EP2895508A1 - Inhibiteurs des interactions de liaison de la sous-unité alpha de protéine g-intégrine bêta - Google Patents

Inhibiteurs des interactions de liaison de la sous-unité alpha de protéine g-intégrine bêta

Info

Publication number
EP2895508A1
EP2895508A1 EP13783707.6A EP13783707A EP2895508A1 EP 2895508 A1 EP2895508 A1 EP 2895508A1 EP 13783707 A EP13783707 A EP 13783707A EP 2895508 A1 EP2895508 A1 EP 2895508A1
Authority
EP
European Patent Office
Prior art keywords
peptide
integrin
seq
antibody
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP13783707.6A
Other languages
German (de)
English (en)
Inventor
Xiaoping Du
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Illinois
Original Assignee
University of Illinois
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US13/621,064 external-priority patent/US8685921B2/en
Application filed by University of Illinois filed Critical University of Illinois
Publication of EP2895508A1 publication Critical patent/EP2895508A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70546Integrin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70546Integrin superfamily
    • C07K14/70557Integrin beta3-subunit-containing molecules, e.g. CD41, CD51, CD61

Definitions

  • Integrins are heterodimer transmembrane receptors for the extracellular matrix and are composed of an alpha and beta subunit.
  • Naturally-occurring integrin ligands include laminin, fibronectin, and vitronectin, and also include fibrinogen and fibrin, thrombospondin, and von Willebrand factor, and fibroblast growth factor 2.
  • Integrins bind ligands by recognizing short amino acid stretches on exposed loops, particularly the arginine-glycine-aspartic acid (RGD) or like sequences.
  • Integrins Upon ligation, integrins mediate cell adhesion, and initiate complex signaling events, alone or in combination with other types of receptors (such as growth factor receptors), and regulate cell spreading, retraction, adhesion, proliferation, survival, and migration. Integrin signaling is bi-directional. Intracellular signals mediates so-called “inside-out” signaling, which induces activation of the ligand binding functions of integrins. Integrin ligation activate "outside- in” signaling pathways, including, for example Src family kinases (SFK), the phosphoinositide 3-kinase, protein kinase B (PKB/Akt), mitogen-activated protein kinase (MAPK), and Rac. See, e.g., Li Z, Delaney MK, O'Brien KA, Du X., Arterioscler Thromb Vase Biol. 30(12):2341-2349, 2010.
  • SFK Src family kinases
  • Integrins are expressed and serve as major adhesion receptors on the surface of blood platelets, a type of blood cells that are critical in thrombosis and hemostasis.
  • the major integrin expressed on platelet surface is the integrin ⁇ 3, also called glycoprotein Ilb-IIIa (GPIIa-IIIa).
  • GPIIa-IIIa glycoprotein Ilb-IIIa
  • integrin ⁇ 3 mediates stable platelet adhesion, spreading and aggregation. This process normally serves to stop bleeding and prevent loss of blood (that is called hemostasis).
  • platelets Under certain conditions, such as at sites of atherosclerosis, platelets form a occlusive thrombus that block blood vessels, leading to ischemia of organs and tissues, causing such as heart attack and thrombotic stroke etc(Li Z, Delaney MK, O'Brien KA, Du X.,
  • inhibitors of integrin function are clinically used to prevent and treat thrombotic diseases. Integrins are also important in other physiological and pathological processes such as immunity, inflammation, angiogenesis and tumor progression and metastasis.
  • monoclonal antibodies targeting the extracellular ligand binding domain of the heterodimer eg, Reopro, Eli Lilly, Indiapolis, Vitaxin; Medlmmune, Gaithersburg, MD
  • synthetic peptides containing an RGD or KGD sequences eg, Integrillin, Millennium Pharmaceuticals; cilengitide; Merck KGaA, Darmstadt, Germany
  • peptidomimetics eg, aggrestat (Tirofiban), Merck, White House Station, NJ; S247; Pfizer, St Louis, MO).
  • the first integrin-specific drugs targeted the integrin ⁇ 3 ⁇ 4 ⁇ 3 , which is central to hemostasis and plays an important role in platelet adhesion and thrombus formation. ⁇ 3 ⁇ 4 ⁇ 3 also funtions in the inflammatory response.
  • the first FDA-approved ⁇ 3 ⁇ 4 ⁇ 3 antagonists have proven benefit for indications, including acute coronary syndromes and prevention of myocardial infarction. However, the use of some of these drugs are limited due to their pharmacokinetic profiles - some drugs demonstrate rapid plasma clearance, rapid metabolism, poor oral bioavailability, and/or large variation in plasma levels. Also, some antagonists of ⁇ 3 ⁇ 4 ⁇ 3 integrin induced thrombocytopenia.
  • the invention provides a compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit.
  • the compound takes form of an antibody, or antibody analog, a peptide, or peptide analog.
  • the invention provides, antibodies, antibody analogs, peptides, and peptide analogs.
  • composition e.g., a pharmaceutical composition, comprising a compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit.
  • Kits comprising one or more compounds of the are also provided herein.
  • the invention provides a method of inhibiting a binding interaction between an integrin and a G protein subunit in a cell.
  • the method comprises the step of contacting the cell with a compound or composition of the invention in an amount effective to inhibit the binding interaction.
  • the invention further provides a method of inhibiting integrin-dependent Src activation in a cell.
  • the method comprises the step of contacting the cells with a compound or composition of the invention in an amount effective to inhibiting the Src activation.
  • a method of activating a GTPase is furthermore provided by the invention.
  • the method comprises the step of contacting a G protein subunit with a compound or composition in an amount effective to activate a GTPase.
  • the invention moreover provides a method of inhibiting spreading or migration of a cell.
  • the method comprises the step of contacting the cell with a compound or composition of the invention in an amount effective to inhibit spreading and migration.
  • Also provided by the invention is a method of inhibiting platelet adhesion.
  • the method comprises the step of contacting a platelet with a compound or composition of the invention in an amount effective to inhibit platelet adhesion.
  • the invention further provides a method of inhibiting platelet granule secretion and platelet aggregation.
  • the method comprises the step of contacting a platelet with a compound or composition of the invention in an amount effective to inhibit granule secretion and aggregation.
  • the compounds and compositions of the invention are additionally contemplated for therapeutic purposes.
  • the compounds and compositions of the invention may be used to enhance blood clot retraction or inhibit thrombosis in a subject in need thereof.
  • the invention provides a method of enhancing blood clot retraction in a subject in need thereof.
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to enhance blood clot retraction.
  • a method of inhibiting thrombosis in a subject in need thereof comprises the step of administering to the subject a compound or composition of the invention in an amount effective to inhibit thrombosis.
  • the invention furthermore provides a method of treating or preventing a stroke or a heart attack in a subject in need thereof.
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to treat or prevent stroke or heart attack.
  • the invention also provides a method of inhibiting metastasis of a tumor cell.
  • the method comprises the step of contacting a tumor cell with a compound or composition of the invention in an amount effective to inhibit metastasis.
  • the compounds and compositions are also contemplated for use in inhibiting angiogenesis. Accordingly, the invention provides a method of inhibiting
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to inhibit
  • the invention moreover provides a method of treating or preventing cancer in a subject in need thereof.
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to treat or prevent cancer.
  • the compounds and compositions provided herein also may be used for affecting leukocyte function.
  • the invention accordingly provides a method of inhibiting leukocyte adhesion, spreading, migration, or chemotaxis.
  • the method comprises the step of contacting a leukocyte with a compound or composition of the invention in an amount effective to inhibit leukocyte adhesion, spreading, migration, or chemotaxis. Since these leukocyte functions are related to inflammation, the invention additionally provides a method of inhibiting or treating inflammation in a subject in need thereof.
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to inhibit or treat inflammation.
  • Figure 1 demonstrates a role of Gcc 13 in integrin outside-in signaling.
  • A Confocal microscopy images of spreading scrambled siRNA control platelets or Gcc 13 -depleted platelets (Gcc 13 -siRNA) on fibrinogen, without or with Y27632. Merged EGFP (green) fluorescence and Alex Fluor 546-conjugated phalloidin (Red) fluorescence.
  • B Western blot comparison of Gcc 13 abundance in platelets from mice innoculated with control siRNA- or Gcc 13 -siRNA-transfected bone marrow stem cells.
  • Figure 2 demonstrates the binding of Gcc 13 to ⁇ and the inhibitory effect of mSRI peptide.
  • A Proteins from platelet lysates were immunoprecipitated with control IgG or antibody to Gcc 13 with or without 1 ⁇ GDP, 1 ⁇ GTP or 30 ⁇ A1F 4 ⁇ . Immunoprecipitates were immunoblotted with anti-Gcc 13 or anti- (MAbl5). See Fig. S4 for quantitation.
  • Immunoprecipitates were immunoblotted with anti-G(Xi3, anti-p 3 , or anti-GPIb antibodies.
  • C, D Purified GST- 3CD (C) or GST- lCD (D) bound to glutathione beads was mixed with purified Gcc 13 with or without 1 ⁇ GDP, 1 ⁇ GTPyS or 30 ⁇ A1F 4 " . Bound proteins were immunoblotted with anti-Gcc 13 . Quantitative data are shown as mean+SD and p value (t-test).
  • G Protein from platelet lysates treated with 0.1% DMSO, 250 ⁇ scrambled control peptide (Ctrl) or mSRI were immunoprecipitated with anti-Gcc 13 . Immunoprecipitates were immunoblotted with anti- Gcc 13 or anti-p . See Fig. S4 for quantitation.
  • Figure 3 demonsrates the effects of mSRI on integrin-induced c-Src phosphorylation, RhoA activity and platelet spreading.
  • Washed human platelets pre-treated with DMSO, mSRI, or scrambled control peptide were allowed to adhere to fibrinogen and then solubilized at indicated time points. Proteins from lysates were immunoblotted with antibodies to c-Src phosphorylated at Tyr 416 , c-Src, or RhoA. GTP-bound RhoA was measured by association with GST-RBD beads (25). See Fig. S4 for quantitative data.
  • Figure 4 demonstrates a role of Gcc 13 in clot retraction and dynamic RhoA regulation.
  • C, D, E, F Platelets were stimulated with thrombin with or without 2 mM RGDS, and monitored for turbidity changes of platelet suspension caused by shape change and aggregation (C). The platelets were then solubilized at indicated time points and analyzed for amount of ⁇ coimmunoprecipitated with Gcc 13 (D) and amount of GTP-RhoA bound to GST-RBD beads (E) by immunoblot. (F) quantitative data (mean+SD) from 3 experiments.
  • G A schematic illustrating Gcc 13 -dependent dynamic regulation of RhoA and crosstalk between GPCR and integrin signaling.
  • Figure 5 demonstrates the efficiency of platelet replacement by irradiation and transplantation of lentivirus-infected bone marrow stem cells.
  • Green EGFP fluorescence indicating that platelets are derived from transplanted stem cells.
  • Red Alexa Fluor 546-conjugated phalloidin staining indicating total platelets.
  • Figure 6 demonstrates the similar effects of two different G l 3 siRNA on platelet spreading, c-Src activation and RhoA activity, and the effect of aspirin on platelet spreading.
  • A Confocal microscopy images of spreading mouse platelets transfected with scrambled siRNA, Ga 13 siRNA #1- and Ga 13 siRNA #2 on immobilized fibrinogen. Merged EGFP green fluorescence and Alexa Fluor 546-conjugated phalloidin red fluorescence.
  • B Scrambled siRNA, Ga 13 siRNA#l- and Ga 13 siRNA#2-transfected platelets were allowed to adhere to immobilized fibrinogen for the indicated time, and analyzed for c-Src activation and RhoA activity.
  • Figure 7 demonstrates the inhibitory effects of Ga 13 siRNA on cell spreading and c-Src phosphorylation in CHO cells expressing integrin ⁇ 3 ⁇ 4 ⁇ 3 and its rescue by an siRNAresistant mutant of Ga 13 .
  • Stable CHO cell line expressing integrin ⁇ 3 ⁇ 4 ⁇ 3 (123 cells) were transfected with cDNA constructs encoding EGFP and scrambled control siRNA or Ga 13 siRNA with or without co-transfection of Flag-tagged siRNA-resistant mutants of Ga 13 cDNA constructs (Flag-G13- Mutl).
  • FIG. 2A Quantitative data for Fig. 2A, showing that co-immunoprecipitation of ⁇ 3 with Ga 13 was enhanced by GTP and A1F4 " .
  • B Quantitative data for Fig. 2G, showing mSRI inhibited co-immunoprecipitation of ⁇ with Ga 13 .
  • C and D Quantitative data from Fig. 3 A showing that mSRI inhibited c-Src activation (C) and accelerated RhoA activation (D). All data are expressed as mean+SD from 3 experiments. Statistical significance was determined using Student t-test.
  • Figure 9 represents a schematic of Gal3 with switch regions indicated.
  • Two Ga 13 truncation mutants were developed to map the ⁇ binding site (See Fig 2F): (1) the mutant encoding a Ga 13 fragment containing residues 1-196 lacking switch region I, and (2) the mutant encoding a Ga 13 fragment (residues 1-212) containing switch region I.
  • Figure 10 provides typical images of clot retraction showing the effects of mSRI and G( i3- knockdown on platelet-mediated clot retraction.
  • A Effect of 250 ⁇ mSRI peptide on clot retraction of human platelet-rich plasma compared with DMSO and scrambled peptide. Quantitative data are shown in Fig. 4A.
  • B Comparison of clot retraction mediated by control siRNA platelets and Ga 13 -knockdown platelets. Quantitative data are shown in Fig. 4B.
  • Figure 11 demonstrates the Gcc 13 binding site in the integrin ⁇ 3 cytoplasmic domain.
  • A Amino acid sequence alignment of the cytoplasmic domains of various human integrins ⁇ subunits. Key sequences critical in Gccl3 binding is marked as red. Synthetic peptides corresponding to the Gal 3 binding region of ⁇ 3 were synthesized
  • B Lysates from CHO cells expressing a similar level of wild type and truncated integrin ⁇ 3 were immunoprecipitated with anti-Gcc 13 antibody or equal amount of control rabbit IgG. Lysates and immunoprecipitates were immunoblotted with anti-Gcc 13 or anti ⁇ (MAbl5) antibody.
  • Figure 13 demonstrates the identification of a Gcc 13 binding motif in integrins.
  • A Platelets were treated with 500 ⁇ control peptide, mP 1 , mP 7 , or mPs peptides, then
  • Figure 14 demonstrates the inhibition of integrin outside-in and inside-out signalling by mP 1 peptide.
  • A Fluorescence microscopy images of human platelets adherent on fibrinogen that were pretreated with 250 ⁇ myristoylated scrambled control peptide, 250 ⁇ mP 13 peptide.
  • B Flow cytometry analysis of PAR4AP (50 ⁇ M)-induced Oregon-green labelled fibrinogen binding to human platelets pre-treated with DMSO, 150 ⁇ Myr-Scrambled peptide or 150 ⁇ mP 13 peptide.
  • Figure 15 demonstrates that the EEE motif of integrin p 3 -CD is important for Ga 13 binding and cell spreading.
  • A Human integrin cytoplasmic domain sequences were aligned manually. The conserved ExE motifs are highlighted in red. The conserved NxxY and
  • HDR[R/K] motifs are highlighted in bold. Residues are numbered according to the National Center for Biotechnology Information (NCBI) sequence. The sequences of the peptide inhibitors developed in this study are shown below the corresponding ⁇ 3 cytoplasmic domain sequences.
  • B Lysates from truncated integrin p -stable expression 123 cells were precipitated with anti- Ga 13 antibody or equal amount of normal rabbit IgG as control. Immunoprecipitates were immunoblotted with anti-Ga 13 or anti-p 3 (M15) antibody.
  • C Lysates from E to A mutant integrin p 3 -stable expression 123 cells were precipitated with anti-Ga 13 antibody or equal amount of normal rabbit IgG as control.
  • Immunoprecipitates were immunoblotted with anti-Ga 13 or anti- ⁇ 3 (Ml 5) antibody.
  • D Confocal microscopy images of spreading ⁇ _/ ⁇ control platelets or platelets express wide type or AAA-mutant integrin ⁇ 3 on fibrinogen. Merged integrin ⁇ 3 (green) fluorescence and Alex Fluor 546- conjugated phalloidin (Red) fluorescence.
  • E Flow cytometry analysis of wide type and AAA-mutant integrin ⁇ expression level. Human platelets and ⁇ 3 _/ ⁇ platelets serve as positive and negative control.
  • FIG. 16 demonstrates that the AAA mutation is responsible for increased RhoA activity and decreased c-Src activity without affecting integrin inside-out signaling.
  • A Wide type or AAA-mutant 123 cells were allowed to adhere to immobilized fibrinogen, solubilized and analyzed for RhoA activation and c-Src Tyr 416 phosphorylation.
  • B Quantitation of RhoA activity for A.
  • C Quantitation of c-Src Tyr 416 phosphorylation for A.
  • D Lysates from wide type or AAA-mutant integrin ⁇ 3 -stable expression 123 cells were precipitated with anti-c-Src antibody or equal amount of normal rabbit IgG as control. Immunoprecipitates were
  • FIG. 17 Flow cytometry analysis of wide type and AAA-mutant integrin ⁇ 3 - ⁇ 88 ⁇ platelets fibrinogen binding ability induced by Par 4 -. Human platelets and ⁇ 3 _/ ⁇ platelets serve as positive and negative control.
  • Figure 17 demonstrates that the Myr-P 5 peptide inhibited platelets aggregation and spreading due to increased RhoA activity and decreased c-Src activity without affecting integrin inside-out signaling.
  • A Human platelets treated with DMSO, Myr-Scramble or Myr-Ps peptide were allowed to adhere to immobilized fibrinogen, solubilized and analyzed for RhoA activation and c-Src Tyr 416 phosphorylation.
  • FIG. 18 demonstrates that Talin head could not rescue AAA-mutant 123 cells spreading-deficiency on fibrinogen.
  • A C
  • Purified GST-p 3 -CD bound to glutathione beads was mixed with purified Ga 13 with or without purified talin. Bound proteins were immunoblotted with anti-Ga 13 or anti-talin antibody. Quantitative data are shown as mean+SD and p value (t- test).
  • B, D Quantitation of GST-p 3 -CD bound Ga 13 or talin for A and C.
  • E Lysates from 123 or AAA cells transfected with talin head were precipitated with anti-p 3 rabbit serum (8053) or eqival amount of pre-immune serum. Immunoprecipitates were immunoblotted with anti-Ga 13 or anti-p antibody.
  • F Quantitation of integrin p -bound talin for E.
  • FIG. 19 demonstrates that the EEE motif of integrin ⁇ is important for mediating cell spreading on immobilized fibrinogen.
  • A Spreading of washed human platelets pre-treated with vehicle (PBS), scramble peptide or P13 peptide on immobilized fibrinogen for 1 hour. Platelets were stained with Alexa Fluor 546— conjugated phalloidin.
  • B Microscopy images of spreading 123 cells or E to A-mutant cells on fibrinogen for 1 hour. Merged integrin ⁇ (green) fluorescence and talin head (Red) fluorescence.
  • Figure 20 demonstrates that the effect of P 5 peptide on platelets spreading and aggregation .
  • A Confocal microscopy images of DMSO, Myr-Scramble or Myr-P 5 peptide treated platelets spreading on fibrinogen for 60 minutes, without or with Y27632. Merged integrin ⁇ 3 (green) fluorescence and Alex Fluor 546-conjugated phalloidin (Red) fluorescence.
  • B, C 30 ⁇ Myr-P 5 peptide inhibited both human (B) and mouse (C) platelets aggregation induced by thrombin.
  • Figure 21 A represents a set of Western blots using ⁇ 3, talin or G l 3 specific antibodies. Platelets treated with DMSO, 500 ⁇ mP13, mP5, or corresponding control peptides were stimulated with 0.025 U/ml thrombin at 22°C, solubilized and immunoprecipitated with anti- or preimmune rabbit serum. Lysates and immunoprecipitates were immunoblotted with anti-Gcc 13 , anti-talin or anti- antibody
  • Figure 21B represents a set of graphs of a flow cytometrical analysis of PAR4AP (50 ⁇ M)-induced Oregon-Green labelled fibrinogen binding to human platelets pre-treated with DMSO, 250 ⁇ Myr-Scrambled peptide or 250 ⁇ mP5 (upper panel), or pre-treated with DMSO, 150 ⁇ Myr-Scrambled peptide or 150 ⁇ mP13 (lower panel).
  • Figure 21C representas a set of fluorescence microscopy images of phalloidin- stained human platelets spreading on fibrinogen for lhr. Platelets were pre-treated with 0.05% DMSO, 250 ⁇ myristoylated mP5, mP13, or the corresponding control peptides for 5 min at room temperature.
  • Figure 22 is an illustration depicting the differences in inside-out and outside-in signaling upon vascular injury treated with a current integrin antagonist vs. an outside-in signal inhibitor.
  • Figure 23 is an illustration depicting signal transduction pathways of integrins.
  • Figure 24 represents a set of graphs depicting the differences in platelet ATP secretion (top) and % platelet aggregation (bottom) upon treatment with mP5 peptide or a scambled control peptide thereof.
  • Figure 25 is an illustration of a micelle.
  • Figure 26 represents a graph of tail bleeding time in mice treated with Integrillin or a saline control. Median value indicated.
  • Figure 27 represents a graph of ATP secretion by platelets stimulated with 0.1 U thrombin and pre-incubated with FEEERA (SEQ ID NO: 87) peptide dissolved in DMSO or the control scrambled peptide thereof.
  • FEEERA SEQ ID NO: 87
  • Figure 28 represents a graph of the occlusion time of mice treated with an EXE motif peptide or the scrambled peptide control.
  • Figure 29 represents a graph of turbidometric measurement of platelet aggregation showing the doses of the EXE motif peptide FEEERA (SEQ ID NO: 87) required for inhibition of platelet aggregation among the peptide in micellar form vs. the peptide dissolved in DMSO.
  • Figure 30A represents a table including scores of ability to inhibit platelet aggregation, for varying doses of mP5 or the peptide of SEQ ID NO: 87.
  • Figure 30B represents a set of graphs depicting platelet aggregation traces for the mP5 peptide, the scambled control of mP5, the peptide of SEQ ID NO: 87 or its scrambled control.
  • Fig ure 31 demonstrates the mutually exclusive binding nature of talin and Gcc 13 to ⁇ 3 .
  • Immunoprecipitates and CHO cell lysates (10% of that used in immunoprecipitation) were immunoblotted (IB) with indicated antibodies, (c) Binding of purified recombinant Gcc 13 to glutathione bead-bound glutathione S-transferase (GST), GST ⁇
  • cytoplasmic domain fusion protein (d) Coimmunoprecipitation of CHO cell-expressed Wt or ExE motif mutated ⁇ with Gcc 13 and talin using anti ⁇ 3 or pre-immune rabbit serum, (e) Coimmunoprecipitation of CHO cell-expressed integrin ( ⁇ 3 ⁇ 4 ⁇ 3 with Gcc 13 and THD following transfection with cDNA encoding THD. (f, g) Inhibition of the binding of THD (20 nM) (f) or Gcc 13 (40 nM) (g) to immobilized GST ⁇ 3 CD proteins (Wt and negative control mutants) by increasing concentrations of Gcc 13 (f) or THD (g). Bound Ga 13 or THD was detected using anti-Gcc 13 or anti-talin.
  • Figure 32 demonstrates the dynamics of talin and Gcc 13 binding to ⁇ and the role of talin in integrin signaling
  • (a, b and c) Human platelets were stimulated with 0.025 U/ml a- thrombin (in an aggregometer) with or without 2 mM integrin inhibitor RGDS, solubilized at various time points, immunoprecipitated with anti ⁇ or pre-immune rabbit serum, and immunoblotted for Gcc 13 , talin, and ⁇ (additional controls in E.D. Fig 3d), (a) Typical immunoblots.
  • Figure 33 demonstrates the selective role of Gcc 13 -ExE binding in integrin outside-in signaling
  • (a) Flow cytometric analysis of ⁇ expression in platelets from ⁇ _/ ⁇ mice transplanted with Wt or AAA-mutant -transfected bone marrow stem cells.
  • ⁇ _/ ⁇ platelets served as negative control
  • (b) Mouse platelets expressing Wt or AAA mutant ⁇ 3 were stimulated with 0.025 U/ml a-thrombin, solubilized at various time points, immunoprecipitated with anti ⁇ 3 or pre-immune rabbit serum, and immunoblotted for Gcc 13 , talin, and ⁇ 3 .
  • Figure 34 demonstrates that the new anti-thrombotic that does not cause bleeding, (a) The effects of 500 ⁇ mP 13 , mP 6 , or mPs on co-immunoprecipitation of ⁇ 3 with Gcc 13 or talin in thrombin- stimulated platelets in comparison with scrambled controls (also see E.D. Fig 7).
  • Figure 35 represents a schematic showing how selective inhibitors of integrin outside- in signaling work as anti-thrombotics. Blue arrows indicate steps that are inhibited.
  • Figure 36 demonstrates the importance of the conserved ExE motif in integrin' s interaction with Gcc 13 .
  • Immunoprecipitates and lysates were immunoblotted with anti-Ga 13 and anti- 3 antibodies, (c and d) GST- 3 CD (Wt) or GST- 3AAACD (AAA mutant) proteins immobilized in Glutathione-coated microtiter wells were incubated with increasing concentrations of Gcc 13 (c), or increasing concentrations of talin head domain (THD) (d).
  • Gcc 13 and THD were respectively detected using anti- Gcc 13 or anti-talin (mouse IgG was used as a specificity control) followed by secondary HRP- labeled anti-IgG antibody, (e)
  • conserved mutations of EEE to DED and EEE to QSE were introduced to the ⁇ 3 cytoplasmic domain. These mutants were co-transfected with Wt ⁇ 3 ⁇ 4 3 ⁇ 4 into CHO cells, which were sorted to achieve comparable expression levels with Wt ccnb 3 -expressing cells (as shown in E.D. Fig. 4e).
  • Lysates from these cells were immunoprecipitated with anti- 3 or equal amount of pre-immune rabbit serum. Lysates (10%) and immunoprecipitates were immunoblotted with anti-Gcc 13 or anti- .
  • Lysates from human platelets (with or without stimulation with 0.025 u/ml thrombin) were immunoprecipitated with anti-Gcc 13 antibody or equal amount of control rabbit IgG.
  • Immunoprecipitates were immunoblotted with anti-Gcc 13 and anti- i antibodies.
  • Gcc 13 is associated with ⁇ 1; which is significantly increased following thrombin stimulation.
  • Figure 37 demonstrates that ligand occupancy induces switch of integrin ( ⁇ 3 ⁇ 4 ⁇ 3 from talin-bound to the Gcc 13 -bound state
  • integrin activation and ligand occupancy induces switch of integrin ( ⁇ 3 ⁇ 4 ⁇ 3 from talin-bound to the Gcc 13 -bound state
  • human platelets were incubated with or without 1 mM MnCl 2 and 30 g/ml fibrinogen for 5 minutes at 22°C. Platelet lysates were then immunoprecipitated with anti- 3 or pre-immune rabbit serum. Lysates (10%) and immunoprecipitates were immunoblotted with anti- 3 or anti-Gcc 13 .
  • Pellets were dissolved in SDS sample buffer to the same volume as the lysates after diluting them 1: 1 with 2x SDS sample buffer, and both were immunoblotted with anti ⁇ and anti-talin antibodies. Note that the levels of talin and ⁇ in platelet lysates kept essentially constant during the course of platelet aggregation and, with low concentrations of thrombin used to stimulate platelets, very little insoluble ⁇ and talin were present in the pellet, which were detectable only after prolonged exposure (5 min exposure compared to 10 sec of normal exposure time) and with no obvious variation during the course of platelet aggregation.
  • Figure 39 demonstrates the effects of AAA mutation on integrin outside-in signaling in platelets
  • (a) Flow cytometric analysis of integrin ⁇ 3 ⁇ 4 3 ⁇ 4 / ⁇ 3 expression levels in ⁇ _/ ⁇ mouse platelets transfected with Wt or AAA mutant ⁇ using bone marrow stem cell transplantation technology in comparison with C57BL/6 mouse platelets.
  • ⁇ _/ ⁇ platelets were used as a negative control.
  • ( ⁇ 3 ⁇ 4 ⁇ 3 complex was detected using an anti-mouse ⁇ 3 ⁇ 4 3 ⁇ 4 antibody
  • (b) Mouse platelets expressing recombinant Wt or AAA mutant ⁇ 3 as in (a) were lysed and immunoprecipitated with anti ⁇ 3 or equal amounts of pre-immune rabbit serum.
  • Figure 40 demonstrates the effects of mutational disruption of the ExE motif on integrin outside-in signaling,
  • Mouse IgG was used as a negative control
  • Wt or AAA-mutant OCm ⁇ -expressing CHO-lb9 cells were allowed to adhere to immobilized fibrinogen, solubilized at various time points, and analyzed for RhoA activation and c-Src Tyr416 phosphorylation.
  • Figure 41 demonstrates that mP 6 selectively inhibits integrin outside-in signaling without affecting inside-out signaling
  • Washed human platelets were stimulated with 0.025 U/ml a-thrombin in the absence or presence of 250 ⁇ myristoylated peptides, mP 13 (a, b) and mP 6 (c, d) with stirring (1000 rpm) at 37°C, and then solubilized at various time points. Lysates were immunoprecipitated with anti ⁇ 3 rabbit serum or equal amounts of pre- immune serum.
  • Integrilin-treated platelets were used as negative control, (h) Flow cytometric analysis of PAR4AP-induced Oregon-Green labeled soluble fibrinogen binding to human platelets pre-treated with solvent DMSO, mP 13 Scr or mP 13 . Resting platelets were used as a negative control.
  • Figure 42 demonstrates the in vivo effect of mP 6 : selective inhibition of thrombosis but not hemostasis.
  • (a) Representative images of laser-induced mouse cremaster arteriolar thrombosis (red) in the context of the bright-field microvascular histology, visualized by infusion of nonblocking rat anti-mouse GPIbp antibody conjugated to DyLight 649. The C57BL/6 mice were injected with 5 ⁇ /kg micellar formulated mP 6 or mPeSrc (negative control), 12 ⁇ /kg Integrillin or buffer, 3 minutes before laser- induced arteriolar wall injury. White arrows indicate the directions of the blood flow, (b) The mean platelet fluorescence intensity for 30 thrombi (performed in 3 mice) for each treatment at selected time points (mean ⁇ SE, n 30, t-test).
  • Figure 43 demonstrates platelet uptake of mP 6 and mP 6 Scr, and no effect of mP6 on hemogram.
  • (a) Estimation of intracellular levels of PDAM-conjugated mP 6 and mP 6 Scr following incubation with platelets for 5 minutes. Platelets were pelleted by centrifugation, and the amounts of PDAM-conjugated peptides in platelet lysates were estimated (mean ⁇ SD, n 3).
  • Figure 44 is a collection of graphs illustrating fibrinogen binding to integrins on platelets.
  • the platelets were pre-incubated with myristoylated peptides mP5 (P5), mP6 (P6), mP7 (P7), or mP13 (P13), or their scrambled counterparts (mP5Scr (P5Scr), mP6Scr (P6Scr), mP7Scr (P7Scr), or mP13Scr (P13Scr)) or Integrilin, prior to being stimulated with PAR4.
  • the fibrinogen binding to resting platelets is shown (line marked as Resting) and served as a baseline control.
  • Figure 45 is a collection of graphs illustrating thrombin- stimulated platelet aggregation of platelets preincubated with myristoylated peptides mP6 (P6), mP7 (P7), or mP13 (P13), or their scrambled counterparts (mP6Scr (P6Scr), mP7Scr (P7Scr), or mP13Scr (P13Scr)).
  • Figure 46 is a graph illustrating platelet adhesion to immobilized fibrinogen of platelets preincubated with myristoylated peptides mP6 (P6), mP7 (P7), or mP13 (P13), or their scrambled counterparts (mP6Scr (P6Scr), mP7Scr (P7Scr), or mP13Scr (P13Scr)) or with HEPES buffer or Integrilin.
  • the compounds of the invention may be considered as inhibitors of a ⁇ integrin binding to a G protein a subunit and/or inhibitors of a G protein a subunit binding to a ⁇ integrin.
  • the compounds are competitive binding inhibitors.
  • the compounds bind to the site of a ⁇ integrin to which a G protein a subunit binds.
  • the compounds bind to the site of a G protein a subunit to which a ⁇ integrin binds.
  • the compounds are noncompetitive binding inhibitors.
  • the compounds inhibit the binding interaction between a ⁇ integrin and a G protein a subunit, yet the compounds bind to a site of ⁇ integrin other than the site to which G protein a subunit binds or the compounds bind to a site of a G protein a subunit other than the site to which ⁇ integrin binds.
  • the inhibition provided by the compounds of the invention may not be a 100% or complete inhibition or abrogation of the binding interaction between the ⁇ integrin and G protein a subunit. Rather, there are varying degrees of inhibition of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect. In this respect, the compounds of the invention may inhibit the binding interaction between a ⁇ integrin and a G protein a subunit to any amount or level.
  • the compound provides at least or about a 10% inhibition (e.g., at least or about a 20% inhibition, at least or about a 30% inhibition, at least or about a 40% inhibition, at least or about a 50% inhibition, at least or about a 60% inhibition, at least or about a 70% inhibition, at least or about a 80% inhibition, at least or about a 90% inhibition, at least or about a 95% inhibition, at least or about a 98% inhibition) of the binding between a ⁇ integrin and G protein a subunit.
  • a 10% inhibition e.g., at least or about a 20% inhibition, at least or about a 30% inhibition, at least or about a 40% inhibition, at least or about a 50% inhibition, at least or about a 60% inhibition, at least or about a 70% inhibition, at least or about a 80% inhibition, at least or about a 90% inhibition, at least or about a 95% inhibition, at least or about a 98% inhibition
  • the compound completely abrogates the binding interaction between the ⁇ integrin and the G protein a subunit, such that no ⁇ integrin-G protein a subunit binding complexes are detectable in a sample obtained from a subject, as measured by, for example, immunoprecipitation, Western blotting,
  • the compounds inhibit the binding interaction between a wild-type human ⁇ integrin and a wild-type human G protein a subunit.
  • the wild-type human G protein a subunit is a wild-type human Ga 12 or a wild-type human Ga 13 .
  • the wild- type human ⁇ integrin is a wildtype human ⁇ 1 ⁇ integrin, wild-type human ⁇ integrin, wild-type human ⁇ 2 integrin, wildtype human ⁇ 3 integrin, wildtype human ⁇ 5 integrin, wildtype human ⁇ 6 integrin, or wildtype human ⁇ 7 integrin.
  • amino acid sequences of these wild-type human proteins are known in the art and are available in the Protein database of the National Center for Biotechnology Information (NCBI) website as NCBI Reference Sequence Nos. NP_006563.2 (Ga 13 ), NP_031379.2 (Ga 12 ), NP_002202 ( ⁇ ⁇ integrin), NP_391988 ( ⁇ 10 integrin), NP_000202 ( ⁇ 2 integrin), NP_000203 ( ⁇ 3 integrin), NP_002204.2 ( ⁇ 5 integrin), NP_000879.2 ( ⁇ 6 integrin), and NP_000770.1 ( ⁇ 7 integrin).
  • the amino acid sequences also are provided herein as SEQ ID NOs: 1-4 and 10-18.
  • the compounds inhibit the binding interaction between a ⁇ integrin and a G protein a subunit, wherein one or both of the ⁇ integrin and the G protein a subunit is/are not-wild-type, e.g., mutant.
  • the amino acid sequence of the mutant ⁇ integrin differs at one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, or more) positions from the amino acid sequence of a wild-type human ⁇ integrin recognized in the art (e.g., ⁇ 1 ⁇ integrin, ⁇ 10 integrin ⁇ 2 integrin, ⁇ 3 integrin, ⁇ 5 integrin, ⁇ 6 integrin, ⁇ 7 integrin).
  • a wild-type human ⁇ integrin recognized in the art (e.g., ⁇ 1 ⁇ integrin, ⁇ 10 integrin ⁇ 2 integrin, ⁇ 3 integrin, ⁇ 5 integrin, ⁇ 6 integrin, ⁇ 7 integrin).
  • the amino acid sequence of the mutant ⁇ integrin in exemplary aspects is about 98% or less (e.g., about 95% or less, about 90% or less, about 85% or less, about 80% or less, about 75% or less, about 70% or less, about 65% or less, about 60% or less, about 55% or less, about 50% or less) identical to that of a wild-type ⁇ integrin.
  • the amino acid sequence of the mutant G protein a subunit differs at one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, or more) positions from the amino acid sequence of a wild-type human G protein a subunit recognized in the art (e.g., Ga 12 , Ga 13 ).
  • the amino acid sequence of the mutant G protein a subunit in exemplary aspects is about 98% or less (e.g., about 95% or less, about 90% or less, about 85% or less, about 80% or less, about 75% or less, about 70% or less, about 65% or less, about 60% or less, about 55% or less, about 50% or less) identical to that of a wild-type G protein a subunit.
  • the compound is an antibody, an antibody analog, a peptide, a peptide analog (e.g., peptoid, peptidomimetic), a nucleic acid molecule encoding any of the antibodies or peptides, or analogs thereof, or a small molecule compound (e.g., small molecule compound rationally designed based on any of the antibodies or peptides described herein).
  • a peptide e.g., peptoid, peptidomimetic
  • nucleic acid molecule e.g., a nucleic acid molecule encoding any of the antibodies or peptides, or analogs thereof
  • a small molecule compound e.g., small molecule compound rationally designed based on any of the antibodies or peptides described herein.
  • the compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit comprises an antibody, or antigen binding fragment thereof.
  • the compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit is an antibody, or antigen binding fragment thereof.
  • the antibody may be any type of immunoglobulin known in the art.
  • the antibody is an antibody of isotype IgA, IgD, IgE, IgG, or IgM.
  • the antibody in some embodiments is a monoclonal antibody. In other embodiments, the antibody is a polyclonal antibody.
  • the antibody is a naturally-occurring antibody, e.g., an antibody isolated and/or purified from a mammal, e.g., mouse, rabbit, goat, horse, chicken, hamster, human, and the like.
  • the antibody may be considered as a mammalian antibody, e.g., a mouse antibody, rabbit antibody, goat antibody, horse antibody, chicken antibody, hamster antibody, human antibody, and the like.
  • Methods of producing naturally- occurring antibodies are known in the art, some of which are described further herein under the section entitled "Methods of Antibody Production.”
  • the antibody is a genetically-engineered antibody, e.g., a single chain antibody, a humanized antibody, a chimeric antibody, a CDR-grafted antibody, an antibody which includes portions of CDR sequences specific for a ⁇ integrin or a G protein a subunit, a humaneered antibody, a bispecific antibody, a trispecific antibody, and the like.
  • a genetically-engineered antibody e.g., a single chain antibody, a humanized antibody, a chimeric antibody, a CDR-grafted antibody, an antibody which includes portions of CDR sequences specific for a ⁇ integrin or a G protein a subunit, a humaneered antibody, a bispecific antibody, a trispecific antibody, and the like.
  • the genetically-engineered antibody is a single chain antibody (SCA) specific for a ⁇ integrin or a G protein a subunit.
  • SCA single chain antibody
  • the SCA binds to the site of a ⁇ integrin to which G protein a subunit binds or the SCA binds to the site of a G protein a subunit to which a ⁇ integrin binds.
  • the SCA binds to an epitope as further described herein under the section entitled "Epitopes.” Methods of making SCAs are known in the art. See, for example, Davis et al., Nature Biotechnology 9: 165-169 (1991).
  • the antibody is a chimeric antibody.
  • the term "chimeric antibody” is used herein to refer to an antibody containing constant domains from one species and the variable domains from a second, or more generally, containing stretches of amino acid sequence from at least two species.
  • the chimeric antibody binds to the site of a ⁇ integrin to which a G protein a subunit binds or the chimeric antibody binds to the site of a G protein a subunit to which a ⁇ integrin binds.
  • the chimeric antibody binds to an epitope as further described herein under the section entitled "Epitopes.”
  • the antibody is a humanized antibody.
  • humanized when used in relation to antibodies refers to antibodies having at least CDR regions from a non-human source which are engineered to have a structure and immunological function more similar to true human antibodies than the original source antibodies.
  • humanizing can involve grafting CDR from a non-human antibody, such as a mouse antibody, into a human antibody.
  • Humanizing also can involve select amino acid substitutions to make a non-human sequence look more like a human sequence.
  • the humanized antibody binds to the site of a ⁇ integrin to which a G protein a subunit binds or the humanized antibody binds to the site of a G protein a subunit to which a ⁇ integrin binds.
  • the humanized antibody binds to an epitope as further described herein under the section entitled "Epitopes.”
  • chimeric or humanized herein is not meant to be mutually exclusive, and rather, is meant to encompass chimeric antibodies, humanized antibodies, and chimeric antibodies that have been further humanized. Except where context otherwise indicates, statements about (properties of, uses of, testing of, and so on) chimeric antibodies of the invention apply to humanized antibodies of the invention, and statements about humanized antibodies of the invention pertain also to chimeric antibodies. Likewise, except where context dictates, such statements also should be understood to be applicable to antibodies and antigen binding fragments of such antibodies of the invention.
  • the antibody is a CDR-grafted antibody specific for a ⁇ integrin or a G protein a subunit.
  • the CDR-grafted antibody binds to the site of a ⁇ integrin to which a G protein a subunit binds or the CDR-grafted antibody binds to the site of a G protein a subunit to which a ⁇ integrin binds.
  • the CDR-grafted antibody binds to an epitope as further described herein under the section entitled "Epitopes.” Methods of making CDR-grafted antibodies are known in the art. See, for example, Lo, Benny, Antibody
  • the antibody is a bispecific or trispecific antibody specific for a ⁇ integrin or a G protein a subunit.
  • the bispecific or trispecific antibody binds to the site of a ⁇ integrin to which a G protein a subunit binds or the bispecific or trispecific antibody binds to the site of a G protein a subunit to which a ⁇ integrin binds.
  • the bispecific or trispecific antibody binds to an epitope as further described herein under the section entitled "Epitopes.” Methods of making bispecific or trispecific antibodies are known in the art. See, for example, Marvin and Zhu, Acta Pharmacologica Sinica 26: 649-658 (2005) and U.S. Patent 6,551,592.
  • the antibody is a humaneeredTM antibody.
  • Humaneering technology is a proprietary method of KaloBios Pharmaceuticals, Inc. (San Francisco, California) for converting non human antibodies into engineered human antibodies.
  • HumaneeredTM antibodies are high affinity, and highly similar to human germline antibody sequences.
  • the antibody has a level of affinity or avidity for the ⁇ integrin which is sufficient to prevent the G protein a subunit from binding to the ⁇ integrin. In some embodiments, the antibody has a level of affinity or avidity for the G protein a subunit which is sufficient to prevent a ⁇ integrin from binding G protein a subunit. Therefore in some embodiments, the affinity constant, K a , (which is the inverterd dissocation constant, K ⁇ j) of the antibody of the invention for the a ⁇ integrin is greater than the K a of G protein a subunit for the ⁇ integrin.
  • the K a of the antibody of the invention for the G protein a subunit is greater than that of the ⁇ integrin for the G protein a subunit.
  • Binding constants may be determined by methods known in the art, including, for example, methods which utilize the principles of surface plasmon resonance, e.g., methods utilizing a BiacoreTM system.
  • the antibody is in monomeric form, while in other words
  • the antibody is conjugated to one or more antibodies (e.g., each of which recognize the same epitope of the first antibody). Accordingly, in some aspects, the antibody is in polymeric, oligomeric, or multimeric form. In certain embodiments in which the antibody comprises two or more distinct antigen binding regions fragments, the antibody is considered bispecific, trispecific, or multi- specific, or bivalent, trivalent, or multivalent, depending on the number of distinct epitopes that are recognized and bound by the antibody.
  • the compound which inhibits a binding interaction between an a ⁇ integrin and a G protein a subunit is an antigen binding fragment of an antibody.
  • the antigen binding fragment (also referred to herein as "antigen binding portion") may be an antigen binding fragment of any of the antibodies described herein.
  • the antigen binding fragment can be any part of an antibody that has at least one antigen binding site, including, but not limited to, Fab, F(ab') 2 , dsFv, sFv, diabodies, triabodies, bis-scFvs, fragments expressed by a Fab expression library, domain antibodies, VhH domains, V-NAR domains, VH domains, VL domains, and the like.
  • Antibody fragments of the invention are not limited to these exemplary types of antibody fragments.
  • a domain antibody comprises a functional binding unit of an antibody, and can correspond to the variable regions of either the heavy (V H ) or light (V L ) chains of antibodies.
  • a domain antibody can have a molecular weight of approximately 13 kDa, or approximately one- tenth of a full antibody. Domain antibodies may be derived from full antibodies such as those described herein.
  • the antigen binding fragments in some embodiments are monomeric or polymeric, bispecific or trispecific, bivalent or trivalent.
  • Antibody fragments that contain the antigen binding, or idiotype, of the antibody molecule may be generated by techniques known in the art.
  • fragments include, but are not limited to, the F(ab') 2 fragment which may be produced by pepsin digestion of the antibody molecule; the Fab' fragments which may be generated by reducing the disulfide bridges of the F(ab') 2 fragment, and the two Fab' fragments which may be generated by treating the antibody molecule with papain and a reducing agent.
  • a single-chain variable region fragment (sFv) antibody fragment which consists of a truncated Fab fragment comprising the variable (V) domain of an antibody heavy chain linked to a V domain of a light antibody chain via a synthetic peptide, can be generated using routine recombinant DNA technology techniques (see, e.g., Janeway et al., supra).
  • dsFv disulfide- stabilized variable region fragments
  • dsFv can be prepared by recombinant DNA technology (see, e.g., Reiter et al., Protein Engineering, 7, 697-704 (1994)).
  • Recombinant antibody fragments e.g., scFvs
  • scFvs can also be engineered to assemble into stable multimeric oligomers of high binding avidity and specificity to different target antigens.
  • diabodies dimers
  • triabodies trimers
  • tetrabodies tetramers
  • Bispecific antibodies are molecules comprising two single-chain Fv fragments joined via a glycine- serine linker using recombinant methods.
  • the V light-chain (V L ) and V heavy-chain (V H ) domains of two antibodies of interest in exemplary embodiments are isolated using standard PCR methods.
  • the V L and V H CDNA'S obtained from each hybridoma are then joined to form a single-chain fragment in a two-step fusion PCR.
  • Bispecific fusion proteins are prepared in a similar manner.
  • Bispecific single-chain antibodies and bispecific fusion proteins are antibody substances included within the scope of the present invention. Exemplary bispecific antibodies are taught in U.S. Patent Application Publication No. 2005-0282233A1 and
  • Suitable methods of making antibodies are known in the art. For instance, standard hybridoma methods are described in, e.g., Harlow and Lane (eds.), Antibodies: A Laboratory Manual, CSH Press (1988), and CA. Janeway et al. (eds.), Immunobiology, 5 th Ed., Garland Publishing, New York, NY (2001)). [0098] Briefly, a polyclonal antibody is prepared by immunizing an animal with an immunogen comprising a polypeptide of the present invention and collecting antisera from that immunized animal. A wide range of animal species can be used for the production of antisera.
  • an animal used for production of anti-antisera is a non-human animal including rabbits, mice, rats, hamsters, goat, sheep, pigs or horses. Because of the relatively large blood volume of rabbits, a rabbit is a preferred choice for production of polyclonal antibodies.
  • 50 ⁇ g of ⁇ integrin antigen is emulsified in Freund's Complete Adjuvant for immunization of rabbits. At intervals of, for example, 21 days, 50 ⁇ g of epitope are emulsified in Freund's Incomplete Adjuvant for boosts.
  • Polyclonal antisera may be obtained, after allowing time for antibody generation, simply by bleeding the animal and preparing serum samples from the whole blood.
  • Monoclonal antibodies for use in the invention may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include but are not limited to the hybridoma technique originally described by Koehler and Milstein (Nature 256: 495-497, 1975), the human B-cell hybridoma technique (Kosbor et al., Immunol Today 4:72, 1983; Cote et al., Proc Natl Acad Sci 80: 2026-2030, 1983) and the EBV- hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R Liss Inc, New York N.Y., pp 77-96, (1985).
  • a mouse is injected periodically with recombinant ⁇ integrin against which the antibody is to be raised (e.g., 10-20 ⁇ g emulsified in Freund's Complete Adjuvant).
  • ⁇ integrin against which the antibody is to be raised e.g. 10-20 ⁇ g emulsified in Freund's Complete Adjuvant.
  • the mouse is given a final pre-fusion boost of an ⁇ integrin polypeptide in PBS, and four days later the mouse is sacrificed and its spleen removed.
  • the spleen is placed in 10 ml serum- free RPMI 1640, and a single cell suspension is formed by grinding the spleen between the frosted ends of two glass microscope slides submerged in serum-free RPMI 1640, supplemented with 2 mM L-glutamine, 1 mM sodium pyruvate, 100 units/ml penicillin, and 100 ⁇ g/ml streptomycin (RPMI) (Gibco, Canada).
  • RPMI streptomycin
  • the cell suspension is filtered through sterile 70-mesh Nitex cell strainer (Becton Dickinson, Parsippany, N.J.), and is washed twice by centrifuging at 200 g for 5 minutes and resuspending the pellet in 20 ml serum-free RPMI.
  • Splenocytes taken from three naive Balb/c mice are prepared in a similar manner and used as a control.
  • NS-1 myeloma cells kept in log phase in RPMI with 11% fetal bovine serum (FBS) (Hyclone Laboratories, Inc., Logan, Utah) for three days prior to fusion, are centrifuged at 200 g for 5 minutes, and the pellet is washed twice.
  • FBS fetal bovine serum
  • Spleen cells (1 x 10 8 ) are combined with 2.0 x 10 7 NS-1 cells and centrifuged, and the supernatant is aspirated. The cell pellet is dislodged by tapping the tube, and 1 ml of 37 °C. PEG 1500 (50% in 75 mM Hepes, pH 8.0) (Boehringer Mannheim) is added with stirring over the course of 1 minute, followed by the addition of 7 ml of serum-free RPMI over 7 minutes. An additional 8 ml RPMI is added and the cells are centrifuged at 200 g for 10 minutes.
  • the pellet After discarding the supernatant, the pellet is resuspended in 200 ml RPMI containing 15% FBS, 100 ⁇ sodium hypoxanthine, 0.4 ⁇ aminopterin, 16 ⁇ thymidine (HAT) (Gibco), 25 units/ml IL-6 (Boehringer Mannheim) and 1.5 x 10 6 splenocytes/ml and plated into 10 Corning flat- bottom 96-well tissue culture plates (Corning, Corning N.Y.).
  • Plates are washed three times with PBS with 0.05% Tween 20 (PBST) and 50 ⁇ culture supernatant is added. After incubation at 37° C. for 30 minutes, and washing as above, 50 ⁇ of horseradish peroxidase conjugated goat anti-mouse IgG(fc) (Jackson ImmunoResearch, West Grove, Pa.) diluted 1:3500 in PBST is added. Plates are incubated as above, washed four times with PBST, and 100 ⁇ substrate, consisting of 1 mg/ml o-phenylene diamine (Sigma) and 0.1 ⁇ /ml 30% ⁇ 2 0 2 in 100 mM Citrate, pH 4.5, are added. The color reaction is stopped after 5 minutes with the addition of 50 ⁇ of 15% H 2 S0 4 . A 490 is read on a plate reader (Dynatech).
  • Selected fusion wells are cloned twice by dilution into 96-well plates and visual scoring of the number of colonies/well after 5 days.
  • the monoclonal antibodies produced by hybridomas are isotyped using the Isostrip system (Boehringer Mannheim, Indianapolis, Ind.).
  • myeloma cell lines may be used.
  • Such cell lines suited for use in hybridoma-producing fusion procedures preferably are non-antibody- producing, have high fusion efficiency, and enzyme deficiencies that render them incapable of growing in certain selective media which support the growth of only the desired fused cells (hybridomas).
  • the immunized animal is a mouse
  • rats one may use R210.RCY3, Y3-Ag 1.2.3, IR983F and 4B210
  • U-266, GM1500-GRG2, LICR-LON-HMy2 and UC729-6 are all useful in connection with cell fusions.
  • the hybridomas and cell lines produced by such techniques for producing the monoclonal antibodies are contemplated to be novel compositions of the invention.
  • adjuvants include but are not limited to Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
  • BCG Bacilli Calmette-Guerin
  • Corynebacterium parvum are potentially useful human adjuvants.
  • Antibodies may also be produced by inducing in vivo production in the lymphocyte population or by screening recombinant immunoglobulin libraries or panels of highly specific binding reagents as disclosed in Orlandi et al (Proc Natl Acad Sci 86: 3833-3837; 1989), and Winter G and Milstein C (Nature 349: 293-299, 1991).
  • Phage display furthermore can be used to generate the antibody of the invention.
  • phage libraries encoding antigen-binding variable (V) domains of antibodies can be generated using standard molecular biology and recombinant DNA techniques (see, e.g.,
  • Phage encoding a variable region with the desired specificity are selected for specific binding to the desired antigen, and a complete or partial antibody is reconstituted comprising the selected variable domain.
  • Nucleic acid sequences encoding the reconstituted antibody are introduced into a suitable cell line, such as a myeloma cell used for hybridoma production, such that antibodies having the characteristics of
  • Antibodies can be produced by transgenic mice that are transgenic for specific heavy and light chain immunoglobulin genes. Such methods are known in the art and described in, for example U.S. Patents 5,545,806 and 5,569,825, and Janeway et al., supra.
  • Humanized antibodies can also be generated using the antibody resurfacing technology described in U.S. Patent 5,639,641 and Pedersen et al., J. Mol. Biol, 235, 959-973 (1994).
  • a preferred chimeric or humanized antibody has a human constant region, while the variable region, or at least a CDR, of the antibody is derived from a non-human species.
  • a humanized antibody has one or more amino acid residues introduced into its framework region from a source which is non-human. Humanization can be performed, for example, using methods described in Jones et al. ⁇ Nature 321: 522-525, 1986), Riechmann et al., (Nature, 332: 323-327, 1988) and Verhoeyen et al. (Science 239: 1534- 1536, 1988), by substituting at least a portion of a rodent complementarity-determining region (CDRs) for the corresponding regions of a human antibody. Numerous techniques for preparing engineered antibodies are described, e.g., in Owens and Young, J. Immunol. Meth., 168: 149-165 (1994). Further changes can then be introduced into the antibody framework to modulate affinity or immunogenicity.
  • CDRs rodent complementarity-determining region
  • compositions comprising CDRs are generated.
  • Complementarity determining regions are characterized by six polypeptide loops, three loops for each of the heavy or light chain variable regions.
  • the amino acid position in a CDR is defined by Kabat et al., "Sequences of Proteins of Immunological Interest," U.S. Department of Health and Human Services, (1983), which is incorporated herein by reference.
  • hypervariable regions of human antibodies are roughly defined to be found at residues 28 to 35, from 49-59 and from residues 92-103 of the heavy and light chain variable regions (Janeway and Travers, Immunobiology, 2 nd Edition, Garland Publishing, New York, (1996)).
  • the murine CDR also are found at approximately these amino acid residues. It is understood in the art that CDR regions may be found within several amino acids of these approximated residues set forth above.
  • An immunoglobulin variable region also consists of four "framework" regions surrounding the CDRs (FR1-4). The sequences of the framework regions of different light or heavy chains are highly conserved within a species, and are also conserved between human and murine sequences.
  • compositions comprising one, two, and/or three CDRs of a heavy chain variable region or a light chain variable region of a monoclonal antibody are generated.
  • Techniques for cloning and expressing nucleotide and polypeptide sequences are well-established in the art (see e.g. Sambrook et al., Molecular Cloning: A Laboratory Manual, 2 nd Edition, Cold Spring Harbor, New York (1989)).
  • the amplified CDR sequences are ligated into an appropriate plasmid.
  • the plasmid comprising one, two, three, four, five and/or six cloned CDRs optionally contains additional polypeptide encoding regions linked to the CDR.
  • Framework regions (FR) of a murine antibody are humanized by substituting compatible human framework regions chosen from a large database of human antibody variable sequences, including over twelve hundred human V H sequences and over one thousand V L sequences.
  • the database of antibody sequences used for comparison is downloaded from Andrew C. R. Martin's KabatMan web page (http://www.rubic.rdg.ac.uk/abs/).
  • the Kabat method for identifying CDR provides a means for delineating the approximate CDR and framework regions from any human antibody and comparing the sequence of a murine antibody for similarity to determine the CDRs and FRs.
  • Best matched human V H and V L sequences are chosen on the basis of high overall framework matching, similar CDR length, and minimal mismatching of canonical and V H / V L contact residues.
  • Human framework regions most similar to the murine sequence are inserted between the murine CDR.
  • the murine framework region may be modified by making amino acid substitutions of all or part of the native framework region that more closely resemble a framework region of a human antibody.
  • Another useful technique for generating antibodies for use in the present invention may be one which uses a rational design type approach.
  • the goal of rational design is to produce structural analogs of biologically active polypeptides or compounds with which they interact (agonists, antagonists, inhibitors, peptidomimetics, binding partners, etc.).
  • An alternative approach, "alanine scan” involves the random replacement of residues throughout molecule with alanine, and the resulting affect on function determined.
  • Chemically constructed bispecific antibodies may be prepared by chemically cross- linking heterologous Fab or F(ab') 2 fragments by means of chemicals such as heterobifunctional reagent succinimidyl-3-(2-pyridyldithiol)-propionate (SPDP, Pierce Chemicals, Rockford, ⁇ 1.).
  • the Fab and F(ab') 2 fragments can be obtained from intact antibody by digesting it with papain or pepsin, respectively (Karpovsky et al., J. Exp. Med. 160: 1686-701, 1984; Titus et al., J.
  • Methods of testing antibodies for the ability to bind to the epitope of the ⁇ integrin regardless of how the antibodies are produced are known in the art and include any antibody- antigen binding assay, such as, for example, radioimmunoassay (RIA), ELISA, Western blot, immunoprecipitation, and competitive inhibition assays (see, e.g., Janeway et al., infra, and U.S. Patent Application Publication No. 2002/0197266 Al).
  • RIA radioimmunoassay
  • ELISA ELISA
  • Western blot Western blot
  • immunoprecipitation immunoprecipitation
  • competitive inhibition assays see, e.g., Janeway et al., infra, and U.S. Patent Application Publication No. 2002/0197266 Al.
  • the compound that inhibits a binding interaction between ⁇ integrin and G protein a subunit is an analog of an antibody.
  • the compound is an aptamer.
  • SELEX technology has been used to identify DNA and RNA aptamers with binding properties that rival mammalian antibodies, the field of immunology has generated and isolated antibodies or antibody fragments which bind to a myriad of compounds and phage display has been utilized to discover new peptide sequences with very favorable binding properties.
  • a loop structure is often involved with providing the desired binding attributes as in the case of: aptamers which often utilize hairpin loops created from short regions without complimentary base pairing, naturally derived antibodies that utilize combinatorial arrangement of looped hyper- variable regions and new phage display libraries utilizing cyclic peptides that have shown improved results when compared to linear peptide phage display results.
  • aptamers which often utilize hairpin loops created from short regions without complimentary base pairing
  • naturally derived antibodies that utilize combinatorial arrangement of looped hyper- variable regions
  • new phage display libraries utilizing cyclic peptides that have shown improved results when compared to linear peptide phage display results.
  • molecular evolution techniques can be used to isolate compounds specific for the ⁇ integrins or G protein a subunits described herein that inhibit the binding interaction between ⁇ integrin and G protein a subunit.
  • aptamers see, generally, Gold, L., Singer, B., He, Y. Y., Brody. E., "Aptamers As Therapeutic And Diagnostic Agents," J. Biotechnol. 74:5-13 (2000).
  • Relevant techniques for generating aptamers may be found in U.S. Pat. No. 6,699,843, which is incorporated by reference in its entirety.
  • epitope as used herein is meant the region of or within the ⁇ integrin or G protein a subunit which is bound by the compound, e.g., the antibody, the antigen binding fragment, the aptamer.
  • the epitope is a linear epitope.
  • linear epitope refers to the region of or within the ⁇ integrin or G protein a subunit which is bound by the compound, which region is composed of contiguous amino acids of the amino acid sequence of the ⁇ integrin or G protein a subunit.
  • the amino acids of a linear epitope are located in close promity to each other in the primary structure of the antigen and the secondary and/or tertiary structure(s) of the antigen.
  • the antigen e.g., ⁇ integrin or G protein a subunit
  • the properly folded state e.g., its native conformation
  • the contiguous amino acids of the linear epitope are located in close proximity to one another.
  • the epitope of the binding construct is a conformational epitope.
  • conformational epitope is meant an epitope which is composed of amino acids which are located in close proximity to one another only when the ⁇ integrin or G protein a subunit is in its properly folded state, but are not contiguous amino acids of the amino acid sequence of the ⁇ integrin or G protein a subunit.
  • the compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit binds to an epitope of a ⁇ integrin.
  • the epitope to which the compound binds is within the cytoplasmic domain of a ⁇ integrin.
  • the epitope to which the compound binds is within the cytoplasmic domain of a ⁇ 1 ⁇ integrin, ⁇ integrin, ⁇ 2 integrin, ⁇ 3 integrin, ⁇ 5 integrin, ⁇ 6 integrin, or ⁇ 7 integrin.
  • the epitope to which the compound binds is within amino acids 738-777 of a ⁇ 1 ⁇ integrin (SEQ ID NO: 12) or amino acids 732-778 of a ⁇ 1 ⁇ integrin (SEQ ID NO: 12). In exemplary aspects, the epitope to which the compound binds is within amino acids 738-776 of a ⁇ 10 integrin (SEQ ID NO: 13) or amino acids 732-781 of a ⁇ 10 integrin (SEQ ID NO: 13).
  • the epitope to which the compound binds is within amino acids 702-746 of a ⁇ 2 integrin (SEQ ID NO: 14) or amino acids 702-747 of a ⁇ 2 integrin (SEQ ID NO: 14). In exemplary aspects, the epitope to which the compound binds is within amino acids 722-761 of a ⁇ 3 integrin (SEQ ID NO: 15) or amino acids 716-762 of a ⁇ 3 integrin (SEQ ID NO: 15).
  • the epitope to which the compound binds is within amino acids 720- 765 of a ⁇ 5 integrin (SEQ ID NO: 16) or amino acids 720-776 of a ⁇ 5 integrin (SEQ ID NO: 16). In exemplary aspects, the epitope to which the compound binds is within amino acids 710-755 of a ⁇ 6 integrin (SEQ ID NO: 17) or amino acids 710-767 of a ⁇ 6 integrin (SEQ ID NO: 17).
  • the epitope to which the compound binds is within amino acids 728-773 of a ⁇ 7 integrin (SEQ ID NO: 18) or amino acids 728-779 of a ⁇ 7 integrin (SEQ ID NO: 18).
  • the compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit binds to an epitope of the G protein a subunit.
  • the epitope to which the compound binds is within the Switch Region I of a G protein a subunit.
  • the epitope to which the compound binds is within the Switch Region I of G protein a subunit Ga 12 or Ga 13 .
  • the compound binds to an epitope within amino acids 201-216 of Ga 12 (SEQ ID NO: 11) or amino acids 197-212 of Ga 13 (SEQ ID NO: 10).
  • the compound binds to an epitope within amino acids 197-209 of Ga 13 (SEQ ID NO: 10) or amino acids 198-206 of Ga 13 (SEQ ID NO: 10). In exemplary aspects, the compound binds to an epitope within amino acids 203-211 of Ga i2 (SEQ ID NO: 11).
  • the compound that inhibits the binding interaction between a ⁇ integrin and a G protein a subunit binds to an epitope comprising the amino acid sequence of any of the peptides or peptide analogs described herein. See, e.g., the section entitled
  • the compound that inhibits the binding interaction between a ⁇ integrin and a G protein a subunit binds to an epitope comprising an amino acid sequence of any of SEQ ID NOs: 19-40.
  • the compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit is a peptide comprising at least four amino acids connected via peptide bonds. Accordingly, the invention provides a peptide. In some aspects, the peptide is about 4 to about 50 amino acids in length. In some aspects, the compound is about 5 to about 25 amino acids in length. In some aspects, the compound is about 5 to 20 amino acids in length. In some aspects, the peptide is 5-15 amino acids in length. In some aspects, the peptide is 5-9 or 5-8 or 5-7 amino acids in length.
  • the peptide is a 5-mer, 6-mer, 7-mer, 8-mer, 9-mer-10-mer, 11-mer, 12-mer, 13-mer, 14-mer, 15- mer, 16-mer, 17-mer, 18-mer, 19-mer, or 20-mer.
  • the peptide that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit comprises a fragment or is a fragment of a human wild-type ⁇ integrin, e.g., any of those disclosed herein.
  • the term "fragment" does not encompass a full length ⁇ integrin or a full length G protein a subunit.
  • the compound comprises or is a fragment of a ⁇ 1 ⁇ integrin, ⁇ 10 integrin, ⁇ 2 integrin, ⁇ 3 integrin, ⁇ 5 integrin, ⁇ 6 integrin, or ⁇ 7 integrin.
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of a cytoplasmic domain of a ⁇ integrin.
  • the compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of the cytoplasmic domain of a ⁇ 1 ⁇ integrin, ⁇ integrin ⁇ 2 integrin, ⁇ 3 integrin, ⁇ 5 integrin, ⁇ 6 integrin, or ⁇ 7 integrin.
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 738-777 of a ⁇ 1 ⁇ integrin (SEQ ID NO: 12) or amino acids 732-778 of a ⁇ 1 ⁇ integrin (SEQ ID NO: 12).
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 738-776 of a ⁇ integrin (SEQ ID NO: 13) or amino acids 732-781 of a ⁇ 1 ⁇ integrin (SEQ ID NO: 13).
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 702-746 of a ⁇ 2 integrin (SEQ ID NO: 14) or amino acids 702-747 of a ⁇ 2 integrin (SEQ ID NO: 14).
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 722-761 of a ⁇ 3 integrin (SEQ ID NO: 15) or amino acids 716-762 of a ⁇ integrin (SEQ ID NO: 15).
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 720-765 of a ⁇ 5 integrin (SEQ ID NO: 16) or amino acids 720-776 of a ⁇ 5 integrin (SEQ ID NO: 16).
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 710-755 of a ⁇ 6 integrin (SEQ ID NO: 17) or amino acids 710-767 of a ⁇ 6 integrin (SEQ ID NO: 17).
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 728-773 of a ⁇ 7 integrin (SEQ ID NO: 18) or amino acids 728-779 of a ⁇ 7 integrin (SEQ ID NO: 18).
  • the compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of the G protein a subunit. In some aspects, the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of the Switch Region I of a G protein a subunit. In exemplary aspects, the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of the Switch Region I of G protein a subunit Ga 12 or Ga 13 .
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 201-216 of Ga 12 (SEQ ID NO: 11) or amino acids 197-212 of Ga 13 (SEQ ID NO: 10). In exemplary aspects, the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 197-209 of Ga 13 (SEQ ID NO: 10) or amino acids 198-206 of Ga 13 (SEQ ID NO: 10). In exemplary aspects, the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 203-211 of Ga 12 (SEQ ID NO: 11).
  • the compound comprises a core sequence of three amino acids which is a portion or fragment of a cytoplasmic domain of the ⁇ integrin.
  • the compound comprises a core sequence identical to amino acids 731 to 733 of the amino acid sequence of ⁇ integrin (SEQ ID NO: 15), which is EEE.
  • the compound comprises a core sequence identical to amino acids 747-749 of the amino acid sequence of ⁇ integrin (SEQ ID NO: 12) or ⁇ 1 ⁇ integrin (SEQ ID NO: 13), amino acids 717-719 of the amino acid sequence of ⁇ 2 integrin (SEQ ID NO: 14), or amino acids 743-745 of the amino acid sequence of ⁇ 7 integrin (SEQ ID NO: 18), each of which is EKE.
  • the compound comprises a core sequence identical amino acids 725-727 of the amino acid sequence of ⁇ 6 integrin (SEQ ID NO: 17), which is EAE.
  • the compound comprises a core sequence identical amino acids 735-737 of the amino acid sequence of ⁇ 5 integrin (SEQ ID NO: 16), which is QSE.
  • the compound comprises additional amino acids N- terminal and/or C-terminal to the core sequence.
  • the additional amino acids e.g., the non-core sequence(s) may represent amino acids which are N-terminal and/or C-terminal to the core sequence of the amino acid sequence of the ⁇ integrin.
  • the compound may comprise a core sequence of EEE (amino acids 731 to 733 of the amino acid sequence of ⁇ 3 integrin (SEQ ID NO: 15) and may additionally comprise an N-terminal non-core sequence comprising KF (Lys-Phe) and/or a C-terminal non-core sequence comprising RA (Arg-Ala).
  • the compound in exemplary aspects comprises the amino acid sequence of KFEEE (SEQ ID NO: 19), KFEEERA (SEQ ID NO: 20), EEERA (SEQ ID NO: 21).
  • the compound comprises an amino acid sequence of KFEEERARAKWDT (SEQ ID NO: 22).
  • the compound comprises a core sequence of EKE and comprises a N-terminal non-core sequence comprising KF (Lys-Phe) or RF (Arg-Phe) and/or a C-terminal non-core sequence comprising KM (Lys-Met), KL (Lys-Leu), or QQ (Gln-Gln).
  • the compound in exemplary aspects comprises the amino acid sequence of KFEKE (SEQ ID NO: 23), RFEKE (SEQ ID NO: 24), KFEKEKM (SEQ ID NO: 25), KFEKEKL (SEQ ID NO: 26), KFEKEQQ (SEQ ID NO: 27), RFEKEKM (SEQ ID NO: 28), RKFEKEKL (SEQ ID NO: 29), RFEKEQQ (SEQ ID NO: 30), EKEKM (SEQ ID NO: 31), EKEKL (SEQ ID NO: 32), or EKEQQ (SEQ ID NO: 33).
  • the compound comprises a core sequence of EAE and comprises a N-terminal non-core sequence comprising KF (Lys-Phe) and/or a C-terminal non- core sequence comprising RS (Arg-Ser). Accordingly, the compound in exemplary aspects comprises the amino acid sequence of KFEAE (SEQ ID NO: 34), KFEAERS (SEQ ID NO: 35), or EAERS (SEQ ID NO: 36).
  • the compound comprises a core sequence of QSE and comprises a N-terminal non-core sequence comprising KF (Lys-Phe) and/or a C-terminal non- core sequence comprising RS (Arg-Ser). Accordingly, the compound in exemplary aspects comprises the amino acid sequence of KFQSE (SEQ ID NO: 37), KFQSERS (SEQ ID NO: 38), or QSERS (SEQ ID NO: 39).
  • the compound comprises a non-core sequence which is not based on the wild-type sequence of the ⁇ integrin.
  • the the compound may comprise a core sequence of EEE (amino acids 731 to 733 of the amino acid sequence of ⁇ 3 integrin (SEQ ID NO: 15) and may additionally comprise a N-terminal non- core sequence other than KF (Lys-Phe) and/or a C-terminal non-core sequence other than RA (Arg-Ala).
  • the peptide that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit comprises a fragment or is a fragment of a human wild-type G protein a subunit, e.g., Ga 12 , Ga 13 .
  • the compound comprises or is a fragment of Ga 12 or Ga 13 .
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of the Switch Region I of the G protein a subunit.
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of the Switch Region I of G protein a subunit Ga 12 or Ga 13 .
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 201-216 of Ga 12 (SEQ ID NO: 11) or amino acids 197-212 of Ga 13 (SEQ ID NO: 10).
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 197-209 of Ga 13 (SEQ ID NO: 10) or amino acids 198-206 of Ga 13 (SEQ ID NO: 10).
  • the compound comprises 4 to 50 (e.g., 5 to 25) consecutive amino acids of amino acids 203-211 of Ga 12 (SEQ ID NO: 11). In some aspects, the compound comprises an amino acid sequence of LLARRPTKGIHEY (SEQ ID NO: 40).
  • the peptide that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit comprises an amino acid sequence which is based on the amino acid sequence of a human wild-type ⁇ integrin, or a fragment thereof, but differs at one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, or more) amino acid positions, when aligned with the human wild-type ⁇ integrin sequence, or fragment thereof.
  • the peptide that inhibits a binding interaction between an a ⁇ integrin and a G protein a subunit comprises an amino acid sequence which has at least 25% sequence identity to the amino acid sequence of a human wild-type ⁇ integrin, e.g., SEQ ID NO: 15, or a fragment thereof (e.g., a fragment of about 4 to about 50 contiguous amino acids of SEQ ID NO: 15).
  • the compound comprises an amino acid sequence which is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or has greater than 95% sequence identity to SEQ ID NO: 15, or a fragment thereof (e.g., a fragment of about 4 to about 25 contiguous amino acids of SEQ ID NO: 15).
  • the compound comprises an amino acid sequence which is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or has greater than 95% sequence identity to any of SEQ ID NOs: 12-18, or a fragment thereof (e.g., a fragment of about 4 to about 25 contiguous amino acids of any one of SEQ ID NOs: 12- 18).
  • the compound comprises an amino acid sequence which is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or has greater than 95% sequence identity to any one of SEQ ID NOs: 19-39.
  • the peptide that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit comprises an amino acid sequence which is based on the amino acid sequence of a human wild-type G protein a subunit, or a fragment thereof, but differs at one or more (e.g., two, three, four, five, six, seven, eight, nine, ten, or more) amino acid positions, when aligned with the human wild-type G protein a subunit sequence, or fragment thereof.
  • the peptide that inhibits a binding interaction between an a ⁇ integrin and a G protein a subunit comprises an amino acid sequence which has at least 25% sequence identity to the amino acid sequence of a human wild-type G protein a subunit, e.g., SEQ ID NO: 10 or 11 or a fragment thereof (e.g., a fragment of about 4 to about 15 contiguous amino acids of SEQ ID NO: 10 or 11).
  • the compound comprises an amino acid sequence which is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or has greater than 95% sequence identity to SEQ ID NO: 10 or 11 or a fragment thereof (e.g., a fragment of about 4 to about 10 contiguous amino acids of SEQ ID NO: 10 or 11).
  • the compound comprises an amino acid sequence which is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or has greater than 95% sequence identity to amino acids 201-216 of Ga 12 (SEQ ID NO: 11) or amino acids 197-212 of Ga 13 (SEQ ID NO: 10), amino acids 197-209 of Ga 13 (SEQ ID NO: 10) or amino acids 198-206 of Ga 13 (SEQ ID NO: 10), amino acids 203-211 of Ga 12 (SEQ ID NO: 11).
  • the compound comprises an amino acid sequence which is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or has greater than 95% sequence identity to SEQ ID NOs: 40.
  • the compound comprises an amino acid sequence:
  • Xaai is Glu or Gin and Xaa 2 is Glu, Lys, Ser, or Ala.
  • (i) Xaai is Glu and Xaa 2 is Glu, Lys, or Ala or
  • Xaai is Gin and Xaa 2 is Ser.
  • each of Xaai and Xaa 2 is Glu.
  • the peptide comprises Phe N-terminal to Xaai Xaa 2 Glu.
  • the peptide comprises Arg C-terminal to Xaai Xaa 2 Glu.
  • the peptide comprises Phe N-terminal to Xaai Xaa 2 Glu and Arg C-terminal to Xaai Xaa 2 Glu.
  • the compound comprises an extension of two amino acids, Xaa_i Xaa 0 N-terminal to Xaai Xaa 2 Glu.
  • Xaa ⁇ of the N-terminal extension is Lys or Arg.
  • Xaao of the N-terminal extension is Phe.
  • the compound comprises an extension of two amino acids Xaa 3 Xaa 4 C terminal to Xaai Xaa 2 Glu.
  • Xaa of the C-terminal extension is Lys, Arg, or Gin.
  • Xaa 4 of the C-terminal extension is Met, Leu, Ala, Ser, or Gin.
  • Xaa 3 is Arg and Xaa 4 is Ala. Accordingly, in exemplary aspects of the invention the compound comprises a sequence selected from the group consisting of:
  • the peptide comprises, consists essentially of, or consists of FEEERA (SEQ ID NO: 87).
  • the peptide comprises a sequence of KFEEE (SEQ ID NO: 19), FEEER (SEQ ID NO: 84), AKFEEE (SEQ ID NO: 85), KFEEER (SEQ ID NO: 86), FEEERA (SEQ ID NO: 87), EEERAR (SEQ ID NO: 88), EEERARA (SEQ ID NO: 89), or EEERARAK (SEQ ID NO: 90).
  • the peptide comprises a core of EXE (SEQ ID NO: 93), wherein X is any amino acid.
  • X is Glu, Lys, Ser, or Ala.
  • X is Ala, Gly, Val, Leu, or He.
  • X is Glu or Asp.
  • X is Ser or Thr.
  • X is Lys or ornithine or Arg.
  • X is Pro, Phe, Tyr, Trp, Asn, Gin, His, Cys, or Met.
  • the peptide comprises FEXE (SEQ ID NO: 94, wherein X is any amino acid.
  • X is Glu or Asp.
  • X is Ala, Gly, Val, Leu, or He.
  • X is Glu or Asp.
  • X is Ser or Thr.
  • X is Lys or ornithine or Arg.
  • X is Pro, Phe, Tyr, Trp, Asn, Gin, His, Cys, or Met.
  • the peptide comprises FEXiEX 2 wherein each of XI and X2 is independently any amino acid.
  • the peptide comprises FEXiEX 2 (SEQ ID NO: 95), wherein XI is any amino acid and X2 is Lys, Arg, or Gin.
  • XI is Glu or Asp.
  • XI is Ala, Gly, Val, Leu, or He.
  • XI is Glu or Asp.
  • XI is Ser or Thr.
  • XI is Lys or ornithine or Arg.
  • XI is Pro, Phe, Tyr, Trp, Asn, Gin, His, Cys, or Met.
  • the peptide comprises EX1EX2 wherein each of XI and X2 is independently any amino acid.
  • the peptide comprises EX1EX2 (SEQ ID NO: 96), wherein XI is any amino acid and X2 is Lys, Arg, or Gin.
  • XI is Glu or Asp.
  • XI is Ala, Gly, Val, Leu, or He.
  • XI is Glu or Asp.
  • XI is Ser or Thr.
  • XI is Lys or ornithine or Arg.
  • XI is Pro, Phe, Tyr, Trp, Asn, Gin, His, Cys, or Met.
  • the peptide comprises X1FEX2E wherein each of XI and X2 is independently any amino acid.
  • the peptide comprises X1FEX2E (SEQ ID NO: 97), wherein XI is Lys or Arg and X2 is any amino acid.
  • X2 is Glu or Asp.
  • X2 is Ala, Gly, Val, Leu, or He.
  • X2 is Glu or Asp.
  • X2 is Ser or Thr.
  • X2 is Lys or ornithine or Arg.
  • X2 is Pro, Phe, Tyr, Trp, Asn, Gin, His, Cys, or Met.
  • the peptide comprises X1FEX2EX3 wherein each of XI, X2, and X3 is independently any amino acid.
  • the peptide comprises
  • X1FEX2EX3 (SEQ ID NO: 98), wherein XI is Lys or Arg, wherein X2 is any amino acid, and X3 is Lys, Arg, or Gin.
  • X2 is Glu or Asp.
  • X2 is Ala, Gly, Val, Leu, or He.
  • X2 is Glu or Asp.
  • X2 is Ser or Thr.
  • X2 is Lys or ornithine or Arg.
  • X2 is Pro, Phe, Tyr, Trp, Asn, Gin, His, Cys, or Met.
  • the peptide comprises X1FEX2EX3X4 wherein each of XI, X2, X3, and X4 is independently any amino acid.
  • the peptide comprises X1FEX2EX3X4 (SEQ ID NO: 99), wherein XI is Lys or Arg, X2 is any amino acid, X3 is Lys, Arg, or Gin, and X4 is any amino acid, optionally, Met, Ala, Leu, Ser, or Gin.
  • X2 is Glu or Asp.
  • X2 is Ala, Gly, Val, Leu, or He.
  • X2 is Glu or Asp.
  • X2 is Ser or Thr. In exemplary aspects, X2 is Lys or ornithine or Arg. In exemplary aspects, X2 is Pro, Phe, Tyr, Trp, Asn, Gin, His, Cys, or Met.
  • the peptide comprises any of SEQ ID NOs: 100-122.
  • the compound comprises the amino acid sequence X 1 X 2 X3X4 5 6 7 X 8 9 io n i2 (SEQ ID NO: 44).
  • each of X 1; X 2 , X 3 , Xg, and X 9 is independently an aliphatic amino acid.
  • each of X 4 , X 5 , and X 10 is independently a basic amino acid.
  • X 6 is Pro.
  • X 7 and X 12 is independently a hydroxyl-containing amino acid.
  • Xn is an acidic amino acid.
  • each of X 1; X 2 , X 3 , Xg, and Xg is independently selected from the group consisting of L, A, G, and I.
  • each of X 4 , X5, and X 10 is independently selected from the group consisting of R and H.
  • X 7 and X 12 is independently a T or Y.
  • Xn is an E or D.
  • the compound comprises the amino acid sequence of LLARRPTKGIHEY (SEQ ID NO: 45).
  • the peptide is lipidated (e.g., myritoylated, palmitoylated), glycosylated, amidated, carboxylated, phosphorylated, esterified, acylated, acetylated, cyclized, or converted into an acid addition salt and/or optionally dimerized or polymerized, or conjugated, as further described herein.
  • lipidated e.g., myritoylated, palmitoylated
  • glycosylated e.g., amidated, carboxylated, phosphorylated, esterified, acylated, acetylated, cyclized, or converted into an acid addition salt and/or optionally dimerized or polymerized, or conjugated, as further described herein.
  • the peptide is lipidated, or otherwise, attached to a lipid.
  • the lipid in some embodiments, is a fatty acid, eicosanoid, prostaglandin, leukotriene, thromboxane, N-acyl ethanolamine), glycerolipid (e.g., mono-, di-, tri- substituted glycerols),
  • glycerophospholipid e.g., phosphatidylcholine, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine
  • sphingolipid e.g., sphingosine, ceramide
  • sterol lipid e.g., steroid, cholesterol
  • prenol lipid saccharolipid, or a polyketide, oil, wax, cholesterol, sterol, fat-soluble vitamin, monoglyceride, diglyceride, triglyceride, a phospholipid.
  • the peptide is covalently attached to a fatty acid.
  • the fatty acid is a C4 to C30 fatty acid.
  • the fatty acid in exemplary aspects is any of a C4 fatty acid, C6 fatty acid, C8 fatty acid, CIO fatty acid, C12 fatty acid, C14 fatty acid, C16 fatty acid, C18 fatty acid, C20 fatty acid, C22 fatty acid, C24 fatty acid, C26 fatty acid, C28 fatty acid, or a C30 fatty acid.
  • the fatty acid is a C8 to C20 fatty acid, a C12 to C29 fatty acid, or a C14 to C18 fatty acid, e.g., a C14 fatty acid or a C16 fatty acid.
  • the peptide is covalently attached to a fatty acid and the fatty acid is attached to the N-terminal amino acid or the C-terminal amino acid.
  • the peptide is covalently attached to a fatty acid and the fatty acid is attached to an internal amino acid of the peptide, e.g., via a functional group off of a side chain of the internal amino acid.
  • the fatty acid may be attached to an amine, hydroxyl, or thiol of a side chain of an internal amino acid.
  • the peptide is covalently attached to a fatty acid and the fatty acid is attached to the second third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, eleventh, or twelvth amino acid.
  • the peptide comprises the amino acid sequence of any one of SEQ ID NOs: 19-39, 84-90, and 93-122 and the peptide is covalently attached to a C4-C30 fatty acid.
  • the peptide is any one of SEQ ID NOs: 355-412.
  • the first amino acid of the peptide is myristoylated at the N-terminus in which the N-terminal alpha -NH 2 group of an unmodified peptide is attached to a fatty acid.
  • the peptide is any one of SEQ ID NOs: 123-180.
  • the first amino acid of the peptide is myristoylated at the N-terminus in which the N-terminal alpha -NH 2 group of an unmodified peptide is attached to a C4-C30 fatty acid.
  • the peptide is any one of SEQ ID NOs: 181-238.
  • the first amino acid of the peptide is myristoylated at the N-terminus in which the N-terminal alpha -NH 2 group of an unmodified peptide is attached to a C12-C18 fatty acid.
  • the peptide is any one of SEQ ID NOs: 239-296.
  • the first amino acid of the peptide is myristoylated at the N-terminus in which the N-terminal alpha -NH 2 group of an unmodified peptide is attached to a myristate.
  • the peptide is any one of SEQ ID NOs: 297-354.
  • the peptide comprises a my
  • the lipid attached to the peptide facilitates micelle formation of the peptide.
  • micellar forms of peptides see the descriptions below under “Micelles. "
  • the peptide is cyclized.
  • the peptide may comprise two Cys residues, the sulfur atoms of which participate in the formation of a disulfide bridge.
  • the peptide comprises a Cys residue as the terminal residues. Suitable methods of modifying peptides with disulfide bridges or sulfur-based cyclization are described in, for example, Jackson et al., /. Am. Chem. Soc. 113: 9391-9392 (1991) and Rudinger and Jost, Experientia l ⁇ : 570-571 (1964).
  • the alpha helix of the analog can alternatively be stabilized through other means of peptide cyclizing, which means are reviewed in Davies, J. Peptide. Sci. 9: 471-501 (2003).
  • the alpha helix can be stabilized via the formation of an amide bridge, thioether bridge, thioester bridge, urea bridge, carbamate bridge, sulfonamide bridge, and the like.
  • a thioester bridge can be formed between the C-terminus and the side chain of a Cys residue.
  • a thioester can be formed via side chains of amino acids having a thiol (Cys) and a carboxylic acid (e.g., Asp, Glu).
  • a cross-linking agent such as a dicarboxylic acid, e.g., suberic acid (octanedioic acid), etc. can introduce a link between two functional groups of an amino acid side chain, such as a free amino, hydroxyl, thiol group, and combinations thereof.
  • the compound is a peptide analog having a structure based on one of the peptides disclosed herein (the "parent peptide") but differs from the parent peptide in one or more respects. Accordingly, as appreciated by one of ordinary skill in the art the teachings of the parent peptides provided herein may also be applicable the peptide analogs.
  • the peptide analog comprises the structure of a parent peptide, except that the peptide analog comprises one or more non-peptide bonds in place of peptide bond(s).
  • the peptide analog comprises in place of a peptide bond, an ester bond, an ether bond, a thioether bond, an amide bond, and the like.
  • the peptide analog is a depsipeptide comprising an ester linkage in place of a peptide bond.
  • the peptide analog comprises the structure of a parent peptide described herein, except that the peptide analog comprises one or more amino acid substitutions, e.g., one or more conservative amino acid substitutions.
  • Conservative amino acid substitutions are known in the art, and include amino acid substitutions in which one amino acid having certain physical and/or chemical properties is exchanged for another amino acid that has the same chemical or physical properties.
  • the conservative ammo acid substitution may be an acidic amino acid substituted for another acidic amino acid (e.g., Asp or Glu), an amino acid with a nonpolar side chain substituted for another amino acid with a nonpolar side chain (e.g., Ala, Gly, Val, He, Leu, Met, Phe, Pro, Trp, Val, etc.), a basic amino acid substituted for another basic amino acid (Lys, Arg, etc.), an amino acid with a polar side chain substituted for another amino acid with a polar side chain (Asn, Cys, Gin, Ser, Thr, Tyr, etc.), etc.
  • an amino acid with a nonpolar side chain substituted for another amino acid with a nonpolar side chain e.g., Ala, Gly, Val, He, Leu, Met, Phe, Pro, Trp, Val, etc.
  • a basic amino acid substituted for another basic amino acid Lys, Arg, etc.
  • the peptide analog comprises one or more synthetic amino acids, e.g., an amino acid non-native to a mammal.
  • Synthetic amino acids include ⁇ -alanine ( ⁇ -Ala), N-a-methyl-alanine (Me-Ala), aminobutyric acid (Abu), ⁇ -aminobutyric acid ( ⁇ -Abu), aminohexanoic acid ( ⁇ -Ahx), aminoisobutyric acid (Aib), aminomethylpyrrole carboxylic acid, aminopiperidinecarboxylic acid, aminoserine (Ams), aminotetrahydropyran-4-carboxylic acid, arginine N-methoxy-N-methyl amide, ⁇ -aspartic acid ( ⁇ -Asp), azetidine carboxylic acid, 3- (2- benzothiazolyl)alanine, cc-iert-butylglycine, 2-amino-5-ureido-n-valeric acid ( ⁇ -Ala),
  • pyrrolidinylalanine sarcosine (Sar), selenocysteine (Sec), O-Benzyl-phosphoserine, 4-amino-3- hydroxy-6-methylheptanoic acid (Sta), 4-amino-5-cyclohexyl-3-hydroxypentanoic acid (ACHPA), 4-amino-3-hydroxy-5-phenylpentanoic acid (AHPPA), 1,2,3,4,-tetrahydro- isoquinoline-3-carboxylic acid (Tic), tetrahydropyranglycine, thienylalanine (Thi) , O-benzyl- phosphotyrosine, O-Phosphotyrosine, methoxytyrosine, ethoxytyrosine, 0-(bis-dimethylamino- phosphono)-tyrosine, tyrosine sulfate tetrabutylamine, methyl-valine (MeVal),
  • the peptide analog comprises one or more non-conservative amino acid substitutions and the peptide analog still functions to a similar extent, the same extent, or an improved extent as the parent peptide.
  • the peptide analog comprising one or more non-conservative amino acid substitutions inhibits the binding interaction between ⁇ integrin and G protein a subunit to an extent better than the parent peptide.
  • the peptide analog comprises an insertion of one or more amino acids at the N- or C-terminus in reference to the parent peptide. In some embodiments, the peptide analog comprises a deletion of one or more amino acids at the N- or C-terminus in reference to the parent peptide. In these aspects, the peptide analog still functions to a similar extent, the same extent, or an improved extent as the parent peptide to inhibit the binding interaction between ⁇ integrin and G protein a subunit.
  • the peptide analog is a peptidomimetic.
  • Peptidomimetics as well as methods of making the same are known in the art. See, for example, Advances in Amino Acid Mimetics and Peptidomimetics, Volumes 1 and 2, ed., Abell, A., JAI Press Inc., Greenwich, CT, 2006.
  • the peptidomimetic is a D-peptide peptidomimetic comprising D-isomer amino acids.
  • the peptidomimetic is a peptoid in which the side chain of an amino acid is connected to the alpha nitrogen atom of the peptide backbone. Methods of making peptoids are known in the art. See, e.g., Zuckermann et al., J ACS 114(26): 10646-10647 (1992) and Design, Synthesis, and Evaluation of Novel Peptoids, Fowler, Sarah, University of
  • the peptidomimetic is a ⁇ -peptide comprising ⁇ amino acids which have their amino group bonded to the ⁇ -cargon rather than the alpha carbon.
  • Methods of making ⁇ -peptides are known in the art. See, for example, Seebach et al., Helvetica Chimica Acta 79(4): 913-941 (1996).
  • the compounds that inhibit a binding interaction between a ⁇ integrin and a G protein a subunit (which compounds are collectively referred to hereinafter as "active agents") in some aspects is in the form of a salt, e.g., a pharmaceutically acceptable salt.
  • a salt e.g., a pharmaceutically acceptable salt.
  • Such salts can be prepared in situ during the final isolation and purification of the active agent or separately prepared by reacting a free base function with a suitable acid.
  • acids which can be employed to form pharmaceutically acceptable acid addition salts include, for example, an inorganic acid, e.g., hydrochloric acid, hydrobromic acid, sulphuric acid, and phosphoric acid, and an organic acid, e.g., oxalic acid, maleic acid, succinic acid, and citric acid.
  • an inorganic acid e.g., hydrochloric acid, hydrobromic acid, sulphuric acid, and phosphoric acid
  • organic acid e.g., oxalic acid, maleic acid, succinic acid, and citric acid.
  • Representative acid addition salts include, but are not limited to acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphor sulfonate, digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate (isothionate), lactate, maleate, methane sulfonate, nicotinate, 2-naphthalene sulfonate, oxalate, palmitoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, phosphate, glutamate, bicarbonate, p-toluenesulfonate
  • Basic addition salts also can be prepared in situ during the final isolation and purification of the active agent, or by reacting a carboxylic acid-containing moiety with a suitable base such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • a suitable base such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • Pharmaceutically acceptable salts include, but are not limited to, cations based on alkali metals or alkaline earth metals such as lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like, and nontoxic quaternary ammonia and amine cations including ammonium, tetramethylammonium, tetraethylammonium, methylammonium, dimethylammonium, trimethylammonium, triethylammonium, diethylammonium, and ethylammonium, amongst others.
  • Other representative organic amines useful for the formation of base addition salts include, for example, ethylenediamine, ethanolamine, diethanolamine, piperidine, piperazine, and the like.
  • basic nitrogen-containing groups can be quaternized with such active agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; long chain halides such as decyl, lauryl, myristyl, and stearyl chlorides, bromides, and iodides; arylalkyl halides like benzyl and phenethyl bromides and others. Water or oil- soluble or dispersible products are thereby obtained.
  • the compounds of the invention that inhibit a binding interaction between a ⁇ integrin and a G protein a subunit can be isolated and/or purified.
  • isolated as used herein means having been removed from its natural environment.
  • purified as used herein means having been increased in purity, wherein “purity” is a relative term, and not to be necessarily construed as absolute purity.
  • the purity of the compound is at least or about 50%, at least or about 60%, at least or about 70%, at least or about 80%, at least or about 90%, at least or about 95%, or at least or about 98% or is about 100%.
  • the peptides of the present disclosure may be obtained by methods known in the art. Suitable methods of de novo synthesizing peptides are described in, for example, Chan et al., Fmoc Solid Phase Peptide Synthesis, Oxford University Press, Oxford, United Kingdom, 2005; Peptide and Protein Drug Analysis, ed. Reid, R., Marcel Dekker, Inc., 2000; Epitope Mapping, ed. Westwood et al., Oxford University Press, Oxford, United Kingdom, 2000; and U.S. Patent No. 5,449,752. Additional exemplary methods of making the peptides of the invention are set forth herein.
  • the peptides described herein are commercially synthesized by companies, such as Synpep (Dublin, CA), Peptide Technologies Corp. (Gaithersburg, MD), Multiple Peptide Systems (San Diego, CA), Peptide 2.0 Inc. (Chantilly, VA), and American Peptide Co. (Sunnyvale, CA).
  • the peptides can be synthetic, recombinant, isolated, and/or purified.
  • the peptides are recombinantly produced using a nucleic acid encoding the amino acid sequence of the peptide using standard recombinant methods. See, for instance, Sambrook et al., Molecular Cloning: A Laboratory Manual. 3rd ed., Cold Spring Harbor Press, Cold Spring Harbor, NY 2001; and Ausubel et al., Current Protocols in Molecular Biology, Greene Publishing Associates and John Wiley & Sons, NY, 1994.
  • the compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit comprises or is a nucleic acid comprising a nucleotide sequence encoding any of the antibodies or peptides described herein (including analogs thereof).
  • the nucleic acid can comprise any nucleotide sequence which encodes any of the antibodies, peptides, or analogs thereof.
  • the compound is a nucleic acid which inhibits expression of the ⁇ integrin or the G protein a subunit.
  • the compound is an antisense molecule, a microRNA (miRNA), small hairpin (shRNA), and the like.
  • the compound is a nucleic acid which inhibits expression of any of the ⁇ integrins or G protein a subunits described herein.
  • the compound is a small interfering RNA molecule (siRNA).
  • the siRNA inhibits the expression Ga 13 .
  • the siRNA comprises the sequence of SEQ ID NO: 7 or 8.
  • nucleic acid includes “polynucleotide,” “oligonucleotide,” and “nucleic acid molecule,” and generally means a polymer of DNA or RNA, which can be single- stranded or double- stranded, synthesized or obtained (e.g., isolated and/or purified) from natural sources, which can contain natural, non-natural or altered nucleotides, and which can contain a natural, non-natural or altered inter-nucleotide linkage, such as a phosphoroamidate linkage or a phosphorothioate linkage, instead of the phosphodiester found between the nucleotides of an unmodified oligonucleotide.
  • the nucleic acid does not comprise any insertions, deletions, inversions, and/or substitutions. In other embodiments, the nucleic acid comprises one or more insertions, deletions, inversions, and/or substitutions.
  • the nucleic acids of the invention are recombinant.
  • the term “recombinant” refers to (i) molecules that are constructed outside living cells by joining natural or synthetic nucleic acid segments to nucleic acid molecules that can replicate in a living cell, or (ii) molecules that result from the replication of those described in (i) above.
  • the replication can be in vitro replication or in vivo replication.
  • nucleic acids in some aspects are constructed based on chemical synthesis and/or enzymatic ligation reactions using procedures known in the art. See, for example, Sambrook et al., supra; and Ausubel et al., supra.
  • a nucleic acid can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed upon hybridization (e.g., phosphorothioate derivatives and acridine substituted nucleotides).
  • modified nucleotides that can be used to generate the nucleic acids include, but are not limited to, 5-fluorouracil, 5-bromouracil, 5-chIorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxymethyl) uracil, 5- carboxymethylaminomethyl-2- thiouridme, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N 6 -isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, N -substituted adenine, 7- methylguanine, 5-methylammomethyluracil, 5- methoxyaminomethyl-2-thiouracil, beta-D- man
  • nucleic acids of the invention can be purchased from any source.
  • nucleic acids of the invention can be purchased from any source.
  • nucleic acids of the invention in some aspects are incorporated into a
  • the invention provides recombinant expression vectors comprising any of the presently disclosed nucleic acids.
  • the term "recombinant expression vector” means a genetically-modified oligonucleotide or polynucleotide construct that permits the expression of an mRNA, protein, polypeptide, or peptide by a host cell, when the construct comprises a nucleotide sequence encoding the mRNA, protein, polypeptide, or peptide, and the vector is contacted with the cell under conditions sufficient to have the mRNA, protein, polypeptide, or peptide expressed within the cell.
  • the vectors of the invention are not naturally- occurring as a whole.
  • the presently disclosed recombinant expression vectors may comprise any type of nucleotides, including, but not limited to DNA and RNA, which may be single- stranded or double- stranded, synthesized or obtained in part from natural sources, and which can contain natural, non-natural or altered nucleotides.
  • the recombinant expression vectors may comprise naturally- occurring or non-naturally-occuring internucleotide linkages, or both types of linkages.
  • the altered nucleotides or non-naturally occurring internucleotide linkages do not hinder the transcription or replication of the vector.
  • the recombinant expression vector of the invention can be any suitable recombinant expression vector, and can be used to transform or transfect any suitable host. Suitable vectors include those designed for propagation and expansion or for expression or both, such as plasmids and viruses.
  • the vector can be selected from the group consisting of the pUC series (Fermentas Life Sciences), the pBluescript series (Stratagene, LaJoIIa, CA), the pET series (Novagen, Madison, WI), the pGEX series (Pharmacia Biotech, Uppsala, Sweden), and the pEX series (Clontech, Palo Alto, CA).
  • Bacteriophage vectors such as ⁇ , ⁇ 1, ZapII (Stratagene), EMBL4, and ⁇ 149, also can be used.
  • plant expression vectors include pBIOl, pBI101.2, pBI101.3, pBI121 and pBIN19 (Clontech).
  • animal expression vectors include pEUK-Cl, pMAM and pMAMneo (Clontech).
  • the recombinant expression vector is a viral vector, e.g., a retroviral vector.
  • the recombinant expression vectors of the invention can be prepared using standard recombinant DNA techniques described in, for example, Sambrook et al., supra, and Ausubel et al., supra.
  • Constructs of expression vectors, which are circular or linear, can be prepared to contain a replication system functional in a prokaryotic or eukaryotic host cell.
  • Replication systems can be derived, e.g., from CoIEl, 2 ⁇ plasmid, ⁇ , SV40, bovine papilloma virus, and the like.
  • the recombinant expression vector comprises regulatory sequences, such as transcription and translation initiation and termination codons, which are specific to the type of host (e.g., bacterium, fungus, plant, or animal) into which the vector is to be introduced, as appropriate and taking into consideration whether the vector is DNA- or RNA- based.
  • regulatory sequences such as transcription and translation initiation and termination codons, which are specific to the type of host (e.g., bacterium, fungus, plant, or animal) into which the vector is to be introduced, as appropriate and taking into consideration whether the vector is DNA- or RNA- based.
  • the recombinant expression vector may include one or more marker genes, which allow for selection of transformed or transfected hosts.
  • Marker genes include biocide resistance, e.g., resistance to antibiotics, heavy metals, etc., complementation in an auxotrophic host to provide prototrophy, and the like.
  • Suitable marker genes for the presently disclosed expression vectors include, for instance, neomycin/G418 resistance genes, hygromycin resistance genes, histidinol resistance genes, tetracycline resistance genes, and ampicillin resistance genes.
  • the recombinant expression vector can comprise a native or normative promoter operably linked to the nucleotide sequence encoding the polypeptide (including functional portions and functional variants thereof), or to the nucleotide sequence which is complementary to or which hybridizes to the nucleotide sequence encoding the polypeptide.
  • a native or normative promoter operably linked to the nucleotide sequence encoding the polypeptide (including functional portions and functional variants thereof), or to the nucleotide sequence which is complementary to or which hybridizes to the nucleotide sequence encoding the polypeptide.
  • the promoter can be a non- viral promoter or a viral promoter, e.g., a cytomegalovirus (CMV) promoter, an SV40 promoter, an RSV promoter, and a promoter found in the long-terminal repeat of the murine stem cell virus.
  • a viral promoter e.g., a cytomegalovirus (CMV) promoter, an SV40 promoter, an RSV promoter, and a promoter found in the long-terminal repeat of the murine stem cell virus.
  • CMV cytomegalovirus
  • the presently disclosed recombinant expression vectors may be designed for either transient expression, for stable expression, or for both. Also, the recombinant expression vectors may be made for constitutive expression or for inducible expression. Further, the recombinant expression vectors may be made to include a suicide gene.
  • suicide gene refers to a gene that causes the cell expressing the suicide gene to die.
  • the suicide gene in some aspects is a gene that confers sensitivity to an agent, e.g., a drug, upon the cell in which the gene is expressed, and causes the cell to die when the cell is contacted with or exposed to the agent.
  • agent e.g., a drug
  • HSV Herpes Simplex Virus
  • TK thymidine kinase
  • the compound is a cell expressing the nucleic acid of the invention, optionally, wherein the nucleic acid is pary of a recombinant expression vector.
  • the term "host cell” refers to any type of cell that can contain the presently disclosed recombinant expression vector.
  • the host cell in some aspects is a eukaryotic cell, e.g., plant, animal, fungi, or algae, or can be a prokaryotic cell, e.g., bacteria or protozoa.
  • the host cell in some aspects is a cultured cell or a primary cell, i.e., isolated directly from an organism, e.g., a human.
  • the host cell in some aspects is an adherent cell or a suspended cell, i.e., a cell that grows in suspension.
  • Suitable host cells are known in the art and include, for instance, DH5a E. coli cells, Chinese hamster ovarian cells, monkey VERO cells, COS cells, HEK293 cells, and the like.
  • the host cell is in some aspects is a prokaryotic cell, e.g., a DH5a cell.
  • the host cell is in some aspects a mammalian cell, e.g., a human cell.
  • the host cell may be of any cell type, can originate from any type of tissue, and can be of any developmental stage.
  • Also provided by the invention is a population of cells comprising at least one host cell described herein.
  • the population of cells in some aspects is a heterogeneous population comprising the host cell comprising any of the recombinant expression vectors described, in addition to at least one other cell, which does not comprise any of the recombinant expression vectors.
  • the population of cells is a substantially homogeneous population, in which the population comprises mainly of host cells (e.g., consisting essentially of) comprising the recombinant expression vector.
  • the population in some aspects is a clonal population of cells, in which all cells of the population are clones of a single host cell comprising a recombinant expression vector, such that all cells of the population comprise the recombinant expression vector.
  • the population of cells is a clonal population comprising host cells comprising a recombinant expression vector as described herein.
  • the compounds of the invention are attached or linked or conjugated to a second moiety (e.g., a heterologous moiety, a conjugate moiety).
  • a second moiety e.g., a heterologous moiety, a conjugate moiety.
  • heterologous moiety is synonomous with “conjugate moiety” and refers to any molecule (chemical or biochemical, naturally- occurring or non-coded) which is different from the compounds of the invention.
  • heterologous moieties include, but are not limited to, a polymer, a carbohydrate, a lipid, a nucleic acid, an oligonucleotide, a DNA or RNA, an amino acid, peptide, polypeptide, protein, therapeutic agent, (e.g., a cytotoxic agent, cytokine), or a diagnostic agent.
  • a polymer e.g., a polymer, a carbohydrate, a lipid, a nucleic acid, an oligonucleotide, a DNA or RNA, an amino acid, peptide, polypeptide, protein, therapeutic agent, (e.g., a cytotoxic agent, cytokine), or a diagnostic agent.
  • the compounds are chemically modified with various substituents.
  • the chemical modifications impart additional desirable characteristics as discussed herein.
  • Chemical modifications in some aspects take a number of different forms such as heterologous peptides, polysaccarides, lipids, radioisotopes, non-standard amino acid resides and nucleic acids, metal chelates, and various cytotoxic agents.
  • the compounds are fused to heterologous peptides to confer various properties, e.g., increased solubility and/or stability and/or half-life, resistance to proteolytic cleavage, modulation of clearance, targeting to particular cell or tissue types.
  • the compound is linked to a Fc domain of IgG or other immunoglobulin.
  • the compound is fused to alkaline phosphatase (AP). Methods for making Fc or AP fusion constructs are found in WO 02/060950. By fusing the compound with protein domains that have specific properties (e.g. half life, bioavailability) it is possible to confer these properties to the the compound of the invention.
  • the peptides also can be modified to create peptide derivatives by forming covalent or noncovalent complexes with other moieties.
  • Covalently bound complexes can be prepared by linking the chemical moieties to functional groups on the side chains of amino acids comprising the peptides, or at the N- or C-terminus.
  • Peptides can be conjugated to a reporter group, including, but not limited to a radiolabel, a fluorescent label, an enzyme (e.g., that catalyzes a calorimetric or fluorometric reaction), a substrate, a solid matrix, or a carrier (e.g., biotin or avidin).
  • a reporter group including, but not limited to a radiolabel, a fluorescent label, an enzyme (e.g., that catalyzes a calorimetric or fluorometric reaction), a substrate, a solid matrix, or a carrier (e.g., biotin or avidin).
  • a carrier e.g., biotin or avidin
  • Cysteinyl residues most commonly are reacted with haloacetates (and corresponding amines), such as chloroacetic acid or chloroacetamide, to give carboxymethyl or carbocyamidomethyl derivatives. Cysteinyl residues also are derivatized by reaction with bromotrifluoroacetone, a-bromo-.beta.(5-imidozoyl)propionic acid, chloroacetyl phosphate, N- alkylmaleimides, 3-nitro-2-pyridyl disulfide, methyl 2-pyridyl disulfide, p- chloromercuribenzoate, 2-chloromercuri-4-nitrophenol, orchloro-7-nitrobenzo-2-oxa- 1 ,3-diazole.
  • Histidyl residues are derivatized by reaction with diethylprocarbonate at pH 5.5-7.0 because this agent is relatively specific for the histidyl side chain.
  • Para-bromophenacyl bromide also is useful; the reaction is preferably performed in 0.1M sodium cacodylate at pH 6.0.
  • Lysinyl and amino terminal residues are reacted with succinic or carboxylic acid anhydrides. Derivatization with these agents has the effect of reversing the charge of the lysinyl residues.
  • Other suitable reagents for derivatizing a-amino-containing residues include imidoesters such as methyl picolinimidate; pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylissurea; 2,4 pentanedione; and transaminase catalyzed reaction with glyoxylate.
  • Arginyl residues are modified by reaction with one or several conventional reagents, among them phenylglyoxal, 2,3-butanedione, 1,2-cyclohexanedione, and ninhydrin.
  • arginine residues requires that the reaction be performed in alkaline conditions because of the high pK of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the arginine epsilon-amino group.
  • tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively.
  • Carboxyl side groups are selectively modified by reaction with carbodiimides (Rl) such as l-cyclohexyl-3-(2-morpholinyl-(4-ethyl) carbodiimide or l-ethyl-3 (4 azonia 4,4-dimethylpentyl)carbodiimide. Furthermore, aspartyl and glutamyl residues are converted to asparaginyl and glutaminyl residues by reaction with ammonium ions.
  • Derivatization with bifunctional agents is useful for crosslinking the binding construct to water-insoluble support matrixes. Such derivation may also provide the linker that may connect adjacent binding elements in a binding construct, or a binding elements to a
  • heterologous peptide e.g., a Fc fragment.
  • crosslinking agents include, e.g., 1,1- bis(diazoacetyl)-2-phenylethane, glutaraldehyde, N-hydroxysuccinimide esters, for example, esters with 4-azidosalicylic acid, homo-bifunctional imidoesters, including disuccinimidyl esters such as 3,3'-dithiiobis(succinimidylpropioonate), and bifunctional maleimides such as bis-N- maleimido-l,8-octane.
  • Derivatizing agents such as methyl-3-[(p-azidophenyl) dithio] propioimidate yield photoactivatable intermediates that are capable of forming cross links in the presence of light.
  • reactive water-insoluble matrices such as cyanogen bromide- activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287;
  • Glutaminyl and asparaginyl residues are frequently deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues falls within the scope of this invention.
  • modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the a-amino groups of lysine, arginine, and histidine side chains (T. E. Creighton, Proteins: Structure and Molecule Properties, W. H. Freeman & Co., San Francisco, pp. 79-86,1983), acetylation of the N-terminal amine, and, in some instances, amidation of the C-terminal carboxyl groups.
  • Such derivatives are chemically modified polypeptide compositions in which the binding construct polypeptide is linked to a polymer.
  • chemical derivatization may be performed under any suitable condition used to react a protein with an activated polymer molecule.
  • Methods for preparing chemical derivatives of polypeptides will generally comprise the steps of (a) reacting the polypeptide with the activated polymer molecule (such as a reactive ester or aldehyde derivative of the polymer molecule) under conditions whereby the binding construct becomes attached to one or more polymer molecules, and (b) obtaining the reaction product(s).
  • the optimal reaction conditions will be determined based on known parameters and the desired result. For example, the larger the ratio of polymer molecules:protein, the greater the amount of attached polymer molecule.
  • the compound may have a single polymer molecule moiety at the amino terminus. (See, e.g., U.S. Pat. No. 5,234,784).
  • Derivatized binding constructs disclosed herein may have additional activities, enhanced or reduced biological activity, or other characteristics, such as increased or decreased half-life, as compared to the non-derivatized molecules.
  • the compound is directly joined to a conjugate moiety in the absence of a linker.
  • the compound is indirectly connected to the conjugate moiety via one or more linkers.
  • the compound may be connected through covalent bonds (e.g., a peptide, ester, amide, or sulfhydryl bond) or non-covalent bonds (e.g., via hydrophobic interaction, hydrogen bond, van der Waals bond, electrostatic or ionic interaction), or a combination thereof.
  • the compound of the invention and conjugate moiety may be connected via any means known in the art, including, but not limited to, via a linker of any of the invention. See, for example, the section herein entitled "Linkers. "
  • the fusion can be fused directly to a compound of the invention or fused through an intervening sequence.
  • a human IgG hinge, CH2 and CH3 region may be fused at either the N-terminus or C-terminus of a binding construct to attach the Fc region.
  • the resulting Fc-fusion construct enables purification via a Protein A affinity column (Pierce, Rockford, 111.).
  • Peptide and proteins fused to an Fc region can exhibit a substantially greater half-life in vivo than the unfused counterpart.
  • a fusion to an Fc region allows for dimerization/multimerization of the fusion polypeptide.
  • the Fc region may be a naturally occurring Fc region, or may be modified for superior characteristics, e.g., therapeutic qualities, circulation time, reduced aggregation. As noted above, in some embodiments thereof.
  • the compounds are conjugated, e.g., fused to an immunoglobulin or portion thereof (e.g., variable region, CDR, or Fc region).
  • immunoglobulins e.g., variable region, CDR, or Fc region.
  • immunoglobulins include IgG, IgA, IgE, IgD or IgM.
  • the Fc region is a C-terminal region of an Ig heavy chain, which is responsible for binding to Fc receptors that carry out activities such as recycling (which results in prolonged half-life), antibody dependent cell-mediated cytotoxicity (ADCC), and complement dependent cytotoxicity (CDC).
  • the human IgG heavy chain Fc region stretches from Cys226 to the C-terminus of the heavy chain.
  • the "hinge region” generally extends from Glu216 to Pro230 of human IgGl (hinge regions of other IgG isotypes may be aligned with the IgGl sequence by aligning the cysteines involved in cysteine bonding).
  • the Fc region of an IgG includes two constant domains, CH2 and CH3.
  • the CH2 domain of a human IgG Fc region usually extends from amino acids 231 to amino acid 341.
  • the CH3 domain of a human IgG Fc region usually extends from amino acids 342 to 447.
  • the Fc region may comprise one or more native or modified constant regions from an immunoglobulin heavy chain, other than CHI, for example, the CH2 and CH3 regions of IgG and IgA, or the CH3 and CH4 regions of IgE.
  • Suitable conjugate moieties include portions of immunoglobulin sequence that include the FcRn binding site.
  • FcRn a salvage receptor, is responsible for recycling
  • the region of the Fc portion of IgG that binds to the FcRn receptor has been described based on X-ray crystallography (Burmeister et al. 1994, Nature 372:379).
  • the major contact area of the Fc with the FcRn is near the junction of the CH2 and CH3 domains. Fc-FcRn contacts are all within a single Ig heavy chain.
  • the major contact sites include amino acid residues 248, 250-257, 272, 285, 288, 290-291, 308-311, and 314 of the CH2 domain and amino acid residues 385-387, 428, and 433-436 of the CH3 domain.
  • FcyR are responsible for ADCC and CDC.
  • positions within the Fc region that make a direct contact with FcyR are amino acids 234-239 (lower hinge region), amino acids 265-269 (B/C loop), amino acids 297-299 (C'/E loop), and amino acids 327-332 (F/G) loop (Sondermann et al., Nature 406: 267-273, 2000).
  • the lower hinge region of IgE has also been implicated in the FcRI binding (Henry, et al., Biochemistry 36, 15568-15578, 1997).
  • Amino acid modifications may be made to the Fc region of an immunoglobulin.
  • Such variant Fc regions comprise at least one amino acid modification in the CH3 domain of the Fc region (residues 342-447) and/or at least one amino acid modification in the CH2 domain of the Fc region (residues 231-341).
  • Mutations believed to impart an increased affinity for FcRn include T256A, T307A, E380A, and N434A (Shields et al. 2001, J. Biol. Chem. 276:6591).
  • FcyRI FcyRIIA, FcyRIIB, and/or FcyRIIIA
  • substitution of the Asn at position 297 of the Fc region with Ala or another amino acid removes a highly conserved N- glycosylation site and may result in reduced immunogenicity with concomitant prolonged half- life of the Fc region, as well as reduced binding to FcyRs (Routledge et al. 1995, Transplantation 60:847; Friend et al. 1999, Transplantation 68: 1632; Shields et al. 1995, J. Biol. Chem.
  • the heterologous moiety is a polymer.
  • the polymer may be branched or unbranched.
  • the polymer may be of any molecular weight.
  • the polymer in some embodiments has an average molecular weight of between about 2 kDa to about 100 kDa (the term "about” indicating that in preparations of a water soluble polymer, some molecules will weigh more, some less, than the stated molecular weight).
  • the average molecular weight of the polymer is in some aspect between about 5 kDa and about 50 kDa, between about 12 kDa to about 40 kDa or between about 20 kDa to about 35 kDa.
  • the polymer is modified to have a single reactive group, such as an active ester for acylation or an aldehyde for alkylation, so that the degree of polymerization may be controlled.
  • the polymer in some embodiments is water soluble so that the protein to which it is attached does not precipitate in an aqueous environment, such as a physiological environment.
  • the polymer when, for example, the composition is used for therapeutic use, the polymer is pharmaceutically acceptable.
  • the polymer is a mixture of polymers, e.g., a co-polymer, a block co-polymer.
  • the polymer is selected from the group consisting of:
  • the polymer is a biodegradable polymer, including a synthetic biodegradable polymer (e.g., polymers of lactic acid and glycolic acid, polyanhydrides, poly(ortho)esters, polyurethanes, poly(butic acid), poly(valeric acid), and poly(lactide- cocaprolactone)), and a natural biodegradable polymer (e.g., alginate and other polysaccharides including dextran and cellulose, collagen, chemical derivatives thereof (substitutions, additions of chemical groups, for example, alkyl, alkylene, hydroxylations, oxidations, and other modifications routinely made by those skilled in the art), albumin and other hydrophilic proteins (e.g., zein and other prolamines and hydrophobic proteins)), as well as any copolymer or mixture thereof.
  • these materials degrade either by enzymatic hydrolysis or exposure to water in vivo, by surface or bulk erosion.
  • the polymer is a bioadhesive polymer, such as a bioerodible hydrogel described by H. S. Sawhney, C. P. Pathak and J. A. Hubbell in Macromolecules, 1993, 26, 581-587, the teachings of which are incorporated herein, polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly(isobutyl methacrylate),
  • a bioadhesive polymer such as a bioerodible hydrogel described by H. S. Sawhney, C. P. Pathak and J. A. Hubbell in Macromolecules, 1993, 26, 581-587, the teachings of which are incorporated herein, polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chi
  • the polymer is a water-soluble polymer or a hydrophilic polymer.
  • Suitable water-soluble polymers are known in the art and include, for example, polyvinylpyrrolidone, hydroxypropyl cellulose (HPC; Klucel), hydroxypropyl methylcellulose (HPMC; Methocel), nitrocellulose, hydroxypropyl ethylcellulose, hydroxypropyl butylcellulose, hydroxypropyl pentylcellulose, methyl cellulose, ethylcellulose (Ethocel), hydroxyethyl cellulose, various alkyl celluloses and hydroxyalkyl celluloses, various cellulose ethers, cellulose acetate, carboxymethyl cellulose, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, vinyl acetate/crotonic acid copolymers, poly-hydroxyalkyl methacrylate,
  • polymethylmethacrylate polymethylmethacrylate, maleic anhydride/methyl vinyl ether copolymers, poly vinyl alcohol, sodium and calcium polyacrylic acid, polyacrylic acid, acidic carboxy polymers,
  • carboxypolymethylene carboxypolymethylene, carboxyvinyl polymers, polyoxyethylene polyoxypropylene copolymer, polymethylvinylether co-maleic anhydride, carboxymethylamide, potassium methacrylate divinylbenzene co-polymer, polyoxyethyleneglycols, polyethylene oxide, and derivatives, salts, and combinations thereof.
  • the water soluble polymers or mixtures thereof include, but are not limited to, N-linked or O-linked carbohydrates, sugars, phosphates, carbohydrates; sugars; phosphates; polyethylene glycol (PEG) (including the forms of PEG that have been used to derivatize proteins, including mono-(Cl-C 10) alkoxy- or aryloxy- polyethylene glycol); monomethoxy-polyethylene glycol; dextran (such as low molecular weight dextran, of, for example about 6 kD), cellulose; cellulose; other carbohydrate-based polymers, poly-(N-vinyl pyrrolidone)polyethylene glycol, propylene glycol homopolymers, a
  • polypropylene oxide/ethylene oxide co-polymer polypropylene oxide/ethylene oxide co-polymer
  • polyoxyethylated polyols e.g., glycerol
  • polyvinyl alcohol polyvinyl alcohol
  • bifunctional crosslinking molecules which may be used to prepare covalently attached multimers.
  • a particularly preferred water-soluble polymer for use herein is polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • polyethylene glycol is meant to encompass any of the forms of PEG that can be used to derivatize other proteins, such as mono-(Cl-ClO) alkoxy- or aryloxy-polyethylene glycol.
  • PEG is a linear or branched neutral polyether, available in a broad range of molecular weights, and is soluble in water and most organic solvents. PEG is effective at excluding other polymers or peptides when present in water, primarily through its high dynamic chain mobility and hydrophibic nature, thus creating a water shell or hydration sphere when attached to other proteins or polymer surfaces.
  • PEG is nontoxic, non-immunogenic, and approved by the Food and Drug Administration for internal consumption.
  • Proteins or enzymes when conjugated to PEG have demonstrated bioactivity, non- antigenic properties, and decreased clearance rates when administered in animals. F. M.
  • Veronese et al. Preparation and Properties of Monomethoxypoly(ethylene glycol) -modified Enzymes for Therapeutic Applications, in J. M. Harris ed., Poly(Ethylene Glycol) Chemistry— Biotechnical and Biomedical Applications, 127-36, 1992, incorporated herein by reference. These phenomena are due to the exclusion properties of PEG in preventing recognition by the immune system. In addition, PEG has been widely used in surface modification procedures to decrease protein adsorption and improve blood compatibility. S. W. Kim et al., Ann. N.Y. Acad. Sci. 516: 116-30 1987; Jacobs et al., Artif.
  • Hydrophobic polymer surfaces such as polyurethanes and polystyrene can be modified by the grafting of PEG (MW 3,400) and employed as nonthrombogenic surfaces.
  • Surface properties can be more consistent with hydrophilic surfaces, due to the hydrating effect of PEG. More importantly, protein (albumin and other plasma proteins) adsorption can be greatly reduced, resulting from the high chain motility, hydration sphere, and protein exclusion properties of PEG.
  • PEG MW 3,400 was determined as an optimal size in surface immobilization studies, Park et al., J. Biomed. Mat. Res. 26:739-45, 1992, while PEG (MW 5,000) was most beneficial in decreasing protein antigenicity.
  • Methods for preparing pegylated compounds may comprise the steps of (a) reacting the compound with polyethylene glycol (such as a reactive ester or aldehyde derivative of PEG) under conditions whereby the compound becomes attached to one or more PEG groups, and (b) obtaining the reaction product(s).
  • polyethylene glycol such as a reactive ester or aldehyde derivative of PEG
  • the optimal reaction conditions for the acylation reactions will be determined based on known parameters and the desired result. For example, the larger the ratio of PEG: compound, the greater the percentage of poly-pegylated product.
  • the compound will have a single PEG moiety at the N-terminus. See U.S. Pat. No. 8,234,784, herein incorporated by reference.
  • the heterologous moiety is a carbohydrate.
  • the carbohydrate is a monosaccharide (e.g., glucose, galactose, fructose), a disaccharide (e.g., sucrose, lactose, maltose), an oligosaccharide (e.g., raffinose, stachyose), a polysaccharide (a starch, amylase, amylopectin, cellulose, chitin, callose, laminarin, xylan, mannan, fucoidan, galactomannan.
  • a monosaccharide e.g., glucose, galactose, fructose
  • a disaccharide e.g., sucrose, lactose, maltose
  • an oligosaccharide e.g., raffinose, stachyose
  • a polysaccharide a starch,
  • the heterologous moiety is a lipid.
  • the lipid in some embodiments, is a fatty acid, eicosanoid, prostaglandin, leukotriene, thromboxane, N-acyl ethanolamine), glycerolipid (e.g., mono-, di-, tri- substituted glycerols), glycerophospholipid (e.g., phosphatidylcholine, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine), sphingolipid (e.g., sphingosine, ceramide), sterol lipid (e.g., steroid, cholesterol), prenol lipid, saccharolipid, or a polyketide, oil, wax, cholesterol, sterol, fat-soluble vitamin, monoglyceride, diglyceride, triglyceride, a phospholipid.
  • glycerolipid e.g
  • the heterologous moiety is a therapeutic agent.
  • the therapeutic agent may be any of those known in the art.
  • therapeutic agents that are contemplated herein include, but are not limited to, natural enzymes, proteins derived from natural sources, recombinant proteins, natural peptides, synthetic peptides, cyclic peptides, antibodies, receptor agonists, cytotoxic agents, immunoglobins, beta-adrenergic blocking agents, calcium channel blockers, coronary vasodilators, cardiac glycosides, antiarrhythmics, cardiac sympathomemetics, angiotensin converting enzyme (ACE) inhibitors, diuretics, inotropes, cholesterol and triglyceride reducers, bile acid sequestrants, fibrates, 3-hydroxy-3-methylgluteryl (HMG)-CoA reductase inhibitors, niacin derivatives, antiadrenergic agents, alpha-adrenergic blocking agents, centrally acting antiadren
  • HMG 3-hydroxy-3
  • cytoprotectives histamine H2-receptor antagonists, mucosal protective agent, proton pump inhibitors, H. pylori eradication therapy, erythropoieses stimulants, hematopoietic agents, anemia agents, heparins, antifibrinolytics, hemostatics, blood coagulation factors, adenosine diphosphate inhibitors, glycoprotein receptor inhibitors, fibrinogen-platelet binding inhibitors, thromboxane- A 2 inhibitors, plasminogen activators, antithrombotic agents, glucocorticoids, mineralcorticoids, corticosteroids, selective immunosuppressive agents, antifungals, drugs involved in prophylactic therapy, AIDS-associated infections, cytomegalovirus, non-nucleoside reverse transcriptase inhibitors, nucleoside analog reverse transcriptse inhibitors, protease inhibitors, anemia, Kaposi's sarcoma, aminoglycosides, carbapenems, cephal
  • benzodiazepines gamma aminobutyric acid derivatives, hydantoin derivatives, iminostilbene derivatives, succinimide derivatives, anticonvulsants, ergot alkaloids, antimigrane preparations, biological response modifiers, carbamic acid eaters, tricyclic derivatives, depolarizing agents, nondepolarizing agents, neuromuscular paralytic agents, CNS stimulants, dopaminergic reagents, monoamine oxidase inhibitors, COMT inhibitors, alkyl sulphonates, ethylenimines,
  • imidazotetrazines nitrogen mustard analogs, nitrosoureas, platinum-containing compounds, antimetabolites, purine analogs, pyrimidine analogs, urea derivatives, antracyclines,
  • actinomycinds camptothecin derivatives, epipodophyllotoxins, taxanes, vinca alkaloids and analogs, antiandrogens, antiestrogens, nonsteroidal aromatase inhibitors, protein kinase inhibitor antineoplastics, azaspirodecanedione derivatives, anxiolytics, stimulants, monoamind reuptake inhibitors, selective serotonin reuptake inhibitors, antidepressants, benzisooxazole derivatives, butyrophenone derivatives, dibenzodiazepine derivatives, dibenzothiazepine derivatives, diphenylbutylpiperidine derivatives, phenothiazines, thienobenzodiazepine derivatives, thioxanthene derivatives, allergenic extracts, nonsteroidal agents, leukotriene receptor antagonists, xanthines, endothelin receptor antagonist, prostaglandins, lung surfactants, mucolytics, antimit
  • parasympathomimetic agents halogenated hydrocarbons, esters of amino benzoic acid, amides (e.g. lidocaine, articaine hydrochloride, bupivacaine hydrochloride), antipyretics, hynotics and sedatives, cyclopyrrolones, pyrazolopyrimidines, nonsteroidal anti-inflammatory drugs, opioids, para-aminophenol derivatives, alcohol dehydrogenase inhibitor, heparin antagonists, adsorbents, emetics, opoid antagonists, cholinesterase reactivators, nicotine replacement therapy, vitamin A analogs and antagonists, vitamin B analogs and antagonists, vitamin C analogs and antagonists, vitamin D analogs and antagonists, vitamin E analogs and antagonists, vitamin K analogs and antagonists.
  • amides e.g. lidocaine, articaine hydrochloride, bupivacaine hydrochloride
  • antipyretics e.g
  • the compounds of the invention may be conjugated to one or more cytokines and growth factors that are effective in inhibiting tumor metastasis, and wherein the cytokine or growth factor has been shown to have an antiproliferative effect on at least one cell population.
  • Such cytokines, lymphokines, growth factors, or other hematopoietic factors include, but are not limited to: M-CSF, GM-CSF, TNF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IFN, TNFa, TNF1, TNF2, G-CSF, Meg- CSF, GM-CSF, thrombopoietin, stem cell factor, and erythropoietin.
  • Additional growth factors for use herein include angiogenin, bone morphogenic protein- 1, bone morphogenic protein-2, bone morphogenic protein-3, bone morphogenic protein-4, bone morphogenic protein-5, bone morphogenic protein-6, bone morphogenic protein-7, bone morphogenic protein-8, bone morphogenic protein-9, bone morphogenic protein- 10, bone morphogenic protein-11, bone morphogenic protein- 12, bone morphogenic protein- 13, bone morphogenic protein- 14, bone morphogenic protein- 15, bone morphogenic protein receptor IA, bone morphogenic protein receptor IB, brain derived neurotrophic factor, ciliary neutrophic factor, ciliary neutrophic factor receptor a, cytokine-induced neutrophil chemotactic factor 1, cytokine-induced neutrophil, chemotactic factor 2 a, cytokine-induced neutrophil chemotactic factor 2 ⁇ , ⁇ endothelial cell growth factor, endothelin 1, epithelial-derived neutrophil attractant, glial cell line-derived neutrophic factor
  • the conjugate comprises a compound as described herein and a cytotoxic agent.
  • the cytotoxic agent is any molecule (chemical or biochemical) which is toxic to a cell.
  • the results obtained are synergistic. That is to say, the effectiveness of the combination therapy of a compound and the cytotoxic agent is synergistic, i.e., the effectiveness is greater than the effectiveness expected from the additive individual effects of each. Therefore, the dosage of the cytotoxic agent can be reduced and thus, the risk of the toxicity problems and other side effects is concomitantly reduced.
  • the cytotoxic agent is a chemotherapeutic agent.
  • Chemotherapeutic agents are known in the art and include, but not limited to, platinum coordination compounds, topoisomerase inhibitors, antibiotics, antimitotic alkaloids and difluoronucleosides, as described in U.S. Pat. No. 6,630,124.
  • the chemotherapeutic agent is a platinum coordination compound.
  • platinum coordination compound refers to any tumor cell growth inhibiting platinum coordination compound that provides the platinum in the form of an ion.
  • the platinum coordination compound is cis-diamminediaquoplatinum (II)- ion ; chloro (diethylenetriamine) -platinum(II)chloride ; dichloro (ethylenediamine) -platinum(II) , diammine(l,l-cyclobutanedicarboxylato) platinum(II) (carboplatin); spiroplatin; iproplatin; diammine(2-ethylmalonato)-platinum(II); ethylenediaminemalonatoplatinum(II); aqua( 1 ,2- diaminodyclohexane)-sulfatoplatinum(II); (l,2-diaminocyclohexan
  • cisplatin is the platinum coordination compound employed in the compositions and methods of the present invention.
  • Cisplatin is commercially available under the name PLATINOLTM from Bristol Myers-Squibb Corporation and is available as a powder for constitution with water, sterile saline or other suitable vehicle.
  • Other platinum coordination compounds suitable for use in the present invention are known and are available commercially and/or can be prepared by conventional techniques.
  • Cisplatin, or cis- dichlorodiammineplatinum II has been used successfully for many years as a chemotherapeutic agent in the treatment of various human solid malignant tumors.
  • diamino- platinum complexes have also shown efficacy as chemotherapeutic agents in the treatment of various human solid malignant tumors.
  • diamino-platinum complexes include, but are not limited to, spiroplatinum and carboplatinum.
  • cisplatin and other diamino-platinum complexes have been widely used as chemotherapeutic agents in humans, they have had to be delivered at high dosage levels that can lead to toxicity problems such as kidney damage.
  • the chemotherapeutic agent is a topoisomerase inhibitor.
  • Topoisomerases are enzymes that are capable of altering DNA topology in eukaryotic cells. They are critical for cellular functions and cell proliferation. Generally, there are two classes of topoisomerases in eukaryotic cells, type I and type II. Topoisomerase I is a monomeric enzyme of approximately 100,000 molecular weight. The enzyme binds to DNA and introduces a transient single-strand break, unwinds the double helix (or allows it to unwind), and
  • the topoisomerase inhibitor is camptothecin or a camptothecin analog.
  • Camptothecin is a water-insoluble, cytotoxic alkaloid produced by Camptotheca accuminata trees indigenous to China and Nothapodytes foetida trees indigenous to India.
  • Camptothecin exhibits tumor cell growth inhibiting activity against a number of tumor cells.
  • Compounds of the camptothecin analog class are typically specific inhibitors of DNA
  • topoisomerase I By the term “inhibitor of topoisomerase” is meant any tumor cell growth inhibiting compound that is structurally related to camptothecin. Compounds of the
  • camptothecin analog class include, but are not limited to; topotecan, irinotecan and 9-amino- camptothecin.
  • the cytotoxic agent is any tumor cell growth inhibiting camptothecin analog claimed or described in: U.S. Pat. No. 5,004,758, issued on Apr. 2, 1991 and European Patent Application Number 88311366.4, published on Jun. 21, 1989 as 20' Publication Number EP 0 321 122; U.S. Pat. No. 4,604,463, issued on Aug. 5, 1986 and
  • CPT-11 is a camptothecin analog with a 4-(piperidino)-piperidine side chain joined through a carbamate linkage at C-10 of 10-hydroxy-7 -ethyl camptothecin.
  • CPT-11 is currently undergoing human clinical trials and is also referred to as irinotecan; Wani et al, J. Med. Chem., 23, 554 (1980); Wani et. al., J. Med. Chem., 30, 1774 (1987); U.S. Pat. No.
  • the chemotherapeutic agent is an antibiotic compound.
  • Suitable antibiotic include, but are not limited to, doxorubicin, mitomycin, bleomycin, daunorubicin and streptozocin.
  • the chemotherapeutic agent is an antimitotic alkaloid.
  • antimitotic alkaloids can be extracted from Cantharanthus roseus, and have been shown to be efficacious as anticancer chemotherapy agents.
  • a great number of semi- synthetic derivatives have been studied both chemically and pharmacologically (see, O. Van Tellingen et al, Anticancer Research, 12, 1699-1716 (1992)).
  • the antimitotic alkaloids of the present invention include, but are not limited to, vinblastine, vincristine, vindesine, Taxol and vinorelbine.
  • the latter two antimitotic alkaloids are commercially available from Eli Lilly and Company, and Pierre Fabre Laboratories, respectively (see, U.S. Pat. No. 5,620,985).
  • the antimitotic alkaloid is vinorelbine.
  • the chemotherapeutic agent is a
  • 2'-deoxy-2',2'-difluoronucleosides are known in the art as having antiviral activity. Such compounds are disclosed and taught in U.S. Pat. Nos. 4,526,988 and 4,808614. European Patent Application Publication 184,365 discloses that these same difluoronucleosides have oncolytic activity.
  • the 2'-deoxy-2',2'-difluoronucleoside used in the compositions and methods of the present invention is 2'-deoxy-2',2'-difluorocytidine hydrochloride, also known as gemcitabine hydrochloride.
  • Gemcitabine is commercially available or can be synthesized in a multi-step process as disclosed and taught in U.S. Pat. Nos. 4,526,988, 4,808,614 and 5,223,608, the teachings of which are incorporated herein by reference.
  • the compounds of the present disclosure can be modified in any number of ways, such that the therapeutic or prophylactic efficacy of the compound of the invention is increased through the modification.
  • the compound of the present disclosure can be conjugated either directly or indirectly through a linker to a targeting moiety.
  • the practice of conjugating compounds to targeting moieties is known in the art. See, for instance, Wadhwa et al., J Drug Targeting, 3, 111-127 (1995) and U.S. Patent No. 5,087,616.
  • targeting moiety refers to any molecule or agent that specifically recognizes and binds to a cell-surface receptor, such that the targeting moiety directs the delivery of the compound of the invention to a population of cells on which surface the receptor is expressed.
  • Targeting moieties include, but are not limited to, antibodies, or fragments thereof, peptides, hormones, growth factors, cytokines, and any other natural or non-natural ligands, which bind to cell surface receptors (e.g., Epithelial Growth Factor Receptor (EGFR), T-cell receptor (TCR), B-cell receptor (BCR), CD28, Platelet-derived Growth Factor Receptor (PDGF), nicotinic acetylcholine receptor (nAChR), etc.).
  • EGFR Epithelial Growth Factor Receptor
  • TCR T-cell receptor
  • BCR B-cell receptor
  • CD28 CD28
  • PDGF Platelet-derived Growth Factor Receptor
  • nAChR nicotinic acetylcholine receptor
  • Linkers may provide for optimal spacing of the two entities or may further supply a labile linkage that allows the two entities to be separated from each other.
  • Labile linkages include photocleavable groups, acid-labile moieties, base-labile moieties and enzyme-cleavable groups.
  • the term "linker” in some embodiments refers to any agent or molecule that bridges the compound of the invention to the targeting moiety.
  • the linker may be any of those described herein under the section entitled "Linkers.”
  • sites on the compound of the invention, which are not necessary for the function of the compound are ideal sites for attaching a linker and/or a targeting moiety, provided that the linker and/or targeting moiety, once attached to the compound, do(es) not interfere with the function of the compound, i.e., the ability to inhibit the binding interaction between ⁇ integrin and G protein a subunit, as described herein.
  • the conjugate comprises a linker that joins the compound of the invention to the heterologous moiety.
  • the linker comprises a chain of atoms from 1 to about 60, or 1 to 30 atoms or longer, 2 to 5 atoms, 2 to 10 atoms, 5 to 10 atoms, or 10 to 20 atoms long.
  • the chain atoms are all carbon atoms.
  • the chain atoms in the backbone of the linker are selected from the group consisting of C, O, N, and S. Chain atoms and linkers may be selected according to their expected solubility (hydrophilicity) so as to provide a more soluble conjugate.
  • the linker provides a functional group that is subject to cleavage by an enzyme or other catalyst or hydrolytic conditions found in the target tissue or organ or cell.
  • the length of the linker is long enough to reduce the potential for steric hindrance.
  • the linker is an amino acid or a peptidyl linker. Such peptidyl linkers may be any length. Exemplary linkers are from about 1 to 50 amino acids in length, 5 to 50, 3 to 5, 5 to 10, 5 to 15, or 10 to 30 amino acids in length.
  • the compound is provided as a dimer or a multimer in which more than one compound of the invention are linked together.
  • the dimer in some aspects is a homodimer comprising two compounds of the same type (e.g., same structure) linked together.
  • the dimer is a heterodimer comprising two compounds of the invention, wherein the two compounds are structurally distinct from each other.
  • the multimer in some aspects is a homomultimer comprising more than one compound of the invention and each compound are of the same type (e.g., same structure).
  • the multimer is a heteromultimer comprising more than one compound of the invention and wherein at least two compounds of the heteromultimer are structurally distinct from the other.
  • the linker connecting the two (or more) compounds is a linker as described in the section entitled "Linkers.”
  • the linker is a disulfide bond.
  • each monomer of the dimer may comprise a sulfhydryl and the sulfur atom of each participates in the formation of the disulfide bond.
  • compositions comprising a compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit, e.g., an antibody, antigen binding fragment, aptamer, peptide, peptide analog, pharmaceutically acceptable salt, conjugate, multimer, dimer, as described herein.
  • the compositions in some aspects comprise the compounds in isolated and/or purified form.
  • the composition comprises a single type (e.g., structure) of a compound of the invention or comprises a combination of two or more compounds of the invention, wherein the combination comprises two or more compounds of different types (e.g., structures).
  • the composition comprises agents which enhance the chemico- physico features of the compound, e.g., via stabilizing the compound at certain temperatures, e.g., room temperature, increasing shelf life, reducing degradation, e.g., oxidation protease mediated degradation, increasing half-life of the compound, etc.
  • the composition comprises any of the agents disclosed herein as a heterologous moiety or conjugate moiety, optionally in admixture with the compounds of the invention or conjugated to the compounds.
  • the composition comprises a delivery agent which aids in localizing the compound of the invention to the appropriate place.
  • the composition comprises a vehicle which aids in getting the compound of the invention inside a cell.
  • the vehicle is covalently attached to the compound.
  • the composition comprises a vehicle in admixture with the compound.
  • the vehicle comprises or is any of the heterologous moieties described herein with regard to conjugates.
  • the vehicle may be a polymer, e.g., water soluble polymer, which may be linear or branched.
  • the water soluble polymer is selected from the group consisting of polyethylene glycol (PEG), branched PEG, polysialic acid (PSA), carbohydrate, polysaccharides, pullulane, chitosan, hyaluronic acid, chondroitin sulfate, dermatan sulfate, starch or a starch derivative, dextran, carboxymethyl-dextran, polyalkylene oxide (PAO), polyalkylene glycol (PAG), polypropylene glycol (PPG), polyoxazoline, poly acryloylmorpholine, polyvinyl alcohol (PVA), polycarboxylate, polyvinylpyrrolidone, polyphosphazene, polyoxazoline, polyethylene-co-maleic acid anhydride, polystyrene-co-maleic acid anhydride, poly(l-hydroxymethylethylene hydroxymethylformal) (PHF), and 2- methacryloyloxy-2' -ethacrylate, polyethylene
  • the vehicle comprises a lipophilic moiety.
  • the vehicle comprises a fatty acid.
  • the fatty acid may be a C4 to C30 fatty acid, e.g., C4 fatty acid, C6 fatty acid, C8 fatty acid, CIO fatty acid, C12 fatty acid, C14 fatty acid, C16 fatty acid, CI 8 fatty acid, C20 fatty acid, C22 fatty acid, C24 fatty acid, C26 fatty acid, C28 fatty acid, or a C30 fatty acid.
  • the fatty acid is a C 12 to C30 fatty acid.
  • the fatty acid is a myristoyl group or a palmitoyl group.
  • the compound is a peptide or peptide analog and the fatty acid is covalently attached to the peptide or peptide analog.
  • the fatty acid is covalently attached to the peptide or peptide analog at the N-terminus or C-terminus or via a side chain of a non-terminal amino acid of the peptide or peptide analog.
  • the vehicle comprises a polypeptide, which when in the composition improves the ability of the composition to enter a cell compared to the ability of the composition in the absence of the polypeptide.
  • the composition comprises a compound of the invention (e.g., a peptide that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit) and a peptide delivery agent.
  • the peptide delivery agent is a cell penetrating peptide (CPP).
  • the composition comprises a CPP fused to the compound, e.g., the composition comprises a fusion peptide product comprising a peptide of the invention that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit fused to a CPP.
  • the composition comprises a compound that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit and additionally comprises a pharmaceutically acceptable carrier, diluents, or excipient.
  • the compound of the invention, the pharmaceutically acceptable salt, the conjugate, the dimer or multimer, of the invention (hereinafter referred to as "active agents") is formulated into a pharmaceutical composition comprising the the active agent, along with a pharmaceutically acceptable carrier, diluent, or excipient.
  • active agents is formulated into a pharmaceutical composition comprising the the active agent, along with a pharmaceutically acceptable carrier, diluent, or excipient.
  • compositions comprising an active agent that inhibits a binding interaction between a ⁇ integrin and a G protein a subunit which is intended for administration to a subject, e.g., a mammal.
  • the active agent is present in the pharmaceutical composition at a purity level suitable for administration to a patient.
  • the active agent has a purity level of at least about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98% or about 99%, and a pharmaceutically acceptable diluent, carrier or excipient.
  • the pharmaceutical composition in some aspects comprises the active agent of the present disclosure at a concentration of at least A, wherein A is about 0.001 mg/ml, about 0.01 mg/ml, 0 about 1 mg/ml, about 0.5 mg/ml, about 1 mg/ml, about 2 mg/ml, about 3 mg/ml, about 4 mg/ml, about 5 mg/ml, about 6 mg/ml, about 7 mg/ml, about 8 mg/ml, about 9 mg/ml, about 10 mg/ml, about 11 mg/ml, about 12 mg/ml, about 13 mg/ml, about 14 mg/ml, about 15 mg/ml, about 16 mg/ml, about 17 mg/ml, about 18 mg/ml, about 19 mg/ml, about 20 mg/ml, about 21 mg/ml, about 22 mg/ml, about 23 mg/ml, about 24 mg/ml, about 25 mg/ml or higher.
  • A is about 0.001 mg/ml, about 0.01
  • the pharmaceutical composition comprises the active agent at a concentration of at most B, wherein B is about 30 mg/ml, about 25 mg/ml, about 24 mg/ml, about 23, mg/ml, about 22 mg/ml, about 21 mg/ml, about 20 mg/ml, about 19 mg/ml, about 18 mg/ml, about 17 mg/ml, about 16 mg/ml, about 15 mg/ml, about 14 mg/ml, about 13 mg/ml, about 12 mg/ml, about 11 mg/ml, about 10 mg/ml, about 9 mg/ml, about 8 mg/ml, about 7 mg/ml, about 6 mg/ml, about 5 mg/ml, about 4 mg/ml, about 3 mg/ml, about 2 mg/ml, about 1 mg/ml, or about 0.1 mg/ml.
  • the compositions may contain an active agent at a concentration range of A to B mg/ml, for example, about 0.001 to about 30.0 mg
  • the pharmaceutical composition may comprise additional
  • pharmaceutically acceptable ingredients including, for example, acidifying agents, additives, adsorbents, aerosol propellants, air displacement agents, alkalizing agents, anticaking agents, anticoagulants, antimicrobial preservatives, antioxidants, antiseptics, bases, binders, buffering agents, chelating agents, coating agents, coloring agents, desiccants, detergents, diluents, disinfectants, disintegrants, dispersing agents, dissolution enhancing agents, dyes, emollients, emulsifying agents, emulsion stabilizers, fillers, film forming agents, flavor enhancers, flavoring agents, flow enhancers, gelling agents, granulating agents, humectants, lubricants,
  • acidifying agents including, for example, acidifying agents, additives, adsorbents, aerosol propellants, air displacement agents, alkalizing agents, anticaking agents, anticoagulants, antimicrobial preservatives, antioxidants, antiseptics, bases, bind
  • mucoadhesives ointment bases, ointments, oleaginous vehicles, organic bases, pastille bases, pigments, plasticizers, polishing agents, preservatives, sequestering agents, skin penetrants, solubilizing agents, solvents, stabilizing agents, suppository bases, surface active agents, surfactants, suspending agents, sweetening agents, therapeutic agents, thickening agents, tonicity agents, toxicity agents, viscosity-increasing agents, water-absorbing agents, water-miscible cosolvents, water softeners, or wetting agents.
  • the pharmaceutical composition comprises any one or a combination of the following components: acacia, acesulfame potassium, acetyltributyl citrate, acetyltriethyl citrate, agar, albumin, alcohol, dehydrated alcohol, denatured alcohol, dilute alcohol, aleuritic acid, alginic acid, aliphatic polyesters, alumina, aluminum hydroxide, aluminum stearate, amylopectin, a-amylose, ascorbic acid, ascorbyl palmitate, aspartame, bacteriostatic water for injection, bentonite, bentonite magma, benzalkonium chloride, benzethonium chloride, benzoic acid, benzyl alcohol, benzyl benzoate, bronopol, butylated hydroxyanisole, butylated hydroxytoluene, butylparaben, butylparaben sodium, calcium alginate,
  • CFC chlorophenoxyethanol, chloroxylenol, corn syrup solids, anhydrous citric acid, citric acid monohydrate, cocoa butter, coloring agents, corn oil, cottonseed oil, cresol, m-cresol, o-cresol, p- cresol, croscarmellose sodium, crospovidone, cyclamic acid, cyclodextrins, dextrates, dextrin, dextrose, dextrose anhydrous, diazolidinyl urea, dibutyl phthalate, dibutyl sebacate,
  • HFC heptafluoropropane
  • HC hydrocarbons
  • dilute hydrochloric acid hydrogenated vegetable oil, type II, hydroxyethyl cellulose, 2 -hydroxyethyl- ⁇ -cyclodextrin, hydroxypropyl cellulose, low- substituted hydroxypropyl cellulose, 2-hydroxypropyl-P-cyclodextrin, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate, imidurea, indigo carmine, ion exchangers, iron oxides, isopropyl alcohol, isopropyl myristate, isopropyl palmitate, isotonic saline, kaolin, lactic acid, lactitol, lactose, lanolin, lanolin alcohols, anhydrous lanolin, lecithin, magnesium aluminum silicate, magnesium carbonate, normal
  • trimethyltetradecylammonium bromide tris buffer, trisodium edentate, vanillin, type I hydrogenated vegetable oil, water, soft water, hard water, carbon dioxide-free water, pyrogen- free water, water for injection, sterile water for inhalation, sterile water for injection, sterile water for irrigation, waxes, anionic emulsifying wax, carnauba wax, cationic emulsifying wax, cetyl ester wax, microcrystalline wax, nonionic emulsifying wax, suppository wax, white wax, yellow wax, white petrolatum, wool fat, xanthan gum, xylitol, zein, zinc propionate, zinc salts, zinc stearate, or any excipient in the Handbook of Pharmaceutical Excipients, Third Edition, A. H. Kibbe (Pharmaceutical Press, London, UK, 2000), which is incorporated by reference in its entirety. Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin
  • the foregoing component(s) may be present in the pharmaceutical composition at any concentration, such as, for example, at least A, wherein A is 0.0001% w/v, 0.001% w/v, 0.01% w/v, 0.1% w/v, 1% w/v, 2% w/v, 5% w/v, 10% w/v, 20% w/v, 30% w/v, 40% w/v, 50% w/v, 60% w/v, 70% w/v, 80% w/v, or 90% w/v. In some embodiments, the foregoing component(s) may be present in the pharmaceutical composition at any concentration, such as, for example, at least A, wherein A is 0.0001% w/v, 0.001% w/v, 0.01% w/v, 0.1% w/v, 1% w/v, 2% w/v, 5% w/v, 10% w/v, 20% w/v, 30% w/v, 40% w/v
  • concentration such as, for example, at most B, wherein B is 90% w/v, 80% w/v, 70% w/v, 60% w/v, 50% w/v, 40% w/v, 30% w/v, 20% w/v, 10% w/v, 5% w/v, 2% w/v, 1% w/v, 0.1% w/v, 0.001% w/v, or 0.0001%.
  • the foregoing component(s) may be present in the pharmaceutical composition at any concentration range, such as, for example from about A to about B. In some embodiments, A is 0.0001% and B is 90%.
  • the pharmaceutical compositions may be formulated to achieve a physiologically compatible pH.
  • the pH of the pharmaceutical composition may be at least 5, at least 5.5, at least 6, at least 6.5, at least 7, at least 7.5, at least 8, at least 8.5, at least 9, at least 9.5, at least 10, or at least 10.5 up to and including pH 11, depending on the formulation and route of administration.
  • the pharmaceutical compositions may comprise buffering agents to achieve a physiological compatible pH.
  • the buffering agents may include any compounds capabale of buffering at the desired pH such as, for example, phosphate buffers (e.g.,PBS), triethanolamine, Tris, bicine, TAPS, tricine, HEPES, TES, MOPS, PIPES, cacodylate, MES, and others.
  • the strength of the buffer is at least 0.5 mM, at least 1 mM, at least 5 mM, at least 10 mM, at least 20 mM, at least 30 mM, at least 40 mM, at least 50 mM, at least 60 mM, at least 70 mM, at least 80 mM, at least 90 mM, at least 100 mM, at least 120 mM, at least 150 mM, or at least 200 mM.
  • the strength of the buffer is no more than 300 mM (e.g., at most 200 mM, at most 100 mM, at most 90 mM, at most 80 mM, at most 70 mM, at most 60 mM, at most 50 mM, at most 40 mM, at most 30 mM, at most 20 mM, at most 10 mM, at most 5 mM, at most 1 mM).
  • the active agent pharmaceutical composition
  • Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the active agent of the present disclosure dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions.
  • Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant.
  • diluents such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant.
  • Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and corn starch.
  • Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and other pharmacologically compatible excipients.
  • Lozenge forms can comprise the active agent of the present disclosure in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active agent of the present disclosure in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to, such excipients as are known in the art.
  • an inert base such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to, such excipients as are known in the art.
  • the active agents of the present disclosure can be delivered via pulmonary administration and can be made into aerosol formulations to be administered via inhalation.
  • aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. Such spray formulations also may be used to spray mucosa.
  • the active agent is formulated into a powder blend or into microparticles or nanoparticles. Suitable pulmonary formulations are known in the art.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.
  • parenteral means not through the alimentary canal but by some other route such as subcutaneous, intramuscular, intraspinal, or intravenous.
  • the active agent of the present disclosure can be administered with a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol or hexadecyl alcohol, a glycol, such as propylene glycol or polyethylene glycol, dimethylsulfoxide, glycerol, ketals such as 2,2- dimethyl-153-dioxolane-4-methanol, ethers, poly(ethyleneglycol) 400, oils, fatty acids, fatty acid esters or glycerides, or acetylated fatty acid glycerides with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or
  • a pharmaceutically acceptable surfactant such as a soap or a detergent, suspend
  • carboxymethylcellulose or emulsifying agents and other pharmaceutical adjuvants.
  • Oils which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters.
  • Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts
  • suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylenepolypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-P-aminopropionates, and 2-alkyl -imidazoline quaternary ammonium salts, and (e) mixtures thereof.
  • the parenteral formulations in some embodiments contain from about 0.5% to about 25% by weight of the active agent of the present disclosure in solution. Preservatives and buffers may be used. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations will typically range from about 5% to about 15% by weight. Suitable surfactants include polyethylene glycol sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the
  • parenteral formulations in some aspects are presented in unit-dose or multi-dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions in some aspects are prepared from sterile powders, granules, and tablets of the kind previously described.
  • injectable formulations are in accordance with the invention.
  • the requirements for effective pharmaceutical carriers for injectable compositions are well-known to those of ordinary skill in the art (see, e.g., Pharmaceutics and Pharmacy Practice, J. B. Lippincott Company, Philadelphia, PA, Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986)).
  • the active agent of the invention can be made into suppositories for rectal administration by mixing with a variety of bases, such as emulsifying bases or water- soluble bases.
  • bases such as emulsifying bases or water- soluble bases.
  • Formulations suitable for vaginal administration can be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate.
  • the active agent of the disclosure can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes.
  • the active agents of the disclosure are believed to be useful in methods of inhibiting a binding interaction between ⁇ integrin and G protein a subunit, as well as other methods, as further described herein, including methods of treating or preventing stroke, heart attack, cancer, or inflammation.
  • the amount or dose of the active agent administered should be sufficient to effect, e.g., a therapeutic or prophylactic response, in the subject or animal over a reasonable time frame.
  • the dose of the active agent of the present disclosure should be sufficient to treat cancer as described herein in a period of from about 1 to 4 minutes, 1 to 4 hours or 1 to 4 weeks or longer, e.g., 5 to 20 or more weeks, from the time of administration. In certain embodiments, the time period could be even longer.
  • the dose will be determined by the efficacy of the particular active agent and the condition of the animal (e.g., human), as well as the body weight of the animal (e.g., human) to be treated.
  • an assay which comprises comparing the extent to which cancer is treated upon administration of a given dose of the active agent of the present disclosure to a mammal among a set of mammals, each set of which is given a different dose of the active agent, could be used to determine a starting dose to be administered to a mammal.
  • the extent to which cancer is treated upon administration of a certain dose can be represented by, for example, the cytotoxicity of the active agent or the extent of tumor regression achieved with the active agent in a mouse xenograft model.
  • Methods of measuring cytotoxicity of compounds and methods of assaying tumor regression are known in the art, including, for instance, the methods described in the EXAMPLES set forth below.
  • the dose of the active agent of the present disclosure also will be determined by the existence, nature and extent of any adverse side effects that might accompany the administration of a particular active agent of the present disclosure.
  • the attending physician will decide the dosage of the active agent of the present disclosure with which to treat each individual patient, taking into consideration a variety of factors, such as age, body weight, general health, diet, sex, active agent of the present disclosure to be administered, route of administration, and the severity of the condition being treated.
  • the dose of the active agent of the present disclosure can be about 0.0001 to about 1 g/kg body weight of the subject being treated/day, from about 0.0001 to about 0.001 g/kg body weight/day, or about 0.01 mg to about 1 g/kg body weight/day.
  • the active agents described herein can be modified into a depot form, such that the manner in which the active agent of the invention is released into the body to which it is administered is controlled with respect to time and location within the body (see, for example, U.S. Patent No. 4,450,150).
  • Depot forms of active agents of the invention can be, for example, an implantable composition comprising the active agents and a porous or non-porous material, such as a polymer, wherein the active agent is encapsulated by or diffused throughout the material and/or degradation of the non-porous material.
  • the depot is then implanted into the desired location within the body of the subject and the active agent is released from the implant at a predetermined rate.
  • the pharmaceutical composition comprising the active agent in certain aspects is modified to have any type of in vivo release profile.
  • the pharmaceutical composition is an immediate release, controlled release, sustained release, extended release, delayed release, or bi-phasic release formulation.
  • Methods of formulating peptides for controlled release are known in the art. See, for example, Qian et al., J Pharm 374: 46-52 (2009) and International Patent Application Publication Nos. WO 2008/130158, WO2004/033036;
  • compositions may further comprise, for example, micelles or liposomes, or some other encapsulated form, or may be administered in an extended release form to provide a prolonged storage and/or delivery effect.
  • the invention also provides a micelle comprising a peptide or peptide analog of the invention and at least one lipid, optionally, wherein the lipid is covalently attached to a water soluble polymer.
  • the peptide or peptide analog of the micelle is covalently attached to a fatty acid or other lipid moiety.
  • the peptide of the micelle consists of FEEERA (SEQ ID NO: 87), wherein the Phe at position 1 is covalently attached to a C16 fatty acid.
  • the micelle comprises a lipid covalently attached to a water soluble polymer and a lipid free of a water soluble polymer.
  • lipids for use in micelle synthesis is known in the art. See, e.g., Banerjee and Onyuksel, Peptide Delivery Using Phospholipid Micelles, WIREs Nanomed Nanobiotechnol 4:562-574 (2012).
  • the lipid that is covalently attached to a water soluble polymer is l,2-distearoyl-s7i- glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] and the lipid that is free of a water soluble polymer is phophatidylcholine.
  • the invention further comprises a composition comprising any of the micelles described herein and an aqueous solution.
  • compositions and formulations may be administered according to any regimen including, for example, daily (1 time per day, 2 times per day, 3 times per day, 4 times per day, 5 times per day, 6 times per day), every two days, every three days, every four days, every five days, every six days, weekly, bi-weekly, every three weeks, monthly, or bi-monthly.
  • Timing like dosing can be fine-tuned based on dose-response studies, efficacy, and toxicity data, and initially guaged based on timing used for other antibody therapeutics.
  • the active agents described herein are administered alone, and in alternative embodiments, the active agents described herein are administered in combination with another therapeutic agent, e.g., another active agent of the invention of different type (e.g., structure), or another therapeutic which does not inhibit a binding interaction between ⁇ integrin and G protein a subunit.
  • another therapeutic agent e.g., another active agent of the invention of different type (e.g., structure), or another therapeutic which does not inhibit a binding interaction between ⁇ integrin and G protein a subunit.
  • the other therapeutic aims to treat or prevent cancer.
  • the other therapeutic is one listed under the section entitled "Heterologous Moieties: Therapeutic Agents.”
  • the other therapeutic is a
  • the other therapeutic is an agent used in radiation therapy for the treatment of cancer.
  • the active agents described herein are administered or packaged in combination with an anti-thrombotic agent.
  • the anti-thrombotic agent is an anticoagulant, e.g., fondaparinux and bivalirudin.
  • the anti-thrombotic agent is an anti-platelet agent, e.g., aspirin, clopidogrel, dipyridamole, and abciximab.
  • the active agent described herein is administered or packaged in combination with an anti-platelet drug.
  • the antiplatelet drug is an irreversible cyclooxygenase inhibitor (e.g., aspirin), an adenosine diphosphate (ADP) receptor inhibitor (e.g., clopidogrel, prasugrel, ticagrelor, ticlopidine), a phosphodiesterase inhibitor (e.g., cilostazol), a glycoprotein Ilb/IIIa inhibitor (e.g., abciximab, eptifibatide), tirofiban), an adenosine reuptake inhibitor (e.g., dipyridamole), or a thromboxane inhibitor (e.g., a thromboxane synthase inhibitor, a thromboxane receptor antagonist (e.g., terutroban).
  • the anti-platelet drug is an irreversible cyclooxygenase inhibitor (
  • the active agent described herein is administered or packaged in combination with an integrin antagonist or integrin inhibitor.
  • the integrin inhibitor in exemplary aspects is eptifibatide
  • the active agent is administered simultaneously as the other therapeutic. In alternative embodiments, the active agent is administered either before or after the other therapeutic.
  • the active agents of the invention are useful for a number of applications in a variety of settings. For example and most simplistically, the active agents of the invention are useful for inhibiting a binding interaction between a ⁇ integrin and a G protein a subunit in a cell.
  • the invention provide a method of inhibiting a binding interaction between a ⁇ integrin and a G protein a subunit in a cell. The method comprises contacting the cell with a compound or composition of the invention, in an amount effective to inhibit the binding interaction.
  • the cell is part of an in vitro or ex vivo cell culture or in vitro or ex vivo tissue sample. In some aspects, the cell is an in vivo cell. In certain embodiments, the method is intended for research purposes, and, in other embodiments, the method is intended for therapeutic purposes.
  • the invention also provides a method of inhibiting integrin-dependent Src activation in a cell.
  • the method comprises the step of contacting the cells with a compound or composition of the invention in an amount effective to inhibiting the Src activation.
  • Methods of measuring integrin-dependent Src activation are known in the art, and include, for example, those set forth herein in EXAMPLES and use of the ProFluor ® Src-Family Kinase Assay (Promega, Madison, WI) or one of the Src activity assay kits available from Millipore (Billerica, MA).
  • a method of activating a small GTPase is furthermore provided by the invention.
  • the method comprises the step of contacting a G protein subunit with a compound or composition in an amount effective to activate the small GTPase.
  • the small GTPase of the method of the invention is RhoA.
  • Methods of measuring GTPase activity are known in the art, and include, for example, those set forth herein in EXAMPLES.
  • GTPase activity levels may be measured using commercially available kits (Thermo Fisher Scientific, Inc. (Rockford, IL), Innova Biosciences (Cambridge, UK), Cell Biolabs, Inc. (San Diego, CA).
  • the invention moreover provides a method of inhibiting spreading or migration of a cell.
  • the method comprises the step of contacting the cell with a compound or composition of the invention in an amount effective to inhibit spreading and migration.
  • the cell may be any cell that undergoes integrin-dependent adhesion, spreading, retraction, or migration, or any cell that undergoes anchorage-dependent survival and proliferation.
  • the cell is a platelet, leukocyte, endothelial cell, fibroblast, epithelial cell. Methods of measuring cell spreading or cell migration are known in the art. See, for example, the methods described herein in EXAMPLES.
  • Also provided by the invention is a method of inhibiting platelet adhesion.
  • the method comprises the step of contacting a platelet with a compound or composition of the invention in an amount effective to inhibit platelet adhesion.
  • the invention further provides a method of inhibiting platelet granule secretion and platelet aggregation.
  • the method comprises the step of contacting a platelet with a compound or composition of the invention in an amount effective to inhibit granule secretion and aggregation. Methods of measuring platelet granule secretion or platelet aggregation are known in the art.
  • platelet aggregation may be measured in platelet rich-plasma (PRP) using a turbidomatric platelet aggregometer to analyze washed platelets. Aggregation may be indicated by an increase in light transmission through PRP or platelet suspension. Platelet aggregation also may be measured in whole blood using a whole blood platelet aggregometer, in which platelet aggregation is indicated by an increase in electric resistance of electrodes. See, for example, the methods described herein in
  • the compounds and compositions of the invention are additionally contemplated for therapeutic purposes.
  • the compounds and compositions of the invention may be used to enhance blood clot retraction or inhibit thrombosis in a subject in need thereof.
  • the invention provides a method of enhancing blood clot retraction in a subject in need thereof.
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to enhance blood clot retraction.
  • a method of inhibiting thrombosis in a subject in need thereof comprises the step of administering to the subject a compound or composition of the invention in an amount effective to inhibit thrombosis.
  • the invention furthermore provides a method of treating or preventing a stroke or a heart attack in a subject in need thereof.
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to treat or prevent stroke or heart attack.
  • the compounds and compositions provided herein relate to the coordinated cell spreading-retraction process, which in turn, is important in cell migration
  • the invention also provides a method of inhibiting metastasis of a tumor cell.
  • the method comprises the step of contacting a tumor cell with a compound or composition of the invention in an amount effective to inhibit metastasis.
  • the compounds and compositions are also contemplated for use in inhibiting angiogenesis. Accordingly, the invention provides a method of inhibiting
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to inhibit angiogenesis.
  • the invention provides a method of facilitating wound healing in a subject in need of thereof.
  • the method comprises the step of administrating to the subject of a compound or composition of of the invention in an amount effective to facilitate the wound healing.
  • the compound or composition is administered to the subject topically near or at the wound site.
  • the rate of wound healing is increased by at least 10%, 25%, 50%, 75%, 90% or more, as compared to the time at which the wound would heal without administration of the compound or composition of the invention.
  • the invention moreover provides a method of treating or preventing cancer in a subject in need thereof.
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to treat or prevent cancer.
  • the subject has a solid tumor and the compound or composition of the invention is administered near or at the tumor site.
  • the compound or composition of the invention is administered via injection at the tumor site.
  • the compounds and compositions provided herein also may be used for affecting leukocyte function.
  • the invention accordingly provides a method of inhibiting leukocyte adhesion, spreading, migration, or chemotaxis.
  • the method comprises the step of contacting a leukocyte with a compound or composition of the invention in an amount effective to inhibit leukocyte adhesion, spreading, migration, or chemotaxis. Since these leukocyte functions are related to inflammation, the invention additionally provides a method of inhibiting or treating inflammation in a subject in need thereof.
  • the method comprises the step of administering to the subject a compound or composition of the invention in an amount effective to inhibit or treat inflammation.
  • the compound or composition is administered to the subject systemically, e.g., parenterally (e.g., via intravenous injection).
  • the term "treat,” as well as words related thereto, do not necessarily imply 100% or complete treatment. Rather, there are varying degrees of treatment of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect.
  • the methods of treating a stroke or a heart attack or cancer or inflammation of the invention can provide any amount or any level of treatment.
  • the treatment provided by the method of the invention may include treatment of one or more conditions or symptoms or signs of the stroke, heart attack, cancer or inflammation, being treated.
  • the treatment provided by the methods of the invention may encompass slowing the progression of the stroke, heart attack, cancer or inflammation.
  • the methods can treat cancer by virtue of reducing tumor or cancer growth, reducing metastasis of tumor cells, increasing cell death of tumor or cancer cells, and the like.
  • the term "prevent” and words stemming therefrom encompasses delaying the onset of the medical condition being prevented.
  • the method delays the onset of the medical condition by 1 day, 2 days, 4 days, 6 days, 8 days, 10 days, 15 days, 30 days, two months, 4 months, 6 months, 1 year, 2 years, 4 years, or more.
  • the term “prevent” and words stemming therefrom encompasses reducing the risk of the medical condition being prevented.
  • the method reduces the risk of SCD 2- fold, 5-fold, 10-fold, 20-fold, 50-fold, 100-fold, or more.
  • the term "inhibit” and words stemming therefrom may not be a 100% or complete inhibition or abrogation. Rather, there are varying degrees of inhibition of which one of ordinary skill in the art recognizes as having a potential benefit or therapeutic effect.
  • the compounds of the invention may inhibit the binding interaction between a ⁇ integrin and a G protein a subunit to any amount or level.
  • the inhibition provided by the methods of the invention is at least or about a 10% inhibition (e.g., at least or about a 20% inhibition, at least or about a 30% inhibition, at least or about a 40% inhibition, at least or about a 50% inhibition, at least or about a 60% inhibition, at least or about a 70% inhibition, at least or about a 80% inhibition, at least or about a 90% inhibition, at least or about a 95% inhibition, at least or about a 98% inhibition).
  • a 10% inhibition e.g., at least or about a 20% inhibition, at least or about a 30% inhibition, at least or about a 40% inhibition, at least or about a 50% inhibition, at least or about a 60% inhibition, at least or about a 70% inhibition, at least or about a 80% inhibition, at least or about a 90% inhibition, at least or about a 95% inhibition, at least or about a 98% inhibition.
  • the cancer treatable by the methods disclosed herein may be any cancer, e.g., any malignant growth or tumor caused by abnormal and uncontrolled cell division that may spread to other parts of the body through the lymphatic system or the blood stream.
  • any cancer e.g., any malignant growth or tumor caused by abnormal and uncontrolled cell division that may spread to other parts of the body through the lymphatic system or the blood stream.
  • the cancer is a cancer in which an integrin and a G protein a subunit are expressed on the surface of the cells.
  • the cancer in some aspects is one selected from the group consisting of acute lymphocytic cancer, acute myeloid leukemia, alveolar rhabdomyosarcoma, bone cancer, brain cancer, breast cancer, cancer of the anus, anal canal, or anorectum, cancer of the eye, cancer of the intrahepatic bile duct, cancer of the joints, cancer of the neck, gallbladder, or pleura, cancer of the nose, nasal cavity, or middle ear, cancer of the oral cavity, cancer of the vulva, chronic lymphocytic leukemia, chronic myeloid cancer, colon cancer, esophageal cancer, cervical cancer, gastrointestinal carcinoid tumor, Hodgkin lymphoma, hypopharynx cancer, kidney cancer, larynx cancer, liver cancer, lung cancer, malignant mesothelioma, melanoma, multiple myeloma, nasopharynx cancer, non-Hodgkin lymphoma, ova
  • the cancer is selected from the group consisting of: head and neck, ovarian, cervical, bladder and oesophageal cancers, pancreatic, gastrointestinal cancer, gastric, breast, endometrial and colorectal cancers, hepatocellular carcinoma, glioblastoma, bladder, lung cancer, e.g., non-small cell lung cancer (NSCLC), bronchioloalveolar carcinoma.
  • NSCLC non-small cell lung cancer
  • the subject is a mammal, including, but not limited to, mammals of the order Rodentia, such as mice and hamsters, and mammals of the order Logomorpha, such as rabbits, mammals from the order Carnivora, including Felines (cats) and Canines (dogs), mammals from the order Artiodactyla, including Bovines (cows) and Swines (pigs) or of the order Perssodactyla, including Equines (horses).
  • the mammals are of the order Primates, Ceboids, or Simoids (monkeys) or of the order Anthropoids (humans and apes).
  • the mammal is a human.
  • the human is an adult aged 18 years or older.
  • the human is a child aged 17 years or less.
  • the subject has a medical history of taking an integrin antagonist or an anti-platelet drug, or the subject has been prescribed an integrin antagonist or an anti-platelet drug.
  • the subject has a medical history of coronary heart disease, heart attack, angina, stroke, transient ischemic attack, peripheral artery disease, myocardial infarction, atrial fibrillation, and/or ischaemic stroke.
  • the subject has a medical history comprising angioplasty, stent placement, and/or heart bypass or valve replacement surgery.
  • the subject suffers from acute coronary syndrome, unstable angin, or non-ST-segment elevation myocardial infarction.
  • the composition comprising a compound of the invention, a pharmaceutically acceptable salt thereof, a conjugate comprising the compound, or a multimer or dimer comprising the compound, is provided as a kit or package or unit dose.
  • "Unit dose” is a discrete amount of a therapeutic composition dispersed in a suitable carrier. Accordingly, provided herein are kits comprising a compound of the invention, a pharmaceutically acceptable salt thereof, a conjugate comprising the compound, or a multimer or dimer comprising the compound.
  • the components of the kit/unit dose are packaged with instructions for administration to a patient.
  • the kit comprises one or more devices for administration to a patient, e.g., a needle and syringe, a dropper, a measuring spoon or cup or like device, an inhaler, and the like.
  • the compound of the invention, a pharmaceutically acceptable salt thereof, a conjugate comprising the compound, or a multimer or dimer comprising the compound is pre-packaged in a ready to use form, e.g., a syringe, an intravenous bag, an inhaler, a tablet, capsule, etc.
  • the kit further comprises other therapeutic or diagnostic agents or pharmaceutically acceptable carriers (e.g., solvents, buffers, diluents, etc.), including any of those described herein.
  • the kit comprises a compound of the invention, a pharmaceutically acceptable salt thereof, a conjugate comprising the compound, or a multimer or dimer comprising the compound, along with an agent, e.g., a therapeutic agent, used in chemotherapy or radiation therapy.
  • Myristoylated Ga 13 SRI peptide, Myr-LLARRPTKGIHEY (mSRI; SEQ ID NO: 46), and myristoylated- scrambled peptide (Myr-LIRYALHRPTKEG; SEQ ID NO: 47) were synthesized and purified at the Research Resource Center at University of Illinois, Chicago (SI). Expression and purification of recombinant Ga 13 was described previously (S2).
  • Anti-RhoA antibody and cell permeable C3 transferase (C3 toxin) were purchased from Cytoskeleton, Inc.; anti- Ga 13 (SC410), anti-c-Src (sc-18) and anti-mouse integrin ⁇ 3 (sc-6627) antibodies were from Santa Cruz Biotechnology, Inc; anti-phospho-Src Y antibody was obtained from Cell
  • anti-human integrin ⁇ 3 antibody, MAb 15 and anti-au b ⁇ 3 antibody, D57 were kindly provided by Dr. Mark Ginsberg (University of California, San Diego, La Jolla, CA); anti-GPIb monoclonal antibody LJP3 was kindly provided by Dr.
  • Zaverio Ruggeri the Scripps Research Institute, La, Jolla, CA
  • anti-tubulin and anti-flag antibodies were purchased from Sigma- Aldrich
  • lipofectamine 2000, viraPower lentivirus expression system, Alexa Fluor 546- conjugated phalloidin, Alexa Fluor 633-conjugated phalloidin, and Alexa Fluor 546-conjugated anti-mouse IgG antibody were from Invitrogen
  • Y27632 was purchased from Calbiochem.
  • Mouse platelets were isolated and washed using methods previously described (S4, S5). For analyzing platelet spreading on integrin ligand fibrinogen, washed platelets were allowed to spread on 100 ⁇ g/ml fibrinogen-coated coverslips at 37°C for 90 minutes, stained and viewed with a Leica RMI RB microscope or Zeiss LSM510 META confocal microscope as previously described (S5). Clot retraction was analyzed with the previously described method (S6, S7). Briefly, mouse platelets (6 x 10 /ml) were resuspended in platelet-depleted human plasma, and 0.4 U/ml a-thrombin was added to initiate coagulation. The clots were photographed at various time points. Sizes of retracted clots on photographs were quantified using NIH Image J software. Statistical significance was determined using t-test.
  • Human platelets or CHO cells expressing recombinant integrin ⁇ 3 ⁇ 4 ⁇ 3 were solubilized in modified RIPA Buffer (50 mM Tris, pH 7.4, 10 mM MgCl 2 , 150 mM NaCl, 1% NP-40, 1 mM sodium orthovanadate, 1 mM NaF), or RIPA buffer (25 mM Tris, pH 7.6, 150 mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS) and complete protease inhibitor cocktail tablets (1 tablet/5 ml buffer, Roche).
  • modified RIPA Buffer 50 mM Tris, pH 7.4, 10 mM MgCl 2 , 150 mM NaCl, 1% NP-40, 1 mM sodium orthovanadate, 1 mM NaF
  • RIPA buffer 25 mM Tris, pH 7.6, 150 mM NaCl, 1% NP-40, 1% sodium deoxycholate, 0.1% SDS
  • cell lysates were incubated with 2 ⁇ g/ml of D57 (antibody to integrin ⁇ 3 ⁇ 4 ⁇ 3 ), LJP3(antibody against GPIb), or mouse IgG, and further incubated with protein G-conjugated Sepharose beads.
  • Cell lysates were also incubated with rabbit anti-Ga 13 IgG antibody (1.5 ⁇ g/ml) or an equal amount of rabbit IgG and further incubated with protein A-conjugated Sepharose beads.
  • Human Ga 13 cDNA (S9) was tagged with Flag-epitope at N-tenninus using PCR with the Flag cDNA sequence incorporated into the forward primer, and then subcloned into pCDEF3 vector using Kpn I and Not I restriction sites.
  • the forward primer sequence is 5'- GCGGGTACCGCCATGGACTACAAGGACGACGATGACAAGGCGGACTTCC- TGCCGTCGCGGTCCGT-3' (SEQ ID NO: 52).
  • the reverse primer sequence is 5'- GGCCGGCGGCCGCTC ACTGTAGC ATA AGCTGCTTG AGGTT- 3 ' (SEQ ID NO: 53).
  • Truncation mutants of Ga 13 were generated using PCR with reverse primer sequences 5'- GGCCGGCGGCCGCTCAAATATCTTGTTGTGATGGAAT- ATA ATCTGGTTCTCC A AGTTTATCC A AG- 3 ' (SEQ ID NO: 54). for mutant 1-196; and 5'- GGCCGGCGGCCGCTCATTCAAAGTCGTATTCATGGATGCC-3' (SEQ ID NO: 55). for mutant 1-212.
  • cDNA encoding Flag-tagged wild type Ga 13 or Ga 13 mutants were transfected into 293FT cells using lipofectamine 2000. Cell lysates were prepared 48 hours after transfection. Flag-tagged wild type or mutant Ga 13 in 293FT cell lysates were incubated with glutathione bead-bound GST or GST-p CD at 4°C overnight. After centrifugation and washing, bead-bound proteins were immunoblotted with anti-Flag antibody.
  • Platelets in modified Tyrode's buffer or adherent on immobilized fibrinogen were lysed quickly in 0.8 ml lysis buffer (50 mM Tris, pH 7.4, 10 mM MgCl 2 , 500 mM NaCl, 1% Triton X-100, 0.1% SDS, 0.5% deoxycholate, 10 ⁇ g/ml each of aprotinin and leupeptin, 1 mM phenylmethylsulfonyl fluoride, and 200 ⁇ sodium vanadate).
  • lysis buffer 50 mM Tris, pH 7.4, 10 mM MgCl 2 , 500 mM NaCl, 1% Triton X-100, 0.1% SDS, 0.5% deoxycholate, 10 ⁇ g/ml each of aprotinin and leupeptin, 1 mM phenylmethylsulfonyl fluoride, and 200 ⁇ sodium vanadate).
  • Lysates were cleared by centrifugation at 18,000 g for 2 minutes at 4°C, and the supernatant was incubated for 1 hour with 30 ⁇ g purified GST-Rhotekin RhoA-binding domain fusion protein (GST-RBD) bound to glutathione-Sepharose beads (S10). Samples were washed three times using washing buffer (50 mM Tris, pH 7.4, 10 mM MgCl 2 , 150 mM NaCl, 1% Triton X-100) and then immunoblotted with an anti-RhoA monoclonal antibody. Cell lysates were also immunoblotted with anti-RhoA as loading control.
  • washing buffer 50 mM Tris, pH 7.4, 10 mM MgCl 2 , 150 mM NaCl, 1% Triton X-100
  • siRNA #1 5'- GTCCACCTTCCTGAAGCAG (SEQ ID NO: 56).
  • siRNAi #2 5'- GGAGATCGACAAATGCCTG (SEQ ID NO: 57).
  • Scrambled siRNA sequence is: 5'- GAGGAGCCGACGCTTAATA-3 ' (SEQ ID NO: 58). These sequences are conserved in hamsters and mice.
  • Lentivirus was prepared by co-transfection of pLL3.7-scrambled siRNA or pLL3.7-Ga 13 siRNA (#1 and #2) with pLPl, pLP2 and pLP/VSVG plasmids (Invitrogen) into -90% confluent 293FT cells using Lipofectamine 2000. 48 hours after transfection, cell culture medium containing virus was filtered, titered and stored at -80°C. Bone manow cells from 6-8 week old healthy C57/BL mice were isolated aseptically from femurs and tibias.
  • Stem cells were negatively selected by MACS Lineage cell depletion kit (Miltenyi Biotec) and cultured in RPMI 1640 complete medium with 10 ng/ml interleukin-3, 10 ng/ml interleukin-6, 10 ng/ml granulocyte-macrophage colony stimulating factor (GM-CSF), and 100 ng/ml stem cell factor (SCF). 50 multiplicity of infection (MOI) lenti- virus was used to infect mice bone marrow stem cells twice with 6 ⁇ g/ml polybrene.
  • MOI multiplicity of infection
  • mutants changed the Ga 13 siRNA #1 target sequence to 5'- GTCC ACCTTttTaA AGC AG- 3 ' (SEQ ID NO: 59). or siRNA #2 target sequence to 5'- GGAGATCGAtAAgTGCCTG-3' (SEQ ID NO: 60). without changing the amino acid sequence of Ga 13 .
  • the mutants were subcloned into pLenti6/V5-Dest vector using EcoR I and Sal I restriction sites in PCR fragments and EcoRI and Xho I restriction sites in the vector.
  • the primer sequences are as follows: (1) Flag tagged forward primer: 5'-CGGAATTCG- CCATGGACTACAAGGACGACGATGACAAGGCGGACTTCCTGCCGTCGCGGTCCGT-3' (SEQ ID NO: 61); (2) reverse primer: 5'-
  • CHO cells expressing human ⁇ 3 ⁇ 4 ⁇ 3 cells were transfected with Ga 13 siRNA construct with or without cotransfection of Flag-tagged siRNA resistant-Ga 13 plasmid using lipofectamine 2000. After 30 hours, the cells were detached by 0.53 mM EDTA in phosphate-buffered saline, and allowed to spread on 100 ⁇ g/ml fibrinogen.
  • c-Src c-Src
  • Integrins mediate cell adhesion and transmit signals within the cell that lead to cell spreading, retraction, migration, and proliferation (1).
  • integrins have pivotal roles in biological processes such as development, immunity, cancer, wound healing, hemostasis and thrombosis.
  • the platelet integrin, ⁇ 3 ⁇ 4 3 ⁇ 4 ⁇ 3 typically displays bidirectional signaling function (2, 3). Signals from within the cell activate binding of ⁇ 3 ⁇ 4] 3 ⁇ 4 ⁇ 3 to extracellular ligands, which in turn triggers signaling within the cell initiated by the occupied receptor (so-called "outside-in” signaling).
  • G proteins Heterotrimeric guanine nucleotide-binding proteins (G proteins) consist of Gcc, ⁇ and Gy subunits (9). G proteins bind to the intracellular side of G-protein coupled receptors (GPCR) and transmit signals that are important in many intracellular events (9-11). Gcc 13 , when activated by GPCRs, interacts with Rho guanine-nucleotide exchange factors (RhoGEF) and thus activates RhoA (11-14), facilitating contractility and rounding of discoid platelets (shape change).
  • RhoGEF Rho guanine-nucleotide exchange factors
  • siRNA small interfering RNA
  • Gcc 13 siRNA inhibited spreading of Chinese hamster ovary (CHO) cells expressing human ⁇ 3 ⁇ 4 3 ⁇ 4 ⁇ 3 (123 cells) (18), which was rescued by an siRNA -resistant Gcc 13 ( Figure 7).
  • Gcc 13 appears to be important in integrin "outside-in" signaling leading to cell spreading.
  • RhoA activity was suppressed to baseline 15 minutes after platelet adhesion, and became activated at 30 minutes (Figure 1C), which is consistent with transient inhibition of RhoA by c-Src (7).
  • the integrin-dependent delayed activation of RhoA was not inhibited by depletion of Gcc 13 , indicating its independence of the GPCR-Gcc 13 -RhoGEF pathway ( Figure 1C).
  • depletion of Gcc 13 accelerated RhoA activation ( Figure 1C).
  • Gcc 13 appears to mediate inhibition of RhoA.
  • ⁇ 3 is present in a complex with Gcc 13 , preferably the active GTP- bound Gcc 13 .
  • Gcc 13 directly binds to the integrin cytoplasmic domain.
  • GST glutathione S- transferase
  • GST ⁇ 3 CD GST ⁇ 3 cytoplasmic domain fusion protein
  • ⁇ 13 - ⁇ 3 binding was assessed in the presence of a myristoylated synthetic peptide, Myr- LLARRPTKGIHEY (mSRI; SEQ ID NO: 45), corresponding to the SRI sequence of Gcc 13 (197- 209) (21).
  • Gcc 13 in mediating the integrin-dependent inhibition of RhoA contrasts with the traditional role of Gcc 13 , which is to mediate GPCR-induced activation of RhoA.
  • GPCR-mediated activation of RhoA is transient, peaking at 1 minute after exposure of platelets to thrombin, indicating the presence of a negative regulatory signal ( Figure 4, D and F).
  • thrombin- stimulated activation of RhoA occurs during platelet shape change before substantial ligand binding to integrins ( Figure 4, C, D and F).
  • Integrin ⁇ 3 _/ ⁇ mice were obtained from the Jackson Laboratory. Myristoylated peptides were synthesized and purified at the Research Resource Center at University of Illinois. These peptides include: mPi 3 (Myr-KFEEERARAKWDT; SEQ ID NO: 67), mP 7 (Myr-KFEEERA) and mP 5 (Myr-EEERA; SEQ I DNO: 68) and myristoylated scrambled peptide for mP 13 (Myr- EE ARERKD W AKFT ; SEQ ID NO: 69); myristoylated scrambled peptide for mP 7 (Myr- EAREKFE; SEQ ID NO: 70) and myristoylated scrambled peptide for mP 5 (Myr-EEARE; SEQ ID NO: 71). Human integrin ⁇ cDNA was cloned into pCDNA3.1 vector following digestion with Hind III and Xho
  • Truncation mutants and integrin E to A mutants were either previously reported 23 or generated using PCR and cloned into pCDNA3.1 vector by Bam HI and Xho I.
  • the primer sequences used are: (1): ITGB -UP: 5'-
  • GCG A AGCTTGCCGCC ATGG ACCG AGCGCGGCCGCGCGGCCCCGGCCGCTCT- 3 ' (SEQ ID NO: 72); (2): ITGB 3 -728DN: 5'-
  • Anti-RhoA antibody was purchased from Cytoskeleton, Inc.; anti-Gcc 13 (sc410), anti-total c-Src (scl8), anti-talin (sc7534), and anti- integrin ⁇ (sc6627) antibodies were from Santa Cruz Biotechnology, Inc; anti-Gcc 13 (26004) was from NewEast; anti-phospho-Src Y 416 antibody was obtained from Cell Signaling; anti-talin (TA205) was from Millipore; anti-human integrin ⁇ 3 antibody, MAb 15 and 8053 rabbit serum were kindly provided by Dr.
  • mice were anesthetized by isoflurane (Pharmaceutical, Inc) and platelets were prepared from freshly drawn blood from the inferior vena cava and washed using the previously described method 25.
  • washed platelets were allowed to spread on 100 ⁇ g/ml fibrinogen-coated coverslips at 37°C for different time points, fixed, permeabilized, stained and viewed with a Leica RMI RB microscope or Zeiss LSM510 META confocal microscope as previously described .
  • Fibrinogen binding assay Washed human or mouse platelets resuspended in modified Tyrode's buffer (3 x
  • Platelets or CHO-lb9 cells expressing recombinant integrin ⁇ 3 ⁇ 4 3 ⁇ 4 ⁇ 3 14 ' 23 were solubilized in modified RIPA Buffer (50 mM Tris, pH 7.4, 150 mM NaCl, 1% NP-40, 1 mM sodium orthovanadate, 1 mM NaF) with complete protease inhibitor cocktail tablets.
  • Cell lysates were incubated with rabbit anti-Gcc 13 IgG (1.5 ⁇ g/ml), anti-integrin ⁇ 3 rabbit serum (5 ⁇ /ml) or an equal amount of rabbit IgG or pre-immune serum at 4°C overnight, and subsequently with protein A-conjugated Sepharose beads for 1 hour.
  • integrin ⁇ 3 clustering experiment human platelets (3 x 10 ) were stimulated with 0.025 U/ml a-thrombin for 2 minutes at room temperature (to avoid platelet aggregation) with or without 2mM RGDS, then incubated with 5 ⁇ anti-integrin ⁇ 3 8053 rabbit serum at room temperature for 1 hour, and 5 ⁇ g goat anti-rabbit secondary antibody for another 1 hour. Platelets were then solubilized in modified RIPA Buffer. For the un-clustering control and pre-immune serum control, cells were solubilized in modified RIPA Buffer first, than incubated with 5 ⁇ 8053 serum or pre-immune serum.
  • Platelets or CHO cells in modified Tyrode's buffer or adherent on immobilized fibrinogen were solubilized in 0.8 ml lysis buffer (50 mM Tris, pH 7.4, 10 mM MgCl 2 , 500 mM Nacl, 1% Triton X-100, 0.1% SDS, 0.5% deoxycholate, 10 ⁇ each of aprotinin and leupeptin, 1 mM phenylmethylsulfonyl fluoride, and 200 ⁇ sodium vanadate).
  • 0.8 lysis buffer 50 mM Tris, pH 7.4, 10 mM MgCl 2 , 500 mM Nacl, 1% Triton X-100, 0.1% SDS, 0.5% deoxycholate, 10 ⁇ each of aprotinin and leupeptin, 1 mM phenylmethylsulfonyl fluoride, and 200 ⁇ sodium vanadate).
  • Lysates were cleared at 14,000 rpm for 2 minutes at 4°C, and the supernatant was incubated for 1 hour with 30 ⁇ g purified GST-Rhotekin RhoA-binding domain fusion protein (GST-RBD) bound to glutathione-Sepharose beads as previously described 27. Samples were washed three times with 50 mM Tris, pH 7.4, 10 mM MgCl 2 , 150 mM NaCl, 1% Triton X-100, and then immunoblotted with an anti-RhoA monoclonal antibody. Cell lysates were also immunoblotted with anti-RhoA as loading control.
  • GST-Rhotekin RhoA-binding domain fusion protein GST-Rhotekin RhoA-binding domain fusion protein
  • Lenti-virus was prepared by co-transfection of pLenti6/V5-Dest vector inserted with integrin ⁇ 3 or AAA mutant ⁇ 3 cDNA with pLPl, pLP2 and pLP/VSVG plasmids (In vitro gen) into -90% confluent 293FT cells using Lipofectamine 2000. 48-72 hours after transfection, cell culture medium containing virus was concentrated, titered and stored at -80°C. Bone marrow cells from 6-8 week old integrin ⁇ _/ ⁇ mice (Jackson Laboratories) were isolated aseptically from femurs and tibias.
  • Stem cells were negatively selected by MACS Lineage cell depletion kit (Miltenyi Biotech) and cultured in RPMI 1640 complete medium with 10 ng/ml interleukin-3, 10 ng/ml interleukin-6, 10 ng/ml granulocyte-macrophage colony stimulating factor (GM-CSF), and 100 ng/ml stem cell factor (SCF).
  • 50 multiplicity of infection (MOI) lenti-virus was used to infect mice bone marrow stem cells twice with 6 ⁇ g/ml polybrene. 48 hours after infection, 5xl0 6 stem cells resuspended in PBS were transplanted by retrobulbar injection into irradiated (5Gy) integrin ⁇ _/ ⁇ mice one day after irradiation 8 .
  • Coverslides were pre-coated with 100 ⁇ g/ml fibrinogen and blocked with 5% BSA in PBS.
  • 300 ⁇ ⁇ 3 ⁇ 4 ⁇ 3 -expressing CHO cells (expressing WT or mutant integrins; 1 x 10 5 /ml) or platelets (1 x 10 /ml) suspended in Tyrode's buffer were added to coverslides and incubated at 37°C for various lengths of time.
  • Cells were fixed with 4% paraformaldehyde (PFA) for 10 minutes and permeabilized with 0.1% Triton X-100 in PBS for 2 minutes.
  • PFA paraformaldehyde
  • was immuno-stained with the anti ⁇ 3 monoclonal antibody MAb 15 and Alexa Fluor-546 conjugated phalloidin. The slides were scanned with a Zeiss LSM510 META confocal microscope as previously described 11
  • Image J software was used for quantitation of uncalibrated optical density of Western blot bands. Paired i-test was used for statistic analysis (mean ⁇ SD).
  • Gcc 13 a G protein subunit, directly binds to the cytoplasmic domain of ⁇ 3 , and is required for integrin outside-in signaling leading to c-Src activation, RhoA inhibition, and cell spreading .
  • Wild type ⁇ and the AAA mutant ⁇ in lenti- virus vectors were transfected into bone marrow stem cells isolated from ⁇ _/ ⁇ mice.
  • the transfected bone marrow stem cells were transplanted into ⁇ _/ ⁇ mice following high dose irradiation.
  • Flow cytometric analysis showed that platelets from the recipient mice express similar levels of wild type or AAA mutant ⁇ 3 (Figure 1 IE).
  • Figure 1 IE When the platelets were plated on the integrin ligand fibrinogen, most wild type ⁇ - ⁇ 88 ⁇ 3 ⁇ 4 platelets spread, compared to the ⁇ _/ ⁇ platelets that do not spread. In contrast to platelets expressing wild type ⁇ 3 , the spreading of AAA mutant platelets on fibrinogen was diminished (Figure 11D).
  • CHO cells expressing ⁇ 3 ⁇ 4 3 ⁇ 4 / ⁇ mutant ⁇ 3 were also defective in spreading on fibrinogen compared with wild type ⁇ 3 ⁇ 4 3 ⁇ 4 ⁇ 3 expressing cells ( Figure 12A).
  • CHO cells expressing the ⁇ 3 AAA mutant also showed a defect in the integrin-dependent activation of c-Src, as shown by phosphorylation at Tyr 416 , and abolished the integrin-dependent early-phase transient inhibition of RhoA during cell spreading ( Figure 12B and C).
  • the peptide mP 1 also inhibited integrin outside-in signaling (Figure 14A), but also significantly affected inside-out signaling as indicated by reduced fibrinogen binding (Figure 14B), which is consistent with the previous observations that some residues (particularly F 730 , and W 739 ) in this peptide are critical for talin interaction.
  • Figure 14A integrin outside-in signaling
  • Figure 14B reduced fibrinogen binding
  • These results demonstrate the selectively inhibition of integrin outside-in signaling by the membrane-permeable peptide inhibitors of Gccl3-integrin interaction.
  • These data also demonstrate that these inhibitors also inhibit platelet spreading and aggregation and therefore useful in treating thrombosis.
  • Such selective inhibitors may allow platelet adhesion without dramatic amplification effect of outside-in signaling, and thus are potentially useful as antithrombotics without profound bleeding side effect, compared to currently used integrin inhibitors.
  • Platelet aggregation assays were carried out as follows: Platelet aggregation and secretion was measured in a turbidometric platelet aggregometer (Chronolog) at 37 °C with stirring (1000 rpm). Washed platelets (3 x 10 /ml) in modified Tyrode's buffer were stimulated with thrombin (Enzyme Research Laboratories). For talin knockdown platelet aggregation assay stimulated with manganese and ADP, manganese and ADP was mixed prior to experiment to achieve final concentration of 1 mM manganese and 5 ⁇ ADP in reaction tube. Aggregation traces shown are representative of at least three independent experiments.
  • Clot Retraction Assays were carried out as follows: Similar as described before(2, 3), freshly prepared human whole blood was citrated with 1/10 volume of 3.8% sodium citrate. After centrifugation at 1300rpm for 22min with break, platelet rich plasm (PRP) was collected. Pre-incubated of PRP with 0.05% DMSO (vehicle), 250 ⁇ mP5 peptide, 250 ⁇ mP13 peptide, or their corresponding scrambled control peptides mP5Scr or mP13Scr for 5 minutes at room temperature. After that, 0.5 U/ml thrombin was added into PRP and mixed gently.
  • PRP platelet rich plasm
  • the clots were formed and allowed to retract at 37 °C incubator with C0 2 and were photographed at various times.
  • Myristoylated peptides were synthesized: mPs (Myr-EEERA) and mP 13 (Myr- KFEEERARAKWDT). Control peptides comprising the scrambled sequence mP5 and mP13 were also made. The peptides were tested for inhibiting binding between in Gcc 13 and ⁇ and for inhibiting binding between talin and ⁇ . Both peptides inhibited co-immunoprecipitation between Gcc 13 and ⁇ (Fig 21A), indicating that the minimal sequence EEERA, which includes the ExE motif, is sufficient to bind Gcc 13 .
  • the peptides were also tested for inhibition of platelet spreading on fibrinogen and for inhibition of platelet adhesion to immobilized fibrinogen.
  • the mPs peptide inhibited platelet spreading on fibrinogen (Fig 21C).
  • the inhibitory effect of mPs on platelet spreading was reversed by the Rho kinase inhibitor, Y27632 (Fig. 21C), suggesting that mP 5 inhibited platelet spreading mainly by blocking the Gcc 13 - and c-Src-dependent RhoA inhibitory signaling pathway as characterized in our recent study(S).
  • mP 5 had no effect on agonist-induced fibrinogen binding to platelets (Fig. 21B) nor does it affect platelet adhesion to immobilized fibrinogen (Fig. 21D).
  • peptides were furthermore analyzed for their ability to inhibit clot retraction mediated by platelets.
  • mP5 did not inhibit but rather accelerated integrin-dependent clot retraction mediated by platelets, which requires late phase outside-in signaling.
  • This example demonstrates the design of modified forms of the mP5 peptide (Myr- EEERA).
  • a first set of modified forms of the mP5 peptide are made wherein each peptide of the set retains the EEE motif, but adds 1, 2, 3, 4, 5, or more flanking residues N-terminal to the EEE motif or C-terminal to the EEE motif.
  • the flanking residues are based on the flanking sequences that naturally occur in the ⁇ 3 integrin sequence, or other beta integrin sequences.
  • the modified peptides include, for example, KFEEE, FEEER, AKFEEE, KFEEER, FEEERA, EEERAR, EEERARA, and EEERARAK.
  • two control peptides are synthesized: (1) a scrambled peptide having the same amino acid composition but having a different amino acid sequence, and (2) a loss of function peptide in which the sequence is identical except for each of the ExE residues are changed to alanine.
  • Each peptide is tested for the ability to inhibit talin binding or until the affinity of the peptide peaks.
  • the peptides of the first set are modified to contain the second glutamic acid in the EEE motif to another amino acid. Some will be changed to EAE or EKE. These peptides are subsequently tested for their affinity for Gal3.
  • a third set of modified forms of the mP5 peptide are made wherein the peptide is cyclized. Cyclization can increase the efficiency of delivery and minimize extracellular cleavage of the peptide.
  • the peptides are made with a Cys at each termini (Island C-termini) and reacted under conditions to form a disulfide bridge. The cyclic peptides are subsequently tested for inhibitory effect on Gal3-integrin interaction, biochemical markers of integrin outside-in signaling, platelet adhesion, platelet aggregation, and thrombus formation in vitro.
  • the above sets of peptides are tested for their ability to inhibit Gal3-integrin interaction by an in vitro binding assay using purified G l 3 and purified integrin ⁇ 3 cytoplasmic domain [14] .
  • the recombinant ⁇ 3 cytoplasmic domain-GST fusion protein and control GST protein are immobilized to glutathione-beads.
  • the beads are mixed with recombinant Gal3 in the presence or absence of GTPyS. After washes, bound Gal3 is analyzed by western blot.
  • the modified peptides as well as each of the control peptides are added to the reaction in order to determine the inhibitory effects of these peptides.
  • Modified forms of mP5 are additionally screened by an assay in which modified or control peptides are added to microtiter wells to which the recombinant ⁇ 3 cytoplasmic domain is immobilized. After addition of the peptide to each well, biotinylated Gal 3 is added to each well. HRP-labeled streptoavidin in an ELISA assay is used to determine which peptides inhibited the interaction between Gal3 and the immobilized ⁇ 3 cytoplasmic domain, as those wells containing HRP-labeled streptoavidin bound to biotinylated Gal 3 indicates that the peptide successfully inhibited the binding between Gal3 and the immobilized ⁇ 3 cytoplasmic domain.
  • ExE peptides are additionally tested by constructing a phage library expressing the ExE motif peptides with random flanking amino acid residues, and screen for high affinity binding using microtiter wells coated with Gal3 protein.
  • the phage clones with high affinity binding sequences are sequenced and the corresponding peptide are synthesized for further testing, as described herein.
  • mice are nano-sized particles formed by aggregation of amphiphilic molecules in water (Fig 25). Peptides of the invention attached to a fatty acid may be used to formulate the peptides into micelles.
  • the ExE motif peptide of SEQ ID NO: 87 was made into a micellar formulation, as follows: l,2-distearoyl-s7i-glycero-3-phosphoethanolamine-N- [amino(polyethylene glycol)-2000] (PEG 2000 -DSPE; Northern Lipids Inc., Vancouver, BC), L-a- phosphatidylcholine (egg PC, Type XI-E, Sigma- Aldrich, St.
  • mice were prepared using a film rehydration method, as previously described [21] .
  • the lipid film was rehydrated with 10 mM isotonic HEPES buffer (HEPES 10 mM, NaCl 135 mM, pH 7.4) to form the micelle colloid.
  • the mixture of the lipids and peptides was dissolved in methanol and chloroform, followed by evaporation using a rotary- evaporator R-215 (40 mbar, 45°C, Buchi, New Castle, DE) to form a thin lipid layer.
  • the lipid film was desiccated under vacuum (in the dark) overnight and then rehydrated with 10 mM isotonic HEPES buffer (HEPES 10 mM, NaCl 135 mM, pH 7.4) to form micelle colloid.
  • micellar formulation of FEEERA (SEQ ID NO: 87) was compared to the same peptide dissolved in DMSO. Whereas 250 ⁇ of this peptide dissolved in DMSO was required for the maximal effect on in vitro platelet aggregation, only 4 ⁇ of this peptide was required for the maximal effect when made into a micellar formulation (Figure 29).
  • ACD acid citrate dextrose
  • PRP 3.2% sodium citrate
  • micellar ExE motif peptides or control scrambled peptides Either isolated platelets or PRP is preincubated with increasing concentrations of micellar ExE motif peptides or control scrambled peptides, and, after adding luciferase/luciferin reagents, the peptides are tested for platelet aggregation and simultaneously recorded for secretion of ATP from dense granules using a Lumi-aggregometer (Chronolog).
  • Adenosine triphosphate (ADP), thrombin, PAR4AP, PARIAP, U46619, ristocetin/botrocetin and collagen are used to stimulate platelets.
  • P-selectin exposure which is an indicator of cc-granule secretion, is measured using flow cytometry using anti-p-selectin antibodies [22] .
  • Isolated platelets or PRP were pre-incubated with various concentrations of either a peptide of FEEERA (SEQ ID NO: 87), or a scrambled control thereof, dissolved in DMSO. 0.1 U thrombin was then added to the cells to stimulate ATP secretion. ATP secretion by the cells was then measured using a lumi-aggregometer. % secretion relative to the scrambled control peptide is shown in Figure 27.
  • micellar ExE motif peptides such as mP5 and the peptide of FEEERA (SEQ ID NO: 87)
  • FEEERA peptide of FEEERA
  • Two different types of flow adhesion assays are set up in the laboratory using: (1) a laminar flow chamber and (2) a cone- plate rheometer.
  • Laminar flow chambers are coated with subendothelial matrix proteins, such as collagen, von Willebrand Factor (VWF) or both.
  • VWF von Willebrand Factor
  • Platelets are labeled with a fluorescent dye (mepacrine or 5-chloromethylfluorescein diacetate (CMFDA)), and anti-coagulated blood, PRP or isolated platelets are infused into the chamber at defined flow shear rates with a syringe pump. Adhesion and the formation of platelet-rich thrombi are monitored using an inverted fluorescent dye (mepacrine or 5-chloromethylfluorescein diacetate (CMFDA)), and anti-coagulated blood, PRP or isolated platelets are infused into the chamber at defined flow shear rates with a syringe pump. Adhesion and the formation of platelet-rich thrombi are monitored using an inverted
  • thrombus size is quantitated using a confocal microscope (similar to that described below).
  • PFA-100 is a clinically used test of platelet function that allows passage of blood through a cartridge coated with platelet agonists/adhesive proteins (such as epinephrine/collagen) until the time of thrombotic occlusion occurs in the cartridge [27] .
  • ExE motif peptides like mP5 and the peptide of FEEERA SEQ ID NO: 87) on thrombosis are tested in this apparatus.
  • Blood from healthy donors who have not taken platelet inhibitors for two weeks is anticoagulated with 3.2% sodium citrate, and micellar ExE motif peptides or control peptides are added. The blood samples are then analyzed by PFA-100 assay as essentially described in [27].
  • Bleeding time analysis is an indicator of overall in vivo hemostatic function.
  • the ligand-binding function of integrin is critical for primary platelet adhesion and aggregation and thus important for hemostasis.
  • Bleeding time analysis is carried out as follows: C57BL/6 mice are retro-orbitally injected with micelle formulated ExE motif peptides (such as mP5) and scrambled controls. Distal portions of the mouse tail (5 mm) are amputated with a scalpel and the tail immersed in 0.15 M NaCl at 37 °C as previously described [28] . Time to cessation of bleeding is recorded.
  • the assay is stopped and pressure is applied to the tail to prevent excessive loss of blood.
  • the assay is performed in a double-blinded fashion.
  • bleeding time of wild type mice treated with current integrin antagonists, reopro and integrillin are also tested in comparison with the ExE motif peptides. Integrillin was tested in this fashion (with a saline control and 4 mice per group). The results are shown in Figure 26 with the median value indicated. Bleeding time for mice treated with integrillin exceeded the upper limit of the assay (900 s). Consequently, the assay was terminated and bleeding was stopped via pressure application.
  • ExE motif peptides do not affect the ligand binding function of integrin ⁇ 3 ⁇ 4 ⁇ 3, it is expected that the effect of these peptides on bleeding time should be significantly reduced as compared to current integrin antagonists, which abolish ligand binding to integrins.
  • micellar ExE motif peptides for in vivo use
  • the maximal tolerated doses (MTD) in C57BL mice are determined in order to assess acute toxicity of the ExE motif peptide.
  • An initial dose of 5X the maximal effective dose (as determined by ex vivo study) is injected into 2 mice via tail vein.
  • a lower dose is administered if mice die or show clinical sign of intolerance.
  • a higher dose is administrated. This process is repeated until the MTD is approached.
  • MTD is tested in additional mice for two weeks (5 mice of each gender). It is possible that mice tolerate these micellar peptides well even at high doses well above the effective dose. If this is the case, the peptides are considered safe for in vivo use.
  • ExE motif peptides e.g., mP5
  • FEEERA the peptide of FEEERA (SEQ ID NO: 87)
  • Negative controls include vehicle controls. Additional tests are performed to see if the use of the ExE motif peptides in
  • combination with one or more existing integrin antagonists or anti-platelet drugs produce an additive or synergistic effect on thrombosis.
  • the ferric chloride injury-induced carotid artery thrombosis model has been carried out in C57BL/6 mice [29 ' 30] .
  • FeCl 3 -induced thrombosis is widely used to reflect the role of platelets in the formation of occlusive arterial thrombosis [31] .
  • adult mice are anesthetized by intraperitoneal injection of pentobarbital (120 mg/kg).
  • pentobarbital 120 mg/kg
  • the left common carotid artery is surgically exposed and a miniature Doppler flow probe (Model 0.5VB; Transonic Systems, Ithaca, NY) is placed on the surface of the artery.
  • This assay was carried out with two EXE peptides: FEEERA and the scrambled control thereof (ERAFEE). As shown in Figure 28, the time to occlusion was increased when the mice were given FEEERA as compared to its scrambled control.
  • the cremaster muscle is exteriorized by removing connective tissues.
  • the muscle is fixed as a single sheet on a glass slide on an intravital microscopy tray.
  • Rat anti-mouse CD42b antibody conjugated with Dylight 649 is infused through the jugular cannula in mice for 5 minutes.
  • Platelet thrombus formation is visualized using an Olympus fluorescence microscope with a 60X water immersion objective lens and recorded using a high speed digital camera.
  • Vaso-occlusive state is determined by monitoring red cell flow velocity. It is expected that the ExE motif peptide-treated mice show a significant reduction in occlusive thrombus formation, but minimally affect initial platelet adhesion in response to laser-induced injury.
  • mice The effect of in vivo injection ofmicellar ExE motif peptides on ex vivo platelet function
  • Micelles containing the ExE motif peptides are injected through the tail vein of mice, and after a defined time (such as 5 min, 10 min and 30 min), the mice are anesthetized and their blood drawn. Platelet function is tested in vitro as described herein. These experiments allow the determination of whether injection of ExE motif peptides have an effect on overall platelet function.
  • Platelet aggregation assays were performed with myristoylated peptide mP5, myristoylated peptide of SEQ: ID NO: 87, or their respective scarmbled control peptides, as essentially described herein. Briefly, washed human platelets were preincubated with 25, 50, 100, 250, or 500 ⁇ peptide solubilized in DMSO. The platelets were subsequently induced with 0.09 U/ml thrombin and platelet aggregation was measured in a turbidometric platelet aggregometer.
  • Figure 30A provides a table of scores for each test peptide (mP5, myristoylated peptide of SEQ ID NO: 87) at the indicated dose, wherein the score indicates how well the peptide inhibited platelet aggregation.
  • the myristoylated peptide of SEQ ID NO: 87 was able to inhibit platelet aggregation at lower doses, as compared to the mP5 peptide.
  • the aggregation traces for each test peptide at 250 ⁇ are shown in Figure 30B.
  • Myristoylated peptide inhibitors were synthesized and purified at the Research Resource Center at University of Illinois at Chicago. These include: mPs, mP 6 , mP 13 , and respective scrambled controls: mP 13 Scr (Myr-EEARERKDWAKFT; SEQ ID NO: 69), mP 5 Scr (Myr-EEARE; SEQ ID NO: 71), and mP 6 Scr (Myr-ERAFEE; SEQ ID NO: 413).
  • the peptides were prepared in DMSO for in vitro, and in micellar formulation for in vivo (and in vitro) uses.
  • micellar formulation has a molar ratio of PEG 2000 -DSPE, L-a-phosphatidylcholine, and peptides of 45:5:2, and prepared as previously described 24.
  • mP 6 is similar to mP 6 Scr in uptake by platelets (Figure 43a) and does not cause significant changes in hemogram in vivo ( Figure 43b).
  • Bone marrow stem cells from 6-8 week old integrin ⁇ 3 ⁇ " or C57 mice were infected twice with concentrated lenti-virus containing shRNA or cDNA constructs, then retro-orbitally injected into irradiated recipient mice (5Gy for integrin ⁇ _/ ⁇ mice and 9.6Gy for C57 mice) one day after irradiation .
  • Platelet functional analyses 13 25 flow cytometry 26 , laser-induced cremaster muscle arterial thrombosis 27 and FeC ⁇ -induced carotid arterial thrombosis 28 were performed as previously described. Data were analyzed using i-test or one-way ANOVA.
  • Tail bleeding time analysis was performed as previously described 29 . Time to stable cessation of the bleeding is defined as no rebleeding for 60 seconds. Bleeding exceeding 15 minutes was immediately stopped. Data were analyzed using Mann-Whitney test.
  • Integrin ⁇ _/ ⁇ mice were obtained from the Jackson Laboratory. Talin-l ifl fi , PF4-C mice were kindly provided by Dr. Brian Petrich and Dr. David Critchley' 6 . Animal usage and protocol were approved by the institutional animal care committee of the University of Illinois at Chicago. For all animal experiments, mice with similar age, weight, and sex ratio (1: 1, except for laser-induced thrombosis) were used for control and specific treatment. The individual mice chosen for specific treatment were decided randomly.
  • ITGB 8 -UP 5'- CGTGGATCC ATTAGACAGGTGATACTACAATGG -3';
  • GST- 3 CD and recombinant Gcc 13 purification was described previously 8 .
  • Human talin head domain (THD) cDNA corresponding to N-terminal talin amino acid residues 1-433, was cloned into pcDNA3.1 vector and pMal-C2 vector between EcoR I and Xho I sites.
  • Anti- RhoA antibody was purchased from Cytoskeleton, Inc.; anti-Ga 1 3(sc410), anti-c-Src (scl8), anti- talin (sc7534), and anti-integrin ⁇ 3 (sc6627) antibodies were from Santa Cruz Biotechnology, Inc.; anti-Gcc 13 (26004) was from NewEast; anti-phospho-Src Y 416 antibody was obtained from Cell Signaling; anti-talin (TA205) was from Millipore; anti-talin antibody 8d4 (T3287) was obtained from Sigma; PAC1 antibody (340507) and anti-mouse ⁇ 3 ⁇ 4 3 ⁇ 4 antibody MWReg3 (14- 0411) were obtained from BD Biosciences; anti-human integrin ⁇ antibody MAbl5, LIBS6 and 8053 rabbit serum were kindly provided by Dr.
  • Bound THD or Gcc 13 was estimated with anti-talin or anti-Gcc 13 antibody, horse radish peroxide (HRP)-conjugated secondary antibody, and 3,3',5,5'- Tetramethylbenzidine Substrates (Pierce, 34021). The wells were washed three times with NP40 buffer between each of these steps. The reactions were terminated with 1 M sulphuric acid and measured for OD 45 o. For the competitive inhibition assay, increasing concentrations of THD or Gcc 13 was added to the reactions.
  • HRP horse radish peroxide
  • Platelet aggregation and secretion were measured in a turbidometric platelet aggregometer (Chronolog) at 37 °C with stirring (1000 rpm). Washed platelets (3 x 10 /ml) in modified Tyrode's buffer were stimulated with thrombin (Enzyme Research Laboratories). Aggregation traces shown are representative of at least three independent experiments.
  • platelets or CHO-lb9 cells expressing recombinant integrin CCiib 3 23 were solubilized in NP40 lysis buffer (50 mM Tris, pH 7.4, 10 mM MgCl 2 , 150 mM NaCl, 1% NP-40, 1 mM sodium orthovanadate, 1 mM NaF), with complete protease inhibitor cocktail tablets (1 tablet/5 ml buffer, Roche). Lysis debris was cleared after centrifugation at 14,000g for 10 min.
  • NP40 lysis buffer 50 mM Tris, pH 7.4, 10 mM MgCl 2 , 150 mM NaCl, 1% NP-40, 1 mM sodium orthovanadate, 1 mM NaF
  • Lysates were then immunoprecipitated with rabbit anti-Gcc 13 IgG, anti- integrin ⁇ 3 rabbit serum or an equal amount of rabbit IgG or pre-immune serum for 2 hours before Protein A/G sepharose beads were added. After incubation of Protein A/G sepharose beads for 45min at 4 °C, beads were centrifuged down and washed for six times with NP40 lysis buffer. Immunoprecipitates were analyzed by immunoblotting.
  • Platelets or ccnb 3 -expressing CHO cells in modified Tyrode's buffer or adherent on immobilized fibrinogen were solubilized in cold NP40 lysis buffer at 4°C, and debris-cleared lysates were incubated for 1 hour with purified GST-RBD beads, washed, and then
  • Bone marrow transplantation As previously described , bone marrow stem cells were isolated from femur and tibias of 6-8 week old integrin ⁇ 3 _/ ⁇ , or C57/BL6 mice using the MACS lineage cell depletion kit (Miltenyi Biotec). Stem cells were subsequently infected twice with concentrated lenti- virus containing shRNA or cDNA constructs, as described in Animals and Reagents section, using a Lenti-X concentrator (Clontech). The cells were then retro-orbitally injected into irradiated recipient mice (5Gy for integrin ⁇ _/ ⁇ mice and 9.6Gy for C57/BL6 mice, one million cells per recipient mice) one day after irradiation.
  • irradiated recipient mice 5Gy for integrin ⁇ _/ ⁇ mice and 9.6Gy for C57/BL6 mice, one million cells per recipient mice
  • Washed platelets or ⁇ 3 ⁇ 4i b 3 -expressing CHO cells suspended in modified Tyrode's buffer were added to 100 ⁇ g/ml fibrinogen (Enzyme Research Laboratories)-coated cover slides and incubated at 37°C for various lengths of time.
  • Cells were fixed, permeabilized, blocked with 0.5% bovine serum albumin in modified Tyrode's buffer, stained with mAbl5 (followed by Fluor 488-conjugated anti-mouse secondary antibody) and/or Alexa Fluor-546 conjugated phalloidin, and viewed with a Zeiss LSM510 META confocal microscope, as previously described or with Leica DM IRB fluorescence microscope, Photometries CoolSNAP HQ camera and ⁇ Manager software. Cell surface area was measured by NIH ImageJ analysis of 5-10 random images. Statistical significance was determined using i-test.
  • Myristoylated peptides were synthesized and purified at the Research Resource Center at the University of Illinois at Chicago. These peptides include: mP 13 (Myr- KFEEERARAKWDT (SEQ ID NO: 67)), mP 5 (Myr-EEERA (SEQ ID NO: 414)), mP 6 (Myr- FEEERA (SEQ ID NO: 415)), and the corresponding control peptides mP 13 Scr (Myr- EEARERKDWAKFT (SEQ ID NO: 69)), mP 5 Scr (Myr-EEARE (SEQ ID NO: 71)), and mP 6 Scr (Myr-ERAFEE (SEQ ID NO: 413)).
  • micellar formulation for in vivo (and in vitro) use.
  • micellar formulation PEG 2000 -DSPE, L-a-phosphatidylcholine, and peptides were mixed at a molar ratio of 45:5:2.
  • the micelles were suspended to form micelle colloid in HEPES-saline buffer (10 mM HEPES, 150 mM NaCl, pH
  • peptide concentration 1 mM as previously described 24.
  • mP 6 is similar to mP 6 Scr in uptake by platelets (Figure 43a) and does not cause significant changes in hemogram in vivo ( Figure 43b).
  • mP 6 and mP 6 Scr peptides were dissolved and conjugated with 1-Pyrenyldiazomethane (PDAM) overnight in the dark in DMSO, or conjugated in methanol and incorporated into the micelle as described above. Platelets were incubated with the PDAM-conjugated peptides for 5 minutes at room temperature, pelleted via centrifugation, washed and lysed with NP40 lysis buffer, and the concentration of PDAM-conjugated peptide was estimated by measuring fluorescence intensity (absorption 340 nm/emission 395 nm) as previously described 30. Platelet lysates (without peptide incubation) were used as a blank control. Standard curve was obtained using known concentrations of peptides added to platelet lysates.
  • PDAM 1-Pyrenyldiazomethane
  • thermo-controlled mice Similar to the methods described previously 27 , Wt male mice (6-8 weeks old) were anesthetized via IP injection of ketamine and xylazine and placed on a thermo-controlled blanket (37 °C). The cremaster muscle was exteriorized and superfused with thermo-controlled (37 °C) bicarbonate -buffered saline for the duration of experiments. Fluorescence and brightfield images were recorded using an Olympus BX61W microscope with a 60 x/1.0 NA water immersion objective and a high speed camera (Hamamatsu C9300) through an intensifier (Video Scope International). Fluorescence images were captured at 20 frames per second, and data were analyzed using Slidebook v5.5 (Intelligent Imaging Innovations).
  • Integrins are critical in thrombosis and hemostasis 1 .
  • Antagonists of the platelet integrin ⁇ 3 ⁇ 4 ⁇ 3 are potent anti-thrombotic drugs, but also have the life-threatening adverse effect of bleeding 2 ' 3. It is thus desirable to develop new antagonists that do not cause bleeding.
  • Integrins transmit signals bidirectionally 4 ' 5 .
  • Inside-out signaling activates integrins via a talin- dependent mechanism 6 ' 7 .
  • Integrin ligation mediates thrombus formation and outside-in signaling 8 ' 9 , which requires Gcc 13 and greatly expands thrombi.
  • Gcc 13 and talin bind to mutually exclusive, but distinct sites within the integrin ⁇ 3 cytoplasmic domain in opposing waves.
  • the first talin binding wave mediates inside-out signaling and also "ligand- induced integrin activation", but is not required for outside-in signaling.
  • Integrin ligation induces transient talin dissociation and Gcc 13 binding to an ExE motif, which selectively mediates outside-in signaling and platelet spreading.
  • the second talin binding wave is associated with clot retraction.
  • An ExE motif-based inhibitor of Gcc 13 -integrin interaction selectively abolishes outside-in signaling without affecting integrin ligation, and suppresses occlusive arterial thrombosis without affecting bleeding time.
  • Integrin signaling involves the binding of several molecules to the cytoplasmic domain of integrin ⁇ subunits including talin 6 ' 7 , kindlins 10 ' 11 , c-Src 12 ' 13 , and Gcc 13 8 (Figure 31a).
  • Co-immunoprecipitation of Gcc 13 with various ⁇ C-terminal truncation mutants suggests that Gcc 13 binding involves the ⁇ sequence between K and T ( Figure 31b, Figure 36a), but not the kindlin-/c-Src-binding sequences ( Figure 31a, b).
  • the ExE motif is located in a talin-binding region ( Figure 31a) 14 ' 15 .
  • Purified recombinant THD and Gcc 13 directly competed for binding to purified GST ⁇ 3 cytoplasmic domain fusion protein (GST ⁇ 3 CD) ( Figure 3 If, g), indicating that Gcc 13 and talin are mutually exclusive in binding to ⁇ 3 .
  • inside-out signaling is not the only pathway of ⁇ 3 ⁇ 4 3 ⁇ 4 ⁇ 3 activation. Integrin-fibrinogen interaction may occur independently of inside-out signaling when fibrinogen changes conformation, either by immobilization or conversion to fibrin 18 ' 19 . This is because the initial contact of the exposed ligand recognition sequence, RGD, with resting integrins triggers "ligand-induced integrin activation" 20. Interestingly, adhesion of resting talin- knockout or -knockdown platelets to immobilized fibrinogen was defective ( Figure 32f, Figure 38b), indicating the importance of talin in platelet adhesion to immobilized fibrinogen in the absence of inside-out signaling.
  • the ExE motif is not required for talin-dependent inside-out signaling.
  • the AAA mutant ⁇ - ⁇ 88 ⁇ 3 ⁇ 4 platelets were defective in spreading on immobilized fibrinogen ( Figure 33d, Figure 39c, d).
  • Gcc 13 binding deficiency in ⁇ 3 causes a selective defect in integrin outside-in signaling and platelet spreading.
  • AAA mutant ⁇ expressed in CHO cells had no negative effect on THD binding, in contrast to the Y747A mutant ( Figure 40d, e).
  • AAA-expressing cells showed defects in integrin-dependent activation of c-Src (as shown by phosphorylation at Tyr 416 ) and transient inhibition of RhoA during cell spreading (Figure 33g, Figure 40f), both of which are important elements of outside-in signaling.
  • ExE-based inhibitor ⁇ ⁇ ⁇
  • mP 1 inhibited inside-out and outside-in signaling, as it inhibited fibrinogen binding (Figure 41h), platelet adhesion (Figure 34d) and clot retraction (Figure 34e) (not reversed by manganese, as previously shown using talin "7" platelets 16 ).
  • mP 6 selectively interferes with the early phase of outside-in signaling, but mP 1 affects all phases of integrin signaling.
  • mP 6 inhibited the second wave of thrombin-induced platelet aggregation in vitro ( Figure 34f), and when injected into mice as micelles, was as potent as the currently used integrin antagonist Integrilin in inhibiting laser-induced arteriolar thrombosis ( Figure 34g, h, Figure 42a, b) and FeCl 3 -induced occlusive carotid artery thrombosis (Figure 34i, Figure 42c).
  • This example demonstrates selective inhibition of outside-in signaling of shorter peptides, mP5 and mP6, in comparison to longer peptides, mP7 and mP13.
  • the structures of mP5, mP6, and mP13 are as described in Examples 20 and 21.
  • the structure of mP7 is KFEEERA (SEQ ID NO: 68), wherein the Lys at the C-terminus is myristoylated in similar fashion to myristoylated mP5, mP6, and mP13.
  • the structure of the scrambled versions of mP5, mP6, mP7, and mP13 are Myr-EEARE (SEQ ID NO: 71) , Myr-ERAFEE (SEQ ID NO: 413), Myr-ERKAFEE (SEQ ID NO: 416), and Myr-EEARERKDWAKFT (SEQ ID NO: 69).
  • the second wave of platelet aggregation requires integrin outside-in signaling (a consequence of first wave fibrinogen binding and platelet aggregation), platelet granule secretion, and consequential amplification of platelet activation and inside-out signaling and further fibrinogen binding.
  • mP5 and mP6 selectively inhibits the early phase of outside-in signaling without affecting talin-dependent inside-out signaling, ligand induced integrin activation, or the late phase of outside-in signaling associated with the second wave of talin binding.
  • mP7 and mP13 inhibited inside-out and outside-in signaling , as it inhibited fibrinogen binding (Figure 44), totally inhibited platelet aggregation ( Figure 45), and inhibited platelet adhesion (Figure 46).
  • mP13 have the intermediate effects between mP6 and mP13, partially inhibiting fibrinogen binding and platelet adhesion. Therefore, use of shorter peptides, like mP5 and mP6, optimizes the selective inhibition of occlusive thrombosis without a bleeding risk.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des composés qui inhibent une interaction de liaison entre une intégrine β et une sous-unité de protéine G, ainsi que des compositions, par exemple des compositions pharmaceutiques, comportant celles-ci, et des nécessaires associés. Dans certains modes de réalisation, le composé est un anticorps ou un analogue d'anticorps et, dans d'autres modes de réalisation, le composé est un peptide ou un analogue de peptide. L'invention concerne également des procédés d'utilisation des composés, comprenant des méthodes de traitement ou de prévention d'un état médical, tel qu'un accident vasculaire cérébral, une crise cardiaque, le cancer ou une inflammation.
EP13783707.6A 2012-09-15 2013-09-13 Inhibiteurs des interactions de liaison de la sous-unité alpha de protéine g-intégrine bêta Withdrawn EP2895508A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US13/621,064 US8685921B2 (en) 2010-03-15 2012-09-15 Inhibitors of beta integrin-G protein alpha subunit binding interactions
PCT/US2013/059833 WO2014043594A1 (fr) 2010-03-15 2013-09-13 Inhibiteurs des interactions de liaison de la sous-unité alpha de protéine g-intégrine bêta

Publications (1)

Publication Number Publication Date
EP2895508A1 true EP2895508A1 (fr) 2015-07-22

Family

ID=49510486

Family Applications (1)

Application Number Title Priority Date Filing Date
EP13783707.6A Withdrawn EP2895508A1 (fr) 2012-09-15 2013-09-13 Inhibiteurs des interactions de liaison de la sous-unité alpha de protéine g-intégrine bêta

Country Status (2)

Country Link
EP (1) EP2895508A1 (fr)
CN (1) CN104995207B (fr)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106226535B (zh) * 2016-08-01 2018-03-09 中国科学院广州生物医药与健康研究院 Cd61作为生血内皮细胞标志物的用途
WO2018231567A1 (fr) * 2017-06-13 2018-12-20 Croda, Inc. Compositions d'électrolyte agrochimiques
WO2022183360A1 (fr) * 2021-03-02 2022-09-09 Tsao Yeou Ping Peptides synthétiques courts et leurs utilisations pour le traitement de la sécheresse oculaire
CN113171440A (zh) * 2021-04-26 2021-07-27 石家庄喜得宝医疗器械有限公司 一种生物蛋白敷料组合物及其制备方法

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2575160T3 (es) * 2010-03-15 2016-06-24 The Board Of Trustees Of The University Of Illinois Inhibidores de las interacciones que unen la subunidad alfa de la beta integrina-proteína G

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2014043594A1 *

Also Published As

Publication number Publication date
CN104995207A (zh) 2015-10-21
CN104995207B (zh) 2018-08-07

Similar Documents

Publication Publication Date Title
US20210054026A1 (en) Inhibitors of Beta Integrin-G Protein Alpha Subunit Binding Interactions
US9029502B2 (en) Inhibitors of the epidermal growth factor receptor-heat shock protein 90 binding interaction
US11827712B2 (en) IL13Rα2 binding agents and use thereof
EP3250611B1 (fr) Lymphocytes t à récepteur d'antigène chimérique (car) reconnaissant le récepteur il 13r-alpha2 spécifique au cancer
BR112020017709A2 (pt) Anticorpo anti-sirpa humana, fragmento de ligação a antígeno do mesmo ou anticorpo modificado do mesmo, composição farmacêutica, uso de um polipeptídeo, métodos para preparar um anticorpo anti-sirpa humana v1, para aumentar a apresentação cruzada de um antígeno e para avaliar a probabilidade de eficácia de um tratamento, método in vitro ou ex vivo para avaliar a probabilidade de eficácia de um tratamento, e, combinação de compostos
JP2020055885A (ja) 赤血球レベルを増加させ、そして鎌状赤血球症を処置するための方法
US10611798B2 (en) PAR1 and PAR2 c-tail peptides and peptide mimetics
JP2016539121A (ja) 血管新生抑制活性を有するペプチド、及びそれを含む組成物
US11999798B2 (en) Anti-factor IX padua antibodies
EP2895508A1 (fr) Inhibiteurs des interactions de liaison de la sous-unité alpha de protéine g-intégrine bêta
JP2021512919A (ja) 異所性骨化を処置するための方法
WO2011080401A1 (fr) Compositions de liaison à un récepteur tyrosine kinase
WO2018112226A1 (fr) Polypeptides à base de sharpin et leurs utilisations
CN108290930A (zh) Pink1 c末端结构域多肽及其用于癌症治疗的方法

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20150414

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20160627

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20161108