WO2000009143A1 - Ligands multivalents du recepteur de avb3 et des recepteurs associes aux metastases - Google Patents

Ligands multivalents du recepteur de avb3 et des recepteurs associes aux metastases Download PDF

Info

Publication number
WO2000009143A1
WO2000009143A1 PCT/US1999/004296 US9904296W WO0009143A1 WO 2000009143 A1 WO2000009143 A1 WO 2000009143A1 US 9904296 W US9904296 W US 9904296W WO 0009143 A1 WO0009143 A1 WO 0009143A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
gly
seq
group
amino
Prior art date
Application number
PCT/US1999/004296
Other languages
English (en)
Inventor
Kam F. Fok
Foe S. Tjoeng
Original Assignee
G.D. Searle & Co.
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 G.D. Searle & Co. filed Critical G.D. Searle & Co.
Priority to EP99916118A priority Critical patent/EP1104304A1/fr
Priority to AU34498/99A priority patent/AU3449899A/en
Priority to CA002338283A priority patent/CA2338283A1/fr
Priority to JP2000564645A priority patent/JP2002522504A/ja
Publication of WO2000009143A1 publication Critical patent/WO2000009143A1/fr

Links

Classifications

    • 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
    • A61K47/6425Drug-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 the peptide or protein in the drug conjugate being a receptor, e.g. CD4, a cell surface antigen, i.e. not a peptide ligand targeting the antigen, or a cell surface determinant, i.e. a part of the surface of a cell
    • 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
    • A61K47/642Drug-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 the peptide or protein in the drug conjugate being a cytokine, e.g. IL2, chemokine, growth factors or interferons being the inactive part of the conjugate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to pharmaceutical compounds which are multivalent a v D3 receptor/metastasis-associated receptor ligands.
  • the use of these multivalent ligands alone or in conjunction with other agents in pharmaceutical compositions, and in methods for treating conditions mediated by a v D3 for the treatment of cancer and other angiogenic diseases, such diabetic retinopathy, arthritis, hemangiomas, and psoriasis, are also disclosed.
  • Angiogenesis the growth of new blood vessels, plays an important role in cancer growth and metastasis.
  • the extent of vasculature in a tumor has been shown to correlate with the patient prognosis for a variety of cancers (Folkman, J., Seminars in Medicine of the Beth Israel Hospital, Boston 333(26): 1757-1763, 1995; Gasparini, G., European Journal of Cancer 32A(14): 2485-2493, 1996; Pluda, J. ML, Seminars in Oncology 24(2): 203-218, 1997).
  • angiogenesis is limited to well controlled situations, such as wound healing and the female reproductive system (Battegay, E.J., J Mol Med 73:-333-346, 1995; Dvorak, H.F, New Engl J Med, 315: 1650-1659, 1986).
  • angiogenesis The control of angiogenesis is thought to be a balance between factors that promote new vessel formation and anti-angiogenic factors that suppress the formation of a neovasculature (Bouck, N. et al., Advances in Cancer Research 69: 135-173, 1996; O'Reilly et al., Cell 79(2): 315-328, 1994).
  • Disruptions in these forces, through alterations in cell adhesion factors, are implicated in a variety of disorders, including cancer, stroke, osteoporosis, restenosis, and rheumatoid arthritis (A. F. Horwitz, Scientific American, 276(5): 68-75, 1997).
  • Integrins are a large family of cell surface glycoproteins which mediate cell adhesion and play central roles in many adhesion phenomena. Integrins are heterodimers composed of noncovalently linked ⁇ and ⁇ polypeptide subunits. Currently eleven different subunits have been identified and six different ⁇ subunits have been identified. The various ⁇ subunits can combine with various ⁇ subunits to form distinct integrins.
  • v b3 integrin receptor
  • vitronectin receptor integrin receptor
  • a v b 3 integrins can promote the formation of blood vessels (angiogenesis) in tumors. These vessels nourish the tumors and provide access routes into the bloodstream for metastatic cells.
  • the a v b3 integrin is also known to play a role in various other disease states or conditions including tumor metastasis, solid tumor growth (neoplasia), osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, angiogenesis, including tumor angiogenesis, retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, and smooth muscle cell migration (e.g., restenosis).
  • Tumor cell invasion occurs by a three step process: 1) tumor cell attachment to extracellular matrix; 2) proteolytic dissolution of the matrix; and 3) movement of the cells through the dissolved barrier. This process can occur repeatedly and can result in metastases at sites distant from the original tumor.
  • a v b3 integrin and a variety of other alpha v-containing integrins bind to a number of Arg-Gly-Asp (RGD)-containing matrix macromolecules.
  • RGD Arg-Gly-Asp
  • Compounds containing the RGD sequence mimic extracellular matrix ligands and bind to cell surface receptors. Fibronectin and vitronectin are among the major binding partners of a v b3 integrin.
  • Other proteins and peptides have also bind the a v b3 ligand. These include the disintegrins (M. Pfaff et al., Cell Adhes. Commun. 2(6): 491-501, 1994), peptides derived from phage display libraries (Healy, J.M.
  • the monoclonal antibody LM609 is also an a v b 3 integrin antagonist (D.A. Cheresh et al., J. Biol. Chem., 262(36): 17703-17711, 1987).
  • Avb ⁇ inhibitors are being developed as potential anti-cancer agents. Compounds that impair endothelial cell adhesion via the a v b3 integrin induce improperly proliferating endothelial cells to die. Avb ⁇ inhibitors can also help patients suffering from proliferative retinopathy, a complication of diabetes in which the retina sprouts weak and leaky blood vessels that can destroy the retina and causes blindness, or from restenosis, a process in which blood vessels in many patients can become occluded again over a period of time after the balloon angioplastic surgery.
  • Osteoporosis is a disorder involving loss of bone and increased risk of bone fracture, that occurs with age, particularly in women. This disease can result from the overactivity of osetoclast cells that bind to bone and degrade bone. The osteoclast binding occurs via the a v b3 integrin. Specific inhibitors that shield a v b3 integrins can prevent the destructive cells from adhering to bone. Additionally, it has been found that such agents can be useful as antivirals, antifungals and antimicrobials. Thus, compounds which selectively inhibit or antagonize a v b3 can be beneficial for treating such conditions.
  • the avb ⁇ integrin has been shown to play a role in melanoma cell invasion (Seftor et al., Proc. Natl. Acad. Sci. USA, 89: 1557-1561, 1992).
  • the a v b 3 integrin expressed on human melanoma cells has also been shown to promote a survival signal, protecting the cells from apoptosis (Montgomery et al., Proc. Natl. Acad. Sci. USA, 91: 8856-8860, 1994).
  • the adhesion receptor identified as integrin a v b3 is a marker of angiogenic blood vessels in chick and man. This receptor plays a critical role in angiogenesis or neovascularization.
  • Angiogenesis is characterized by the invasion, migration and proliferation of smooth muscle and endothelial cells by new blood vessels. Antagonists of a v b3 inhibit this process by selectively promoting apoptosis of cells in the neovasculature. The growth of new blood vessels, also contributes to pathological conditions such as diabetic retinopathy (Adonis et al., Amer. J.
  • a v b3 antagonists can be useful therapeutic targets for treating such conditions associated with neovascularization (Brooks et al., Science, 264: 569-571, 1994).
  • the avb ⁇ cell surface receptor is also the major integrin on osteoclasts responsible for the attachment to the matrix of bone. Osteoclasts cause bone resorption and when such bone resorbing activity exceeds bone forming activity, osteoporosis (a loss of bone) results, which leads to an increased number of bone fractures, incapacitation and increased mortality. Antagonists of a v b3 have been shown to be potent inhibitors of osteoclastic activity both in vitro (Sato et al., J. Cell. Biol , 111: 1713-1723, 1990) and in vivo (Fisher et al., Endocrinology, 132: 1411-1413, 1993).
  • Antagonism of a v b3 leads to decreased bone resorption and therefore assists in restoring a normal balance of bone forming and resorbing activity.
  • antagonists of osteoclast a v b3 which are effective inhibitors of bone resorption and therefore are useful in the treatment or prevention of osteoporosis.
  • avb ⁇ integrin in smooth muscle cell migration also makes an a v b3 a therapeutic target for prevention or inhibition of neointimal hyperplasia which is a leading cause of restenosis after vascular procedures (Choi et al., J.
  • PCT Int. Appl. WO 97/08145 by Sikorski et al. discloses meta-guanidine, urea, thiourea or azacyclic amino benzoic acid derivatives as highly specific a v b3 integrin antagonists.
  • PCT Int. Appl. WO 96/00574 Al 960111 by Cousins, R.D. et al., describe preparation of 3-oxo-2,3,4,5-tetrahydro-lH-l,4-benzodiazepine and -2-benzazepine derivatives and analogs as vitronectin receptor antagonists.
  • Selected compounds were shown to bind to human integin a v b3 with EIB ⁇ 1000 nM and claimed as compds. useful for inhibiting the binding of fibrinogen to blood platelets and for inhibiting the aggregation of blood platelets.
  • PCT Int. Appl. WO 96/16983 Al 960606. by Vuori, K. and Ruoslahti, E. describe cooperative combinations of a b3 integrin ligand and second ligand contained within a matrix, and use in wound healing and tissue regeneration.
  • the compounds contain a ligand for the a v b3 integrin and a ligand for the insulin receptor, the PDGF receptor, the IL-4 receptor, or the IGF receptor, combined in a biodegradable polymeric (e.g. hyaluronic acid) matrix.
  • PCT Int. Appl. WO 97/10507 Al 970320 by Ruoslahti, E; and Pasqualini, R. describe peptides that home to a selected organ or tissue in vivo, and methods of identifying them.
  • a brain-homing peptide nine amino acid residues long, for example, directs red blood cells to the brain.
  • Thomas S. describes bifunctional ligands for specific tumor inhibition by blood coagulation in tumor vasculature.
  • the disclosed bispecific binding ligands bind through a first binding region to a disease-related target cell, e.g. a tumor cell or tumor vasculature; the second region has coagulation-promoting activity or is a binding region for a coagulation factor.
  • the disclosed bispecific binding ligand may be a bispecific (monoclonal) antibody, or the two ligands may be connected by a (selectively cleavable) covalent bond, a chemical linking agent, an avidin-biotin linkage, and the like.
  • the target of the first binding region can be a cytokine- inducible component, and the cytokine can be released in response to a leukocyte- activating antibody; this may be a bispecific antibody which crosslinks activated leukocytes with tumor cells.
  • Interferon alpha is a family of highly homologous, species- specific proteins that possess complex antiviral, antineoplastic and immunomodulating activities (Extensively reviewed in the monograph "Antineoplastic agents, interferon alfa," American Society of Hospital Pharmacists, Inc., 1996, LEXIS file GENMED [General Medicine], DIF [Drug Information file]). Interferon alpha also has anti-proliferative, and anti-angiogenic properties, and has specific effects on cellular differentiation (Sreevalsan, in "Biologic Therapy of Cancer", pp. 347-364, (eds. V.T. DeVita Jr., S. Hellman, and S.A. Rosenberg), J.B. Lippincott Co, Philadelphia, PA, 1995).
  • Interferon alpha is effective against a variety of cancers including hairy cell leukemia, chronic myelogenous leukemia, malignant melanoma, and Kaposi's sarcoma.
  • the precise mechanism by which IFN alpha exerts its anti-tumor activity is not entirely clear, and may differ based on the tumor type or stage of disease.
  • the anti-proliferative properties of IFN alpha which may result from the modulation of the expression of oncogenes and/or proto-oncogenes, have been demonstrated on both tumor cell lines and human tumors growing in nude mice (Gutterman, J. U., Proc. Natl. Acad. Sci., USA 91: 1198-1205, 1994).
  • Interferon is also considered an anti-angiogenic factor, as demonstrated through the successful treatment of hemangiomas in infants (Ezekowitz et al, N. Engl. J. Med., May 28, 326(22) 1456-1463, 1992) and the effectiveness of IF ⁇ alpha against Kaposi's sarcoma (Krown, Semin Oncol 14(2 Suppl 3): 27-33, 1987).
  • the mechanism underlying these anti-angiogenic effects is not clear, and may be the result of IF ⁇ alpha action on the tumor (decreasing the secretion of pro-angiogenic factors) or on the neovasculature.
  • IF ⁇ receptors have been identified on a variety of cell types ( ⁇ avarro et al., Modern Pathology 9(2): 150-156, 1996). A general review on the structure and function of type I and type II interferon receptors has been recently published (Pestka, S., Semin. Oncol. 24(3, Suppl. 9), 18-40, 1997).
  • United States Patent 4,530,901 to Weissmann describes the cloning and expression of IF ⁇ alpha-type molecules in transformed host strains.
  • United States Patent 4,503,035, Pestka describes an improved processes for purifying 10 species of human leukocyte interferon using preparative high performance liquid chromatography.
  • United States Patent 5,231,176 to Goeddel describes the cloning of a novel distinct family of human leukocyte interferons containing in their mature form greater than 166 and no more than 172 amino acids.
  • Physiologically-active immunological interferon-polyethylene glycol conjugates have been described that appear to have improved circulating half-life, water solubility and immunological properties (see, for example, U.S. 4,179,337).
  • Long-acting alpha-interferon compositions containing 1-4 polyalkene oxide moieties conjugated to interferon U.S. 5,711,944
  • methods for preparing mono- and bis-interferon polymer (polyethylene glycol) conjugates U.S. 5,738,846)
  • interferon alpha polyethylene glycol conjugates containing a polymer moiety from 300 to 30,000 daltons, for example have been described (U.S. 5,595,732).
  • proteins purified from recombinant organisms are comprised of the 20 naturally-occurring L-amino acids, occasionally modified at certain positions by post-translational processing.
  • Fragment condensation where two protected peptides are prepared, purified, and then coupled to form a longer peptide, is an alternative method (H. Kuroda et al.,
  • an unprotected peptide with C-terminal carboxylthioester can be linked to the N-terminus of the N-terminal cysteine containing unprotected peptide selectively (M. Schnolzer, and S. B. H. Kent, Science 256: 221, 1992; C. F Liu, and J. P. Tarn, J. Am. Chem. Soc. 116: 4149, 1994).
  • Several biologically active peptides and proteins have been prepared (L. E. Canne et al., J. Am. Chem. Soc. 117: 2998, 1995). This method is not limited to peptide ligation and has been used for preparation of cyclic proteins or peptides (L. Zhang and J. P. Tarn, J. Am. Chem. Soc. 119: 2363, 1997).
  • Another semisynthetic method of assembling proteins involves the chemoselective addition of a peptide to a recombinant protein (Muir et al., Proc. Natl. Acad. Sci. USA 95: 6705-6710, 1998).
  • a thioester generated in the C-terminus of recombinant tyrosine kinase C-terminal Src kinase (Csk) was ligated to a synthetic phosphotyrosine peptide containing an N-terminal cysteine.
  • This method permits the introduction of unnatural amino acids, biophysical probes, and post-translational modifications into proteins of any size, overcoming the -15 kDa upper limit currently reached by the chemoselective method.
  • v b3 antagonists conjugated to a metastasis-associated receptor ligand can have improved biological properties by acting through several mechanisms.
  • the two moieties can have distinct mechanisms, permitting synergy or increased efficacy, or both, as a result of blocking the angiogenic process at two separate points.
  • the two moieties may act on the same type of cell, resulting in an increased binding, or other action, by virtue of increased avidity.
  • Each moiety in this case participates in the targeting the other moiety to the appropriate site of action. Dimers or higher order multimers of these moieties with themselves or other chemical groups, including proteins, can have increased efficacy or potency, or both, by virtue of either of these mechanisms.
  • Avb3 bioconjugates can have improved activities due to the ability to bind to two distinct receptors on the same cell type and thus demonstrate improved activity due to interactions with receptors on a single cell. These a v b3 bioconjugates can have improved therapeutic properties through a variety of mechanisms such as: (1) alterations in the overall on- or off-rates or Ka or Kd of the ligand(s) on the target cell, (2) activation or blockade of complementary receptor signaling pathways, and/or (3) more specific targeting of one or both of the components to the cell of interest.
  • a v b3 bioconjugates are expected to possess a unique pharmacokinetic distribution and clearance profile (Dehmer et al., Circulation, 91, 2188-2194, 1995; Tanaka et al., Nature Medicine, 3, 437-442, 1997).
  • a v b3 bioconjugates can also have improved properties in vivo, compared to the two components individually, as a result of alterations in biodistribution or half-life.
  • the improved properties can also result from the binding of the a v b3 bioconjugate to one or more of the receptors, pharmacokinetics, or uptake of the bioconjugate is altered in a favorable manner. These moieties are likely to act through complementary mechanisms. Therefore, bioconjugates containing anti- proliferative and anti-angiogenic activities can provide improved anti-tumor activity by using two distinct mechanisms to decrease tumor growth.
  • the anti- proliferative moiety can act directly on the tumor to decrease its growth whereas the anti-angiogenic factor acts indirectly by preventing the growth of the neovasculature required for rapid tumor growth.
  • Avb ⁇ bioconjugates illustrate one example of a compound by this mechanism.
  • a chemoselective ligation method can be used to conjugate the a v b3 antagonists to the protein to improve the antitumor properties of interferon alpha.
  • Small RGD-containing peptides or peptidomimetic a v b3 antagonists can also be conjugated to interferon by different linking methods (G.T.
  • the present invention relates to bioconjugates comprising one or more a v b3 antagonist moieties coupled to an amide or to a metastasis-associated receptor ligand by a covalent bond or by a linear or branched linker.
  • the a v b3 bioconjugates are prepared by conjugating an a v b3 integrin antagonist with a peptide or a polypeptide, processes for their preparation, pharmaceutical compositions containing them, and methods for their use.
  • a compound contemplated by this invention is represented by the group formulas I, II, III, IV, V, VI, and VII consisting of:
  • a 1 separately or in combination with L 1 is an a v b3 antagonist
  • T 1 is selected from the group consisting of metastasis-associated receptor ligands and amides,
  • L 1 is a covalent bond or a linker that covalently bonds A 1 to R 2 , R 4 , R 6 , or R 8 , R 2 , R 4 , R 6 , and R 8 are branched linkers,
  • R 3 , R 5 , and R 7 are covalent bonds or linkers that covalently bond R 2 to R 4 , R 4 to R 6 , and R 6 to R 8 , respectfully, and
  • R 1 is a covalent bond or a linker that covalently bonds R 2 to T 1 .
  • the ligand portion of a compound is an agonist.
  • the ligand portion of a compound can also be an antagonist.
  • T 1 is a polypeptide. More preferably, the polypeptide is selected from the group consisting of natural cytokines, synthetic cytokines, and anti-angiogenic proteins.
  • the anti-angiogenic protein is selected from the group consisting of angiostatin and endostatin.
  • the polypeptide is selected from the group consisting of interleukin-2, interleukin-7, interleukin-12, interleukin-15, interferons and progenipoietin-G, erythropoietin, erythropoietin receptor agonists, colony stimulating factors, and hematopoietic growth factors.
  • the cytokine is human interferon.
  • the interferon is interferon alpha. Most preferably, wherein the interferon alpha is interferon alpha 2b. Most preferably, the interferon alpha is interferon alpha A/D hybrid.
  • polypeptide is selected from the group consisting of SEQ ID NO: 1
  • SEQ ID NO: 15 SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 24, SEQ ID NO: 25, SEQ ID NO: 26, SEQ ID NO: 27, and SEQ ID NO: 28.
  • L 1 is a peptide linker. More preferably the peptide linker is a peptide ranging in length from 2 through 10 amino acids.
  • the peptide linker is Gly-Asp
  • the peptide linker is Gly-Asp-L 2 , wherein; L 2 is selected from the group consisting of Ala, Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, He, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Try, Val. Even more preferably, L 2 is selected from the group consisting of Ala, Ser, Val.
  • the peptide linker is one or more peptide sequences selected from the group consisting of; -Gly-Asp-Ala- (SEQ ID NO: 29); -Gly-Asp-Ser- (SEQ ID NO: 30); -Gly-Gly-Gly-Gly-Ala- (SEQ ID NO: 31); -Gly-Gly-Gly-Gly-Ser- (SEQ ID NO: 32); -Gly-Asp-Ala-Gly-Gly-Gly-Gly-Ala- (SEQ ID NO: 33); -Gly-Asp-Ala-Gly-Gly-Gly-Gly-Ser- (SEQ ID NO: 34); -Gly-Asp- Ala-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Gly- Ser- (SEQ ID NO: 35); and -Gly-Asp- Ala-Gly-
  • L 1 is a peptide linker, said peptide linker is one or more peptide sequences selected from the group consisting of GDA (SEQ ID NO: 29); GDS (SEQ ID NO: 30); GDSLA (SEQ ID NO: 37) GDSGA (SEQ ID NO: 38) GDSGGGGA (SEQ ID NO: 39); GDSGGGGGA(SEQ ID NO: 40); GDSGGGGAS (SEQ ID NO: 41);GDS(GGGGS) 2 ; (SEQ ID NO: 42); and GDS(GGGG) 4 ; (SEQ ID NO: 43).
  • GDA SEQ ID NO: 29
  • GDS SEQ ID NO: 30
  • GDSLA SEQ ID NO: 37
  • GDSGA SEQ ID NO: 38
  • GDSGGGGA SEQ ID NO: 39
  • GDSGGGGAS SEQ ID NO: 41
  • GDS(GGGGS) 2 ;
  • SEQ ID NO: 42 and G
  • R 2 , R 4 , R 6 , and R 8 are branched linkers. More preferably the branched linker is lysine.
  • R 1 is a covalent bond or a linker that covalently bonds R 2 to T 1 and R 1 is selected from the group consisting of peptides and amino-alkyl carboxylic acids.
  • R 1 is a covalent bond or a linker that covalently bonds R 2 to T 1 and R 1 is selected from the group consisting of
  • AGAGA-C 0-S-CH 2 CH 2 CONH 2
  • AGAGA SEQ ID NO 40
  • GAGAG ( SEQ ID NO 43) .
  • GAGAG ( SEQ ID NO 4 ) .
  • a 1 is selected from the group consisting of ,
  • T 1 is selected from the group consisting of amides and metastasis-associated receptor ligands, or peptide fragments thereof.
  • a 1 is a compound of the formula XI,
  • X is C or N
  • R 4 is one or more substituents independently selected from the group consisting of hydrogen, alkyl, hydroxy, alkoxy, aryloxy, halogen, haloalkyl, haloalkoxy, nitro, amino, alkylamino, acylamino, dialkylamino, cyano, alkylthio, alkylsulfonyl, carboxyl moieties, trihaloacetamide, acetamide, aryl, fused aryl, cycloalkyl, thio, monocyclic heterocycle, and fused monocyclic heterocycle;
  • R is R", R 12 , or R 13 , wherein
  • Y is O or S
  • R 8 is a substituent independently selected from the group of the following substituents:
  • halogen haloalkyl, lower alkyl, alkoxy, methylenedioxy, ethylenedioxy, alkylthio, haloalkylthio, mercapto, hydroxy, cyano,
  • R 13 is
  • R 7 is hydrogen or hydroxyl
  • R 5 and R 6 are each substituents independently selected from the group consisting of hydrogen, lower alkyl, hydroxy, alkoxy, halogen, phenyl, amino, carboxyl or carboxyl ester, and fused phenyl;
  • n is an integer 1, 2, 3,or 4.
  • X is C
  • R 4 is one or more substituents independently selected from the group 15 consisting of hydrogen, hydroxy, halogen, and haloalkyl.
  • R 13 is
  • R 4 is one or more substituents independently selected from the group consisting of hydrogen, hydroxy, halogen, and haloalkyl.
  • R 7 is hydrido or hydroxyl
  • R 5 and R 6 are one or more substituents independently selected from the group consisting of hydrogen, lower alkyl, and hydroxy;
  • A is nitrogen
  • n is an integer 1 or 2.
  • a 1 is a compound of the formula XIII,
  • R is selected from R», R ⁇ R"; wherein
  • the a v b3 bioconjugate is selected from the group consisting of Compound 100, Compound 101, Compound 102, Compound 103, Compound 201, Compound 202, Compound 203, Compound 204, Compound 205, Compound 206, Compound 207, Compound 208, Compound 209, Compound 210, Compound 211, Compound 212.
  • a 1 is a compound of the formula XIV,
  • J 1 is selected from the group consisting of Gly-Asp-Ala- NH 2 , and Gly-Asp-Ser-NH 2 .
  • R 4 is one or more substituents independently selected from the group consisting of hydrogen, hydroxy, halogen, and haloalkyl.
  • R 7 is hydrogen or hydroxyl
  • R 5 and R 6 are one or more substituents independently selected from the group consisting of hydrogen, lower alkyl, hydroxy;
  • A is nitrogen
  • n is an integer 1 or 2.
  • the a v b3 bioconjugate is selected from the group consisting of Compound 301, Compound 302, Compound 303, Compound 304, Compound 305, Compound 306.
  • the present invention relates to recombinant expression vectors comprising nucleotide sequences encoding the metastasis-associated receptor proteins, related microbial and eukaryotic expression systems, and processes for making the a v b3 precursors and the metastasis proteins.
  • the invention also relates to pharmaceutical compositions containing the a v b3 bioconjugates, and methods for using the a v b3 bioconjugates, including use of the compounds for the manufacture of a medicament for therapeutic application to inhibit tumor growth.
  • Benefits of the invention are the provision of a pharmaceutical composition comprising a compound of the formulas I-VII. Such compounds and compositions are useful in inhibiting or antagonizing the ⁇ v ⁇ integrin and in targeting angiogenesis.
  • Another embodiment of the present invention relates to a method of selectively inhibiting or antagonizing the ⁇ v ⁇ 3 integrin and tumor angiogenesis.
  • the invention further contemplates treating a disease or inhibiting pathological conditions associated selected from the group consisting of osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, thrombosis, angina, atherosclerosis, smooth muscle cell migration and restenosis in a mammal in need of such treatment.
  • a disease or inhibiting pathological conditions associated selected from the group consisting of osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis,
  • compositions comprising a therapeutically-effective amount of an avb ⁇ bioconjugate in admixture with a pharmaceutically-acceptable carrier are contemplated.
  • the composition can further comprise one or more of the following: an adjunctive agent, a chemotherapeutic agent, and an immunotherapeutic agent.
  • the invention contemplates a process for treating a human patient with an angiogenesis-mediated disease, by administering an effective amount of an avb ⁇ bioconjugate.
  • the angiogenesis-mediated disease is selected from the group consisting of cancer, arthritis, and macular degeneration.
  • the invention contemplates a process for treating cancer comprising administering to a mammalian host suffering therefrom a therapeutically effective amount of a bioconjugate in unit dosage form. It also contemplates a process of inhibiting elevated levels of tumor antigens comprising administering to a host in need thereof a therapeutically effective amount of an a v b3 bioconjugate in unit dosage form. It also contemplates a process of modulating tumors in a patient comprising administering an angiogenesis-inhibiting effective amount of a bioconjugate to such a patient.
  • the invention contemplates a process of treating inhibiting the proliferation of tumor cells in a patient comprising administering an angiogenesis- inhibiting effective amount of an avb ⁇ bioconjugate to said patient.
  • the tumor cells are selected from the group consisting of lung cancer, breast cancer, ovarian cancer, prostate cancer, pancreatic cancer, gastric cancer, colon cancer, renal cancer, bladder cancer, melanoma, hepatoma, sarcoma, and lymphoma.
  • the invention contemplates a process for the treatment of a patient with a solid tumor, that comprises the steps of: (a) administering an effective dose of angiogenesis-inhibiting effective amount of the a v b3 bioconjugate to a patient in a pharmaceutically acceptable vehicle; and (b) maintaining said patient for a time period sufficient to cause a reduction in tumor size.
  • steps (a) and (b) are repeated.
  • BH3-THF borane-tetrahydrofuran complex
  • CH3CN acetonitrile
  • CH3I iodomethane
  • CHNCl analysis carbon/hydrogen/nitrogen/chlorine elemental analysis
  • CHNS analysis carbon/hydrogen/nitrogen/sulfur elemental analysis
  • DCC 1,3-dicyclohexylcarbodiimide
  • DIBAL diisobutylaluminum hydride
  • EDCl l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • FAB MS fast atom bombardment mass spectroscopy
  • g gram(s)
  • GIHA et ⁇ -guanidinohippuric acid
  • GIHA HC1 meta-guanidinohippuric acid hydrochloride
  • HBTU 2-(lH-benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate
  • HPLC high performance liquid chromatography
  • IBCF isobutylchloroformate
  • i-Pr iso propyl
  • i-Prop iso propyl
  • K2CO3 potassium carbonate
  • KOH potassium hydroxide
  • KSCN potassium thiocyanate
  • MBHA methoxybenzhydrylamine
  • MCPBA m-chloroperoxybenzoic acid or m-chloroperbenzoic acid
  • NMR nuclear magnetic resonance
  • RPHPLC reverse phase high performance liquid chromatography
  • TBTU 2-Cl-H-benzotriazole-lyl)-l,l,3,3-tetramethyluronium tetrafluoro- borate
  • TFA trifluoroacetic acid
  • THF tetrahydrofuran
  • TMEDA trimethylethylenediamine
  • chemical ligation and “conjugation” mean a chemical reaction which covalently links two similar or dissimilar functional groups together intramolecularly or intermolecularly.
  • peptide linker means a compound which forms a carboxamide bond between two groups having one or more peptide linkages (CONH-) and serves as a connector for the propose of amelioration of the distance or space orientation between two molecules.
  • multi-functional bioconjugate means organic compounds consisting of two or more different types of biomolecules and at least one of the biomolecules has more than one copy of a specific structural or functional moiety.
  • peptide means organic compounds consisting of two or more aminoacyl residues covalently linked by carboxamide functional groups.
  • polypeptide means organic compounds consisting of more than two aminoacyl residues linked by carboxamide functional groups.
  • v ⁇ 3 v ⁇ 3
  • a v b3 v b3
  • alpha v beta 3 alpha v beta 3
  • a v b3 precursor means a compound, separately or in combination with a linker, is an a v b3 antagonist which can be conjugated to other small molecules, peptides, or polypeptides to form an a v b3 bioconjugate.
  • a v b3 bioconjugate means a molecule composed of one or more a v b3 antagonists fused directly, or indirectly through a linker to one or more other small molecules, peptides, or polypeptides, that retains a v b3 antagonist activity.
  • multi-functional protein means a single polypeptide which inherently possesses two distinct activities. In the context of the current invention, anti-angiogenic and/or anti-tumor activities are contemplated.
  • the polypeptide can be formed by the covalent union of two distinct proteins or portions thereof, or two copies of the same protein, or portions thereof.
  • anti-tumor means possessing an activity which slows or abolishes the growth of, or which kills, or otherwise harms tumors in vivo.
  • ligand means a molecule that binds to a receptor to initiate (agonist) or block (antagonist) a response.
  • Ligands may be small natural or synthetic molecules (e.g., neurotransmitters and their analogues) or may be large proteins or protein nucleic acid associates (e.g., viruses).
  • receptor means a component of a cell that interacts specifically with (receives) other molecules and, in appropriate combination, initiates a biological response.
  • Receptors may be protein, lipid, nucleic acid, or carbohydrate. The possess the fundamental property, when combined with the appropriate ligand, of expressing the information content of the receptor or the ligand.
  • endogenous means a cellular component that interacts with a specific receptor.
  • endogenous ligand describes cellular molecules that interact with receptors that may have been defined earlier by synthetic or other approaches. A classic example is the discovery of endogenous opioid peptides for the opiate receptor.
  • mutant sequence refers to an amino acid or nucleic acid sequence which is identical to a wild-type or native form of a gene or protein.
  • mutant amino acid sequence refers to a polypeptide having an amino acid sequence which varies from a native sequence due to amino acid additions, deletions, substitutions, or all three, or is encoded by a nucleotide sequence from an intentionally-made variant derived from a native sequence.
  • interferon includes one of a group of species specific proteins which will induce antiviral and anti-proliferative responses in cells including type I interferon, type I interferon variants, interferon alpha 2a, interferon alpha 2b, interferon alpha hybrid A/D, consensus interferon, functional homologues thereof, and those encoded by a DNA sequences related to those in SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, and SEQ ID NO: 22.
  • composition means a product which results from the mixing or combining of more than one element or ingredient.
  • pharmaceutically-acceptable carrier means a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting a chemical agent.
  • therapeutically-effective amount means that amount of drug or pharmaceutical agent that elicits the biological or medical response of a tissue, system or animal that is being sought by a researcher or clinician.
  • alkyl and “lower alkyl”, refer to a straight chain or branched chain hydrocarbon radical having 1 to about 10 carbon atoms, and 1 to about 6 carbon atoms, respectively.
  • alkyl radicals and lower alkyl radicals are methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec- butyl, t-butyl, pentyl, neopentyl, hexyl, isohexyl, octyl, nonyl, decyl, and the like.
  • alkenyl or “lower alkenyl” refer to unsaturated acyclic hydrocarbon radicals containing at least one double bond and 2 to about 10 carbon atoms, and 2 to about 6 carbon atoms, respectively, which carbon-carbon double bond may have either cis or trans geometry within the alkenyl moiety, relative to groups substituted on the double bond carbons. Examples of such groups are ethenyl, propenyl, butenyl, isobutenyl, pentenyl, hexenyl, octenyl, nonenyl, decenyl, and the like.
  • alkynyl or “lower alkynyl”, refer to acyclic hydrocarbon radicals containing one or more triple bonds and 2 to about 10 carbon atoms, and 2 to about 6 carbon atoms, respectively. Examples of such groups are ethynyl, propynyl, butynyl, pentynyl, hexynyl, octynyl, nonynyl, decynyl, and the like.
  • cycloalkyl as used herein means saturated or partially unsaturated cyclic radicals containing 3 to about 8 carbon atoms and more preferably 4 to about 6 carbon atoms.
  • examples of such cycloalkyl radicals include cyclopropyl, cyclopropenyl, cyclobutyl, cyclopentyl, cyclohexyl, 2-cyclohexen-l-yl, and the like.
  • aryl denotes aromatic ring systems composed of one or more aromatic rings. Preferred aryl groups are those consisting of one, two or three aromatic rings. The term embraces aromatic radicals such as phenyl, pyridyl, naphthyl, thiophene, furan, biphenyl and the like.
  • cyano is represented by a radical of the formula
  • hydroxy and "hydroxyl” as used herein are synonymous and are represented by a radical of the formula -OH.
  • lower alkylene or “alkylene” as used herein refers to divalent linear or branched saturated hydrocarbon radicals of 1 to about 6 carbon atoms.
  • alkoxy refers to straight or branched chain oxy containing radicals of the formula -OR 20 , wherein R 20 is an alkyl group as defined above.
  • alkoxy groups encompassed include methoxy, ethoxy, n- propoxy, n-butoxy, isopropoxy, isobutoxy, sec-butoxy, t-butoxy, octyloxy, nonyloxy, decyloxy, and the like.
  • arylalkyl or “aralkyl” refer to a radical of the formula -R 22 -R 21 wherein R 21 is aryl as defined above and R 22 is an alkylene as defined above.
  • aralkyl groups include benzyl, pyridylmethyl, naphthylpropyl, phenethyl and the like.
  • nitro is represented by a radical of the formula -N0 2 .
  • halogen refers to bromo, chloro, fluoro, or iodo.
  • haloalkyl refers to alkyl groups as defined above substituted with one or more of the same or different halo groups at one or more carbon atom.
  • haloalkyl groups include trifluoromethyl, dichloroethyl, fluoropropyl and the like.
  • carboxyl or “carboxy” refers to a radical of the formula -COOH.
  • carboxyl ester refers to a radical of the formula -COOR 23 wherein R 23 is selected from the group consisting of H, alkyl, alkenyl, alkynyl, aralkyl or aryl as defined above.
  • amino is represented by a radical of the formula -NH
  • alkylsulfonyl As used herein the terms “alkylsulfonyl,” “alkylsulfone,” “alkenylsulfonyl,”
  • alkenylfone alkynylsulfonyl
  • alkynylsulfone refer to a radical of the formula -S0 2 -R 24 wherein R 24 is alkyl, alkenyl, alkynyl, as defined above.
  • alkylthio alkenylthio
  • alkynylthio refers to a radical of the formula -SR 24 wherein R 24 is alkyl, alkenyl, alkynyl as defined above.
  • sulfonic acid refers to a radical of the formula -S0 2 -R 25 wherein R 25 is H, alkyl or aryl as defined above.
  • sulfonamide refers to a radical of the formula -S0 2 -NR 7 R 8 wherein R 7 and R 8 are as defined above.
  • fused aryl refers to an aromatic ring such as the aryl groups defined above fused to one or more phenyl rings. Embraced by the term “fused aryl” is the radical naphthyl.
  • monocyclic heterocycle or “monocyclic heterocyclic” refer to a monocyclic ring containing from 4 to about 12 atoms, and more preferably from 5 to about 10 atoms, wherein 1 to 3 of the atoms are heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur with the understanding that if two or more different heteroatoms are present at least one of the heteroatoms must be nitrogen.
  • monocyclic heterocycles are imidazole, furan, pyridine, oxazole, pyran, triazole, thiophene, pyrazole, thiazole, thiadiazole, and the like.
  • fused monocyclic heterocycle refers to a monocyclic heterocycle as defined above with a benzene fused thereto.
  • fused monocyclic heterocycles include benzofuran, benzopyran, benzodioxole, benzothiazole, benzothiophene, benzimidazole and the like.
  • methylenedioxy refers to the radical -OCH 2 0- and the term “ethylenedioxy” refers to the radical -OCH 2 CH 2 0-.
  • m is 1 or 2 and R 19 is H, alkyl, alkenyl, alkynyl, aryl, or aralkyl, and X is H or halide. More preferably the term refers to a 4-9 membered ring and includes rings such as imidazoline.
  • 5-membered optionally substituted heteroaromatic ring includes for example a radical of the formula
  • 5-membered heteroaromatic ring fused with a phenyl refers to such a “5- membered heteroaromatic ring" with a phenyl fused thereto.
  • Representative of such 5-membered heteroaromatic rings fused with a phenyl is benzimidazole.
  • bicycloalkyl refers to a bicyclic hydrocarbon radical containing 6 to about 12 carbon atoms which is saturated or partially unsaturated.
  • acyl refers to a radical of the formula -COR 26 wherein R 26 is alkyl, alkenyl, alkynyl, aryl or aralkyl and optionally substituted thereon as defined above. Encompassed by such radical are the groups acetyl, benzoyl and the like.
  • thio and mercapto refer to a radical of the formula -SH.
  • sulfonyl refers to a radical of the formula -S0 2 R 27 wherein R 27 is alkyl, alkenyl, alkynyl, aryl, or aralkyl as defined above.
  • haloalkylthio refers to a radical of the formula -SR 28 wherein R 28 is haloalkyl as defined above.
  • aryloxy refers to a radical of the formula -OR 29 wherein R is aryl as defined above.
  • acylamino refers to a radical of the formula
  • R 30 CONH- wherein R 30 is alkyl, alkenyl, alkynyl, aralkyl or aryl as defined above.
  • alkylamino refers to a radical of the formula - NHR 32 wherein R 32 is alkyl as defined above.
  • dialkylamino refers to a radical of the formula - NR 33 R 34 wherein R 33 and R 34 are the same or different alkyl groups as defined above.
  • trifluoromethyl refers to a radical of the formula
  • trifluoroalkoxy refers to a radical of the formula F 3 C-R 35 -0- wherein R 35 is a bond or an alkylene group as defined above.
  • alkylaminosulfonyl refers to a radical of the formula R 36 -NH-S0 2 - wherein R 36 is alkyl as defined above.
  • alkylsulfonylamino refers to a radical of the formula R 36 -S0 2 -NH- wherein R 36 is alkyl as defined above.
  • trifluoromethylthio refers to a radical of the formula F 3 C-S-.
  • trifluoromethylsulfonyl refers to a radical of the formula F 3 C-S0 2 -.
  • 4-12 membered mono-nitrogen-containing monocyclic or bicyclic ring refers to a saturated or partially unsaturated monocyclic or bicyclic ring of 4-12 atoms and more preferably a ring of 4-9 atoms wherein one atom is nitrogen. Such rings may optionally contain additional heteroatoms selected from nitrogen, oxygen or sulfur. Included within this group are morpholine, piperidine, piperazine, thiomorpholine, pyrrolidine, proline, azacycloheptene and the like.
  • benzyl refers to the radical -CH 2 -Ph.
  • phenethyl refers to the radical -CH 2 CH 2 -Ph.
  • the term "4-12 membered mono-nitrogen-containing monosulfur- or monooxygen-containing heterocyclic ring” refers to a ring of 4 to about 12 atoms, and more preferably 4 to about 9 atoms wherein at least one atom is a nitrogen and at least one atom is oxygen or sulfur. Encompassed within this definition are rings such as thiazoline and the like.
  • arylsulfonyl or “arylsulfone” refers to a radical of the formula R 37 -S0 2 - wherein R 37 is aryl as defined above.
  • alkylsulfoxide or arylsulfoxide refer to radicals of the formula R 38 -SO- wherein R 38 is, respectively, alkyl or aryl as defined above.
  • arylthio refers to a radical of the formula -SR 42 wherein R 42 is aryl as defined above.
  • monocyclic heterocycle thio refers to a radical of the formula -SR 43 wherein R 43 is a monocyclic heterocycle radical as defined above.
  • monocyclic heterocycle sulfone refer, respectively, to radicals of the formula -SO-R 43 and -S0 2 -R 43 wherein R 43 is a monocyclic heterocycle radical as defined above.
  • alkylcarbonyl refers to a radical of the formula R 50 -CO- wherein R 50 is alkyl as defined above.
  • arylcarbonyl refers to a radical of the formula R 5! -CO- wherein R 51 is aryl as defined above.
  • alkoxycarbonyl refers to a radical of the formula R 52 -CO- wherein R 52 is alkoxy as defined above.
  • aryloxycarbonyl refers to a radical of the formula
  • R 51 -0-CO- wherein R 51 is aryl as defined above.
  • haloalkylcarbonyl refers to a radical of the formula R 53 -CO- wherein R 53 is haloalkyl as defined above.
  • haloalkoxycarbonyl refers to a radical of the formula R o -0-CO- wherein R 53 is haloalkyl as defined above.
  • alkylthiocarbonyl refers to a radical of the formula R 50 -S-CO- wherein R 50 is alkyl as defined above.
  • arylthiocarbonyl refers to a radical of the formula R 5I -S-CO- wherein R o1 is aryl as defined above.
  • acyloxymethoxycarbonyl refers to a radical of the formula R ⁇ -O-CH j -CO- wherein R 54 is acyl as defined above.
  • arylamino refers to a radical of the formula R 51 -NH- wherein R 51 is aryl as defined above.
  • polyalkylether refers to commonly used glycols such as triethyleneglycol, tetraethylene glycol, polyethylene glycol and the like.
  • alkylamido refers to a radical of the formula R 50 -NH-CO- wherein R 50 is alkyl as defined above.
  • N,N-dialkylamido refers to a radical of the formula (R 50 ) 2 -N-CO- wherein R 50 is the same or different alkyl group as defined above.
  • pivaloyloxymethyl refers to a radical of the formula (Me) 3 C-CO-0-CH2-.
  • acyloxy refers to a radical of the formula R 55 -0- wherein R 55 is acyl as defined above.
  • the compounds as shown in the present invention can exist in various isomeric forms and all such isomeric forms are meant to be included. Tautomeric forms are also included as well as pharmaceutically acceptable salts of such isomers and tautomers.
  • a bond drawn across a bond of a ring can be to any available atom on the ring.
  • pharmaceutically acceptable salt refers to a salt prepared by contacting a compound of an a v b3 precursor or an a v b3 bioconjugate with an acid whose anion is generally considered suitable for human consumption.
  • pharmacologically acceptable salts include the hydrochloride, hydrobromide, hydroiodide, sulfate, phosphate, acetate, propionate, lactate, maleate, malate, succinate, tartrate salts and the like. All of the pharmacologically acceptable salts may be prepared by conventional means. See Berge et al., (J Pharm. Sci., 66(1): 1- 19, 1977) for additional examples of pharmaceutically acceptable salts.
  • a compound of the present invention can be administered orally, parenterally, or by inhalation spray, or topically in unit dosage formulations containing conventional pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral includes, for example, subcutaneous, intravenous, intramuscular, intrasternal, infusion techniques or intraperitonally. See Wong and Parasrampuria [Biopharm 10(11): 52-61 1997) for a current review of pharmaceutical excipients for the stabilization of proteins. See D.W. Osborne and J.J. Henke (Pharmaceutical Technology 21(11): 58-67, 1997) for an extensive list of chemical compounds used as skin penetration enhancers.
  • a compound of the present invention is administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • Therapeutically effective doses of a compound required to prevent or arrest the progress of or to treat the medical condition are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts.
  • the present invention provides a method of treating conditions mediated by selectively inhibiting or antagonizing the a v b3 cell surface receptor which method comprises administering a therapeutically effective amount of a compound selected from the class of compounds depicted herein as a v b3 precursors or a v b3 bioconjugates, wherein one or more these compounds is administered in association with one or more non-toxic, pharmaceutically acceptable carriers and or diluents and/or adjuvants (collectively referred to herein as "carrier” materials) and if desired other active ingredients.
  • carrier non-toxic, pharmaceutically acceptable carriers and or diluents and/or adjuvants
  • the present invention provides a method for inhibition of the a v b3 cell surface receptor. Most preferably the present invention provides a method for one or more of the following: inhibiting bone resorption, treating osteoporosis, inhibiting humoral hypercalcemia of malignancy, treating Paget's disease, inhibiting tumor metastasis, inhibiting neoplasia (solid tumor growth), inhibiting angiogenesis including tumor angiogenesis, treating diabetic retinopathy, inhibiting arthritis, psoriasis and periodontal disease, and inhibiting smooth muscle cell migration including restenosis.
  • the general synthetic sequences for preparing the compounds useful in the present invention are outlined in Scheme I. Both an explanation of, and the actual procedures for, the various aspects of the present invention are described where appropriate.
  • Peptides conjugated to a resin are covalently coupled to an avb ⁇ antagonist- linker and ligated to the reduced form of Cys-interferon alpha.
  • the bioconjugate is then released from the resin, refolded, and purified to homogeneity.
  • One general scheme for the production of bioconjugates of the present invention is shown below.
  • Boc-NH-a.a.C 0-S-CH 2 CH 2 -CONH-Resin
  • avb antagonist-linker-a.a.-C 0-S-CH 2 CH2-CONH2 t s-
  • v b3 antagonist-linker-a.a.-C 0- NH-CH(SH)-CO-interferon ⁇ (avb ⁇ antagonist-linker Cys interferon ⁇ , reduced form)
  • the present invention encompasses bioconjugates which inherently possess two distinct activities.
  • Bioconjugates with multi-functional a b3 receptor antagonist/metastasis receptor-associated ligand activities are contemplated.
  • anti-angiogenic or anti-tumor activities, or both are contemplated.
  • a contemplated bioconjugate can be formed by the covalent union of two distinct moieties, such as a small chemical and a polypeptide. Each of these distinct moieties may act through a different and specific cell receptor to initiate complementary biological activities.
  • Bioconjugates can also be formed by the covalent union of a small chemical and a peptide, a peptide and a polypeptide, two distinct polypeptides, two distinct peptides, and two distinct small molecules, provided that one of the two moieties alone or in combination with the other moiety possesses a v b3 antagonist activity, are also contemplated by this invention.
  • Peptides in this sense are comprised of two to about 10 amino acid residues.
  • Polypeptides are larger, comprised of from about 11 to about 500 amino acid residues.
  • Bioconjugates containing more than one copy of a moiety achieved through the covalent attachment of more than one copy of that moiety to itself and then to the other moiety, or by covalent attachment of more than one copy of that moiety to the other moiety at multiple sites, are also contemplated by this invention.
  • the present invention relates to bioconjugates comprising one or more a b ⁇ antagonist moieties coupled to an amide or to a metastasis-associated receptor ligand by a covalent bond or by a linear or branched linker.
  • An a v b3 bioconjugate can be composed of a small chemical moiety linked to a polypeptide moiety, for example, (A 1 -L 1 -R 1 -T 1 ) preferably has a polypeptide moiety (T 1 ) with a different but complementary activity than the small chemical moiety (A 1 ).
  • Complementary activity is meant to be activity which enhances or changes the response to another cell modulator.
  • a 1 separately or in combination with L 1 is an a v b3 antagonist
  • T 1 is selected from the group consisting of metastasis-associated receptor ligands and amides,
  • L 1 is a covalent bond or a linker that covalently bonds A 1 to R 2 , R 4 , R 6 , or R 8 ,
  • R 2 , R 4 , R 6 , and R 8 are branched linkers
  • R 3 , R 5 , and R 7 are covalent bonds or linkers that covalently bond R 2 to R 4 , R 4 to R «, and R 6 to R 8 , respectfully, and
  • R 1 is a covalent bond or a linker that covalently bonds R 2 to T 1 .
  • T 1 is a polypeptide. More preferably, the polypeptide is selected from the group consisting of natural cytokines, synthetic cytokines, and anti-angiogenic proteins. Most preferably, the polypeptide is interferon alpha.
  • this invention encompasses the use of modified T 1 molecules or mutated or modified DNA sequences encoding these molecules.
  • the present invention also includes bioconjugates in which T 1 is a variant.
  • R 2 , R 4 , R 6 , and R 8 are branched linkers. More preferably the branched linker is lysine.
  • R 1 is a covalent bond or a linker that covalently bonds R 2 to T 1 and R 1 is selected from the group consisting of peptides and amino-alkyl carboxylic acids.
  • the polypeptide can be joined either directly or through a linker segment to the small chemical moiety.
  • the term "directly” defines bioconjugates in which the polypeptide is joined without a linker.
  • L 1 represents a chemical bond or a linker, preferably a polypeptide segment to which both A 1 and T 1 are joined in a structure represented by the formula I, A 1 -L 1 -R 1 -T 1 , for example, where R 1 is a chemical bond.
  • L 1 is a linear peptide in which A 1 and L 1 are joined by amide bonds, linking A 1 to the amino-terminus of L 1 and carboxy-terminus of L 1 to the amino terminus of T 1 .
  • Compounds of the formulas A 1 -T 1 , ⁇ -L -A 1 , and T 1 - A 1 are also contemplated. The foregoing discussion applies equally to R 1 and when R 1 is a linking group.
  • the linking groups (L 1 and R 1 ) are generally polypeptides of between 1 and 500 amino acid residues in length. More preferably, the peptide linker is a peptide ranging in length from 2 through 10 amino acids.
  • the linkers joining the two molecules are preferably designed to (1) permit the two molecules to fold and act independently of each other, (2) being free of ordered secondary structure which could interfere with the functional domains of the two proteins, (3) can exhibit minimal hydrophobic characteristics that could interact with the functional domains of the linked moieties and (4) provide steric separation of A 1 and T 1 such that A 1 and T 1 can interact simultaneously with their corresponding receptors on a single cell.
  • surface amino acids in flexible protein regions include Gly, Asn and Ser.
  • linkers with the following formulas:
  • -CO-(aa) n - such as -CO-Gly-Gly-Gly-Gly-Ala-, and
  • each linker comprises four or more amino acid or carbon linking units. Additional peptide sequences may also be added to facilitate purification or identification of bioconjugates (e.g., poly-His). A highly antigenic peptide may also be added that would enable rapid assay and facile purification of the bioconjugates by a specific monoclonal antibody.
  • Multi-functional bioconjugates of the present invention can exhibit useful properties such as having similar or greater biological activity when compared to a single factor or by having improved half-life or decreased adverse side effects, or a combination of these properties.
  • Multi-functional bioconjugates which have little or no activity are useful as antigens for the production of antibodies for use in immunology or immunotherapy, as genetic probes or as intermediates used to construct other useful bioconjugates.
  • Biological activity of the multi-functional bioconjugates of the present invention can be determined by tumor cell proliferation assays, endothelial cell proliferation assays, endothelial cell migration assays, endothelial cell tube formation assays, mouse corneal micro-pocket angiogenesis assays, and tumor growth assays.
  • Syngeneic models of mouse tumor growth such as the Lewis Lung carcinoma assay (Sugiura and Stock, Cancer Res.
  • the biological activity of individual moieties can be performed using specific assays.
  • the antiviral activity of interferon for example, can be carried out by titering the potency of interferon preparations on Madin Darby bovine kidney cells infected with vesicular stomatitis virus (Rubinstein et al., J. Virol. 37(2): 755-
  • the a v b3 activity can be carried out using solid state binding assays.
  • the multi-functional bioconjugates of the present invention may have an improved therapeutic profile as compared to single-acting anti-angiogenic or anti- tumor compounds or proteins.
  • some multi-functional bioconjugates of the present invention may have a similar or more potent anti-tumor activity relative to other anti-tumor compounds or proteins without having a similar or corresponding increase in side-effects.
  • the multi-functional bioconjugates of the present invention are useful in the treatment of angiogenic-mediated diseases such as cancer, diabetic retinopathy, and macular degeneration.
  • cancers susceptible to treatment with the polypeptides of the present invention are lung cancer, breast cancer, ovarian cancer, prostate cancer, pancreatic cancer, gastric cancer, colon cancer, renal cancer, bladder cancer, melanoma, hepatoma, sarcoma, and lymphoma.
  • the present invention provides an improvement to the existing methods of treating solid tumors, in that it provides a process utilizing multi-functional bioconjugates that have improved biological activities.
  • Therapeutic treatment of tumors with these multi-functional bioconjugates of the present invention can avoid undesirable side effects caused by treatment with presently available drugs.
  • the treatment of solid tumors can include administration of a pharmaceutical composition containing the multi-functional bioconjugates to a patient.
  • compositions for treating the conditions referred to above.
  • Such compositions comprise a therapeutically effective amount of one or more of the multi-functional bioconjugates of the present invention in a mixture with a pharmaceutically acceptable carrier.
  • the compositions can also be admixtures containing adjunctive agents, such as chemotherapeutic or immunotherapeutic agents.
  • This composition can be administered either parenterally, intravenously, or subcutaneously. Other routes of administration are also contemplated, including intranasal and transdermal routes, and by inhalation.
  • the therapeutic composition for use in this invention is preferably in the form of a pyrogen-free, parenterally-acceptable aqueous solution.
  • the preparation of such a parenterally- acceptable protein solution having due regard to pH, isotonicity, stability and the like, is within the skill of the art.
  • the compounds depicted herein as a v b3 precursors or a v b3 bioconjugates can be used in the treatment of patients suffering from the above pathological conditions.
  • selection of the most appropriate compound of the invention is within the ability of one with ordinary skill in the art and depend on a variety of factors including assessment of results obtained in standard assay and animal models.
  • Treatment of a patient afflicted with one of the pathological conditions comprises administering to such a patient an amount of compound any of the formulas I- VII which is therapeutically effective in controlling the condition or in prolonging the survivability of the patient beyond that expected in the absence of such treatment.
  • the term "inhibition" of the condition refers to slowing, interrupting, arresting, or stopping the condition and does not necessarily indicate a total elimination of the condition. It is believed that prolonging the survivability of a patient, beyond being a significant advantageous effect in and of itself, also indicates that the condition is beneficially controlled to some extent.
  • a compound of the present invention can be used in a variety of biological, prophylactic, or therapeutic areas. It is contemplated that these compounds are useful in prevention or treatment of any disease state or condition wherein the a v b3 integrin plays a role.
  • the dosage regimen for the compounds and/or compositions containing the compounds is based on a variety of factors, including the type, age, weight, sex and medical condition of the patient; the severity of the condition; the route of administration; and the activity of the particular compound employed. Thus the dosage regimen may vary widely. Dosage levels of the order from about 0.01 mg to about 100 mg per kilogram of body weight per day are useful in the treatment of the above-indicated conditions.
  • the active ingredient administered by injection is formulated as a composition wherein, for example, saline, dextrose or water may be used as a suitable carrier.
  • a suitable daily dose would typically be about 0.01 to 10 mg/kg body weight injected per day in multiple doses depending on the factors listed above.
  • the compounds in a therapeutically effective amount are ordinarily combined with one or more excipients appropriate to the indicated route of administration.
  • the compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulphuric acids, gelatin, acacia, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and tableted or encapsulated for convenient administration.
  • a compound can be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, and/or various buffers.
  • Other excipients and modes of administration are well and widely known in the pharmaceutical art.
  • compositions useful in the present invention may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional pharmaceutical adjuvants such as preservatives, stabilizers, wetting agents, emulsifiers, buffers, and the like.
  • a daily regimen may be about 10 ⁇ g/kg of multi- functional bioconjugates per kilogram of body weight. Dosages would be adjusted relative to the activity of a given multi-functional bioconjugates and it would not be unreasonable to note that dosage regimens may include doses as low as 0.1 microgram and as high as 100 milligrams per kilogram of body weight per day.
  • multi- functional bioconjugates there may exist specific circumstances where dosages of multi- functional bioconjugates would be adjusted higher or lower than the range of 0.2 - 100,000 micrograms per kilogram of body weight. These include co-administration with other anti-angiogenic or antitumor proteins or variants; co-administration with adjunctive agents such as chemotherapeutic or immunotherapeutic agents, co-administration with chemotherapeutic drugs and/or radiation; the use of glycosylated multi-functional bioconjugates; and various patient-related issues mentioned earlier in this section. As indicated above, the therapeutic method and compositions may also include co-administration with other human anti-tumor proteins, compounds, or bioconjugates.
  • a non-exclusive list of other appropriate anti-angiogenic or anti-tumor agents or treatments includes chemotherapy, radiation therapy, hormonal therapy, or interleukin-2, or combinations thereof.
  • the dosage recited above would be adjusted to compensate for such additional components in the therapeutic composition.
  • the present invention also includes the DNA sequences which code for the protein portions of multi-functional bioconjugates, DNA sequences which are substantially similar and perform substantially the same function, and DNA sequences which differ from the DNAs encoding the protein portions of multifunctional bioconjugates of the invention only due to the degeneracy of the genetic code. Also included in the present invention are the oligonucleotide intermediates used to construct the mutant DNAs and the polypeptides coded for by these oligonucleotides.
  • Pairs of complementary synthetic oligonucleotides encoding the desired gene can be made and annealed to each other.
  • the DNA sequence of the oligonucleotide would encode sequence for amino acids of desired gene with the exception of those substituted and/or deleted from the sequence.
  • Plasmid DNA can be treated with the chosen restriction endonucleases then ligated to the annealed oligonucleotides.
  • the ligated mixtures can be used to transform competent E. coli cells to resistance to an appropriate antibiotic.
  • Single colonies can be picked and the plasmid DNA examined by restriction analysis and/or DNA sequencing to identify plasmids with the desired genes.
  • Cloning of DNA sequences encoding the polypeptide component of these multi-functional bioconjugates may be accomplished by the use of intermediate vectors.
  • one gene can be cloned directly into a vector containing the other gene.
  • Linkers and adapters can be used for joining the DNA sequences, as well as replacing lost sequences, where a restriction site was internal to the region of interest.
  • genetic material (DNA) encoding one polypeptide, peptide linker, and the other polypeptide is inserted into a suitable expression vector which is used to transform bacteria, yeast, insect cells or mammalian cells.
  • the transformed organism or cell line is grown and the protein isolated by standard techniques.
  • the resulting product is therefore a new protein which has all or a portion of one protein joined by a linker region to all or a portion of second protein.
  • Another aspect of the present invention includes plasmid DNA vectors for use in the expression of the polypeptide component of these multi-functional bioconjugates.
  • These vectors contain the novel DNA sequences described above which code for the novel polypeptides of the invention.
  • Appropriate vectors which can transform microorganisms or cell lines capable of expressing the polypeptide component of these novel multi-functional bioconjugates include expression vectors comprising nucleotide sequences coding for the polypeptide component of these novel multi-functional bioconjugates joined to transcriptional and translational regulatory sequences which are selected according to the host cells used.
  • Vectors incorporating modified sequences as described above are included in the present invention and are useful in the production of the protein component of these multi-functional bioconjugates.
  • the vector employed in the method also contains selected regulatory sequences in operative association with the DNA coding sequences of the invention and which are capable of directing the replication and expression thereof in selected host cells.
  • a method for producing the polypeptide component of these multifunctional bioconjugates is another aspect of the present invention.
  • a method of the present invention comprises culturing suitable cells or cell lines, which have been transformed with a vector containing a DNA sequence coding for expression of the protein portion of a novel multi-functional bioconjugate.
  • Suitable cells or cell lines can be bacterial cells.
  • the various strains of E. coli are well- known as host cells in the field of biotechnology. Examples of such strains include E. coli strains JM101 (Yanisch-Perron et al., Gene 33: 103-119, 1985) and MON105 (Obukowicz et al., Applied Environmental Microbiology 58: 1511-1523, 1992).
  • DH5 alpha and DH10B from Life Technologies, Inc., Rockville, MD
  • DH5 alpha and DH10B from Life Technologies, Inc., Rockville, MD
  • DH10B from Life Technologies, Inc., Rockville, MD
  • Various strains of B. subtilis can also be employed in this method.
  • Many strains of yeast cells known to those skilled in the art are also available as host cells for expression of the polypeptides of the present invention.
  • the gene encoding the protein component of these multi-functional bioconjugates of the present invention can also be constructed such that at the 5' end of the gene codons are added to encode Met- 2 -Ala- 1 , Met- 2 -Ser 1 , Met ⁇ -Cys- 1 , or Met- 1 at the N-terminus of the protein.
  • the N-termini of proteins made in the cytoplasm of E. coli are affected by post- translational processing by methionine aminopeptidase (Ben Bassat et al., J. Bacteriol.
  • polypeptide component of these multi-functional bioconjugates of the present invention may include polypeptides having Met 1 , Ala- 1 , Ser 1 , Cys 1 , Met- 2 -Ala- 1 , Met-2-Ser 1 , or Met- 2 -Cys 1 at the N-terminus. These polypeptides may also be expressed in E. coli by fusing a secretion signal peptide to the N-terminus. This signal peptide is cleaved from the polypeptide as part of the secretion process.
  • Free forms of a v b3 antagonists were prepared by the solid phase method using FMOC chemistry. When completed, avb ⁇ precursors were released from the solid support with trifluoroacetic acid treatment followed by reversed phase HPLC.
  • Avb ⁇ antagonists was prepared as thioesters as described (H. Hojo and S. Aimoto, Bull. Chem. Soc. Jpn. 64: 111, 1991; L.E. Canne et al., J. Am. Chem. Soc. 118: 5891, 1996).
  • Boc-Ala-S-CH 2 CH 2 COOH was prepared and coupled on methylbenzhydryl- amine resin (Peptide International, Louisville, KY) as described (H. Hojo and S. Aimoto, Bull. Chem. Soc. Jpn. 64: 111, 1991). After deprotection with 50% triflouroacetic acid in methylene chloride (2 X 1 min, 1 X 30 min) the remaining amino acid residue and the N-terminal arginine analog was attached on the solid support by N,N'-diisoproplycarbodiimide/l-hydroxybenzotriazole) as coupling agent as described by L. Zhang and J. P. Tarn, J. Am. Chem. Soc. 119: 2363, 1997).
  • the purified a v b3 antagonist thioester was conjugated with amino acids 1-5 of interferon-alpha using an orthogonal coupling method (J.P. Tam et al., Proc. Natl. Acad. Sci. USA 92: 12485, 1995).
  • the a v b3 thioester was reacted with proteins or peptides containing a free N-terminal cysteine residue using a chemoselective method (P. E. Dawson et al., Science 266: 776, 1994).
  • the bioconjugate was purified by reverse-phase HPLC or ionic exchange chromatography.
  • Example 2 Preparation of branched forms ofa ⁇ b3 antagonist interferon conjugates
  • a branched form of an avb ⁇ antagonist interferon-alpha hybrid has been designed for increasing the potency and selectivity. Lysine branching method is adopted as general method described by Tam (J. P. Tam, Proc. Natl. Acad. Sci USA 85: 5409, 1988). A typical example is that exemplified by formulas I-IV.
  • Avb3 antagonists were prepared as thioesters as described (H. Hojo and S.
  • Boc-Ala-(3-thiopropionic acid) ester was coupled into benzylhydryl amine resin.
  • Di-Boc-lysine is used as the branched amino acids.
  • the amino acid residues of spacers and the RGD portion of the molecules, and di-Boc-lysine are coupled by the solid phase method.
  • the arginine analog of the RGA moiety was coupled using TBTU as coupling agent.
  • the crude thioester was purified by reversed HPLC. The purified thioester was coupled with cys- interferon-alpha using the chemoselective method as described above.
  • Avb ⁇ antagonists were prepared as thioesters as described (H. Hojo and S. Aimoto, Bull Chem Soc. Jpn 64: 111, 1991; L. E. Canne et al., J. Am. Chem. Soc. 118: 5891, 1996).
  • Boc-Ala-S-CH 2 CH 2 COOH was prepared and coupled on 4-methyl benzhydrylamine resin (Peptide International, Louisville, KY) as described (H. Hojo & S. Aimoro, 1991). After deprotection with 50% triflouroacetic acid in methylene chloride (2 X 1 min, 1 X 30 min) the remaining amino acid residues were attached on the solid support by N,N'-dicyclohexylcarbodiimide as coupling agent and the N-terminal arginine analog was coupled with TBTU as coupling agent.
  • a v b3 thioesters were reacted with protein or peptide with free N-terminal cysteine residue by using chemoselective method (P. E. Dawson et al., Science 266: 776, 1994).
  • the conjugate was purified by reversed phase HPLC or ionic exchange chromatography.
  • N,N'-di-[3-[(l,4,5,6-tetrahydro-2-pyrimidyl)amino benzoyl glycyl-aspartyl seryl-leucyl- alanyl]- lysyl alanyl glycyl alanyl glycyl alanyl thioester was prepared by the solid-phase method as described in the general methods. Interferon alpha in 6 M guanidine hydrochloride, 0.1 M sodium phosphate, 50 mM sodium chloride, pH 8.2, was first reduced with dithiothreitol and then coupled with the thioester with native chemical ligation as described by S. Kent et al., in WO 96/34878. Folding of the bioconjugate was performed as described above.
  • N,N'-di-[3-[(l,4,5,6-tetrahydro-2-pyrimidyl)amino benzoyl glycyl-aspartyl seryl-glycyl-alanyl]- lysyl alanyl glycyl alanyl tyrosyl glycyl alanyl thioester was prepared by the solid-phase method as described in the general methods. Interferon alpha in 6 M guanidine hydrochloride, 0.1 M sodium phosphate, 50 mM sodium chloride, pH 8.2, was first reduced with dithiothreitol and then coupled with the thioester with native chemical ligation as described by S. Kent et al., in WO 96/34878. Folding of the bioconjugate was performed as described above.
  • N,N'-Di(N,N'-Di-(N,N'-di-[3-[(l,4,5,6-tetrahydro-2-pyrimidyl)amino benzoyl glycyl-aspartyl seryl-leucyl-alanyl]- lysyl) ⁇ lysyl lysyl alanyl glycyl alanyl glycyl alanyl thioester was prepared by the solid-phase method as described in the general methods.
  • N, N'-Di N, N'-Di(3-[(l,4,5,6-tetrahydro-2-pyrimidyl)amino benzoyl glycycl- aspartyl serinyl glycyl glycyl glycyl glycyl alanyl] lysinyl) lysinyl alanyl glycyl alanyl glycyl alanyl (3-thiopropioanmide) thioester was prepared by the solid-phase method as described in the general methods.
  • N, N'-Di N, N'-Di (N, N'-Di(3-[(l,4,5,6-tetrahydro-5-hydroxyl 2-pyrimidyl)amino 5- hydroxyl benzoyl glycycl-aspartyl serinyl glycyl glycyl glycyl alanyl ] lysinyl) lysinyl alanyl glycyl alanyl glycyl alanyl (3-thiopropionamide) thioester was prepared by the solid-phase method as described in the general methods.
  • Fmoc-Lys (Boc)-GAGAG-Lys(Boc)-GAGA was first assembled on Fmoc-Gly- Wang Resin stepwise by the solid phase method. After acid cleavage from the Wang Resin it is purified and then protected with t-Boc groups before coupling to the Ala-thioester resin. After deprotection with 50% TFA [(l,4,5,6-tetrahydro-2- pyrimidyDamino benzoyl glycycl-aspartyl seryl leucyl alanyl residues are then assembled stepwise as described previously. After cleavage by hydrogen fluoride the crude thioester was purified by reversed HPLC. The purified thioester was coupled with cys-interferon-a using the chemoselective method as described earlier.
  • Fmoc-Lys (Boc)-GAGAG-Lys (Boc)-GAGAG-Lys(Boc)-GAGA was first assembled on Fmoc-Gly-Wang Resin stepwise by the solid phase method. After acid cleavage from the Wang Resin it is purified and then protected with t-Boc groups before coupling to the Ala-thioester resin. After deprotection with 50% TFA 3- [(l,4,5,6-tetrahydro-2-pyrimidyl)amino benzoyl glycycl-aspartyl seryl leucyl alanyl residues are then assembled stepwise as described previously. After cleavage by hydrogen fluoride the crude thioester was purified by reversed HPLC. The purified thioester was coupled with cys-interferon-a using the chemoselective method as described earlier.
  • Fmoc- Lys (Boc)-GAGAG Lys (Boc)-GAGAG-Lys (Boc)-GAGAG-Lys(Boc)- GAGA was first assembled on Fmoc-Gly-Wang Resin stepwise by the solid phase method. After acid cleavage from the Wang Resin it is purified and then protected with t-Boc groups before coupling to the Ala-thioester resin. After deprotection with 50% TFA 3-[(l,4,5,6-tetrahydro-2-pyrimidyl)amino benzoyl glycycl-aspartyl seryl leucyl alanyl residues are then assembled stepwise as described previously. After cleavage by hydrogen fluoride the crude thioester was purified by reversed HPLC. The purified thioester was coupled with cys-interferon-alpha using the chemoselective method as described earlier.
  • Compound 304 (A2-GDSLA-N ⁇ (A2-GDSLA)Lys-GAGAG-N ⁇ (A2-GDSLA)Lys- GAGAG-interferon alpha)
  • Fmoc-Lys (Boc)-GAGAG-Lys(Boc)-GAGA was first assembled on Fmoc-
  • Gly-Wang Resin stepwise by the solid phase method. After acid cleavage from the Wang Resin it is purified and then protected with t-Boc groups before coupling to the Ala-thioester resin. After deprotection with 50% TFA 3-[(l,4,5,6-tetrahydro — 5-hydroxyl) 2-pyrimidyl)amino 5-hydroxyl benzoyl glycycl-aspartyl seryl leucyl alanyl residues are then assembled stepwise as described previously. After cleavage by hydrogen fluoride the crude thioester was purified by reversed HPLC. The purified thioester was coupled with cys-interferon-a using the chemoselective method as described earlier.
  • Fmoc-Lys (Boc)-GAGAG-Lys (Boc)-GAGAG-Lys(Boc)-GAGA was first assembled on Fmoc-Gly-Wang Resin stepwise by the solid phase method. After acid cleavage from the Wang Resin it is purified and then protected with t-Boc groups before coupling to the Ala-thioester resin. After deprotection with 50% TFA 3-[(l,4,5,6-tetrahydro-5-hydroxyl 2-pyrimidyl)amino 5-hydroxyl benzoyl glycycl- aspartyl seryl leucyl alanyl residues are then assembled stepwise as described previously. After cleavage by hydrogen fluoride the crude thioester was purified by reversed HPLC. The purified thioester was coupled with cys-interferon-a using the chemoselective method as described earlier.
  • GAGA was first assembled on Fmoc-Gly-Wang Resin stepwise by the solid phase method. After acid cleavage from the Wang Resin it is purified and then protected with t-Boc groups before coupling to the Ala-thioester resin. After deprotection with 50% TFA 3-[(l,4,5,6-tetrahydro-5-hydroxyl 2-pyrimidyl)amino 5-hydroxyl benzoyl glycycl-aspartyl seryl leucyl alanyl residues are then assembled stepwise as described previously. After cleavage by hydrogen fluoride the crude thioester was purified by reversed HPLC. The purified thioester was coupled with cys- interferon-a using the chemoselective method as described earlier.
  • Example 5 Preparation of purified interferon for conjugation to a ⁇ b3 antagonists
  • E. coli strains such as DH5 TM (Life Technologies, Gaithersburg, MD) and TGI (Amersham Corp., Arlington Heights, IL) are used for transformation of ligation reactions and are the hosts used to prepare plasmid
  • E. coli strains such as JM101 (Yanisch- Perron et al., Gene, 33: 103-119, 1985) and MON105 (Obukowicz et al., Appl. and En ⁇ ir. Micr., 58: 1511-1523, 1992) can be used for expressing the protein component of multi-functional bioconjugates of the present invention in the cytoplasm or periplasmic space.
  • JM101 (ATCC#33876) delta (pro lac), supE, thi, V Yanisch-Perron et al.,
  • DH5 ⁇ subcloning efficiency cells are purchased as competent cells and are ready for transformation using the manufacturer's protocol, while both E. coli strains TGI and MON105 are rendered competent to take up DNA using a CaCl 2 method.
  • 20 to 50 mL of cells are grown in LB medium (1% Bacto- tryptone, 0.5% Bacto-yeast extract, 150 mM NaCl) to a density of approximately 1.0 optical density unit at 600 nanometers (OD ⁇ oo) as measured by a Baush & Lomb Spectronic spectrophotometer (Rochester, NY).
  • the cells are collected by centrifugation and resuspended in one-fifth culture volume of CaCl 2 solution (50 mM CaCl 2 , 10 mM Tris-Cl, pH7.4) and are held at 4°C for 30 minutes.
  • the cells are again collected by centrifugation and resuspended in one-tenth culture volume of CaCl 2 solution.
  • Ligated DNA is added to 0.2 mL of these cells, and the samples are held at 4°C for 30-60 minutes.
  • the samples are shifted to 42°C for two minutes and 1.0 mL of LB is added prior to shaking the samples at 37°C for one hour.
  • Cells from these samples are spread on plates (LB medium plus 1.5% Bacto-agar) containing either ampicillin (100 micrograms/mL, ug/mL) when selecting for ampicillin-resistant transformants, or spectinomycin (75 ug/mL) when selecting for spectinomycin-resistant transformants. The plates are incubated overnight at 37°C.
  • Colonies are picked and inoculated into LB plus appropriate antibiotic (100 ug/mL ampicillin or 75 ug/mL spectinomycin) and are grown at 37°C while shaking.
  • appropriate antibiotic 100 ug/mL ampicillin or 75 ug/mL spectinomycin
  • IFN ⁇ -2b interferon alpha 2b gene was amplified from plasmid DNA from ATCC clone no. 67979 using the primer set, IFStart (SEQ ID NO. 1) and IFStop (SEQ ID NO. 2).
  • Oligo IFStart (SEQ ID NO:l) GATCGACCAT GGCTTGTGAT CTGCCTCAAA CC 32
  • Oligo IFStop (SEQ ID NO: 2 ) CGATCGAAGC TTATTATTCC TTACTTCTTA AACTTT 3 6
  • the primers were designed to include the appropriate restriction enzyme recognition sites which allow cloning of the gene into expression plasmids.
  • Conditions for polymerase chain reaction (PCR) amplification were 35 cycles, using settings of 92°C denaturation for one minute, 40°C annealing for one minute, and 72°C extension for one minute.
  • a 100 ul reaction contained 100 pmol of each primer and one ug of template DNA (isolated by Qiagen Miniprep); and IX PCR reaction buffer, 200 uM dNTPs and 0.6 unit Taq DNA polymerase (Boehringer Mannheim).
  • the PCR product was digested with restriction endonucleases Ncol and HmdIII and gel-purified.
  • the vector pMO ⁇ 6875 encoding a Ptac promoter, G10L ribosome binding site and P22 terminator, was digested with restriction endonucleases Ncol and HmdIII. The digested PCR product and vector fragment were combined and ligated. A portion of the ligation reaction was used to transform E. coli strain MO ⁇ 208. Transformant bacteria were selected on spectinomycin-containing plates. Plasmid DNA was isolated and sequenced to confirm the correct insert. The resulting plasmid was designated pMON30422. Construction of pMON30426 (encoding IFNa2b with optimized amino terminal codons)
  • a new gene with an optimized N-terminus was amplified from plasmid DNA from pMON30422 using the primer set, New IF-A (SEQ ID NO. 3) and IFStop (SEQ ID NO. 2).
  • Oligo IFstop (SEQ ID N0: 2 )
  • the primers were designed to include the appropriate restriction enzyme recognition sites which allow cloning of the gene into expression plasmids.
  • Conditions for polymerase chain reaction (PCR) amplification were 35 cycles, using settings of 92°C denaturation for one minute, 40°C annealing for one minute, and 72°C extension for one minute.
  • a 100 ul reaction contained 100 pmol of each primer and one ug of template DNA; and IX PCR reaction buffer, 200 uM dNTPs and 0.6 unit Taq DNA polymerase (Boehringer Mannheim).
  • the PCR product was digested with restriction endonucleases Ncol and HmdIII and gel-purified.
  • Vector D ⁇ A encoding a Ptac promoter, G10L ribosome binding site and P22 terminator, pMO ⁇ 6875 was digested with restriction endonucleases Ncol and HmdIII. The digested PCR product and vector fragment were combined and ligated. A portion of the ligation reaction was used to transform E. coli strain MO ⁇ 208. Transformed bacteria were selected on spectinomycin-containing plates. Plasmid DNA was isolated and sequenced to confirm the correct insert. The resulting plasmid was designated pMON30426. Both pMON30422 and pMON30426 encode the same peptide.
  • the DNA sequence encoding the N-terminus, Met-Ala-Cys-, of pMON30426 was changed to the DNA sequence encoding the N-terminus, Met-Cys-, of pMON20442 by mutagenesis.
  • the QuikChange Site-Directed Mutagenesis Kit of Stratagene was used employing oligos IFcys.for (SEQ ID NO: 12) and IFcys.rev (SEQ ID NO: 13).
  • IFcys.for GAG ATA TAT cca tgT GTG ATC TGC CGC SEQ ID NO: 12
  • Plasmid pMON30426 DNA was digested with the restriction enzymes Ncol and Hindlll resulting in a 3207 bp vector fragment. Plasmid D ⁇ A from pMO ⁇ 30426 was digested with Ncol and Bgl ⁇ l resulting in a 192 bp fragment. The 192 bp fragment along with a 315 bp Bgl HindUI fragment that was assembled from synthetic oligonucleotides IF ⁇ D1 (SEQ ID NO. 4), IFND2 (SEQ ID NO. 5), IFND3X (SEQ ID NO. 6), IFND4X (SEQ ID NO. 7), IFND5 (SEQ ID NO.
  • IFND6 SEQ ID NO. 9
  • IFND7 SEQ ID NO. 10
  • IFND8 SEQ ID NO. 11
  • the genetic elements derived from plasmid pMON20405 are the pBR327 origin of replication, the tac promoter, the gene 10 leader (glO-L) ribosome binding site joined to human interferon (hlFN) alpha A/D hybrid, the P22 transcriptional terminator, and the streptomycin adenyltransferase gene.
  • the DNA sequence encoding the N-terminus, Met-Ala-Cys-, of pMON20405 was changed to the DNA sequence encoding the N-terminus, Met-Cys-, of pMON20433 by mutagenesis.
  • the QuikChange Site-Directed Mutagenesis Kit of Stratagene was used employing oligos IFcys.for (SEQ ID NO: 12) and IFcys.rev (SEQ ID NO: 13).
  • the vector pMON20433 (SEQ ID NO: 22) encodes interferon A D (SEQ ID NO: 23, 28).
  • Plasmid DNA can be isolated by a number of different methods and using commercially available kits known to those skilled in the art. Plasmid DNA is isolated using the Promega WizardTM Miniprep kit (Madison, WI), the Qiagen QIAwell Plasmid isolation kits (Chatsworth, CA) or Qiagen Plasmid Midi or Mini kit. These kits follow the same general procedure for plasmid DNA isolation. Briefly, cells are pelleted by centrifugation (5000 x g), the plasmid DNA released with sequential NaO ⁇ /acid treatment, and cellular debris is removed by centrifugation (10000 x g).
  • the supernatant (containing the plasmid DNA) is loaded onto a column containing a DNA-binding resin, the column is washed, and plasmid DNA eluted. After screening for the colonies with the plasmid of interest, the E. coli cells are inoculated into 50-100 ml of LB plus appropriate antibiotic for overnight growth at 37°C in an air incubator while shaking.
  • the purified plasmid DNA is used for DNA sequencing, further restriction enzyme digestion, additional subcloning of DNA fragments and transfection into E. coli, mammalian cells, or other cell types.
  • Purified plasmid DNA is resuspended in dH 2 0 and its concentration is determined by measuring the absorbance at 260/280 nm in a Bausch and Lomb Spectronic 601 UV spectrometer.
  • DNA samples are sequenced using ABI PRISMTM DyeDeoxyTM terminator sequencing chemistry (Applied Biosystems Division of Perkin Elmer Corporation, Lincoln City, CA) kits (Part Number 401388 or 402078) according to the manufacturer's suggested protocol usually modified by the addition of 5% DMSO to the sequencing mixture. Sequencing reactions are performed in a DNA thermal cycler (Perkin Elmer Corporation, Norwalk, CT) following the recommended amplification conditions.
  • Samples are purified to remove excess dye terminators with Centri-SepTM spin columns (Princeton Separations, Adelphia, NJ) and lyophilized. Fluorescent dye labeled sequencing reactions are resuspended in deionized formamide, and sequenced on denaturing 4.75% polyacrylamide-8M urea gels using ABI Model 373A and Model 377 automated DNA sequencers. Overlapping DNA sequence fragments are analyzed and assembled into master DNA contigs using Sequencher DNA analysis software (Gene Codes Corporation, Ann Arbor, MI).
  • the BHK-21 cell line can be obtained from the ATCC (Rockville, MD). The cells are cultured in Dulbecco's modified Eagle media (DMEM/high-glucose), supplemented to 2 mM (mM) L-glutamine and 10% fetal bovine serum (FBS). This formulation is designated BHK growth media. Selective media is BHK growth media supplemented with 453 units/mL hygromycin B (CalBiochem, San Diego,
  • the BHK-21 cell line was previously stably transfected with the HSV transactivating protein VP16, which transactivates the IE110 promoter found on the plasmid pMON3359 and pMON3633 and the IE175 promoter found in the plasmid pMON3360B (See Hippenmeyer et al., Bio /Technology, pp.1037-1041,
  • the VP16 protein drives expression of genes inserted behind the IE 110 or
  • BHK-21 cells expressing the transactivating protein VP16 are designated BHK-VP16.
  • the plasmid pMONlll ⁇ (See Highkin et al., Poultry Sci., 70: 970-981, 1991) expresses the hygromycin resistance gene from the SV40 promoter.
  • a similar plasmid, pSV2-hph, is available from ATCC.
  • BHK-VP16 cells are seeded into a 60 millimeter (mm) tissue culture dish at 3 x 10 5 cells per dish 24 hours prior to transfection.
  • Cells are transfected for 16 hours in 3 mL of "OPTIMEM”TM (Gibco-BRL, Gaithersburg, MD) containing 10 ug of plasmid DNA containing the gene of interest, 3 ug hygromycin resistance plasmid, pMONlll ⁇ , and 80 ug of Gibco-BRL "LIPOFECTAMINE”TM per dish.
  • the media is subsequently aspirated and replaced with 3 mL of growth media.
  • media from each dish is collected and assayed for activity (transient conditioned media).
  • the cells are removed from the dish by trypsin-EDTA, diluted 1:10, and transferred to 100 mm tissue culture dishes containing 10 mL of selective media. After approximately 7 days in selective media, resistant cells grow into colonies several millimeters in diameter. The colonies are removed from the dish with filter paper (cut to approximately the same size as the colonies and soaked in trypsin/EDTA) and transferred to individual wells of a 24 well plate containing 1 mL of selective media. After the clones are grown to confluence, the conditioned media is re-assayed, and positive clones are expanded into growth media.
  • E. coli strain MON105 or JM101 harboring the plasmid of interest are grown at 37°C in M9 plus casamino acids medium with shaking in an air incubator Model G2 ⁇ from New Brunswick Scientific (Edison, NJ). Growth is monitored at OD ⁇ oo until it reaches a value of 1.0 at which time nalidixic acid (10 mg/mL) in 0.1 N NaOH is added to a final concentration of ⁇ 0 ⁇ g/mL. The cultures are then shaken at 37°C for three to four additional hours. A high degree of aeration is maintained throughout culture period in order to achieve maximal production of the desired gene product. The cells are examined under a light microscope for the presence of inclusion bodies (IB).
  • IB inclusion bodies
  • One mL aliquots of the culture are removed for analysis of protein content by boiling the pelleted cells, treating them with reducing buffer and electrophoresis via SDS-PAGE (see Maniatis et al., "Molecular Cloning: A Laboratory Manual", 1982).
  • the culture is centrifuged (5000 x g) to pellet the cells.
  • Isolation of Inclusion Bodies The cell pellet from a 330 mL E. coli culture is resuspended in 15 mL of sonication buffer (10 mM 2-amino-2-(hydroxymethyl) 1,3-propanediol hydrochloride (Tris-HCl), pH 8.0, 1 mM ethylenediaminetetraacetic acid (EDTA). These resuspended cells are sonicated using the microtip probe of a Sonicator Cell Disruptor (Model W-375, Heat Systems-Ultrasonics, Inc., Farmingdale, New York). Three rounds of sonication in sonication buffer followed by centrifugation are employed to disrupt the cells and wash the inclusion bodies (IB). The first round of sonication is a 3 minute burst followed by a 1 minute burst, and the final two rounds of sonication are for 1 minute each.
  • Tris-HCl 2-amino-2-(hydroxymethyl) 1,3-propanedi
  • E. coli inclusion bodies were dissolved in 6 M Gn-HCl, 10 mM EDTA, pH 8. ⁇ -9; stirred at 4°C for 1 hr and then diluted 4-fold with 0.1M Tris/HCl pH 8.1. The solution is left to oxidize, while stirring, for 48-72 hrs at 4°C. The structure of refolded molecules is monitored by reverse-phase HPLC.
  • the sample was dialyzed against 20 volumes of 0.1 M NaCl at 4°C and the precipitate removed by centrifugation.
  • the pH of the sample was adjusted to pH 4.5 by addition of NaP/phosphoric acid.
  • the sample was dialyzed against 50 mM NaP, pH 4.5, O.l ⁇ M NaCl. Precipitates are removed from the sample by centrifugation and the sample was applied to Mono S or S-sepharose and eluted with 1 M NaCl in ⁇ O mM NaP, pH 4.5.
  • the pH of the eluted sample was adjusted to pH 7 by the addition of dibasic NaP and then loaded onto a Pharmacia Cu- chelate resin, washed and eluted in 50 mM acidic acid, 0.25 M NaCl, pH 3.
  • the folded proteins can be affinity-purified using affinity reagents such as monoclonal antibodies or receptor subunits attached to a suitable matrix. Purification can also be accomplished using any of a variety of chromatographic methods such as: ion exchange, gel filtration or hydrophobic chromatography or reversed phase HPLC. These and other protein purification methods are described in detail in Methods in Enzymology, Volume 182 "Guide to Protein Purification” edited by Murray Deutscher, Academic Press, San Diego, California, 1990. Protein Characterization
  • Protein quantitation is done by amino acid composition, RP-HPLC, and Bradford protein determination. In some cases tryptic
  • 5 peptide mapping is performed in conjunction with electrospray mass spectrometry to confirm the identity of the protein.
  • bioconjugates containing a v b3 inhibitors conjugated to interferon were characterized by electrospray mass spectrometry, SDS-PAGE, mapping of trypsin-digested fragments, and amino acid composition analysis. 0 Endotoxin levels in recombinant protein samples were determined by standard assays.
  • Antiviral activity induced by human interferon is measured spectrophotometrically as inhibition of the cytopathic effect that normally results from infection of Madin Darby bovine kidney cells, (ATCC CCL #22), with vesicular stomatitis virus (VSV) (ATCC VR-158) (Rubinstein et al., J. Virol. 37(2): 7 ⁇ -758, 0 1981).
  • VSV stocks are prepared on mouse L cells (L929) (ATCC CCL# 1). Samples of interferon are serially titrated in a 96-well plate format and incubated with 4 x 10 4 cells per well, 6 hours prior to addition of virus at a multiplicity of infection of 0.1 plaque forming units per cell.
  • the cells are stained with crystal violet at 20-24 hours postinfection and staining is measured spectrophotometrically at 680 nm.
  • the relative potencies of the samples are compared with Intron A, a recombinant interferon produced by Schering Plough and obtained by prescription.
  • Human Burkitt's lymphoma Daudi cells (ATCC) are seeded at 2 x 10 4 cells/well into 96-well tissue culture plates and cells are cultured in the presence or 0 absence of serial doses of rhIFN alpha 2b for 3 days. Cultures are pulsed with 3 H- thymidine for the last hour of the culture period, and the 3 H-thymidine uptake, counts per minute (cpm), measured on a Beta-plate reader. All samples are assayed in triplicate. Endothelial cell proliferation assay
  • HdMVEC human dermal microvascular endothelial cells
  • BocEnd, Incell bovine microvascular 5 endothelial cells
  • FBS heat-inactivated fetal bovine serum
  • antibiotics 100 ug/ml heparin (Sigma) and 100 ug/ml endothelial mitogen (Biomedical Technologies).
  • Confluent monolayers at passages 2-5 were dispersed in .05% trypsin and resuspended in complete medium.
  • Five hundred ul of complete media containing 1.25 x 10 4 cells 0 were seeded into wells of a 24-well tissue culture plate coated with 0.1% gelatin (Sigma). The cells were incubated overnight at 3775% CO2 at which time the media was replaced with 250 ul of media containing 5% FBS and various concentrations of inhibitors. After 30 minutes of incubation, 250 ul of media containing 1 ng/ml bFGF (R&D Systems) was added and the cells were incubated 5 for an additional 72 hours, at which time they were trypsinized and counted with a Coulter counter.
  • the endothelial cell migration assay is performed essentially as previous described (Gately et al., Cancer Res. 56:4887-4890, 1996). To determine the ability 0 of angiostatin to inhibit the migration of endothelial cells, migration assays were performed in a transwell chamber (Costar) containing 8 mm pore size polycarbonate membranes. The cells utilized in the assay were either human microvascular endothelial cells from Emory or bovine endothelial cells (kindly provided by Gately Northwestern University, Evanston, IL).
  • the cells were 5 starved overnight before use in MCDB131 + 0.1% BSA (human cells) or DMEM + 0.1% BSA (bovine cells), harvested, and resuspended in the same media at 10 6 cells/ml.
  • the lower side of the transwells were coated with 0.1% gelatin for 30 minutes at 37°C before addition of 2 x 10 5 cells to the upper chamber.
  • the transwell was moved to a well containing the chemoattractant (bFGF or VEGF) in 0 the lower chamber. Migration was allowed to occur overnight at 37°C.
  • the membranes were then fixed and stained, and the number of cells that migrated to the lower side of the membrane counted in 3 high powered fields.
  • the corneal micropocket assay has been developed to evaluate the anti- ⁇ angiogenic activity of test compounds in mice.
  • BALBc or C57BL strains of mice are anesthetized with avertin (tribromoethanol, 125 mpk, 0.3-.4 ml/mouse, i.p., 25 ga needle).
  • the eyes are topically anesthetized with 0.5% proparacaine. Only one eye is used.
  • the eye is proptosed with a small forceps and under an operating microscope, a central, intrastromal linear keratotomy is performed with a #15 blade parallel to the insertion of the lateral rectus muscle.
  • a modified cataract knife (1 x 20 mm) is then inserted to dissect a lamellar micropocket to within 1 mm of the temporal limbus.
  • a single Hydron pellet containing either basic fibroblastic growth factor or vascular endothelial growth factor (bFGF or VEGF) is placed on the eye and pushed into the pocket with one arm of the forceps.
  • the flap is self- sealing.
  • Antibiotic ointment (Neobacimyx) is applied once to prevent infection and to decrease irritation of the irregular ocular surface.
  • Compounds are administered immediately post-operatively. They can be administered either orally, intraperitoneally, subcutaneously or intravenously, depending on bioavailability and potency of the compound. Dosing is from one to three times daily for oral compounds, one or two per day for i.p. or s.q., and once per day via the tail vein for i.v. delivery. Volumes do not exceed 5ml/kg orally, lOml/kg i.p. or s.q. or 2.5 ml/kg i.v. All injections are done with a 25 guage needle.
  • mice On post-operative day 5 or 6 the mice are anesthetized with avertin (125 mpk, i.p.), the eyes proptosed, and the degree of neovascularization assessed by determining the maximum vessel length, and the contiguous circumferential zone involved. Using the formula for the area of an ellipse, the neovascular area is measured. The animals receive a thoracotomy while still anesthetized, to assure euthanasia. Some of the eyes are removed for histology. If blood samples are required for compound blood levels, the mice are bled by cardiac puncture immediately following the corneal neovascularization assessment. This is done via a substernal approach with a 1 inch, 23 guage needle, and the animal is subsequently euthanized.
  • avertin 125 mpk, i.p.
  • Topical proparicaine is used as necessary to relieve irritation of the affected eye.
  • the maximum number of bleeds per rat is four, every third day, although typically only two are required, one at day 4 or 6 and one at completion.
  • mice can be used to evaluate the anti-tumor activity of the chimeric proteins; either direct effects on the growth of the primary tumor or effects on metastasis.
  • These can be divided into two broad classes: syngeneic models of mouse tumor in mice, such as the Lewis Lung Carcinoma (Sugiura and Stock, Cancer Res., 15: 38-51, 1955; O'Reilly et al., Cell 79(2): 315-328, 1994) and xenograft models of human tumors in nude or severe combined immunodeficiency (SCID) mice.
  • SCID severe combined immunodeficiency
  • Examples of the human tumor xenografts include: the breast cancer cell lines, MDA-MB-435 (Price, Breast Cancer Research and Treatment, 39: 93-102, 1996) and MDA-231 (Sasaki et al., Can. Res. 55: 3551-3557, 1995), the human prostate carcinoma cell line, PC-3 (Pretlow et al, Can. Res. 51: 3814-3817, 1991; Passaniti et al., Int. J. Cancer, 51: 318-324, 1992) and the human melanoma line M21 (Felding-Habermann et al., J. Clin. Invest, 89: 2018-2022, 1992).
  • Human vitronectin receptor ( ⁇ v ⁇ 3) was purified from human placenta as previously described (Pytela et al., Methods in Enzymology, 144: 475-489, 1987). Human vitronectin was purified from fresh frozen plasma as previously described (Yatohgo et al., Cell Structure and Function, 13: 281-292, 1988)] . Biotinylated human vitronectin was prepared by coupling NHS-biotin from Pierce Chemical Company (Rockford, IL) to purified vitronectin as previously described (Charo et al., J. Biol. Chem., 266(3): 1415-1421 1991). Assay buffer, OPD substrate tablets, and RIA grade BSA were obtained from Sigma (St. Louis, MO). Anti-biotin antibody was obtained from CalBiochem (La Jolla, CA). Linbro microtiter plates were obtained from Flow Labs (McLean, VA). ADP reagent was obtained from Sigma (St. Louis, MO).
  • the purified human vitronectin receptor ( v ⁇ ) was diluted from stock solutions to 1.0 g/mL in Tris-buffered saline containing 1.0 mM Ca ++ , Mg ⁇ , and Mn ++ , pH 7.4 (TBS +++ ). The diluted receptor was immediately transferred to Linbro microtiter plates at 100 uL/well (100 ng receptor/well). The plates were sealed and incubated overnight at 4 C to allow the receptor to bind to the wells. All remaining steps were at room temperature.
  • the assay plates were emptied and 200 uL of 1% RIA grade BSA in TBS +++ (TBS + -VBSA) were added to block exposed plastic surfaces. Following a 2 hour incubation, the assay plates were washed with TBS +++ using a 96 well plate washer. Logarithmic serial dilution of the test compound and controls were made starting at a stock concentration of 2 mM and using 2 nM biotinylated vitronectin in TBS +++ /BSA as the diluent.
  • the plates were washed and incubated with OPD/H 2 0 2 substrate in 100 mM L Citrate buffer, 5 pH 5.0.
  • the plate was read with a microtiter plate reader at a wavelength of 450 nm and when the maximum-binding control wells reached an absorbance of about 1.0, the final A450 were recorded for analysis.
  • the data were analyzed using a macro written for use with the EXCELTM spreadsheet program.
  • the mean, standard deviation, and %CV were determined for duplicate concentrations.
  • the 0 mean A 45 0 values were normalized to the mean of four maximum-binding controls (no competitor added)(B-MAX).
  • Human fibrinogen receptor ( ⁇ b ⁇ a) was purified from outdated platelets. (Pytela, R., Pierschbacher, M.D., Argraves, S., Suzuki, S., and Rouslahti, E. "Arginine-Glycine-Aspartic acid adhesion receptors", Methods in Enzymology 144: ⁇ 47 ⁇ -489, 1987) Human vitronectin was purified from fresh frozen plasma as described (Yatohgo, T., Izumi, M., Kashiwagi, H., and Hayashi, M., "Novel purification of vitronectin from human plasma by heparin affinity chromatography," Cell Structure and Function 13:):281-292, 1988).
  • Biotinylated human vitronectin was prepared by coupling NHS-biotin from Pierce Chemical 0 Company (Rockford, IL) to purified vitronectin as previously described. (Charo, I.F., Nannizzi, L., Phillips, D.R., Hsu, M.A., Scarborough, R.M., "Inhibition of fibrinogen binding to GP Ilb/IIIa by a GP Ilia peptide", J. Biol. Chem. 266(3): 1415-1421, 1991). Assay buffer, OPD substrate tablets, and RIA grade BSA were obtained from Sigma (St. Louis, MO). Anti-biotin antibody was obtained from 5 CalBiochem (La Jolla, CA).
  • Linbro microtiter plates were obtained from Flow Labs (McLean, VA).
  • ADP reagent was obtained from Sigma (St. Louis, MO). This assay is essentially the same reported in Niiya, K., Hodson, E., Bader, R., Byers-Ward, V. Koziol, J.A., Plow, E.F. and Ruggeri, Z.M., "Increased surface expression of the membrane glycoprotein Ilb/IIIa complex induced by platelet activation: Relationships to the binding of fibrinogen and platelet aggregation", Blood 70: 475-483, 1987).
  • the purified human fibrinogen receptor (allbb3) was diluted from stock solutions to 1.0 ⁇ g/mL in Tris-buffered saline containing 1.0 M Ca ++ , Mg+ + , and Mn ++ , pH 7.4 (TBS +++ ).
  • the diluted receptor was immediately transferred to Linbro microtiter plates at 100 uL/well (100 ng receptor/well). The plates were sealed and incubated overnight at 4 C to allow the receptor to bind to the wells. All remaining steps were at room temperature.
  • the assay plates were emptied and 200 ⁇ L of 1% RIA grade BSA in TBS +++ (TBS +++ /BSA) were added to block exposed plastic surfaces.
  • the assay plates were washed with TBS +++ using a 96 well plate washer.
  • Logarithmic serial dilution of the test compound and controls were made starting at a stock concentration of 2 mM and using 2 nM biotinylated vitronectin in TBS +++ /BSA as the diluent.
  • This premixing of labeled ligand with test (or control) ligand, and subsequent transfer of 50 uL aliquots to the assay plate was carried out with a CETUS Propette robot; the final concentration of the labeled ligand was 1 nM and the highest concentration of test compound was 1.0 x 10- 4 M.
  • the mean, standard deviation, and %CV were determined for duplicate concentrations.
  • the mean A450 values were normalized to the mean of four maximum-binding controls (no competitor added)(B-MAX).
  • the normalized values were subjected to a four parameter curve fit algorithm, (Robard et al., Int. Atomic Energy Agency, Vienna, pp 469, 1977), plotted on a semi-log scale, and the computed concentration corresponding to inhibition of 50% of the maximum binding of biotinylated vitronectin (IC ⁇ O) and corresponding R 2 was reported for those compounds exhibiting greater than ⁇ 0% inhibition at the highest concentration tested; otherwise the IC ⁇ O is reported as being greater than the highest concentration tested.
  • IC ⁇ O biotinylated vitronectin
  • a 1 is:
  • a 1 is:
  • a 1 is:
  • X is succinyl (-C(0)-CH 2 -CH 2 -C(0)- ), amino acids, or peptides served as linker.
  • Table 6 SEQ ID Correlation table

Abstract

La présente invention se rapporte à des composés pharmaceutiques qui sont des ligands multivalents du récepteur de avb3 et des récepteurs associés aux métastases. L'invention se rapporte également à l'utilisation de ces ligands multivalents, seuls ou associés à d'autres agents, dans des compositions pharmaceutiques et dans des méthodes de traitement des troubles à médiation assurée par avb3 permettant le traitement du cancer et d'autres maladies angiogéniques, telles que la rétinopathie diabétique, l'arthrite, les hémangiomes et le psoriasis.
PCT/US1999/004296 1998-08-13 1999-04-07 Ligands multivalents du recepteur de avb3 et des recepteurs associes aux metastases WO2000009143A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP99916118A EP1104304A1 (fr) 1998-08-13 1999-04-07 Ligands multivalents du recepteur de avb3 et des recepteurs associes aux metastases
AU34498/99A AU3449899A (en) 1998-08-13 1999-04-07 Multivalent avb3 and Metastasis-Associated Receptor Ligands
CA002338283A CA2338283A1 (fr) 1998-08-13 1999-04-07 Ligands multivalents du recepteur de avb3 et des recepteurs associes aux metastases
JP2000564645A JP2002522504A (ja) 1998-08-13 1999-04-07 多価avb3および転移関連レセプター・リガンド

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US9644298P 1998-08-13 1998-08-13
US60/096,442 1998-08-13

Publications (1)

Publication Number Publication Date
WO2000009143A1 true WO2000009143A1 (fr) 2000-02-24

Family

ID=22257375

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/004296 WO2000009143A1 (fr) 1998-08-13 1999-04-07 Ligands multivalents du recepteur de avb3 et des recepteurs associes aux metastases

Country Status (5)

Country Link
EP (1) EP1104304A1 (fr)
JP (1) JP2002522504A (fr)
AU (1) AU3449899A (fr)
CA (1) CA2338283A1 (fr)
WO (1) WO2000009143A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003228057B2 (en) * 2002-04-30 2008-08-28 Molmed Spa Immunoconjugates for the treatment of tumours
WO2009046080A1 (fr) 2007-10-01 2009-04-09 Pharmaessentia Corp. Interféron-alpha modifié n-terminal
US8877158B2 (en) 2009-06-12 2014-11-04 Fujifilm Corporation Targeting agent to newly formed blood vessels
US10662247B2 (en) 2014-10-08 2020-05-26 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993020229A1 (fr) * 1992-04-03 1993-10-14 Genentech, Inc. ANTICORPS DE L'INTEGRINE ALPHA v BETA 3
WO1995025543A1 (fr) * 1994-03-18 1995-09-28 The Scripps Research Institute Compositions inhibitrices de l'angiogenese et procedes correspondants
WO1997036858A1 (fr) * 1996-03-29 1997-10-09 G.D. Searle & Co. Derives de l'acide alcanoique de cyclopropyle

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1993020229A1 (fr) * 1992-04-03 1993-10-14 Genentech, Inc. ANTICORPS DE L'INTEGRINE ALPHA v BETA 3
WO1995025543A1 (fr) * 1994-03-18 1995-09-28 The Scripps Research Institute Compositions inhibitrices de l'angiogenese et procedes correspondants
WO1997036858A1 (fr) * 1996-03-29 1997-10-09 G.D. Searle & Co. Derives de l'acide alcanoique de cyclopropyle

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2003228057B2 (en) * 2002-04-30 2008-08-28 Molmed Spa Immunoconjugates for the treatment of tumours
US9119886B2 (en) 2002-04-30 2015-09-01 Molmed Spa Immunoconjugates for the treatment of tumours
US9782496B2 (en) 2002-04-30 2017-10-10 Molmed Spa Immunoconjugates for the treatment of tumours
WO2009046080A1 (fr) 2007-10-01 2009-04-09 Pharmaessentia Corp. Interféron-alpha modifié n-terminal
EP2195338A1 (fr) * 2007-10-01 2010-06-16 Pharmaessentia Corp. Interféron-alpha modifié n-terminal
EP2195338A4 (fr) * 2007-10-01 2010-11-03 Pharmaessentia Corp Interféron-alpha modifié n-terminal
US8106160B2 (en) * 2007-10-01 2012-01-31 Pharmaessentia Corp. N-terminal modified interferon-alpha
AU2008308805B2 (en) * 2007-10-01 2013-05-30 Pharmaessentia Corp. N-terminal modified interferon-alpha
US8877158B2 (en) 2009-06-12 2014-11-04 Fujifilm Corporation Targeting agent to newly formed blood vessels
US10662247B2 (en) 2014-10-08 2020-05-26 Novartis Ag Compositions and methods of use for augmented immune response and cancer therapy

Also Published As

Publication number Publication date
JP2002522504A (ja) 2002-07-23
CA2338283A1 (fr) 2000-02-24
EP1104304A1 (fr) 2001-06-06
AU3449899A (en) 2000-03-06

Similar Documents

Publication Publication Date Title
US8759285B2 (en) VEGF superagonists and methods of use
EP1705248A1 (fr) Protéines de fusion comprenant une fraction d'angiostatine et leur utilisation dans un traitement ant-tumeur
JP4332163B2 (ja) Mpl受容体に結合し血小板生成活性を有する二量体トロンボポエチンペプチド模倣体
CZ2001643A3 (cs) Exprese a export inhibitorů angiogeneze ve formě imunofúzinů
AU2005317684B2 (en) Conjugation product
EP0407008A2 (fr) Fragment de fibronectine
US20060172941A1 (en) Anti-angiogenic peptides and methods of use thereof
JPH06509551A (ja) Gp II↓bIII↓aに対する高い特異性を有する血小板凝集阻害剤
JP2008231107A (ja) 抗血管新生性ポリペプチド及び血管新生を抑制するための方法
WO1994012219A2 (fr) Facteur de croissance insulinoide modifie
EP1070085A1 (fr) LIGANDS DOUBLES DU RECEPTEUR A v?b 3? ET DU RECEPTEUR ASSOCIE AUX METASTASES
JP2014051498A (ja) 修飾基をポリペプチドおよびその他の高分子に結合するための窒素ベースのリンカー
JP2018505850A (ja) 皮膚状態改善活性を有するペプチド及びその用途
CA2837858A1 (fr) Blocage de proteases inflammatoires par des theta - defensines
CA2355874A1 (fr) Inhibiteurs de l'integrine $g(a)v$g(b)6
TW201838647A (zh) 對胰島素受體有降低親和性之胰島素類似物之接合物及其用途
JP2011523634A (ja) ペプチド、ペプチド模倣物およびそれらの誘導体、それらの製造、ならびに治療および/または予防活性のある医薬組成物を調製するためのそれらの使用
WO2000009143A1 (fr) Ligands multivalents du recepteur de avb3 et des recepteurs associes aux metastases
CA2333929A1 (fr) Derives de polypeptides
EP1542730B1 (fr) Agents therapeutiques modifies
AU2012202866A1 (en) Conjugation product
MXPA01006229A (en) &agr;v
MXPA01001970A (en) Expression and export of angiostatin and endostatin as immunofusis

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK 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 MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN 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)
ENP Entry into the national phase

Ref document number: 2338283

Country of ref document: CA

Ref country code: CA

Ref document number: 2338283

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1999916118

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1999916118

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 1999916118

Country of ref document: EP