fermentation process of preparing demethyl maytansinoids
US4265814A (en) 1978-03-24 1981-05-05 Takeda Chemical Industries Matansinol 3-n-hexadecanoate
US4307016A (en) 1978-03-24 1981-12-22 Takeda Chemical Industries, Ltd. Demethyl maytansinoids
US4248870A (en) 1978-10-27 1981-02-03 Takeda Chemical Industries, Ltd. Maytansinoids and use
US4256746A (en) 1978-11-14 1981-03-17 Takeda Chemical Industries Dechloromaytansinoids, their pharmaceutical compositions and method of use
US4260608A (en) 1978-11-14 1981-04-07 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and methods of use thereof
US4294757A (en) 1979-01-31 1981-10-13 Takeda Chemical Industries, Ltd 20-O-Acylmaytansinoids
US4322348A (en) 1979-06-05 1982-03-30 Takeda Chemical Industries, Ltd. Maytansinoids
US4317821A (en) 1979-06-08 1982-03-02 Takeda Chemical Industries, Ltd. Maytansinoids, their use and pharmaceutical compositions thereof
US4308269A (en) 1979-06-11 1981-12-29 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and method of use thereof
US4308268A (en) 1979-06-11 1981-12-29 Takeda Chemical Industries, Ltd. Maytansinoids, pharmaceutical compositions thereof and method of use thereof
US4309428A (en) 1979-07-30 1982-01-05 Takeda Chemical Industries, Ltd. Maytansinoids
US4331598A (en) 1979-09-19 1982-05-25 Takeda Chemical Industries, Ltd. Maytansinoids
US4362663A (en) 1979-09-21 1982-12-07 Takeda Chemical Industries, Ltd. Maytansinoid compound
US4364866A (en) 1979-09-21 1982-12-21 Takeda Chemical Industries, Ltd. Maytansinoids
WO1981001145A1 (en) 1979-10-18 1981-04-30 Univ Illinois Hydrolytic enzyme-activatible pro-drugs
US4371533A (en) 1980-10-08 1983-02-01 Takeda Chemical Industries, Ltd. 4,5-Deoxymaytansinoids, their use and pharmaceutical compositions thereof
US4450254A (en) 1980-11-03 1984-05-22 Standard Oil Company Impact improvement of high nitrile resins
US4315929A (en) 1981-01-27 1982-02-16 The United States Of America As Represented By The Secretary Of Agriculture Method of controlling the European corn borer with trewiasine
US4313946A (en) 1981-01-27 1982-02-02 The United States Of America As Represented By The Secretary Of Agriculture Chemotherapeutically active maytansinoids from Trewia nudiflora
US4424219A (en) 1981-05-20 1984-01-03 Takeda Chemical Industries, Ltd. 9-Thiomaytansinoids and their pharmaceutical compositions and use
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4970198A (en) 1985-10-17 1990-11-13 American Cyanamid Company Antitumor antibiotics (LL-E33288 complex)
US6982321B2 (en) 1986-03-27 2006-01-03 Medical Research Council Altered antibodies
US5500362A (en) 1987-01-08 1996-03-19 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5079233A (en) 1987-01-30 1992-01-07 American Cyanamid Company N-acyl derivatives of the LL-E33288 antitumor antibiotics, composition and methods for using the same
US5648260A (en) 1987-03-18 1997-07-15 Scotgen Biopharmaceuticals Incorporated DNA encoding antibodies with altered effector functions
US5624821A (en) 1987-03-18 1997-04-29 Scotgen Biopharmaceuticals Incorporated Antibodies with altered effector functions
US5770701A (en) 1987-10-30 1998-06-23 American Cyanamid Company Process for preparing targeted forms of methyltrithio antitumor agents
US5606040A (en) 1987-10-30 1997-02-25 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methyl-trithio group
US5770710A (en) 1987-10-30 1998-06-23 American Cyanamid Company Antitumor and antibacterial substituted disulfide derivatives prepared from compounds possessing a methlytrithio group
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
US5053394A (en) 1988-09-21 1991-10-01 American Cyanamid Company Targeted forms of methyltrithio antitumor agents
US6248516B1 (en) 1988-11-11 2001-06-19 Medical Research Council Single domain ligands, receptors comprising said ligands methods for their production, and use of said ligands and receptors
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
EP0404097A2 (en) 1989-06-22 1990-12-27 BEHRINGWERKE Aktiengesellschaft Bispecific and oligospecific, mono- and oligovalent receptors, production and applications thereof
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5416064A (en) 1989-10-25 1995-05-16 Immunogen, Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
EP0425235B1 (en) 1989-10-25 1996-09-25 Immunogen Inc Cytotoxic agents comprising maytansinoids and their therapeutic use
US5208020A (en) 1989-10-25 1993-05-04 Immunogen Inc. Cytotoxic agents comprising maytansinoids and their therapeutic use
US6417429B1 (en) 1989-10-27 2002-07-09 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5750373A (en) 1990-12-03 1998-05-12 Genentech, Inc. Enrichment method for variant proteins having altered binding properties, M13 phagemids, and growth hormone variants
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
US5821337A (en) 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1993001161A1 (en) 1991-07-11 1993-01-21 Pfizer Limited Process for preparing sertraline intermediates
US6313276B1 (en) * 1991-07-12 2001-11-06 Shionogi Sieyaku Kabushiki Kaisha Human endothelin receptor
US5648237A (en) 1991-09-19 1997-07-15 Genentech, Inc. Expression of functional antibody fragments
US5362852A (en) 1991-09-27 1994-11-08 Pfizer Inc. Modified peptide derivatives conjugated at 2-hydroxyethylamine moieties
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
WO1993016185A2 (en) 1992-02-06 1993-08-19 Creative Biomolecules, Inc. Biosynthetic binding protein for cancer marker
WO1993021232A1 (en) 1992-04-10 1993-10-28 Research Development Foundation IMMUNOTOXINS DIRECTED AGAINST c-erbB-2 (HER-2/neu) RELATED SURFACE ANTIGENS
WO1994011026A2 (en) 1992-11-13 1994-05-26 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human b lymphocyte restricted differentiation antigen for treatment of b cell lymphoma
US5635483A (en) 1992-12-03 1997-06-03 Arizona Board Of Regents Acting On Behalf Of Arizona State University Tumor inhibiting tetrapeptide bearing modified phenethyl amides
US5780588A (en) 1993-01-26 1998-07-14 Arizona Board Of Regents Elucidation and synthesis of selected pentapeptides
WO1994029351A2 (en) 1993-06-16 1994-12-22 Celltech Limited Antibodies
US5739116A (en) 1994-06-03 1998-04-14 American Cyanamid Company Enediyne derivatives useful for the synthesis of conjugates of methyltrithio antitumor agents
US5877296A (en) 1994-06-03 1999-03-02 American Cyanamid Company Process for preparing conjugates of methyltrithio antitumor agents
US5773001A (en) 1994-06-03 1998-06-30 American Cyanamid Company Conjugates of methyltrithio antitumor agents and intermediates for their synthesis
US5767285A (en) 1994-06-03 1998-06-16 American Cyanamid Company Linkers useful for the synthesis of conjugates of methyltrithio antitumor agents
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5663149A (en) 1994-12-13 1997-09-02 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide heterocyclic and halophenyl amides
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5714586A (en) 1995-06-07 1998-02-03 American Cyanamid Company Methods for the preparation of monomeric calicheamicin derivative/carrier conjugates
US5712374A (en) 1995-06-07 1998-01-27 American Cyanamid Company Method for the preparation of substantiallly monomeric calicheamicin derivative/carrier conjugates
US6267958B1 (en) 1995-07-27 2001-07-31 Genentech, Inc. Protein formulation
WO1997030087A1 (en) 1996-02-16 1997-08-21 Glaxo Group Limited Preparation of glycosylated antibodies
WO1997038731A1 (en) 1996-04-18 1997-10-23 The Regents Of The University Of California Immunoliposomes that optimize internalization into target cells
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
WO1998058964A1 (en) 1997-06-24 1998-12-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
WO1999022764A1 (en) 1997-10-31 1999-05-14 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
US7189826B2 (en) 1997-11-24 2007-03-13 Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
US7087409B2 (en) 1997-12-05 2006-08-08 The Scripps Research Institute Humanization of murine antibody
WO1999051642A1 (en) 1998-04-02 1999-10-14 Genentech, Inc. Antibody variants and fragments thereof
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6602684B1 (en) 1998-04-20 2003-08-05 Glycart Biotechnology Ag Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US7371826B2 (en) 1999-01-15 2008-05-13 Genentech, Inc. Polypeptide variants with altered effector function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7332581B2 (en) 1999-01-15 2008-02-19 Genentech, Inc. Polypeptide variants with altered effector function
WO2000061739A1 (en) 1999-04-09 2000-10-19 Kyowa Hakko Kogyo Co., Ltd. Method for controlling the activity of immunologically functional molecule
US20100003240A1 (en) * 1999-05-04 2010-01-07 New York University Cancer treatment with endothelin receptor antagonists
US6420548B1 (en) 1999-10-04 2002-07-16 Medicago Inc. Method for regulating transcription of foreign genes
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2001029246A1 (en) 1999-10-19 2001-04-26 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
US20070117126A1 (en) 1999-12-15 2007-05-24 Genentech, Inc. Shotgun scanning
US6630579B2 (en) 1999-12-29 2003-10-07 Immunogen Inc. Cytotoxic agents comprising modified doxorubicins and daunorubicins and their therapeutic use
US7097840B2 (en) 2000-03-16 2006-08-29 Genentech, Inc. Methods of treatment using anti-ErbB antibody-maytansinoid conjugates
US20060025576A1 (en) 2000-04-11 2006-02-02 Genentech, Inc. Multivalent antibodies and uses therefor
US20030115614A1 (en) 2000-10-06 2003-06-19 Yutaka Kanda Antibody composition-producing cell
US20020164328A1 (en) 2000-10-06 2002-11-07 Toyohide Shinkawa Process for purifying antibody
WO2002031140A1 (en) 2000-10-06 2002-04-18 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions
US20070061900A1 (en) 2000-10-31 2007-03-15 Murphy Andrew J Methods of modifying eukaryotic cells
US7041870B2 (en) 2000-11-30 2006-05-09 Medarex, Inc. Transgenic transchromosomal rodents for making human antibodies
WO2002088172A2 (en) 2001-04-30 2002-11-07 Seattle Genetics, Inc. Pentapeptide compounds and uses related thereto
US6441163B1 (en) 2001-05-31 2002-08-27 Immunogen, Inc. Methods for preparation of cytotoxic conjugates of maytansinoids and cell binding agents
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20030157108A1 (en) 2001-10-25 2003-08-21 Genentech, Inc. Glycoprotein compositions
US20040093621A1 (en) 2001-12-25 2004-05-13 Kyowa Hakko Kogyo Co., Ltd Antibody composition which specifically binds to CD20
US20040132140A1 (en) 2002-04-09 2004-07-08 Kyowa Hakko Kogyo Co., Ltd. Production process for antibody composition
WO2003085107A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. Cells with modified genome
WO2003084570A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. DRUG CONTAINING ANTIBODY COMPOSITION APPROPRIATE FOR PATIENT SUFFERING FROM FcϜRIIIa POLYMORPHISM
WO2003085119A1 (en) 2002-04-09 2003-10-16 Kyowa Hakko Kogyo Co., Ltd. METHOD OF ENHANCING ACTIVITY OF ANTIBODY COMPOSITION OF BINDING TO FcϜ RECEPTOR IIIa
US20040110704A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells of which genome is modified
US20040109865A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Antibody composition-containing medicament
US20040110282A1 (en) 2002-04-09 2004-06-10 Kyowa Hakko Kogyo Co., Ltd. Cells in which activity of the protein involved in transportation of GDP-fucose is reduced or lost
US20050119455A1 (en) 2002-06-03 2005-06-02 Genentech, Inc. Synthetic antibody phage libraries
US6913748B2 (en) 2002-08-16 2005-07-05 Immunogen, Inc. Cross-linkers with high reactivity and solubility and their use in the preparation of conjugates for targeted delivery of small molecule drugs
EP1391213A1 (en) 2002-08-21 2004-02-25 Boehringer Ingelheim International GmbH Compositions and methods for treating cancer using maytansinoid CD44 antibody immunoconjugates and chemotherapeutic agents
US20050014934A1 (en) 2002-10-15 2005-01-20 Hinton Paul R. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
WO2004056312A2 (en) 2002-12-16 2004-07-08 Genentech, Inc. Immunoglobulin variants and uses thereof
US20050079574A1 (en) 2003-01-16 2005-04-14 Genentech, Inc. Synthetic antibody phage libraries
US20050260186A1 (en) 2003-03-05 2005-11-24 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US20050016993A1 (en) 2003-04-17 2005-01-27 Koskey James Donald Heated pet mat
US7276497B2 (en) 2003-05-20 2007-10-02 Immunogen Inc. Cytotoxic agents comprising new maytansinoids
WO2005035586A1 (en) 2003-10-08 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Fused protein composition
WO2005035778A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. PROCESS FOR PRODUCING ANTIBODY COMPOSITION BY USING RNA INHIBITING THE FUNCTION OF α1,6-FUCOSYLTRANSFERASE
WO2005037992A2 (en) 2003-10-10 2005-04-28 Immunogen, Inc. Method of targeting specific cell populations using cell-binding agent maytansinoid conjugates linked via a non-cleavable linker, said conjugates, and methods of making said conjugates
US20050123546A1 (en) 2003-11-05 2005-06-09 Glycart Biotechnology Ag Antigen binding molecules with increased Fc receptor binding affinity and effector function
US7498298B2 (en) 2003-11-06 2009-03-03 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
US20050238649A1 (en) 2003-11-06 2005-10-27 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2005053742A1 (en) 2003-12-04 2005-06-16 Kyowa Hakko Kogyo Co., Ltd. Medicine containing antibody composition
US20050166993A1 (en) 2004-01-29 2005-08-04 Viken James P. Automatic fluid exchanger
US20050256030A1 (en) 2004-02-23 2005-11-17 Bainian Feng Heterocyclic self-immolative linkers and conjugates
US7527791B2 (en) 2004-03-31 2009-05-05 Genentech, Inc. Humanized anti-TGF-beta antibodies
US20050266000A1 (en) 2004-04-09 2005-12-01 Genentech, Inc. Variable domain library and uses
WO2005100402A1 (en) 2004-04-13 2005-10-27 F.Hoffmann-La Roche Ag Anti-p-selectin antibodies
US20050276812A1 (en) 2004-06-01 2005-12-15 Genentech, Inc. Antibody-drug conjugates and methods
WO2006029879A2 (en) 2004-09-17 2006-03-23 F.Hoffmann-La Roche Ag Anti-ox40l antibodies
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2006044908A2 (en) 2004-10-20 2006-04-27 Genentech, Inc. Antibody formulation in histidine-acetate buffer
US20090226925A1 (en) * 2005-05-20 2009-09-10 Grebe Stefan K G Methods for Detecting Circulating Tumor Cells
WO2007008848A2 (en) 2005-07-07 2007-01-18 Seattle Genetics, Inc. Monomethylvaline compounds having phenylalanine carboxy modifications at the c-terminus
WO2007008603A1 (en) 2005-07-07 2007-01-18 Seattle Genetics, Inc. Monomethylvaline compounds having phenylalanine side-chain modifications at the c-terminus
US20100249096A1 (en) * 2005-10-07 2010-09-30 Exelixis, Inc. Azetidines as MEK Inhibitors for the Treatment of Proliferative Diseases
US20070160598A1 (en) 2005-11-07 2007-07-12 Dennis Mark S Binding polypeptides with diversified and consensus vh/vl hypervariable sequences
US20070237764A1 (en) 2005-12-02 2007-10-11 Genentech, Inc. Binding polypeptides with restricted diversity sequences
US20070292936A1 (en) 2006-05-09 2007-12-20 Genentech, Inc. Binding polypeptides with optimized scaffolds
US20080069820A1 (en) 2006-08-30 2008-03-20 Genentech, Inc. Multispecific antibodies
WO2008077546A1 (en) 2006-12-22 2008-07-03 F. Hoffmann-La Roche Ag Antibodies against insulin-like growth factor i receptor and uses thereof
US20090002360A1 (en) 2007-05-25 2009-01-01 Innolux Display Corp. Liquid crystal display device and method for driving same
WO2009089004A1 (en) 2008-01-07 2009-07-16 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
US20110105521A1 (en) * 2008-07-11 2011-05-05 Novartis Ag Combination of (a) a phosphoinositide 3-kinase inhibitor and (b) a modulator of ras/raf/mek pathway
US20110206702A1 (en) * 2010-02-23 2011-08-25 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor

Non-Patent Citations (175)

* Cited by examiner, † Cited by third party
Title
ALLEY, S.C. ET AL.: "Controlling the location of drug attachment in antibody-drug conjugates", AMERICAN ASSOCIATION FOR CANCER RESEARCH, 2004 ANNUAL MEETING, MARCH 27-31, 2004, PROCEEDINGS OF THE AACR, vol. 45, 27 March 2004 (2004-03-27)
ALMAGRO; FRANSSON, FRONT. BIOSCI., vol. 13, 2008, pages 1619 - 1633
AMSBERRY ET AL., J. ORG. CHEM., vol. 55, 1990, pages 5867
ASCIERTO PA ET AL.: "Melanoma: a model for testing new agents in combination therapies", J TRANSL MED, vol. 8, 2010, pages 38 - 45
BACA ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 10678 - 10684
BALDWIN ET AL., LANCET, 15 March 1986 (1986-03-15), pages 603 - 05
BEERAM ET AL.: "A phase I study of trastuzumab-MCC-DMl (T-DM1), a first-in-class HER2 antibody-drug conjugate (ADC), in patients (pts) with HER2+ metastatic breast cancer (BC", AMERICAN SOCIETY OF CLINICAL ONCOLOGY 43RD, 2 June 2007 (2007-06-02)
BOERNER ET AL., J. IMMUNOL., vol. 147, 1991, pages 86
BRENNAN ET AL., SCIENCE, vol. 229, no. 81, 1985
BRODEUR ET AL.: "Monoclonal Antibody Production Techniques and Applications", 1987, MARCEL DEKKER, INC., pages: 51 - 63
BRUGGEMANN, M. ET AL., J. EXP. MED., vol. 166, 1987, pages 1351 - 1361
CARLSSON ET AL., BIOCHEM. J., vol. 173, 1978, pages 723 - 737
CARTER ET AL., PROC. NATL. ACAD. SCI. USA, vol. , 89, 1992, pages 4285
CHARI ET AL., CANCER RES., vol. 52, 1992, pages 127 - 131
CHARI ET AL., CANCER RESEARCH, vol. 52, 1992, pages 127 - 131
CHARI, R.V.J. ET AL.: "Immunoconjugates Containing Novel Maytansinoids: Promising Anticancer Drugs.", CANCER RES., vol. 52, no. 1, 1992, pages 127 - 131, XP000453560 *
CHARLTON: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, pages: 245 - 254
CHEN ET AL., J. MOL. BIOL., vol. 293, 1999, pages 865 - 881
CHOTHIA; LESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CHOWDHURY, METHODS MOL. BIOL., vol. 207, 2008, pages 179 - 196
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
CLARKSON ET AL., NATURE, vol. 352, 1991, pages 624 - 628
CLYNES ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 95, 1998, pages 652 - 656
CRAGG, M.S. ET AL., BLOOD, vol. 101, 2003, pages 1045 - 1052
CRAGG, M.S.; M.J. GLENNIE, BLOOD, vol. 103, 2004, pages 2738 - 2743
CUNNINGHAM; WELLS, SCIENCE, vol. 244, 1989, pages 1081 - 1085
DALL' ACQUA ET AL., METHODS, vol. 36, 2005, pages 43 - 60
DORONINA ET AL., BIOCONJ. CHEM., vol. 17, 2006, pages 114 - 124
DORONINA ET AL., BIOCONJUGATE CHEM., vol. 17, 2006, pages 114 - 124
DORONINA ET AL., NAT. BIOTECH., vol. 21, 2003, pages 778 - 784
DORONINA ET AL., NAT. BIOTECHNOL., vol. 21, 2003, pages 778 - 784
DORONINA ET AL., NATURE BIOTECHNOL., vol. 21, no. 7, 2003, pages 778 - 784
DORONINA, NAT. BIOTECHNOL., vol. 21, no. 7, 2003, pages 778 - 784
DRUGS OF THE FUTURE, vol. 25, no. 7, 2000, pages 686
DUBOWCHIK ET AL., BIOORG. & MED. CHEM. LETTERS, vol. 12, 2002, pages 1529 - 1532
DUNCAN; WINTER, NATURE, vol. 322, 1988, pages 738 - 40
E. SCHRODER; K. LUBKE: "The Peptides", vol. 1, 1965, ACADEMIC PRESS, pages: 76 - 136
EPSTEIN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 3688
ERICKSON ET AL., CANCER RES., vol. 66, no. 8, 2006, pages 1 - 8
FELLOUSE, PROC. NATL. ACAD. SCI. USA, vol. 101, no. 34, 2004, pages 12467 - 12472
FLATMAN ET AL., J. CHROMATOGR. B, vol. 848, 2007, pages 79 - 87
FRAKER ET AL., BIOCHEM. BIOPHYS. RES. COMMUN., vol. 80, 1978, pages 49 - 57
GABIZON ET AL., J. NATIONAL CANCER INST., vol. 81, no. 19, 1989, pages 1484
GAZZANO-SANTORO ET AL., J. IMMUNOL. METHODS, vol. 202, 1996, pages 163
GEOGHEGAN; STROH, BIOCONJUGATE CHEM., vol. 3, 1992, pages 138 - 146
GERNGROSS, NAT. BIOTECH., vol. 22, 2004, pages 1409 - 1414
GRAHAM ET AL., J. GEN VIROL., vol. 36, 1977, pages 59
GRIFFITHS ET AL., EMBO J, vol. 12, 1993, pages 725 - 734
GRUBER ET AL., J. IMMUNOL., vol. 152, 1994, pages 5368
GUYER ET AL., J. IMMUNOL., vol. 117, 1976, pages 587
HALABAN R ET AL.: "PLX4032, a Selective BRAF (V600E) Kinase Inhibitor, Activates the ERK Pathway and Enhances Cell Migration and Proliferation of BRAF(WT) Melanoma Cells", PIGMENT CELL MELANOMA RES, vol. 23, no. 2, February 2010 (2010-02-01), pages 190 - 200
HAMANN ET AL., EXPERT OPIN. THER. PATENTS, vol. 15, 2005, pages 1087 - 1103
HAMBLETT ET AL., CLIN. CANCER RES., vol. 10, 2004, pages 7063 - 7070
HAMBLETT, K.J. ET AL.: "Effect of drug loading on the pharmacology, pharmacokinetics, and toxicity of an anti-CD30 antibody-drug conjugate", AMERICAN ASSOCIATION FOR CANCER RESEARCH, 2004 ANNUAL MEETING, MARCH 27-31, 2004, PROCEEDINGS OF THE AACR, vol. 45, 27 March 2004 (2004-03-27)
HARLOW; LANE: "Antibodies: A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY, article "ch.14"
HATZIVASSILIOU G ET AL.: "RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth.", NATURE, vol. 464, no. 7287, pages 431 - 5, XP055126892, DOI: doi:10.1038/nature08833
HAY ET AL., BIOORG. MED. CHEM. LETT., vol. 9, 1999, pages 2237
HELLSTROM, I ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 82, 1985, pages 1499 - 1502
HELLSTROM, I. ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 83, 1986, pages 7059 - 7063
HINMAN ET AL., CANCER RES., vol. 53, 1993, pages 3336 - 3342
HINMAN ET AL., CANCER RESEARCH, vol. 53, 1993, pages 3336 - 3342
HOLLINGER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 90, 1993, pages 6444 - 6448
HOOGENBOOM ET AL., METHODS IN MOLECULAR BIOLOGY, vol. 178, pages 1 - 37
HOOGENBOOM ET AL.: "Methods in Molecular Biology", vol. 178, 2001, HUMAN PRESS, pages: 1 - 37
HOOGENBOOM; WINTER, J. MOL. BIOL., vol. 227, 1992, pages 381 - 388
HUDSON ET AL., NAT. MED., vol. 9, 2003, pages 129 - 134
HWANG ET AL., PROC. NATL ACAD. SCI. USA, vol. 77, 1980, pages 4030
IDUSOGIE ET AL., J. IMMUNOL., vol. 164, 2000, pages 4178 - 4184
JEFFREY ET AL., BIOORGANIC & MED. CHEM. LETTERS, vol. 16, 2006, pages 358 - 362
JEFFREY ET AL., J. MED. CHEM., vol. 48, 2005, pages 1344 - 1358
JOHNSTON S.: "XL518, a potent, selective, orally bioavailable MEK1 inhibitor, downregulates the Ras/Raf/MEKJERK pathway in vivo, resulting in tumor growth inhibition and regression in preclinical models", AACR-NCI-EORTC SYMPOSIUM ON MOLECULAR TARGETS AND CANCER THERAPEUTICS, 22 October 2007 (2007-10-22)
KABAT ET AL.: "Sequences of Proteins of Immunological Interest, 5th Ed.", 1991, PUBLIC HEALTH SERVICE, NATIONAL INSTITUTES OF HEALTH
KABAT ET AL.: "Sequences of Proteins of Immunological Interest, Fifth Edition,", vol. 1-3, 1991, NIH PUBLICATION, pages: 91 - 3242
KAM ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 11600 - 11605
KANDA, Y. ET AL., BIOTECHNOL. BIOENG., vol. 94, no. 4, 2006, pages 680 - 688
KASHMIRI ET AL., METHODS, vol. 36, 2005, pages 25 - 34
KIM ET AL., J. IMMUNOL., vol. 24, 1994, pages 249
KINDT ET AL.: "Kuby Immunology", 2007, W.H. FREEMAN AND CO, pages: 91
KING ET AL., J. MED. CHEM., vol. 45, 2002, pages 4336 - 4343
KINGSBURY ET AL., J. MED. CHEM., vol. 27, 1984, pages 1447
KLIMKA ET AL., BR. J. CANCER, vol. 83, 2000, pages 252 - 260
KOSTELNY ET AL., J. IMMUNOL., vol. 148, no. 5, 1992, pages 1547 - 1553
KOVTUN ET AL., CANCER RES., vol. 66, no. 6, 2006, pages 3214 - 3121
KOZBOR, J. IMMUNOL., vol. 133, 1984, pages 3001
KRATZ ET AL., CURRENT MED. CHEM., vol. 13, 2006, pages 477 - 523
KROP ET AL., EUROPEAN CANCER CONFERENCE ECCO, 23 September 2007 (2007-09-23)
LAMBERT, J., CURR. OPINION IN PHARMACOLOGY, vol. 5, 2005, pages 543 - 549
LAW ET AL., CANCER RES., vol. 66, no. 4, 2006, pages 2328 - 2337
LEE ET AL., J. IMMUNOL. METHODS, vol. 284, no. 1-2, 2004, pages 119 - 132
LEE ET AL., J. MOL. BIOL., vol. 340, no. 5, 2004, pages 1073 - 1093
LI ET AL., NAT. BIOTECH., vol. 24, 2006, pages 210 - 215
LI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 3557 - 3562
LIU ET AL., PROC. NATL. ACAD. SCI USA, vol. 93, 1996, pages 8618 - 8623
LIU ET AL., PROC. NATL. ACAD. SCI. USA, vol. 93, 1996, pages 8618 - 8623
LODE ET AL., CANCER RES., vol. 58, 1998, pages 2928
LODE ET AL., CANCER RESEARCH, vol. 58, 1998, pages 2925 - 2928
LONBERG, CURR. OPIN. IMMUNOL., vol. 20, 2008, pages 450 - 459
LONBERG, NAT. BIOTECH., vol. 23, 2005, pages 1117 - 1125
MANDLER ET AL., BIOCONJUGATE CHEM., vol. 13, 2002, pages 786 - 791
MANDLER ET AL., BIOORGANIC & MED. CHEM. LETTERS, vol. 10, 2000, pages 1025 - 1028
MANDLER ET AL., J. NAT. CANCER INST., vol. 92, no. 19, 2000, pages 1573 - 1581
MARKS ET AL., J. MOL. BIOL., vol. 222, 1992, pages 581 - 597
MARKS; BRADBURY, METHODS IN MOLECULAR BIOLOGY, vol. 248, pages 161 - 175
MARTIN ET AL., J. BIOL. CHEM., vol. 257, 1982, pages 286 - 288
MATHER ET AL., ANNALS N.Y. ACAD. SCI., vol. 383, 1982, pages 44 - 68
MATHER, BIOL. REPROD., vol. 23, 1980, pages 243 - 251
MCCAFFERTY ET AL., NATURE, vol. 348, pages 552 - 554
MCDONAGH ET AL., PROT. ENGR. DESIGN & SELECTION, vol. 19, no. 7, 2006, pages 299 - 307
MCDONAGH, PROTEIN ENG. DESIGN & SEL, 2006
MILSTEIN; CUELLO, NATURE, vol. 305, 1983, pages 537
MORRIS: "Methods in Molecular Biology", vol. 66, 1996, HUMANA PRESS, article "Epitope Mapping Protocols"
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
NAGY ET AL., PROC. NATL. ACAD. SCI., vol. 97, 2000, pages 829 - 834
NAKAMUTA M ET AL.: "Cloning and Sequence Analysis of a cDNA encoding Human non-selective type of endothelin receptor", BIOCHEM BIOPHYS RES COMMUN., vol. 177, no. l, 31 May 1991 (1991-05-31), pages 34 - 9, XP024771029, DOI: doi:10.1016/0006-291X(91)91944-8
NAZARIAN R. ET AL.: "Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation", NATURE, vol. 468, 16 December 2010 (2010-12-16), pages 973 - 977
NEUBERGER ET AL., NATURE, vol. 312, 1984, pages 604 - 608
NI, XIANDAI MIANYIXUE, vol. 26, no. 4, 2006, pages 265 - 268
NICULESCU-DUVAZ; SPRINGER, ADV. DRUG DELIV. REV., vol. 26, 1997, pages 151 - 172
OKAZAKI ET AL., J. MOL. BIOL., vol. 336, 2004, pages 1239 - 1249
OSBOURN ET AL., METHODS, vol. 36, 2005, pages 61 - 68
OSOL, A.: "Remington's Pharmaceutical Sciences 16th edition,", 1980
PADLAN, MOL. IMMUNOL., vol. 28, 1991, pages 489 - 498
PETKOVA, S.B. ET AL., INT'L. IMMUNOL., vol. 18, no. 12, 2006, pages 1759 - 1769
PETTIT ET AL., ANTI-CANCER DRUG DESIGN, vol. 13, 1998, pages 243 - 277
PETTIT ET AL., ANTIMICROB. AGENTS CHEMOTHER., vol. 42, 1998, pages 2961 - 2965
PETTIT ET AL., J. AM. CHEM. SOC., vol. 111, 1989, pages 5463 - 5465
PETTIT ET AL., J. CHEM. SOC. PERKIN TRANS., vol. 1, no. 5, 1996, pages 859 - 863
PETTIT, G.R. ET AL., SYNTHESIS, 1996, pages 719 - 725
PLUCKTHUN: "The Pharmacology of Monoclonal Antibodies,", vol. 113, 1994, SPRINGER-VERLAG, pages: 269 - 315
PORTOLANO ET AL., J. IMMUNOL., vol. 150, 1993, pages 880 - 887
PRESTA ET AL., CANCER RES., vol. 57, 1997, pages 4593 - 4599
PRESTA ET AL., J. IMMUNOL., vol. 151, 1993, pages 2623
QUEEN ET AL., PROC. NAT'L ACAD. SCI. USA, vol. 86, 1989, pages 10029 - 10033
RAVETCH; KINET, ANNU. REV. IMMUNOL., vol. 9, 1991, pages 457 - 492
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 329
RIPKA ET AL., ARCH. BIOCHEM. BIOPHYS., vol. 249, 1986, pages 533 - 545
RODRIGUES ET AL., CHEMISTRY BIOLOGY, vol. 2, 1995, pages 223
ROSOK ET AL., J. BIOL. CHEM., vol. 271, 1996, pages 22611 - 22618
ROWLAND ET AL., CANCER IMMUNOL. IMMUNOTHER., vol. 21, 1986, pages 183 - 87
SALA E ET AL.: "BRAF silencing by short hairpin RNA or chemical blockade by PLX4032 leads to different responses in melanoma and thyroid carcinoma cells", MOL. CANCER RES., vol. 6, no. 5, May 2008 (2008-05-01), pages 751 - 9, XP055267102, DOI: doi:10.1158/1541-7786.MCR-07-2001
SANDERSON ET AL., CLIN. CANCER RES., vol. 11, 2005, pages 843 - 852
SEBOLT-LEOPOLD JS; HERRERA R.: "Targeting the mitogen-activated protein kinase cascade to treat cancer", NAT REV CANCER., vol. 4, 2004, pages 937 - 947, XP002446982, DOI: doi:10.1038/nrc1503
See also references of EP2776051A4
SHIELDS ET AL., J. BIOL. CHEM., vol. 9, no. 2, 2001, pages 6591 - 6604
SIDHU ET AL., J. MOL. BIOL., vol. 338, no. 2, 2004, pages 299 - 310
SIMS ET AL., J. IMMUNOL., vol. 151, 1993, pages 2296
STORM ET AL., J. AMER. CHEM. SOC., vol. 94, 1972, pages 5815
SUN ET AL., BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 12, 2002, pages 2213 - 2215
SUN ET AL., BIOORGANIC & MEDICINAL CHEMISTRY, vol. 11, 2003, pages 1761 - 1768
SYRIGOS; EPENETOS, ANTICANCER RESEARCH, vol. 19, 1999, pages 605 - 614
THOMPSON JF ET AL.: "Cutaneous melanoma in the era of molecular profiling", LANCET, vol. 374, 2009, pages 362 - 5, XP026395827, DOI: doi:10.1016/S0140-6736(09)61397-0
THOMPSON, LANCET, 2009
THORPE ET AL.: "Monoclonal Antibodies '84: Biological And Clinical Applications", 1985, article "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review", pages: 475 - 506
TORGOV ET AL., BIOCONJ. CHEM., vol. 16, 2005, pages 717 - 721
TRAUNECKER ET AL., EMBO J., vol. 10, 1991, pages 3655
TUTT ET AL., J. IMMUNOL., vol. 147, 1991, pages 60
URLAUB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216
VAN DIJK; VAN DE WINKEL, CURR. OPIN. PHARMACOL., vol. 5, 2001, pages 368 - 74
VITETTA ET AL., SCIENCE, vol. 238, 1987, pages 1098
VOLLMERS; BRANDLEIN, HISTOLOGY AND HISTOPATHOLOGY, vol. 20, no. 3, 2005, pages 927 - 937
VOLLMERS; BRANDLEIN, METHODS AND FINDINGS IN EXPERIMENTAL AND CLINICAL PHARMACOLOGY, vol. 27, no. 3, 2005, pages 185 - 91
WIDDISON ET AL., J. MED. CHEM., vol. 49, 2006, pages 4392 - 4408
WINTER ET AL., ANN. REV. IMMUNOL., vol. 12, 1994, pages 433 - 455
WISEMAN ET AL., BLOOD, vol. 99, no. 12, 2002, pages 4336 - 42
WISEMAN ET AL., EUR. JOUR. NUCL. MED., vol. 27, no. 7, 2000, pages 766 - 77
WITZIG ET AL., J. CLIN. ONCOL., vol. 20, no. 10, 2002, pages 2453 - 63
WITZIG ET AL., J. CLIN. ONCOL., vol. 20, no. 15, 2002, pages 3262 - 69
WONG K-K ET AL.: "Recent developments in anti-cancer agents targeting the Ras/Raf/MEKJERK pathway", RECENT PAT ANTICANCER DRUG DISCOV., vol. 4, 2009, pages 28 - 35, XP055056888, DOI: doi:10.2174/157489209787002461
WOYKE ET AL., ANTIMICROB. AGENTS AND CHEMOTHER., vol. 45, no. 12, 2001, pages 3580 - 3584
WRIGHT ET AL., TIBTECH, vol. 15, 1997, pages 26 - 32
WU ET AL., NATURE BIOTECHNOLOGY, vol. 23, no. 9, 2005, pages 1137 - 1146
XIE ET AL., EXPERT. OPIN. BIOL. THER., vol. 6, no. 3, 2006, pages 281 - 291
YAMANE-OHNUKI ET AL., BIOTECH. BIOENG., vol. 87, 2004, pages 614
YAZAKI; WU: "Methods in Molecular Biology", vol. 248, 2003, HUMANA PRESS, pages: 255 - 268
YU ET AL., PNAS, vol. 99, 2002, pages 7968 - 7973

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9724413B2 (en) 2011-08-01 2017-08-08 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
US10646567B2 (en) 2011-08-01 2020-05-12 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
US9216170B2 (en) 2012-03-19 2015-12-22 Hoffmann-La Roche Inc. Combination therapy for proliferative disorders
US9486445B2 (en) 2012-03-19 2016-11-08 Hoffmann-La Roche Inc. Combination therapy for proliferative disorders
US10946093B2 (en) 2014-07-15 2021-03-16 Genentech, Inc. Methods of treating cancer using PD-1 axis binding antagonists and MEK inhibitors
WO2016055907A1 (en) * 2014-10-10 2016-04-14 Pfizer Inc. Synergistic auristatin combinations
EP3549583A1 (en) * 2014-10-10 2019-10-09 Pfizer Inc Synergistic auristatin combinations
US10617670B2 (en) 2014-10-10 2020-04-14 Pfizer Inc. Synergistic auristatin combinations
WO2017220739A1 (en) 2016-06-24 2017-12-28 Commissariat A L'energie Atomique Et Aux Energies Alternatives Antibody directed against the endothelin receptor beta sub-type
FR3053042A1 (en) * 2016-06-24 2017-12-29 Commissariat Energie Atomique ANTIBODIES AGAINST ENDOTHELIN RECEPTOR B SUBTYPE B AND USES THEREOF
US11312780B2 (en) 2016-06-24 2022-04-26 Commissariat à l'énergie atomique et aux énergies alternatives Antibody directed against the endothelin receptor beta sub-type
US11040027B2 (en) 2017-01-17 2021-06-22 Heparegenix Gmbh Protein kinase inhibitors for promoting liver regeneration or reducing or preventing hepatocyte death

Also Published As

Publication number Publication date
TW201325613A (en) 2013-07-01
EP2776051A4 (en) 2015-06-17
JP2014532648A (en) 2014-12-08
CO7151543A2 (en) 2014-12-29
MX2014004991A (en) 2014-05-22
SG11201401815XA (en) 2014-05-29
CN104039340B (en) 2017-04-05
CN104039340A (en) 2014-09-10
IL232135A0 (en) 2014-05-28
EP2776051A1 (en) 2014-09-17
KR20140097205A (en) 2014-08-06
AR088509A1 (en) 2014-06-11
JP2018027948A (en) 2018-02-22
AU2012328980A1 (en) 2014-04-24
IN2014CN03062A (en) 2015-07-31
CL2014001092A1 (en) 2014-07-25
CA2850034A1 (en) 2013-05-02
JP6251682B2 (en) 2017-12-20
EA201490879A1 (en) 2014-08-29
PE20142312A1 (en) 2015-01-25
BR112014009953A2 (en) 2017-12-05

Similar Documents

Publication Publication Date Title
US11352431B2 (en) Anti-FCRH5 antibodies
US10653792B2 (en) Anti-Ly6E antibodies and immunoconjugates and methods of use
US9175089B2 (en) Anti-LGR5 antibodies and immunoconjugates
US20180312602A1 (en) Anti-gpc3 antibodies and immunoconjugates
JP6251682B2 (en) Combinations and methods of treatment for melanoma treatment
US9926377B2 (en) Anti-GPC3 antibodies and immunoconjugates
US20170043034A1 (en) Methods of using anti-steap1 antibodies and immunoconjugates
EP3046940B1 (en) Methods of using anti-lgr5 antibodies
US20140341916A1 (en) Therapeutic combinations and methods of treating melanoma
NZ715125B2 (en) Anti-fcrh5 antibodies

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12842985

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2850034

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2012842985

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 14083118

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 232135

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2012328980

Country of ref document: AU

Date of ref document: 20121024

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2014/004991

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 000603-2014

Country of ref document: PE

Ref document number: 14354362

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2014538892

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20147013878

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201490879

Country of ref document: EA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014009953

Country of ref document: BR

REG Reference to national code

Ref country code: BR

Ref legal event code: B01E

Ref document number: 112014009953

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112014009953

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20140425