US20080152660A1 - Anti-Angiogenic Compounds - Google Patents

Anti-Angiogenic Compounds Download PDF

Info

Publication number
US20080152660A1
US20080152660A1 US11/817,668 US81766806A US2008152660A1 US 20080152660 A1 US20080152660 A1 US 20080152660A1 US 81766806 A US81766806 A US 81766806A US 2008152660 A1 US2008152660 A1 US 2008152660A1
Authority
US
United States
Prior art keywords
pro
substituted
ile
arg
thr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/817,668
Other languages
English (en)
Inventor
Curt W. Bradshaw
Venkata Ramana Doppalapudi
Jing-Yu Lai
John Rizzo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Covx Technologies Ireland Ltd
Original Assignee
Covx Technologies Ireland Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Covx Technologies Ireland Ltd filed Critical Covx Technologies Ireland Ltd
Priority to US11/817,668 priority Critical patent/US20080152660A1/en
Assigned to COVX TECHNOLOGIES IRELAND LIMITED reassignment COVX TECHNOLOGIES IRELAND LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRADSHAW, CURT W., DOPPALAPUDI, VENKATA RAMANA, LAI, JING-YU, RIZZO, JOHN
Publication of US20080152660A1 publication Critical patent/US20080152660A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/02Linear peptides containing at least one abnormal peptide link
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • 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
    • A61K39/44Antibodies bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates to novel compounds that possess anti-angiogenic activity and methods of making and using these compounds.
  • Angiogenesis is the fundamental process by which new blood vessels are formed and is essential to a variety of normal body activities such as reproduction, development and wound repair.
  • angiogenesis is a highly regulated process under normal conditions, many diseases (characterized as “angiogenic diseases”) are caused or exacerbated by unregulated angiogenesis.
  • angiogenic diseases characterized as “angiogenic diseases”
  • ocular neovascularization has been implicated as the most common cause of blindness.
  • new capillaries formed in the retina invade the vitreous, bleed, and cause blindness.
  • Growth and metastasis of solid tumors are also angiogenesis-dependent (J. Folkman, Cancer Res., 46:467-473 (1986), J. Folkman, J. Natl.
  • Thrombospondin-1 is an extracellular matrix protein secreted in response to activation of platelets by thrombin.
  • TSP-1 Thrombospondin-1
  • Various studies have demonstrated that certain peptide analogs of TSP-1 possess antiangiogenesic activity. See, e.g., WO 01/38397, WO 01/38347, WO 99/61476, U.S. Patent Application Pub. No. 2003/0045477, U.S. Patent Application Pub. No. 2002/0183242, U.S. Pat. No. 6,774,211, U.S. Pat. No. 6,716,963, U.S. Pat. No. 6,753,408, and U.S. Pat. No. 5,932,545.
  • the present invention provides thrombospondin receptor targeting compounds (AA targeting compounds) with unique specificity and biological properties which are useful in many applications.
  • the thrombospondin targeting compounds of the invention are formed by covalently linking a thrombospondin targeting agent to a combining site of an antibody.
  • Pharmaceutical compositions comprising targeting compounds of the invention and a pharmaceutically acceptable carrier are also provided.
  • a first aspect of the invention is an AA targeting agent-linker conjugate having Formula I:
  • [AA targeting agent] is a peptide selected from the group consisting of:
  • X is:
  • X is attached to an amino acid residue in [AA targeting agent], and is an optionally substituted —R 22 —[CH 2 —CH 2 —O] t —R 23 —, —R 22 -cycloalkyl- R 23 —, —R 22 -aryl-R 23 —, or —R 22 -heterocyclyl-R 23 —, wherein t is 0 to 50.
  • R 22 is —(CH 2 ) v —, —(CH 2 ) u —C(O)—(CH 2 ) v —, —(CH 2 ) u —C(O)—O—(CH 2 ) v —, —(CH 2 ) u —C(S)—NR b —(CH 2 ) v —, —(CH 2 ) u —C(O)—NR b —(CH 2 ) v —, —(CH 2 ) u —NR b —(CH 2 ) v —, —(CH 2 ) u —O—(CH 2 ) v —, —(CH 2 ) u —S(O) 0-2 —(CH 2 ) v —, —(CH 2 ) u —S(O) 0-2 —NR b —(CH 2 ) v —,
  • R 21 and R 23 are independently —(CH 2 ) s —, —(CH 2 ) r —C(O)—(CH 2 ) s —, —(CH 2 ) r —C(O)—O—(CH 2 ) v —, —(CH 2 ) r —C(S)—NR b —(CH 2 ) s —, —(CH 2 ) r ,C(O)—NR b —(CH 2 ) s —, (CH 2 ) r —NR b (CH 2 ) s —, —(CH 2 ) r —O—(CH 2 ) s —, —(CH 2 ) r —S(O) 0-2 —(CH 2 ) s , —(CH 2 ) n —S(O) 0-2 —NR b —(CH 2 ) s —, or
  • 1A and 1B illustrate two embodiments according to Formula I that employ Ac-Sar-Gly-Val-(D-alloIle)-Thr-Nva-Ile-Arg-Pro (SEQ ID NO:1, wherein R 1 is Ac and R 3 is absent) and Sar-Gly-Val-(D-alloIle)-Thr-Nva-Ile-Arg-Pro-NHEt (SEQ ID NO:1, wherein R 1 is absent and R 3 is NHEt), respectively, as targeting agents.
  • SEQ ID NO:1 wherein R 1 is Ac and R 3 is absent
  • SEQ ID NO:1 wherein R 1 is absent and R 3 is NHEt
  • FIG. 2 illustrates other embodiments according to Formula I that employ Ac-Sar-Gly-Val-(D-alloIle)-Thr-Nva-Lys-Arg-Pro-NHEt (SEQ ID NO:3, wherein R 1 is Ac and R 3 is NHEt) as a targeting agent.
  • an AA targeting compound comprising an AA targeting agent covalently linked to a combining site of an Antibody via an intervening linker L′.
  • the Antibody portion of an AA targeting compound can include whole (full length) antibody, unique antibody fragments, or any other forms of an antibody as this term is used herein.
  • the Antibody is a humanized version of a murine aldolase antibody comprising a constant region from a human IgG, IgA, IgM, IgD, or IgE antibody.
  • the Antibody is a chimeric antibody comprising the variable region from a murine aldolase antibody and a constant region from a human IgG, IgA, IgM, IgD, or IgE antibody.
  • the Antibody is a fully human version of a murine aldolase antibody comprising a polypeptide sequence from natural or native human IgG, IgA, IgM, IgD, or IgE antibody
  • [AA targeting agent] is a peptide selected from the group consisting of:
  • X is:
  • X is attached to an amino acid residue in [AA targeting agent], and is an optionally substituted —R 22 —[CH 2 —CH 2 —O] t —R 23 —, —R 22 -cycloalkyl- R 23 —, —R 22 -aryl-R 23 —, or —R 22 -heterocyclyl-R 23 —, wherein t is 0 to 50.
  • R 22 is —(CH 2 ) v —, —(CH 2 ) u —C(O)—(CH 2 ) v —, —(CH 2 ) u —C(O)—O—(CH 2 ) v —, —(CH 2 ) u —C(S)—NR b —(CH 2 ) v —, —(CH 2 ) n —C(O)—NR b —(CH 2 ) v —, —(CH 2 ) u —NR b —(CH 2 ) v —, —(CH 2 ) u —O—(CH 2 ) v —, —(CH 2 ) u —S(O) 0-2 —(CH 2 ) v —, —(CH 2 ) u —S(O) 0-2 —NR b —(CH 2 ) v —,
  • R 21 and R 23 are independently —(CH 2 ) s —, —(CH 2 ) r ,C(O)—(CH 2 ) s —, (CH 2 ) r ,C(O)—O—(CH 2 ) v —, —(CH 2 ) r —C(S)—NR b —(CH 2 ) s —, (CH 2 ) r ,C(O)—NR b (CH 2 ) s —, —(CH 2 ) r —NR b (CH 2 ) v —, —(CH 2 ) r —O—(CH 2 ) s , —(CH 2 ) r —S(O) 0-2 —(CH 2 ) s —, —(CH 2 ) r —S(O) 0-2 —NR b —(CH 2 ) s —, or —(CH 2 ) s —,
  • Antibody is the humanized aldolase antibody h38c2 IgG1
  • Antibody is the humanized aldolase antibody h38c2 IgG1
  • an AA targeting compound in which two AA targeting agents, which may be the same or different, are each covalently linked to a combining site of an antibody.
  • the Antibody portion of an AA targeting compound can include whole (full length) antibody, unique antibody fragments, or any other forms of an antibody as this term is used herein.
  • the Antibody is a humanized version of a murine aldolase antibody comprising a constant region from a human IgG, IgA, IgM, IgD, or IgE antibody.
  • the Antibody is a chimeric antibody comprising the variable region from a murine aldolase antibody and a constant region from a human IgG, IgA, IgM, IgD, or IgE antibody.
  • the Antibody is a fully human version of a murine aldolase antibody comprising a polypeptide sequence from natural or native human IgG, IgA, IgM, IgD, or IgE antibody.
  • FIG. 3 illustrates embodiments according to Formula III that employ Sar-Gly-Val-(D-alloIle)-Thr-Nva-Ile-Arg-Pro-NHEt (SEQ ID NO:1, wherein R 1 is absent and R 3 is NHEt) as a targeting agent.
  • FIG. 4 illustrates other embodiments according to Formula III that employ Ac-Sar-Gly-Val-(D-alloIle)-Thr-Nva-Ile-Arg-Pro (SEQ ID NO:1) as a targeting agent.
  • FIG. 5 illustrates other embodiments according to Formula III that employ Ac-Sar-Gly-Val-(D-alloIle)-Thr-Nva-Lys-Arg-Pro-NHEt (SEQ ID NO:3) as a targeting agent.
  • methods of delivering or administering AA targeting compounds of the invention and methods of treatment using AA targeting compounds of the invention include administering a therapeutically effective amount of an AA targeting compound of the invention to the subject.
  • Diseases and conditions that may be treated include cancer, arthritis, hypertension, kidney disease, psoriasis, angiogenesis of the eye associated with ocular disorder, infection or surgical intervention, macular degeneration, diabetic retinopathy, and the like.
  • AA targeting compounds of the invention can be used for the diagnosis of a disease or condition associated with abnormal angiogenesis, including cancer, arthritis, psoriasis, angiogenesis of the eye associated with an ocular disorder, infection or surgical intervention, macular degeneration, diabetic retinopathy, and the like.
  • FIGS. 1A and B illustrate embodiments according to Formula I.
  • FIG. 2 illustrate additional embodiments according to Formula I.
  • FIG. 3 illustrates embodiments according to Formula III.
  • FIG. 4 illustrates additional embodiments according to Formula III.
  • FIG. 5 illustrates additional embodiments according to Formula III.
  • Antibody-N- represents a covalent bond to a side of an amino acid in a combining site of an antibody.
  • FIG. 6A and FIG. 6B illustrate the solid phase synthesis of targeting agent-linker conjugates of the present invention.
  • FIG. 7A illustrates the amino acid sequence alignment of the variable domains of m38c2, h38c2, and human germlines.
  • Framework regions (FR) and complementarity determining regions (CDR) are defined according to Kabat et al. Asterisks mark differences between m38c2 and h38c2 or between h38c2 and the human germlines.
  • FIG. 7B illustrates the amino acid sequence of the light and heavy chains of h38c2 IgG1.
  • FIG. 8 shows various structures that may serve as linker reactive groups.
  • Structures A-C form reversible covalent bonds with surface accessible reactive nucleophilic groups (e.g., lysine or cysteine side chain) of a combining site of an antibody.
  • R′ 1 , R′ 2 , R′ 3 , and R 4 in structures A-C represent substituents which include, for example, C, H, N, O, P, S, halogen (F, Cl, Br, I) or a salt thereof.
  • X is N, C, or any other heteroatom.
  • substituents may also include a group such as an alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl group.
  • R′ 2 and R′ 3 could be cyclic as exemplified in structures B and C while X could be a heteroatom.
  • structure A could form an irreversible covalent bond with a reactive nucleophile if X is N and if R′ 1 , and R 3 form part of a cyclic structure.
  • Structures D-G may form nonreversible covalent bonds with reactive nucleophilic groups in a combining site of an antibody.
  • R′′ 1 , and R′′ 2 represent C, O, N, halide or leaving groups such as mesyl or tosyl.
  • FIG. 9 shows various electrophiles that are suitable for reactive modification with a reactive amino acid side chain in a combining site of an antibody and thus may serve as linker reactive groups.
  • the squiggle line indicates the point of attachment to the rest of the linker or targeting agent.
  • X refers to a halogen.
  • FIG. 10 shows the addition of a nucleophilic (“nu”) side chain in an antibody combining site to compounds A-G in FIG. 8 .
  • Antibody-Nu- refers to a covalent bond to an amino acid side chain bearing a nucleophile in a combining site of an antibody.
  • FIG. 11 shows the addition of a nucleophilic side chain in an antibody combining to compounds A-H in FIG. 9 .
  • Antibody-Nu- refers to a covalent bond to an amino acid side chain bearing a nucleophile in a combining site of an antibody.
  • FIG. 12 shows a synthesis of:
  • FIG. 13 shows a synthesis of:
  • FIG. 14 shows a synthesis of:
  • FIG. 15 shows a synthesis of:
  • FIG. 16 shows a synthesis of:
  • FIG. 17 shows a synthesis of:
  • FIG. 18 shows a synthesis of:
  • FIG. 19 shows a synthesis of:
  • FIG. 20 shows a synthesis of:
  • FIG. 21 shows syntheses of:
  • FIG. 22 shows a synthesis of:
  • FIG. 23 shows a synthesis of:
  • FIG. 24 shows a synthesis of:
  • FIG. 25 shows a synthesis of:
  • FIG. 26 shows a synthesis of:
  • FIG. 27 shows a synthesis of:
  • FIG. 28 shows a synthesis of:
  • Every amino-bearing side chain of a targeting agent can be terminated by R 1 or R 2 as defined herein.
  • Every COOH/COO ⁇ — bearing side chain of a targeting agent can be terminated by R 3 as defined herein.
  • Sarcosine refers to N-methyl glycine.
  • 3-(4-thiazolyl)-L-Alanine or 3-(4-thiazolyl)-L-Ala refers to:
  • 4-Cyanophenylalanine or 4-Cyano-Phe refers to:
  • D-alloisoleucine or D-alloIle or D-aIle refers to:
  • ⁇ -ally-glycine or ⁇ -ally-Gly refers to:
  • Cycloleucine or Cyclo-Leu refers to:
  • 2-furyl-alanine or 2-furyl-Ala refers to:
  • Polypeptide “peptide,” and “protein” are used interchangeably to refer to a polymer of amino acid residues. As used herein, these terms apply to amino acid polymers in which one or more amino acid residues is an artificial chemical analog of a corresponding naturally occurring amino acid. These terms also apply to naturally occurring amino acid polymers.
  • Amino acids can be in the L or D form as long as the binding function of the peptide is maintained.
  • Peptides may be cyclic, having an intramolecular bond between two non-adjacent amino acids within the peptide, e.g., backbone to backbone, side-chain to backbone and side-chain to side-chain cyclization. Cyclic peptides can be prepared by methods well know in the art. See e.g., U.S. Pat. No. 6,013,625.
  • N-terminus refers to the free alpha-amino group of an amino acid in a peptide
  • C-terminus refers to the free ⁇ -carboxylic acid terminus of an amino acid in a peptide.
  • a peptide which is N-terminated with a group refers to a peptide bearing a group on the alpha-amino nitrogen of the N-terminal amino acid residue.
  • An amino acid which is N-terminated with a group refers to an amino acid bearing a group on the alpha-amino nitrogen.
  • substituted refers to a group as defined below in which one or more bonds to a hydrogen atom contained therein are replaced by a bond to non-hydrogen or non-carbon atoms such as, but not limited to, a halogen atom such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, and ester groups; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as in trialkylsilyl groups, dialkylarylsilyl groups, alkyldiary
  • Substituted alkyl groups and also substituted cycloalkyl groups and others also include groups in which one or more bonds to a carbon(s) or hydrogen(s) atom is replaced by a bond to a heteroatom such as oxygen in carbonyl, carboxyl, and ester groups; nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • a group which is “optionally substituted” may be substituted or unsubstituted.
  • optionally substituted alkyl refers to both substituted alkyl groups and unsubstituted alkyl groups.
  • unsubstituted alkyl refers to alkyl groups that do not contain heteroatoms.
  • the phrase includes straight chain alkyl groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like.
  • the phrase also includes branched chain isomers of straight chain alkyl groups, including but not limited to, the following which are provided by way of example: —CH(CH 3 ) 2 , —CH(CH 3 )(CH 2 CH 3 ), —CH(CH 2 CH 3 ) 2 , —C(CH 3 ) 3 , —C(CH 2 CH 3 ) 3 , —CH 2 CH(CH 3 ) 2 , —CH 2 CH(CH 3 )(CH 2 CH 3 ), —CH 2 CH(CH 2 CH 3 ) 2 , —CH 2 C(CH 3 ) 3 , —CH 2 C(CH 2 CH 3 ) 3 , —CH(CH 3 )CH(CH 3 )(CH 2 CH 3 ), —CH 2 CH 2 CH(CH 3 ) 2 , —CH 2 CH 2 CH(CH 3 )(CH 2 CH 3 ), —CH 2 CH 2 CH(CH 3 ) 2 , —CH 2 CH 2 CH(CH 3 ) 2
  • unsubstituted alkyl groups includes primary alkyl groups, secondary alkyl groups, and tertiary alkyl groups.
  • Unsubstituted alkyl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound.
  • Possible unsubstituted alkyl groups include straight and branched chain alkyl groups having 1 to 20 carbon atoms. Alternatively, such unsubstituted alkyl groups have from 1 to 10 carbon atoms or are lower alkyl groups having from 1 to about 6 carbon atoms.
  • Other unsubstituted alkyl groups include straight and branched chain alkyl groups having from 1 to 3 carbon atoms and include methyl, ethyl, propyl, and —CH(CH 3 ) 2 .
  • substituted alkyl refers to an alkyl group in which one or more bonds to a carbon(s) or hydrogen(s) are replaced by a bond to non-hydrogen and non-carbon atoms such as, but not limited to, a halogen atom in halides such as F, Cl, Br, and I; an oxygen atom in groups such as hydroxyl groups, alkoxy groups, aryloxy groups, and ester groups; a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfone groups, sulfonyl groups, and sulfoxide groups; a nitrogen atom in groups such as amines, amides, alkylamines, dialkylamines, arylamines, alkylarylamines, diarylamines, N-oxides, imides, and enamines; a silicon atom in groups such as in trialkylsilyl groups, dialkylarylsilyl
  • Substituted alkyl groups also include groups in which one or more bonds to a carbon(s) or hydrogen(s) atom is replaced by a bond to a heteroatom such as oxygen in carbonyl, carboxyl, and ester groups; nitrogen in groups such as imines, oximes, hydrazones, and nitriles.
  • Substituted alkyl groups include, among others, alkyl groups in which one or more bonds to a carbon or hydrogen atom is/are replaced by one or more bonds to fluorine atoms.
  • One example of a substituted alkyl group is the trifluoromethyl group and other alkyl groups that contain the trifluoromethyl group.
  • alkyl groups include those in which one or more bonds to a carbon or hydrogen atom is replaced by a bond to an oxygen atom such that the substituted alkyl group contains a hydroxyl, alkoxy, aryloxy group, or heterocyclyloxy group.
  • Still other alkyl groups include alkyl groups that have an amine, alkylamine, dialkylamine, arylamine, (alkyl)(aryl)amine, diarylamine, heterocyclylamine, (alkyl)(heterocyclyl)amine, (aryl)(heterocyclyl)amine, or diheterocyclylamine group.
  • unsubstituted alkylene refers to a divalent unsubstituted alkyl group as defined above.
  • methylene, ethylene, and propylene are each examples of unsubstituted alkylenes.
  • substituted alkylene refers to a divalent substituted alkyl group as defined above.
  • Substituted or unsubstituted lower alkylene groups have from 1 to about 6 carbons.
  • unsubstituted cycloalkyl refers to cyclic alkyl groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl and such rings substituted with straight and branched chain alkyl groups as defined above.
  • the phrase also includes polycyclic alkyl groups such as, but not limited to, adamantyl norbornyl, and bicyclo[2.2.2]octyl and the like, as well as such rings substituted with straight and branched chain alkyl groups as defined above.
  • the phrase would include methylcylcohexyl groups among others.
  • Unsubstituted cycloalkyl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound. In some embodiments unsubstituted cycloalkyl groups have from 3 to 20 carbon atoms. In other embodiments, such unsubstituted alkyl groups have from 3 to 8 carbon atoms while in others, such groups have from 3 to 7 carbon atoms.
  • substituted cycloalkyl has the same meaning with respect to unsubstituted cycloalkyl groups that substituted alkyl groups have with respect to unsubstituted alkyl groups.
  • the phrase includes, but is not limited to, oxocyclohexyl, chlorocyclohexyl, hydroxycyclopentyl, and chloromethylcyclohexyl groups.
  • unsubstituted aryl refers to aryl groups that do not contain heteroatoms.
  • the phrase includes, but is not limited to, groups such as phenyl, biphenyl, anthracenyl, and naphthenyl by way of example.
  • unsubstituted aryl includes groups containing condensed rings such as naphthalene, it does not include aryl groups that have other groups such as alkyl or halo groups bonded to one of the ring members, as aryl groups such as tolyl are considered herein to be substituted aryl groups as described below.
  • an unsubstituted aryl may be a lower aryl, having from 6 to about 10 carbon atoms.
  • One unsubstituted aryl group is phenyl.
  • Unsubstituted aryl groups may be bonded to one or more carbon atom(s), oxygen atom(s), nitrogen atom(s), and/or sulfur atom(s) in the parent compound, however.
  • substituted aryl group has the same meaning with respect to unsubstituted aryl groups that substituted alkyl groups have with respect to unsubstituted alkyl groups.
  • a substituted aryl group also includes aryl groups in which one of the aromatic carbons is bonded to one of the non-carbon or non-hydrogen atoms described above and also includes aryl groups in which one or more aromatic carbons of the aryl group is bonded to a substituted and/or unsubstituted alkyl, alkenyl, or alkynyl group as defined herein.
  • unsubstituted alkenyl refers to straight and branched chain and cyclic groups such as those described with respect to unsubstituted alkyl groups as defined above, except that at least one double bond exists between two carbon atoms.
  • Examples include, but are not limited to vinyl, —CH ⁇ C(H)(CH 3 ), —CH ⁇ C(CH 3 ) 2 , —C(CH 3 ) ⁇ C(H) 2 , —C(CH 3 ) ⁇ C(H)(CH 3 ), —C(CH 2 CH 3 ) ⁇ CH 2 , cyclohexenyl, cyclopentenyl, cyclohexadienyl, butadienyl, pentadienyl, and hexadienyl among others.
  • Lower unsubstituted alkenyl groups have from 1 to about 6 carbons.
  • substituted alkenyl has the same meaning with respect to unsubstituted alkenyl groups that substituted alkyl groups have with respect to unsubstituted alkyl groups.
  • a substituted alkenyl group includes alkenyl groups in which a non-carbon or non-hydrogen atom is bonded to a carbon double bonded to another carbon and those in which one of the non-carbon or non-hydrogen atoms is bonded to a carbon not involved in a double bond to another carbon.
  • —CH ⁇ CH—OCH 3 and —CH ⁇ CH—CH 2 —OH are both substituted alkenyls.
  • Oxoalkenyls wherein a CH 2 group is replaced by a carbonyl such as —CH ⁇ CH—C(O)—CH 3 , are also substituted alkenyls.
  • unsubstituted alkenylene refers to a divalent unsubstituted alkenyl group as defined above.
  • —CH ⁇ CH— is an exemplary unsubstituted alkenylene.
  • substituted alkenylene refers to a divalent substituted alkenyl group as defined above.
  • unsubstituted alkynyl refers to straight and branched chain groups such as those described with respect to unsubstituted alkyl groups as defined above, except that at least one triple bond exists between two carbon atoms. Examples include, but are not limited to, —C ⁇ C(H), —C ⁇ C(CH 3 ), —C ⁇ C(CH 2 CH 3 ), —C(H 2 )C ⁇ C(H), —C(H) 2 C ⁇ C(CH 3 ), and —C(H) 2 C ⁇ C(CH 2 CH 3 ) among others. Unsubstituted lower alkynyl groups have from 1 to about 6 carbons.
  • substituted alkynyl has the same meaning with respect to unsubstituted alkynyl groups that substituted alkyl groups have with respect to unsubstituted alkyl groups.
  • a substituted alkynyl group includes alkynyl groups in which a non-carbon or non-hydrogen atom is bonded to a carbon triple bonded to another carbon and those in which a non-carbon or non-hydrogen atom is bonded to a carbon not involved in a triple bond to another carbon.
  • Examples include, but are not limited to, oxoalkynyls wherein a CH 2 group is replaced by a carbonyl, such as —C(O)—CH ⁇ CH—CH 3 and —C(O)—CH 2 —CH ⁇ CH.
  • unsubstituted alkynylene refers to a divalent unsubstituted alkynyl group as defined above.
  • A-C ⁇ C— is an example of an unsubstituted alkynylene.
  • substituted alkynylene refers to a divalent substituted alkynyl group as defined above.
  • unsubstituted aralkyl refers to unsubstituted alkyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkyl group is replaced with a bond to an aryl group as defined above.
  • methyl —CH 3
  • a hydrogen atom of the methyl group is replaced by a bond to a phenyl group, such as if the carbon of the methyl were bonded to a carbon of benzene, then the compound is an unsubstituted aralkyl group (i.e., a benzyl group).
  • the phrase includes, but is not limited to, groups such as benzyl, diphenylmethyl, and 1-phenylethyl (—CH(C 6 H 5 )(CH 3 )).
  • substituted aralkyl has the same meaning with respect to unsubstituted aralkyl groups that substituted aryl groups have with respect to unsubstituted aryl groups.
  • substituted aralkyls also include groups in which a carbon or hydrogen bond of the alkyl part of the group is replaced by a bond to a non-carbon or a non-hydrogen atom. Examples of substituted aralkyl groups include, but are not limited to, —CH 2 C( ⁇ O)(C 6 Hs), and —CH 2 (2-methylphenyl).
  • unsubstituted aralkenyl refers to unsubstituted alkenyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkenyl group is replaced with a bond to an aryl group as defined above.
  • vinyl is an unsubstituted alkenyl group. If a hydrogen atom of the vinyl group is replaced by a bond to a phenyl group, such as if a carbon of the vinyl were bonded to a carbon of benzene, then the compound is an unsubstituted aralkenyl group (i.e., a styryl group).
  • the phrase includes, but is not limited to, groups such as styryl, diphenylvinyl, and 1-phenylethenyl (—C(C 6 H 5 )(CH 2 )).
  • substituted aralkenyl has the same meaning with respect to unsubstituted aralkenyl groups that substituted aryl groups have with respect to unsubstituted aryl groups.
  • a substituted aralkenyl group also includes groups in which a carbon or hydrogen bond of the alkenyl part of the group is replaced by a bond to a non-carbon or a non-hydrogen atom. Examples of substituted aralkenyl groups include, but are not limited to, —CH ⁇ C(Cl)(C 6 H 5 ), and —CH ⁇ CH(2-methylphenyl).
  • unsubstituted aralkynyl refers to unsubstituted alkynyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkynyl group is replaced with a bond to an aryl group as defined above.
  • acetylene is an unsubstituted alkynyl group. If a hydrogen atom of the acetylene group is replaced by a bond to a phenyl group, such as if a carbon of the acetylene were bonded to a carbon of benzene, then the compound is an unsubstituted aralkynyl group.
  • the phrase includes, but is not limited to, groups such as —C ⁇ -phenyl and —CH 2 —C ⁇ C-phenyl.
  • substituted aralkynyl has the same meaning with respect to unsubstituted aralkynyl groups that substituted aryl groups have with respect to unsubstituted aryl groups.
  • a substituted aralkynyl group also includes groups in which a carbon or hydrogen bond of the alkynyl part of the group is replaced by a bond to a non-carbon or a non-hydrogen atom. Examples of substituted aralkynyl groups include, but are not limited to, —C ⁇ C—C(Br)(C 6 H 5 ) and —C ⁇ C(2-methylphenyl).
  • unsubstituted heteroalkyl refers to unsubstituted alkyl groups as defined above in which the carbon chain is interrupted by one or more heteroatoms chosen from N, O, and S.
  • Unsubstituted heteroalkyls containing N may have NH or N (unsubstituted alkyl) in the carbon chain.
  • unsubstituted heteroalkyls include alkoxy, alkoxyalkyl, alkoxyalkoxy, thioether, alkylaminoalkyl, aminoalkyloxy, and other such groups.
  • unsubstituted heteroalkyl groups contain 1-5 heteroatoms, and particularly 1-3 heteroatoms.
  • unsubstituted heteroalkyls include, for example, alkoxyalkoxyalkoxy groups such as ethyloxyethyloxyethyloxy.
  • substituted heteroalkyl has the same meaning with respect to unsubstituted heteroalkyl groups that substituted alkyl groups have with respect to unsubstituted alkyl groups.
  • unsubstituted heteroalkylene refers to a divalent unsubstituted heteroalkyl group as defined above.
  • —CH 2 —O—CH 2 — and —CH 2 —NH—CH 2 CH 2 — are both exemplary unsubstituted heteroalkylenes.
  • substituted heteroalkylene refers to a divalent substituted heteroalkyl group As defined above.
  • unsubstituted heteroalkenyl refers to unsubstituted alkene groups as defined above in which the carbon chain is interrupted by one or more heteroatoms chosen from N, O, and S. Unsubstituted heteroalkenyls containing N may have NH or N (unsubstituted alkyl or alkene) in the carbon chain.
  • substituted heteroalkenyl has the same meaning with respect to unsubstituted heteroalkenyl groups that substituted heteroalkyl groups have with respect to unsubstituted heteroalkyl groups.
  • unsubstituted heteroalkenylene refers to a divalent unsubstituted heteroalkenyl group as defined above.
  • —CH 2 —O—CH ⁇ CH— is an example of an unsubstituted heteroalkenylene.
  • substituted heteroalkenylene refers to a divalent substituted heteroalkenyl group as defined above.
  • unsubstituted heteroalkynyl refers to unsubstituted alkynyl groups as defined above in which the carbon chain is interrupted by one or more heteroatoms chosen from N, O, and S.
  • Unsubstituted heteroalkynyls containing N may have NH or N (unsubstituted alkyl, alkene, or alkyne) in the carbon chain.
  • substituted heteroalkynyl has the same meaning with respect to unsubstituted heteroalkynyl groups that substituted heteroalkyl groups have with respect to unsubstituted heteroalkyl groups.
  • unsubstituted heteroalkynylene refers to a divalent unsubstituted heteroalkynyl group as defined above.
  • —CH 2 —O—CH 2 —C ⁇ C— is an example of an unsubstituted heteroalkynylene.
  • substituted heteroalkynylene refers to a divalent substituted heteroalkynyl group as defined above.
  • unsubstituted heterocyclyl refers to both aromatic and nonaromatic ring compounds including monocyclic, bicyclic, and polycyclic ring compounds such as, but not limited to, quinuclidyl, containing 3 or more ring members of which one or more is a heteroatom such as, but not limited to, N, O, and S.
  • unsubstituted heterocyclyl includes condensed heterocyclic rings such as benzimidazolyl, it does not include heterocyclyl groups that have other groups such as alkyl or halo groups bonded to one of the ring members as compounds such as 2-methylbenzimidazolyl are substituted heterocyclyl groups.
  • heterocyclyl groups include, but are not limited to: unsaturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, dihydropyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g., 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl etc.), tetrazolyl, (e.g., 1H-tetrazolyl, 2H tetrazolyl, etc.); saturated 3 to 8 membered rings containing 1 to 4 nitrogen atoms such as, but not limited to, pyrrolidinyl, imidazolidinyl, piperidinyl, piperazinyl; condensed unsaturated heterocyclic groups containing
  • Heterocyclyl group also include those described above in which one or more S atoms in the ring is double-bonded to one or two oxygen atoms (sulfoxides and sulfones).
  • heterocyclyl groups include tetrahydrothiophene, tetrahydrothiophene oxide, and tetrahydrothiophene 1,1-dioxide. In some embodiments heterocyclyl groups contain 5 or 6 ring members.
  • heterocyclyl groups include morpholine, piperazine, piperidine, pyrrolidine, imidazole, pyrazole, 1,2,3-triazole, 1,2,4-triazole, tetrazole, thiomorpholine, thiomorpholine in which the S atom of the thiomorpholine is bonded to one or more O atoms, pyrrole, homopiperazine, oxazolidin-2-one, pyrrolidin-2-one, oxazole, quinuclidine, thiazole, isoxazole, furan, and tetrahydrofuran.
  • substituted heterocyclyl refers to an unsubstituted heterocyclyl group as defined above in which one of the ring members is bonded to a non-hydrogen atom such as described above with respect to substituted alkyl groups and substituted aryl groups. Examples include, but are not limited to, 2-methylbenzimidazolyl, 5-methylbenzimidazolyl, 5-chlorobenzthiazolyl, 1-methyl piperazinyl, and 2-chloropyridyl.
  • unsubstituted heteroaryl refers to unsubstituted aromatic heterocyclyl groups as defined above.
  • unsubstituted heteroaryl groups include but are not limited to furyl, imidazolyl, oxazolyl, isoxazolyl, pyridinyl, benzimidazolyl, and benzothiazolyl.
  • substituted heteroaryl refers to substituted aromatic heterocyclyl groups as defined above.
  • unsubstituted heterocyclylalkyl refers to unsubstituted alkyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkyl group is replaced with a bond to a heterocyclyl group as defined above.
  • methyl —CH 3
  • a hydrogen atom of the methyl group is replaced by a bond to a heterocyclyl group, such as if the carbon of the methyl were bonded to carbon 2 of pyridine (one of the carbons bonded to the N of the pyridine) or carbons 3 or 4 of the pyridine, then the compound is an unsubstituted heterocyclylalkyl group.
  • substituted heterocyclylalkyl has the same meaning with respect to unsubstituted heterocyclylalkyl groups that substituted aralkyl groups have with respect to unsubstituted aralkyl groups.
  • a substituted heterocyclylalkyl group also includes groups in which a non-hydrogen atom is bonded to a heteroatom in the heterocyclyl group of the heterocyclylalkyl group such as, but not limited to, a nitrogen atom in the piperidine ring of a piperidinylalkyl group.
  • unsubstituted heterocyclylalkenyl refers to unsubstituted alkenyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkenyl group is replaced with a bond to a heterocyclyl group as defined above.
  • vinyl is an unsubstituted alkenyl group. If a hydrogen atom of the vinyl group is replaced by a bond to a heterocyclyl group, such as if the carbon of the vinyl were bonded to carbon 2 of pyridine or carbons 3 or 4 of the pyridine, then the compound is an unsubstituted heterocyclylalkenyl group.
  • substituted heterocyclylalkenyl has the same meaning with respect to unsubstituted heterocyclylalkenyl groups that substituted aralkenyl groups have with respect to unsubstituted aralkenyl groups.
  • a substituted heterocyclylalkenyl group also includes groups in which a non-hydrogen atom is bonded to a heteroatom in the heterocyclyl group of the heterocyclylalkenyl group such as, but not limited to, a nitrogen atom in the piperidine ring of a piperidinylalkenyl group.
  • unsubstituted heterocyclylalkynyl refers to unsubstituted alkynyl groups as defined above in which a hydrogen or carbon bond of the unsubstituted alkynyl group is replaced with a bond to a heterocyclyl group as defined above.
  • acetylene is an unsubstituted alkynyl group. If a hydrogen atom of the acetylene group is replaced by a bond to a heterocyclyl group, such as if the carbon of the acetylene were bonded to carbon 2 of pyridine or carbons 3 or 4 of the pyridine, then the compound is an unsubstituted heterocyclylalkynyl group.
  • substituted heterocyclylalkynyl has the same meaning with respect to unsubstituted heterocyclylalkynyl groups that substituted aralkynyl groups have with respect to unsubstituted aralkynyl groups.
  • a substituted heterocyclylalkynyl group also includes groups in which a non-hydrogen atom is bonded to a heteroatom in the heterocyclyl group of the heterocyclylalkynyl group such as, but not limited to, a nitrogen atom in the piperidine ring of a piperidinylalkynyl group.
  • unsubstituted alkoxy refers to a hydroxyl group (—OH) in which the bond to the hydrogen atom is replaced by a bond to a carbon atom of an otherwise unsubstituted alkyl group as defined above.
  • substituted alkoxy refers to a hydroxyl group (—OH) in which the bond to the hydrogen atom is replaced by a bond to a carbon atom of an otherwise substituted alkyl group as defined above.
  • a “pharmaceutically acceptable salt” includes a salt with an inorganic base, organic base, inorganic acid, organic acid, or basic or acidic amino acid.
  • Salts of inorganic bases include, for example, alkali metals such as sodium or potassium; alkaline earth metals such as calcium and magnesium or aluminum; and ammonia.
  • Salts of organic bases include, for example, trimethylamine, triethylamine, pyridine, picoline, ethanolamine, diethanolamine, and triethanolamine.
  • Salts of inorganic acids include for example, hydrochloric acid, hydroboric acid, nitric acid, sulfuric acid, and phosphoric acid.
  • Salts of organic acids include for example, formic acid, acetic acid, trifluoroacetic acid, fumaric acid, oxalic acid, tartaric acid, maleic acid, citric acid, succinic acid, malic acid, methanesulfonic acid, benzenesulfonic acid, and p-toluenesulfonic acid.
  • Salts of basic amino acids include, for example, arginine, lysine and ornithine.
  • Acidic amino acids include, for example, aspartic acid and glutamic acid.
  • Tautomers refers to isomeric forms of a compound that are in equilibrium with each other.
  • concentrations of the isomeric forms will depend on the environment the compound is found in and may be different depending upon, for example, whether the compound is a solid or is in an organic or aqueous solution.
  • ketones are typically in equilibrium with their enol forms.
  • ketones and their enols are referred to as tautomers of each other.
  • tautomers of each other As readily understood by one skilled in the art, a wide variety of functional groups and other structures may exhibit tautomerism, and all tautomers of compounds of Formulas I, II, and III are within the scope of the present invention.
  • the compounds according to the invention may be solvated, especially hydrated. Hydration may occur during manufacturing of the compounds or compositions comprising the compounds, or the hydration may occur over time due to the hygroscopic nature of the compounds.
  • prodrugs denotes a derivative of a pharmaceutically or therapeutically active drug, e.g., esters and amides, wherein the derivative has an enhanced characteristic such as, for example, enhanced delivery and therapeutic value as compared to the drug and can be transformed into the drug by an enzymatic or chemical process.
  • an enhanced characteristic such as, for example, enhanced delivery and therapeutic value as compared to the drug and can be transformed into the drug by an enzymatic or chemical process.
  • the prodrug may be designed to alter the metabolic stability or transport characteristics of a drug, mask side effects or toxicity of a drug, improve the flavor of a drug, or to alter other characteristics or properties of a drug.
  • Compounds of the present invention include enriched or resolved optical isomers at any or all asymmetric atoms as are apparent from the depictions. Both racemic and diastereomeric mixtures, as well as the individual optical isomers can be isolated or synthesized so as to be substantially free of their enantiomeric or diastereomeric partners. All such stereoisomers are within the scope of the invention.
  • carboxy protecting group refers to a carboxylic acid protecting ester group employed to block or protect the carboxylic acid functionality while the reactions involving other functional sites of the compound are carried out.
  • Carboxy protecting groups are disclosed in, for example, Greene, Protective Groups in Organic Synthesis , pp. 152-186, John Wiley & Sons, New York (1981), which is hereby incorporated herein by reference.
  • a carboxy protecting group can be used as a prodrug, whereby the carboxy protecting group can be readily cleaved in vivo by, for example, enzymatic hydrolysis to release the biologically active parent.
  • esters useful as prodrugs for compounds containing carboxyl groups can be found, for example, at pp. 14-21 in Bioreversible Carriers in Drug Design: Theory and Application (E. B. Roche, ed.), Pergamon Press, New York (1987), which is hereby incorporated herein by reference.
  • carboxy protecting groups are C 1 to C 8 alkyl (e.g., methyl, ethyl or tertiary butyl and the like); haloalkyl; alkenyl; cycloalkyl and substituted derivatives thereof such as cyclohexyl, cyclopentyl and the like; cycloalkylalkyl and substituted derivatives thereof such as cyclohexylmethyl, cyclopentylmethyl and the like; arylalkyl, for example, phenethyl or benzyl and substituted derivatives thereof such as alkoxybenzyl or nitrobenzyl groups and the like; arylalkenyl, for example, phenylethenyl and the like; aryl and substituted derivatives thereof, for example, 5-indanyl and the like; dialkylaminoalkyl (e.g., dimethylaminoethyl and the like); alkanoyloxyalkyl groups such as acetoxy
  • N-protecting group or “N-protected” as used herein refers to those groups intended to protect the N-terminus of an amino acid or peptide or to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in, for example, Greene, Protective Groups in Organic Synthesis , John Wiley & Sons, New York (1981), which is hereby incorporated by reference.
  • N-protecting groups can comprise acyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and the like; sulfonyl groups such as benzenesulfonyl, p-toluenesulfonyl and the like; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-brom
  • N-protecting groups are formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, phenylsulfonyl, benzyl, 9-fluorenylmethyloxycarbonyl (Fmoc), t-butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).
  • halo refers to F, Cl, Br or I.
  • substantially pure means sufficiently homogeneous to appear free of readily detectable impurities as determined by standard methods of analysis, such as thin layer chromatography (TLC), gel electrophoresis, and high performance liquid chromatography (HPLC), used by those of skill in the art to assess such purity, or sufficiently pure such that further purification would not detectably alter the physical and chemical properties, such as enzymatic and biological activities, of the substance.
  • Substantially pure includes compositions in which the AA targeting agent or AA targeting compound forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, or about 95% or more of the substances in the composition.
  • a substantially chemically pure compound may, however, be a mixture of stereoisomers. In such instances, further purification may increase the specific activity of the compound.
  • AA targeting agents need not always be provided in a specific purified state. Partially purified compositions will have utility in certain embodiments and depending on the desired use. For example, purification methods that may yield a greater total recovery of AA-targeting agent may produce a lower degree of relative purification.
  • biological activity refers to the in vivo activities of a compound, composition, or other mixture, or physiological responses that result upon in vivo administration of a compound, composition or other mixture. Biological activity thus encompasses therapeutic effects, diagnostic effects and pharmaceutical activity of such compounds, compositions, and mixtures.
  • biologically active or “functional” when used as a modifier of invention AA targeting agent containing polypeptides or compositions thereof refers to a polypeptide that exhibits at least one activity that is characteristic of or similar to an AA targeting agent.
  • pharmacokinetics refers to the concentration of an administered compound in the serum over time.
  • Pharmacodynamics refers to the concentration of an administered compound in target and nontarget tissues over time and the effects on the target tissue (e.g., efficacy) and the non-target tissue (e.g., toxicity). Improvements in, for example, pharmacokinetics or pharmacodynamics can be designed for a particular targeting agent or biological agent, such as by using labile linkages or by modifying the chemical nature of any linker (e.g., changing solubility, charge, and the like).
  • an effective amount and “therapeutically effective amount” refer to an amount of an AA targeting agent or compound comprising an AA targeting agent that is useful or able to support an observable change in the level of one or more biological activity characteristic of an AA targeting agent, or a dose sufficient to impart a beneficial effect, e.g., an amelioration of a symptom on the recipient thereof.
  • a therapeutically effective dose level for any particular subject will depend upon a variety of factors including the symptom or disorder being treated, the severity of the symptom or disorder, the activity of the specific compound, the route of administration, the rate of clearance of the compound, the duration of treatment, the drugs used in combination or coincident with the compound, the age, body weight, sex, diet, and general health of the subject, and like, as well as other factors well known in the medical arts and sciences.
  • a therapeutically effective amount can be an amount of AA targeting compound sufficient to produce a measurable inhibition of angiogenesis in the tissue being treated, i.e., an angiogenesis-inhibiting amount. Inhibition of angiogenesis can be measured in situ by immunohistochemistry, or by other methods known to one skilled in the art.
  • the present invention provides various targeting compounds in which AA targeting agents are covalently linked to a combining site of an antibody.
  • the present invention includes methods of altering at least one physical or biological characteristic of an AA targeting agent.
  • the methods include covalently linking an AA targeting agent to a combining site of an antibody, either directly or though a linker.
  • Characteristics of an AA targeting agent that may be modified include, but are not limited to, binding affinity, susceptibility to degradation (e.g., by proteases), pharmacokinetics, pharmacodynamics, immunogenicity, solubility, lipophilicity, hydrophilicity, hydrophobicity, stability (either more or less stable, as well as planned degradation), rigidity, flexibility, modulation of antibody binding, and the like.
  • the biological potency of a particular AA targeting agent may be increased by the addition of the effector function(s) provided by the antibody.
  • an antibody provides effector functions such as complement mediated effector functions.
  • the antibody portion of an AA targeting compound may generally extend the half-life of a smaller sized AA targeting agent in vivo.
  • the invention provides a method for increasing the effective circulating half-life of an AA targeting agent.
  • the present invention provides methods for modulating the binding activity of an antibody by covalently attaching an AA targeting agent to a combining site of the antibody.
  • substantially reduced antibody binding to an antigen may result from the linked AA targeting agent(s) sterically hindering the antigen from contacting the antibody combining site.
  • substantially reduced antigen binding may result if the amino acid side chain of the antibody combining site modified by covalent linkage is important for binding to the antigen.
  • substantially increased antibody binding to an antigen may result when a linked AA targeting agent(s) does not sterically hinder the antigen from contacting the antibody combining site and/or when the amino acid side chain of the antibody combining site modified by covalent linkage is not important for binding to the antigen.
  • the present invention includes methods of modifying a combining site of an antibody to generate binding specificity for the thrombospondin binding cognate.
  • Such methods include covalently linking a reactive amino acid side chain in a combining site of the antibody to a chemical moiety on a linker of an AA targeting agent-linker compound as described herein where an AA targeting agent is based upon a thrombospondin peptide.
  • the chemical moiety of the linker is sufficiently distanced from the AA targeting agent so that an AA targeting agent can bind its cognate when an AA targeting agent-linker compound is covalently linked to an antibody combining site.
  • the antibody will not be considered specific for the target molecule.
  • an antibody prior to covalent linking would have an affinity for the thrombospondin binding cognate of less than about 1 ⁇ 10 ⁇ 5 moles/liter.
  • the modified antibody preferably has an affinity for the target molecule of at least about 1 ⁇ 10 ⁇ 6 moles/liter, alternatively, at least about 1 ⁇ 10 ⁇ 7 moles/liter, alternatively, at least 1 ⁇ 10 ⁇ 8 moles/liter, alternatively at least 1 ⁇ 10 ⁇ 9 moles/liter, or alternatively, at least about 1 ⁇ 10 ⁇ 10 moles/liter.
  • An AA targeting agent is a peptide selected from the group consisting of:
  • R 2 is NH 2 , NHC(O)CH 3 , NHC(O)CH 2 CH 3 , NHC(O)CH 2 CH 2 CH 3 , NHC(O)CH(CH 3 )CH 3 , NHC(O)CH 2 CH 2 CH 2 CH 3 , NHC(O)CH(CH 3 )CH 2 CH 3 , NHC(O)C 6 H 5 , NH(CH 3 )C(O)CH 2 CH 2 (CH 2 CH 2 O) 1-5 Me, an amino protecting group, a lipid fatty acid group or a carbohydrate; and
  • An AA targeting compound can be prepared using techniques well known in the art. Typically, synthesis of the peptidyl AA targeting agent is the first step and is carried out as described herein. The targeting agent is then derivatized for linkage to a connecting component (the linker), which is then combined with the antibody.
  • the linker a connecting component
  • One of skill in the art will readily appreciate that the specific synthetic steps used depend upon the exact nature of the three components. Thus, AA targeting agent-linker conjugates and AA targeting compounds described herein can be readily synthesized.
  • AA targeting agent peptides may be synthesized by many techniques that are known to those skilled in the art. For solid phase peptide synthesis, a summary of exemplary techniques may be found in Chemical Approaches to the Synthesis of Peptides and Proteins (Williams et al., eds.), CRC Press, Boca Raton, Fla. (1997).
  • the desired peptidic AA targeting agent is synthesized sequentially on solid phase according to procedures well known in the art. See, e.g., U.S. Patent Application No. 2003/0045477).
  • the linker may be attached to the peptide in part or in full on the solid phase, or may be added using solution phase techniques after the removal of the peptide from the resin (see FIGS. 6A and 6B ).
  • an N-protected amino and carboxylic acid-containing linking moiety may be attached to a resin such as 4-hydroxymethyl-phenoxymethyl-poly(styrene-1% divinylbenzene).
  • the N-protecting group may be removed by the appropriate acid (e.g., TFA for Boc) or base (e.g., piperidine for Fmoc), and the peptide sequence developed in the normal C-terminus to N-terminus fashion (see FIG. 6A ).
  • the peptide sequence may be synthesized first and the linker added to the N-terminal amino acid residue last (see FIG. 6B ).
  • Yet another method entails deprotecting an appropriate side chain during synthesis and derivatizing with a suitably reactive linker. For example, a lysine side chain may be deprotected and reacted with a linker having an active ester.
  • an amino acid derivative with a suitably protected linker moiety already attached to the side chain see FIG. 6B ) or, in some cases, the alpha-amino nitrogen, may be added as part of the growing peptide sequence.
  • the targeting agent-linker conjugate is removed from the resin and deprotected, either in succession or in a single operation. Removal of the targeting agent-linker conjugate and deprotection can be accomplished in a single operation by treating the resin-bound peptide-linker conjugate with a cleavage reagent, for example, trifluoroacetic acid containing scavengers such as thianisole, water, or ethanedithiol. After deprotection and release of the targeting agent, further derivatization of the targeting agent peptide may be carried out.
  • a cleavage reagent for example, trifluoroacetic acid containing scavengers such as thianisole, water, or ethanedithiol.
  • the fully deprotected targeting agent-linker conjugate is purified by a sequence of chromatographic steps employing any or all of the following types: ion exchange on a weakly basic resin in the acetate form; hydrophobic adsorption chromatography on underivatized polystyrene-divinylbenzene (e.g., AMBERLITE XAD); silica gel adsorption chromatography; ion exchange chromatography on carboxymethylcellulose; partition chromatography, e.g., on SEPHADEX G-25, LH-20 or countercurrent distribution; high performance liquid chromatography (HPLC), especially reverse-phase HPLC on octyl- or octadecylsilyl-silica bonded phase column packing.
  • HPLC high performance liquid chromatography
  • Antibody as used herein includes polypeptide molecules comprising heavy and/or light chains which have immunoreactive activity. Antibodies include immunoglobulins which are the product of B cells and variants thereof, as well as the T cell receptor (TcR) which is the product of T cells and variants thereof.
  • An immunoglobulin is a protein comprising one or more polypeptides substantially encoded by the immunoglobulin kappa and lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda.
  • Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD, and IgE, respectively. Subclasses of heavy chains are also known. For example, IgG heavy chains in humans can be any of IgG1, IgG2, IgG3, and IgG4 subclasses.
  • a typical immunoglobulin structural unit is known to comprise a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD).
  • the N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the terms variable light chain (V L ) and variable heavy chain (V H ) refer to these light and heavy chains respectively.
  • the amino acids of an antibody may be naturally or nonnaturally occurring.
  • Antibodies that contain two combining sites are bivalent in that they have two complementarity or antigen recognition sites.
  • a typical natural bivalent antibody is an IgG.
  • vertebrate antibodies generally comprise two heavy chains and two light chains, heavy chain only antibodies are also known. See Muyldermans et al., TRENDS in Biochem. Sci. 26(4):230-235 (1991). Such antibodies are bivalent and are formed by the pairing of heavy chains.
  • Antibodies may also be multi-valent, as in the case of dimeric forms of IgA and the pentameric IgM molecule.
  • Antibodies also include hybrid antibodies wherein the antibody chains are separately homologous with referenced mammalian antibody chains.
  • One pair of heavy and light chain has a combining site specific to one antigen and the other pair of heavy and light chains has a combining site specific to a different antigen.
  • Such antibodies are referred to as bi-specific because they are able to bind two different antigens at the same time.
  • Antibodies may also be univalent, such as, for example, in the case of Fab or Fab′ fragments.
  • Antibodies exist as full length intact antibodies or as a number of well-characterized fragments produced by digestion with various peptidases or chemicals.
  • pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab′) 2 , a dimer of Fab which itself is a light chain joined to V H —CH 1 by a disulfide bond.
  • F(ab′) 2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab′) 2 dimer into a Fab′ monomer.
  • the Fab′ monomer is essentially a Fab fragment with part of the hinge region (see, e.g., Fundamental Immunology (W. E.
  • antibody fragments are defined in terms of the digestion of an intact antibody, one of skill in the art will appreciate that any of a variety of antibody fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology.
  • the term antibody as used herein also includes antibody fragments produced by the modification of whole antibodies, synthesized de novo, or obtained from recombinant DNA methodologies.
  • the smaller size of the antibody fragments allows for rapid clearance and may lead to improved access to solid tumors.
  • Recombinant antibodies may be conventional full length antibodies, hybrid antibodies, heavy chain antibodies, antibody fragments known from proteolytic digestion, antibody fragments such as Fv or single chain Fv (scFv), single domain fragments such as V H or V L , diabodies, domain deleted antibodies, minibodies, and the like.
  • An Fv antibody is about 50 kD in size and comprises the variable regions of the light and heavy chain.
  • the light and heavy chains may be expressed in bacteria where they assemble into an Fv fragment. Alternatively, the two chains can be engineered to form an interchain disulfide bond to give a dsFv.
  • a single chain Fv (“scFv”) is a single polypeptide comprising V H and V L sequence domains linked by an intervening linker sequence, such that when the polypeptide folds the resulting tertiary structure mimics the structure of the antigen binding site.
  • scFv single chain Fv
  • One skilled in the art will recognize that depending on the particular expression method and/or antibody molecule desired, appropriate processing of the recombinant antibodies may be performed to obtain a desired reconstituted or reassembled antibody. See, e.g., Vallejo and Rinas, Biomed Central., available at world wide web URL microbialcellfactories.com/content/3/1/11.
  • Single domain antibodies are the smallest functional binding units of antibodies (approximately 13 kD in size), corresponding to the variable regions of either the heavy V H or light V L chains. See U.S. Pat. No. 6,696,245, WO04/058821, WO04/003019 and WO03/002609. Single domain antibodies are well expressed in bacteria, yeast, and other lower eukaryotic expression systems. Domain deleted antibodies have a domain, such as CH 2 , deleted relative to the full length antibody. In many cases such domain deleted antibodies, particularly CH 2 deleted antibodies, offer improved clearance relative to their full length counterparts. Diabodies are formed by the association of a first fusion protein comprising two V H domains with a second fusion protein comprising two V L domains.
  • Diabodies like full length antibodies, are bivalent and may be bi-specific.
  • Minibodies are fusion proteins comprising a V H , V L , or scFv linked to CH 3 , either directly or via an intervening IgG hinge. See T. Olafsen et al., Protein Eng. Des. Sel. 17:315-323 (2004).
  • Minibodies, like domain deleted antibodies, are engineered to preserve the binding specificity of full-length antibodies but with improved clearance due to their smaller molecular weight.
  • the T cell receptor is a disulfide linked heterodimer composed of two chains.
  • the two chains are generally disulfide-bonded just outside the T cell plasma membrane in a short extended stretch of amino acids resembling the antibody hinge region.
  • Each TcR chain is composed of one antibody-like variable domain and one constant domain.
  • the full TcR has a molecular mass of about 95 kD, with the individual chains varying in size from 35 to 47 kD.
  • portions of the receptor such as, for example, the variable region, which can be produced as a soluble protein using methods well known in the art. For example, U.S. Pat. No. 6,080,840 and A. E. Slanetz and A. L.
  • soluble T cell receptor prepared by splicing the extracellular domains of a TcR to the glycosyl phosphatidylinositol (GPI) membrane anchor sequences of Thy-1.
  • the molecule is expressed in the absence of CD3 on the cell surface, and can be cleaved from the membrane by treatment with phosphatidylinositol specific phospholipase C(PI-PLC).
  • the soluble TcR also may be prepared by coupling the TcR variable domains to an antibody heavy chain CH 2 or CH 3 domain, essentially as described in U.S. Pat. No. 5,216,132 and G. S.
  • TcR tet al.
  • S. Immunol. Methods 155:175-191 (1992) or as soluble TcR single chains, as described by E. V. Shusta et al., Nat. Biotechnol. 18:754-759 (2000) or P. D. Holler et al., Proc. Natl. Acad. Sci. U.S.A. 97:5387-5392 (2000).
  • Certain embodiments of the invention use TcR “antibodies” as a soluble antibody.
  • the combining site of the TcR can be identified by reference to CDR regions and other framework residues using the same methods discussed above for antibodies.
  • the combining site refers to the part of an antibody molecule that participates in antigen binding.
  • the antigen binding site is formed by amino acid residues of the N-terminal variable (“V”) regions of the heavy (“H”) and light (“L”) chains.
  • the antibody variable regions comprise three highly divergent stretches referred to as “hypervariable regions” or “complementarity determining regions” (CDRs), which are interposed between more conserved flanking stretches known as “framework regions” (FRs).
  • the three hypervariable regions of a light chain (LCDR1, LCDR2, and LCDR3) and the three hypervariable regions of a heavy chain (HCDR1, HCDR2, and HCDR3) are disposed relative to each other in three dimensional space to form an antigen binding surface or pocket.
  • the antigen binding site is formed by the three hypervariable regions of the heavy chains.
  • V L domains the antigen binding site is formed by the three hypervariable regions of the light chain.
  • antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. See E. A. Kabat et al., Sequences of Proteins of Immunological Interest, 5 th ed., Public Health Service, NIH, Washington D.C. (1992).
  • the positions of the CDRs may also be identified as the structural loop structures originally described by Chothia and others. See, e.g., C. Chothia and A. M. Lesk, J. Mol. Biol. 196:901-917 (1987); C.
  • Residue before is always a Cys.
  • Residue after is always a Trp, typically followed by Tyr-Gln, but also followed by Leu-Gln, Phe-Ghn, or Tyr-Leu. Length is 10 to 17 residues.
  • the identity of the amino acid residues in a particular antibody that are outside the CDRs, but nonetheless make up part of the combining site by having a side chain that is part of the lining of the combining site (i.e., that is available to linkage through the combining site), can be determined using methods well known in the art, such as molecular modeling and X-ray crystallography. See, e.g., L. Riechmann et al., Nature 332:323-327 (1988).
  • antibodies that can be used in preparing antibody-based AA targeting compounds require a reactive side chain in the antibody combining site.
  • a reactive side chain may be present naturally or may be placed in an antibody by mutation.
  • the reactive residue of the antibody combining site may be associated with the antibody, such as when the residue is encoded by nucleic acid present in the lymphoid cell first identified to make the antibody.
  • the amino acid residue may arise by purposely mutating the DNA so as to encode the particular residue (see, e.g., WO 01/22922 to Meares et al.).
  • the reactive residue may be a non-natural residue arising, for example, by biosynthetic incorporation using a unique codon, tRNA, and aminoacyl-tRNA as discussed herein.
  • the amino acid residue or its reactive functional groups may be attached to an amino acid residue in the antibody combining site.
  • covalent linkage with the antibody occurring “through an amino acid residue in a combining site of an antibody” as used herein means that linkage can be directly to an amino acid residue of an antibody combining site or through a chemical moiety that is linked to a side chain of an amino acid residue of an antibody combining site.
  • Catalytic antibodies are one source of antibodies with combining sites that comprise one or more reactive amino acid side chains.
  • Such antibodies include aldolase antibodies, beta lactamase antibodies, esterase antibodies, amidase antibodies, and the like.
  • One embodiment comprises an aldolase antibody such as the mouse monoclonal antibody mAb 38C2 or nab 33F12, as well as suitably humanized and chimeric versions of such antibodies.
  • Mouse mAb 38C2 has a reactive lysine near to but outside HCDR3, and is the prototype of a new class of catalytic antibodies that were generated by reactive immunization and mechanistically mimic natural aldolase enzymes. See C. F. Barbas 3 rd et al., Science 278:2085-2092 (1997)).
  • aldolase catalytic antibodies that may be used include the antibodies produced by the hybridoma 85A2, having ATCC accession number PTA-1015; hybridoma 85C7, having ATCC accession number PTA-1014; hybridoma 92F9, having ATCC accession number PTA-1017; hybridoma 93F3, having ATCC accession number PTA-823; hybridoma 84G3, having ATCC accession number PTA-824; hybridoma 84G11, having ATCC accession number PTA-1018; hybridoma 84H9, having ATCC accession number PTA-1019; hybridoma 85H6, having ATCC accession number PTA-825; hybridoma 90G8, having ATCC accession number PTA-1016.
  • AA targeting compounds may also be formed by linking an AA targeting agent to a reactive cysteine, such as those found in the combining sites of thioesterase and esterase catalytic antibodies.
  • a reactive cysteine such as those found in the combining sites of thioesterase and esterase catalytic antibodies.
  • Suitable thioesterase catalytic antibodies are described by K. D. Janda et al., Proc. Natl. Acad. Sci. U.S.A. 91:2532-2536 (1994).
  • Suitable esterase antibodies are described by P. Wirsching et al., Science 270:1775-1782 (1995).
  • Reactive amino acid-containing antibodies may be prepared by means well known in the art, including mutating an antibody combining site residue to encode for the reactive amino acid or chemically derivatizing an amino acid side chain in an antibody combining site with a linker that contains the reactive group.
  • Antibodies suitable for use herein may be obtained by conventional immunization, reactive immunization in vivo, or by reactive selection in vitro, such as with phage display. Antibodies may also be obtained by hybridoma or cell fusion methods or in vitro host cells expression system. Antibodies may be produced in humans or in other animal species. Antibodies from one species of animal may be modified to reflect another species of animal. For example, human chimeric antibodies are those in which at least one region of the antibody is from a human immunoglobulin.
  • a human chimeric antibody is typically understood to have variable region amino acid sequences homologous to a non-human animal, e.g., a rodent, with the constant region having amino acid sequence homologous to a human immunoglobulin
  • a humanized antibody uses CDR sequences from a non-human antibody with most or all of the variable framework region sequence and all the constant region sequence from a human immunoglobulin.
  • Chimeric and humanized antibodies may be prepared by methods well known in the art including CDR grafting approaches (see, e.g., N. Hardman et al., Int. J. Cancer 44:424-433 (1989); C. Queen et al., Proc. Natl. Acad. Sci. U.S.A.
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is nonhuman. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the methods of Winter and colleagues (see, e.g., P. T. Jones et al., Nature 321:522-525 (1986); L. Riechmann et al., Nature 332:323-327 (1988); M.
  • humanized antibodies are chimeric antibodies wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies are typically human antibodies in which some hypervariable region residues and possibly some framework (FR) residues are substituted by residues from analogous sites in rodent antibodies.
  • human variable domains both light and heavy
  • HAMA human anti-mouse antibody
  • the human variable domain utilized for humanization is selected from a library of known domains based on a high degree of homology with the rodent variable region of interest (M. J. Sims et al., J. Immunol., 151:2296-2308 (1993); M. Chothia and A. M. Lesk, J. Mol. Biol. 196:901-917 (1987)).
  • Another method uses a framework region derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains.
  • the same framework may be used for several different humanized antibodies (see, e.g., P. Carter et al., Proc. Natl. Acad. Sci. U.S.A. 89:4285-4289 (1992); L. G. Presta et al., J. Immunol., 151:2623-2632 (1993)).
  • humanized antibodies are prepared by analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence with respect to linking to the Z group. In this way, FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved.
  • humanized murine aldolase antibodies are contemplated.
  • One embodiment uses the humanized aldolase catalytic antibody h38c2 IgG1 or h38c2 Fab with human constant domains C ⁇ and C ⁇ 1 1.
  • C. Rader et al., J. Mol. Bio. 332:889-899 (2003) discloses the gene sequences and vectors that may be used to produce h38c2 Fab and h38c2 IgG1.
  • FIG. 7A illustrates a sequence alignment between the variable light and heavy chains in m38c2 (SEQ ID NOs: 32 and 33, respectively), h38c2 (SEQ ID NOs: 34 and 35, respectively), and human germlines.
  • h38c2 may utilize IgG1, IgG2, IgG3, or IgG4 constant domains, including any of the allotypes thereof.
  • FIG. 7B illustrates one embodiment of h38c2 IgG1 using the Glm(f) allotype.
  • the light and heavy chain amino acid sequences of this h38c2 IgG1 are set forth in SEQ ID NOs:40 and 41, respectively.
  • Z binds to the side chain of the lysine residue at position 99 of SEQ ID NO:41. This residue is denoted by bold print in FIG. 7B .
  • Another embodiment uses a chimeric antibody comprising the variable domains (V L and V H ) of h38c2 and the constant domains from an IgG1, IgG2, IgG3, or IgG4.
  • h38c2 F(ab′) 2 may be produced by the proteolytic digestion of h38c2 IgG1.
  • Another embodiment uses an h38c2 scFv comprising the V L and V H domains from h38c2 which are optionally connected by the intervening linker (Gly 4 Ser) 3 .
  • human antibodies can be generated.
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • immunization or reactive immunization in the case of catalytic antibodies
  • J H antibody heavy-chain joining region
  • phage display technology can be used to produce human antibodies and antibody fragments in vitro using immunoglobulin variable (V) domain gene repertoires from unimmunized donors.
  • antibody V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M13 or fd, and displayed as functional antibody fragments on the surface of the phage particle.
  • a filamentous bacteriophage such as M13 or fd
  • the filamentous particle contains a single-stranded DNA copy of the phage genome
  • selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties.
  • the phage mimics some of the properties of the B-cell.
  • Phage display can be performed in a variety of formats, and is reviewed in, e.g., K. S. Johnson and D. J. Chiswell, Curr. Opin. Struct. Biol. 3:564-571 (1993).
  • V-gene segments can be used for phage display.
  • T. Clackson et al., Nature, 352:624-628 (1991) isolated a diverse array of anti-oxazolone antibodies from a small random combinatorial library of V genes derived from the spleens of immunized mice.
  • a repertoire of V genes from unimmunized human donors can be constructed and antibodies to a diverse array of antigens (including self-antigens) can be isolated essentially following the techniques described by J. D. Marks et al., J. Mol. Biol. 222:581-597 (1991) or A. D. Griffiths et al., EMBO J. 12:725-734 (1993). See also U.S. Pat. Nos. 5,565,332 and 5,573,905; and L. S. Jespers et al., Biotechnology 12:899-903 (1994).
  • human antibodies may also be generated by in vitro activated B cells. See, e.g., U.S. Pat. Nos. 5,567,610 and 5,229,275; and C. A. K. Borrebaeck et al., Proc. Natl. Acad. Sci. U.S.A. 85:3995-3999 (1988).
  • Amino acid sequence modification(s) of the antibodies described 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 are prepared by introducing appropriate nucleotide changes into the antibody nucleic acid, or by peptide synthesis. Such modifications include, for example, deletions from, insertions into, and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution is made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • the amino acid changes also may alter post-translational processes of the antibody, such as changing the number or position of glycosylation sites.
  • a useful method for identification of certain residues or regions of an antibody that are preferred locations for mutagenesis is called “alanine scanning mutagenesis,” as described in B. C. Cunningham and J. A. Wells, Science 244:1081-1085 (1989).
  • a residue or group of target residues are identified (e.g., charged residues such as Arg, Asp, His, Lys, and Glu) and replaced by a neutral or negatively charged amino acid (most preferably Ala or Polyalanine) to affect the interaction of the amino acids with the Z group of the linker.
  • Those amino acid locations demonstrating functional sensitivity to the substitutions are then refined by introducing further or other variants at, or for, the sites of substitution.
  • the site for introducing an amino acid sequence variation is predetermined, the nature of the mutation per se need not be predetermined.
  • alanine scanning or random mutagenesis is conducted at the target codon or region and the expressed antibody variants are screened for the ability to form a covalent bond with Z.
  • 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 or the antibody fused to a cytotoxic polypeptide.
  • Other insertional variants of an antibody molecule include the fusion to the N- or C-terminus of an anti-antibody to an enzyme or a polypeptide which increases the serum half-life of the antibody.
  • variants are an amino acid substitution variant. These variants have at least one amino acid residue in an antibody molecule replaced by a different residue.
  • the sites of greatest interest for substitutional mutagenesis include the hypervariable regions, but FR alterations are also contemplated. Conservative substitutions are shown in Table 3 below under the heading of “preferred substitutions.” If such substitutions result in a change in biological activity, then more substantial changes, denominated “exemplary substitutions” as further described below in reference to amino acid classes, may be introduced and the products screened.
  • Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • Naturally occurring residues are divided into groups based on common side-chain properties:
  • Non-conservative substitutions will entail exchanging a member of one of these classes for a member of another class.
  • cysteine residues not involved in maintaining the proper conformation of the antibody may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking.
  • cysteine bond(s) may be added to the antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment).
  • 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 development will have improved biological properties relative to the parent antibody from which they are generated.
  • a convenient way for generating such substitutional variants involves affinity maturation using phage display. Briefly, several hypervariable region sites (e.g., 6-7 sites) are mutated to generate all possible amino substitutions at each site.
  • the antibody variants thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle.
  • the phage-displayed variants are then screened for their biological activity (e.g., binding affinity) as herein disclosed.
  • alanine scanning mutagenesis can be performed to identify hypervariable region residues contributing significantly to antigen binding.
  • Another type of amino acid variant of the antibody alters the original glycosylation pattern of the antibody by deleting one or more carbohydrate moieties found in the antibody and/or adding one or more glycosylation sites that are not present in the antibody.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences Asn-X′′-Ser and Asn-X′′-Thr, where X′′ is any amino acid except proline, are generally the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • X′′ is any amino acid except proline
  • O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration may also be made by the addition of or substitution by one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
  • an antibody may be desirable to modify an antibody with respect to effector function, for example to enhance antigen-dependent cell-mediated cytotoxicity (ADCC) and/or complement dependent cytotoxicity (CDC) of the antibody. This may be achieved by introducing one or more amino acid substitutions in an Fc region of the antibody.
  • an antibody can be engineered which has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities. See G. T. Stevenson et al., Anticancer Drug Des. 3:219-230 (1989).
  • a salvage receptor binding epitope refers to an epitope of the Fc region of an IgG molecule (e.g., IgG 1 , IgG 2 , IgG 3 , or IgG 4 ) that is responsible for increasing the in vivo serum half-life of the IgG molecule.
  • Fab′-SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab′) 2 fragments (P. Carter et al., Biotechnology 10:163-167 (1992)).
  • F(ab′) 2 fragments can be isolated directly from recombinant host cell culture.
  • a variety of expression vector/host systems may be utilized to express antibodies. These systems include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with bacterial expression vectors (e.g., Ti or pBR322 plasmid); or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with bacterial expression
  • Mammalian cells that are useful in recombinant antibody expression include but are not limited to VERO cells, HeLa cells, Chinese hamster ovary (CHO) cell lines, COS cells (such as COS-7), W138, BHK, HepG2, 3T3, RIN, MDCK, A549, PC12, K562 and 293 cells, as well as hybridoma cell lines as described herein. Mammalian cells are preferred for preparation of those antibodies that are typically glycosylated and require proper refolding for activity. Preferred mammalian cells include CHO cells, hybridoma cells, and myeloid cells.
  • expression vector refers to a plasmid, phage, virus or vector, for expressing a polypeptide from a DNA (RNA) sequence.
  • An expression vector may comprise a transcriptional unit comprising (1) one or more regulatory sequences controlling gene expression, for example, promoters or enhancers, (2) one or more sequences that encode one or more polypeptides, and (3) appropriate transcription initiation and termination sequences.
  • Expression vectors intended for use in yeast or eukaryotic expression systems preferably include a leader sequence enabling extracellular secretion of translated protein by a host cell.
  • an antibody polypeptide(s) is expressed without a leader or transport sequence, it may include an amino terminal methionine residue. This residue may or may not be subsequently cleaved from the expressed recombinant protein to provide a final antibody product.
  • Antibodies, specifically antibody fragments may be expressed in prokaryotic systems such as E. coli .
  • the DNA sequence encoding the specific binding agent peptide can be amplified by PCR and cloned into an appropriate vector, such as for example pGEX-3 ⁇ (Pharmacia).
  • the pGEX vector is designed to produce a fusion protein comprising glutathione-5-transferase (GST), encoded by the vector, and a peptide encoded by a DNA fragment inserted into the vector's cloning site.
  • the primers for PCR can be generated to include for example, an appropriate cleavage site.
  • the pGEX-3 ⁇ antibody peptide construct is transformed into E. coli XL-1 Blue cells (Stratagene, La Jolla Calif.), and individual transformants are isolated and grown.
  • the expressed peptide fusion protein may then be cleaved from the GST portion of the fusion protein.
  • Antibodies, specifically antibody fragments, made in bacterial cells may be produced as an insoluble inclusion body in the bacteria.
  • Such antibodies can be purified as follows. Host cells can be sacrificed by centrifugation; washed in 0.15 M NaCl, 10 mM Tris, pH 8, 1 mM EDTA; and treated with 0.1 mg/ml lysozyme (Sigma, St. Louis, Mo.) for 15 minutes at room temperature. The lysate can be cleared by sonication, and cell debris can be pelleted by centrifugation for 10 minutes at 12,000 ⁇ g.
  • the antibody containing pellet can be resuspended in 50 mM Tris, pH 8, and 10 mM EDTA, layered over 50% glycerol, and centrifuged for 30 min. at 6000 ⁇ g.
  • the pellet can be resuspended in standard phosphate buffered saline solution (PBS) free of Mg and Ca ions.
  • PBS phosphate buffered saline solution
  • the antibody can be further purified by fractionating the resuspended pellet in a denaturing SDS polyacrylamide gel (Sambrook et al., supra). The gel can be soaked in 0.4 M KCl to visualize the protein, which can be excised and electroeluted in gel-running buffer lacking SDS.
  • Mammalian host systems for the expression of antibodies are well known to those of skill in the art. Host cell strains can be chosen for a particular ability to process the expressed protein or produce certain post-translation modifications that will be useful in providing protein activity. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation. Different host cells such as CHO, HeLa, MDCK, 293, W138, as well as hybridoma cell lines, and the like have specific cellular machinery and characteristic mechanisms for such post-translational activities and can be chosen to ensure the correct modification and processing of the introduced, foreign protein.
  • selection systems can be used to recover the cells that have been transformed for recombinant antibody production.
  • selection systems include, but are not limited to, HSV thymidine kinase, hypoxanthine-guanine phosphoribosyltransferase and adenine phosphoribosyltransferase genes, in tk ⁇ , hgprt ⁇ or aprt-cells, respectively.
  • anti-metabolite resistance can be used as the basis of selection for DHFR which confers resistance to methotrexate; gpt which confers resistance to mycophenolic acid; neo which confers resistance to the aminoglycoside G418 and confers resistance to chlorsulfuron; and hygro which that confers resistance to hygromycin.
  • Additional selectable genes that may be useful include trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine.
  • Markers that give a visual indication for identification of transformants include anthocyanins, beta.-glucuronidase and its substrate, GUS, and luciferase and its substrate, luciferin.
  • antibodies produced using procedures described above may need to be “refolded” and oxidized into a proper tertiary structure and allowed to generate disulfide linkages in order to be biologically active.
  • Refolding can be accomplished using a number of procedures well known in the art. Such methods include, for example, exposing the solubilized polypeptide agent to a pH usually above 7 in the presence of a chaotropic agent.
  • a chaotrope is similar to the choices used for inclusion body solubilization. However a chaotrope is typically used at a lower concentration.
  • An exemplary chaotropic agent is guanidine.
  • the refolding/oxidation solution will also contain a reducing agent plus its oxidized form in a specific ratio to generate a particular redox potential which allows for disulfide shuffling to occur for the formation of cysteine bridges.
  • Some commonly used redox couples include cysteine/cystamine, glutathione/dithiobisGSH, cupric chloride, dithiothreitol DTT/dithiane DTT, and 2-mercaptoethanol (bME)/dithio-bME.
  • a co-solvent may be used to increase the efficiency of the refolding.
  • cosolvents include glycerol, polyethylene glycol of various molecular weights, and arginine.
  • An AA targeting agent may be covalently linked to a combining site in an antibody either directly or via a linker.
  • An appropriate linker can be chosen to provide sufficient distance between the targeting agent and the antibody.
  • the general design of an embodiment of a linker for use in preparing AA targeting compounds is represented by the formula: —X—Y-Z, wherein X is a connecting chain, Y is a recognition group and Z is a reactive group.
  • the linker may be linear or branched, and optionally includes one or more carbocyclic or heterocyclic groups. Linker length may be viewed in terms of the number of linear atoms, with cyclic moieties such as aromatic rings and the like to be counted by taking the shortest route around the ring.
  • the linker has a linear stretch of between 5-15 atoms, in other embodiments 15-30 atoms, in still other embodiments 30-50 atoms, in still other embodiments 50-100 atoms, and in still other embodiments 100-200 atoms.
  • Other linker considerations include the effect on physical or pharmacokinetic properties of the resulting AA targeting compound or AA targeting agent-linker, such as solubility, lipophilicity, hydrophilicity, hydrophobicity, stability (more or less stable as well as planned degradation), rigidity, flexibility, immunogenicity, modulation of antibody binding, the ability to be incorporated into a micelle or liposome, and the like.
  • the connecting chain X of the linker includes any atom from the group C, H, N, O, P, S, halogen (F, Cl, Br, I), or a salt thereof.
  • X also may include a group such as an alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, sulfoalkynyl, phosphoalkyl, phosphoalkenyl, or phosphoalkynyl group.
  • X may include one or more ring structures.
  • the linker is a repeating polymer such as polyethylene glycol comprising 2-100 units.
  • the recognition group Y of the linker is optional, and if present is located between the reactive group and the connecting chain. In some embodiments, Y is located from 1-20 atoms from Z. Although not wishing to be bound by any theory, it is believed that the recognition group acts to properly position the reactive group into the antibody combining site so that it may react with a reactive amino acid side chain. Exemplary recognition groups include carbocyclic and heterocyclic rings, preferably having five or six atoms. However, larger ring structures also may be used. In some embodiments, an AA targeting agent is linked directly to Y without the use of an intervening linker.
  • Z is capable of forming a covalent bond with a reactive side chain in an antibody combining site.
  • Z includes one or more C ⁇ O groups arranged to form a diketone, an acyl beta-lactam, an active ester, a haloketone, a cyclohexyl diketone group, an aldehyde, a maleimide, an activated alkene, an activated alkyne or, in general, a molecule comprising a leaving group susceptible to nucleophilic or electrophilic displacement.
  • Other groups may include a lactone, an anhydride, an alpha-haloacetamide, an imine, a hydrazide, or an epoxide.
  • Exemplary linker electrophilic reactive groups that can covalently bond to a reactive nucleophilic group (e.g., a lysine or cysteine side chain) in a combining site of antibody include acyl beta-lactam, simple diketone, succinimide active ester, maleimide, haloacetamide with linker, haloketone, cyclohexyl diketone, aldehyde, amidine, guanidine, imine, eneamine, phosphate, phosphonate, epoxide, aziridine, thioepoxide, a masked or protected diketone (a ketal for example), lactam, sulfonate, and the like, masked C ⁇ O groups such as imines, ketals, acetals, and any other known electrophilic group.
  • acyl beta-lactam simple diketone
  • succinimide active ester maleimide
  • the reactive group includes one or more C ⁇ O groups arranged to form an acyl beta-lactam, simple diketone, succinimide active ester, maleimide, haloacetamide with linker, haloketone, cyclohexyl diketone, or aldehyde.
  • a chemical moiety for modification by an aldolase antibody may be a ketone, diketone, beta lactam, active ester haloketone, lactone, anhydride, maleimide, alpha-haloacetamide, cyclohexyl diketone, epoxide, aldehyde, amidine, guanidine, imine, eneamine, phosphate, phosphonate, epoxide, aziridine, thioepoxide, masked or protected diketone (ketal for example), lactam, haloketone, aldehyde, and the like.
  • a linker reactive group chemical moiety suitable for covalent modification by a reactive sulfhydryl group in an antibody may be a disulfide, aryl halide, maleimide, alpha-haloacetamide, isocyanate, epoxide, thioester, active ester, amidine, guanidine, imine, eneamine, phosphate, phosphonate, epoxide, aziridine, thioepoxide, masked or protected diketone (ketal for example), lactam, haloketone, aldehyde, and the like.
  • reactive amino acid side chains in antibody combining sites may possess an electrophilic group that reacts with a nucleophilic group on an AA targeting agent or its linker, whereas in other embodiments a reactive nucleophilic group in an amino acid side chain reacts with an electrophilic group in an AA targeting agent or linker.
  • An AA targeting compound may be prepared by several approaches.
  • an AA targeting agent-linker compound is synthesized with a linker that includes one or more reactive groups designed for covalent reaction with a side chain of an amino acid in a combining site of an antibody.
  • the targeting agent-linker compound and antibody are combined under conditions where the linker reactive group forms a covalent bond with the amino acid side chain.
  • linking can be achieved by synthesizing an antibody-linker compound comprising an antibody and a linker wherein the linker includes one or more reactive groups designed for covalent reaction with an appropriate chemical moiety of an AA targeting agent.
  • An AA targeting agent may need to be modified to provide the appropriate moiety for reaction with the linker reactive group.
  • the antibody-linker and AA targeting agent are combined under conditions where the linker reactive group covalently links to the targeting and/or biological agent.
  • a further approach for forming an antibody-AA targeting compound uses a dual linker design.
  • an AA targeting agent-linker compound is synthesized which comprises an AA targeting agent and a linker with a reactive group.
  • An antibody-linker compound is synthesized which comprises an antibody and a linker with a chemical group susceptible to reactivity with the reactive group of the AA targeting agent-linker of the first step. These two linker containing compounds are then combined under conditions whereby the linkers covalently link, forming the antibody-AA-targeting compound.
  • Exemplary functional groups that can be involved in the linkage include, for example, esters, amides, ethers, phosphates, amino, keto, amidine, guanidine, imines, eneamines, phosphates, phosphonates, epoxides, aziridines, thioepoxides, masked or protected diketones (ketals for example), lactams, haloketones, aldehydes, thiocarbamate, thioamide, thioester, sulfide, disulfide, phosphoramide, sulfonamide, urea, thioruea, carbamate, carbonate, hydroxamide, and the like.
  • the linker includes any atom from the group C, H, N, O, P, S, halogen (F, Cl, Br, I), or a salt thereof.
  • the linker also may include a group such as an alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, or phosphoalkynyl group.
  • the linker also may include one or more ring structures.
  • a “ring structure” includes saturated, unsaturated, and aromatic carbocyclic rings and saturated, unsaturated, and aromatic heterocyclic rings.
  • the ring structures may be mono-, bi-, or polycyclic, and include fused or unfused rings.
  • the ring structures are optionally substituted with functional groups well known in the art, including but not limited to halogen, oxo, —OH, —CHO, —COOH, —NO 2 , —CN, —NH 2 , —C(O)NH 2 , C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, sulfoalkynyl, phosphoalkyl, phosphoalkenyl, or phosphoalkynyl group. Combinations of the above groups and rings may also be present in the linkers of AA targeting compounds.
  • One aspect of the invention is an AA targeting agent-linker conjugate having Formula I:
  • [AA targeting agent] is an AA targeting agent peptide
  • the linker moiety L in compounds of Formula I may be attached to the amino terminus, carboxy terminus or any amino acid side chain of an AA targeting agent.
  • L is linked to the carboxy terminus of an AA targeting agent.
  • L is linked to the amino terminus of an AA targeting agent.
  • L is linked to either a nucleophilic or electrophilic side chain.
  • L should possess a nucleophilic group susceptible to covalent reaction with the electrophilic side chain.
  • Exemplary electrophilic side chains are Asp and Glu.
  • Exemplary nucleophilic side chains are Cys, Lys, Ser, Thr, and Tyr.
  • L should comprise an electrophilic group susceptible to covalent reaction with the nucleophilic side chain.
  • a nucleophilic amino acid is added to either the carboxy terminus or the amino terminus of an AA targeting agent and the linker L is covalently attached to the side chain of this additional amino acid.
  • Lys is added to the amino terminus of an AA targeting agent. In certain other embodiments, Lys is added to the carboxy terminus of an AA targeting agent.
  • exemplary compounds of Formula I formed by linking to either i) the side chains of D, E, C, K, S, T, and Y or ii) the amino or carboxy termini include:
  • exemplary compounds of Formula I formed by linking to either i) the side chains of D, E, C, K, S, T, and Y or ii) the amino or carboxy termini include:
  • L is a linker moiety having the formula —X—Y-Z, wherein:
  • X is:
  • R 22 is —(CH 2 ) v —, —(CH 2 ) u —C(O)—(CH 2 ) v —, —(CH 2 ) u —C(O)—O—(CH 2 ) v — —(CH 2 ) u —C(S)—NR b —(CH 2 ) v —, —(CH 2 ) u —C(O)—NR b —(CH 2 ) v —, —(CH 2 ) u —NR b —(CH 2 ) v —, —(CH 2 ) u —O—(CH 2 ) v —, —(CH 2 ) u —S(O) 0-2 —(CH 2 ) v —, —(CH 2 ) u —S(O) 0-2 —NR b —(CH 2 ) v —, or
  • R 22 is —(CH 2 ) v —, —(CH 2 ) u —C(O)—(CH 2 ) v —, —(CH 2 ) u —C(O)—O—(CH 2 ) v —, —(CH 2 ) u —C(O)—NR b —(CH 2 ) v —, or —(CH 2 ) u , —NR b —(CH 2 ) v .
  • R 12 is —(CH 2 ) u —C(O)—NR b —(CH 2 ) v —.
  • R 21 and R 23 are each independently —(CH 2 ) s —, —(CH 2 ) r —C(O)—(CH 2 ) s —, (CH 2 ) r —C(O)—O—(CH 2 ) v —, —(CH 2 ) r —C(S)—NR b —(CH 2 ) s —, (CH 2 ) r ,C(O)—NR b (CH 2 ) s —, —(CH 2 ) r —NR b —(CH 2 ) s —, —(CH 2 ) r —O—(CH 2 ) s —, (CH 2 ) r —S(O) 0-2 —(CH 2 ) s —, —(CH 2 ) r —S(O) 0-2 —NR b —(CH 2 ) s —, or ——(CH 2 ) s
  • R 21 and R 23 are each independently —(CH 2 ) s —, —(CH 2 ) r —C(O)—(CH 2 ) s —, (CH 2 ) r ,C(O)—O—(CH 2 ) s —, —(CH 2 ) r —C(O)—NR b —(CH 2 ) s —, or —(CH 2 ) r —NR b —(CH 2 ) s , and —(CH 2 ) r ,C(O)—NR b (CH 2 ) s .
  • R 21 and R 23 each independently have the structure:
  • p is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 32, 43, 44, or 45;
  • w, r, and s are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20;
  • R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • X has the structure:
  • X has the structure:
  • H 1 and H 1′ at each occurrence are independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; t and t′ are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 32, 43, 44, 45, 46, 47, 48, 49 or 50; and R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • X has the structure:
  • H 1 and H 1′ at each occurrence are independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; t and t′ are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 32, 43, 44, 45, 46, 47, 48, 49 or 50; and R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • X has the structure:
  • H 1 and H 1′ at each occurrence are independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; t and t′ are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 32, 43, 44, 45, 46, 47, 48, 49 or 50; and R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • X has the structure:
  • H 1 and H 1′ at each occurrence are independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20; t and t′ are each independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 32, 43, 44, 45, 46, 47, 48, 49 or 50; and R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • X has the structure:
  • X has the structure:
  • X has the structure:
  • v and w are each independently 1, 2, 3, 4, or 5 and R b is hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • v is 1, 2 or 3
  • w is 1, 2, or 3
  • R b is hydrogen.
  • L is a linker moiety having the formula —X—Y-Z, wherein:
  • X is:
  • X has the formula:
  • the values of v, t, w, and p are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of v, t, r, and s are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, w, and p are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, r, and s are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, w, and p are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, r, and s are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, w, and p are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, r, and s are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, w, and p are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, r, and s are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, w, and p are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • X has the structure:
  • the values of u, v, t, r, and s are selected such that the backbone length of X is less than 200 atoms, alternatively is less than 100 atoms, alternatively, is less than 75 atoms, alternatively is less than 50 atoms, alternatively is less than 25 atoms, or alternatively is less than 15 atoms.
  • the ring structure of Y includes saturated, unsaturated, and aromatic carbocyclic rings and saturated, unsaturated, and aromatic heterocyclic rings.
  • the ring structure(s) may be mono-, bi-, or polycyclic, and include fused or unfused rings.
  • the ring structure(s) is optionally substituted with functional groups well known in the art including, but not limited to halogen, oxo, —OH, —CHO, —COOH, —NO 2 , —CN, —NH 2 , amidine, guanidine, hydroxylamine, —C(O)NH 2 , secondary and tertiary amides, sulfonamides, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, sulfoalkynyl, phosphoalkyl, phosphoalkenyl, and phosphoalkynyl groups.
  • functional groups well known in the
  • the ring structure of Y has the optionally substituted structure:
  • a, b, c, d, and e are each independently carbon or nitrogen; f is carbon, nitrogen, oxygen, or sulfur; Y is attached to X and Z independently at any two ring positions of sufficient valence; and no more than four of a, b, c, d, e, or f are simultaneously nitrogen.
  • any open valences remaining on atoms constituting the ring structure may be filled by hydrogen or other substituents, or by the covalent attachments to X and Z.
  • b is carbon
  • its valence may be filled by hydrogen, a substituent such as halogen, a covalent attachment to X, or a covalent attachment to Z.
  • a, b, c, d, and e are each carbon, while in others, a, c, d and f are each carbon.
  • at least one of a, b, c, d, or e is nitrogen, and in still others, f is oxygen or sulfur.
  • the ring structure of Y is unsubstituted.
  • Y is phenyl.
  • X—Y has the structure:
  • v is 1, 2 or 3 and w is 1, 2, or 3.
  • v is 1 or 2 and w is 1 or 2.
  • X has the structure:
  • H 1 and H 1′ are each independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, or 5; and t and t′ are each independently 0, 1, 2, 3, 4, or 5.
  • H 1 and H 1′ are each independently O or CH 2 ; r and s are each independently 1 or 2; and t and t′ are each independently 0 or 1.
  • X has the structure:
  • H 1 and H 1′ are each independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, or 5; t and t′ are each independently 0, 1, 2, 3, 4, or 5, and R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • H 1 and H 1′ are each independently O or CH 2 ; r and s are each independently 1 or 2; and t and t′ are each independently 0 or 1.
  • X has the structure:
  • H 1 and H 1′ are each independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, or 5; t and t′ are each independently 0, 1, 2, 3, 4, or 5, and R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • H 1 and H 1′ are each independently O or CH 2 ; r and s are each independently 1 or 2; and t and t′ are each independently 0 or 1.
  • X has the structure:
  • H 1 and H 1′ are each independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, or 5; t and t′ are each independently 0, 1, 2, 3, 4, or 5, and R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • H 1 and H 1′ are each independently O or CH 2 ; r and s are each independently 1 or 2; and t and t′ are each independently 0 or 1.
  • X has the structure:
  • H 1 and H 1′ are each independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, or 5; t and t′ are each independently 0, 1, 2, 3, 4, or 5, and R b at each occurrence is independently hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • H 1 and H 1′ are each independently O or CH 2 ; r and s are each independently 1 or 2; and t and t′ are each independently 0 or 1.
  • X has the structure:
  • H 1 and H 1′ are each independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, or 5; t and t′ are each independently 0, 1, 2, 3, 4, or 5, and R b is hydrogen, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 3-7 cycloalkyl-C 0-6 alkyl, or substituted or unsubstituted aryl-C 0-6 alkyl.
  • H 1 and H 1′ are each independently O or CH 2 ; r and s are each independently 1 or 2; and t and t′ are each independently 0 or 1.
  • X has the structure:
  • H 1 and H 1′ are each independently N, O, S, or CH 2 ; r and s are each independently 1, 2, 3, 4, or 5; and t and t′ are each independently 0, 1, 2, 3, 4, or 5.
  • H 1 and H 1′ are each independently O or CH 2 ; r and s are each independently 1 or 2; and t and t′ are each independently 0 or 1.
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • X—Y has the structure:
  • the reactive group Z contains a moiety capable of forming a covalent linkage with an amino acid in a combining site of an antibody.
  • Z may be substituted alkyl, substituted cycloalkyl, substituted aryl, substituted arylalkyl, substituted heterocyclyl, or substituted heterocyclylalkyl, wherein at least one substituent is a 1,3-diketone moiety, an acyl beta-lactam, an active ester, an alpha-haloketone, an aldehyde, a maleimide, a lactone, an anhydride, an alpha-haloacetamide, an amine, a hydrazide, or an epoxide.
  • Z is substituted alkyl.
  • Z may be a group that forms a reversible or irreversible covalent bond.
  • reversible covalent bonds may be formed using diketone Z groups such as those shown in FIG. 8 .
  • structures A-C may form reversible covalent bonds with reactive nucleophilic groups (e.g. lysine or cysteine side chain) in a combining site of an antibody.
  • R′ 1 , R′ 2 , R′ 3 , and R 4 in structures A-C of FIG. 8 represent substituents which can be C, H, N, O, P, S, halogen (F, Cl, Br, I) or a salt thereof.
  • substituents also may include a group such as an alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl group.
  • R′ 2 and R′ 3 also could from a ring structure as exemplified in structures B and C.
  • X in FIG. 8 could be a heteroatom.
  • FIG. 9 includes the structures of other linker reactive groups that form reversible covalent bonds, e.g., structures B, G, H, and, where X is not a leaving group, E and F.
  • Z reactive groups that form an irreversible covalent bond with a combining site of an antibody include structures D-G in FIG. 8 (e.g., when G is an imidate) and structures A, C and D of FIG. 9 .
  • structures E and F of FIG. 9 may also form irreversible covalent bonds.
  • Such structures are useful for irreversibly attaching a targeting agent-linker to a reactive nucleophilic group to a combining site of an antibody.
  • Z is a 1,3-diketone moiety. In still other such embodiments, Z is alkyl substituted by a 1,3-diketone moiety. In certain embodiments, Z has the structure:
  • Z has the structure:
  • One linker for use in AA targeting compounds and for preparing AA targeting agent-linker compounds includes a 1,3-diketone reactive group as Z.
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • u is 0; v iso; t is 1, 2, 3, 4, 5, or 6; w is 1; p is 3; and q iso, 1,2, or 3.
  • u is 0 or 1; v is 0; t is 1 or 2; w is 1; p is 1 or 2; and q is 1 or 2.
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • u is 0; v iso; t is 1, 2, 3, 4, 5, or 6; r is 1 or 2; s is 3; and q iso, 1,2, or 3.
  • L has the structure:
  • L has the structure:
  • u is O; v is 0; t is 1, 2, 3, 4, 5, or 6; w is 1; p is 3; and q is 0, 1, 2, or 3.
  • u is 0 or 1; v is 0; t is 1 or 2; w is 1; p is 1 or 2; and q is 1 or 2.
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • L has the structure:
  • AA 1 -AA 2 -AA n refers to an AA targeting agent wherein “AA 1 ” is the first amino acid in the AA targeting agent sequence, as measured from the N-terminus, “AA 2 ” is the second amino acid in the AA targeting agent sequence, as measured from the N-terminus, and “AA n ” is the n th amino acid in the AA targeting agent sequence, as measured from the N-terminus.
  • v is 1, 2, or 3; w is 1, 2, or 3; and q is 0, 1, 2, or 3.
  • v is 1,2, or 3; w is 1,2, or 3; and q is 0, 1, 2, 3.
  • v is 1, 2, or 3; w is 1, 2, or 3; and q is 0, 1, 2, 3.
  • AA targeting agent refers to an AA targeting agent wherein “AA 1 ” is the first amino acid in an AA targeting agent sequence as measured from the N-terminus, “AA 2 ” is the second amino acid in an AA targeting agent sequence as measured from the N-terminus, and “AA n ” is the n th amino acid in an AA targeting agent sequence as measured from the N-terminus.
  • the targeting agent further comprises a Lys residue at arbitrary position m+1 as measured from the N-terminus. It will be appreciated that in addition to linking to a Lys side chain in the body of an AA targeting agent, it is also possible to link to a Lys side chain on the N-terminus or C-terminus of an AA targeting agent.
  • an AA targeting compound administered to an immunocompetent individual may result in the production of antibodies against the conjugate.
  • Such antibodies may be directed to the variable region, including the antibody idiotype, as well as to the targeting agent or any linker used to conjugate the targeting agent to the antibody.
  • Reducing the immunogenicity of an AA targeting compound can be accomplished by methods well known in the art, such as by attaching long chain polyethylene glycol (PEG)-based spacers and the like to the AA targeting compound.
  • PEG polyethylene glycol
  • Long chain PEG and other polymers are known for their ability to mask foreign epitopes, resulting in the reduced immunogenicity of therapeutic proteins that display foreign epitopes (N. V. Katre, J. Immunol. 144:209-213 (1990); G. E.
  • the individual administered the antibody-AA targeting agent conjugate may be administered an immunosuppressant such as cyclosporin A, anti-CD3 antibody, and the like.
  • an AA targeting compound is as shown by Formula II, and includes stereoisomers, tautomers, solvates, prodrugs, and pharmaceutically acceptable salts thereof.
  • [AA targeting agent] is defined as in Formula I.
  • L is a linker moiety linking an antibody to the targeting agent and having the formula —X—Y-Z-.
  • X and Y are defined as in Formula I, and
  • Antibody is an antibody as defined herein.
  • FIGS. 10 and 11 respectively, illustrate the addition mechanism of a reactive, nucleophilic side chain in a combining site of an antibody to the Z moieties illustrated in FIGS. 8 and 9 .
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z′-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z′-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z′-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z′-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z′-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z′-Antibody has the structure:
  • HN-Antibody refers to an arbitrary side chain in the combining site of an antibody bearing an amino group.
  • Z′ is an attachment moiety comprising a covalent bond and 0-20 carbon atoms to which the Antibody is attached. This is shown below for the case where the linker has a diketone moiety as the reactive group and linkage occurs with the side chain amino group of a lysine residue in the antibody combining site.
  • the Antibody is shown schematically as bivalent with a reactive amino acid side chain for each combining site indicated.
  • linker has a beta lactam moiety as the reactive group and linkage occurs with the side chain amino group of a lysine residue in the antibody combining site.
  • the Antibody is shown schematically as bivalent with a reactive amino acid side chain for each combining site indicated.
  • v is 1, 2, or 3; w is 1, 2, or 3; and q is 0, 1, 2, 3.
  • v is 1, 2, or 3; w is 1, 2, or 3; and q is 0, 1, 2, 3.
  • v is 1 or 2 and w is 1 or 2.
  • the linker may have an amine or hydrazide as the reactive group and the Antibody may be engineered to have a diketone moiety.
  • An unnatural diketone-containing amino acid may be readily incorporated into an antibody combining site using techniques well known in the art; proteins containing unnatural amino acids have been produced in yeast and bacteria. See, e.g., J. W. Chin et al., Science 301:964-966 (2003); L. Wang et al., Science 292:498-500 (2001); J. W. Chin et al., J. Am. Chem. Soc. 124:9026-9027 (2002); L. Wang, et al., J. Am. Chem. Soc.
  • an unnatural amino acid containing a diketone moiety into the yeast Saccharomyces cerevisiae requires the addition of new components to the protein biosynthetic machinery including a unique codon, tRNA, and aminoacyl-tRNA synthetase (aa RS).
  • aa RS aminoacyl-tRNA synthetase
  • the amber suppressor tyrosyl-tRNA synthetase (TyrRS)-tRNA CUA pair from E. coli may be used as reported for eukaryotes in J. W. Chin et al., Science 301:964-966 (2003).
  • the amber codon is used to code for the unnatural amino acid of interest.
  • mutant TyrRS and tRNA CUA may then be produced and selected for those aaRS-tRNA CUA pairs in which the TyrRS charges the tRNA CUA with the unnatural amino acid of interest, e.g., the diketone-containing amino acid.
  • antibodies incorporating the diketone-containing amino acid may be produced by cloning and expressing a gene containing the amber codon at one or more antibody combining sites.
  • the Antibody is a full length antibody. In other embodiments, the Antibody is Fab, Fab′ F(ab′) 2 , Fv, V H , V L , or scFv. In certain embodiments, the Antibody is a human antibody, humanized antibody or chimeric human antibody. In certain embodiments, the Antibody is a catalytic antibody. In one embodiment, the Antibody is a humanized version of a murine 38c2 comprising a constant region from a human IgG, IgA, IgM, IgD, or IgE antibody. In another embodiment, Antibody is a chimeric antibody comprising the variable region from murine 38c2 and a constant region from a human IgG, IgA, IgM, IgD, or IgE antibody.
  • two or more AA targeting agents may be linked to a single full length bivalent Antibody. This is shown below as Formula III:
  • stereoisomers are also provided.
  • tautomers are also provided.
  • solvates are also provided.
  • [AA targeting agent], L′ and Antibody are each defined as in Formula II.
  • Targeting compounds such as those of Formula II may also be readily synthesized by covalently linking a targeting agent-linker compound as described herein to a combining site of a multivalent antibody.
  • a targeting agent-linker compound as described herein for example, an AA targeting-agent linker conjugate, where the linker includes a diketone reactive moiety, can be incubated with 0.5 equivalents of an aldolase antibody, such as h38C2 IgG1 to produce an AA targeting compound.
  • an AA targeting compound such as those of Formula III may be produced by covalently linking an AA targeting agent-linker compound as described herein to each combining site of a bivalent antibody.
  • One aspect of the invention provides methods for modulating thrombospondin activity in vivo comprising administering an effective amount of an AA targeting compound as described herein to a subject.
  • methods for treating abnormal angiogenesis or an angiogenesis-mediated condition in a subject include administering to the subject a therapeutically effective amount of an AA targeting compound as described herein.
  • an angiogenesis-mediated condition is a condition that is caused by abnormal angiogenesis activity or one in which compounds that modulate angiogenesis activity have therapeutic use.
  • Diseases and conditions that may be treated include cancer, arthritis, psoriasis, angiogenesis of the eye associated with infection or surgical intervention, macular degeneration or diabetic retinopathy.
  • methods of treating cancer include carcinomas of the breast, colon, rectum, lung, oropharynx, hypopharynx, esophagus, stomach, pancreas, liver, gallbladder and bile ducts, small intestine, urinary tract, female genital tract, male, genital tract, endocrine glands, and skin; hemangiomas; melanomas; sarcomas; tumors of the brain, nerves, eyes, and meninges; leukemia; or lymphoma.
  • compositions of the AA targeting compounds can be mixed with pharmaceutically-acceptable carriers to form a pharmaceutical composition for administration to a cell or subject, either alone, or in combination with one or more other modalities of therapy.
  • a pharmaceutical composition is generally formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral (e.g. intravenous, intramuscular, intramedullary, intradermal, subcutaneous), oral (e.g. inhalation, ingestion), intranasal, transdermal (e.g. topical), transmucosal, and rectal administration.
  • Administration routes of AA targeting compounds may also include intrathecal, direct intraventricular and intraperitoneal delivery.
  • the AA targeting compounds may be administered through any of the parenteral routes either by direct injection of the formulation or by infusion of a mixture of the targeting AA compound formulation with an infusion matrix such as normal saline, D5W, lactated Ringers solution or other commonly used infusion media.
  • an infusion matrix such as normal saline, D5W, lactated Ringers solution or other commonly used infusion media.
  • AA targeting compounds may be administered using techniques well known to those in the art.
  • agents are formulated and administered systemically. Techniques for formulation and administration may be found in “Remington's Pharmaceutical Sciences,” 18 th Ed., 1990, Mack Publishing Co., Easton, Pa.
  • AA targeting compounds may be formulated in aqueous solutions, emulsions or suspensions.
  • AA targeting compounds are preferably formulated in aqueous solutions containing physiologically compatible buffers such as citrate, acetate, histidine or phosphate.
  • physiologically compatible buffers such as citrate, acetate, histidine or phosphate.
  • such formulations may also contain various tonicity adjusting agents, solubilizing agents and/or stabilizing agents (e.g.
  • salts such as sodium chloride or sugars such as sucrose, mannitol, and trehalose, or proteins such as albumin or amino acids such as glycine and histidine or surfactants such as polysorbates (Tweens) or cosolvents such as ethanol, polyethylene glycol and propylene glycol.
  • the pharmaceutical composition may contain formulation materials for modifying, maintaining or preserving, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition.
  • formulation materials for modifying, maintaining or preserving for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the composition.
  • Suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine or lysine); antimicrobials; antioxidants (such as ascorbic acid, sodium sulfite or sodium hydrogen-sulfite); buffers (such as borate, bicarbonate, Tris-HCl, citrates, phosphates, other organic acids, chelating agents [such as ethylenediamine tetraacetic acid (EDTA)]; solvents (such as glycerin, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); suspending agents; surfactants or wetting agents (such as pluronics, PEG, sorbitan esters, polysorbates such as polysorbate 20, polysorbate 80, triton, tromethamine, lecithin, cholesterol, tyloxapal); stability enhancing agents (sucrose or sorbitol); tonicity enhancing
  • the therapeutic compositions may be in the form of a pyrogen-free, parenterally acceptable aqueous solution comprising an AA targeting compound in a pharmaceutically acceptable vehicle.
  • a vehicle for parenteral injection is sterile distilled water in which an AA targeting compound is formulated as a sterile, isotonic solution.
  • another formulation can involve the formulation an AA targeting compound with an agent, such as injectable microspheres, bio-degradable particles, polymeric compounds (polylactic acid, polyglycolic acid), beads, or liposomes, that provides for the controlled or sustained release of the product which may then be delivered via a depot injection.
  • Hyaluronic acid may also be used, and this may have the effect of promoting sustained duration in the circulation.
  • Other suitable means for the introduction of the desired molecule include implantable drug delivery devices.
  • compositions suitable for parenteral administration may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or a physiologically buffered saline.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • Suitable lipophilic solvents or vehicles include fatty oils, such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate, triglycerides, or liposomes.
  • Non-lipid polycationic amino polymers may also be used for delivery.
  • the suspension may also contain suitable stabilizers or agents to increase the solubility of the compounds and allow for the preparation of highly concentrated solutions.
  • compositions to be used for in vivo administration typically must be sterile. This may be accomplished by filtration through sterile filtration membranes. Where the composition is lyophilized, sterilization using this method may be conducted either prior to or following lyophilization and reconstitution.
  • the composition for parenteral administration may be stored in lyophilized form or in solution.
  • parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • the pharmaceutical composition may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or a dehydrated or lyophilized powder.
  • Such formulations may be stored either in a ready-to-use form or in a form (e.g., lyophilized) requiring reconstitution prior to administration.
  • kits for producing a single-dose administration unit may each contain both a first container having an AA targeting compound and a second container having an aqueous formulation. Also included within the scope of this invention are kits containing single and multi-chambered pre-filled syringes.
  • a therapeutically effective amount of an AA targeting compound or a pharmaceutically acceptable derivative is administered.
  • the frequency of dosing will depend upon the pharmacokinetic parameters of the AA targeting compound in the formulation used.
  • a composition is administered until a dosage is reached that achieves the desired effect.
  • the composition may therefore be administered as a single dose, or as multiple doses (at the same or different concentrations/dosages) over time, or as a continuous infusion.
  • Routes and frequency of administration of a composition as well as dosage may vary from individual to individual and may be readily established using standard techniques. Further refinement of the appropriate dosage is routinely made. Appropriate dosages may be developed by one skilled in the art through the use of appropriate dose-response data.
  • An appropriate dosage and treatment regimen provides the active compound(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit.
  • a response can be monitored by establishing an improved clinical outcome (e.g. reduced number of blood vessels in a target area, decreased tumor size or volume, in treated patients as compared to non-treated patients.
  • a suitable dose is an amount of a compound that, when administered as described herein, is capable of promoting an anti-angiogenesis response, and/or is at least 10-50% above the basal or untreated level.
  • the most effective mode of administration and dosage regimen for the invention compositions depends upon the severity and course of the disease, the patient's health and response to treatment, and the judgment of the treating physician. Accordingly, the dosages of the invention compositions should be titrated to the individual patient.
  • An effective dose of the compounds is in the range of from about 0.1 ug to about 40 mg per kilogram per day.
  • An AA targeting compound may be administered as a daily intravenous infusion from about 0.1 mg/kg body weight to about 15 mg/kg body weight. Accordingly, one embodiment provides a dose of about 0.5 mg/kg body weight. Another embodiment provides a dose of about 0.75 mg/kg body weight. Another embodiment provides a dose of about 1.0 mg/kg body weight.
  • Another embodiment provides a dose of about 2.5 mg/kg body weight. Another embodiment provides a dose of about 5 mg/kg body weight. Another embodiment provides a dose of about 10.0 mg/kg body weight. Another embodiment provides a dose of about 15.0 mg/kg body weight.
  • Doses of an AA targeting compound or a pharmaceutically acceptable derivative should be administered in intervals of from about once per day to 2 times per week, or alternatively, from about once every week to once per month. In one embodiment, a dose is administered to achieve peak plasma concentrations of an AA targeting compound or a pharmaceutically acceptable derivative thereof from about 0.002 mg/ml to 30 mg/ml.
  • Desirable blood levels may be maintained by a continuous infusion of an AA targeting compound as ascertained by plasma levels measured by a validated analytical methodology.
  • One method for administering an AA targeting compound to an individual comprises administering an AA targeting agent-linker conjugate to the individual and allowing it to form a covalent compound with a combining site of an appropriate antibody in vivo.
  • the antibody portion of an AA targeting compound that forms in vivo may be administered to the individual before, at the same time, or after administration of the targeting agent-linker conjugate.
  • an AA targeting agent may include a linker/reactive moiety, or the antibody combining site may be suitably modified to covalently link to the targeting agent.
  • an antibody may be present in the circulation of the individual following immunization with an appropriate immunogen.
  • catalytic antibodies may be generated by immunizing with a reactive intermediate of the substrate conjugated to a carrier protein. See R. A. Lerner and C. F. Barbas 3 rd , Acta Chem. Scand. 50:672-678 (1996).
  • aldolase catalytic antibodies may be generated by administering with keyhole limpet hemocyanin linked to a diketone moiety as described by P. Wirsching et al., Science 270:1775-1782 (1995) (commenting on J. Wagner et al., Science 270:1797-1800 (1995)).
  • the invention also provides a method of visualizing or localizing a thrombospondin receptor or anti-angiogenesis target (i.e. AA-targeting agent receptor) in tissues and cells.
  • biopsied tissues may be examined for presence of AA-targeting agent receptor.
  • neovascularization in a subject may be imaged by administering to the subject an AA targeting agent or compound including a detectable label.
  • detectable label refers to any molecule which can be administered in vivo and subsequently detected.
  • Exemplary detectable labels include radiolabels and fluorescent molecules.
  • Exemplary radionuclides include indium-111, technetium-99, carbon-11, and carbon-13. Fluorescent molecules include, without limitation, fluorescein, allophycocyanin, phycoerythrin, rhodamine, and Texas red.
  • vasculature within a tumor generally undergoes active angiogenesis, resulting in the continual formation of new blood vessels to support the growing tumor.
  • angiogenic blood vessels are distinguishable from mature vasculature in that angiogenic vasculature expresses unique endothelial cell surface markers, including the .alpha.sub.v.beta.sub.3 integrin. (Brooks, Cell 79:1157-1164 (1994); WO 95/14714, Int. Filing Date Nov. 22, 1994) and receptors for angiogenic growth factors (Mustonen and Alitalo, J. Cell Biol. 129:895-898 (1995); Lappi, Semin. Cancer Biol. 6:279-288 (1995)).
  • the invention also includes administration of one or more AA targeting agents in combination with one or more oncology therapeutics, each being administered according to a regimen suitable for that therapeutic.
  • the components of the combination therapy may be administered concurrently or non-concurrently.
  • concurrently administered and non-concurrent administration encompass substantially simultaneous administration of one or more AA targeting compounds and one other oncology therapeutic.
  • non-concurrent administration encompasses administering one or more AA targeting compounds at different times, in any order, whether overlapping or not. This includes, but is not limited to, sequential treatment (such as pretreatment, post-treatment, or overlapping treatment) with the components of the combination, as well as regimens in which the drugs are alternated, or wherein one component is administered long-term and the other(s) are administered intermittently. Components may be administered in the same or in separate compositions, and by the same or different routes of administration.
  • Suitable oncology therapeutics and combinations that may be used in combination with an AA targeting compounds are listed in Tables 4-6.
  • Aldesleukin Proleukin Proleukin is indicated for the treatment of Chiron Corp adults with metastatic renal cell carcinoma (metastatic RCC) and for the treatment of adults with metastatic melanoma.
  • Alemtuzumab Campath Campath is indicated for the treatment of B- Millennium and cell chronic lymphocytic leukemia (B-CLL) ILEX Partners, in patients who have been treated with LP alkylating agents and who have failed fludarabine therapy.
  • Palonosetron Aloxi For the treatment of nausea MGI Pharmaceuticals Altretamine Hexalen Single agent palliative treatment of patients US Bioscience with persistent or recurrent ovarian cancer following first-line therapy with a cisplatin and/or alkylating agent based combination.
  • Amifostine Ethyol To reduce the cumulative renal toxicity US Bioscience associated with repeated administration of cisplatin in patients with advanced ovarian cancer Amifostine Ethyol Reduces platinum toxicity in non-small cell US Bioscience lung cancer Amifostine Ethyol To reduce post-radiation xerostomia for US Bioscience head and neck cancer where the radiation port includes a substantial portion of the parotid glands.
  • Anastrozole Arimidex Adjuvant treatment of postmenopausal AstraZeneca women with hormone receptor positive early breast cancer
  • Anastrozole Arimidex Treatment of advanced breast cancer in AstraZeneca postmenopausal women with disease Pharmaceuticals progression following tamoxifen therapy.
  • Anastrozole Arimidex For first-line treatment of postmenopausal AstraZeneca women with hormone receptor positive or Pharmaceuticals hormone receptor unknown locally advanced or metastatic breast cancer.
  • Nelarabine Arranon For the treatement of T cell acute GlaxoSmithKline lymphoblatic leukemia
  • Arsenic Trisenox Second line treatment of relapsed or Cell Therapeutic trioxide refractory APL following ATRA plus an anthracycline.
  • Asparaginase Elspar ELSPAR is indicated in the therapy of Merck & Co, Inc patients with acute lymphocytic leukemia. This agent is useful primarily in combination with other chemotherapeutic agents in the induction of remissions of the disease in pediatric patients.
  • Bevacizumab Avastin For the treatment of metastatic colorectal Genentech cancer Bexarotene Targretin
  • Bexarotene gel Targretin For the topical treatment of cutaneous Ligand manifestations of cutaneous T-cell Pharmaceuticals lymphoma in patients who are refractory to at least one prior systemic therapy.
  • Squibb Squamous Cell Carcinoma head and neck including mouth, tongue, tonsil, nasopharynx, oropharynx, sinus, palate, lip, buccal mucosa, gingivae, epiglottis, skin, larynx, penis, cervix, and vulva.
  • Lymphomas Hodgkin's Disease, non- Hodgkin's lymphoma.
  • Testicular Carcinoma Embryonal cell, choriocarcinoma, and teratocarcinoma).
  • Bleomycin Blenoxane Sclerosing agent for the treatment of Bristol-Myers malignant pleural effusion (MPE) and Squibb prevention of recurrent pleural effusions.
  • Busulfan Busulfex Use in combination with cyclophoshamide Orphan Medical, intravenous as conditioning regimen prior to allogeneic Inc. hematopoietic progenitor cell transplantation for chronic myelogenous leukemia.
  • Upjohn Company Capecitabine Xeloda Treatment of metastatic breast cancer Roche resistant to both paclitaxel and an anthracycline containing chemotherapy regimen or resistant to paclitaxel and for whom further anthracycline therapy may be contraindicated, e.g., patients who have received cumulative doses of 400 mg/m2 of doxorubicin or doxorubicin equivalents Capecitabine Xeloda Initial therapy of patients with metastatic Roche colorectal carcinoma when treatment with fluoropyrimidine therapy alone is preferred.
  • Combination chemotherapy has shown a survival benefit compared to 5-FU/LV alone.
  • a survival benefit over 5_FU/LV has not been demonstrated with Xeloda monotherapy.
  • Capecitabine Xeloda Treatment in combination with docetaxel of Roche patients with metastatic breast cancer after failure of prior anthracycline containing chemotherapy Carboplatin Paraplatin Palliative treatment of patients with ovarian Bristol-Myers carcinoma recurrent after prior Squibb chemotherapy, including patients who have been previously treated with cisplatin.
  • Carboplatin Paraplatin Initial chemotherapy of advanced ovarian Bristol-Myers carcinoma in combination with other Squibb approved chemotherapeutic agents.
  • Carmustine BCNU Palliative therapy as a single agent or in Bristol-Myers BiCNU established combination therapy with other Squibb approved chemotherapeutic agents in the following: Brain tumors (glioblastoma, brainstem glioma, medulloblastoma, astrocytoma, ependymoma, and metastatic brain tumors); Multiple myeloma; Hodgkin's Disease; and Non-Hodgkin's lymphomas.
  • Carmustine Giladel For use in addition to surgery to prolong Guilford with Wafer survival in patients with recurrent Pharmaceuticals Polifeprosan glioblastoma multiforme who qualify for Inc. 20 Implant surgery.
  • Celecoxib Celebrex Reduction of polyp number in patients with Searle the rare genetic disorder of familial adenomatous polyposis.
  • Cetuximab Erbitux For the treatement of EGFR expressing metastatic colorectal cancer Chlorambucil Leukeran Chronic Lymphocytic Leukemia - palliative GlaxoSmithKline therapy Chlorambucil Leukeran Treatment for CLL or indolent NHL.
  • An established combination therapy consists of Platinol, Blenoxane and Velbam.
  • Cisplatin Platinol Metastatic ovarian tumors - in established Bristol-Myers combination therapy with other approved Squibb chemotherapeutic agents Ovarian-in established combination therapy with other approved chemotherapeutic agents in patients with metastatic ovarian tumors who have already received appropriate surgical and/or radiotherapeutic procedures.
  • Platinol As a single agent, is indicated as secondary therapy in patients with metastatic ovarian tumors refractory to standard chemotherapy who have not previously received Platinol therapy.
  • Cisplatin Platinol Transitional cell bladder cancer which is no Bristol-Myers longer amenable to local treatments such as Squibb surgery and/or radiotherapy.
  • Cladribine Leustatin 2- Treatment of active hairy cell leukemia.
  • R.W. Johnson CdA Pharmaceutical Research Institute Clofarabine Clolar Treatment for acute lymphblastic leukemia Genzyme Cyclophosphamide Cytoxan, Treatment for ovary, breast, bladder and Bristol-Myers Neosar CLL.
  • Daunorubicin Daunorubicin Leukemia/myelogenous/monocytic/erythoid Bedford Labs' daunomycin of adults/remission induction in acute lymphocytic leukemia of children and adults.
  • Daunorubicin, Cerubidine In combination with approved anticancer Wyeth Ayerst daunomycin drugs for induction of remission in adult ALL.
  • Docetaxel Taxotere Treatment of patients with locally advanced Aventis or metastatic breast cancer who have Pharmaceutical progressed during anthracycline-based therapy or have relapsed during anthracycline-based adjuvant therapy.
  • Docetaxel Taxotere For the treatment of locally advanced or Aventis metastatic breast cancer which has Pharmaceutical progressed during anthracycline-based treatment or relapsed during anthracycline- based adjuvant therapy.
  • Docetaxel Taxotera For locally advanced or metastatic non-small Aventis cell lung cancer after failure of prior Pharmaceutical platinum-based chemotherapy.
  • Docetaxel Taxotere Aventis Pharmaceutical Docetaxel Taxotere Used in combination with cisplatin for the Aventis treatment of patients with unresectable, Pharmaceutical locally advanced or metastatic non-small cell lung cancer who have not previously received chemotherapy for this condition.
  • Elliott's B Elliott's B Diluent for the intrathecal administration of Orphan Medical, Solution Solution methotrexate sodium and cytarabine for the Inc. prevention or treatment of meningeal leukemia or lymphocytic lymphoma.
  • Epoetin Epogen EPOGEN is indicated for the treatment of Amgen, Inc. alfa/beta anemia.
  • Erlotinib Tarceva For the treatment of advanced metatstaic OSI non-small cell lung cancer Pharmaceuticals Estramustine Emcyt Palliation of prostate cancer Pharmacia & Upjohn Company Etoposide Etopophos Management of refractory testicular tumors, Bristol-Myers phosphate in combination with other approved Squibb chemotherapeutic agents. Etoposide Etopophos Management of small cell lung cancer, first- Bristol-Myers phosphate line, in combination with other approved Squibb chemotherapeutic agents. Etoposide Etopophos Management of refractory testicular tumors Bristol-Myers phosphate and small cell lung cancer.
  • Squibb Etoposide, VP- Vepesid Refractory testicular tumors-in combination Bristol-Myers 16 therapy with other approved Squibb chemotherapeutic agents in patients with refractory testicular tumors who have already received appropriate surgical, chemotherapeutic and radiotherapeutic therapy.
  • etoposide, VP- VePesid In combination with other approved Bristol-Myers 16 chemotherapeutic agents as first line Squibb treatment in patients with small cell lung cancer.
  • Etoposide, VP- Vepesid In combination with other approved Bristol-Myers 16 chemotherapeutic agents as first line Squibb treatment in patients with small cell lung cancer.
  • Filgrastim Neupogen NEUPOGEN is indicated for reducing the Amgen, Inc. time to neutrophil recovery and the duration of fever, following induction or consolidation hemotherapy treatment of adults with AML.
  • Floxuridine FUDR An analog for 5-flurouracil. FUDR has been Roche (intraarterial) approved in the directed treatment of liver metastases using hepatic arterial infusion. Fludarabine Fludara Palliative treatment of patients with B-cell Berlex lymphocytic leukemia (CLL) who have not Laboratories Inc.
  • CLL B-cell Berlex lymphocytic leukemia
  • Gemcitabine Gemzar For use in combination with cisplatin for the Eli Lilly first-line treatment of patients with inoperable, locally advanced (Stage IIIA or IIIB) or metastatic (Stage IV) non-small cell lung cancer.
  • Gemtuzumab Mylotarg Treatment of CD33 positive acute myeloid Wyeth Ayerst ozogamicin leukemia in patients in first relapse who are 60 years of age or older and who are not considered candidates for cytotoxic chemotherapy.
  • Idarubicin Idamycin For use in combination with other approved Adria antileukemic drugs for the treatment of acute Laboratories myeloid leukemia (AML) in adults. Idarubicin Idamycin In combination with other approved Pharmacia & antileukemic drugs for the treatment of acute Upjohn Company non-lymphocytic leukemia in adults. Ifosfamide IFEX Third line chemotherapy of germ cell Bristol-Myers testicular cancer when used in combination Squibb with certain other approved antineoplastic agents.
  • Imatinib Gleevec Initial therapy of chronic myelogenous Novartis mesylate leukemia
  • Imatrinib Gleevac Treatment of metastatic or unresectable Novartis mesylate malignant gastrointestinal stromal tumors
  • Imatinib Gleevec Initial treatment of newly diagnosed Ph+ Novartis mesylate chronic myelogenous leukemia (CML).
  • Interferon alfa- Roferon-A Treatment of chronic Hoffmann-La 2a hepatitis C, hairy cell leukemia and AIDS- Roche Inc.
  • Interferon alfa- Intron A Interferon alfa-2b, recombinant for injection Schering Corp. 2b is indicated as adjuvant to surgical treatment in patients 18 years of age or older with malignant melanoma who are free of disease but at high risk for systemic recurrence within 56 days of surgery.
  • Interferon alfa-2b recombinant for Injection is indicated for the initial treatment of clinically aggressive follicular Non- Hodgkin's Lymphoma in conjunction with anthracycline-containing combination chemotherapy in patients 18 years of age or older.
  • Interferon alfa-2b, recombinant for Injection is indicated for intralesional treatment of selected patients 18 years of age or older with condylomata acuminata involving external surfaces of the genital and perianal areas.
  • Interferon alfa-2b, recombinant for Injection is indicated for the treatment of patients 18 years of age or older with hairy cell leukemia.
  • Interferon alfa-2b, recombinant for Injection is indicated for the treatment of selected patients 18 years of age or older with AIDS- Related Kaposi's Sarcoma.
  • the likelihood of response to INTRON A therapy is greater in patients who are without systemic symptoms, who have limited lymphadenopathy and who have a relatively intact immune system as indicated by total CD4 count.
  • Leucovorin calcium is indicated fro use in Immunex Leucovorin combination with 5-fluorouracil to prolong Corporation survival in the palliative treatment of patients with advanced colorectal cancer.
  • Leucovorin Leucovorin In combination with fluorouracil to prolong Lederle survival in the palliative treatment of laboratories patients with advanced colorectal cancer.
  • Levamisole Ergamisol Adjuvant treatment in combination with 5- Janssen Research fluorouracil after surgical resection in Foundation patients with Dukes' Stage C colon cancer.
  • Mercaptopurine, Purinethol Purinethol is indicated for remission GlaxoSmithKline 6-MP induction and maintenance therapy of acute lymphatic leukemia.
  • Methoxsalen Uvadex For the use of UVADEX with the UVAR Therakos Photopheresis System in the palliative treatment of the skin manifestations of cutaneous T-cell lymphoma (CTCL) that is unresponsive to other forms of treatment.
  • CTCL cutaneous T-cell lymphoma
  • Mitotane Lysodren Used for the treatment of adrenal cancers.
  • Bristol-Myers Squibb Mitoxantrone Novantrone For use in combination with corticosteroids Immunex as initial chemotherapy for the treatment of Corporation patients with pain related to advanced hormone-refractory prostate cancer.
  • Mitoxantrone Novantrone For use with other approved drugs in the Laderle initial therapy for acute nonlymphocytic Laboratories leukemia (ANLL) in adults.
  • Nandrolone Durabolin- It is indicated as a treatment for palliation of Organon phenpropionate 509 inoperable metastatic breast cancer in postmenopausal women.
  • Nofetumomab Verluma Verluma is a monoclonal antibody Fab Boehringer fragment linked to 99m Tc. Verluma identifies Ingelheim advanced-stage disease in patients with Pharma KG small-cell lung cancer (SCLC). (formerly Dr. Karl Thomae GmbH) Oprelvekin Neumega Neumega is indicated for the prevention of Genetics severe thrombocytopenia and the reduction Institute, Inc.
  • Oxaliplatin Eloxatin Used in combination with infusional 5- Sanofi FU/LV, is indicated for the treatment of Synthelabo patient with metastatic carcinoma of the colon or rectum whose disease has recurred or progressed during or within 6 months of completion of first line therapy with the combination of bolus 5-FU/LV and irinotecan.
  • Paclitaxel Taxol Treatment of patients with metastatic Bristol-Myers carcinoma of the ovary after failure of first- Squibb line or subsequent chemotherapy. Treatment of breast cancer after failure of combination chemotherapy for metastatic disease or relapse within 6 months of adjuvant chemotherapy. Prior therapy should have included an anthracycline unless clinically contraindicated. New dosing regimen for patients who have failed initial or subsequent chemotherapy for metastatic carcinoma of the ovary Second line therapy for AIDS related Kaposi's sarcoma. For first-line therapy for the treatment of advanced carcinoma of the ovary in combination with cisplatin. For use in combination with cisplatin, for the first-line treatment of non-small cell lung cancer in patients who are not candidates for potentially curative surgery and/or radiation therapy.
  • Pfizer Labs mithramycin Porfimer Photofrin For use in photodynamic therapy (PDT) for QLT sodium palliation of patients with completely Phototherapeutics obstructing esophageal cancer, or patients Inc. with partially obstructing esophageal cancer who cannot be satisfactorily treated with ND-YAG laser therapy.
  • photodynamic therapy for use in photodynamic therapy for treatment of microinvasive endobronchial nonsmall cell lung cancer in patients for whom surgery and radiotherapy are not indicated.
  • photodynamic therapy (PDT) for reduction of obstruction and palliation of symptoms in patients with completely or partially obstructing endobroncial nonsmall cell lung cancer (NSCLC). Procarbazine Matulane One component of the MOPP regime.
  • Tamoxifen Nolvadex As a single agent to delay breast cancer AstraZeneca recurrence following total mastectomy and Pharmaceuticals axillary dissection in postmenopausal women with breast cancer (T1-3, N1, M0). For use in premenopausal women with metastatic breast cancer as an alternative to oophorectomy or ovarian irradiation. For use in women with axillary node- negative breast cancer adjuvant therapy. Metastatic breast cancer in men.
  • Temozolomide Temodar For treatment of adult patients with Scherine refractory anaplastic astrocytoma, i.e., patients at first relapse with disease progression on a nitrosourea and procarbazine containing regimen
  • Teniposide, Vumon In combination with other approved Bristol-Myers VM-26 anticancer agents for induction therapy in Squibb patients with refractory childhood acute lymphoblastic leukemia (all).
  • Testolactone Teslac Used in the treatment of breast cancer. Bristol-Myers Squibb Thioguanine, Thioguanine Antimetabolite used in the treatment of GlaxoSmithKline 6-TG AML, CML, CLL.
  • Thiotepa Thioplex Thiotepa is a cytotoxic agent of the Immunex polyfunctional type, related chemically and Corporation pharmacologically to nitrogen mustard. Thiotepa has been tried with varying results in the palliation of a wide variety of neoplastic diseases. However, the most consistent results have been seen in the following tumors: 1. Adenocarcinoma of the breast. 2. Adenocarcinoma of the ovary. 3. For controlling intracavitary effusions secondary to diffuse or localized neoplastic diseases of various serosal cavities. 4. For the treatment of superficial papillary carcinoma of the urinary bladder.
  • lymphomas such as lymphosarcoma and Hodgkin's disease.
  • Topotecan Hycamtin Treatment of patients with metastatic GlaxoSmithKline carcinoma of the ovary after failure of initial or subsequent chemotherapy. Treatment of small cell lung cancer sensitive disease after failure of first-line chemotherapy.
  • Toremifene Fareston Treatment of advanced breast cancer in Chiron Corp. postmenopausal women. Tositumomab Bexxar Accel. Approv.
  • Herceptin HERCEPTIN as a single agent is indicated Genentech, Inc. for the treatment of patients with metastatic breast cancer whose tumors overexpress the HER2 protein and who have received one or more chemotherapy regimens for their metastatic disease.
  • Herceptin in combination with paclitaxel is indicated for treatment of patients with metastatic breast cancer whose tumors.
  • Tretinoin Vesanoid Induction of remission in patients with acute Roche ATRA promyelocytic leukemia (APL) who are refractory to or unable to tolerate anthracycline based cytotoxic chemotherapeutic regimens.
  • APL promyelocytic leukemia
  • Capsules Valrubicin Valstar For intravesical therapy of BCG-refractory Anthra ⁇ carcinoma in situ (CIS) of the urinary Medeva bladder in patients for whom immediate cystectomy would be associated with unacceptable morbidity or mortality.
  • Vinorelbine Navelbine Navelbine is indicated as a single agent or in GlaxoSmithKline combination with cisplatin for the first-line treatment of ambulatory patients with unreseactable, advanced non-small cell lung cancer (NSCLC).
  • Navelbine is indicated as a single agent or in combination with cisplatin.
  • Stage III NSCLC Navelbine is indicated in combination with cisplatin.
  • Zoledronate Zometa Used in the treatment of patients with Novartis multiple myeloma and patients with documented bone metastases from solid tumors, in conjunction with standard antineoplastic therapy. Prostate cancer should have progressed after treatment with at least one hormonal therapy
  • Endothelial cell migration is performed as described in P. J. Polverini et al., Methods Enzymol. 198:440-450 (1991).
  • the BAMVECs bovine adrenal microvascular endothelial cells, VEC Technologies, Rensselaer, N.Y.
  • EBM endothelial basal medium
  • CellstripperTM Mediatech, Herndon, Va.
  • the chamber is assembled, inverted, and the cells are allowed to adhere for 90 minutes.
  • the test compounds are added to the top part of the wells and incubated 3-4 hours.
  • Membranes are recovered, fixed, stained, and the cells migrated through the filter.
  • the cells are counted at (100 ⁇ ) using 10 fields.
  • FBS fetal bovine serum
  • BSA fetal bovine serum
  • Background migration is subtracted and the data presented is a percentage of FBS-induced migration (% maximal migration).
  • 500 ⁇ l of growth factor reduced Matrigel (BD Bioscience), containing 100 ng/ml bFGF (R&D systems), is prepared on ice and injected in the left chest area of nude mouse anesthetized with isofluorane (5 mice per group).
  • the test compounds are dosed i.v., at 30 mg/kg twice a week. After one week, the plugs are extracted and photographed. Five plugs of the same group are aligned together and snap-frozen in one OTC compound block. Five 5 ⁇ m sections in different depths are obtained from each block using a Leica CM1850 Cryostat. The slides are immediately fixed in cold acetone for 2 minutes and air dried.
  • CD31 immunohistochemical staining of blood vessels is carried out by using an Anti-Rat IG HRP detection kit (BD Pharmingen) and using the methods provided in the manufacturer's instruction manual.
  • the primary CD31 antibody used is Rat IgG2a, Clone MEC13.3 (BD Pharmingen, cat# 550274,).
  • the CD31 antibody is diluted 1: 30-50.
  • the CD31 positive area of every plug is photographed using a Qimaging Micropublisher 5.0 RTV camera coupled with a Nikon Eclipse 80i microscope (20 ⁇ ). ImagePro 5.1 software is used to quantify the CD31 positive area using a common macro throughout the experiment. The total CD31 positive area of five sections of each plug is calculated.
  • Solid phase peptide synthesis of the modified peptide on a 100 ⁇ mole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA.
  • the following protected amino acids are sequentially added to resin: Fmoc-Pro-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Nva-OH, Fmoc-Thr(tBu)—OH, Fmoc-(D-alloIle)-OH, Fmoc-Val-OH, Fmoc-Gly-OH, and Fmoc-Sar-OH.
  • N,N-dimethylformamide DMF
  • HBTU O-benzotriazol-1-yl-N, N, N 1 , N 1 -tetramethyl-uronium hexafluorophosphate
  • DIEA Diisopropylethylamine
  • the peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et 2 0.
  • the product is purified by a reverse phase HPLC using a C 18 column.
  • Solid phase peptide synthesis of the modified peptide on a 100 ⁇ mole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA.
  • the following protected amino acids are sequentially added to resin: Fmoc-Pro-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Lys(Boc)-OH, Fmoc-Nva-OH, Fmoc-Thr(tBu)-OH, Fmoc-(D-allo-Ile)-OH, Fmoc-Val-OH, Fmoc-Gly-OH, and Fmoc-Sar-OH.
  • N,N-dimethylformamide DMF
  • HBTU O-benzotriazol-1-yl-N,N, N 1 , N 1 -tetramethyl-uronium hexafluorophosphate
  • DIEA Diisopropylethylamine
  • the peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et 2 0.
  • the product is purified by a reverse phase HPLC using a C 18 column.
  • Solid phase peptide synthesis of the modified peptide on a 100 ⁇ mole scale is performed using manual solid-phase synthesis, a Symphony Peptide Synthesizer and Fmoc protected Rink Amide MBHA.
  • the following protected amino acids are sequentially added to resin: Fmoc-Pro-OH, Fmoc-Arg(Pbf)-OH, Fmoc-Ile-OH, Fmoc-Nva-OH, Fmoc-Thr(tBu)—OH, Fmoc-(D-allo-Ile)-OH, Fmoc-Val-OH, Fmoc-Gly-OH, and Fmoc-Pro-OH.
  • N,N-dimethylformamide DMF
  • HBTU O-benzotriazol-1-yl-N,N, N 1 , N 1 -tetramethyl-uronium hexafluorophosphate
  • DIEA Diisopropylethylamine
  • the peptide is cleaved from the resin using 85% TFA/5% TIS/5% thioanisole and 5% phenol, followed by precipitation by dry-ice cold Et 2 0.
  • the product is purified by a reverse phase HPLC using a C 18 column.
  • FIG. 12 is provided in FIG. 12 .
  • FIG. 13 is provided in FIG. 13 .
  • FIG. 14 is provided in FIG. 14 .
  • FIG. 15 is provided in FIG. 15 .
  • FIG. 16 is provided in FIG. 16 .
  • FIG. 17 is provided in FIG. 17 .
  • FIG. 18 is provided in FIG. 18 .
  • FIG. 19 is provided in FIG. 19 .
  • FIG. 20 is provided in FIG. 20 .

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Cardiology (AREA)
  • Diabetes (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Urology & Nephrology (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Dermatology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Vascular Medicine (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
US11/817,668 2005-03-03 2006-03-03 Anti-Angiogenic Compounds Abandoned US20080152660A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/817,668 US20080152660A1 (en) 2005-03-03 2006-03-03 Anti-Angiogenic Compounds

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US65865405P 2005-03-03 2005-03-03
US67708905P 2005-05-02 2005-05-02
US11/817,668 US20080152660A1 (en) 2005-03-03 2006-03-03 Anti-Angiogenic Compounds
PCT/US2006/007865 WO2006094269A2 (fr) 2005-03-03 2006-03-03 Composes antiangiogeniques

Publications (1)

Publication Number Publication Date
US20080152660A1 true US20080152660A1 (en) 2008-06-26

Family

ID=36941895

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/817,668 Abandoned US20080152660A1 (en) 2005-03-03 2006-03-03 Anti-Angiogenic Compounds
US11/368,338 Expired - Fee Related US7521425B2 (en) 2005-03-03 2006-03-03 Anti-angiogenic compounds
US12/897,594 Abandoned US20110087010A1 (en) 2005-03-03 2010-10-04 Anti-angiogenic compounds

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/368,338 Expired - Fee Related US7521425B2 (en) 2005-03-03 2006-03-03 Anti-angiogenic compounds
US12/897,594 Abandoned US20110087010A1 (en) 2005-03-03 2010-10-04 Anti-angiogenic compounds

Country Status (6)

Country Link
US (3) US20080152660A1 (fr)
EP (2) EP1853294A4 (fr)
JP (2) JP5167473B2 (fr)
KR (1) KR101373140B1 (fr)
CA (1) CA2598833A1 (fr)
WO (1) WO2006094269A2 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011094671A2 (fr) * 2010-01-29 2011-08-04 The Uab Research Foundation Polypeptides à conjugaison n-terminale pour la thérapie et le diagnostic ciblés

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100560131C (zh) * 2001-10-22 2009-11-18 斯克里普斯研究学院 抗体靶向化合物
JP5167473B2 (ja) * 2005-03-03 2013-03-21 コヴェックス・テクノロジーズ・アイルランド・リミテッド 抗血管新生化合物
AU2011254001B2 (en) * 2007-01-05 2012-08-02 Covx Technologies Ireland Limited Glucagon-like protein-1 receptor (GLP-1R) agonist compounds
US20090098130A1 (en) * 2007-01-05 2009-04-16 Bradshaw Curt W Glucagon-like protein-1 receptor (glp-1r) agonist compounds
US8293714B2 (en) * 2008-05-05 2012-10-23 Covx Technology Ireland, Ltd. Anti-angiogenic compounds
CN102076331B (zh) 2008-06-26 2013-12-18 普罗林科斯有限责任公司 具有可控的药物释放速率的前药和药物-大分子轭合物
EP2395992A2 (fr) * 2009-02-10 2011-12-21 The Scripps Research Institute Vaccination programmée chimiquement
US8980268B2 (en) 2009-07-29 2015-03-17 Regeneron Pharamceuticals, Inc. Methods for treating cancer by administering an anti-Ang-2 antibody
US20120189635A1 (en) 2009-07-29 2012-07-26 Regeneron Pharmaceuticals, Inc. Methods for Treating or Preventing Malaria by Administering an Antibody that Specifically Binds Angiopoietin-2 (Ang-2)
JO3182B1 (ar) 2009-07-29 2018-03-08 Regeneron Pharma مضادات حيوية بشرية عالية الالفة مع تولد الاوعية البشرية - 2
WO2011143614A1 (fr) 2010-05-14 2011-11-17 Amgen Inc. Agonistes des récepteurs de mort améliorés
BR112013000951A2 (pt) 2010-07-12 2016-05-17 Covx Technologies Ireland Ltd conjungados de anticorpo multifuncionais
CA2815967A1 (fr) * 2010-11-05 2012-05-10 Covx Technologies Ireland Limited Composes antidiabetiques
KR20130113493A (ko) 2011-01-03 2013-10-15 에프. 호프만-라 로슈 아게 항―dig 항체 및 펩티드에 접합되어 있는 디그옥시게닌의 복합체의 약학적 조성물
PE20141790A1 (es) 2011-11-17 2014-11-27 Pfizer Peptidos citotoxicos y conjugados de anticuerpo-farmaco de los mismos
EP2793950A2 (fr) 2011-12-20 2014-10-29 Pfizer Inc Procédés améliorés pour la préparation de conjugués peptidiques et de lieurs
TWI560202B (en) * 2011-12-22 2016-12-01 Pfizer Anti-diabetic compounds
ES2721882T3 (es) 2011-12-23 2019-08-06 Pfizer Regiones constantes de anticuerpo modificadas por ingeniería genética para conjugación específica de sitio y procedimientos y usos de las mismas
CA2863216C (fr) 2012-01-09 2020-06-02 Covx Technologies Ireland Limited Anticorps mutants et leur conjugaison
JP6219923B2 (ja) 2012-03-28 2017-10-25 アムジェン インコーポレイテッド Dr5受容体アゴニストの組み合わせ
CN105121421B (zh) 2012-11-05 2020-10-02 辉瑞公司 司普力西欧他汀类似物
PE20150892A1 (es) 2012-11-07 2015-06-11 Pfizer Anticuerpos anti-notch y conjugados de anticuerpo-farmaco
RU2669807C2 (ru) 2014-01-27 2018-10-16 Пфайзер Инк. Бифункциональные цитотоксические агенты
US10647750B2 (en) * 2015-01-09 2020-05-12 Ipsen Bioinnovation Limited Cationic neurotoxins
MX2017012102A (es) 2015-03-20 2018-01-12 Pfizer Agentes citotoxicos bifuncionales que contienen el farmacoforo de cti.
EP3349795B1 (fr) * 2015-09-17 2021-03-17 The Scripps Research Institute Immunoconjugués à double domaine variable et leurs utilisations
TWI778491B (zh) 2015-11-30 2022-09-21 美商輝瑞股份有限公司 位點專一性her2抗體藥物共軛體
EP3411371A1 (fr) 2016-02-01 2018-12-12 Pfizer Inc Analogues de la tubulysine et leurs procédés de préparation
US11364303B2 (en) 2017-09-29 2022-06-21 Pfizer Inc. Cysteine engineered antibody drug conjugates
CN108066285B (zh) * 2017-11-30 2019-06-07 江南大学 一种肝靶向共输送基因/药物的整合纳米系统及制备方法
WO2023212725A2 (fr) * 2022-04-29 2023-11-02 University Of Florida Research Foundation, Incorporated Composés d'anticorps avec cystéine réactive et conjugués anticorps-médicament associés

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6051375A (en) * 1995-09-26 2000-04-18 Rose; Timothy M. Glycoprotein B of the RFHV/KSHV subfamily of herpes viruses
US20050004044A1 (en) * 2003-04-07 2005-01-06 Board Of Regents Use of orsaponin [3beta, 16beta, 17 alpha-trihydroxycholost-5-en-22-one 16-0-(2-0-4-methoxybenzoyl-beta-D-xylopyranosyl)-(1->3)-(2-0-acetyl-alpha-L-arabinopyranoside)] or OSW-1 and its derivatives for cancer therapeutics

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3719667A (en) * 1970-08-24 1973-03-06 Lilly Co Eli Epimerization of 6-acylamido and 6-imido penicillin sulfoxide esters
US3840556A (en) 1971-05-28 1974-10-08 Lilly Co Eli Penicillin conversion by halogen electrophiles and anti-bacterials derived thereby
US5567610A (en) 1986-09-04 1996-10-22 Bioinvent International Ab Method of producing human monoclonal antibodies and kit therefor
US5216132A (en) 1990-01-12 1993-06-01 Protein Design Labs, Inc. Soluble t-cell antigen receptor chimeric antigens
AU660926B2 (en) * 1990-04-06 1995-07-13 La Jolla Cancer Research Foundation Method and composition for treating thrombosis
US5229275A (en) 1990-04-26 1993-07-20 Akzo N.V. In-vitro method for producing antigen-specific human monoclonal antibodies
ES2136092T3 (es) * 1991-09-23 1999-11-16 Medical Res Council Procedimientos para la produccion de anticuerpos humanizados.
US6080840A (en) 1992-01-17 2000-06-27 Slanetz; Alfred E. Soluble T cell receptors
US5573905A (en) * 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5981478A (en) 1993-11-24 1999-11-09 La Jolla Cancer Research Foundation Integrin-binding peptides
US5733757A (en) 1995-12-15 1998-03-31 The Scripps Research Institute Aldolase catalytic antibody
US5739277A (en) * 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
DE19544393A1 (de) 1995-11-15 1997-05-22 Hoechst Schering Agrevo Gmbh Synergistische herbizide Mischungen
US5932545A (en) 1997-03-17 1999-08-03 Abbott Laboratories Antiangiogenic drug to treat cancer, arthritis and retinopathy
GB9722131D0 (en) * 1997-10-20 1997-12-17 Medical Res Council Method
US6326176B1 (en) * 1997-12-18 2001-12-04 The Scripps Research Institute Aldol condensations by catalytic antibodies
EP1078002B1 (fr) 1998-05-22 2008-05-21 Abbott Laboratories Medicaments peptidiques anti-angiogeniques
US6716963B1 (en) * 1998-05-22 2004-04-06 Abbott Laboratories Peptide antiangiogenic drugs
US6774211B1 (en) 1998-05-22 2004-08-10 Abbott Laboratories Peptide antiangiogenic drugs
EP1218413A4 (fr) 1999-09-27 2005-06-15 Univ California Manipulations d'anticorps a liaison irreversible
US6294374B1 (en) * 1999-10-08 2001-09-25 The Scripps Research Institute Use of catalytic antibodies for synthesizing epothilone
CA2391386A1 (fr) 1999-11-22 2001-05-31 Abbott Laboratories Peptides presentant une activite anti-angiogenique
CO5261544A1 (es) 1999-11-22 2003-03-31 Abbott Lab Peptidos n-alquilados que tienen actividad antiangiogenica
US6753408B1 (en) 1999-11-22 2004-06-22 Fortuna Haviv Peptides having antiangiogenic activity
DE60041351D1 (de) * 1999-11-24 2009-02-26 Transave Inc Gssytem
US20020183242A1 (en) 2001-04-11 2002-12-05 Jack Henkin Peptide antiangiogenic drugs
AU2002319402B2 (en) 2001-06-28 2008-09-11 Domantis Limited Dual-specific ligand and its use
US20030045477A1 (en) 2001-07-26 2003-03-06 Fortuna Haviv Peptides having antiangiogenic activity
CN100560131C (zh) * 2001-10-22 2009-11-18 斯克里普斯研究学院 抗体靶向化合物
EP2366718A3 (fr) 2002-06-28 2012-05-02 Domantis Limited Ligand
JP2006523090A (ja) 2002-12-27 2006-10-12 ドマンティス リミテッド リガンドに、そしてリガンド受容体に特異的な二重特異性単一ドメイン抗体
WO2004091542A2 (fr) * 2003-04-15 2004-10-28 Covx Pharmaceuticals, Inc. Composes de ciblage d'integrine contenant de l'azote
JP5167473B2 (ja) * 2005-03-03 2013-03-21 コヴェックス・テクノロジーズ・アイルランド・リミテッド 抗血管新生化合物

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6051375A (en) * 1995-09-26 2000-04-18 Rose; Timothy M. Glycoprotein B of the RFHV/KSHV subfamily of herpes viruses
US20050004044A1 (en) * 2003-04-07 2005-01-06 Board Of Regents Use of orsaponin [3beta, 16beta, 17 alpha-trihydroxycholost-5-en-22-one 16-0-(2-0-4-methoxybenzoyl-beta-D-xylopyranosyl)-(1->3)-(2-0-acetyl-alpha-L-arabinopyranoside)] or OSW-1 and its derivatives for cancer therapeutics

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011094671A2 (fr) * 2010-01-29 2011-08-04 The Uab Research Foundation Polypeptides à conjugaison n-terminale pour la thérapie et le diagnostic ciblés
WO2011094671A3 (fr) * 2010-01-29 2011-12-22 The Uab Research Foundation Polypeptides à conjugaison n-terminale pour la thérapie et le diagnostic ciblés

Also Published As

Publication number Publication date
EP2292248A2 (fr) 2011-03-09
US7521425B2 (en) 2009-04-21
JP5167473B2 (ja) 2013-03-21
US20060205670A1 (en) 2006-09-14
AU2006218437A1 (en) 2006-09-08
EP1853294A2 (fr) 2007-11-14
CA2598833A1 (fr) 2006-09-08
KR101373140B1 (ko) 2014-03-12
EP2292248A3 (fr) 2011-06-29
WO2006094269A3 (fr) 2007-01-11
JP2013056895A (ja) 2013-03-28
EP1853294A4 (fr) 2010-01-27
US20110087010A1 (en) 2011-04-14
KR20070108958A (ko) 2007-11-14
WO2006094269A2 (fr) 2006-09-08
JP2008531734A (ja) 2008-08-14

Similar Documents

Publication Publication Date Title
US7521425B2 (en) Anti-angiogenic compounds
US8912148B2 (en) Methods of treating a disease or condition associated with abnormal angiogenesis
AU2008203641B2 (en) Glucagon-like protein-1 receptor (GLP-1R) agonist compounds
US8937048B2 (en) Anti-angiogenic compounds
CN101287485A (zh) 抗血管生成化合物
AU2011202645B2 (en) Anti-angiogenic compounds
ES2369587T3 (es) Compuestos anti-angiogénicos.
AU2006218437B2 (en) Anti-angiogenic compounds
AU2011254001B2 (en) Glucagon-like protein-1 receptor (GLP-1R) agonist compounds

Legal Events

Date Code Title Description
AS Assignment

Owner name: COVX TECHNOLOGIES IRELAND LIMITED, IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRADSHAW, CURT W.;DOPPALAPUDI, VENKATA RAMANA;LAI, JING-YU;AND OTHERS;REEL/FRAME:019838/0410

Effective date: 20070907

STCB Information on status: application discontinuation

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