WO2003034995A2 - Integrin targeting compounds - Google Patents

Integrin targeting compounds Download PDF

Info

Publication number
WO2003034995A2
WO2003034995A2 PCT/US2002/033866 US0233866W WO03034995A2 WO 2003034995 A2 WO2003034995 A2 WO 2003034995A2 US 0233866 W US0233866 W US 0233866W WO 03034995 A2 WO03034995 A2 WO 03034995A2
Authority
WO
WIPO (PCT)
Prior art keywords
linker
targeting
compound
component
integrin
Prior art date
Application number
PCT/US2002/033866
Other languages
French (fr)
Other versions
WO2003034995A3 (en
Inventor
Carlos F. Barbas
Christoph Rader
Subhash C. Sinha
Original Assignee
The Scripps Research Institute
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 The Scripps Research Institute filed Critical The Scripps Research Institute
Priority to EP02773859A priority Critical patent/EP1446418B1/en
Priority to AU2002337954A priority patent/AU2002337954C1/en
Priority to JP2003537564A priority patent/JP5021152B2/en
Priority to KR1020047005978A priority patent/KR101085784B1/en
Priority to CA002464271A priority patent/CA2464271A1/en
Priority to ES02773859T priority patent/ES2384235T3/en
Publication of WO2003034995A2 publication Critical patent/WO2003034995A2/en
Publication of WO2003034995A3 publication Critical patent/WO2003034995A3/en

Links

Classifications

    • 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
    • A61K38/06Tripeptides
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/66Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid the modifying agent being a pre-targeting system involving a peptide or protein for targeting specific cells
    • A61K47/67Enzyme prodrug therapy, e.g. gene directed enzyme drug therapy [GDEPT] or VDEPT
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6891Pre-targeting systems involving an antibody for targeting specific cells
    • A61K47/6899Antibody-Directed Enzyme Prodrug Therapy [ADEPT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the invention relates to integrin targeting compounds and methods of making and using the compounds.
  • potent chemotherapeutics has some distinct limitations mainly related to the toxicity ofthe drug.
  • the dose of drug that is required for therapy often renders patients susceptible to potentially fatal infections, cardiac toxicity and other side effects. Improved understanding ofthe biology of cancer has led to more specific "targeted therapies.”
  • Antigens expressed on the surface of diseased cells e.g., tumor cells
  • tumor antigens are often down regulated due to drug resistance: this limits the effectiveness ofthe "monoclonal antibody directed drug delivery” approaches.
  • tumor endothelial cells are readily accessible to targeting molecules circulating in the blood. Furthermore, in contrast to tumor cells, tumor endothelial cells are genetically stable and are unlikely to be down regulated due to drug resistance. Hence, molecules expressed on the surface of tumor endothelial cells appear to be an appropriate receptor for "directed drug delivery.”
  • the present invention solves problems ofthe art by providing novel compounds for targeting integrin expressing cells. Such compounds have diagnostic and therapeutic applications in cancer and other diseases.
  • the present invention is directed to targeting compounds comprising at least one integrin targeting component covalently linked via a linker to at least one functional component, wherein said integrin targeting component is selected from a group that includes:
  • the integrin targeting component is specific for an integrin such as ⁇ v ⁇ or ⁇ v ⁇ 5 .
  • the core structures of preferred RGD peptidomimetics are disclosed for use in particular targeting compound embodiments.
  • the integrin targeting compounds ofthe invention confer various benefits over the components themselves.
  • the functional component may generally extend the half-life of a smaller sized targeting component in vivo.
  • the biological potency or other biological feature of an integrin targeting component may be modified by the addition of effector function(s) provided by a functional component such as an antibody.
  • the integrin targeting component through its increased size conferred by linkage to the functional component, may enable the targeting agent to function in new capacities.
  • an integrin targeting component-linker compound is prepared which includes an integrin targeting component and a linker that includes a reactive group for covalent reaction with a susceptible reactive moiety ofthe functional component.
  • a functional component-linker compound is prepared that includes a functional component and a linker that includes a reactive group for covalent reaction with a susceptible reactive moiety of an integrin targeting component.
  • the targeting component and the functional component are linked to one of a linker with a reactive groups or linker with a susceptible moiety so that the targeting compound forms when the two linkers covalently bond.
  • the linker includes a reactive group for covalently linking to the other ofthe components.
  • the linker reactive group is a ketone, a diketone, a beta lactam, a succinimide active ester, haloketone, a lactone, an anhydride, an epoxide, an aldehyde or a maleimide.
  • the linker has the general formula X - Y - Z wherein X is a linear or branched connecting chain of atoms comprising any of C, H, N, O, P, S, Si, F, CI, Br, and I, or a salt thereof, and comprising a repeating ether unit of between 2-100 units; Y is optional and is a single or fused 5 or 6 membered homo- or heterocarbocylic saturated or unsaturated ring located within 1-20 atoms of Z; and Z is a reactive group for covalently linking the one or more targeting agents to susceptible moiety such as a side chain of a reactive amino acid.
  • the targeting component or functional component may be linked to X or Y or to X and Y when multiple components are included in the targeting compound.
  • the method includes administering to the individual an integrin targeting compound ofthe invention.
  • the functional component is a therapeutic agent which includes the functional component.
  • the methods includes administering to the individual a therapeutically effective amount of an integrin targeting compound ofthe invention that includes a functional component that is a therapeutic agent.
  • the disease or condition is susceptible to the therapeutic agent such that a reduction or prevention ofthe symptoms associated with the disease or condition is effected.
  • the disease or condition involves a defect in angiogenesis, bone metabolism, inflammation or cell growth.
  • the invention further provides pharmaceutical compositions or medicaments that include an integrin targeting compound ofthe invention and a pharmaceutically acceptable carrier.
  • FIG. 1 shows exemplary integrin targeting agents of which Panels A-E are RGD peptidomimetic while Panel F is an RGD peptide.
  • the core structures are from the following: U.S. Patent No. 6,335,330 (Panel A), U.S. Patent No. 5,693,636 (Panel B), U.S. Patent No. 6,040,311 (Panel C), and U.S. Patent No. 6,001,117 (Panel E).
  • FIG. 2 is a general linker design (Panel A) and specific embodiment (Panel B; SCS-873) shown in association with a targeting agent.
  • FIG. 3 shows a general scheme of an embodiment of a targeting agent-linker compound with a branched linker and two identical targeting agents (Panel A) with specific embodiments in Panel B (integrin targeting agent diketo linker; compound 26), and Panel C (integrin targeting agent diketo linker; compound 27).
  • the branch point is in the connecting chain portion ofthe linker.
  • FIG. 4 shows a general scheme of an embodiment of a targeting agent-linker compound with a branched linker and two different targeting agents (Panel A) with a specific embodiment in Panel B (integrin targeting and folate targeting agent diketo linker; compound 28).
  • the branch point is in the connecting chain portion ofthe linker.
  • TI is an integrin targeting component while T2 is a folate receptor targeting component.
  • FIG. 5 shows a general scheme of an embodiment of a targeting agent-linker compound with a branched linker and two different targeting agents (Panel A) with a specific embodiment in Panel B (integrin targeting agent diketo linker; compound 29).
  • the branch point is in the recognition group portion ofthe linker.
  • FIG. 6 shows the structure of linker reactive groups.
  • Structures A-C form reversible covalent bonds with reactive nucleophilic group (e.g. lysine or cysteine side chain) in the combining site of an antibody (structure A could form an irreversible covalent bond
  • X is N and if R ⁇ and R form part of a cyclic structure).
  • Ri and R 2 and R 3 in structures A-C represent substituents which can be C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof.
  • X is N, C, Si, 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. Structures D-G form nonreversible covalent bonds with reactive nucleophilic group (e.g.
  • FIG. 7 shows various electrophiles suitable for reactive modification with a reactive amino acid side chain of an antibody.
  • A acyl beta-lactam
  • B simple diketone
  • C succinimide active ester
  • D maleimide
  • E haloacetamide with linker
  • F haloketone
  • G cyclohexyl diketone
  • H aldehyde.
  • R refers to other structure that may include a targeting agent, linker or antibody, while X refers to halogen.
  • FIG. 8 shows the structure of linker recognition group (Y), situated between the reactive group portion and the connecting chain portion ofthe linker.
  • Panel A shows the relationship ofthe recognition group Y within the linker (see FIG. 2).
  • Panels B-D show distance of Y from Z, substituents on the ring and ring member atoms.
  • FIG. 9 shows the structure ofthe linker connecting chain (X), which directly attaches at one end to the targeting agent as shown in Panel A (see FIG. 2).
  • Substituents R 2 to R 4 can be C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof, and may include a group such as an alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl as well as a carbocyclic or heterocyclic mono or fused saturated or unsaturated ring structure.
  • n, r or m is 1-100
  • FIG. 10 shows Scheme 1, a synthetic scheme for the amine precursor of SCS-873, targeting agent 3 or SCS-amine. Key: (a) BBr 3 , CH 2 C1 2 , -20 °C, 2h; (b) DMF, rt to 80°C, 3h; (c) BnCOCl, sat. aq.
  • FIG. 13 shows Scheme 4, a synthetic scheme for making Compounds SCS-864 and SCS-789. Key: (a) Et3N, DMF, rt, 16h.
  • FIG. 14 shows an embodiment whereby two targeting components are linked to a single linker.
  • TI is an integrin targeting component and T2 is biotin (compound 30).
  • the present invention provides integrin targeting compounds useful for presentation to a particular integrin target such as a cell or tissue.
  • the targeting component operates to situate the compound at the target site.
  • Integrin targeting compounds comprise at least two components: a targeting component and a functional component. The components are operatively linked such that each component retains its activity.
  • the targeting component specifically binds to the integrin target typically through a ligand/receptor relationship. Such ligand/receptor relationships are well known in the art.
  • the integrin target is on the surface ofthe target cell or tissue.
  • the integrin target may also be associated with a particular condition such a pathological condition.
  • a preferred targeting component targets to one or more integrins.
  • the targeting component can be either an agonist or antagonist to the integrin or bind without any biological activity.
  • the integrin is ⁇ v ⁇ or ⁇ v ⁇ 5 .
  • Integrins are heterodimeric transmembrane glycoprotein complexes that function in cellular adhesion events and signal transduction processes. Integrin ⁇ v ⁇ 3 is expressed on numerous cells and has been shown to mediate several biologically relevant processes, including adhesion of osteoclasts to the bone matrix, migration of vascular smooth muscle cells, and angiogenesis. Integrin ⁇ v ⁇ 3 antagonists may be employed in the treatment of several human diseases, including diseases involving neovascularization, such as rheumatoid arthritis, cancer, ocular diseases, and the like.
  • the integrin targeting component of integrin targeting compounds ofthe invention may be a small molecule organic compound of about 5,000 daltons or less such as a drug or pharmaceutical, which is an integrin antagonist or agonist.
  • An integrin agonist or antagonist also can be a protein, peptide, peptidomimetic, glycoprotein, proteoglycan, lipid, phospholipid, lipopolysaccharide, glycolipid, nucleic acid, proteoglycan, carbohydrate, and the like.
  • polypeptide polypeptide
  • peptide and “protein” are used interchangeably to refer to a polymer of amino acid residues.
  • amino acid polymers in which one or more amino acid residue is a synthetic chemical analogue (e.g., para-methyl-tyrosine, para- chloro-phenylanine, and the like) of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers.
  • Amino acids can be in the L or D form so long as the binding function ofthe peptide is maintained.
  • Peptides can be of variable length, generally between about 4 and 200 amino acids.
  • 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.
  • the integrin targeting component is not an antibody, although the targeting compound may include an antibody as the functional component and such antibody may have a targeting capability. If such antibody has an integrin targeting capability, the targeting compound will include a non-antibody component that is integrin specific.
  • Arg-Gly-Asp peptide or "RGD peptide” is intended to refer to a peptide having one or more Arg-Gly-Asp containing sequences which may function as a binding site for a receptor ofthe "Arg-Gly-Asp family of receptors", e.g., an integrin.
  • Integrins which comprise and alpha and a beta subunit, include numerous types including ⁇ , ⁇ 2 ⁇ , ⁇ 3 ⁇ , ⁇ 4 ⁇ 1; ⁇ 5 ⁇ , ⁇ 6 ⁇ , ⁇ 7 ⁇ , ⁇ 8 ⁇ , ⁇ 9 ⁇ ⁇ 1; ⁇ 6 ⁇ 4 ⁇ ⁇ 4 ⁇ 7 , ⁇ D ⁇ 2, ⁇ D ⁇ 2.
  • RGD is present in several matrix proteins and is the target for cell binding to matrix by integrins.
  • Platelets contain a large amount of RGD-cell surface receptors ofthe protein GP II b / ⁇ i a , which is primarily responsible, through interaction with other platelets and with the endothelial surface of injured blood vessels, for the development of coronary artery thrombosis.
  • the term RGD peptide also includes peptides with amino acids that are functional equivalents (e.g., RLD or KGD) of RGD peptide provided that they interact with the same RGD receptor.
  • Peptides containing RGD sequences can be synthesized from amino acids by means well known in the art, using, for example, an automated peptide synthesizer, such as those manufactured by Applied Biosystems, Inc., Foster City, California.
  • the integrin targeting component ofthe targeting compounds ofthe invention can be a non-RGD peptide agonist or antagonist or peptidomimetic thereof.
  • non-RGD peptide refers to a peptide that is an antagonist or agonist of integrin binding to its ligand (e.g. fibronectin, vitronectin, laminin, collagen etc.), but does not involve an RGD binding site.
  • ligand e.g. fibronectin, vitronectin, laminin, collagen etc.
  • Such non-RGD integrin targeting compounds are known for v ⁇ (see, e.g., U.S. Pat. Nos.
  • the integrin targeting component is a peptidomimetic agonist or antagonist, which preferably is a peptidomimetic agonist or antagonist of an RGD peptide or non-RGD peptide.
  • peptidomimetic is a compound containing non-peptidic structural elements that are capable of mimicking or antagonizing the biological action(s) of a natural parent peptide.
  • a peptidomimetic of an RGD peptide is an organic molecule that retains similar peptide chain pharmacophore groups ofthe RGD amino acid sequence but lacks amino acids or peptide bonds in the binding site sequence.
  • a peptidomimetic of a non-RGD peptide is an organic molecule that retains similar peptide chain pharmacophore groups ofthe non-RGD binding site sequence but lacks amino acids or peptide bonds in the binding site sequence.
  • a "pharmacophore” is a particular three- dimensional arrangement of functional groups that are required for a compound to produce a particular response or have a desired activity.
  • the term "RGD peptidomimetic" is intended to refer to a compound that comprises a molecule containing the RGD pharmacophores supported by an organic/non-peptide structure. It will be understood that an RGD peptidomimetic (or non-RGD peptidomimetic) may be part of a larger molecule that itself includes conventional or modified amino acids linked by peptide bonds.
  • RGD peptidomimetics are well known in the art, and have been described with respect to integrins such as GPIIb/IIIa, ⁇ v ⁇ 3 and ⁇ v ⁇ 5 (See, e.g., Miller et al., J. Med. Chem. 2000, 43:22-26; and International Patent Publications WO 0110867, WO 9915178, WO 9915170, WO 9815278, WO 9814192, WO 0035887, WO 9906049, WO 9724119 and WO 9600730; see also Kumar et al., Cancer Res. 61:2232-2238 (2000)). Many such compounds are specific for more than one integrin.
  • RGD peptidomimetics are generally based on a core or template (also referred to as "fibrinogen receptor antagonist template"), to which are linked by way of spacers to an acidic group at one end and a basic group at the other end ofthe core.
  • the acidic group is generally a carboxylic acid functionality while the basic group is generally a N-containing moiety such as an amidine or guanidine.
  • the core structure adds a form of rigid spacing between the acidic moiety and the basic nitrogen moiety, and contains one or more ring structures (e.g., pyridine, indazole, etc.) or amide bonds for this purpose.
  • a fibrinogen receptor antagonist generally, about twelve to fifteen, more preferably thirteen or fourteen, intervening covalent bonds are present (via the shortest intramolecular path) between the acidic group ofthe RGD peptidomimetic and a nitrogen ofthe basic group.
  • the number of intervening covalent bonds between the acidic and basic moiety is generally shorter, two to five, preferably three or four, for a vitronectin receptor antagonist.
  • the particular core may be chosen to obtain the proper spacing between the acidic moiety ofthe fibrinogen antagonist template and the nitrogen atom ofthe pyridine.
  • a fibrinogen antagonist will have an intramolecular distance of about 16 angstroms (1.6 nm) between the acidic moiety (e.g., the atom which gives up the proton or accepts the electron pair) and the basic moiety (e.g., which accepts a proton or donates an electron pair), while a vitronectin antagonist will have about 14 angstroms (1.4 nm) between the respective acidic and basic centers. Further description for converting from a fibrinogen receptor mimetic to a vitronectin receptor mimetic can be found in U.S. Pat. No. 6,159,964.
  • the peptidomimetic RGD core can comprise a 5-11 membered aromatic or nonaromatic mono- or polycyclic ring system containing 0 to 6 double bonds, and containing 0 to 6 heteroatoms chosen from N, O and S.
  • the ring system may be unsubstituted or may be substituted on a carbon or nitrogen atom.
  • Preferred core structures with suitable substituents useful for vitronectin binding include monocyclic and bicyclic groups, such as benzazapine described in WO 98/14192, benzdiazapine described in U.S. 6,239,168, and fused tricyclics described in U.S. 6,008,213.
  • U.S. Pat. No. 6,159,964 contains an extensive list of references in Table 1 of that document which disclose RGD peptidomimetic cores structures (referred to as fibrinogen templates) which can be used for prepraring RGD peptidomimetics.
  • RGD peptidomimetic cores structures referred to as fibrinogen templates
  • Preferred vitronectin RGD and fibronectin RGD peptidomimetics are disclosed in U.S. Patent Nos.
  • RGD peptidomimetic integrin targeting agents are shown below as compounds 1, 2, and 3 can be used for preparing an intregrin targeting compound ofthe present invention.
  • the linker is attached as indicated to the nitrogen ofthe seven membered ring.
  • Other RGD peptidomimetic integrin targeting agents include compound 31, wherein P and L or carbon or nitrogen.
  • the linker may be Rl or R2 while the R3 group includes a basic group such as an -NH group.
  • the R3 group is as shown in structures 1, 2, or 3.
  • the R3 group includes a heterocyclic group such a benzimidazole, imidazole, pyridine group, or the like.
  • the R3 group is a alkoxy group, such as a propoxy group or the like, that is substituted with a heterocarbyl group that is substituted with an alkylamine group, such as a methylamino group or the like, whereas in other embodiments, the R3 group is an alkoxy group, such as a propoxy group or the like, substituted with a heterocyclylamino group, such as with a pyridinylamino group or the like such as a 2-pyridinylamino group.
  • the linker may be any of Ri, R 2 , R , while R 4 may be a linker or a hydrolyzable group such as alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl group, and the like.
  • a targeting compound ofthe invention can contain more than one targeting component.
  • one ofthe targeting components targets an integrin as disclosed above.
  • the second targeting component can target another entity.
  • chemokine receptors such as CCR5, interleukin or cytokine receptors, vitamin receptors such as the folate receptor, cancer markers such as prostate specific antigen, carcinoembryonic antigen, HER-2, viral markers such as HIV gp41, or a peptide hormone receptor.
  • Linear or branched linkers may be used for preparing dual agent targeting compounds. Exemplary branched linker designs are shown in FIGs. 3-5.
  • Compound 32 An exemplary such dual targeting compound (Compound 32) is shown below, where the compound targets both integrin and the folate receptor.
  • Another dual targeting compound that uses a single linear linker and targets integrin and avidin is shown in FIG. 14 (compound 30).
  • Compound 30 which has a shorter linker than in other examples, has several uses.
  • compound 30 can be used in conjunction with a detectably labeled avidin or streptavidin to test cells for expression ofthe integrin target reactive with TI.
  • Compound 30 can be administered in vivo together with avidin or streptavidin labeled with a suitable therapeutic or imaging agent.
  • the targeting component is linked to a functional component in that it may have one or more biological activities, each activity characterized as a detectable biological affect on the functioning of a cell organ or organism.
  • a targeting compound may, however, be a pure binding compound without biological activity. In the case where the targeting compound has functional activity, the compound would include a functional component that is distinct from the targeting component.
  • the functional component of a targeting compound can be any structure having a desired biological activity toward a cell or tissue associated with integrin.
  • the functional component is not a metal chelating structure with or without the associated metal ion, e.g., a radiometal ion.
  • the functional component is not a lipid.
  • the functional component may include any of a number of biologically active structures well known in the art.
  • Biological agents suitable as the functional component of the targeting compounds include, but are not limited to, small molecule drugs (a pharmaceutical organic compound of about 5,000 daltons or less), organic molecules, proteins, peptides, peptidomimetics, glycoproteins, proteoglycans, lipids, phospholipids, lipopolysaccharides, glycolipids, nucleic acids, proteoglycans, carbohydrates, and the like.
  • the biological agent functional component may be anti-neoplastic, antimicrobial, a hormone, an effector, and the like.
  • Suitable functional components include well known therapeutic compounds such as anti-neoplastic agents include paclitaxel, daunorubicin, doxorubicin, carminomycin, 4'-epiadriamycin, 4-demethoxy-daunomycin, 11 - deoxydaunorubicin, 13-deoxydaunorubicin, adriamycin-14-benzoate, adriamycin-14- octanoate, adriamycin-14-naphthaleneacetate, vinblastine, vincristine, mitomycin C, N- methyl mitomycin C, bleomycin A2, dideazatetrahydrofolic acid, aminopterin, methotrexate, cholchicine and cisplatin, and the like.
  • anti-neoplastic agents include paclitaxel, daunorubicin, doxorubicin, carminomycin, 4'-epiadriamycin
  • Suitable anti-microbial agent functional components include aminoglycosides including gentamicin, antiviral compounds such as rifampicin, 3'- azido-3'-deoxythymidine (AZT) and acylovir, antifungal agents such as azoles including fluconazole, plyre macrolides such as amphotericin B, and candicidin, anti-parasitic compounds such as antimonials, and the like.
  • Suitable hormone functional components may include toxins such as diphtheria toxin, cytokine such as CSF, GSF, GMCSF, TNF, erythropoietin, immunomodulators or cytokines such as the interferons or interleukins, a neuropeptide, reproductive hormone such as HGH, FSH, or LH, thyroid hormone, neurotransmitters such as acetylcholine, hormone receptors such as the estrogen receptor.
  • Suitable functional components also include non-steroidal anti-inflammatories such as indomethacin, salicylic acid acetate, ibuprofen, sulindac, piroxicam, and naproxen, and anesthetics or analgesics.
  • the functional component is not a radioisotope.
  • Functional components can be naturally occurring or synthetic and may be biologically active in their native state where they can act, for example, at the surface of a target cell or can be transported into the target cell to act intracellularly.
  • the functional component can also be an "antibody” which as used herein includes 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. Also subclasses ofthe heavy chain are known. For example, IgG heavy chains in humans can be any of IgGl, IgG2, IgG3 and IgG4 subclass.
  • 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 ) and variable heavy chain (V H ) refer to these light and heavy chains respectively.
  • 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 I by a disulfide bond.
  • the 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 an Fab' monomer.
  • the Fab' monomer is essentially a Fab fragment with part ofthe hinge region (see, Fundamental Immunology, W. E.
  • antibody fragments are defined in terms ofthe digestion of an intact antibody, one of skill will appreciate that any of a variety of antibody fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology.
  • antibody as used herein also includes antibody fragments either produced by the modification of whole antibodies or synthesized de novo or antibodies and fragments obtained by using recombinant DNA methodologies.
  • the T cell receptor is a disulfide linked heterodimer composed of ⁇ or ⁇ chains or , on a minority of T cells, ⁇ or ⁇ 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 (V ⁇ or V ⁇ ) and one constant domain (C ⁇ or C ⁇ ).
  • V ⁇ or V ⁇ Antibody-like variable domain
  • C ⁇ or C ⁇ constant domain
  • the full TcR has a molecular mass of about 95 kDa with the individual chains varying in size from 35 to 47 kDa.
  • TCR soluble T cell receptor
  • GPI glycosyl phosphatidylinositol
  • PI-PLC phosphatidylinositol specific phospholipase C
  • 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. Patent No. 5,216,132 or as soluble TcR single chains as described by Schusta et al. Nature Biotech. 18,754-759 (2000) or Holler et al. Proc. Natl. Acad. Sci (USA) 97:5387-5392 (2000).
  • the TcR "antibodies" as soluble products may be used in place of antibody for making the compounds ofthe invention.
  • the combining site ofthe TcR can be identified by reference to CDR regions and other framework residues using the same methods discussed above for antibodies.
  • Recombinant antibodies may be conventional full length antibodies, antibody fragments known from proteolytic digestion, unique antibody fragments such as Fv or single chain Fv (scFv), domain deleted antibodies, and the like. Fragments may include a domains or polypeptides with as little as one or a few amino acid deleted or mutated while more extensive deletion is possible such as deletion of one or more domains.
  • An Fv antibody is about 50 Kd in size and comprises the variable regions ofthe light and heavy chain.
  • a single chain Fv (“scFv”) polypeptide is a covalently linked VH-V L heterodimer which may be expressed from a nucleic acid including V H - and VL-encoding sequences either joined directly or joined by a peptide-encoding linker.
  • V H - and VL-encoding sequences either joined directly or joined by a peptide-encoding linker.
  • Huston, et al. (1988) Proc. Nat. Acad. Sci. USA, 85:5879-5883 A number of structures for converting the naturally aggregated, but chemically separated light and heavy polypeptide chains from an antibody V region into an scFv molecule which will fold into a three dimensional structure substantially similar to the structure of an antigen-binding site. See, e.g. U.S. Patent Nos. 5,091,513, 5,132,405 and 4,956,778.
  • An exemplary functional component which is an antibody is one that recognizes and binds to the target ofthe targeting component.
  • the target is an integrin
  • exemplary and preferred such antibodies are LM609 and its humanized form known as Vitaxin (See, e.g. Publications WO 89/05155 and WO 01/30393.
  • Another class of antibodies that can be inco ⁇ orated into a targeting compound of the invention is an antibody with a reactive amino acid side chain in the combining site.
  • 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 ofthe N-terminal variable ("V") regions ofthe heavy ("H") and light (“L”) chains.
  • V N-terminal variable
  • H heavy
  • L light 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).
  • CDRs complementarity determining regions
  • the three hypervariable regions of a light chain (LCDR1, LCDR2, and LCDR3) and the three hypervariable regions of a heavy chain (HCDRl, HCDR2 and HCDR3) are disposed relative to each other in three dimensional space to form an antigen binding surface or pocket.
  • the antibody combining site therefore represents the amino acids that make up the CDRs of an antibody and any framework residues that make up the binding site pocket.
  • antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. (see, "Sequences of Proteins of Immunological Interest," E. Kabat et al., U.S. Department of Health and Human Services; Johnson, G and Wu, TT (2001) Kabat Database and its applications: future directions. Nucleic Acids Research, 29: 205-206; http://immuno.bme.nwa.edu).
  • the positions ofthe CDRs may also be identified as the structural loop structures originally described by Chothia and others, (see Chothia and Lesk, J. Mol. Biol.
  • LCDR1 Start - Approximately residue 24. Residue before is always a Cys. Residue after is always a T ⁇ . Typically TRP is followed with TYR-GLN, but also may be followed by LEU-GLN, PHE-GLN, or TYR-LEU. Length is 10 to 17 residues.
  • Sequence before is generally ILE-TYR, but also may be VAL-TYR, ILE-LYS, or ILE-PHE.
  • Length is generally 7 residues.
  • HCDRl Start - at approximately residue 26 (four residues after a CYS) [Chothia / AbM definition] Kabat definition starts 5 residues later. Sequence before is CYS-X-X-X.
  • Residues after is a TRP, typically followed by VAL, but also followed by ILE, or ALA.
  • Length is 10 to 12 residues under AbM definition while Chothia definition excludes the last 4 residues.
  • HCDR2 Start - 15 residues after the end of Kabat /AbM definition of CDR-H1. Sequence before typically LEU-GLU-TRP-ILE-GLY (SEQ ID NO: 1), but a number of variations are possible.
  • HCDR3 Start -33 residues after end of CDR-H2 (two residues after a CYS). Sequence before is CYS-X-X (typically CYS-ALA-ARG). Sequence after is TRP-GLY-X-GLY. Length is 3 to 25 residues.
  • the identity ofthe amino acid residues in a particular antibody that are outside the CDRs, but nonetheless make up part ofthe combining site by having a side chain that is part ofthe lining ofthe combining site (i.e., it 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., Riechmann et al., (1988) Nature, 332:;323-327.
  • the aldolase antibody mouse mAb 38C2 which has a reactive lysine near to but outside HCDR3, is an example of such an antibody.
  • the reactive residue ofthe antibody combining site may be naturally 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 so as to encode the particular residue (see, e.g., WO 01/22922 to Meares et al.).
  • the amino acid residue or its reactive elements e.g., a nucleophilic amino group or sulfhydryl group
  • covalent linkage with the antibody occurring "through an amino acid residue in the combining site ofthe antibody” as used herein means that linkage can be made directly to an amino acid residue of an antibody combining site or indirectly through a chemical moiety that is linked to a side chain of an amino acid residue of an antibody combining site.
  • Functional components that are proteins can be linked to targeting components via a reactive side chain in the protein.
  • the reactive side chain may be present or may arise by mutation.
  • the reactive side chain in lysine (epsilon amino group) may be covalently linked to a linker comprising a ketone, diketone, beta lactam, active ester haloketone, lactone, anhydride, maleimide, epoxide, aldehyde amidine, guanidine, imines, eneamines, phosphates, phosphonates, epoxides, aziridines, thioepoxides, masked or protected diketones (ketals for example), lactams, haloketones, aldehydes, and the like.
  • Such a reactive lysine side chain is present in the combining site of an aldolase antibody e.g., mouse monoclonal antibody mAb 38C2 and other like catalytic antibodies as well as suitably humanized and chimeric versions of such antibodies.
  • Mouse mAb 38C2 is the prototype of a new class of catalytic antibodies that were generated by reactive immunization and mechanistically mimic natural aldolase enzymes (Barbas et al., 1997, Science 278, 2085-2092).
  • these antibodies catalyze aldol and retro-aldol reactions using the enamine mechanism of natural aldolases (Wagner et al., 1995, Science 270, 1797-1800; Barbas et al., 1997, Science 278, 2085-2092; Zhong et al., 1999, Angew. Chem. Int. Ed. 38, 3738-3741; Karlstrom et al., 2000, Proc. Natl. Acad. Sci. U.S.A., 973878-3883).
  • synthetic organic chemistry e.g., Hoffmann et al., 1998, J. Am. Chem. Soc.
  • aldolase antibodies have been used to activate camptothecin, doxorubicin, and etoposide prodrugs in vitro and in vivo as an anti-cancer strategy (Shabat et al., 1999, Proc. Natl. Acad. Sci. U.S.A. 96, 6925- 6930 and ,2001, Proc. Natl. Acad. Sci. U.S.A. 98, 7528-7533 ).
  • Catalytic antibodies are a preferred source of such functional component antibodies include aldolase antibodies, beta lactamase antibodies, esterase antibodies, amidase antibodies, and the like.
  • Suitable such linkers may include a ketone, a diketone, a beta lactam, a succinamide haloketone, a lactone, an anhydride, a maleimide, an alpha-haloacetamide, a cyclohexyl diketone group, and the like (see above). Examples of such linker compounds are shown in FIGs. 2-5.
  • the reactive amino acid of a functional component such as an antibody may be a reactive cysteine, serine or tyrosine residue.
  • cysteines the resulting antibody may form a covalent linkage with maleimide-containing components or other thiol- reactive groups such as iodoacetamides, aryl halides, disulfhydryls and the like.
  • Reactive cysteines may be found in thioesterase catalytic antibodies as described by Janda et al., Proc. Natl. Acad. Sci. (USA) 91:2532-2536, (1994). For other esterase antibodies, see Wirsching et al., Science 270:1775-82 (1995).
  • Reactive amino acid-containing functional components may be prepared by means well known in the art including mutating the amino acid residue to encode for the reactive amino acid or chemically derivatizing an amino acid side chain that contains the reactive group.
  • the components of the targeting compound are linked preferably covalently and preferably with a linker or branched linker.
  • the targeting component may be covalently linked to the antibody combining site by using a linker.
  • An appropriate linker can be chosen to provide sufficient distance between the targeting component and the functional component in order for the targeting component to be able to bind to its target molecule.
  • an appropriate linker can be chosen to provide sufficient distance between the targeting component and the antibody combining site in order for the targeting component to be able to bind to its target molecule. This distance depends on several factors including, for example, the distance from the outermost surface ofthe antibody combining site to the reactive side chain in the combining site, and the nature ofthe targeting agent.
  • the linker will be between about 5 to 10 angstroms (0.5 to 1 nm) in length, with 10 or more angstroms (1.0 nm) being more preferred, although shorter linkers of about 3 angstroms (0.3 nm) in length may be sufficient if the amino acid side chain is very near to the outermost portion ofthe combining site and/or the targeting component includes a segment that can function as a part of a linker.
  • Linker length may also be viewed in terms of the number of linear atoms (cyclic moieties such as aromatic rings and the like to be counted by taking the shortest route). Linker length under this measure is generally about 10 to 200 atoms and more typically about 30 or more atoms, although shorter linkers of two or more atoms may be sufficient if the reactive amino acid side chain is very near to the outermost portion ofthe antibody combining site. Generally, linkers with a linear stretch of at least about 9 atoms are sufficient.
  • the above linker lengths for linking to antibody combining sites are generally applicable to linking integrin targeting components to non-antibody functional components.
  • linker considerations include the effect ofthe linker on physical or pharmacokinetic properties ofthe resulting targeting compound such as, solubility, lipophilicity, hydrophilicity, hydrophobicity, stability (more or less stable as well as planned degradation), rigidity, flexibility, immunogenicity, modulation of binding, chemical compatibility with targeting agent, ability to be inco ⁇ orated into a micelle or liposome, and the like.
  • the linker may be attached to the spacer between the core ofthe molecule and the basic or acid group. Alternatively, the linker can be attached to the core itself.
  • the linker includes any atom from the group C, H, N, O, P, S, Si, halogen (F, CI, 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, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl group, as well as a carbocyclic or heterocyclic mono or fused saturated or unsaturated ring structure. Combinations ofthe above groups and rings may also be present in the linkers ofthe targeting compounds ofthe invention.
  • FIG. 2 The general design of a embodiment of a linear linker for use in targeting compounds ofthe present invention is shown in FIG. 2.
  • the linker includes three functional areas identified starting from the targeting agent (T), being the connecting chain (X), the recognition group (Y) and the reactive group (Z).
  • the integrin targeting agent-linker compound SCS-873 is shown in FIG. 2 with the linker portions X, Y and Z identified. In some embodiments, the recognition group may not be needed.
  • Other groups may include lactone, anhydride, and alpha- haloacetamide and epoxide.
  • Exemplary linker electrophilic reactive groups that can covalently bond to a reactive nucleophilic group (e.g.
  • Linker diketone reactive groups which form reversible covalent bonds with the reactive lysine or cysteine of a functional component or integrin targeting component are shown in FIG. 6.
  • Ri and R 2 and R 3 in structures A-C represent substituents which can be C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof.
  • 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.
  • Other diketone linker reactive groups are shown in FIG. 7 as structures B and G.
  • FIG. 7 also includes the structures of other preferred linker reactive groups.
  • the linker may include a reactive group that forms a nonreversible covalent bond with the combining site of an antibody.
  • exemplary such reactive groups shown as structures D-G in FIG. 6, are useful for nonreversibly attaching a targeting component-linker to a reactive nucleophilic group (e.g. lysine or cysteine side chain) in the functional component (or vice versa).
  • a reactive nucleophilic group e.g. lysine or cysteine side chain
  • FIG. 7 Other diketone linker reactive groups that form nonreversible covalent bonds are shown in FIG. 7 as structures A, C and D.
  • Linkers may optionally contain a recognition group Y situated between the reactive group portion and the connecting chain portion ofthe linker such as shown in FIG. 2. While not wishing to be bound by any theory, the recognition group, if present, may work to properly position the reactive group into a binding site such an antibody combining site so that it may react with a reactive amino acid side chain.
  • FIG. 8 shows a variety of exemplary recognition groups with one or more homo or hetero ring structures of five or six atoms. Larger ring structures are also possible.
  • FIG. 9 Various embodiments ofthe connecting chain X portion ofthe general linker design (FIG. 2) is shown in FIG. 9. As shown, the connecting chain may vary considerably in length with both straight chain and branched chain structures possible.
  • a preferred linker for use in targeting compounds ofthe invention and for preparing targeting agent-linker compounds or functional component-linker compounds is a linker with a 1,3 -diketone reactive group have the structure 33 as shown below where n is from 1-100 or more and preferably is 1, 2, or 4, and more preferably is 3.
  • the linker is a repeating polymer such as polyethylene glycol.
  • the linker reactive group or similar such reactive group that may be inherent in the targeting component can be chosen for linkage with a particular functional component.
  • 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 1,3-diketone configuration such as the diketone shown in Compound SCS-873 (see below) or SCS-864 (see below), is especially preferred as a substrate for modification by an aldolase antibody.
  • a linker reactive group chemical moiety suitable for covalent modification by a reactive sulfhydryl group of a functional component or integrin targeting component 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 protein functional components may possess an electrophilic group that reacts with a nucleophilic group on the targeting component or its linker, whereas in other embodiments a reactive nucleophilic group in an amino acid side chain of a protein functional group reacts with an electrophilic group in a targeting component or linker.
  • protein component side chains may be substituted with an electrophile (e.g., FIGs. 6 and 7) and this group may be used to react with a nucleophile on the targeting component or its linker (e.g., NH 2 ).
  • the functional and targeting components each have a partial linker with appropriate reactive moieties at each end so that the two ends ofthe partial linker can form the full linker, thus creating the complete targeting compound.
  • Integrin targeting compounds may be prepared by several approaches.
  • a targeting component-linker compound is synthesized with a linker that includes one or more reactive groups designed for covalent reaction with a susceptible reactive moiety on the functional component.
  • the suitable reactive moiety can be a side chain of an amino acid.
  • the component-linker compound and functional component are then combined under conditions where the linker reactive group forms a covalent bond with the functional component.
  • "Susceptible" as used herein with reference to a chemical moiety indicates that the chemical moiety will covalently bond with a compatible reactive group.
  • an electrophilic group is susceptible to covalent bonding with a nucleophillic group and vice versa.
  • linking can be achieved by synthesizing a functional component-linker compound comprising the functional component and a linker where the linker includes one or more reactive groups designed for covalent reaction with a susceptibe chemical moiety ofthe targeting component.
  • the targeting component may need to be modified to provide the appropriate reactive moiety for reaction with the linker reactive group.
  • the functional component-linker and targeting component are combined under conditions where the linker reactive group covalently links to the targeting component.
  • a further approach for forming targeting compounds ofthe invention uses a dual linker design.
  • a targeting component-linker compound is synthesized which comprises a targeting component and a linker with a reactive group.
  • a functional component-linker compound also is synthesized which comprises a functional component and a linker, the latter with a chemical moiety susceptible to reactivity with the reactive group ofthe component-linker ofthe first step. These two linker containing compounds are then combined under conditions whereby the linkers covalently link, forming the targeting compound.
  • a functional component-linker compound is synthesized which comprises a functional component and a linker with a reactive group.
  • a targeting component-linker compound is also prepared which comprises the component and a linker, the latter with a chemical moiety susceptible to reactivity with the reactive group of the antibody-linker ofthe first step. These two linker containing compounds are then combined under conditions whereby the linkers covalently link, forming the targeting compound.
  • linker to the targeting agent or antibody combining site.
  • 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.
  • a functional component can be linked to a targeting component using a linker moiety that is labile under certain conditions.
  • the labile linkage may be between the functional component and the linker, between the targeting component and the linker, or within the linker, or combinations thereof.
  • the linker may be labile when subjected to a certain pH.
  • the linker may also be a substrate for a particular enzyme, such as an enzyme present in body fluids.
  • the particular design ofthe labile linker may be used to direct the release ofthe biological agent functional component after it has reached it intended target.
  • a labile linker may be a reversibly covalent bond.
  • Such linker may be an acid-labile linker such as a cis-aconitic acid linker that takes advantage ofthe acidic environment of different intracellular compartments such as the endosomes encountered during receptor mediated endocytosis and the lysosomes. See Shen et al., Biochem. Biophys. Res. Commun. (1981) 102:1048-1054; Yang et al., J. Natl. Cane. Inst. (1988) 80: 1154-1159.
  • a peptide spacer arm is employed as the linker so that the functional component can be released by the action of a peptidase such as a lysosomal peptidase.
  • Labile linkers include, reversible covalent bonds, pH sensitive linkages (acid or base sensitive), enzyme sensitive linkages, degradation sensitive linkers, photosensitive linkers, sand the like, and combinations thereof. These features are also characteristic of a prodrug which can be considered as a type of labile linker.
  • a variety of labile linkers have been previously moieties designed.
  • prodrugs can be formed using compounds having carboxylic acid that slowly degrade by hydrolysis as described in U.S. Patent No. 5,498,729.
  • the functional component can be a "prodrug,” meaning that the functional component is essentially therapeutically inactive, but becomes active upon some modification.
  • the prodrug can be delivered at the surface of a cell or intracellulary using antibody targeting compounds ofthe invention where it can then be activated.
  • site-specific drug delivery can be obtained from tissue-specific activation of a prodrug, which is the result of metabolism by an enzyme that is either unique for the tissue or present at a higher concentration (compared with other tissues); thus, it activates the prodrug more efficiently.
  • Photodynamic treatment may be used to activate a prodrug by cleaving a photosenitive linker or by activating a photoresponsive enzyme (acyl enzyme hydrolysis) as described previously (see U.S. Patent No. 5,114,851 and 5,218,137). Photodynamic treatment also may be used to rapidly inactivate a drug in sites where the drug activity is not desired (e.g. in non-target tissues).
  • a photoresponsive enzyme acyl enzyme hydrolysis
  • the functional component is a therapeutic agent such as drug. Any suitable drug can be used. Selection ofthe therapeutic agent depends upon the desired activity and target ofthe present compound. Where the target is an integrin, a preferred therapeutic is an agent having biological activity directed against the integrin. For example, in the case of Kaposi's Sarcoma, a cancer associated with angiogenesis of cancerous lesions, one can chose any of various drugs such as, for example, the three drugs paclitaxel, doxorubicin, and etoposide with demonstrated therapeutic efficacy in this disease.
  • a derivative of doxorubicin, 2-pyrrolinodoxorubicin is 500-1000 times more potent than doxorubicin itself and it has been extensively studied in other drug targeting strategies (for a recent review see Schally and Nagy, Eur. J. Endocrinology 141, 1-14, 1999).
  • a compound comprising an integrin targeting component with any of these compounds can be used to treat the abnormal angiogenesis of Kaposi's Sarcoma. Synthesis of these compounds with the integrin targeting component SCS-873 is described in the Examples.
  • integrin targeting compounds ofthe present invention have many uses.
  • a functional component can be delivered to integrin associated with cells, tissue or fluid macromolecule of an individual by administering the targeting compound.
  • the functional component is a therapeutic agent.
  • the integrin targeting invention compounds have particular utility for treating a pathological condition associated with integrin expression. Accordingly, a method is provided for treating or preventing a disease or condition in an individual wherein said disease or condition involves an integrin, the method comprising administering to the individual a therapeutically effective amount of an invention targeting compound comprising a therapeutic component effective against the disease or condition.
  • the condition is a carcinoma.
  • the association of integrin expression in carcinomas is well known in the art (See, e.g., U.S. Pat. Nos. 5,753,230 and 5,766,591).
  • the integrin targeting compounds ofthe present invention may be used for treating any disease or condition that is associated with the integrin being targeted.
  • the vitronectin receptor on osteoclasts is known to inhibits osteoclastic bone reso ⁇ tion.
  • diseases or conditions in which bone reso ⁇ tion is associated with a pathology such as osteoporosis and osteoarthritis
  • a vitronectin targeting compound ofthe invention can be treated by administering a vitronectin targeting compound ofthe invention.
  • a vitronectin targeting compound with an appropriate functional component can be used to stimulate bone formation by increasing osteocalcin release by osteoclasts.
  • Increased bone production is a clear benefit in disease states wherein there is a deficiency of mineralized bone mass or remodeling of bone is desired, such as fracture healing and the prevention of bone fractures. Diseases and metabolic disorders which result in loss of bone structure would also benefit from such treatment.
  • the integrin targeting compounds ofthe present invention also may be used for treating any inflammatory disorders, such as rheumatoid arthritis and psoriasis, and cardiovascular diseases, such as atherosclerosis and restenosis, which involve vitronectin expressing cells.
  • vitronectin targeting compounds ofthe invention which comprise an appropriate functional component can be used to treat these disorders.
  • This approach also applies to the treatment or prevention of other diseases including, but not limited to, thromboembolic disorders, asthma, allergies, adult respiratory distress syndrome, graft versus host disease, organ transplant rejection, septic shock, eczema, contact dermatitis, inflammatory bowel disease, and other autoimmune diseases.
  • the compounds ofthe present invention may also be useful for wound healing.
  • the integrin targeting compounds ofthe present invention further find use in treating angiogenic disorders.
  • Such disorders involve abnormal neovascularization where growth of new blood vessels is the cause of, or contributes to, the pathology associated with a disease. In these situations, inhibition of angiogenesis will reduce the deleterious effects of the disease.
  • Other therapeutic targets for the compounds ofthe instant invention are eye diseases characterized by neovascularization.
  • eye diseases include corneal neovascular disorders, such as corneal transplantation, he ⁇ etic keratitis, luetic keratitis, pterygium and neovascular pannus associated with contact lens use.
  • Additional eye diseases include age- related macular degeneration, presumed ocular histoplasmosis, retinopathy of prematurity, neovascular glaucoma, and the like.
  • the integrin targeting compounds ofthe present invention inhibit tumor tissue angiogenesis, thereby preventing tumor metastasis and tumor growth.
  • integrin targeting compounds ofthe invention also may be used for imaging of cells or tissues such as tumor cells as is well known in the art. Accordingly, provided is a method of imaging cells or tissue in an individual wherein said cells or tissue expresses an integrin target molecule, said method comprising administering to the individual an effective amount ofthe integrin targeting compound linked to a suitable radioisotope or detectable label.
  • the radioisotope or label may be attached to the targeting component or the functional component.
  • An integrin targeting compound ofthe present invention can be administered as a pharmaceutical composition wherein the invention compound is formulated with a pharmaceutically acceptable carrier. Accordingly, the invention compounds may be used in the manufacture of a medicament.
  • Pharmaceutical compositions ofthe invention compounds may be formulated as solutions or lyophilized powders for parenteral administration. Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use. Powders also may be sprayed in dry form.
  • the liquid formulation may be a buffered, isotonic, aqueous solution. Examples of suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or buffered sodium or ammonium acetate solution.
  • Such formulation is especially suitable for parenteral administration, but may also be used for oral administration or contained in a metered dose inhaler or nebulizer for insufflation. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride or sodium citrate.
  • excipients such as polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride or sodium citrate.
  • integrin targeting compounds may be encapsulated, tableted or prepared in a emulsion or syrup for oral administration.
  • Pharmaceutically acceptable solid or liquid carriers may be added to enhance or stabilize the composition, or to facilitate preparation ofthe composition.
  • Solid carriers include starch, lactose, calcium sulfate dihydrate, terra alba, magnesium stearate or stearic acid, talc, pectin, acacia, agar or gelatin.
  • Liquid carriers include syrup, peanut oil, olive oil, saline and water.
  • the carrier may also include a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax.
  • the amount of solid carrier varies but, preferably, will be between about 20 mg to about 1 g per dosage unit.
  • the pharmaceutical preparations are made following the conventional techniques of pharmacy involving milling, mixing, granulating, and compressing, when necessary, for tablet forms; or milling, mixing and filling for hard gelatin capsule forms.
  • a liquid carrier When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion or an aqueous or non-aqueous suspension.
  • the invention compounds may be combined with excipients such as cocoa butter, glycerin, gelatin or polyethylene glycols and molded into a suppository.
  • Integrin targeting compounds ofthe invention may be formulated to include other medically useful drugs or biological agents.
  • the compounds also may be administered in conjunction with the administration of other drugs or biological agents useful for the disease or condition that the invention compounds are directed (see e.g., U.S. Pat. No. 6,413,955 for active ingredients useful for osteoporosis).
  • an effective amount refers to a dose sufficient to provide concentrations high enough to impart a beneficial effect on the recipient thereof.
  • the specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated, the severity ofthe disorder, the activity ofthe specific compound, the route of administration, the rate of clearance ofthe compound, the duration of treatment, the drugs used in combination or coincident with the compound, the age, body weight, sex, diet and general health ofthe subject, and like factors well known in the medical arts and sciences.
  • a compound can be administered parenterally, such as intravascularly, intravenously, intraarterially, intramuscularly, subcutaneously, or the like. Administration can also be orally, nasally, rectally, transdermally or inhalationally via an aerosol.
  • the composition may be administered as a bolus, or slowly infused.
  • the administration ofthe targeting compound to an immunocompetent individual may result in the production of antibodies against the compound.
  • Such antibodies may be directed to the targeting component, the functional component or any other entity associated with the compound.
  • the immunogenicity of such compound may be addressed by methods well known in the art such as by attaching long chain polyethylene glycol (PEG)-based spacers, and the like, to one or more components ofthe 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 (Katre et al., 1990, J. Immunol. 144, 209-213; Francis et al., 1998, Int. J. Hematol. 68, 1-18).
  • PEG can serve as a linker in targeting compounds ofthe invention, thus providing both linker function and reduced immunogenicity.
  • the individual may be administered an immunosuppressent drug such as cyclosporin A, anti-CD3 antibody, and the like, to reduce the likelihood that an immune response to the targeting compound will develop.
  • RGD and non-RGD peptidomimetic or non-RGD peptide antagonists or agonists can be used to target liposomes. This is achieved by chemically linking such targeting components to a lipid moiety that enables the targeting component to associate with lipids ofthe liposome.
  • the liposome encapsulated with an appropriate drug can then be targeted more effectively in vivo by aid ofthe targeting component.
  • treatment of Kaposi's sarcoma with sterically stabilized stealth liposomes containing doxorubicin (DoxilTM) is one ofthe most effective ofthe approved KS therapies. This liposomal formulation provides several advantages over administration of free doxorubicin.
  • DoxilTM In addition to a reduction in cardiac toxicity, vomiting, alopecia, peripheral neuropathy, and mucositis provided by the liposomal formulation, DoxilTM possesses an intrinsic capacity for passive targeting to the tumor as a result of particulate size and the enhanced permeability and retention phenomenon (Matsumura et al., Cancer Res. 46, 6387- 6392, 1986) that is further aided by the extended serum half-life ofthe drug.
  • Active targeting as disclosed herein can augment the passive targeting capacity of DoxilTM thereby more efficiently delivering the cytotoxic payload ofthe liposomes to the KS tumor and its vasculature. Such active targeting of DoxilTM likely will reduce the dosage needed for clinical response thereby limiting non-specific toxicities.
  • Administration of targeted liposomes may be achieved as previously described (See, e.g., Allen et al., Adv. Drug Deliv. Rev. 21, 117-133).
  • the targeting compounds ofthe invention may be used in combination with other compounds or therapies such as radiation therapy.
  • EXAMPLE 1 Antibody targeting compound comprising an RGD peptidomimetic targeting agent covalently linked to the combining site of aldolase monoclonal antibody 38C2.
  • An integrin targeting compound was formed based on the formation of a reversible covalent bond between a diketone linker derivative of an RGD peptidomimetic and the reactive lysine of mouse mAb 38C2.
  • Mouse mAb 38C2 is the prototype for a new class of catalytic antibodies generated by reactive immunization and mechanistically mimic natural aldolase enzymes (Barbas et al., Science 278, 2085-2092, 1997).
  • aldolase antibodies Through a reactive lysine, these antibodies catalyze aldol and retro-aldol reactions using the enamine mechanism of natural aldolases (Wagner et al., Science 270, 1797-1800, 1995; Barbas et al., Science 278, 2085-2092, 1997; Zhong et al., Angew. Chem. Int. Ed. 38, 3738-3741, 1999).
  • aldolase antibodies In addition to their versatility and efficacy in synthetic organic chemistry, aldolase antibodies have been used in the activation of camptothecin, doxorubicin, and etoposide prodrugs in vitro and in vivo as an anti-cancer strategy (Shabat et al., Proc. Natl.
  • RGD peptidomimetic used (see Compound 1) is specific for human integrin with a high binding affinity for ⁇ v ⁇ at 0.9 nM and ⁇ v ⁇ 5 at 0.6 nM (specificity exhibited by minimal a ⁇ n,b 3 binding) (Miller et al., supra).
  • a diketone linker modified version of Compound 1, designated SCS-873, was prepared as described in Example 3. SCS-873 is shown below with the targeting component and the linker separately identified.
  • a peptidomimetic RGD antagonist with known activity for both ⁇ v ⁇ 3 or ⁇ v ⁇ 5 binding is desirable because some of these compounds bind both murine and human integrins. Such species cross reactivity affords preclinical in vivo studies in animal angiogenesis models prior to human trials.
  • the targeting compound may be used for the therapy of Kaposi's sarcoma which is associated with ⁇ v ⁇ 3 integrin.
  • SCS-873 was linked to antibody 38C2 by the following procedure: One milliliter antibody 38C2 in phosphate buffered saline (lOmg/ml) was added to 12 microliters of a 10 mg/mL stock solution of SCS-873 and the resulting mixture was maintained at room temperature for 2 hours prior to use. [0099] The binding of a mixture of SCS-873 and 38C2 to SLK cells was evaluated. SCS- 873 effectively mediated cell surface binding of 38C2. No binding of 38C2 was detectable in the absence of SCS-873. Control experiments confirmed that the diketone moiety ofthe linker is required for binding of SCS-873 to 38C2.
  • SCS-873 retains the integrin specificity ofthe integrin targeting component, i.e., no binding to am,b 3 in ELISA was detected while binding to ⁇ v ⁇ 3 and ⁇ v ⁇ was found to be strong.
  • independent i.p. and i.v. injections ofthe targeting compound prepared with SCS-873 and 38C2 versus each component alone into mice demonstrated integrin targeting in vivo.
  • the serum half-life of SCS-873 was extended by more than two orders of magnitude through binding to 38C2. Free SCS-873 not bound to antibody had a serum half-life of only minutes while the combination of antibody and small molecule could be detected in the serum sampled from eye bleeds after several days.
  • EXAMPLE 2 Integrin Targeting with Protein Functional Components.
  • the integrin targeting component can be covalently attached to a functional component such as a protein in order to channel effector functions triggered by these proteins.
  • a functional component such as a protein
  • Such linking can be achieved by a lysine-reactive N-hydroxysuccinimide or a cysteine-reactive maleimide functionality.
  • conjugation ofthe integrin targeting compound to an IgM antibody induces complement-mediated cytotoxicity; conjugation to IL-2 results in cell-mediated cytotoxicity.
  • a variety of antibodies that neutralize growth factors involved in tumor angiogenesis can be modified with the integrin targeting compound in order to increase their selectivity.
  • a monoclonal antibody that neutralizes VEGF is highly selective if conjugated to the integrin targeting compound. Taking the potential immunogenicity of these conjugates into consideration, long chain polyethylene glycol (PEG)-based spacers can be introduced between the protein-reactive and the integrin targeting functionality.
  • PEG polyethylene glycol
  • Integrin targeting components shown as compounds 15 and 4 were synthesized as shown in the FIG. 10 (Scheme 1) and FIG. 11 (Scheme 2), respectively.
  • a linker with a diketone reactive moiety was added to these targeting molecules as shown in Scheme 3 (FIG. 12) to form targeting compound-linker molecules SCS-873 and SCS-1655.
  • Synthesis of SCS-873 was achieved starting from compound 14 in three steps.
  • Compound 14 was converted to 15 as shown in Scheme 1 and the crude product was reacted with an N- hydroxysuccinimide (NHS) -ester ofthe diketone compound 23 in CH 3 CN-DMF in the presence of Et 3 N.
  • Purification over silica gel CH 2 Cl 2 -MeOH, 9:1) afforded pure SCS-873.
  • Compound SCS-1655 was synthesized from 14 in five steps (Schemes 2 and 3). Deprotection ofthe BOC group in compound 14 followed by reaction with the NHS ester of the bivalent linker 24 afforded compound 25, which was then deprotected and reacted with 23 as above to afford SCS-1655.
  • Synthesis of integrin targeting component-linker molecules SCS-864 and SCS- 789 is shown in Scheme 4 (FIG. 13). SCS-864 and SCS-789 were each synthesized in one step from compound 4 (FIG. 13, scheme 4). Linking of Compound 4 was achieved with the appropriate activated NHS-ester. SCS-864 is shown below with the targeting component and linker separately identified.
  • Paclitaxel-SCS-873 is synthesized starting from taxol-succinate prepared as previously described (Deutsch et al., J. Med. Chem. 32, 788-792, 1989). Following activation ofthe carboxy group with PyBOP in DMF, the SCS-amine is directly coupled (Huang et al., Chemistry & Biology 7, 453-461, 2000) providing Paclitaxel-SCS-873. This derivative is analogous to a previously described somatostatin antagonist peptide targeted paclitaxel derivative (Huang et al., Chemistry & Biology 7, 453-461, 2000) that demonstrated good activity and targeting, thereby validating the succinate-based linker strategy for this drug.
  • Paclitaxel-SCS-873 is considerably more soluble than paclitaxel itself which suffers from poor solubility.
  • the structure of Paclitaxel-SCS-873 is shown below.
  • Doxorubicin-SCS-873 is prepared using a synthetic scheme similar to that described for doxorubicin derivatives of luteinizing hormone-releasing hormone and somatostatin conjugates (Nagy et al., Proc. Natl. Acad. Sci. U.S.A. 93, 7269-7273, 1996; Nagy et al., Proc. Natl. Acad. Sci. U.S.A. 95, 1794-1799, 1998).
  • N-Fmoc-DOX-14-O- hemiglutarate are prepared as described by Nagy et al. (Proc Natl. Acad. Sci.
  • Doxorubicin-SCS-873 The structure of Doxorubicin-SCS-873 is shown below.
  • 2-Pyrrolinodoxorubicin-SCS-873 can be prepared from Doxorubicin-SCS-873 by reaction with 4-iodobutyraldehyde as described for LH-RH-2 -Pyrrolinodoxorubicin conjugates (Nagy et al., Proc Natl. Acad. Sci. 93, 2464-2469, 1996). A variety of other synthetic approaches are also available. The proven activity of LH-RH-2- pyrrolinodoxorubicin and doxorubicin conjugates of similar design supports the design of our integrin targeted derivatives (See, e.g., Schally and Nagy, Eur. J. Endocrinology 141, 1-14, 1999 and references therein for a review peptide targeted drug conjugates). The structure of 2-Pyrrolinodoxorubicin-SCS-873 is shown below.
  • Etoposide-SCS-873 is be prepared from the p-nitrophenylcarbonate recently described for structurally similar etoposide prodrugs that are activated by catalytic antibody (Shabat et al, Proc. Natl. Acad. Sci. U.S.A. 98, 7528-7533, 2001).
  • the retro-Michael step was readily catalyzed by endogenous cellular enzymes as well as by the catalytic antibody (Shabat et al., Proc. Natl. Acad. Sci. U.S.A. 96, 6925-6930, 1999).
  • Etoposide- SCS-873 is based on endogenous enzyme or general acid base activation via a retro-Michael reaction that is followed by spontaneous decarboxylation and lactamization reactions that provide active etoposide.
  • the structure of Etoposide-SCS-873 is shown below.
  • DoxilTM containing liposomes are associated with a targeting compound ofthe invention as described.
  • the targeting component is attached to maleimide-terminated PEG2000-DSPE (MAL-PEG2000-DSPE).
  • MAL-PEG2000-DSPE is commercially available through Shearwater Polymers, Inc. Attachment of thiol terminated targeting molecules to the maleimide moiety is spontaneous.
  • the PEG-lipid derivative is then transferred into the DoxilTM liposome with negligible drug leakage in a simple incubation step.
  • Targeted binding of liposomes to cells is studied by FACS analysis by co- inco ⁇ oration of biotin labeled MAL-PEG2000-DSPE and staining with FITC-labeled streptavidin.
  • Various control cell lines are also studied to assess non-specific binding. Cytotoxicity assays are performed as described (Moase et al., 2001) with a variety of different loadings ofthe targeting molecules into the liposome using the three KS cell lines.
  • animals are treated 1 day after cell implantation and after tumors have established (200 mm 3 ).
  • a single dosing regimen is studied initially to assess the relative efficacy of targeted DoxilTM vs.
  • DoxilTM untargeted DoxilTM. Drug dosing ranges from 0.5 mg/kg to 5 mg/kg i.v.. Other control groups are be treated with empty, but targeted liposomes to assess the effect ofthe multivalent liposome itself on the disease. A buffer control group is also included. In multiple treatment studies, drug is injected at 14-day intervals. Systemic toxicity is assessed as described above. [00111 ] It should be noted that DoxilTM is also approved for the treatment of refractory ovarian cancers. A recent study of 25 permanent human cell lines established from advanced ovarian cancer demonstrated that all lines were positive for integrin expression (Bruning et al., Hum. Gene Ther. 12, 391-399, 2001).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nanotechnology (AREA)
  • Immunology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Genetics & Genomics (AREA)
  • Rheumatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medical Informatics (AREA)
  • Biophysics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention is directed to integrin targeting compounds comprising an integrin targeting component linked to a functio nal component such as a therapeutic agent or antibody. Structures of various integrin targeting compounds are provided. Additionally provided are methods of delivering a functional component to integrin associated with cells or tissue of an individual using the integrin targeting compounds. Also provided are methods of treating or preventing a disease or condition in an individual using the wherein said disease or condition involves an integrin using the integrin targeting compounds.

Description

INTEGRIN TARGETING COMPOUNDS
BACKGROUND OF THE INVENTION
[0001] The invention relates to integrin targeting compounds and methods of making and using the compounds. The use of potent chemotherapeutics has some distinct limitations mainly related to the toxicity ofthe drug. The dose of drug that is required for therapy often renders patients susceptible to potentially fatal infections, cardiac toxicity and other side effects. Improved understanding ofthe biology of cancer has led to more specific "targeted therapies." Antigens expressed on the surface of diseased cells (e.g., tumor cells) form the basis of "monoclonal antibody directed drug delivery" approaches. Unfortunately, however, tumor antigens are often down regulated due to drug resistance: this limits the effectiveness ofthe "monoclonal antibody directed drug delivery" approaches. On the other hand, target molecules expressed on the surface of tumor endothelial cells are readily accessible to targeting molecules circulating in the blood. Furthermore, in contrast to tumor cells, tumor endothelial cells are genetically stable and are unlikely to be down regulated due to drug resistance. Hence, molecules expressed on the surface of tumor endothelial cells appear to be an appropriate receptor for "directed drug delivery."
[0002] The present invention solves problems ofthe art by providing novel compounds for targeting integrin expressing cells. Such compounds have diagnostic and therapeutic applications in cancer and other diseases.
BRIEF SUMMARY OF THE INVENTION
[0003] The present invention is directed to targeting compounds comprising at least one integrin targeting component covalently linked via a linker to at least one functional component, wherein said integrin targeting component is selected from a group that includes:
(a) a RGD peptidomimetic, and
(b) a non-RGD peptide, peptidomimetic, or organic molecule integrin agonist or antagonist. [0004] In some embodiments, the integrin targeting component is specific for an integrin such as αvβ or αvβ5. The core structures of preferred RGD peptidomimetics are disclosed for use in particular targeting compound embodiments.
[0005] The integrin targeting compounds ofthe invention confer various benefits over the components themselves. For example, the functional component may generally extend the half-life of a smaller sized targeting component in vivo. Also, the biological potency or other biological feature of an integrin targeting component may be modified by the addition of effector function(s) provided by a functional component such as an antibody. In addition, the integrin targeting component, through its increased size conferred by linkage to the functional component, may enable the targeting agent to function in new capacities.
[0006] Also provided are methods of producing the integrin targeting compounds ofthe invention. In one embodiment, an integrin targeting component-linker compound is prepared which includes an integrin targeting component and a linker that includes a reactive group for covalent reaction with a susceptible reactive moiety ofthe functional component. In another approach, a functional component-linker compound is prepared that includes a functional component and a linker that includes a reactive group for covalent reaction with a susceptible reactive moiety of an integrin targeting component. In yet another approach, the targeting component and the functional component are linked to one of a linker with a reactive groups or linker with a susceptible moiety so that the targeting compound forms when the two linkers covalently bond.
[0007] Further provided are integrin targeting component-linker compounds and functional component-linker compounds for covalently linking an integrin targeting component to a functional component. In some embodiments, the linker includes a reactive group for covalently linking to the other ofthe components. In some embodiments, the linker reactive group is a ketone, a diketone, a beta lactam, a succinimide active ester, haloketone, a lactone, an anhydride, an epoxide, an aldehyde or a maleimide.
[0008] Various chemical features ofthe integrin targeting component- linker compounds and functional component linker compounds are described. In one embodiment, the linker has the general formula X - Y - Z wherein X is a linear or branched connecting chain of atoms comprising any of C, H, N, O, P, S, Si, F, CI, Br, and I, or a salt thereof, and comprising a repeating ether unit of between 2-100 units; Y is optional and is a single or fused 5 or 6 membered homo- or heterocarbocylic saturated or unsaturated ring located within 1-20 atoms of Z; and Z is a reactive group for covalently linking the one or more targeting agents to susceptible moiety such as a side chain of a reactive amino acid. The targeting component or functional component may be linked to X or Y or to X and Y when multiple components are included in the targeting compound.
[0009] Additionally provided are methods of delivering a functional component to integrin associated cells, tissue of an individual. The method includes administering to the individual an integrin targeting compound ofthe invention. In some embodiments ofthe method, the functional component is a therapeutic agent which includes the functional component.
[0010] Still further provided are methods of treating or preventing a disease or condition that involves integrin in an individual. The methods includes administering to the individual a therapeutically effective amount of an integrin targeting compound ofthe invention that includes a functional component that is a therapeutic agent. The disease or condition is susceptible to the therapeutic agent such that a reduction or prevention ofthe symptoms associated with the disease or condition is effected. In some embodiments ofthe method, the disease or condition involves a defect in angiogenesis, bone metabolism, inflammation or cell growth.
[0011] The invention further provides pharmaceutical compositions or medicaments that include an integrin targeting compound ofthe invention and a pharmaceutically acceptable carrier.
BRIEF DESCRIPTION OF THE DRAWINGS
[0012] FIG. 1 shows exemplary integrin targeting agents of which Panels A-E are RGD peptidomimetic while Panel F is an RGD peptide. The core structures are from the following: U.S. Patent No. 6,335,330 (Panel A), U.S. Patent No. 5,693,636 (Panel B), U.S. Patent No. 6,040,311 (Panel C), and U.S. Patent No. 6,001,117 (Panel E). [0013] FIG. 2 is a general linker design (Panel A) and specific embodiment (Panel B; SCS-873) shown in association with a targeting agent.
[0014] FIG. 3 shows a general scheme of an embodiment of a targeting agent-linker compound with a branched linker and two identical targeting agents (Panel A) with specific embodiments in Panel B (integrin targeting agent diketo linker; compound 26), and Panel C (integrin targeting agent diketo linker; compound 27). The branch point is in the connecting chain portion ofthe linker.
[0015] FIG. 4 shows a general scheme of an embodiment of a targeting agent-linker compound with a branched linker and two different targeting agents (Panel A) with a specific embodiment in Panel B (integrin targeting and folate targeting agent diketo linker; compound 28). The branch point is in the connecting chain portion ofthe linker. TI is an integrin targeting component while T2 is a folate receptor targeting component.
[0016] FIG. 5 shows a general scheme of an embodiment of a targeting agent-linker compound with a branched linker and two different targeting agents (Panel A) with a specific embodiment in Panel B (integrin targeting agent diketo linker; compound 29). The branch point is in the recognition group portion ofthe linker.
[0017] FIG. 6 shows the structure of linker reactive groups. Structures A-C form reversible covalent bonds with reactive nucleophilic group (e.g. lysine or cysteine side chain) in the combining site of an antibody (structure A could form an irreversible covalent bond X is N and if R\ and R form part of a cyclic structure). Ri and R2 and R3 in structures A-C represent substituents which can be C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof. X is N, C, Si, or any other heteroatom. These 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. R2 and R3 could be cyclic as exemplified in structures B and C while X could be a heteroatom. Structures D-G form nonreversible covalent bonds with reactive nucleophilic group (e.g. lysine or cysteine side chain) in the combining site of an antibody. In these structures, R\ and R2 represent C, O N, halide and leaving groups such as mesyl or tosyl. [0018] FIG. 7 shows various electrophiles suitable for reactive modification with a reactive amino acid side chain of an antibody. Key: (A) acyl beta-lactam; (B) simple diketone; (C) succinimide active ester; (D) maleimide; (E) haloacetamide with linker; (F) haloketone; (G) cyclohexyl diketone; and (H) aldehyde. R refers to other structure that may include a targeting agent, linker or antibody, while X refers to halogen.
[0019] FIG. 8 shows the structure of linker recognition group (Y), situated between the reactive group portion and the connecting chain portion ofthe linker. Panel A shows the relationship ofthe recognition group Y within the linker (see FIG. 2). Panels B-D show distance of Y from Z, substituents on the ring and ring member atoms.
[0020] FIG. 9 shows the structure ofthe linker connecting chain (X), which directly attaches at one end to the targeting agent as shown in Panel A (see FIG. 2). Substituents R2 to R4 can be C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof, and may include a group such as an alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl as well as a carbocyclic or heterocyclic mono or fused saturated or unsaturated ring structure. In the connecting chain in structures B and C, n, r or m is 1-100. In structures D and E, n is 1, 2, 4, or more preferably is 3.
[0021] FIG. 10 shows Scheme 1, a synthetic scheme for the amine precursor of SCS-873, targeting agent 3 or SCS-amine. Key: (a) BBr3, CH2C12, -20 °C, 2h; (b) DMF, rt to 80°C, 3h; (c) BnCOCl, sat. aq. NaHCO3, ether; (d) TBDPSiCl, imidazole, DMF, 16h; (e) Pd(OAc)2, (o-tol)3P, /-Pr2EtN, CH3CH2CN, reflux, 3h; (f) 20 % (w/w) Pd-C (10%), H2, EtOH-AcOH (1 :1), 36h; (g) TBAF, THF, rt, lh; (h) DEAD, PPh3, THF-benzene (3: 1), 16h; (i) 20 % (w/w) Pd-C (10%), cyclohexene-t-PrOH (1:1), 90°C, 12h; ( ) i. aq. 2N NaOH, MeOH-THF (1 : 1), 16h, ii. TFAA, anisole, CH2C12, 0°C, 2h.
[0022] FIG. 11 shows Scheme 2, a synthetic scheme for making Compound 4, (R = Butoxycarboxyaminohexanoyl-derivative). Key: (a) DMF, rt; (b) EDC, HOBT, DMF; (c) 0.01 M in DMSO, 130 °C; (d) TFAA, anisole, dichloromethane; (e) DMF; (f) EDC, HOBT, DMF; (g) (i) step d, (ii) 2M NaOH, MeOH-THF (1 :1). [0023] FIG. 12 shows Scheme 3, a synthetic scheme for making compounds SCS-873 and SCS-1655.
[0024] FIG. 13 shows Scheme 4, a synthetic scheme for making Compounds SCS-864 and SCS-789. Key: (a) Et3N, DMF, rt, 16h.
[0025] FIG. 14 shows an embodiment whereby two targeting components are linked to a single linker. TI is an integrin targeting component and T2 is biotin (compound 30).
DETAILED DESCRIPTION OF THE INVENTION
[0026] The present invention provides integrin targeting compounds useful for presentation to a particular integrin target such as a cell or tissue. The targeting component operates to situate the compound at the target site. Integrin targeting compounds comprise at least two components: a targeting component and a functional component. The components are operatively linked such that each component retains its activity. Typically, the targeting component specifically binds to the integrin target typically through a ligand/receptor relationship. Such ligand/receptor relationships are well known in the art. Preferably the integrin target is on the surface ofthe target cell or tissue. The integrin target may also be associated with a particular condition such a pathological condition.
[0027] A preferred targeting component targets to one or more integrins. The targeting component can be either an agonist or antagonist to the integrin or bind without any biological activity. In one embodiment, the integrin is αvβ or αvβ5. Integrins are heterodimeric transmembrane glycoprotein complexes that function in cellular adhesion events and signal transduction processes. Integrin αvβ3 is expressed on numerous cells and has been shown to mediate several biologically relevant processes, including adhesion of osteoclasts to the bone matrix, migration of vascular smooth muscle cells, and angiogenesis. Integrin αvβ3 antagonists may be employed in the treatment of several human diseases, including diseases involving neovascularization, such as rheumatoid arthritis, cancer, ocular diseases, and the like.
[0028] The integrin targeting component of integrin targeting compounds ofthe invention may be a small molecule organic compound of about 5,000 daltons or less such as a drug or pharmaceutical, which is an integrin antagonist or agonist. An integrin agonist or antagonist also can be a protein, peptide, peptidomimetic, glycoprotein, proteoglycan, lipid, phospholipid, lipopolysaccharide, glycolipid, nucleic acid, proteoglycan, carbohydrate, and the like. The terms "polypeptide", "peptide," and "protein" are used interchangeably to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is a synthetic chemical analogue (e.g., para-methyl-tyrosine, para- chloro-phenylanine, and the like) of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers. Amino acids can be in the L or D form so long as the binding function ofthe peptide is maintained. Peptides can be of variable length, generally between about 4 and 200 amino acids. 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.
[0029] The integrin targeting component is not an antibody, although the targeting compound may include an antibody as the functional component and such antibody may have a targeting capability. If such antibody has an integrin targeting capability, the targeting compound will include a non-antibody component that is integrin specific.
[0030] As used herein, reference to "Arg-Gly-Asp peptide" or "RGD peptide" is intended to refer to a peptide having one or more Arg-Gly-Asp containing sequences which may function as a binding site for a receptor ofthe "Arg-Gly-Asp family of receptors", e.g., an integrin. Integrins, which comprise and alpha and a beta subunit, include numerous types including αιβι, α2βι, α3βι, α4β1; α5βι, α6βι, α7βι, α8βι, α9β αιβ1; α6β α4β7, αDβ2, αDβ2. αLβ2, otMβ2,ocvβι ,αvβ3, ocvβ5> ocvβ6, αvβ8, αxβ2, ocnbβ3, !ELbβ7, and the like. The sequence RGD is present in several matrix proteins and is the target for cell binding to matrix by integrins. Platelets contain a large amount of RGD-cell surface receptors ofthe protein GP IIb/πia, which is primarily responsible, through interaction with other platelets and with the endothelial surface of injured blood vessels, for the development of coronary artery thrombosis. The term RGD peptide also includes peptides with amino acids that are functional equivalents (e.g., RLD or KGD) of RGD peptide provided that they interact with the same RGD receptor. Peptides containing RGD sequences can be synthesized from amino acids by means well known in the art, using, for example, an automated peptide synthesizer, such as those manufactured by Applied Biosystems, Inc., Foster City, California.
[0031] In other embodiments, the integrin targeting component ofthe targeting compounds ofthe invention can be a non-RGD peptide agonist or antagonist or peptidomimetic thereof. As used herein, "non-RGD peptide" refers to a peptide that is an antagonist or agonist of integrin binding to its ligand (e.g. fibronectin, vitronectin, laminin, collagen etc.), but does not involve an RGD binding site. Such non-RGD integrin targeting compounds are known for vβ (see, e.g., U.S. Pat. Nos. 5,767,071 and 5,780,426) as well as for other integrins such as α4βι (VLA-4), α4β7 (see, e.g., U.S. Pat. Nos 6,365,619; Chang et al., Bioorganic & Medicinal Chem Lett, 12:159-163 (2002); Lin et al., Bioorganic & Medicinal Chem Lett, 12:133-136 (2002)), and the like.
[0032] In one embodiment, the integrin targeting component is a peptidomimetic agonist or antagonist, which preferably is a peptidomimetic agonist or antagonist of an RGD peptide or non-RGD peptide. As used herein the term "peptidomimetic" is a compound containing non-peptidic structural elements that are capable of mimicking or antagonizing the biological action(s) of a natural parent peptide. A peptidomimetic of an RGD peptide is an organic molecule that retains similar peptide chain pharmacophore groups ofthe RGD amino acid sequence but lacks amino acids or peptide bonds in the binding site sequence. Likewise, a peptidomimetic of a non-RGD peptide is an organic molecule that retains similar peptide chain pharmacophore groups ofthe non-RGD binding site sequence but lacks amino acids or peptide bonds in the binding site sequence. A "pharmacophore" is a particular three- dimensional arrangement of functional groups that are required for a compound to produce a particular response or have a desired activity. The term "RGD peptidomimetic" is intended to refer to a compound that comprises a molecule containing the RGD pharmacophores supported by an organic/non-peptide structure. It will be understood that an RGD peptidomimetic (or non-RGD peptidomimetic) may be part of a larger molecule that itself includes conventional or modified amino acids linked by peptide bonds.
[0033] RGD peptidomimetics are well known in the art, and have been described with respect to integrins such as GPIIb/IIIa, αvβ3 and αvβ5 (See, e.g., Miller et al., J. Med. Chem. 2000, 43:22-26; and International Patent Publications WO 0110867, WO 9915178, WO 9915170, WO 9815278, WO 9814192, WO 0035887, WO 9906049, WO 9724119 and WO 9600730; see also Kumar et al., Cancer Res. 61:2232-2238 (2000)). Many such compounds are specific for more than one integrin. RGD peptidomimetics are generally based on a core or template (also referred to as "fibrinogen receptor antagonist template"), to which are linked by way of spacers to an acidic group at one end and a basic group at the other end ofthe core. The acidic group is generally a carboxylic acid functionality while the basic group is generally a N-containing moiety such as an amidine or guanidine. Typically, the core structure adds a form of rigid spacing between the acidic moiety and the basic nitrogen moiety, and contains one or more ring structures (e.g., pyridine, indazole, etc.) or amide bonds for this purpose. For a fibrinogen receptor antagonist, generally, about twelve to fifteen, more preferably thirteen or fourteen, intervening covalent bonds are present (via the shortest intramolecular path) between the acidic group ofthe RGD peptidomimetic and a nitrogen ofthe basic group. The number of intervening covalent bonds between the acidic and basic moiety is generally shorter, two to five, preferably three or four, for a vitronectin receptor antagonist. The particular core may be chosen to obtain the proper spacing between the acidic moiety ofthe fibrinogen antagonist template and the nitrogen atom ofthe pyridine. Generally, a fibrinogen antagonist will have an intramolecular distance of about 16 angstroms (1.6 nm) between the acidic moiety (e.g., the atom which gives up the proton or accepts the electron pair) and the basic moiety (e.g., which accepts a proton or donates an electron pair), while a vitronectin antagonist will have about 14 angstroms (1.4 nm) between the respective acidic and basic centers. Further description for converting from a fibrinogen receptor mimetic to a vitronectin receptor mimetic can be found in U.S. Pat. No. 6,159,964.
[0034] The peptidomimetic RGD core can comprise a 5-11 membered aromatic or nonaromatic mono- or polycyclic ring system containing 0 to 6 double bonds, and containing 0 to 6 heteroatoms chosen from N, O and S. The ring system may be unsubstituted or may be substituted on a carbon or nitrogen atom. Preferred core structures with suitable substituents useful for vitronectin binding include monocyclic and bicyclic groups, such as benzazapine described in WO 98/14192, benzdiazapine described in U.S. 6,239,168, and fused tricyclics described in U.S. 6,008,213.
[0035] U.S. Pat. No. 6,159,964 contains an extensive list of references in Table 1 of that document which disclose RGD peptidomimetic cores structures (referred to as fibrinogen templates) which can be used for prepraring RGD peptidomimetics. Preferred vitronectin RGD and fibronectin RGD peptidomimetics are disclosed in U.S. Patent Nos. 6,335,330; 5,977,101; 6,088,213; 6,069,158; 6,191,304; 6,239,138; 6,159,964; 6,117,910; 6,117,866; 6,008,214; 6,127,359; 5,939,412; 5,693,636; 6,403,578; 6,387,895; 6,268,378; 6,218,387; 6,207,663; 6,011,045; 5,990,145; 6,399,620; 6,322,770; 6,017,925; 5,981,546; 5,952,341; 6,413,955; 6,340,679; 6313,119; 6,268,378; 6,211,184; 6,066,648; 5,843,906; 6,251,944; 5,952,381; 5,852,210; 5,811,441; 6,114,328; 5,849,736; 5,446,056; 5,756,441; 6,028,087; 6,037,343; 5,795,893; 5,726,192; 5,741,804; 5,470,849; 6,319,937; 6,172,256; 5,773,644; 6,028,223; 6,232, 308; 6,322,770; 5,760,028.
[0036] Exemplary RGD peptidomimetic integrin targeting agents are shown below as compounds 1, 2, and 3 can be used for preparing an intregrin targeting compound ofthe present invention. In the three compounds, the linker is attached as indicated to the nitrogen ofthe seven membered ring. Other RGD peptidomimetic integrin targeting agents include compound 31, wherein P and L or carbon or nitrogen. The linker may be Rl or R2 while the R3 group includes a basic group such as an -NH group. In some embodiments, the R3 group is as shown in structures 1, 2, or 3. In some embodiments, the R3 group includes a heterocyclic group such a benzimidazole, imidazole, pyridine group, or the like. In some such embodiments, the R3 group is a alkoxy group, such as a propoxy group or the like, that is substituted with a heterocarbyl group that is substituted with an alkylamine group, such as a methylamino group or the like, whereas in other embodiments, the R3 group is an alkoxy group, such as a propoxy group or the like, substituted with a heterocyclylamino group, such as with a pyridinylamino group or the like such as a 2-pyridinylamino group. In other embodiments R3 is a group of formula -C(=O)Rb where Rb is selected from -N(alkyl)-alkyl- heterocyclyl groups such as -N(Me)-CH2 -benzimidazole groups and the like.
Figure imgf000013_0001
Figure imgf000013_0002
Figure imgf000013_0003
Figure imgf000013_0004
31
[0037] Other exemplary integrin peptidomimetic targeting agents and a peptide targeting agent are shown in FIG. 1. The linker may be any of Ri, R2, R , while R4 may be a linker or a hydrolyzable group such as alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl group, and the like. One of skill in the art will readily appreciate that other integrin agonist and antagonist mimetics can also be used in targeting compounds ofthe present invention. [0038] A targeting compound ofthe invention can contain more than one targeting component. In such an embodiment, one ofthe targeting components targets an integrin as disclosed above. The second targeting component can target another entity. Exemplary such entities are chemokine receptors such as CCR5, interleukin or cytokine receptors, vitamin receptors such as the folate receptor, cancer markers such as prostate specific antigen, carcinoembryonic antigen, HER-2, viral markers such as HIV gp41, or a peptide hormone receptor. Linear or branched linkers may be used for preparing dual agent targeting compounds. Exemplary branched linker designs are shown in FIGs. 3-5.
[0039] An exemplary such dual targeting compound (Compound 32) is shown below, where the compound targets both integrin and the folate receptor. Another dual targeting compound that uses a single linear linker and targets integrin and avidin is shown in FIG. 14 (compound 30). Compound 30, which has a shorter linker than in other examples, has several uses. For example, compound 30 can be used in conjunction with a detectably labeled avidin or streptavidin to test cells for expression ofthe integrin target reactive with TI. In another embodiment, Compound 30 can be administered in vivo together with avidin or streptavidin labeled with a suitable therapeutic or imaging agent.
Figure imgf000014_0001
32
[0040] As described herein the targeting component is linked to a functional component in that it may have one or more biological activities, each activity characterized as a detectable biological affect on the functioning of a cell organ or organism. A targeting compound may, however, be a pure binding compound without biological activity. In the case where the targeting compound has functional activity, the compound would include a functional component that is distinct from the targeting component.
[0041] The functional component of a targeting compound can be any structure having a desired biological activity toward a cell or tissue associated with integrin. In some embodiments, the functional component is not a metal chelating structure with or without the associated metal ion, e.g., a radiometal ion. In some embodiments, the functional component is not a lipid. The functional component may include any of a number of biologically active structures well known in the art. Biological agents suitable as the functional component of the targeting compounds include, but are not limited to, small molecule drugs (a pharmaceutical organic compound of about 5,000 daltons or less), organic molecules, proteins, peptides, peptidomimetics, glycoproteins, proteoglycans, lipids, phospholipids, lipopolysaccharides, glycolipids, nucleic acids, proteoglycans, carbohydrates, and the like. In some embodiments, the biological agent functional component may be anti-neoplastic, antimicrobial, a hormone, an effector, and the like. Suitable functional components include well known therapeutic compounds such as anti-neoplastic agents include paclitaxel, daunorubicin, doxorubicin, carminomycin, 4'-epiadriamycin, 4-demethoxy-daunomycin, 11 - deoxydaunorubicin, 13-deoxydaunorubicin, adriamycin-14-benzoate, adriamycin-14- octanoate, adriamycin-14-naphthaleneacetate, vinblastine, vincristine, mitomycin C, N- methyl mitomycin C, bleomycin A2, dideazatetrahydrofolic acid, aminopterin, methotrexate, cholchicine and cisplatin, and the like. Suitable anti-microbial agent functional components include aminoglycosides including gentamicin, antiviral compounds such as rifampicin, 3'- azido-3'-deoxythymidine (AZT) and acylovir, antifungal agents such as azoles including fluconazole, plyre macrolides such as amphotericin B, and candicidin, anti-parasitic compounds such as antimonials, and the like. Suitable hormone functional components may include toxins such as diphtheria toxin, cytokine such as CSF, GSF, GMCSF, TNF, erythropoietin, immunomodulators or cytokines such as the interferons or interleukins, a neuropeptide, reproductive hormone such as HGH, FSH, or LH, thyroid hormone, neurotransmitters such as acetylcholine, hormone receptors such as the estrogen receptor. Suitable functional components also include non-steroidal anti-inflammatories such as indomethacin, salicylic acid acetate, ibuprofen, sulindac, piroxicam, and naproxen, and anesthetics or analgesics. In some embodiments, the functional component is not a radioisotope.
[0042] Functional components can be naturally occurring or synthetic and may be biologically active in their native state where they can act, for example, at the surface of a target cell or can be transported into the target cell to act intracellularly.
[0043] The functional component can also be an "antibody" which as used herein includes 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. Also subclasses ofthe heavy chain are known. For example, IgG heavy chains in humans can be any of IgGl, IgG2, IgG3 and IgG4 subclass.
[0044] 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 ) and variable heavy chain (VH) refer to these light and heavy chains respectively.
[0045] Antibodies exist as full length intact antibodies or as a number of well- characterized fragments produced by digestion with various peptidases or chemicals. Thus, for example, 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 VH-CHI by a disulfide bond. The 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 an Fab' monomer. The Fab' monomer is essentially a Fab fragment with part ofthe hinge region (see, Fundamental Immunology, W. E. Paul, ed., Raven Press, N.Y. (1993), for a more detailed description of other antibody fragments). While various antibody fragments are defined in terms ofthe digestion of an intact antibody, one of skill will appreciate that any of a variety of antibody fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term antibody, as used herein also includes antibody fragments either produced by the modification of whole antibodies or synthesized de novo or antibodies and fragments obtained by using recombinant DNA methodologies.
[0046] The T cell receptor (TcR) is a disulfide linked heterodimer composed of α or β chains or , on a minority of T cells, γ or δ 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 (Vα or Vβ) and one constant domain (Cα or Cβ). The full TcR has a molecular mass of about 95 kDa with the individual chains varying in size from 35 to 47 kDa. Also encompassed within the meaning of TCR are portions ofthe receptor such as the variable regions of this receptor that can be produced as a soluble protein using methods well known in the art. For example, U.S. Patent No. 6,080,840 describes a soluble T cell receptor (TcR) 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 CH2 or CH3 domain, essentially as described in U.S. Patent No. 5,216,132 or as soluble TcR single chains as described by Schusta et al. Nature Biotech. 18,754-759 (2000) or Holler et al. Proc. Natl. Acad. Sci (USA) 97:5387-5392 (2000). The TcR "antibodies" as soluble products may be used in place of antibody for making the compounds ofthe invention. The combining site ofthe TcR can be identified by reference to CDR regions and other framework residues using the same methods discussed above for antibodies.
[0047] Recombinant antibodies may be conventional full length antibodies, antibody fragments known from proteolytic digestion, unique antibody fragments such as Fv or single chain Fv (scFv), domain deleted antibodies, and the like. Fragments may include a domains or polypeptides with as little as one or a few amino acid deleted or mutated while more extensive deletion is possible such as deletion of one or more domains. [0048] An Fv antibody is about 50 Kd in size and comprises the variable regions ofthe light and heavy chain. A single chain Fv ("scFv") polypeptide is a covalently linked VH-VL heterodimer which may be expressed from a nucleic acid including VH- and VL-encoding sequences either joined directly or joined by a peptide-encoding linker. See Huston, et al. (1988) Proc. Nat. Acad. Sci. USA, 85:5879-5883. A number of structures for converting the naturally aggregated, but chemically separated light and heavy polypeptide chains from an antibody V region into an scFv molecule which will fold into a three dimensional structure substantially similar to the structure of an antigen-binding site. See, e.g. U.S. Patent Nos. 5,091,513, 5,132,405 and 4,956,778.
[0049] An exemplary functional component which is an antibody is one that recognizes and binds to the target ofthe targeting component. Where the target is an integrin, exemplary and preferred such antibodies are LM609 and its humanized form known as Vitaxin (See, e.g. Publications WO 89/05155 and WO 01/30393.
[0050] Another class of antibodies that can be incoφorated into a targeting compound of the invention is an antibody with a reactive amino acid side chain in the combining site. 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 ofthe N-terminal variable ("V") regions ofthe 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). In an antibody molecule, the three hypervariable regions of a light chain (LCDR1, LCDR2, and LCDR3) and the three hypervariable regions of a heavy chain (HCDRl, HCDR2 and HCDR3) are disposed relative to each other in three dimensional space to form an antigen binding surface or pocket. The antibody combining site therefore represents the amino acids that make up the CDRs of an antibody and any framework residues that make up the binding site pocket.
[0051] The identity ofthe amino acid residues in a particular antibody that make up the combining site can be determined using methods well known in the art. For example, antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. (see, "Sequences of Proteins of Immunological Interest," E. Kabat et al., U.S. Department of Health and Human Services; Johnson, G and Wu, TT (2001) Kabat Database and its applications: future directions. Nucleic Acids Research, 29: 205-206; http://immuno.bme.nwa.edu). The positions ofthe CDRs may also be identified as the structural loop structures originally described by Chothia and others, (see Chothia and Lesk, J. Mol. Biol. 196, 901 (1987), Chothia et al., Nature 342, 877 (1989), and Tramontano et al., J. Mol. Biol. 215, 175 (1990)). Other methods include the "AbM definition" wliich is a compromise between Kabat and Chothia and is derived using Oxford Molecular's AbM antibody modeling software (now Accelrys) or the "contact definition" of CDRs by Macallum et al., ("Antibody-antigen interactions: contact analysis and binding site topography," J Mol Biol. 1996 Oct 11;262(5):732-45). The following chart identifies CDRs based upon various known definitions.
Loop Kabat AbM Chothia Contact
LI L24 - L34 L24 - L34 L24 - L34 L30 - L36
L2 L50 - L56 L50 - L56 L50 - L56 L46 - L55
L3 L89 - L97 L89 - L97 L89 ~ L97 L89 - L96
HI H31 -- H35B H26 - H35B H26 - H32..34 H30 - H35B
(Kabat Numbering) HI H31 -- H35 H26 - H35 H26 - H32 H30 - H35
(Chothia Numbering) H2 H50 - H65 H50 -- H58 H52 - H56 H47 - H58
H3 H95 -- H102 H95 - H102 H95 - H102 H93 -- H101
General guidelines by which one may identify the CDRs in an antibody from sequence alone are as follows:
LCDR1: Start - Approximately residue 24. Residue before is always a Cys. Residue after is always a Tφ. Typically TRP is followed with TYR-GLN, but also may be followed by LEU-GLN, PHE-GLN, or TYR-LEU. Length is 10 to 17 residues.
LCDR2: Start - 16 residues after the end of LI.
Sequence before is generally ILE-TYR, but also may be VAL-TYR, ILE-LYS, or ILE-PHE. Length is generally 7 residues.
LCDR3:
Start - generally 33 residues after end of L2. Residue before is a Cys. Sequence after is PHE-GLY-X-GLY. Length is 7 to 11 residues.
HCDRl: Start - at approximately residue 26 (four residues after a CYS) [Chothia / AbM definition] Kabat definition starts 5 residues later. Sequence before is CYS-X-X-X.
Residues after is a TRP, typically followed by VAL, but also followed by ILE, or ALA. Length is 10 to 12 residues under AbM definition while Chothia definition excludes the last 4 residues.
HCDR2: Start - 15 residues after the end of Kabat /AbM definition of CDR-H1. Sequence before typically LEU-GLU-TRP-ILE-GLY (SEQ ID NO: 1), but a number of variations are possible.
Sequence after is LYS/ARG-LEU/ILE/VAL/PHE/THR ALA-THR SER/ILE/ALA Length is 16 to 19 residues under Kabat definition (AbM definition ends 7 residues earlier).
HCDR3: Start -33 residues after end of CDR-H2 (two residues after a CYS). Sequence before is CYS-X-X (typically CYS-ALA-ARG). Sequence after is TRP-GLY-X-GLY. Length is 3 to 25 residues.
[0052] The identity ofthe amino acid residues in a particular antibody that are outside the CDRs, but nonetheless make up part ofthe combining site by having a side chain that is part ofthe lining ofthe combining site (i.e., it 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., Riechmann et al., (1988) Nature, 332:;323-327. The aldolase antibody mouse mAb 38C2, which has a reactive lysine near to but outside HCDR3, is an example of such an antibody.
[0053] The reactive residue ofthe antibody combining site may be naturally 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. Alternatively, the amino acid residue may arise by purposely mutating so as to encode the particular residue (see, e.g., WO 01/22922 to Meares et al.). In another approach, the amino acid residue or its reactive elements (e.g., a nucleophilic amino group or sulfhydryl group) may be attached to an amino acid residue in the antibody combining site. Thus, covalent linkage with the antibody occurring "through an amino acid residue in the combining site ofthe antibody" as used herein means that linkage can be made directly to an amino acid residue of an antibody combining site or indirectly through a chemical moiety that is linked to a side chain of an amino acid residue of an antibody combining site.
[0054] Functional components that are proteins can be linked to targeting components via a reactive side chain in the protein. The reactive side chain may be present or may arise by mutation. The reactive side chain in lysine (epsilon amino group) may be covalently linked to a linker comprising a ketone, diketone, beta lactam, active ester haloketone, lactone, anhydride, maleimide, epoxide, aldehyde amidine, guanidine, imines, eneamines, phosphates, phosphonates, epoxides, aziridines, thioepoxides, masked or protected diketones (ketals for example), lactams, haloketones, aldehydes, and the like. Such a reactive lysine side chain is present in the combining site of an aldolase antibody e.g., mouse monoclonal antibody mAb 38C2 and other like catalytic antibodies as well as suitably humanized and chimeric versions of such antibodies. Mouse mAb 38C2 is the prototype of a new class of catalytic antibodies that were generated by reactive immunization and mechanistically mimic natural aldolase enzymes (Barbas et al., 1997, Science 278, 2085-2092). Through a reactive lysine, these antibodies catalyze aldol and retro-aldol reactions using the enamine mechanism of natural aldolases (Wagner et al., 1995, Science 270, 1797-1800; Barbas et al., 1997, Science 278, 2085-2092; Zhong et al., 1999, Angew. Chem. Int. Ed. 38, 3738-3741; Karlstrom et al., 2000, Proc. Natl. Acad. Sci. U.S.A., 973878-3883). In addition to their versatility and efficacy in synthetic organic chemistry (e.g., Hoffmann et al., 1998, J. Am. Chem. Soc. 120, 2768-2779 ; Sinha et al., 1998, Proc. Natl. Acad. Sci. U.S.A. 95, 14603-14608 ), aldolase antibodies have been used to activate camptothecin, doxorubicin, and etoposide prodrugs in vitro and in vivo as an anti-cancer strategy (Shabat et al., 1999, Proc. Natl. Acad. Sci. U.S.A. 96, 6925- 6930 and ,2001, Proc. Natl. Acad. Sci. U.S.A. 98, 7528-7533 ).
[0055] Catalytic antibodies are a preferred source of such functional component antibodies include aldolase antibodies, beta lactamase antibodies, esterase antibodies, amidase antibodies, and the like. In one approach, a targeting component is derivatized with a linker having one or more C=O groups, preferably configured as an aldolase antibody. Suitable such linkers may include a ketone, a diketone, a beta lactam, a succinamide haloketone, a lactone, an anhydride, a maleimide, an alpha-haloacetamide, a cyclohexyl diketone group, and the like (see above). Examples of such linker compounds are shown in FIGs. 2-5.
[0056] In another example, the reactive amino acid of a functional component such as an antibody may be a reactive cysteine, serine or tyrosine residue. For cysteines, the resulting antibody may form a covalent linkage with maleimide-containing components or other thiol- reactive groups such as iodoacetamides, aryl halides, disulfhydryls and the like. Reactive cysteines may be found in thioesterase catalytic antibodies as described by Janda et al., Proc. Natl. Acad. Sci. (USA) 91:2532-2536, (1994). For other esterase antibodies, see Wirsching et al., Science 270:1775-82 (1995). Reactive amino acid-containing functional components may be prepared by means well known in the art including mutating the amino acid residue to encode for the reactive amino acid or chemically derivatizing an amino acid side chain that contains the reactive group. [0057] The components of the targeting compound are linked preferably covalently and preferably with a linker or branched linker. In some embodiments where an antibody is the functional component, the targeting component may be covalently linked to the antibody combining site by using a linker. An appropriate linker can be chosen to provide sufficient distance between the targeting component and the functional component in order for the targeting component to be able to bind to its target molecule. Where linkage to the combining site of an antibody is desired, an appropriate linker can be chosen to provide sufficient distance between the targeting component and the antibody combining site in order for the targeting component to be able to bind to its target molecule. This distance depends on several factors including, for example, the distance from the outermost surface ofthe antibody combining site to the reactive side chain in the combining site, and the nature ofthe targeting agent. Generally, the linker will be between about 5 to 10 angstroms (0.5 to 1 nm) in length, with 10 or more angstroms (1.0 nm) being more preferred, although shorter linkers of about 3 angstroms (0.3 nm) in length may be sufficient if the amino acid side chain is very near to the outermost portion ofthe combining site and/or the targeting component includes a segment that can function as a part of a linker.
[0058] Linker length may also be viewed in terms of the number of linear atoms (cyclic moieties such as aromatic rings and the like to be counted by taking the shortest route). Linker length under this measure is generally about 10 to 200 atoms and more typically about 30 or more atoms, although shorter linkers of two or more atoms may be sufficient if the reactive amino acid side chain is very near to the outermost portion ofthe antibody combining site. Generally, linkers with a linear stretch of at least about 9 atoms are sufficient. The above linker lengths for linking to antibody combining sites are generally applicable to linking integrin targeting components to non-antibody functional components.
[0059] Other linker considerations include the effect ofthe linker on physical or pharmacokinetic properties ofthe resulting targeting compound such as, solubility, lipophilicity, hydrophilicity, hydrophobicity, stability (more or less stable as well as planned degradation), rigidity, flexibility, immunogenicity, modulation of binding, chemical compatibility with targeting agent, ability to be incoφorated into a micelle or liposome, and the like. In the case of RGD peptidomimetic targeting components, the linker may be attached to the spacer between the core ofthe molecule and the basic or acid group. Alternatively, the linker can be attached to the core itself.
[0060] In some embodiments, the linker includes any atom from the group C, H, N, O, P, S, Si, halogen (F, CI, 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, or sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, phosphoalkynyl group, as well as a carbocyclic or heterocyclic mono or fused saturated or unsaturated ring structure. Combinations ofthe above groups and rings may also be present in the linkers ofthe targeting compounds ofthe invention.
[0061 ] The general design of a embodiment of a linear linker for use in targeting compounds ofthe present invention is shown in FIG. 2. The linker includes three functional areas identified starting from the targeting agent (T), being the connecting chain (X), the recognition group (Y) and the reactive group (Z). The integrin targeting agent-linker compound SCS-873 is shown in FIG. 2 with the linker portions X, Y and Z identified. In some embodiments, the recognition group may not be needed.
[0062] Linker reactive group Z may include one or more C=O, groups arranged to form a diketone, an acyl beta-lactam, an active ester, haloketone, a cyclohexyl diketone group, an aldehyde or maleimide. Other groups may include lactone, anhydride, and alpha- haloacetamide and epoxide. Exemplary linker electrophilic reactive groups that can covalently bond to a reactive nucleophilic group (e.g. lysine or cysteine side chain) of a functional component or integrin targeting component 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, sulfonate, a masked or protected diketone (a ketal for example), lactam, and the like, masked C=O groups such as imine, ketal, acetal and any other known electrophilic group. A preferred linker reactive group includes one or more C=O, groups arranged to form a acyl beta-lactam, simple diketone, succinimide active ester, maleimide, haloacetamide with linker, haloketone, cyclohexyl diketone, or aldehyde. [0063] Linker diketone reactive groups which form reversible covalent bonds with the reactive lysine or cysteine of a functional component or integrin targeting component are shown in FIG. 6. Ri and R2 and R3 in structures A-C represent substituents which can be C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof. These 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. R2 and R3 could be cyclic as exemplified in structures B and C while X could be a heteroatom. Other diketone linker reactive groups are shown in FIG. 7 as structures B and G. FIG. 7 also includes the structures of other preferred linker reactive groups.
[0064] In some embodiments, the linker may include a reactive group that forms a nonreversible covalent bond with the combining site of an antibody. Exemplary such reactive groups, shown as structures D-G in FIG. 6, are useful for nonreversibly attaching a targeting component-linker to a reactive nucleophilic group (e.g. lysine or cysteine side chain) in the functional component (or vice versa). Other diketone linker reactive groups that form nonreversible covalent bonds are shown in FIG. 7 as structures A, C and D.
[0065] Linkers may optionally contain a recognition group Y situated between the reactive group portion and the connecting chain portion ofthe linker such as shown in FIG. 2. While not wishing to be bound by any theory, the recognition group, if present, may work to properly position the reactive group into a binding site such an antibody combining site so that it may react with a reactive amino acid side chain. FIG. 8 shows a variety of exemplary recognition groups with one or more homo or hetero ring structures of five or six atoms. Larger ring structures are also possible.
[0066] Various embodiments ofthe connecting chain X portion ofthe general linker design (FIG. 2) is shown in FIG. 9. As shown, the connecting chain may vary considerably in length with both straight chain and branched chain structures possible.
[0067] A preferred linker for use in targeting compounds ofthe invention and for preparing targeting agent-linker compounds or functional component-linker compounds is a linker with a 1,3 -diketone reactive group have the structure 33 as shown below where n is from 1-100 or more and preferably is 1, 2, or 4, and more preferably is 3. In some embodiments, the linker is a repeating polymer such as polyethylene glycol.
Figure imgf000026_0001
33
[0068] The linker reactive group or similar such reactive group that may be inherent in the targeting component can be chosen for linkage with a particular functional component. For example, 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 1,3-diketone configuration such as the diketone shown in Compound SCS-873 (see below) or SCS-864 (see below), is especially preferred as a substrate for modification by an aldolase antibody.
[0069] A linker reactive group chemical moiety suitable for covalent modification by a reactive sulfhydryl group of a functional component or integrin targeting component 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. One of skill in the art will readily appreciate that reactive amino acid side chains in protein functional components may possess an electrophilic group that reacts with a nucleophilic group on the targeting component or its linker, whereas in other embodiments a reactive nucleophilic group in an amino acid side chain of a protein functional group reacts with an electrophilic group in a targeting component or linker. Thus, protein component side chains may be substituted with an electrophile (e.g., FIGs. 6 and 7) and this group may be used to react with a nucleophile on the targeting component or its linker (e.g., NH2). In this embodiment, the functional and targeting components each have a partial linker with appropriate reactive moieties at each end so that the two ends ofthe partial linker can form the full linker, thus creating the complete targeting compound.
[0070] Integrin targeting compounds may be prepared by several approaches. In one approach, a targeting component-linker compound is synthesized with a linker that includes one or more reactive groups designed for covalent reaction with a susceptible reactive moiety on the functional component. In a preferred embodiment, the suitable reactive moiety can be a side chain of an amino acid. The component-linker compound and functional component are then combined under conditions where the linker reactive group forms a covalent bond with the functional component. "Susceptible" as used herein with reference to a chemical moiety indicates that the chemical moiety will covalently bond with a compatible reactive group. Thus, an electrophilic group is susceptible to covalent bonding with a nucleophillic group and vice versa.
[0071] In another approach, linking can be achieved by synthesizing a functional component-linker compound comprising the functional component and a linker where the linker includes one or more reactive groups designed for covalent reaction with a susceptibe chemical moiety ofthe targeting component. The targeting component may need to be modified to provide the appropriate reactive moiety for reaction with the linker reactive group. The functional component-linker and targeting component are combined under conditions where the linker reactive group covalently links to the targeting component.
[0072] A further approach for forming targeting compounds ofthe invention uses a dual linker design. In one embodiment, a targeting component-linker compound is synthesized which comprises a targeting component and a linker with a reactive group. A functional component-linker compound also is synthesized which comprises a functional component and a linker, the latter with a chemical moiety susceptible to reactivity with the reactive group ofthe component-linker ofthe first step. These two linker containing compounds are then combined under conditions whereby the linkers covalently link, forming the targeting compound. [0073] In yet another embodiment, a functional component-linker compound is synthesized which comprises a functional component and a linker with a reactive group. A targeting component-linker compound is also prepared which comprises the component and a linker, the latter with a chemical moiety susceptible to reactivity with the reactive group of the antibody-linker ofthe first step. These two linker containing compounds are then combined under conditions whereby the linkers covalently link, forming the targeting compound.
[0074] Numerous means well known in the art can be used to attach a linker to the targeting agent or antibody combining site. 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.
[0075] A functional component can be linked to a targeting component using a linker moiety that is labile under certain conditions. The labile linkage may be between the functional component and the linker, between the targeting component and the linker, or within the linker, or combinations thereof. For example, the linker may be labile when subjected to a certain pH. The linker may also be a substrate for a particular enzyme, such as an enzyme present in body fluids. Thus, the particular design ofthe labile linker may be used to direct the release ofthe biological agent functional component after it has reached it intended target. A labile linker may be a reversibly covalent bond. Such linker may be an acid-labile linker such as a cis-aconitic acid linker that takes advantage ofthe acidic environment of different intracellular compartments such as the endosomes encountered during receptor mediated endocytosis and the lysosomes. See Shen et al., Biochem. Biophys. Res. Commun. (1981) 102:1048-1054; Yang et al., J. Natl. Cane. Inst. (1988) 80: 1154-1159. In other embodiments, a peptide spacer arm is employed as the linker so that the functional component can be released by the action of a peptidase such as a lysosomal peptidase. See e.g., Trouet et al., Proc. Natl. Acad. Sci. (1982) 79: 626-629. Shown below (Compound 34) is an exemplary targeting compound of this invention wherein the targeting moiety is an integrin antagonist and the functional component is propyrrolinodoxorubicin (R=peptides) and the two are linked by a pH sensitive labile linker.
[0076] Labile linkers include, reversible covalent bonds, pH sensitive linkages (acid or base sensitive), enzyme sensitive linkages, degradation sensitive linkers, photosensitive linkers, sand the like, and combinations thereof. These features are also characteristic of a prodrug which can be considered as a type of labile linker. A variety of labile linkers have been previously moieties designed. For example, prodrugs can be formed using compounds having carboxylic acid that slowly degrade by hydrolysis as described in U.S. Patent No. 5,498,729.
Figure imgf000029_0001
[0077] In this regard, the functional component can be a "prodrug," meaning that the functional component is essentially therapeutically inactive, but becomes active upon some modification. The prodrug can be delivered at the surface of a cell or intracellulary using antibody targeting compounds ofthe invention where it can then be activated. In the prodrug approach, site-specific drug delivery can be obtained from tissue-specific activation of a prodrug, which is the result of metabolism by an enzyme that is either unique for the tissue or present at a higher concentration (compared with other tissues); thus, it activates the prodrug more efficiently.
[0078] Photodynamic treatment may be used to activate a prodrug by cleaving a photosenitive linker or by activating a photoresponsive enzyme (acyl enzyme hydrolysis) as described previously (see U.S. Patent No. 5,114,851 and 5,218,137). Photodynamic treatment also may be used to rapidly inactivate a drug in sites where the drug activity is not desired (e.g. in non-target tissues). Various means of covalently modifying a drug to form a prodrug are well known in the art.
[0079] One desiring to make a compound ofthe present invention simply selects a structure having a pre-selected activity. In one embodiment, the functional component is a therapeutic agent such as drug. Any suitable drug can be used. Selection ofthe therapeutic agent depends upon the desired activity and target ofthe present compound. Where the target is an integrin, a preferred therapeutic is an agent having biological activity directed against the integrin. For example, in the case of Kaposi's Sarcoma, a cancer associated with angiogenesis of cancerous lesions, one can chose any of various drugs such as, for example, the three drugs paclitaxel, doxorubicin, and etoposide with demonstrated therapeutic efficacy in this disease. A derivative of doxorubicin, 2-pyrrolinodoxorubicin is 500-1000 times more potent than doxorubicin itself and it has been extensively studied in other drug targeting strategies (for a recent review see Schally and Nagy, Eur. J. Endocrinology 141, 1-14, 1999). A compound comprising an integrin targeting component with any of these compounds can be used to treat the abnormal angiogenesis of Kaposi's Sarcoma. Synthesis of these compounds with the integrin targeting component SCS-873 is described in the Examples.
[0080] The integrin targeting compounds ofthe present invention have many uses. In one approach, a functional component can be delivered to integrin associated with cells, tissue or fluid macromolecule of an individual by administering the targeting compound. In a preferred approach, the functional component is a therapeutic agent.
[0081] The integrin targeting invention compounds have particular utility for treating a pathological condition associated with integrin expression. Accordingly, a method is provided for treating or preventing a disease or condition in an individual wherein said disease or condition involves an integrin, the method comprising administering to the individual a therapeutically effective amount of an invention targeting compound comprising a therapeutic component effective against the disease or condition. In one such embodiment, the condition is a carcinoma. The association of integrin expression in carcinomas is well known in the art (See, e.g., U.S. Pat. Nos. 5,753,230 and 5,766,591). [0082] The integrin targeting compounds ofthe present invention may be used for treating any disease or condition that is associated with the integrin being targeted. For example, the vitronectin receptor on osteoclasts is known to inhibits osteoclastic bone resoφtion. Thus, diseases or conditions in which bone resoφtion is associated with a pathology, such as osteoporosis and osteoarthritis, can be treated by administering a vitronectin targeting compound ofthe invention. Alternatively, a vitronectin targeting compound with an appropriate functional component can be used to stimulate bone formation by increasing osteocalcin release by osteoclasts. Increased bone production is a clear benefit in disease states wherein there is a deficiency of mineralized bone mass or remodeling of bone is desired, such as fracture healing and the prevention of bone fractures. Diseases and metabolic disorders which result in loss of bone structure would also benefit from such treatment. For instance, hypeφarathyroidism, Paget's disease, hypercalcemia of malignancy, osteolytic lesions produced by bone metastasis, bone loss due to immobilization or sex hormone deficiency, Behcet's disease, osteomalacia, hyperostosis and osteopetrosis, could benefit from administering a compound of this invention.
[0083] The integrin targeting compounds ofthe present invention also may be used for treating any inflammatory disorders, such as rheumatoid arthritis and psoriasis, and cardiovascular diseases, such as atherosclerosis and restenosis, which involve vitronectin expressing cells. Accordingly, vitronectin targeting compounds ofthe invention which comprise an appropriate functional component can be used to treat these disorders. This approach also applies to the treatment or prevention of other diseases including, but not limited to, thromboembolic disorders, asthma, allergies, adult respiratory distress syndrome, graft versus host disease, organ transplant rejection, septic shock, eczema, contact dermatitis, inflammatory bowel disease, and other autoimmune diseases. The compounds ofthe present invention may also be useful for wound healing.
[0084] The integrin targeting compounds ofthe present invention further find use in treating angiogenic disorders. Such disorders involve abnormal neovascularization where growth of new blood vessels is the cause of, or contributes to, the pathology associated with a disease. In these situations, inhibition of angiogenesis will reduce the deleterious effects of the disease. Other therapeutic targets for the compounds ofthe instant invention are eye diseases characterized by neovascularization. Such eye diseases include corneal neovascular disorders, such as corneal transplantation, heφetic keratitis, luetic keratitis, pterygium and neovascular pannus associated with contact lens use. Additional eye diseases include age- related macular degeneration, presumed ocular histoplasmosis, retinopathy of prematurity, neovascular glaucoma, and the like.
[0085] Where the growth of new blood vessels is required to support growth of a deleterious tissue, inhibition of angiogenisis will reduce the blood supply to the tissue and thereby contribute to reduction in tissue mass based on blood supply requirements. Cancer is an example where neovascularization is a continual requirement in order for the tumor to grow and establish tumor metastases. Thus, the integrin targeting compounds ofthe present invention inhibit tumor tissue angiogenesis, thereby preventing tumor metastasis and tumor growth.
[0086] In addition to therapeutic applications, integrin targeting compounds ofthe invention also may be used for imaging of cells or tissues such as tumor cells as is well known in the art. Accordingly, provided is a method of imaging cells or tissue in an individual wherein said cells or tissue expresses an integrin target molecule, said method comprising administering to the individual an effective amount ofthe integrin targeting compound linked to a suitable radioisotope or detectable label. The radioisotope or label may be attached to the targeting component or the functional component.
[0087] An integrin targeting compound ofthe present invention can be administered as a pharmaceutical composition wherein the invention compound is formulated with a pharmaceutically acceptable carrier. Accordingly, the invention compounds may be used in the manufacture of a medicament. Pharmaceutical compositions ofthe invention compounds may be formulated as solutions or lyophilized powders for parenteral administration. Powders may be reconstituted by addition of a suitable diluent or other pharmaceutically acceptable carrier prior to use. Powders also may be sprayed in dry form. The liquid formulation may be a buffered, isotonic, aqueous solution. Examples of suitable diluents are normal isotonic saline solution, standard 5% dextrose in water or buffered sodium or ammonium acetate solution. Such formulation is especially suitable for parenteral administration, but may also be used for oral administration or contained in a metered dose inhaler or nebulizer for insufflation. It may be desirable to add excipients such as polyvinylpyrrolidone, gelatin, hydroxy cellulose, acacia, polyethylene glycol, mannitol, sodium chloride or sodium citrate.
[0088] Alternately, integrin targeting compounds may be encapsulated, tableted or prepared in a emulsion or syrup for oral administration. Pharmaceutically acceptable solid or liquid carriers may be added to enhance or stabilize the composition, or to facilitate preparation ofthe composition. Solid carriers include starch, lactose, calcium sulfate dihydrate, terra alba, magnesium stearate or stearic acid, talc, pectin, acacia, agar or gelatin. Liquid carriers include syrup, peanut oil, olive oil, saline and water. The carrier may also include a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax. The amount of solid carrier varies but, preferably, will be between about 20 mg to about 1 g per dosage unit. The pharmaceutical preparations are made following the conventional techniques of pharmacy involving milling, mixing, granulating, and compressing, when necessary, for tablet forms; or milling, mixing and filling for hard gelatin capsule forms. When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion or an aqueous or non-aqueous suspension. For rectal administration, the invention compounds may be combined with excipients such as cocoa butter, glycerin, gelatin or polyethylene glycols and molded into a suppository.
[0089] Integrin targeting compounds ofthe invention may be formulated to include other medically useful drugs or biological agents. The compounds also may be administered in conjunction with the administration of other drugs or biological agents useful for the disease or condition that the invention compounds are directed (see e.g., U.S. Pat. No. 6,413,955 for active ingredients useful for osteoporosis).
[0090] As employed herein, the phrase "an effective amount," refers to a dose sufficient to provide concentrations high enough to impart a beneficial effect on the recipient thereof. The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including the disorder being treated, the severity ofthe disorder, the activity ofthe specific compound, the route of administration, the rate of clearance ofthe compound, the duration of treatment, the drugs used in combination or coincident with the compound, the age, body weight, sex, diet and general health ofthe subject, and like factors well known in the medical arts and sciences. Various general considerations taken into account in determining the "therapeutically effective amount" are known to those of skill in the art and are described, e.g., in Gilman et al., eds., Goodman And Gilman's: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press, 1990; and Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa., 1990. Dosage levels typically fall in the range of about 0.001 up to 100 mg/kg/day; with levels in the range of about 0.05 up to 10 mg/kg/day are generally applicable. A compound can be administered parenterally, such as intravascularly, intravenously, intraarterially, intramuscularly, subcutaneously, or the like. Administration can also be orally, nasally, rectally, transdermally or inhalationally via an aerosol. The composition may be administered as a bolus, or slowly infused.
[0091] The administration ofthe targeting compound to an immunocompetent individual may result in the production of antibodies against the compound. Such antibodies may be directed to the targeting component, the functional component or any other entity associated with the compound. The immunogenicity of such compound may be addressed by methods well known in the art such as by attaching long chain polyethylene glycol (PEG)-based spacers, and the like, to one or more components ofthe compound. 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 (Katre et al., 1990, J. Immunol. 144, 209-213; Francis et al., 1998, Int. J. Hematol. 68, 1-18). As noted, PEG can serve as a linker in targeting compounds ofthe invention, thus providing both linker function and reduced immunogenicity. Alternatively or in addition, the individual may be administered an immunosuppressent drug such as cyclosporin A, anti-CD3 antibody, and the like, to reduce the likelihood that an immune response to the targeting compound will develop.
[0092] In some embodiments, RGD and non-RGD peptidomimetic or non-RGD peptide antagonists or agonists can be used to target liposomes. This is achieved by chemically linking such targeting components to a lipid moiety that enables the targeting component to associate with lipids ofthe liposome. The liposome encapsulated with an appropriate drug can then be targeted more effectively in vivo by aid ofthe targeting component. For example, treatment of Kaposi's sarcoma with sterically stabilized stealth liposomes containing doxorubicin (DoxilTM) is one ofthe most effective ofthe approved KS therapies. This liposomal formulation provides several advantages over administration of free doxorubicin. In addition to a reduction in cardiac toxicity, vomiting, alopecia, peripheral neuropathy, and mucositis provided by the liposomal formulation, DoxilTM possesses an intrinsic capacity for passive targeting to the tumor as a result of particulate size and the enhanced permeability and retention phenomenon (Matsumura et al., Cancer Res. 46, 6387- 6392, 1986) that is further aided by the extended serum half-life ofthe drug.
[0093] Active targeting as disclosed herein can augment the passive targeting capacity of DoxilTM thereby more efficiently delivering the cytotoxic payload ofthe liposomes to the KS tumor and its vasculature. Such active targeting of DoxilTM likely will reduce the dosage needed for clinical response thereby limiting non-specific toxicities. Administration of targeted liposomes may be achieved as previously described (See, e.g., Allen et al., Adv. Drug Deliv. Rev. 21, 117-133). The targeting compounds ofthe invention may be used in combination with other compounds or therapies such as radiation therapy.
[0094] It would be readily evident that the compounds ofthe invention find use not only in human medical therapy and diagnosis but also in vitro diagnostics, veterinary, agricultural, environmental and other disciplines. The versatility ofthe invention is illustrated by the following Examples which illustrate preferred embodiments ofthe invention and are not limiting ofthe claims or specification in any way.
EXAMPLE 1: Antibody targeting compound comprising an RGD peptidomimetic targeting agent covalently linked to the combining site of aldolase monoclonal antibody 38C2. [0095] An integrin targeting compound was formed based on the formation of a reversible covalent bond between a diketone linker derivative of an RGD peptidomimetic and the reactive lysine of mouse mAb 38C2. Mouse mAb 38C2 is the prototype for a new class of catalytic antibodies generated by reactive immunization and mechanistically mimic natural aldolase enzymes (Barbas et al., Science 278, 2085-2092, 1997). Through a reactive lysine, these antibodies catalyze aldol and retro-aldol reactions using the enamine mechanism of natural aldolases (Wagner et al., Science 270, 1797-1800, 1995; Barbas et al., Science 278, 2085-2092, 1997; Zhong et al., Angew. Chem. Int. Ed. 38, 3738-3741, 1999). In addition to their versatility and efficacy in synthetic organic chemistry, aldolase antibodies have been used in the activation of camptothecin, doxorubicin, and etoposide prodrugs in vitro and in vivo as an anti-cancer strategy (Shabat et al., Proc. Natl. Acad. Sci. U.S.A. 96, 6925-6930, 1999); Shabat, D. et al. Proc. Natl. Acad. Sci. U.S.A. 98, 7528-7533, 2001). Yet another feature of these antibodies, namely their ability to bind diketones covalently, has remained largely unexplored.
[0096] The RGD peptidomimetic used (see Compound 1) is specific for human integrin with a high binding affinity for αvβ at 0.9 nM and αvβ5 at 0.6 nM (specificity exhibited by minimal aιn,b3 binding) (Miller et al., supra). A diketone linker modified version of Compound 1, designated SCS-873, was prepared as described in Example 3. SCS-873 is shown below with the targeting component and the linker separately identified.
Figure imgf000036_0001
Targeting Agent Linker with 1 ,3 Diketone Group
SCS873
[0097] A peptidomimetic RGD antagonist with known activity for both αvβ3 or αvβ5 binding is desirable because some of these compounds bind both murine and human integrins. Such species cross reactivity affords preclinical in vivo studies in animal angiogenesis models prior to human trials. In addition, the targeting compound may be used for the therapy of Kaposi's sarcoma which is associated with αvβ3 integrin.
[0098] SCS-873 was linked to antibody 38C2 by the following procedure: One milliliter antibody 38C2 in phosphate buffered saline (lOmg/ml) was added to 12 microliters of a 10 mg/mL stock solution of SCS-873 and the resulting mixture was maintained at room temperature for 2 hours prior to use. [0099] The binding of a mixture of SCS-873 and 38C2 to SLK cells was evaluated. SCS- 873 effectively mediated cell surface binding of 38C2. No binding of 38C2 was detectable in the absence of SCS-873. Control experiments confirmed that the diketone moiety ofthe linker is required for binding of SCS-873 to 38C2. It was determined that SCS-873 retains the integrin specificity ofthe integrin targeting component, i.e., no binding to am,b3 in ELISA was detected while binding to αvβ3 and αvβ was found to be strong. Independent i.p. and i.v. injections ofthe targeting compound prepared with SCS-873 and 38C2 versus each component alone into mice demonstrated integrin targeting in vivo. In these experiments, the serum half-life of SCS-873 was extended by more than two orders of magnitude through binding to 38C2. Free SCS-873 not bound to antibody had a serum half-life of only minutes while the combination of antibody and small molecule could be detected in the serum sampled from eye bleeds after several days.
EXAMPLE 2: Integrin Targeting with Protein Functional Components. [00100] The integrin targeting component can be covalently attached to a functional component such as a protein in order to channel effector functions triggered by these proteins. Such linking can be achieved by a lysine-reactive N-hydroxysuccinimide or a cysteine-reactive maleimide functionality.
[00101] For example, conjugation ofthe integrin targeting compound to an IgM antibody induces complement-mediated cytotoxicity; conjugation to IL-2 results in cell-mediated cytotoxicity. A variety of antibodies that neutralize growth factors involved in tumor angiogenesis can be modified with the integrin targeting compound in order to increase their selectivity. For example, a monoclonal antibody that neutralizes VEGF is highly selective if conjugated to the integrin targeting compound. Taking the potential immunogenicity of these conjugates into consideration, long chain polyethylene glycol (PEG)-based spacers can be introduced between the protein-reactive and the integrin targeting functionality. 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 (Katre et al., 1990, J. Immunol. 144, 209-213; Francis et al., 1998, Int. J. Hematol 68, 1-18). EXAMPLE 3: Synthesis of Targeting Components-Linkers Molecules
[00102] Integrin targeting components shown as compounds 15 and 4 were synthesized as shown in the FIG. 10 (Scheme 1) and FIG. 11 (Scheme 2), respectively. A linker with a diketone reactive moiety was added to these targeting molecules as shown in Scheme 3 (FIG. 12) to form targeting compound-linker molecules SCS-873 and SCS-1655. Synthesis of SCS-873 was achieved starting from compound 14 in three steps. Compound 14 was converted to 15 as shown in Scheme 1 and the crude product was reacted with an N- hydroxysuccinimide (NHS) -ester ofthe diketone compound 23 in CH3CN-DMF in the presence of Et3N. Purification over silica gel (CH2Cl2-MeOH, 9:1) afforded pure SCS-873.
[00103] Compound SCS-1655 was synthesized from 14 in five steps (Schemes 2 and 3). Deprotection ofthe BOC group in compound 14 followed by reaction with the NHS ester of the bivalent linker 24 afforded compound 25, which was then deprotected and reacted with 23 as above to afford SCS-1655.
[00104] Synthesis of integrin targeting component-linker molecules SCS-864 and SCS- 789 is shown in Scheme 4 (FIG. 13). SCS-864 and SCS-789 were each synthesized in one step from compound 4 (FIG. 13, scheme 4). Linking of Compound 4 was achieved with the appropriate activated NHS-ester. SCS-864 is shown below with the targeting component and linker separately identified.
Figure imgf000038_0001
Targeting Agent Linker with 1 ,3 Diketone Group
SCS864 EXAMPLE 4: Synthesis of an Integrin Targeting Compound with Paclitaxel as the
Functional Component
[00105] Paclitaxel-SCS-873 is synthesized starting from taxol-succinate prepared as previously described (Deutsch et al., J. Med. Chem. 32, 788-792, 1989). Following activation ofthe carboxy group with PyBOP in DMF, the SCS-amine is directly coupled (Huang et al., Chemistry & Biology 7, 453-461, 2000) providing Paclitaxel-SCS-873. This derivative is analogous to a previously described somatostatin antagonist peptide targeted paclitaxel derivative (Huang et al., Chemistry & Biology 7, 453-461, 2000) that demonstrated good activity and targeting, thereby validating the succinate-based linker strategy for this drug. Others have described paclitaxel prodrugs utilizing a 2'-carbamate instead of a succinate-based linker with success (de Groot et al., J. Medicinal Chem. 43, 3093-3102, 2000). Paclitaxel-SCS-873 is considerably more soluble than paclitaxel itself which suffers from poor solubility. The structure of Paclitaxel-SCS-873 is shown below.
Figure imgf000039_0001
Paclitaxel-SCS-873
EXAMPLE 5: Synthesis of an Integrin Targeting Compound with Doxorubicin or 2-
Pyrrolinodoxorubicin as the Functional Component
[00106] Doxorubicin-SCS-873 is prepared using a synthetic scheme similar to that described for doxorubicin derivatives of luteinizing hormone-releasing hormone and somatostatin conjugates (Nagy et al., Proc. Natl. Acad. Sci. U.S.A. 93, 7269-7273, 1996; Nagy et al., Proc. Natl. Acad. Sci. U.S.A. 95, 1794-1799, 1998). N-Fmoc-DOX-14-O- hemiglutarate are prepared as described by Nagy et al. (Proc Natl. Acad. Sci. 93, 2464-2469, 1996) and subsequently activated with PyBOP in DMF followed by addition ofthe SCS- amine and removal ofthe Fmoc protective group affording Doxorubicin-SCS-873. The structure of Doxorubicin-SCS-873 is shown below.
[00107] 2-Pyrrolinodoxorubicin-SCS-873 can be prepared from Doxorubicin-SCS-873 by reaction with 4-iodobutyraldehyde as described for LH-RH-2 -Pyrrolinodoxorubicin conjugates (Nagy et al., Proc Natl. Acad. Sci. 93, 2464-2469, 1996). A variety of other synthetic approaches are also available. The proven activity of LH-RH-2- pyrrolinodoxorubicin and doxorubicin conjugates of similar design supports the design of our integrin targeted derivatives (See, e.g., Schally and Nagy, Eur. J. Endocrinology 141, 1-14, 1999 and references therein for a review peptide targeted drug conjugates). The structure of 2-Pyrrolinodoxorubicin-SCS-873 is shown below.
Figure imgf000040_0001
EXAMPLE 6: Synthesis of an Integrin Targeting Compound with Etoposide as the Functional Component
[00108] Etoposide-SCS-873 is be prepared from the p-nitrophenylcarbonate recently described for structurally similar etoposide prodrugs that are activated by catalytic antibody (Shabat et al, Proc. Natl. Acad. Sci. U.S.A. 98, 7528-7533, 2001). The retro-Michael step was readily catalyzed by endogenous cellular enzymes as well as by the catalytic antibody (Shabat et al., Proc. Natl. Acad. Sci. U.S.A. 96, 6925-6930, 1999). The design of Etoposide- SCS-873 is based on endogenous enzyme or general acid base activation via a retro-Michael reaction that is followed by spontaneous decarboxylation and lactamization reactions that provide active etoposide. The structure of Etoposide-SCS-873 is shown below.
Figure imgf000041_0001
Etoposide-SCS-873
EXAMPLE 7: Preparation of an Integrin Targeting Compound for Liposome Targeting
[00109] DoxilTM containing liposomes are associated with a targeting compound ofthe invention as described. The targeting component is attached to maleimide-terminated PEG2000-DSPE (MAL-PEG2000-DSPE). MAL-PEG2000-DSPE is commercially available through Shearwater Polymers, Inc. Attachment of thiol terminated targeting molecules to the maleimide moiety is spontaneous. The PEG-lipid derivative is then transferred into the DoxilTM liposome with negligible drug leakage in a simple incubation step.
[00110] Targeted binding of liposomes to cells is studied by FACS analysis by co- incoφoration of biotin labeled MAL-PEG2000-DSPE and staining with FITC-labeled streptavidin. Various control cell lines are also studied to assess non-specific binding. Cytotoxicity assays are performed as described (Moase et al., 2001) with a variety of different loadings ofthe targeting molecules into the liposome using the three KS cell lines. To study the efficacy of this approach in an SLK animal model, animals are treated 1 day after cell implantation and after tumors have established (200 mm3). A single dosing regimen is studied initially to assess the relative efficacy of targeted Doxil™ vs. untargeted Doxil™. Drug dosing ranges from 0.5 mg/kg to 5 mg/kg i.v.. Other control groups are be treated with empty, but targeted liposomes to assess the effect ofthe multivalent liposome itself on the disease. A buffer control group is also included. In multiple treatment studies, drug is injected at 14-day intervals. Systemic toxicity is assessed as described above. [00111 ] It should be noted that Doxil™ is also approved for the treatment of refractory ovarian cancers. A recent study of 25 permanent human cell lines established from advanced ovarian cancer demonstrated that all lines were positive for integrin expression (Bruning et al., Hum. Gene Ther. 12, 391-399, 2001). This result suggests that the targeting of both tumor and its supporting vasculature also has utility in the treatment of ovarian cancer. Recent studies have indicated that αvβ3 is highly expressed on malignant human cervical tumor tissues (Chattopadhyay and Chatterjee, J. Exp. Clin. Cancer Res. 20, 269-275, 2001), suggesting that the Doxil™ based strategy disclosed herein have utility in the treatment of cervical cancer. Other polymerized liposome assemblies such as those described in Bruehl et al. (Biochemistry, 40:5964-5971, 2001) can also be used.
[00112] The invention thus has been disclosed broadly and illustrated in reference to representative embodiments described above. Those skilled in the art will recognize that various modifications can be made to the present invention without departing from the spirit and scope thereof. All publications, patent applications, and issued patents, are herein incoφorated by reference to the same extent as if each individual publication, patent application or issued patent were specifically and individually indicated to be incoφorated by reference in its entirety. Definitions that are contained in text incoφorated by reference are excluded to the extent that they contradict definitions in this disclosure. All structures shown herein are contemplated to provide all enantiomers and tautomers.

Claims

WHAT IS CLAIMED IS:
1. An integrin targeting compound, comprising at least one integrin targeting component covalently linked to a linear or branched linker which is covalently linked to at least one functional component, wherein said integrin targeting component is selected from the group consisting of:
(a) a RGD peptidomimetic, and
(b) a non-RGD peptide, peptidomimetic or organic molecule integrin agonist or antagonist.
2. The targeting compound of claim 1 wherein said integrin targeting component is a RGD peptidomimetic.
3. The targeting compound of claim 1 wherein said integrin targeting component is a non-RGD peptide.
4. The targeting compound of claim 1 wherein said integrin targeting component is an organic molecule integrin agonist or antagonist.
5. The targeting compound of claim 1 wherein said integrin targeting component is a non-RGD peptidomimetic.
6. The targeting compound of claim 1 wherein the integrin targets o÷iβi,
Figure imgf000043_0001
α3βι, α4βι, αsβ!, 6β!, α^, α8βι, α9βi, ctiβi, α6β4, α4β7, αDβ2, αDβ2,αLβ2, αMβ2 αvβι αvβ3, αvβ5, αvβ6, αvβ8> αxβ2, αnbβ3, or αιELbβ7-
7. The targeting compound of claim 1 wherein said at least one functional component is a therapeutic agent.
8. The targeting compound of claim 7 wherein the therapeutic agent is selected from the group consisting of paclitaxel, doxorubicin, 2-pyyrolinodoxorubicin, and etoposide.
9. The targeting compound of claim 1 wherein said at least one functional component is an antibody.
10. The targeting compound of claim 9 wherein said antibody is full length.
11. The targeting compound of claim 9 wherein said antibody is a fragment of a full length antibody.
12. The targeting compound of claim 11 wherein said fragment of a full length antibody is Fab, Fab' F(ab')2, Fv or sFv.
13. The targeting compound of claim 9 wherein said antibody is a human antibody, humanized antibody or chimeric human antibody.
14. The targeting compound of claim 9 wherein at least one integrin targeting component is covalently linked via a linker to the combining site ofthe antibody.
15. The targeting compound of claim 9 wherein the antibody is a catalytic antibody.
16. The targeting compound of claim 15 wherein said catalytic antibody is selected from the group consisting of an aldolase antibody, a beta lactamase antibody and an esterase antibody or an amidase antibody.
17. The targeting compound of claim 1 wherein the integrin targeting component is linked to two or more functional components.
18. The targeting compound of claim 17 wherein at least one of said two or more functional components is an antibody.
19. The targeting compound of claim 18 wherein said at least one of said two or more functional components is a therapeutic agent.
20. The targeting compound of claim 1 wherein the integrin targeting component comprises two or more targeting components.
21. The targeting compound of claim 1 wherein said linker comprises a linear stretch of between 5-100 atoms selected from the group consisting of C, H, N, O, P, S, Si, F, CI, Br, and I, or a salt thereof.
22. The targeting compound of claim 1 wherein said linker comprises one or more groups selected from alkyl, alkenyl, alkynyl, oxoalkyl, oxoalkenyl, oxoalkynyl, aminoalkyl, aminoalkenyl, aminoalkynyl, sulfoalkyl, sulfoalkenyl, sulfoalkynyl group, phosphoalkyl, phosphoalkenyl, and phosphoalkynyl.
23. The targeting compound of claim 1 wherein said linker comprises a repeating ether unit of between 2-100 units.
24. The targeting compound of claim 1 wherein said linker comprises a heterocarbyl structure ofthe formula
Figure imgf000045_0001
wherein
R2 to R4 is C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof;
n is 1-100; and
m is 1-100.
25. The targeting compound of claim 1 wherein said linker comprises one or more ring structures.
26. The targeting compound of claim 25 wherein said one or more ring structures includes one or more six membered rings ofthe formula
Figure imgf000046_0001
wherein
A, Z, Y, X or W are independently C or N.
27. The targeting compound of claim 25 wherein said one or more ring structures includes one or more five membered rings ofthe formula
X
wherein
A, Z, Y or X are independently C, O, N or S.
28. The targeting compound of claim 1 wherein said integrin targeting component is an RGD peptidomimetic shown as compounds 1, 2 or 3, below.
Figure imgf000047_0001
Figure imgf000047_0002
Figure imgf000047_0003
29. The targeting compound of claim 1 wherein said covalent linkage between said targeting component and said linker or between said linker and said functional component or both is nonreversible.
30. The targeting compound of claim 1 wherein said covalent linkage between said targeting component and said linker or between said linker and said functional component or both is reversible.
31. The targeting compound of claim 1 wherein said covalent linkage between said targeting component and said linker or between said linker and said functional component or both is labile.
32. The targeting compound of claim 31 wherein said labile linkage is a pH sensitive linkage, is a substrate for an enzyme, or is susceptible to degradation by radiation.
33. A method of producing an integrin targeting compound, comprising covalently linking at least one integrin targeting component via a linear or branched linker to at least one functional component, wherein said integrin targeting component is selected from the group consisting of:
(a) a RGD peptidomimetic, and
(b) a non-RGD peptide, peptidomimetic or organic molecule integrin agonist or antagonist.
34. The method of claim 33 wherein said at least one targeting component is linked to said at least one functional component in such a way as to retain the binding function of the targeting component and the biological activity ofthe functional component.
35. The method of claim 33 wherein said linking is achieved by preparing an integrin targeting component-linker compound comprising said at least one integrin targeting component and a linker, said linker comprising a reactive group for reaction with the functional component, and linking said reactive group of said linker covalently to the functional component.
36. The method of claim 33 wherein said linking is achieved by preparing a functional component-linker compound comprising a functional component and a linker, said linker comprising a reactive group for reaction said at least one integrin targeting component, and linking the reactive group of said linker covalently to said at least one integrin targeting component.
37. The method of claim 33 wherein said linking is achieved by:
(a) preparing an integrin targeting component-linker compound comprising an integrin targeting component and a linker with comprising a reactive group; and
(b) preparing a functional component-linker comprising a functional compound and a linker comprising a chemical moiety susceptible to reaction with the reactive group of step (a); or (c) preparing a functional component-linker compound comprising a functional component and a linker comprising a reactive group; and
(d) preparing an integrin targeting component-linker compound comprising an integrin targeting component and a linker comprising a chemical moiety susceptible to reaction with said reactive group of step (c); and
(e) linking the linkers of steps (a) and (b) or steps (c) and (d) covalently together through said reactive and susceptible groups to form the integrin targeting compound.
38. An integrin targeting compound produced by the method of claim 37.
39. An integrin targeting component-linker compound or functional component- linker compound for covalently linking an integrin targeting component to a functional component, said linker ofthe formula
X - Y - Z
wherein
X is a linear or branched connecting chain of atoms comprising any of C, H, N, O, P, S, Si, F, CI, Br, and I, or a salt thereof,
Y if present is a single or fused 5 or 6 membered homo- or heterocarbocylic saturated or unsaturated ring; and
Z is a ketone, diketone, beta lactam, active ester, haloketone, lactone, anhydride, epoxide, aldehyde, maleimide disulfide, or aryl halide; and
wherein Z is a reactive group for covalently linking one ofthe components to reactive amino acid or other susceptible moiety in the other ofthe components, said targeting component or functional component linked to X or Y if present or both X and Y if Y is present.
40. The integrin targeting component-linker compound or functional component- linker compound of claim 39 wherein said components are linked in such a way to retain the ability to bind the target and exhibit functional activity.
41. The integrin targeting component- linker compound or functional component- linker compound of claim 39 wherein X comprises a linear stretch of between 5-200 atoms.
42. The integrin targeting component-linker compound or functional component- linker compound of claim 39 wherein X is a heterocarbyl structure ofthe formula
Figure imgf000050_0001
wherein
R2 to Rt is C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof;
n is 1-100; and
m is 1-100.
43. The integrin targeting component-linker compound or functional component- linker compound of claim 39 wherein Y is a six membered ring ofthe formula
Figure imgf000050_0002
wherein A, Z, Y, X or W are independently C or N.
44. The integrin targeting component-linker compound or functional component- linker compound of claim 39 wherein Y is a five membered ring ofthe formula
Figure imgf000051_0001
wherein
A, Z, Y or X are independently C, O, N or S.
45. The integrin targeting component-linker compound or functional component- linker compound of claim 39 wherein said linker is branched by addition of one or more connecting chains, said linker comprises more than one recognition group, said linker comprises more than one reactive group, or combinations thereof.
46. The integrin targeting component-linker compound or functional component- linker compound of claim 39 wherein said linker has the structure below wherein n is from 1- 100.
Figure imgf000051_0002
33
47. An integrin targeting component- linker compound or functional component- linker compound for covalently linking an integrin targeting component to a functional component, said, said linker ofthe formula
X - Y - Z wherein
X is a linear or branched connecting chain of atoms comprising any of C, H, N, O, P, S, Si, F, CI, Br, and I, or a salt thereof, and comprising a repeating ether unit of between 0- 100 units;
Y is a single or fused 5 or 6 membered homo- or heterocarbocylic saturated or unsaturated ring located within 1-20 atoms of Z; and
Z is a reactive group for covalently linking one ofthe components to a reactive amino acid or other susceptible moiety in the other ofthe components, said targeting component or functional component linked to X or Y.
48. The integrin targeting component-linker compound or functional component- linker compound of claim 47 wherein said agents are linked in such a way as to retain the ability to bind the target and exhibit functional activity.
49. The integrin targeting component-linker compound or functional component- linker compound of claim 47 wherein Z is selected from the group consisting of a ketone, diketone, beta lactam, active ester, haloketone, lactone, anhydride, epoxide, aldehyde, maleimide, disulfide, and aryl halide.
50. The integrin targeting component-linker compound or functional component- linker compound of claim 47 wherein X comprises a linear stretch of between 10-200 atoms.
51. The integrin targeting component- linker compound or functional component- linker compound of claim 47 wherein X is a heterocarbyl ofthe formula
Figure imgf000052_0001
wherein R2 to R4 is C, H, N, O, P, S, Si, halogen (F, CI, Br, I) or a salt thereof
n is 1-100 and
m is 1-100
52. The integrin targeting component-linker compound or functional component- linker compound of claim 47 wherein Y is a six membered ring ofthe formula
wherein
A, Z, Y, X or W are independently C or N.
53. The integrin targeting component- linker compound or functional component- linker compound of claim 47 wherein Y is a five membered ring ofthe formula
wherein
A, Z, Y or X are independently C, O, N or S.
54. The integrin targeting component-linker compound or functional component- linker compound of claim 47 wherein said linker comprises more than one connecting chain, more than one recognition group or more than one reactive group, or combinations thereof.
55. A method of delivering a functional component to integrin expressing cells or tissue of an individual, said method comprising administering to the individual the targeting compound of claim 1.
56. The method of claim 55 wherein said functional component is a therapeutic agent.
57. A method of delivering a functional component to integrin expressing cells or tissue of an individual, said method comprising administering to the individual the targeting compound of claim 38.
58. The method of claim 57 wherein said functional component is a therapeutic agent.
59. A method of treating or preventing a disease or condition that involves integrin in an individual, said method comprising administering to the individual a therapeutically effective amount ofthe targeting compound of claim 7, wherein said therapeutic component reduces the symptoms associated with said disease or condition.
60. The method of claim 59 wherein said disease or condition involves a defect in angiogenesis, bone metabolism, inflammation or cell growth.
61. The method of claim 59 wherein said disease or condition is cancer.
62. A pharmaceutical formulation comprising the targeting compound of claim 1 and a pharmaceutically acceptable carrier.
63. A pharmaceutical formulation comprising the targeting compound of claim 38 and a pharmaceutically acceptable carrier.
PCT/US2002/033866 2001-10-22 2002-10-22 Integrin targeting compounds WO2003034995A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP02773859A EP1446418B1 (en) 2001-10-22 2002-10-22 Integrin targeting compounds
AU2002337954A AU2002337954C1 (en) 2001-10-22 2002-10-22 Integrin targeting compounds
JP2003537564A JP5021152B2 (en) 2001-10-22 2002-10-22 Integrin targeting compounds
KR1020047005978A KR101085784B1 (en) 2001-10-22 2002-10-22 Integrin targeting compounds
CA002464271A CA2464271A1 (en) 2001-10-22 2002-10-22 Integrin targeting compounds
ES02773859T ES2384235T3 (en) 2001-10-22 2002-10-22 Integrin-directed compounds

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US34379901P 2001-10-22 2001-10-22
US60/343,799 2001-10-22
US41251902P 2002-09-19 2002-09-19
US60/412,519 2002-09-19

Publications (2)

Publication Number Publication Date
WO2003034995A2 true WO2003034995A2 (en) 2003-05-01
WO2003034995A3 WO2003034995A3 (en) 2003-09-12

Family

ID=26993632

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2002/033866 WO2003034995A2 (en) 2001-10-22 2002-10-22 Integrin targeting compounds

Country Status (9)

Country Link
US (1) US20030129188A1 (en)
EP (1) EP1446418B1 (en)
JP (1) JP5021152B2 (en)
KR (1) KR101085784B1 (en)
CN (1) CN1606566A (en)
AU (1) AU2002337954C1 (en)
CA (1) CA2464271A1 (en)
ES (1) ES2384235T3 (en)
WO (1) WO2003034995A2 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1443963A2 (en) * 2001-10-22 2004-08-11 The Scripps Research Institute Antibody targeting compounds
US7208145B2 (en) 2002-12-31 2007-04-24 Nektar Therapeutics Al, Corporation Polymeric reagents comprising a ketone or a related functional group
WO2010093706A3 (en) * 2009-02-10 2011-11-10 The Scripps Research Institute Chemically programmed vaccination
US8106131B2 (en) 2002-12-31 2012-01-31 Nektar Therapeutics Hydrolytically stable maleimide-terminated polymers
WO2014124227A1 (en) * 2013-02-07 2014-08-14 Immunomedics, Inc. Pro-drug form (p2pdox) of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer
CN109503700A (en) * 2017-09-14 2019-03-22 南京安吉生物科技有限公司 The blood vessel formation inhibitor IIM 3-1 of maleimide base group modification and its application
US11571480B2 (en) 2015-08-11 2023-02-07 Coherent Biopharma I, Limited Multi-ligand drug conjugates and uses thereof

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20040106547A (en) * 2002-05-15 2004-12-17 엔도사이트, 인코포레이티드 Vitamin-mitomycin conjugates
US7271245B2 (en) * 2004-02-13 2007-09-18 The Scripps Research Institute Methods and compositions for inhibition of metastasis
US20090298195A1 (en) 2005-01-05 2009-12-03 F-Star Biotechnologische Forschungs-Und Entwicklun Gseges M.B.H. Synthetic immunoglobulin domains with binding properties engineered in regions of the molecule different from the complementarity determining regions
US20070122408A1 (en) * 2005-10-20 2007-05-31 The Scripps Research Institute Fc Labeling for Immunostaining and Immunotargeting
AT503889B1 (en) 2006-07-05 2011-12-15 Star Biotechnologische Forschungs Und Entwicklungsges M B H F MULTIVALENT IMMUNE LOBULINE
JP5098011B2 (en) * 2006-11-20 2012-12-12 国立大学法人山口大学 Wound healing promoter
CN102702317A (en) * 2006-11-30 2012-10-03 首都医科大学 RGDSYIGSR with target antithrombotic activity, preparation and application thereof
CN101240031B (en) * 2007-02-07 2010-10-13 首都医科大学 Conjugate constructed from normal heptadecyl and RGD peptide, and its preparation method and application
CN101240028B (en) * 2007-02-07 2010-08-18 首都医科大学 Conjugate constructed from normal pentadecyl and RGD peptide, and its synthesis and application in medicine
CN101240030B (en) * 2007-02-07 2010-08-11 首都医科大学 Conjugate constructed from normal undecyl and RGD peptide, and its synthesis and application in medicine
CN101240029B (en) * 2007-02-07 2010-08-11 首都医科大学 Conjugate constructed from normal heptyl and RGD peptide, and its synthesis method and application
CN101240026B (en) * 2007-02-07 2010-04-21 首都医科大学 Conjugate constructed from normal tridecyl and RGD peptide, and its synthesis and application in medicine
US8921279B2 (en) 2007-06-26 2014-12-30 F-Star Biotechnologische Forschungs—und Entwicklungsges. m.b.H Display of binding agents
EP2113255A1 (en) 2008-05-02 2009-11-04 f-star Biotechnologische Forschungs- und Entwicklungsges.m.b.H. Cytotoxic immunoglobulin
EP2258719A1 (en) * 2009-05-19 2010-12-08 Max-Delbrück-Centrum für Molekulare Medizin (MDC) Multiple target T cell receptor
WO2011094671A2 (en) * 2010-01-29 2011-08-04 The Uab Research Foundation N-terminally conjugated polypeptides for targeted therapy and diagnosis
JP6104287B2 (en) * 2012-03-06 2017-03-29 天津和美生物技術有限公司 Tetracyclic anthraquinone derivatives
CN105451743B (en) * 2013-08-07 2019-11-19 箭头研究公司 For delivering the poly- conjugate of RNAi triggering to tumour cell in the living body
WO2019210308A1 (en) * 2018-04-27 2019-10-31 Arrowhead Pharmaceuticals, Inc. Integrin targeting ligands and uses thereof
WO2022114919A2 (en) * 2020-11-30 2022-06-02 (주)한국바이오셀프 Integrin-binding peptide and pharmaceutical composition containing same

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5114851A (en) 1989-08-29 1992-05-19 Duke University Light activated acyl-enzymes
US5218137A (en) 1989-08-29 1993-06-08 Duke University Light activated acyl-enzymes
US5498729A (en) 1989-12-26 1996-03-12 Domb; Abraham J. Prodrug compositions
US6013625A (en) 1990-04-06 2000-01-11 La Jolla Cancer Research Foundation Method and composition for treating thrombosis
WO2001022922A2 (en) 1999-09-27 2001-04-05 The Regents Of The University Of California Engineering antibodies that bind irreversibly

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2148299B (en) * 1983-09-01 1988-01-06 Hybritech Inc Antibody compositions of therapeutic agents having an extended serum half-life
US4722892A (en) * 1984-08-31 1988-02-02 Meares Claude F Monoclonal antibodies against metal chelates
US5215889A (en) * 1988-11-18 1993-06-01 The Regents Of The University Of California Catalytic and reactive polypeptides and methods for their preparation and use
US5162218A (en) * 1988-11-18 1992-11-10 The Regents Of The University Of California Conjugated polypeptides and methods for their preparation
US5612034A (en) * 1990-10-03 1997-03-18 Redcell, Inc. Super-globuling for in vivo extended lifetimes
JP2579730B2 (en) * 1993-01-22 1997-02-12 株式会社ディ・ディ・エス研究所 Peptide-lipid derivatives and liposomes
US5780426A (en) * 1995-06-07 1998-07-14 Ixsys, Incorporated Fivemer cyclic peptide inhibitors of diseases involving αv β3
US5767071A (en) * 1995-06-07 1998-06-16 Ixsys Incorporated Sevenmer cyclic peptide inhibitors of diseases involving αv β3
US6008213A (en) * 1995-06-29 1999-12-28 Smithkline Beecham Corporation Integrin receptor antagonists
AU713676B2 (en) * 1996-01-16 1999-12-09 Merck & Co., Inc. Integrin receptor antagonists
AU727722B2 (en) * 1996-10-25 2000-12-21 Kyowa Hakko Kogyo Co. Ltd. Radicicol derivatives
US6045774A (en) * 1997-01-10 2000-04-04 Epicyte Pharmaceutical Inc. J chain polypeptide targeting molecule linked to an imaging agent
CA2284726A1 (en) * 1997-04-11 1998-10-22 G.D. Searle & Co. Antagonistic anti-avb3 integrin antibodies
WO1998052966A1 (en) * 1997-05-19 1998-11-26 The Johns Hopkins University School Of Medecine Tissue specific prodrug
US6096725A (en) * 1997-07-02 2000-08-01 Neose Technologies, Inc. Methods of using αGal oligosaccharides as immune system targeting agents
US6251392B1 (en) * 1997-10-20 2001-06-26 Epicyte Pharmaceuticals, Inc. Epithelial cell targeting agent
AU3545399A (en) * 1998-04-08 1999-10-25 G.D. Searle & Co. Dual avb3 and metastasis-associated receptor ligands
WO2000069472A2 (en) * 1999-05-14 2000-11-23 Boehringer Ingelheim Pharmaceuticals, Inc. Enzyme-activated anti-tumor prodrug compounds
US6365619B1 (en) * 1999-07-22 2002-04-02 Novartis Ag Treatment of arteriosclerosis
DE19939980A1 (en) * 1999-08-24 2001-03-01 Merck Patent Gmbh Inhibitors of the integrin alphavbeta¶3¶
CA2405299C (en) * 2000-03-31 2014-07-22 Purdue Research Foundation Method of treatment using ligand-immunogen conjugates
CN100560131C (en) * 2001-10-22 2009-11-18 斯克里普斯研究学院 Antibody targeting compounds

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5114851A (en) 1989-08-29 1992-05-19 Duke University Light activated acyl-enzymes
US5218137A (en) 1989-08-29 1993-06-08 Duke University Light activated acyl-enzymes
US5498729A (en) 1989-12-26 1996-03-12 Domb; Abraham J. Prodrug compositions
US6013625A (en) 1990-04-06 2000-01-11 La Jolla Cancer Research Foundation Method and composition for treating thrombosis
WO2001022922A2 (en) 1999-09-27 2001-04-05 The Regents Of The University Of California Engineering antibodies that bind irreversibly

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
"Goodman And Gilman's: The Pharmacological Bases of Therapeutics", 1990, PERGAMON PRESS
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING CO.
BRUEHL ET AL., BIOCHEMISTRY, vol. 40, 2001, pages 5964 - 5971
BRUNING ET AL., HUM. GENE THER, vol. 12, 2001, pages 391 - 399
CHATTOPADHYAY, CHATTERJEE, J. EXP. CLIN. CANCER RES., vol. 20, 2001, pages 269 - 275
FRANCIS ET AL., INT. J. HEMATOL., vol. 68, 1998, pages 1 - 18
KATRE ET AL., J. IMMUNOL., vol. 144, 1990, pages 209 - 213
NAGY ET AL., PROC NATL. ACAD. SCI., vol. 93, 1996, pages 2464 - 2469
RIECHMANN ET AL., NATURE, vol. 332, 1988, pages 323 - 327
SCHALLY, NAGY, EUR. J. ENDOCRINOLOGY, vol. 141, 1999, pages 1 - 14
See also references of EP1446418A4
SHABAT ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 96, 1999, pages 6925 - 6930
SHABAT ET AL., PROC. NATL. ACAD. SCI. U.S.A., vol. 98, 2001, pages 7528 - 7533

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1443963A4 (en) * 2001-10-22 2007-09-19 Scripps Research Inst Antibody targeting compounds
EP1443963A2 (en) * 2001-10-22 2004-08-11 The Scripps Research Institute Antibody targeting compounds
EP2428226A1 (en) * 2001-10-22 2012-03-14 The Scripps Research Institute Antibody targeting compounds
US8865149B2 (en) 2002-12-31 2014-10-21 Nektar Therapeutics Polymeric reagents comprising a ketone or a related functional group
US7208145B2 (en) 2002-12-31 2007-04-24 Nektar Therapeutics Al, Corporation Polymeric reagents comprising a ketone or a related functional group
US8106131B2 (en) 2002-12-31 2012-01-31 Nektar Therapeutics Hydrolytically stable maleimide-terminated polymers
US8227555B2 (en) 2002-12-31 2012-07-24 Nektar Therapeutics Hydrolytically stable maleimide-terminated polymers
US9132188B2 (en) 2009-02-10 2015-09-15 The Scripps Research Institute Chemically Programmed Vaccination
US8518927B2 (en) 2009-02-10 2013-08-27 The Scripps Research Institute Chemically programmed vaccination
WO2010093706A3 (en) * 2009-02-10 2011-11-10 The Scripps Research Institute Chemically programmed vaccination
WO2014124227A1 (en) * 2013-02-07 2014-08-14 Immunomedics, Inc. Pro-drug form (p2pdox) of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer
US8877202B2 (en) 2013-02-07 2014-11-04 Immunomedics, Inc. Pro-drug form (P2PDOX) of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer
US9095628B2 (en) 2013-02-07 2015-08-04 Immunomedics, Inc. Pro-drug form (P2PDOX) of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer
US9283286B2 (en) 2013-02-07 2016-03-15 Immunomedics, Inc. Pro-drug form (P2PDox) of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer
US9486536B2 (en) 2013-02-07 2016-11-08 Immunomedics, Inc. Pro-drug form (P2PDOX) of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer
US9694088B2 (en) 2013-02-07 2017-07-04 Immunomedics, Inc. Pro-drug form (P2PDox) of the highly potent 2-pyrrolinodoxorubicin conjugated to antibodies for targeted therapy of cancer
US11571480B2 (en) 2015-08-11 2023-02-07 Coherent Biopharma I, Limited Multi-ligand drug conjugates and uses thereof
CN109503700A (en) * 2017-09-14 2019-03-22 南京安吉生物科技有限公司 The blood vessel formation inhibitor IIM 3-1 of maleimide base group modification and its application

Also Published As

Publication number Publication date
AU2002337954B2 (en) 2008-05-29
KR20040048982A (en) 2004-06-10
CN1606566A (en) 2005-04-13
AU2002337954C1 (en) 2008-10-23
WO2003034995A3 (en) 2003-09-12
KR101085784B1 (en) 2011-11-25
JP2005532252A (en) 2005-10-27
CA2464271A1 (en) 2003-05-01
EP1446418A2 (en) 2004-08-18
ES2384235T3 (en) 2012-07-02
EP1446418B1 (en) 2012-05-23
US20030129188A1 (en) 2003-07-10
EP1446418A4 (en) 2006-05-17
JP5021152B2 (en) 2012-09-05

Similar Documents

Publication Publication Date Title
EP1446418B1 (en) Integrin targeting compounds
AU2002337954A1 (en) Integrin targeting compounds
CN100560131C (en) Antibody targeting compounds
JP3592711B2 (en) Cytotoxic drug treatment
ES2352180T3 (en) MONOCLONAL ANTIBODY OF HUMAN ANTI-TENASCINE.
CN101663317A (en) glucagon-like protein-1 receptor (glp-1r) agonist compounds
CN112601522A (en) antibody-ALK 5 inhibitor conjugates and uses thereof
US20110142756A1 (en) Method And Composition For The Treatment Of Cancer By The Enzymatic Conversion Of Soluble Radioactive Toxic Precipitates In The Cancer
JP5191036B2 (en) Anti-human tenascin monoclonal antibody
CN114269388A (en) antibody-ALK 5 inhibitor conjugates and uses thereof
WO2004091542A2 (en) Nitrogen containing integrin targeting compounds
JP2023509760A (en) ALK5 inhibitor complexes and uses thereof
AU2002365182B2 (en) Antibody targeting compounds
Wang et al. Small Molecule–Drug Conjugates Emerge as a New Promising Approach for Cancer Treatment
KR20220082846A (en) B-lymphocyte-specific amatoxin antibody conjugate
Vaklavas et al. CDX-011 (Glembatumumab Vedotin, CR011-vcMMAE)

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR IE IT LU MC NL PT SE SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2464271

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2002337954

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2003537564

Country of ref document: JP

Ref document number: 1020047005978

Country of ref document: KR

Ref document number: KR

WWE Wipo information: entry into national phase

Ref document number: 2002773859

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 20028255887

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2002773859

Country of ref document: EP